Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
Front Oncol ; 11: 662723, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35223446

RESUMO

Myeloid-derived suppressor cells (MDSC) and tumor-associated macrophages (TAM) contribute to cancer-related inflammation and tumor progression. While several myeloid molecules have been ascribed a regulatory function in these processes, the triggering receptors expressed on myeloid cells (TREMs) have emerged as potent modulators of the innate immune response. While various TREMs amplify inflammation, others dampen it and are emerging as important players in modulating tumor progression-for instance, soluble TREM-1 (sTREM-1), which is detected during inflammation, associates with disease progression, while TREM-2 expression is associated with tumor-promoting macrophages. We hypothesized that TREM-1 and TREM-2 might be co-expressed on tumor-infiltrating myeloid cells and that elevated sTREM-1 associates with disease outcomes, thus representing a possibility for mutual modulation in cancer. Using the 4T1 breast cancer model, we found TREM-1 and TREM-2 expression on MDSC and TAM and that sTREM-1 was elevated in tumor-bearing mice in multiple models and correlated with tumor volume. While TREM-1 engagement enhanced TNF, a TREM-2 ligand was detected on MDSC and TAM, suggesting that both TREM could be functional in the tumor setting. Similarly, we detected TREM-1 and Trem2 expression in myeloid cells in the RENCA model of renal cell carcinoma (RCC). We confirmed these findings in human disease by demonstrating the expression of TREM-1 on tumor-infiltrating myeloid cells from patients with RCC and finding that sTREM-1 was increased in patients with RCC. Finally, The Cancer Genome Atlas analysis shows that TREM1 expression in tumors correlates with poor outcomes in RCC. Taken together, our data suggest that manipulation of the TREM-1/TREM-2 balance in tumors may be a novel means to modulate tumor-infiltrating myeloid cell phenotype and function.

2.
J Natl Cancer Inst Monogr ; 2017(52)2017 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-29140488

RESUMO

Since 2007, the US National Cancer Institute (NCI) Office of Cancer Complementary and Alternative Medicine (OCCAM), together with the Cancer Institute of the China Academy of Chinese Medical Sciences (CICACMS), institutes at China Academy of Sciences and Chinese Academy of Medical Sciences, have engaged in collaborations on Chinese medicine (CM) and cancer research. Through these collaborations, CM drugs and compounds have been studied at NCI labs. This paper summarizes the discoveries and progress on these research projects, exploring the aspects of cancer prevention, botanical drug mechanisms of action and component analysis/quality control (QC), and anticancer activity screening. These and other related projects have been presented in various jointly convened workshops and have provided the backdrop for establishing a new organization, the International Consortium for CM and Cancer, to promote international collaborations in this field.


Assuntos
Medicina Tradicional Chinesa , Neoplasias/terapia , China , Medicamentos de Ervas Chinesas/química , Medicamentos de Ervas Chinesas/farmacologia , Medicamentos de Ervas Chinesas/uso terapêutico , Humanos , Medicina Tradicional Chinesa/métodos , National Cancer Institute (U.S.) , Neoplasias/diagnóstico , Neoplasias/prevenção & controle , Pesquisa , Estados Unidos
3.
Biomaterials ; 53: 545-53, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25890750

RESUMO

DNA that encodes tumor-specific antigens represents potential immunostimulatory agents. However, rapid enzymatic degradation and fragmentation of DNA during administration can result in limited vector expression and, consequently, poor efficacy. These challenges have necessitated the use of novel strategies for DNA delivery. Herein, we study the ability of cationic self-assembling peptide hydrogels to encapsulate plasmid DNA, and enhance its immunostimulatory potential in vivo. The effect of network charge on the gel's ability to retain the DNA was assessed employing three gel-forming peptides that vary systematically in formal charge. The peptide HLT2, having a formal charge of +5 at neutral pH, was optimal in encapsulating microgram quantities of DNA with little effect on its rheological properties, allowing its effective syringe delivery in vivo. The plasmid, DNA(TA), encapsulated within these gels encodes for a melanoma-specific gp100 antigen fused to the alarmin protein adjuvant HMGN1. Implantation of DNA(TA)-loaded HLT2 gels into mice resulted in an acute inflammatory response with the presence of polymorphonuclear cells, which was followed by infiltrating macrophages. These cellular infiltrates aid in the processing of encapsulated DNA, promoting increased lymphoproliferation and producing an enhanced immune response mediated by CD4+/IFNγ+ expressing Th1 cells, and complemented by the formation of gp100-specific antibodies.


Assuntos
Adjuvantes Imunológicos/farmacologia , DNA/química , Hidrogéis , Peptídeos/farmacologia , Adjuvantes Imunológicos/química , Sequência de Aminoácidos , Animais , Feminino , Inflamação/patologia , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Peptídeos/química
4.
Cancer Lett ; 355(2): 232-41, 2014 Dec 28.
Artigo em Inglês | MEDLINE | ID: mdl-25242356

RESUMO

Cancer pain is a deleterious consequence of tumor growth and related inflammation. Opioids and anti-inflammatory drugs provide first line treatment for cancer pain, but both are limited by side effects. Fufang Kushen injection (FKI) is GMP produced, traditional Chinese medicine used alone or with chemotherapy to reduce cancer-associated pain. FKI limited mouse sarcoma growth both in vivo and in vitro, in part, by reducing the phosphorylation of ERK and AKT kinases and BAD. FKI inhibited TRPV1 mediated capsaicin-induced ERK phosphorylation and reduced tumor-induced proinflammatory cytokine production. Thus, FKI limited cancer pain both directly by blocking TRPV1 signaling and indirectly by reducing tumor growth.


Assuntos
Medicamentos de Ervas Chinesas/farmacologia , Hiperalgesia/tratamento farmacológico , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sarcoma/complicações , Sarcoma/tratamento farmacológico , Canais de Cátion TRPV/metabolismo , Animais , Capsaicina/metabolismo , Linhagem Celular Tumoral , Citocinas/metabolismo , Feminino , Hiperalgesia/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Sarcoma/enzimologia , Sarcoma/metabolismo
5.
Cancer Res ; 74(21): 5989-98, 2014 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-25205103

RESUMO

Alarmins are endogenous mediators that are elicited rapidly in response to danger signals, enhancing innate and adaptive immune responses by promoting the recruitment and maturation of antigen-presenting cells (APC). The nucleosome-binding protein HMGN1 is a potent alarmin that binds TLR4 and induces antigen-specific Th1 immune responses, but its contributions to antitumor immunity have not been explored. We found that ovalbumin (OVA)-expressing EG7 mouse thymoma cells grew much faster in Hmgn1-deficient mice than littermate-matched controls. Tumor-bearing Hmgn1(-/-) mice generated fewer OVA-specific CD8 cells in the spleen than EG7-bearing Hmgn1(+/+) mice, suggesting that HMGN1 supported T cell-mediated antitumor immunity. In addition, EG7 tumors expressing HMGN1 grew more slowly than control EG7 tumors, suggesting greater resistance to HMGN1-expressing tumors. This resistance relied on T cell-mediated immunity because it was abolished by in vivo depletion of CD4(+) and CD8(+) T cells. Moreover, mice vaccinated with a DNA vector expressing an HMGN1-gp100 fusion protein manifested gp100-specific, Th1-polarized immune responses, acquiring resistance to challenge with mouse B16F1 melanoma. Overall, our findings show that HMGN1 contributes to antitumor immunity and it may offer an effective adjuvant to heighten responses to cancer vaccines.


Assuntos
Células Apresentadoras de Antígenos/imunologia , Proteína HMGN1/genética , Imunidade Celular/genética , Melanoma Experimental/imunologia , Adjuvantes Imunológicos/genética , Animais , Células Apresentadoras de Antígenos/metabolismo , Linfócitos T CD8-Positivos/imunologia , Proteína HMGN1/imunologia , Humanos , Melanoma Experimental/genética , Melanoma Experimental/patologia , Camundongos , Proteínas Recombinantes de Fusão/uso terapêutico , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/metabolismo , Vacinação , Antígeno gp100 de Melanoma/genética , Antígeno gp100 de Melanoma/imunologia
6.
Cytokine ; 65(1): 56-64, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24080164

RESUMO

CD4(+) T cells stimulate immune responses through distinct patterns of cytokine produced by Th1, Th2 or Th17 cells, or inhibit immune responses through Foxp3-expressing regulatory T cells (Tregs). Paradoxically, effector T cells were recently shown to activate Tregs, however, it remains unclear which Th subset is responsible for this effect. In this study, we found that Th17 cells expressed the highest levels of TNF among in vitro generated Th subsets, and most potently promoted expansion and stabilized Foxp3 expression by Tregs when co-transferred into Rag1(-/-) mice. Both TNF and IL-2 produced by Th17 cells contributed to this effect. The stimulatory effect of Th17 cells on Tregs was largely abolished when co-transferred with TNFR2-deficient Tregs. Furthermore, Tregs deficient in TNFR2 also supported a much lower production of IL-17A and TNF expression by co-transferred Th17 cells. Thus, our data indicate that the TNF-TNFR2 pathway plays a crucial role in the reciprocal stimulatory effect of Th17 cells and Tregs. This bidirectional interaction should be taken into account when designing therapy targeting Th17 cells, Tregs, TNF and TNFR2.


Assuntos
Ativação Linfocitária/imunologia , Receptores Tipo II do Fator de Necrose Tumoral/imunologia , Linfócitos T Reguladores/imunologia , Células Th17/imunologia , Fator de Necrose Tumoral alfa/imunologia , Transferência Adotiva , Animais , Antígenos CD4/imunologia , Citocinas/imunologia , Fatores de Transcrição Forkhead/imunologia , Interleucina-17/biossíntese , Interleucina-17/imunologia , Interleucina-2/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores Tipo II do Fator de Necrose Tumoral/biossíntese , Células Th1/imunologia , Células Th2/imunologia , Fator de Necrose Tumoral alfa/biossíntese
7.
J Immunol ; 191(2): 865-74, 2013 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-23776172

RESUMO

Alarmins are a group of structurally diverse host defense antimicrobial peptides that are important immune activators. In this article, we present a novel role for two potent alarmins, human ß-defensin 2 and 3 (HBD2 and 3), in promoting IFN-α production by human plasmacytoid dendritic cells. We demonstrate that HBD2 and 3 activate pDCs by enhancing the intracellular uptake of CpG and self DNA and promote DNA-induced IFN-α production in a TLR9-dependent manner. Both CpG and host DNA form aggregates that resemble DNA nets when combined with HBD2 and 3. Isothermal titration calorimetry studies to elucidate the nature of HBD3/CpG complexes demonstrate involvement of enthalpy-driven interactions, in addition to hydrophobic interactions, with the formation of complexes at a molar ratio of 2:1 defensin/CpG. The i.v. administration of HBD3/CpG complexes induced proinflammatory cytokines like IL-12, IFN-γ, IL-6, IFN-α, and IL-10 in serum, associated with an increased recruitment of APCs in the spleen. Subcutaneous injections of these complexes showed enhanced infiltration of inflammatory cells at the injection site, indicating a potential pathophysiological role for alarmin/DNA complexes in contributing to inflammation. Intraperitoneal immunization of HBD3/CpG complexes with OVA enhanced both cellular and humoral responses to OVA, compared with OVA/HBD3 or OVA/CPG alone, indicative of a much more potent adjuvant effect of the HBD3/CpG complexes. Thus, the ability of defensins to enhance cellular uptake of nucleic acids can lead to improved vaccine formulations by promoting their uptake by various cells, resulting in an enhanced immune response.


Assuntos
Células Dendríticas/metabolismo , Inflamação/imunologia , Interferon-alfa/biossíntese , beta-Defensinas/metabolismo , Adjuvantes Imunológicos/farmacologia , Animais , Células Apresentadoras de Antígenos/imunologia , Transporte Biológico , Células Cultivadas , Ilhas de CpG , DNA/metabolismo , Células Dendríticas/imunologia , Feminino , Humanos , Interações Hidrofóbicas e Hidrofílicas , Interferon-alfa/sangue , Interferon gama/sangue , Interleucina-12/sangue , Interleucina-6/sangue , Camundongos , Camundongos Endogâmicos C57BL , Ovalbumina/imunologia , Receptor Toll-Like 9/metabolismo
8.
PLoS One ; 8(3): e58791, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23527025

RESUMO

MCP-1/CCL2 plays an important role in the initiation and progression of cancer. Since tumor cells produce MCP-1, they are considered to be the main source of this chemokine. Here, we examined whether MCP-1 produced by non-tumor cells affects the growth and lung metastasis of 4T1 breast cancer cells by transplanting them into the mammary pad of WT or MCP-1(-/-) mice. Primary tumors at the injected site grew similarly in both mice; however, lung metastases were markedly reduced in MCP-1(-/-) mice, with significantly longer mouse survival. High levels of MCP-1 mRNA were detected in tumors growing in WT, but not MCP-1(-/-) mice. Serum MCP-1 levels were increased in tumor-bearing WT, but not MCP-1(-/-) mice. Transplantation of MCP-1(-/-) bone marrow cells into WT mice did not alter the incidence of lung metastasis, whereas transplantation of WT bone marrow cells into MCP-1(-/-) mice increased lung metastasis. The primary tumors of MCP-1(-/-) mice consistently developed necrosis earlier than those of WT mice and showed decreased infiltration by macrophages and reduced angiogenesis. Interestingly, 4T1 cells that metastasized to the lung constitutively expressed elevated levels of MCP-1, and intravenous injection of 4T1 cells producing a high level of MCP-1 resulted in increased tumor foci in the lung of WT and MCP-1(-/-) mice. Thus, stromal cell-derived MCP-1 in the primary tumors promotes lung metastasis of 4T1 cells, but tumor cell-derived MCP-1 can also contribute once tumor cells enter the circulation. A greater understanding of the source and role of this chemokine may lead to novel strategies for cancer treatment.


Assuntos
Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Quimiocina CCL2/genética , Neoplasias Pulmonares/secundário , Células Estromais/metabolismo , Animais , Células Sanguíneas/metabolismo , Células da Medula Óssea/metabolismo , Neoplasias da Mama/metabolismo , Neoplasias da Mama/mortalidade , Linhagem Celular Tumoral , Quimiocina CCL2/metabolismo , Modelos Animais de Doenças , Feminino , Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Leucócitos/imunologia , Leucócitos/patologia , Macrófagos/imunologia , Macrófagos/patologia , Camundongos , Camundongos Knockout , Necrose/genética , Neovascularização Patológica/genética
9.
J Immunol ; 190(3): 1076-84, 2013 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-23277487

RESUMO

Several lines of evidence indicate the instability of CD4(+)Foxp3(+) regulatory T cells (Tregs). We have therefore investigated means of promoting the stability of Tregs. In this study, we found that the proportion of Tregs in mouse strains deficient in TNFR2 or its ligands was reduced in the thymus and peripheral lymphoid tissues, suggesting a potential role of TNFR2 in promoting the sustained expression of Foxp3. We observed that upon in vitro activation with plate-bound anti-CD3 Ab and soluble anti-CD28 Ab, Foxp3 expression by highly purified mouse Tregs was markedly downregulated. Importantly, TNF partially abrogated this effect of TCR stimulation and stabilized Foxp3 expression. This effect of TNF was blocked by anti-TNFR2 Ab, but not by anti-TNFR1 Ab. Furthermore, TNF was not able to maintain Foxp3 expression by TNFR2-deficient Tregs. In a mouse colitis model induced by transfer of naive CD4 cells into Rag1(-/-) mice, the disease could be inhibited by cotransfer of wild-type Tregs, but not by cotransfer of TNFR2-deficient Tregs. Furthermore, in the lamina propria of the colitis model, most wild-type Tregs maintained Foxp3 expression. In contrast, an increased number of TNFR2-deficient Tregs lost Foxp3 expression. Thus, our data clearly show that TNFR2 is critical for the phenotypic and functional stability of Tregs in the inflammatory environment. This effect of TNF should be taken into account when designing future therapy of autoimmunity and graft-versus-host disease by using TNF inhibitors.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Colite/imunologia , Ativação Linfocitária/imunologia , Receptores Tipo II do Fator de Necrose Tumoral/imunologia , Linfócitos T Reguladores/imunologia , Transferência Adotiva , Animais , Animais Congênicos , Linfócitos T CD4-Positivos/transplante , Colite/microbiologia , Colite/patologia , Fatores de Transcrição Forkhead/análise , Fatores de Transcrição Forkhead/biossíntese , Fatores de Transcrição Forkhead/genética , Regulação da Expressão Gênica/imunologia , Proteínas de Homeodomínio/genética , Mucosa Intestinal/imunologia , Linfonodos/imunologia , Linfonodos/patologia , Linfopenia/etiologia , Linfopenia/imunologia , Linfopenia/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Baço/imunologia , Baço/patologia , Linfócitos T Reguladores/transplante , Timo/imunologia , Timo/patologia , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/farmacologia
10.
Immunol Lett ; 148(1): 34-8, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22898052

RESUMO

To clarify controversies in the literature of the field, we have purified and characterized B16F1 melanoma cell derived exosomes (mcd-exosomes) then we attempted to dissect their immunological activities. We tested how mcd-exosomes influence CD4+ T cell proliferation induced by bone marrow derived dendritic cells; we quantified NF-κB activation in mature macrophages stimulated with mcd-exosomes, and we compared the cytokine profile of LPS-stimulated, IL-4 induced, and mcd-exosome treated macrophages. We observed that mcd-exosomes helped the maturation of dendritic cells, enhancing T cell proliferation induced by the treated dendritic cells. The exosomes also activated macrophages, as measured by NF-κB activation. The cytokine and chemokine profile of macrophages treated with tumor cell derived exosomes showed marked differences from those induced by either LPS or IL-4, and it suggested that exosomes may play a role in the tumor progression and metastasis formation through supporting tumor immune escape mechanisms.


Assuntos
Células Dendríticas/imunologia , Exossomos/imunologia , Macrófagos/imunologia , Melanoma/imunologia , Animais , Linhagem Celular , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Quimiocinas/imunologia , Quimiocinas/metabolismo , Técnicas de Cocultura , Citocinas/imunologia , Citocinas/metabolismo , Células Dendríticas/metabolismo , Exossomos/metabolismo , Exossomos/ultraestrutura , Feminino , Interleucina-4/farmacologia , Lipopolissacarídeos/farmacologia , Ativação de Macrófagos/efeitos dos fármacos , Ativação de Macrófagos/imunologia , Macrófagos/citologia , Macrófagos/metabolismo , Melanoma/metabolismo , Melanoma/patologia , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Eletrônica de Transmissão , NF-kappa B/imunologia , NF-kappa B/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo
11.
Proc Natl Acad Sci U S A ; 109(16): 6130-5, 2012 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-22474389

RESUMO

Nuclear translocation of cytosolic CLIC4 is an essential feature of its proapoptotic and prodifferentiation functions. Here we demonstrate that CLIC4 is induced concurrently with inducible nitric oxide synthase (iNOS) and S-nitrosylated in proinflammatory peritoneal macrophages. Chemical inhibition or genetic ablation of iNOS inhibits S-nitrosylation and nuclear translocation of CLIC4. In macrophages, iNOS-induced nuclear CLIC4 coincides with the pro- to anti-inflammatory transition of the cells because IL-1ß and CXCL1 mRNA remain elevated in CLIC4 and iNOS knockout macrophages at late time points, whereas TNFα mRNA is elevated only in the iNOS knockout macrophages. Active IL-1ß remains elevated in CLIC4 knockout macrophages and in macrophages in which CLIC4 nuclear translocation is prevented by the NOS inhibitor l-NAME. Moreover, overexpression of nuclear-targeted CLIC4 down-regulates IL-1ß in stimulated macrophages. In mice, genetically null for CLIC4, the number of phagocytosing macrophages stimulated by LPS is reduced. Thus, iNOS-induced nuclear CLIC4 is an essential part of the macrophage deactivation program.


Assuntos
Núcleo Celular/metabolismo , Canais de Cloreto/metabolismo , Macrófagos/metabolismo , Proteínas Mitocondriais/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Animais , Linhagem Celular , Células Cultivadas , Canais de Cloreto/genética , Expressão Gênica/efeitos dos fármacos , Immunoblotting , Interferon gama/farmacologia , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Lipopolissacarídeos/farmacologia , Ativação de Macrófagos/genética , Ativação de Macrófagos/imunologia , Macrófagos/citologia , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Mitocondriais/genética , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo II/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
12.
Proc Natl Acad Sci U S A ; 108(24): 9798-803, 2011 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-21628584

RESUMO

Significant efforts have been devoted to the development of nanoparticular delivering systems targeting tumors. However, clinical application of nanoparticles is hampered by insufficient size homogeneity, difficulties in reproducible synthesis and manufacturing, frequent high uptake in the liver, systemic toxicity of the carriers (particularly for inorganic nanoparticles), and insufficient selectivity for tumor cells. We have found that properly modified synthetic analogs of transmembrane domains of membrane proteins can self-assemble into remarkably uniform spherical nanoparticles with innate biological activity. Self-assembly is driven by a structural transition of the peptide that adopts predominantly a beta-hairpin conformation in aqueous solutions, but folds into an alpha-helix upon spontaneous fusion of the nanoparticles with cell membrane. A 24-amino acid peptide corresponding to the second transmembrane helix of the CXCR4 forms self-assembled particles that inhibit CXCR4 function in vitro and hamper CXCR4-dependent tumor metastasis in vivo. Furthermore, such nanoparticles can encapsulate hydrophobic drugs, thus providing a delivery system with the potential for dual biological activity.


Assuntos
Membrana Celular/química , Proteínas de Membrana/química , Nanopartículas/química , Peptídeos/química , Sequência de Aminoácidos , Animais , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Dicroísmo Circular , Relação Dose-Resposta a Droga , Feminino , Humanos , Espectroscopia de Ressonância Magnética , Neoplasias Mamárias Experimentais/patologia , Neoplasias Mamárias Experimentais/prevenção & controle , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Nus , Microscopia Confocal , Microscopia Eletrônica de Transmissão , Dados de Sequência Molecular , Nanopartículas/ultraestrutura , Tamanho da Partícula , Peptídeos/metabolismo , Peptídeos/farmacologia , Ligação Proteica , Receptores CXCR4/antagonistas & inibidores , Receptores CXCR4/química , Receptores CXCR4/metabolismo
13.
J Immunol ; 186(11): 6417-26, 2011 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-21515789

RESUMO

We hypothesize that innate immune signals from infectious organisms and/or injured tissues may activate peripheral neuronal pain signals. In this study, we demonstrated that TLRs 3, 7, and 9 are expressed by human dorsal root ganglion neurons (DRGNs) and in cultures of primary mouse DRGNs. Stimulation of murine DRGNs with TLR ligands induced expression and production of proinflammatory chemokines and cytokines CCL5 (RANTES), CXCL10 (IP-10), IL-1α, IL-1ß, and PGE(2), which have previously been shown to augment pain. Further, TLR ligands upregulated the expression of a nociceptive receptor, transient receptor potential vanilloid type 1 (TRPV1), and enhanced calcium flux by TRPV1-expressing DRGNs. Using a tumor-induced temperature sensitivity model, we showed that in vivo administration of a TLR9 antagonist, known as a suppressive oligodeoxynucleotide, blocked tumor-induced temperature sensitivity. Taken together, these data indicate that stimulation of peripheral neurons by TLR ligands can induce nerve pain.


Assuntos
Neurônios/efeitos dos fármacos , Dor/fisiopatologia , Transdução de Sinais/fisiologia , Receptores Toll-Like/metabolismo , Aminoquinolinas/farmacologia , Anilidas/farmacologia , Animais , Ácidos Araquidônicos/farmacologia , Western Blotting , Cálcio/metabolismo , Capsaicina/farmacologia , Células Cultivadas , Cinamatos/farmacologia , Citocinas/genética , Citocinas/metabolismo , Dinoprostona/metabolismo , Relação Dose-Resposta a Droga , Endocanabinoides , Gânglios Espinais/citologia , Gânglios Espinais/efeitos dos fármacos , Gânglios Espinais/metabolismo , Humanos , Imidazóis/farmacologia , Camundongos , Microscopia Confocal , Neurônios/metabolismo , Poli I-C/farmacologia , Alcamidas Poli-Insaturadas/farmacologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/efeitos dos fármacos , Canais de Cátion TRPV/antagonistas & inibidores , Canais de Cátion TRPV/metabolismo , Receptor 3 Toll-Like/metabolismo , Receptor 7 Toll-Like/metabolismo , Receptor Toll-Like 9/metabolismo , Receptores Toll-Like/agonistas
14.
Sci Transl Med ; 2(61): 61ra90, 2010 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-21148126

RESUMO

Psoriasis is a common complex genetic disease characterized by hyperplasia and inflammation in the skin; however, the relative contributions of epidermal cells and the immune system to disease pathogenesis remain unclear. Linkage studies have defined a psoriasis susceptibility locus (PSORS4) on 1q21, the epidermal differentiation complex, which includes genes for small S100 calcium-binding proteins. These proteins are involved in extracellular and intracellular signaling during epithelial host defense, linking innate and adaptive immunity. Inflammation-prone psoriatic skin constitutively expresses elevated concentrations of S100A7 (psoriasin) and S100A15 (koebnerisin) in the epidermis. Here, we report that genetically modified mice expressing elevated amounts of doxycycline-regulated mS100a7a15 in skin keratinocytes demonstrated an exaggerated inflammatory response when challenged by exogenous stimuli such as abrasion (Koebner phenomenon). This immune response was characterized by immune cell infiltration and elevated concentrations of T helper 1 (T(H)1) and T(H)17 proinflammatory cytokines, which have been linked to the pathogenesis of psoriasis and were further amplified upon challenge. Both inflammation priming and amplification required mS100a7a15 binding to the receptor of advanced glycation end products (RAGE). mS100a7a15 potentiated inflammation by acting directly as a chemoattractant for leukocytes, further increasing the number of inflammatory cells infiltrating the skin. This study provides a pathogenetic psoriasis model using a psoriasis candidate gene to link the epidermis and innate immune system in inflammation priming, highlighting the S100A7A15-RAGE axis as a potential therapeutic target.


Assuntos
Inflamação/imunologia , Inflamação/patologia , Psoríase/genética , Psoríase/patologia , Proteínas S100/genética , Pele/patologia , Animais , Células Cultivadas , Citocinas/imunologia , Predisposição Genética para Doença , Humanos , Queratinócitos/citologia , Queratinócitos/imunologia , Camundongos , Camundongos Transgênicos , Psoríase/imunologia , Receptor para Produtos Finais de Glicação Avançada , Receptores Imunológicos/metabolismo , Proteína A7 Ligante de Cálcio S100 , Proteínas S100/imunologia , Pele/imunologia , Fator de Necrose Tumoral alfa/imunologia
15.
J Immunol ; 185(1): 174-82, 2010 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-20525892

RESUMO

Our previous study showed that TNFR2 is preferentially expressed by CD4(+)FoxP3(+) regulatory T cells (Tregs), and expression of this receptor identified maximally suppressive Tregs. TNFR2 is also expressed by a small fraction of CD4(+)FoxP3(-) conventional T cells (Tconvs) in normal mice, and its expression is upregulated by T cell activation. This raises questions about the role of TNFR2 signaling in the function of Tconv cells. In this study, by using FoxP3/gfp knock-in mice, we showed that TNFR2 signaling did not induce FoxP3(-) CD4 cells to become suppressive. Ki-67, a marker of proliferation, was concomitantly expressed with TNFR2 by CD4 cells, independent of forkhead box P3 expression, in normal mice and Lewis lung carcinoma-bearing mice. TNFR2 is associated with greater suppressive functions when expressed by Tregs and is associated with greater resistance to suppression when expressed by Tconv cells. In mice bearing 4T1 breast tumor or Lewis lung carcinoma, intratumoral Tconv cells expressing elevated levels of TNFR2 acquired the capacity to resist suppression by lymph node-derived Tregs. However, they remained susceptible to inhibition by more suppressive tumor-infiltrating Tregs, which expressed higher levels of TNFR2. Our data indicate that TNFR2 also costimulates Tconv cells. However, intratumoral Tregs expressing more TNFR2 are able to overcome the greater resistance to suppression of intratumoral Tconv cells, resulting in a dominant immunosuppressive tumor environment.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Proliferação de Células , Fatores de Transcrição Forkhead , Imunidade Inata , Ativação Linfocitária/imunologia , Receptores Tipo II do Fator de Necrose Tumoral/biossíntese , Receptores Tipo II do Fator de Necrose Tumoral/fisiologia , Linfócitos T Reguladores/imunologia , Animais , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/metabolismo , Carcinoma Pulmonar de Lewis/imunologia , Carcinoma Pulmonar de Lewis/patologia , Carcinoma Pulmonar de Lewis/prevenção & controle , Linhagem Celular Tumoral , Células Cultivadas , Anergia Clonal/genética , Anergia Clonal/imunologia , Técnicas de Cocultura , Feminino , Fatores de Transcrição Forkhead/biossíntese , Fatores de Transcrição Forkhead/deficiência , Técnicas de Introdução de Genes , Imunidade Inata/genética , Ativação Linfocitária/genética , Neoplasias Mamárias Experimentais/imunologia , Neoplasias Mamárias Experimentais/patologia , Neoplasias Mamárias Experimentais/prevenção & controle , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Receptores Tipo II do Fator de Necrose Tumoral/genética , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/metabolismo
16.
Eur J Immunol ; 40(4): 1099-106, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20127680

RESUMO

Previously, we found that co-expression of CD25 and TNFR2 identified the most suppressive subset of mouse Treg. In this study, we report that human peripheral blood (PB) FOXP3(+) cells present in CD25(high), CD25(low) and even CD25(-) subsets of CD4(+) cells expressed high levels of TNFR2. Consequently, TNFR2-expressing CD4(+)CD25(+) Treg included all of the FOXP3(+) cells present in the CD4(+)CD25(high) subset as well as a substantial proportion of the FOXP3(+) cells present in the CD4(+)CD25(low) subset. Flow cytometric analysis of PB identified five-fold more Treg, determined by FOXP3 expression, in the CD4(+)CD25(+)TNFR2(+) subset than in the CD4(+)CD25(high) subset. In addition, similar levels of FOXP3(+) cells were identified in both the CD4(+)CD25(+)TNFR2(+) and CD4(+)CD25(+)CD127(low/-) subsets. Furthermore, the CD4(+)CD25(+)TNFR2(+) subset expressed high levels of CTLA-4, CD45RO, CCR4 and low levels of CD45RA and CD127, a phenotype characteristic of Treg. Upon TCR stimulation, human PB CD4(+)CD25(+)TNFR2(+) cells were anergic and markedly inhibited the proliferation and cytokine production of co-cultured T-responder cells. In contrast, CD4(+)CD25(+)TNFR2(-) and CD4(+)CD25(-)TNFR2(+) T cells did not show inhibitory activity. As some non-Treg express TNFR2, the combination of CD25 and TNFR2 must be used to identify a larger population of human Treg, a population that may prove to be of diagnostic and therapeutic benefit in cancer and autoimmune diseases.


Assuntos
Fatores de Transcrição Forkhead/análise , Subunidade alfa de Receptor de Interleucina-2/biossíntese , Receptores Tipo II do Fator de Necrose Tumoral/biossíntese , Subpopulações de Linfócitos T/imunologia , Linfócitos T Reguladores/classificação , Adulto , Apresentação de Antígeno , Antígenos CD/análise , Antígeno CTLA-4 , Células Cultivadas/imunologia , Células Cultivadas/metabolismo , Técnicas de Cocultura , Citometria de Fluxo , Humanos , Imunofenotipagem , Interferon gama/biossíntese , Subunidade alfa de Receptor de Interleucina-2/análise , Subunidade alfa de Receptor de Interleucina-7/análise , Antígenos Comuns de Leucócito/análise , Ativação Linfocitária/efeitos dos fármacos , Receptores de Antígenos de Linfócitos T/imunologia , Receptores Tipo II do Fator de Necrose Tumoral/análise , Subpopulações de Linfócitos T/metabolismo , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo
17.
Carcinogenesis ; 30(2): 348-55, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19037090

RESUMO

The G protein-coupled formylpeptide receptor (FPR), known to mediate phagocytic leucocyte chemotaxis in response to bacterial- and host-derived agonists, was expressed by tumor cells in specimens of surgically removed more highly malignant human gliomas. In human glioblastoma cell lines, FPR activation increased cell motility, tumorigenicity and production of angiogenic factors. In studies of the mechanistic basis for the selective expression of FPR in more highly malignant gliomas, we found that the DNA methyltransferase inhibitor 5-Aza-2'-deoxycytidine (Aza), while promoting the differentiation of human glioblastoma cells, downregulated FPR expression. Aza also reduced the global methylation levels in glioblastoma cells and activated the pathway of p53 tumor suppressor. Methylation-specific polymerase chain reaction revealed that Aza treatment of tumor cells reduced the methylation of p53 promoter, which was accompanied by increased expression of p53 gene and protein. In addition, overexpression of p53 in glioblastoma cells mimicked the effect of Aza treatment as shown by increased cell differentiation but reduction in FPR expression, the capacity of tumor sphere formation in soft agar and tumorigenesis in nude mice. Furthermore, Aza treatment or overexpression of the wild-type p53 in glioblastoma cells increased the binding of p53 to FPR promoter region shown by chromatin immunoprecipitation. These results indicate that increased methylation of p53 gene retains human glioblastoma cells at a more poorly differentiated phase associated with the aberrant expression of FPR as a tumor-promoting cell surface receptor.


Assuntos
Neoplasias Encefálicas/metabolismo , Glioblastoma/metabolismo , Receptores de Formil Peptídeo/biossíntese , Animais , Azacitidina/análogos & derivados , Azacitidina/farmacologia , Neoplasias Encefálicas/patologia , Diferenciação Celular , Linhagem Celular Tumoral , Proliferação de Células , Quimiotaxia , Metilases de Modificação do DNA/antagonistas & inibidores , Decitabina , Regulação Neoplásica da Expressão Gênica , Glioblastoma/patologia , Humanos , Metilação , Camundongos , Camundongos Nus , Transplante de Neoplasias , Regiões Promotoras Genéticas , Transplante Heterólogo , Proteína Supressora de Tumor p53/metabolismo
18.
J Immunol ; 180(10): 6467-71, 2008 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-18453563

RESUMO

TNFR2 is predominantly expressed by a subset of human and mouse CD4(+)CD25(+)FoxP3(+) T regulatory cells (Tregs). In this study, we characterized the phenotype and function of TNFR2(+) Tregs in peripheral lymphoid tissues of normal and tumor-bearing C57BL/6 mice. We found that TNFR2 was expressed on 30-40% of the Tregs of the peripheral activated/memory subset that were most highly suppressive. In contrast, TNFR2(-) Tregs exhibited the phenotype of naive cells and only had minimal suppressive activity. Although not typically considered to be Tregs, CD4(+)CD25(-)TNFR2(+) cells nevertheless possessed moderate suppressive activity. Strikingly, the suppressive activity of TNFR2(+) Tregs was considerably more potent than that of reportedly highly suppressive CD103(+) Tregs. In the Lewis lung carcinoma model, more highly suppressive TNFR2(+) Tregs accumulated intratumorally than in the periphery. Thus, TNFR2 identifies a unique subset of mouse Tregs with an activated/memory phenotype and maximal suppressive activity that may account for tumor-infiltrating lymphocyte-mediated immune evasion by tumors.


Assuntos
Carcinoma Pulmonar de Lewis/imunologia , Linfócitos do Interstício Tumoral/imunologia , Receptores Tipo II do Fator de Necrose Tumoral/biossíntese , Subpopulações de Linfócitos T/imunologia , Linfócitos T Reguladores/imunologia , Animais , Antígenos CD4/metabolismo , Feminino , Citometria de Fluxo , Fatores de Transcrição Forkhead/metabolismo , Subunidade alfa de Receptor de Interleucina-2/metabolismo , Ativação Linfocitária/imunologia , Linfócitos do Interstício Tumoral/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Fenótipo , Receptores Tipo II do Fator de Necrose Tumoral/imunologia , Subpopulações de Linfócitos T/metabolismo , Linfócitos T Reguladores/metabolismo , Evasão Tumoral/imunologia
19.
Biochem Biophys Res Commun ; 362(1): 44-50, 2007 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-17706604

RESUMO

FOXP3, a forkhead transcription factor is essential for the development and function of CD4(+)CD25(+) regulatory T cells (Tregs). In contrast to conversion of murine naive T cells to Tregs by transduction of Foxp3, it is controversial whether ectopic expression of FOXP3 in human CD4(+) T cells is sufficient for acquisition of suppressive activity. Here, we show that retroviral transduction of FOXP3 induces a Treg phenotype in human leukemic CD4(+) Jurkat-T cells, evidenced by increased expression of Treg-associated cell surface markers as well as inhibition of cytokine production. Furthermore, FOXP3-transduced Jurkat-T cells suppress the proliferation of human CD4(+)CD25(-) T cells. Additionally, DNA microarray analysis identifies Treg-related genes regulated by FOXP3. Our study demonstrates that enforced expression of FOXP3 confers Treg-like properties on Jurkat-T cells, which can be a convenient and efficient Treg-like cell model for further study to identify Treg cell surface markers and target genes regulated by FOXP3.


Assuntos
Linfócitos T CD4-Positivos/metabolismo , Fatores de Transcrição Forkhead/química , Regulação da Expressão Gênica , Linfócitos T Reguladores/metabolismo , Proliferação de Células , Separação Celular , Citometria de Fluxo , Fatores de Transcrição Forkhead/metabolismo , Humanos , Subunidade alfa de Receptor de Interleucina-2/biossíntese , Células Jurkat , NF-kappa B/metabolismo , Fenótipo , Retroviridae/metabolismo , Linfócitos T/metabolismo , Transfecção
20.
Cancer Res ; 67(12): 5906-13, 2007 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-17575160

RESUMO

The G protein-coupled formylpeptide receptor (FPR), which mediates leukocyte migration in response to bacterial and host-derived chemotactic peptides, promotes the chemotaxis, survival, and tumorigenesis of highly malignant human glioblastoma cells. Because glioblastoma cells may also express other receptors for growth signals, such as the epidermal growth factor (EGF) receptor (EGFR), we investigated the role of EGFR in the signaling cascade of FPR and how two receptors cross-talk to exacerbate tumor growth. We found that N-formyl-methionyl-leucyl-phenylalanine, an FPR agonist peptide, rapidly induced EGFR phosphorylation at tyrosine residue (Tyr) 992, but not residues 846, 1068, or 1173, in glioblastoma cells, whereas all these residues were phosphorylated after only EGF treatment. The FPR agonist-induced EGFR phosphorylation in tumor cells was dependent on the presence of FPR as well as Galphai proteins, and was controlled by Src tyrosine kinase. The transactivation of EGFR contributes to the biological function of FPR in glioblastoma cells because inhibition of EGFR phosphorylation significantly reduced FPR agonist-induced tumor cell chemotaxis and proliferation. Furthermore, depletion of both FPR and EGFR by short interference RNA abolished the tumorigenesis of the glioblastoma cells. Our study indicates that the glioblastoma-promoting activity of FPR is mediated in part by transactivation of EGFR and the cross-talk between two receptors exacerbates the malignant phenotype of tumor cells. Thus, targeting both receptors may yield antiglioblastoma agents superior to those targeting one of them.


Assuntos
Neoplasias Encefálicas/metabolismo , Receptores ErbB/metabolismo , Glioblastoma/metabolismo , Receptores de Formil Peptídeo/metabolismo , Transdução de Sinais/fisiologia , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Receptores ErbB/genética , Glioblastoma/patologia , Humanos , Immunoblotting , Receptor Cross-Talk , Ativação Transcricional
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA