Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters











Publication year range
1.
J Physiol ; 600(16): 3795-3817, 2022 08.
Article in English | MEDLINE | ID: mdl-35844058

ABSTRACT

Adipose tissue secretes numerous cytokines (termed 'adipokines') that have known or hypothesized actions on skeletal muscle. The majority of adipokines have been implicated in the pathological link between excess adipose and muscle insulin resistance, but approximately half also have documented in vitro effects on myogenesis and/or hypertrophy. This complexity suggests a potential dual role for adipokines in the regulation of muscle mass in homeostasis and the development of pathology. In this study, we used lipodystrophic 'fat-free' mice to demonstrate that adipose tissue is indeed necessary for the development of normal muscle mass and strength. Fat-free mice had significantly reduced mass (∼15%) and peak contractile tension (∼20%) of fast-twitch muscles, a slowing of contractile dynamics and decreased cross-sectional area of fast twitch fibres compared to wild-type littermates. These deficits in mass and contractile tension were fully rescued by reconstitution of ∼10% of normal adipose mass, indicating that this phenotype is the direct consequence of absent adipose. We then showed that the rescue is solely mediated by the adipokine leptin, as similar reconstitution of adipose from leptin-knockout mice fails to rescue mass or strength. Together, these data indicate that the development of muscle mass and strength in wild-type mice is dependent on adipose-secreted leptin. This finding extends our current understanding of the multiple roles of adipokines in physiology as well as disease pathophysiology to include a critical role for the adipokine leptin in muscle homeostasis. KEY POINTS: Adipose-derived cytokines (adipokines) have long been implicated in the pathogenesis of insulin resistance in obesity but likely have other under-appreciated roles in muscle physiology. Here we use a fat-free mouse to show that adipose tissue is necessary for the normal development of muscle mass and strength. Through add-back of genetically modified adipose tissue we show that leptin is the key adipokine mediating this regulation. This expands our understanding of leptin's role in adipose-muscle signalling to include development and homeostasis and adds the surprising finding that leptin is the sole mediator of the maintenance of muscle mass and strength by adipose tissue.


Subject(s)
Insulin Resistance , Leptin , Adipokines , Adipose Tissue/physiology , Animals , Cytokines , Mice , Muscle, Skeletal
2.
Elife ; 102021 08 11.
Article in English | MEDLINE | ID: mdl-34378533

ABSTRACT

Bone marrow adipocytes accumulate with age and in diverse disease states. However, their origins and adaptations in these conditions remain unclear, impairing our understanding of their context-specific endocrine functions and relationship with surrounding tissues. In this study, by analyzing bone and adipose tissues in the lipodystrophic 'fat-free' mouse, we define a novel, secondary adipogenesis pathway that relies on the recruitment of adiponectin-negative stromal progenitors. This pathway is unique to the bone marrow and is activated with age and in states of metabolic stress in the fat-free mouse model, resulting in the expansion of bone marrow adipocytes specialized for lipid storage with compromised lipid mobilization and cytokine expression within regions traditionally devoted to hematopoiesis. This finding further distinguishes bone marrow from peripheral adipocytes and contributes to our understanding of bone marrow adipocyte origins, adaptations, and relationships with surrounding tissues with age and disease.


Subject(s)
Adipocytes/physiology , Adipogenesis/physiology , Bone Marrow/physiology , Hematopoiesis/physiology , Adipocytes/metabolism , Adipose Tissue/metabolism , Adipose Tissue/physiology , Age Factors , Animals , Bone Marrow/metabolism , Bone Marrow Cells/metabolism , Bone Marrow Cells/physiology , Bone and Bones/metabolism , Bone and Bones/physiology , Cell Differentiation , Female , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Osteoblasts/metabolism , Osteoblasts/physiology
3.
Sci Rep ; 11(1): 14560, 2021 07 15.
Article in English | MEDLINE | ID: mdl-34267289

ABSTRACT

Lipodystrophic mice are protected from cartilage damage following joint injury. This protection can be reversed by the implantation of a small adipose tissue graft. The purpose of this study was to evaluate the relationship between the gut microbiota and knee cartilage damage while controlling for adiposity, high fat diet, and joint injury using lipodystrophic (LD) mice. LD and littermate control (WT) mice were fed a high fat diet, chow diet, or were rescued with fat implantation, then challenged with destabilization of the medial meniscus surgery to induce osteoarthritis (OA). 16S rRNA sequencing was conducted on feces. MaAslin2 was used to determine associations between taxonomic relative abundance and OA severity. While serum LPS levels between groups were similar, synovial fluid LPS levels were increased in both limbs of HFD WT mice compared to all groups, except for fat transplanted animals. The Bacteroidetes:Firmicutes ratio of the gut microbiota was significantly reduced in HFD and OA-rescued animals when compared to chow. Nine novel significant associations were found between gut microbiota taxa and OA severity. These findings suggest the presence of causal relationships the gut microbiome and cartilage health, independent of diet or adiposity, providing potential therapeutic targets through manipulation of the microbiome.


Subject(s)
Cartilage/physiopathology , Diet, High-Fat/adverse effects , Gastrointestinal Microbiome/physiology , Osteoarthritis/microbiology , Adiposity , Animals , Bacteroidetes/genetics , Female , Firmicutes/genetics , Gastrointestinal Microbiome/genetics , Lipodystrophy/microbiology , Lipopolysaccharides/blood , Male , Meniscus/surgery , Mice, Transgenic , Obesity/microbiology , Osteoarthritis/etiology , RNA, Ribosomal, 16S/genetics , Synovial Fluid/metabolism
4.
Proc Natl Acad Sci U S A ; 118(1)2021 01 05.
Article in English | MEDLINE | ID: mdl-33443201

ABSTRACT

Osteoarthritis (OA), the leading cause of pain and disability worldwide, disproportionally affects individuals with obesity. The mechanisms by which obesity leads to the onset and progression of OA are unclear due to the complex interactions among the metabolic, biomechanical, and inflammatory factors that accompany increased adiposity. We used a murine preclinical model of lipodystrophy (LD) to examine the direct contribution of adipose tissue to OA. Knee joints of LD mice were protected from spontaneous or posttraumatic OA, on either a chow or high-fat diet, despite similar body weight and the presence of systemic inflammation. These findings indicate that adipose tissue itself plays a critical role in the pathophysiology of OA. Susceptibility to posttraumatic OA was reintroduced into LD mice using implantation of a small adipose tissue depot derived from wild-type animals or mouse embryonic fibroblasts that undergo spontaneous adipogenesis, implicating paracrine signaling from fat, rather than body weight, as a mediator of joint degeneration.


Subject(s)
Adipose Tissue/metabolism , Lipodystrophy/metabolism , Osteoarthritis, Knee/metabolism , Adipose Tissue/physiopathology , Adipose Tissue/transplantation , Adiposity , Animals , Body Weight , Cartilage/pathology , Cytokines/metabolism , Diet, High-Fat/adverse effects , Disease Models, Animal , Disease Susceptibility/complications , Disease Susceptibility/metabolism , Female , Fibroblasts/metabolism , Hyperplasia/complications , Inflammation/metabolism , Lipodystrophy/diagnostic imaging , Lipodystrophy/genetics , Lipodystrophy/physiopathology , Locomotion , Male , Mice , Muscle Strength , Osteoarthritis, Knee/complications , Osteoarthritis, Knee/diagnostic imaging , Osteoarthritis, Knee/prevention & control , Pain/complications , Paracrine Communication/physiology
5.
JCI Insight ; 5(17)2020 09 03.
Article in English | MEDLINE | ID: mdl-32879136

ABSTRACT

Obesity predisposes to cancer and a virtual universality of nonalcoholic fatty liver disease (NAFLD). However, the impact of hepatic steatosis on liver metastasis is enigmatic. We find that while control mice were relatively resistant to hepatic metastasis, those which were lipodystrophic or obese, with NAFLD, had a dramatic increase in breast cancer and melanoma liver metastases. NAFLD promotes liver metastasis by reciprocal activation initiated by tumor-induced triglyceride lipolysis in juxtaposed hepatocytes. The lipolytic products are transferred to cancer cells via fatty acid transporter protein 1, where they are metabolized by mitochondrial oxidation to promote tumor growth. The histology of human liver metastasis indicated the same occurs in humans. Furthermore, comparison of isolates of normal and fatty liver established that steatotic lipids had enhanced tumor-stimulating capacity. Normalization of glucose metabolism by metformin did not reduce steatosis-induced metastasis, establishing the process is not mediated by the metabolic syndrome. Alternatively, eradication of NAFLD in lipodystrophic mice by adipose tissue transplantation reduced breast cancer metastasis to that of control mice, indicating the steatosis-induced predisposition is reversible.


Subject(s)
Lipolysis , Liver Neoplasms/metabolism , Non-alcoholic Fatty Liver Disease/complications , Animals , Female , Glucose/metabolism , Hep G2 Cells , Hepatocytes/metabolism , Humans , Liver Neoplasms/complications , Liver Neoplasms/pathology , Mice , Mice, Inbred C57BL , Mitochondria, Liver/metabolism , Neoplasm Metastasis
6.
J Physiol ; 598(13): 2669-2683, 2020 07.
Article in English | MEDLINE | ID: mdl-32358797

ABSTRACT

KEY POINTS: Muscle infiltration with adipose tissue (IMAT) is common and associated with loss of skeletal muscle strength and physical function across a diverse set of pathologies. Whether the association between IMAT and muscle weakness is causative or simply correlative remains an open question that needs to be addressed to effectively guide muscle strengthening interventions in people with increased IMAT. In the present studies, we demonstrate that IMAT deposition causes decreased muscle strength using mouse models. These findings indicate IMAT is a novel therapeutic target for muscle dysfunction. ABSTRACT: Intramuscular adipose tissue (IMAT) is associated with deficits in strength and physical function across a wide array of conditions, from injury to ageing to metabolic disease. Due to the diverse aetiologies of the primary disorders involving IMAT and the strength of the associations, it has long been proposed that IMAT directly contributes to this muscle dysfunction. However, infiltration of IMAT and reduced strength could both be driven by muscle disuse, injury and systemic disease, making IMAT simply an 'innocent bystander.' Here, we utilize novel mouse models to evaluate the direct effect of IMAT on muscle contraction. First, we utilize intramuscular glycerol injection in wild-type mice to evaluate IMAT in the absence of systemic disease. In this model we find that, in isolation from the neuromuscular and circulatory systems, there remains a muscle-intrinsic association between increased IMAT volume and decreased contractile tension (r2  > 0.5, P < 0.01) that cannot be explained by reduction in contractile material. Second, we utilize a lipodystrophic mouse model which cannot generate adipocytes to 'rescue' the deficits. We demonstrate that without IMAT infiltration, glycerol treatment does not reduce contractile force (P > 0.8). Taken together, this indicates that IMAT is not an inert feature of muscle pathology but rather has a direct impact on muscle contraction. This finding suggests that novel strategies targeting IMAT may improve muscle strength and function in a number of populations.


Subject(s)
Adipose Tissue , Muscle Contraction , Adipocytes , Animals , Mice , Muscle Strength , Muscle, Skeletal
7.
Endocrinology ; 161(2)2020 02 01.
Article in English | MEDLINE | ID: mdl-32036385

ABSTRACT

Increased visceral adiposity and hyperglycemia, 2 characteristics of metabolic syndrome, are also present in conditions of excess glucocorticoids (GCs). GCs are hormones thought to act primarily via the glucocorticoid receptor (GR). GCs are commonly prescribed for inflammatory disorders, yet their use is limited due to many adverse metabolic side effects. In addition to GR, GCs also bind the mineralocorticoid receptor (MR), but there are many conflicting studies about the exact role of MR in metabolic disease. Using MR knockout mice (MRKO), we find that both white and brown adipose depots form normally when compared with wild-type mice at P5. We created mice with adipocyte-specific deletion of MR (FMRKO) to better understand the role of MR in metabolic dysfunction. Treatment of mice with excess GCs for 4 weeks, via corticosterone in drinking water, induced increased fat mass and glucose intolerance to similar levels in FMRKO and floxed control mice. Separately, when fed a high-fat diet for 16 weeks, FMRKO mice had reduced body weight, fat mass, and hepatic steatosis, relative to floxed control mice. Decreased adiposity likely resulted from increased energy expenditure since food intake was not different. RNA sequencing analysis revealed decreased enrichment of genes associated with adipogenesis in inguinal white adipose of FMRKO mice. Differentiation of mouse embryonic fibroblasts (MEFs) showed modestly impaired adipogenesis in MRKO MEFs compared with wild type, but this was rescued upon the addition of peroxisome proliferator-activated receptor gamma (PPARγ) agonist or PPARγ overexpression. Collectively, these studies provide further evidence supporting the potential value of MR as a therapeutic target for conditions associated with metabolic syndrome.


Subject(s)
Adipocytes/metabolism , Adipogenesis , Metabolic Syndrome/etiology , Obesity, Abdominal/etiology , Receptors, Mineralocorticoid/metabolism , Animals , Energy Metabolism , Glucocorticoids , Male , Metabolic Syndrome/metabolism , Mice, Inbred C57BL , Mice, Knockout , Obesity, Abdominal/metabolism , PPAR gamma
8.
Sci Rep ; 9(1): 17427, 2019 11 22.
Article in English | MEDLINE | ID: mdl-31758074

ABSTRACT

Adipocytes within the skeleton are collectively termed bone marrow adipose tissue (BMAT). BMAT contributes to peripheral and local metabolism, however, its capacity for cell-autonomous expression of uncoupling protein 1 (UCP1), a biomarker of beige and brown adipogenesis, remains unclear. To overcome this, Ucp1-Cre was used to drive diphtheria toxin expression in cells expressing UCP1 (Ucp1Cre+/DTA+). Despite loss of brown adipose tissue, BMAT volume was not reduced in Ucp1Cre+/DTA+ mice. Comparably, in mTmG reporter mice (Ucp1Cre+/mTmG+), Ucp1-Cre expression was absent from BMAT in young (3-weeks) and mature (16-weeks) male and female mice. Further, ß3-agonist stimulation failed to induce Ucp1-Cre expression in BMAT. This demonstrates that BMAT adipocytes are not UCP1-expressing beige/brown adipocytes. Thus, to identify novel and emerging roles for BMAT adipocytes in skeletal and whole-body homeostasis, we performed gene enrichment analysis of microarray data from adipose tissues of adult rabbits. Pathway analysis revealed genetic evidence for differences in BMAT including insulin resistance, decreased fatty acid metabolism, and enhanced contributions to local processes including bone mineral density through candidate genes such as osteopontin. In sum, this supports a paradigm by which BMAT adipocytes are a unique subpopulation that is specialized to support cells within the skeletal and hematopoietic niche.


Subject(s)
Adipocytes/metabolism , Adipose Tissue/metabolism , Bone Marrow/metabolism , Gene Expression , Uncoupling Protein 1/genetics , Adipose Tissue/pathology , Adrenergic Agents/pharmacology , Animals , Bone Marrow/pathology , Cell Lineage/genetics , Female , Fluorescent Antibody Technique , Gene Expression Regulation/drug effects , Male , Mice , Skeleton/metabolism
9.
Immunity ; 51(1): 119-130.e5, 2019 07 16.
Article in English | MEDLINE | ID: mdl-31231034

ABSTRACT

Tissue-resident macrophages require specific milieus for the maintenance of defining gene-expression programs. Expression of the transcription factor GATA6 is required for the homeostasis, function and localization of peritoneal cavity-resident macrophages. Gata6 expression is maintained in a non-cell autonomous manner and is elicited by the vitamin A metabolite, retinoic acid. Here, we found that the GATA6 transcriptional program is a common feature of macrophages residing in all visceral body cavities. Retinoic acid-dependent and -independent hallmark genes of GATA6+ macrophages were induced by mesothelial and fibroblastic stromal cells that express the transcription factor Wilms' Tumor 1 (WT1), which drives the expression of two rate-limiting enzymes in retinol metabolism. Depletion of Wt1+ stromal cells reduced the frequency of GATA6+ macrophages in the peritoneal, pleural and pericardial cavities. Thus, Wt1+ mesothelial and fibroblastic stromal cells constitute essential niche components supporting the tissue-specifying transcriptional landscape and homeostasis of cavity-resident macrophages.


Subject(s)
GATA6 Transcription Factor/metabolism , Macrophages/physiology , Pericardium/immunology , Peritoneal Cavity/physiology , Pleural Cavity/immunology , Repressor Proteins/metabolism , Stromal Cells/physiology , Animals , Cell Differentiation , Cells, Cultured , GATA6 Transcription Factor/genetics , Homeostasis , Mice , Mice, Inbred C57BL , Mice, Transgenic , Repressor Proteins/genetics , Tretinoin/metabolism , WT1 Proteins
10.
PLoS Genet ; 15(6): e1008244, 2019 06.
Article in English | MEDLINE | ID: mdl-31233501

ABSTRACT

Berardinelli-Seip congenital generalized lipodystrophy is associated with increased bone mass suggesting that fat tissue regulates the skeleton. Because there is little mechanistic information regarding this issue, we generated "fat-free" (FF) mice completely lacking visible visceral, subcutaneous and brown fat. Due to robust osteoblastic activity, trabecular and cortical bone volume is markedly enhanced in these animals. FF mice, like Berardinelli-Seip patients, are diabetic but normalization of glucose tolerance and significant reduction in circulating insulin fails to alter their skeletal phenotype. Importantly, the skeletal phenotype of FF mice is completely rescued by transplantation of adipocyte precursors or white or brown fat depots, indicating that adipocyte derived products regulate bone mass. Confirming such is the case, transplantation of fat derived from adiponectin and leptin double knockout mice, unlike that obtained from their WT counterparts, fails to normalize FF bone. These observations suggest a paucity of leptin and adiponectin may contribute to the increased bone mass of Berardinelli-Seip patients.


Subject(s)
Adiponectin/genetics , Leptin/genetics , Lipodystrophy, Congenital Generalized/genetics , Osteosclerosis/genetics , Adipocytes/metabolism , Adipose Tissue, Brown/metabolism , Animals , Bone Density/genetics , Disease Models, Animal , Female , Glucose/genetics , Glucose/metabolism , Humans , Insulin/genetics , Intra-Abdominal Fat/metabolism , Lipodystrophy, Congenital Generalized/complications , Lipodystrophy, Congenital Generalized/pathology , Mice , Mice, Knockout , Osteosclerosis/etiology , Osteosclerosis/metabolism , Osteosclerosis/pathology , Skeleton/metabolism , Skeleton/pathology , Subcutaneous Fat/metabolism
11.
Cell Rep ; 27(10): 2809-2816.e3, 2019 06 04.
Article in English | MEDLINE | ID: mdl-31167128

ABSTRACT

We explored the relationship of obesity and inflammatory arthritis (IA) by selectively expressing diphtheria toxin in adipose tissue yielding "fat-free" (FF) mice completely lacking white and brown fat. FF mice exhibit systemic neutrophilia and elevated serum acute phase proteins suggesting a predisposition to severe IA. Surprisingly, FF mice are resistant to K/BxN serum-induced IA and attendant bone destruction. Despite robust systemic basal neutrophilia, neutrophil infiltration into joints of FF mice does not occur when challenged with K/BxN serum. Absence of adiponectin, leptin, or both has no effect on joint disease, but deletion of the adipokine adipsin (complement factor D) completely prevents serum-induced IA. Confirming that fat-expressed adipsin modulates the disorder, transplantation of wild-type (WT) adipose tissue into FF mice restores susceptibility to IA, whereas recipients of adipsin-deficient fat remain resistant. Thus, adipose tissue regulates development of IA through a pathway in which adipocytes modify neutrophil responses in distant tissues by producing adipsin.


Subject(s)
Adipose Tissue/metabolism , Arthritis/etiology , Arthritis/metabolism , Neutrophils/metabolism , Adipocytes/metabolism , Adipose Tissue/immunology , Animals , Arthritis/chemically induced , Arthritis/immunology , Complement Factor D/genetics , Complement Factor D/metabolism , Inflammation/immunology , Inflammation/metabolism , Leptin/genetics , Leptin/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Neutrophil Infiltration/genetics , Neutrophil Infiltration/immunology , Neutrophils/cytology , Neutrophils/immunology
12.
Article in English | MEDLINE | ID: mdl-30310815

ABSTRACT

Glucocorticoids are steroid hormones that play a key role in metabolic adaptations during stress, such as fasting and starvation, in order to maintain plasma glucose levels. Excess and chronic glucocorticoid exposure, however, causes metabolic syndrome including insulin resistance, dyslipidemia, and hyperglycemia. Studies in animal models of metabolic disorders frequently demonstrate that suppressing glucocorticoid signaling improves insulin sensitivity and metabolic profiles. Glucocorticoids convey their signals through an intracellular glucocorticoid receptor (GR), which is a transcriptional regulator. The adipocyte is one cell type that contributes to whole body metabolic homeostasis under the influence of GR. Glucocorticoids' functions on adipose tissues are complex. Depending on various physiological or pathophysiological states as well as distinct fat depots, glucocorticoids can either increase or decrease lipid storage in adipose tissues. In rodents, glucocorticoids have been shown to reduce the thermogenic activity of brown adipocytes. However, in human acute glucocorticoid exposure, glucocorticoids act to promote thermogenesis. In this article, we will review the recent studies on the mechanisms underlying the complex metabolic functions of GR in adipocytes. These include studies of the metabolic outcomes of adipocyte specific GR knockout mice and identification of novel GR primary target genes that mediate glucocorticoid action in adipocytes.

13.
Endocrinology ; 159(9): 3275-3286, 2018 09 01.
Article in English | MEDLINE | ID: mdl-30085057

ABSTRACT

The global obesity epidemic is fueling alarming rates of diabetes, associated with increased risk of cardiovascular disease and cancer. Leptin is a hormone secreted by adipose tissue that is a key regulator of body weight (BW) and energy expenditure. Leptin-deficient humans and mice are obese, diabetic, and infertile and have hepatic steatosis. Although leptin replacement therapy can alleviate the pathologies seen in leptin-deficient patients and mouse models, treatment is costly and requires daily injections. Because adipocytes are the source of leptin secretion, we investigated whether mouse embryonic fibroblasts (MEFs), capable of forming adipocytes, could be injected into ob/ob mice and prevent the metabolic phenotype seen in these leptin-deficient mice. We performed a single subcutaneous injection of MEFs into leptin-deficient ob/ob mice. The MEF injection formed a single fat pad that is histologically similar to white adipose tissue. The ob/ob mice receiving MEFs (obRs) had significantly lower BW compared with nontreated ob/ob mice, primarily because of decreased adipose tissue mass. Additionally, obR mice had significantly less liver steatosis and greater glucose tolerance and insulin sensitivity. obR mice also manifested lower food intake and greater energy expenditure than ob/ob mice, providing a mechanism underlying their metabolic improvement. Furthermore, obRs have sustained metabolic protection and restoration of fertility. Collectively, our studies show the importance of functional adipocytes in preventing metabolic abnormalities seen in leptin deficiency and point to the possibility of cell-based therapies for the treatment of leptin-deficient states.


Subject(s)
Adipocytes, White/metabolism , Cell Transplantation , Eating , Energy Metabolism , Fatty Liver/metabolism , Fibroblasts/metabolism , Insulin Resistance , Obesity/metabolism , Adipocytes, White/cytology , Adipogenesis , Adipose Tissue, White , Animals , Cell Differentiation , Fibroblasts/cytology , Leptin/genetics , Mice , Mice, Obese , Mutation , Obesity/genetics
14.
J Immunol ; 200(8): 2786-2797, 2018 04 15.
Article in English | MEDLINE | ID: mdl-29531168

ABSTRACT

Factor D (FD) is an essential component of the complement alternative pathway (AP). It is an attractive pharmaceutical target because it is an AP-specific protease circulating in blood. Most components of the complement activation pathways are produced by the liver, but FD is highly expressed by adipose tissue. Two critical questions are: 1) to what degree does adipose tissue contribute to circulating FD levels and 2) what quantity of FD is sufficient to maintain a functional AP? To address these issues, we studied a novel mouse strain with complete lipodystrophy (LD), the fld mouse with partial LD, an FD-deficient mouse, and samples from lipodystrophic patients. FD was undetectable in the serum of LD mice, which also showed minimal AP function. Reconstitution with purified FD, serum mixing experiments, and studies of partial LD mice all demonstrated that a low level of serum FD is sufficient for normal AP activity in the mouse system. This conclusion was further supported by experiments in which wild-type adipose precursors were transplanted into LD mice. Our results indicate that almost all FD in mouse serum is derived from adipose tissue. In contrast, FD levels were reduced ∼50% in the sera of patients with congenital generalized LD. Our studies further demonstrate that a relatively small amount of serum FD is sufficient to facilitate significant time-dependent AP activity in humans and in mice. Furthermore, this observation highlights the potential importance of obtaining nearly complete inhibition of FD in treating alternative complement activation in various autoimmune and inflammatory human diseases.


Subject(s)
Adipose Tissue/metabolism , Complement Factor D/metabolism , Lipodystrophy/blood , Animals , Complement Factor D/analysis , Humans , Mice
15.
Endocrinology ; 159(5): 2050-2061, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29579167

ABSTRACT

Regulation of adipogenesis is of major interest given that adipose tissue expansion and dysfunction are central to metabolic syndrome. Glucocorticoids (GCs) are important for adipogenesis in vitro. However, establishing a role for GCs in adipogenesis in vivo has been difficult. GC receptor (GR)‒null mice die at birth, a time at which wild-type (WT) mice do not have fully developed white adipose depots. We conducted several studies aimed at defining the role of GC signaling in adipogenesis in vitro and in vivo. First, we showed that GR-null mouse embryonic fibroblasts (MEFs) have compromised ability to form adipocytes in vitro, a phenotype that can be partially rescued with a peroxisome proliferator-activated receptor γ agonist. Next, we demonstrated that MEFs are capable of forming de novo fat pads in mice despite the absence of GR or circulating GCs [by bilateral adrenalectomy (ADX)]. However, ADX and GR-null fat pads and their associated adipocyte areas were smaller than those in controls. Second, using adipocyte-specific luciferase reporter mice, we identified adipocytes in both WT and GR-null embryonic day (E)18 mouse embryos. Lastly, positive perilipin staining in WT and GR-null E18 embryos confirmed the presence of early white inguinal and brown adipocytes. Taken together, these results provide compelling evidence that GCs and GR augment but are not required for the development of functional adipose tissue in vivo.


Subject(s)
Adipogenesis/genetics , Adipose Tissue, Brown/embryology , Adipose Tissue, White/embryology , Fibroblasts/metabolism , Glucocorticoids/metabolism , Receptors, Glucocorticoid/genetics , Adipocytes, Brown , Adipocytes, White , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Adrenalectomy , Animals , In Vitro Techniques , Mice , PPAR gamma/agonists , Perilipin-1/metabolism , Signal Transduction
16.
Endocrinology ; 159(2): 609-621, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29126303

ABSTRACT

Several recent studies have suggested that compounds known as endocrine-disrupting chemicals (EDCs) can promote obesity by serving as ligands for nuclear receptors, including the peroxisome proliferator-activated receptor γ (PPARγ) and the glucocorticoid receptor (GR). Thiazolidinedione insulin sensitizers, which act as ligands for PPARγ, also interact with and regulate the activity of the mitochondrial pyruvate carrier (MPC). We evaluated whether several EDCs might also affect MPC activity. Most of the EDCs evaluated did not acutely affect pyruvate metabolism. However, the putative endocrine disruptors tributyltin (TBT) and tolylfluanid (TF) acutely and markedly suppressed pyruvate metabolism in isolated mitochondria. Using mitochondria isolated from brown adipose tissue in mice with adipocyte-specific deletion of the MPC2 protein, we determined that the effect of TF on pyruvate metabolism required MPC2, whereas TBT did not. We attempted to determine whether the obesogenic effects of TF might involve MPC2 in adipose tissue. However, we were unable to replicate the published effects of TF on weight gain and adipose tissue gene expression in wild-type or fat-specific MPC2 knockout mice. Treatment with TF modestly enhanced adipogenic gene expression in vitro but had no effect on GR activation or phosphorylation in cultured cells. These data suggest that TF may affect mitochondrial pyruvate metabolism via the MPC complex but also call into question whether this compound affects GR activity and is obesogenic in mice.


Subject(s)
Endocrine Disruptors/pharmacology , Membrane Transport Proteins/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Proprotein Convertase 2/metabolism , Sulfonamides/pharmacology , Toluidines/pharmacology , Adipocytes/drug effects , Adipocytes/metabolism , Adipose Tissue, Brown , Animals , Anion Transport Proteins , Biological Transport/drug effects , Female , Membrane Transport Proteins/genetics , Mice , Mice, Inbred C57BL , Mitochondria/genetics , Mitochondrial Membrane Transport Proteins , Monocarboxylic Acid Transporters , Proprotein Convertase 2/genetics , Pyruvic Acid/metabolism
17.
F1000Res ; 6: 1208, 2017.
Article in English | MEDLINE | ID: mdl-28781762

ABSTRACT

Glucocorticoids via the glucocorticoid receptor (GR) have effects on a variety of cell types, eliciting important physiological responses via changes in gene expression and signaling. Although decades of research have illuminated the mechanism of how this important steroid receptor controls gene expression using in vitro and cell culture-based approaches, how GR responds to changes in external signals in vivo under normal and pathological conditions remains elusive. The goal of this review is to highlight recent work on GR action in fat cells and liver to affect metabolism in vivo and the role GR ligands and receptor phosphorylation play in calibrating signaling outputs by GR in the brain in health and disease. We also suggest that both the brain and fat tissue communicate to affect physiology and behavior and that understanding this "brain-fat axis" will enable a more complete understanding of metabolic diseases and inform new ways to target them.

18.
Endocrinology ; 157(12): 4943-4960, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27754788

ABSTRACT

Exogenous glucocorticoid administration results in hyperglycemia, insulin resistance, hepatic dyslipidemia, and hypertension, a constellation of findings known as Cushing's syndrome. These effects are mediated by the glucocorticoid receptor (GR). Because GR activation in liver and adipose has been implicated in metabolic syndrome (MS), we wanted to determine the role of GR in these tissues in the development of MS. Because GR knockout (KO) mice (whole-body KO) exhibit perinatal lethality due to respiratory failure, we generated tissue-specific (liver or adipose) GRKO mice using cre-lox technology. Real-time PCR analysis of liver mRNA from dexamethasone-treated wildtype (WT) and liver GRKO mice indicated that hepatic GR regulates the expression of key genes involved in gluconeogenesis and glycogen metabolism. Interestingly, we have observed that liver-specific deletion of GR resulted in a significant increase in mRNA expression of key genes involved in gluconeogenesis and glycogen metabolism in kidney tissue, indicating a compensatory mechanism to maintain glucose homeostasis. We have also observed that GR plays an important role in regulating the mRNA expression of key genes involved in lipid metabolism. Liver GRKO mice demonstrated decreased fat mass and liver glycogen content compared with WT mice administered dexamethasone for 2 weeks. Adipose-specific deletion of GR did not alter glucose tolerance or insulin sensitivity of adipose GRKO mice compared with WT mice administrated dexamethasone. This indicates that liver GR might be more important in development of MS in dexamethasone-treated mice, whereas adipose GR plays a little role in these paradigms.


Subject(s)
Adipose Tissue/metabolism , Kidney/metabolism , Liver/metabolism , Metabolic Syndrome/metabolism , Receptors, Glucocorticoid/metabolism , Triglycerides/metabolism , Animals , Body Composition/physiology , Gluconeogenesis/genetics , Insulin Resistance/genetics , Lipid Metabolism/genetics , Metabolic Syndrome/genetics , Mice , Mice, Knockout , Receptors, Glucocorticoid/genetics
19.
Mol Cell Endocrinol ; 380(1-2): 79-88, 2013 Nov 05.
Article in English | MEDLINE | ID: mdl-23523565

ABSTRACT

Glucocorticoids (GCs) exert key metabolic influences on skeletal muscle. GCs increase protein degradation and decrease protein synthesis. The released amino acids are mobilized from skeletal muscle to liver, where they serve as substrates for hepatic gluconeogenesis. This metabolic response is critical for mammals' survival under stressful conditions, such as fasting and starvation. GCs suppress insulin-stimulated glucose uptake and utilization and glycogen synthesis, and play a permissive role for catecholamine-induced glycogenolysis, thus preserving the level of circulating glucose, the major energy source for the brain. However, chronic or excess exposure of GCs can induce muscle atrophy and insulin resistance. GCs convey their signal mainly through the intracellular glucocorticoid receptor (GR). While GR can act through different mechanisms, one of its major actions is to regulate the transcription of its primary target genes through genomic glucocorticoid response elements (GREs) by directly binding to DNA or tethering onto other DNA-binding transcription factors. These GR primary targets trigger physiological and pathological responses of GCs. Much progress has been made to understand how GCs regulate protein and glucose metabolism. In this review, we will discuss how GR primary target genes confer metabolic functions of GCs, and the mechanisms governing the transcriptional regulation of these targets. Comprehending these processes not only contributes to the fundamental understanding of mammalian physiology, but also will provide invaluable insight for improved GC therapeutics.


Subject(s)
Glucocorticoids/physiology , Muscle, Skeletal/metabolism , Receptors, Glucocorticoid/physiology , Animals , Carbohydrate Metabolism , Gene Expression Regulation , Humans , Insulin/physiology , Muscle Proteins/genetics , Muscle Proteins/metabolism , Protein Biosynthesis , Response Elements
20.
Proc Natl Acad Sci U S A ; 109(28): 11160-5, 2012 Jul 10.
Article in English | MEDLINE | ID: mdl-22733784

ABSTRACT

Glucocorticoids elicit a variety of biological responses in skeletal muscle, including inhibiting protein synthesis and insulin-stimulated glucose uptake and promoting proteolysis. Thus, excess or chronic glucocorticoid exposure leads to muscle atrophy and insulin resistance. Glucocorticoids propagate their signal mainly through glucocorticoid receptors (GR), which, upon binding to ligands, translocate to the nucleus and bind to genomic glucocorticoid response elements to regulate the transcription of nearby genes. Using a combination of chromatin immunoprecipitation sequencing and microarray analysis, we identified 173 genes in mouse C2C12 myotubes. The mouse genome contains GR-binding regions in or near these genes, and gene expression is regulated by glucocorticoids. Eight of these genes encode proteins known to regulate distinct signaling events in insulin/insulin-like growth factor 1 pathways. We found that overexpression of p85α, one of these eight genes, caused a decrease in C2C12 myotube diameters, mimicking the effect of glucocorticoids. Moreover, reducing p85α expression by RNA interference in C2C12 myotubes significantly compromised the ability of glucocorticoids to inhibit Akt and p70 S6 kinase activity and reduced glucocorticoid induction of insulin receptor substrate 1 phosphorylation at serine 307. This phosphorylation is associated with insulin resistance. Furthermore, decreasing p85α expression abolished glucocorticoid inhibition of protein synthesis and compromised glucocorticoid-induced reduction of cell diameters in C2C12 myotubes. Finally, a glucocorticoid response element was identified in the p85α GR-binding regions. In summary, our studies identified GR-regulated transcriptional networks in myotubes and showed that p85α plays a critical role in glucocorticoid-induced insulin resistance and muscle atrophy in C2C12 myotubes.


Subject(s)
Gene Expression Regulation , Insulin/metabolism , Muscle Fibers, Skeletal/metabolism , Receptors, Glucocorticoid/chemistry , Animals , Atrophy , Binding Sites , Chromatin Immunoprecipitation , Disease Models, Animal , Genome-Wide Association Study , Genomics , Glucocorticoids/metabolism , Mice , Muscle, Skeletal/metabolism , Muscles/pathology , Oligonucleotide Array Sequence Analysis , Proteolysis , RNA Interference , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL