Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Mol Immunol ; 158: 79-90, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37172353

RESUMO

BACKGROUND: Head and neck squamous cell carcinoma (HNSCC) is a ferroptosis sensitive tumor type with high mortality rate. However, it remains largely unknown whether ferroptosis influences the tumor cell in HNSCC. MATERIALS AND METHODS: To investigate how ferroptosis regulators were differentially expressed between normal and tumor tissue, data related to HNSCC was downloaded from The Cancer Genome Atlas. The expression levels of key factors in HNSCC and the relationship between key factors and ferroptosis in HNSCC were conducted in vitro, and then analyzed to correlate with the differences in prognosis and survival. This was then combined with TNM staging data, and the migration effects of key factors in HNSCC were verified by scratch test and transwell test. RESULTS: In this study, gene expression analysis and correlation studies between genes showed that HSPA5 was a potentially key associated ferroptosis regulator in HNSCC. Bioinformatics analysis showed that high expression of HSPA5 in HNSCC was positively correlated with poor prognosis and distal metastasis of HNSCC. In vitro immunohistochemistry and western blot tests confirmed that HSPA5 was highly expressed in HNSCC tissues and cell lines. In vitro inhibition of HSPA5 reduced the viability of HNSCC cells and increased ferroptosis. The results of scratch, transwell, and immunofluorescence tests showed that HSPA5 was related to the migration of HNSCC. In addition, a pan-cancer analysis showed that HSPA5 was also overexpressed in many types of cancer with poor prognoses. CONCLUSION: In total, our study demonstrates the critical role of ferroptosis regulators in HNSCC and that HSPA5, as a ferroptosis regulator, can be regarded as a key molecular target for designing new therapeutic regimens to control HNSCC metastasis and progression.


Assuntos
Ferroptose , Neoplasias de Cabeça e Pescoço , Humanos , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Neoplasias de Cabeça e Pescoço/genética , Carcinoma de Células Escamosas de Cabeça e Pescoço/genética
2.
mSystems ; 7(5): e0046922, 2022 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-36000726

RESUMO

Oral squamous cell carcinoma (OSCC) is a fatal disease, and periodontitis is associated with OSCC development. However, the pathogenesis in the context of OSCC with periodontitis has not been fully understood. Here, we demonstrated that periodontitis promoted OSCC development, accompanied by alterations in the oral bacterial community and the tumor immune microenvironment. The oral microbiota from periodontitis maintained the dominant position throughout the whole process of OSCC with periodontitis, of which Porphyromonas was the most abundant genus. The oral microbiota from periodontitis could activate interleukin-17-positive (IL-17+) γδ T cells directly. The activated γδ T cells were necessary for the IL-17/signal transducer and activator of transcription 3 (STAT3) pathway and promoted M2-tumor-associated macrophage (TAM) infiltration in OSCC proliferation. Our data provide insight into the carcinogenesis of OSCC with periodontitis by outlining the tumor-associated immune response shaped by the oral microbiota from periodontitis. Thus, oral commensal bacteria and IL-17+ γδ T cells might be potential targets for monitoring and treating OSCC. IMPORTANCE The work reveals the role of the oral microbiota from periodontitis in carcinogenesis. Furthermore, our study provides insight into the pathogenesis of OSCC with periodontitis by outlining the tumor-associated immune response shaped by the oral microbiota from periodontitis, which might identify new research and intervention targets for OSCC with periodontitis.


Assuntos
Carcinoma de Células Escamosas , Neoplasias de Cabeça e Pescoço , Microbiota , Neoplasias Bucais , Periodontite , Humanos , Carcinoma de Células Escamosas de Cabeça e Pescoço , Interleucina-17 , Periodontite/complicações , Linfócitos T , Carcinogênese , Microambiente Tumoral
3.
Mol Immunol ; 91: 65-74, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28886588

RESUMO

Human ß-defensin 3 (hBD3) is a cationic peptide with immunomodulatory effects on both innate and acquired immune responses. Periodontitis, an inflammatory disease that extends deep into periodontal tissues, causes the loss of supporting structures around the tooth. The present study assessed the effects of hBD3 as a monotherapy for periodontitis in mice and explored its potential mechanism. In vivo, hBD3 inhibited the levels of tumour necrosis factor (TNF)-α, interleukin-6, and matrix metalloprotease-9 in periodontium exposed to Porphyromonas gingivalis (P.g) in a mouse periodontitis model; reduced osteoclast formation and lower alveolar bone loss were also observed. In addition, hBD3 was related to the expression of polarization signature molecules in circulating monocytes. In vitro, hBD3 notably suppressed the production of TNF-α and interleukin-6 in RAW 264.7 cells stimulated by the lipopolysaccharide of P.g. Moreover, hBD3 attenuated polarization of RAW 264.7 cells into the M1 phenotype, with reduced activation of nuclear factor-κB signal transduction. In conclusion, hBD3 exhibits potent anti-periodontitis properties both in vitro and in vivo, and this effect may be correlated to inhibition of the nuclear factor-κB pathway and macrophage polarization.


Assuntos
Perda do Osso Alveolar/imunologia , Infecções por Bacteroidaceae/imunologia , Osteoclastos/imunologia , Periodontite/imunologia , Porphyromonas gingivalis/imunologia , beta-Defensinas/farmacologia , Perda do Osso Alveolar/tratamento farmacológico , Perda do Osso Alveolar/genética , Perda do Osso Alveolar/patologia , Animais , Infecções por Bacteroidaceae/tratamento farmacológico , Infecções por Bacteroidaceae/genética , Infecções por Bacteroidaceae/patologia , Modelos Animais de Doenças , Humanos , Interleucina-6/genética , Interleucina-6/imunologia , Masculino , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/imunologia , Camundongos , Camundongos Knockout , Osteoclastos/patologia , Periodontite/tratamento farmacológico , Periodontite/genética , Periodontite/patologia , Células RAW 264.7 , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/imunologia
4.
Cytokine ; 92: 48-54, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28092794

RESUMO

ß-defensin 3, a multifunctional antimicrobial peptide, has immuno-regulatory activities. We investigated the modulatory mechanism of human ß-defensin 3 (hBD3) on acute inflammatory response resulted from Porphyromonas gingivalis lipopolysaccharide (P.g-LPS), which plays a pro-inflammatory role in periodontal infection and its derived systemic inflammation. P.g-LPS was administrated to mice and murine macrophages alone or along with hBD3. P.g-LPS could lead to acute inflammation as soon as 2h. And it was observed that hBD3 significantly decreased the production of pro-inflammatory biomarkers of in response to P.g-LPS in vivo and in vitro in the early stage. Interestingly, although hBD3 as well as P.g-LPS stimulated the expression of TLR2 mRNA in macrophages in this study, hBD3 exhibited suppressive effect on the downstream NF-κB signaling pathway activated by P.g-LPS. And above all, hBD3 could polarize macrophages into M2 phenotype and this contributed to its anti-inflammatory property. These results indicated that hBD3 could have therapeutic effect on systemic inflammation associated with periodontal infections via modulating macrophage activation and orientation.


Assuntos
Lipopolissacarídeos/toxicidade , Ativação de Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Porphyromonas gingivalis/química , Transdução de Sinais/efeitos dos fármacos , beta-Defensinas/farmacologia , Doença Aguda , Animais , Humanos , Inflamação/induzido quimicamente , Inflamação/genética , Inflamação/imunologia , Lipopolissacarídeos/química , Ativação de Macrófagos/genética , Masculino , Camundongos , Camundongos Knockout , NF-kappa B/genética , NF-kappa B/imunologia , Transdução de Sinais/genética , Transdução de Sinais/imunologia
5.
Peptides ; 82: 92-100, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27298203

RESUMO

Human beta-defensin 3 (hBD3) is an antimicrobial peptide showing immunomodulatory effect on both innate and acquired immune response. Atherosclerosis is an inflammatory disease characterized by accumulation of lipids in the vascular wall. In this study, we evaluated whether hBD3 could attenuate the atherosclerosis development accelerated by Porphyromonas gingivalis lipopolysaccharide (Pg-LPS) with apolipoprotein E-deficient (ApoE(-/-)) mice. We observed that, in vivo, hBD3 inhibited serum MCP-1, sICAM-1 levels of ApoE-deficient mice exposed to Pg-LPS in a chronic inflammation model. Serum levels of total cholesterol (TC) and low-density lipoprotein (LDL) were also markedly reduced with hBD3 intervention. In addition, thinned vascular walls, less macrophage infiltration and the formation of atherosclerotic lesions were observed in the hBD3-treated group. Furthermore, in vitro, hBD3 profoundly suppressed the production of TNF-α and IL-6 in RAW 264.7 cells induced by Pg-LPS in a dose-dependent manner. Moreover, hBD3 attenuated the phosphorylation of p38 and ERK1/2 in the mitogen-activated protein kinase (MAPK) pathway. Taken together, our work has revealed that hBD3 exhibits potent anti-inflammatory properties both in vitro and in vivo, and this effect might be correlated with inhibition of MAPK pathway.


Assuntos
Aterosclerose/tratamento farmacológico , Inflamação/tratamento farmacológico , beta-Defensinas/administração & dosagem , Animais , Apolipoproteínas E/genética , Aterosclerose/sangue , Aterosclerose/induzido quimicamente , Aterosclerose/microbiologia , Quimiocina CCL2/sangue , Humanos , Inflamação/sangue , Inflamação/induzido quimicamente , Molécula 1 de Adesão Intercelular/sangue , Interleucina-6/genética , Lipopolissacarídeos/química , Lipopolissacarídeos/toxicidade , Camundongos , Quinases de Proteína Quinase Ativadas por Mitógeno/efeitos dos fármacos , Quinases de Proteína Quinase Ativadas por Mitógeno/genética , Porphyromonas gingivalis/química , Porphyromonas gingivalis/patogenicidade , Fator de Necrose Tumoral alfa/genética , beta-Defensinas/metabolismo
6.
Int Immunopharmacol ; 38: 204-13, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27289299

RESUMO

BACKGROUND AND AIM: Porphyromonas gingivalis (P.gingivalis) lipopolysaccharide (LPS) is reported to be associated with the progression of atherosclerosis (AS). In this study, we explored the potential of human ß-defensin-3 (hBD3), an antimicrobial peptide with immunomodulatory properties, to alleviate AS progression accelerated by P.gingivalis LPS and the mechanism underlying this effect. MATERIALS AND METHODS: Apolipoprotein E-deficient mice were injected intraperitoneally with hBD3, P.gingivalis LPS, or hBD3+P.gingivalis LPS. The aorta was assessed immunohistologically and mRNA levels of inflammatory cytokines were determined by quantitative PCR. Macrophages and vascular endothelial cells were stimulated in vitro to investigate the hBD3 target cells. Inflammatory cytokines in serum and cell culture supernatants were detected using cytometric bead arrays. Signaling pathways were investigated by Western blotting. RESULTS: In P.gingivalis LPS-treated mice, hBD3 significantly reduced serum IL-6 and TNF-α levels and aortic expression of ICAM-1, IL-6, and MCP-1 (mRNA and protein). The area and severity of atherosclerotic lesions were also diminished, with less advanced plaque formation, more continuous and distinct elastic lamina, and more normal smooth muscle cells arranged along the tunica media layer. In vitro, hBD3 decreased TNF-α, IL-1ß, IL-6 secretion and downregulated TNF-α, IL-1ß, IL-6, IL-8, VCAM-1, and IL-10 mRNA levels in macrophages. hBD3 did not influence TNF-α, IL-6, and IL-8 levels in HUVECs culture supernatants. Furthermore, hBD3 suppressed P.gingivalis LPS-induced activation of the NF-κB, p38 and JNK pathways. CONCLUSION: hBD3 alleviates AS progression accelerated by P.gingivalis LPS in apolipoprotein E-deficient mice by downregulating the cytokine expression in macrophages via the MAPK and NF-κB signaling pathways.


Assuntos
Anti-Infecciosos/uso terapêutico , Aorta/efeitos dos fármacos , Aterosclerose/tratamento farmacológico , Células Endoteliais/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Porphyromonas gingivalis/imunologia , beta-Defensinas/uso terapêutico , Animais , Aorta/patologia , Apolipoproteínas/genética , Aterosclerose/microbiologia , Citocinas/genética , Citocinas/metabolismo , Progressão da Doença , Células Endoteliais/imunologia , Células Endoteliais da Veia Umbilical Humana , Humanos , Mediadores da Inflamação/metabolismo , Lipopolissacarídeos/imunologia , Lipopolissacarídeos/metabolismo , Macrófagos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transdução de Sinais/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA