Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
2.
J Mol Endocrinol ; 70(3)2023 04 01.
Article in English | MEDLINE | ID: mdl-36748836

ABSTRACT

Human genome-wide association studies found single-nucleotide polymorphisms (SNPs) near LYPLAL1 (Lysophospholipase-like protein 1) that have sex-specific effects on fat distribution and metabolic traits. To determine whether altering LYPLAL1 affects obesity and metabolic disease, we created and characterized a mouse knockout (KO) of Lyplal1. We fed the experimental group of mice a high-fat, high-sucrose (HFHS) diet for 23 weeks, and the controls were fed regular chow diet. Here, we show that CRISPR-Cas9 whole-body Lyplal1 KO mice fed an HFHS diet showed sex-specific differences in weight gain and fat accumulation as compared to chow diet. Female, not male, KO mice weighed less than WT mice, had reduced body fat percentage, had white fat mass, and had adipocyte diameter not accounted for by changes in the metabolic rate. Female, but not male, KO mice had increased serum triglycerides, decreased aspartate, and decreased alanine aminotransferase. Lyplal1 KO mice of both sexes have reduced liver triglycerides and steatosis. These diet-specific effects resemble the effects of SNPs near LYPLAL1 in humans, suggesting that LYPLAL1 has an evolutionary conserved sex-specific effect on adiposity. This murine model can be used to study this novel gene-by-sex-by-diet interaction to elucidate the metabolic effects of LYPLAL1 on human obesity.


Subject(s)
Genome-Wide Association Study , Lysophospholipase , Obesity , Animals , Female , Humans , Male , Mice , Diet, High-Fat/adverse effects , Mice, Inbred C57BL , Mice, Knockout , Obesity/genetics , Obesity/metabolism , Triglycerides , Lysophospholipase/genetics
3.
JCI Insight ; 7(21)2022 11 08.
Article in English | MEDLINE | ID: mdl-36048537

ABSTRACT

BM adipocytes (BMAd) are a unique cell population derived from BM mesenchymal progenitors and marrow adipogenic lineage precursors. Although they have long been considered to be a space filler within bone cavities, recent studies have revealed important physiological roles in hematopoiesis and bone metabolism. To date, the approaches used to study BMAd function have been confounded by contributions by nonmarrow adipocytes or by BM stromal cells. To address this gap in the field, we have developed a BMAd-specific Cre mouse model to deplete BMAds by expression of diphtheria toxin A (DTA) or by deletion of peroxisome proliferator-activated receptor gamma (Pparg). We found that DTA-induced loss of BMAds results in decreased hematopoietic stem and progenitor cell numbers and increased bone mass in BMAd-enriched locations, including the distal tibiae and caudal vertebrae. Elevated bone mass appears to be secondary to enhanced endosteal bone formation, suggesting a local effect caused by depletion of BMAd. Augmented bone formation with BMAd depletion protects mice from bone loss induced by caloric restriction or ovariectomy, and it facilitates the bone-healing process after fracture. Finally, ablation of Pparg also reduces BMAd numbers and largely recapitulates high-bone mass phenotypes observed with DTA-induced BMAd depletion.


Subject(s)
Bone Marrow , Mesenchymal Stem Cells , Female , Mice , Animals , Bone Marrow/metabolism , Osteogenesis , Bone Marrow Cells , PPAR gamma/genetics , PPAR gamma/metabolism , Mesenchymal Stem Cells/metabolism , Adipocytes/metabolism
4.
Elife ; 112022 06 22.
Article in English | MEDLINE | ID: mdl-35731039

ABSTRACT

To investigate roles for bone marrow adipocyte (BMAd) lipolysis in bone homeostasis, we created a BMAd-specific Cre mouse model in which we knocked out adipose triglyceride lipase (ATGL, Pnpla2 gene). BMAd-Pnpla2-/- mice have impaired BMAd lipolysis, and increased size and number of BMAds at baseline. Although energy from BMAd lipid stores is largely dispensable when mice are fed ad libitum, BMAd lipolysis is necessary to maintain myelopoiesis and bone mass under caloric restriction. BMAd-specific Pnpla2 deficiency compounds the effects of caloric restriction on loss of trabecular bone in male mice, likely due to impaired osteoblast expression of collagen genes and reduced osteoid synthesis. RNA sequencing analysis of bone marrow adipose tissue reveals that caloric restriction induces dramatic elevations in extracellular matrix organization and skeletal development genes, and energy from BMAd is required for these adaptations. BMAd-derived energy supply is also required for bone regeneration upon injury, and maintenance of bone mass with cold exposure.


Subject(s)
Bone Marrow , Lipolysis , Adipocytes/metabolism , Adipose Tissue/metabolism , Animals , Bone Marrow/metabolism , Lipase/metabolism , Lipolysis/genetics , Male , Mice
5.
Diabetes ; 70(7): 1419-1430, 2021 07.
Article in English | MEDLINE | ID: mdl-34155042

ABSTRACT

Wnt signaling is an ancient and evolutionarily conserved pathway with fundamental roles in the development of adipose tissues. Roles of this pathway in mesenchymal stem cell fate determination and differentiation have been extensively studied. Indeed, canonical Wnt signaling is a significant endogenous inhibitor of adipogenesis and promoter of other cell fates, including osteogenesis, chondrogenesis, and myogenesis. However, emerging genetic evidence in both humans and mice suggests central roles for Wnt signaling in body fat distribution, obesity, and metabolic dysfunction. Herein, we highlight recent studies that have begun to unravel the contributions of various Wnt pathway members to critical adipocyte functions, including carbohydrate and lipid metabolism. We further explore compelling evidence of complex and coordinated interactions between adipocytes and other cell types within adipose tissues, including stromal, immune, and endothelial cells. Given the evolutionary conservation and ubiquitous cellular distribution of this pathway, uncovering the contributions of Wnt signaling to cell metabolism has exciting implications for therapeutic intervention in widespread pathologic states, including obesity, diabetes, and cancers.


Subject(s)
Adipocytes/physiology , Lipogenesis/physiology , Mesenchymal Stem Cells/physiology , Wnt Signaling Pathway/physiology , Adipogenesis/physiology , Animals , Humans , Metabolic Diseases/etiology , Mice , Osteoblasts/physiology , Transcription Factor 7-Like 2 Protein/genetics , beta Catenin/physiology
6.
Diabetes ; 70(9): 1970-1984, 2021 09.
Article in English | MEDLINE | ID: mdl-34088712

ABSTRACT

Mechanisms by which autosomal recessive mutations in Lmna cause familial partial lipodystrophy type 2 (FPLD2) are poorly understood. To investigate the function of lamin A/C in adipose tissue, we created mice with an adipocyte-specific loss of Lmna (Lmna ADKO). Although Lmna ADKO mice develop and maintain adipose tissues in early postnatal life, they show a striking and progressive loss of white and brown adipose tissues as they approach sexual maturity. Lmna ADKO mice exhibit surprisingly mild metabolic dysfunction on a chow diet, but on a high-fat diet they share many characteristics of FPLD2 including hyperglycemia, hepatic steatosis, hyperinsulinemia, and almost undetectable circulating adiponectin and leptin. Whereas Lmna ADKO mice have reduced regulated and constitutive bone marrow adipose tissue with a concomitant increase in cortical bone, FPLD2 patients have reduced bone mass and bone mineral density compared with controls. In cell culture models of Lmna deficiency, mesenchymal precursors undergo adipogenesis without impairment, whereas fully differentiated adipocytes have increased lipolytic responses to adrenergic stimuli. Lmna ADKO mice faithfully reproduce many characteristics of FPLD2 and thus provide a unique animal model to investigate mechanisms underlying Lmna-dependent loss of adipose tissues.


Subject(s)
Adipocytes/metabolism , Adipogenesis/physiology , Lamin Type A/genetics , Lipodystrophy, Familial Partial/genetics , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Animals , Bone Density/physiology , Disease Models, Animal , Lamin Type A/metabolism , Lipodystrophy, Familial Partial/metabolism , Mice , Mice, Knockout
7.
PLoS Biol ; 19(5): e3000988, 2021 05.
Article in English | MEDLINE | ID: mdl-33979328

ABSTRACT

Although visceral adipocytes located within the body's central core are maintained at approximately 37°C, adipocytes within bone marrow, subcutaneous, and dermal depots are found primarily within the peripheral shell and generally exist at cooler temperatures. Responses of brown and beige/brite adipocytes to cold stress are well studied; however, comparatively little is known about mechanisms by which white adipocytes adapt to temperatures below 37°C. Here, we report that adaptation of cultured adipocytes to 31°C, the temperature at which distal marrow adipose tissues and subcutaneous adipose tissues often reside, increases anabolic and catabolic lipid metabolism, and elevates oxygen consumption. Cool adipocytes rely less on glucose and more on pyruvate, glutamine, and, especially, fatty acids as energy sources. Exposure of cultured adipocytes and gluteal white adipose tissue (WAT) to cool temperatures activates a shared program of gene expression. Cool temperatures induce stearoyl-CoA desaturase-1 (SCD1) expression and monounsaturated lipid levels in cultured adipocytes and distal bone marrow adipose tissues (BMATs), and SCD1 activity is required for acquisition of maximal oxygen consumption at 31°C.


Subject(s)
Adipocytes, White/metabolism , Body Temperature Regulation/physiology , Adaptation, Physiological , Adipocytes/metabolism , Adipocytes/physiology , Adipocytes, Brown/metabolism , Adipocytes, White/physiology , Adipose Tissue/metabolism , Adipose Tissue, White/metabolism , Animals , Cold Temperature , Fatty Acids/metabolism , Female , Lipid Metabolism/physiology , Male , Mice , Mice, Inbred C57BL , Oxygen Consumption , Rats , Rats, Sprague-Dawley , Stearoyl-CoA Desaturase/metabolism
8.
Diabetes ; 70(2): 400-414, 2021 02.
Article in English | MEDLINE | ID: mdl-33214137

ABSTRACT

Mice lacking SH2B1 and humans with variants of SH2B1 display severe obesity and insulin resistance. SH2B1 is an adapter protein that is recruited to the receptors of multiple hormones and neurotrophic factors. Of the four known alternatively spliced SH2B1 isoforms, SH2B1ß and SH2B1γ exhibit ubiquitous expression, whereas SH2B1α and SH2B1δ are essentially restricted to the brain. To understand the roles for SH2B1α and SH2B1δ in energy balance and glucose metabolism, we generated mice lacking these brain-specific isoforms (αδ knockout [αδKO] mice). αδKO mice exhibit decreased food intake, protection from weight gain on standard and high-fat diets, and an adiposity-dependent improvement in glucose homeostasis. SH2B1 has been suggested to impact energy balance via the modulation of leptin action. However, αδKO mice exhibit leptin sensitivity that is similar to that of wild-type mice by multiple measures. Thus, decreasing the abundance of SH2B1α and/or SH2B1δ relative to the other SH2B1 isoforms likely shifts energy balance toward a lean phenotype via a primarily leptin-independent mechanism. Our findings suggest that the different alternatively spliced isoforms of SH2B1 perform different functions in vivo.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Brain/metabolism , Obesity/genetics , Protein Isoforms/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Energy Metabolism/drug effects , Energy Metabolism/genetics , Insulin Resistance/genetics , Leptin/pharmacology , Mice , Mice, Knockout , Obesity/metabolism , Protein Isoforms/metabolism
9.
Mol Metab ; 42: 101078, 2020 12.
Article in English | MEDLINE | ID: mdl-32919095

ABSTRACT

OBJECTIVE: Canonical Wnt/ß-catenin signaling is a well-studied endogenous regulator of mesenchymal cell fate determination, promoting osteoblastogenesis and inhibiting adipogenesis. However, emerging genetic evidence in humans links a number of Wnt pathway members to body fat distribution, obesity, and metabolic dysfunction, suggesting that this pathway also functions in adipocytes. Recent studies in mice have uncovered compelling evidence that the Wnt signaling pathway plays important roles in adipocyte metabolism, particularly under obesogenic conditions. However, complexities in Wnt signaling and differences in experimental models and approaches have thus far limited our understanding of its specific roles in this context. METHODS: To investigate roles of the canonical Wnt pathway in the regulation of adipocyte metabolism, we generated adipocyte-specific ß-catenin (ß-cat) knockout mouse and cultured cell models. We used RNA sequencing, ChIP sequencing, and molecular approaches to assess expression of Wnt targets and lipogenic genes. We then used functional assays to evaluate effects of ß-catenin deficiency on adipocyte metabolism, including lipid and carbohydrate handling. In mice maintained on normal chow and high-fat diets, we assessed the cellular and functional consequences of adipocyte-specific ß-catenin deletion on adipose tissues and systemic metabolism. RESULTS: We report that in adipocytes, the canonical Wnt/ß-catenin pathway regulates de novo lipogenesis (DNL) and fatty acid monounsaturation. Further, ß-catenin mediates effects of Wnt signaling on lipid metabolism in part by transcriptional regulation of Mlxipl and Srebf1. Intriguingly, adipocyte-specific loss of ß-catenin is sensed and defended by CD45-/CD31- stromal cells to maintain tissue-wide Wnt signaling homeostasis in chow-fed mice. With long-term high-fat diet, this compensatory mechanism is overridden, revealing that ß-catenin deletion promotes resistance to diet-induced obesity and adipocyte hypertrophy and subsequent protection from metabolic dysfunction. CONCLUSIONS: Taken together, our studies demonstrate that Wnt signaling in adipocytes is required for lipogenic gene expression, de novo lipogenesis, and lipid desaturation. In addition, adipose tissues rigorously defend Wnt signaling homeostasis under standard nutritional conditions, such that stromal-vascular cells sense and compensate for adipocyte-specific loss. These findings underscore the critical importance of this pathway in adipocyte lipid metabolism and adipose tissue function.


Subject(s)
Adipocytes/metabolism , Adipose Tissue/metabolism , Wnt Signaling Pathway/physiology , Adipocytes/physiology , Adipogenesis/physiology , Adipose Tissue/physiology , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Cell Differentiation , Cells, Cultured , Gene Expression/genetics , Gene Expression Regulation/genetics , Lipid Metabolism , Lipogenesis/physiology , Mice , Mice, Knockout , Obesity , Sterol Regulatory Element Binding Protein 1 , Stromal Cells/metabolism , Wnt Signaling Pathway/drug effects , Wnt Signaling Pathway/genetics , Wnt1 Protein/metabolism , beta Catenin/genetics , beta Catenin/metabolism
10.
Mol Metab ; 39: 100992, 2020 09.
Article in English | MEDLINE | ID: mdl-32325263

ABSTRACT

OBJECTIVE: Obesity is a key risk factor for many secondary chronic illnesses, including type 2 diabetes and cardiovascular disease. Canonical Wnt/ß-catenin signaling is established as an important endogenous inhibitor of adipogenesis. This pathway is operative in mature adipocytes; however, its roles in this context remain unclear due to complexities of Wnt signaling and differences in experimental models. In this study, we used novel cultured cell and mouse models to investigate functional roles of Wnts secreted from adipocytes. METHODS: We generated adipocyte-specific Wntless (Wls) knockout mice and cultured cell models to investigate molecular and metabolic consequences of disrupting Wnt secretion from mature adipocytes. To characterize Wls-deficient cultured adipocytes, we evaluated the expression of Wnt target and lipogenic genes and the downstream functional effects on carbohydrate and lipid metabolism. We also investigated the impact of adipocyte-specific Wls deletion on adipose tissues and global glucose metabolism in mice fed normal chow or high-fat diets. RESULTS: Many aspects of the Wnt signaling apparatus are expressed and operative in mature adipocytes, including the Wnt chaperone Wntless. Deletion of Wntless in cultured adipocytes results in the inhibition of de novo lipogenesis and lipid monounsaturation, likely through repression of Srebf1 (SREBP1c) and Mlxipl (ChREBP) and impaired cleavage of immature SREBP1c into its active form. Adipocyte-specific Wls knockout mice (Wls-/-) have lipogenic gene expression in adipose tissues and isolated adipocytes similar to that of controls when fed a normal chow diet. However, closer investigation reveals that a subset of Wnts and downstream signaling targets are upregulated within stromal-vascular cells of Wls-/- mice, suggesting that adipose tissues defend loss of Wnt secretion from adipocytes. Interestingly, this compensation is lost with long-term high-fat diet challenges. Thus, after six months of a high-fat diet, Wls-/- mice are characterized by decreased adipocyte lipogenic gene expression, reduced visceral adiposity, and improved glucose homeostasis. CONCLUSIONS: Taken together, these studies demonstrate that adipocyte-derived Wnts regulate de novo lipogenesis and lipid desaturation and coordinate the expression of lipogenic genes in adipose tissues. In addition, we report that Wnt signaling within adipose tissues is defended, such that a loss of Wnt secretion from adipocytes is sensed and compensated for by neighboring stromal-vascular cells. With chronic overnutrition, this compensatory mechanism is lost, revealing that Wls-/- mice are resistant to diet-induced obesity, adipocyte hypertrophy, and metabolic dysfunction.


Subject(s)
Adipocytes/metabolism , Gene Expression Regulation , Lipogenesis/genetics , Metabolic Diseases/etiology , Metabolic Diseases/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Biomarkers , Cells, Cultured , Diet/adverse effects , Disease Models, Animal , Disease Susceptibility , Glucose/metabolism , Immunohistochemistry , Insulin/metabolism , Lipid Metabolism/genetics , Metabolic Diseases/diagnosis , Mice , Mice, Knockout , Receptors, G-Protein-Coupled/genetics , Wnt Signaling Pathway
11.
J Biol Chem ; 294(48): 18408-18420, 2019 11 29.
Article in English | MEDLINE | ID: mdl-31615896

ABSTRACT

Although adipogenesis is mainly controlled by a small number of master transcription factors, including CCAAT/enhancer-binding protein family members and peroxisome proliferator-activated receptor γ (PPARγ), other transcription factors also are involved in this process. Thyroid cancer cells expressing a paired box 8 (PAX8)-PPARγ fusion oncogene trans-differentiate into adipocyte-like cells in the presence of the PPARγ ligand pioglitazone, but this trans-differentiation is inhibited by the transcription factor NK2 homeobox 1 (NKX2-1). Here, we tested whether NKX family members may play a role also in normal adipogenesis. Using quantitative RT-PCR (RT-qPCR), we examined the expression of all 14 NKX family members during 3T3-L1 adipocyte differentiation. We found that most NKX members, including NKX2-1, are expressed at very low levels throughout differentiation. However, mRNA and protein expression of a related family member, NKX1-2, was induced during adipocyte differentiation. NKX1-2 also was up-regulated in cultured murine ear mesenchymal stem cells (EMSCs) during adipogenesis. Importantly, shRNA-mediated NKX1-2 knockdown in 3T3-L1 preadipocytes or EMSCs almost completely blocked adipocyte differentiation. Furthermore, NKX1-2 overexpression promoted differentiation of the ST2 bone marrow-derived mesenchymal precursor cell line into adipocytes. Additional findings suggested that NKX1-2 promotes adipogenesis by inhibiting expression of the antiadipogenic protein COUP transcription factor II. Bone marrow mesenchymal precursor cells can differentiate into adipocytes or osteoblasts, and we found that NKX1-2 both promotes ST2 cell adipogenesis and inhibits their osteoblastogenic differentiation. These results support a role for NKX1-2 in promoting adipogenesis and possibly in regulating the balance between adipocyte and osteoblast differentiation of bone marrow mesenchymal precursor cells.


Subject(s)
Adipocytes/metabolism , Adipogenesis/genetics , Cell Differentiation/genetics , Homeodomain Proteins/genetics , Nuclear Proteins/genetics , Osteoblasts/metabolism , Transcription Factors/genetics , 3T3-L1 Cells , Adipocytes/cytology , Animals , Cell Line , Cells, Cultured , Gene Expression Regulation , HEK293 Cells , Homeodomain Proteins/metabolism , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Nuclear Proteins/metabolism , Osteoblasts/cytology , PAX8 Transcription Factor/genetics , PAX8 Transcription Factor/metabolism , PPAR gamma/genetics , PPAR gamma/metabolism , RNA Interference , Transcription Factors/metabolism
12.
J Vis Exp ; (149)2019 07 11.
Article in English | MEDLINE | ID: mdl-31355801

ABSTRACT

Adipose tissues are complex organs with a wide array of functions, including storage and mobilization of energy in response to local and global needs, uncoupling of metabolism to generate heat, and secretion of adipokines to regulate whole-body homeostasis and immune responses. Emerging research is identifying important regional differences in the developmental, molecular, and functional profiles of adipocytes located in discrete depots throughout the body. Different properties of the depots are medically relevant since metabolic diseases often demonstrate depot-specific effects. This protocol will provide investigators with a detailed anatomic atlas and dissection guide for the reproducible and accurate identification and excision of diverse mouse adipose tissues. Standardized dissection of discrete adipose depots will allow detailed comparisons of their molecular and metabolic characteristics and contributions to local and systemic pathologic states under various nutritional and environmental conditions.


Subject(s)
Adipocytes/metabolism , Adipose Tissue/metabolism , Dissection/methods , Obesity/metabolism , Animals , Humans , Mice
13.
Nutrients ; 11(6)2019 Jun 21.
Article in English | MEDLINE | ID: mdl-31234301

ABSTRACT

Overnutrition during critical windows of development plays a significant role in life-long metabolic disease risk. Early exposure to excessive nutrition may result in altered programming leading to increased susceptibility to obesity, inflammation, and metabolic complications. This study investigated the programming effects of high-fat diet (HFD) exposure during the lactation period on offspring adiposity and inflammation. Female C57Bl/6J dams were fed a normal diet or a 60% HFD during lactation. Offspring were weaned onto a normal diet until 12 weeks of age when half were re-challenged with HFD for 12 weeks. Metabolic testing was performed throughout adulthood. At 24 weeks, adipose depots were isolated and evaluated for macrophage profiling and inflammatory gene expression. Males exposed to HFD during lactation had insulin resistance and glucose intolerance as adults. After re-introduction to HFD, males had increased weight gain and worsened insulin resistance and hyperglycemia. There was increased infiltration of pro-inflammatory CD11c+ adipose tissue macrophages, and bone marrow was primed to produce granulocytes and macrophages. Bone density was lower due to enhanced marrow adiposity. This study demonstrates that maternal HFD exposure during the lactational window programs offspring adiposity, inflammation, and impaired glucose homeostasis.


Subject(s)
Adipose Tissue/physiopathology , Adiposity , Bone Marrow/physiopathology , Diet, High-Fat/adverse effects , Hyperglycemia/etiology , Inflammation/etiology , Lactation , Maternal Exposure/adverse effects , Maternal Nutritional Physiological Phenomena , Obesity/etiology , Adipose Tissue/metabolism , Age Factors , Animals , Biomarkers/blood , Blood Glucose/metabolism , Bone Marrow/metabolism , Female , Hyperglycemia/blood , Hyperglycemia/physiopathology , Inflammation/blood , Inflammation/physiopathology , Inflammation Mediators/blood , Insulin Resistance , Male , Mice, Inbred C57BL , Myeloid Cells/metabolism , Nutritional Status , Obesity/blood , Obesity/physiopathology , Risk Factors , Sex Factors , Time Factors , Weight Gain
14.
J Clin Invest ; 129(6): 2404-2416, 2019 05 06.
Article in English | MEDLINE | ID: mdl-31063988

ABSTRACT

Bariatric surgeries are integral to the management of obesity and its metabolic complications. However, these surgeries cause bone loss and increase fracture risk through poorly understood mechanisms. In a mouse model, vertical sleeve gastrectomy (VSG) caused trabecular and cortical bone loss that was independent of sex, body weight, and diet, and this loss was characterized by impaired osteoid mineralization and bone formation. VSG had a profound effect on the bone marrow niche, with rapid loss of marrow adipose tissue, and expansion of myeloid cellularity, leading to increased circulating neutrophils. Following VSG, circulating granulocyte-colony stimulating factor (G-CSF) was increased in mice, and was transiently elevated in a longitudinal study of humans. Elevation of G-CSF was found to recapitulate many effects of VSG on bone and the marrow niche. In addition to stimulatory effects of G-CSF on myelopoiesis, endogenous G-CSF suppressed development of marrow adipocytes and hindered accrual of peak cortical and trabecular bone. Effects of VSG on induction of neutrophils and depletion of marrow adiposity were reduced in mice deficient for G-CSF; however, bone mass was not influenced. Although not a primary mechanism for bone loss with VSG, G-CSF plays an intermediary role for effects of VSG on the bone marrow niche.


Subject(s)
Adipocytes/metabolism , Bone Marrow Cells/metabolism , Bone Resorption/blood , Gastroplasty , Granulocyte Colony-Stimulating Factor/blood , Obesity/blood , Postoperative Complications/blood , Adipocytes/pathology , Adolescent , Adult , Animals , Bone Marrow/pathology , Bone Marrow Cells/pathology , Bone Resorption/etiology , Bone Resorption/genetics , Bone Resorption/pathology , Female , Gastrectomy , Humans , Longitudinal Studies , Mice , Mice, Knockout , Obesity/genetics , Obesity/pathology , Obesity/surgery , Postoperative Complications/genetics , Postoperative Complications/pathology
15.
Article in English | MEDLINE | ID: mdl-30564437

ABSTRACT

Sri Lanka is experiencing a rapid increase in the number of people with diabetes mellitus (DM) due to population growth and aging. Physician shortages, outdated technology, and insufficient health education have contributed to the difficulties associated with managing the burden of disease. New models of chronic disease management are needed to address the increasing prevalence of DM. Medical students, business students, and faculty members from the University of Michigan partnered with the Grace Girls' Home, Trincomalee General Hospital, and Selvanayakapuram Central Hospital to identify and train diabetes-focused medical assistants (MAs) to collect and enter patient data and educate patients about their disease. Return visits to these MAs were encouraged so that patient progress and disease progression could be tracked longitudinally. Data entry was conducted through a cloud-based mechanism, facilitating patient management and descriptive characterization of the population. We implemented this pilot program in June 2016 in coordination with Trincomalee General Hospital and Selvanayakapuram Central Hospital. Over a 12-month period, 93 patients were systematically assessed by the medical assistants. All patients received education and were provided materials after the visit to better inform them about the importance of controlling their disease. Fifteen percent (14/93) of patients returned for follow-up consultation. Trained MAs have the potential to provide support to physicians working in congested health systems in low-resource settings. Public investment in training programs for MAs and greater acceptance by physicians and patients will be essential for handling the growing burden associated with chronic illnesses like DM. Trained MAs may also play a role in improved patient education and awareness regarding diabetes self-management.

16.
Cell Metab ; 27(1): 264-264.e1, 2018 01 09.
Article in English | MEDLINE | ID: mdl-29320707

ABSTRACT

Adipose tissues are complex organs, with central roles in energy homeostasis as well as local functions. Adipocytes develop in diverse, discrete locations throughout the body. Important regional differences in adipocytes exist, and diseases that affect adipose tissues often demonstrate depot-specific effects. Herein, we depict the widespread locations of major and minor rodent adipose depots. Depot-specific molecular and functional characteristics will be described in Part II.


Subject(s)
Adipocytes/cytology , Adipose Tissue/cytology , Animals , Humans , Mice
17.
Cell Metab ; 27(1): 266-266.e1, 2018 01 09.
Article in English | MEDLINE | ID: mdl-29320708

ABSTRACT

The intrinsic cellular and metabolic properties of an adipocyte are shaped by the specific niche in which it resides. The diverse and discrete locations of major and minor rodent adipose depots are depicted in Part I. In Part II, the molecular and functional characteristics of four major types of adipocytes are described. Identified functions of relatively understudied but undoubtedly important depots are also highlighted.


Subject(s)
Adipocytes/cytology , Adipocytes/metabolism , Adipogenesis/genetics , Adipose Tissue/cytology , Animals , Humans , Mice , Organ Specificity
18.
Bone ; 110: 134-140, 2018 05.
Article in English | MEDLINE | ID: mdl-29343445

ABSTRACT

Most adipocytes exist in discrete depots throughout the body, notably in well-defined white and brown adipose tissues. However, adipocytes also reside within specialized niches, of which the most abundant is within bone marrow. Whereas bone marrow adipose tissue (BMAT) shares many properties in common with white adipose tissue, the distinct functions of BMAT are reflected by its development, regulation, protein secretion, and lipid composition. In addition to its potential role as a local energy reservoir, BMAT also secretes proteins, including adiponectin, RANK ligand, dipeptidyl peptidase-4, and stem cell factor, which contribute to local marrow niche functions and which may also influence global metabolism. The characteristics of BMAT are also distinct depending on whether marrow adipocytes are contained within yellow or red marrow, as these can be thought of as 'constitutive' and 'regulated', respectively. The rBMAT for instance can be expanded or depleted by myriad factors, including age, nutrition, endocrine status and pharmaceuticals. Herein we review the site specificity, age-related development, regulation and metabolic characteristics of BMAT under various metabolic conditions, including the functional interactions with bone and hematopoietic cells.


Subject(s)
Adipose Tissue/growth & development , Adipose Tissue/metabolism , Bone Marrow/growth & development , Bone Marrow/metabolism , Adipocytes/cytology , Adipokines/metabolism , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Animals , Bone Marrow Cells/metabolism , Caloric Restriction , Female , Hematopoietic Stem Cells/cytology , Humans , Lipids/chemistry , Male , Mice , Mice, Inbred C3H , Physical Conditioning, Animal
19.
J Biol Chem ; 292(22): 9034-9050, 2017 06 02.
Article in English | MEDLINE | ID: mdl-28373279

ABSTRACT

The accumulation of α-synuclein (α-syn) fibrils in neuronal inclusions is the defining pathological process in Parkinson's disease (PD). A pathogenic role for α-syn fibril accumulation is supported by the identification of dominantly inherited α-syn (SNCA) gene mutations in rare cases of familial PD. Fibril formation involves a spontaneous nucleation event in which soluble α-syn monomers associate to form seeds, followed by fibril growth during which monomeric α-syn molecules sequentially associate with existing seeds. To better investigate this process, we developed sensitive assays that use the fluorescein arsenical dye FlAsH (fluorescein arsenical hairpin binder) to detect soluble oligomers and mature fibrils formed from recombinant α-syn protein containing an N-terminal bicysteine tag (C2-α-syn). Using seed growth by monomer association (SeGMA) assays to measure fibril growth over 3 h in the presence of C2-α-syn monomer, we observed that some familial PD-associated α-syn mutations (i.e. H50Q and A53T) greatly increased growth rates, whereas others (E46K, A30P, and G51D) decreased growth rates. Experiments with wild-type seeds extended by mutant monomer and vice versa revealed that single-amino acid differences between seed and monomer proteins consistently decreased growth rates. These results demonstrate that α-syn monomer association during fibril growth is a highly ordered process that can be disrupted by misalignment of individual amino acids and that only a subset of familial-PD mutations causes fibril accumulation through increased fibril growth rates. The SeGMA assays reported herein can be utilized to further elucidate structural requirements of α-syn fibril growth and to identify growth inhibitors as a potential therapeutic approach in PD.


Subject(s)
Amyloid/chemistry , Mutation, Missense , Parkinson Disease , Protein Aggregation, Pathological , alpha-Synuclein/chemistry , Amino Acid Substitution , Amyloid/genetics , Amyloid/metabolism , Humans , alpha-Synuclein/genetics , alpha-Synuclein/metabolism
20.
J Med Chem ; 58(15): 6002-17, 2015 Aug 13.
Article in English | MEDLINE | ID: mdl-26177091

ABSTRACT

A series of 3-(benzylidine)indolin-2-one derivatives were synthesized and evaluated for their in vitro binding to alpha synuclein (α-syn), beta amyloid (Aß), and tau fibrils. Compounds with a single double bond in the 3-position had only a modest affinity for α-syn and no selectivity for α-syn versus Aß or tau fibrils. Homologation to the corresponding diene analogues yielded a mixture of Z,E and E,E isomers; substitution of the indoline nitrogen with an N-benzyl group resulted in increased binding to α-syn and reasonable selectivity for α-syn versus Aß and tau. Introduction of a para-nitro group into the benzene ring of the diene enabled separation of the Z,E and E,E isomers and led to the identification of the Z,E configuration as the more active regioisomer. The data described here provide key structural information in the design of probes which bind preferentially to α-syn versus Aß or tau fibrils.


Subject(s)
Drug Design , Indoles/chemistry , alpha-Synuclein/chemistry , Indoles/chemical synthesis , Ligands , Microscopy, Fluorescence
SELECTION OF CITATIONS
SEARCH DETAIL
...