Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
FASEB J ; 33(5): 5850-5863, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30726112

RESUMO

Sarcopenic obesity, the combination of skeletal muscle mass and function loss with an increase in body fat, is associated with physical limitations, cardiovascular diseases, metabolic stress, and increased risk of mortality. Cannabinoid receptor type 1 (CB1R) plays a critical role in the regulation of whole-body energy metabolism because of its involvement in controlling appetite, fuel distribution, and utilization. Inhibition of CB1R improves insulin secretion and insulin sensitivity in pancreatic ß-cells and hepatocytes. We have now developed a skeletal muscle-specific CB1R-knockout (Skm-CB1R-/-) mouse to study the specific role of CB1R in muscle. Muscle-CB1R ablation prevented diet-induced and age-induced insulin resistance by increasing IR signaling. Moreover, muscle-CB1R ablation enhanced AKT signaling, reducing myostatin expression and increasing IL-6 secretion. Subsequently, muscle-CB1R ablation increased myogenesis through its action on MAPK-mediated myogenic gene expression. Consequently, Skm-CB1R-/- mice had increased muscle mass and whole-body lean/fat ratio in obesity and aging. Muscle-CB1R ablation improved mitochondrial performance, leading to increased whole-body muscle energy expenditure and improved physical endurance, with no change in body weight. These results collectively show that CB1R in muscle is sufficient to regulate whole-body metabolism and physical performance and is a novel target for the treatment of sarcopenic obesity. -González-Mariscal, I., Montoro, R. A., O'Connell, J. F., Kim, Y., Gonzalez-Freire, M., Liu, Q.-R., Alfaras, I., Carlson, O. D., Lehrmann, E., Zhang, Y., Becker, K. G., Hardivillé, S., Ghosh, P., Egan, J. M. Muscle cannabinoid 1 receptor regulates Il-6 and myostatin expression, governing physical performance and whole-body metabolism.


Assuntos
Interleucina-6/metabolismo , Músculo Esquelético/metabolismo , Miostatina/metabolismo , Receptor CB1 de Canabinoide/metabolismo , Transdução de Sinais , Envelhecimento , Animais , Composição Corporal , Peso Corporal , Linhagem Celular , Dieta , Feminino , Hepatócitos/metabolismo , Insulina/metabolismo , Resistência à Insulina , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Masculino , Camundongos , Camundongos Knockout , Análise de Sequência com Séries de Oligonucleotídeos , Fosfoproteínas/metabolismo
2.
Diabetologia ; 61(6): 1470-1483, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29497784

RESUMO

AIMS/HYPOTHESIS: The cannabinoid 1 receptor (CB1R) regulates insulin sensitivity and glucose metabolism in peripheral tissues. CB1R is expressed on pancreatic beta cells and is coupled to the G protein Gαi, suggesting a negative regulation of endogenous signalling in the beta cell. Deciphering the exact function of CB1R in beta cells has been confounded by the expression of this receptor on multiple tissues involved in regulating metabolism. Thus, in models of global genetic or pharmacological CB1R blockade, it is difficult to distinguish the indirect effects of improved insulin sensitivity in peripheral tissues from the direct effects of inhibiting CB1R in beta cells per se. To assess the direct contribution of beta cell CB1R to metabolism, we designed a mouse model that allows us to determine the role of CB1R specifically in beta cells in the context of whole-body metabolism. METHODS: We generated a beta cell specific Cnr1 (CB1R) knockout mouse (ß-CB1R-/-) to study the long-term consequences of CB1R ablation on beta cell function in adult mice. We measured beta cell function, proliferation and viability in these mice in response to a high-fat/high-sugar diet and induction of acute insulin resistance with the insulin receptor antagonist S961. RESULTS: ß-CB1R-/- mice had increased fasting (153 ± 23% increase at 10 weeks of age) and stimulated insulin secretion and increased intra-islet cAMP levels (217 ± 33% increase at 10 weeks of age), resulting in primary hyperinsulinaemia, as well as increased beta cell viability, proliferation and islet area (1.9-fold increase at 10 weeks of age). Hyperinsulinaemia led to insulin resistance, which was aggravated by a high-fat/high-sugar diet and weight gain, although beta cells maintained their insulin secretory capacity in response to glucose. Strikingly, islets from ß-CB1R-/- mice were protected from diet-induced inflammation. Mechanistically, we show that this is a consequence of curtailment of oxidative stress and reduced activation of the NLRP3 inflammasome in beta cells. CONCLUSIONS/INTERPRETATION: Our data demonstrate CB1R to be a negative regulator of beta cell function and a mediator of islet inflammation under conditions of metabolic stress. Our findings point to beta cell CB1R as a therapeutic target, and broaden its potential to include anti-inflammatory effects in both major forms of diabetes. DATA AVAILABILITY: Microarray data have been deposited at GEO (GSE102027).


Assuntos
Células Secretoras de Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Receptor CB1 de Canabinoide/genética , Animais , Peso Corporal , Proliferação de Células , Sobrevivência Celular , Dieta Hiperlipídica/efeitos adversos , Carboidratos da Dieta/efeitos adversos , Inflamação/patologia , Insulina/metabolismo , Células Secretoras de Insulina/patologia , Ilhotas Pancreáticas/fisiopatologia , Masculino , Camundongos , Camundongos Knockout , Estresse Oxidativo
3.
Eur J Immunol ; 46(1): 212-22, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26514620

RESUMO

The TORC1 pathway is necessary for ribosomal biogenesis and initiation of protein translation. Furthermore, the differentiation of Th1 and Th17 cells requires TORC1 activity. To investigate the role of the TORC1 pathway in the differentiation of Th1 and/or Th17 cells in more detail, we compared the differentiation capacity of naïve T cells from wild type and p70(S6K1) knockout mice. Expression of many of the genes associated with Th17-cell differentiation, such as IL17a, IL17f, and IL-23R, were reduced in p70(S6K1) knockout mice. In contrast, the development of Th1, Th2, and Treg cells was unaffected in the absence of p70(S6K1) . Furthermore, expression of the major transcription factor in Th17-cell differentiation, retinoic acid receptor-related orphan receptor gamma T, remained unchanged. However, the acetylation of histone 3 at the promoters of IL17a and IL17f was reduced in the absence of p70(S6K1) . In accordance with the in vitro data, the kinetics, but not the development, of EAE was affected with the loss of p70(S6K1) expression. Collectively, our findings suggested that both in vitro and in vivo differentiation of Th17 cells were positively regulated by p70(S6K1) .


Assuntos
Diferenciação Celular/imunologia , Encefalomielite Autoimune Experimental/imunologia , Proteínas Quinases S6 Ribossômicas 70-kDa/imunologia , Subpopulações de Linfócitos T/imunologia , Células Th17/citologia , Animais , Imunoprecipitação da Cromatina , Feminino , Citometria de Fluxo , Immunoblotting , Ativação Linfocitária/imunologia , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Camundongos Knockout , Complexos Multiproteicos/imunologia , Subpopulações de Linfócitos T/citologia , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/imunologia , Serina-Treonina Quinases TOR/imunologia , Células Th1/citologia , Células Th1/imunologia , Células Th17/imunologia , Células Th2/citologia , Células Th2/imunologia
4.
J Immunol ; 195(12): 5780-6, 2015 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-26566676

RESUMO

Mitofusin 2 (Mfn2), a mitochondrial protein, was shown to have antiproliferative properties when overexpressed. In this article, we show that activation of resting human peripheral blood T cells caused downregulation of Mfn2 levels. This downregulation of Mfn2 was blocked by different inhibitors (mTOR inhibitor rapamycin, PI3K inhibitor LY294002, and Akt inhibitor A443654), producing cells that were arrested in the G0/G1 stage of the cell cycle. Furthermore, the activation-induced downregulation of Mfn2 preceded the entry of the cells into the cell cycle, suggesting that Mfn2 downregulation is a prerequisite for activated T cell entry into the cell cycle. Accordingly, small interfering RNA-mediated knockdown of Mfn2 resulted in increased T cell proliferation. Overexpression of constitutively active AKT resulted in the downregulation of Mfn2, which can be blocked by a proteasome inhibitor. Akt-mediated downregulation of Mfn2 was via the mTORC1 pathway because this downregulation was blocked by rapamycin, and overexpression of wild-type, but not kinase-dead mTOR, caused Mfn2 downregulation. Our data suggested that activation-induced reactive oxygen species production plays an important role in the downregulation of Mfn2. Collectively, our data suggest that the PI3K-AKT-mTOR pathway plays an important role in activation-induced downregulation of Mfn2 and subsequent proliferation of resting human T cells.


Assuntos
GTP Fosfo-Hidrolases/metabolismo , Leucócitos Mononucleares/imunologia , Proteínas Mitocondriais/metabolismo , Linfócitos T/fisiologia , Ciclo Celular/genética , Linhagem Celular Tumoral , Proliferação de Células/genética , Cromonas/farmacologia , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , GTP Fosfo-Hidrolases/genética , Humanos , Indazóis/farmacologia , Indóis/farmacologia , Ativação Linfocitária , Proteínas Mitocondriais/genética , Morfolinas/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Interferente Pequeno/genética , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/genética , Sirolimo/farmacologia , Linfócitos T/efeitos dos fármacos , Serina-Treonina Quinases TOR/metabolismo
5.
FASEB J ; 28(1): 382-94, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24081906

RESUMO

It has been reported that Mitofusin2 (Mfn2) inhibits cell proliferation when overexpressed. We wanted to study the role of endogenous Mfn2 in cell proliferation, along with the structural features of Mfn2 that influence its mitochondrial localization and control of cell proliferation. Mfn2-knockdown clones of a B-cell lymphoma cell line BJAB exhibited an increased rate of cell proliferation. A 2-fold increase in cell proliferation was also observed in Mfn2-knockout mouse embryonic fibroblast (MEF) cells as compared with the control wild-type cells, and the proliferative advantage of the knockout MEF cells was blocked on reintroduction of the Mfn2 gene. Mfn2 exerts its antiproliferative effect by acting as an effector molecule of Ras, resulting in the inhibition of the Ras-Raf-ERK signaling pathway. Furthermore, both the N-terminal (aa 1-264) and the C-terminal (aa 265-757) fragments of Mfn2 blocked cell proliferation through distinct mechanisms: the N-terminal-mediated inhibition was due to its interaction with Raf-1, whereas the C-terminal fragment of Mfn2 inhibited cell proliferation by interacting with Ras. The inhibition of proliferation by the N-terminal fragment was independent of its mitochondrial localization. Collectively, our data provide new insights regarding the role of Mfn2 in controlling cellular proliferation.


Assuntos
GTP Fosfo-Hidrolases/metabolismo , Proteínas Mitocondriais/metabolismo , Ciclo Celular/genética , Ciclo Celular/fisiologia , Linhagem Celular Tumoral , Proliferação de Células , Citometria de Fluxo , GTP Fosfo-Hidrolases/genética , Humanos , Lentivirus/genética , Proteínas Mitocondriais/genética , Consumo de Oxigênio/genética , Consumo de Oxigênio/fisiologia
6.
Life Sci ; 340: 122460, 2024 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-38286207

RESUMO

AIMS: Extracellular vesicles (EVs) are involved in intercellular communication and are a topic of increasing interest due to their therapeutic potential. The aim of this study was to determine whether human islet-derived EVs contain insulin, and if so, what role do they play in glucose stimulated insulin secretion. MAIN METHODS: We isolated EVs from human islets culture and plasma to probe for insulin. Plasma from hyperglycemic glucose clamp experiments were also used to isolate and measure EV insulin content in response to a secretory stimulus. We performed immunogold electron microscopy for insulin presence in EVs. Co-culture experiments of isolated EVs with fresh islets were performed to examine the effect of EV cargo on insulin receptor signaling. KEY FINDINGS: EVs isolated from culture medium contained insulin. Glucose treatment of islets increased the level of EV insulin. Hyperglycemic glucose clamp experiments in humans also lead to increased levels of insulin in plasma-derived EVs. Immunogold electron microscopy and proteinase K-digestion experiments demonstrated that insulin in EVs predominantly associated with the exterior surface of EVs while western blot analyses uncovered the presence of only preproinsulin in EVs. Membrane-bound preproinsulin in EVs was capable of activating insulin signaling pathway in an insulin receptor-dependent manner. The physiological relevance of this finding was observed in priming of human naïve islets by EVs during glucose stimulated insulin secretion. SIGNIFICANCE: Our data suggest that (1) human islets secret insulin via an alternate pathway (EV-mediated) other than conventional granule-mediated insulin secretion, and (2) EV membrane bound preproinsulin is biologically active.


Assuntos
Vesículas Extracelulares , Células Secretoras de Insulina , Ilhotas Pancreáticas , Precursores de Proteínas , Humanos , Células Secretoras de Insulina/metabolismo , Secreção de Insulina , Receptor de Insulina/metabolismo , Glucose/metabolismo , Insulina/metabolismo , Vesículas Extracelulares/metabolismo , Ilhotas Pancreáticas/metabolismo
7.
J Biol Chem ; 287(27): 23184-95, 2012 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-22621932

RESUMO

Previously we reported that TGF-ß1-induced growth suppression was associated with a decrease in mutant p53 levels in B-cell lymphoma cells. The goal of the present study was to understand the mechanism involved in TGF-ß1-mediated down-regulation of mutant p53. In RL and CA46, two B-cell lymphoma cell lines, TGF-ß1 treatment caused down-regulation of E2F-1 transcription factor resulting in the down-regulation of both p14(ARF) and mutant p53, leading to growth arrest. Experimental overexpression of E2F-1 increased p14(ARF) level and blocked TGF-ß1-induced down-regulation of p14(ARF). Overexpression of p14(ARF) blocked the down-regulation of mutant p53 and prevented growth arrest. p14(ARF) also attenuated TGF-ß1-induced p21(Cip1/WAF1) induction, which was reversible by p53 siRNA, indicating the involvement of mutant p53 in controlling the TGF-ß1-induced expression of p21(Cip1/WAF1). The interaction observed between phospho-Smad2 and mutant p53 in the nucleus could be the mechanism responsible for blocking the growth-suppressive effects of TGF-ß1. In RL cells, p14(ARF) is present in a trimer consisting of mutant p53-Mdm2-p14(ARF) and in a dimer consisting of Mdm2-p14(ARF). Because it is known that Mdm2 can degrade p53, it is possible that, in its trimeric form, p14(ARF) is able to stabilize mutant p53 by inhibiting Mdm2. In its dimeric form, p14(ARF) may be sequestering Mdm2, limiting its ability to degrade p53. Collectively, these data demonstrate a unique mechanism in which the inhibition of TGF-ß1-mediated growth suppression by mutant p53 can be reversed by the down-regulation of its stabilizing protein p14(ARF). This work suggests that the high levels of p14(ARF) often found in tumor cells could be a potential therapeutic target.


Assuntos
Divisão Celular/efeitos dos fármacos , Linfoma de Células B/tratamento farmacológico , Fator de Crescimento Transformador beta1/farmacologia , Proteína Supressora de Tumor p14ARF/metabolismo , Proteína Supressora de Tumor p53/genética , Antineoplásicos/farmacologia , Divisão Celular/fisiologia , Inibidor de Quinase Dependente de Ciclina p21/genética , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/fisiologia , Desenho de Fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/fisiologia , Humanos , Linfoma de Células B/genética , Linfoma de Células B/patologia , Regiões Promotoras Genéticas/fisiologia , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/genética , Proteína Smad2/metabolismo , Células Tumorais Cultivadas , Proteína Supressora de Tumor p14ARF/genética , Proteína Supressora de Tumor p53/metabolismo
8.
J Biol Chem ; 286(2): 1093-102, 2011 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-21071440

RESUMO

Tumor necrosis factor (TNF) induces expression of granulocyte-macrophage colony-stimulating factor (GM-CSF) but lymphotoxin ß (LTß) does not. Here we report that priming of cells with agonistic LTß receptor antibody synergistically enhanced TNF-induced GM-CSF expression. The LTß priming process was not due to an increase in TNF-mediated nuclear translocation of p65, p65 DNA binding, or NF-κB transactivational activity. The synergistic effect of LTß priming was not observed with other TNF-responsive genes such as Ccl2 or RelB, which suggested that this effect was not a general increase in TNF signaling. Furthermore, RelB and p65 were both independently recruited to the GM-CSF promoter when cells were primed with LTß followed by TNF treatment. As a consequence, an increase in both chromatin accessibility and the recruitment of RNA polymerase II were observed to the GM-CSF promoter. Taken together, these findings suggested that LTß signaling amplified TNF-mediated GM-CSF expression by facilitating chromatin access and the co-recruitment of RNA polymerase II to increase gene transcription. Moreover, the novel priming process described here underscores the complexity of the interactions between the classical and alternative NF-κB signaling pathways.


Assuntos
Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Transdução de Sinais/fisiologia , Fator de Transcrição RelA/metabolismo , Fator de Transcrição RelB/metabolismo , Transporte Ativo do Núcleo Celular/fisiologia , Animais , Anticorpos Monoclonais/farmacologia , Receptor beta de Linfotoxina/imunologia , Receptor beta de Linfotoxina/metabolismo , Camundongos , Células NIH 3T3 , Fosforilação/fisiologia , Regiões Promotoras Genéticas/fisiologia , Proteínas Serina-Treonina Quinases/metabolismo , RNA Polimerase II/metabolismo , Transcrição Gênica/fisiologia , Fator de Necrose Tumoral alfa/metabolismo , Quinase Induzida por NF-kappaB
9.
Sci Rep ; 12(1): 14049, 2022 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-35982068

RESUMO

Even before the COVID-19 pandemic declines in life expectancy in the United States were attributed to increased mortality rates in midlife adults across racial and ethnic groups, indicating a need for markers to identify individuals at risk for early mortality. Extracellular vesicles (EVs) are small, lipid-bound vesicles capable of shuttling functional proteins, nucleic acids, and lipids. Given their role as intercellular communicators and potential biomarkers of disease, we explored whether circulating EVs may be markers of mortality in a prospective, racially, and socioeconomically diverse middle-aged cohort. We isolated plasma EVs from 76 individuals (mean age = 59.6 years) who died within a 5 year period and 76 surviving individuals matched by age, race, and poverty status. There were no significant differences in EV concentration, size, or EV-associated mitochondrial DNA levels associated with mortality. We found that several EV-associated inflammatory proteins including CCL23, CSF-1, CXCL9, GDNF, MCP-1, STAMBP, and 4E-BP1 were significantly associated with mortality. IL-10RB and CDCP1 were more likely to be present in plasma EVs from deceased individuals than in their alive counterparts. We also report differences in EV-associated inflammatory proteins with poverty status, race, and sex. Our results suggest that plasma EV-associated inflammatory proteins are promising potential clinical biomarkers of mortality.


Assuntos
COVID-19 , Vesículas Extracelulares , Adulto , Antígenos de Neoplasias/metabolismo , Biomarcadores , Proteínas Sanguíneas/metabolismo , Moléculas de Adesão Celular/metabolismo , Vesículas Extracelulares/metabolismo , Humanos , Pessoa de Meia-Idade , Pandemias , Estudos Prospectivos
10.
Front Pharmacol ; 12: 809965, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35115945

RESUMO

Type 1 diabetes mellitus (T1DM) is an autoimmune disease resulting from loss of insulin-secreting ß-cells in islets of Langerhans. The loss of ß-cells is initiated when self-tolerance to ß-cell-derived contents breaks down, which leads to T cell-mediated ß-cell damage and, ultimately, ß-cell apoptosis. Many investigations have demonstrated the positive effects of antagonizing cannabinoid receptor 1 (CB1R) in metabolic diseases such as fatty liver disease, obesity, and diabetes mellitus, but the role of cannabinoid receptor 2 (CB2R) in such diseases is relatively unknown. Activation of CB2R is known for its immunosuppressive roles in multiple sclerosis, rheumatoid arthritis, Crohn's, celiac, and lupus diseases, and since autoimmune diseases can share common environmental and genetic factors, we propose CB2R specific agonists may also serve as disease modifiers in diabetes mellitus. The CNR2 gene, which encodes CB2R protein, is the result of a gene duplication of CNR1, which encodes CB1R protein. This ortholog evolved rapidly after transitioning from invertebrates to vertebrate hundreds of million years ago. Human specific CNR2 isoforms are induced by inflammation in pancreatic islets, and a CNR2 nonsynonymous SNP (Q63R) is associated with autoimmune diseases. We collected evidence from the literature and from our own studies demonstrating that CB2R is involved in regulating the inflammasome and especially release of the cytokine interleukin 1B (IL-1ß). Furthermore, CB2R activation controls intracellular autophagy and may regulate secretion of extracellular vesicles from adipocytes that participate in recycling of lipid droplets, dysregulation of which induces chronic inflammation and obesity. CB2R activation may play a similar role in islets of Langerhans. Here, we will discuss future strategies to unravel what roles, if any, CB2R modifiers potentially play in T1DM.

11.
Aging Cell ; 20(1): e13283, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33355987

RESUMO

The mitochondrial free radical theory of aging suggests that accumulating oxidative damage to mitochondria and mitochondrial DNA (mtDNA) plays a central role in aging. Circulating cell-free mtDNA (ccf-mtDNA) isolated from blood may be a biomarker of disease. Extracellular vesicles (EVs) are small (30-400 nm), lipid-bound vesicles capable of shuttling proteins, nucleic acids, and lipids as part of intercellular communication systems. Here, we report that a portion of ccf-mtDNA in plasma is encapsulated in EVs. To address whether EV mtDNA levels change with human age, we analyzed mtDNA in EVs from individuals aged 30-64 years cross-sectionally and longitudinally. EV mtDNA levels decreased with age. Furthermore, the maximal mitochondrial respiration of cultured cells was differentially affected by EVs from old and young donors. Our results suggest that plasma mtDNA is present in EVs, that the level of EV-derived mtDNA is associated with age, and that EVs affect mitochondrial energetics in an EV age-dependent manner.


Assuntos
DNA Mitocondrial/genética , Vesículas Extracelulares/metabolismo , Adulto , Envelhecimento , Humanos , Pessoa de Meia-Idade
12.
Cell Death Dis ; 11(12): 1044, 2020 12 09.
Artigo em Inglês | MEDLINE | ID: mdl-33298885

RESUMO

Cannabinoid 1 receptor (CB1R) expression is upregulated in the liver with viral hepatitis, cirrhosis, and both alcoholic and non-alcoholic fatty liver disease (FLD), whereas its expression is muted under usual physiological conditions. Inhibiting CB1R has been shown to be beneficial in preserving hepatic function in FLD but it is unclear if inhibiting CB1R during an inflammatory response to an acute hepatic injury, such as toxin-induced injury, would also be beneficial. We found that intrinsic CB1R in hepatocytes regulated liver inflammation-related gene transcription. We tested if nullification of hepatocyte-specific CB1R (hCNR1-/-) in mice protects against concanavalin A (Con A)-induced liver injury. We looked for evidence of liver damage and markers of inflammation in response to Con A by measuring liver enzyme levels and proinflammatory cytokines (e.g., TNF-α, IL-1ß, IL-6, IL-17) in serum collected from hCNR1-/- and control mice. We observed a shift to the right in the dose-response curve for liver injury and inflammation in hCNR1-/- mice. We also found less inflammatory cell infiltration and focal necrosis in livers of hCNR1-/- mice compared to controls, resulting from downregulated apoptotic markers. This anti-apoptotic mechanism results from increased activation of nuclear factor kappa B (NF-κB), especially cAMP-dependent cannabinoid signaling and membrane-bound TNF-α, via downregulated TNF-α receptor 2 (TNFR2) transcription levels. Collectively, these findings provide insight into involvement of CB1R in the pathogenesis of acute liver injury.


Assuntos
Concanavalina A/toxicidade , Hepatócitos/metabolismo , Fígado/metabolismo , Fígado/patologia , NF-kappa B/metabolismo , Receptor CB1 de Canabinoide/deficiência , Transdução de Sinais , Animais , Apoptose/efeitos dos fármacos , Membrana Celular/metabolismo , AMP Cíclico/metabolismo , Hepatócitos/efeitos dos fármacos , Inflamação/patologia , Fígado/efeitos dos fármacos , Masculino , Camundongos , Modelos Biológicos , Ligação Proteica , Receptor CB1 de Canabinoide/genética , Receptor CB1 de Canabinoide/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fator de Necrose Tumoral alfa/metabolismo
13.
Mol Immunol ; 45(6): 1799-806, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17983655

RESUMO

Activation-induced deaminase (AID) is expressed in activated B lymphocytes and initiates somatic hypermutation and class switch recombination. To determine if different stimuli affect the expression and function of AID, we monitored AID activity in murine B cells stimulated ex vivo with various ligands. AID was rapidly expressed at both the RNA and protein levels following stimulation with LPS, LPS plus IL-4, and anti-CD40 plus IL-4, but was delayed after stimulation with anti-IgM plus IL-4. By day 4, AID was expressed in all groups; however, cells stimulated with anti-IgM plus IL-4 did not undergo switch recombination. These cells expressed normal levels of gamma 1 germline transcripts, implying that the gamma 1 switch region was accessible. Furthermore, switching was suppressed by the addition of anti-IgM to cells stimulated with LPS plus IL-4 or anti-CD40 plus IL-4, even though AID was expressed. The lack of class switch recombination could be reversed by inhibition of phosphatidylinositol 3-kinase (PI3K). This suggests that activation through the B cell receptor induces PI3K, which interferes with the function of AID.


Assuntos
Linfócitos B/imunologia , Citidina Desaminase/imunologia , Switching de Imunoglobulina , Imunoglobulina M/imunologia , Fosfatidilinositol 3-Quinases/fisiologia , Animais , Linfócitos B/citologia , Antígenos CD40/fisiologia , Diferenciação Celular/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Receptores de Antígenos de Linfócitos B/fisiologia , Transdução de Sinais , Hipermutação Somática de Imunoglobulina/imunologia , Receptores Toll-Like/fisiologia
14.
Cancer Res ; 64(22): 8199-207, 2004 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-15548685

RESUMO

The mechanisms for "gain-of-function" phenotypes produced by mutant p53s such as enhanced proliferation, resistance to transforming growth factor-beta-mediated growth suppression, and increased tumorigenesis are not known. One theory is that these phenotypes are caused by novel transcriptional regulatory events acquired by mutant p53s. Another explanation is that these effects are a result of an imbalance of functions caused by the retention of some of the wild-type transcriptional regulatory events in the context of a loss of other counterbalancing activities. An analysis of the ability of DNA-binding domain mutants A138P and R175H, and wild-type p53 to regulate the expression levels of 6.9 x 10(3) genes revealed that the mutants retained only <5% of the regulatory activities of the wild-type protein. A138P p53 exhibited mostly retained wild-type regulatory activities and few acquired novel events. However, R175H p53 possessed an approximately equal number of wild-type regulatory events and novel activities. This is the first report that, after examination of the regulation of a large unfocused set of genes, provides data indicating that remaining wild-type transcriptional regulatory functions existing in the absence of counterbalancing activities as well as acquired novel events both contribute to the gain-of-function phenotypes produced by mutant p53s. However, mutant p53s are likely to be distinct in terms of the extent to which each mechanism contributes to their gain-of-function phenotypes.


Assuntos
Expressão Gênica/fisiologia , Mutação , Proteína Supressora de Tumor p53/fisiologia , Sequência de Bases , Linhagem Celular Tumoral , Primers do DNA , Expressão Gênica/genética , Humanos , Proteína Supressora de Tumor p53/genética
15.
Exp Neurol ; 273: 151-60, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26277686

RESUMO

OBJECTIVE: Multiple sclerosis (MS) is a debilitating neurological disorder involving an autoimmune reaction to oligodendrocytes and degeneration of the axons they ensheath in the CNS. Because the damage to oligodendrocytes and axons involves local inflammation and associated oxidative stress, we tested the therapeutic efficacy of combined treatment with a potent anti-inflammatory thalidomide analog (lenalidomide) and novel synthetic anti-oxidant cerium oxide nanoparticles (nanoceria) in the experimental autoimmune encephalomyelitis (EAE) mouse model of MS. METHODS: C57BL/6 mice were randomly assigned to a control (no EAE) group, or one of the four myelin oligodendrocyte glycoprotein-induced EAE groups: vehicle, lenalidomide, nanoceria, or lenalidomide plus nanoceria. During a 23 day period, clinical EAE symptoms were evaluated daily, and MRI brain scans were performed at 11-13 days and 20-22 days. Histological and biochemical analyses of brain tissue samples were performed to quantify myelin loss and local inflammation. RESULTS: Lenalidomide treatment alone delayed symptom onset, while nanoceria treatment had no effect on symptom onset or severity, but did promote recovery; lenalidomide and nanoceria each significantly attenuated white matter pathology and associated inflammation. Combined treatment with lenalidomide and nanoceria resulted in a near elimination of EAE symptoms, and reduced white matter pathology and inflammatory cell responses to a much greater extent than either treatment alone. INTERPRETATION: By suppressing inflammation and oxidative stress, combined treatment with lenalidomide and nanoceria can reduce demyelination and associated neurological symptoms in EAE mice. Our preclinical data suggest a potential application of this combination therapy in MS.


Assuntos
Autoimunidade/efeitos dos fármacos , Sistema Nervoso Central/efeitos dos fármacos , Cério/uso terapêutico , Encefalomielite Autoimune Experimental/tratamento farmacológico , Fatores Imunológicos/uso terapêutico , Talidomida/análogos & derivados , Análise de Variância , Animais , Células Cultivadas , Sistema Nervoso Central/imunologia , Sistema Nervoso Central/patologia , Citocinas/genética , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Citometria de Fluxo , Regulação da Expressão Gênica/efeitos dos fármacos , Lenalidomida , Imageamento por Ressonância Magnética , Camundongos , Camundongos Endogâmicos C57BL , RNA Mensageiro , Talidomida/uso terapêutico , Fatores de Tempo
16.
Neurobiol Aging ; 35(5): 975-89, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24325797

RESUMO

Cellular damage by reactive oxygen species and altered neurogenesis are implicated in the etiology of AD and the pathogenic actions of amyloid ß-peptide (Aß); the underlying mechanisms and the early oxidative intracellular events triggered by Aß are not established. In the present study, we found that mouse embryonic cortical neural progenitor cells exhibit intermittent spontaneous mitochondrial superoxide (SO) flashes that require transient opening of mitochondrial permeability transition pores (mPTPs). The incidence of mitochondria SO flash activity in neural progenitor cells (NPCs) increased during the first 6-24 hours of exposure to aggregating amyloid ß-peptide (Aß1-42), indicating an increase in transient mPTP opening. Subsequently, the SO flash frequency progressively decreased and ceased between 48 and 72 hours of exposure to Aß1-42, during which time global cellular reactive oxygen species increased, mitochondrial membrane potential decreased, cytochrome C was released from mitochondria and the cells degenerated. Inhibition of mPTPs and selective reduction in mitochondrial SO flashes significantly ameliorated the negative effects of Aß1-42 on NPC proliferation and survival. Our findings suggest that mPTP-mediated bursts of mitochondrial SO production is a relatively early and pivotal event in the adverse effects of Aß1-42 on NPCs. If Aß inhibits NPC proliferation in the brains of AD patients by a similar mechanism, then interventions that inhibit mPTP-mediated superoxide flashes would be expected to protect NPCs against the adverse effects of Aß.


Assuntos
Peptídeos beta-Amiloides/efeitos adversos , Proliferação de Células/efeitos dos fármacos , Mitocôndrias/metabolismo , Neurônios/citologia , Poro Nuclear/metabolismo , Fragmentos de Peptídeos/efeitos adversos , Células-Tronco/citologia , Superóxidos/efeitos adversos , Superóxidos/metabolismo , Doença de Alzheimer/etiologia , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Animais , Células Cultivadas , Citocromos c/metabolismo , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Camundongos , Permeabilidade , Espécies Reativas de Oxigênio/efeitos adversos , Espécies Reativas de Oxigênio/metabolismo
17.
Avicenna J Med Biotechnol ; 4(3): 148-54, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23407303

RESUMO

BACKGROUND: In this study, we evaluated the protective effects of water Hyacinth Root Powder (HRP) on arsenic-mediated toxic effects in mice. METHODS: Swiss albino mice, used in this study, were divided into four different groups (for each group n=5). The control group was supplied with normal feed and water, Arsenic group (As-group) was supplied with normal feed plus arsenic (sodium arsenite)-containing water, and arsenic+hyacinth group (As+Hy group) was supplied with feed supplemented with HRP plus arsenic water. The remaining Hy-group was supplied with feed supplemented with HRP plus normal water. RESULTS: Oral administration of arsenic reduced the normal growth of the mice as evidenced by weight loss. Interestingly, tip of the tails of these mice developed wound that caused gradual reduction of the tail length. Supplementation of HRP in feed significantly prevented mice growth retardation and tail wounding in As+Hy group mice. However, the growth pattern in Hy-group mice was observed to be almost similar to that of the control group indicating that HRP itself has no toxic or negative effect in mice. Ingested arsenic also distorted the shape of the blood cells and elevated the serum enzymes such as lactate dehydrogenase (LDH), alkaline phosphatase (ALP) and serum glutamic pyruvic transaminase (SGPT). Importantly, elevation of these enzymes and distortion of blood cell shape were partially reduced in mice belong to As+Hy group, indicating HRP-mediated reduction of arsenic toxicity. CONCLUSION: Therefore, the preventive effect of hyacinth root on arsenic-poisoned mice suggested the future application of hyacinth to reduce arsenic toxicity in animal and human.

18.
Mol Cancer Res ; 9(1): 78-89, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21131601

RESUMO

There are multiple mechanisms by which cells evade TGF-ß-mediated growth inhibitory effects. In this report, we describe a novel mechanism by which cells become resistant to TGF-ß-mediated growth suppression. Although having all the components of the TGF-ß signaling pathway, different cell lines, RL, HaCaT, and BJAB, have different sensitivities toward TGF-ß-induced growth suppression. The TGF-ß resistance of RL, a B-cell lymphoma cell line, was due to ligand-induced downregulation of TGF-ß receptor II (TßRII) and only transient TGF-ß induced nuclear translocation of Smad2 and Smad3. With low-dose phorbol 12-myristate 13-acetate (PMA) or anti-IgM treatment, TGF-ß sensitivity was restored by stabilizing TßRII expression and sustaining TGF-ß signaling. The MEK inhibitor, U0126, blocked both PMA- and anti-IgM-induced upregulation of TßRII. In HaCaT and BJAB, two TGF-ß-sensitive cell lines, which had higher basal levels of phospho-MEK and TßRII compared with RL, U0126 induced downregulation of TßRII and blocked subsequent TGF-ß signaling. Similar results were also obtained with normal B cells, where MEK1 inhibitor downregulated TßRII and subsequent TGF-ß signaling. Constitutively active MEK1, but not constitutively active ERK2, induced upregulation of TßRII. Furthermore, TßRII physically interacted with the constitutively active MEK1, but not with wild-type MEK1, indicating involvement of active MEK1 in stabilizing TßRII. Collectively, our data suggest a novel mechanism for MEK1 in regulating the sensitivity to TGF-ß signaling by stabilizing TßRII.


Assuntos
Proliferação de Células , MAP Quinase Quinase 1/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta1/metabolismo , Western Blotting , Butadienos/farmacologia , Linhagem Celular , Linhagem Celular Tumoral , Resistência a Medicamentos , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Células HEK293 , Humanos , MAP Quinase Quinase 1/antagonistas & inibidores , MAP Quinase Quinase 1/genética , Nitrilas/farmacologia , Ligação Proteica/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/genética , Receptor do Fator de Crescimento Transformador beta Tipo II , Receptores de Fatores de Crescimento Transformadores beta/genética , Acetato de Tetradecanoilforbol/análogos & derivados , Acetato de Tetradecanoilforbol/farmacologia , Fator de Crescimento Transformador beta1/farmacologia
19.
PLoS One ; 6(9): e24667, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21931802

RESUMO

BACKGROUND: CXCL12 is a pleiotropic chemokine involved in multiple different processes such as immune regulation, inflammatory responses, and cancer development. CXCL12 is also a potent chemokine involved in chemoattraction of T cells to the site of infection or inflammation. Mammalian target of rapamycin (mTOR) is a serine-threonine kinase that modulates different cellular processes, such as metabolism, nutrient sensing, protein translation, and cell growth. The role of mTOR in CXCL12-mediated resting T cell migration has yet to be elucidated. METHODOLOGY/PRINCIPAL FINDINGS: Rapamycin, an inhibitor of mTOR, significantly inhibits CXCL12 mediated migration of both primary human resting T cells and human T cell leukemia cell line CEM. p70(S6K1), an effector molecule of mTOR signaling pathway, was knocked down by shRNA in CEM cells using a lentiviral gene transfer system. Using p70(S6K1) knock down cells, we demonstrate the role of mTOR signaling in T cell migration both in vitro and in vivo. CONCLUSIONS: Our data demonstrate a new role for mTOR in CXCL12-induced T cell migration, and enrich the current knowledge regarding the clinical use of rapamycin.


Assuntos
Quimiocina CXCL12/metabolismo , Linfócitos T/citologia , Linfócitos T/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Animais , Linhagem Celular Tumoral , Movimento Celular/genética , Movimento Celular/fisiologia , Células Cultivadas , Quimiocina CXCL12/genética , Humanos , Camundongos , Microscopia Confocal , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/antagonistas & inibidores , Serina-Treonina Quinases TOR/genética
20.
PLoS One ; 6(5): e18553, 2011 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-21572994

RESUMO

The role of T cells in innate immunity is not well defined. In this report, we show that a subset of human peripheral blood CD4(+) T cells responds to IL-12 plus IL-18, but not to IL-12 or IL-18 alone, by producing IFN-γ in the absence of any antigenic stimulation or cell proliferation. Intracellular staining reveals a small percentage of resting CD4(+) T cells (0.5 to 1.5%) capable of producing IFN-γ in response to IL-12 plus IL-18. Interestingly, both naïve (CD45RA(+)) and memory (CD45RO(+)) CD4(+) populations were responsive to IL-12 plus IL-18 stimulation in producing IFN-γ. The expression of IFN-γinduced by IL-12 and IL-18 is sensitive to rapamycin and SB203580, indicating the possible involvement of mTOR and p38 MAP kinase, respectively, in this synergistic pathway. While p38MAP kinase is involved in transcription, mTOR is involved in message stabilization. We have also shown that NFκB family member, cRel, but not GADD45ß and GADD45γ, plays an important role in IL-12 plus IL-18-induced IFN-γ transcription. Thus, the present study suggests that naïve CD4(+) T cells may participate in innate immunity or amplify adaptive immune responses through cytokine-induced antigen-independent cytokine production.


Assuntos
Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/metabolismo , Interferon gama/metabolismo , Interleucina-12/farmacologia , Interleucina-18/farmacologia , Animais , Antígenos de Diferenciação/metabolismo , Células Cultivadas , Humanos , Imidazóis/farmacologia , Interferon gama/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Masculino , Camundongos , Fosforilação/efeitos dos fármacos , Reação em Cadeia da Polimerase , Piridinas/farmacologia , Fator de Transcrição STAT4/metabolismo , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/antagonistas & inibidores , Serina-Treonina Quinases TOR/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA