Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Blood Adv ; 8(2): 309-323, 2024 01 23.
Article in English | MEDLINE | ID: mdl-37967356

ABSTRACT

ABSTRACT: Ca2+/calmodulin-dependent protein kinase II γ (CAMKIIγ) has been identified as a potential target for treating cancer. Based on our previous study of berbamine (BBM) as a CAMKIIγ inhibitor, we have synthesized a new BBM derivative termed PA4. Compared with BBM, PA4 showed improved potency and specificity and was more cytotoxic against lymphoma and leukemia than against other types of cancer. In addition to indirectly targeting c-Myc protein stability, we demonstrated that its cytotoxic effects were also mediated via increased reactive oxygen species production in lymphoma cells. PA4 significantly impeded tumor growth in vivo in a xenograft T-cell lymphoma mouse model. Pharmacokinetics studies demonstrated quick absorption into plasma after oral administration, with a maximum concentration of 1680 ± 479 ng/mL at 5.33 ± 2.31 hours. The calculated oral absolute bioavailability was 34.1%. Toxicity assessment of PA4 showed that the therapeutic window used in our experiments was safe for future development. Given its efficacy, safety, and favorable pharmacokinetic profile, PA4 is a potential lead candidate for treating lymphoma.


Subject(s)
Antineoplastic Agents , Benzylisoquinolines , Leukemia , Lymphoma, T-Cell , Humans , Mice , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Benzylisoquinolines/pharmacology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use
2.
Hepatology ; 79(5): 1005-1018, 2024 May 01.
Article in English | MEDLINE | ID: mdl-37820064

ABSTRACT

BACKGROUND AND AIMS: Although the benefits of vertical sleeve gastrectomy (VSG) surgery are well known, the molecular mechanisms by which VSG alleviates obesity and its complications remain unclear. We aim to determine the role of CYP8B1 (cytochrome P450, family 8, subfamily B, polypeptide 1) in mediating the metabolic benefits of VSG. APPROACH AND RESULTS: We found that expression of CYP8B1, a key enzyme in controlling the 12α-hydroxylated (12α-OH) bile acid (BA) to non-12α-OH BA ratio, was strongly downregulated after VSG. Using genetic mouse models of CYP8B1 overexpression, knockdown, and knockout, we demonstrated that overexpression of CYP8B1 dampened the metabolic improvements associated with VSG. In contrast, short hairpin RNA-mediated CYP8B1 knockdown improved metabolism similar to those observed after VSG. Cyp8b1 deficiency diminished the metabolic effects of VSG. Further, VSG-induced alterations to the 12α-OH/non-12α-OH BA ratio in the BA pool depended on CYP8B1 expression level. Consequently, intestinal lipid absorption was restricted, and the gut microbiota (GM) profile was altered. Fecal microbiota transplantation from wild type-VSG mice (vs. fecal microbiota transplantation from wild-type-sham mice) improved metabolism in recipient mice, while there were no differences between mice that received fecal microbiota transplantation from knockout-sham and knockout-VSG mice. CONCLUSIONS: CYP8B1 is a critical downstream target of VSG. Modulation of BA composition and gut microbiota profile by targeting CYP8B1 may provide novel insight into the development of therapies that noninvasively mimic bariatric surgery to treat obesity and its complications.


Subject(s)
Bariatric Surgery , Steroid 12-alpha-Hydroxylase , Mice , Animals , Steroid 12-alpha-Hydroxylase/metabolism , Down-Regulation , Obesity/metabolism , Gastrectomy , Mice, Inbred C57BL
3.
Molecules ; 28(14)2023 Jul 11.
Article in English | MEDLINE | ID: mdl-37513205

ABSTRACT

The regulation of bile acid pathways has become a particularly promising therapeutic strategy for a variety of metabolic disorders, cancers, and diseases. However, the hydrophobicity of bile acids has been an obstacle to clinical efficacy due to off-target effects from rapid drug absorption. In this report, we explored a novel strategy to design new structure fragments based on lithocholic acid (LCA) with improved hydrophilicity by introducing a polar "oxygen atom" into the side chain of LCA, then (i) either retaining the carboxylic acid group or replacing the carboxylic acid group with (ii) a diol group or (iii) a vinyl group. These novel fragments were evaluated using luciferase-based reporter assays and the MTS assay. Compared to LCA, the result revealed that the two lead compounds 1a-1b were well tolerated in vitro, maintaining similar potency and efficacy to LCA. The MTS assay results indicated that cell viability was not affected by dose dependence (under 25 µM). Additionally, computational model analysis demonstrated that compounds 1a-1b formed more extensive hydrogen bond networks with Takeda G protein-coupled receptor 5 (TGR5) than LCA. This strategy displayed a potential approach to explore the development of novel endogenous bile acids fragments. Further evaluation on the biological activities of the two lead compounds is ongoing.


Subject(s)
Bile Acids and Salts , Lithocholic Acid , Lithocholic Acid/pharmacology , Bile Acids and Salts/pharmacology
4.
Commun Biol ; 6(1): 105, 2023 01 27.
Article in English | MEDLINE | ID: mdl-36707678

ABSTRACT

Long non-coding RNAs (lncRNAs) are emerging important epigenetic regulators in metabolic processes. Whether they contribute to the metabolic effects of vertical sleeve gastrectomy (VSG), one of the most effective treatments for sustainable weight loss and metabolic improvement, is unknown. Herein, we identify a hepatic lncRNA Gm19619, which is strongly repressed by VSG but highly up-regulated by diet-induced obesity and overnight-fasting in mice. Forced transcription of Gm19619 in the mouse liver significantly promotes hepatic gluconeogenesis with the elevated expression of G6pc and Pck1. In contrast, AAV-CasRx mediated knockdown of Gm19619 in high-fat diet-fed mice significantly improves hepatic glucose and lipid metabolism. Mechanistically, Gm19619 is enriched along genomic regions encoding leptin receptor (Lepr) and transcription factor Foxo1, as revealed in chromatin isolation by RNA purification (ChIRP) assay and is confirmed to modulate their transcription in the mouse liver. In conclusion, Gm19619 may enhance gluconeogenesis and lipid accumulation in the liver.


Subject(s)
Lipogenesis , RNA, Long Noncoding , Animals , Mice , Diet, High-Fat , Down-Regulation , Gastrectomy , Gluconeogenesis/genetics , Lipogenesis/genetics , Liver/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism
5.
Mol Ther Nucleic Acids ; 30: 115-130, 2022 Dec 13.
Article in English | MEDLINE | ID: mdl-36250205

ABSTRACT

We investigated the role of microRNA (miR-379) in the pathogenesis of obesity, adipose tissue dysfunction, and insulin resistance (IR). We used miR-379 knockout (miR-379KO) mice to test whether loss of miR-379 affects high-fat diet (HFD)-induced obesity and IR via dysregulation of key miR-379 targets in adipose tissue. Increases in body weight, hyperinsulinemia, and IR in wild-type (WT)-HFD mice were significantly attenuated in miR-379KO-HFD mice with some sex differences. Relative to control chow-fed mice, in WT-HFD mice, expression of miR-379 and C/EBP homologous protein (Chop) (pro-endoplasmic reticulum [ER] stress) and inflammation in perigonadal white adipose tissue (gWAT) were increased, whereas adipogenic genes and miR-379 target genes (Vegfb and Edem3) were decreased. These changes, as well as key parameters of brown adipose tissue dysfunction (including mitochondrial defects), were significantly attenuated in miR-379KO-HFD mice. WAT from obese human subjects with and without type 2 diabetes showed increased miR-379 and decreased miR-379 target genes. In cultured 3T3L1 pre-adipocytes, miR-379 inhibitors increased miR-379 targets and adipogenic genes. These data suggest that miR-379 plays an important role in HFD-induced obesity through increased adipose inflammation, mitochondrial dysfunction, and ER stress as well as impaired adipogenesis and angiogenesis. miR-379 inhibitors may be developed as novel therapies for obesity and associated complications.

6.
Front Endocrinol (Lausanne) ; 13: 929530, 2022.
Article in English | MEDLINE | ID: mdl-36072923

ABSTRACT

Metabolic surgery, or bariatric surgery, is currently the most effective approach for treating obesity and its complications. Vertical sleeve gastrectomy (VSG) and Roux-en-Y gastric bypass (RYGB) are the top two types of commonly performed metabolic surgery now. The precise mechanisms of how the surgeries work are still unclear, therefore much research has been conducted in this area. Gut hormones such as GLP-1 and PYY have been studied extensively in the context of metabolic surgery because they both participate in satiety and glucose homeostasis. Bile acids, whose functions cover intestinal lipid absorption and various aspects of metabolic regulation via the action of FXR, TGR5, and other bile acid receptors, have also been actively investigated as potential mediators of metabolic surgery. Additionally, gut microbiota and their metabolites have also been studied because they can affect metabolic health. The current review summarizes and compares the recent scientific progress made on identifying the mechanisms of RYGB and VSG. One of the long-term goals of metabolic/bariatric surgery research is to develop new pharmacotherapeutic options for the treatment of obesity and diabetes. Because obesity is a growing health concern worldwide, there is a dire need in developing novel non-invasive treatment options.


Subject(s)
Bariatric Surgery , Gastric Bypass , Gastrointestinal Microbiome , Bile Acids and Salts , Gastrointestinal Microbiome/physiology , Humans , Obesity/metabolism , Obesity/surgery
7.
Mol Ther Nucleic Acids ; 30: 1-12, 2022 Dec 13.
Article in English | MEDLINE | ID: mdl-36158629

ABSTRACT

MicroRNAs (miRNAs) are small noncoding RNAs that regulate a variety of physiological and pathological functions. miR-26a is one of the many miRNAs that have been identified as regulators of cancer development and as potential anticancer drug targets. However, the specific cellular and molecular mechanisms by which miR-26a attenuates hepatocarcinogenesis are still elusive. Here, we interrogated mouse models with miR-26a cell-specific overexpression in either hepatocytes or myeloid cells to show that miR-26a strongly attenuated the chemical-induced hepatocellular carcinoma (HCC). miR-26a overexpression broadly inhibited the inflammatory response in both hepatocytes and macrophages by decreasing several key oncogenic signaling pathways in HCC promotion. These findings thus reveal new insights into a concerted role of miR-26a in both hepatocytes and Kupffer cells to suppress hepatocarcinogenesis, thereby highlighting the potential use of miR-26a mimetics as potential approaches for the prevention and treatment of HCC.

8.
Nat Commun ; 13(1): 1135, 2022 03 03.
Article in English | MEDLINE | ID: mdl-35241650

ABSTRACT

The energy-dissipating capacity of brown adipose tissue through thermogenesis can be targeted to improve energy balance. Mammalian 5'-AMP-activated protein kinase, a key nutrient sensor for maintaining cellular energy status, is a known therapeutic target in Type II diabetes. Despite its well-established roles in regulating glucose metabolism in various tissues, the functions of AMPK in the intestine remain largely unexplored. Here we show that AMPKα1 deficiency in the intestine results in weight gain and impaired glucose tolerance under high fat diet feeding, while metformin administration fails to ameliorate these metabolic disorders in intestinal AMPKα1 knockout mice. Further, AMPKα1 in the intestine communicates with brown adipose tissue to promote thermogenesis. Mechanistically, we uncover a link between intestinal AMPKα1 activation and BAT thermogenic regulation through modulating anti-microbial peptide-controlled gut microbiota and the metabolites. Our findings identify AMPKα1-mediated mechanisms of intestine-BAT communication that may partially underlie the therapeutic effects of metformin.


Subject(s)
Diabetes Mellitus, Type 2 , Gastrointestinal Microbiome , Metformin , AMP-Activated Protein Kinases/metabolism , Adipose Tissue, Brown/metabolism , Animals , Diabetes Mellitus, Type 2/metabolism , Diet, High-Fat , Energy Metabolism , Gastrointestinal Microbiome/physiology , Intestines , Mammals/metabolism , Metformin/pharmacology , Mice , Thermogenesis/physiology
9.
Cell Mol Gastroenterol Hepatol ; 13(3): 809-826, 2022.
Article in English | MEDLINE | ID: mdl-34896286

ABSTRACT

BACKGROUND & AIMS: Alcohol-associated liver disease (AALD) is one of the most common causes of liver injury and failure. Limited knowledge of the mechanisms underlying AALD impedes the development of efficacious therapies. Bile acid (BA) signaling was shown to participate in the progression of AALD. However, the mechanisms remain poorly understood. METHODS: C57BL/6J wild-type (WT), Takeda G-protein-coupled bile acid receptor 5 (TGR5) knockout (KO) and brown adipose tissue (BAT)-specific TGR5 knockdown mice were subjected to ethanol feeding-induced AALD. Liver samples from alcoholic hepatitis patients were used to examine the BA circulation signaling. Human Embryonic Kidney Cells 293 were used for the TGR5 reporter assay. 23(S)-methyl-lithocholic acid was used as a molecular tool to confirm the regulatory functions of BAT in the AALD mouse model. RESULTS: Ethanol feeding increased the expression of the thermogenesis genes downstream of TGR5 in BAT of WT, but not TGR5 KO, mice. TGR5 deficiency significantly blocked BAT activity and energy expenditure in mice after ethanol feeding. Alcohol increased serum BA levels in mice and human beings through altering BA transportation, and the altered BAs activated TGR5 signaling to regulate metabolism. Compared with ethanol-fed WT mice, ethanol-fed TGR5 KO mice showed less free fatty acid (FFA) ß-oxidation in BAT, leading to higher levels of FFA in the circulation, increased liver uptake of FFAs, and exacerbated AALD. BAT-specific TGR5 knockdown mice showed similar results with TGR5 KO mice in AALD. Agonist treatment significantly activated TGR5 signaling in BAT, increased thermogenesis, reduced serum FFA level, and ameliorated hepatic steatosis and injury in AALD mice, while these effects were lost in TGR5 KO mice. CONCLUSIONS: BA signaling plays a protective role in AALD by enhancing BAT thermogenesis. Targeting TGR5 in BAT may be a promising approach for the treatment of AALD.


Subject(s)
Adipose Tissue, Brown , Bile Acids and Salts , Animals , Bile Acids and Salts/metabolism , Ethanol/toxicity , Humans , Liver/metabolism , Mice , Mice, Inbred C57BL
10.
Acta Pharm Sin B ; 11(6): 1541-1554, 2021 Jun.
Article in English | MEDLINE | ID: mdl-34221867

ABSTRACT

Obesity and its associated complications are highly related to a current public health crisis around the world. A growing body of evidence has indicated that G-protein coupled bile acid (BA) receptor TGR5 (also known as Gpbar-1) is a potential drug target to treat obesity and associated metabolic disorders. We have identified notoginsenoside Ft1 (Ft1) from Panax notoginseng as an agonist of TGR5 in vitro. However, the pharmacological effects of Ft1 on diet-induced obese (DIO) mice and the underlying mechanisms are still elusive. Here we show that Ft1 (100 mg/100 diet) increased adipose lipolysis, promoted fat browning in inguinal adipose tissue and induced glucagon-like peptide-1 (GLP-1) secretion in the ileum of wild type but not Tgr5 -/- obese mice. In addition, Ft1 elevated serum free and taurine-conjugated bile acids (BAs) by antagonizing Fxr transcriptional activities in the ileum to activate Tgr5 in the adipose tissues. The metabolic benefits of Ft1 were abolished in Cyp27a1 -/- mice which have much lower BA levels. These results identify Ft1 as a single compound with opposite activities on two key BA receptors to alleviate high fat diet-induced obesity and insulin resistance in mice.

11.
Proc Natl Acad Sci U S A ; 118(6)2021 02 09.
Article in English | MEDLINE | ID: mdl-33526687

ABSTRACT

Vertical sleeve gastrectomy (VSG) is one of the most effective and durable therapies for morbid obesity and its related complications. Although bile acids (BAs) have been implicated as downstream mediators of VSG, the specific mechanisms through which BA changes contribute to the metabolic effects of VSG remain poorly understood. Here, we confirm that high fat diet-fed global farnesoid X receptor (Fxr) knockout mice are resistant to the beneficial metabolic effects of VSG. However, the beneficial effects of VSG were retained in high fat diet-fed intestine- or liver-specific Fxr knockouts, and VSG did not result in Fxr activation in the liver or intestine of control mice. Instead, VSG decreased expression of positive hepatic Fxr target genes, including the bile salt export pump (Bsep) that delivers BAs to the biliary pathway. This reduced small intestine BA levels in mice, leading to lower intestinal fat absorption. These findings were verified in sterol 27-hydroxylase (Cyp27a1) knockout mice, which exhibited low intestinal BAs and fat absorption and did not show metabolic improvements following VSG. In addition, restoring small intestinal BA levels by dietary supplementation with taurocholic acid (TCA) partially blocked the beneficial effects of VSG. Altogether, these findings suggest that reductions in intestinal BAs and lipid absorption contribute to the metabolic benefits of VSG.


Subject(s)
Cholestanetriol 26-Monooxygenase/genetics , Gastrectomy/methods , Obesity, Morbid/surgery , Receptors, Cytoplasmic and Nuclear/genetics , Animals , Bile Acids and Salts/biosynthesis , Bile Acids and Salts/metabolism , Diet, High-Fat/adverse effects , Humans , Lipid Metabolism/genetics , Lipids/genetics , Mice , Mice, Knockout , Obesity, Morbid/metabolism , Obesity, Morbid/physiopathology , Weight Loss/genetics
12.
Theranostics ; 9(5): 1369-1384, 2019.
Article in English | MEDLINE | ID: mdl-30867837

ABSTRACT

Nuclear factor-kappa B (NF-κB) activation is critical for innate immune responses. However, cellular-intrinsic regulation of NF-κB activity during inflammatory diseases remains incompletely understood. Ubiquitin-like protein 4A (UBL4A, GdX) is a small adaptor protein involved in protein folding, biogenesis and transcription. Yet, whether GdX has a role during innate immune response is largely unknown. Methods: To investigate the involvement of GdX in innate immunity, we challenged GdX-deficient mice with lipopolysaccharides (LPS). To investigate the underlying mechanism, we performed RNA sequencing, real-time PCR, ELISA, luciferase reporter assay, immunoprecipitation and immunoblot analyses, flow cytometry, and structure analyses. To investigate whether GdX functions in inflammatory bowel disease, we generated dendritic cell (DC), macrophage (Mφ), epithelial-cell specific GdX-deficient mice and induced colitis with dextran sulfate sodium. Results: GdX enhances DC and Mφ-mediated innate immune defenses by positively regulating NF-κB signaling. GdX-deficient mice were resistant to LPS-induced endotoxin shock and DSS-induced colitis. DC- or Mφ- specific GdX-deficient mice displayed alleviated mucosal inflammation. The production of pro-inflammatory cytokines by GdX-deficient DCs and Mφ was reduced. Mechanistically, we found that tyrosine-protein phosphatase non-receptor type 2 (PTPN2, TC45) and protein phosphatase 2A (PP2A) form a complex with RelA (p65) to mediate its dephosphorylation whereas GdX interrupts the TC45/PP2A/p65 complex formation and restrict p65 dephosphorylation by trapping TC45. Conclusion: Our study provides a mechanism by which NF-κB signaling is positively regulated by an adaptor protein GdX in DC or Mφ to maintain the innate immune response. Targeting GdX could be a strategy to reduce over-activated immune response in inflammatory diseases.


Subject(s)
Colitis/pathology , Dendritic Cells/immunology , Immunity, Innate , Macrophages/immunology , NF-kappa B/metabolism , Signal Transduction , Ubiquitins/metabolism , Animals , Colitis/chemically induced , Dextran Sulfate/administration & dosage , Disease Models, Animal , Lipopolysaccharides/administration & dosage , Lipopolysaccharides/immunology , Mice , Mice, Knockout , Ubiquitins/deficiency
13.
Sci Rep ; 8(1): 11001, 2018 Jul 20.
Article in English | MEDLINE | ID: mdl-30030465

ABSTRACT

Various physiological stimuli, such as cold environment, diet, and hormones, trigger brown adipose tissue (BAT) to produce heat through sympathetic nervous system (SNS)- and ß-adrenergic receptors (ßARs). The ßAR stimulation increases intracellular cAMP levels through heterotrimeric G proteins and adenylate cyclases, but the processes by which cAMP modulates brown adipocyte function are not fully understood. Here we described that specific ablation of cAMP production in brown adipocytes led to reduced lipolysis, mitochondrial biogenesis, uncoupling protein 1 (Ucp1) expression, and consequently defective adaptive thermogenesis. Elevated cAMP signaling by sympathetic activation inhibited Salt-inducible kinase 2 (Sik2) through protein kinase A (PKA)-mediated phosphorylation in brown adipose tissue. Inhibition of SIKs enhanced Ucp1 expression in differentiated brown adipocytes and Sik2 knockout mice exhibited enhanced adaptive thermogenesis at thermoneutrality in an Ucp1-dependent manner. Taken together, our data indicate that suppressing Sik2 by PKA-mediated phosphorylation is a requisite for SNS-induced Ucp1 expression and adaptive thermogenesis in BAT, and targeting Sik2 may present a novel therapeutic strategy to ramp up BAT thermogenic activity in humans.


Subject(s)
Adipose Tissue, Brown/physiology , Sympathetic Nervous System/physiology , Thermogenesis/physiology , Animals , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Humans , Mice , Phosphorylation , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Receptors, Adrenergic, beta/metabolism , Uncoupling Protein 1/metabolism
14.
Cell Metab ; 27(3): 616-629.e4, 2018 03 06.
Article in English | MEDLINE | ID: mdl-29514069

ABSTRACT

Brown adipose tissue (BAT) thermogenesis is critical for thermoregulation and contributes to total energy expenditure. However, whether BAT has non-thermogenic functions is largely unknown. Here, we describe that BAT-specific liver kinase b1 knockout (Lkb1BKO) mice exhibited impaired BAT mitochondrial respiration and thermogenesis but reduced adiposity and liver triglyceride accumulation under high-fat-diet feeding at room temperature. Importantly, these metabolic benefits were also present in Lkb1BKO mice at thermoneutrality, where BAT thermogenesis was not required. Mechanistically, decreased mRNA levels of mtDNA-encoded electron transport chain (ETC) subunits and ETC proteome imbalance led to defective BAT mitochondrial respiration in Lkb1BKO mice. Furthermore, reducing mtDNA gene expression directly in BAT by removing mitochondrial transcription factor A (Tfam) in BAT also showed ETC proteome imbalance and the trade-off between BAT thermogenesis and systemic metabolism at room temperature and thermoneutrality. Collectively, our data demonstrate that ETC proteome imbalance in BAT regulates systemic metabolism independently of thermogenesis.


Subject(s)
Adipocytes, Brown/metabolism , Adipose Tissue, Brown/metabolism , Mitochondria/metabolism , Mitochondrial Membranes/metabolism , Proteome/metabolism , AMP-Activated Protein Kinases , Adiposity , Animals , DNA-Binding Proteins/metabolism , Diet, High-Fat , Electron Transport , Liver/metabolism , Mice, Inbred C57BL , Mitochondrial Proteins/metabolism , Oxygen/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Thermogenesis , Transcription Factors/metabolism , Triglycerides/metabolism
15.
Diabetes ; 66(12): 2952-2963, 2017 12.
Article in English | MEDLINE | ID: mdl-28882900

ABSTRACT

Uncoupling protein 1+ beige adipocytes are dynamically regulated by environment in rodents and humans; cold induces formation of beige adipocytes, whereas warm temperature and nutrient excess lead to their disappearance. Beige adipocytes can form through de novo adipogenesis; however, how "beiging" characteristics are maintained afterward is largely unknown. In this study, we show that beige adipocytes formed postnatally in subcutaneous inguinal white adipose tissue lost thermogenic gene expression and multilocular morphology at the adult stage, but cold restored their beiging characteristics, a phenomenon termed beige adipocyte renaissance. Ablation of these postnatal beige adipocytes inhibited cold-induced beige adipocyte formation in adult mice. Furthermore, we demonstrated that beige adipocyte renaissance was governed by liver kinase b1 and histone deacetylase 4 in white adipocytes. Although neither presence nor thermogenic function of uncoupling protein 1+ beige adipocytes contributed to metabolic fitness in adipocyte liver kinase b1-deficient mice, our results reveal an unexpected role of white adipocytes in maintaining properties of preexisting beige adipocytes.


Subject(s)
Adipocytes, Beige/metabolism , Histone Deacetylases/physiology , Protein Serine-Threonine Kinases/physiology , AMP-Activated Protein Kinases , Animals , Cold Temperature , Diet, High-Fat , Female , Male , Mice , Mice, Inbred C57BL , Thermogenesis , Uncoupling Protein 1/analysis , Uncoupling Protein 1/physiology
16.
FEBS Lett ; 589(15): 1890-6, 2015 Jul 08.
Article in English | MEDLINE | ID: mdl-26026268

ABSTRACT

STAT3 is associated with embryo development and survival as well as proliferation and metastasis of tumor cells. In a previous study, we demonstrated that STAT3-Interacting Protein As a Repressor (SIPAR) enhances the dephosphorylation of STAT3 and negatively regulates its activity. However, it remains unclear how SIPAR inhibits phosphorylation of STAT3. Here we demonstrate that SIPAR directly interacts with T cell protein tyrosine phosphatase TC45 and enhances its association with STAT3. This interaction triggers an accelerated dephosphorylation process for STAT3. Furthermore, SIPAR inhibits the transcriptional activity of STAT3 in wild-type MEF cells but not in TC45 null MEF cells. These results suggest that SIPAR terminates the activation of STAT3 through a dephosphorylation process that is dependent upon interaction with TC45 in the nucleus.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Nucleus/metabolism , Nuclear Proteins/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 2/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , Animals , Cells, Cultured , HEK293 Cells , Humans , Mice
17.
Mol Cell ; 53(5): 752-65, 2014 Mar 06.
Article in English | MEDLINE | ID: mdl-24530303

ABSTRACT

Impaired phosphatase activity contributes to the persistent activation of STAT3 in tumors. Given that STAT family members with various or even opposite functions are often phosphorylated or dephosphorylated by the same enzymes, the mechanism for STAT3-specific dephosphorylation in cells remains largely unknown. Here, we report that GdX (UBL4A) promotes STAT3 dephosphorylation via mediating the interaction between TC45 (the nuclear isoform of TC-PTP) and STAT3 specifically. GdX stabilizes the TC45-STAT3 complex to bestow upon STAT3 an efficient dephosphorylation by TC45. Inasmuch, GdX suppresses tumorigenesis and tumor development by reducing the level of phospho-STAT3 (p-STAT3), whereas deletion of GdX results in a high level of p-STAT3 and accelerated colorectal tumorigenesis induced by AOM/DSS. Thus, GdX converts TC45, a nonspecific phosphatase, into a STAT3-specific phosphatase by bridging an association between TC45 and STAT3.


Subject(s)
Carcinogenesis , Gene Expression Regulation, Neoplastic , Protein Tyrosine Phosphatase, Non-Receptor Type 2/chemistry , STAT3 Transcription Factor/chemistry , Ubiquitins/chemistry , Animals , COS Cells , Cell Transformation, Neoplastic , Chlorocebus aethiops , Cytokines/metabolism , Fibroblasts/metabolism , Gene Deletion , Humans , MCF-7 Cells , Melanoma, Experimental , Mice , Mice, Inbred BALB C , Neoplasm Metastasis , Neoplasm Transplantation , Phosphorylation , Protein Binding , Ubiquitins/genetics
18.
Carcinogenesis ; 35(5): 983-91, 2014 May.
Article in English | MEDLINE | ID: mdl-24302614

ABSTRACT

The carboxyl terminus of Hsc70-interacting protein (CHIP, also named Stub1), a U-box containing E3 ubiquitin ligase, is involved in degradation of certain oncogenic proteins. Recent studies indicated that CHIP suppresses tumor progression in human cancers by targeting Src-3, hypoxia inducible factor 1α, NF-κB, ErbB2 and c-Myc. Here, we report that CHIP was downregulated, predominantly, in the late stages of human colorectal cancer (CRC), and that the CHIP promoter was hypermethylated in CRC specimens. Overexpression of CHIP in HCT-116 cells resulted in impaired tumor growth in nude mice and decreased abilities of tumor cell migration and invasion. Conversely, depletion of CHIP in HCT-116 cells promoted tumor growth and increased tumor cell migration and invasion. CHIP was further found to negatively regulate NF-κB signaling in HCT-116 cells by promoting ubiquitination and degradation of p65, a subunit of the NF-κB complex. The suppressive effect of CHIP led to decreased expression of NF-κB-targeted oncogenes including Cyclin D1, c-Myc, MMP-2, VEGF and IL-8. We proposed that CHIP inhibits the malignancy of CRC cells, possibly through targeting NF-κB signaling. This study provides functional evidence for CHIP as a potential tumor suppressor in CRC, and CHIP expression may be a marker for stages of CRC.


Subject(s)
Colorectal Neoplasms/metabolism , NF-kappa B/metabolism , Signal Transduction , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Aged , Aged, 80 and over , Animals , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , DNA Methylation , Disease Models, Animal , Female , Gene Expression Regulation, Neoplastic , HCT116 Cells , Heterografts , Humans , Male , Mice , Middle Aged , Neoplasm Staging , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Promoter Regions, Genetic , Transcription Factor RelA/metabolism , Tumor Burden , Tumor Suppressor Proteins/genetics , Ubiquitin-Protein Ligases/genetics
19.
Cell Signal ; 25(11): 2272-80, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23917203

ABSTRACT

Persistently activated STAT3 is important for tumorigenesis in a variety of cancers, including melanoma. Although many co-factors in the regulation of STAT3 activity have been identified, it remains unclear how STAT3 phosphorylation is negatively regulated. Here, we report that SIPAR (STAT3-Interacting Protein As a Repressor) inhibits STAT3 activity by accelerating its dephosphorylation. We observed that SIPAR directly interacted with STAT3 upon IL-6 stimulation. Moreover, over-expression of SIPAR reduced, whereas depletion enhanced, the level of phosphorylated STAT3. We further demonstrated that SIPAR inhibited the growth of melanoma cells by decreasing the level of phosphorylated STAT3 and the expression of its target genes. These results suggest that SIPAR, functioning as a new negative regulator, inhibits STAT3 activity by enhancing its dephosphorylation and represses melanoma progression.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Gene Expression Regulation, Neoplastic , Melanoma, Experimental/genetics , Nuclear Proteins/genetics , STAT3 Transcription Factor/genetics , Skin Neoplasms/genetics , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/metabolism , Amino Acid Sequence , Animals , Cell Line, Tumor , Disease Progression , Genes, Reporter , Humans , Interleukin-6/pharmacology , Luciferases/genetics , Luciferases/metabolism , Male , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Mice, Nude , Molecular Sequence Data , NIH 3T3 Cells , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/metabolism , Phosphorylation , STAT3 Transcription Factor/metabolism , Sequence Homology, Amino Acid , Signal Transduction/drug effects , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Tumor Burden
20.
Breast Cancer Res ; 14(2): R38, 2012 Mar 06.
Article in English | MEDLINE | ID: mdl-22394684

ABSTRACT

INTRODUCTION: Signal transducer and activator of transcription 3 (STAT3) is over-activated or phosphorylated in breast cancers. The hyper-phosphorylation of STAT3 was attributed to either up-regulated phosphorylation by several tyrosine-kinases or down-regulated activity of phosphatases. Although several factors have been identified to phosphorylate STAT3, it remains unclear how STAT3 is dephosphorylated by PTPMeg2. The aim of this study was to determine the role of PTPMeg2 as a phosphatase in regulation of the activity of STAT3 in breast cancers. METHODS: Immunoprecipitation assays were used to study the interaction of STAT3 with PTPMeg2. A series of biochemistry experiments were performed to evaluate the role of PTPMeg2 in the dephosphorylation of STAT3. Two breast cancer cell lines MCF7 (PTPMeg2 was depleted as it was endogenously high) and MDA-MB-231 (PTPMeg2 was overexpressed as it was endogenously low) were used to compare the level of phosphorylated STAT3 and the tumor growth ability in vitro and in vivo. Samples from breast carcinoma (n = 73) were subjected to a pair-wise Pearson correlation analysis for the correlation of levels of PTPMeg2 and phosphorylated STAT3. RESULTS: PTPMeg2 directly interacts with STAT3 and mediates its dephosphorylation in the cytoplasm. Over-expression of PTPMeg2 decreased tyrosine phosphorylation of STAT3 while depletion of PTPMeg2 increased its phosphorylation. The decreased tyrosine phosphorylation of STAT3 is coupled with suppression of STAT3 transcriptional activity and reduced tumor growth in vitro and in vivo. Levels of PTPMeg2 and phosphorylated STAT3 were inversely correlated in breast cancer tissues (P = 0.004). CONCLUSIONS: PTPMeg2 is an important phosphatase for the dephosphorylation of STAT3 and plays a critical role in breast cancer development.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , STAT3 Transcription Factor/metabolism , Animals , Base Sequence , Cell Line, Tumor , Cell Proliferation , Female , Gene Knockout Techniques , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Molecular Sequence Data , Phosphorylation , Protein Tyrosine Phosphatases, Non-Receptor/genetics , STAT3 Transcription Factor/genetics , Signal Transduction , Tyrosine/metabolism , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...