Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Res Sq ; 2024 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-38766212

RESUMO

Understanding the molecular mechanisms underlying tumorigenesis is crucial for developing effective cancer therapies. Here, we investigate the co-amplification of MED30 and MYC across diverse cancer types and its impact on oncogenic transcriptional programs. Transcriptional profiling of MYC and MED30 single or both overexpression/amplification revealed the over amount of MED30 lead MYC to a new transcriptional program that associate with poor prognosis. Mechanistically, MED30 overexpression/amplification recruits other Mediator components and binding of MYC to a small subset of novel genomic regulatory sites, changing the epigenetic marks and inducing the formation of new enhancers, which drive the expression of target genes crucial for cancer progression. In vivo studies in pancreatic ductal adenocarcinoma (PDAC) further validate the oncogenic potential of MED30, as its overexpression promotes tumor growth and can be attenuated by knockdown of MYC. Using another cancer type as an example, MED30 knockdown reduces tumor growth particularly in MYC high-expressed glioblastoma (GBM) cell lines. Overall, our study elucidates the critical role of MED30 overexpression in orchestrating oncogenic transcriptional programs and highlights its potential as a therapeutic target for MYC-amplified cancer.

2.
Nat Cancer ; 2024 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-38519786

RESUMO

Cancers commonly reprogram translation and metabolism, but little is known about how these two features coordinate in cancer stem cells. Here we show that glioblastoma stem cells (GSCs) display elevated protein translation. To dissect underlying mechanisms, we performed a CRISPR screen and identified YRDC as the top essential transfer RNA (tRNA) modification enzyme in GSCs. YRDC catalyzes the formation of N6-threonylcarbamoyladenosine (t6A) on ANN-decoding tRNA species (A denotes adenosine, and N denotes any nucleotide). Targeting YRDC reduced t6A formation, suppressed global translation and inhibited tumor growth both in vitro and in vivo. Threonine is an essential substrate of YRDC. Threonine accumulated in GSCs, which facilitated t6A formation through YRDC and shifted the proteome to support mitosis-related genes with ANN codon bias. Dietary threonine restriction (TR) reduced tumor t6A formation, slowed xenograft growth and augmented anti-tumor efficacy of chemotherapy and anti-mitotic therapy, providing a molecular basis for a dietary intervention in cancer treatment.

3.
Cell Rep ; 42(7): 112690, 2023 07 25.
Artigo em Inglês | MEDLINE | ID: mdl-37384528

RESUMO

AKT kinase is a key regulator in cell metabolism and survival, and its activation is strictly modulated. Herein, we identify XAF1 (XIAP-associated factor) as a direct interacting protein of AKT1, which strongly binds the N-terminal region of AKT1 to block its K63-linked poly-ubiquitination and subsequent activation. Consistently, Xaf1 knockout causes AKT activation in mouse muscle and fat tissues and reduces body weight gain and insulin resistance induced by high-fat diet. Pathologically, XAF1 expression is low and anti-correlated with the phosphorylated p-T308-AKT signal in prostate cancer samples, and Xaf1 knockout stimulates the p-T308-AKT signal to accelerate spontaneous prostate tumorigenesis in mice with Pten heterozygous loss. And ectopic expression of wild-type XAF1, but not the cancer-derived P277L mutant, inhibits orthotopic tumorigenesis. We further identify Forkhead box O 1 (FOXO1) as a transcriptional regulator of XAF1, thus forming a negative feedback loop between AKT1 and XAF1. These results reveal an important intrinsic regulatory mechanism of AKT signaling.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular , Neoplasias , Animais , Masculino , Camundongos , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Apoptose/fisiologia , Proteínas Reguladoras de Apoptose/metabolismo , Carcinogênese , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo
4.
Nature ; 617(7962): 818-826, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-37198486

RESUMO

Cancer cells rewire metabolism to favour the generation of specialized metabolites that support tumour growth and reshape the tumour microenvironment1,2. Lysine functions as a biosynthetic molecule, energy source and antioxidant3-5, but little is known about its pathological role in cancer. Here we show that glioblastoma stem cells (GSCs) reprogram lysine catabolism through the upregulation of lysine transporter SLC7A2 and crotonyl-coenzyme A (crotonyl-CoA)-producing enzyme glutaryl-CoA dehydrogenase (GCDH) with downregulation of the crotonyl-CoA hydratase enoyl-CoA hydratase short chain 1 (ECHS1), leading to accumulation of intracellular crotonyl-CoA and histone H4 lysine crotonylation. A reduction in histone lysine crotonylation by either genetic manipulation or lysine restriction impaired tumour growth. In the nucleus, GCDH interacts with the crotonyltransferase CBP to promote histone lysine crotonylation. Loss of histone lysine crotonylation promotes immunogenic cytosolic double-stranded RNA (dsRNA) and dsDNA generation through enhanced H3K27ac, which stimulates the RNA sensor MDA5 and DNA sensor cyclic GMP-AMP synthase (cGAS) to boost type I interferon signalling, leading to compromised GSC tumorigenic potential and elevated CD8+ T cell infiltration. A lysine-restricted diet synergized with MYC inhibition or anti-PD-1 therapy to slow tumour growth. Collectively, GSCs co-opt lysine uptake and degradation to shunt the production of crotonyl-CoA, remodelling the chromatin landscape to evade interferon-induced intrinsic effects on GSC maintenance and extrinsic effects on immune response.


Assuntos
Histonas , Lisina , Neoplasias , Processamento de Proteína Pós-Traducional , Cromatina/química , Cromatina/genética , Cromatina/metabolismo , Glutaril-CoA Desidrogenase/metabolismo , Histonas/química , Histonas/metabolismo , Lisina/deficiência , Lisina/metabolismo , Neoplasias/tratamento farmacológico , Neoplasias/imunologia , Neoplasias/metabolismo , Neoplasias/patologia , RNA de Cadeia Dupla/imunologia , Humanos , Animais , Camundongos , Interferon Tipo I/imunologia
5.
Cancer Discov ; 13(4): 974-1001, 2023 04 03.
Artigo em Inglês | MEDLINE | ID: mdl-36649564

RESUMO

Glioblastoma (GBM) constitutes the most lethal primary brain tumor for which immunotherapy has provided limited benefit. The unique brain immune landscape is reflected in a complex tumor immune microenvironment (TIME) in GBM. Here, single-cell sequencing of the GBM TIME revealed that microglia were under severe oxidative stress, which induced nuclear receptor subfamily 4 group A member 2 (NR4A2)-dependent transcriptional activity in microglia. Heterozygous Nr4a2 (Nr4a2+/-) or CX3CR1+ myeloid cell-specific Nr4a2 (Nr4a2fl/flCx3cr1Cre) genetic targeting reshaped microglia plasticity in vivo by reducing alternatively activated microglia and enhancing antigen presentation capacity for CD8+ T cells in GBM. In microglia, NR4A2 activated squalene monooxygenase (SQLE) to dysregulate cholesterol homeostasis. Pharmacologic NR4A2 inhibition attenuated the protumorigenic TIME, and targeting the NR4A2 or SQLE enhanced the therapeutic efficacy of immune-checkpoint blockade in vivo. Collectively, oxidative stress promotes tumor growth through NR4A2-SQLE activity in microglia, informing novel immune therapy paradigms in brain cancer. SIGNIFICANCE: Metabolic reprogramming of microglia in GBM informs synergistic vulnerabilities for immune-checkpoint blockade therapy in this immunologically cold brain tumor. This article is highlighted in the In This Issue feature, p. 799.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Humanos , Glioblastoma/tratamento farmacológico , Glioblastoma/genética , Microglia , Inibidores de Checkpoint Imunológico/uso terapêutico , Macrófagos , Encéfalo/patologia , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/genética , Microambiente Tumoral/fisiologia
6.
Mol Cell ; 81(13): 2736-2751.e8, 2021 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-33932349

RESUMO

Cholesterol metabolism is tightly associated with colorectal cancer (CRC). Nevertheless, the clinical benefit of statins, the inhibitor of cholesterol biogenesis mevalonate (MVA) pathway, is inconclusive, possibly because of a lack of patient stratification criteria. Here, we describe that YAP-mediated zinc finger MYND-type containing 8 (ZMYND8) expression sensitizes intestinal tumors to the inhibition of the MVA pathway. We show that the oncogenic activity of YAP relies largely on ZMYND8 to enhance intracellular de novo cholesterol biogenesis. Disruption of the ZMYND8-dependent MVA pathway greatly restricts the self-renewal capacity of Lgr5+ intestinal stem cells (ISCs) and intestinal tumorigenesis. Mechanistically, ZMYND8 and SREBP2 drive the enhancer-promoter interaction to facilitate the recruitment of Mediator complex, thus upregulating MVA pathway genes. Together, our results establish that the epigenetic reader ZMYND8 endows YAP-high intestinal cancer with metabolic vulnerability.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Neoplasias Colorretais/metabolismo , Ácido Mevalônico/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Linhagem Celular Tumoral , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Camundongos , Camundongos Transgênicos , Proteínas Supressoras de Tumor/genética , Proteínas de Sinalização YAP
7.
Cancer Cell ; 38(3): 350-365.e7, 2020 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-32619406

RESUMO

The level of SETD2-mediated H3K36me3 is inversely correlated with that of EZH2-catalyzed H3K27me3. Nevertheless, it remains unclear whether these two enzymatic activities are molecularly intertwined. Here, we report that SETD2 delays prostate cancer (PCa) metastasis via its substrate EZH2. We show that SETD2 methylates EZH2 which promotes EZH2 degradation. SETD2 deficiency induces a Polycomb-repressive chromatin state that enables cells to acquire metastatic traits. Conversely, mice harboring nonmethylated EZH2 mutant or SETD2 mutant defective in binding to EZH2 develop metastatic PCa. Furthermore, we identify that metformin-stimulated AMPK signaling converges at FOXO3 to stimulate SETD2 expression. Together, our results demonstrate that the SETD2-EZH2 axis integrates metabolic and epigenetic signaling to restrict PCa metastasis.


Assuntos
Proteínas Quinases Ativadas por AMP/genética , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Regulação Neoplásica da Expressão Gênica , Histona-Lisina N-Metiltransferase/genética , Neoplasias da Próstata/genética , Transdução de Sinais/genética , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Linhagem Celular Tumoral , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Histona-Lisina N-Metiltransferase/metabolismo , Humanos , Estimativa de Kaplan-Meier , Masculino , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos Nus , Camundongos SCID , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Transplante Heterólogo
9.
J Clin Invest ; 129(2): 759-773, 2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30496141

RESUMO

Loss of phosphatase and tensin homolog (PTEN) represents one hallmark of prostate cancer (PCa). However, restoration of PTEN or inhibition of the activated PI3K/AKT pathway has shown limited success, prompting us to identify obligate targets for disease intervention. We hypothesized that PTEN loss might expose cells to unique epigenetic vulnerabilities. Here, we identified a synthetic lethal relationship between PTEN and Brahma-related gene 1 (BRG1), an ATPase subunit of the SWI/SNF chromatin remodeling complex. Higher BRG1 expression in tumors with low PTEN expression was associated with a worse clinical outcome. Genetically engineered mice (GEMs) and organoid assays confirmed that ablation of PTEN sensitized the cells to BRG1 depletion. Mechanistically, PTEN loss stabilized BRG1 protein through the inhibition of the AKT/GSK3ß/FBXW7 axis. Increased BRG1 expression in PTEN-deficient PCa cells led to chromatin remodeling into configurations that drove a protumorigenic transcriptome, causing cells to become further addicted to BRG1. Furthermore, we showed in preclinical models that BRG1 antagonist selectively inhibited the progression of PTEN-deficient prostate tumors. Together, our results highlight the synthetic lethal relationship between PTEN and BRG1 and support targeting BRG1 as an effective approach to the treatment of PTEN-deficient PCa.


Assuntos
DNA Helicases , Neoplasias Experimentais , Proteínas Nucleares , PTEN Fosfo-Hidrolase , Neoplasias da Próstata , Transdução de Sinais/genética , Fatores de Transcrição , Animais , Proteínas Cromossômicas não Histona/genética , Proteínas Cromossômicas não Histona/metabolismo , DNA Helicases/genética , DNA Helicases/metabolismo , Humanos , Masculino , Camundongos , Camundongos Nus , Camundongos Transgênicos , Neoplasias Experimentais/genética , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/mortalidade , Neoplasias Experimentais/patologia , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Células PC-3 , PTEN Fosfo-Hidrolase/genética , PTEN Fosfo-Hidrolase/metabolismo , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/mortalidade , Neoplasias da Próstata/patologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
10.
Nat Cell Biol ; 20(3): 320-331, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29403037

RESUMO

Growth signals, such as extracellular nutrients and growth factors, have substantial effects on genome integrity; however, the direct underlying link remains unclear. Here, we show that the mechanistic target of rapamycin (mTOR)-ribosomal S6 kinase (S6K) pathway, a central regulator of growth signalling, phosphorylates RNF168 at Ser60 to inhibit its E3 ligase activity, accelerate its proteolysis and impair its function in the DNA damage response, leading to accumulated unrepaired DNA and genome instability. Moreover, loss of the tumour suppressor liver kinase B1 (LKB1; also known as STK11) hyperactivates mTOR complex 1 (mTORC1)-S6K signalling and decreases RNF168 expression, resulting in defects in the DNA damage response. Expression of a phospho-deficient RNF168-S60A mutant rescues the DNA damage repair defects and suppresses tumorigenesis caused by Lkb1 loss. These results reveal an important function of mTORC1-S6K signalling in the DNA damage response and suggest a general mechanism that connects cell growth signalling to genome stability control.


Assuntos
Proliferação de Células , Reparo do DNA , Neoplasias/enzimologia , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Proteínas Quinases S6 Ribossômicas 90-kDa/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Células A549 , Quinases Proteína-Quinases Ativadas por AMP , Proteínas Quinases Ativadas por AMP , Animais , Quebras de DNA de Cadeia Dupla , Feminino , Células HCT116 , Células HEK293 , Humanos , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina/genética , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Camundongos Transgênicos , Neoplasias/genética , Neoplasias/patologia , Fosforilação , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Quinases S6 Ribossômicas 70-kDa/genética , Proteínas Quinases S6 Ribossômicas 90-kDa/genética , Transdução de Sinais , Serina-Treonina Quinases TOR/genética , Carga Tumoral , Ubiquitina-Proteína Ligases/genética
11.
J Clin Invest ; 127(9): 3375-3391, 2017 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-28825595

RESUMO

The histone H3K36 methyltransferase SETD2 is frequently mutated or deleted in a variety of human tumors. Nevertheless, the role of SETD2 loss in oncogenesis remains largely undefined. Here, we found that SETD2 counteracts Wnt signaling and its inactivation promotes intestinal tumorigenesis in mouse models of colorectal cancer (CRC). SETD2 was not required for intestinal homeostasis under steady state; however, upon irradiation, genetic inactivation of Setd2 in mouse intestinal epithelium facilitated the self-renewal of intestinal stem/progenitor cells as well as tissue regeneration. Furthermore, depletion of SETD2 enhanced the susceptibility to tumorigenesis in the context of dysregulated Wnt signaling. Mechanistic characterizations indicated that SETD2 downregulation affects the alternative splicing of a subset of genes implicated in tumorigenesis. Importantly, we uncovered that SETD2 ablation reduces intron retention of dishevelled segment polarity protein 2 (DVL2) pre-mRNA, which would otherwise be degraded by nonsense-mediated decay, thereby augmenting Wnt signaling. The signaling cascades mediated by SETD2 were further substantiated by a CRC patient cohort analysis. Together, our studies highlight SETD2 as an integral regulator of Wnt signaling through epigenetic regulation of RNA processing during tissue regeneration and tumorigenesis.


Assuntos
Processamento Alternativo , Carcinogênese/genética , Neoplasias Colorretais/genética , Histona-Lisina N-Metiltransferase/genética , Animais , Diferenciação Celular , Transformação Celular Neoplásica , Modelos Animais de Doenças , Proteínas Desgrenhadas/genética , Epigênese Genética , Regulação Neoplásica da Expressão Gênica , Humanos , Íntrons , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Regeneração , Transdução de Sinais , Células-Tronco/citologia , Proteínas Wnt/metabolismo
12.
Proc Natl Acad Sci U S A ; 114(19): E3796-E3805, 2017 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-28439030

RESUMO

Epithelial barrier disruption is a major cause of inflammatory bowel disease (IBD); however, the mechanism through which epigenetic regulation modulates intestinal epithelial integrity remains largely undefined. Here we show that EZH2, the catalytic subunit of polycomb repressive complex (PRC2), is indispensable for maintaining epithelial cell barrier integrity and homeostasis under inflammatory conditions. In accordance with reduced EZH2 expression in patients, the inactivation of EZH2 in IECs sensitizes mice to DSS- and TNBS-induced experimental colitis. Conversely, EZH2 overexpression in the intestinal epithelium renders mice more resistant to colitis. Mechanistically, the genes encoding TRAF2/5 are held in a finely tuned bivalent status under inflammatory conditions. EZH2 deficiency potentiates the expression of these genes to enhance TNFα-induced NF-κB signaling, thereby leading to uncontrolled inflammation. More importantly, we show that EZH2 depletion compromises the protective role of NF-κB signaling in cell survival by directly up-regulating ITCH, a well-known E3 ligase that degrades the c-FLIP protein. Thus, our findings highlight an epigenetic mechanism by which EZH2 integrates the multifaceted effects of TNFα signaling to promote the inflammatory response and apoptosis in colitis.


Assuntos
Apoptose , Colite/metabolismo , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Epigênese Genética , Mucosa Intestinal/metabolismo , Transdução de Sinais , Fator de Necrose Tumoral alfa/metabolismo , Animais , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/genética , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/metabolismo , Colite/induzido quimicamente , Colite/genética , Colite/patologia , Sulfato de Dextrana/toxicidade , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Humanos , Inflamação/induzido quimicamente , Inflamação/genética , Inflamação/metabolismo , Inflamação/patologia , Mucosa Intestinal/patologia , Camundongos , Camundongos Knockout , NF-kappa B/genética , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Fator 2 Associado a Receptor de TNF/genética , Fator 2 Associado a Receptor de TNF/metabolismo , Fator 5 Associado a Receptor de TNF/genética , Fator 5 Associado a Receptor de TNF/metabolismo , Fator de Necrose Tumoral alfa/genética , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo
13.
J Clin Invest ; 127(4): 1284-1302, 2017 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-28319045

RESUMO

Loss of phosphatase and tensin homolog (PTEN) and activation of the PI3K/AKT signaling pathway are hallmarks of prostate cancer (PCa). However, these alterations alone are insufficient for cells to acquire metastatic traits. Here, we have shown that the histone dimethyl transferase WHSC1 critically drives indolent PTEN-null tumors to become metastatic PCa. In a PTEN-null murine PCa model, WHSC1 overexpression in prostate epithelium cooperated with Pten deletion to produce a metastasis-prone tumor. Conversely, genetic ablation of Whsc1 prevented tumor progression in PTEN-null mice. Molecular characterization revealed that increased AKT activity due to PTEN loss directly phosphorylates WHSC1 at S172, preventing WHSC1 degradation by CRL4Cdt2 E3 ligase. Increased WHSC1 expression transcriptionally upregulates expression of RICTOR, a pivotal component of mTOR complex 2 (mTORC2), to further enhance AKT activity. Therefore, the AKT/WHSC1/mTORC2 signaling cascade represents a vicious feedback loop that elicits unrestrained AKT signaling. Furthermore, we determined that WHSC1 positively regulates Rac1 transcription to increase tumor cell motility. The biological importance of a WHSC1-mediated signaling cascade is substantiated by patient sample analysis in which WHSC1 signaling is tightly correlated with disease progression and recurrence. Taken together, our findings highlight a pivotal link between an epigenetic regulator, WHSC1, and key intracellular signaling molecules, AKT, RICTOR, and Rac1, to drive PCa metastasis.


Assuntos
Movimento Celular , Histona-Lisina N-Metiltransferase/metabolismo , Neoplasias da Próstata/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Animais , Proteínas de Transporte/biossíntese , Proteínas de Transporte/genética , Estabilidade Enzimática/genética , Epigênese Genética , Regulação Neoplásica da Expressão Gênica , Histona-Lisina N-Metiltransferase/genética , Masculino , Alvo Mecanístico do Complexo 2 de Rapamicina , Camundongos , Camundongos Transgênicos , Complexos Multiproteicos/genética , Complexos Multiproteicos/metabolismo , Metástase Neoplásica , Neuropeptídeos/genética , Neuropeptídeos/metabolismo , PTEN Fosfo-Hidrolase/genética , PTEN Fosfo-Hidrolase/metabolismo , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação/genética , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Proteínas Proto-Oncogênicas c-akt/genética , Proteína Companheira de mTOR Insensível à Rapamicina , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo , Transcrição Gênica , Proteínas rac1 de Ligação ao GTP/genética , Proteínas rac1 de Ligação ao GTP/metabolismo
14.
Acta Histochem ; 119(1): 26-31, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27884396

RESUMO

This study was undertaken to determine if visfatin is involved in the inflammation or apoptosis introduced by LPS in rats. Forty 8-week old Wistar rats were divided into four groups (n=10 in each group) and injected with saline, visfatin, LPS and visfatin+LPS co-stimulated via caudal vein. The duodenum, jejunum and ileum were harvested from all the rats. Compared to the saline treated group, visfatin significantly increased the number of TUNEL-positive apoptotic cells and the expression of caspase-3 protein in intestinal mucosa. Similarly, ELISA and western blot analysis also showed the up-regulation of pro-caspase-3 and cleaved caspase-3 expression in the visfatin group compared to the control group. In contrast to LPS, visfatin down-regulated the expression of cleaved-caspase-3 in the visfatin+LPS co-stimulated group, resulting in a significant decrease in apoptosis in intestinal mucosal cells. We observed more pro-caspase-3 positive cells in the visfatin+LPS co-stimulated group. The results indicate that, in the presence of LPS, visfatin plays an important role in the regulation of cell apoptosis and inflammation.


Assuntos
Apoptose/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Imunidade nas Mucosas/efeitos dos fármacos , Mucosa Intestinal/efeitos dos fármacos , Lipopolissacarídeos/farmacologia , Nicotinamida Fosforribosiltransferase/farmacologia , Animais , Caspase 3/genética , Caspase 3/metabolismo , Combinação de Medicamentos , Duodeno/citologia , Duodeno/efeitos dos fármacos , Duodeno/imunologia , Células Epiteliais/citologia , Células Epiteliais/imunologia , Expressão Gênica , Íleo/citologia , Íleo/efeitos dos fármacos , Íleo/imunologia , Marcação In Situ das Extremidades Cortadas , Mucosa Intestinal/citologia , Mucosa Intestinal/imunologia , Jejuno/citologia , Jejuno/efeitos dos fármacos , Jejuno/imunologia , Ratos , Ratos Wistar
15.
Histol Histopathol ; 31(9): 987-1000, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26857566

RESUMO

Visfatin is an adipocytokine displaying multiple functional properties, which plays a role in the regulation of cell apoptosis and inflammation by an as yet unidentified mechanism. The aim of the present study was to determine if visfatin is involved in apoptosis pathway induced by LPS in rat Mesenteric lymph nodes (MLNs). Experimental rats were divided into four groups and MLNs samples were collected from each group. The morphological changes of the MLNs were examined by histological imaging. CD68 and ENPP1 were detected with immunohistochemistry and Western Blot. Apoptosis was evaluated with TUNEL and Flow Cytometry, the mRNA levels of the apoptosis-related genes were detected by qRT-PCR, and the protein levels of the apoptotic-related factors were detected by western blot. The main results showed that visfatin could significantly increase the macrophages in MLNs and prevent cell apoptosis from LPS-induced mesenteric lymph nodes, activate apoptotic signaling pathways and regulate the mRNA levels of the apoptosis-related genes. Visfatin had a pro-apoptotic effect on normal MLNs, whereas it exerted an anti-apoptotic effect during LPS-induced cell apoptosis in rat MLNs. In short, visfatin plays a dual role in the apoptosis in rat MLNs, which is mediated by both the mitochondrial apoptotic pathway and the death-receptor apoptotic pathway.


Assuntos
Apoptose/fisiologia , Linfonodos/patologia , Nicotinamida Fosforribosiltransferase/metabolismo , Animais , Apoptose/efeitos dos fármacos , Western Blotting , Citometria de Fluxo , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Lipopolissacarídeos/toxicidade , Linfonodos/efeitos dos fármacos , Nicotinamida Fosforribosiltransferase/farmacologia , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase em Tempo Real
16.
Int. j. morphol ; 33(4): 1502-1509, Dec. 2015. ilus
Artigo em Inglês | LILACS | ID: lil-772345

RESUMO

This study investigated the effects of visfatin on the structure and the immunity levels in the small intestine of LPS-induced rats. Forty Wistar male and female SPF rats were randomly and equally divided into four groups: the saline (control), vistfatin, lipopolysaccharide (LPS), and visfatin+LPS co-stimulated. The functions of visfatin in the intestinal mucosal immunity were investigated by examining the variation of tissue structure, inflammation and immunity-related proteins in the intestine of immunologically stressed rats using HE staining, ELISA, immunohistochemistry and Western Blot. The results showed that, when compared with the control group, the visfatin-treated group showed a decrease in the intestinal villus height and width, and a significant increase in the levels of IL-6 and TNF-ð as well as Immunoglobulin A (IgA) positive cells. Additionally, when compared with the LPS-treated group, the visfatin+LPS co-stimulated group showed a decrease in the villus height and width as well as the levels of IL-6 and TNF-ð, and an increase in IgA levels, implying a shrinking response to LPS injection. All the results suggest that, under normal physiological conditions, visfatin disturbs the body's homeostasis and causes intestinal villus atrophy by increasing IgA expression. While under immune response conditions, LPS acts as an exogenous antigen to promote visfatin against LPS-induced inflammation by decreasing the expression of IgA. Under immune stress conditions, visfatin as an exogenous stimulus promotes the immune response by regulating the protein levels of IL-6, TNF-ð and IgA.


Este estudio investigó los efectos de la visfatina sobre la estructura y los niveles de inmunidad en el intestino delgado de ratas inducidas por lipopolisacáridos (LPS). Cuarenta ratas Wistar se dividieron aleatoriamente e igualmente en cuatro grupos: solución salina (control), vistafin, LPS y visfatina + LPS co-estimuladas. Las funciones de la visfatina en la inmunidad de la mucosa intestinal se investigaron mediante el examen de variación de la estructura del tejido, la inflamación y las proteínas relacionadas con la inmunidad en el intestino de ratas estresadas inmunológicamente; usando tinción HE, ELISA, inmunohistoquímica y Western Blot. Los resultados mostraron que, en comparación con el grupo control, el grupo tratado con visfatina presentó una disminución en la altura y ancho de las vellosidades intestinales, y un aumento significativo en los niveles de IL-6 y TNF-ð, así como inmunoglobulina A (IgA células positivas). Además, al comparar este grupo con el grupo tratado con LPS- el grupo visfatina + LPS co-estimulado mostró una disminución en la altura y ancho de las vellosidades, así como en los niveles de IL-6 y TNF-ð, y un aumento en los niveles de IgA, lo que implica reducción de una respuesta a la inyección LPS. Todos los resultados sugieren que, en condiciones fisiológicas normales, la visfatina perturba la homeostasis del cuerpo y provoca la atrofia de las vellosidades intestinales mediante el aumento de la expresión de IgA. Mientras que bajo condiciones de la respuesta inmune, LPS actúa como un antígeno exógeno para promover visfatina contra la inflamación inducida por LPS por la disminución de la expresión de IgA. En condiciones de estrés inmunológico, la visfatina como estímulo exógeno promueve la respuesta inmune mediante la regulación de los niveles de proteína de IL-6, TNF-ð e IgA.


Assuntos
Animais , Masculino , Feminino , Ratos , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/imunologia , Nicotinamida Fosforribosiltransferase/administração & dosagem , Western Blotting , Ensaio de Imunoadsorção Enzimática , Imunidade nas Mucosas/efeitos dos fármacos , Imunoglobulina A/análise , Imuno-Histoquímica , Ratos Wistar
17.
Cell Tissue Res ; 359(2): 605-618, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25358398

RESUMO

The purpose of the present study is to determine if visfatin is involved in inflammation or apoptosis induced by LPS in rat. Forty Wistar rats were divided into four groups: saline group, LPS group, visfatin group and Visfatin + LPS co-stimulated group. Spleen samples from each group of rats were collected for study. The spleen structure was examined by histological imaging. Apoptosis was evaluated with TUNEL reaction. Caspase-3 was detected with immunohistochemistry and western blot. The apoptosis-related genes were detected by qPCR and inflammatory cytokines were tested by ELISA. Our main findings were as follows. (1) Macrophages were markedly increased in the visfatin group compared with the saline group. This finding was confirmed when spleen samples were examined with western blot using CD68 antibody. (2) Visfatin promoted the expression of CD68 and caspase-3 in rat spleen, whereas visfatin could inhibit the expression of CD68 and activated caspase-3 in spleen of LPS-induced acute inflammation. (3) Visfatin had a pro-apoptotic effect on normal rat spleen, whereas it exerted an anti-apoptotic effect during LPS-induced lymphocytes apoptosis in rat spleen. Moreover, the effect of visfatin on cell apoptosis was mediated by the mitochondrial pathway. (4) Visfatin could modulate both the anti-inflammatory cytokines and pro-inflammatory cytokines in rat spleen, such as IL-10, IL-4, IL-6, TNF-α and IL-1ß. Taken together, we demonstrate that visfatin could participate in the inflammatory process in rat spleen by modulating the macrophages and inflammatory cytokines. Also, visfatin plays a dual role in the apoptosis in rat spleen, which is mediated by the mitochondrial pathway.


Assuntos
Apoptose , Inflamação/patologia , Lipopolissacarídeos/farmacologia , Nicotinamida Fosforribosiltransferase/metabolismo , Baço/enzimologia , Baço/patologia , Animais , Antígenos CD/metabolismo , Antígenos de Diferenciação Mielomonocítica/metabolismo , Caspase 3/genética , Caspase 3/metabolismo , Citocinas/metabolismo , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Imuno-Histoquímica , Mediadores da Inflamação/metabolismo , Linfócitos/efeitos dos fármacos , Linfócitos/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Ratos Wistar , Transdução de Sinais/efeitos dos fármacos , Baço/efeitos dos fármacos , Proteína X Associada a bcl-2/genética , Proteína X Associada a bcl-2/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA