Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 14(1): 11179, 2024 05 16.
Artigo em Inglês | MEDLINE | ID: mdl-38750069

RESUMO

During a SARS-CoV-2 infection, macrophages recognize viral components resulting in cytokine production. While this response fuels virus elimination, overexpression of cytokines can lead to severe COVID-19. Previous studies suggest that the spike protein (S) of SARS-CoV-2 can elicit cytokine production via the transcription factor NF-κB and the toll-like receptors (TLRs). In this study, we found that: (i) S and the S2 subunit induce CXCL10, a chemokine implicated in severe COVID-19, gene expression by human macrophage cells (THP-1); (ii) a glycogen synthase kinase-3 inhibitor attenuates this induction; (iii) S and S2 do not activate NF-κB but do activate the transcription factor IRF; (iv) S and S2 do not require TLR2 to elicit CXCL10 production or activate IRF; and (v) S and S2 elicit CXCL10 production by peripheral blood mononuclear cells (PBMCs). We also discovered that the cellular response, or lack thereof, to S and S2 is a function of the recombinant S and S2 used. While such a finding raises the possibility of confounding LPS contamination, we offer evidence that potential contaminating LPS does not underly induced increases in CXCL10. Combined, these results provide insights into the complex immune response to SARS-CoV-2 and suggest possible therapeutic targets for severe COVID-19.


Assuntos
COVID-19 , Quimiocina CXCL10 , SARS-CoV-2 , Glicoproteína da Espícula de Coronavírus , Humanos , Quimiocina CXCL10/metabolismo , COVID-19/virologia , COVID-19/imunologia , COVID-19/metabolismo , Leucócitos Mononucleares/metabolismo , Leucócitos Mononucleares/virologia , Macrófagos/metabolismo , Macrófagos/imunologia , Macrófagos/virologia , NF-kappa B/metabolismo , Glicoproteína da Espícula de Coronavírus/metabolismo , Glicoproteína da Espícula de Coronavírus/imunologia , Células THP-1
2.
Biochem Biophys Res Commun ; 605: 171-176, 2022 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-35367865

RESUMO

A key component of severe COVID-19 is a "cytokine storm" i.e., the excessive expression of unneeded cytokines. Previous studies suggest that SARS-CoV-2 proteins can induce macrophages to secrete pro-inflammatory cytokines; a process that may involve Toll-like receptors (TLRs). Glycogen synthase kinase-3 (GSK-3) has been implicated in TLR signal transduction and a selective GSK-3 inhibitor, termed COB-187, dramatically attenuates cytokine expression induced by the TLR ligand lipopolysaccharide (LPS). In the present study, we provide evidence that the SARS-CoV-2 spike protein (S) and the S2 subunit (S2) induce production of CXCL10 (a chemokine elevated in severe COVID-19) by a human macrophage cell line. Further, we report that two clinically relevant GSK-3 inhibitors and COB-187 attenuate S and S2 protein-induced CXCL10 production. Combined, our observations provide impetus for investigating GSK-3 inhibitors as potential therapeutics for severe COVID-19.


Assuntos
Tratamento Farmacológico da COVID-19 , Quinase 3 da Glicogênio Sintase , Citocinas/metabolismo , Humanos , SARS-CoV-2 , Glicoproteína da Espícula de Coronavírus
3.
Exp Cell Res ; 323(1): 7-27, 2014 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-24569142

RESUMO

Dendritic cells (DCs) are immune cells found in the peripheral tissues where they sample the organism for infections or malignancies. There they take up antigens and migrate towards immunological organs to contact and activate T lymphocytes that specifically recognize the antigen presented by these antigen presenting cells. In the steady state there are several types of resident DCs present in various different organs. For example, in the mouse, splenic DC populations characterized by the co-expression of CD11c and CD8 surface markers are specialized in cross-presentation to CD8 T cells, while CD11c/SIRP-1α DCs seem to be dedicated to activating CD4 T cells. On the other hand, DCs have also been associated with the development of various diseases such as cancer, atherosclerosis, or inflammatory conditions. In such disease, DCs can participate by inducing angiogenesis or immunosuppression (tumors), promoting autoimmune responses, or exacerbating inflammation (atherosclerosis). This change in DC biology can be prompted by signals in the microenvironment. We have previously shown that the interaction of DCs with various extracellular matrix components modifies the immune properties and angiogenic potential of these cells. Building on those studies, herewith we analyzed the angiogenic profile of murine myeloid DCs upon interaction with 2D and 3D type-I collagen environments. As determined by PCR array technology and quantitative PCR analysis we observed that interaction with these collagen environments induced the expression of particular angiogenic molecules. In addition, DCs cultured on collagen environments specifically upregulated the expression of CXCL-1 and -2 chemokines. We were also able to establish DC cultures on type-IV collagen environments, a collagen type expressed in pathological conditions such as atherosclerosis. When we examined DC populations in atherosclerotic veins of Apolipoprotein E deficient mice we observed that they expressed adhesion molecules capable of interacting with collagen. Finally, to further investigate the interaction of DCs with collagen in other pathological conditions, we determined that both murine ovarian and breast cancer cells express several collagen molecules that can contribute to shape their particular tumor microenvironment. Consistently, tumor-associated DCs were shown to express adhesion molecules capable of interacting with collagen molecules as determined by flow cytometry analysis. Of particular relevance, tumor-associated DCs expressed high levels of CD305/LAIR-1, an immunosuppressive receptor. This suggests that signaling through this molecule upon interaction with collagen produced by tumor cells might help define the poorly immunogenic status of these cells in the tumor microenvironment. Overall, these studies demonstrate that through interaction with collagen proteins, DCs can be capable of modifying the microenvironments of inflammatory disease such as cancer or atherosclerosis.


Assuntos
Aterosclerose/metabolismo , Neoplasias da Mama/metabolismo , Células Dendríticas/metabolismo , Neoplasias Ovarianas/metabolismo , Receptores de Colágeno/metabolismo , Animais , Apolipoproteínas E/genética , Aterosclerose/imunologia , Neoplasias da Mama/imunologia , Antígeno CD11c/metabolismo , Moléculas de Adesão Celular/metabolismo , Técnicas de Cultura de Células , Linhagem Celular Tumoral , Proliferação de Células , Quimiocina CXCL1/biossíntese , Quimiocina CXCL2/biossíntese , Quimiotaxia , Colágeno/metabolismo , Feminino , Integrina alfa1beta1/biossíntese , Integrina alfa1beta1/metabolismo , Integrina alfa2beta1/biossíntese , Integrina alfa2beta1/metabolismo , Integrina alfa3beta1/biossíntese , Integrina alfa3beta1/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/metabolismo , Neovascularização Fisiológica , Neoplasias Ovarianas/imunologia , Receptores de Colágeno/biossíntese , Receptores Imunológicos/biossíntese , Receptores Imunológicos/metabolismo , Receptores Depuradores Classe A/biossíntese , Receptores Depuradores Classe A/metabolismo , Microambiente Tumoral , Regulação para Cima
4.
Immunobiology ; 218(1): 64-75, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22551928

RESUMO

Dendritic cells (DCs) are antigen presenting cells capable of inducing specific immune responses against microbial infections, transplant antigens, or tumors. DCs have been shown to possess a high plasticity showing different phenotypes in response to their microenvironment. For example, tumor-associated DCs can acquire an angiogenic phenotype thus promoting tumor growth. Further, DCs cultured in vitro under different conditions are able to upregulate the expression of endothelial markers and to express angiogenic factors. Indeed, it has been shown that soluble factors such as VEGF of PGE-2, that are present in the microenvironment of several tumors, affect the biology of these cells. We hypothesize that in addition to soluble factors the adhesion to different substrates will also define the phenotype and function of DCs. Herewith we demonstrate that murine myeloid(m) DCs upregulate endothelial markers such as VE-Cadherin, and to a lesser extent TIE-2, and decrease their immune capabilities when cultured on solid surfaces as compared with the same cells cultured on ultra-low binding (ULB) surfaces. On the other hand, the expression of angiogenic molecules at the level of RNA was not different among these cultures. In order to further investigate this phenomenon we used the murine ID8 model of ovarian cancer which can generate solid tumors when cancer cells are injected subcutaneously or a malignant ascites when they are injected intraperitoneally. This model gave us the unique opportunity to investigate DCs in suspension or attached to solid surfaces under the influence of the same tumor cells. We were able to determine that DCs present in solid tumors showed higher levels of expression of endothelial markers and angiogenic molecules but were not able to respond to inflammatory stimuli at the same extent as DCs recovered from ascites. Moreover, mDCs cultured on ULB surfaces in the presence of tumor factors do not expressed endothelial markers. Taking into account all these data we consider that tumor factors might be responsible for inducing angiogenic properties in DCs, but that in some settings the expression of endothelial markers such as VE-Cadherin and TIE-2 might be a function of attachment to solid surfaces and independent of the angiogenic properties of these cells.


Assuntos
Células Dendríticas/imunologia , Endotélio/metabolismo , Neoplasias Ovarianas/imunologia , Animais , Antígenos CD/genética , Antígenos CD/metabolismo , Caderinas/genética , Caderinas/metabolismo , Adesão Celular/imunologia , Diferenciação Celular , Linhagem Celular Tumoral , Endotélio/imunologia , Feminino , Humanos , Interleucina-6/genética , Interleucina-6/metabolismo , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Receptor TIE-2/genética , Receptor TIE-2/metabolismo , Microambiente Tumoral , Regulação para Cima , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
5.
J Virol ; 81(8): 3769-77, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17267484

RESUMO

Mouse mammary tumor virus (MMTV) is a milk-transmitted betaretrovirus that causes mammary tumors in mice. Although mammary epithelial cells are the ultimate targets of MMTV, the virus utilizes components of the host immune system to establish infection. Previous studies indicated that dendritic cells play a role in MMTV infection. Here we show that dendritic cells are the first cells to be infected by MMTV in vivo and that they are capable of producing infectious virus that can be transmitted to other cell types. Moreover, upon contact with the virus, dendritic cells became more mature and migrated in response to the chemokine macrophage inflammatory protein 3beta. Finally, we demonstrate that targeted ablation of dendritic cells in vivo dramatically attenuated MMTV infection. These data indicate that MMTV infection of dendritic cells is critical to initial propagation of the virus in vivo.


Assuntos
Células Dendríticas/virologia , Vírus do Tumor Mamário do Camundongo/fisiologia , Infecções por Retroviridae/virologia , Infecções Tumorais por Vírus/virologia , Animais , Movimento Celular , Células Dendríticas/fisiologia , Linfonodos/virologia , Proteínas Inflamatórias de Macrófagos/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Camundongos Transgênicos
6.
Nat Med ; 10(9): 950-8, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15334073

RESUMO

The involvement of immune mechanisms in tumor angiogenesis is unclear. Here we describe a new mechanism of tumor vasculogenesis mediated by dendritic cell (DC) precursors through the cooperation of beta-defensins and vascular endothelial growth factor-A (Vegf-A). Expression of mouse beta-defensin-29 recruited DC precursors to tumors and enhanced tumor vascularization and growth in the presence of increased Vegf-A expression. A new leukocyte population expressing DC and endothelial markers was uncovered in mouse and human ovarian carcinomas coexpressing Vegf-A and beta-defensins. Tumor-infiltrating DCs migrated to tumor vessels and independently assembled neovasculature in vivo. Bone marrow-derived DCs underwent endothelial-like differentiation ex vivo, migrated to blood vessels and promoted the growth of tumors expressing high levels of Vegf-A. We show that beta-defensins and Vegf-A cooperate to promote tumor vasculogenesis by carrying out distinct tasks: beta-defensins chemoattract DC precursors through CCR6, whereas Vegf-A primarily induces their endothelial-like specialization and migration to vessels, which is mediated by Vegf receptor-2.


Assuntos
Indutores da Angiogênese/imunologia , Células Dendríticas/imunologia , Neovascularização Patológica/imunologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , beta-Defensinas/imunologia , Animais , Diferenciação Celular/imunologia , Movimento Celular/imunologia , Colágeno , Células Dendríticas/metabolismo , Combinação de Medicamentos , Citometria de Fluxo , Técnicas Histológicas , Immunoblotting , Laminina , Camundongos , Proteoglicanas , Receptores de Quimiocinas/metabolismo , beta-Defensinas/metabolismo
7.
Virology ; 309(1): 75-84, 2003 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-12726728

RESUMO

Here we study the role of nitric oxide in the vaginal infection of Balb/c mice with herpes simplex virus type 2. Inducible nitric oxide synthase (iNOS) mRNA was detected by RT-PCR in vaginal tissue and inguinal lymph nodes early postinfection. iNOS was also found to be activated in cells recovered from vaginal washings of infected animals. Animals treated with aminoguanidine (AG), an iNOS inhibitor, showed a dose-dependent increase in vaginal pathology after viral infection compared to controls. Viral titers in vaginal washings and vaginas were higher in AG-treated mice. Treated animals presented higher PMN counts in vaginal washings compared to controls. Histopathology studies revealed a profound inflammatory exudate in vaginal tissue of treated animals. Finally, RT-PCR analysis showed increased expression of the chemokines MIP-2 and RANTES in vaginal tissue and inguinal lymph nodes of these animals.


Assuntos
Herpes Genital/fisiopatologia , Herpesvirus Humano 2/patogenicidade , Óxido Nítrico Sintase/genética , Óxido Nítrico/fisiologia , Animais , Sequência de Bases , Divisão Celular/efeitos dos fármacos , Quimiocina CCL5/genética , Quimiocina CXCL2 , Quimiocinas/genética , Chlorocebus aethiops , Primers do DNA , Feminino , Herpes Genital/imunologia , Herpesvirus Humano 2/fisiologia , Linfonodos/enzimologia , Linfonodos/virologia , Camundongos , Camundongos Endogâmicos BALB C , Doadores de Óxido Nítrico/farmacologia , Óxido Nítrico Sintase Tipo II , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , S-Nitroso-N-Acetilpenicilamina/farmacologia , Vagina/enzimologia , Vagina/virologia , Células Vero , Replicação Viral
8.
Curr Gene Ther ; 3(2): 113-25, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12653405

RESUMO

The biological therapy of tumors using live viruses was first proposed a century ago but was abandoned due to potential virulence of wild-type strains. Thanks to advances in recombinant technology, replication-restricted strains have been genetically engineered, which replicate selectively within tumor cells. Examples include replication-competent mutants of herpes simplex virus (HSV), adenovirus, vesicular stomatitis virus, reovirus and measles virus. Replication-restricted oncolytic viruses are able to propagate selectively within solid tumor nodules exerting direct antitumor activity by killing infected tumor cells at the completion of a replicative cycle. In the process, they generate an intratumoral inflammatory response, which under the appropriate circumstances, may trigger the activation of an adaptive antitumor immune response, a process that has been named in situ tumor vaccination. Recombinant HSV may offer distinct advantages in oncolytic therapy of epithelial tumors. HSV is highly infectious to tumors of epithelial origin, resulting in high efficacy, there is considerable redundancy in HSV receptors, which makes the loss of HSV receptors by tumors due to mutations less likely and potent anti-herpetic drugs are commercially available, which may be used clinically to control undesired side effects. Herewith we describe the use of oncolytic viral therapy against intraperitoneal malignancies with special emphasis on oncolytic herpes simplex virus. We review the preclinical evidence on the efficacy and safety of intraperitoneal applications of HSV and discuss the rationale for its use for oncolytic therapy and in situ tumor vaccination of intraperitoneal tumors.


Assuntos
Vacinas Anticâncer , Terapia Genética/métodos , Neoplasias Epiteliais e Glandulares/terapia , Neoplasias/terapia , Simplexvirus/genética , Animais , Citocinas/metabolismo , Células Dendríticas/virologia , Humanos , Inflamação , Camundongos , Mutação , Neoplasias Peritoneais/terapia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA