1
|
Shalitin S, Gat-Yablonski G. Associations of Obesity with Linear Growth and Puberty. Horm Res Paediatr 2022; 95:120-136. [PMID: 34130293 DOI: 10.1159/000516171] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 03/27/2021] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND The prevalence of obesity in childhood has increased dramatically in recent decades with increased risk of developing cardiometabolic and other comorbidities. Childhood adiposity may also influence processes of growth and puberty. SUMMARY Growth patterns of obesity during childhood have been shown to be associated with increased linear growth in early childhood, leading to accelerated epiphyseal growth plate (EGP) maturation. Several hormones secreted by the adipose tissue may affect linear growth in the context of obesity, both via the growth hormone IGF-1 axis and via a direct effect on the EGP. The observation that children with obesity tend to mature earlier than lean children has led to the assumption that the degree of body fatness may trigger the neuroendocrine events that lead to pubertal onset. The most probable link between obesity and puberty is leptin and its interaction with the kisspeptin system, which is an important regulator of puberty. However, peripheral action of adipose tissue could also be involved in changes in the onset of puberty. In addition, nutritional factors, epigenetics, and endocrine-disrupting chemicals are potential mediators linking pubertal onset to obesity. In this review, we focused on interactions of obesity with linear growth and pubertal processes, based on basic research and clinical data in humans. KEY MESSAGE Children with obesity are subject to accelerated linear growth with risk of impaired adult height and early puberty, with its psychological consequences. The data highlight another important objective in combatting childhood obesity, for the prevention of abnormal growth and pubertal patterns.
Collapse
Affiliation(s)
- Shlomit Shalitin
- National Center for Childhood Diabetes, Schneider Children's Medical Center of Israel, The Jesse Z and Sara Lea Shafer Institute for Endocrinology and Diabetes, Petach Tikva, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Galia Gat-Yablonski
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Felsenstein Medical Research Center, Petach Tikva, Israel
| |
Collapse
|
2
|
The Effects of Nutrition on Linear Growth. Nutrients 2022; 14:nu14091752. [PMID: 35565716 PMCID: PMC9100533 DOI: 10.3390/nu14091752] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 04/10/2022] [Accepted: 04/17/2022] [Indexed: 12/16/2022] Open
Abstract
Linear growth is a complex process and is considered one of the best indicators of children’s well-being and health. Genetics, epigenetics and environment (mainly stress and availability of nutrients) are the main regulators of growth. Nutrition exerts its effects on growth throughout the course of life with different, not completely understood mechanisms. Cells have a sophisticated sensing system, which allows growth processes to occur in the presence of an adequate nutrient availability. Most of the nutritional influence on growth is mediated by hormonal signals, in turn sensitive to nutritional cues. Both macro- and micro-nutrients are required for normal growth, as demonstrated by the impairment of growth occurring when their intake is insufficient. Clinical conditions characterized by abnormal nutritional status, including obesity and eating disorders, are associated with alterations of growth pattern, confirming the tight link between growth and nutrition. The precise molecular mechanisms connecting nutrition to linear growth are far from being fully understood and further studies are required. A better understanding of the interplay between nutrients and the endocrine system will allow one to develop more appropriate and effective nutritional interventions for optimizing child growth.
Collapse
|
3
|
Serum IGF1 and linear growth in children with congenital leptin deficiency before and after leptin substitution. Int J Obes (Lond) 2021; 45:1448-1456. [PMID: 34002033 PMCID: PMC8236407 DOI: 10.1038/s41366-021-00809-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 02/19/2021] [Accepted: 03/22/2021] [Indexed: 02/04/2023]
Abstract
BACKGROUND Evidence from in vitro and rodent studies suggests that leptin, a key signal of long-term energy reserves, promotes IGF1 synthesis and linear growth. This effect of leptin has not been fully investigated in humans. The aim of our study was to investigate the effect of leptin substitution on growth factors and linear growth in children with congenital leptin deficiency (CLD). METHODS In this cohort study we included eight pediatric patients (six males), age 0.9-14.8 years, who were diagnosed with CLD and received leptin substitution at our University Medical Center. We calculated standard deviation scores (SDS) for serum levels of IGF1 and IGFBP3, IGF1/IGFBP3 molar ratio, and height at baseline (T0) and 12 months (T12) after the initiation of substitution with metreleptin. RESULTS All patients had severe obesity (BMI-SDS mean ± SD: 4.14 ± 1.51) at T0 and significant BMI-SDS reduction to 2.47 ± 1.05 at T12. At T0, all patients were taller than the mid-parental median, yet had low IGF1 and IGF1/IGFBP3 molar ratios (IGF1-SDS[Formula: see text]T0: -1.58 ± 0.92, IGF1/IGFBP3 molar ratio-SDS[Formula: see text]T0: -1.58 ± 0.88). At T12, IGF1-SDS increased significantly (∆T0-12: 1.63 ± 1.40, p = 0.01), and IGFBP3-SDS and IGF1/IGFBP3 molar ratio-SDS showed a trend toward an increase. In the three children within the childhood growth period (post-infancy, pre-puberty) height-SDS increased (∆height-SDST0-12: 0.57 ± 0.06, p = 0.003) despite substantial weight loss. CONCLUSIONS These results in CLD patients are contrary to observations in children with idiopathic obesity who typically have above-mean IGF1 levels that decrease with weight loss, and therefore suggest that leptin increases IGF1 levels and promotes linear growth.
Collapse
|
4
|
Abstract
The well-balanced nourishment during "the first 1000 days," the period between conception (day 18) and the age of two years, is quite important for two main reasons. Firstly, the nutritive requirement is high due to the rapid physiological growth and functional development. Then, this period is characterized by extreme susceptibility to external stimuli such as inadequate maternal and infant nutritional status which they can interfere with the different stages of the development process leading to short and long-term consequences for health. Linear growth and brain development are particularly impaired from not sufficient nutrition. In consideration of the irreversible damage of malnutrition, especially on developing brain, an adequate nutrition during the first 1000 days of life is paramount. The aim of this review was to overview the latest scientific evidences on the relationship between nutrition and growth, focusing on nutritional requirements during the first 1000 days, and the impact of inadequate nutrition on brain development and linear growth.
Collapse
Affiliation(s)
- Lorena Matonti
- Department of Pediatrics, University of Chieti, Chieti, Italy
| | | | | |
Collapse
|
5
|
Hossain M, Nahar B, Haque MA, Mondal D, Mahfuz M, Naila NN, Gazi MA, Hasan MM, Haque NMS, Haque R, Arndt MB, Walson JL, Ahmed T. Serum Adipokines, Growth Factors, and Cytokines Are Independently Associated with Stunting in Bangladeshi Children. Nutrients 2019; 11:nu11081827. [PMID: 31394828 PMCID: PMC6723106 DOI: 10.3390/nu11081827] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Revised: 08/06/2019] [Accepted: 08/06/2019] [Indexed: 02/07/2023] Open
Abstract
Growth in young children is controlled through the release of several hormonal signals, which are affected by diet, infection, and other exposures. Stunting is clearly a growth disorder, yet limited evidence exists documenting the association of different growth biomarkers with child stunting. This study explored the association between different growth biomarkers and stunting in Bangladeshi children. A quasi-experimental study was conducted among 50 stunted (length-for-age Z-score (LAZ) < -2 SD) and 50 control (LAZ ≥ -2 SD) children, aged 12-18 months, residing in a Bangladeshi slum. The enrolled stunted children received an intervention package, which included food supplementation for three months, psychosocial stimulation for six months, and routine clinical care on community nutrition center at the study field site. The controls received routine clinical care only. All children were clinically screened over the study period. Length, weight, fasting blood and fecal biomarkers were measured. All biomarkers levels were similar in both groups except for oxyntomodulin at enrolment. Leptin (adjusted odds ratio, AOR: 4.0, p < 0.01), leptin-adiponectin ratio (AOR 5.07 × 108, p < 0.01), insulin-like growth factor-1 (IGF-1) (AOR 1.02, p < 0.05), and gamma interferon (IFN-γ) (AOR 0.92, p < 0.05) levels were independently associated with stunting at enrolment. Serum leptin, leptin-adiponectin ratio, interleukin-6 (IL-6), IL-10, tumor necrosis factor-alpha (TNF-α), and fecal alpha-1-antitrypsin (AAT) levels increased significantly (p < 0.001), while IFN-γ levels significantly decreased among stunted children after six months of intervention. Leptin, leptin-adiponectin ratio, IGF-1, and IFN-γ are independently associated with stunting in Bangladeshi children. This trial was registered at clinicaltrials.gov as NCT02839148.
Collapse
Affiliation(s)
- Muttaquina Hossain
- Nutrition and Clinical Services Division, icddr,b, Dhaka 1212, Bangladesh.
| | - Baitun Nahar
- Nutrition and Clinical Services Division, icddr,b, Dhaka 1212, Bangladesh
| | - Md Ahshanul Haque
- Nutrition and Clinical Services Division, icddr,b, Dhaka 1212, Bangladesh
| | - Dinesh Mondal
- Nutrition and Clinical Services Division, icddr,b, Dhaka 1212, Bangladesh
| | - Mustafa Mahfuz
- Nutrition and Clinical Services Division, icddr,b, Dhaka 1212, Bangladesh
| | - Nurun Nahar Naila
- Nutrition and Clinical Services Division, icddr,b, Dhaka 1212, Bangladesh
| | - Md Amran Gazi
- Nutrition and Clinical Services Division, icddr,b, Dhaka 1212, Bangladesh
| | - Md Mehedi Hasan
- Nutrition and Clinical Services Division, icddr,b, Dhaka 1212, Bangladesh
| | | | - Rashidul Haque
- Enteric and Respiratory Infections, icddr,b, Dhaka 1212, Bangladesh
| | - Michael B Arndt
- PATH, Seattle, WA 98109, USA
- Department of Global Health, University of Washington, Seattle, WA 98109, USA
| | - Judd L Walson
- Department of Global Health, University of Washington, Seattle, WA 98109, USA
- Department of Pediatrics, University of Washington, Seattle, WA 98109, USA
- Childhood Acute Illness and Nutrition Network, Nairobi 00200, Kenya
- Departments of Medicine, University of Washington, Seattle, WA 98109, USA
- Department of Epidemiology, University of Washington, Seattle, WA 98109, USA
| | - Tahmeed Ahmed
- Nutrition and Clinical Services Division, icddr,b, Dhaka 1212, Bangladesh
- Department of Global Health, University of Washington, Seattle, WA 98109, USA
- James P. Grant School of Public Health, BRAC University, Dhaka 1212, Bangladesh
| |
Collapse
|
6
|
Affiliation(s)
- André Briend
- Center for Child Health Research, Faculty of Medicine and Medical Technology, Tampere University, Tampere, Finland; and Department of Nutrition, Exercise and Sports, University of Copenhagen, Fredericksberg, Denmark
| |
Collapse
|
7
|
Isanaka S, Hitchings MDT, Berthé F, Briend A, Grais RF. Linear growth faltering and the role of weight attainment: Prospective analysis of young children recovering from severe wasting in Niger. MATERNAL AND CHILD NUTRITION 2019; 15:e12817. [PMID: 30903806 PMCID: PMC6849732 DOI: 10.1111/mcn.12817] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 01/11/2019] [Accepted: 03/15/2019] [Indexed: 12/17/2022]
Abstract
Efforts to reduce the impact of stunting have been largely independent of interventions to reduce the impact of wasting, despite the observation that the conditions can coexist in the same child and increase risk of death. To optimize the management of malnourished children—who can be wasted, stunted, or both—the relationship between stunting and wasting should be elaborated. We aimed to describe the relationship between concurrent weight and height gain during and after rehabilitation from severe wasting. We conducted a secondary analysis of a randomized trial for the outpatient treatment of severe wasting, including 1,542 children who recovered and were followed for 12 weeks. We described the overlap of stunting and severe wasting and the change in stunting over time. We showed the relationship between concurrent weight and height gain using adjusted generalized estimating equations and calculated the mean rate of change in weight‐for‐height z score (WHZ) and height‐for‐age z score (HAZ) during and after rehabilitation. At baseline, 79% (n = 1,223/1,542) and 49% (n = 757/1,542) of children were stunted and severely stunted, respectively. Prevalence increased over time among children <24 months. During rehabilitation when weight was not yet fully recovered, we found rapid WHZ gain but limited HAZ gain. Following successful rehabilitation, WHZ gain slowed. The rate of HAZ gain was negative after rehabilitation but increased relative to the period during treatment. The potential relationship between weight and height gain calls for increased coverage of wasting treatment to not only prevent child mortality but also reduce linear growth faltering.
Collapse
Affiliation(s)
- Sheila Isanaka
- Departments of Nutrition and Global Health and Population, Harvard T.H. Chan School of Public Health, Boston, United States.,Department of Research, Epicentre, Paris, France
| | - Matt D T Hitchings
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, United States
| | | | - André Briend
- Center for Child Health Research, University of Tampere School of Medicine, Tampere, Finland.,Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | | |
Collapse
|
8
|
Reid IR, Baldock PA, Cornish J. Effects of Leptin on the Skeleton. Endocr Rev 2018; 39:938-959. [PMID: 30184053 DOI: 10.1210/er.2017-00226] [Citation(s) in RCA: 109] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 06/26/2018] [Indexed: 12/12/2022]
Abstract
Leptin originates in adipocytes, including those in bone marrow, and circulates in concentrations 20 to 90 times higher than those in the cerebrospinal fluid. It has direct anabolic effects on osteoblasts and chondrocytes, but it also influences bone indirectly, via the hypothalamus and sympathetic nervous system, via changes in body weight, and via effects on the production of other hormones (e.g., pituitary). Leptin's role in bone physiology is determined by the balance of these conflicting effects. Reflecting this inconsistency, the leptin-deficient mouse has reduced length and bone mineral content of long bones but increased vertebral trabecular bone. A consistent bone phenotype in human leptin deficiency has not been established. Systemic leptin administration in animals and humans usually exerts a positive effect on bone mass, and leptin administration into the cerebral ventricles usually normalizes the bone phenotype in leptin-deficient mice. Reflecting the role of the sympathetic nervous system in mediating the central catabolic effects of leptin on the skeleton, β-adrenergic agonists and antagonists have major effects on bone in mice, but this is not consistently seen in humans. The balance of the central and peripheral effects of leptin on bone remains an area of substantial controversy and might vary between species and according to other factors such as body weight, baseline circulating leptin levels, and the presence of specific pathologies. In humans, leptin is likely to contribute to the positive relationship observed between adiposity and bone density, which allows the skeleton to respond appropriately to changes in soft tissue mass.
Collapse
Affiliation(s)
- Ian R Reid
- University of Auckland, Auckland, New Zealand.,Department of Endocrinology, Auckland District Health Board, Auckland, New Zealand
| | - Paul A Baldock
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | | |
Collapse
|
9
|
Masarwi M, Shamir R, Phillip M, Gat-Yablonski G. Leptin stimulates aromatase in the growth plate: limiting catch-up growth efficiency. J Endocrinol 2018; 237:229-242. [PMID: 29615477 DOI: 10.1530/joe-18-0028] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Accepted: 04/03/2018] [Indexed: 12/15/2022]
Abstract
Catch-up growth (CUG) in childhood is defined as periods of growth acceleration, after the resolution of growth attenuation causes, bringing the children back to their original growth trajectory. Sometimes, however, CUG is incomplete, leading to permanent growth deficit and short stature. The aim of this study was to investigate the mechanisms that limit nutritional-CUG. Specifically, we focused on the crosstalk between leptin, increased by re-feeding, and sex hormones, which increase with age. In vivo studies were performed in young male Sprague Dawley rats fed ad libitum or subjected to 10/36 days of 40% food restriction followed by 90-120 days of re-feeding. In vitro studies were performed on ATDC5 cells. Analyses of mRNA and protein levels were done using qPCR and Western blot, respectively. CUG was complete in body weight and humerus length in animals that were food-restricted for 10 days but not for those food-restricted for 36 days. In vitro studies showed that leptin significantly increased aromatase gene expression and protein level as well as the expression of estrogen and leptin receptors in a dose- and time-dependent manner. The effect of leptin on aromatase was direct and was mediated through the MAPK/Erk, STAT3 and PI3K pathways. The crosstalk between leptin and aromatase in the growth plate suggests that re-feeding during puberty may lead to increased estrogen level and activity, and consequently, irreversible premature epiphyseal growth plate closure. These results may have important implications for the development of novel treatment strategies for short stature in children.
Collapse
Affiliation(s)
- Majdi Masarwi
- Sackler Faculty of MedicineTel Aviv University, Tel Aviv, Israel
- Felsenstein Medical Research CenterPetach Tikva, Israel
| | - Raanan Shamir
- Sackler Faculty of MedicineTel Aviv University, Tel Aviv, Israel
- Felsenstein Medical Research CenterPetach Tikva, Israel
- Institute of GastroenterologyNutrition and Liver Diseases, Schneider Children's Medical Center of Israel, Petach Tikva, Israel
| | - Moshe Phillip
- Sackler Faculty of MedicineTel Aviv University, Tel Aviv, Israel
- Felsenstein Medical Research CenterPetach Tikva, Israel
- The Jesse Z and Sara Lea Shafer Institute for Endocrinology and DiabetesNational Center for Childhood Diabetes, Schneider Children's Medical Center of Israel, Petach Tikva, Israel
| | - Galia Gat-Yablonski
- Sackler Faculty of MedicineTel Aviv University, Tel Aviv, Israel
- Felsenstein Medical Research CenterPetach Tikva, Israel
- The Jesse Z and Sara Lea Shafer Institute for Endocrinology and DiabetesNational Center for Childhood Diabetes, Schneider Children's Medical Center of Israel, Petach Tikva, Israel
| |
Collapse
|
10
|
Gat-Yablonski G, De Luca F. Effect of Nutrition on Statural Growth
. Horm Res Paediatr 2018; 88:46-62. [PMID: 28365689 DOI: 10.1159/000456547] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 01/11/2017] [Indexed: 12/14/2022] Open
Abstract
In children, proper growth and development are often regarded as a surrogate marker for good health. A complex system controls the initiation, rate, and cessation of growth, and thus gives a wonderful example of the interactions between genetics, epigenetics, and environmental factors (especially stress and nutrition). Malnutrition is considered a leading cause of growth attenuation in children. This review summarizes our current knowledge regarding the mechanisms linking nutrition and skeletal growth, including systemic factors, such as insulin, growth hormone, insulin-like growth factor-1, fibroblast growth factor-21, etc., and local mechanisms, including mTOR, miRNAs, and epigenetics. Studying the molecular mechanisms regulating skeletal growth may lead to the establishment of better nutritional and therapeutic regimens for more effective linear growth in children with malnutrition and growth abnormalities.
.
Collapse
Affiliation(s)
- Galia Gat-Yablonski
- The Jesse Z and Sara Lea Shafer Institute for Endocrinology and Diabetes, National Center for Children's Diabetes, Schneider Children's Medical Center of Israel, Petach Tikva, Israel.,Felsenstein Medical Research Center, Petach Tikva, Israel.,Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Francesco De Luca
- Section of Endocrinology and Diabetes, St. Christopher's Hospital for Children, Department of Pediatrics, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
11
|
De Blasio MJ, Lanham SA, Blache D, Oreffo ROC, Fowden AL, Forhead AJ. Sex- and bone-specific responses in bone structure to exogenous leptin and leptin receptor antagonism in the ovine fetus. Am J Physiol Regul Integr Comp Physiol 2018; 314:R781-R790. [PMID: 29443548 DOI: 10.1152/ajpregu.00351.2017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Widespread expression of leptin and its receptor in developing cartilage and bone suggests that leptin may regulate bone growth and development in the fetus. Using microcomputed tomography, this study investigated the effects of exogenous leptin and leptin receptor antagonism on aspects of bone structure in the sheep fetus during late gestation. From 125 to 130 days of gestation (term ~145 days), chronically catheterized singleton sheep fetuses were infused intravenously for 5 days with either saline (0.9% saline, n = 13), recombinant ovine leptin at two doses (0.6 mg·kg-1·day-1 LEP1, n = 10 or 1.4 mg·kg-1·day-1 LEP2, n = 7), or recombinant superactive ovine leptin receptor antagonist (4.6 mg·kg-1·day-1 SOLA, n = 6). No significant differences in plasma insulin-like growth factor-I, osteocalcin, calcium, inorganic phosphate, or alkaline phosphatase were observed between treatment groups. Total femur midshaft diameter and metatarsal lumen diameter were narrower in male fetuses treated with exogenous leptin. In a fixed length of femur midshaft, total and bone volumes were reduced by the higher dose of leptin; nonbone space volume was lower in both groups of leptin-treated fetuses. Leptin infusion caused increments in femur porosity and connectivity density, and vertebral trabecular thickness. Leptin receptor antagonism decreased trabecular spacing and increased trabecular number, degree of anisotrophy, and connectivity density in the lumbar vertebrae. The increase in vertebral porosity observed following leptin receptor antagonism was greater in the malecompared with female, fetuses. Therefore, leptin may have a role in the growth and development of the fetal skeleton, dependent on the concentration of leptin, sex of the fetus, and bone type examined.
Collapse
Affiliation(s)
- Miles J De Blasio
- Department of Physiology, Development, and Neuroscience, University of Cambridge , Cambridge , United Kingdom
| | - Stuart A Lanham
- Bone and Joint Research Group, Centre for Human Development, Stem Cells, and Regeneration, Institute of Developmental Sciences, University of Southampton , Southampton , United Kingdom
| | - Dominique Blache
- School of Animal Biology, University of Western Australia , Crawley , Australia
| | - Richard O C Oreffo
- Bone and Joint Research Group, Centre for Human Development, Stem Cells, and Regeneration, Institute of Developmental Sciences, University of Southampton , Southampton , United Kingdom
| | - Abigail L Fowden
- Department of Physiology, Development, and Neuroscience, University of Cambridge , Cambridge , United Kingdom
| | - Alison J Forhead
- Department of Physiology, Development, and Neuroscience, University of Cambridge , Cambridge , United Kingdom.,Department of Biological and Medical Sciences, Oxford Brookes University , Oxford , United Kingdom
| |
Collapse
|
12
|
Philbrick KA, Martin SA, Colagiovanni AR, Branscum AJ, Turner RT, Iwaniec UT. Effects of hypothalamic leptin gene therapy on osteopetrosis in leptin-deficient mice. J Endocrinol 2018; 236:57-68. [PMID: 29191939 PMCID: PMC5771473 DOI: 10.1530/joe-17-0524] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2017] [Accepted: 11/20/2017] [Indexed: 12/26/2022]
Abstract
Impaired resorption of cartilage matrix deposited during endochondral ossification is a defining feature of juvenile osteopetrosis. Growing, leptin-deficient ob/ob mice exhibit a mild form of osteopetrosis. However, the extent to which the disease is (1) self-limiting and (2) reversible by leptin treatment is unknown. We addressed the first question by performing histomorphometric analysis of femurs in rapidly growing (2-month-old), slowly growing (4-month-old) and skeletally mature (6-month-old) wild-type (WT) and ob/ob male mice. Absent by 6 months of age in WT mice, cartilage matrix persisted to varying extents in distal femur epiphysis, metaphysis and diaphysis in ob/ob mice, suggesting that the osteopetrotic phenotype is not entirely self-limiting. To address the second question, we employed hypothalamic recombinant adeno-associated virus (rAAV) gene therapy to restore leptin signaling in ob/ob mice. Two-month-old mice were randomized to one of the three groups: (1) untreated control, (2) rAAV-Leptin or (3) control vector rAAV-green fluorescent protein and vectors injected intracerebroventricularly. Seven months later, rAAV-leptin-treated mice exhibited no cartilage in the metaphysis and greatly reduced cartilage in the epiphysis and diaphysis. At the cellular level, the reduction in cartilage was associated with increased bone turnover. These findings (1) support the concept that leptin is important for normal replacement of cartilage by bone, and (2) demonstrate that osteopetrosis in ob/ob mice is bone-compartment-specific and reversible by leptin at skeletal sites capable of undergoing robust bone turnover.
Collapse
Affiliation(s)
- Kenneth A Philbrick
- Skeletal Biology LaboratorySchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USA
| | - Stephen A Martin
- Skeletal Biology LaboratorySchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USA
| | - Amy R Colagiovanni
- Skeletal Biology LaboratorySchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USA
| | - Adam J Branscum
- Biostatistics ProgramSchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USA
| | - Russell T Turner
- Skeletal Biology LaboratorySchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USA
- Center for Healthy Aging ResearchOregon State University, Corvallis, Oregon, USA
| | - Urszula T Iwaniec
- Skeletal Biology LaboratorySchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USA
- Center for Healthy Aging ResearchOregon State University, Corvallis, Oregon, USA
| |
Collapse
|
13
|
|
14
|
Firth EC, Gamble GD, Cornish J, Vickers MH. Neonatal leptin treatment reverses the bone-suppressive effects of maternal undernutrition in adult rat offspring. Sci Rep 2017; 7:7686. [PMID: 28794412 PMCID: PMC5550441 DOI: 10.1038/s41598-017-07500-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 06/29/2017] [Indexed: 11/19/2022] Open
Abstract
Alterations in the early life environment, including maternal undernutrition (UN) during pregnancy, can lead to increased risk of metabolic and cardiovascular disorders in offspring. Leptin treatment of neonates born to UN rats reverses the programmed metabolic phenotype, but the possible benefits of this treatment on bone tissue have not been defined. We describe for the first time the effects of neonatal leptin treatment on bone in adult offspring following maternal UN. Offspring from either UN or ad libitum-fed (AD) rats were treated with either saline or leptin (2.5 µg/ g.d on postnatal days (D)3–13) and were fed either a chow or high fat (HF) diet from weaning until study completion at D170. Analysis of micro-tomographic data of the left femur showed highly significant effects of UN on cortical and trabecular bone tissue indices, contributing to inferior microstructure and bone strength, almost all of which were reversed by early leptin life treatment. The HF fat diet negatively affected trabecular bone tissue, but the effects of only trabecular separation and number were reversed by leptin treatment. The negative effects of maternal UN on skeletal health in adult offspring might be prevented or attenuated by various interventions including leptin. Establishment of a minimal efficacious leptin dose warrants further study.
Collapse
Affiliation(s)
- Elwyn C Firth
- Liggins Institute, University of Auckland, Auckland, New Zealand. .,Department of Exercise Sciences, Faculty of Science, University of Auckland, Auckland, New Zealand.
| | - Greg D Gamble
- Bone and Joint Research Group, Department of Medicine, University of Auckland, Auckland, New Zealand
| | - Jillian Cornish
- Bone and Joint Research Group, Department of Medicine, University of Auckland, Auckland, New Zealand
| | - Mark H Vickers
- Liggins Institute, University of Auckland, Auckland, New Zealand
| |
Collapse
|
15
|
Oral Supplementation with a Special Additive of Retinyl Palmitate and Alpha Tocopherol Reduces Growth Retardation in Young Pancreatic Duct Ligated Pigs Used as a Model for Children Suffering from Exocrine Pancreatic Insufficiency. Int J Mol Sci 2016; 17:ijms17101642. [PMID: 27690005 PMCID: PMC5085675 DOI: 10.3390/ijms17101642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 09/07/2016] [Accepted: 09/18/2016] [Indexed: 11/17/2022] Open
Abstract
Pancreatic exocrine insufficiency (PEI) is a disease of diverse aetiology-e.g., majority of patients suffering from cystic fibrosis (CF) show PEI congenitally. Malnutrition and malabsorption of nutrients impair growth and nutritional status. As reduced fat digestion leads to a deficiency of fat-soluble vitamins the supplementation is standard, but absorption is a critical point in PEI-patients. The pancreatic duct ligated (PL) pig is an established model for PEI in humans and has been proven to be a suitable model to compare different vitamin additives for supplementation. In a former study, PEI caused distinct growth retardation in young piglets, but did not affect growth in older ones. Our study hypothesised that this age-dependent effect is caused by exhausted body reserves of fat-soluble vitamins and, therefore, extra supply reduces growth retardation. PEI was induced by PL at the age of seven (PL-7) or 16 weeks (PL-16). Controls (C) underwent a sham surgery. Some PL-7 pigs (PL-7 + Vit) were fed a special vitamin additive. PEI reduced the mean final body weight (kg) at 26 weeks of age significantly with lower effect in PL-16-pigs (C:117; PL-7:49.5; PL-7 + Vit:77.1; PL-16:96.4). Extra vitamin supply resulted in an increased growth and normalised serum concentration of alpha-tocopherol, underlining the importance of special supplementation in PEI-patients.
Collapse
|
16
|
Iwaniec UT, Turner RT. Influence of body weight on bone mass, architecture and turnover. J Endocrinol 2016; 230:R115-30. [PMID: 27352896 PMCID: PMC4980254 DOI: 10.1530/joe-16-0089] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 06/27/2016] [Indexed: 12/11/2022]
Abstract
Weight-dependent loading of the skeleton plays an important role in establishing and maintaining bone mass and strength. This review focuses on mechanical signaling induced by body weight as an essential mechanism for maintaining bone health. In addition, the skeletal effects of deviation from normal weight are discussed. The magnitude of mechanical strain experienced by bone during normal activities is remarkably similar among vertebrates, regardless of size, supporting the existence of a conserved regulatory mechanism, or mechanostat, that senses mechanical strain. The mechanostat functions as an adaptive mechanism to optimize bone mass and architecture based on prevailing mechanical strain. Changes in weight, due to altered mass, weightlessness (spaceflight), and hypergravity (modeled by centrifugation), induce an adaptive skeletal response. However, the precise mechanisms governing the skeletal response are incompletely understood. Furthermore, establishing whether the adaptive response maintains the mechanical competence of the skeleton has proven difficult, necessitating the development of surrogate measures of bone quality. The mechanostat is influenced by regulatory inputs to facilitate non-mechanical functions of the skeleton, such as mineral homeostasis, as well as hormones and energy/nutrient availability that support bone metabolism. Although the skeleton is very capable of adapting to changes in weight, the mechanostat has limits. At the limits, extreme deviations from normal weight and body composition are associated with impaired optimization of bone strength to prevailing body size.
Collapse
Affiliation(s)
- Urszula T Iwaniec
- Skeletal Biology LaboratorySchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USA Center for Healthy Aging ResearchOregon State University, Corvallis, Oregon, USA
| | - Russell T Turner
- Skeletal Biology LaboratorySchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USA Center for Healthy Aging ResearchOregon State University, Corvallis, Oregon, USA
| |
Collapse
|
17
|
Relationships between Wasting and Stunting and Their Concurrent Occurrence in Ghanaian Preschool Children. J Nutr Metab 2016; 2016:4654920. [PMID: 27379184 PMCID: PMC4917721 DOI: 10.1155/2016/4654920] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2016] [Revised: 05/02/2016] [Accepted: 05/15/2016] [Indexed: 01/12/2023] Open
Abstract
Objective. The main aim of the study was to assess the magnitude of concurrent wasting and stunting among Ghanaian preschool children. Secondly, we investigated the relationship between wasting and stunting as well as factors associated with these conditions. Methods. This paper is based on reanalysis of anthropometric and other relevant data which was collected in the 2014 Ghana Demographic and Health Survey. The data set consisted of 2,720 preschool children aged 0-59 months. We conducted three-step moderated hierarchical multiple regression analyses to determine independent predictors and moderators of height-for-age Z-score. Results. Nationally, the prevalence of concurrent wasting and stunting among children aged 0-59 months was low at 1.4% but it varied geographically with the Upper East Region having the highest prevalence of 3.2% (95% CI: 1.7-5.8). Children who had low weight-for-height Z-scores were at a higher risk of linear growth retardation (stunting) especially among children aged less than three years. A 1-unit increase in weight-for-height Z-score (WHZ) was associated with 0.07 standard units' increase in height-for-age Z-score (HAZ) [β = 0.071 (95% CI: 0.03, 0.15)]. Conclusions. The study results suggest that weight-for-height relates to linear growth but this relationship is moderated by age of child. Stunting and wasting share some common risk factors. Therefore, measures to prevent wasting may positively influence linear growth.
Collapse
|
18
|
Devlin MJ, Brooks DJ, Conlon C, Vliet MV, Louis L, Rosen CJ, Bouxsein ML. Daily leptin blunts marrow fat but does not impact bone mass in calorie-restricted mice. J Endocrinol 2016; 229:295-306. [PMID: 27340200 PMCID: PMC5171226 DOI: 10.1530/joe-15-0473] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 04/11/2016] [Indexed: 01/02/2023]
Abstract
Starvation induces low bone mass and high bone marrow adiposity in humans, but the underlying mechanisms are poorly understood. The adipokine leptin falls in starvation, suggesting that hypoleptinemia may be a link between negative energy balance, bone marrow fat accumulation, and impaired skeletal acquisition. In that case, treating mice with leptin during caloric restriction (CR) should reduce marrow adipose tissue (MAT) and improve bone mass. To test this hypothesis, female C57Bl/6J mice were fed a 30% CR or normal (N) diet from 5 to 10 weeks of age, with daily injections of vehicle (VEH), 1mg/kg leptin (LEP1), or 2mg/kg leptin (LEP2) (N=6-8/group). Outcomes included body mass, body fat percentage, and whole-body bone mineral density (BMD) via peripheral dual-energy X-ray absorptiometry, cortical and trabecular microarchitecture via microcomputed tomography (μCT), and MAT volume via μCT of osmium tetroxide-stained bones. Overall, CR mice had lower body mass, body fat percentage, BMD, and cortical bone area fraction, but more connected trabeculae, vs N mice (P<0.05 for all). Most significantly, although MAT was elevated in CR vs N overall, leptin treatment blunted MAT formation in CR mice by 50% vs VEH (P<0.05 for both leptin doses). CR LEP2 mice weighed less vs CR VEH mice at 9-10 weeks of age (P<0.05), but leptin treatment did not affect body fat percentage, BMD, or bone microarchitecture within either diet. These data demonstrate that once daily leptin bolus during CR inhibits bone marrow adipose expansion without affecting bone mass acquisition, suggesting that leptin has distinct effects on starvation-induced bone marrow fat formation and skeletal acquisition.
Collapse
Affiliation(s)
- M J Devlin
- Department of AnthropologyUniversity of Michigan, Ann Arbor, Michigan, USA
| | - D J Brooks
- Center for Advanced Orthopedic StudiesBeth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - C Conlon
- Center for Advanced Orthopedic StudiesBeth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - M van Vliet
- Center for Advanced Orthopedic StudiesBeth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - L Louis
- Center for Advanced Orthopedic StudiesBeth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - C J Rosen
- Maine Medical Center Research InstituteScarborough, Maine, USA
| | - M L Bouxsein
- Center for Advanced Orthopedic StudiesBeth Israel Deaconess Medical Center, Boston, Massachusetts, USA Harvard Medical SchoolBoston, Massachusetts, USA
| |
Collapse
|
19
|
Abstract
The regulation of organ size is essential to human health and has fascinated biologists for centuries. Key to the growth process is the ability of most organs to integrate organ-extrinsic cues (eg, nutritional status, inflammatory processes) with organ-intrinsic information (eg, genetic programs, local signals) into a growth response that adapts to changing environmental conditions and ensures that the size of an organ is coordinated with the rest of the body. Paired organs such as the vertebrate limbs and the long bones within them are excellent models for studying this type of regulation because it is possible to manipulate one member of the pair and leave the other as an internal control. During development, growth plates at the end of each long bone produce a transient cartilage model that is progressively replaced by bone. Here, we review how proliferation and differentiation of cells within each growth plate are tightly controlled mainly by growth plate-intrinsic mechanisms that are additionally modulated by extrinsic signals. We also discuss the involvement of several signaling hubs in the integration and modulation of growth-related signals and how they could confer remarkable plasticity to the growth plate. Indeed, long bones have a significant ability for "catch-up growth" to attain normal size after a transient growth delay. We propose that the characterization of catch-up growth, in light of recent advances in physiology and cell biology, will provide long sought clues into the molecular mechanisms that underlie organ growth regulation. Importantly, catch-up growth early in life is commonly associated with metabolic disorders in adulthood, and this association is not completely understood. Further elucidation of the molecules and cellular interactions that influence organ size coordination should allow development of novel therapies for human growth disorders that are noninvasive and have minimal side effects.
Collapse
Affiliation(s)
- Alberto Roselló-Díez
- Developmental Biology Program, Sloan Kettering Institute, New York, New York 10065
| | - Alexandra L Joyner
- Developmental Biology Program, Sloan Kettering Institute, New York, New York 10065
| |
Collapse
|
20
|
Liu A, Li Y, Wang Y, Liu L, Shi H, Qiu Y. Exogenous Parathyroid Hormone-Related Peptide Promotes Fracture Healing in Lepr(-/-) Mice. Calcif Tissue Int 2015; 97:581-91. [PMID: 26314884 DOI: 10.1007/s00223-015-0041-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 07/15/2015] [Indexed: 10/23/2022]
Abstract
Diabetic osteoporosis continues to surge worldwide, increasing the risk of fracture. We have previously demonstrated that haploinsufficiency of endogenous parathyroid hormone-related peptide (PTHrP) impairs fracture healing. However, whether an exogenous supply of PTHrP can repair bone damage and accelerate fracture healing remains unclear. This study aimed to assess the efficacy and safety of PTHrP in healing fractures. Standardized mid-diaphyseal femur fractures were generated in 12-week-old wild-type and leptin receptor null Lepr(-/-) mice. After administration of PTHrP for 2 weeks, callus tissue properties were analyzed by radiography, micro-computed tomography, histology, histochemistry, immunohistochemistry, and molecular biology techniques. At 2 weeks post-fracture, cartilaginous callus areas were reduced, while total callus and bony callus areas were increased in PTHrP-treated Lepr(-/-) animals and control wild-type mice, compared with vehicle-treated Lepr(-/-) mice. The following parameters were enhanced both in Lepr(-/-) mice after treatment with PTHrP and vehicle-treated wild-type animals, compared with vehicle-treated Lepr(-/-) mice: osteoblast numbers; tissue alkaline phosphatase (ALP) and Type I collagen immunopositive areas; mRNA levels of ALP, Type I collagen, osteoprotegerin, and receptor activator for nuclear factor-κ B ligand; protein levels of Runt-related transcription factor 2 and insulin-like growth factor-1; and the number and surface of osteoclasts. In conclusion, exogenous PTHrP by subcutaneous injection promotes fracture repair in Lepr(-/-) mice by increasing callus formation and accelerating cell transformation: upregulated osteoblastic gene and protein expression, increased endochondral bone formation, osteoblastic bone formation, and osteoclastic bone resorption. However, complete repair was not obtained in PTHrP-treated Lepr(-/-) mice as in control wild-type animals.
