Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Int J Mol Sci ; 22(7)2021 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-33805189

RESUMO

Exposure to particulate matter (PM) is becoming a major global health issue. The amount and time of exposure to PM are known to be closely associated with cardiovascular diseases. However, the mechanism through which PM affects the vascular system is still not clear. Endothelial cells line the interior surface of blood vessels and actively interact with plasma proteins, including the complement system. Unregulated complement activation caused by invaders, such as pollutants, may promote endothelial inflammation. In the present study, we sought to investigate whether urban PM (UPM) acts on the endothelial environment via the complement system. UPM-treated human endothelial cells with normal human serum showed the deposition of membrane attack complexes (MACs) on the cell surface via the alternative pathway of the complement system. Despite the formation of MACs, cell death was not observed, and cell proliferation was increased in UPM-mediated complement activation. Furthermore, complement activation on endothelial cells stimulated the production of inflammation-related proteins. Our results revealed that UPM could activate the complement system in human endothelial cells and that complement activation regulated inflammatory reaction in microenvironment. These findings provide clues with regard to the role of the complement system in pathophysiologic events of vascular disease elicited by air pollution.


Assuntos
Poluentes Atmosféricos/efeitos adversos , Ativação do Complemento , Proteínas do Sistema Complemento/metabolismo , Células Endoteliais/metabolismo , Inflamação/metabolismo , Vasos Sanguíneos/patologia , Morte Celular , Proliferação de Células , Sobrevivência Celular , Células Cultivadas , Citocinas/metabolismo , Células Endoteliais da Veia Umbilical Humana , Humanos , Material Particulado/efeitos adversos , Fosforilação , Espécies Reativas de Oxigênio/metabolismo , Fator de Transcrição STAT3/metabolismo
2.
Int J Mol Sci ; 21(2)2020 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-31963761

RESUMO

The demyelinating diseases of the central nervous system involve myelin abnormalities, oligodendrocyte damage, and consequent glia activation. Neurotoxicant cuprizone (CPZ) was used to establish a mouse model of demyelination. However, the effects of CPZ on microRNA (miRNA) expression and behavior have not been clearly reported. We analyzed the behavior of mice administered a diet containing 0.2% CPZ for 6 weeks, followed by 6 weeks of recovery. Rotarod analysis demonstrated that the treated group had poorer motor coordination than control animals. This effect was reversed after 6 weeks of CPZ withdrawal. Open-field tests showed that CPZ-treated mice exhibited significantly increased anxiety and decreased exploratory behavior. CPZ-induced demyelination was observed to be alleviated after 4 weeks of CPZ treatment, according to luxol fast blue (LFB) staining and myelin basic protein (MBP) expression. miRNA expression profiling showed that the expression of 240 miRNAs was significantly changed in CPZ-fed mice compared with controls. Furthermore, miR-155-5p and miR-20a-5p upregulations enhanced NgR induction through Smad 2 and Smad 4 suppression in demyelination. Taken together, our results demonstrate that CPZ-mediated demyelination induces behavioral deficits with apparent alterations in miRNA expression, suggesting that differences in miRNA expression in vivo may be new potential therapeutic targets for remyelination.


Assuntos
Cuprizona/efeitos adversos , Doenças Desmielinizantes/psicologia , Comportamento Exploratório/efeitos dos fármacos , Redes Reguladoras de Genes/efeitos dos fármacos , Animais , Doenças Desmielinizantes/induzido quimicamente , Doenças Desmielinizantes/genética , Modelos Animais de Doenças , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/efeitos dos fármacos , MicroRNAs/genética , Teste de Desempenho do Rota-Rod
3.
Tumour Biol ; 37(2): 2587-96, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26392109

RESUMO

Existing evidence suggests a possible role of viruses in human bladder cancer development. Recently, Kaposi's sarcoma-associated herpesvirus (KSHV) was reported to be the most frequently detected virus in bladder cancer tissue from Croatian patients on screening with the Lawrence Livermore Microbial Detection Array. In the current study, to investigate the functional roles of KSHV in bladder cancer, five bladder cancer cell lines were infected with KSHV and their tumour progression-associated changes investigated. Four KSHV-infected bladder cancer cell lines were established; two invasive bladder cancer cell lines showed higher proliferation rates than uninfected cells. Additionally, these KSHV-infected invasive bladder cancer cells showed a greater number of colonies, which were also significantly larger than those of uninfected cells, in a soft agar colony formation assay. cDNA microarray analysis showed that various genes associated with cell proliferation and cancer development were upregulated in these KSHV-infected bladder cancer cells. Taken together, we suggest that KSHV infection affects the proliferation of a subset of invasive bladder cancer cells and may therefore play a role in their oncogenic progression. Further studies are required to elucidate the exact mechanism used by KSHV to promote bladder cancer progression.


Assuntos
Proliferação de Células/fisiologia , Infecções por Herpesviridae/patologia , Infecções por Herpesviridae/virologia , Herpesvirus Humano 8/patogenicidade , Sarcoma de Kaposi/virologia , Neoplasias da Bexiga Urinária/patologia , Neoplasias da Bexiga Urinária/virologia , Linhagem Celular , Células Endoteliais da Veia Umbilical Humana , Humanos , Células Tumorais Cultivadas
4.
J Extracell Vesicles ; 13(9): e12495, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39254228

RESUMO

Mesenchymal stem cell (MSC)-derived extracellular vesicles (EVs) have shown anti-inflammatory potential in multiple inflammatory diseases. In the March 2022 issue of the Journal of Extracellular Vesicles, it was shown that EVs from human MSCs can suppress severe acute respiratory distress syndrome, coronavirus 2 (SARS-CoV-2) replication and can mitigate the production and release of infectious virions. We therefore hypothesized that MSC-EVs have an anti-viral effect in SARS-CoV-2 infection in vivo. We extended this question to ask whether also other respiratory viral infections could be treated by MSC-EVs. Adipose stem cell-derived EVs (ASC-EVs) were isolated using tangential flow filtration from conditioned media obtained from a multi-flask cell culture system. The effects of the ASC-EVs were tested  in Vero E6 cells in vitro. ASC-EVs were also given i.v. to SARS-CoV-2 infected Syrian Hamsters, and H1N1 influenza virus infected mice. The ASC-EVs attenuated SARS-CoV-2 virus replication in Vero E6 cells and reduced body weight and signs of lung injury in infected Syrian hamsters. Furthermore, ASC-EVs increased the survival rate of influenza A-infected mice and attenuated signs of lung injury. In summary, this study suggests that ASC-EVs can have beneficial therapeutic effects in models of virus-infection-associated acute lung injury and may potentially be developed to treat lung injury in humans.


Assuntos
Lesão Pulmonar Aguda , COVID-19 , Vesículas Extracelulares , Vírus da Influenza A Subtipo H1N1 , Células-Tronco Mesenquimais , SARS-CoV-2 , Animais , Vesículas Extracelulares/metabolismo , Células-Tronco Mesenquimais/metabolismo , SARS-CoV-2/fisiologia , COVID-19/terapia , Lesão Pulmonar Aguda/terapia , Lesão Pulmonar Aguda/virologia , Camundongos , Células Vero , Humanos , Chlorocebus aethiops , Infecções por Orthomyxoviridae/terapia , Replicação Viral , Mesocricetus , Modelos Animais de Doenças , Masculino , Influenza Humana/terapia , Feminino
5.
J Microbiol ; 59(5): 522-529, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33877577

RESUMO

Extracellular vesicles (EVs) play a crucial role in cell-to-cell communication. EVs and viruses share several properties related to their structure and the biogenesis machinery in cells. EVs from virus-infected cells play a key role in virus spread and suppression using various loading molecules, such as viral proteins, host proteins, and microRNAs. However, it remains unclear how and why viruses regulate EV production inside host cells. The purpose of this study is to investigate the molecular mechanisms underlying EV production and their roles in Kaposi's sarcoma-associated herpesvirus (KSHV)-infected cells. Here, we found that KSHV induced EV production in human endothelial cells via Rab-27b upregulation. The suppression of Rab27b expression in KSHV-infected cells enhanced cell death by increasing autophagic flux and autolysosome formation. Our results indicate that Rab27b regulates EV biogenesis to promote cell survival and persistent viral infection during KSHV infection, thereby providing novel insights into the crucial role of Rab-27b in the KSHV life cycle.


Assuntos
Vesículas Extracelulares/metabolismo , Infecções por Herpesviridae/metabolismo , Herpesvirus Humano 8 , Proteínas rab de Ligação ao GTP/metabolismo , Autofagia , Morte Celular , Sobrevivência Celular , Células Endoteliais/virologia , Infecções por Herpesviridae/virologia , Herpesvirus Humano 8/genética , Humanos , MicroRNAs/metabolismo , Nanopartículas , Regulação para Cima , Proteínas Virais/metabolismo
6.
PLoS One ; 15(7): e0235793, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32634162

RESUMO

Extracellular vesicles (EVs) are small vesicles secreted from cells. They have crucial biological functions in intercellular communications and may even be biomarkers for cancer. The various methods used to isolate EVs from body fluid and cell culture supernatant have been compared in prior studies, which determined that the component yield and physical properties of isolated EVs depend largely on the isolation method used. Several novel and combined methods have been recently developed, which have not yet been compared to the established methods. Therefore, the purpose of this study is to compare the physical and functional differences in EVs isolated using a differential centrifugation method, the precipitation-based Invitrogen kit, the ExoLutE kit, and the Exodisc, of which the latter two were recently developed. We investigated the properties of EVs isolated from non-infected and Kaposi's sarcoma-associated herpesvirus-infected human umbilical vein endothelial cells using each method and determined the yields of DNA, RNA, and proteins using quantitative polymerase chain reaction and bicinchoninic acid assays. Additionally, we determined whether the biological activity of EVs correlated with the quantity or physical properties of the EVs isolated using different methods. We found that Exodisc was the most suitable method for obtaining large quantities of EVs, which might be useful for biomarker investigations, and that the EVs separated using Exodisc exhibited the highest complement activation activity. However, we also found that the functional properties of EVs were best maintained when differential centrifugation was used. Effective isolation is necessary to study EVs as tools for diagnosing cancer and our findings may have relevant implications in the field of oncology by providing researchers with data to assist their selection of a suitable isolation method.


Assuntos
Fracionamento Celular/métodos , Células Endoteliais/química , Vesículas Extracelulares/química , Biomarcadores/análise , Centrifugação/métodos , Precipitação Química , DNA/análise , Células Endoteliais/virologia , Vesículas Extracelulares/virologia , Infecções por Herpesviridae/virologia , Herpesvirus Humano 8/isolamento & purificação , Células Endoteliais da Veia Umbilical Humana , Humanos , Proteínas/análise , RNA/análise
7.
Front Immunol ; 10: 876, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31068945

RESUMO

Kaposi's Sarcoma-associated herpesvirus (KSHV) is the etiologic agent of Kaposi's sarcoma, which is the most common cancer in acquired immune deficiency syndrome patients. KSHV contains a variety of immunoregulatory proteins. There have been many studies on the modulation of antiviral response by these immunoregulatory proteins of KSHV. However, the antiviral effects of extracellular vesicles (EVs) during de novo KSHV infection have not been investigated to our best knowledge. In this study, we showed that KSHV-infected cells induce interferon-stimulated genes (ISGs) response but not type I interferon in uninfected bystander cells using EVs. mRNA microarray analysis showed that ISGs and IRF-activating genes were prominently activated in EVs from KSHV-infected cells (KSHV EVs)-treated human endothelial cells, which were validated by RT-qPCR and western blot analysis. We also found that this response was not associated with cell death or apoptosis by virus infection. Mechanistically, the cGAS-STING pathway was linked with these KSHV EVs-mediated ISGs expressions, and mitochondrial DNA on the surface of KSHV EVs was one of the causative factors. Besides, KSHV EVs-treated cells showed lower infectivity for KSHV and viral replication activity than mock EVs-treated cells. Our results indicate that EVs from KSHV-infected cells could be an initiating factor for the innate immune response against viral infection, which may be critical to understanding the microenvironment of virus-infected cells.


Assuntos
DNA Mitocondrial , Vesículas Extracelulares/metabolismo , Infecções por Herpesviridae/etiologia , Infecções por Herpesviridae/metabolismo , Herpesvirus Humano 8/fisiologia , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Animais , Transporte Biológico , Linhagem Celular , Chlorocebus aethiops , Biologia Computacional/métodos , Células Epiteliais/metabolismo , Células Epiteliais/virologia , Perfilação da Expressão Gênica , Infecções por Herpesviridae/patologia , Humanos , Transcriptoma , Células Vero
8.
Exp Mol Med ; 51(10): 1-15, 2019 10 24.
Artigo em Inglês | MEDLINE | ID: mdl-31649250

RESUMO

Nogo receptor (NgR) has been shown to inhibit the migration and invasion of human glioma cells. However, little is known regarding the regulatory mechanisms of NgR in glioblastoma multiforme (GBM). In this study, we propose a novel mechanism that regulates the maturation process of NgR through an interaction with vimentin. The inhibition of TGFß1 activity by LY2109761 attenuated the migration/invasion of GBM cells by upregulating cell-surface NgR. Conversely, the treatment of GBM cells with TGFß1 suppressed NgR maturation. We showed that NgR and vimentin interact, which could be a possible mechanism for the suppression of NgR maturation. The knockdown of vimentin suppressed the migration/invasion of GBM cells through the increased maturation of NgR. Finally, TCGA (The Cancer Genome Atlas) analysis also supported the association of NgR and vimentin. The maturation of NgR is regulated by the interaction of vimentin and NgR, which attenuates the invasive activity of GBM, and might be a potential therapeutic target for brain cancer.


Assuntos
Proteínas da Matriz Extracelular/genética , Glioblastoma/genética , Receptor Nogo 1/genética , Fator de Crescimento Transformador beta/genética , Vimentina/genética , Biomarcadores Tumorais/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica/genética , Glioblastoma/patologia , Humanos , Invasividade Neoplásica/genética
9.
In Vitro Cell Dev Biol Anim ; 54(6): 423-429, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29748909

RESUMO

Extracellular vesicles (EVs) are mediators of intercellular communication by transporting cargo containing proteins, lipids, mRNA, and miRNA. There is increasing evidence that EVs have various roles in regulating migration, invasion, stemness, survival, and immune functions. Previously, we have found that EVs from Kaposi's sarcoma-associated herpesvirus (KSHV)-infected human endothelial cells have the potential to activate the complement system. Although many studies have shown that the physical properties of EVs can be changed by their storage condition, there have been few studies for the stability of biological activity of EVs in various storage conditions. In this study, we investigated various conditions to identify the best conditions to store EVs with functional stability for 25 d. Furthermore, the correlation between the function and other characteristics of EVs, including the expression of EV markers, size distribution, and particle number, were also analyzed. Our results demonstrated that storage temperature is an important factor to maintain the activity of EVs and would be useful information for basic research and clinical application using EVs.


Assuntos
Vesículas Extracelulares/fisiologia , Manejo de Espécimes/métodos , Biomarcadores/metabolismo , Herpesvirus Humano 8 , Células Endoteliais da Veia Umbilical Humana , Humanos , Nanopartículas , Temperatura , Tetraspanina 28/metabolismo , Tetraspanina 30/metabolismo , Fatores de Tempo
10.
Sci Rep ; 8(1): 5415, 2018 04 03.
Artigo em Inglês | MEDLINE | ID: mdl-29615744

RESUMO

There is increasing evidence that the complement system is activated in various cancer tissues. Besides being involved in innate immunity against pathogens, the complement system also participates in inflammation and the modulation of tumor microenvironment. Recent studies suggest that complement activation promotes tumor progression in various ways. Among some cancer cell lines, we found that human bone osteosarcoma epithelial cells (U2-OS) can activate the alternative pathway of the complement system by pooled normal human serum. Interestingly, U2-OS cells showed less expression of complement regulatory proteins, compared to other cancer cell lines. Furthermore, the activated complement system enhanced the production of growth factors, which promoted angiogenesis of human endothelial cells. Our results demonstrated a direct linkage between the complement system and angiogenesis using the in vitro model, which suggest the complement system and related mechanisms might be potential targets for cancer treatment.


Assuntos
Neoplasias Ósseas/patologia , Proteínas do Sistema Complemento/metabolismo , Fator 1 de Crescimento de Fibroblastos/metabolismo , Neovascularização Patológica/metabolismo , Osteossarcoma/patologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Neoplasias Ósseas/irrigação sanguínea , Neoplasias Ósseas/metabolismo , Linhagem Celular Tumoral , Células Endoteliais/patologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Osteossarcoma/irrigação sanguínea , Osteossarcoma/metabolismo , Fosforilação
11.
J Microbiol Biotechnol ; 28(1): 165-174, 2018 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-29032648

RESUMO

Glioblastoma multiforme is the most lethal malignant brain tumor. Despite many intensive studies, the prognosis of glioblastoma multiforme is currently very poor, with a median overall survival duration of 14 months and 2-year survival rates of less than 10%. Although viral infections have been emphasized as potential cofactors, their influences on pathways that support glioblastoma progression are not known. Some previous studies indicated that human Kaposi's sarcoma-associated herpesvirus (KSHV) was detected in healthy brains, and its microRNA was also detected in glioblastoma patients' plasma. However, a direct link between KSHV infection and glioblastoma is currently not known. In this study, we infected glioblastoma cells and glioma stem-like cells (GSCs) with KSHV to establish an in vitro cell model for KSHV-infected glioblastoma cells and glioma stem-like cells in order to identify virologic outcomes that overlap with markers of aggressive disease. Latently KSHV-infected glioblastoma cells and GSCs were successfully established. Additionally, using these cell models, we found that KSHV infection modulates the proliferation of glioma stem-like cells.


Assuntos
Proliferação de Células , Glioma/virologia , Herpesvirus Humano 8/crescimento & desenvolvimento , Células-Tronco/fisiologia , Células-Tronco/virologia , Células Tumorais Cultivadas/fisiologia , Células Tumorais Cultivadas/virologia , Células Cultivadas , Humanos
12.
Oncotarget ; 8(59): 99841-99860, 2017 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-29245944

RESUMO

Extracellular vesicles (EVs), released by cells, are associated with cell-to-cell communication and regulate various cellular processes. EVs draw parallels with viruses for their similar structures and functions. Increasing evidences from recent studies indicate that cells infected with viruses release a variety of EVs. Delineating the functions and mechanisms of EVs released during virus infection is essential for understanding the molecular basis of viral infection and replication as well as associated pathogenesis. The most challenging obstacle for these studies is the separation of EVs from viruses. In this study, we successfully isolated the EVs from de novo Kaposi's sarcoma-associated herpesvirus (KSHV) infected-human endothelial cells during the period between virus entry and production. Intriguingly, a proteomics analysis of these EVs has revealed alterations of the complement system. Additionally, we have discovered that the EVs from KSHV-infected endothelial cells are potent activators of an alternative pathway of the complement system via exploitation of the endogenous C3 complement protein and properdin. Furthermore, we have found that complement activation promotes KSHV persistent latent infection by activating the NF-κB pathway, which enhances the survival of KSHV-infected cells and inhibits viral lytic replication. Our work identifies a novel role of EVs induced by KSHV during de novo infection and the underlying mechanism of complement activation by EVs.

13.
J Microbiol Biotechnol ; 26(3): 618-26, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26699745

RESUMO

Pleural effusion lymphoma (PEL) is a rare B-cell lymphoma that has a very poor prognosis with a median survival time of around 6 months. PEL is caused by Kaposi's sarcoma-associated herpesvirus, and is often co-infected with the Epstein Barr virus. The complement system is fundamental in the innate immune system against pathogen invasion and tumor development. In the present study, we investigated the activation of the complement system in PEL cells using human serum complements. Interestingly, two widely used PEL cell lines, BCP-1 and BCBL-1, showed different susceptibility to the complement system, which may be due to CD46 expression on their cell membranes. Complement activation did not induce apoptosis but supported cell survival considerably. Our results demonstrated the susceptibility of PEL to the complement system and its underlying mechanisms, which would provide insight into understanding the pathogenesis of PEL.


Assuntos
Proteínas do Sistema Complemento/imunologia , Herpesvirus Humano 8/fisiologia , Linfoma/imunologia , Derrame Pleural Maligno/imunologia , Linhagem Celular , Herpesvirus Humano 8/imunologia , Humanos , Linfoma/virologia , Proteína Cofatora de Membrana/imunologia , Derrame Pleural Maligno/virologia
14.
J Microbiol ; 54(11): 782-788, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27796928

RESUMO

Kaposi's sarcoma-associated herpesvirus (KSHV) is the major etiologic agent of Kaposi's sarcoma, primary effusion lymphoma, and multicentric Castleman's disease. Recent studies have indicated that KSHV can be detected at high frequency in patient-derived bladder cancer tissue and might be associated with the pathogenesis of bladder cancer. Bladder cancer is the second most common cancer of the genitourinary tract, and it has a high rate of recurrence. Because drug resistance is closely related to chemotherapy failure and cancer recurrence, we investigated whether KSHV infection is associated with drug resistance of bladder cancer cells. Some KSHV-infected bladder cancer cell lines showed resistance to an anti-cancer drug, cisplatin, possibly as a result of down-regulation of reactive oxygen species. Additionally, drug resistance acquired from KSHV infection could partly be overcome by HDAC1 inhibitors. Taken together, the data suggest the possible role of KSHV in chemo-resistant bladder cancer, and indicate the therapeutic potential of HDAC1 inhibitors in drug-resistant bladder cancers associated with KSHV infection.


Assuntos
Resistencia a Medicamentos Antineoplásicos , Herpesvirus Humano 8/fisiologia , Espécies Reativas de Oxigênio , Sarcoma de Kaposi/virologia , Neoplasias da Bexiga Urinária/virologia , Linhagem Celular Tumoral , Cisplatino/farmacologia , Regulação para Baixo , Infecções por Herpesviridae/metabolismo , Infecções por Herpesviridae/virologia , Herpesvirus Humano 8/genética , Histona Desacetilase 1 , Inibidores de Histona Desacetilases/farmacologia , Humanos , Análise Serial de Proteínas , Espécies Reativas de Oxigênio/metabolismo , Sarcoma de Kaposi/complicações
15.
mBio ; 7(1): e02109-15, 2016 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-26814175

RESUMO

UNLABELLED: Kaposi's sarcoma (KS), a highly angiogenic and invasive tumor often involving different organ sites, including the oral cavity, is caused by infection with Kaposi's sarcoma-associated herpesvirus (KSHV). Diverse cell markers have been identified on KS tumor cells, but their origin remains an enigma. We previously showed that KSHV could efficiently infect, transform, and reprogram rat primary mesenchymal stem cells (MSCs) into KS-like tumor cells. In this study, we showed that human primary MSCs derived from diverse organs, including bone marrow (MSCbm), adipose tissue (MSCa), dental pulp, gingiva tissue (GMSC), and exfoliated deciduous teeth, were permissive to KSHV infection. We successfully established long-term cultures of KSHV-infected MSCa, MSCbm, and GMSC (LTC-KMSCs). While LTC-KMSCs had lower proliferation rates than the uninfected cells, they expressed mixtures of KS markers and displayed differential angiogenic, invasive, and transforming phenotypes. Genetic analysis identified KSHV-derived microRNAs that mediated KSHV-induced angiogenic activity by activating the AKT pathway. These results indicated that human MSCs could be the KSHV target cells in vivo and established valid models for delineating the mechanism of KSHV infection, replication, and malignant transformation in biologically relevant cell types. IMPORTANCE: Kaposi's sarcoma is the most common cancer in AIDS patients. While KSHV infection is required for the development of Kaposi's sarcoma, the origin of KSHV target cells remains unclear. We show that KSHV can efficiently infect human primary mesenchymal stem cells of diverse origins and reprogram them to acquire various degrees of Kaposi's sarcoma-like cell makers and angiogenic, invasive, and transforming phenotypes. These results indicate that human mesenchymal stem cells might be the KSHV target cells and establish models for delineating the mechanism of KSHV-induced malignant transformation.


Assuntos
Transformação Celular Neoplásica , Herpesvirus Humano 8/crescimento & desenvolvimento , Herpesvirus Humano 8/fisiologia , Células-Tronco Mesenquimais/virologia , Neovascularização Patológica , Proliferação de Células , Células Cultivadas , Regulação da Expressão Gênica , Interações Hospedeiro-Patógeno , Humanos , MicroRNAs/metabolismo , RNA Viral/metabolismo , Cultura de Vírus
16.
J Immunol Methods ; 415: 57-62, 2014 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-25260423

RESUMO

The complement system is an important aspect of immune defense against microbial invasion. Eukaryotic cells express various complement regulatory proteins to protect them from uncontrolled complement activation. However, some eukaryotic cells possess constitutive complement system activation that does not require specific triggering factors, which is known to have unexpected effects on cell proliferation and survival. This area of research is still preliminary and a standard method to measure complement system activation in eukaryotic cells has yet to be identified. Here, we present a quantitative in vitro method to measure complement system activation in eukaryotic cells by detecting C5b-9, the membrane attack complex, on cell surfaces. The results obtained using this assay correlated with C3b deposition measured using flow cytometry and C5b-9 deposition detected using an immunofluorescence assay. Furthermore, we showed that various cancer cell lines displayed different levels of complement system activation by using this assay.


Assuntos
Membrana Celular/química , Ativação do Complemento , Complexo de Ataque à Membrana do Sistema Complemento/análise , Ensaio de Imunoadsorção Enzimática/métodos , Anticorpos/farmacologia , Antígenos CD/genética , Antígenos CD/imunologia , Moléculas de Adesão Celular Neuronais/antagonistas & inibidores , Moléculas de Adesão Celular Neuronais/genética , Moléculas de Adesão Celular Neuronais/imunologia , Linhagem Celular Tumoral , Membrana Celular/imunologia , Complemento C3b/farmacologia , Endoglina , Proteínas Fetais/antagonistas & inibidores , Proteínas Fetais/genética , Proteínas Fetais/imunologia , Citometria de Fluxo , Expressão Gênica , Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/imunologia , Humanos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/imunologia , Especificidade de Órgãos , Receptores de Superfície Celular/antagonistas & inibidores , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA