Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Am J Pathol ; 180(1): 390-8, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22067905

RESUMO

Exosomes participate in intercellular communication, but most data published are based on exosomes released from in vitro cultured cells that do not communicate with neighboring cells located in the same microenvironment as the exosomal-producing cells in vivo. In this study, our data show that co-culture of leukocytes isolated from breast tumor tissue leads to uptake of fibronectin (FN) on or in the tumor exosomes (Exo(fib+)). The induction of FN and exosomal uptake is tumor tissue derived and leukocyte specific, because leukocytes isolated from the peripheral blood of naïve mice failed to induce FN uptake by tumor exosomes. Furthermore, depletion of both CD25(+) cells and Gr-1(+) cells from tumor-associated leukocytes causes a reduction of Exo(fib+), suggesting that tumor-associated CD25(+) cells and Gr-1(+) cells participate in FN production and uptake by tumor exosomes, resulting in Exo(fib+). As a result of tumor cells absorbing Exo(fib+), two major events are induced: focal adhesion kinase/Src-dependent signaling pathways are activated, and the production of proinflammatory cytokines and metalloproteinase 9 is enhanced in response to absorbing exosomes. This, in turn, enhances tumor cell invasion in vitro and in vivo. Collectively, our findings provide evidence that exosomes released from freshly excised tumor tissue cells that have communicated/interacted with immune cells gain new immune evasion capacity.


Assuntos
Comunicação Celular/fisiologia , Neoplasias do Colo/fisiopatologia , Exossomos/fisiologia , Fibronectinas/metabolismo , Leucócitos/fisiologia , Animais , Neoplasias da Mama/fisiopatologia , Linhagem Celular Tumoral , Progressão da Doença , Feminino , Neoplasias Pulmonares/fisiopatologia , Neoplasias Pulmonares/secundário , Camundongos , Camundongos Endogâmicos BALB C , Invasividade Neoplásica/fisiopatologia , Metástase Neoplásica , Transplante de Neoplasias , Microambiente Tumoral/fisiologia
3.
Am J Pathol ; 176(5): 2490-9, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20348242

RESUMO

In this study we observed that mice pretreated with tumor exosomes had a significant acceleration of tumor metastasis in the lung. Tumor metastasis correlated significantly with an increase in recruitment of more Myeloid-derived suppressor cells (MDSCs) in the lung of C57BL/6j (B6) mice pretreated with tumor exosomes. These effects were blunted when MyD88 knockout (KO) mice were pretreated with tumor exosomes. MDSCs induced by tumor exosomes and isolated from wild-type B6 mice also more potently inhibited T cell activation and induction of interleukin-6 and tumor necrosis factor-alpha than MDSCs isolated from the lung of MyD88 KO mice. In vitro, addition of tumor exosomes to bone marrow-derived CD11b(+)Gr-1(+) cells isolated from wild-type B6 mice resulted in more cytokine production, including tumor necrosis factor-alpha, interleukin-6, and the chemokine CCL2, than CD11b(+)Gr-1(+) cells isolated from MyD88 KO mice. Moreover, lower levels of CCL2 were observed in the lungs in MyD88 KO mice pretreated with tumor exosomes than that in wild-type mice. Together these data demonstrate a pivotal role for MyD88 in tumor exosome-mediated expansion of MDSCs and tumor metastasis.


Assuntos
Exossomos/metabolismo , Células Mieloides/citologia , Fator 88 de Diferenciação Mieloide/metabolismo , Neoplasias/imunologia , Animais , Células da Medula Óssea/citologia , Quimiocina CCL2/metabolismo , Fibroblastos/metabolismo , Interleucina-6/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Metástase Neoplásica , Fator de Necrose Tumoral alfa/metabolismo
4.
Mol Ther ; 18(9): 1606-14, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20571541

RESUMO

Monocyte-derived myeloid cells play vital roles in inflammation-related autoimmune/inflammatory diseases and cancers. Here, we report that exosomes can deliver anti-inflammatory agents, such as curcumin, to activated myeloid cells in vivo. This technology provides a means for anti-inflammatory drugs, such as curcumin, to target the inflammatory cells as well as to overcome unwanted off-target effects that limit their utility. Using exosomes as a delivery vehicle, we provide evidence that curcumin delivered by exosomes is more stable and more highly concentrated in the blood. We show that the target specificity is determined by exosomes, and the improvement of curcumin activity is achieved by directing curcumin to inflammatory cells associated with therapeutic, but not toxic, effects. Furthermore, we validate the therapeutic relevance of this technique in a lipopolysaccharide (LPS)-induced septic shock mouse model. We further show that exosomes, but not lipid alone, are required for the enhanced anti-inflammatory activity of curcumin. The specificity of using exosomes as a drug carrier creates opportunities for treatments of many inflammation-related diseases without significant side effects due to innocent bystander or off-target effects.


Assuntos
Anti-Inflamatórios/administração & dosagem , Anti-Inflamatórios/uso terapêutico , Curcumina/administração & dosagem , Curcumina/uso terapêutico , Exossomos/química , Nanopartículas/uso terapêutico , Animais , Anti-Inflamatórios/química , Western Blotting , Linhagem Celular Tumoral , Células Cultivadas , Curcumina/química , Portadores de Fármacos/administração & dosagem , Portadores de Fármacos/uso terapêutico , Feminino , Citometria de Fluxo , Lipopolissacarídeos/toxicidade , Camundongos , Camundongos Endogâmicos C57BL , Nanopartículas/administração & dosagem , Choque Séptico/induzido quimicamente , Choque Séptico/tratamento farmacológico
5.
Am J Pathol ; 174(4): 1415-25, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19246649

RESUMO

Ubiquitinated endosomal proteins that are deposited into the lumens of multivesicular bodies are either sorted for lysosomal-mediated degradation or secreted as exosomes into the extracellular milieu. The mechanisms that underlie the sorting of cellular cargo proteins are currently unknown. In this study, we show that the COP9 signalosome (CSN)-associated protein CSN5 quantitatively regulated proteins that were sorted into exosomes. Western blot analysis of exosomal proteins indicated that small interfering (si)RNA knockdown of CSN5 results in increased levels of both ubiquitinated and non-ubiquitinated exosomal proteins, including heat shock protein 70, in comparison with exosomes isolated from the supernatants of 293 cells transfected with scrambled siRNA. Furthermore, 293 cells transfected with JAB1/MPN/Mov34 metalloenzyme domain-deleted CSN5 produced exosomes with higher levels of ubiquitinated heat shock protein 70, which did not affect non-ubiquitinated heat shock protein 70 levels. The loss of COP9-associated deubiquitin activity of CSN5 also led to the enhancement of HIV Gag that was sorted into exosomes as well as the promotion of HIV-1 release, suggesting that COP9-associated CSN5 regulates the sorting of a number of exosomal proteins in both a CSN5 JAB1/MPN/Mov34 metalloenzyme domain-dependent and -independent manner. We propose that COP9-associated CSN5 regulates exosomal protein sorting in both a deubiquitinating activity-dependent and -independent manner, which is contrary to the current idea of ubiquitin-dependent sorting of proteins to exosomes.


Assuntos
Exossomos/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Complexos Multiproteicos/metabolismo , Peptídeo Hidrolases/metabolismo , Proteínas/metabolismo , Ubiquitinação/fisiologia , Western Blotting , Complexo do Signalossomo COP9 , Proteína do Núcleo p24 do HIV/metabolismo , Infecções por HIV/metabolismo , HIV-1 , Proteínas de Choque Térmico HSP70/metabolismo , Humanos , Imunoprecipitação , Fragmentos de Peptídeos/metabolismo , Transporte Proteico/fisiologia , RNA Interferente Pequeno , Transfecção
6.
Hepatology ; 50(5): 1412-20, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19708080

RESUMO

UNLABELLED: Chronic inflammation plays a critical role in promoting obesity-related disorders, such as fatty liver disease. The inflammatory cells that mediate these effects remain unknown. This study investigated the accumulation of immature myeloid cells in the liver and their role in liver inflammation. We found that the accumulation of immature myeloid cells, i.e., CD11b(+)Ly6C(hi)Ly6G(-) cells, in the liver of B6 mice fed a high-fat diet contribute to liver inflammation. Adoptive transfer of CD11b(+)Ly6C(hi)Ly6G(-) cells isolated from the liver of obese B6 mice, but not from lean B6 mice, resulted in liver damage that was evident by an increase in the activity of liver transferases in serum. CD11b(+)Ly6C(hi)Ly6G(-) cells isolated from the liver of obese mice are more easily activated by way of Toll-like receptor (TLR) stimulation resulting in interleukin 12 and other inflammatory cytokine expression in an MyD88-dependent fashion. TLR7-activated CD11b(+)Ly6C(hi)Ly6G(-) cells also enhance liver natural killer T cell (NKT) death in an Fas-dependent manner. Experiments using mice depleted of Gr-1(+) immature myeloid cells demonstrated the important role of CD11b(+)Ly6C(hi)Ly6G(-) in liver inflammation. Repeated injection of exosome-like particles causes CD11b(+) cell activation and subsequent homing to and accumulation of the cells in the liver. CONCLUSION: Consumption of a high-fat diet by B6 mice triggers an accumulation of immature myeloid cells in the liver. The immature myeloid cells release proinflammatory cytokines and induce NKT cell apoptosis. Activation-induced NKT apoptosis further promotes excessive production of Th-1 cytokines. This diet-induced accumulation of immature myeloid cells may contribute to obesity-related liver disease.


Assuntos
Gorduras na Dieta/efeitos adversos , Hepatite/etiologia , Hepatite/patologia , Fígado/patologia , Células Mieloides/patologia , Obesidade/complicações , Animais , Apoptose/fisiologia , Antígeno CD11b/metabolismo , Proliferação de Células , Modelos Animais de Doenças , Hepatite/metabolismo , Interleucina-12/metabolismo , Células Matadoras Naturais/metabolismo , Células Matadoras Naturais/patologia , Fígado/metabolismo , Camundongos , Camundongos Obesos , Células Mieloides/metabolismo , Fator 88 de Diferenciação Mieloide/metabolismo , Óxido Nítrico/metabolismo , Receptor 7 Toll-Like/metabolismo , Transferases/metabolismo
7.
J Immunol ; 181(8): 5242-8, 2008 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-18832678

RESUMO

Exosomes released from different types of cells have been proposed to contribute to intercellular communication. We report that thymic exosome-like particles (ELPs) released from cells of the thymus can induce the development of Foxp3(+) regulatory T (Treg) cells in the lung and liver. Thymic ELPs also induce the conversion of thymic CD4(+)CD25(-) T cells into Tregs. Tregs induced by thymic ELPs suppress the proliferation of CD4(+)CD25(-) T cells in vitro and in vivo. We further show that neutralization of TGF-beta in ELPs partially reverses thymic ELP-mediated induction of CD4(+)Foxp3(+) T cells in the lung and liver. This study demonstrates that thymic ELPs participate in the induction of Foxp3(+) Tregs. Also, TGF-beta of thymic ELPs might be required for the generation of Tregs in the peripheral tissues.


Assuntos
Fatores de Transcrição Forkhead , Fígado/imunologia , Pulmão/imunologia , Linfócitos T Reguladores/imunologia , Timo , Fator de Crescimento Transformador beta/imunologia , Animais , Feminino , Fígado/citologia , Pulmão/citologia , Camundongos , Camundongos Endogâmicos BALB C , Linfócitos T Reguladores/citologia
8.
Sci Rep ; 10(1): 21364, 2020 12 07.
Artigo em Inglês | MEDLINE | ID: mdl-33288795

RESUMO

Previously, we showed that embryonic deletion of TGF-ß type 2 receptor in mouse sclerotome resulted in defects in fibrous connective tissues in the spine. Here we investigated how TGF-ß regulates expression of fibrous markers: Scleraxis, Fibromodulin and Adamtsl2. We showed that TGF-ß stimulated expression of Scleraxis mRNA by 2 h and Fibromodulin and Adamtsl2 mRNAs by 8 h of treatment. Regulation of Scleraxis by TGF-ß did not require new protein synthesis; however, protein synthesis was required for expression of Fibromodulin and Adamtsl2 indicating the necessity of an intermediate. We subsequently showed Scleraxis was a potential intermediate for TGF-ß-regulated expression of Fibromodulin and Adamtsl2. The canonical effector Smad3 was not necessary for TGF-ß-mediated regulation of Scleraxis. Smad3 was necessary for regulation of Fibromodulin and Adamtsl2, but not sufficient to super-induce expression with TGF-ß treatment. Next, the role of several noncanonical TGF-ß pathways were tested. We found that ERK1/2 was activated by TGF-ß and required to regulate expression of Scleraxis, Fibromodulin, and Adamtsl2. Based on these results, we propose a model in which TGF-ß regulates Scleraxis via ERK1/2 and then Scleraxis and Smad3 cooperate to regulate Fibromodulin and Adamtsl2. These results define a novel signaling mechanism for TGFß-mediated fibrous differentiation in sclerotome.


Assuntos
RNA Mensageiro/genética , RNA Interferente Pequeno/genética , Esqueleto/metabolismo , Fator de Crescimento Transformador beta/farmacologia , Proteínas ADAMTS/genética , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Western Blotting , Desenvolvimento Ósseo/efeitos dos fármacos , Desenvolvimento Ósseo/genética , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Proteínas da Matriz Extracelular/genética , Feminino , Fibromodulina/genética , Fibromodulina/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Gravidez , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Esqueleto/efeitos dos fármacos
9.
Int J Cancer ; 124(11): 2621-33, 2009 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-19235923

RESUMO

Myeloid-derived suppressor cells (MDSCs) promote tumor progression. The mechanisms of MDSC development during tumor growth remain unknown. Tumor exosomes (T-exosomes) have been implicated to play a role in immune regulation, however the role of exosomes in the induction of MDSCs is unclear. Our previous work demonstrated that exosomes isolated from tumor cells are taken up by bone marrow myeloid cells. Here, we extend those findings showing that exosomes isolated from T-exosomes switch the differentiation pathway of these myeloid cells to the MDSC pathway (CD11b(+)Gr-1(+)). The resulting cells exhibit MDSC phenotypic and functional characteristics including promotion of tumor growth. Furthermore, we demonstrated that in vivo MDSC mediated promotion of tumor progression is dependent on T-exosome prostaglandin E2 (PGE2) and TGF-beta molecules. T-exosomes can induce the accumulation of MDSCs expressing Cox2, IL-6, VEGF, and arginase-1. Antibodies against exosomal PGE2 and TGF-beta block the activity of these exosomes on MDSC induction and therefore attenuate MDSC-mediated tumor-promoting ability. Exosomal PGE2 and TGF-beta are enriched in T-exosomes when compared with exosomes isolated from the supernatants of cultured tumor cells (C-exosomes). The tumor microenvironment has an effect on the potency of T-exosome mediated induction of MDSCs by regulating the sorting and the amount of exosomal PGE2 and TGF-beta available. Together, these findings lend themselves to developing specific targetable therapeutic strategies to reduce or eliminate MDSC-induced immunosuppression and hence enhance host antitumor immunotherapy efficacy.


Assuntos
Exossomos/fisiologia , Células Mieloides/fisiologia , Neoplasias/patologia , Animais , Antígeno CD11b/análise , Linhagem Celular Tumoral , Dinoprostona/fisiologia , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Receptores de Quimiocinas/análise , Fator de Crescimento Transformador beta/fisiologia
10.
Biochim Biophys Acta ; 1773(7): 1116-23, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17555831

RESUMO

An important characteristic of tumors is that they at some point in their development overcome the surveillance of the immune system. Tumors secrete exosomes, multivesicular bodies containing a distinct set of proteins that can fuse with cells of the circulating immune system. Purified exosomes from TS/A breast cancer cells, but not non-exosomal fractions, inhibit (at concentrations of nanograms per ml protein) IL-2-induced natural killer (NK) cell cytotoxicity. The dietary polyphenol, curcumin (diferuloylmethane), partially reverses tumor exosome-mediated inhibition of natural killer cell activation, which is mediated through the impairment of the ubiquitin-proteasome system. Exposure of mouse breast tumor cells to curcumin causes a dose-dependent increase in ubiquitinated exosomal proteins compared to those in untreated TS/A breast tumor cells. Furthermore, exosomes isolated from tumor cells pretreated with curcumin have a much attenuated inhibition of IL-2 stimulated NK cell activation. Jak3-mediated activation of Stat5 is required for tumor cytotoxicity of IL-2 stimulated NK cells. TS/A tumor exosomes strongly inhibit activation of Stat5, whereas the tumor exosomes isolated from curcumin-pretreated tumor cells have a lowered potency for inhibition of IL-2 stimulated NK cell cytotoxicity. These data suggest that partial reversal of tumor exosome-mediated inhibition of NK cell tumor cytotoxicity may account for the anti-cancer properties of curcumin.


Assuntos
Antineoplásicos/metabolismo , Neoplasias da Mama , Curcumina/metabolismo , Células Matadoras Naturais/imunologia , Animais , Neoplasias da Mama/imunologia , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Ativação Enzimática , Feminino , Humanos , Sistema Imunitário/fisiologia , Interleucina-2/imunologia , Janus Quinase 3/metabolismo , Camundongos , Proteínas de Neoplasias/metabolismo , Ubiquitina/metabolismo
11.
JOR Spine ; 1(2)2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30662980

RESUMO

BACKGROUND: Members of the transforming growth factor beta (TGF-ß) family are secreted proteins that regulate skeletal development. TGF-ß signaling is critical in embryonic development of the annulus fibrosus (AF) of the intervertebral disc (IVD). To address the question of the role of TGF-ß signaling in postnatal development and maintenance of the skeleton, we generated mice in which Tgfbr2 was deleted at 2-weeks of age in Aggrecan (Acan)-expressing cells using inducible Cre/LoxP recombination. METHODS: Localization of Cre recombination was visualized by crossing Acantm1(cre/ERT2)Crm mice to fluorescent mTmG reporter mice. Acantm1(cre/ERT2)Crm mice were mated to Tgfbr2 LoxP/LoxP mice and Cre recombinase was activated by tamoxifen injection at 2-weeks postnatally. Following tamoxifen injection, mice were aged to 3, 6, and 12-months and control mice were compared to the experimental (cKO) group. Mice were initially analyzed using X-ray and skeletal preparations. Sternocostal joints and IVD tissues were further analyzed histologically by hematoxylin and eosin (H&E), Safranin O, and Picrosirius Red staining as well as Col10 immunostaining. RESULTS: Cre recombination was observed in the IVD and sternocostal joints. X-ray analysis revealed osteophyte formation within the disc space of 12-month-old cKO mice. Skeletal preparations confirmed calcification within the IVD and the sternocostal joints in cKO mice. H&E staining of cKO IVD revealed disorganized growth plates, delay in the formation of the bony endplate, and Col10 staining in the AF indicative of ectopic endochondral bone formation. Furthermore, proteoglycan loss was observed and collagen bundles within the inner AF were thinner and less organized. Alterations in the IVD were apparent beginning at 3 months and were progressively more visible at 6 and 12 months. Similarly, histological analysis of cKO sternocostal joints revealed joint calcification, proteoglycan loss, and disorganization of the collagen architecture at 12 months of age. CONCLUSIONS: TGF-ß signaling is important for postnatal development and maintenance of fibrocartilaginous IVD and sternocostal joints.

12.
J Clin Invest ; 112(9): 1332-41, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-14597760

RESUMO

Previously, we described an APC-adenovirus (APC-Ad) FasL cell gene therapy method which could be used to deplete autoreactive T cells in vivo. FasL was toxic, however, and controlled regulation of FasL was not achieved. Here we describe an improved approach to delivering TNF-related apoptosis-inducing ligand (TRAIL) in vivo in which collagen II-induced (CII-induced) arthritis-susceptible (CIA-susceptible) DBA/1j mice were treated with CII-pulsed DCs that had been transfected with a novel Ad system. The Ad was engineered to exhibit inducible TRAIL under the control of the doxycycline-inducible (DOX-inducible) tetracycline response element (TRE). Four groups of mice were treated with CII-DC-AdTRAIL+DOX, CII-DC-AdTRAIL (no DOX), CII-DC-AdGFP+DOX, or DC-AdTRAIL+DOX (no CII), beginning 2 weeks after priming with CII in CFA. The incidence of arthritis and infiltration of T cells in the joint was significantly decreased in CII-DC-AdTRAIL+DOX-treated mice. The in vitro splenic T cell proliferative response and induction of IFN-gamma to bovine CII stimulation were also significantly reduced in mice treated with CII-DC-AdTRAIL+DOX. AdTRAIL+DOX was not toxic to DCs or mice but could induce activated T cells to undergo apoptosis in the spleen. Our results suggest that CII-DC-AdTRAIL+DOX cell gene therapy is a safe and effective method for inhibiting the development of CIA.


Assuntos
Artrite Reumatoide/terapia , Terapia Baseada em Transplante de Células e Tecidos , Colágeno Tipo II/imunologia , Células Dendríticas/fisiologia , Terapia Genética , Glicoproteínas de Membrana/genética , Linfócitos T/fisiologia , Fator de Necrose Tumoral alfa/genética , Adenoviridae/genética , Animais , Anticorpos/imunologia , Formação de Anticorpos , Apoptose , Proteínas Reguladoras de Apoptose , Movimento Celular , Doxorrubicina/farmacologia , Feminino , Ativação Linfocitária , Glicoproteínas de Membrana/fisiologia , Camundongos , Camundongos Endogâmicos DBA , Baço/patologia , Ligante Indutor de Apoptose Relacionado a TNF , Transfecção , Fator de Necrose Tumoral alfa/fisiologia
13.
Mech Ageing Dev ; 128(11-12): 672-80, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-18036633

RESUMO

In this study, our data show that in young BXD12 mice, the implanted TS/A tumor regressed in 4 weeks after implantation, and this regression was associated with extensive T cell infiltration. In contrast, in old BXD12 mice, it was observed that there was rapid tumor growth by 7 weeks. T cell cytotoxicity against TS/A tumor cells exhibited a significant age-related decline, which was correlated with a decline in CD3(+) T cell infiltration of the tumor. Furthermore, the decline of T cell tumor cytotoxicity in aged BXD12 mice was also correlated with the accumulation of CD11b(+)Gr1(+) myeloid-derived suppressor cells in the spleen. Adoptive transfer of these accumulated CD11b(+)Gr1(+)cells from aged mice to 2-month-old BXD12 mice led to the delay of the rejection of implanted tumor cells. The depletion of CD11b(+)Gr1(+)cells from aged BXD12 mice led to the slower growth of tumor. Induction of arginase 1 in myeloid cells isolated from aged mice plays a partial role in immune suppression of T cell cytotoxicity. Thus, the accumulation of immunosuppresssing myeloid cells appears to contribute to the increase of tumor susceptibility as the age of mice increases.


Assuntos
Envelhecimento/imunologia , Tolerância Imunológica , Linfócitos do Interstício Tumoral/imunologia , Neoplasias Mamárias Animais/imunologia , Células Mieloides/imunologia , Linfócitos T Citotóxicos/imunologia , Linfócitos T Reguladores/imunologia , Evasão Tumoral , Transferência Adotiva , Fatores Etários , Envelhecimento/patologia , Animais , Arginase/biossíntese , Antígenos CD11/análise , Complexo CD3/análise , Linhagem Celular Tumoral , Indução Enzimática , Feminino , Linfócitos do Interstício Tumoral/patologia , Neoplasias Mamárias Animais/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Células Mieloides/enzimologia , Células Mieloides/patologia , Baço/imunologia , Linfócitos T Citotóxicos/patologia , Linfócitos T Reguladores/enzimologia , Linfócitos T Reguladores/patologia , Fatores de Tempo
14.
Diabetes ; 58(11): 2498-505, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19675137

RESUMO

OBJECTIVE: We sought to determine whether exosome-like vesicles (ELVs) released from adipose tissue play a role in activation of macrophages and subsequent development of insulin resistance in a mouse model. RESEARCH DESIGN AND METHODS: ELVs released from adipose tissue were purified by sucrose gradient centrifugation and labeled with green fluorescent dye and then intravenously injected into B6 ob/ob mice (obese model) or B6 mice fed a high-fat diet. The effects of injected ELVs on the activation of macrophages were determined through analysis of activation markers by fluorescence-activated cell sorter and induction of inflammatory cytokines using an ELISA. Glucose tolerance and insulin tolerance were also evaluated. Similarly, B6 mice with different gene knockouts including TLR2, TLR4, MyD88, and Toll-interleukin-1 receptor (TIR) domain-containing adaptor protein inducing interferon-beta (TRIF) were also used for testing their responses to the injected ELVs. RESULTS: ELVs are taken up by peripheral blood monocytes, which then differentiate into activated macrophages with increased secretion of tumor necrosis factor-alpha (TNF-alpha) and interleukin-6 (IL-6). Injection of obELVs into wild-type C57BL/6 mice results in the development of insulin resistance. When the obELVs were intravenously injected into TLR4 knockout B6 mice, the levels of glucose intolerance and insulin resistance were much lower. RBP4 is enriched in the obELVs. Bone marrow-derived macrophages preincubated with recombinant RBP4 led to attenuation of obELV-mediated induction of IL-6 and TNF-alpha. CONCLUSIONS: ELVs released by adipose tissue can act as a mode of communication between adipose tissues and macrophages. The obELV-mediated induction of TNF-alpha and IL-6 in macrophages and insulin resistance requires the TLR4/TRIF pathway.