Collapse
Affiliation(s)
- Anlong Liu
- Department of Orthopaedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, Jiangsu, China
| | - Yishan Li
- Department of International Training, PLA University of Science and Technology, Nanjing, 210007, Jiangsu, China
| | - Yinhe Wang
- Department of Orthopaedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, Jiangsu, China.
| | - Li Liu
- Department of Orthopaedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, Jiangsu, China
| | - Hongfei Shi
- Department of Orthopaedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, Jiangsu, China
| | - Yong Qiu
- Department of Orthopaedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, Jiangsu, China.
| |
Collapse
|
21
|
Turner RT, Dube M, Branscum AJ, Wong CP, Olson DA, Zhong X, Kweh MF, Larkin IV, Wronski TJ, Rosen CJ, Kalra SP, Iwaniec UT. Hypothalamic leptin gene therapy reduces body weight without accelerating age-related bone loss. J Endocrinol 2015; 227:129-41. [PMID: 26487675 PMCID: PMC4917201 DOI: 10.1530/joe-15-0280] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/14/2015] [Indexed: 02/04/2023]
Abstract
Excessive weight gain in adults is associated with a variety of negative health outcomes. Unfortunately, dieting, exercise, and pharmacological interventions have had limited long-term success in weight control and can result in detrimental side effects, including accelerating age-related cancellous bone loss. We investigated the efficacy of using hypothalamic leptin gene therapy as an alternative method for reducing weight in skeletally-mature (9 months old) female rats and determined the impact of leptin-induced weight loss on bone mass, density, and microarchitecture, and serum biomarkers of bone turnover (CTx and osteocalcin). Rats were implanted with cannulae in the 3rd ventricle of the hypothalamus and injected with either recombinant adeno-associated virus encoding the gene for rat leptin (rAAV-Leptin, n=7) or a control vector encoding green fluorescent protein (rAAV-GFP, n=10) and sacrificed 18 weeks later. A baseline control group (n=7) was sacrificed at vector administration. rAAV-Leptin-treated rats lost weight (-4±2%) while rAAV-GFP-treated rats gained weight (14±2%) during the study. At study termination, rAAV-Leptin-treated rats weighed 17% less than rAAV-GFP-treated rats and had lower abdominal white adipose tissue weight (-80%), serum leptin (-77%), and serum IGF1 (-34%). Cancellous bone volume fraction in distal femur metaphysis and epiphysis, and in lumbar vertebra tended to be lower (P<0.1) in rAAV-GFP-treated rats (13.5 months old) compared to baseline control rats (9 months old). Significant differences in cancellous bone or biomarkers of bone turnover were not detected between rAAV-Leptin and rAAV-GFP rats. In summary, rAAV-Leptin-treated rats maintained a lower body weight compared to baseline and rAAV-GFP-treated rats with minimal effects on bone mass, density, microarchitecture, or biochemical markers of bone turnover.
Collapse
Affiliation(s)
- Russell T Turner
- Skeletal Biology LaboratorySchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon 97331, USACenter for Healthy Aging ResearchOregon State University, Corvallis, Oregon, USADepartment of NeuroscienceMcKnight Brain Institute, University of Florida, Gainesville, Florida, USABiostatisticsSchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USADepartment of Physiological SciencesUniversity of Florida, Gainesville, Florida, USADepartment of Large Animal Clinical SciencesUniversity of Florida, Gainesville, Florida, USAMaine Medical Center Research InstituteScarborough, Maine, USA Skeletal Biology LaboratorySchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon 97331, USACenter for Healthy Aging ResearchOregon State University, Corvallis, Oregon, USADepartment of NeuroscienceMcKnight Brain Institute, University of Florida, Gainesville, Florida, USABiostatisticsSchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USADepartment of Physiological SciencesUniversity of Florida, Gainesville, Florida, USADepartment of Large Animal Clinical SciencesUniversity of Florida, Gainesville, Florida, USAMaine Medical Center Research InstituteScarborough, Maine, USA
| | - Michael Dube
- Skeletal Biology LaboratorySchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon 97331, USACenter for Healthy Aging ResearchOregon State University, Corvallis, Oregon, USADepartment of NeuroscienceMcKnight Brain Institute, University of Florida, Gainesville, Florida, USABiostatisticsSchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USADepartment of Physiological SciencesUniversity of Florida, Gainesville, Florida, USADepartment of Large Animal Clinical SciencesUniversity of Florida, Gainesville, Florida, USAMaine Medical Center Research InstituteScarborough, Maine, USA
| | - Adam J Branscum
- Skeletal Biology LaboratorySchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon 97331, USACenter for Healthy Aging ResearchOregon State University, Corvallis, Oregon, USADepartment of NeuroscienceMcKnight Brain Institute, University of Florida, Gainesville, Florida, USABiostatisticsSchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USADepartment of Physiological SciencesUniversity of Florida, Gainesville, Florida, USADepartment of Large Animal Clinical SciencesUniversity of Florida, Gainesville, Florida, USAMaine Medical Center Research InstituteScarborough, Maine, USA
| | - Carmen P Wong
- Skeletal Biology LaboratorySchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon 97331, USACenter for Healthy Aging ResearchOregon State University, Corvallis, Oregon, USADepartment of NeuroscienceMcKnight Brain Institute, University of Florida, Gainesville, Florida, USABiostatisticsSchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USADepartment of Physiological SciencesUniversity of Florida, Gainesville, Florida, USADepartment of Large Animal Clinical SciencesUniversity of Florida, Gainesville, Florida, USAMaine Medical Center Research InstituteScarborough, Maine, USA
| | - Dawn A Olson
- Skeletal Biology LaboratorySchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon 97331, USACenter for Healthy Aging ResearchOregon State University, Corvallis, Oregon, USADepartment of NeuroscienceMcKnight Brain Institute, University of Florida, Gainesville, Florida, USABiostatisticsSchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USADepartment of Physiological SciencesUniversity of Florida, Gainesville, Florida, USADepartment of Large Animal Clinical SciencesUniversity of Florida, Gainesville, Florida, USAMaine Medical Center Research InstituteScarborough, Maine, USA
| | - Xiaoying Zhong
- Skeletal Biology LaboratorySchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon 97331, USACenter for Healthy Aging ResearchOregon State University, Corvallis, Oregon, USADepartment of NeuroscienceMcKnight Brain Institute, University of Florida, Gainesville, Florida, USABiostatisticsSchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USADepartment of Physiological SciencesUniversity of Florida, Gainesville, Florida, USADepartment of Large Animal Clinical SciencesUniversity of Florida, Gainesville, Florida, USAMaine Medical Center Research InstituteScarborough, Maine, USA
| | - Mercedes F Kweh
- Skeletal Biology LaboratorySchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon 97331, USACenter for Healthy Aging ResearchOregon State University, Corvallis, Oregon, USADepartment of NeuroscienceMcKnight Brain Institute, University of Florida, Gainesville, Florida, USABiostatisticsSchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USADepartment of Physiological SciencesUniversity of Florida, Gainesville, Florida, USADepartment of Large Animal Clinical SciencesUniversity of Florida, Gainesville, Florida, USAMaine Medical Center Research InstituteScarborough, Maine, USA
| | - Iske V Larkin
- Skeletal Biology LaboratorySchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon 97331, USACenter for Healthy Aging ResearchOregon State University, Corvallis, Oregon, USADepartment of NeuroscienceMcKnight Brain Institute, University of Florida, Gainesville, Florida, USABiostatisticsSchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USADepartment of Physiological SciencesUniversity of Florida, Gainesville, Florida, USADepartment of Large Animal Clinical SciencesUniversity of Florida, Gainesville, Florida, USAMaine Medical Center Research InstituteScarborough, Maine, USA
| | - Thomas J Wronski
- Skeletal Biology LaboratorySchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon 97331, USACenter for Healthy Aging ResearchOregon State University, Corvallis, Oregon, USADepartment of NeuroscienceMcKnight Brain Institute, University of Florida, Gainesville, Florida, USABiostatisticsSchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USADepartment of Physiological SciencesUniversity of Florida, Gainesville, Florida, USADepartment of Large Animal Clinical SciencesUniversity of Florida, Gainesville, Florida, USAMaine Medical Center Research InstituteScarborough, Maine, USA
| | - Clifford J Rosen
- Skeletal Biology LaboratorySchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon 97331, USACenter for Healthy Aging ResearchOregon State University, Corvallis, Oregon, USADepartment of NeuroscienceMcKnight Brain Institute, University of Florida, Gainesville, Florida, USABiostatisticsSchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USADepartment of Physiological SciencesUniversity of Florida, Gainesville, Florida, USADepartment of Large Animal Clinical SciencesUniversity of Florida, Gainesville, Florida, USAMaine Medical Center Research InstituteScarborough, Maine, USA
| | - Satya P Kalra
- Skeletal Biology LaboratorySchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon 97331, USACenter for Healthy Aging ResearchOregon State University, Corvallis, Oregon, USADepartment of NeuroscienceMcKnight Brain Institute, University of Florida, Gainesville, Florida, USABiostatisticsSchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USADepartment of Physiological SciencesUniversity of Florida, Gainesville, Florida, USADepartment of Large Animal Clinical SciencesUniversity of Florida, Gainesville, Florida, USAMaine Medical Center Research InstituteScarborough, Maine, USA
| | - Urszula T Iwaniec
- Skeletal Biology LaboratorySchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon 97331, USACenter for Healthy Aging ResearchOregon State University, Corvallis, Oregon, USADepartment of NeuroscienceMcKnight Brain Institute, University of Florida, Gainesville, Florida, USABiostatisticsSchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USADepartment of Physiological SciencesUniversity of Florida, Gainesville, Florida, USADepartment of Large Animal Clinical SciencesUniversity of Florida, Gainesville, Florida, USAMaine Medical Center Research InstituteScarborough, Maine, USA Skeletal Biology LaboratorySchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon 97331, USACenter for Healthy Aging ResearchOregon State University, Corvallis, Oregon, USADepartment of NeuroscienceMcKnight Brain Institute, University of Florida, Gainesville, Florida, USABiostatisticsSchool of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USADepartment of Physiological SciencesUniversity of Florida, Gainesville, Florida, USADepartment of Large Animal Clinical SciencesUniversity of Florida, Gainesville, Florida, USAMaine Medical Center Research InstituteScarborough, Maine, USA
| |
Collapse
|
22
|
Hara ES, Ono M, Pham HT, Sonoyama W, Kubota S, Takigawa M, Matsumoto T, Young MF, Olsen BR, Kuboki T. Fluocinolone Acetonide Is a Potent Synergistic Factor of TGF-β3-Associated Chondrogenesis of Bone Marrow-Derived Mesenchymal Stem Cells for Articular Surface Regeneration. J Bone Miner Res 2015; 30:1585-96. [PMID: 25753754 PMCID: PMC5569386 DOI: 10.1002/jbmr.2502] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Revised: 02/09/2015] [Accepted: 03/04/2015] [Indexed: 12/18/2022]
Abstract
Articular cartilage repair remains a challenging problem. Based on a high-throughput screening and functional analysis, we found that fluocinolone acetonide (FA) in combination with transforming growth factor beta 3 (TGF-β3) strongly potentiated chondrogenic differentiation of human bone marrow-derived mesenchymal stem cells (hBMSCs). In an in vivo cartilage defect model in knee joints of immunocompromised mice, transplantation of FA/TGF-β3-treated hBMSCs could completely repair the articular surface. Analysis of the intracellular pathways revealed that FA enhanced TGF-β3-induced phosphorylation of Smad2 and Smad3. Additionally, we performed a pathway array and found that FA activates the mTORC1/AKT pathway. Chemical inhibition of mTORC1 with rapamycin substantially suppressed FA effect, and inhibition of AKT completely repressed chondrogenesis of hBMSCs. Inhibition of glucocorticoid receptor with mifepristone also suppressed FA effect, suggesting that FA involves binding to the glucocorticoid receptor. Comparative analysis with other glucocorticoids (triamcinolone acetonide [TA] and dexamethasone [DEX]) revealed the unique ability of FA to repair articular cartilage surgical defects. Analysis of intracellular pathways showed that the mTORC1/AKT pathway and the glucocorticoid receptor was highly activated with FA and TA, but to a lesser extent with DEX. Collectively, these results show a unique ability of FA to enhance TGF-β3-associated chondrogenesis, and suggest that the FA/TGF-β3 combination may be used as major inducer of chondrogenesis in vitro. Additionally, FA/TGF-β3 could be potentially applied in a clinical setting to increase the efficiency of regenerative approaches based on chondrogenic differentiation of stem cells.
Collapse
Affiliation(s)
- Emilio Satoshi Hara
- Department of Oral Rehabilitation and Regenerative Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan.,Department of Biomaterials, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Mitsuaki Ono
- Department of Oral Rehabilitation and Regenerative Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Hai Thanh Pham
- Department of Oral Rehabilitation and Regenerative Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Wataru Sonoyama
- Department of Oral Rehabilitation and Regenerative Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Satoshi Kubota
- Department of Biochemistry and Molecular Dentistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Masaharu Takigawa
- Advanced Research Center for Oral and Craniofacial Sciences, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Takuya Matsumoto
- Department of Biomaterials, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Marian F Young
- Craniofacial and Skeletal Diseases Branch, National Institutes of Craniofacial and Dental Research, National Institutes of Health, Bethesda, MD,, USA
| | - Bjorn R Olsen
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA,, USA
| | - Takuo Kuboki
- Department of Oral Rehabilitation and Regenerative Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| |
Collapse
|
23
|
Abstract
Muscle- and liver-derived IGF-1 play important roles in muscle anabolism throughout growth and aging. Yet, prolonged food restriction is thought to increase longevity in part by lowering levels of IGF-1, which in turn reduces the risk for developing various cancers. The dietary factors that modulate IGF-1 levels are, however, poorly understood. We tested the hypothesis that the adipokine leptin, which is elevated with food intake and suppressed during fasting, is a key mediator of IGF-1 levels with aging and food restriction. First, leptin levels in peripheral tissues were measured in young mice fed ad libitum, aged mice fed ad libitum, and aged calorie-restricted (CR) mice. A group of aged CR mice were also treated with recombinant leptin for 10 days. Later, aged mice fed ad libitum were treated with saline (VEH) or with a novel leptin receptor antagonist peptide (Allo-aca) and tissue-specific levels of IGF-1 were determined. On one hand, recombinant leptin induced a three-fold increase in liver-derived IGF-1 and a two-fold increase in muscle-derived IGF-1 in aged, CR mice. Leptin also significantly increased serum growth hormone levels in the aged, CR mice. On the other, the leptin receptor antagonist Allo-aca did not alter body weight or muscle mass in treated mice compared to VEH mice. Allo-aca did, however, produce a significant (20%) decline in liver-derived IGF-1 as well as an even more pronounced (>50%) decrease in muscle-derived IGF-1 compared to VEH-treated mice. The reduced IGF-1 levels in Allo-aca treated mice were not accompanied by any significant change in growth hormone levels compared to VEH mice. These findings suggest that leptin receptor antagonists may represent novel therapeutic agents for attenuating IGF-1 signaling associated with aging, and could potentially mimic some of the positive effects of calorie restriction on longevity.
Collapse
|
24
|
Mitchell SE, Tang Z, Kerbois C, Delville C, Konstantopedos P, Bruel A, Derous D, Green C, Aspden RM, Goodyear SR, Chen L, Han JJ, Wang Y, Promislow DE, Lusseau D, Douglas A, Speakman JR. The effects of graded levels of calorie restriction: I. impact of short term calorie and protein restriction on body composition in the C57BL/6 mouse. Oncotarget 2015; 6:15902-30. [PMID: 26079539 PMCID: PMC4599246 DOI: 10.18632/oncotarget.4142] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 03/31/2015] [Indexed: 12/18/2022] Open
Abstract
Faced with reduced levels of food, animals must adjust to the consequences of the shortfall in energy. We explored how C57BL/6 mice withdrew energy from different body tissues during three months of food restriction at graded levels up to 40% (calorie restriction: CR). We compared this to the response to equivalent levels of protein restriction (PR) without a shortfall in calories. Under CR there was a dynamic change in body mass over 30 days and thereafter it stabilized. The time to reach stability was independent of the level of restriction. At the end of three months whole body dissections revealed differential utilization of the different tissues. Adipose tissue depots were the most significantly utilized tissue, and provided 55.8 to 60.9% of the total released energy. In comparison, reductions in the sizes of structural tissues contributed between 29.8 and 38.7% of the energy. The balance was made up by relatively small changes in the vital organs. The components of the alimentary tract grew slightly under restriction, particularly the stomach, and this was associated with a parallel increase in assimilation efficiency of the food (averaging 1.73%). None of the changes under CR were recapitulated by equivalent levels of PR.
Collapse
Affiliation(s)
- Sharon E. Mitchell
- Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, Scotland, UK
| | - Zhanhui Tang
- Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, Scotland, UK
| | - Celine Kerbois
- Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, Scotland, UK
| | - Camille Delville
- Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, Scotland, UK
| | - Penelope Konstantopedos
- Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, Scotland, UK
| | - Aurélie Bruel
- Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, Scotland, UK
| | - Davina Derous
- Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, Scotland, UK
| | - Cara Green
- Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, Scotland, UK
| | - Richard M. Aspden
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, Scotland, UK
| | - Simon R. Goodyear
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, Scotland, UK
| | - Luonan Chen
- Key laboratory of Systems Biology, Shanghai Institute of Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Jackie J.D. Han
- Chinese Academy of Sciences Key Laboratory of Computational Biology, Chinese Academy of Sciences, Max Planck Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yingchun Wang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Chaoyang, Beijing, China
| | - Daniel E.L. Promislow
- Department of Pathology and Department of Biology, University of Washington, Seattle, WA, USA
| | - David Lusseau
- Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, Scotland, UK
| | - Alex Douglas
- Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, Scotland, UK
| | - John R. Speakman
- Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, Scotland, UK
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Chaoyang, Beijing, China
| |
Collapse
|
25
|
Briend A, Khara T, Dolan C. Wasting and Stunting—Similarities and Differences: Policy and Programmatic Implications. Food Nutr Bull 2015; 36:S15-23. [DOI: 10.1177/15648265150361s103] [Citation(s) in RCA: 119] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Wasting and stunting are often presented as two separate forms of malnutrition requiring different interventions for prevention and/or treatment. These two forms of malnutrition, however, are closely related and often occur together in the same populations and often in the same children. Wasting and stunting are both associated with increased mortality, especially when both are present in the same child. A better understanding of the pathophysiology of these two different forms of malnutrition is needed to design efficient programs. A greatly reduced muscle mass is characteristic of severe wasting, but there is indirect evidence that it also occurs in stunting. A reduced muscle mass increases the risk of death during infections and also in many other different pathological situations. Reduced muscle mass may represent a common mechanism linking wasting and stunting with increased mortality. This suggests that to decrease malnutrition-related mortality, interventions should aim at preventing both wasting and stunting, which often share common causes. Also, this suggests that treatment interventions should focus on children who are both wasted and stunted and therefore have the greatest deficits in muscle mass, instead of focusing on one or the other form of malnutrition. Interventions should also focus on young infants and children, who have a low muscle mass in relation to body weight to start with. Using mid-upper-arm circumference (MUAC) to select children in need of treatment may represent a simple way to target young wasted and stunted children efficiently in situations where these two conditions are present. Wasting is also associated with decreased fat mass. A decreased fat mass is frequent but inconsistent in stunting. Fat secretes multiple hormones, including leptin, which may have a stimulating effect on the immune system. Depressed immunity resulting from low fat stores may also contribute to the increased mortality observed in wasting. This may represent another common mechanism linking wasting and stunting with increased mortality in situations where stunting is associated with reduced fat mass. Leptin may also have an effect on bone growth. This may explain why wasted children with low fat stores have reduced linear growth when their weight-for-height remains low. It may also explain the frequent association of stunting with previous episodes of wasting. Stunting, however, can occur in the absence of wasting and even in overweight children. Thus, food supplementation should be used with caution in populations where stunting is not associated with wasting and low fat stores.
Collapse
|
26
|
Gat-Yablonski G, Phillip M. Nutritionally-induced catch-up growth. Nutrients 2015; 7:517-51. [PMID: 25594438 PMCID: PMC4303852 DOI: 10.3390/nu7010517] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Accepted: 12/31/2014] [Indexed: 12/17/2022] Open
Abstract
Malnutrition is considered a leading cause of growth attenuation in children. When food is replenished, spontaneous catch-up (CU) growth usually occurs, bringing the child back to its original growth trajectory. However, in some cases, the CU growth is not complete, leading to a permanent growth deficit. This review summarizes our current knowledge regarding the mechanism regulating nutrition and growth, including systemic factors, such as insulin, growth hormone, insulin- like growth factor-1, vitamin D, fibroblast growth factor-21, etc., and local mechanisms, including autophagy, as well as regulators of transcription, protein synthesis, miRNAs and epigenetics. Studying the molecular mechanisms regulating CU growth may lead to the establishment of better nutritional and therapeutic regimens for more effective CU growth in children with malnutrition and growth abnormalities. It will be fascinating to follow this research in the coming years and to translate the knowledge gained to clinical benefit.
Collapse
Affiliation(s)
- Galia Gat-Yablonski
- The Jesse Z and Sara Lea Shafer Institute for Endocrinology and Diabetes, National Center for Children's Diabetes, Schneider Children's Medical Center of Israel, and Felsenstein Medical Research Center, Petach Tikva 49100, and Sackler School of Medicine, Tel Aviv University, Tel Aviv, 69978, Israel.
| | - Moshe Phillip
- The Jesse Z and Sara Lea Shafer Institute for Endocrinology and Diabetes, National Center for Children's Diabetes, Schneider Children's Medical Center of Israel, and Felsenstein Medical Research Center, Petach Tikva 49100, and Sackler School of Medicine, Tel Aviv University, Tel Aviv, 69978, Israel.
| |
Collapse
|
27
|
Pando R, Masarwi M, Shtaif B, Idelevich A, Monsonego-Ornan E, Shahar R, Phillip M, Gat-Yablonski G. Bone quality is affected by food restriction and by nutrition-induced catch-up growth. J Endocrinol 2014; 223:227-39. [PMID: 25248555 DOI: 10.1530/joe-14-0486] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Growth stunting constitutes the most common effect of malnutrition. When the primary cause of malnutrition is resolved, catch-up (CU) growth usually occurs. In this study, we have explored the effect of food restriction (RES) and refeeding on bone structure and mechanical properties. Sprague-Dawley male rats aged 24 days were subjected to 10 days of 40% RES, followed by refeeding for 1 (CU) or 26 days long-term CU (LTCU). The rats fed ad libitum served as controls. The growth plates were measured, osteoclasts were identified using tartrate-resistant acid phosphatase staining, and micro-computed tomography (CT) scanning and mechanical testing were used to study structure and mechanical properties. Micro-CT analysis showed that RES led to a significant reduction in trabecular BV/TV and trabecular number (Tb.N), concomitant with an increase in trabecular separation (Tb.Sp). Trabecular BV/TV and Tb.N were significantly greater in the CU group than in the RES in both short- and long-term experiments. Mechanical testing showed that RES led to weaker and less compliant bones; interestingly, bones of the CU group were also more fragile after 1 day of CU. Longer term of refeeding enabled correction of the bone parameters; however, LTCU did not achieve full recovery. These results suggest that RES in young rats attenuated growth and reduced trabecular bone parameters. While nutrition-induced CU growth led to an immediate increase in epiphyseal growth plate height and active bone modeling, it was also associated with a transient reduction in bone quality. This should be taken into consideration when treating children undergoing CU growth.
Collapse
Affiliation(s)
- Rakefet Pando
- Felsenstein Medical Research CenterPetach Tikva, IsraelSackler Faculty of MedicineTel Aviv University, Tel Aviv, IsraelRobert H. Smith Faculty of AgricultureFood and Environment, Institute of Biochemistry and NutritionFaculty of AgriculturalFood and Environmental Quality Sciences, Koret School of Veterinary Medicine, The Hebrew University of Jerusalem, Rehovot, IsraelThe Jesse Z and Sara Lea Shafer Institute for Endocrinology and DiabetesNational Center for Childhood Diabetes, Schneider Children's Medical Center of Israel, 14 Kaplan Street, Petach Tikva 49202, Israel Felsenstein Medical Research CenterPetach Tikva, IsraelSackler Faculty of MedicineTel Aviv University, Tel Aviv, IsraelRobert H. Smith Faculty of AgricultureFood and Environment, Institute of Biochemistry and NutritionFaculty of AgriculturalFood and Environmental Quality Sciences, Koret School of Veterinary Medicine, The Hebrew University of Jerusalem, Rehovot, IsraelThe Jesse Z and Sara Lea Shafer Institute for Endocrinology and DiabetesNational Center for Childhood Diabetes, Schneider Children's Medical Center of Israel, 14 Kaplan Street, Petach Tikva 49202, Israel
| | - Majdi Masarwi
- Felsenstein Medical Research CenterPetach Tikva, IsraelSackler Faculty of MedicineTel Aviv University, Tel Aviv, IsraelRobert H. Smith Faculty of AgricultureFood and Environment, Institute of Biochemistry and NutritionFaculty of AgriculturalFood and Environmental Quality Sciences, Koret School of Veterinary Medicine, The Hebrew University of Jerusalem, Rehovot, IsraelThe Jesse Z and Sara Lea Shafer Institute for Endocrinology and DiabetesNational Center for Childhood Diabetes, Schneider Children's Medical Center of Israel, 14 Kaplan Street, Petach Tikva 49202, Israel Felsenstein Medical Research CenterPetach Tikva, IsraelSackler Faculty of MedicineTel Aviv University, Tel Aviv, IsraelRobert H. Smith Faculty of AgricultureFood and Environment, Institute of Biochemistry and NutritionFaculty of AgriculturalFood and Environmental Quality Sciences, Koret School of Veterinary Medicine, The Hebrew University of Jerusalem, Rehovot, IsraelThe Jesse Z and Sara Lea Shafer Institute for Endocrinology and DiabetesNational Center for Childhood Diabetes, Schneider Children's Medical Center of Israel, 14 Kaplan Street, Petach Tikva 49202, Israel
| | - Biana Shtaif
- Felsenstein Medical Research CenterPetach Tikva, IsraelSackler Faculty of MedicineTel Aviv University, Tel Aviv, IsraelRobert H. Smith Faculty of AgricultureFood and Environment, Institute of Biochemistry and NutritionFaculty of AgriculturalFood and Environmental Quality Sciences, Koret School of Veterinary Medicine, The Hebrew University of Jerusalem, Rehovot, IsraelThe Jesse Z and Sara Lea Shafer Institute for Endocrinology and DiabetesNational Center for Childhood Diabetes, Schneider Children's Medical Center of Israel, 14 Kaplan Street, Petach Tikva 49202, Israel Felsenstein Medical Research CenterPetach Tikva, IsraelSackler Faculty of MedicineTel Aviv University, Tel Aviv, IsraelRobert H. Smith Faculty of AgricultureFood and Environment, Institute of Biochemistry and NutritionFaculty of AgriculturalFood and Environmental Quality Sciences, Koret School of Veterinary Medicine, The Hebrew University of Jerusalem, Rehovot, IsraelThe Jesse Z and Sara Lea Shafer Institute for Endocrinology and DiabetesNational Center for Childhood Diabetes, Schneider Children's Medical Center of Israel, 14 Kaplan Street, Petach Tikva 49202, Israel
| | - Anna Idelevich
- Felsenstein Medical Research CenterPetach Tikva, IsraelSackler Faculty of MedicineTel Aviv University, Tel Aviv, IsraelRobert H. Smith Faculty of AgricultureFood and Environment, Institute of Biochemistry and NutritionFaculty of AgriculturalFood and Environmental Quality Sciences, Koret School of Veterinary Medicine, The Hebrew University of Jerusalem, Rehovot, IsraelThe Jesse Z and Sara Lea Shafer Institute for Endocrinology and DiabetesNational Center for Childhood Diabetes, Schneider Children's Medical Center of Israel, 14 Kaplan Street, Petach Tikva 49202, Israel
| | - Efrat Monsonego-Ornan
- Felsenstein Medical Research CenterPetach Tikva, IsraelSackler Faculty of MedicineTel Aviv University, Tel Aviv, IsraelRobert H. Smith Faculty of AgricultureFood and Environment, Institute of Biochemistry and NutritionFaculty of AgriculturalFood and Environmental Quality Sciences, Koret School of Veterinary Medicine, The Hebrew University of Jerusalem, Rehovot, IsraelThe Jesse Z and Sara Lea Shafer Institute for Endocrinology and DiabetesNational Center for Childhood Diabetes, Schneider Children's Medical Center of Israel, 14 Kaplan Street, Petach Tikva 49202, Israel
| | - Ron Shahar
- Felsenstein Medical Research CenterPetach Tikva, IsraelSackler Faculty of MedicineTel Aviv University, Tel Aviv, IsraelRobert H. Smith Faculty of AgricultureFood and Environment, Institute of Biochemistry and NutritionFaculty of AgriculturalFood and Environmental Quality Sciences, Koret School of Veterinary Medicine, The Hebrew University of Jerusalem, Rehovot, IsraelThe Jesse Z and Sara Lea Shafer Institute for Endocrinology and DiabetesNational Center for Childhood Diabetes, Schneider Children's Medical Center of Israel, 14 Kaplan Street, Petach Tikva 49202, Israel
| | - Moshe Phillip
- Felsenstein Medical Research CenterPetach Tikva, IsraelSackler Faculty of MedicineTel Aviv University, Tel Aviv, IsraelRobert H. Smith Faculty of AgricultureFood and Environment, Institute of Biochemistry and NutritionFaculty of AgriculturalFood and Environmental Quality Sciences, Koret School of Veterinary Medicine, The Hebrew University of Jerusalem, Rehovot, IsraelThe Jesse Z and Sara Lea Shafer Institute for Endocrinology and DiabetesNational Center for Childhood Diabetes, Schneider Children's Medical Center of Israel, 14 Kaplan Street, Petach Tikva 49202, Israel Felsenstein Medical Research CenterPetach Tikva, IsraelSackler Faculty of MedicineTel Aviv University, Tel Aviv, IsraelRobert H. Smith Faculty of AgricultureFood and Environment, Institute of Biochemistry and NutritionFaculty of AgriculturalFood and Environmental Quality Sciences, Koret School of Veterinary Medicine, The Hebrew University of Jerusalem, Rehovot, IsraelThe Jesse Z and Sara Lea Shafer Institute for Endocrinology and DiabetesNational Center for Childhood Diabetes, Schneider Children's Medical Center of Israel, 14 Kaplan Street, Petach Tikva 49202, Israel Felsenstein Medical Research CenterPetach Tikva, IsraelSackler Faculty of MedicineTel Aviv University, Tel Aviv, IsraelRobert H. Smith Faculty of AgricultureFood and Environment, Institute of Biochemistry and NutritionFaculty of AgriculturalFood and Environmental Quality Sciences, Koret School of Veterinary Medicine, The Hebrew University of Jerusalem, Rehovot, IsraelThe Jesse Z and Sara Lea Shafer Institute for Endocrinology and DiabetesNational Center for Childhood Diabetes, Schneider Children's Medical Center of Israel, 14 Kaplan Street, Petach Tikva 49202, Israel
| | - Galia Gat-Yablonski
- Felsenstein Medical Research CenterPetach Tikva, IsraelSackler Faculty of MedicineTel Aviv University, Tel Aviv, IsraelRobert H. Smith Faculty of AgricultureFood and Environment, Institute of Biochemistry and NutritionFaculty of AgriculturalFood and Environmental Quality Sciences, Koret School of Veterinary Medicine, The Hebrew University of Jerusalem, Rehovot, IsraelThe Jesse Z and Sara Lea Shafer Institute for Endocrinology and DiabetesNational Center for Childhood Diabetes, Schneider Children's Medical Center of Israel, 14 Kaplan Street, Petach Tikva 49202, Israel Felsenstein Medical Research CenterPetach Tikva, IsraelSackler Faculty of MedicineTel Aviv University, Tel Aviv, IsraelRobert H. Smith Faculty of AgricultureFood and Environment, Institute of Biochemistry and NutritionFaculty of AgriculturalFood and Environmental Quality Sciences, Koret School of Veterinary Medicine, The Hebrew University of Jerusalem, Rehovot, IsraelThe Jesse Z and Sara Lea Shafer Institute for Endocrinology and DiabetesNational Center for Childhood Diabetes, Schneider Children's Medical Center of Israel, 14 Kaplan Street, Petach Tikva 49202, Israel Felsenstein Medical Research CenterPetach Tikva, IsraelSackler Faculty of MedicineTel Aviv University, Tel Aviv, IsraelRobert H. Smith Faculty of AgricultureFood and Environment, Institute of Biochemistry and NutritionFaculty of AgriculturalFood and Environmental Quality Sciences, Koret School of Veterinary Medicine, The Hebrew University of Jerusalem, Rehovot, IsraelThe Jesse Z and Sara Lea Shafer Institute for Endocrinology and DiabetesNational Center for Childhood Diabetes, Schneider Children's Medical Center of Israel, 14 Kaplan Street, Petach Tikva 49202, Israel
| |
Collapse
|
28
|
FTO is a relevant factor for the development of the metabolic syndrome in mice. PLoS One 2014; 9:e105349. [PMID: 25144618 PMCID: PMC4140775 DOI: 10.1371/journal.pone.0105349] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Accepted: 07/23/2014] [Indexed: 12/19/2022] Open
Abstract
The metabolic syndrome is a worldwide problem mainly caused by obesity. FTO was found to be a obesity-risk gene in humans and FTO deficiency in mice led to reduction in adipose tissue. Thus, FTO is an important factor for the development of obesity. Leptin-deficient mice are a well characterized model for analysing the metabolic syndrome. To determine the relevance of FTO for the development of the metabolic syndrome we analysed different parameters in combined homozygous deficient mice (Lep(ob/ob);Fto(-/-)). Lep(ob/ob);Fto(-/-) mice showed an improvement in analysed hallmarks of the metabolic syndrome in comparison to leptin-deficient mice wild type or heterozygous for Fto. Lep(ob/ob);Fto(-/-) mice did not develop hyperglycaemia and showed an improved glucose tolerance. Furthermore, extension of beta-cell mass was prevented in Lep(ob/ob);Fto(-/-)mice and accumulation of ectopic fat in the liver was reduced. In conclusion this study demonstrates that FTO deficiency has a protective effect not only on the development of obesity but also on the metabolic syndrome. Thus, FTO plays an important role in the development of metabolic disorders and is an interesting target for therapeutic agents.
Collapse
|
29
|
Briend A. Retards de croissance d’origine nutritionnelle Quelles données et quelles solutions en 2014 ? Arch Pediatr 2014. [DOI: 10.1016/s0929-693x(14)71480-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
30
|
Pando R, Shtaif B, Phillip M, Gat-Yablonski G. A serum component mediates food restriction-induced growth attenuation. Endocrinology 2014; 155:932-40. [PMID: 24456162 DOI: 10.1210/en.2013-1610] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Proper nutrition in terms of calories and essential food components is required to maximize longitudinal growth in children. Our previous study showed that prepubertal male rats subjected to 10 days of 40% food restriction (RES) exhibited a dramatic reduction in weight and epiphyseal growth plate height, as well as changes in gene expression and microRNAs (miRNAs) in the epiphyseal growth plate. These findings reversed rapidly after renewal of the regular food supply (catch-up [CU]). To further elucidate the mechanisms underlying the nutrition-growth association, serum collected from the RES and CU rats and control rats fed ad libitum (AL) was added to the culture medium of the chondrocyte cell line ATDC5 (instead of fetal calf serum). Serum from the RES group induced a reduction in cell viability (25%, P < .05) concomitant with an increase in cell differentiation compared with that for the AL group serum. The most interesting observation, in our opinion, was the significant reduction in the expression of specific miRNAs, including the chondro-specific miR-140. These effects were not observed for serum from refed (CU) rats. Serum levels of IGF-I, leptin, and fibroblast growth factor 21 were reduced by food restriction. The addition of IGF-I and leptin to the culture increased cell viability, whereas fibroblast growth factor 21 reduced it, suggesting the involvement of IGF-I, leptin, and possibly other still unidentified serum factors in chondrocyte cell growth. In conclusion, specific miRNAs respond to nutritional cues, and these effects are mediated by serum-borne factors. These results may promote the development of superior interventions for children with malnutrition and growth abnormalities.
Collapse
Affiliation(s)
- Rakefet Pando
- Sackler School of Medicine (R.P., B.S., M.P., G.G.-Y.), Tel Aviv University, Tel Aviv 6997801, Israel; Felsentein Medical Research Center (B.S., M.P., G.G.-Y.), Petach Tikva 4945102, Israel; and The Jesse Z. and Sara Lea Shafer Institute for Endocrinology and Diabetes (M.P., G.G.-Y.), National Center for Childhood Diabetes, Schneider Children's Medical Center of Israel, Petach Tikva 4920235, Israel
| | | | | | | |
Collapse
|
31
|
Lee HJ, Kim H, Ku SY, Choi YM, Kim JH, Kim JG. Association between polymorphisms in leptin, leptin receptor, and β-adrenergic receptor genes and bone mineral density in postmenopausal Korean women. Menopause 2014; 21:67-73. [PMID: 23760429 DOI: 10.1097/gme.0b013e31829366ed] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
OBJECTIVE The purpose of this study was to investigate the association between single nucleotide polymorphisms in leptin (LEP), leptin receptor (LEPR), and β-adrenergic receptor (ADRB) genes and bone mineral density (BMD) in postmenopausal Korean women. METHODS LEP c.280G>A, LEPR c.326A>G, LEPR c.668A>G, LEPR c.1968G>C, LEPR c.2096C>T, ADRB2 c.46A>G, ADRB2 c.79C>G, ADRB2 c.718T>C, ADRB2 c.741G>T, ADRB2 c.769G>A, and ADRB3 c.190T>C polymorphisms were analyzed in 592 postmenopausal Korean women. Serum levels of leptin, soluble leptin receptor, osteoprotegerin, soluble receptor activator of the nuclear factor-κB ligand, bone alkaline phosphatase, and carboxy-terminal telopeptide of type I collagen were measured, and BMDs at the lumbar spine and femoral neck were also examined. RESULTS Among the polymorphisms measured, only the LEPR c.1968G>C polymorphism was found to be associated with BMD at the femoral neck, and higher BMD was observed with increasing number of G alleles (P = 0.04). Osteoporosis at the femoral neck was 3.27 and 3.89 times more frequently observed in the AG and GG genotypes than in the AA genotype in the ADRB2 c.46A>G polymorphism (P = 0.024 and P = 0.015, respectively). However, no significant differences in serum levels of leptin, soluble leptin receptor, free leptin index, osteoprotegerin, soluble receptor activator of the nuclear factor-κB ligand, and bone turnover markers were detected among single and haplotype genotypes. CONCLUSIONS These results suggest that the LEPR c.1968G>C polymorphism may be one of the genetic factors affecting femoral neck BMD in postmenopausal Korean women and that an analysis of the ADRB2 c.46A>G polymorphism may be useful in identifying women at risk for osteoporosis at the femoral neck.
Collapse
Affiliation(s)
- Hee Jun Lee
- From the 1Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Korea; 2Clinical Research Institute, Seoul National University Hospital, Seoul, Korea; and 3Department of Anesthesiology and Pain Medicine, School of Medicine, Ewha Woman's University, Seoul, Korea
| | | | | | | | | | | |
Collapse
|
32
|
Martín AE, del R Pani M, Holgado NR, López Miranda LI, Meheris HE, Garat JA. Facial development disorders due to inhibition to endochondral ossification of mandibular condyle process caused by malnutrition. Angle Orthod 2013; 84:473-8. [PMID: 24160994 DOI: 10.2319/042013-307.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
OBJECTIVE To analyze the effect of protein restriction on histomorphometric parameters of bone remodeling in mandibular condyle process and its possible influence in facial development in growing rats. MATERIALS AND METHODS Wistar rats weaned at the age of 21 days were assigned to one of the following groups: control (fed a regular hard diet ad libitum) and protein restricted (PR) (fed a hard diet lacking in protein ad libitum). The animals were euthanized on day 35 after the onset of the experiment. Mandibles were resected, fixed in 10% formalin, hemisected at the symphysis, and then radiographed in order to perform cephalometric studies of the condylar process length and the height of the lower alveolar process. Mandibles were then processed for light microscopy, and histomorphometric determinations were performed on histologic sections of the condylar process subchondral bone. RESULTS The PR group showed a significantly lower body weight than control group at the end of the experiment. The length of the condylar process was lower in the PR group; however, the diet used in this study did not affect the height of the lower alveolar process. The histomorphometric analysis showed that the PR group exhibited a statistically significant decrease in bone formation and bone volume in condylar process subchondral bone. CONCLUSION Protein restriction inhibits bone formation and longitudinal growth in the mandibular condylar process. This result suggest that protein restriction can alter normal facial development.
Collapse
Affiliation(s)
- Adrián E Martín
- a Assistant Professor, Department of Histology, School of Dentistry, National University of Tucumán, Tucumán, Argentina
| | | | | | | | | | | |
Collapse
|
33
|
Turner RT, Kalra SP, Wong CP, Philbrick KA, Lindenmaier LB, Boghossian S, Iwaniec UT. Peripheral leptin regulates bone formation. J Bone Miner Res 2013; 28:22-34. [PMID: 22887758 PMCID: PMC3527690 DOI: 10.1002/jbmr.1734] [Citation(s) in RCA: 184] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2012] [Revised: 06/30/2012] [Accepted: 07/16/2012] [Indexed: 12/15/2022]
Abstract
Substantial evidence does not support the prevailing view that leptin, acting through a hypothalamic relay, decreases bone accrual by inhibiting bone formation. To clarify the mechanisms underlying regulation of bone architecture by leptin, we evaluated bone growth and turnover in wild-type (WT) mice, leptin receptor-deficient db/db mice, leptin-deficient ob/ob mice, and ob/ob mice treated with leptin. We also performed hypothalamic leptin gene therapy to determine the effect of elevated hypothalamic leptin levels on osteoblasts. Finally, to determine the effects of loss of peripheral leptin signaling on bone formation and energy metabolism, we used bone marrow (BM) from WT or db/db donor mice to reconstitute the hematopoietic and mesenchymal stem cell compartments in lethally irradiated WT recipient mice. Decreases in bone growth, osteoblast-lined bone perimeter and bone formation rate were observed in ob/ob mice and greatly increased in ob/ob mice following subcutaneous administration of leptin. Similarly, hypothalamic leptin gene therapy increased osteoblast-lined bone perimeter in ob/ob mice. In spite of normal osteoclast-lined bone perimeter, db/db mice exhibited a mild but generalized osteopetrotic-like (calcified cartilage encased by bone) skeletal phenotype and greatly reduced serum markers of bone turnover. Tracking studies and histology revealed quantitative replacement of BM cells following BM transplantation. WT mice engrafted with db/db BM did not differ in energy homeostasis from untreated WT mice or WT mice engrafted with WT BM. Bone formation in WT mice engrafted with WT BM did not differ from WT mice, whereas bone formation in WT mice engrafted with db/db cells did not differ from the low rates observed in untreated db/db mice. In summary, our results indicate that leptin, acting primarily through peripheral pathways, increases osteoblast number and activity.
Collapse
Affiliation(s)
- Russell T. Turner
- Skeletal Biology Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR, 97331, USA
| | - Satya P. Kalra
- Department of Neuroscience, University of Florida McKnight Brain Institute, Gainesville, FL, 32610, USA
| | - Carmen P. Wong
- Molecular and Cellular Nutrition Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR, 97331, USA
| | - Kenneth A. Philbrick
- Skeletal Biology Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR, 97331, USA
| | - Laurence B. Lindenmaier
- Skeletal Biology Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR, 97331, USA
| | - Stephane Boghossian
- Department of Neuroscience, University of Florida McKnight Brain Institute, Gainesville, FL, 32610, USA
| | - Urszula T. Iwaniec
- Skeletal Biology Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR, 97331, USA
| |
Collapse
|
34
|
Baek K, Bloomfield SA. Blocking β-adrenergic signaling attenuates reductions in circulating leptin, cancellous bone mass, and marrow adiposity seen with dietary energy restriction. J Appl Physiol (1985) 2012; 113:1792-801. [PMID: 22995391 DOI: 10.1152/japplphysiol.00187.2012] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
We tested whether β-adrenergic blockade attenuates bone loss and increased marrow adiposity during energy restriction (ER) and whether such an effect is associated with changes in serum leptin and leptin expression in bone and marrow tissues. Female 4-mo-old Sprague-Dawley rats were assigned into four groups (n = 10 each): two groups of 40% ER treated with vehicle (ERVEH; saline) or β-blocker (ERBB; DL-propranolol; 250 μg · kg(-1) · h(-1)) during 12 wk, and two groups of ad libitum-fed controls treated with the same two agents (CONVEH, CONBB, respectively). Over 84 days, CONVEH and CONBB rats gained but ERVEH and ERBB rats lost body fat mass; lean mass did not change in any group. Reduction in serum leptin in ERVEH rats was mitigated in ERBB rats (-5.32 vs. -1.15 ng/ml, respectively). The decline in proximal tibia cancellous vBMD observed in ERVEH rats was attenuated in ERBB rats (-85.24 vs. -53.94 mg/cm(3), respectively). Adipocyte number in ERVEH rats was dramatically higher vs. CON rats at week 12, but this increment was abolished by β-blockade in ERBB animals. The number of osteoblastic cells and marrow adipocytes staining positively for leptin in ERVEH rats tended to be lower vs. that of both CON groups, but β-blockade appears to reverse this effect in ERBB rats. In summary, β-adrenergic blockade mitigated metaphyseal bone loss and bone marrow adiposity during energy restriction and attenuated reductions in serum leptin. These data suggest an important role for β-adrenoreceptor signaling pathway in the cancellous bone and marrow fat response to energy restriction.
Collapse
Affiliation(s)
- Kyunghwa Baek
- Department of Health and Kinesiology and Intercollegiate Graduate Faculty of Nutrition, Texas A&M University, College Station, Texas 77843, USA
| | | |
Collapse
|
35
|
Modan-Moses D, Yaroslavsky A, Kochavi B, Toledano A, Segev S, Balawi F, Mitrany E, Stein D. Linear growth and final height characteristics in adolescent females with anorexia nervosa. PLoS One 2012; 7:e45504. [PMID: 23029058 PMCID: PMC3445517 DOI: 10.1371/journal.pone.0045504] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2012] [Accepted: 08/21/2012] [Indexed: 01/29/2023] Open
Abstract
OBJECTIVE Growth retardation is an established complication of anorexia nervosa (AN). However, findings concerning final height of AN patients are inconsistent. The aim of this study was to assess these phenomena in female adolescent inpatients with AN. METHODS We retrospectively studied all 211 female adolescent AN patients hospitalized in an inpatient eating disorders department from 1/1/1987 to 31/12/99. Height and weight were assessed at admission and thereafter routinely during hospitalization and follow-up. Final height was measured in 69 patients 2-10 years after discharge. Pre-morbid height data was available in 29 patients. RESULTS Patients' height standard deviation scores (SDS) on admission (-0.285±1.0) and discharge (-0.271±1.02) were significantly (p<0.001) lower than expected in normal adolescents. Patients admitted at age ≤13 years, or less than 1 year after menarche, were more severely growth-impaired than patients admitted at an older age, (p = 0.03). Final height SDS, available for 69 patients, was -0.258±1.04, significantly lower than expected in a normal population (p = 0.04), and was more severely compromised in patients who were admitted less than 1 year from their menarche. In a subgroup of 29 patients with complete growth data (pre-morbid, admission, discharge, and final adult height), the pre-morbid height SDS was not significantly different from the expected (-0.11±1.1), whereas heights at the other time points were significantly (p = 0.001) lower (-0.56±1.2, -0.52±1.2, and -0.6±1.2, respectively). CONCLUSIONS Our findings suggest that whereas the premorbid height of female adolescent AN patients is normal, linear growth retardation is a prominent feature of their illness. Weight restoration is associated with catch-up growth, but complete catch-up is often not achieved.
Collapse
Affiliation(s)
- Dalit Modan-Moses
- Pediatric Endocrinology and Diabetes Unit, The Edmond and Lily Safra Children's Hospital, The Chaim Sheba Medical Center, Tel-Hashomer, Ramat Gan, Israel.
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Carlton ED, Demas GE, French SS. Leptin, a neuroendocrine mediator of immune responses, inflammation, and sickness behaviors. Horm Behav 2012; 62:272-9. [PMID: 22561456 DOI: 10.1016/j.yhbeh.2012.04.010] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2011] [Revised: 04/05/2012] [Accepted: 04/18/2012] [Indexed: 01/27/2023]
Abstract
Effective immune responses are coordinated by interactions among the nervous, endocrine, and immune systems. Mounting immune, inflammatory, and sickness responses requires substantial energetic investments, and as such, an organism may need to balance energy allocation to these processes with the energetic demands of other competing physiological systems. The metabolic hormone leptin appears to be mediating trade-offs between the immune system and other physiological systems through its actions on immune cells and the brain. Here we review the evidence in both mammalian and non-mammalian vertebrates that suggests leptin is involved in regulating immune responses, inflammation, and sickness behaviors. Leptin has also been implicated in the regulation of seasonal immune responses, including sickness; however, the precise physiological mechanisms remain unclear. Thus, we discuss recent data in support of leptin as a mediator of seasonal sickness responses and provide a theoretical model that outlines how seasonal cues, leptin, and proinflammatory cytokines may interact to coordinate seasonal immune and sickness responses.
Collapse
Affiliation(s)
- Elizabeth D Carlton
- Department of Biology, Program in Neuroscience and Center for the Integrative Study of Animal Behavior, Indiana University, Bloomington, IN 47405, USA.
| | | | | |
Collapse
|
37
|
Stunes AK, Westbroek I, Gordeladze JO, Gustafsson BI, Reseland JE, Syversen U. Systemic leptin administration in supraphysiological doses maintains bone mineral density and mechanical strength despite significant weight loss. Endocrinology 2012; 153:2245-53. [PMID: 22374968 DOI: 10.1210/en.2011-1848] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The effects of leptin on bone are controversial. Although in vitro studies have shown that leptin stimulates osteoblast differentiation and mineralization and inhibits osteoclastogenesis, some rodent studies have shown that leptin administered centrally might result in decreased bone formation. In the present study we have investigated the skeletal effects of supraphysiological concentrations of leptin administered sc to rats. Female Fischer rats were given leptin 100 μg/d, 200 μg/d, or saline by continuous infusion for 9 wk. Bone mineral density (BMD) was measured by dual energy x-ray absorptiometry, bone microarchitecture was analyzed by micro-computed tomography, and biomechanical properties were tested by three-point bending experiments. At the end of the study, the body weight was significantly lower in rats receiving leptin compared with controls (-10.8% and -12.0% in low- and high-dose leptin groups, respectively). The high-dose leptin group also significantly lost weight compared with baseline. The plasma leptin concentration was 14- and 33-fold increased in the low- and high-dose groups, respectively. No significant differences in femoral BMD were observed. Whole-body BMD was significantly lower in the low-dose leptin group, whereas there was no difference between the high-dose leptin group and the control. Mechanical strength and microarchitecture were similar in the high-dose and the control group. The low-dose group, however, had decreased cortical volume in the femoral metaphysis, lowered bone strength, and altered moment of inertia. In conclusion, leptin given at very high doses maintains BMD, microarchitecture, and mechanical strength in female rats, despite a significant decrease in body weight.
Collapse
Affiliation(s)
- Astrid K Stunes
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, N-7491 Trondheim, Norway.
| | | | | | | | | | | |
Collapse
|
38
|
Pando R, Even-Zohar N, Shtaif B, Edry L, Shomron N, Phillip M, Gat-Yablonski G. MicroRNAs in the growth plate are responsive to nutritional cues: association between miR-140 and SIRT1. J Nutr Biochem 2012; 23:1474-81. [PMID: 22402365 DOI: 10.1016/j.jnutbio.2011.09.010] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2011] [Revised: 07/17/2011] [Accepted: 09/27/2011] [Indexed: 12/18/2022]
Abstract
MicroRNAs (miRNAs) have been reported to be involved in a variety of functions, including skeletal development and longitudinal growth. The aim of this study was to investigate the role of miRNAs in food-restriction-induced growth attenuation and nutrition-induced catch-up growth in the epiphyseal growth plate (EGP). Prepubertal rats were fed ad libitum or were subjected to 40% food restriction for 10 days followed by a renewal of the regular food supply. At sacrifice, tibial EGPs were excised, and the total RNA was extracted and loaded on miRNA microarrays. The miRNA microarray yielded more than 400 miRNAs that are expressed in the EGP of mature animals. Results were confirmed by quantitative polymerase chain reaction. Chondrocyte-specific miR-140-3p showed the highest expression in the mature EGP, and it was one of the few miRNAs that were significantly reduced following nutrition restriction. Changes in predicted miRNA targets were then followed with Western immunoblotting. Direct binding was demonstrated using exogenous miRNA, the 3'UTR of the target mRNA and a luciferase reporter assay. Nutrition restriction induced an increase in the level of the miR-140-3p target, NAD+-dependent SIRT1. This study is the first to show that SIRT1 and miRNAs expressed in the mature EGP are responsive to nutritional cues. Nutrition-induced epigenetic regulation of growth activates two parts of the epigenetic world - miRNAs and histone deacetylases - that are interconnected. Deciphering the role of epigenetic regulation in growth may open a new era of research and pave the way for the development of new treatments for children with growth disorders.
Collapse
Affiliation(s)
- Rakefet Pando
- Felsenstein Medical Research Center, Petach Tikva, National Center for Childhood Diabetes, Schneider Children's Medical Center of Israel, Petach Tikva, Israel
| | | | | | | | | | | | | |
Collapse
|
39
|
Burgos-Ramos E, González-Rodríguez A, Canelles S, Baquedano E, Frago LM, Revuelta-Cervantes J, Gómez-Ambrosi J, Frühbeck G, Chowen JA, Argente J, Valverde AM, Barrios V. Differential insulin receptor substrate-1 (IRS1)-related modulation of neuropeptide Y and proopiomelanocortin expression in nondiabetic and diabetic IRS2-/- mice. Endocrinology 2012; 153:1129-40. [PMID: 22210743 DOI: 10.1210/en.2011-1278] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Insulin resistance and type 2 diabetes correlate with impaired leptin and insulin signaling. Insulin receptor substrate-2 deficient (IRS2(-/-)) mice are an accepted model for the exploration of alterations in these signaling pathways and their relationship with diabetes; however, disturbances in hypothalamic signaling and the effect on neuropeptides controlling food intake remain unclear. Our aim was to analyze how leptin and insulin signaling may differentially affect the expression of hypothalamic neuropeptides regulating food intake and hypothalamic inflammation in diabetic (D) and nondiabetic (ND) IRS2(-/-) mice. We analyzed the activation of leptin and insulin targets by Western blotting and their association by immunoprecipitation, as well as the mRNA levels of neuropeptide Y (NPY), proopiomelanocortin, and inflammatory markers by real-time PCR and colocalization of forkhead box protein O1 (FOXO1) and NPY by double immunohistochemistry in the hypothalamus. Serum leptin and insulin levels and hypothalamic Janus kinase 2 and signal transducer and activator of transcription factor 3 activation were increased in ND IRS2(-/-) mice. IRS1 levels and its association with Janus kinase 2 and p85 and protein kinase B activation were increased in ND IRS2(-/-). Increased FOXO1 positively correlated with NPY mRNA levels in D IRS2(-/-) mice, with FOXO1 showing mainly nuclear localization in D IRS2(-/-) and cytoplasmic in ND IRS2(-/-) mice. D IRS2(-/-) mice exhibited higher hypothalamic inflammation markers than ND IRS2(-/-) mice. In conclusion, differential activation of these pathways and changes in the expression of NPY and inflammation may exert a protective effect against hypothalamic deregulation of appetite, suggesting that manipulation of these targets could be of interest in the treatment of insulin resistance and type 2 diabetes.
Collapse
Affiliation(s)
- Emma Burgos-Ramos
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Avda. Menéndez Pelayo, 65; E-28009 Madrid, Spain
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Gat-Yablonski G, Yackobovitch-Gavan M, Phillip M. Nutrition and bone growth in pediatrics. Pediatr Clin North Am 2011; 58:1117-40, ix. [PMID: 21981952 DOI: 10.1016/j.pcl.2011.07.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Children's growth is a hallmark of their normal development and the association between nutrition and linear growth in children is well accepted. Growth requires an adequate supply of many different nutritional factors, some form the "building materials," whereas others play regulatory roles. In this article we describe the growth of the growth plate and discuss the role of nutritional affected hormones on this process. In addition we describe the effect of local regulators and nutritional factors on the growth process and suggest the involvement of new regulatory factors in the translation of nutrition to growth.
Collapse
Affiliation(s)
- Galia Gat-Yablonski
- The Jesse Z. and Sara Lea Shafer Institute for Endocrinology and Diabetes, National Center for Childhood Diabetes, Schneider Children's Medical Center of Israel, 14 Kaplan Street, Petah Tikva 49202, Israel
| | | | | |
Collapse
|
41
|
Bar-El Dadon S, Shahar R, Katalan V, Monsonego-Ornan E, Reifen R. Leptin administration affects growth and skeletal development in a rat intrauterine growth restriction model: Preliminary study. Nutrition 2011; 27:973-7. [DOI: 10.1016/j.nut.2010.10.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2010] [Revised: 10/25/2010] [Accepted: 10/26/2010] [Indexed: 01/21/2023]
|
42
|
|
43
|
Wang L, Shao YY, Ballock RT. Leptin synergizes with thyroid hormone signaling in promoting growth plate chondrocyte proliferation and terminal differentiation in vitro. Bone 2011; 48:1022-7. [PMID: 21349356 DOI: 10.1016/j.bone.2011.02.012] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2010] [Revised: 02/14/2011] [Accepted: 02/15/2011] [Indexed: 12/22/2022]
Abstract
Leptin and thyroid hormone are two hormones that regulate energy balance through central signaling mechanisms. Recent studies in leptin-deficient ob/ob mice indicate that leptin also has peripheral effects in modulating the function of the growth plate, perhaps in terms of proliferation and differentiation enhancement. Thyroid hormone has been well-described as a potent stimulator of growth plate chondrocyte maturation. The objective of this study was therefore to investigate the interaction between leptin and thyroid hormone signaling in growth plate chondrocyte proliferation and terminal differentiation. Our in vitro data demonstrate that leptin synergistically functions with thyroid hormone through activation of both IGF-1/IGF1R signaling and Wnt/β-catenin signaling, two pathways that have been previously described as downstream effectors of thyroid hormone action. Leptin increases thyroid hormone receptor-α (TRα) expression and thyroid hormone receptor transcriptional activity. Thyroid hormone also activates leptin signaling in growth plate cells undergoing proliferation and hypertrophy. We conclude that leptin synergically interacts with thyroid hormone in promoting growth plate chondrocyte proliferation and terminal differentiation.
Collapse
Affiliation(s)
- Lai Wang
- Department of Biomedical Engineering, The Lerner Research Institute, The Cleveland Clinic Foundation, Cleveland, OH, USA.
| | | | | |
Collapse
|
44
|
Palsdottir V, Wickman A, Andersson N, Hezaveh R, Olsson B, Gabrielsson BG, Strandvik B. Postnatal deficiency of essential fatty acids in mice results in resistance to diet-induced obesity and low plasma insulin during adulthood. Prostaglandins Leukot Essent Fatty Acids 2011; 84:85-92. [PMID: 21177089 DOI: 10.1016/j.plefa.2010.11.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2010] [Revised: 11/16/2010] [Accepted: 11/17/2010] [Indexed: 01/21/2023]
Abstract
Our objective was to investigate the long-term metabolic effects of postnatal essential fatty acid deficiency (EFAD). Mouse dams were fed an EFAD diet or an isoenergetic control diet 4 days before delivery and throughout lactation. The pups were weaned to standard diet (STD) and were later subdivided into two groups: receiving high fat diet (HFD) or STD. Body composition, energy expenditure, food intake and leptin levels were analyzed in adult offspring. Blood glucose and plasma insulin concentrations were measured before and during a glucose tolerance test. EFAD offspring fed STD were leaner with lower plasma leptin and insulin concentrations compared to controls. EFAD offspring fed HFD were resistant to diet-induced obesity, had higher energy expenditure and lower levels of plasma leptin and insulin compared to controls. These results indicate that the fatty acid composition during lactation is important for body composition and glucose tolerance in the adult offspring.
Collapse
|
45
|
Lorentzon M, Norjavaara E, Kindblom JM. Pubertal timing predicts leg length and childhood body mass index predicts sitting height in young adult men. J Pediatr 2011; 158:452-7. [PMID: 20961561 DOI: 10.1016/j.jpeds.2010.09.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2010] [Revised: 08/12/2010] [Accepted: 09/02/2010] [Indexed: 01/26/2023]
Abstract
OBJECTIVE To investigate the impact of pubertal timing and childhood body mass index (BMI), both within normal range, on adult anthropometrics. STUDY DESIGN Detailed growth charts were retrieved for the men participating in the population-based Gothenburg Osteoporosis and Obesity Determinants study. Age at peak height velocity and childhood BMI were calculated (n = 527), and anthropometric measurements were performed. RESULTS Analysis of variance analysis of tertiles according to age at peak height velocity demonstrated that the early peak height velocity tertile had a lower adult height (180.9 ± 6.8 cm) compared with the middle tertile group (182.7 ± 6.9 cm, P < .05), and this difference was attributable to shorter leg length. No difference was seen for sitting height. In contrast, analysis of tertiles according to childhood BMI demonstrated low sitting height in the low BMI tertile (93.7 ± 3.3 cm for low, 94.6 ± 3.3, for middle, and 94.8 ± 3.3 cm for high childhood BMI tertiles, P < .05 and P < .01, respectively), but childhood BMI did not affect adult height and leg length. CONCLUSION We demonstrate that subjects with early pubertal timing have reduced adult height and leg length, and subjects with low childhood BMI have reduced adult sitting height. Thus childhood body composition and pubertal timing have different impact on trunk growth and growth of long bones.
Collapse
Affiliation(s)
- Mattias Lorentzon
- Center for Bone and Arthritis Research, Institute of Medicine, The Sahlgrenska Academy at Gothenburg University, Gothenburg, Sweden
| | | | | |
Collapse
|
46
|
Anorexia und Bulimia nervosa im Kindes- und Jugendalter. Monatsschr Kinderheilkd 2011. [DOI: 10.1007/s00112-010-2315-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
47
|
|
48
|
Devlin MJ, Cloutier AM, Thomas NA, Panus DA, Lotinun S, Pinz I, Baron R, Rosen CJ, Bouxsein ML. Caloric restriction leads to high marrow adiposity and low bone mass in growing mice. J Bone Miner Res 2010; 25:2078-88. [PMID: 20229598 PMCID: PMC3127399 DOI: 10.1002/jbmr.82] [Citation(s) in RCA: 253] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2009] [Revised: 02/11/2010] [Accepted: 03/04/2010] [Indexed: 12/12/2022]
Abstract
The effects of caloric restriction (CR) on the skeleton are well studied in adult rodents and include lower cortical bone mass but higher trabecular bone volume. Much less is known about how CR affects bone mass in young, rapidly growing animals. This is an important problem because low caloric intake during skeletal acquisition in humans, as in anorexia nervosa, is associated with low bone mass, increased fracture risk, and osteoporosis in adulthood. To explore this question, we tested the effect of caloric restriction on bone mass and microarchitecture during rapid skeletal growth in young mice. At 3 weeks of age, we weaned male C57Bl/6J mice onto 30% caloric restriction (10% kcal/fat) or normal diet (10% kcal/fat). Outcomes at 6 (n = 4/group) and 12 weeks of age (n = 8/group) included body mass, femur length, serum leptin and insulin-like growth factor 1 (IGF-1) values, whole-body bone mineral density (WBBMD, g/cm(2)), cortical and trabecular bone architecture at the midshaft and distal femur, bone formation and cellularity, and marrow fat measurement. Compared with the normal diet, CR mice had 52% and 88% lower serum leptin and 33% and 39% lower serum IGF-1 at 6 and 12 weeks of age (p < .05 for all). CR mice were smaller, with lower bone mineral density, trabecular, and cortical bone properties. Bone-formation indices were lower, whereas bone-resorption indices were higher (p < .01 for all) in CR versus normal diet mice. Despite having lower percent of body fat, bone marrow adiposity was elevated dramatically in CR versus normal diet mice (p < .05). Thus we conclude that caloric restriction in young, growing mice is associated with impaired skeletal acquisition, low leptin and IGF-1 levels, and high marrow adiposity. These results support the hypothesis that caloric restriction during rapid skeletal growth is deleterious to cortical and trabecular bone mass and architecture, in contrast to potential skeletal benefits of CR in aging animals.
Collapse
Affiliation(s)
- Maureen J Devlin
- Center for Advanced Orthopedic Studies, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Growth-plate cartilage in chronic renal failure. Pediatr Nephrol 2010; 25:643-9. [PMID: 19816714 DOI: 10.1007/s00467-009-1307-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2009] [Revised: 08/05/2009] [Accepted: 08/05/2009] [Indexed: 12/13/2022]
Abstract
Bone growth occurs in the growth-plate cartilage located at the ends of long bones. Changes in the architecture, abnormalities in matrix organization, reduction in protein staining and RNA expression of factors involved in cell signaling have been described in the growth-plate cartilage of nephrectomized animals. These changes can lead to a smaller growth plate associated with decrease in chondrocyte proliferation, delayed hypertrophy, and prolonged initiation of mineralization and vascular invasion. As a result, chronic renal failure can result in stunted body growth and skeletal deformities. Multiple etiologic factors can contribute to impaired bone growth in renal failure, including suboptimal nutrition, metabolic acidosis, and secondary hyperparathyroidism. Recent findings have also shown the tight connection between chondro/osteogenesis, hematopoiesis, and immunogenesis.
Collapse
|
50
|
Gat-Yablonski G, Yackobovitch-Gavan M, Phillip M. Nutrition and bone growth in pediatrics. Endocrinol Metab Clin North Am 2009; 38:565-86. [PMID: 19717005 DOI: 10.1016/j.ecl.2009.07.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Children's growth is a hallmark of their normal development and the association between nutrition and linear growth in children is well accepted. Growth requires an adequate supply of many different nutritional factors, some form the "building materials," whereas others play regulatory roles. In this article we describe the growth of the growth plate and discuss the role of nutritional affected hormones on this process. In addition we describe the effect of local regulators and nutritional factors on the growth process and suggest the involvement of new regulatory factors in the translation of nutrition to growth.
Collapse
Affiliation(s)
- Galia Gat-Yablonski
- The Jesse Z. and Sara Lea Shafer Institute for Endocrinology and Diabetes, National Center for Childhood Diabetes, Schneider Children's Medical Center of Israel, 14 Kaplan Street, Petah Tikva 49202, Israel
| | | | | |
Collapse
|