1
|
Fan S, Cai Y, Wei Y, Yang J, Gao J, Yang Y. Sarcopenic obesity and osteoporosis: Research progress and hot spots. Exp Gerontol 2024; 195:112544. [PMID: 39147076 DOI: 10.1016/j.exger.2024.112544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 07/17/2024] [Accepted: 08/11/2024] [Indexed: 08/17/2024]
Abstract
Sarcopenic obesity (SO) and osteoporosis (OP) are associated with aging and obesity. The pathogenesis of SO is complex, including glucolipid and skeletal muscle metabolic disorders caused by inflammation, insulin resistance, and other factors. Growing evidence links muscle damage to bone loss. Muscle-lipid metabolism disorders of SO disrupt the balance between bone formation and bone resorption, increasing the risk of OP. Conversely, bones also play a role in fat and muscle metabolism. In the context of aging and obesity, the comprehensive review focuses on the effects of mechanical stimulation, mesenchymal stem cells (MSCs), chronic inflammation, myokines, and adipokines on musculoskeletal, at the same time, the impact of osteokines on muscle-lipid metabolism were also analyzed. So far, exercise combined with diet therapy is the most effective strategy for increasing musculoskeletal mass. A holistic treatment of musculoskeletal diseases is still in the preliminary exploration stage. Therefore, this article aims to improve the understanding of musculoskeletal -fat interactions in SO and OP, explores targets that can provide holistic treatment for SO combined with OP, and discusses current limitations and challenges. We hope to provide relevant ideas for developing specific therapies and improving disease prognosis in the future.
Collapse
Affiliation(s)
- Shangheng Fan
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, China; Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China; Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Department of Pharmacology, Zunyi Medical University, Zunyi, China
| | - Yulan Cai
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Yunqin Wei
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Jia Yang
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Jianmei Gao
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China; Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Department of Pharmacology, Zunyi Medical University, Zunyi, China.
| | - Yan Yang
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, China.
| |
Collapse
|
2
|
Zhang X, Tian L, Majumdar A, Scheller EL. Function and Regulation of Bone Marrow Adipose Tissue in Health and Disease: State of the Field and Clinical Considerations. Compr Physiol 2024; 14:5521-5579. [PMID: 39109972 PMCID: PMC11725182 DOI: 10.1002/cphy.c230016] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2025]
Abstract
Bone marrow adipose tissue (BMAT) is a metabolically and clinically relevant fat depot that exists within bone. Two subtypes of BMAT, regulated and constitutive, reside in hematopoietic-rich red marrow and fatty yellow marrow, respectively, and exhibit distinct characteristics compared to peripheral fat such as white and brown adipose tissues. Bone marrow adipocytes (BMAds) are evolutionally preserved in most vertebrates, start development after birth and expand throughout life, and originate from unique progenitor populations that control bone formation and hematopoiesis. Mature BMAds also interact closely with other cellular components of the bone marrow niche, serving as a nearby energy reservoir to support the skeletal system, a signaling hub that contributes to both local and systemic homeostasis, and a final fuel reserve for survival during starvation. Though BMAT and bone are often inversely correlated, more BMAT does not always mean less bone, and the prevention of BMAT expansion as a strategy to prevent bone loss remains questionable. BMAT adipogenesis and lipid metabolism are regulated by the nervous systems and a variety of circulating hormones. This contributes to the plasticity of BMAT, including BMAT expansion in common physiological or pathological conditions, and BMAT catabolism under certain extreme circumstances, which are often associated with malnutrition and/or systemic inflammation. Altogether, this article provides a comprehensive overview of the local and systemic functions of BMAT and discusses the regulation and plasticity of this unique adipose tissue depot in health and disease. © 2024 American Physiological Society. Compr Physiol 14:5521-5579, 2024.
Collapse
Affiliation(s)
- Xiao Zhang
- Division of Bone and Mineral Diseases, Department of Medicine, Washington University, St. Louis, Missouri, USA
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri, USA
| | - Linda Tian
- Division of Bone and Mineral Diseases, Department of Medicine, Washington University, St. Louis, Missouri, USA
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri, USA
| | - Anurag Majumdar
- Division of Bone and Mineral Diseases, Department of Medicine, Washington University, St. Louis, Missouri, USA
| | - Erica L. Scheller
- Division of Bone and Mineral Diseases, Department of Medicine, Washington University, St. Louis, Missouri, USA
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri, USA
- Department of Cell Biology and Physiology, Washington University, St. Louis, Missouri, USA
| |
Collapse
|
3
|
Turner RT, Branscum AJ, Iwaniec UT. Long-duration leptin transgene expression in dorsal vagal complex does not alter bone parameters in female Sprague Dawley rats. Bone Rep 2024; 21:101769. [PMID: 38706522 PMCID: PMC11067478 DOI: 10.1016/j.bonr.2024.101769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 04/21/2024] [Accepted: 04/23/2024] [Indexed: 05/07/2024] Open
Abstract
The hypothalamus and dorsal vagal complex (DVC) are both important for integration of signals that regulate energy balance. Increased leptin transgene expression in either the hypothalamus or DVC of female rats was shown to decrease white adipose tissue and circulating levels of leptin and adiponectin. However, in contrast to hypothalamus, leptin transgene expression in the DVC had no effect on food intake, circulating insulin, ghrelin and glucose, nor on thermogenic energy expenditure. These findings imply different roles for hypothalamus and DVC in leptin signaling. Leptin signaling is required for normal bone accrual and turnover. Leptin transgene expression in the hypothalamus normalized the skeletal phenotype of leptin-deficient ob/ob mice but had no long-duration (≥10 weeks) effects on the skeleton of leptin-replete rats. The goal of this investigation was to determine the long-duration effects of leptin transgene expression in the DVC on the skeleton of leptin-replete rats. To accomplish this goal, we analyzed bone from three-month-old female rats that were microinjected with recombinant adeno-associated virus encoding either rat leptin (rAAV-Leptin, n = 6) or green fluorescent protein (rAAV-GFP, control, n = 5) gene. Representative bones from the appendicular (femur) and axial (3rd lumbar vertebra) skeleton were evaluated following 10 weeks of treatment. Selectively increasing leptin transgene expression in the DVC had no effect on femur cortical or cancellous bone microarchitecture. Additionally, increasing leptin transgene expression had no effect on vertebral osteoblast-lined or osteoclast-lined bone perimeter or marrow adiposity. Taken together, the findings suggest that activation of leptin receptors in the DVC has minimal specific effects on the skeleton of leptin-replete female rats.
Collapse
Affiliation(s)
- Russell T. Turner
- Skeletal Biology Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR 97331, USA
- Center for Healthy Aging Research, Oregon State University, Corvallis, OR 97331, USA
| | - Adam J. Branscum
- Biostatistics Program, School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR 97331, USA
| | - Urszula T. Iwaniec
- Skeletal Biology Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR 97331, USA
- Center for Healthy Aging Research, Oregon State University, Corvallis, OR 97331, USA
| |
Collapse
|
4
|
Shen F, Shi Y. Recent Advances in Single-Cell View of Mesenchymal Stem Cell in Osteogenesis. Front Cell Dev Biol 2022; 9:809918. [PMID: 35071243 PMCID: PMC8766509 DOI: 10.3389/fcell.2021.809918] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 12/10/2021] [Indexed: 02/05/2023] Open
Abstract
Osteoblasts continuously replenished by osteoblast progenitor cells form the basis of bone development, maintenance, and regeneration. Mesenchymal stem cells (MSCs) from various tissues can differentiate into the progenitor cell of osteogenic lineage and serve as the main source of osteoblasts. They also respond flexibly to regenerative and anabolic signals emitted by the surrounding microenvironment, thereby maintaining bone homeostasis and participating in bone remodeling. However, MSCs exhibit heterogeneity at multiple levels including different tissue sources and subpopulations which exhibit diversified gene expression and differentiation capacity, and surface markers used to predict cell differentiation potential remain to be further elucidated. The rapid advancement of lineage tracing methods and single-cell technology has made substantial progress in the characterization of osteogenic stem/progenitor cell populations in MSCs. Here, we reviewed the research progress of scRNA-seq technology in the identification of osteogenic markers and differentiation pathways, MSC-related new insights drawn from single-cell technology combined with experimental technology, and recent findings regarding the interaction between stem cell fate and niche in homeostasis and pathological process.
Collapse
Affiliation(s)
- Fangyuan Shen
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yu Shi
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
5
|
Wang L, Zhang H, Wang S, Chen X, Su J. Bone Marrow Adipocytes: A Critical Player in the Bone Marrow Microenvironment. Front Cell Dev Biol 2021; 9:770705. [PMID: 34912805 PMCID: PMC8667222 DOI: 10.3389/fcell.2021.770705] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Accepted: 10/29/2021] [Indexed: 12/01/2022] Open
Abstract
Recognized for nearly 100 years, bone marrow adipocytes (BMAs) form bone marrow niches that contain hematopoietic and bone cells, the roles of which have long been underestimated. Distinct from canonical white, brown, and beige adipocytes, BMAs derived from bone marrow mesenchymal stromal cells possess unique characteristics and functions. Recent single-cell sequencing studies have revealed the differentiation pathway, and seminal works support the tenet that BMAs are critical regulators in hematopoiesis, osteogenesis, and osteoclastogenesis. In this review, we discuss the origin and differentiation of BMAs, as well as the roles of BMAs in hematopoiesis, osteogenesis, osteoclastogenesis, and immune regulation. Overall, BMAs represent a novel target for bone marrow-related diseases, including osteoporosis and leukemia.
Collapse
Affiliation(s)
- Lipeng Wang
- Institute of Translational Medicine, Shanghai University, Shanghai, China
| | - Hao Zhang
- Department of Orthopedics Trauma, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Sicheng Wang
- Department of Orthopedics, Shanghai Zhongye Hospital, Shanghai, China
| | - Xiao Chen
- Department of Orthopedics Trauma, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| | - Jiacan Su
- Institute of Translational Medicine, Shanghai University, Shanghai, China.,Department of Orthopedics Trauma, Shanghai Changhai Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
6
|
Abstract
Bone marrow adipose tissue (BMAT) is an important cellular component of the skeleton. Understanding how it is regulated by the nervous system is crucial to the study of bone and bone marrow related diseases. BMAT is innervated by sympathetic and sensory axons in bone and fluctuations in local nerve density and function may contribute to its distinct physiologic adaptations at various skeletal sites. BMAT is directly responsive to adrenergic signals. In addition, neural regulation of surrounding cells may modify BMAT-specific responses, providing many potential avenues for both direct and indirect neural regulation of BMAT metabolism. Lastly, BMAT and peripheral adipose tissues share the same autonomic pathways across the central neuraxis and regulation of BMAT may occur in diverse clinical settings of neurologic and metabolic disease. This review will highlight what is known and unknown about the neural regulation of BMAT and discuss opportunities for future research in the field.
Collapse
Affiliation(s)
- Xiao Zhang
- Division of Bone and Mineral Diseases, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, 63110, USA; Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO, 63130, USA
| | - Mohamed G Hassan
- Department of Orthodontics, Faculty of Oral and Dental Medicine, South Valley University, Qena, Egypt; Department of Orthodontics, Faculty of Dentistry, October 6 University, Giza, Egypt
| | - Erica L Scheller
- Division of Bone and Mineral Diseases, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, 63110, USA; Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO, 63130, USA.
| |
Collapse
|
7
|
Piotrowska K, Tarnowski M. Bone Marrow Adipocytes-Role in Physiology and Various Nutritional Conditions in Human and Animal Models. Nutrients 2021; 13:nu13051412. [PMID: 33922353 PMCID: PMC8146898 DOI: 10.3390/nu13051412] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/19/2021] [Accepted: 04/20/2021] [Indexed: 02/06/2023] Open
Abstract
In recent years, adipose tissue has attracted a lot of attention. It is not only an energy reservoir but also plays important immune, paracrine and endocrine roles. BMAT (bone marrow adipose tissue) is a heterogeneous tissue, found mostly in the medullary canal of the long bones (tibia, femur and humerus), in the vertebrae and iliac crest. Adipogenesis in bone marrow cavities is a consequence of ageing or may accompany pathologies like diabetes mellitus type 1 (T1DM), T2DM, anorexia nervosa, oestrogen and growth hormone deficiencies or impaired haematopoiesis and osteoporosis. This paper focuses on studies concerning BMAT and its physiology in dietary interventions, like obesity in humans and high fat diet in rodent studies; and opposite: anorexia nervosa and calorie restriction in animal models.
Collapse
|
8
|
Abstract
The skeleton harbors an array of lineage cells that have an essential role in whole body homeostasis. Adipocytes start the colonization of marrow space early in postnatal life, expanding progressively and influencing other components of the bone marrow through paracrine signaling. In this unique, closed, and hypoxic environment close to the endosteal surface and adjacent to the microvascular space the marrow adipocyte can store or provide energy, secrete adipokines, and target neighboring bone cells. Adipocyte progenitors can also migrate from the bone marrow to populate white adipose tissue, a process that accelerates during weight gain. The marrow adipocyte also has an endocrine role in whole body homeostasis through its varied secretome that targets distant adipose depots, skeletal muscle, and the nervous system. Further insights into the biology of this unique and versatile cell will undoubtedly lead to novel therapeutic approaches to metabolic and age-related disorders such as osteoporosis and diabetes mellitus.
Collapse
Affiliation(s)
- Francisco J A de Paula
- Department of Internal Medicine, Ribeirao Preto Medical School, University of São Paulo, São Paulo 14049-900, Brazil;
| | - Clifford J Rosen
- Center for Clinical and Translational Research, Maine Medical Center Research Institute, Scarborough, Maine 04074, USA;
| |
Collapse
|
9
|
Sebo ZL, Rendina-Ruedy E, Ables GP, Lindskog DM, Rodeheffer MS, Fazeli PK, Horowitz MC. Bone Marrow Adiposity: Basic and Clinical Implications. Endocr Rev 2019; 40:1187-1206. [PMID: 31127816 PMCID: PMC6686755 DOI: 10.1210/er.2018-00138] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 04/03/2019] [Indexed: 12/14/2022]
Abstract
The presence of adipocytes in mammalian bone marrow (BM) has been recognized histologically for decades, yet, until recently, these cells have received little attention from the research community. Advancements in mouse transgenics and imaging methods, particularly in the last 10 years, have permitted more detailed examinations of marrow adipocytes than ever before and yielded data that show these cells are critical regulators of the BM microenvironment and whole-body metabolism. Indeed, marrow adipocytes are anatomically and functionally separate from brown, beige, and classic white adipocytes. Thus, areas of BM space populated by adipocytes can be considered distinct fat depots and are collectively referred to as marrow adipose tissue (MAT) in this review. In the proceeding text, we focus on the developmental origin and physiologic functions of MAT. We also discuss the signals that cause the accumulation and loss of marrow adipocytes and the ability of these cells to regulate other cell lineages in the BM. Last, we consider roles for MAT in human physiology and disease.
Collapse
Affiliation(s)
- Zachary L Sebo
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, Connecticut.,Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, Connecticut
| | | | - Gene P Ables
- Orentreich Foundation for the Advancement of Science, Cold Spring, New York
| | - Dieter M Lindskog
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, New Haven, Connecticut
| | - Matthew S Rodeheffer
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, Connecticut.,Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, Connecticut
| | - Pouneh K Fazeli
- Neuroendocrine Unit, Massachusetts General Hospital, Boston, Massachusetts
| | - Mark C Horowitz
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|
10
|
Suzuki T, Fujiwara S, Kinoshita S, Butovich IA. Cyclic Change of Fatty Acid Composition in Meibum During the Menstrual Cycle. Invest Ophthalmol Vis Sci 2019; 60:1724-1733. [PMID: 31013345 PMCID: PMC6736406 DOI: 10.1167/iovs.18-26390] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Purpose To evaluate the fatty acid (FA) composition in the meibum of pre- and postmenopausal women and age-matched men. Methods This prospective study involved 24 healthy subjects; six premenopausal women in their 30s with a regular menstrual cycle (young-female [yF] group), six postmenopausal women in their 60s (elderly-female [eF] group), and 12 age-matched men (i.e., young-male [yM] and elderly-male [eM] groups, respectively). The menstrual cycle was divided into six phases (phase I–VI). Meibum was obtained from the meibomian gland orifices via a Daviel spoon, and its FA composition was then analyzed via gas chromatography mass spectrometry (GC-MS). Principal component analysis (PCA) was performed on the GC-MS results. Results The mean FA composition of all subjects was 40% saturated FAs (SFA) and 60% unsaturated FAs (UFAs). The PCA results of all groups indicated two categories (PC1 [77.5%] and PC2 [12.4%]); one consisting of yF-group samples of mainly phase II and III and the other consisting of the yF-group samples of the rest of the cycle, as well as from eF-group, yM-group, and eM-group samples. Each group had a distinctive nature. The FAs that most contributed to PC1 were C14:0, C16:0, and C18:0 in a positive correlation, and C18:1n9 in a negative correlation. Conclusions FA composition noticeably changes during the menstrual cycle and is somewhat affected by sex and age. The ratio of SFAs (C16:0, C18:0) to mono-UFAs (C18:1n9) in the FA composition might have an impact on the lipid quality of meibum, thus suggesting alteration of its melting temperature and viscosity.
Collapse
Affiliation(s)
- Tomo Suzuki
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, Kyoto, Japan.,Department of Ophthalmology, Kyoto City Hospital Organization, Kyoto, Japan
| | | | - Shigeru Kinoshita
- Department of Frontier Medical Science and Technology for Ophthalmology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Igor A Butovich
- Department of Ophthalmology, The University of Texas Southwestern Medical Center, Dallas, Texas, United States
| |
Collapse
|
11
|
Abstract
PURPOSE OF REVIEW Mesenchymal stem cells (MSCs) located in the bone marrow have the capacity to differentiate into multiple cell lineages, including osteoblast and adipocyte. Adipocyte density within marrow is inversely associated with bone mass during aging and in some pathological conditions, contributing to the prevailing view that marrow adipocytes play a largely negative role in bone metabolism. However, a negative association between marrow adipocytes and bone balance is not universal. Although MAT levels appear tightly regulated, establishing the precise physiological significance of MAT has proven elusive. Here, we review recent literature aimed at delineating the function of MAT. RECENT FINDINGS An important physiological function of MAT may be to provide an expandable/contractible fat depot, which is critical for minimization of energy requirements for sustaining optimal hematopoiesis. Because the energy requirements for storing fat are negligible compared to those required to maintain hematopoiesis, even small reductions in hematopoietic tissue volume to match a reduced requirement for hematopoiesis could represent an important reduction in energy cost. Such a physiological function would require tight coupling between hematopoietic stem cells and MSCs to regulate the balance between MAT and hematopoiesis. Kit-ligand, an important regulator of proliferation, differentiation, and survival of hematopoietic cells, may function as a prototypic factor coupling MAT and hematopoiesis. Crosstalk between hematopoietic and mesenchymal cells in the bone marrow may contribute to establishing the balance between MAT levels and hematopoiesis.
Collapse
Affiliation(s)
- Russell T Turner
- Skeletal Biology Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR, 97331, USA
- Center for Healthy Aging Research, Oregon State University, Corvallis, OR, 97331, USA
| | - Stephen A Martin
- Skeletal Biology Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR, 97331, USA
| | - Urszula T Iwaniec
- Skeletal Biology Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR, 97331, USA.
- Center for Healthy Aging Research, Oregon State University, Corvallis, OR, 97331, USA.
| |
Collapse
|
12
|
|
13
|
Scheller EL, Cawthorn WP, Burr AA, Horowitz MC, MacDougald OA. Marrow Adipose Tissue: Trimming the Fat. Trends Endocrinol Metab 2016; 27:392-403. [PMID: 27094502 PMCID: PMC4875855 DOI: 10.1016/j.tem.2016.03.016] [Citation(s) in RCA: 153] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 03/25/2016] [Accepted: 03/29/2016] [Indexed: 02/07/2023]
Abstract
Marrow adipose tissue (MAT) is a unique fat depot, located in the skeleton, that has the potential to contribute to both local and systemic metabolic processes. In this review we highlight several recent conceptual developments pertaining to the origin and function of MAT adipocytes; consider the relationship of MAT to beige, brown, and white adipose depots; explore MAT expansion and turnover in humans and rodents; and discuss future directions for MAT research in the context of endocrine function and metabolic disease. MAT has the potential to exert both local and systemic effects on metabolic homeostasis, skeletal remodeling, hematopoiesis, and the development of bone metastases. The diversity of these functions highlights the breadth of the potential impact of MAT on health and disease.
Collapse
Affiliation(s)
- Erica L Scheller
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA; Division of Bone and Mineral Diseases, Department of Internal Medicine, Washington University, Saint Louis, MO 63110, USA.
| | - William P Cawthorn
- University/BHF Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Aaron A Burr
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Mark C Horowitz
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Ormond A MacDougald
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA; Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
14
|
Rial SA, Karelis AD, Bergeron KF, Mounier C. Gut Microbiota and Metabolic Health: The Potential Beneficial Effects of a Medium Chain Triglyceride Diet in Obese Individuals. Nutrients 2016; 8:nu8050281. [PMID: 27187452 PMCID: PMC4882694 DOI: 10.3390/nu8050281] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 05/05/2016] [Accepted: 05/09/2016] [Indexed: 02/07/2023] Open
Abstract
Obesity and associated metabolic complications, such as non-alcoholic fatty liver disease (NAFLD) and type 2 diabetes (T2D), are in constant increase around the world. While most obese patients show several metabolic and biometric abnormalities and comorbidities, a subgroup of patients representing 3% to 57% of obese adults, depending on the diagnosis criteria, remains metabolically healthy. Among many other factors, the gut microbiota is now identified as a determining factor in the pathogenesis of metabolically unhealthy obese (MUHO) individuals and in obesity-related diseases such as endotoxemia, intestinal and systemic inflammation, as well as insulin resistance. Interestingly, recent studies suggest that an optimal healthy-like gut microbiota structure may contribute to the metabolically healthy obese (MHO) phenotype. Here, we describe how dietary medium chain triglycerides (MCT), previously found to promote lipid catabolism, energy expenditure and weight loss, can ameliorate metabolic health via their capacity to improve both intestinal ecosystem and permeability. MCT-enriched diets could therefore be used to manage metabolic diseases through modification of gut microbiota.
Collapse
Affiliation(s)
- Sabri Ahmed Rial
- BioMed Research Center, Biological Sciences Department, University of Quebec at Montreal, Montreal, QC H2X 1Y4, Canada.
| | - Antony D Karelis
- Department of Exercise Science, University of Quebec at Montreal, Montreal, QC H2X 1Y4, Canada.
| | - Karl-F Bergeron
- BioMed Research Center, Biological Sciences Department, University of Quebec at Montreal, Montreal, QC H2X 1Y4, Canada.
| | - Catherine Mounier
- BioMed Research Center, Biological Sciences Department, University of Quebec at Montreal, Montreal, QC H2X 1Y4, Canada.
| |
Collapse
|
15
|
Yue R, Zhou BO, Shimada IS, Zhao Z, Morrison SJ. Leptin Receptor Promotes Adipogenesis and Reduces Osteogenesis by Regulating Mesenchymal Stromal Cells in Adult Bone Marrow. Cell Stem Cell 2016; 18:782-796. [PMID: 27053299 DOI: 10.1016/j.stem.2016.02.015] [Citation(s) in RCA: 336] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 01/25/2016] [Accepted: 02/25/2016] [Indexed: 12/24/2022]
Abstract
Skeletal stem cells (SSCs) that are the major source of osteoblasts and adipocytes in adult bone marrow express leptin receptor (LepR). To test whether LepR regulates SSC function, we conditionally deleted Lepr from limb bone marrow stromal cells, but not from the axial skeleton or hypothalamic neurons, using Prx1-Cre. Prx1-Cre;Lepr(fl/fl) mice exhibited normal body mass and normal hematopoiesis. However, limb bones from Prx1-Cre;Lepr(fl/fl) mice exhibited increased osteogenesis, decreased adipogenesis, and accelerated fracture healing. Leptin increased adipogenesis and reduced osteogenesis by activating Jak2/Stat3 signaling in bone marrow stromal cells. A high-fat diet increased adipogenesis and reduced osteogenesis in limb bones from wild-type mice, but not from Prx1-Cre;Lepr(fl/fl) mice. This reflected local effects of LepR on osteogenesis and adipogenesis by bone marrow stromal cells and systemic effects on bone resorption. Leptin/LepR signaling regulates adipogenesis and osteogenesis by mesenchymal stromal cells in the bone marrow in response to diet and adiposity.
Collapse
Affiliation(s)
- Rui Yue
- Department of Pediatrics and Children's Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Bo O Zhou
- Department of Pediatrics and Children's Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Issei S Shimada
- Department of Pediatrics and Children's Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Zhiyu Zhao
- Department of Pediatrics and Children's Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Sean J Morrison
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Pediatrics and Children's Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
16
|
Cawthorn WP, Scheller EL, Parlee SD, Pham HA, Learman BS, Redshaw CMH, Sulston RJ, Burr AA, Das AK, Simon BR, Mori H, Bree AJ, Schell B, Krishnan V, MacDougald OA. Expansion of Bone Marrow Adipose Tissue During Caloric Restriction Is Associated With Increased Circulating Glucocorticoids and Not With Hypoleptinemia. Endocrinology 2016; 157:508-21. [PMID: 26696121 PMCID: PMC4733126 DOI: 10.1210/en.2015-1477] [Citation(s) in RCA: 109] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Bone marrow adipose tissue (MAT) accounts for up to 70% of bone marrow volume in healthy adults and increases further in clinical conditions of altered skeletal or metabolic function. Perhaps most strikingly, and in stark contrast to white adipose tissue, MAT has been found to increase during caloric restriction (CR) in humans and many other species. Hypoleptinemia may drive MAT expansion during CR but this has not been demonstrated conclusively. Indeed, MAT formation and function are poorly understood; hence, the physiological and pathological roles of MAT remain elusive. We recently revealed that MAT contributes to hyperadiponectinemia and systemic adaptations to CR. To further these observations, we have now performed CR studies in rabbits to determine whether CR affects adiponectin production by MAT. Moderate or extensive CR decreased bone mass, white adipose tissue mass, and circulating leptin but, surprisingly, did not cause hyperadiponectinemia or MAT expansion. Although this unexpected finding limited our subsequent MAT characterization, it demonstrates that during CR, bone loss can occur independently of MAT expansion; increased MAT may be required for hyperadiponectinemia; and hypoleptinemia is not sufficient for MAT expansion. We further investigated this relationship in mice. In females, CR increased MAT without decreasing circulating leptin, suggesting that hypoleptinemia is also not necessary for MAT expansion. Finally, circulating glucocorticoids increased during CR in mice but not rabbits, suggesting that glucocorticoids might drive MAT expansion during CR. These observations provide insights into the causes and consequences of CR-associated MAT expansion, knowledge with potential relevance to health and disease.
Collapse
Affiliation(s)
- William P Cawthorn
- Departments of Molecular and Integrative Physiology (W.P.C., E.L.S., S.D.P., H.A.P., B.S.L., A.A.B., H.M., A.J.B., B.S., O.A.M.) and Internal Medicine (A.K.D., O.A.M.), and Program in Cellular and Molecular Biology (B.R.S., O.A.M.), University of Michigan Medical School, Ann Arbor, Michigan 48109; Musculoskeletal Research (W.P.C., V.K.), Lilly Research Laboratories, Indianapolis, Indiana 46285; and University/British Heart Foundation Centre for Cardiovascular Science (W.P.C., C.M.H.R., R.J.S.), The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom EH16 4TJ
| | - Erica L Scheller
- Departments of Molecular and Integrative Physiology (W.P.C., E.L.S., S.D.P., H.A.P., B.S.L., A.A.B., H.M., A.J.B., B.S., O.A.M.) and Internal Medicine (A.K.D., O.A.M.), and Program in Cellular and Molecular Biology (B.R.S., O.A.M.), University of Michigan Medical School, Ann Arbor, Michigan 48109; Musculoskeletal Research (W.P.C., V.K.), Lilly Research Laboratories, Indianapolis, Indiana 46285; and University/British Heart Foundation Centre for Cardiovascular Science (W.P.C., C.M.H.R., R.J.S.), The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom EH16 4TJ
| | - Sebastian D Parlee
- Departments of Molecular and Integrative Physiology (W.P.C., E.L.S., S.D.P., H.A.P., B.S.L., A.A.B., H.M., A.J.B., B.S., O.A.M.) and Internal Medicine (A.K.D., O.A.M.), and Program in Cellular and Molecular Biology (B.R.S., O.A.M.), University of Michigan Medical School, Ann Arbor, Michigan 48109; Musculoskeletal Research (W.P.C., V.K.), Lilly Research Laboratories, Indianapolis, Indiana 46285; and University/British Heart Foundation Centre for Cardiovascular Science (W.P.C., C.M.H.R., R.J.S.), The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom EH16 4TJ
| | - H An Pham
- Departments of Molecular and Integrative Physiology (W.P.C., E.L.S., S.D.P., H.A.P., B.S.L., A.A.B., H.M., A.J.B., B.S., O.A.M.) and Internal Medicine (A.K.D., O.A.M.), and Program in Cellular and Molecular Biology (B.R.S., O.A.M.), University of Michigan Medical School, Ann Arbor, Michigan 48109; Musculoskeletal Research (W.P.C., V.K.), Lilly Research Laboratories, Indianapolis, Indiana 46285; and University/British Heart Foundation Centre for Cardiovascular Science (W.P.C., C.M.H.R., R.J.S.), The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom EH16 4TJ
| | - Brian S Learman
- Departments of Molecular and Integrative Physiology (W.P.C., E.L.S., S.D.P., H.A.P., B.S.L., A.A.B., H.M., A.J.B., B.S., O.A.M.) and Internal Medicine (A.K.D., O.A.M.), and Program in Cellular and Molecular Biology (B.R.S., O.A.M.), University of Michigan Medical School, Ann Arbor, Michigan 48109; Musculoskeletal Research (W.P.C., V.K.), Lilly Research Laboratories, Indianapolis, Indiana 46285; and University/British Heart Foundation Centre for Cardiovascular Science (W.P.C., C.M.H.R., R.J.S.), The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom EH16 4TJ
| | - Catherine M H Redshaw
- Departments of Molecular and Integrative Physiology (W.P.C., E.L.S., S.D.P., H.A.P., B.S.L., A.A.B., H.M., A.J.B., B.S., O.A.M.) and Internal Medicine (A.K.D., O.A.M.), and Program in Cellular and Molecular Biology (B.R.S., O.A.M.), University of Michigan Medical School, Ann Arbor, Michigan 48109; Musculoskeletal Research (W.P.C., V.K.), Lilly Research Laboratories, Indianapolis, Indiana 46285; and University/British Heart Foundation Centre for Cardiovascular Science (W.P.C., C.M.H.R., R.J.S.), The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom EH16 4TJ
| | - Richard J Sulston
- Departments of Molecular and Integrative Physiology (W.P.C., E.L.S., S.D.P., H.A.P., B.S.L., A.A.B., H.M., A.J.B., B.S., O.A.M.) and Internal Medicine (A.K.D., O.A.M.), and Program in Cellular and Molecular Biology (B.R.S., O.A.M.), University of Michigan Medical School, Ann Arbor, Michigan 48109; Musculoskeletal Research (W.P.C., V.K.), Lilly Research Laboratories, Indianapolis, Indiana 46285; and University/British Heart Foundation Centre for Cardiovascular Science (W.P.C., C.M.H.R., R.J.S.), The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom EH16 4TJ
| | - Aaron A Burr
- Departments of Molecular and Integrative Physiology (W.P.C., E.L.S., S.D.P., H.A.P., B.S.L., A.A.B., H.M., A.J.B., B.S., O.A.M.) and Internal Medicine (A.K.D., O.A.M.), and Program in Cellular and Molecular Biology (B.R.S., O.A.M.), University of Michigan Medical School, Ann Arbor, Michigan 48109; Musculoskeletal Research (W.P.C., V.K.), Lilly Research Laboratories, Indianapolis, Indiana 46285; and University/British Heart Foundation Centre for Cardiovascular Science (W.P.C., C.M.H.R., R.J.S.), The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom EH16 4TJ
| | - Arun K Das
- Departments of Molecular and Integrative Physiology (W.P.C., E.L.S., S.D.P., H.A.P., B.S.L., A.A.B., H.M., A.J.B., B.S., O.A.M.) and Internal Medicine (A.K.D., O.A.M.), and Program in Cellular and Molecular Biology (B.R.S., O.A.M.), University of Michigan Medical School, Ann Arbor, Michigan 48109; Musculoskeletal Research (W.P.C., V.K.), Lilly Research Laboratories, Indianapolis, Indiana 46285; and University/British Heart Foundation Centre for Cardiovascular Science (W.P.C., C.M.H.R., R.J.S.), The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom EH16 4TJ
| | - Becky R Simon
- Departments of Molecular and Integrative Physiology (W.P.C., E.L.S., S.D.P., H.A.P., B.S.L., A.A.B., H.M., A.J.B., B.S., O.A.M.) and Internal Medicine (A.K.D., O.A.M.), and Program in Cellular and Molecular Biology (B.R.S., O.A.M.), University of Michigan Medical School, Ann Arbor, Michigan 48109; Musculoskeletal Research (W.P.C., V.K.), Lilly Research Laboratories, Indianapolis, Indiana 46285; and University/British Heart Foundation Centre for Cardiovascular Science (W.P.C., C.M.H.R., R.J.S.), The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom EH16 4TJ
| | - Hiroyuki Mori
- Departments of Molecular and Integrative Physiology (W.P.C., E.L.S., S.D.P., H.A.P., B.S.L., A.A.B., H.M., A.J.B., B.S., O.A.M.) and Internal Medicine (A.K.D., O.A.M.), and Program in Cellular and Molecular Biology (B.R.S., O.A.M.), University of Michigan Medical School, Ann Arbor, Michigan 48109; Musculoskeletal Research (W.P.C., V.K.), Lilly Research Laboratories, Indianapolis, Indiana 46285; and University/British Heart Foundation Centre for Cardiovascular Science (W.P.C., C.M.H.R., R.J.S.), The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom EH16 4TJ
| | - Adam J Bree
- Departments of Molecular and Integrative Physiology (W.P.C., E.L.S., S.D.P., H.A.P., B.S.L., A.A.B., H.M., A.J.B., B.S., O.A.M.) and Internal Medicine (A.K.D., O.A.M.), and Program in Cellular and Molecular Biology (B.R.S., O.A.M.), University of Michigan Medical School, Ann Arbor, Michigan 48109; Musculoskeletal Research (W.P.C., V.K.), Lilly Research Laboratories, Indianapolis, Indiana 46285; and University/British Heart Foundation Centre for Cardiovascular Science (W.P.C., C.M.H.R., R.J.S.), The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom EH16 4TJ
| | - Benjamin Schell
- Departments of Molecular and Integrative Physiology (W.P.C., E.L.S., S.D.P., H.A.P., B.S.L., A.A.B., H.M., A.J.B., B.S., O.A.M.) and Internal Medicine (A.K.D., O.A.M.), and Program in Cellular and Molecular Biology (B.R.S., O.A.M.), University of Michigan Medical School, Ann Arbor, Michigan 48109; Musculoskeletal Research (W.P.C., V.K.), Lilly Research Laboratories, Indianapolis, Indiana 46285; and University/British Heart Foundation Centre for Cardiovascular Science (W.P.C., C.M.H.R., R.J.S.), The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom EH16 4TJ
| | - Venkatesh Krishnan
- Departments of Molecular and Integrative Physiology (W.P.C., E.L.S., S.D.P., H.A.P., B.S.L., A.A.B., H.M., A.J.B., B.S., O.A.M.) and Internal Medicine (A.K.D., O.A.M.), and Program in Cellular and Molecular Biology (B.R.S., O.A.M.), University of Michigan Medical School, Ann Arbor, Michigan 48109; Musculoskeletal Research (W.P.C., V.K.), Lilly Research Laboratories, Indianapolis, Indiana 46285; and University/British Heart Foundation Centre for Cardiovascular Science (W.P.C., C.M.H.R., R.J.S.), The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom EH16 4TJ
| | - Ormond A MacDougald
- Departments of Molecular and Integrative Physiology (W.P.C., E.L.S., S.D.P., H.A.P., B.S.L., A.A.B., H.M., A.J.B., B.S., O.A.M.) and Internal Medicine (A.K.D., O.A.M.), and Program in Cellular and Molecular Biology (B.R.S., O.A.M.), University of Michigan Medical School, Ann Arbor, Michigan 48109; Musculoskeletal Research (W.P.C., V.K.), Lilly Research Laboratories, Indianapolis, Indiana 46285; and University/British Heart Foundation Centre for Cardiovascular Science (W.P.C., C.M.H.R., R.J.S.), The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom EH16 4TJ
| |
Collapse
|
17
|
Ropka-Molik K, Knapik J, Pieszka M, Szmatoła T. The expression of the SCD1 gene and its correlation with fattening and carcass traits in sheep. Arch Anim Breed 2016. [DOI: 10.5194/aab-59-37-2016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Abstract. Stearoyl-CoA desaturase 1 (SCD1) is a critical enzyme that catalyzes the synthesis of monounsaturated fatty acids and is involved in several signaling pathways related to lipid metabolism. The objective of the present study was to estimate the expression of the SCD1 gene in three different ovine tissues strongly associated with lipid homeostasis. The SCD1 gene expression measurement was performed on three tissues (liver, subcutaneous fat, perirenal fat) originated from 15 old-type Polish Merino sheep. The SCD1 transcript abundance was evaluated based on the two most stable endogenous controls (RPS2 – ribosomal protein S2; ATP5G2 – H(+)-transporting ATP synthase). The highest expression of the SCD1 gene was observed in ovine subcutaneous fat compared to perirenal fat and liver. Furthermore, the present research indicated the significant correlation between ovine SCD1 transcript abundance and several important production traits. The expression of the SCD1 gene in liver and perirenal fat highly positively correlated with the feed : gain ratio, test of daily gain and age of the animals at slaughter. Moreover, in both tissues, the SCD1mRNA level positively correlated with weight and content of perirenal fat and subcutaneous fat (R = 0.64, 0.8, 0.6, respectively) and negatively with assessment of external fat content with the use of the EUROP scale (R = −0.64). The SCD1 expression in subcutaneous fat also corresponds with back fat of blade chop and thickness of longissimus dorsi muscles evaluated using USG (ultrasonography) (R = −0.6 and 0.62, respectively). The significant correlation between SCD1 transcript abundance and fattening and slaughtering traits indicate the ability to improve important production traits in sheep via modification of expression of the SCD1 gene.
Collapse
|
18
|
Lindenmaier LB, Philbrick KA, Branscum AJ, Kalra SP, Turner RT, Iwaniec UT. Hypothalamic Leptin Gene Therapy Reduces Bone Marrow Adiposity in ob/ob Mice Fed Regular and High-Fat Diets. Front Endocrinol (Lausanne) 2016; 7:110. [PMID: 27579023 PMCID: PMC4985531 DOI: 10.3389/fendo.2016.00110] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 08/02/2016] [Indexed: 12/14/2022] Open
Abstract
Low bone mass is often associated with elevated bone marrow adiposity. Since osteoblasts and adipocytes are derived from the same mesenchymal stem cell (MSC) progenitor, adipocyte formation may increase at the expense of osteoblast formation. Leptin is an adipocyte-derived hormone known to regulate energy and bone metabolism. Leptin deficiency and high-fat diet-induced obesity are associated with increased marrow adipose tissue (MAT) and reduced bone formation. Short-duration studies suggest that leptin treatment reduces MAT and increases bone formation in leptin-deficient ob/ob mice fed a regular diet. Here, we determined the long-duration impact of increased hypothalamic leptin on marrow adipocytes and osteoblasts in ob/ob mice following recombinant adeno-associated virus (rAAV) gene therapy. Eight- to 10-week-old male ob/ob mice were randomized into four groups: (1) untreated, (2) rAAV-Lep, (3) rAAV-green fluorescent protein (rAAV-GFP), or (4) pair-fed to rAAV-Lep. For vector administration, mice were injected intracerebroventricularly with either rAAV-leptin gene therapy (rAAV-Lep) or rAAV-GFP (9 × 10(7) particles) and maintained for 30 weeks. In a second study, the impact of increased hypothalamic leptin levels on MAT was determined in mice fed high-fat diets; ob/ob mice were randomized into two groups and treated with either rAAV-Lep or rAAV-GFP. At 7 weeks post-vector administration, half the mice in each group were switched to a high-fat diet for 8 weeks. Wild-type (WT) controls included age-matched mice fed regular or high-fat diet. High-fat diet resulted in a threefold increase in MAT in WT mice, whereas MAT was increased by leptin deficiency up to 50-fold. Hypothalamic leptin gene therapy increased osteoblast perimeter and osteoclast perimeter with minor change in cancellous bone architecture. The gene therapy decreased MAT levels in ob/ob mice fed regular or high-fat diet to values similar to WT mice fed regular diet. These findings suggest that leptin plays an important role in regulating the differentiation of MSCs to adipocytes and osteoblasts, a process that may be dysregulated by high-fat diet. However, the results also illustrate that reducing MAT by increasing leptin levels does not necessarily result in increased bone mass.
Collapse
Affiliation(s)
- Laurence B. Lindenmaier
- Skeletal Biology Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR, USA
| | - Kenneth A. Philbrick
- Skeletal Biology Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR, USA
| | - Adam J. Branscum
- Biostatistics Program, School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR, USA
| | - Satya P. Kalra
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Russell T. Turner
- Skeletal Biology Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR, USA
- Center for Healthy Aging Research, Oregon State University, Corvallis, OR, USA
| | - Urszula T. Iwaniec
- Skeletal Biology Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR, USA
- Center for Healthy Aging Research, Oregon State University, Corvallis, OR, USA
- *Correspondence: Urszula T. Iwaniec,
| |
Collapse
|
19
|
Solomon G, Atkins A, Shahar R, Gertler A, Monsonego-Ornan E. Effect of peripherally administered leptin antagonist on whole body metabolism and bone microarchitecture and biomechanical properties in the mouse. Am J Physiol Endocrinol Metab 2014; 306:E14-27. [PMID: 24169045 DOI: 10.1152/ajpendo.00155.2013] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Leptin's in vivo effect on the rodent skeleton depends on the model used and the mode of administration. Superactive mouse leptin antagonist (SMLA) was produced and then pegylated (PEG) to prolong and enhance its in vivo activity. We blocked leptin signaling by injecting this antagonist peripherally into normal mice at various time points and studied their metabolic and skeletal phenotypes. Subcutaneous PEG-SMLA injections into 4-wk-old female C57BL/6J mice increased weight gain and food consumption significantly after only 1 mo, and the effect lasted for the 3 mo of the experiment, proving its central inhibiting activity. Mice showed a significant increase in serum glucose, cholesterol, triglycerides, insulin, and HOMA-IR throughout the experiment. Quantification of gene expression in "metabolic" tissues also indicated the development of insulin resistance. Bone analyses revealed a significant increase in trabecular and cortical parameters measured in both the lumbar vertebrae and tibiae in PEG-SMLA-treated mice in the 1st and 3rd months as well as a significant increase in tibia biomechanical parameters. Interestingly, 30 days of treatment with the antagonist in older mice (aged 3 and 6 mo) affected body weight and eating behavior, just as they had in the 1-mo-old mice, but had no effect on bone parameters, suggesting that leptin's effect on bones, either directly or through its obesogenic effect, is dependent upon stage of skeletal development. This potent and reversible antagonist enabled us to study leptin's in vivo role in whole body and bone metabolism and holds potential for future therapeutic use in diseases involving leptin signaling.
Collapse
|
20
|
Choi HJ, Ki KH, Yang JY, Jang BY, Song JA, Baek WY, Kim JH, An JH, Kim SW, Kim SY, Kim JE, Shin CS. Chronic central administration of Ghrelin increases bone mass through a mechanism independent of appetite regulation. PLoS One 2013; 8:e65505. [PMID: 23843943 PMCID: PMC3699588 DOI: 10.1371/journal.pone.0065505] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2013] [Accepted: 04/25/2013] [Indexed: 12/19/2022] Open
Abstract
Leptin plays a critical role in the central regulation of bone mass. Ghrelin counteracts leptin. In this study, we investigated the effect of chronic intracerebroventricular administration of ghrelin on bone mass in Sprague-Dawley rats (1.5 μg/day for 21 days). Rats were divided into control, ghrelin ad libitum-fed (ghrelin ad lib-fed), and ghrelin pair-fed groups. Ghrelin intracerebroventricular infusion significantly increased body weight in ghrelin ad lib-fed rats but not in ghrelin pair-fed rats, as compared with control rats. Chronic intracerebroventricular ghrelin infusion significantly increased bone mass in the ghrelin pair-fed group compared with control as indicated by increased bone volume percentage, trabecular thickness, trabecular number and volumetric bone mineral density in tibia trabecular bone. There was no significant difference in trabecular bone mass between the control group and the ghrelin ad-lib fed group. Chronic intracerebroventricular ghrelin infusion significantly increased the mineral apposition rate in the ghrelin pair-fed group as compared with control. In conclusion, chronic central administration of ghrelin increases bone mass through a mechanism that is independent of body weight, suggesting that ghrelin may have a bone anabolic effect through the central nervous system.
Collapse
Affiliation(s)
- Hyung Jin Choi
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Kyoung Ho Ki
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Jae-Yeon Yang
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Bo Young Jang
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Jung Ah Song
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Wook-Young Baek
- Department of Molecular Medicine, Cell and Matrix Research Institute, Kyungpook National University School of Medicine, Daegu, Korea
| | - Jung Hee Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Jee Hyun An
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Sang Wan Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Seong Yeon Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Jung-Eun Kim
- Department of Molecular Medicine, Cell and Matrix Research Institute, Kyungpook National University School of Medicine, Daegu, Korea
| | - Chan Soo Shin
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
- * E-mail:
| |
Collapse
|
21
|
Bartell SM, Rayalam S, Ambati S, Gaddam DR, Hartzell DL, Hamrick M, She JX, Della-Fera MA, Baile CA. Central (ICV) leptin injection increases bone formation, bone mineral density, muscle mass, serum IGF-1, and the expression of osteogenic genes in leptin-deficient ob/ob mice. J Bone Miner Res 2011; 26:1710-20. [PMID: 21520275 DOI: 10.1002/jbmr.406] [Citation(s) in RCA: 122] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Both central and peripheral leptin administrations reduce body weight, food intake, and adiposity in ob/ob mice. In this study we compared effects of intracerebroventricular (ICV) and subcutaneous (SC) administration of leptin on bone metabolism in the appendicular and axial skeleton and adipose tissue gene expression and determined the effects of ICV leptin on bone marrow gene expression in ob/ob mice. In experiment 1, leptin (1.5 or 0.38 µg/d) or control was continuously injected ICV for 12 days. Gene expression analysis of femoral bone marrow stromal cells showed that expression of genes associated with osteogenesis was increased after ICV injection, whereas those associated with osteoclastogenesis, adipogenesis, and adipocyte lipid storage were decreased. In experiment 2, leptin was injected continuously ICV (0.0 or 1.5 µg/d) or SC (0.0 or 10 µg/d) for 12 days. In both experiments, regardless of mode of administration, leptin decreased body weight, food intake, and body fat and increased muscle mass, bone mineral density, bone mineral content, bone area, marrow adipocyte number, and mineral apposition rate. Serum insulin was decreased, whereas serum osteocalcin, insulin-like growth factor 1, osteoprotegerin, pyridinoline, and receptor activator of nuclear factor κB ligand concentrations were increased. In experiment 2, expression of genes in adipose tissue associated with apoptosis, lipid mobilization, insulin sensitivity, and thermogenesis was increased, whereas expression of genes associated with cell differentiation and maturation was decreased regardless of mode of administration. Thus ICV injection of leptin promotes expression of pro-osteogenic factors in bone marrow, leading to enhanced bone formation in ob/ob mice.
Collapse
Affiliation(s)
- Shoshana M Bartell
- Department of Animal and Dairy Science, University of Georgia, Athens, GA 30602, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Iwaniec UT, Boghossian S, Trevisiol CH, Wronski TJ, Turner RT, Kalra SP. Hypothalamic leptin gene therapy prevents weight gain without long-term detrimental effects on bone in growing and skeletally mature female rats. J Bone Miner Res 2011; 26:1506-16. [PMID: 21328617 PMCID: PMC3129999 DOI: 10.1002/jbmr.365] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Hypothalamic leptin gene therapy normalizes the mosaic skeletal phenotype of leptin-deficient ob/ob mice. However, it is not clear whether increased hypothalamic leptin alters bone metabolism in animals already producing the hormone. The objective of this study was to evaluate the long duration effects of recombinant adeno-associated virus-rat leptin (rAAV-Lep) hypothalamic gene therapy on weight gain and bone metabolism in growing and skeletally mature leptin-replete female Sprague-Dawley rats. Rats were either unoperated or implanted with cannulas in the third ventricle of the hypothalamus and injected with either rAAV-Lep or rAAV-GFP (control vector encoding green fluorescent protein) and maintained on standard rat chow fed ad libitum for either 5 or 10 weeks (starting at 3 months of age) or 18 weeks (starting at 9 months of age). Tibias, femurs, or lumbar vertebrae were analyzed by micro-computed tomography and/or histomorphometry. In comparison with age-matched rAAV-GFP rats, rAAV-Lep rats maintained a lower body weight for the duration of studies. At 5 weeks after vector administration, rAAV-Lep rats had lower cancellous bone volume and bone marrow adiposity but higher osteoblast perimeter compared with nonoperated controls. However, these values did not differ between the two groups at 10 weeks after vector administration. Differences in cancellous bone volume and architecture were not detected between the rAAV-Lep and rAAV-GFP groups at either time point. Also, rAAV-Lep had no negative effects on bone in the 9-month-old skeletally mature rats at 18 weeks after vector administration. We hypothesize that the transient reductions in bone mass and bone marrow adiposity at 5 weeks after vector administration were due to hypothalamic surgery. We conclude that increased hypothalamic leptin, sufficient to prevent weight gain, has minimal specific effects (rAAV-Lep versus rAAV-GFP) on bone metabolism in normal female rats.
Collapse
Affiliation(s)
- Urszula T Iwaniec
- Department of Nutrition and Exercise Sciences, Oregon State University, Corvallis, OR 97331, USA.
| | | | | | | | | | | |
Collapse
|
23
|
Current world literature. Curr Opin Endocrinol Diabetes Obes 2011; 18:83-98. [PMID: 21178692 DOI: 10.1097/med.0b013e3283432fa7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|