当前位置: 首页 > 医学版 > 期刊论文 > 内科学 > 内分泌进展 > 2005年 > 第1期 > 正文
编号:11168582
Endocrine Control of Body Composition in Infancy, Childhood, and Puberty
http://www.100md.com 内分泌进展 2005年第1期
     Division of Endocrinology and Metabolism (J.D.V.), Department of Internal Medicine, Mayo Medical and Graduate Schools of Medicine, General Clinical Research Center, Mayo Clinic, Rochester, Minnesota 55905

    Department of Pediatrics (J.N.R.), State University of New York at Buffalo, Buffalo, New York 14214-3000

    Departments of Pediatrics and Internal Medicine (E.J.R., A.D.R.), General Clinical Research Center, University of Virginia School of Medicine, Charlottesville, Virginia 22903

    Pennington Biomedical Research Center (J.C.L.), Baton Rouge, Louisiana 70808; Division of Endocrinology (M.S.-M.), Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas 77555

    Department of Pediatrics (N.M.), Nemours Children’s Clinic, Jacksonville, Florida 32207

    Division of Endocrinology and Metabolism (C.Y.B.), Department of Internal Medicine, Tulane University Medical Center, New Orleans, Louisiana 70112

    Abstract

    Body composition exhibits marked variations across the early human lifetime. The precise physiological mechanisms that drive such developmental adaptations are difficult to establish. This clinical challenge reflects an array of potentially confounding factors, such as marked intersubject differences in tissue compartments; the incremental nature of longitudinal intrasubject variations in body composition; technical limitations in quantitating the unobserved mass of mineral, fat, water, and muscle ad seriatim; and the multifold contributions of genetic, dietary, environmental, hormonal, nutritional, and behavioral signals to physical and sexual maturation. From an endocrine perspective (reviewed here), gonadal sex steroids and GH/IGF-I constitute prime determinants of evolving body composition. The present critical review examines hormonal regulation of body composition in infancy, childhood, and puberty.

    I. Timing and Tempo of Normal Human Growth

    A. Linear growth and body weight

    B. Body composition in childhood and puberty

    II. Measurement of Body Composition

    A. Body mass index

    B. Two-compartment models

    C. Four-compartment models

    D. Two- and three-compartment models

    E. Fat topography

    F. Clinic and field methods

    III. Sex-Steroid and GH Interactions on Target Tissues in Puberty

    A. Overview

    B. Actions of androgen, estrogen, GH, and IGF-I on bone

    C. Adiposity and sex-steroid hormones

    D. Adiposity and GH/IGF-I

    E. Control of muscle by sex steroids and GH/IGF-I

    IV. Energy Expenditure in Puberty

    V. Summary

    I. Timing and Tempo of Normal Human Growth

    A. Linear growth and body weight

    AUXOLOGY [Gk. auxesis, to increase or grow; logos, study] is the scientific analysis of physical growth and development. Growth is a complex process that is sustained throughout in utero development, infancy, childhood, puberty, and early adulthood. Dynamic control of growth is endowed by age- and gender-dependent interactions among key genetic, environmental, dietary, socioeconomic, developmental, behavioral, nutritional, metabolic, biochemical, and hormonal factors. Thus, normative data must be developed from age-specific, gender-matched, and genetically comparable healthy populations.

    Although normative isobars are widely used for comparisons of static height, the endocrinologist and pediatrician should also evaluate the velocity of linear growth velocity (annual increment in height), chronological and apparent biological age, pubertal status, family history, and psychosocial adjustment. From a clinical perspective, biological age is often assessed indirectly as radiographic bone age.

    The velocity of in utero linear growth is maximal at about 18 wk of gestational age in the human. At this time, the fetus grows four times more rapidly than at any time postnatally. Increases in body weight follow a similar temporal pattern, except that the zenith occurs at about 34 wk. The growth rate declines sharply during the last weeks of gestation. The maternal-placental environment dictates the infant’s birth weight more than the fetal genotype (1). In the newborn, height velocity adjusts toward the genetically predicted trajectory. Linear growth averages approximately 25 cm in the first year and 12.5 cm in the second year of life (see Refs.2 and 3 for distributional estimates). The annual height velocity decreases to 8 cm (ages 2–4 yr) and 6 cm (ages 4–6 yr) during childhood. A plateau-like phase emerges in midchildhood, wherein height velocity approaches 5.5 cm/yr before puberty. Especially in the male, there is an incompletely understood decline in height velocity before onset of the pubertal growth spurt.

    1. Sex differences in the fetal period.

    Unborn humans exhibit two gender-related auxological distinctions: 1) males exhibit more rapid linear growth than females early in utero; and 2) girls manifest greater skeletal maturation than boys after 15 wk of gestational development. For example, the crown-rump length in boys exceeds that in girls by 1.0 mm at 8 wk and by 2.6 mm at 14 wk gestation (4). Ultrasonographic records of fetal head circumference show an analogous gender difference early in development. At term, the foregoing sex-related distinctions approach 2% of the population mean. Conversely, skeletal maturation (e.g., defined by radiological bone age) proceeds more rapidly in the female than male fetus, which disparity yields a bone age advance of 1.5 wk in girls by the early third trimester of pregnancy (5). Weight diverges in the sexes at approximately 24 wk of gestational age, such that boys weigh 70 g more than girls at 30–32 wk of in utero life. The absolute male-female weight difference approximates 130 g (4% of the mean) at birth.

    2. Sex differences in the postnatal period.

    Figure 1 presents population-based projections of linear growth velocity by gender in North American children. Healthy cohorts are heterogeneous in genetic background, biological development, nutrition, exercise, and psychosocial adaptation. Accordingly, in an effort to incorporate expected genetic nonuniformity in height trajectories, normative data include observations in children destined to become relatively tall or short as adults.

    In the first one-half year of life, boys gain height more rapidly than girls. This velocity difference recedes after 8 months of age (2). During the age interval of 1–4 yr, girls increase in stature slightly more rapidly than boys. Thereafter, mean linear growth velocities converge in the sexes until approximately age 9, when girls (but not boys) begin a pubertal growth spurt. In North American and West European cohorts, during the interval of 9–14 yr of age, girls on average are taller than boys (3).

    Girls attain a peak height velocity of 8.3 cm/yr at an average chronological age of 11.5 yr. This growth milestone corresponds to pubertal Tanner breast stages 2 and 3. Boys gain height at a prepubertal rate until age 11 (instead of 9), when testis volume begins to increase beyond 7–10 ml. Adolescent males then achieve a peak height velocity of 9.5 cm/yr at about 13.5 yr of age. The latter chronology coincides with pubertal genital stages 3 and 4. Maximal height velocity, but not total duration of linear growth, tends to be greater in youths who mature early. In both sexes, the pubertal growth rate declines rapidly after the gender-specific zenith; e.g., girls gain 1 cm/yr or less in height after age 14.5 yr, and boys gain 1 cm/yr or less after age 17 yr. The net pubertal increment in stature in the male exceeds that in the female by 3–5 cm in Western cultures. Accordingly, the mean adult height difference of 13 cm between the sexes primarily reflects the gain of an additional 8–11 cm during a more prolonged prepubertal interval (2 additional years) in boys.

    3. Interindividual auxological variations.

    Height isobar projections (static distance curves) are shown for both sexes in Figure 2. Such population-defined data belie significant nonuniformities among individual children in the timing (onset) and tempo (rate) of sexual maturation and attendant physical development. Known genetic and environmental factors predispose to pubertal pathophysiology (6, 7, 8, 9, 10, 11, 12). However, precisely how heredity and environment control normal variations in physical maturation in healthy individuals is less well understood (13). Mechanistic considerations include mutations or microsequence polymorphisms of genes encoding (at least) the LH ?-subunit, the aromatase enzyme, and the GH, LH, leptin, glucocorticoid, estrogen, and androgen receptors (14, 15, 16, 17, 18, 19). Additional studies will be important to elucidate the impact of molecular diversity on physical, sexual, and psychological phenotypes.

    Standardized growth curves assume a population-based mean timing of pubertal onset and progress. However, any given child may exhibit a delay or advance in sexual maturation and thus diverge at least temporarily from group predictions (2, 20, 21). A relevant family history should help in interpreting the clinical significance of serial growth measurements. As an additional aid, reference height-velocity predictions are available for average, early-, and late-maturing children in Northern Europe (3).

    Normative weight trajectories are illustrated in Figure 3. Newborns lose approximately 10% of birth weight over the first 7–10 d of life. The exact adaptive processes that mediate evident extracellular fluid loss and inferred tissue catabolism at this time have not been articulated fully, but presumptively entail combined nutritional and endocrine factors. Healthy neonates overcome the expected weight deficit within several weeks, and then gain approximately 30 g/d during the first 3 months of life. The latter mean increment declines to 20, 15, and 12 g/d over successive quarterly intervals until age 1 yr.

    In the first year of life, male infants gain weight slightly more rapidly than female counterparts, such that at 12 months of age boys weigh an average of 10 kg and girls 9.5 kg. Over the next 2 yr, mean weight velocity approximates 8 g/d in both sexes. Weight gain diminishes to 6–7 g/d (2 or 2.5 kg/yr) in midchildhood. At age 7 yr, boys usually weigh 23 kg and girls 22 kg. Weight velocities accelerate by nearly 2-fold in the gender-specific years of puberty, wherein males gain 5 kg/yr (13.7 g/d) and girls 4.2 kg/yr (11.5 g/d).

    B. Body composition in childhood and puberty

    Extended, prospective, ethnicity-specific, and population-based normative body composition data stratified by gender in childhood are lacking. However, important (albeit longitudinally delimited and/or cross-sectional) observations are available in the fetus, neonate, child, and adolescent (22, 23, 24, 25, 26, 27, 28). Comprehensive body-compositional investigations will require the use of validated quantitative procedures, minimal (if any) radiation exposure, high procedural reproducibility, and repeated application in randomly selected cohorts of healthy children.

    Accurate estimates of and (population-based) statistical boundaries for fat mass (FM) are crucial to classify children accurately as lean, normal, overweight, or obese. Analogously, reliable quantitation of fat-free mass (FFM) is important to identify relative or absolute sarcopenia and osteopenia. Valid measures of regional adiposity (e.g., sc and visceral fat) are essential to elucidate the pathophysiological basis and clinical impact of hyposomatotropism, insulin resistance, dyslipidemia, obesity, and cardiovascular morbidity (see Section II.A).

    1. FM and FFM accrual.

    Projections of the gender-specific evolution of FM, FFM, and percentage body fat in Caucasian children are given in Fig. 4. These predictions aggregate the results of accurate multicompartmental analyses performed cross-sectionally at selected stages in infancy, childhood, puberty, and early adulthood (22, 29, 30). Interpolations are required to supplement incomplete body-compositional data in midchildhood and early adolescence. Although ethnic comparisons are limited, one pediatric investigation compared FM and FFM estimates among African-, European-, and Mexican-American children at or over the age of 4 yr (31) (Fig. 5). This analysis like several recent other studies reported a higher mean FM value than that typically observed earlier in children (32). Whether the latter (possible) increase in absolute FM reflects de facto historical trends, population selection, and/or technical differences is not clear.

    The newborn boy has about 6.5% more (absolute) FFM than the newborn girl (33, 34). This gender difference mirrors the relative paucity of total body fat in the infant male (13%), compared with the female (15%), and the male’s higher birth weight. FM increases to 25–30% of total body weight by age 6 months. Thereafter, FFM begins to accumulate preferentially. For example, 85% of the total weight gain over the second 6 months of life comprises FFM. Although fractional FFM remains comparable by gender across midchildhood, boys accrue about 1 kg more absolute FFM than girls before puberty (22, 29, 30). In puberty, boys acquire FFM at a greater rate (kilograms per year) and for a longer period than girls. In one analysis, stable (adult) values of FFM were attained by approximately 15–16 yr of age in girls and 2–3 yr later in boys (35).

    In absolute terms, FM (kilograms) is comparable by sex in children ages 3–5 yr. Girls accumulate FM more rapidly than boys in midchildhood, such that 10-yr-old females have approximately 2 kg (6%) more FM than males. In adolescence, girls gain absolute FM at an average annual rate of 1.14 kg, whereas boys maintain a relatively fixed absolute FM. Hence, percentage body fat declines in pubertal boys (27).

    2. Water, protein, and mineral accrual.

    Primary components of FFM (water, protein, and mineral) vary markedly in infancy and adapt further in childhood and adolescence (Fig. 6). The percentage of total body water (TBW) normalized to FFM (TBW/FFM) exceeds 80% at birth. The latter value decreases by 1% over the first year of life. TBW/FFM falls to 77% in boys and 78% in girls in early childhood and to 76% by age 10 in both sexes (22, 25, 29).

    During the age interval of 10.5–12.5 yr, girls maintain a lower mean TBW/FFM than boys. This sex difference wanes until boys begin (and girls complete) puberty (25, 29, 30). Protein constitutes 15.7% of FFM at birth. The latter value increases to 18 and 19% at 2 and 10 yr of age, respectively, and approximates 20% in late adolescence (22, 25, 29).

    Mineral comprises primarily (82%) bone salts. The mineral fraction in FFM remains stable in infancy and early childhood, and then rises disproportionately (over protein and water) in midchildhood and early puberty (22, 25, 29). Bone mineral density (BMD) determined at near-peak height velocity is greater in boys than girls (25, 29, 30). BMD is higher in African-American than Caucasian individuals before and after puberty in both sexes (36, 37). The precise endocrine determinants of this consistent ethnic difference are not known. Nonendocrine genetic and environmental factors may contribute to some differences. One analysis revealed higher (overnight) serum concentrations of GH and estradiol in African-American than Caucasian men, which levels correlated positively with BMD (36). No comparable distinction was evident before puberty or in women (37). Other clinical studies have reported ethnic contrasts in plasma IGF-I/IGF binding protein (IGFBP) concentrations in the female (37, 38, 39). The foregoing epidemiological observations highlight the need to better understand the specific mechanisms by which ethnicity, gender, and developmental age modulate the endocrine control of human growth and body composition (40).

    II. Measurement of Body Composition

    Body composition evolves dramatically in utero and across infancy, childhood, puberty, and adulthood and appears to be conditional on early developmental events (28, 34, 35, 41, 42, 43, 44, 45). Quantitation of body composition relies on auxological or anthropological attributes [e.g., body mass index (BMI), skinfold thickness, abdominal girth], physical properties (e.g., total body volume, bioelectrical impedance, dual-energy x-ray absorbance), and/or biochemical markers (e.g., TBW, calcium, potassium, or nitrogen). Clinical measures are then applied in empirically constructed regression (correlation or prediction) equations to estimate unobserved features of body composition (26, 35, 46, 47, 48, 49, 50).

    Primary estimates of body composition were derived by chemical analyses of adult cadaveric tissues (27, 31). Such data, albeit limited, provide validation for secondary estimates based on densitometry (e.g., underwater weighing), dual-energy x-ray absorptiometry (DEXA), isotope dilution, bioelectrical impedance, BMI, and skinfold thickness (22, 24, 26, 27, 29, 31, 46, 47, 51, 52).

    A. Body mass index

    Height (meters) and weight (kilograms) are simple anthropological attributes. Algebraic combinations of these two measures are used to compute the BMI (kilograms per square meter), ponderal index (kilograms per cubic meter) or Benn index (kilograms per meter) (53). BMI has been applied to categorize children as lean, normal, overweight, or obese (54, 55). However, this metric varies with developmental age, gender, and ethnicity (27, 55, 56, 57, 58). For example, BMI is high in the first year of life, decreases in early childhood (ages 2–5 yr), and then increases in puberty (54, 59). Accordingly, BMI should be compared via age-stratified standardized z-scores (or percentiles) defined in healthy populations, e.g., as reported in North America, Holland, United Kingdom, France, and China (60, 61, 62, 63).

    BMI does not quantitate body composition. Indeed, this metric amalgamates frame size (mineral content), total FM (visceral and sc) and lean tissue (27, 28, 30, 52, 64). Thus, a short, muscular adolescent could be assigned a high BMI spuriously suggestive of obesity (47, 59). Moreover, treatment with recombinant human (rh) GH often reduces FM by 2–3 kg and increases lean body mass comparably in the hypopituitary adult, while leaving BMI unchanged (65).

    Indices like BMI also do not monitor the regional fat distribution (e.g., visceral vs. sc) (57, 66, 67, 68). This distinction is significant epidemiologically, because visceral fat accumulation predicts higher risk of peripheral insulin resistance, dyslipidemia, adult cardiovascular disease, hypoandrogenemia, elevated free (salivary) cortisol, reduced concentrations of SHBG, IGFBP-1, LH, and high-density lipoprotein, and impoverished daily GH production (22, 26, 28, 69, 70, 71, 72, 73, 74, 75, 76). Recent investigations suggest that deficiency of intrauterine growth factors, degree of fetal stress, low birth weight, relative hypercortisolemia, impaired glucose disposal in midchildhood, and premature adrenarche further forecast greater risk of insulin resistance, cardiovascular disease, dyslipidemia, and abdominal obesity in adulthood (39, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86).

    B. Two-compartment models

    1. Densitometry.

    Densitometric methods partition body composition into two mutually exclusive compartments, viz., FM and FFM. Calculations relate whole-body density (weight divided by volume) to FM and FFM by way of average tissue-density constants (24, 87). To estimate whole-body density, weight is quantitated accurately on a dry scale, and volume is estimated by underwater weighing, clinical volumetry, or air plethysmography (26).

    Water-displacement procedures are based on the principle of Archimedes, and thus require: 1) complete submersion of the volunteer in a suitable water-filled chamber to record underwater weight (hydrodensitometry) or quantitate water overflow into a burrette (clinical volumetry); and 2) accurate measurement of functional residual lung capacity by nitrogen washout to correct for the thoracic gas space. The latter determination introduces the majority of technical variability into the final estimate of percentage body fat. Within-subject coefficients of variation are approximately 3–4% of total body weight (26, 88). Limitations of hydrodensitometry include the requirement for a water tank, variable subject reluctance, and multiple (up to 10) submersions to ensure technical reproducibility.

    Air-displacement plethysmography provides a complementary volumetric approach based on Boyle’s law of the partial pressure of gases. This procedure may be less stressful to the subject than repeated immersion in a water chamber (89, 90). One plethysmographic unit comprises a sealed fiberglass capsule (or pod). The volunteer enters the chamber wearing a tightly fitting swimsuit and swim cap, views the room through a small window, and breathes quietly for several minutes while an internal diaphragm is oscillated to generate small changes in air pressure. The air-displacement estimate is also corrected for thoracic gas volume (above). Cross-validating analyses indicate that air- and water-displacement methods perform comparably in young adults. However, air plethysmography may underestimate percentage body fat by 2–7% of total body weight (2–6 kg absolute FM) in children and older individuals (45, 52, 89, 90, 91).

    In densitometric techniques, one calculates percentage body fat from the density estimate using an empirical regression model, such as that of Brozek et al. (51) or Siri (92). Both sets of equations assume a nominal adult tissue density of 0.9 g/ml for fat and 1.1 g/ml for FFM (24, 31). However, the use of adult tissue-density constants forces an overestimate of percentage body fat in children (Fig. 7). This artifact arises because the true density of FFM is as low as 1.063 g/ml in early childhood, whereas the contribution of water (density, 0.9937 g/ml) and mineral (density, 3.0 g/ml) to body density is higher and lower, respectively (27, 30). Accordingly, Lohman and colleagues suggest the use of age-specific tissue-density constants in the Siri model (24, 27). This adjustment obviates systematic overestimation bias in younger subjects. However, compared with multicompartmental methods (below), densitometry may yield inconsistent individual predictions (random procedural bias) (30).

    2. Isotope dilution methods.

    Accurate quantitation of TBW facilitates reliable determination of body composition, because water represents 74–80% of FFM depending on age and gender (29). TBW is quantitated by the degree of dilution in the aqueous compartment of a known amount of a stable or radioactive isotope of water (e.g., 2H2O, H218O, or 3H2O) administered orally. Isotope concentrations are monitored in one or more timed (postequilibration) samples of serum, urine, saliva, or expired air by liquid scintigraphy (3H2O), infrared spectrometry (2H2O), or isotope-ratio mass spectroscopy (2H2O or H218O) (68). Estimates are corrected for nonaqueous loss, because 4% of labeled hydrogen exchanges with nonaqueous hydrogen and 1% of labeled oxygen are removed via metabolic oxidation. The degree of final isotope dilution is proportionate to TBW. Given an estimate of TBW, one may calculate FFM and percentage body fat from age- and sex-specific constants for TBW/FFM (above). In isotope dilution studies, the coefficient of variation in the calculation of percentage body fat approximates 2–3% of body weight (31, 92).

    C. Four-compartment models

    Multicompartmental models are used to quantitate FM and the principal components of FFM (water, mineral, and protein) (29, 30, 59, 93, 94, 95, 96, 97, 98, 99). For example, one method determines TBW by isotope dilution (above) and quantitates FM and mineral mass by DEXA. Some compartmental models include the determination of body cell mass (e.g., appraised by nonradioactive potassium spectrometry) or total body nitrogen and calcium (e.g., assessed by whole-body neutron activation analysis) (100, 101, 102, 103, 104, 105).

    D. Two- and three-compartment models

    Two-compartment analyses of body composition use several means to evaluate the components of FFM (26, 46). In the water-density model, TBW is quantitated first to calculate FM (total weight minus TBW). Protein and mineral content of FFM are estimated secondly from age- and gender-specific prediction equations. In the mineral-density model, bone mineral content is determined so as to compute summed water and protein (mineral-free lean tissue) and FM (29). The water-density model performs more reliably in pediatric age groups, because water represents 73–80% (and mineral only 5%) of FFM in children (30). The mean bias of the water-density calculation of FM is approximately 0.75% when calibrated against four-compartment methods. On the other hand, the mineral-density model may overestimate percentage body fat by as much as 5–7.5% in individual children and adolescents (Fig. 8).

    DEXA scanning is a contemporary three-compartment model. DEXA quantitates FM, mineral and mineral-free lean tissue (protein and water) based on differential tissue absorption of two distinct x-ray energy peaks (31, 35, 43, 50). The volunteer lies supine on a table under a detector panel placed over the x-ray source. X-rays are fractionated into 40 keV and 70–100 keV energy streams. Approximately 40–45% of recorded pixels (unit absorbance ratios) monitor x-ray attenuation by bone and soft tissue (combined), and 55–60% monitor x-ray absorbance by fat and mineral-free lean tissue (combined) (31). Prediction bias arises from DEXA determinations made near the surface of the body (due to disproportionate proximity of mineral and sc fat) and the appendicular skeleton (due to undue contiguity of muscle and connective tissue).

    DEXA precision is higher when applied to calibration phantoms than to the human skeleton or the whole body (96, 106). In adults, the reproducibility of DEXA-based quantitation of BMD averages 0.7% or 0.01 g/cm2; and, the absolute error in percentage body fat approaches ± 1.4% of body weight. The latter precision compares well with a value of ±1% in predicting absolute FM by four-compartment models (96, 107). In adults, estimates of percentage body fat based on DEXA usually fall within 3% of those determined by more complex models. Discrepancies typically reflect technical uncertainty in the DEXA calculation of body weight (which should agree with the scale weight within 1 kg) and/or errors in the isotopic determination of TBW.

    DEXA scanning tends to predict falsely high percentage body fat in children (and older adults) (30, 95) (Fig. 8). DEXA likewise overestimated FM in two recent primary validation studies using the whole carcass of immature swine (108, 109). Practical limitations include equipment and technician costs and low-dose radiation exposure (1–3 mrad, or less than that contributed by cosmic background during a single 4000-km air flight). Nonetheless, DEXA technology offers a valuable means to estimate body composition. Additional important insights are achievable by way of computed tomography (CT) and magnetic resonance imaging (MRI), because these techniques allow one to appraise the regional distribution of fat.

    E. Fat topography

    Intraabdominal fat is a key epidemiological determinant of insulin resistance and cardiovascular risk (110). CT provides one well-validated means to quantitate intraabdominal adiposity. CT is technically precise in discriminating adipose tissue and affords a brief scan time that obviates motion artifact (111). To estimate abdominal fat, the CT examination is performed at the level of the fourth or fifth lumbar vertebrae, the corresponding intervertebral disc space, or (somewhat less reliably) the umbilicus. Data are expressed as the cross-sectional area (square centimeters) of a demarcated region of adipose tissue, such as visceral (mesenteric, pericolic, and perirenal), retroperitoneal, and sc fat (112, 113). A recent distinction between superficial and deep sc FM suggests that the latter may also predict increased cardiovascular health risk (114). MRI offers a complementary method to quantify regional FM that does not require x-ray exposure. Although not evaluated exhaustively in children, MRI outcomes correlate with those of CT (115). Table 1 summarizes available CT and MRI data in children as distinguished by peripubertal age, gender, and ethnicity in cross-sectional analyses. However, appropriately stratified longitudinal comparisons will be required to definitively assess the transpubertal control of regional fat distribution in girls and boys. Concomitant metabolic implications of visceral and sc accumulation and dissipation (e.g., peripheral insulin sensitivity and lipoprotein composition) will be important to quantitate so well as practicable in pediatric populations. Finally, anatomic and metabolic adaptations across puberty need to be correlated with changing hormone outflow (viz., GH, testosterone, estradiol, IGF-I, insulin, and leptin) or resting energy expenditure.

    F. Clinic and field methods

    Body composition may be assessed clinically by physical anthropometry (e.g., BMI, the waist-to-hip ratio, anteroposterior abdominal dimension, skinfold thickness) and/or bioelectrical impedance (34, 49, 116, 117, 118, 119). In some studies, average predictions of percentage body fat derived from summed skinfold thickness agree well with multicompartmental analysis (Fig. 9). However, anthropometric assessments may exhibit significant inter- and intraindividual variability (random bias), and bioelectrical impedance estimates may manifest marked (>25%) systematic bias compared with multicompartmental analyses (34, 75, 119, 120, 121).

    III. Sex-Steroid and GH Interactions on Target Tissues in Puberty

    A. Overview

    From an endocrine vantage, normal physical growth and sexual maturation require time-evolving coordination among the somatotropic, gonadotropic, and adipostat systems (122, 123, 124, 125, 126). Time-varying somatic, visceral, endocrine, and metabotropic signals are integrated to a significant degree in hypothalamic centers (127, 128, 129). Additional interaxis control is accomplished by convergent and divergent actions of the corresponding hormones on the pituitary gland and peripheral target tissues (40, 130, 131, 132, 133). States of mono- and bihormonal deficiency underscore the inferred interplay between somatotropic peptides and gonadal sex steroids in directing adult body composition and sexual maturation (134, 135, 136, 137). For example, in the human, mouse, and rat, isolated GH deficiency reduces the production of major GH-dependent hepatic proteins, viz., IGF-I, IGFBP-3, acid-labile subunit, and IGFBP-5, and retards the initiation of sexual development (138, 139). In particular, deprivation of GH and IGF-I: 1) delays the timely onset of puberty (in all species studied); 2) slows the pace of pubertal maturation (all species); 3) attenuates phallic growth (human); 4) reduces adult testicular size (rodent); and 5) impairs sperm motility (mouse); but 6) does not abrogate fertility in the mature individual (all species) (131, 135, 140, 141, 142, 143, 144, 145).

    Clinical treatment of precocious puberty highlights the inference that stimulatory effects of gonadal sex steroids on the GH/IGF-I axis are reversed in part when ovarian or testicular secretion is decreased medically (40, 146, 147). In particular, therapy with a GnRH analog suppresses concentrations of estradiol and testosterone profoundly and those of GH, IGF-I, and IGFBP-3 significantly, but does not affect measurements of cortisol or adrenal androgens (148, 149). Gonadal-axis down-regulation may thereby obviate rapid skeletal maturation not only by sex-steroid depletion but also by secondary inhibition of the somatotropic axis (150). Albeit originally hypothesized as a means to stimulate growth in the face of bone-age delay, combining rh GH supplementation with GnRH agonist therapy in children with sexual precocity may enhance predicted final stature (147, 151, 152).

    From a simplified viewpoint, the timely onset and effectual progress of puberty would require, at a minimum, interaxis coordination of GH/IGF-I and GnRH/LH/sex-steroid production. Several mechanistic insights are relevant to this network-like concept. First, IGF-I and/or insulin act in an apparently species-specific manner to: 1) enhance hypothalamic GnRH outflow in vivo in the juvenile female monkey and rat and stimulate GnRH secretion in vitro by murine GT1–7 cells; 2) promote normal reproductive hormone secretion in the male and female mouse in part via the central nervous system insulin receptor substrate-2 signaling pathway; 3) potentiate GnRH-stimulated LH release in vitro; and 4) synergize with LH and FSH in stimulating ovarian and testicular steroidogenesis in vitro and in vivo (12, 132, 153, 154, 155, 156, 157, 158, 159, 160, 161, 162, 163). Second, endogenous gonadal sex steroids amplify the synthesis of GH and IGF-I and regulate the availability of IGFBPs and cognate proteases (90, 131, 132, 133, 164, 165, 166, 167, 168, 169, 170). Third, GH, IGF-I, IGF-II, insulin, and sex steroids interact via complex heterologous control of receptor-effector signaling pathways (135, 165, 171, 172, 173, 174, 175, 176, 177, 178, 179, 180, 181, 182). And, fourth, sex steroids and insulinomimetic peptides act in combination to govern appetite, thermoregulation, behavior, and energy expenditure via central and peripheral pathways (183, 184, 185, 186). Comprehensive formal integration of the foregoing multivalent mechanisms is not yet possible.

    B. Actions of androgen, estrogen, GH, and IGF-I on bone

    1. Hypogonadism overview.

    Prolonged deprivation of sex-steroid hormones at or after the time of expected puberty predisposes to reduced peak bone mass, attendant osteopenia, osteoporosis, and major fractures in the adult (56, 187, 188, 189, 190). Cross-sectional epidemiological analyses demonstrate that total and bioavailable (non-SHBG-bound) estradiol concentrations predict bone mass in women and men more accurately than total or bioavailable testosterone concentrations (188, 191, 192, 193, 194). Data from four longitudinal investigations corroborate the fundamental association between peripheral estrogen concentrations and bone mass in the aging individual (56, 188). Testosterone, GH, IGF-I, and (in some studies) leptin concentrations also correlate with TBW in some analyses (195, 196). Albeit incompletely defined, heterogeneous genetic factors are prominent determinants of bone mass in healthy individuals (197, 198). In addition, ethnicity may influence bone density by as much as 6–11% (199).

    2. Male hypogonadism.

    Testosterone replacement in hypogonadal boys and men increases TBW incrementally in proportion to the degree of androgen deficiency at presentation (168, 200, 201, 202). The anabolic effects of testosterone in vivo are not fully understood but are associated with augmentation of at least: 1) pulsatile GH secretion, which drives longitudinal bone growth (166, 169, 170, 203, 204); 2) IGF-I synthesis in both liver and bone cells (169, 170, 205, 206, 207); 3) gastrointestinal absorption and skeletal retention of calcium and magnesium (201, 208, 209, 210, 211, 212, 213, 214, 215); 4) muscle mass, mechanical loading, and energy expenditure, which in turn correlate with bone mineral content and density (90, 162, 164, 165, 168, 216, 217, 218, 219, 220); 5) biochemical markers of osteoblastic activity, such as osteocalcin (221); and 6) epiphyseal growth-plate maturation, which culminates in mineralization-dependent cessation of skeletal elongation (15, 222, 223, 224). In vitro studies affirm these inferences and further illustrate that (in rodent species) testosterone and 5-DHT can stimulate osteoblastic activity, inhibit apoptosis of osteoblasts and osteocytes, suppress osteoclastogenesis, and promote cortical (periosteal) bone apposition (225, 226, 227, 228). Androgen- and estrogen-dependent stimulation of epiphyseal mineralization underscores the clinical challenge of tailoring sex-hormone replacement in hypogonadal children to optimize total skeletal growth without inducing premature fusion of the growth plate (229, 230, 231, 232, 233, 234).

    Androgen receptors are expressed in human osteoblastic cells and mature osteocytes (235). A normal linear growth spurt is described in 46XY patients with complete androgen insensitivity (testicular-feminization syndrome) due to inactivating mutation of the cognate receptor (145, 236). Nonetheless, loss of androgen-receptor function limits adult height and skeletal volume (bone size) in the genotypic male to values intermediate between those of the unaffected male and female (237). A reduction in bone mineral content is reported in some (but not other) patients with testicular feminization syndrome. Low bone mineral content may reflect: 1) a younger age at prophylactic orchidectomy; 2) suboptimal estrogen replacement; 3) the postgonadectomy fall in IGF-I availability; 4) a role for the androgen receptor in early bone development; 5) reduced supplementation with aromatizable androgens, which provide substrate for estrogen synthesis in situ; and/or 6) more severe inactivation of androgen-receptor function (135, 223, 238, 239, 240).

    Supraphysiological amounts of aromatizable and nonaromatizable androgens stimulate osteoblast proliferation, antagonize the osteoclast-activating effect of PTH, and elevate markers of bone growth (228, 241, 242, 243, 244). In experimental animals, 5-DHT especially stimulates periosteal (appositional) skeletal growth and thereby increases cortical bone formation (228, 245, 246, 247). However, available data are not facile to interpret, because 5-reduced products of testosterone activate the androgen receptor and simultaneously impede estrogen action in some tissues (248, 249, 250). In the human, the androgen receptor may mediate up to 30% of sex steroid-induced skeletal remodeling, as inferred by combined administration of a down-regulating dose of a GnRH agonist, testosterone, and placebo, or an aromatase-enzyme inhibitor in healthy older men. In the sex steroid-depleted setting, transdermal repletion of testosterone or estradiol alone suppressed indices of bone resorption, increased markers of bone formation, and stimulated production of osteoprotegerin, a potent inhibitor of osteoclastogenesis (below). Each of the effects of testosterone was blunted by pharmacological aromatase blockade, with the exception of increased synthesis of osteocalcin, a marker of osteoblast function. Comparable mechanistic investigations of sex steroid-specific control of skeletal development are not available in childhood. Moreover, no studies have extended discrete receptor agonist and antagonist analyses over prolonged intervals (years) in the human.

    3. Estrogenic effects.

    Estrogenic steroids repress osteoclastogenesis, promote epiphyseal maturation, stimulate endosteal and trabecular bone formation, augment mineralization, and increase tensile bone strength (136, 137, 211, 240, 251, 252, 253, 254). Selective estrogen receptor (ER) modulators (e.g., raloxifene) appear to act analogously (but not necessarily identically) to enhance overall bone mineral content. Estrogen supplementation also stimulates the intestinal absorption and skeletal retention of calcium, which processes contribute to bone mineralization (255, 256, 257). Estrogens drive proliferation and differentiation of the entire osteoblastic-cell lineage; enhance the anabolic actions of other trophic signals (e.g., PTH, GH, IGF-I, and prostaglandin E2); limit osteocyte apoptosis; inhibit osteoclastic resorption under osteolytic stress (e.g., by PTH, prostaglandin F2, interferon , IL-1, and TNF-); and induce osteoblast synthesis of osteoprotegerin. The last-named glycoprotein is a potent inhibitor of osteoclastogenesis and inducer of osteoblast cytodifferentiation (77, 136, 245, 251, 258, 259, 260, 261, 262).

    4. ER subtype and aromatase-enzyme expression.

    Gene transcripts encoding truncated and full-length ER and ER? are detectable in osteoprogenitor cells, differentiated osteoblasts, and mature osteocytes (259, 263, 264). Expression of ER? predominates in immature bone and wanes with skeletal maturation (265, 266). As highlighted in Table 2, inactivating mutations of ER or the aromatase gene (but not ER?) cause severe osteoporosis and impair epiphyseal mineralization in the human and mouse (10, 237, 240, 243, 244, 246, 267, 268, 269, 270). In several patients with rare inborn aromatase deficiency, repletion of estradiol stimulated prompt epiphyseal maturation and bone mineralization, whereas testosterone supplementation did not (243, 271). Albeit less well studied, certain molecular polymorphisms of the estrogen-receptor gene also predict reduced BMD epidemiologically.

    Experiments based on short-term pharmacological inhibition of the aromatase enzyme are consistent with genetic inferences. In older men and aged male rats, administration of specific aromatase antagonists increased biochemical markers of bone resorption and (where assessed in the rodent) impaired the pubertal gain in skeletal calcium and (in the human and rodent) accelerated the age-related decline in mineral density (242, 246, 272, 273). One prospectively randomized study in boys with constitutionally delayed puberty combined placebo or a potent, orally active, selective aromatase-enzyme inhibitor (letrozole) with testosterone supplementation. The combination delayed radiographic bone maturation significantly compared with testosterone administration alone.

    Interpretation of target-tissue responses to ostensibly isolated interruption of a single sex-steroid signaling pathway in vivo is not straightforward, as indicated by the following considerations. First, in one analysis, supplementation with testosterone partially restored appendicular skeletal size in the orchidectomized mouse harboring transgenetic inactivation (knockout) of the ER subtype (-ERKO model) (240). In a strict technical context, this novel finding might be explained by androgen-receptor and/or ER?-mediated drive of longitudinal bone growth; confounding by supraphysiological androgen addback; and/or species, gene-dosage, or strain effects inherent in the transgenic model (101, 165, 174, 235, 243, 253, 264, 274). Second, pharmacological muting of sex-steroid negative feedback in the human and rodent stimulates (systemic) testosterone and estradiol secretion by 1.5- to 3-fold, thereby secondarily altering the systemic sex-hormone milieu (275, 276, 277, 278, 279). Third, androgen and estrogen exert both delayed genomic and rapid nongenomic effects on diverse target cells. Such bipartite actions mediate an array of complementary neuronal and extraneuronal effects. For example, in the central nervous system, estrogen acts on membrane receptors that facilitate IGF-I signaling via Akt and MAPK, thereby plausibly altering negative feedback by peripheral IGF-I (280, 281, 282). Fourth, androgens and estrogens regulate sex-steroid metabolism by inducing or inhibiting aromatase, 5- reductase and 17?-hydroxysteroid dehydrogenase isoenzymes, which interconvert androgens and estrogens. Fifth, age and gender appear to influence the skeletal effects of aromatase deficiency in transgenic murine models (246). Sixth, species modulates neuroendocrine adaptations to the sex-steroid milieu; e.g., estradiol but not 5-DHT in the human (and, conversely, in the rodent) drives GH secretion (127, 128). GH output is significant as a stimulus of both systemic and skeletal synthesis of IGF-I (166). Seventh, inactivation of ER in the mouse depletes systemic IGF-I concentrations (237). Transgenic depletion of blood-borne IGF-I indicates that this peripheral source of growth-factor drive also contributes to adult bone growth (283). Eighth, androgen depletion heightens the capacity of estrogen to stimulate osteoblastic synthesis of the potent osteoclastogenesis-inhibiting peptide, osteoprotegerin (251, 259, 284, 285, 286) (Fig. 10). And, lastly, the relative availabilities of estrogen and androgen can determine promoter-specific gene transcription due to incompletely characterized heterologous interactions among ER, truncated ER, ER?, and the androgen receptor (237, 240, 243, 287, 288, 289, 290, 291, 292). In view of extensive complementation of osteogenic and osteolytic signals, the biological effects of interrupting the action of a single agonist-receptor linkage, such as disabling ER, could reflect nonexclusively: 1) impairment of ER-dependent drive; 2) collateral actions via ER? and/or the androgen receptor; 3) reduced availability of systemic and in situ IGF-I; 4) altered sex-steroid synthesis and metabolism; and/or 5) heterologous receptor-receptor interactions.

    5. GH and IGF-I.

    GH, IGF-I, IGF-II, and IGFBPs control growth, remodeling, and mineralization of the skeleton in part via direct actions on bone (7, 241, 293, 294, 295, 296, 297, 298, 299, 300, 301, 302). A classic study showed that unilateral infusion of GH into the tibial artery of the GH-deficient male rat stimulates ipsilateral longitudinal bone growth. Mechanistically, GH drives a number of local bone effects; viz., skeletal IGF-I synthesis; proliferation of prechondrocytes; hypertrophy of osteoblasts; bone remodeling; and net mineralization (after a time lag of 1–2 yr in the human) (101, 303, 304, 305). In addition, exogenous GH suppresses osseous production of IGFBP-4 (which antagonizes the actions of IGF-I in bone) and stimulates in situ synthesis of IGFBP-2, -3, and -5 (which stimulate bone cells directly and/or via IGF-I) (306, 307, 308, 309).

    Sex steroids, IGF-II, T4, and glucocorticoids not only modulate the secretion of GH and IGF-I (127, 128), but also impact the direct effects of GH and IGF-I on skeletal growth (146, 147, 310, 311). For example, testosterone stimulates GH and IGF-I production systemically; induces IGF-I synthesis in the skeleton; enhances GH-driven IGF-I accumulation in osteoblasts; promotes epiphyseal cartilage growth; increases mineralization of bone matrix; and, augments net trophic effects of selected IGFBPs (306, 307, 308, 312, 313). Estradiol amplifies GH receptor-mediated signaling in osteocytes, up-regulates osteoblast IGF-I production, down-regulates inhibitory binding proteins (IGFBP-4 and -6), induces the type I IGF receptor in bone, and uniquely stimulates osteoblastic synthesis of osteoprotegerin, a potent antiresorptive signal that is not induced by nonaromatizable androgens (15, 136, 165, 174, 207, 237, 265, 308, 309, 313, 314, 315, 316, 317, 318). Apparently joint trophic roles of GH and estradiol in bone accrual are inferable indirectly in clinical studies. In particular, maximal BMD correlates with 24-h GH concentrations in young men and with overnight GH and estradiol concentrations in the African-American (but not Caucasian) male (36, 37, 221).

    Height, weight, and genetic endowment are strong epidemiological determinants of bone mineral content (191, 199). However, height and weight mirror multiple convergent genetic, environmental, and trophic-hormone interactions (319, 320, 321). The rate of skeletal calcium and magnesium accretion is maximal at ages 11–14 yr in girls and 16–18 yr in boys (322, 323, 324). On the other hand, total IGF-I, but not sex-steroid, concentrations reach a zenith 1.5–2 yr later. In young women, 99% of maximal BMD and 99% of total mineral content are attained at ages 22 ± 2.5 yr and 26 ± 3.7 yr, respectively (325). Skeletal mass at age 20, which amalgamates the conjoint impact of height, weight, environment, and genetics, predicts more than 50% of the statistical variability in bone mineral content in later adulthood (326). Nonetheless, some bone growth and mineralization continue in selected skeletal sites into the fourth or fifth decade of life (56, 188, 221, 323, 325, 327).

    In hyposomatotropic children and adults, GH replacement therapy facilitates the timely onset of sexual development and therewith increased sex-steroid secretion (140). GH treatment in such patients uniformly elevates biochemical indices of bone remodeling (within weeks), promotes marked (so-called catch-up) linear growth in the first year, augments skeletal mineralization after 1.5 to 2 yr, and (in children) increases final adult stature (97, 241, 298, 328, 329, 330, 331, 332, 333, 334) (Table 3). Albeit less well documented, administration of IGF-I also stimulates bone growth, skeletal remodeling, and mineral deposition in the IGF-I-deficient setting in man and animals. Estrogen blunts the actions of GH on biochemical markers of skeletal remodeling in the hypopituitary female, postmenopausal woman, and male-to-female transsexual patient (335, 336, 337). Estradiol replacement also attenuates the rh GH-induced rise in IGF-I concentrations and decline in visceral FM (127, 337). Whether the foregoing GH/sex-steroid interactions apply equally to other long-term tissue effects is not known.

    Genetic GH receptor defects and primary IGF-I deficiency states are associated with osteopenia, sarcopenia, and visceral adiposity in the adult (338). In the GH receptor-defective patient, replacement therapy with rh IGF-I facilitates the onset of pubertal development and stimulates musculoskeletal growth (339). Nonetheless, systemic delivery of IGF-I does not normalize growth velocity or body composition in children with inborn GH receptor defects. The precise factors that account for incomplete tissue responses in this setting are not yet evident (141, 340, 341, 342, 343, 344, 345, 346, 347, 348). One consideration is that GH and IGF-I exert both singular and combined trophic effects in a target tissue-specific fashion (349, 350, 351, 352, 353, 354, 355, 356, 357). For example, GH but not IGF-I induces synthesis of the complete 150-kDa ternary complex comprising IGF-I, IGFBP-3, and the acid-labile subunit (358). Systemic concentrations of the ternary complex correlate well with somatic growth in normal puberty and during GH treatment in hyposomatotropic children (338). Conversely, hepatic-specific IGF-I-deficient transgenic mice exhibit diminished BMD in adulthood despite elevated GH concentrations (283). The latter important observation suggests that postnatally induced IGF-I deficiency impairs skeletal growth in the rodent. In support of this experimental inference, peripheral IGF-I administration can stimulate markers of skeletal remodeling and increase BMD.

    6. Multisignal endocrine control.

    Sex steroids, GH, IGF-I, cortisol, T4, and other systemic hormones act on bone collaboratively via potent local effector molecules, such as IGF-I/IGFBPs, cytokines, prostaglandins, and osteoprotegerin. This nonexclusive ensemble of in situ regulators directs skeletal growth (increased volume), remodeling, and mineralization (259). The importance of multihormonal trophic control of bone growth and maturation is illustrated in children with Turner syndrome. Osteopenia in this setting is attributable to 3-fold deficiency of estrogen, GH/IGF-I, and androgen along with important but incompletely characterized genetic factors that disrupt bone development. TBW in gonadal dysgenesis is reduced detectably in the third decade, and fracture risk is increased significantly by the fourth decade of life (40, 77, 136, 137, 201, 243, 359, 360, 361, 362, 363). Clinical interventional trials have combined physiological estrogen replacement (based on developmental age), dose-titrated repletion of androgen, and supraphysiological amounts of GH to accelerate height velocity. Final statural gain in Turner syndrome is influenced principally by age at initial treatment, duration of hormonal intervention, doses of GH (higher) and androgen (low), degree of growth failure, and incompletely defined genetic factors (98, 175, 176, 177, 209, 210, 232, 233, 364, 365).

    C. Adiposity and sex-steroid hormones

    Sex-steroid hormones, GH, insulin, glucocorticoids, and ?-3 adrenergic agonists are dominant determinants of adipocyte mass (Fig. 11). A corollary thesis is that fat topography is controlled by regionalized expression and activity of sex steroid-metabolizing enzymes, growth factors, and cognate receptors (366, 367, 368). For example, estradiol receptors predominate in mammary and gluteofemoral fat, whereas androgen receptors are more abundant in intraabdominal (omental) fat (367). At the level of target cells, GH, ?-3-adrenergic agonists, and testosterone induce greater lipolysis of visceral than sc adipose tissue. In contradistinction, insulin and cortisol stimulate lipogenesis in diverse fat depots (76, 100, 365, 366, 367, 369, 370, 371, 372).

    The liporegressive actions of testosterone reflect the 4-fold ability of androgens to: 1) amplify the direct lipolytic effects of GH and adrenergic agonists; 2) up-regulate androgen receptors homologously (positive autofeedback effect) and ?-3 adrenergic receptors heterologously (sensitization effect); 3) oppose the lipogenetic effects of insulin and cortisol; and 4) inhibit lipoprotein lipase-dependent esterification of triglycerides, which biochemical step is required to enlarge fat cells (253, 351, 365, 373, 374, 375, 376, 377). Collectively, such mechanisms could contribute to pubertal redistribution of fat stores. However, the details of such putative actions have not been mapped.

    Estradiol up-regulates its own receptor and that of insulin in fat cells in vitro and in vivo (378, 379). These effects would be consistent with the statistical association between (unopposed) estrogen replacement therapy and intraabdominal adiposity in postmenopausal women (366, 380). However, correlations may be invidious, inasmuch as the route of estrogen supplementation also determines the magnitude of metabotropic effects; e.g., oral compared with transdermal estradiol replenishment inhibits lipid oxidation more (thereby predisposing to fat retention) and blocks glucose disposal less (thus facilitating insulin action and fat synthesis) (98, 381). Conversely, a history of combined use of estrogen and a synthetic progestin postmenopausally predicts less visceral fat accumulation (98, 175, 177, 180, 382, 383). The apparent lipolytic effect of adding a synthetic progestin may be due to weak intrinsic androgenicity of such agents. According to this reasoning, greater availability of androgens in menstruating than ovariprival women may contribute to relatively less intraabdominal (visceral) fat (365, 383, 384). In addition, nonwithdrawal of adrenal androgenic sex steroids during long-term GnRH agonist therapy of precocious puberty may account for some changes in total body fat despite estrogen depletion (73, 385).

    Estradiol inhibits proliferation of immature fat cells via ER and stimulates growth of preadipocytes via ER? (366). The foregoing distinction applies in the rodent, inasmuch as transgenetic -ERKO and aromatase knockout induce hyperplasia and hypertrophy of (white) adipocytes with a resultant 80–100% increase in total body fat (368, 386). Conversely, high-dose estradiol administration in the immature mouse, rat, and cow reduces FM. The topography of adipose tissue presumably depends further on (nonexclusively) tissue-specific differences in the expression of - (inhibitory) and ?- (stimulatory) ER, aromatase enzyme, 11-hydroxysteroid dehydrogenase (types I and II), and 17?-hydroxysteroid dehydrogenase (isotypes 2 and 3) (384). The foregoing enzymes control interconversion of testosterone and estradiol, as well as cortisol and (inactive) cortisone. Understanding how the ensemble of IGF-I, GH, insulin, cortisol, sex steroids, adipocyte topography, gender, and species determines regional and total FM in pubertal development presents a daunting investigative challenge.

    D. Adiposity and GH/IGF-I

    GH increases lipolysis acutely (within minutes), and decreases adiposity over the short-term (days to months) in children and adults (387, 388) (Table 3). The whole-body lipolytic effect of a given dose of GH is attenuated in estrogen-replete young or postmenopausal women compared with estrogen-insufficient women and androgen-sufficient men (175, 180, 345). Such distinctions suggest that estrogen induces partial resistance of adipose tissue to GH. In one mechanistic analysis of this relationship, estradiol down-regulated GH signaling in vitro by inducing cellular expression of the negative-feedback signal, suppressor of cytokine signaling (389).

    GH induces rapid loss of fat due to stimulation of lipolysis and reciprocal antagonism of the lipogenetic actions of insulin (97, 119, 175, 299, 339, 353, 390, 391, 392, 393, 394, 395). Although the initial reduction of adipose-tissue mass persists during continued GH replacement in hyposomatotropic patients, the rate of decrease in FM slows over time. The basis for evident down-regulation of fat-cell responsiveness to the lipolytic action of GH is not well defined.

    GH replacement therapy in the adult initially elevates (days to weeks) and then suppresses (months to years) insulin and leptin concentrations. The delayed decline in insulin and leptin concentrations parallels a gradual reduction in visceral fat and total adiposity induced by exogenous GH (98, 144, 180, 299, 351, 391, 396, 397, 398, 399, 400). The key lipolytic role of GH is indicated in a murine model of transgenically enforced postnatal depletion of liver-derived IGF-I concentrations. In this experimental context, hypersecretion of GH reduces total-body fat in the face of elevated leptin and insulin concentrations. Hepatic IGF-I gene silencing also impairs muscle sensitivity to insulin and stimulates hepatic gluconeogenesis. Such outcomes would secondarily modulate in vivo glucose and fat metabolism (401, 402).

    Mature adipocytes express GH, but not IGF-I, receptors (338). Indeed, in clinical studies, combining rh IGF-I and GH replacement fails to reduce FM further (355). In addition, long-term rh IGF-I treatment of children with GH receptor defects decreases intraabdominal FM only over the first 6 months of therapy (339). A plausible notion is that this short-lived liporegressive effect reflects IGF-I action on less mature fat cells and/or the known capability of exogenous IGF-I to suppress endogenous insulin secretion (403).

    Adiposity suppresses GH production markedly in the human and experimental animal (59, 72, 404, 405, 406, 407, 408). In men and women, intraabdominal FM is a primary negative determinant of pulsatile GH secretion (71, 409, 410). On the other hand, for reasons that are not known, sc rather than visceral adiposity is a major negative correlate of GH production in children (93). However, as observed in the adult, intraabdominal adiposity in the child presages insulin resistance and dyslipidemia, and presumptively elevates long-term atherosclerotic risk (47, 76, 411). In fact, hyposomatotropism, topography of fat distribution, ethnicity, gender, sex hormones, IGF-I, IGFBP-1, and insulin concentrations jointly influence predicted risk of cardiovascular disease (58, 366).

    Low GH concentrations in obesity result from reduced secretion and increased elimination rates (412, 413). In particular, adiposity in some manner represses GH secretory-burst mass, impedes the stimulatory effects of secretagogues, expands the GH distribution volume, and accelerates the metabolic clearance of GH (71, 405, 407, 409, 412, 413, 414, 415, 416, 417). Reduced absolute GH secretion constitutes the major (>85%) basis for low GH concentrations in obesity (127). More rapid elimination of GH correlates with intraabdominal (upper body) rather than sc (lower body) adiposity in young women (71). The mechanistic basis for this association has not been delineated.

    In the human and experimental animal, several factors appear to repress GH production in obesity: 1) direct inhibition of somatotrope secretion by elevated systemic concentrations of free fatty acids (FFAs), free IGF-I, insulin, and, less plausibly in the human, leptin (83, 418, 419, 420, 421, 422, 423, 424, 425, 426, 427); 2) proximate suppression of GH release by excessive hypothalamic somatostatin outflow, which in the obese Zucker rat differs by gender; 3) impaired release and blunted actions of GHRH; and 4) an obesity-associated reduction in systemic ghrelin concentrations (17, 409, 414, 428, 429, 430, 431, 432, 433). These considerations are supported in part by the increase in GH secretion in obese subjects following: 1) short-term fasting or weight reduction, which suppresses IGF-I and insulin and elevates ghrelin concentrations; 2) pharmacological inhibition of somatostatin outflow, which drives GH secretion; and 3) infusion of GHRH and/or GH-releasing peptide (GHRP), which stimulates GH release (420, 429, 432, 434, 435, 436, 437). Notably, none of the foregoing individual interventions is able to reinstate GH output equivalently in the obese and lean individual. However, the combination of L-arginine (to repress somatostatin) or acipimox (to block FFA release) and GHRH or GHRP will induce significant GH secretion in obese subjects, which outcome is consistent with a presumptively multifactorial basis for relative hyposomatotropism (127, 128). In fact, all three of the insulin, FFA, and free IGF-I concentrations are elevated in the obese adult, thus conferring potentially combined repression of GH secretion (430).

    Acute nutrient deprivation in obese volunteers fails to stimulate maximal GH release (127). Attempting to interpret this outcome illustrates the complexity of metabolic control of the GH-IGF-I axis. For example, fasting lowers insulin and free IGF-I concentrations, thereby potentially disinhibiting feedback on GH output. At the same time, nutrient deprivation increases FFA concentrations, which presumptively suppress somatotrope secretion (76, 423, 438, 439, 440, 441, 442, 443).

    Linear growth appears to be accelerated in obese children. The precise basis for this observation is unknown. However, unbound IGF-I concentrations are elevated in the obese human, which is presumably due to hyperinsulinemia-dependent suppression of hepatic IGFBP-1 production (100, 439). Elevated total and free IGF-I concentrations correlate with exogenous IGF-I-induced inhibition of GH secretion in individuals of normal body weight (282, 419, 444, 445). Conversely, partial (34%) reduction of total IGF-I concentrations by hepatic GH receptor blockade with pegvisomant amplifies GH secretory-burst mass significantly (by 1.8-fold) in healthy young adults (280, 281). Likewise, liver-specific postnatal IGF-I gene inactivation in mice and a single case of mutational truncation of the IGF-I gene in the human increased GH concentrations by 2- to 10-fold and more than 30-fold, respectively (358, 401). These ensemble data would support the postulate that excessive IGF-I availability contributes to reduced GH secretion in obesity.

    The role of hyperinsulinemia in promoting skeletal growth or in repressing GH secretion in obesity is not clear. In the latter context, free IGF-I and insulin concentrations both correlate inversely with GH output (430). For example, in one study, an acute euglycemic hyperinsulinemic clamp that achieved insulin concentrations observed in fasting obese adults did not inhibit GH secretion in healthy young men (419). This important outcome does not exclude the corollary notions that: 1) insulin may potentiate IGF-I feedback inhibition; and 2) more prolonged hyperinsulinemia may suppress GH secretion. Such issues are relevant in view of indirect clinical data that are consistent with negative feedback by both insulin and IGF-I (426, 427, 440, 444, 446, 447).

    Marked weight loss is required to normalize suppressed GH secretion in obese individuals (448). For example, several weeks of caloric restriction, which were sufficient to reduce visceral FM by more than 30% in obese premenopausal women, failed to reinstate normal 24-h GH production (71). This outcome raises the question whether intraabdominal obesity and hyposomatotropism arise individually, exacerbate each other, or reflect a common defect in metabotrobic or appetite-regulating signals (17, 71, 366, 368, 418, 449, 450, 451). Knowledge of this issue could aid in formulating how long GH replacement should be continued in adult GH-deficient patients with visceral adiposity and increased cardiovascular risk defined on a priori epidemiological grounds. A corollary interventional query in children with idiopathic visceral obesity is the possible utility of short-term supplementation with rh GH. The question is made difficult by possible toxicity, significant cost, uncertain compliance, unknown perpetuity of therapy, and the availability of alternative treatments that reduce intraabdominal FM, enhance peripheral insulin sensitivity, and engender more favorable lipid profiles (32, 65, 98, 144, 348, 351, 391, 396, 399, 452, 453, 454, 455, 456, 457, 458, 459, 460).

    In the human, monkey, sheep, dog, and guinea pig, fasting stimulates GH secretion. These responses contrast with those in the rodent, in which caloric deprivation represses GH production (127, 357, 391, 442, 461, 462, 463, 464, 465, 466, 467, 468, 469, 470). The basis for the latter clarion species difference remains to be clarified.

    E. Control of muscle by sex steroids and GH/IGF-I

    Few prospective interventional studies have examined the impact of sex steroids and GH/IGF-I on muscle mass and function in infancy, childhood, or puberty. In one analysis, short-term testosterone supplementation in prepubertal boys increased whole-body proteolysis by 18%, reduced amino-acid oxidation (catabolism) by 49%, and stimulated net protein synthesis by 35% (211). In laboratory experiments, androgens promote hypertrophy of type IIA (rapid-twitch, glycogenolytic, highly oxidative) muscle fibers and protect against immobilization-induced muscle atrophy (210). Physiological amounts of testosterone stimulate lean-tissue accrual, augment total muscle volume, accelerate protein synthesis, retard protein breakdown, increase isokinetic strength and induce in situ muscle IGF-I gene expression in hypogonadal boys or men (23, 167, 201, 209, 210, 211, 370, 471, 472, 473, 474, 475, 476, 477, 478). However, pharmacological doses of androgen are required to enhance isometric strength or maximal aerobic capacity in eugonadal young adults. This distinction could indicate that euandrogenemia operates near or above the genetically determined upper bound of the physiological testosterone concentration-muscle response function (334, 370, 473). According to the foregoing collective data, increased androgen availability in puberty would provide a proximate (but nonexclusive) stimulus to muscle growth (25, 479) (Table 3). On the other hand, estrogen repletion does not measurably affect whole-body protein synthesis or oxidation in combined estrogen- and androgen-deficient (ovariprival) girls with Turner syndrome (480, 481). These data suggest that endogenous androgens may be required to drive pubertal anabolism in girls.

    Androgens and muscle loading stimulate myofibrillar protein synthesis, myoblast proliferation, and myocyte hypertrophy (211, 392, 471, 482). Anabolic and growth-promoting effects of testosterone and muscle contraction occur in significant part via the induction of in situ IGF-I and the inhibition of IGFBP-4 gene expression (472). One study in older men reported that repletion of testosterone also transiently induced the androgen-receptor gene in muscle (472). The significance of this finding has not been demonstrated. Intramuscular IGF-I accumulation driven by testosterone and other factors promotes myoblast proliferation from satellite cells and stimulates myocyte hypertrophy, as established by direct local infusion of IGF-I peptide into senescent skeletal muscle and transfer of the recombinant IGF-I gene into developing smooth muscle, respectively (212, 213, 214, 483, 484, 485, 486).

    Testosterone induces expression of the myostatin gene in skeletal muscle. Myostatin is a 26-kDa glycoprotein that opposes myocyte apoptosis (215, 486, 487). Mutations of the myostatin gene result in marked muscle hypertrophy in the transgenic mouse and in the Belgian double-muscled Piedmontese cow. Muscle unloading and catabolic syndromes like AIDS-associated wasting also stimulate skeletal-muscle myostatin gene expression (488). The foregoing adaptations were postulated to reflect compensatory autocrine or paracrine mechanisms in muscle. However, the nature of such mechanisms is elusive. In a small number of studies, administration of either GH or IGF-I in elderly humans did not consistently up-regulate this antiapoptotic signal (489, 490). Therefore, additional studies will be required to clarify precisely how testosterone and GH promote myostatin-dependent and -independent muscle growth (392).

    Anabolism occurs when the rate of amino-acid incorporation into proteins exceeds that of oxidative metabolism (392). Net protein accumulation is determined positively by amino-acid availability, muscle loading, and the myotrophic hormones, GH, IGF-I, and testosterone. Protein loss is accentuated variously by: 1) amino-acid depletion (491, 492, 493); 2) aging (472, 493, 494, 495); and 3) systemic inflammatory disease (496). Systemically delivered hormones stimulate protein synthesis (androgen, GH, and high plasma concentrations of IGF-I) and/or retard protein breakdown (testosterone and lower blood-borne concentrations of IGF-I and insulin) (124, 201, 253, 334, 349, 453, 472, 476, 496, 497, 498, 499, 500, 501, 502). In one analysis, exercise enhanced the biosynthesis of myofibrillar proteins in part by enhancing translation of existing mRNA.

    Testosterone supplementation promotes whole-body nitrogen retention in eugonadal and hypogonadal men and in individuals with heightened catabolism; e.g., fasting, AIDS-associated muscle wasting, and severe burns (166). Administration of GH or IGF-I increases lean body mass in organically hyposomatotropic patients (356, 393) and limits protein catabolism in patients with multiorgan failure, major surgery, protracted critical illness, male hypogonadism, and glucocorticoid excess (357, 392, 499, 500, 503, 504, 505, 506, 507, 508, 509). In a recent study in postmenopausal women, supplementation with rh GH for 6 months stimulated whole-body protein synthesis and breakdown by 9 and 8%, respectively; and administration of a high dose of rh IGF-I increased the same measures by 18 and 17%, respectively. Although net protein synthesis rose by 48% (GH) and 196% (IGF-I), exact comparison of GH and IGF-I dosimetry in the human is not yet possible. The importance of continuing trophic peptide drive is evident in young-adult hypopituitary patients, in whom discontinuation of GH replacement therapy results in measurable attrition of muscle mass (510).

    An implicit clinical thesis is that testosterone, GH, and IGF-I promote anabolism synergistically (323, 511, 512, 513, 514, 515). This concept has not been explored definitively in human pubertal physiology (Table 4). In particular, the precise cellular and molecular mechanisms that transduce putative hormonal synergy are unexplained.

    IV. Energy Expenditure in Puberty

    Energy expenditure is quantitated by way of whole-room calorimetry, portable closed- or open-circuit calorimetry, and the metabolism of doubly labeled water (387, 408, 516, 517, 518). The benchmark method has been quantitation in a closed respiratory chamber (519). Calorimetry relies on the respiratory quotient (ratio of oxygen and carbon dioxide content in expired air) (72). Portable systems are used commonly, but may be less reliable in children or apprehensive adults (516). The innovative doubly labeled-water technique can be applied in community-dwelling individuals to monitor total energy utilization during normal daily physical activity and rest (293, 519).

    Energy expenditure over 24 h reflects principally basal metabolic costs (517). Energy is expended at rest to maintain core body temperature via heat generation in internal organs (liver, kidney, muscle, fat, and brain). Heat production proceeds through mitochondrial uncoupling protein (UCP), such as muscle UCP-3, and in the maintenance of transmembrane ionic gradients and other ATP-dependent metabolic reactions. Nonbasal contributions to energy balance arise from the thermogenic effects of metabolizing glucose and mixed nutrients (specific dynamic action of food) and physical activity (520, 521, 522). In correlational studies, FFM and aerobic exercise capacity are the principal positive determinants of resting energy expenditure (518, 523, 524). Fasting leptin concentrations also predict basal energy expenditure to some degree (525). Although a causal relationship is not established in the human, leptin promotes central sympathetic outflow experimentally, which in principle would elevate basal energy expenditure (526). In young adults, acute aerobic exercise increases energy expenditure without altering leptin concentrations (527).

    Healthy aging, food restriction, and limited physical activity lower total energy expenditure (520, 528, 529, 530, 531). Resistance exercise, aerobic physical training, and sympathoadrenal outflow stimulate energy utilization in an age-, nutrient-, gender-, and ethnicity-related fashion (521, 522, 523, 532, 533). In the human, endurance training drives heat production, enhances insulin action, increases noradrenergic outflow, augments maximal oxygen consumption, elevates 24-h GH secretion, facilitates muscle glucose uptake, reduces visceral FM, and lowers leptin concentrations (520, 532, 534, 535).

    Longitudinal analyses will be important to clarify the precise impact of sexual maturation and gender on energy expenditure, inasmuch as current data are limited and contradictory. For example, in a study of adolescent girls, the resting metabolic rate (adjusted for FFM) averaged 1418 ± 186 kcal/d before puberty, tended to decline in early puberty, and then decreased significantly to 1179 ± 189 kcal/d 4 yr after menarche (536). In another comparison of 12- to 14-yr-old children, resting energy expenditure was significantly higher during adolescence than before puberty (537).

    Resting energy utilization under free-living conditions is lower in women than men (293). The gender distinction presumably reflects the capacity of testosterone, nortestosterone, and to a lesser degree androstenedione or DHEA to significantly augment basal energy expenditure (524, 538, 539). In addition, energy expenditure rises consistently (by 4–16%) in the luteal phase of the normal menstrual cycle (540, 541, 542, 543, 544, 545) and falls in amenorrheic states (546). Whether increased progesterone or androgen availability contributes to the former association is not evident. In women, estradiol does not affect the basal metabolic rate or whole-body anabolism (408, 547). However, transgenic disruption of ER in the male mouse significantly (11%) reduced basal energy expenditure via unknown mechanisms (368).

    Energy expenditure is normal in GH-deficient patients, when data are corrected for age, gender, and FFM (398, 548). Nonetheless, GH replacement therapy in hyposomatotropic patients and obese individuals elevates the basal metabolic rate significantly by within-subject comparison (393, 398, 549, 550). GH administration in hyposomatotropic adults is associated with: 1) increased expression of mitochrondrial UCPs (551); 2) early stimulation and delayed suppression of leptin concentrations (387, 525, 552); 3) accrual of lean-body tissue and recession of intraabdominal FM (387, 398, 550, 553); and 4) enhanced conversion of T4 to T3 by 3'-mono-deiodination in peripheral tissues (549, 553, 554). At present, the impact of rh IGF-I on energy expenditure is not well studied. In one investigation, acute infusion of this growth factor did not stimulate energy expenditure in the parenterally fed rat.

    V. Summary

    Body composition adapts across the in utero, neonatal, pubertal, and adult lifetime in an ethnicity- and gender-related fashion. The present review highlights these developmental adaptations and illustrates how signals from the gonadotropic and somatotropic axes singly and jointly govern accrual and depletion of muscle, fat, and bone mass. These emerging concepts should enlarge the platform of critical clinical and basic-science investigations of this developmental theme.

    Acknowledgments

    The authors thank Jean Plote and Kris Nunez for skillful preparation of the manuscript.

    Footnotes

    This work was supported in part by National Center for Research Resources Grant MO1 RR00585 to the General Clinical Research Center at the Mayo Clinic, and National Institutes of Health/National Institute on Aging Grants AG-014779 and AG-019695 (to J.D.V.).

    First Published Online December 7, 2004

    Abbreviations: BMD, Bone mineral density; BMI, body mass index; CT, computed tomography; DEXA, dual-energy x-ray absorptiometry; ER, estrogen receptor; FFA, free fatty acid; FM, fat mass; FFM, fat-free mass; GHRP, GH-releasing peptide; IGFBP, IGF binding protein; MRI, magnetic resonance imaging; rh, recombinant human; TBW, total body water; TBW/FFM, percentage of TBW normalized to FFM; UCP, uncoupling protein.

    References

    Savage MO, Burren CP, Blair JC, Woods KA, Metherell L, Clark AJ, Camacho-Hubner C 2001 Growth hormone insensitivity: pathophysiology, diagnosis, clinical variation and future perspectives. [Review]. Horm Res 55:32–35

    Gasser T, Kneip A, Ziegler P, Largo R, Prader A 1990 A method for determining the dynamics and intensity of average growth. Ann Hum Biol 17:459–474

    Kuczmarski RJ, Ogden CL, Grummer-Strawn LM, Flegal KM, Guo SS, Wei R, Mei Z, Curtin LR, Roche AF, Johnson CL 2000 CDC growth charts: United States. Adv Data 1–27

    Pedersen JF 1982 Fetal crown-rump length measurement by ultrasound in normal pregnancy. Br J Obstet Gynaecol 89:926–930

    Tanner JM 2001 The Growth Process. In: Kostyo J, ed. Handbook of physiology: the endocrine system. New York: Oxford University Press

    Hojbjerg GC, Weis NR 1997 Reference values for body proportions and body composition in adult women with Ullrich-Turner syndrome. Am J Med Genet 72:403–408

    Backeljauw PF, Underwood LE; GHIS Collaborative Group 2001 Therapy for 6.5–7.5 years with recombinant insulin-like growth factor I in children with growth hormone insensitivity syndrome: a clinical research center study. J Clin Endocrinol Metab 86:1504–1510

    Flier JS, Maratos-Flier E 1998 Obesity and the hypothalamus: novel peptides for new pathways. Cell 92:437–440

    Zhou Y, Xu BC, Maheshwari HG, He L, Reed M, Lozykowski M, Okada S, Cataldo L, Coschigamok K, Wagner TE, Baumann G, Kopchick JJ 1997 A mammalian model for Laron syndrome produced by targeted disruption of the mouse growth hormone receptor/binding protein gene (the Laron mouse). Proc Natl Acad Sci USA 94:13215–13220

    Vidal O, Lindberg M, Savendahl L, Lubahn DB, Ritzen EM, Gustafsson JA, Ohlsson C 1999 Disproportional body growth in female estrogen receptor--inactivated mice. Biochem Biophys Res Commun 265:569–571

    Nishimori K, Matzuk MM 1996 Transgenic mice in the analysis of reproductive development and function. Rev Reprod 1:203–212

    Bruning JC, Gautam D, Burks DJ, Gillette J, Schubert M, Orban PC, Klein R, Krone W, Muller-Wieland D, Kahn CR 2000 Role of brain insulin receptor in control of body weight and reproduction. Science 289:2122–2125

    Thalange NK, Price DA, Gill MS, Whatmore AJ, Addison GM, Clayton PE 1996 Insulin-like growth factor binding protein-3 generation: an index of growth hormone insensitivity. Pediatr Res 39:849–855

    Yanovski JA, Yanovski SZ, Filmer KM, Hubbard VS, Avila N, Lewis B, Reynolds JC, Flood M 1996 Differences in body composition of black and white girls. Am J Clin Nutr 64:833–839

    Smith EP, Boyd J, Frank GR, Takahashi H 1994 Estrogen resistance caused by a mutation in the estrogen-receptor gene in a man. N Engl J Med 331:1056–1061

    Lopez-Bermejo A, Buckway CK, Rosenfeld RG 2000 Genetic defects of the growth hormone-insulin-like growth factor axis. Trends Endocrinol Metab 11:39–49 [Review]

    Yiannakouris N, Yannakoulia M, Melistas L, Chan JL, Klimis-Zacas D, Mantzoros CS 2001 The Q223R polymorphism of the leptin receptor gene is significantly associated with obesity and predicts a small percentage of body weight and body composition variability. J Clin Endocrinol Metab 86:4434–4439

    Ellis JA, Stebbing M, Harrap SB 2001 Significant population variation in adult male height associated with the Y chromosome and the aromatase gene. J Clin Endocrinol Metab 86:4147–4150

    Salerno M, Balestrieri B, Matrecano E, Officioso A, Rosenfeld RG, Di Maio S, Fimiani G, Ursini MV, Pignata C 2001 Abnormal GH receptor signaling in children with idiopathic short stature. J Clin Endocrinol Metab 86:3882–3888

    Vilain E, Guo W, Zhang YH, McCabe ER 1997 DAX1 gene expression upregulated by steroidogenic factor 1 in an adrenocortical carcinoma cell line. Biochem Mol Med 61:1–8

    Tanner JM, Davies PS 1985 Clinical longitudinal standards for height and height velocity for North American children. J Pediatr 107:317–329

    Fomom SJ, Haschke F, Ziegler EE, Nelson SE 1982 Body composition of reference children from birth to age 10 years. Am J Clin Nutr 35(5 Suppl):1169–1175

    Gregory JW, Greene SA, Thompson J, Scrimgeour CM, Rennie MJ 1992 Effects of oral testosterone undecanoate on growth, body composition, strength and energy expenditure of adolescent boys. Clin Endocrinol (Oxf) 37:207–213

    Goings SB 1996 Densitometry. In: Roche AF, Heysfiled SB, Lohman TG, eds. Human body composition. Champaign, IL: Human Kinetics Publishers; 205–215

    Haschke F 1983 Body composition of adolescent males. Part I. Total body water in normal adolescent males. Part II. Body composition of the male reference adolescent. Acta Paediatr Scand Suppl 307:1–23

    Clasey JL, Kanaley JA, Wideman.L., Heymsfield SB, Teates CD, Gutgesell ME, Thorner MO, Hartman ML, Weltman A 1999 Validity of methods of body composition assessment in younger and older males and females. J Appl Physiol 86:1728–1739

    2001 Total body composition: Birth to old age. In: Roche AF, Heysfiled SB, Lohman TG, eds. Human body composition. Champaign, IL: Human Kinetics Publishers; 230–245

    Bouchard C, Despres JP 1989 Variation in fat distribution with age and health implications. In: Spirduso WW, Eckert HM, ed. Physical activity and aging. Champaign, IL: Human Kinetics Publishers; 78–106

    Lohman TG 1992 Advances in body composition assessment. Current issues in exercise science, Monograph no. 3. Champaign, IL: Human Kinetics Publishers

    Roemmich JN, Clark PA, Weltman A, Rogol AD 1997 Alterations in growth and body composition during puberty: I. Comparison among 2-, 3-, and 4-compartment models of body composition. J Appl Physiol 83:927–935

    Ellis KJ, Shypailo RJ, Abrams SA, Wong WW 2000 The reference child and adolescent models of body composition. A contemporary comparison. Ann NY Acad Sci 904:374–382

    Codner E, Cassorla F, Tiulpakov AN, Mericq MV, Avila A, Pescovitz OH, Svensson J, Cerchio K, Krupa D, Gertz BJ, Murphy G 2001 Effects of oral administration of ibutamoren mesylate, a nonpeptide growth hormone secretagogue, on the growth hormone-insulin-like growth factor I axis in growth hormone-deficient children. Clin Pharmacol Ther 70:91–98

    Catalano PM, Drago NM, Amini SB 1995 Maternal carbohydrate metabolism and its relationship to fetal growth and body composition. Am J Obstet Gynecol 172:1464–1470

    Catalano PM, Thomas AJ, Avallone DA, Amini SB 1995 Anthropometric estimation of neonatal body composition. Am J Obstet Gynecol 173:1176–1181

    Malina RM, Bouchard C 1991 Models and methods for studying body composition. Growth, maturation, and physical activity. Champaign, IL; Human Kinetics Publishers; 87–100

    Wright NM, Renault J, Willi S, Veldhuis JD, Gordon L, Key LL, Bell NH 1995 Greater secretion of growth hormone in black than in white males: possible factor in greater bone mineral density. J Clin Endocrinol Metab 80:2291–2297

    Wright NM, Papadea N, Willi S, Veldhuis JD, Pandey J, Key LL, Bell NH 1996 Demonstration of a lack of racial differences in secretion of growth hormone despite a racial difference in bone mineral density in premenopausal women –a clinical research study. J Clin Endocrinol Metab 81:1023–1026

    Wong WW, Copeland KC, Hergenroeder AC, Hill RB, Stuff JE, Ellis KJ 1999 Serum concentrations of insulin, insulin-like growth factor-I and insulin-like growth factor binding proteins are different between white and African American girls. J Pediatr 135:296–300

    Cutfield WS, Hofman PL, Vickers M, Breier B, Blum WF, Robinson EM 2002 IGFs and binding proteins in short children with intrauterine growth retardation. J Clin Endocrinol Metab 87:235–239

    Attie KM, Ramirez NR, Conte FA, Kaplan SL, Grumbach MM 1990 The pubertal growth spurt in eight patients with true precocious puberty and growth hormone deficiency: evidence for a direct role of sex steroids. J Clin Endocrinol Metab 71:975–983

    Dietz WH 1994 Critical periods in childhood for the development of obesity. Am J Clin Nutr 59:955–959 [Review]

    Fall CH, Pandit AN, Law CM, Yajnik CS, Clark PM, Breier B, Osmond C, Shiell AW, Gluckman PD, Barker DJ 1995 Size at birth and plasma insulin-like growth factor-1 concentrations. Arch Dis Child 73:287–293

    Mann DR, Akinbami MA, Gould KG, Castracane VD 2000 A longitudinal study of leptin during development in the male rhesus monkey: the effect of body composition and season on circulating leptin levels. Biol Reprod 62:285–291

    Bouchard C, Tremblay A, Despardes JP, Nadeau A, Lupien PJ, Theriault G, Dussault J, Moorjani S, Pinault S, Fournier G 1990 The response to long-term overfeeding in identical twins. N Engl J Med 322:1477–1482

    Biaggi RR, Vollman MW, Nies MA, Brener CE, Flakoll PJ, Levenhagen DK, Sun M, Karabulut Z, Chen KY 1999 Comparison of air-displacement plethysmography with hydrostatic weighing and bioelectrical impedance analysis for the assessment of body composition in healthy adults. Am J Clin Nutr 69:898–903

    Clasey JL, Bouchard C, Teates CD, Riblett JE, Thorner MO, Hartman ML, Weltman A 1999 The use of anthropometric and dual-energy x-ray absorptiometry (DEXA) measures to estimate total abdominal and abdominal visceral fat in men and women. Obes Res 7:256–264

    Lesser G, Deutsch S, Markofsky J 1971 Use of independent measurement of body fat to evaluate overweight and underweight. Metabolism 20:792–804

    van Baak MA 1999 Physical activity and energy balance. Public Health Nutr 2:335–339 [Review]

    Durnin JV, Womersley J 1974 Body fat assessed from total body density and its estimation from skinfold thickness: measurements on 481 men and women aged from 16 to 72 years. Br J Nutr 32:77–97

    Mazess RB, Barden HS, Bisek JP, Hanson J 1990 Dual-energy x-ray absorptiometry for total-body and regional bone-mineral and soft-tissue composition. Am J Clin Nutr 51:1106–1112

    Brozek J, Grande F, Anderson JT, Keys A 1963 Densitometric analysis of body composition: revision of some quantitative assumptions. Ann NY Acad Sci 110:113–140

    Fields DA, Goran MI 2000 Body composition techniques and the four-compartment model in children. J Appl Physiol 89:613–620

    Cole TJ, Henson GL, Tremble JM, Colley NV 1997 Birthweight for length: ponderal index, body mass index or Benn index? Ann Hum Biol 24:289–298

    Rolland-Cachera MF, Cole TJ, Sempe M, Tichet J, Rossignol C, Charraud A 1991 Body mass index variations: centiles from birth to 87 years. Eur J Clin Nutr 45:13–21

    Cole TJ, Bellizzi MC, Flegal KM, Dietz WH 2000 Establishing a standard definition for child overweight and obesity worldwide: international survey. BMJ 320:1240–1243

    Khosla S, Melton III LJ, Atkinson EJ, O’Fallon WM 2001 Relationship of serum sex steroid levels to longitudinal changes in bone density in young versus elderly men. J Clin Endocrinol Metab 86:3555–3561

    Lindsay RS, Hanson RL, Rouman J, Ravussin E, Knowler WC, Tataranni A 2001 Body mass index as a measure of adiposity in children and adolescents: relationship to adiposity by dual energy x-ray absorptiometry and to cardiovascular risk factors. J Clin Endocrinol Metab 86:4061–4067

    Calle EE, Thun MJ, Petrelli JM, Rodriguez C, Heath Jr CW 1999 Body-mass index and mortality in a prospective cohort of U.S. adults. N Engl J Med 341:1097–1105

    Roemmich JN, Rogol AD 1999 Hormonal changes during puberty and their relationship to fat distribution. Am J Human Biol 11:209–224

    Cole TJ, Roede MJ 1999 Centiles of body mass index for Dutch children aged 0–20 years in 1980–a baseline to assess recent trends in obesity. Ann Hum Biol 26:303–308

    Cole TJ, Freeman JV, Preece MA 1995 Body mass index reference curves for the UK, 1990. Arch Dis Child 73:25–29

    Cole TJ, Freeman JV, Preece MA 1998 British 1990 growth reference centiles for weight, height, body mass index and head circumference fitted by maximum penalized likelihood. Stat Med 17:407–429

    Leung SS, Cole TJ, Tse LY, Lau JT 1998 Body mass index reference curves for Chinese children. Ann Hum Biol 25:169–174

    Siervogel RM, Maynard LM, Wisemandle WA, Roche AF, Guo SS, Chumlea WC, Towne B 2000 Annual changes in total body fat and fat-free mass in children from 8 to 18 years in relation to changes in body mass index. The Fels Longitudinal Study. Ann NY Acad Sci 904:420–423

    Johannsson G, Rosen T, Bengtsson BA 1997 Individualized dose titration of growth hormone (GH) during GH replacement in hypopituitary adults. Clin Endocrinol (Oxf) 47:571–581

    Daniels SR, Khoury PR, Morrison JA 1997 The utility of body mass index as a measure of body fatness in children and adolescents: differences by race and gender. Pediatrics 99:804–807

    Malina RM, Katzmarzyk PT 1999 Validity of the body mass index as an indicator of the risk and presence of overweight in adolescents. Am J Clin Nutr 70:131S–136S

    Pietrobelli A, Faith MS, Allison DB, Gallagher D, Chiumello G, Heymsfield SB 1998 Body mass index as a measure of adiposity among children and adolescents: a validation study. J Pediatr 132:204–210

    Plymate SR, Matej LA, Jones RE, Friedl KE 1988 Inhibition of sex hormone-binding globulin production in the human hepatoma (Hep G2) cell line by insulin and prolactin. J Clin Endocrinol Metab 67:460–464

    Fowler PA, Fuller MF, Glasbey CA, Foster MA, Cameron GG, McNeill G, Maughan RJ 1991 Total and subcutaneous adipose tissue in women: the measurement of distribution and accurate prediction of quantity by using magnetic resonance imaging. Am J Clin Nutr 54:18–25

    Pijl H, Langendonk JG, Burggraaf J, Frolich M, Cohen AF, Veldhuis JD, Meinders AE 2001 Altered neuroregulation of GH secretion in viscerally obese premenopausal women. J Clin Endocrinol Metab 86:5509–5515

    Armellini F, Zamboni M, De Pergola G, Bissoli L, Turcato E, Giorgino R, Bosello O 2000 Resting energy expenditure, growth hormone indices, body composition and adipose tissue distribution in premenopausal women. J Intern Med 247:709–714

    Palmert MR, Mansfield MJ, Crowley Jr WF, Crigler Jr JF, Crawford JD, Boepple PA 1999 Is obesity an outcome of gonadotropin-releasing hormone agonist administration? Analysis of growth and body composition in 110 patients with central precocious puberty. J Clin Endocrinol Metab 84:4480–4488

    Owen JB 1999 Genetic aspects of body composition. Nutrition 15:609–613

    Orme SM, Sebastian JP, Oldroyd B, Stewart SP, Grant PJ, Stickland MH, Smith MA, Belchetz PE 1992 Comparison of measures of body composition in a trial of low dose growth hormone replacement therapy. Clin Endocrinol (Oxf) 37:453–459

    Pasquali R, Casimirri F, Cantobelli S, Labate AM, Venturoli S, Paradisi R, Zannarini L 1993 Insulin and androgen relationships with abdominal body fat distribution in women with and without hyperandrogenism. Horm Res 39:179–187

    Mauras N 2001 GH and sex steroids: interactions in puberty. In: Klibanski A, ed. Neuroendocrine clinics of North America. Philadelphia: W. B. Saunders

    Andreassen TT, Melsen F, Oxlund H 1996 The influence of growth hormone on cancellous and cortical bone of the vertebral body in aged rats. J Bone Miner Res 11:1094–1102

    Ashton IK, Zapf J, Einschenk I, MacKenzie IZ 1985 Insulin-like growth factors (IGF) 1 and 2 in human foetal plasma and relationship to gestational age and foetal size during midpregnancy. Acta Endocrinol (Copenh) 110:558–563

    Chard T 1989 Hormonal control of growth in the human fetus. J Endocrinol 123:3–9 [Review]

    Bajoria R, Sooranna SR, Ward S, Hancock M 2002 Placenta as a link between amino acids, insulin-IGF axis, and low birth weight: evidence from twin studies. J Clin Endocrinol Metab 87:308–315

    Stanhope R, Preece MA, Hamill G 1991 Does growth hormone treatment improve final height attainment of children with intrauterine growth retardation? Arch Dis Child 66:1180–1183

    Iqbal J, Pompolo S, Considine RV, Clarke IJ 2000 Localization of leptin receptor-like immunoreactivity in the corticotropes, somatotropes, and gonadotropes in the ovine anterior pituitary. Endocrinology 141:1515–1520

    Jaquet D, Gaboriau A, Czernichow P, Levy-Marchal C 2000 Insulin resistance early in adulthood in subjects born with intrauterine growth retardation. J Clin Endocrinol Metab 85:1401–1406

    Gluckman PD 1997 Endocrine and nutritional regulation of prenatal growth. Acta Paediatr Suppl 423:153–157 [Review]

    Westwood M, Gibson JM, Sooranna SR, Ward S, Neilson JP, Bajoria R 2001 Genes or placenta as modulator of fetal growth: evidence from the insulin-like growth factor axis in twins with discordant growth. Mol Hum Reprod 7:387–395

    Katch FI, Michael ED, Horvath SM 1967 Estimation of body volume by underwater weighing: description of a single method. J Appl Physiol 23:811–816

    Wong WLT, Garg SJ, Woodruff T, Bald L, Fendly B, Lofgren JA 1993 Monoclonal antibody based ELISAs for measurement of activins in biological fluids. J Immunol Meth 165:1–10

    McCrory MA, Gomez TD, Bernauer EM, Mole PA 1995 Evaluation of a new air displacement plethysmograph for measuring human body composition. Med Sci Sports Exerc 27:1686–1691

    Devesa J, Lois N, Arce V, Diaz MJ, Lima L, Tresguerres JA 1991 The role of sexual steroids in the modulation of growth hormone (GH) secretion in humans. J Steroid Biochem Mol Biol 40:165–173

    Bland JM, Altman DG 1986 Statistical methods for assessing agreement between two methods of clinical measurement. Lancet 1:307–310

    Siri WE 1961 Body composition from fluid spaces and density: analysis of methods. In: Brozek J, Henschel A, eds. Techniques for measuring body composition. Washington DC: National Academy of Sciences/National Research Council

    Roemmich JN, Clark PA, Mai V, Berr SS, Weltman A, Veldhuis JD, Rogol AD 1998 Alterations in growth and body composition during puberty. III. Influence of maturation, gender, body composition, fat distribution, aerobic fitness, and energy expenditure on nocturnal growth hormone release. J Clin Endocrinol Metab 83:1440–1447

    Bosaeus I, Johannsson G, Rosen T, Hallgren P, Tolli J, Sjostrom L, Bengtsson BA 1996 Comparison of methods to estimate body fat in growth hormone deficient adults. Clin Endocrinol (Oxf) 44:395–402

    Heymsfield SB, Lichtman S, Baumgartner RN, Wang J, Kamen Y, Aliprantis A, Pierson RN 1990 Body composition of humans: comparison of two improved four-compartment models that differ in expense, technical complexity, and radiation exposure. Am J Clin Nutr 52:52–58

    Johnson J, Dawson-Hughes B 1991 Precision and stability of dual-energy x-ray absorptiometry measurements. Calcif Tissue Int 49:174–178

    Jorgensen JO, Thuesen L, Muller J, Ovesen P, Skakkebaek NE, Christiansen JS 1994 Three years of growth hormone treatment in growth hormone-deficient adults: near normalization of body composition and physical performance. Eur J Endocrinol 130:224–228

    Richelsen B, Pedersen SB, Borglum JD, Moller-Pedesen T, Jorgensen JO, Jorgensen JO 1994 Growth hormone treatment of obese women for 5 wk: effect on body composition and adipose tissue LPL activity. Am J Physiol 266:E211–E216

    Fisker S, Kristensen K, Rosenfalck AM, Pedersen SB, Ebdrup L, Richelsen B, Hilsted J, Christiansen JS, Jorgensen JO 2001 Gene expression of a truncated and the full-length growth hormone (GH) receptor in subcutaneous fat and skeletal muscle in GH-deficient adults: impact of GH treatment. J Clin Endocrinol Metab 86:792–796

    Laursen T, Gravholt CH, Heickendorff L, Drustrup J, Kappelgaard AM, Jorgensen JO, Christiansen JS 2001 Long-term effects of continuous subcutaneous infusion versus daily subcutaneous injections of growth hormone (GH) on the insulin-like growth factor system, insulin sensitivity, body composition, and bone and lipoprotein metabolism in GH-deficient adults. J Clin Endocrinol Metab 86:1222–1228

    Ohlsson C, Bengtsson BA, Isaksson OG, Andreassen TT, Slootweg MC 1998 Growth hormone and bone. Endocr Rev 19:55–79 [Review]

    Gotherstrom G, Svensson J, Koranyi J, Alpsten M, Bosaeus I, Bengtsson B, Johannsson G 2001 A prospective study of 5 years of GH replacement therapy in GH-deficient adults: sustained effects on body composition, bone mass, and metabolic indices. J Clin Endocrinol Metab 86:4657–4665

    Pierson Jr RN, Lin DH, Phillips RA 1974 Total-body potassium in health: effects of age, sex, height, and fat. Am J Physiol 226:206–212

    Landin K, Lindgarde F, Saltin B, Wilhelmsen L 1988 Decreased skeletal muscle potassium in obesity. Acta Med Scand 223:507–513

    Cohn SH, Vaswani AN, Yasumura S, Yuen K, Ellis KJ 1985 Assessment of cellular mass and lean body mass by noninvasive nuclear techniques. J Lab Clin Med 105:305–311

    Diessel E, Shepherd JA, Fuerst T, Gonzalez M, Genant HK, Carew B, Looker A 2000 Comparison of two phantoms for body composition with dual-energy x-ray absorptiometry. Ann NY Acad Sci 904:107–110

    Metzger DL, Kerrigan JR 1994 Estrogen receptor blockade with tamoxifen diminishes growth hormone secretion in boys: evidence for a stimulatory role of endogenous estrogens during male adolescence. J Clin Endocrinol Metab 79:513–518

    Brunton JA, Weiler HA, Atkinson SA 1997 Improvement in the accuracy of dual energy x-ray absorptiometry for whole body and regional analysis of body composition: validation using piglets and methodologic considerations in infants. Pediatr Res 41:590–596

    Pintauro SJ, Nagy TR, Duthie CM, Goran MI 1996 Cross-calibration of fat and lean measurements by dual-energy x-ray absorptiometry to pig carcass analysis in the pediatric body weight range. Am J Clin Nutr 63:293–298

    Albu JB, Kovera AJ, Johnson JA 2000 Fat distribution and health in obesity. Ann NY Acad Sci 904:491–501

    Enzi G, Gasparo M, Biondetti PR, Fiore D, Semisa M, Zurlo F 1986 Subcutaneous and visceral fat distribution according to sex, age, and overweight, evaluated by computed tomography. Am J Clin Nutr 44:739–746

    Borkan GA, Gerzof SG, Robbins AH, Hults DE, Silbert CK, Silbert JE 1982 Assessment of abdominal fat content by computed tomography. Am J Clin Nutr 36:172–177

    Rossner S, Bo WJ, Hiltbrandt E, Hinson W, Karstaedt N, Santago P, Sobol WT, Crouse JR 1990 Adipose tissue determinations in cadavers–a comparison between cross-sectional planimetry and computed tomography. Int J Obes 14:893–902

    Smith SR, Lovejoy JC, Greenway F, Ryan D, deJonge L, de la Bretonne J, Volafova J, Bray GA 2001 Contributions of total body fat, abdominal subcutaneous adipose tissue compartments, and visceral adipose tissue to the metabolic complications of obesity. Metabolism 50:425–435

    Abate N, Burns D, Peshock RM, Garg A, Grundy SM 1994 Estimation of adipose tissue mass by magnetic resonance imaging: validation against dissection in human cadavers. J Lipid Res 35:1490–1496

    Brook CG 1971 Determination of body composition of children from skinfold measurements. Arch Dis Child 46:182–184

    Kushner RF, Schoeller DA 1986 Estimation of total body water by bioelectrical impedance analysis. Am J Clin Nutr 44:417–424

    De Boer H, Blok GJ, Voerman HJ, De Vries PM, Van der Veen EA 1992 Body composition in adult growth hormone-deficient men, assessed by anthropometry and bioimpedance analysis. J Clin Endocrinol Metab 75:833–837

    Janssen YJH, Deurenberg P, Roelfsema F 1997 Using dilution techniques and multifrequency bioelectrical impedance to assess both total body water and extracellular water at baseline and during recombinant human growth hormone (GH) treatment in GH-deficient adults. J Clin Endocrinol Metab 82:3349–3355

    Fulcher GR, Farrer M, Walker M, Rodham D, Clayton B, Alberti KM 1991 A comparison of measurements of lean body mass derived by bioelectrical impedance, skinfold thickness and total body potassium. A study in obese and non-obese normal subjects. Scand J Clin Lab Invest 51:245–253

    Pirlich M, Biering H, Gerl H, Ventz M, Schmidt B, Ertl S, Lochs H 2002 Loss of body cell mass in Cushing’s syndrome: effect of treatment. J Clin Endocrinol Metab 87:1078–1084

    Lee PA, Plotnick LP, Steel PE, Thompson RG, Blizzard RM 1976 Integrated concentrations of luteinizing hormone and puberty. J Clin Endocrinol Metab 43:168–172

    Veldhuis JD 1996 Neuroendocrine mechanisms mediating awakening of the gonadotropic axis in puberty. Pediatr Nephrol 10:304–317

    Zadik Z, Chalew SA, Kowarski A 1990 Assessment of growth hormone secretion in normal stature children using 24-hour integrated concentration of GH and pharmacological stimulation. J Clin Endocrinol Metab 71:932–936

    Wennink JMB, Delemarre-van de Waal HA, Schoemaker R, Blaauw G, van den Brakern C, Schoemaker J 1991 Growth hormone secretion patterns in relation to LH and estradiol secretion throughout normal female puberty. Acta Endocrinol (Copenh) 124:129–135

    Delemarre-van de Waal HA, Wennink JM, Odink RJ 1991 Gonadotrophin and growth hormone secretion throughout puberty. Acta Paediatr Scand Suppl 372:26–31

    Giustina A, Veldhuis JD 1998 Pathophysiology of the neuroregulation of growth hormone secretion in experimental animals and the human. Endocr Rev 19:717–797

    Mueller EE, Locatelli V, Cocchi D 1999 Neuroendocrine control of growth hormone secretion. Physiol Rev 79:511–607

    Inui A 2001 Ghrelin: an orexigenic and somatotrophic signal from the stomach. Nat Rev Neurosci 2:551–560

    Veldhuis JD, Roemmich JN, Rogol AD 2000 Gender and sexual maturation-dependent contrasts in the neuroregulation of growth hormone secretion in prepubertal and late adolescent males and females–a general clinical research center-based study. J Clin Endocrinol Metab 85:2385–2394

    Palmert MR, Boepple PA 2001 Variation in the timing of puberty: clinical spectrum and genetic investigation. J Clin Endocrinol Metab 86:2364–2368 [Review]

    Liu L, Merriam GR, Sherins RJ 1987 Chronic sex steroid exposure increases mean plasma growth hormone concentration and pulse amplitude in men with isolated hypogonadotropic hypogonadism. J Clin Endocrinol Metab 64:651–656

    Bertelloni S, Baroncelli GI, Battini R, Perri G, Saggese G 1995 Short-term effect of testosterone treatment on reduced bone density in boys with constitutional delay of puberty. J Bone Miner Res 10:1488–1495

    Zachmann M, Prader A 1970 Anabolic and androgenic effect of testosterone in sexually immature boys and its dependency on growth hormone. J Clin Endocrinol Metab 30:85–92

    Young IR, Mesiano S, Hintz R, Caddy DJ, Ralph MM, Browne CA, Thorburn GD 1989 Growth hormone and testosterone can independently stimulate the growth of hypophysectomized prepubertal lambs without any alteration in circulating concentrations of insulin-like growth factors. J Endocrinol 121:563–570

    Ernst M, Rodan GA 1991 Estradiol regulation of insulin-like growth factor-I expression in osteoblastic cells: evidence for transcriptional control. Mol Endocrinol 5:1081–1089

    Slootweg MC, Swolin D, Netelenbos JC, Isaksson OG, Ohlsson C 1997 Estrogen enhances growth hormone receptor expression and growth hormone action in rat osteosarcoma cells and human osteoblast-like cells. J Endocrinol 155:159–164

    Baxter RC 2001 Changes in the IGF-IGFBP axis in critical illness. Best Pract Res Clin Endocrinol Metab 15:421–434 [Review]

    Yu H, Mistry J, Nicar MJ, Khosravi MJ, Diamandis A, van Doorn J, Juul A 1999 Insulin-like growth factors (IGF-I, free IGF-I and IGF-II) and insulin-like growth factor binding proteins (IGFBP-2, IGFBP-3, IGFBP-6, and ALS) in blood circulation. J Clin Lab Anal 13:166–172

    Stanhope R, Albanese A, Hindmarsh P, Brook CG 1992 The effects of growth hormone therapy on spontaneous sexual development. Horm Res 38(Suppl-1): 9–13

    Laron Z 1999 Natural history of the classical form of primary growth hormone (GH) resistance (Laron syndrome). J Pediatr Endocrinol Metab [Erratum (2001) 14:568] 12(Suppl 1):231–249 [Review]

    Arsenijevic Y, Wehrenberg WB, Conz A, Eshkol A, Sizonenko PC, Aubert ML 1989 Growth hormone (GH) deprivation induced by passive immunization against rat GH-releasing factor delays sexual maturation in the male rat. Endocrinology 124:3050–3059

    Jansson JO, Eden S, Isaksson O 1993 Sexual dimorphism in the control of growth hormone secretion. Endocr Rev 6:128–150

    Cersosimo E, Danou F, Persson M, Miles JM 1996 Effects of pulsatile delivery of basal growth hormone on lipolysis in humans. Am J Physiol 271:E123–E126

    Jorgensen JO, Moller N, Lauritzen T, Alberti KG, Orskov H, Christiansen JS 1990 Evening versus morning injections of growth hormone (GH) in GH-deficient patients: effects on 24-hour patterns of circulating hormones and metabolites. J Clin Endocrinol Metab 70:207–214

    Neely EK, Bachrach LK, Hintz RL, Habiby RL, Slemenda CW, Feezle L, Pescovitz OH 1995 Bone mineral density during treatment of central precocious puberty. J Pediatr 127:819–822

    Mericq MV, Eggers M, Avila A, Cutler Jr GB, Cassorla F 2000 Near final height in pubertal growth hormone (GH)-deficient patients treated with GH alone or in combination with luteinizing hormone-releasing hormone analog: results of a prospective, randomized trial. J Clin Endocrinol Metab 85:569–573

    Muller J, Juul A, Andersson AM, Sehested A, Skakkebaek NE 2000 Hormonal changes during GnRH analogue therapy in children with central precocious puberty. J Pediatr Endocrinol Metab 13(Suppl 1):739–746 [Review]

    DiMartino-Nardi J, Wu R, Fishman K, Saenger P 1991 The effect of long-acting analog of luteinizing hormone-releasing hormone on growth hormone secretory dynamics in children with precocious puberty. J Clin Endocrinol Metab 73:902–906

    Tanner JM, Whitehouse RH, Hughes PCR, Carter BS 1976 Relative importance of growth hormone and sex steroids for the growth at puberty of trunk length, limb length, and muscle width in growth hormone-deficient children. J Pediatr 89:1000–1008

    Tato L, Saggese G, Cavallo L, Antoniazzi F, Corrias A, Pasquino AM, Cisternino M 1995 Use of combined Gn-RH agonist and hGH therapy for better attaining the goals in precocious puberty treatment. Horm Res 44(Suppl 3):49–54

    Pasquino AM, Pucarelli I, Segni M, Matrunola M, Cerroni F, Cerrone F 1999 Adult height in girls with central precocious puberty treated with gonadotropin-releasing hormone analogues and growth hormone. J Clin Endocrinol Metab 84:449–452

    Daftary SS, Gore AC 2003 Developmental changes in hypothalamic insulin-like growth factor-1: relationship to gonadotropin-releasing hormone neurons. Endocrinology 144:2034–2045

    Hiney JK, Srivastava V, Nyberg CL, Ojeda SR, Dees WL 1996 Insulin-like growth factor I of peripheral origin acts centrally to accelerate the initiation of female puberty. Endocrinology 137:3717–3728

    Burks DJ, de Mora JF, Schubert M, Withers DJ, Myers MG, Towery HH, Altamuro SL, Flint CL, White MF 2000 IRS-2 pathways integrate female reproduction and energy homeostasis. Nature 407:377–382

    Khamsi F, Roberge S, Yavas Y, Lacanna IC, Zhu X, Wong J 2001 Recent discoveries in physiology of insulin-like growth factor-1 and its interaction with gonadotropins in folliculogenesis. Endocrine 16:151–165

    Wilson ME 1998 Premature elevation in serum insulin-like growth factor-I advances first ovulation in rhesus monkeys. J Endocrinol 158:247–257

    Suter KJ, Pohl CR, Wilson ME 2000 Circulating concentrations of nocturnal leptin, growth hormone, and insulin-like growth factor-I increase before the onset of puberty in agonadal male monkeys: potential signals for the initiation of puberty. J Clin Endocrinol Metab 85:808–814

    Wilson ME 1995 IGF-I administration advances the decrease in hypersensitivity to oestradiol negative feedback inhibition of serum LH in adolescent female rhesus monkeys. J Endocrinol 145:121–130

    Adashi EY, Hsueh AJW, Yen SSC 1981 Insulin enhancement of luteinizing hormone and follicle stimulating hormone release by cultured pituitary cells. Endocrinology 108:1441–1449

    Urban RJ, Garmey JC, Shupnik MA, Veldhuis JD 1990 Insulin-like growth factor type I increases concentrations of messenger ribonucleic acid encoding cytochrome P450 cholesterol side-chain cleavage enzyme in primary cultures of porcine granulosa cells. Endocrinology 127:2481–2488

    Keenan BS, Richards GE, Ponder SW, Dallas JS, Nagamani M, Smith ER 1993 Androgen-stimulated pubertal growth: the effects of testosterone and dihydrotestosterone on growth hormone and insulin-like growth factor-I in the treatment of short stature and delayed puberty. J Clin Endocrinol Metab 76:996–1001

    Kulin HE, Samojlik E, Santen RJ, Santner S 1981 The effect of growth hormone on the Leydig cell response to chorionic gonadotropin in boys with hypopituitarism. Clin Endocrinol (Oxf) 15:463–472

    Fisker S, Norrelund H, Juul A, Skakkebaek NE, Christiansen JS, Jorgensen JO 2001 The growth hormone (GH)-insulin-like growth factor axis during testosterone replacement therapy in GH-treated hypopituitary males. Growth Horm IGF Res 11:104–109

    Phillip M, Palese T, Hernandez ER, Roberts Jr CT, LeRoith D, Kowarski AA 1992 Effect of testosterone on insulin-like growth factor-I (IGF-I) and IGF-I receptor gene expression in the hypophysectomized rat. Endocrinology 130:2865–2870

    Fryburg DA, Weltman A, Jahn LA, Weltman JY, Samojlik E, Hintz RL, Veldhuis JD 1999 Androgenic modulation of the growth hormone-IGF axis and its impact on metabolic outcomes. In: Veldhuis JD, Giustina A, eds. Sex-steroid interaction with growth hormone. New York: Springer-Verlag, Inc.; 82–92

    Arslanian S, Suprasongsin C 1997 Testosterone treatment in adolescents with delayed puberty: changes in body composition, protein, fat, and glucose metabolism. J Clin Endocrinol Metab 82:3213–3220

    van Kesteren P, Lips P, Deville W, Popp-Snijders C, Asscheman H, Megens, J, Gooren L 1996 The effect of one-year cross-sex hormonal treatment on bone metabolism and serum insulin-like growth factor-1 in transsexuals. J Clin Endocrinol Metab 81:2227–2232

    Giustina A, Scalvini T, Tassi C, Desenzani P, Poiesi C, Wehrenberg WB, Rogol A, Veldhuis JD 1997 Maturation of the regulation of growth hormone secretion in young males with hypogonadotropic hypogonadism pharmacologically exposed to progressive increments in serum testosterone. J Clin Endocrinol Metab 82:1210–1219

    Fryburg DA, Weltman A, Jahn LA, Weltman JY, Samolijik E, Veldhuis JD 1997 Short-term modulation of the androgen milieu alters pulsatile but not exercise or GHRH-stimulated GH secretion in healthy men. J Clin Endocrinol Metab 82:3710–3719

    Bhasin S, Woodhouse L, Casaburi R, Singh AB, Bhasin D, Berman N, Chen X, Yarasheski KE, Magliano L, Dzekov C, Dzekov J, Bross R, Phillips J, Sinha-Hikim I, Shen R, Storer TW 2001 Testosterone dose-response relationships in healthy young men. Am J Physiol 281:E1172–E1181

    Contreras B, Talamantes F 1999 Growth hormone (GH) and 17?-estradiol regulation of the expression of mouse GH receptor and GH-binding protein in cultured mouse hepatocytes. Endocrinology 140:4725–4731

    Bennett PA, Levy A, Carmignac DF, Robinson IC, Lightman SL 1996 Differential regulation of the growth hormone receptor gene: effects of dexamethasone and estradiol. Endocrinology 137:3891–3896

    Yu YM, Domene HM, Sztein J, Counts DR, Cassorla F 1996 Developmental changes and differential regulation by testosterone and estradiol of growth hormone receptor expression in the rabbit. Eur J Endocrinol 135:583–590

    Vahl N, Moller N, Lauritzen T, Christiansen JS, Jorgensen JO 1997 Metabolic effects and pharmacokinetics of a growth hormone pulse in healthy adults: relation to age, sex, and body composition. J Clin Endocrinol Metab 82:1–7

    Juul A, Bang P, Hertel NT, Main K, Dalgaard P, Jorgensen K, Muller J, Hall K, Skakkebaek NE 1994 Serum insulin-like growth factor-I in 1030 healthy children, adolescents, and adults: relation to age, sex, stage of puberty, testicular size, and body mass index. J Clin Endocrinol Metab 78:744–752

    Johannsson G, Bjarnason R, Bramnert M, Carlsson LM, Degerblad M, Manhem P, Rosen T, Thoren M, Bengtsson BA 1996 The individual responsiveness to growth hormone (GH) treatment in GH-deficient adults is dependent on the level of GH-binding protein, body mass index, age, and gender. J Clin Endocrinol Metab 81:1575–1581

    Janssen YJH, Helmerhorst F, Frolich M, Roelfsema F 2000 A switch from oral (2 mg/d) to transdermal (50 μg/d) 17?-estradiol therapy increases serum insulin-like growth factor-I levels in recombinant human growth hormone (GH)-substituted women with GH deficiency. J Clin Endocrinol Metab 85:464–467

    Hertz P, Silbermann M, Even L, Hochberg Z 1989 Effects of sex steroids on the response of cultured rat pituitary cells to growth hormone-releasing hormone and somatostatin. Endocrinology 125:581–585

    Moller N, Schmitz O, Porksen N, Moller J, Jorgensen JO 1992 Dose-response studies on the metabolic effects of a growth hormone pulse in humans. Metabolism 41:172–175

    Klein NA, Battaglia DE, Miller PB, Soules MR 1996 Circulating levels of growth hormone, insulin-like growth factor-I and growth hormone binding protein in normal women of advanced reproductive age. Clin Endocrinol (Oxf) 44:285–292

    Engstrom BE, Karlsson FA, Wide L 1998 Marked gender differences in ambulatory morning growth hormone values in young adults. Clin Chem 44:1289–1295

    Ostrer H 2001 Invited review: sex-based differences in gene expression. J Appl Physiol 91:2384–2388

    Sivitz WI, Walsh SA, Morgan DA, Thomas MJ, Haynes WG 1997 Effects of leptin on insulin sensitivity in normal rats. Endocrinology 138:3395–3401

    Elias CF, Lee CE, Kelly JF, Ahima RS, Kuhar M, Saper CB, Elmquist JK 2001 Characterization of CART neurons in the rat and human hypothalamus. J Comp Neurol 432:1–19

    Ahima RS, Kelly J, Elmquist JK, Flier JS 1999 Distinct physiologic and neuronal responses to decreased leptin and mild hyperleptinemia. Endocrinology 140:4923–4931

    Jackson JA, Riggs MW, Spiekerman AM 1992 Testosterone deficiency as a risk factor for hip fractures in men: a case-control study. Am J Med Sci 304:4–8

    Melton III LJ, Khosla S, Atkinson EJ, Oconnor MK, Ofallon WM, Riggs BL 2000 Cross-sectional versus longitudinal evaluation of bone loss in men and women. Osteoporos Int 11:592–599

    Finkelstein JS, Klibanski A, Neer RM, Greenspan S, Rosenthal DI, Crowley Jr WF 1987 Osteoporosis in men with idiopathic hypogonadotropic hypogonadism. Ann Intern Med 106:354–361

    Tollefsen SE, Sadow JL, Rotwein P 1989 Coordinate expression of insulin-like growth factor II and its receptor during muscle differentiation. Proc Natl Acad Sci USA 86:1543–1547

    Katzman DK, Bachrach LK, Carter DR, Marcus R 1991 Clinical and anthropometric correlates of bone mineral acquisition in healthy adolescent girls. J Clin Endocrinol Metab 73:1332–1339

    Szulc P, Munoz F, Claustrat B, Garnero P, Marchand F, Duboeuf F, Delmas PD 2001 Bioavailable estradiol may be an important determinant of osteoporosis in men: the MINOS study. J Clin Endocrinol Metab 86:192–199

    van den Beld AW, De Jong FH, Grobbee DE, Pols HA, Lamberts SW 2000 Measures of bioavailable serum testosterone and estradiol and their relationships with muscle strength, bone density, and body composition in elderly men. J Clin Endocrinol Metab 85:3276–3282

    Ongphiphadhanakul B, Rajatanavin R, Chanprasertyothin S, Piaseu N, Chailurkit L 1998 Serum oestradiol and oestrogen-receptor gene polymorphism are associated with bone mineral density independently of serum testosterone in normal males. Clin Endocrinol (Oxf) 49:803–809

    Yanovski JA, Sovik KN, Nguyen TT, Sebring NG 2000 Insulin-like growth factors and bone mineral density in African American and White girls. J Pediatr 137:826–832

    2000 Consensus guidelines for the diagnosis and treatment of growth hormone (GH) deficiency in childhood and adolescence: summary statement of the GH Research Society. GH Research Society. J Clin Endocrinol Metab 85:3990–3993

    Seeman E, Hopper JL, Young NR, Formica C, Goss P, Tsalamandris C 1996 Do genetic factors explain associations between muscle strength, lean mass, and bone density? A twin study. Am J Physiol 270:E320–E327

    Rosen CJ, Kurland ES, Vereault D, Adler RA, Rackoff PJ, Craig WY, Witte S, Rogers J, Bilezikian JP 1998 Association between serum insulin growth factor-I (IGF-I) and a simple sequence repeat in IGF-I gene: implications for genetic studies of bone mineral density. J Clin Endocrinol Metab 83:2286–2290

    Gilsanz V, Skaggs DL, Kovanlikaya A, Sayre J, Loro ML, Kaufman F, Korenman SG 1998 Differential effect of race on the axial and appendicular skeletons of children. J Clin Endocrinol Metab 83:1420–1427

    Guo C-Y, Jones TH, Eastell R 1997 Treatment of isolated hypogonadotropic hypogonadism effect on bone mineral density and bone turnover. J Clin Endocrinol Metab 82:658–665

    Katznelson L, Finkelstein JS, Schoenfeld DA, Rosenthal DI, Anderson EJ, Klibanski A 1996 Increase in bone density and lean body mass during testosterone administration in men with acquired hypogonadism. J Clin Endocrinol Metab 81:4358–4365

    Snyder PJ, Peachey H, Berlin JA, Hannoush P, Haddad G, Dlewati A, Santanna J, Loh L, Lenrow DA, Holmes JH, Kapoor SC, Atkinson LE, Strom BL 2000 Effects of testosterone replacement in hypogonadal men. J Clin Endocrinol Metab 85:2670–2677

    Jansson JO, Albertsson-Wikland K, Eden S, Thorngren KG, Isaksson OGP 1982 Effect of frequency of growth hormone administration on longitudinal bone growth and body weight in hypophysectomized rats. Acta Physiol Scand 114:261–265

    Jansson JO, Eden S, Isaksson O 1983 Sites of action of testosterone and estradiol on longitudinal bone growth. Am J Physiol 244:E135–E140

    Veldhuis JD, Metzger DL, Martha Jr PM, Mauras N, Kerrigan JR, Keenan B, Rogol AD, Pincus SM 1997 Estrogen and testosterone, but not a non-aromatizable androgen, direct network integration of the hypothalamo-somatotrope (growth hormone)-insulin-like growth factor I axis in the human: evidence from pubertal pathophysiology and sex-steroid hormone replacement. J Clin Endocrinol Metab 82:3414–3420

    Gevers EF, Wit JM, Robinson IC 1995 Effect of gonadectomy on growth and GH responsiveness in dwarf rats. J Endocrinol 145:69–79

    Attie KM 2000 The importance of growth hormone replacement therapy for bone mass in young adults with growth hormone deficiency. J Pediatr Endocrinol Metab 13(Suppl 2):1011–1021 [Review]

    Abrams SA 1998 The relationship between magnesium and calcium kinetics in 9- to 14-year-old children. J Bone Miner Res 13:149–153

    Kochakian CD 1937 Testosterone and testosterone acetate and the protein and energy metabolism of castrate dogs. Endocrinol 21:750–755

    Krotkiewski M, Kral JG, Karlsson J 1980 Effects of castration and testosterone substitution on body composition and muscle metabolism in rats. Acta Physiol Scand 109:233–237

    Mauras N, Haymond MW, Darmaun D, Vieira NE, Abrams SA, Yergey AL 1994 Calcium and protein kinetics in prepubertal boys. Positive effects of testosterone. J Clin Invest 93:1014–1019

    Musaro A, McCullagh K, Paul A, Houghton L, Dobrowolny G, Molinaro M, Barton ER, Sweeney HL, Rosenthal N 2001 Localized IGF-1 transgene expression sustains hypertrophy and regeneration in senescent skeletal muscle. Nat Genet 27:195–200

    Barton-Davis ER, Shoturma DI, Musaro A, Rosenthal N, Sweeney HL 1998 Viral mediated expression of insulin-like growth factor I blocks the aging-related loss of skeletal muscle function. Proc Natl Acad Sci USA 95:15603–15607

    Coleman ME, DeMayo F, Yin KC, Lee HM, Geske R, Montgomery C, Schwartz RJ 1995 Myogenic vector expression of insulin-like growth factor I stimulates muscle cell differentiation and myofiber hypertrophy in transgenic mice. J Biol Chem 270:12109–12116

    Kambadur R, Sharma M, Smith TP, Bass JJ 1997 Mutations in myostatin (GDF8) in double-muscled Belgian Blue and Piedmontese cattle. Genome Res 7:910–916

    Illig R, Prader A 1970 Effect of testosterone on growth hormone secretion in patients with anorchia and delayed puberty. J Clin Endocrinol Metab 30:615–618

    Eakman GD, Dallas JS, Ponder SW, Keenan BS 1996 The effects of testosterone and dihydrotestosterone on hypothalamic regulation of growth hormone secretion. J Clin Endocrinol Metab 81:1217–1223

    Maor G, Segev Y, Phillip M 1999 Testosterone stimulates insulin-like growth factor-I and insulin-like growth factor-I-receptor gene expression in the mandibular condyle—a model of endochondral ossification. Endocrinology 140:1901–1910

    Fisker S, Jorgensen JO, Vahl N, Orskov H, Christiansen JS 1999 Impact of gender and androgen status on IGF-I levels in normal and GH-deficient adults. Euro J Endocrinol 141:601–608

    Bailey DA, McKay HA, Mirwald RL, Crocker PR, Faulkner RA 1999 A six-year longitudinal study of the relationship of physical activity to bone mineral accrual in growing children: the university of Saskatchewan bone mineral accrual study. J Bone Miner Res 14:1672–1679

    Soyka LA, Fairfield WP, Klibanski A 2000 Clinical review 117: hormonal determinants and disorders of peak bone mass in children. J Clin Endocrinol Metab 85:3951–3963 [Review]

    Drake WM, Rodriguez-Arnao J, Weaver JU, James IT, Coyte D, Spector TD, Besser GM, Monson JP 2001 The influence of gender on the short and long-term effects of growth hormone replacement on bone metabolism and bone mineral density in hypopituitary adults: a 5-year study. Clin Endocrinol (Oxf) 54:525–532

    Malhotra A, Poon E, Tse WY, Pringle PJ, Hindmarsh PC, Brook CG 1993 The effects of oxandrolone on the growth hormone and gonadal axes in boys with constitutional delay of growth and puberty. Clin Endocrinol (Oxf) 38:393–398

    Stanhope R, Buchanan CR, Fenn GC, Preece MA 1988 Double blind placebo controlled trial of low dose oxandrolone in the treatment of boys with constitutional delay of growth and puberty. Arch Dis Child 63:501–505

    Huber DM, Bendixen AC, Pathrose P, Srivastava S, Dienger KM, Shevde NK, Pike JW 2001 Androgens suppress osteoclast formation induced by RANKL and macrophage-colony stimulating factor. Endocrinology 142:3800–3808

    Bellido T, Jilka RL, Boyce BF, Girasole G, Broxmeyer H, Dalrymple SA, Murray R, Manolagas SC 1995 Regulation of interleukin-6, osteoclastogenesis, and bone mass by androgens. The role of the androgen receptor. J Clin Invest 95:2886–2895

    Kousteni S, Bellido T, Plotkin LI, O’Brien CA, Bodenner DL, Han L, Han K, DiGregorio GB, Katzenellenbogen JA, Katzenellenbogen BS, Roberson PK, Weinstein RS, Jilka RL, Manolagas SC 2001 Nongenotropic, sex-nonspecific signaling through the estrogen or androgen receptors: dissociation from transcriptional activity. Cell 104:719–730

    Marcus R, Leary D, Schneider DL, Shane E, Favus M, Quigley CA 2000 The contribution of testosterone to skeletal development and maintenance: lessons from the androgen insensitivity syndrome. J Clin Endocrinol Metab 85:1032–1037

    Hojbjerg GC, Svenstrup B, Bennett P, Sandahl Christiansen J 1999 Reduced androgen levels in adult Turner syndrome: influence of female sex steroids and growth hormone status. Clin Endocrinol (Oxf) 50:791–800

    Rosenfeld RG, Frane J, Atatie KM, Brasel JA, Burstein S, Cara JF, Chernausek S, Gotlin RW, Kuntze J, Lippe BM 1992 Six-year results of a randomized, prospective trial of human growth hormone and oxandrolone in Turner syndrome. J Pediatr 121:49–55

    Nilsson KO, Albertsson-Wikland K, Alm J, Aronson S, Gustafsson J, Hagenas L, Hager A, Ivarsson SA, Karlberg J, Kristrom B, Marcus C, Moell C, Ritzen M, Tuvemo T, Wattsgard C, Westgren U, Westphal O, Aman J 1996 Improved final height in girls with Turner’s syndrome treated with growth hormone and oxandrolone. J Clin Endocrinol Metab 81:635–640

    Chernausek SD, Attie KM, Cara JF, Rosenfeld RG, Frane J 2000 Growth hormone therapy of Turner syndrome: the impact of age of estrogen replacement on final height. Genentech, Inc., Collaborative Study Group. J Clin Endocrinol Metab 85:2439–2445

    Cacciari E, Mazzanti L 1999 Final height of patients with Turner’s syndrome treated with growth hormone (GH): indications for GH therapy alone at high doses and late estrogen therapy. Italian Study Group for Turner Syndrome. J Clin Endocrinol Metab 84:4510–4515

    Vanderschueren D, Boonen S, Ederveen AG, De Coster R, Van Herck E, Moermans K, Vandenput L, Verstuyf A, Bouillon R 2000 Skeletal effects of estrogen deficiency as induced by an aromatase inhibitor in an aged male rat model. Bone 27:611–617

    Colvard DS, Eriksen EF, Keeting PE, Wilson EM, Lubahn DB, French FS, Riggs BL, Spelsberg TC 1989 Identification of androgen receptors in normal human osteoblast-like cells. Proc Natl Acad Sci USA 86:854–857

    Siiteri PK, Wilson JD 1974 Testosterone formation and metabolism during male sexual differentiation in the human embryo. J Clin Endocrinol Metab 38:113–125

    Vidal O, Lindberg MK, Hollberg K, Baylink DJ, Andersson G, Lubahn DB, Mohan S, Gustafsson JA, Ohlsson C 2000 Estrogen receptor specificity in the regulation of skeletal growth and maturation in male mice. Proc Natl Acad Sci USA 97:5474–5479

    Cicognani A, Cacciari E, Tacconi M, Pascucci MG, Tonioli S, Pirazzoli P, Balsamo A 1989 Effect of gonadectomy on growth hormone, IGF-I and sex steroids in children with complete and incomplete androgen insensitivity. Acta Endocrinol (Copenh) 121:777–783

    Chalbos D, Philips A, Rochefort H 1994 Genomic cross-talk between the estrogen receptor and growth factor regulatory pathways in estrogen target tissues. Semin Cancer Biol 5:361–368 [Review]

    Vandenput L, Ederveen AG, Erben RG, Stahr K, Swinnen JV, Van Herck E, Verstuyf A, Boonen S, Bouillon R, Vanderschueren D 2001 Testosterone prevents orchidectomy-induced bone loss in estrogen receptor- knockout mice. Biochem Biophys Res Commun 285:70–76

    Hansen TB, Brixen K, Vahl N, Jorgensen JO, Christiansen JS, Mosekilde L, Hagen C 1996 Effects of 12 months of growth hormone (GH) treatment on calciotropic hormones, calcium homeostasis, and bone metabolism in adults with acquired GH deficiency: a double blind, randomized, placebo-controlled study. J Clin Endocrinol Metab 81:3352–3359

    Taxel P, Kennedy DG, Fall PM, Willard AK, Clive JM, Raisz LG 2001 The effect of aromatase inhibition on sex steroids, gonadotropins, and markers of bone turnover in older men. J Clin Endocrinol Metab 86:2869–2874

    MacGillivray MH, Morishima A, Conte F, Grumbach M, Smith EP 1998 Pediatric endocrinology update: an overview. The essential roles of estrogens in pubertal growth, epiphyseal fusion and bone turnover: lessons from mutations in the genes for aromatase and the estrogen receptor. Horm Res 49(Suppl 1):2–8 [Review]

    Bilezikian JP, Morishima A, Bell J, Grumbach MM 1998 Increased bone mass as a result of estrogen therapy in a man with aromatase deficiency. N Engl J Med 339:599–603

    Manolagas SC 2000 Birth and death of bone cells: basic regulatory mechanisms and implications for the pathogenesis and treatment of osteoporosis. Endocr Rev 21:115–137

    Miyaura C, Toda K, Inada M, Ohshiba T, Matsumoto C, Okada T, Ito M, Shizuta Y, Ito A 2001 Sex- and age-related response to aromatase deficiency in bone. Biochem Biophys Res Commun 280:1062–1068

    Turner RT, Wakley GK, Hannon KS 1990 Differential effects of androgens on cortical bone histomorphometry in gonadectomized male and female rats. J Orthop Res 8:612–617

    Ly LP, Jimenez M, Zhuang TN, Celermajer DS, Conway AJ, Handelsman DJ 2001 A double-blind, placebo-controlled, randomized clinical trial of transdermal dihydrotestosterone gel on muscular strength, mobility, and quality of life in older men with partial androgen deficiency. J Clin Endocrinol Metab 86:4078–4088

    Eberle AJ, Sparrow JT, Keenan BS 1986 Treatment of persistent pubertal gynecomastia with dihydrotestosterone heptanoate. J Pediatr 109:144–149

    Mahendroo MS, Russell DW 1999 Male and female isoenzymes of steroid 5-reductase. Rev Reprod 4:179–183 [Review]

    Kassem M 1997 Cellular and molecular effects of growth hormone and estrogen on human bone cells. APMIS Suppl 71:1–30 [Review]

    Libanati C, Baylink DJ, Lois-Wenzel E, Srinvasan N, Mohan S 1999 Studies on the potential mediators of skeletal changes occurring during puberty in girls. J Clin Endocrinol Metab 84:2807–2814

    Mauras N, Doi SQ, Shapiro JR 1996 Recombinant human insulin-like growth factor I, recombinant human growth hormone, and sex steroids: effects on markers of bone turnover in humans. J Clin Endocrinol Metab 81:2222–2226

    Mauras N, Klein KO, Hayes V 2001 Estrogen suppression in males: metabolic effects. J Clin Endocrinol Metab 85:2370–2377

    Mauras N, Vieira NE, Yergey AL 1997 Estrogen therapy enhances calcium absorption and retention and diminishes bone turnover in young girls with Turner’s syndrome: a calcium kinetic study. Metabolism 46:908–913

    Turner RT, Riggs BL, Spelsberg TC 1994 Skeletal effects of estrogen. Endocr Rev 15:275–300 [Review]

    Oleksik AM, Duong T, Pliester N, Asma G, Popp-Snijders C, Lips P 2001 Effects of the selective estrogen receptor modulator, raloxifene, on the somatotropic axis and insulin-glucose homeostasis. J Clin Endocrinol Metab 86:2763–2768

    Tomkinson A, Reeve J, Shaw RW, Noble BS 1997 The death of osteocytes via apoptosis accompanies estrogen withdrawal in human bone. J Clin Endocrinol Metab 82:3128–3135

    Hofbauer LC, Khosla S, Dunstan CR, Lacey DL, Spelsberg TC, Riggs BL 1999 Estrogen stimulates gene expression and protein production of osteoprotegerin in human osteoblastic cells. Endocrinology 140:4367–4370

    Shevde NK, Bendixen AC, Dienger KM, Pike JW 2000 Estrogens suppress RANK ligand-induced osteoclast differentiation via a stromal cell independent mechanism involving c-Jun repression. Proc Natl Acad Sci USA 97:7829–7834

    Srivastava S, Toraldo G, Weitzmann MN, Cenci S, Ross FP, Pacifici R 2001 Estrogen decreases osteoclast formation by down-regulating receptor activator of NF-B ligand (RANKL)-induced JNK activation. J Biol Chem 276:8836–8840

    Nasu M, Sugimoto T, Kaji H, Chihara K 2000 Estrogen modulates osteoblast proliferation and function regulated by parathyroid hormone in osteoblastic SaOS-2 cells: role of insulin-like growth factor (IGF)-I and IGF-binding protein-5. J Endocrinol 167:305–313

    Denger S, Reid G, Kos M, Flouriot G, Parsch D, Brand H, Korach KS, Sonntag-Buck V, Gannon F 2001 ER gene expression in human primary osteoblasts: evidence for the expression of two receptor proteins. Mol Endocrinol 15:2064–2077

    Eriksen EF, Colvard DS, Berg NJ, Graham ML, Mann KG, Spelsberg TC, Riggs BL 1988 Evidence of estrogen receptor in normal human osteoblast-like cells. Science 241:84–86

    Waters KM, Rickard DJ, Riggs BL, Khosla S, Katzenellenbogen JA, Katzenellenbogen BS, Moore J, Spelsberg TC 2001 Estrogen regulation of human osteoblast function is determined by the stage of differentiation and the estrogen receptor isoform. J Cell Biochem 83:448–462

    Arts J, Kuiper GG, Janssen JM, Gustafsson JA, Lowik CW, Pols HA, van Leeuwen JP 1997 Differential expression of estrogen receptors and ? mRNA during differentiation of human osteoblast SV-HFO cells. Endocrinology 138:5067–5070

    Morishima A, Grumbach MM, Simpson ER, Fisher C, Qin K 1995 Aromatase deficiency in male and female siblings caused by a novel mutation and the physiological role of estrogens. J Clin Endocrinol Metab 80:3689–3698

    Windahl SH, Hollberg K, Vidal O, Gustafsson JA, Ohlsson C, Andersson G 2001 Female estrogen receptor ?–/– mice are partially protected against age-related trabecular bone loss. J Bone Miner Res 16:1388–1398

    Carani C, Qin K, Simoni M 1997 Effect of testosterone and estradiol in a man with aromatase deficiency. N Engl J Med 337:91–95

    Smith EP, Boyd J, Frank GR, Takahashi H, Cohen RM, Specker B, Williams TC, Lubahn DB, Korach KS 1994 Estrogen resistance caused by a mutation in the estrogen-receptor gene in a man. N Engl J Med 331:1056–1061

    Rochira V, Faustini-Fustini M, Balestrieri A, Carani C 2000 Estrogen replacement therapy in a man with congenital aromatase deficiency: effects of different doses of transdermal estradiol on bone mineral density and hormonal parameters. J Clin Endocrinol Metab 85:1841–1845

    Vanderschueren D, Van Herck E, De Coster R, Bouillon R 1996 Aromatization of androgens is important for skeletal maintenance of aged male rats. Calcif Tissue Int 59:179–183

    Vanderschueren D, Van Herck E, Nijs J, Ederveen AG, De Coster R, Bouillon R 1997 Aromatase inhibition impairs skeletal modeling and decreases bone mineral density in growing male rats. Endocrinology 138:2301–2307

    Schmid C, Ernst M, Zapf J, Froesch ER 1989 Release of insulin-like growth factor carrier proteins by osteoblasts: stimulation by estradiol and growth hormone. Biochem Biophys Res Commun 160:788–794

    Urban RJ, Evans WS, Rogol AD, Kaiser DL, Johnson ML, Veldhuis JD 1988 Contemporary aspects of discrete peak detection algorithms. I. The paradigm of the luteinizing hormone pulse signal in men. Endocr Rev 9:3–37

    Evans WS, Sollenberger MJ, Booth Jr RA, Rogol AD, Urban RJ, Carlsen EC, Johnson ML, Veldhuis JD 1992 Contemporary aspects of discrete peak detection algorithms. II. The paradigm of the luteinizing hormone pulse signal in women. Endocr Rev 13:81–104

    Schnorr JA, Bray MJ, Veldhuis JD 2001 Aromatization mediates testosterone’s short-term feedback restraint of 24-hour endogenously driven and acute exogenous GnRH-stimulated LH and FSH secretion in young men. J Clin Endocrinol Metab 86:2600–2606

    Kerrigan JR, Veldhuis JD, Rogol AD 1994 Androgen-receptor blockade enhances pulsatile luteinizing hormone production in late pubertal males: evidence for a hypothalamic site of physiological androgen feedback action. Pediatr Res 35:102–106

    Urban RJ, Davis MR, Rogol AD, Johnson ML, Veldhuis JD 1988 Acute androgen receptor blockade increases luteinizing-hormone secretory activity in men. J Clin Endocrinol Metab 67:1149–1155

    Veldhuis JD, Bidlingmaier M, Anderson SM, Wu Z, Strassburger CJ 2001 Lowering total plasma insulin-like growth factor I concentrations by way of a novel, potent, and selective growth hormone (GH) receptor antagonist, pegvisomant (B2036-peg), augments the amplitude of GH secretory bursts and elevates basal/nonpulsatile GH release in healthy women and men. J Clin Endocrinol Metab 86:3304–3310

    Veldhuis JD, Bidlingmaier M, Anderson SM, Evans WS, Wu Z, Strassburger CJ 2002 Impact of experimental blockade of peripheral growth hormone (GH) receptors on the kinetics of endogenous and exogenous GH removal in healthy women and men. J Clin Endocrinol Metab 87:5737–5745

    Veldhuis JD, Anderson SM, Kok P, Iranmanesh A, Frystyk J, Orskov H, Keenan DM 2004 Estradiol supplementation modulates growth hormone (GH) secretory-burst waveform and recombinant human insulin-like growth factor-I-enforced suppression of endogenously driven GH release in postmenopausal women. J Clin Endocrinol Metab 89:1312–1318

    Yakar S, Rosen CJ, Beamer WG, Ackert-Bicknell CL, Wu Y, Liu JL, Ooi GT, Setser J, Frystyk J, Boisclair YR, LeRoith D 2002 Circulating levels of IGF-1 directly regulate bone growth and density. J Clin Invest 110:771–781

    Scott CJ, Kuehl DE, Ferreira SA, Jackson GL 1997 Hypothalamic sites of action for testosterone, dihydrotestosterone, and estrogen in the regulation of luteinizing hormone secretion in male sheep. Endocrinology 138:3686–3694

    Resko JA, Connolly PB, Roselli CE, Abdelgadir SE, Choate JV 1993 Differential effects of aromatase inhibition on luteinizing hormone secretion in intact and castrated male cynomolgus macaques. J Clin Endocrinol Metab 77:1529–1534

    Hayes FJ, Seminara SB, DeCruz S, Boepple PA, Crowley Jr WF 2000 Aromatase inhibition in the human male reveals a hypothalamic site of estrogen feedback. J Clin Endocrinol Metab 85:3027–3035

    Hall JM, McDonnell DP 1999 The estrogen receptor ?-isoform (ER?) of the human estrogen receptor modulates ER transcriptional activity and is a key regulator of the cellular response to estrogens and antiestrogens. Endocrinology 140:5566–5578

    Zhou J, Ng S, Adesanya-Famuiya O, Anderson K, Bondy CA 2000 Testosterone inhibits estrogen-induced mammary epithelial proliferation and suppresses estrogen receptor expression. FASEB J 14:1725–1730

    Handa RJ, Resko JA 1988 Effects of gonadectomy and hormone replacement on steroid hormone receptors and 5 -reductase activity in pituitaries of male rhesus macaques. J Clin Endocrinol Metab 66:1251–1258

    Reiter E, Bonnet P, Sente B, Dombrowicz D, De Leval J, Closset J, Hennen G 1992 Growth hormone and prolactin stimulate androgen receptor, insulin-like growth factor-I (IGF-I) and IGF-I receptor levels in the prostate of immature rats. Mol Cell Biol 88:77–87

    Bollig A, Miksicek RJ 2000 An estrogen receptor- splicing variant mediates both positive and negative effects on gene transcription. Mol Endocrinol 14:634–649

    McCarthy TL, Ji C, Shu H, Casinghino S, Crothers K, Rotwein P, Centrella M 1997 17?-Estradiol potently suppresses cAMP-induced insulin-like growth factor-I gene activation in primary rat osteoblast cultures. J Biol Chem 272:18132–18139

    Starling RD, Toth MJ, Matthews DE, Poehlman ET 1998 Energy requirements and physical activity of older free-living African-Americans: a doubly labeled water study. J Clin Endocrinol Metab 83:1529–1534

    Baker J, Liu JP, Robertson EJ, Efstratiadis A 1993 Role of insulin-like growth factors in embryonic and postnatal growth. Cell 75:73–82

    Baxter RC 2000 Insulin-like growth factor (IGF)-binding proteins: interactions with IGFs and intrinsic bioactivities. Am J Physiol 278:E967–E976

    Saggese G, Baroncelli GI, Bertelloni S, Barsanti S 1996 The effect of long-term growth hormone (GH) treatment on bone mineral density in children with GH deficiency. Role of GH in the attainment of peak bone mass. J Clin Endocrinol Metab 81:3077–3083

    Beckett PR, Copeland KC, Flannery TK, Sherman LD, Abrams SA 1999 Combination growth hormone and estrogen increase bone mineralization in girls with Turner syndrome. Pediatr Res 45:709–713

    Brixen K, Nielsen HK, Mosekilde L, Flyvbjerg A 1990 A short course of recombinant human growth hormone treatment stimulates osteoblasts and activates bone remodeling in normal human volunteers. J Bone Miner Res 5:609–618

    Holloway L, Butterfield G, Hintz R, Gesundheit N, Marcus R 1994 Effects of recombinant human growth hormone on metabolic indices, body composition, and bone turnover in healthy elderly women. J Clin Endocrinol Metab 79:470–479

    Aloia JF, Zanzi I, Ellis K 1976 Effects of growth hormone in osteoporosis. J Clin Endocrinol Metab 43:992–999

    Bikle DD, Halloran BP 1999 The response of bone to unloading. J Bone Miner Metab 17:233–244 [Review]

    Canalis E, Centrella M, Burch W, McCarthy TL 1989 Insulin-like growth factor I mediates selective anabolic effects of parathyroid hormone in bone cultures. J Clin Invest 83:60–65

    Schlechter NL, Russell SM, Spencer EM, Nicoll CS 1986 Evidence suggesting that the direct growth-promoting effect of growth hormone on cartilage in vivo is mediated by local production of somatomedin. Proc Natl Acad Sci USA 83:7932–7934

    Ohlsson C, Nilsson A, Isaksson OGP, Lindahl A 1992 Growth hormone induces multiplication of slowly cycling germinal cells of the rat tibial growth plate. Proc Natl Acad Sci USA 89:9826–9830

    Isaksson OGP, Jansson JO, Gause IA 1982 Growth hormone stimulates longitudinal bone growth directly. Science 216:1237–1239

    Slootweg MC, Ohlsson C, Salles JP, de Vries CP, Netelenbos JC 1995 Insulin-like growth factor binding proteins-2 and -3 stimulate growth hormone receptor binding and mitogenesis in rat osteosarcoma cells. Endocrinol 136:4210–4217

    Richman C, Baylink DJ, Lang K, Dony C, Mohan S 1999 Recombinant human insulin-like growth factor-binding protein-5 stimulates bone formation parameters in vitro and in vivo. Endocrinology 140:4699–4705

    Schmid C, Schlapfer I, Keller A, Waldvogel M, Froesch ER, Zapf J 1995 Effects of insulin-like growth factor (IGF) binding proteins (BPs)-3 and -6 on DNA synthesis of rat osteoblasts: further evidence for a role of auto-/paracrine IGF I but not IGF II in stimulating osteoblast growth. Biochem Biophys Res Commun 212:242–248

    Schneider MR, Lahm H, Wu M, Hoeflich A, Wolf E 2000 Transgenic mouse models for studying the functions of insulin-like growth factor-binding proteins. FASEB J 14:629–640 [Review]

    Langdahl BL, Kassem M, Moller MK, Eriksen EF 1998 The effects of IGF-I and IGF-II on proliferation and differentiation of human osteoblasts and interactions with growth hormone. Eur J Clin Invest 28:176–183

    Sims NA, Clement-Lacroix P, Da Ponte F, Bouali Y, Binart N, Moriggl R, Goffin V, Coschigano K, Gaillard-Kelly M, Kopchick J, Baron R, Kelly PA 2000 Bone homeostasis in growth hormone receptor-null mice is restored by IGF-I but independent of Stat5. J Clin Invest 106:1095–1103

    Kasperk CH, Wergedal JE, Farley JR, Linkhart TA, Turner RT, Baylink DJ 1989 Androgens directly stimulate proliferation of bone cells in vitro. Endocrinology 124:1576–1578

    Schwartz Z, Soskolne WA, Neubauer T, Goldstein M, Adi S, Ornoy A 1991 Direct and sex-specific enhancement of bone formation and calcification by sex steroids in fetal mice long bones in vitro (biochemical and morphometric study). Endocrinology 129:167–171

    Copeland KC, Johnson DM, Kuehl TJ, Castracane VD 1984 Estrogen stimulates growth hormone and somatomedin-C in castrate and intact female baboons. J Clin Endocrinol Metab 58:698–703

    Cuttler L, Van Vliet G, Conte FA, Kaplan SL, Grumbach MM 1985 Somatomedin-C levels in children and adolescents with gonadal dysgenesis: differences from age-matched normal females and effect of chronic estrogen replacement therapy. J Clin Endocrinol Metab 60:1087–1092

    Dawson-Hughes B, Stern D, Goldman J, Reichlin S 1986 Regulation of growth hormone and somatomedin-C secretion in postmenopausal women: effect of physiological estrogen replacement. J Clin Endocrinol Metab 63:424–432

    De Boer H, Blok GJ, Popp-Snijders C, Stuurman L, Baxter RC, van der Veen E 1996 Monitoring of growth hormone replacement therapy in adults, based on measurement of serum markers. J Clin Endocrinol Metab 81:1371–1377

    Frantz AG, Rabkin MT 1965 Effects of estrogen and sex difference on secretion of human growth hormone. J Clin Endocrinol Metab 25:1470–1480

    Blumsohn A, Hannon RA, Wrate R, Barton J, al Dehaimi AW, Colwell A, Eastell R 1994 Biochemical markers of bone turnover in girls during puberty. Clin Endocrinol (Oxf) 40:663–670

    Griffin JE 1992 Androgen resistance—the clinical and molecular spectrum. N Engl J Med 326:611–618 [Review]

    Slemenda CW, Reister TK, Hui SL, Miller JZ, Christian JC, Johnston Jr CC 1994 Influences on skeletal mineralization in children and adolescents: evidence for varying effects of sexual maturation and physical activity. J Pediatr 125:201–207

    Wastney ME, Ng J, Smith D, Martin BR, Peacock M, Weaver CM 1996 Differences in calcium kinetics between adolescent girls and young women. Am J Physiol 271:R208–R216

    Matkovic V, Jelic T, Wardlaw GM, Ilich JZ, Goel PK, Wright JK, Andon MB, Smith KT, Heaney RP 1994 Timing of peak bone mass in Caucasian females and its implication for the prevention of osteoporosis. Inference from a cross-sectional model. J Clin Invest 93:799–808

    Warner JT, Cowan FJ, Dunstan FD, Evans WD, Webb DK, Gregory JW 1998 Measured and predicted bone mineral content in healthy boys and girls aged 6–18 years: adjustment for body size and puberty. Acta Paediatr 87:244–249

    Teegarden D, Proulx WR, Martin BR, Zhao J, McCabe GP, Lyle RM, Peacock M, Slemenda C, Johnston CC, Weaver CM 1995 Peak bone mass in young women. J Bone Miner Res 10:711–715

    Louis O, Demeirleir K, Kalender W, Keizer HA, Platen P, Hollmann W, Osteaux M 1991 Low vertebral bone density values in young non-elite female runners. Int J Sports Med 12:214–217

    Bass S, Delmas PD, Pearce G, Hendrich E, Tabensky A, Seeman E 1999 The differing tempo of growth in bone size, mass, and density in girls is region-specific. J Clin Invest 104:795–804

    Mauras N, Attie KM, Reiter EO, Saenger P, Baptista J 2000 High dose recombinant human growth hormone (GH) treatment of GH-deficient patients in puberty increases near-final height: a randomized, multicenter trial. Genentech, Inc., Cooperative Study Group. J Clin Endocrinol Metab 85:3653–3660

    MacGillivray MH, Baptista J, Johanson A 1996 Outcome of a four-year randomized study of daily versus three times weekly somatropin treatment in prepubertal naive growth hormone-deficient children. Genentech Study Group. J Clin Endocrinol Metab 81:1806–1809

    Radetti G, Buzi F, Paganini C, Martelli C, Adami S 2000 A four year dose-response study of recombinant human growth hormone treatment of growth hormone deficient children: effects on growth, bone growth and bone mineralization. Eur J Endocrinol 142:42–46

    Clanget C, Seck T, Hinke V, Wuster C, Ziegler R, Pfeilschifter J 2001 Effects of 6 years of growth hormone (GH) treatment on bone mineral density in GH-deficient adults. Clin Endocrinol (Oxf) 55:93–99

    Cuneo RC, Saloman F, Wiles CM, Hesp R, Sonksen PH 1991 Growth hormone treatment in growth hormone-deficient adults. I. Effects on muscle mass and strength. J Appl Physiol 70:688–694

    Fong Y, Rosenbaum M, Tracey KJ, Raman G, Hesse DG, Matthews DE, Leibel RL, Gertner JM, Fischman DA, Lowry SF 1989 Recombinant growth hormone enhances muscle myosin heavy-chain mRNA accumulation and amino acid accrual in humans. Proc Natl Acad Sci USA 86:3371–3374

    Fryburg DA, Gelfand RA, Barrett EJ 1991 Growth hormone acutely stimulates forearm muscle protein synthesis in normal humans. Am J Physiol 260:E499–E504

    Jorgensen JO, Moller J, George K 1993 Marked effects of sustained low growth hormone (GH) levels on day-to-day fuel metabolism. Studies in GH-deficient patients and healthy untreated subjects. J Clin Endocrinol Metab 77:1589–1596

    Span JP, Pieters GF, Sweep FG, Hermus AR, Smals AG 2001 Gender differences in rhGH-induced changes in body composition in GH-deficient adults. J Clin Endocrinol Metab 86:4161–4165

    Burman P, Johansson AG, Siegbahn A, Vessby B, Karlsson FA 1997 Growth hormone (GH)-deficient men are more responsive to GH replacement therapy than women. J Clin Endocrinol Metab 82:550–555

    Stewart CE, Rotwein P 1996 Growth, differentiation, and survival: multiple physiological functions for insulin-like growth factors. Phys Rev 76:1005–1026

    Laron Z, Klinger B 1993 Body fat in Laron syndrome patients: effects of insulin-like growth factor I treatment. Horm Res 40:16–22

    Mandel S, Moreland E, Nichols V, Hanna C, Lafranchi S 1995 Changes in insulin-like growth factor-I (IGF-I), IGF-binding protein-3, growth hormone (GH)-binding protein, erythrocyte IGF-I receptors, and growth rate during GH treatment. J Clin Endocrinol Metab 80:190–194

    Marzullo P, Di Somma C, Pratt KL, Khosravi J, Diamandis A, Lombardi G, Colao A, Rosenfeld RG 2001 Usefulness of different biochemical markers of the insulin-like growth factor (IGF) family in diagnosing growth hormone excess and deficiency in adults. J Clin Endocrinol Metab 86:3001–3008

    Jorgensen JO, Blum WF, Moller N, Ranke MB, Christiansen JS 1990 Circadian patterns of serum insulin-like growth factor (ICF) II and IGF binding protein 3 in growth hormone-deficient patients and age- and sex-matched normal subjects. Acta Endocrinol (Copenh) 123:257–262

    Laursen T, Flyvbjerg A, Jorgensen JO, Baxter RC, Christiansen JS 2000 Stimulation of the 150-kilodalton insulin-like growth factor-binding protein-3 ternary complex by continuous and pulsatile patterns of growth hormone (GH) administration in GH-deficient patients. J Clin Endocrinol Metab 85:4310–4314

    Aimaretti G, Corneli G, Razzore P, Bellone S, Baffoni C, Bellone J, Camanni F, Ghigo E 1998 Usefulness of IGF-I assay for the diagnosis of GH deficiency in adults. J Endocrinol Invest 21:506–511

    Ghigo E, Aimaretti G, Maccario M, Fanciulli G, Arvat E, Minuto F, Giordano G, Delitala G, Camanni F 1999 Does-response study of GH effects on circulating IGF-I and IGFBP-3 levels in healthy young men and women. Am J Physiol 276:E1009–E1013

    Jorgensen JO, Moller N, Lauritzen T, Christiansen JS 1990 Pulsatile versus continuous intravenous administration of growth hormone (GH) in GH-deficient patients: effects on circulating insulin-like growth factor-I and metabolic indices. J Clin Endocrinol Metab 70:1616–1623

    Devi GR, Yang DH, Rosenfeld RG, Oh Y 2000 Differential effects of insulin-like growth factor (IGF)-binding protein-3 and its proteolytic fragments on ligand binding, cell surface association, and IGF-I receptor signaling. Endocrinology 141:4171–4179

    Eliakim A, Brasel JA, Mohan S, Barstow TJ, Berman N, Cooper DM 1996 Physical fitness, endurance training, and the growth hormone-insulin-like growth factor I system in adolescent females. J Clin Endocrinol Metab 81:3986–3992

    Welle S, Thornton C, Statt M, McHenry B 1996 Growth hormone increases muscle mass and strength but does not rejuvenate myofibrillar protein in healthy subjects over 60 years old. J Clin Endocrinol Metab 81:3239–3243

    Russell-Jones DL, Umpleby M 1996 Protein anabolic action of insulin, growth hormone and insulin-like growth factor I. Eur J Endocrinol 135:631–642

    Ogle GD, Rosenberg AR, Calligeros D, Kainer G 1994 Effects of growth hormone treatment for short stature on calcium homeostasis, bone mineralisation, and body composition. Horm Res 41:16–20

    Russell-Jones DL, Weissberger AJ, Bowes JM, Kelly JM, Thomason M, Umpleby AM, Jones RH, Sonksen PH 1993 The effects of growth hormone on protein metabolism in adult growth hormone-deficient patients. Clin Endocrinol (Oxf) 38:427–431

    Hussain MA, Schmitz O, Mengel A, Glatz Y, Christiansen JS, Zapf J, Froesch ER 1994 Comparison of the effects of growth hormone and insulin-like growth factor I on substrate oxidation and on insulin sensitivity in growth hormone-deficient humans. J Clin Invest 94:1126–1133

    Daugaard JR, Laustsen JL, Hansen BS, Richter EA 1998 Growth hormone induces muscle fibre type transformation in growth hormone-deficient rats. Acta Physiol Scand 164:119–126

    Mauras N 1995 Combined recombinant human growth hormone and recombinant human insulin-like growth factor I: lack of synergy on whole body protein anabolism in normally fed subjects. J Clin Endocrinol Metab 80:2633–2637

    Mauras N, O’Brien KO, Welch S, Rini A, Helgeson K, Vieira NE, Yergey AL 2000 Insulin-like growth factor I and growth hormone (GH) treatment in GH-deficient humans: differential effects on protein, glucose, lipid, and calcium metabolism. J Clin Endocrinol Metab 85:1686–1694

    Clemmons DR, Smith-Banks A, Underwood LE 1992 Reversal of diet-induced catabolism by infusion of recombinant insulin-like growth factor-I in humans. J Clin Endocrinol Metab 75:234–238

    Ueki I, Ooi GT, Tremblay ML, Hurst KR, Bach LA, Boisclair YR 2000 Inactivation of the acid labile subunit gene in mice results in mild retardation of postnatal growth despite profound disruptions in the circulating insulin-like growth factor system. Proc Natl Acad Sci USA 97:6868–6873

    Landin-Wilhelmsen K, Bryman I, Windh M, Wilhelmsen L 1999 Osteoporosis and fractures in Turner syndrome-importance of growth promoting and oestrogen therapy. Clin Endocrinol (Oxf) 51:497–502

    Gabrielsson BG, Carmignac DF, Flavell DM, Robinson IC 1995 Steroid regulation of growth hormone (GH) receptor and GH-binding protein messenger ribonucleic acids in the rat. Endocrinology 136:209–217

    Davenport ML, Punyasavatsut N, Gunther D, Savendahl L, Stewart PW 1999 Turner syndrome: a pattern of early growth failure. Acta Paediatr Suppl 88:118–121

    Lyon AJ, Preece MA, Grant DB 1985 Growth curve for girls with Turner syndrome. Arch Dis Child 60:932–935

    Russell-Aulet M, Shapiro B, Jaffe CA, Gross MD, Barkan AL 1998 Peak bone mass in young healthy men is correlated with the magnitude of endogenous growth hormone secretion. J Clin Endocrinol Metab 83:3463–3468

    Sato N, Nimura A, Horikawa R, Katumata N, Tanae A, Tanaka T 2000 Bone mineral density in Turner syndrome: relation to GH treatment and estrogen treatment. Endocr J 47(Suppl):115–119

    Rebuffe-Scrive M, Andersson B, Olbe L, Bjorntorp P 1989 Metabolism of adipose tissue in intraabdominal depots of non-obese men and women. Metabolism 38:453–458

    Cornelius P, MacDougald OA, Lane MD 1994 Regulation of adipocyte development. Annu Rev Nutr 14:99–129 [Review]

    Elbers JM, de Jong S, Teerlink T, Asscheman H, Seidell JC, Gooren LJ 1999 Changes in fat cell size and in vitro lipolytic activity of abdominal and gluteal adipocytes after a one-year cross-sex hormone administration in transsexuals. Metabolism 48:1371–1377

    Heine PA, Taylor JA, Iwamoto GA, Lubahn DB, Cooke PS 2000 Increased adipose tissue in male and female estrogen receptor- knockout mice. Proc Natl Acad Sci USA 97:12729–12734

    Muller AF, Janssen JA, Hofland LJ, Lamberts SW, Bidlingmaier M, Strassburger CJ, van der Lely AJ 2001 Blockade of the growth hormone (GH) receptor unmasks rapid GH-releasing peptide-6-mediated tissue-specific insulin resistance. J Clin Endocrinol Metab 86:590–593

    Wang C, Swedloff RS, Iranmanesh A, Dobs A, Snyder PJ, Cunningham G, Matsumoto AM, Weber T, Berman N 2000 Transdermal testosterone gel improves sexual function, mood, muscle strength, and body composition parameters in hypogonadal men. Testosterone Gel Study Group. J Clin Endocrinol Metab 85:2839–2853

    Van RL, Roncari DA 1978 Complete differentiation of adipocyte precursors. A culture system for studying the cellular nature of adipose tissue. Cell Tissue Res 195:317–329

    Dieudonne MN, Pecquery R, Leneveu MC, Giudicelli Y 2000 Opposite effects of androgens and estrogens on adipogenesis in rat preadipocytes: evidence for sex and site-related specificities and possible involvement of insulin-like growth factor 1 receptor and peroxisome proliferator-activated receptor 2. Endocrinology 141:649–656

    Pertzelan A, Blum I, Grunebaum M, Laron Z 1977 The combined effect of growth hormone and methandrostenolone on the linear growth of patients with multiple pituitary hormone deficiencies. Clin Endocrinol (Oxf) 6:271–276

    Xu X, De Pergola G, Eriksson PS, Fu L, Carlsson B, Yang S, Eden S, Bjorntorp P 1993 Postreceptor events involved in the up-regulation of ?-adrenergic receptor mediated lipolysis by testosterone in rat white adipocytes. Endocrinology 132:1651–1657

    Beauville M, Harant I, Crampes F, Riviere D, Tauber MT, Tauber JP, Garrigues M 1992 Effect of long-term rhGH administration in GH-deficient adults on fat cell epinephrine response. Am J Physiol Endocrinol Metab 263:E467–E472

    Ottosson M, Vikman-Adolfsson K, Enerback S, Olivecrona G, Bjorntorp P 1994 The effects of cortisol on the regulation of lipoprotein lipase activity in human adipose tissue. J Clin Endocrinol Metab 79:820–825

    Yang S, Xu X, Bjorntorp P, Eden S 1995 Additive effects of growth hormone and testosterone on lipolysis in adipocytes of hypophysectomized rats. J Endocrinol 147:147–152

    Machinal F, Dieudonne MN, Leneveu MC, Pecquery R, Giudicelli Y 1999 In vivo and in vitro ob gene expression and leptin secretion in rat adipocytes: evidence for a regional specific regulation by sex steroid hormones. Endocrinology 140:1567–1574

    Ohlsson C, Hellberg N, Parini P, Vidal O, Bohlooly M, Rudling M, Lindberg MK, Warner M, Angelin B, Gustafsson JA 2000 Obesity and disturbed lipoprotein profile in estrogen receptor--deficient male mice. Biochem Biophys Res Commun 278:640–645

    Eliakim A, Brasel JA, Barstow TJ, Mohan S, Cooper DM 1998 Peak oxygen uptake, muscle volume, and the growth hormone-insulin-like growth factor-I axis in adolescent males. Med Sci Sports Exerc 30:512–517

    O’Sullivan AJ, Crampton LJ, Freund J, Ho KK 1998 The route of estrogen replacement therapy confers divergent effects on substrate oxidation and body composition in postmenopausal women. J Clin Invest 102:1035–1040

    Roncari DA, Van RL 1978 Promotion of human adipocyte precursor replication by 17?-estradiol in culture. J Clin Invest 62:503–508

    Cleland WH, Mendelson CR, Simpson ER 1983 Aromatase activity of membrane fractions of human adipose tissue stromal cells and adipocytes. Endocrinology 113:2155–2160

    Corbould AM, Judd SJ, Rodgers RJ 1998 Expression of types 1, 2, and 3 17?-hydroxysteroid dehydrogenase in subcutaneous abdominal and intra-abdominal adipose tissue of women. J Clin Endocrinol Metab 83:187–194

    Boot AM, Muinck Keizer-Schrama S, Pols HA, Krenning EP, Drop SL 1998 Bone mineral density and body composition before and during treatment with gonadotropin-releasing hormone agonist in children with central precocious and early puberty. J Clin Endocrinol Metab 83:370–373

    Jones ME, Thorburn AW, Britt KL, Hewitt KN, Wreford NG, Proietto J, Oz OK, Leury BJ, Robertson KM, Yao S, Simpson ER 2000 Aromatase-deficient (ArKO) mice have a phenotype of increased adiposity. Proc Natl Acad Sci USA 97:12735–12740

    Karlsson C, Stenlof K, Johannsson G, Marin P, Bjorntorp P, Bengtsson BA, Carlsson B, Carlsson LM, Sjostrom L 1998 Effects of growth hormone treatment on the leptin system and on energy expenditure in abdominally obese men. Eur J Endocrinol 138:408–414

    Tagliaferri M, Scacchi M, Pincelli AI, Berselli ME, Silvestri P, Montesano A, Ortolani S, Dubini A, Cavagnini F 1998 Metabolic effects of biosynthetic growth hormone treatment in severely energy-restricted obese women. Int J Obes Relat Metab Disord 22:836–841

    Leung KC, Doyle N, Ballesteros M, Sjogren K, Watts CK, Low TH, Leong GM, Ross RJ, Ho KK 2003 Estrogen inhibits GH signaling by suppressing GH-induced JAK2 phosphorylation, an effect mediated by SOCS-2. Proc Natl Acad Sci USA 100:1016–1021

    Laursen T, Jorgensen JO, Christiansen JS 1994 Metabolic effects of growth hormone administered subcutaneously once or twice daily to growth hormone deficient adults. Clin Endocrinol (Oxf) 41:337–343

    Snyder DK, Clemmons DR, Underwood LE 1988 Treatment of obese, diet-restricted subjects with growth hormone for 11 weeks: effects on anabolism, lipolysis, and body composition. J Clin Endocrinol Metab 67:54–61

    Hayes VY, Urban RJ, Jiang J, Marcell TJ, Helgeson K, Mauras N 2001 Recombinant human growth hormone and recombinant human insulin-like growth factor I diminish the catabolic effects of hypogonadism in man: metabolic and molecular effects. J Clin Endocrinol Metab 86:2211–2219

    Gregory JW, Greene SA, Jung RT, Scrimgeour CM, Rennie MJ 1991 Changes in body composition and energy expenditure after six weeks’ growth hormone treatment. Arch Dis Child 66:598–602

    Leger J, Carel C, Legrand I, Paulsen A, Hassan M, Czernichow P 1994 Magnetic resonance imaging evaluation of adipose tissue and muscle tissue mass in children with growth hormone (GH) deficiency, Turner’s syndrome, and intrauterine growth retardation during the first year of treatment with GH. J Clin Endocrinol Metab 78:904–909

    Munzer T, Harman SM, Hees P, Shapiro E, Christmas C, Bellantoni MF, Stevens TE, O’Connor KG, Pabst KM, St Clair C, Sorkin JD, Blackman MR 2001 Effects of GH and/or sex steroid administration on abdominal subcutaneous and visceral fat in healthy aged women and men. J Clin Endocrinol Metab 86:3604–3610

    Vaisman N, Zadik Z, Duchan R, Voet H, Lotan D, Drukker A 1994 Changes in body composition of children with chronic renal failure during growth hormone treatment. Pediatr Nephrol 8:201–204

    Wit JM, van’t Hof MA, Van den Brande JL 1988 The effect of human growth hormone therapy on skinfold thickness in growth hormone-deficient children. Eur J Pediatr 147:588–592

    Chong PK, Jung RT, Scrimgeour CM, Rennie MJ, Paterson CR 1994 Energy expenditure and body composition in growth hormone deficient adults on exogenous growth hormone. Clin Endocrinol (Oxf) 40:103–110

    Salomon F, Cuneo RC, Hesp R, Sonksen PH 1989 The effects of treatment with recombinant human growth hormone on body composition and metabolism in adults with growth hormone deficiency. N Engl J Med 321:1797–1803

    Thompson JL, Butterfield GE, Marcus R, Hintz RL, Van Loan M, Ghiron L, Hoffman AR 1995 The effects of recombinant human insulin-like growth factor-I and growth hormone on body composition in elderly women. J Clin Endocrinol Metab 80:1845–1852

    Sjogren K, Wallenius K, Liu JL, Bohlooly Y, Pacini G, Svensson L, Tornell J, Isaksson OG, Ahren B, Jansson JO, Ohlsson C 2001 Liver-derived IGF-I is of importance for normal carbohydrate and lipid metabolism. Diabetes 50:1539–1545

    Di Cola G, Cool MH, Accili D 1997 Hypoglycemic effect of insulin-like growth factor-1 in mice lacking insulin receptors. J Clin Invest 99:2538–2544

    Porksen N, Hussain MA, Bianda TL, Nyholm B, Christiansen JS, Butler PC, Veldhuis JD, Foresch ER, Schmitz O 1997 IGF-I inhibits burst mass of pulsatile insulin secretion at supraphysiological and low IGF-I infusion rates. Am J Physiol 272:E352–E358

    Albertsson-Wikland K, Rosberg S, Karlberg J, Groth T 1994 Analysis of 24-hour growth hormone profiles in healthy boys and girls of normal stature: relation to puberty. J Clin Endocrinol Metab 78:1195–1201

    Martha Jr PM, Goorman KM, Blizzard RM, Rogol AD, Veldhuis JD 1992 Endogenous growth hormone secretion and clearance rates in normal boys as determined by deconvolution analysis: relationship to age, pubertal status and body mass. J Clin Endocrinol Metab 74:336–344

    Rose SR, Municchi G, Barners KM, Kamp GA, Uriarte MM, Ross JL, Cassoria F, Cutler Jr GB 1991 Spontaneous growth hormone secretion increases during puberty in normal girls and boys. J Clin Endocrinol Metab 73:428–435

    Faria ACS, Bekenstein LW, Booth Jr RA, Vaccaro VA, Asplin CM, Veldhuis JD, Thorner MO, Evans WS 1992 Pulsatile growth hormone release in normal women during the menstrual cycle. Clin Endocrinol (Oxf) 36:591–596

    Astrup A, Buemann B, Christensen NJ, Madsen J, Gluud C, Bennett P, Svenstrup B 1992 The contribution of body composition, substrates, and hormones to the variability in energy expenditure and substrate utilization in premenopausal women. J Clin Endocrinol Metab 74:279–286

    Iranmanesh A, South S, Liem AY, Clemmons D, Thorner MO, Weltman A, Veldhuis JD 1998 Unequal impact of age, percentage body fat, and serum testosterone concentrations on the somatotrophic, IGF-I, and IGF-binding protein responses to a three-day intravenous growth hormone-releasing hormone pulsatile infusion in men. Eur J Endocrinol 139:59–71

    Vahl N, Jorgensen JO, Skjaerback C, Veldhuis JD, Orskov H, Christiansen J 1997 Abdominal adiposity rather than age and sex predicts the mass and patterned regularity of growth hormone secretion in mid-life healthy adults. Am J Physiol 272:E1108–E1116

    Travers SH, Jeffers BW, Bloch CA, Hill JO, Eckel RH 1995 Gender and Tanner stage differences in body composition and insulin sensitivity in early pubertal children. J Clin Endocrinol Metab 80:172–178

    Iranmanesh A, Lizarralde G, Veldhuis JD 1991 Age and relative adiposity are specific negative determinants of the frequency and amplitude of growth hormone (GH) secretory bursts and the half-life of endogenous GH in healthy men. J Clin Endocrinol Metab 73:1081–1088

    Hartman ML, Faria AC, Vance ML, Johnson ML, Thorner MO, Veldhuis JD 1991 Temporal structure of in vivo growth hormone secretory events in man. Am J Physiol 260:E101–E110

    Barbarino A, De Marinis L, Troncone L 1978 Growth hormone response to propranolol and L-dopa in obese subjects. Metabolism 27:275–278

    Veldhuis JD 1996 Physiological regulation of the human growth hormone (GH)-insulin-like growth factor type I (IGF-I) axis: predominant impact of age, obesity, gonadal function, and sleep. Sleep 19:S221–S224

    Schaefer F, Baumann G, Faunt LM, Haffner D, Johnson ML, Mercado M, Ritz E, Mehls O, Veldhuis JD 1996 Multifactorial control of the elimination kinetics of unbound (free) GH in the human: regulation by age, adiposity, renal function, and steady-state concentrations of GH in plasma. J Clin Endocrinol Metab 81:22–31

    Kopelman PG, Noonan K, Goulton R, Forrest AJ 1985 Impaired growth hormone response to growth hormone releasing factor and insulin-hypoglycaemia in obesity. Clin Endocrinol (Oxf) 23:87–94

    Ukkola O, Ravussin E, Jacobson P, Snyder EE, Chagnon M, Sjostrom L, Bouchard C 2001 Mutations in the preproghrelin/ghrelin gene associated with obesity in humans. J Clin Endocrinol Metab 86:3996–3999

    Hartman ML, Clayton PE, Johnson ML, Celniker A, Perlman AJ, Alberti KG, Thorner MO 1993 A low dose euglycemic infusion of recombinant human insulin-like growth factor I rapidly suppresses fasting-enhanced pulsatile growth hormone secretion in humans. J Clin Invest 91:2453–2462

    Hartman ML, Veldhuis JD, Johnson ML, Lee MM, Alberti KGMM, Samojlik E, Thorner MO 1992 Augmented growth hormone (GH) secretory burst frequency and amplitude mediate enhanced GH secretion during a two-day fast in normal men. J Clin Endocrinol Metab 74:757–765

    Bergendahl M, Evans WS, Pastor CL, Patel A, Iranmanesh A, Veldhuis JD 1999 Short-term fasting suppresses leptin and (conversely) activates disorderly GH secretion in mid-luteal phase women. J Clin Endocrinol Metab 84:883–894

    Licinio J, Negra AB, Mantzoros C, Kaklamani V, Wong M-L, Bongiorno PB, Negro PP, Mulla A, Veldhuis JD, Cearnal L, Flier JS, Gold PW 1998 Sex differences in circulating human leptin pulse amplitude: clinical implications. J Clin Endocrinol Metab 83:4140–4147

    Pontiroli AE, Lanzi R, Monti LD, Sandoli E, Pozza G 1991 Growth hormone (GH) autofeedback on GH response to GH-releasing hormone. Role of free fatty acids and somatostatin. J Clin Endocrinol Metab 72:492–495

    Melmed S 1984 Insulin suppresses growth hormone secretion by rat pituitary cells. J Clin Invest 73:1425–1433

    Cordido F, Peino R, Penalva A, Alvarez CV, Casanueva FF, Dieguez C 1996 Impaired growth hormone secretion in obese subjects is partially reversed by acipimox-mediated plasma free fatty acid depression. J Clin Endocrinol Metab 81:914–918

    Bermann M, Jaffe CA, Tsai W, DeMott-Friberg R, Barkan AL 1994 Negative feedback regulation of pulsatile growth hormone secretion by insulin-like growth factor I: involvement of hypothalamic somatostatin. J Clin Invest 94:138–145

    Jaffe CA, Ocampo-Lim B, Guo W, Krueger K, Sugahara I, DeMott-Friberg R, Bermann M, Barkan AL 1998 Regulatory mechanisms of growth hormone secretion are sexually dimorphic. J Clin Invest 102:153–164

    Tschop M, Weyer C, Tataranni PA, Devanarayan V, Ravussin E, Heiman ML 2001 Circulating ghrelin levels are decreased in human obesity. Diabetes 50:707–709

    Kelijman M, Frohman LA 1988 Enhanced growth hormone (GH) responsiveness to GH releasing hormone after dietary manipulation in obese and nonobese subjects. J Clin Endocrinol Metab 66:489–494

    Frystyk J, Vestbo E, Skjaerbaek C, Mogensen CE, Orskov H 1995 Free insulin-like growth factors in human obesity. Metabolism 44(10 Suppl 4):37–44

    Bennett PA, Lindell K, Karlsson C, Robinson IC, Carlsson LM, Carlsson B 1998 Differential expression and regulation of leptin receptor isoforms in the rat brain: effects of fasting and oestrogen. Neuroendocrinology 67:29–36

    Cordido F, Penalva A, Dieguez C, Casanueva FF 1993 Massive growth hormone (GH) discharge in obese subjects after the combined administration of GH-releasing hormone and GHRP-6: evidence for a marked somatotroph secretory capability in obesity. J Clin Endocrinol Metab 76:819–823

    Procopio M, Maccario M, Grottoli S, Oleandri SE, Boffano GM, Camanni F, Ghigo E 1995 Short-term fasting in obesity fails to restore the blunted GH responsiveness to GH-releasing hormone alone or combined with arginine. Clin Endocrinol (Oxf) 43:665–669

    Csizmadi I, Brazeau P, Serri O 1989 Effect of dietary restriction and repeated growth hormone-releasing factor injections on growth hormone response to growth hormone-releasing factor in obese subjects. Metabolism 38:1016–1021

    Tanaka K, Inoue S, Numata K, Okazaki H, Nakamura S, Takamura Y 1990 Very-low-calorie diet-induced weight reduction reverses impaired growth hormone secretion response to growth hormone-releasing hormone, arginine, and L-dopa in obesity. Metabolism 39:892–896

    Williams T, Berelowitz M, Joffe SN, Thorner MO, Rivier J, Vale WW, Frohman LA 1984 Impaired growth hormone responses to growth hormone-releasing factor in obesity. A pituitary defect reversed with weight reduction. N Engl J Med 311:1403–1407

    Loche S, Pintor C, Cappa M, Ghigo E, Puggioni R, Locatelli V, Mueller EE 1989 Pyridostigmine counteracts the blunted growth hormone response to growth hormone-releasing hormone of obese children. Acta Endocrinol (Copenh) 120:624–628

    Frystyk J, Skjaerbaek C, Vestbo E, Fisker S, Orskov H 1999 Circulating levels of free insulin-like growth factors in obese subjects: the impact of type 2 diabetes. Diabetes Metab Res Rev 15:314–322

    Lee PD, Jensen MD, Divertie GD, Heiling VJ, Katz HH, Conover CA 1993 Insulin-like growth factor-binding protein-1 response to insulin during suppression of endogenous insulin secretion. Metabolism 42:409–414

    Travers SH, Labarta JI, Gargosky SE, Rosenfeld RG, Jeffers BW, Eckel RH 1998 Insulin-like growth factor binding protein-I levels are strongly associated with insulin sensitivity and obesity in early pubertal children. J Clin Endocrinol Metab 83:1935–1939

    Fliesen T, Maiter D, Gerard G, Underwood LE, Maes M, Ketelslegers JM 1989 Reduction of serum insulin-like growth factor-I by dietary protein restriction is age dependent. Pediatr Res 26:415–419

    Smith WJ, Underwood LE, Clemmons DR 1995 Effects of caloric or protein restriction on insulin-like growth factor-I (IGF-I) and IGF-binding proteins in children and adults. J Clin Endocrinol Metab 80:443–449

    Casanueva FF, Villanueva L, Dieguez C, Diaz Y, Cabranes JA, Szoke B, Scanlon MF, Schally AV, Fernandez-Cruz A 1987 Free fatty acids block growth hormone (GH) release hormone-stimulated GH secretion in man directly at the pituitary. J Clin Endocrinol Metab 65:634–642

    Chapman IM, Hartman ML, Pieper KS, Skiles EH, Pezzoli SS, Hintz RL, Thorner MO 1998 Recovery of growth hormone release from suppression by exogenous insulin-like growth factor I (IGF-I): evidence for a suppressive action of free rather than bound IGF-I. J Clin Endocrinol Metab 83:2836–2842

    Chapman IM, Hartman ML, Pezzoli SS, Harrell Jr FE, Hintz RL, Alberti KGMM, Thorner MO 1997 Effect of aging on the sensitivity of growth hormone secretion to insulin-like growth factor-I negative feedback. J Clin Endocrinol Metab 82:2996–3004

    Ghigo E, Gianotti L, Arvat E, Ramunni J, Valetto MR, Broglio F, Rolla M, Cavagnini F, Muller EE 1999 Effects of recombinant human insulin-like growth factor I administration on growth hormone (GH) secretion, both spontaneous and stimulated by GH-releasing hormone or hexarelin, a peptidyl GH secretagogue, in humans. J Clin Endocrinol Metab 84:285–290

    Gianotti L, Maccario M, Lanfranco F, Ramunni J, Di Vito L, Grottoli S, Mueller EE, Ghigo E, Arvat E 2000 Arginine counteracts the inhibitory effect of recombinant human insulin-like growth factor I on the somatotroph responsiveness to growth hormone-releasing hormone in humans. J Clin Endocrinol Metab 85:3604–3608

    Rasmussen MH, Hvidberg A, Juul A, Main K, Gotfredsen A, Skakkebae NE, Hilsted J 1995 Massive weight loss restores 24-hour growth hormone release profiles and serum insulin-like growth factor-I levels in obese subjects. J Clin Endocrinol Metab 80:1407–1415

    Furuhata Y, Kagaya R, Hirabayashi K, Ikeda A, Chang KT, Nishihara M, Takahashi M 2000 Development of obesity in transgenic rats with low circulating growth hormone levels: involvement of leptin resistance. Eur J Endocrinol 143:535–541

    Weyer C, Funahashi T, Tanaka S, Hotta K, Matsuzawa Y, Pratley RE, Tataranni PA 2001 Hypoadiponectinemia in obesity and type 2 diabetes: close association with insulin resistance and hyperinsulinemia. J Clin Endocrinol Metab 86:1930–1935

    Bowers CY 1993 Editorial: A new dimension on the induced release of growth hormone in obese subjects. J Clin Endocrinol Metab 76:817–818

    Taylor RW, Gold E, Manning P, Goulding A 1997 Gender differences in body fat content are present well before puberty. Int J Obes Relat Metab Disord 21:1082–1084

    Attia N, Tamborlane WV, Heptulla R, Maggs D, Grozman A, Sherwin RS, Caprio S 1998 The metabolic syndrome and insulin-like growth factor I regulation in adolescent obesity. J Clin Endocrinol Metab 83:1467–1471

    Vahl N, Jorgensen JO, Jurik AG, Christiansen JS 1996 Abdominal adiposity and physical fitness are major determinants of the age associated decline in stimulated GH secretion in healthy adults. J Clin Endocrinol Metab 81:2209–2215

    De Boer H, Blok GJ, Voerman HJ, Phillips M, Schouten JA 1994 Serum lipid levels in growth hormone-deficient men. Metabolism 43:199–203

    Murphy MG, Plunkett LM, Gertz BJ, He W, Wittreich J, Polvino WM, Clemmons DR 1998 MK-677, an orally active growth hormone secretagogue, reverses diet-induced catabolism. J Clin Endocrinol Metab 83:320–325

    Rosen T, Bosaeus I, Tolli J, Lindstedt G, Bengtsson BA 1993 Increased body fat mass and decreased extracellular fluid volume in adults with growth hormone deficiency. Clin Endocrinol (Oxf) 38:63–71

    Sklar CA, Ulstrom RA 1984 Effect of human growth hormone on adrenal androgens in children with growth hormone deficiency. Horm Res 20:166–171

    Svensson J, Lonn L, Jansson J-O, Murphy G, Wyss D, Krupa D, Cerchio K, Polvino W, Gertz B, Boseaus I, Sjostrom L, Bengtsson BA 1998 Two-month treatment of obese subjects with the oral growth hormone (GH) secretagogue MK-677 increases GH secretion, fat-free mass, and energy expenditure. J Clin Endocrinol Metab 83:362–369

    Svensson J, Jansson JO, Ottosson M, Johannsson G, Taskinen MR, Wiklund O, Bengtsson BA 1999 Treatment of obese subjects with the oral growth hormone secretagogue MK-677 affects serum concentrations of several lipoproteins, but not lipoprotein(a). J Clin Endocrinol Metab 84:2028–2033

    Fairhall KM, Gabrielsson BG, Robinson ICAF 1990 Effect of food withdrawal and insulin on growth hormone secretion in the guinea pig. Endocrinology 127:716–723

    Eigenmann JE, de Bruijne JJ, Froesch ER 1985 Insulin-like growth factor I and growth hormone in canine starvation. Acta Endocrinol (Copenh) 108:161–166

    Lemozy S, Pucilowska JB, Underwood LE 1994 Reduction of insulin-like growth factor I (IGF-I) in protein-restricted rats is associated with differential regulation of IGF-binding protein messenger ribonucleic acids in liver and kidney, and peptides in liver and serum. Endocrinology 136:617–632

    Pao CL, Farmer PK, Begovic S, Billafuerte BC, Wu G, Robertson DG, Phillips LS 1993 Regulation of insulin-like growth factor-I (IGF-I) and IGF-binding protein I gene transcription by hormones and provision of amino acids in rat hepatocytes. Mol Endocrinol 7:1561–1568

    Osborn BH, Fowlkes J, Han VKM, Freemark M 1992 Nutritional regulation of insulin-like growth factor-binding protein gene expression in the ovine fetus and pregnant ewe. Endocrinology 131:1743–1750

    Bruno JF, Olchovsky D, White JD, Leidy JW, Song J, Berelowitz M 1990 Influence of food deprivation in the rat on hypothalamic expression of growth hormone-releasing factor and somatostatin. Endocrinology 127:2111–2116

    Cunningham MJ, Clifton DK, Steiner RA 1999 Leptin’s actions on the reproductive axis: perspectives and mechanisms. Biol Reprod 60:216–222 [Review]

    Brier BH, Bass JJ, Butler JH, Gluckman PD 1986 The somatotropic axis in young steers: influence of nutritional status on pulsatile release of growth hormone and circulating concentrations of insulin-like growth factor I. J Endocrinol 111:209–215

    Crvyfan Hughes S, Cotterill AM, Molloy AR, Cassell TB, Brayde N, Hinds CJ, Wass JAH, Holly JMP 1992 The induction of specific proteases for insulin-like growth factor-binding proteins following major heart surgery. J Endocrinol 135:135–145

    Davenport ML, Svoboda ME, Koerber KL, Van Wyk JJ, Clemmons DR, Underwood LE 1988 Serum concentrations of insulin-like growth factor II are not changed by short term fasting and refeeding. J Clin Endocrinol Metab 67:1231–1236

    Mauras N, Hayes V, Welch S, Rini A, Helgeson K, Dokler M, Veldhuis JD, Urban RJ 1998 Testosterone deficiency in young men: marked alterations in whole body protein kinetics, strength and adiposity. J Clin Endocrinol Metab 83:1886–1892

    Ferrando AA, Sheffield-Moore M, Yeckel CW, Gilkison C, Jiang J, Achacosa A, Lieberman SA, Tipton K, Wolfe RR, Urban RJ 2002 Testosterone administration to older men improves muscle function: molecular and physiological mechanisms. Am J Physiol Endocrinol Metab 282:E601–E607

    Snyder PJ, Peachey H, Hannoush P, Berlin JA, Loh L, Lenrow DA, Holmes JH, Dlewati A, Santanna J, Rosen CJ, Strom BL 1999 Effect of testosterone treatment on body composition and muscle strength in men over 65 years of age. J Clin Endocrinol Metab 84:2647–2653

    Forbes GB, Porta CR, Herr BE, Griggs RC 1992 Sequence of changes in body composition induced by testosterone and reversal of changes after drug is stopped. JAMA 267:397–399

    Friedl KE, Dettori JR, Hannan CJ, Patience TH, Plymate SR 1991 Comparison of the effects of high dose testosterone and 19-nortestosterone to a replacement dose of testosterone on strength and body composition in normal men. J Steroid Biochem Mol Biol 40:607–612

    Bhasin S, Storer TW, Berman N, Yarasheski KE, Clevenger B, Phillips J, Lee WP, Bunnell TJ, Casaburi R 1997 Testosterone replacement increases fat-free mass and muscle size in hypogonadal men. J Clin Endocrinol Metab 82:407–413

    Bhasin S, Storer TW, Berman N, Callegari C, Clevenger B, Phillips J, Bunnell TJ, Tricker R, Shirazi A, Casaburi R 1996 The effects of supraphysiologic doses of testosterone on muscle size and strength in normal men. N Engl J Med 335:1–7

    Young NR, Baker HW, Liu G, Seeman E 1993 Body composition and muscle strength in healthy men receiving testosterone enanthate for contraception. J Clin Endocrinol Metab 77:1028–1032

    Arslanian SA, Kalhan SC 1996 Protein turnover during puberty in normal children. Am J Physiol 270:E79–E84

    Mauras N 1995 Estrogens do not affect whole-body protein metabolism in the prepubertal female. J Clin Endocrinol Metab 80:2842–2845

    Haarbo J, Marslew U, Gotfredsen A, Christiansen C 1991 Postmenopausal hormone replacement therapy prevents central distribution of body fat after menopause. Metabolism 40:1323–1326

    Evans WJ, Ivy JL 1982 Effects of testosterone propionate on hindlimb-immobilized rats. J Appl Physiol 52:1643–1647

    Wang J, Niu W, Nikiforov Y, Naito S, Chernausek S, Witte D, LeRoith D, Strauch A, Fagin JA 1997 Targeted overexpression of IGF-I evokes distinct patterns of organ remodeling in smooth muscle cell tissue beds of transgenic mice. J Clin Invest 100:1425–1439

    Adams GR, McCue SA 1998 Localized infusion of IGF-I results in skeletal muscle hypertrophy in rats. J Appl Physiol 84:1716–1722

    Willis PE, Chadan SG, Baracos V, Parkhouse WS 1998 Restoration of insulin-like growth factor I action in skeletal muscle of old mice. Am J Physiol 275:E525–E530

    Beguinot R, Kahn CR, Moses AC, Smith RJ 1985 Distinct biologically active receptors for insulin, insulin-like growth factor I and insulin-like growth factor II in cultured skeletal muscle cells. J Biol Chem 260:15892–15898

    Grobet L, Martin LJ, Poncelet D, Pirottin D, Brouwers B, Riquet J, Schoeberlein A, Dunner S, Menissier F, Massabanda J, Fries R, Hanset R, Georges M 1997 A deletion in the bovine myostatin gene causes the double-muscled phenotype in cattle. Nat Genet 17:71–74

    Carlson CJ, Booth FW, Gordon SE 1999 Skeletal muscle myostatin mRNA expression is fiber-type specific and increases during hindlimb unloading. Am J Physiol 277:R601—R606

    Brill KT, Weltman AL, Gentili A, Patrie JT, Fryburg DA, Hanks JB, Urban RJ, Veldhuis JD 2002 Single and combined effects of growth hormone and testosterone administration on measures of body composition, physical performance, mood, sexual function, bone turnover, and muscle gene expression in healthy older men. J Clin Endocrinol Metab 87:5649–5657

    Welle S, Thornton C 1997 Insulin-like growth factor-I, actin, and myosin heavy chain messenger RNAs in skeletal muscle after an injection of growth hormone in subjects over 60 years old. J Endocrinol 155:93–97

    Fryburg DA, Barrett EJ, Louard RJ, Gelfand RA 1990 Effect of starvation on human muscle protein metabolism and its response to insulin. Am J Physiol 259:E477–E482

    Tsalikian E, Howard C, Gerich JE, Haymond MW 1984 Increased leucine flux in short-term fasted human subjects: evidence for increased proteolysis. Am J Physiol 247:E323—E327

    Biolo G, Tipton KD, Klein S, Wolfe RR 1997 An abundant supply of amino acids enhances the metabolic effect of exercise on muscle protein. Am J Physiol 273:E122—E129

    Volpi E, Ferrando AA, Yeckel CW, Tipton KD, Wolfe RR 1998 Exogenous amino acids stimulate net muscle protein synthesis in the elderly. J Clin Invest 101:2000–2007

    Volpi E, Mittendorfer B, Wolf SE, Wolfe RR 1999 Oral amino acids stimulate muscle protein anabolism in the elderly despite higher first-pass splanchnic extraction. Am J Physiol 277:E513–E520

    Ferrando AA, Chinkes DL, Wolf SE, Matin S, Herndon DN, Wolfe RR 1999 A submaximal dose of insulin promotes net skeletal muscle protein synthesis in patients with severe burns. Ann Surg 229:11–18

    Griggs RC, Kingston W, Jozefowicz RF, Herr BE, Forbes G, Halliday D 1989 Effect of testosterone on muscle mass and muscle protein synthesis. J Appl Physiol 66:498–503

    Brodsky IG, Balagopla P, Nair KS 1996 Effects of testosterone replacement on muscle mass and muscle protein synthesis in hypogonadal men—a clinical research center study. J Clin Endocrinol Metab 81:3469–3475

    Mauras N, Beaufrere B 1995 Recombinant human insulin-like growth factor-I enhances whole body protein anabolism and significantly diminishes the protein catabolic effects of prednisone in humans without a diabetogenic effect. J Clin Endocrinol Metab 80:869–874

    Horber FF, Haymond MW 1990 Human growth hormone prevents the protein catabolic side effects of prednisone in humans. J Clin Invest 86:265–272

    Fryburg DA, Barrett EJ 1993 Growth hormone acutely stimulates skeletal muscle but not whole body protein synthesis in humans. Metabolism 42:1223–1227

    Yarasheski KE, Zachwieja JJ, Campbell JA, Bier DM 1995 Effect of growth hormone and resistance exercise on muscle growth and strength in older men. Am J Physiol 268:E268–E276

    Grinspoon SK, Baum HB, Peterson S, Klibanski A 1995 Effects of rhIGF-I administration on bone turnover during short-term fasting. J Clin Invest 96:900–906

    Touati G, Prieur AM, Ruiz JC, Noel M, Czernichow P 1998 Beneficial effects of one-year growth hormone administration to children with juvenile chronic arthritis on chronic steroid therapy. I. Effects on growth velocity and body composition. J Clin Endocrinol Metab 83:403–409

    Van den Berghe G, de Zegher F, Bouillon R 1998 Acute and prolonged critical illness as different neuroendocrine paradigms. J Clin Endocrinol Metab 83:1827–1834

    Sandstrom R, Svanberg E, Hyltander A, Haglind E, Ohlsson C, Zachrisson H, Berglund B, Lindholm E, Brevinge H, Lundholm K 1995 The effect of recombinant human IGF-I on protein metabolism in post-operative patients without nutrition compared to effects in experimental animals. Eur J Clin Invest 25:784–792

    Voerman HJ, Van Schijndel RJMS, Groeneveld ABJ, De Boer H, Nauta JP, Van der Veen EA, Thijs LG 1992 Effects of recombinant human growth hormone in patients with severe sepsis. Ann Surg 216:648–655

    Wolf RF, Pearlstone DB, Newman E, Heslin MJ, Gonenne A, Burt ME, Brennan MF 1992 Growth hormone and insulin reverse net whole body and skeletal muscle protein catabolism in cancer patients. Ann Surg 216:280–288

    Hart DW, Herndon DN, Klein G, Lee SB, Celis M, Mohan S, Chinkes DL, Wolf SE 2001 Attenuation of posttraumatic muscle catabolism and osteopenia by long-term growth hormone therapy. Ann Surg 233:827–834

    Rutherford OM, Jones DA, Round JM, Preece MA 1989 Changes in skeletal muscle after discontinuation of growth hormone treatment in young adults with hypopituitarism. Acta Paediatr Scand Suppl 356:61–63

    Boersma B, Wit JM 1997 Catch-up growth. Endocr Rev 18:646–661 [Review]

    Theintz G, Buchs B, Rizzoli R, Slosman D, Clavien H, Sizonenko PC, Bonjour JP 1992 Longitudinal monitoring of bone mass accumulation in healthy adolescents: evidence for a marked reduction after 16 years of age at the levels of lumbar spine and femoral neck in female subjects. J Clin Endocrinol Metab 75:1060–1065

    Ogle GD, Allen JR, Humphries IR, Lu PW, Briody JN, Morley K, Howman-Giles R, Cowell CT 1995 Body-composition assessment by dual-energy x-ray absorptiometry in subjects aged 4–26 y. Am J Clin Nutr 61:746–753

    Recker RR, Davies KM, Hinders SM, Heaney RP, Stegman MR, Kimmel DB 1992 Bone gain in young adult women. JAMA 268:2403–2408

    Schoenau E, Neu CM, Mokov E, Wassmer G, Manz F 2000 Influence of puberty on muscle area and cortical bone area of the forearm in boys and girls. J Clin Endocrinol Metab 85:1095–1098

    Bandini LG, Schoeller DA, Dietz WH 1990 Energy expenditure in obese and nonobese adolescents. Pediatr Res 27:198–203

    Schutz Y, Jequier E 1997 Resting energy expenditure, thermic effect of food, and total energy expenditure. In: Bray GA, Bouchard C, James WP, eds. Handbook of obesity. New York: Marcel Dekker; 443–456

    Ravussin E, Lillioja S, Anderson TE, Christin L, Bogardus C 1986 Determinants of 24-hour energy expenditure in man. Methods and results using a respiratory chamber. J Clin Invest 78:1568–1578

    Schulz LO, Alger S, Harper I, Wilmore JH, Ravussin E 1992 Energy expenditure of elite female runners measured by respiratory chamber and doubly labeled water. J Appl Physiol 72:23–28

    Nichold JF, Leier SE, Verity LS, Adams PL 1990 Effect of age and aerobic capacity on resting metabolic rate and the thermic effect of food. Nutr Res 10:1161–1170

    Poehlman ET, Denino WF, Beckett T, Kinaman KA, Dionne IJ, Dvorak R, Ades PA 2002 Effects of endurance and resistance training on total daily energy expenditure in young women: a controlled randomized trial. J Clin Endocrinol Metab 87:1004–1009

    Toth MJ, Poehlman ET 1996 Effects of exercise on daily energy expenditure. Nutr Rev 54:S140—S148 [Review]

    Smith DA, Dollman J, Withers RT, Brinkman M, Keeves JP, Clark DG 1997 Relationship between maximum aerobic power and resting metabolic rate in young adult women. J Appl Physiol 82:156–163

    Klausen B, Toubro S, Astrup A 1997 Age and sex effects on energy expenditure. Am J Clin Nutr 65:895–907

    Tuominen JA, Ebeling P, Heiman ML, Stephens T, Koivisto VA 1997 Leptin and thermogenesis in humans. Acta Physiol Scand 160:83–87

    Sell H, Berger JP, Samson P, Castriota G, Lalonde J, Deshaies Y, Richard D 2004 Peroxisome proliferator-activated receptor agonism increases the capacity for sympathetically-mediated thermogenesis in lean and ob/ob mice. Endocrinology 145:3925–3934

    Weltman A, Pritzlaff CJ, Wideman L, Considine RV, Fryburg DA, Gutgesell ME, Hartman ML, Veldhuis JD 2000 Intensity of acute exercise does not affect serum leptin concentrations in young men. Med Sci Sports Exerc 32:1556–1561

    Weltman A, Pritzlaff CJ, Wideman L, Weltman JY, Blumer JL, Abbott RD, Hartman ML, Veldhuis JD 2000 Exercise-dependent growth hormone release is linked to markers of heightened central adrenergic outflow. Am J Physiol 89:629–635

    Benardot D, Czerwinski C 1991 Selected body composition and growth measures of junior elite gymnasts. J Am Diet Assoc 91:29–33

    Fornetti WC, Pivarnik JM, Foley JM, Fiechtner JJ 1999 Reliability and validity of body composition measures in female athletes. J Appl Physiol 87:1114–1122

    Georgopoulos NA, Markou KB, Theodoropoulou A, Vagenakis GA, Benardot D, Leglise M, Dimopoulos JC, Vagenakis AG 2001 Height velocity and skeletal maturation in elite female rhythmic gymnasts. J Clin Endocrinol Metab 86:5159–5164

    Van Etten LM, Westerterp KR, Verstappen FT, Boon BJ, Saris WH 1997 Effect of an 18-wk weight-training program on energy expenditure and physical activity. J Appl Physiol 82:298–304

    Meijer GA, Janssen GM, Westerterp KR, Verhoeven F, Saris WH, ten Hoor F 1991 The effect of a 5-month endurance-training program on physical activity: evidence for a sex-difference in the metabolic response to exercise. Eur J Appl Physiol 62:11–17

    Lawson S, Webster JD, Pacy PJ, Garrow JS 1987 Effect of a 10-week aerobic exercise programme on metabolic rate, body composition and fitness in lean sedentary females. Br J Clin Pract 41:684–688

    Poehlman ET, Dvorak RV, Denino WF, Brochu M, Ades PA 2000 Effects of resistance training and endurance training on insulin sensitivity in nonobese, young women: a controlled randomized trial. J Clin Endocrinol Metab 85:2463–2468

    Spadano JL, Bandini LG, Must A, Dietz WH 2001 Changes in resting metabolic rate relative to menarche. Obes Res 9:73S

    DeLany JP, Bray GA, Harsha DW, Volaufova J, Champagne C 2001 Energy expenditure in African-American and Caucasian boys and girls in the 2-year follow-up of the Baton Rouge Children’s (BAROC) study. Obes Res 3:74S

    Jankowska EA, Rogucka E, Medra SM, Welon Z 2000 Relationships between age-related changes of sex steroids, obesity and body fat distribution among healthy Polish males. Med Sci Monit 6:1159–1164

    Nestler JE, Barlascini CO, Clore JN, Blackard WG 1988 Dehydroepiandrosterone reduces serum low density lipoprotein levels and body fat but does not alter insulin sensitivity in normal men. J Clin Endocrinol Metab 66:57–61

    Solomon SJ, Kurzer MS, Calloway DH 1982 Menstrual cycle and basal metabolic rate in women. Am J Clin Nutr 36:611–616

    Webb P 1986 24-Hour energy expenditure and the menstrual cycle. Am J Clin Nutr 44:614–619

    Bisdee JT, James WP, Shaw MA 1989 Changes in energy expenditure during the menstrual cycle. Br J Nutr 61:187–199

    Howe JC, Rumpler WV, Seale JL 1993 Energy expenditure by indirect calorimetry in premenopausal women: variation within one menstrual cycle. J Nutr Biochem 4:268

    Meijer GA, Westerterp KR, Saris WH, ten Hoor F 1992 Sleeping metabolic rate in relation to body composition and the menstrual cycle. Am J Clin Nutr 55:637–640

    Pelkman CL, Chow M, Heinbach RA, Rolls BJ 2001 Short-term effects of a progestational contraceptive drug on food intake, resting energy expenditure, and body weight in young women. Am J Clin Nutr 73:19–26

    Poehlman ET, Toth MJ, Gardner AW 1995 Changes in energy balance and body composition at menopause: a controlled longitudinal study. Ann Intern Med 123:673–675

    O’Sullivan AJ, Martin A, Brown MA 2001 Efficient fat storage in premenopausal women and in early pregnancy: a role for estrogen. J Clin Endocrinol Metab 86:4951–4956

    Hoffman DM, O’Sullivan AJ, Freund J, Ho KKY 1995 Adults with growth hormone deficiency have abnormal body composition but normal energy metabolism. J Clin Endocrinol Metab 80:72–77

    Jorgensen JO, Moller J, Laursen T, Orskov H, Christiansen JS, Weeke J 1994 Growth hormone administration stimulates energy expenditure and extrathyroidal conversion of thyroxine to triiodothyronine in a dose-dependent manner and suppresses circadian thyrotrophin levels: studies in GH-deficient adults. Clin Endocrinol (Oxf) 41:609–614

    Vaisman N, Zadik Z, Akivias A, Voet H, Katz I, Yair S, Ashkenazi A 1994 Changes in body composition, resting energy expenditure, and thermic effect of food in short children on growth hormone therapy. Metabolism 43:1543–1548

    Pedersen SB, Kristensen K, Fisker S, Jorgensen JO, Christiansen JS, Richelsen B 1999 Regulation of uncoupling protein-2 and -3 by growth hormone in skeletal muscle and adipose tissue in growth hormone-deficient adults. J Clin Endocrinol Metab 84:4073–4078

    Shimon I, Yan X, Magoffin DA, Friedman TC, Melmed S 1998 Intact leptin receptor is selectively expressed in human fetal pituitary and pituitary adenomas and signals human fetal pituitary growth hormone secretion. J Clin Endocrinol Metab 83:4059–4064

    Jorgensen JO, Pedersen SB, Borglum J, Moller N, Schmitz O, Christiansen JS, Richelsen B 1994 Fuel metabolism, energy expenditure, and thyroid function in growth hormone-treated obese women: a double-blind placebo-controlled study. Metabolism 43:872–877

    Wolthers T, Groftne T, Moller N, Christiansen JS, Orskov H, Weeke J, Jorgensen JO 1996 Calorigenic effects of growth hormone: the role of thyroid hormones. J Clin Endocrinol Metab 81:1416–1419

    Goran MI, Kaskoun M, Shuman WP 1995 Intra-abdominal adipose tissue in young children. Int J Obes Relat Metab Disord 19:279–283

    Goran MI, Nagy TR, Treuth MS, Trowbridge C, Dezenberg C, McGloin A, Gower BA 1997 Visceral fat in white and African American prepubertal children. Am J Clin Nutr 65:1703–1708

    Goran MI, Bergman RN, Gower BA 2001 Influence of total versus visceral fat on insulin action and secretion in African American and white children. Obes Res 9:423–431

    Brambilla P, Manzoni P, Sironi S, Simone P, Del Maschio A, di Natale B, Chiumello G 1994 Peripheral and abdominal adiposity in childhood obesity. Int J Obes Relat Metab Disord 18:795–800

    Owens S, Gutin B, Ferguson M, Allison J, Karp W, Le NA 1998 Visceral adipose tissue and cardiovascular risk factors in obese children. J Pediatr 133:41–45

    Roemmich JN, Clark PA, Berr SS, Mai V, Mantzoros CS, Flier JS, Weltman A, Rogol AD 1998 Gender differences in leptin levels during puberty are related to the subcutaneous fat depot and sex steroids. Am J Physiol 275:E543–E551

    de Ridder CM, de Boer RW, Seidell JC, Nieuwenhoff CM, Jeneson JA, Bakker CJ, Zonderland ML, Erich WB 1992 Body fat distribution in pubertal girls quantified by magnetic resonance imaging. Int J Obes Relat Metab Disord 16:443–449

    Alexander MK 1964 The postmortem estimation of total body fat, muscle and bone. Clin Sci (Lond) 26:193–202

    Skerlj B, Brozek J, Hunt EE 1953 Subcutaneous fat and age changes in body build and body form in women. Am J Phys Anthropol 11:577–600

    Gregory JW, Greene SA, Jung RT, Scrimgeour CM, Rennie MJ 1993 Metabolic effects of growth hormone treatment: an early predictor of growth response? Arch Dis Child 68:205–209

    Tanner JM, Whitehouse RH 1967 The effect of human growth hormone on subcutaneous fat thickness in hyposomatotrophic and panhypopituitary dwarfs. J Endocrinol 39:263–275

    Vaisman N, Zadik Z, Shamai Y, Franklin L, Dukhan R 1992 Changes in body composition of patients with subnormal spontaneous secretion of growth hormone, during the first year of treatment with growth hormone. Metabolism 41:483–486

    Falahati-Nini A, Riggs BL, Atkinson EJ, O’Fallon WM, Eastell R, Khosla S 2000 Relative contributions of testosterone and estrogen in regulating bone resorption and formation in normal elderly men. J Clin Invest 106:1553–1560

    Larsson L, Grimby G, Karlsson J 1979 Muscle strength and speed of movement in relation to age and muscle morphology. J Appl Physiol 46:451–456

    Rochira V, Faustini-Fustini M, Balestrieri A, Carani C 2000 Estrogen replacement therapy in a man with congenital aromatase deficiency: effects of different doses of transdermal estradiol on bone mineral density and hormonal parameters. J Clin Endocrinol Metab 85:1841–1845

    Szulc P, Munoz F, Claustrat B, Garnero P, Marchand F, Duboeuf F, Delmas PD 2001 Bioavailable estradiol may be an important determinant of osteoporosis in men: the MINOS study. J Clin Endocrinol Metab 86:192–199

    Forbes GB 1962 Methods for determining composition of the human body. Pediatrics 29:477–494(Johannes D. Veldhuis, Jam)