Assuntos
Tecido Adiposo/fisiologia , Exossomos/fisiologia , Resistência à Insulina/fisiologia , Ativação de Macrófagos/fisiologia , Macrófagos/fisiologia , Tecido Adiposo/citologia , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/fisiologia , Comunicação Celular/fisiologia , Diferenciação Celular , Exossomos/efeitos dos fármacos , Exossomos/ultraestrutura , Glucose/metabolismo , Teste de Tolerância a Glucose , Insulina/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Obesos , Microscopia Eletrônica , Monócitos/citologia , Monócitos/fisiologia , Proteínas Recombinantes/farmacologia , Proteínas Plasmáticas de Ligação ao Retinol/genética , Receptor 4 Toll-Like/deficiência , Receptor 4 Toll-Like/genética
15.
Blood ; 109(10): 4336-42, 2007 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-17244679

RESUMO

Tumor growth promotes the expansion of myeloid suppressor cells. An inverse correlation between natural killer (NK) cell activation and myeloid suppressor cell (MSC) expansion in tumor-bearing patients and mice prompted us to investigate the role of MSCs in controlling NK antitumor cytotocixity. After adoptive transfer to naive recipients, CD11b(+)Gr-1(+) MSCs freshly isolated from spleens of tumor-bearing mice but not naive mice were able to inhibit NK cell cytotoxicity. An in vivo imaging analysis indicates that the removal of tumors resulted in a significant increased ability (P < .05) in NK cell cytotoxicity to eliminate injected YAC-1 cells from the lungs. Fluorescence-activated cell sorter (FACS) analysis of the composition of lung leukocytes further indicates that the removal of tumors also leads to the reduction of MSCs accumulated in the lung. These data suggest that MSCs suppress NK cell cytotoxicity. The inhibition of NK cell cytotoxicity is cell-cell contact dependent. Inhibition of perforin but not granzyme B production was responsible for MSC-mediated inhibition of NK cytotoxicity. Western blot analyses further suggests that MSCs suppress IL-2-mediated NK cell cytotoxicity by affecting the activity of Stat5.


Assuntos
Adenocarcinoma/imunologia , Proliferação de Células , Citotoxicidade Imunológica , Células Matadoras Naturais/imunologia , Neoplasias Mamárias Animais/imunologia , Baço/citologia , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/fisiologia , Animais , Antígeno CD11b/metabolismo , Adesão Celular/imunologia , Células Cultivadas , Feminino , Interleucina-2/farmacologia , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Perforina , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Receptores de Quimiocinas/metabolismo , Fator de Transcrição STAT5/metabolismo , Baço/efeitos dos fármacos , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/metabolismo
16.
J Immunol ; 178(11): 6867-75, 2007 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-17513735

RESUMO

The production of exosomes by tumor cells has been implicated in tumor-associated immune suppression. In this study, we show that, in mice, exosomes produced by TS/A murine mammary tumor cells target CD11b(+) myeloid precursors in the bone marrow (BM) in vivo, and that this is associated with an accumulation of myeloid precursors in the spleen. Moreover, we demonstrate that TS/A exosomes block the differentiation of murine myeloid precursor cells into dendritic cells (DC) in vitro. Addition of tumor exosomes at day 0 led to a significant block of differentiation into DC, whereas addition at later time points was less effective. Similarly, exosomes produced by human breast tumor cells inhibited the differentiation of human monocytes in vitro. The levels of IL-6 and phosphorylated Stat3 were elevated 12 h after the tumor exosome stimulation of murine myeloid precursors, and tumor exosomes were less effective in inhibiting differentiation of BM cells isolated from IL-6 knockout mice. Addition of a rIL-6 to the IL-6 knockout BM cell culture restored the tumor exosome-mediated inhibition of DC differentiation. These data suggest that tumor exosome-mediated induction of IL-6 plays a role in blocking BM DC differentiation.


Assuntos
Células da Medula Óssea/citologia , Diferenciação Celular/imunologia , Células Dendríticas/citologia , Exocitose/imunologia , Inibidores do Crescimento/imunologia , Neoplasias Mamárias Experimentais/imunologia , Melanoma Experimental/imunologia , Vesículas Secretórias/imunologia , Adulto , Animais , Células da Medula Óssea/imunologia , Linhagem Celular Tumoral , Células Cultivadas , Células Dendríticas/imunologia , Feminino , Inibidores do Crescimento/metabolismo , Humanos , Neoplasias Mamárias Experimentais/metabolismo , Neoplasias Mamárias Experimentais/patologia , Melanoma Experimental/metabolismo , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Progenitoras Mieloides/citologia , Células Progenitoras Mieloides/imunologia , Vesículas Secretórias/metabolismo , Vesículas Secretórias/patologia
17.
Am J Pathol ; 169(3): 889-902, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16936264

RESUMO

Fibroblast-like synoviocytes (FLSs) of patients with rheumatoid arthritis (RA FLSs) exhibit prosurvival, rather than apoptotic, response to tumor necrosis factor (TNF)-alpha stimulation. Here, we show that JAB1 is a critical regulator of the TNF-alpha-mediated anti-apo-ptosis pathways in RA FLSs. We found that knockdown of JAB1 using small interfering (si)RNA led to restoration of the TNF-alpha-induced apoptosis response, reduction of nuclear factor-kappaB activity, delayed degradation of IkappaB-alpha, and inhibited phosphorylation of JNK. Analysis of the interactions of JAB1 by reciprocal co-immunoprecipitations and confocal microscopy revealed that JAB1 interacts with TNF receptor-associated-factor 2 (TRAF2). The generation of the anti-apoptotic signal on binding of TNF-alpha to the TNF receptor (TNFR)1 has been shown to be associated with the recruitment of TRAF2 to the TNFR1 in a process that requires ubiquitination of TRAF2 with lysine-63-linked polyubiquitin chains. We found that TNF-alpha stimulation of JAB1 siRNA-transfected RA FLSs failed to stimulate ubiquitination of TRAF2. Thus, we conclude that JAB1-regulated ubiquitination of TRAF2 is a novel mechanism whereby TNF-alpha can induce anti-apoptosis signaling and production of matrix metalloproteinases through activation of nuclear factor-kappaB and JNK in RA FLSs.


Assuntos
Apoptose , Artrite Reumatoide/metabolismo , Fibroblastos/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Peptídeo Hidrolases/metabolismo , Membrana Sinovial/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Apoptose/efeitos dos fármacos , Artrite Reumatoide/patologia , Complexo do Signalossomo COP9 , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Fibroblastos/patologia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , MAP Quinase Quinase 4/metabolismo , Metaloproteinases da Matriz/metabolismo , NF-kappa B/metabolismo , Fosforilação/efeitos dos fármacos , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , RNA Interferente Pequeno/farmacologia , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Transdução de Sinais/efeitos dos fármacos , Líquido Sinovial/metabolismo , Membrana Sinovial/patologia , Fator 2 Associado a Receptor de TNF/metabolismo , Transfecção , Fator de Necrose Tumoral alfa/farmacologia , Ubiquitina/metabolismo
18.
J Immunol ; 176(12): 7385-93, 2006 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-16751383

RESUMO

In common with many other cell types, synovial fibroblasts produce exosomes. In this study, we show that the exosomes produced by synovial fibroblasts obtained from individuals with rheumatoid arthritis (RASF), but not exosomes produced by synovial fibroblasts obtained from individuals with osteoarthritis, contain a membrane bound form of TNF-alpha as demonstrated by colloidal gold immunostaining of TNF-alpha and confirmed by both Western blot and mass spectrometry. The RASF-derived exosomes, but not exosomes derived from fibroblasts obtained from individuals with osteoarthritis, are cytotoxic for the L929 cell, a TNF-alpha-sensitive cell line, and stimulate activation of NF-kappaB and induction of collagenase-1 in RASF. These effects are blocked by addition of soluble TNFR1 (sTNFbp), suggesting that a TNF-alpha-signaling pathway mediates these biological activities. sTNFbp also reduced the production of exosomes by RASF, suggesting the interruption of a positive amplification loop. Exosomes can transmit signals between cells, and RASF exosomes, effectively taken up by anti-CD3-activated T cells, activated AKT and NF-kappaB and rendered these activated T cells resistant to apoptosis. Neutralization of exosomal membrane TNF-alpha by sTNFbp partially reversed this resistance, suggesting that not only TNF-alpha but also additional exosomal proteins may contribute to the development of apoptosis resistance.


Assuntos
Apresentação de Antígeno , Apoptose/imunologia , Exocitose/imunologia , Ativação Linfocitária/imunologia , Proteínas de Membrana/fisiologia , Linfócitos T/imunologia , Fator de Necrose Tumoral alfa/fisiologia , Idoso , Animais , Artrite Reumatoide/imunologia , Artrite Reumatoide/metabolismo , Artrite Reumatoide/patologia , Linhagem Celular , Linhagem Celular Tumoral , Proliferação de Células , Técnicas de Cocultura , Citotoxicidade Imunológica , Feminino , Fibroblastos/imunologia , Fibroblastos/metabolismo , Fibroblastos/patologia , Humanos , Proteínas de Membrana/imunologia , Proteínas de Membrana/metabolismo , Camundongos , Pessoa de Meia-Idade , Osteoartrite/imunologia , Osteoartrite/metabolismo , Osteoartrite/patologia , Membrana Sinovial/imunologia , Membrana Sinovial/metabolismo , Membrana Sinovial/patologia , Linfócitos T/metabolismo , Fator de Necrose Tumoral alfa/imunologia , Fator de Necrose Tumoral alfa/metabolismo
19.
J Immunol ; 176(3): 1375-85, 2006 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-16424164

RESUMO

Many tumor cells shed specialized membrane vesicles known as exosomes. In this study, we show that pretreatment of mice with exosomes produced by TS/A or 4T.1 murine mammary tumor cells resulted in accelerated growth of implanted tumor cells in both syngeneic BALB/c mice and nude mice. As implanted TS/A tumor cells grew more rapidly in mice that had been depleted of NK cells, we analyzed the effects of the tumor-derived exosomes on NK cells. The tumor-derived exosomes inhibit NK cell cytotoxic activity ex vivo and in vitro as demonstrated by chromium release assays. The treatment of mice with TS/A tumor exosomes also led to a reduction in the percentages of NK cells, as determined by FACS analysis, in the lungs and spleens. Key features of NK cell activity were inhibited, including release of perforin but not granzyme B, as well as the expression of cyclin D3 and activation of the Jak3-mediated pathways. Human tumor cell lines also were found to produce exosomes that were capable of inhibiting IL-2-stimulated NK cell proliferation. Exosomes produced by dendritic cells or B cells did not. The presentation of tumor Ags by exosomes is under consideration as a cancer vaccine strategy; however, we found that pretreatment of mice with tumor exosomes blunted the protective effect of syngeneic dendritic cells pulsed ex vivo with tumor exosomes. We propose that tumor exosomes contribute to the growth of tumors by blocking IL-2-mediated activation of NK cells and their cytotoxic response to tumor cells.


Assuntos
Carcinoma/imunologia , Proliferação de Células , Vesículas Citoplasmáticas/imunologia , Citotoxicidade Imunológica/imunologia , Exocitose/imunologia , Terapia de Imunossupressão , Células Matadoras Naturais/imunologia , Neoplasias Mamárias Animais/imunologia , Animais , Carcinoma/patologia , Linhagem Celular Tumoral , Células Dendríticas/imunologia , Feminino , Imunoterapia Adotiva , Células Matadoras Naturais/metabolismo , Contagem de Linfócitos , Subpopulações de Linfócitos/imunologia , Subpopulações de Linfócitos/patologia , Neoplasias Mamárias Animais/metabolismo , Neoplasias Mamárias Animais/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Transdução de Sinais/imunologia
20.
Am J Pathol ; 167(3): 705-19, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16127151

RESUMO

Rheumatoid arthritis synovial fibroblasts (RASFs) contribute to arthritic cartilage degradation. Although RASFs are normally resistant to apoptosis, Apo2L/tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-based gene therapy has been successfully used in a mouse model of arthritis. We investigated this further by treating human RASFs with nontoxic doses of the proteasome inhibitor lactacystin. Treatment induced cytosolic accumulation of p53 and enhanced the susceptibility of RASFs to apoptosis mediated by TRAIL-R2 (DR5) but not Fas. A specific role for p53 in TRAIL-R2-mediated apoptosis was indicated by the ability of p53 siRNA to significantly reduce RASF apoptosis and by the reduced apoptosis of RASFs bearing p53 mutations on treatment with anti-DR5 antibody or anti-DR5 antibody plus lactacystin. p53 immunoprecipitation followed by mass spectrometry identified a vimentin-p53 complex, an interaction that was confirmed by reciprocal vimentin-p53 immunoprecipitation and by co-immunofluorescence. Interestingly, human caspase-4 cleaved human vimentin, and blockade of caspase-4 with a chemical inhibitor or with specific siRNA significantly inhibited TRAIL-R2-mediated apoptosis of RASFs. Furthermore, blockade of caspase-4 was paralleled by persistence of a cytosolic pattern of p53 and absence of p53 translocation to the nucleus. Taken together, our findings suggest a unique role for caspase-4 in cleaving vimentin and releasing cytosolic p53 for nuclear translocation, events that may regulate the sensitivity of RASFs to receptor-mediated apoptosis.


Assuntos
Apoptose , Artrite Reumatoide/fisiopatologia , Fibroblastos , Glicoproteínas de Membrana/metabolismo , Membrana Sinovial/fisiopatologia , Fator de Necrose Tumoral alfa/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Vimentina/metabolismo , Acetilcisteína/análogos & derivados , Acetilcisteína/farmacologia , Anticorpos/farmacologia , Proteínas Reguladoras de Apoptose , Artrite Reumatoide/metabolismo , Transporte Biológico , Inibidores de Caspase , Caspases/metabolismo , Núcleo Celular/metabolismo , Células Cultivadas , Citoplasma/metabolismo , Citosol/metabolismo , Ativação Enzimática , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Humanos , Complexo de Endopeptidases do Proteassoma/metabolismo , Receptores do Ligante Indutor de Apoptose Relacionado a TNF , Receptores do Fator de Necrose Tumoral/imunologia , Receptores do Fator de Necrose Tumoral/metabolismo , Transdução de Sinais , Membrana Sinovial/metabolismo , Membrana Sinovial/patologia , Ligante Indutor de Apoptose Relacionado a TNF , Distribuição Tecidual , Ubiquitina/metabolismo , Vimentina/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA