Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 164
Filtrar
1.
Mol Med Rep ; 25(1)2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34779498

RESUMO

Endocrine drug resistance is common in some patients with estrogen receptor (ER)­positive breast cancer, so it is necessary to identify potential therapeutic targets. The aim of the present study was to investigate the regulatory effect and mechanism of epsin 3 (EPN3) expression level changes on the proliferation and apoptosis of ER­positive breast cancer. Online GEPIA was used to analyze the expression level of EPN3 in breast cancer. The online Kaplan­Meier plotter tool was used to analyze the relationship between EPN3 expression and the prognosis of patients with breast cancer. Reverse transcription­quantitative PCR, immunohistochemistry and western blotting were performed to detect the mRNA and protein expression levels of EPN3 in breast cancer tissues and cells. A lentiviral infection system was used to knockdown the expression of EPN3 in breast cancer cell lines. Cell Counting Kit­8 and flow cytometry assays were conducted to detect the effect of EPN3 knockdown on breast cancer cell proliferation and apoptosis. Western blotting was used to detect the regulation of EPN3 expression on NF­κB, and immunofluorescence was performed to detect the effect of EPN3 expression on NF­κB nuclear translocation. The results demonstrated that the expression level of EPN3 in breast cancer tissues was higher compared with that in adjacent tissues (P<0.05). The expression level of EPN3 in the ER­positive breast cancer cell line, MCF7, was higher compared with that in the other cell lines (MCF10A, ZR75­1, MDA­MB­231, BT549 and SK­BR­3). After knocking down the expression of EPN3 in MCF7 cells, the proliferative ability of the cells was decreased, and the apoptosis rate was increased (P<0.05). After EPN3 knockdown in MCF7 cells, the phosphorylation of NF­κB was decreased (P<0.05), and the nuclear translocation signal was weakened. Thus, it was suggested that EPN3 promoted cell proliferation and inhibited cell apoptosis by regulating the NF­κB signaling pathway in ER­positive breast cancer.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Apoptose/fisiologia , Neoplasias da Mama/metabolismo , Proteínas Adaptadoras de Transporte Vesicular/fisiologia , Adulto , Linhagem Celular Tumoral , Proliferação de Células/genética , China , Feminino , Expressão Gênica/genética , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Células MCF-7 , Pessoa de Meia-Idade , NF-kappa B/metabolismo , Prognóstico , Transdução de Sinais
2.
J Neurosci ; 41(17): 3932-3943, 2021 04 28.
Artigo em Inglês | MEDLINE | ID: mdl-33741721

RESUMO

The Abelson-helper integration site 1 (AHI1) gene encodes for a ciliary transition zone localizing protein that when mutated causes the human ciliopathy, Joubert syndrome. We prepared and examined neuronal cultures derived from male and female embryonic Ahi1+/+ and Ahi1-/- mice (littermates) and found that the distribution of ciliary melanin-concentrating hormone receptor-1 (MchR1) was significantly reduced in Ahi1-/- neurons; however, the total and surface expression of MchR1 on Ahi1-/- neurons was similar to controls (Ahi1+/+). This indicates that a pathway for MchR1 trafficking to the surface plasma membrane is intact, but the process of targeting MchR1 into cilia is impaired in Ahi1-deficient mouse neurons, indicating a role for Ahi1 in localizing MchR1 to the cilium. Mouse Ahi1-/- neurons that fail to accumulate MchR1 in the ciliary membrane have significant decreases in two downstream MchR1 signaling pathways [cAMP and extracellular signal-regulated kinase (Erk)] on MCH stimulation. These results suggest that the ciliary localization of MchR1 is necessary and critical for MchR1 signaling, with Ahi1 participating in regulating MchR1 localization to cilia, and further supporting cilia as critical signaling centers in neurons.SIGNIFICANCE STATEMENT Our work here demonstrates that neuronal primary cilia are powerful and focused signaling centers for the G-protein-coupled receptor (GPCR), melanin-concentrating hormone receptor-1 (MCHR1), with a role for the ciliary transition zone protein, Abelson-helper integration site 1 (AHI1), in mediating ciliary trafficking of MCHR1. Moreover, our manuscript further expands the repertoire of cilia functions on neurons, a cell type that has not received significant attention in the cilia field. Lastly, our work demonstrates the significant influence of ciliary GPCR signaling in the overall signaling of neurons.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/fisiologia , Cílios/fisiologia , Neurônios/fisiologia , Receptores de Somatostatina/fisiologia , Transdução de Sinais/fisiologia , Anormalidades Múltiplas/genética , Anormalidades Múltiplas/fisiopatologia , Proteínas Adaptadoras de Transporte Vesicular/genética , Animais , Membrana Celular/fisiologia , Cerebelo/anormalidades , Cerebelo/fisiopatologia , AMP Cíclico/metabolismo , Anormalidades do Olho/genética , Anormalidades do Olho/fisiopatologia , Feminino , Doenças Renais Císticas/genética , Doenças Renais Císticas/fisiopatologia , Sistema de Sinalização das MAP Quinases/genética , Sistema de Sinalização das MAP Quinases/fisiologia , Camundongos , Camundongos Knockout , Gravidez , Receptores de Somatostatina/genética , Retina/anormalidades , Retina/fisiopatologia , Transdução de Sinais/genética
3.
J Thorac Cardiovasc Surg ; 161(4): e261-e271, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-31924353

RESUMO

BACKGROUND: Aortic valve interstitial cells have been implicated in the pathogenesis of aortic stenosis. In response to proinflammatory stimuli, aortic valve interstitial cells undergo an osteogenic phenotypic change. The purpose of this study was to determine whether the anti-inflammatory effects of statins prevent osteogenic activity in cultured aortic valve interstitial cells. METHODS: Human aortic valve interstitial cells were isolated from hearts explanted for cardiac transplantation. To test whether simvastatin down-regulates TLR4-induced osteogenic response, aortic valve interstitial cells were treated with simvastatin with and without TLR4 agonist lipopolysaccharide (LPS), and osteogenic markers were measured. Simvastatin's influence on in vitro calcium deposition was assessed by alizarin red staining. Knockdown of postreceptor signaling proteins (MyD88 and TRIF) was performed to determine which of 2 TLR4-associated pathways mediates the osteogenic response. Expression levels of TLR4-induced nuclear factor kappa light chain enhancer of activated B cells (NF-κB) and TLR4 expression were assessed after treatment with simvastatin. Statistical testing was done by analysis of variance (P < .05). RESULTS: Simvastatin decreased LPS-induced ALP and Runx2 expression and inhibited in vitro calcium deposition in aortic valve interstitial cells. Knockdown of MyD88 and TRIF attenuated the osteogenic response. Simvastatin attenuated TLR4-dependent NF-κB signaling and down-regulated TLR4 levels. CONCLUSIONS: Simvastatin prevented TLR4-induced osteogenic phenotypic changes in isolated aortic valve interstitial cells via down-regulation of TLR4 and inhibition of NF-κB signaling. These data offer mechanistic insight into a possible therapeutic role for simvastatin in the prevention of aortic stenosis.


Assuntos
Valva Aórtica/efeitos dos fármacos , Valva Aórtica/patologia , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Osteogênese/efeitos dos fármacos , Sinvastatina/farmacologia , Proteínas Adaptadoras de Transporte Vesicular/fisiologia , Fosfatase Alcalina/metabolismo , Valva Aórtica/metabolismo , Técnicas de Cultura de Células , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Humanos , Lipopolissacarídeos/fisiologia , Fator 88 de Diferenciação Mieloide/fisiologia , NF-kappa B/metabolismo , Transdução de Sinais , Receptor 4 Toll-Like/fisiologia
4.
Cancer Immunol Immunother ; 69(10): 2113-2124, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32448982

RESUMO

Metastatic cancer involving spread to the peritoneal cavity is referred to as peritoneal carcinomatosis and has a very poor prognosis. Our previous studies demonstrated a toll-like receptor 4 (TLR4) and C-type lectin receptor (CLR; Mincle/MCL) agonist pairing of monophosphoryl lipid A (MPL) and trehalose-6,6'-dicorynomycolate (TDCM) effectively inhibits peritoneal tumor growth and ascites development through a mechanism dependent upon B1a cell-produced natural IgM, complement, and phagocytes. In the current study, we investigated the requirement for TLR4 and Fc receptor common γ chain (FcRγ), required for Mincle/MCL signaling, in the MPL/TDCM-elicited response. MPL/TDCM significantly increased macrophages and Ly6Chi monocytes in the peritoneal cavity of both TLR4-/- and FcRγ-/- mice, suggesting redundancy in the signals required for monocyte/macrophage recruitment. However, B1 cell activation, antibody secreting cell differentiation, and tumor-reactive IgM production were defective in TLR4-/-, but not FcRγ-/- mice. TRIF was required for production of IgM reactive against tumor- and mucin-related antigens, but not phosphorylcholine, whereas TLR4 was required for production of both types of reactivities. Consistent with this, B1 cells lacking TLR4 or TRIF did not proliferate or differentiate into tumor-reactive IgM-producing cells in vitro and did not reconstitute MPL/TDCM-dependent protection against peritoneal carcinomatosis in CD19-/- mice. Our results indicate a TLR4/TRIF-dependent pathway is required by B1 cells for MPL/TDCM-elicited production of protective tumor-reactive natural IgM. The dependency on TRIF signaling for tumor-reactive, but not phosphorylcholine-reactive, IgM production reveals unexpected heterogeneity in TLR4-dependent regulation of natural IgM production, thereby highlighting important differences to consider when designing vaccines or therapies targeting these specificities.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/fisiologia , Subpopulações de Linfócitos B/imunologia , Fatores Corda/administração & dosagem , Imunoglobulina M/imunologia , Lipídeo A/análogos & derivados , Neoplasias Peritoneais/imunologia , Receptor 4 Toll-Like/fisiologia , Adjuvantes Imunológicos/administração & dosagem , Animais , Subpopulações de Linfócitos B/efeitos dos fármacos , Lipídeo A/administração & dosagem , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/fisiologia , Neoplasias Peritoneais/metabolismo , Neoplasias Peritoneais/patologia
5.
FEBS Open Bio ; 10(3): 407-413, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31961070

RESUMO

Post-transcriptional regulation of cytokine production is crucial to ensure appropriate immune responses. We previously demonstrated that poly-rC-binding protein-1 (PCBP1) can act as a trans-acting factor to stabilize transcripts encoding sortilin, which mediates cytokine trafficking. Here, we report that PCBP2, which strongly resembles PCBP1, can stabilize sortilin transcripts in macrophages using the same mechanism employed by PCBP1. PCBP2 recognized the C-rich element in the 3' UTR of sortilin mRNA, and PCBP2 knockdown decreased sortilin transcripts, indicating that PCBP2 stabilizes sortilin mRNA by binding to its 3' UTR. Zn2+ reversibly inhibited the nucleotide binding ability of PCBP2 in vitro. These findings suggest that both PCBP2 and PCBP1 may control the stability of sortilin transcripts by sensing intracellular Zn2+ levels in immune cells.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Proteínas de Ligação a RNA/metabolismo , Regiões 3' não Traduzidas/genética , Proteínas Adaptadoras de Transporte Vesicular/genética , Proteínas Adaptadoras de Transporte Vesicular/fisiologia , Animais , Técnicas de Cultura de Células , Citoplasma/metabolismo , Proteínas de Ligação a DNA/genética , Feminino , Expressão Gênica/genética , Regulação da Expressão Gênica , Ribonucleoproteínas Nucleares Heterogêneas/genética , Ribonucleoproteínas Nucleares Heterogêneas/metabolismo , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Ligação Proteica , Processamento Pós-Transcricional do RNA/genética , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/fisiologia , Zinco/metabolismo
6.
Cell Biol Int ; 44(3): 894-904, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-31868265

RESUMO

Disabled-2 (Dab2) and PAR-3 (partitioning defective 3) are reported to play critical roles in maintaining retinal microvascular endothelial cells biology by regulating VEGF-VEGFR-2 signaling. The role of Dab2 and PAR-3 in glomerular endothelial cell (GEnC) is unclear. In this study, we found that, no matter whether with vascular endothelial growth factor (VEGF) treatment or not, decreased expression of Dab2 could lead to cell apoptosis by preventing activation of VEGF-VEGFR-2 signaling in GEnC, accompanied by reduced membrane VEGFR-2 expression. And silencing of PAR-3 gene expression caused increased apoptosis of GEnC by inhibiting activation of VEGF-VEGFR-2 signaling and membrane VEGFR-2 expression. In our previous research, we found that the silencing of syndecan-1 gene expression inhibited VEGF-VEGFR-2 signaling by modulating internalization of VEGFR-2. And our further research demonstrated that downregulation of syndecan-1 lead to no significant change in the expression of Dab2 and PAR-3 both at messenger RNA and protein levels in GEnC, while phosphorylation of Dab2 was significantly increased in GEnC transfected with Dab2 small interfering RNA (siRNA) compared with control siRNA. Atypical protein kinase C (aPKC) could induce phosphorylation of Dab2, thus negatively regulating VEGF-VEGFR-2 signaling. And we found that decreased expression of syndecan-1 lead to activation of aPKC, and aPKC inhibitor treatment could block phosphorylation of Dab2 in GEnC. Besides, aPKC inhibitor treatment could activate VEGF-VGEFR-2 signaling in GEnC transfected with syndecan-1 siRNA in a dose-dependent manner. In conclusion, we speculated that phosphorylation of Dab2 is involved in preventing activation of VEGF-VEGFR-2 signaling in GEnC transfected with syndecan-1 siRNA. This provides a new target for the therapy of GEnC injury and kidney disease.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/fisiologia , Células Endoteliais/metabolismo , Glomérulos Renais/metabolismo , Proteínas do Tecido Nervoso/fisiologia , Animais , Apoptose , Células Cultivadas , Células Endoteliais/citologia , Glomérulos Renais/citologia , Proteína Quinase C/metabolismo , Ratos , Sindecana-1/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
7.
Oncogene ; 38(24): 4875-4884, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30770901

RESUMO

Castration-resistant prostate cancer (CRPC) with neuroendocrine differentiation (NED) is a lethal disease for which effective therapies are urgently needed. The mechanism underlying development of CRPC with NED, however, remains largely uncharacterized. In this study, we explored and characterized the functional role of neurotensin (NTS) in cell line and animal models of CRPC with NED. NTS was acutely induced by androgen deprivation in animal models of prostate cancer (PCa) and activated downstream signaling leading to NED through activation of neurotensin receptor 1 (NTSR1) and neurotensin receptor 3 (NTSR3), but not neurotensin receptor 2 (NTSR2). Our findings also revealed the existence of a CK8+/CK14+ subpopulation in the LNCaP cell line that expresses high levels of both NTSR1 and NTSR3, and displays an enhanced susceptibility to develop neuroendocrine-like phenotypes upon treatment with NTS. More importantly, NTSR1 pathway inhibition prevented the development of NED and castration resistance in vivo. We propose a novel role of NTS in the development of CRPC with NED, and a possible strategy to prevent the onset of NED by targeting the NTS signaling pathway.


Assuntos
Transdiferenciação Celular/genética , Células Neuroendócrinas/fisiologia , Neurotensina/fisiologia , Próstata/patologia , Neoplasias de Próstata Resistentes à Castração/patologia , Receptores de Neurotensina/fisiologia , Proteínas Adaptadoras de Transporte Vesicular/fisiologia , Animais , Linhagem Celular Tumoral , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Próstata/metabolismo , Neoplasias de Próstata Resistentes à Castração/genética , Neoplasias de Próstata Resistentes à Castração/fisiopatologia
8.
Exp Mol Pathol ; 106: 17-26, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30439350

RESUMO

Invadopodia, cancer cell protrusions with proteolytic activity, are functionally associated with active remodeling of the extracellular matrix. Here, we show that the invadopodia-related protein TKS5 is expressed in human pancreatic adenocarcinoma lines, and demonstrate that pancreatic cancer cells depend on TKS5 for invadopodia formation and function. Immunofluorescence staining of human pancreatic cancer cells reveals that TKS5 is a marker of mature and immature invadopodia. We also analyze the co-staining patterns of TKS5 and the commonly used invadopodia marker Cortactin, and find only partial co-localization of these two proteins at invadopodia, with a large fraction of TKS5-positive invadopodia lacking detectable levels of Cortactin. Whereas compelling evidence exist on the role of invadopodia as mediators of invasive migration in cultured cells and in animal models of cancer, these structures have never been detected inside human tumors. Here, using antibodies against TKS5 and Cortactin, we describe for the first time structures strongly resembling invadopodia in various paraffin-embedded human tumor surgical specimens from pancreas and other organs. Our results strongly suggest that invadopodia are present inside human tumors, and warrants further investigation on their regulation and occurrence in surgical specimens, and on the value of TKS5 antibodies as pathological research and diagnostic tools.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/fisiologia , Adenocarcinoma/patologia , Proteínas de Neoplasias/fisiologia , Neoplasias Pancreáticas/patologia , Podossomos/fisiologia , Adenocarcinoma/química , Adenocarcinoma/cirurgia , Adenocarcinoma/ultraestrutura , Adulto , Idoso , Linhagem Celular Tumoral , Cortactina/análise , Feminino , Técnica Direta de Fluorescência para Anticorpo , Humanos , Masculino , Pessoa de Meia-Idade , Invasividade Neoplásica , Neoplasias/química , Neoplasias/patologia , Neoplasias Pancreáticas/química , Neoplasias Pancreáticas/cirurgia , Neoplasias Pancreáticas/ultraestrutura , Inclusão em Parafina , Podossomos/química , Podossomos/ultraestrutura , Interferência de RNA , RNA Interferente Pequeno/genética
9.
Gastroenterology ; 155(5): 1539-1552.e12, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30063920

RESUMO

BACKGROUND & AIMS: Activating transcription factor 6 (ATF6) regulates endoplasmic reticulum stress. We studied whether ATF6 contributes to the development of colorectal cancer (CRC) using tissue from patients and transgenic mice. METHODS: We analyzed data from 541 patients with CRC in The Cancer Genome Atlas database for genetic variants and aberrant expression levels of unfolded protein response genes. Findings were validated in a cohort of 83 patients with CRC in Germany. We generated mice with intestinal epithelial cell-specific expression of the active form of Atf6 (nATF6IEC) from 2 alleles (homozygous), mice with expression of nATF6IEC from 1 allele (heterozygous), and nATF6IECfl/fl mice (controls). All nATF6IEC mice were housed under either specific-pathogen-free or germ-free conditions. Cecal microbiota from homozygous nATF6IEC mice or control mice was transferred into homozygous nATF6IEC mice or control mice. nATF6IEC mice were crossed with mice with disruptions in the myeloid differentiation primary response gene 88 and toll-like receptor adaptor molecule 1 gene (Myd88/Trif-knockout mice). Intestinal tissues were collected from mice and analyzed by histology, immunohistochemistry, immunoblots, gene expression profiling of unfolded protein response and inflammatory genes, array-based comparative genome hybridization, and 16S ribosomal RNA gene sequencing. RESULTS: Increased expression of ATF6 was associated with reduced disease-free survival times of patients with CRC. Homozygous nATF6IEC mice developed spontaneous colon adenomas at 12 weeks of age. Compared with controls, homozygous nATF6IEC mice had changes in the profile of their cecal microbiota, increased proliferation of intestinal epithelial cells, and loss of the mucus barrier-all preceding tumor formation. These mice had increased penetration of bacteria into the inner mucus layer and activation of signal transducer and activator of transcription 3, yet inflammation was not observed at the pretumor or tumor stages. Administration of antibiotics to homozygous nATF6IEC mice greatly reduced tumor incidence, and germ-free housing completely prevented tumorigenesis. Analysis of nATF6IEC MyD88/TRIF-knockout mice showed that tumor initiation and growth required MyD88/TRIF-dependent activation of signal transducer and activator of transcription 3. Transplantation of cecal microbiota from nATF6IEC mice and control mice, collected before tumor formation, caused tumor formation in ex-germ-free nATF6IEC mice. CONCLUSIONS: In patients with CRC, ATF6 was associated with reduced time of disease-free survival. In studies of nATF6IEC mice, we found sustained intestinal activation of ATF6 in the colon to promote dysbiosis and microbiota-dependent tumorigenesis.


Assuntos
Fator 6 Ativador da Transcrição/fisiologia , Neoplasias Colorretais/etiologia , Disbiose/etiologia , Imunidade Inata , Intestinos/microbiologia , Proteínas Adaptadoras de Transporte Vesicular/fisiologia , Animais , Neoplasias Colorretais/mortalidade , Progressão da Doença , Humanos , Camundongos , Fator 88 de Diferenciação Mieloide/fisiologia , Fator de Transcrição STAT3/fisiologia , Receptores Toll-Like/fisiologia , Resposta a Proteínas não Dobradas
10.
J Immunol Res ; 2018: 6249085, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29977930

RESUMO

Toll/IL-1R-domain-containing adaptor-inducing IFN-ß (TRIF) is an important adaptor for TLR3- and TLR4-mediated inflammatory signaling pathways. Recent studies have shown that TRIF plays a key role in vessel inflammation and atherosclerosis; however, the precise mechanisms are unclear. We investigated the mechanisms of the TRIF-regulated inflammatory response in RAW264.7 macrophages under oxidized low-density lipoprotein (ox-LDL) stimulation. Our data show that ox-LDL induces TRIF, miR-155, and BIC expression, activates the ERK1/2 and SOCS1-STAT3-NF-κB signaling pathways, and elevates the levels of IL-6 and TNF-α in RAW264.7 cells. Knockdown of TRIF using TRIF siRNA suppressed BIC, miR-155, IL-6, and TNF-α expression and inhibited the ERK1/2 and SOCS1-STAT3-NF-κB signaling pathways. Inhibition of ERK1/2 signaling also suppressed BIC and miR-155 expression. These findings suggest that TRIF plays an important role in regulating the ox-LDL-induced macrophage inflammatory response and that TRIF modulates the expression of BIC/miR-155 and the downstream SOCS1-STAT3-NF-κB signaling pathway via ERK1/2. Therefore, TRIF might be a novel therapeutic target for atherosclerosis.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Lipoproteínas LDL/farmacologia , Sistema de Sinalização das MAP Quinases , Macrófagos/metabolismo , MicroRNAs/metabolismo , Proteínas Adaptadoras de Transporte Vesicular/genética , Proteínas Adaptadoras de Transporte Vesicular/fisiologia , Animais , Inativação Gênica , Mediadores da Inflamação/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/enzimologia , Macrófagos/fisiologia , Camundongos , MicroRNAs/efeitos dos fármacos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/fisiologia , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/fisiologia , NF-kappa B/metabolismo , Células RAW 264.7 , Precursores de RNA/metabolismo , RNA Interferente Pequeno , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Proteína 1 Supressora da Sinalização de Citocina/metabolismo
11.
Brain Behav Immun ; 73: 364-374, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29852290

RESUMO

Hypothalamic inflammation is a key component of acute sickness behavior and cachexia, yet mechanisms of inflammatory signaling in the central nervous system remain unclear. Previous work from our lab and others showed that while MyD88 is an important inflammatory signaling pathway for sickness behavior, MyD88 knockout (MyD88KO) mice still experience sickness behavior after inflammatory stimuli challenge. We found that after systemic lipopolysaccharide (LPS) challenge, MyD88KO mice showed elevated expression of several cytokine and chemokine genes in the hypothalamus. We therefore assessed the role of an additional inflammatory signaling pathway, TRIF, in acute inflammation (LPS challenge) and in a chronic inflammatory state (cancer cachexia). TRIFKO mice resisted anorexia and weight loss after peripheral (intraperitoneal, IP) or central (intracerebroventricular, ICV) LPS challenge and in a model of pancreatic cancer cachexia. Compared to WT mice, TRIFKO mice showed attenuated upregulation of Il6, Ccl2, Ccl5, Cxcl1, Cxcl2, and Cxcl10 in the hypothalamus after IP LPS treatment, as well as attenuated microglial activation and neutrophil infiltration into the brain after ICV LPS treatment. Lastly, we found that TRIF was required for Ccl2 upregulation in the hypothalamus and induction of the catabolic genes, Mafbx, Murf1, and Foxo1 in gastrocnemius during pancreatic cancer. In summary, our results show that TRIF is an important inflammatory signaling mediator of sickness behavior and cachexia and presents a novel therapeutic target for these conditions.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/fisiologia , Caquexia/fisiopatologia , Comportamento de Doença/efeitos dos fármacos , Proteínas Adaptadoras de Transporte Vesicular/imunologia , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Animais , Encéfalo/metabolismo , Citocinas/metabolismo , Feminino , Hipotálamo/metabolismo , Comportamento de Doença/fisiologia , Inflamação/metabolismo , Lipopolissacarídeos/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microglia/metabolismo , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/metabolismo , Neoplasias/metabolismo , Transdução de Sinais/efeitos dos fármacos
12.
Mol Immunol ; 93: 206-215, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29207327

RESUMO

In response to environmental stimuli such as granulocyte-macrophage or macrophage colony stimulating factor (GM-CSF/M-CSF), macrophages (MΦ) can acquire distinct functional phenotypes that control inflammatory processes on the one hand and contribute to a broad spectrum of pathologies on the other. Potential intervention strategies will require an understanding of the signalling processes that are associated with macrophage polarization. In the present study, we show that M-MΦ produce more IFN-ß and IL-10 and a lot less TNF-α than do GM-MΦ in response to LPS. To define the molecular mechanisms that underlie the biosynthesis of TNF-α we carried out a detailed investigation of the LPS-induced activation of the canonical and non-canonical myeloid differentiation primary response 88 (MyD88)-dependent signal transduction pathways as well as the TIR-domain-containing adapter-inducing interferon-ß (TRIF)-dependent pathway. Our results show that all three pathways are activated in both cell types and that the activation is more pronounced in M-MΦ. While IL-10 was found to interfere with TNF-α production in M-MΦ, we exclude a decisive role for IFN-ß in this respect. Furthermore, we demonstrate that TNF-α mRNA is markedly destabilized in M-MΦ and that expression of the mRNA destabilizing protein tristetraprolin is greatly enhanced in these cells. Collectively, our study suggests that differential effects of LPS on TNF-α mRNA turnover and on signal transduction pathways influence the amount of TNF-α finally produced by GM-MΦ and M-MΦ.


Assuntos
Lipopolissacarídeos/farmacologia , Macrófagos/efeitos dos fármacos , Fator de Necrose Tumoral alfa/biossíntese , Proteínas Adaptadoras de Transporte Vesicular/fisiologia , Células Cultivadas , Regulação da Expressão Gênica , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Humanos , Interferon beta/farmacologia , Interleucina-10/farmacologia , Ativação de Macrófagos/efeitos dos fármacos , Fator Estimulador de Colônias de Macrófagos/farmacologia , Macrófagos/metabolismo , Fator 88 de Diferenciação Mieloide/fisiologia , Estabilidade de RNA , RNA Mensageiro/metabolismo , Transdução de Sinais/efeitos dos fármacos , Tristetraprolina/metabolismo , Fator de Necrose Tumoral alfa/genética
13.
J Biol Chem ; 292(1): 146-160, 2017 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-27881673

RESUMO

The liver plays a key role in cholesterol metabolism. Impaired hepatic cholesterol homeostasis causes intracellular free cholesterol accumulation and hepatocyte injury. Sortilin 1 (SORT1) is a lysosomal trafficking receptor that was identified by genome-wide association studies (GWAS) as a novel regulator of cholesterol metabolism in humans. Here we report that SORT1 deficiency protected against cholesterol accumulation-induced liver injury and inflammation in mice. Using an LC-MS/MS-based proteomics approach, we identified liver carboxylesterase 1 (CES1) as a novel SORT1-interacting protein. Mechanistic studies further showed that SORT1 may regulate CES1 lysosomal targeting and degradation and that SORT1 deficiency resulted in higher liver CES1 protein abundance. Previous studies have established an important role of hepatic CES1 in promoting intracellular cholesterol mobilization, cholesterol efflux, and bile acid synthesis. Consistently, high cholesterol atherogenic diet-challenged Sort1 knock-out mice showed less hepatic free cholesterol accumulation, increased bile acid synthesis, decreased biliary cholesterol secretion, and the absence of gallstone formation. SORT1 deficiency did not alter hepatic ceramide and fatty acid metabolism in high cholesterol atherogenic diet-fed mice. Finally, knockdown of liver CES1 in mice markedly increased the susceptibility to high cholesterol diet-induced liver injury and abolished the protective effect against cholesterol lipotoxicity in Sort1 knock-out mice. In summary, this study identified a novel SORT1-CES1 axis that regulates cholesterol-induced liver injury, which provides novel insights that improve our current understanding of the molecular links between SORT1 and cholesterol metabolism. This study further suggests that therapeutic inhibition of SORT1 may be beneficial in improving hepatic cholesterol homeostasis in metabolic and inflammatory liver diseases.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/fisiologia , Hidrolases de Éster Carboxílico/metabolismo , Doença Hepática Induzida por Substâncias e Drogas/patologia , Colesterol/toxicidade , Hepatócitos/patologia , Inflamação/patologia , Animais , Hidrolases de Éster Carboxílico/antagonistas & inibidores , Hidrolases de Éster Carboxílico/genética , Células Cultivadas , Doença Hepática Induzida por Substâncias e Drogas/etiologia , Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Feminino , Células Hep G2 , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Humanos , Inflamação/etiologia , Inflamação/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , RNA Interferente Pequeno/genética
14.
Crit Care Med ; 44(11): e1054-e1066, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27509390

RESUMO

OBJECTIVES: Lung contusion is a major risk factor for the development of acute respiratory distress syndrome. We set to determine the role of toll-like receptor 3 and the binding of double-stranded RNA in the pathogenesis of sterile injury following lung contusion. DESIGN: Toll-like receptor 3 expression was analyzed in postmortem lung samples from patients with lung contusion. Unilateral lung contusion was induced in toll-like receptor 3 (-/-), TIR-domain-containing adapter-inducing interferon-ß (-/-), and wild-type mice. Subsequently, lung injury and inflammation were evaluated. Apoptotic indices, phagocytic activity, and phenotypic characterization of the macrophages were determined. Double-stranded RNA in bronchoalveolar lavage and serum samples following lung contusion was measured. A toll-like receptor 3/double-stranded RNA ligand inhibitor was injected into wild-type mice prior to lung contusion. MEASUREMENTS AND MAIN RESULTS: Toll-like receptor 3 expression was higher in patients and wild-type mice with lung contusion. The degree of lung injury, inflammation, and macrophage apoptosis was reduced in toll-like receptor 3 (-/-), TIR-domain-containing adapter-inducing interferon-ß (-/-), and wild-type mice with toll-like receptor 3 antibody neutralization. Alveolar macrophages from toll-like receptor 3 (-/-) mice had a lower early apoptotic index, a predominant M2 phenotype and increased surface translocation of toll-like receptor 3 from the endosome to the surface. When compared with viral activation pathways, lung injury in lung contusion demonstrated increased p38 mitogen-activated protein kinases, extracellular signal-regulated kinase 1/2 phosphorylation with inflammasome activation without a corresponding increase in nuclear factor-κB or type-1 interferon production. Additionally, pretreatment with toll-like receptor 3/double-stranded RNA ligand inhibitor led to a reduction in injury, inflammation, and macrophage apoptosis. CONCLUSIONS: We conclude that the interaction of double-stranded RNA from injured cells with toll-like receptor 3 drives the acute inflammatory response following lung contusion.


Assuntos
Contusões/metabolismo , Lesão Pulmonar/metabolismo , RNA de Cadeia Dupla/metabolismo , Receptor 3 Toll-Like/metabolismo , Proteínas Adaptadoras de Transporte Vesicular/fisiologia , Albuminas/metabolismo , Animais , Apoptose , Líquido da Lavagem Broncoalveolar , Contusões/patologia , Citocinas/metabolismo , Células Epiteliais/patologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Inflamassomos/metabolismo , Pulmão/metabolismo , Pulmão/patologia , Lesão Pulmonar/patologia , Linfócitos/patologia , Macrófagos Alveolares/patologia , Camundongos , Fator 88 de Diferenciação Mieloide/metabolismo , Fosforilação , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
15.
Clin Chim Acta ; 460: 11-7, 2016 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-27312323

RESUMO

Several lines of evidence have shown that SORT1 gene within 1p13.3 locus is an important modulator of the low-density lipoprotein-cholesterol (LDL-C) level and atherosclerosis risk. Here, we summarize the effects of SORT1, which codes for sortilin, on lipid metabolism and development of atherosclerosis and explore the mechanisms underlying sortilin effects on lipid metabolism especially in hepatocytes and macrophages. Recent epidemiological evidence demonstrated that sortilin has been implicated as the causative factor and regulates lipid metabolism in vivo. Hepatic sortilin overexpression leads to both increased and decreased LDL-C levels by several different mechanisms, suggesting the complex roles of sortilin in hepatic lipid metabolism. Macrophage sortilin causes internalization of LDL and probably a reduction in cholesterol efflux, resulting in the intracellular accumulation of excessive lipids. In addition, sortilin deficiency in an atherosclerotic mouse model results in decreased aortic atherosclerotic lesion. Sortilin involves in lipid metabolism, promotes the development of atherosclerosis, and possibly becomes a potential therapeutic target for atherosclerosis treatment.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/fisiologia , Aterosclerose , Metabolismo dos Lipídeos , Proteínas Adaptadoras de Transporte Vesicular/genética , Animais , LDL-Colesterol/metabolismo , Humanos , Macrófagos/metabolismo
16.
J Leukoc Biol ; 100(6): 1311-1322, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27354411

RESUMO

Treatment with the TLR4 agonist MPLA augments innate resistance to common bacterial pathogens. However, the cellular and molecular mechanisms by which MPLA augments innate immunocyte functions are not well characterized. This study examined the importance of MyD88- and TRIF-dependent signaling for leukocyte mobilization, recruitment, and activation following administration of MPLA. MPLA potently induced MyD88- and TRIF-dependent signaling. A single injection of MPLA caused rapid mobilization and recruitment of neutrophils, a response that was largely mediated by the chemokines CXCL1 and -2 and the hemopoietic factor G-CSF. Rapid neutrophil recruitment and chemokine production were regulated by both pathways although the MyD88-dependent pathway showed some predominance. In further studies, multiple injections of MPLA potently induced mobilization and recruitment of neutrophils and monocytes. Neutrophil recruitment after multiple injections of MPLA was reliant on MyD88-dependent signaling, but effective monocyte recruitment required activation of both pathways. MPLA treatment induced expansion of myeloid progenitors in bone marrow and upregulation of CD11b and shedding of L-selectin by neutrophils, all of which were attenuated in MyD88- and TRIF-deficient mice. These results show that MPLA-induced neutrophil and monocyte recruitment, expansion of bone marrow progenitors and augmentation of neutrophil adhesion molecule expression are regulated by both the MyD88- and TRIF-dependent pathways.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/fisiologia , Imunidade Inata , Lipídeo A/análogos & derivados , Monócitos/imunologia , Fator 88 de Diferenciação Mieloide/fisiologia , Neutrófilos/imunologia , Receptor 4 Toll-Like/agonistas , Proteínas Adaptadoras de Transporte Vesicular/deficiência , Animais , Antígeno CD11b/biossíntese , Antígeno CD11b/genética , Quimiocina CXCL1/fisiologia , Quimiocina CXCL2/fisiologia , Quimiotaxia de Leucócito/efeitos dos fármacos , Fator Estimulador de Colônias de Granulócitos/fisiologia , Selectina L/metabolismo , Lipídeo A/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Fator 88 de Diferenciação Mieloide/deficiência , Mielopoese/efeitos dos fármacos , Neutrófilos/efeitos dos fármacos , Neutrófilos/metabolismo , Receptores de Interleucina-8B/fisiologia , Transdução de Sinais , Receptor 4 Toll-Like/fisiologia
17.
Biochem Biophys Res Commun ; 463(4): 719-25, 2015 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-26047698

RESUMO

Hepatocytes are the target host cells during hepatitis B virus (HBV) infection. Recent studies indicate that the innate immune response of hepatocytes plays an important role in inhibiting HBV replication. TIR-domain-containing adaptor inducing interferon-beta (TRIF) is a key component in innate immune signaling pathways. In this study, we found that the TRIF protein was downregulated in human hepatoma cell lines and liver tissue samples harboring HBV. Hepatitis B virus X protein (HBX) reduced TRIF protein expression in a dose-dependent manner via the proteasomal pathway. HBX appeared to not directly interact with TRIF as these proteins failed to co-immunoprecipitate when overexpressed in hepatoma cells. TRIF upregulation in hepatoma cell lines dramatically inhibited HBV transcription and expression of its viral proteins. Cellular apoptosis induced by TRIF was also confirmed in hepatoma cell lines. Taken together, these findings reveal a new mechanism for HBV evasion of the cellular innate immunity by HBX-mediated degradation of TRIF protein in hepatocytes.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/fisiologia , Regulação para Baixo/fisiologia , Vírus da Hepatite B/fisiologia , Evasão da Resposta Imune/fisiologia , Imunidade Inata , Sequência de Bases , Primers do DNA , Replicação do DNA , DNA Viral/biossíntese , Ensaio de Imunoadsorção Enzimática , Células Hep G2 , Vírus da Hepatite B/genética , Humanos , Reação em Cadeia da Polimerase em Tempo Real , Transativadores/fisiologia , Proteínas Virais Reguladoras e Acessórias
18.
PLoS One ; 10(3): e0121003, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25826475

RESUMO

The ability of cancer cells to invade underlies metastatic progression. One mechanism by which cancer cells can become invasive is through the formation of structures called invadopodia, which are dynamic, actin-rich membrane protrusions that are sites of focal extracellular matrix degradation. While there is a growing consensus that invadopodia are instrumental in tumor metastasis, less is known about whether they are involved in tumor growth, particularly in vivo. The adaptor protein Tks5 is an obligate component of invadopodia, and is linked molecularly to both actin-remodeling proteins and pericellular proteases. Tks5 appears to localize exclusively to invadopodia in cancer cells, and in vitro studies have demonstrated its critical requirement for the invasive nature of these cells, making it an ideal surrogate to investigate the role of invadopodia in vivo. In this study, we examined how Tks5 contributes to human breast cancer progression. We used immunohistochemistry and RNA sequencing data to evaluate Tks5 expression in clinical samples, and we characterized the role of Tks5 in breast cancer progression using RNA interference and orthotopic implantation in SCID-Beige mice. We found that Tks5 is expressed to high levels in approximately 50% of primary invasive breast cancers. Furthermore, high expression was correlated with poor outcome, particularly in those patients with late relapse of stage I/II disease. Knockdown of Tks5 expression in breast cancer cells resulted in decreased growth, both in 3D in vitro cultures and in vivo. Moreover, our data also suggest that Tks5 is important for the integrity and permeability of the tumor vasculature. Together, this work establishes an important role for Tks5 in tumor growth in vivo, and suggests that invadopodia may play broad roles in tumor progression.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/fisiologia , Neoplasias da Mama/patologia , Divisão Celular/fisiologia , Animais , Xenoenxertos , Humanos , Técnicas In Vitro , Camundongos , Camundongos SCID
20.
Circ Res ; 116(5): 789-96, 2015 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-25593281

RESUMO

RATIONALE: Noncoding gene variants at the SORT1 locus are strongly associated with low-density lipoprotein cholesterol (LDL-C) levels, as well as with coronary artery disease. SORT1 encodes a protein called sortilin, and hepatic sortilin modulates LDL metabolism by targeting apolipoprotein B-containing lipoproteins to the lysosome. Sortilin is also expressed in macrophages, but its role in macrophage uptake of LDL and in atherosclerosis independent of plasma LDL-C levels is unknown. OBJECTIVE: To determine the effect of macrophage sortilin expression on LDL uptake, foam cell formation, and atherosclerosis. METHODS AND RESULTS: We crossed Sort1(-/-) mice onto a humanized Apobec1(-/-); hAPOB transgenic background and determined that Sort1 deficiency on this background had no effect on plasma LDL-C levels but dramatically reduced atherosclerosis in the aorta and aortic root. To test whether this effect was a result of macrophage sortilin deficiency, we transplanted Sort1(-/-);LDLR(-/-) or Sort1(+/+);LDLR(-/-) bone marrow into Ldlr(-/-) mice and observed a similar reduction in atherosclerosis in mice lacking hematopoetic sortilin without an effect on plasma LDL-C levels. In an effort to determine the mechanism by which hematopoetic sortilin deficiency reduced atherosclerosis, we found no effect of sortilin deficiency on macrophage recruitment or lipopolysaccharide-induced cytokine release in vivo. In contrast, sortilin-deficient macrophages had significantly reduced uptake of native LDL ex vivo and reduced foam cell formation in vivo, whereas sortilin overexpression in macrophages resulted in increased LDL uptake and foam cell formation. CONCLUSIONS: Macrophage sortilin deficiency protects against atherosclerosis by reducing macrophage uptake of LDL. Sortilin-mediated uptake of native LDL into macrophages may be an important mechanism of foam cell formation and contributor to atherosclerosis development.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/fisiologia , Doenças da Aorta/etiologia , Aterosclerose/etiologia , Células Espumosas/metabolismo , Lipoproteínas LDL/metabolismo , Macrófagos/metabolismo , Desaminase APOBEC-1 , Proteínas Adaptadoras de Transporte Vesicular/deficiência , Proteínas Adaptadoras de Transporte Vesicular/genética , Animais , Doenças da Aorta/genética , Doenças da Aorta/metabolismo , Doenças da Aorta/prevenção & controle , Aterosclerose/genética , Aterosclerose/metabolismo , Aterosclerose/prevenção & controle , Células da Medula Óssea/metabolismo , LDL-Colesterol/sangue , Citidina Desaminase/genética , Dieta Ocidental/efeitos adversos , Feminino , Macrófagos Peritoneais/metabolismo , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Quimera por Radiação , Receptores de LDL/deficiência , Receptores de LDL/genética , Receptores de LDL/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA