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










Publication year range
1.
Mol Metab ; 78: 101821, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37806486

ABSTRACT

The disease progression of the metabolic syndrome is associated with prolonged hyperlipidemia and insulin resistance, eventually giving rise to impaired insulin secretion, often concomitant with hypoadiponectinemia. As an adipose tissue derived hormone, adiponectin is beneficial for insulin secretion and ß cell health and differentiation. However, the down-stream pathway of adiponectin in the pancreatic islets has not been studied extensively. Here, along with the overall reduction of endocrine pancreatic function in islets from adiponectin KO mice, we examine PPARα and HNF4α as additional down-regulated transcription factors during a prolonged metabolic challenge. To elucidate the function of ß cell-specific PPARα and HNF4α expression, we developed doxycycline inducible pancreatic ß cell-specific PPARα (ß-PPARα) and HNF4α (ß-HNF4α) overexpression mice. ß-PPARα mice exhibited improved protection from lipotoxicity, but elevated ß-oxidative damage in the islets, and also displayed lowered phospholipid levels and impaired glucose-stimulated insulin secretion. ß-HNF4α mice showed a more severe phenotype when compared to ß-PPARα mice, characterized by lower body weight, small islet mass and impaired insulin secretion. RNA-sequencing of the islets of these models highlights overlapping yet unique roles of ß-PPARα and ß-HNF4α. Given that ß-HNF4α potently induces PPARα expression, we define a novel adiponectin-HNF4α-PPARα cascade. We further analyzed downstream genes consistently regulated by this axis. Among them, the islet amyloid polypeptide (IAPP) gene is an important target and accumulates in adiponectin KO mice. We propose a new mechanism of IAPP aggregation in type 2 diabetes through reduced adiponectin action.


Subject(s)
Diabetes Mellitus, Type 2 , Insulin-Secreting Cells , Animals , Mice , Adiponectin/genetics , Adiponectin/metabolism , Diabetes Mellitus, Type 2/metabolism , Insulin/metabolism , Insulin-Secreting Cells/metabolism , PPAR alpha/genetics , PPAR alpha/metabolism
2.
EMBO J ; 41(4): e108290, 2022 02 15.
Article in English | MEDLINE | ID: mdl-35028974

ABSTRACT

Nucleotide metabolism fuels normal DNA replication and is also primarily targeted by the DNA replication checkpoint when replication stalls. To reveal a comprehensive interconnection between genome maintenance and metabolism, we analyzed the metabolomic changes upon replication stress in the budding yeast S. cerevisiae. We found that upon treatment of cells with hydroxyurea, glucose is rapidly diverted to the oxidative pentose phosphate pathway (PPP). This effect is mediated by the AMP-dependent kinase, SNF1, which phosphorylates the transcription factor Mig1, thereby relieving repression of the gene encoding the rate-limiting enzyme of the PPP. Surprisingly, NADPH produced by the PPP is required for efficient recruitment of replication protein A (RPA) to single-stranded DNA, providing the signal for the activation of the Mec1/ATR-Rad53/CHK1 checkpoint signaling kinase cascade. Thus, SNF1, best known as a central energy controller, determines a fast mode of replication checkpoint activation through a redox mechanism. These findings establish that SNF1 provides a hub with direct links to cellular metabolism, redox, and surveillance of DNA replication in eukaryotes.


Subject(s)
DNA Replication , Protein Serine-Threonine Kinases/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Checkpoint Kinase 2/genetics , Checkpoint Kinase 2/metabolism , DNA Replication/drug effects , DNA, Single-Stranded/metabolism , Glucose/genetics , Glucose/metabolism , Glycolysis/physiology , Hydroxyurea , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , NADP/metabolism , Pentose Phosphate Pathway , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Replication Protein A/genetics , Replication Protein A/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism
3.
J Endocr Soc ; 3(3): 617-631, 2019 Mar 01.
Article in English | MEDLINE | ID: mdl-30834357

ABSTRACT

Obesity and diabetes are often associated with lipotoxic conditions in multiple tissues. The insulin-producing ß cells are susceptible to elevated lipid levels and the ensuing lipotoxicity. The preservation of ß cell mass and function is one of the main goals of diabetes management under these metabolically stressful conditions. However, the adverse effects from the adaptive signaling pathways that ß cells use to counteract lipotoxic stress have secondary negative effects in their own right. Antilipotoxic signaling cascades in ß cells can contribute to their eventual failure. Such dual roles are seen for many other biological adaptive processes as well.

4.
Am J Pathol ; 189(4): 813-825, 2019 04.
Article in English | MEDLINE | ID: mdl-30653956

ABSTRACT

Syntrophins are a family of proteins forming membrane-anchored scaffolds and serving as adaptors for various transmembrane and intracellular signaling molecules. To understand the physiological roles of ß1 syntrophin, one of the least characterized members, we generated mouse models to eliminate ß1 syntrophin specifically in the endocrine or exocrine pancreas. ß1 syntrophin is dispensable for the morphology and function of insulin-producing ß cells. However, mice with ß1 syntrophin deletion in exocrine acinar cells exhibit increased severity of cerulein-induced acute pancreatitis. Reduced expression of cystic fibrosis transmembrane conductance regulator and dilation of acinar lumen are potential predisposition factors. During the disease progression, a relative lack of autophagy is associated with deficiencies in both actin assembly and endoplasmic reticulum nucleation. Our findings reveal, for the first time, that ß1 syntrophin is a critical regulator of actin cytoskeleton and autophagy in pancreatic acinar cells and is potently protective against cerulein-induced acute pancreatitis.


Subject(s)
Autophagy , Ceruletide/toxicity , Dystrophin-Associated Proteins/physiology , Pancreatitis/prevention & control , Protective Agents , Acinar Cells/drug effects , Acinar Cells/metabolism , Animals , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Pancreas/drug effects , Pancreas/metabolism , Pancreatitis/chemically induced , Pancreatitis/metabolism , Pancreatitis/pathology
5.
Cell Metab ; 28(2): 282-288.e3, 2018 08 07.
Article in English | MEDLINE | ID: mdl-29909970

ABSTRACT

Adipose tissue in the mammary gland undergoes dramatic remodeling during reproduction. Adipocytes are replaced by mammary alveolar structures during pregnancy and lactation, then reappear upon weaning. The fate of the original adipocytes during lactation and the developmental origin of the re-appearing adipocyte post involution are unclear. Here, we reveal that adipocytes in the mammary gland de-differentiate into Pdgfrα+ preadipocyte- and fibroblast-like cells during pregnancy and remain de-differentiated during lactation. Upon weaning, de-differentiated fibroblasts proliferate and re-differentiate into adipocytes. This cycle occurs over multiple pregnancies. These observations reveal the potential of terminally differentiated adipocytes to undergo repeated cycles of de-differentiation and re-differentiation in a physiological setting.


Subject(s)
Adipocytes, White/metabolism , Adipogenesis , Adipose Tissue , Lactation/metabolism , Mammary Glands, Animal , Adipocytes, White/cytology , Adipose Tissue/cytology , Adipose Tissue/metabolism , Animals , Female , Mammary Glands, Animal/cytology , Mammary Glands, Animal/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pregnancy , Weaning
6.
J Clin Invest ; 128(3): 1178-1189, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29457786

ABSTRACT

The compensatory proliferation of insulin-producing ß cells is critical to maintaining glucose homeostasis at the early stage of type 2 diabetes. Failure of ß cells to proliferate results in hyperglycemia and insulin dependence in patients. To understand the effect of the interplay between ß cell compensation and lipid metabolism upon obesity and peripheral insulin resistance, we eliminated LDL receptor-related protein 1 (LRP1), a pleiotropic mediator of cholesterol, insulin, energy metabolism, and other cellular processes, in ß cells. Upon high-fat diet exposure, LRP1 ablation significantly impaired insulin secretion and proliferation of ß cells. The diminished insulin signaling was partly contributed to by the hypersensitivity to glucose-induced, Ca2+-dependent activation of Erk and the mTORC1 effector p85 S6K1. Surprisingly, in LRP1-deficient islets, lipotoxic sphingolipids were mitigated by improved lipid metabolism, mediated at least in part by the master transcriptional regulator PPARγ2. Acute overexpression of PPARγ2 in ß cells impaired insulin signaling and insulin secretion. Elimination of Apbb2, a functional regulator of LRP1 cytoplasmic domain, also impaired ß cell function in a similar fashion. In summary, our results uncover the double-edged effects of intracellular lipid metabolism on ß cell function and viability in obesity and type 2 diabetes and highlight LRP1 as an essential regulator of these processes.


Subject(s)
Diet , Insulin-Secreting Cells/metabolism , Lipid Metabolism , Obesity/metabolism , Receptors, LDL/metabolism , Tumor Suppressor Proteins/metabolism , Alleles , Animals , Blood Glucose/metabolism , Cell Proliferation , Crosses, Genetic , Cytoplasm/metabolism , Female , Gene Expression Profiling , Gene Expression Regulation , Glucose/metabolism , Glucose Tolerance Test , Insulin/blood , Insulin/metabolism , Low Density Lipoprotein Receptor-Related Protein-1 , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , PPAR gamma/metabolism , Sphingolipids/metabolism , Transcription, Genetic
7.
Mol Cell Biol ; 38(10)2018 05 15.
Article in English | MEDLINE | ID: mdl-29483301

ABSTRACT

Adipose tissue is a dynamic organ that makes critical contributions to whole-body metabolic homeostasis. Although recent studies have revealed that different fat depots have distinct molecular signatures, metabolic functions and adipogenic mechanisms, peroxisome proliferator-activated receptor γ (PPARγ) is still widely viewed as the master regulator of adipogenesis and critical for maintaining mature adipocyte function. Using an inducible, adipocyte-specific knockout system, we explored the role of PPARγ in mature adipocytes in vivo Short-term PPARγ deficiency in adipocytes reduces whole-body insulin sensitivity, but adipocytes are viable both in vitro and in vivo However, after exposure to a high-fat diet, even short-term PPARγ deficiency leads to rapid adipocyte death. When mature adipocytes are depleted of both PPARγ and CCAAT-enhancer-binding protein α (C/EBPα), they are rapidly depleted of lipids and undergo adipocyte death, both in vitro and in vivo Surprisingly, although thiazolidinediones (TZDs; PPARγ agonists) are thought to act mainly on PPARγ, PPARγ in adipocytes is not required for the whole-body insulin-sensitizing effect of TZDs. This offers new mechanistic aspects of PPARγ/TZD action and its effect on whole-body metabolic homeostasis.


Subject(s)
Adipocytes/metabolism , Insulin Resistance/physiology , PPAR gamma/physiology , Adipogenesis/physiology , Adipose Tissue/physiology , Animals , CCAAT-Enhancer-Binding Protein-alpha/metabolism , Homeostasis/physiology , Insulin/metabolism , Mice , Mice, Inbred C57BL , Thiazolidinediones/pharmacology , Transcription Factors
8.
Physiol Rep ; 4(18)2016 Sep.
Article in English | MEDLINE | ID: mdl-27664190

ABSTRACT

Acute neonatal hypoxia, a common stressor, causes a spontaneous decrease in body temperature which may be protective. There is consensus that hypothermia should be prevented during acute hypoxia in the human neonate; however, this may be an additional stress with negative consequences. We hypothesize that maintaining body temperature during hypoxia in the first week of postnatal life alters the subsequent insulin, glucose, and glucagon secretion in adult rats. Rat pups were separated from their dam daily from postnatal days (PD) 2-6 for the following 90 min experimental treatments: (1) normoxic separation (control), (2) hypoxia (8% O2) allowing spontaneous hypothermia, (3) normoxic hypothermia with external cold, and (4) exposure to 8% O2 while maintaining body temperature using external heat. An additional normoxic non-separated control group was performed to determine if separation per se changed the adult phenotype. Plasma insulin, glucose, and glucagon responses to arginine stimulation were evaluated from PD105 to PD133. Maternal separation (compared to non-separated neonates) had more pronounced effects on the adult response to arginine compared to the hypoxic, hypothermic, and hypoxic-isothermic neonatal treatments. Adult males exposed to neonatal maternal separation had augmented insulin and glucose responses to arginine compared to unseparated controls. Additionally, neonatal treatment had a significant effect on body weight gain; adults exposed to neonatal maternal separation were significantly heavier. Female adults had significantly smaller insulin and glucose responses to arginine regardless of neonatal treatment. Neonatal maternal separation during the first week of life significantly altered adult beta-cell function in a sexually dimorphic manner.

9.
Mol Metab ; 5(7): 437-448, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27408770

ABSTRACT

BACKGROUND: Evidence hints at the ability of ß-cells to emerge from non-ß-cells upon genetic or pharmacological interventions. However, their quantitative contributions to the process of autonomous ß-cell regeneration without genetic or pharmacological manipulations remain to be determined. METHODS & RESULTS: Using PANIC-ATTAC mice, a model of titratable, acute ß-cell apoptosis capable of autonomous, and effective islet mass regeneration, we demonstrate that an extended washout of residual tamoxifen activity is crucial for ß-cell lineage tracing studies using the tamoxifen-inducible Cre/loxP systems. We further establish a doxycycline-inducible system to label different cell types in the mouse pancreas and pursued a highly quantitative assessment to trace adult ß-cells after various metabolic challenges. Beyond proliferation of pre-existing ß-cells, non-ß-cells contribute significantly to the post-challenge regenerated ß-cell pool. α-cell trans-differentiation is the predominant mechanism upon post-apoptosis regeneration and multiparity. No contributions from exocrine acinar cells were observed. During diet-induced obesity, about 25% of α-cells arise de novo from ß-cells. Ectopic expression of Nkx6.1 promotes α-to-ß conversion and insulin production. CONCLUSIONS: We identify the origins and fates of adult ß-cells upon post-challenge upon autonomous regeneration of islet mass and establish the quantitative contributions of the different cell types using a lineage tracing system with high temporal resolution.

10.
Diabetes ; 65(6): 1534-55, 2016 06.
Article in English | MEDLINE | ID: mdl-26895793

ABSTRACT

Mitochondrial metabolism plays an integral role in glucose-stimulated insulin secretion (GSIS) in ß-cells. In addition, the diabetogenic role of glucagon released from α-cells plays a major role in the etiology of both type 1 and type 2 diabetes because unopposed hyperglucagonemia is a pertinent contributor to diabetic hyperglycemia. Titrating expression levels of the mitochondrial protein mitoNEET is a powerful approach to fine-tune mitochondrial capacity of cells. Mechanistically, ß-cell-specific mitoNEET induction causes hyperglycemia and glucose intolerance due to activation of a Parkin-dependent mitophagic pathway, leading to the formation of vacuoles and uniquely structured mitophagosomes. Induction of mitoNEET in α-cells leads to fasting-induced hypoglycemia and hypersecretion of insulin during GSIS. MitoNEET-challenged α-cells exert potent antiapoptotic effects on ß-cells and prevent cellular dysfunction associated with mitoNEET overexpression in ß-cells. These observations identify that reduced mitochondrial function in α-cells exerts potently protective effects on ß-cells, preserving ß-cell viability and mass.


Subject(s)
Glucagon-Secreting Cells/metabolism , Insulin-Secreting Cells/metabolism , Iron-Binding Proteins/metabolism , Membrane Proteins/metabolism , Mitochondria/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Apoptosis , Cell Survival , Glucagon/biosynthesis , Glucose/metabolism , Glucose Intolerance/etiology , Hyperglycemia/etiology , Insulin/metabolism , Insulin Secretion , Mice , Receptor Cross-Talk
11.
Mol Metab ; 4(11): 771-8, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26629402

ABSTRACT

BACKGROUND: The selective estrogen receptor modulator tamoxifen, in combination with the Cre-ER(T2) fusion protein, has been one of the mainstream methods to induce genetic recombination and has found widespread application in lineage tracing studies. METHODS & RESULTS: Here, we report that tamoxifen exposure at widely used concentrations remains detectable by mass-spectrometric analysis in adipose tissue after a washout period of 10 days. Surprisingly, its ability to maintain nuclear translocation of the Cre-ER(T2) protein is preserved beyond 2 months of washout. Tamoxifen treatment acutely leads to transient lipoatrophy, followed by de novo adipogenesis that reconstitutes the original fat mass. In addition, we find a "synthetically lethal" phenotype for adipocytes when tamoxifen treatment is combined with adipocyte-specific loss-of-function mutants, such as an adipocyte-specific PPARγ knockout. This is observed to a lesser extent when alternative inducible approaches are employed. CONCLUSIONS: These findings highlight the potential for tamoxifen-induced adipogenesis, and the associated drawbacks of the use of tamoxifen in lineage tracing studies, explaining the discrepancy in lineage tracing results from different systems with temporal control of gene targeting.

12.
Nat Cell Biol ; 17(9): 1099-111, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26280538

ABSTRACT

Pathological expansion of adipose tissue contributes to the metabolic syndrome. Distinct depots develop at various times under different physiological conditions. The transcriptional cascade mediating adipogenesis is established in vitro, and centres around a core program involving PPARγ and C/EBPα. We developed an inducible, adipocyte-specific knockout system to probe the requirement of key adipogenic transcription factors at various stages of adipogenesis in vivo. C/EBPα is essential for all white adipogenic conditions in the adult stage, such as adipose tissue regeneration, adipogenesis in muscle and unhealthy expansion of white adipose tissue during high-fat feeding or due to leptin deficiency. Surprisingly, terminal embryonic adipogenesis is fully C/EBPα independent, but does however depend on PPARγ; cold-induced beige adipogenesis is also C/EBPα independent. Moreover, C/EBPα is not vital for adipocyte survival in the adult stage. We reveal a surprising diversity of transcriptional signals required at different stages of adipogenesis in vivo.


Subject(s)
Adipocytes/physiology , Adipogenesis , Adipose Tissue, White/cytology , Animals , CCAAT-Enhancer-Binding Proteins/genetics , Carbohydrate Metabolism , Cell Shape , Diet, High-Fat/adverse effects , Embryo, Mammalian/cytology , Female , Gene Knockout Techniques , Lipid Metabolism , Male , Mice, Obese , Mice, Transgenic , Organ Specificity , PPAR gamma/metabolism , Transcription, Genetic
13.
Endocrinology ; 156(6): 2019-28, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25815422

ABSTRACT

Pathways that stimulate ß-cell regeneration remain of great clinical interest, yet effective therapeutic avenues that promote survival or reconstitution of ß-cell mass remain elusive. Using a mouse model with inducible ß-cell apoptosis followed by adiponectin-mediated regeneration, we aimed to identify key molecules boosting ß-cell viability. In the regenerating pancreatic islets, we examined changes within the transcriptome and observed an extensive up-regulation of genes encoding proteins involved in lipid transport and metabolism. The most prominent targets were further confirmed by quantitative PCR and immunofluorescence. Among the upstream regulators predicted by pathway analysis of the transcriptome, we detected enhanced levels of 2 key transcription factors, Hepatocyte Nuclear Factor 4α and Peroxisome Proliferator-Activated Receptorα. Our data suggest that improving pancreatic islet lipid metabolism as an important antilipotoxic phenomenon to boost ß-cell regeneration. This is primarily mediated by the adipokine adiponectin that exerts its action on both the beta-cell directly as well as on the adipocyte. Adiponectin induces lipid metabolism gene expression in regenerating islets through Hepatocyte Nuclear Factor 4α and Peroxisome Proliferator-Activated Receptorα. Adiponectin also modulates leptin levels via preserving adipose tissue mass in the insulinopenic state.


Subject(s)
Adiponectin/metabolism , Islets of Langerhans/metabolism , Adiponectin/genetics , Animals , Apoptosis/physiology , Cell Proliferation/physiology , Immunohistochemistry , Insulin-Secreting Cells/metabolism , Lipid Metabolism/physiology , Male , Mice , Mice, Knockout , Real-Time Polymerase Chain Reaction , Transcriptome/genetics
14.
Elife ; 32014 Oct 23.
Article in English | MEDLINE | ID: mdl-25339419

ABSTRACT

As an adipokine in circulation, adiponectin has been extensively studied for its beneficial metabolic effects. While many important functions have been attributed to adiponectin under high-fat diet conditions, little is known about its essential role under regular chow. Employing a mouse model with inducible, acute ß-cell ablation, we uncovered an essential role of adiponectin under insulinopenic conditions to maintain minimal lipid homeostasis. When insulin levels are marginal, adiponectin is critical for insulin signaling, endocytosis, and lipid uptake in subcutaneous white adipose tissue. In the absence of both insulin and adiponectin, severe lipoatrophy and hyperlipidemia lead to lethality. In contrast, elevated adiponectin levels improve systemic lipid metabolism in the near absence of insulin. Moreover, adiponectin is sufficient to mitigate local lipotoxicity in pancreatic islets, and it promotes reconstitution of ß-cell mass, eventually reinstating glycemic control. We uncovered an essential new role for adiponectin, with major implications for type 1 diabetes.


Subject(s)
Adiponectin/metabolism , Homeostasis , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Insulin/deficiency , Lipid Metabolism , Regeneration , Adipocytes/metabolism , Adipocytes/ultrastructure , Adipose Tissue, White/metabolism , Adipose Tissue, White/pathology , Adipose Tissue, White/ultrastructure , Animals , Caveolae/metabolism , Caveolin 1/metabolism , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Homeostasis/drug effects , Insulin/metabolism , Lipid Metabolism/drug effects , Lipids/toxicity , Mice , Regeneration/drug effects , Streptozocin , Survival Analysis
15.
Cancer Res ; 74(18): 4996-5007, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-25060520

ABSTRACT

Elevated levels of TGFß are a negative prognostic indicator for patients diagnosed with pancreatic cancer; as a result, the TGFß pathway is an attractive target for therapy. However, clinical application of pharmacologic inhibition of TGFß remains challenging because TGFß has tumor suppressor functions in many epithelial malignancies, including pancreatic cancer. In fact, direct neutralization of TGFß promotes tumor progression of genetic murine models of pancreatic cancer. Here, we report that neutralizing the activity of murine TGFß receptor 2 using a monoclonal antibody (2G8) has potent antimetastatic activity in orthotopic human tumor xenografts, syngeneic tumors, and a genetic model of pancreatic cancer. 2G8 reduced activated fibroblasts, collagen deposition, microvessel density, and vascular function. These stromal-specific changes resulted in tumor cell epithelial differentiation and a potent reduction in metastases. We conclude that TGFß signaling within stromal cells participates directly in tumor cell phenotype and pancreatic cancer progression. Thus, strategies that inhibit TGFß-dependent effector functions of stromal cells could be efficacious for the therapy of pancreatic tumors. Cancer Res; 74(18); 4996-5007. ©2014 AACR.


Subject(s)
Antibodies, Monoclonal/pharmacology , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Animals , Antimetabolites, Antineoplastic/pharmacology , Cell Differentiation/drug effects , Cell Line, Tumor , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Humans , Mice , Mice, Inbred C57BL , NIH 3T3 Cells , Neoplasm Metastasis , Pancreatic Neoplasms/metabolism , Random Allocation , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction , Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta/metabolism , Xenograft Model Antitumor Assays , Gemcitabine
16.
Mol Metab ; 3(4): 347-8, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24944893
17.
Mol Metab ; 2(3): 133-41, 2013 Apr 19.
Article in English | MEDLINE | ID: mdl-24049728

ABSTRACT

Almost 20 years have passed since the first laboratory evidence emerged that an abundant message encoding a protein with homology to the C1q superfamily is highly specifically expressed in adipocytes. At this stage, we refer to this protein as adiponectin. Despite more than 10,000 reports in the literature since its initial description, we seem to have written only the first chapter in the textbook on adiponectin physiology. With every new aspect we learn about adiponectin, a host of new questions arise with respect to the underlying molecular mechanisms. Here, we aim to summarize recent findings in the field and bring the rodent studies that suggest a causal relationship between adiponectin levels in plasma and systemic insulin sensitivity in perspective with the currently available data on the clinical side.

18.
FASEB J ; 27(3): 955-64, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23180827

ABSTRACT

To investigate the role of GRP78 in adipogenesis and metabolic homeostasis, we knocked down GRP78 in mouse embryonic fibroblasts and 3T3-L1 preadipocytes induced to undergo differentiation into adipocytes. We also created an adipose Grp78-knockout mouse utilizing the aP2 (fatty acid binding protein 4) promoter-driven Cre-recombinase. Adipogenesis was monitored by molecular markers and histology. Tissues were analyzed by micro-CT and electron microscopy. Glucose homeostasis and cytokine analysis were performed. Our results indicate that GRP78 is essential for adipocyte differentiation in vitro. aP2-cre-mediated GRP78 deletion leads to lipoatrophy with ∼90% reduction in gonadal and subcutaneous white adipose tissue and brown adipose tissue, severe growth retardation, and bone defects. Despite severe abnormality in adipose mass and function, adipose Grp78-knockout mice showed normal plasma triglyceride levels, and plasma glucose and insulin levels were reduced by 40-60% compared to wild-type mice, suggesting enhanced insulin sensitivity. The endoplasmic reticulum is grossly expanded in the residual mutant white adipose tissue. Thus, these studies establish that GRP78 is required for adipocyte differentiation, glucose homeostasis, and balanced secretion of adipokines. Unexpectedly, the phenotypes and metabolic parameters of the mutant mice, which showed early postnatal mortality, are uniquely distinct from previously characterized lipodystrophic mouse models.


Subject(s)
Adipogenesis/physiology , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Glucose/metabolism , Heat-Shock Proteins/metabolism , Homeostasis/physiology , 3T3-L1 Cells , Adipocytes/cytology , Adipocytes/metabolism , Adipokines/genetics , Adipokines/metabolism , Adipose Tissue, Brown/cytology , Adipose Tissue, White/cytology , Animals , Cell Differentiation/physiology , Disease Models, Animal , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Chaperone BiP , Fatty Acid-Binding Proteins/genetics , Fatty Acid-Binding Proteins/metabolism , Gene Deletion , Glucose/genetics , Heat-Shock Proteins/genetics , Lipodystrophy/genetics , Lipodystrophy/metabolism , Lipodystrophy/pathology , Mice , Mice, Knockout , Triglycerides/blood , Triglycerides/genetics
19.
J Endocrinol ; 210(2): 209-17, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21565852

ABSTRACT

The inositol 1,4,5-trisphosphate receptors (IP3Rs) as ligand-gated Ca(2)(+) channels are key modulators of cellular processes. Despite advances in understanding their critical role in regulating neuronal function and cell death, how this family of proteins impact cell metabolism is just emerging. Unexpectedly, a transgenic mouse line (D2D) exhibited progressive glucose intolerance as a result of transgene insertion. Inverse PCR was used to identify the gene disruption in the D2D mice. This led to the discovery that Itpr1 is among the ten loci disrupted in chromosome 6. Itpr1 encodes for IP3R1, the most abundant IP3R isoform in mouse brain and also highly expressed in pancreatic ß-cells. To study IP3R1 function in glucose metabolism, we used the Itpr1 heterozygous mutant mice, opt/+. Glucose homeostasis in male mice cohorts was examined by multiple approaches of metabolic phenotyping. Under regular diet, the opt/+ mice developed glucose intolerance but no insulin resistance. Decrease in second-phase glucose-stimulated blood insulin level was observed in opt/+ mice, accompanied by reduced ß-cell mass and insulin content. Strikingly, when fed with high-fat diet, the opt/+ mice were more susceptible to the development of hyperglycemia, glucose intolerance, and insulin resistance. Collectively, our studies identify the gene Itpr1 being interrupted in the D2D mice and uncover a novel role of IP3R1 in regulation of in vivo glucose homeostasis and development of diet-induced diabetes.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Glucose/genetics , Homeostasis/genetics , Inositol 1,4,5-Trisphosphate Receptors/genetics , Mutation , Animals , Diabetes Mellitus, Type 2/metabolism , Dietary Fats , Disease Susceptibility , Glucose/metabolism , Glucose Intolerance/genetics , Glucose Intolerance/metabolism , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Insulin/genetics , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Male , Mice , Mice, Transgenic
20.
Cancer Res ; 71(8): 2848-57, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21467168

ABSTRACT

Glucose-regulated protein 78 (GRP78)/BiP is a multifunctional protein which plays a major role in endoplasmic reticulum (ER) protein processing, protein quality control, maintaining ER homeostasis, and controlling cell signaling and viability. Previously, using a transgene-induced mammary tumor model, we showed that Grp78 heterozygosity impeded cancer growth through suppression of tumor cell proliferation and promotion of apoptosis and the Grp78(+/-) mice exhibited dramatic reduction (70%) in the microvessel density (MVD) of the endogenous mammary tumors, while having no effect on the MVD of normal organs. This observation suggests that GRP78 may critically regulate the function of the host vasculature within the tumor microenvironment. In this article, we interrogated the role of GRP78 in the tumor microenvironment. In mouse tumor models in which wild-type (WT), syngeneic mammary tumor cells were injected into the host, we showed that Grp78(+/-) mice suppressed tumor growth and angiogenesis during the early phase but not during the late phase of tumor growth. Growth of metastatic lesions of WT, syngeneic melanoma cells in the Grp78(+/-) mice was potently suppressed. We created conditional heterozygous knockout of GRP78 in the host endothelial cells and showed severe reduction of tumor angiogenesis and metastatic growth, with minimal effect on normal tissue MVD. Furthermore, knockdown of GRP78 expression in immortalized human endothelial cells showed that GRP78 is a critical mediator of angiogenesis by regulating cell proliferation, survival, and migration. Our findings suggest that concomitant use of current chemotherapeutic agents and novel therapies against GRP78 may offer a powerful dual approach to arrest cancer initiation, progression, and metastasis.


Subject(s)
Heat-Shock Proteins/genetics , Mammary Neoplasms, Experimental/blood supply , Mammary Neoplasms, Experimental/genetics , Animals , Apoptosis/physiology , Cell Growth Processes/physiology , Cell Movement/physiology , Endoplasmic Reticulum Chaperone BiP , Endothelial Cells/cytology , Endothelial Cells/physiology , Female , Heterozygote , Humans , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neoplasm Metastasis , Neovascularization, Pathologic/genetics , Tumor Microenvironment
SELECTION OF CITATIONS
SEARCH DETAIL
...