Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
J Immunol ; 183(1): 524-32, 2009 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-19542463

RESUMO

Follicular dendritic cells (FDCs) are located in the lymphoid follicles of secondary lymphoid tissues and play a pivotal role in the selection of memory B lymphocytes within the germinal center, a major site for HIV-1 infection. Germinal centers are composed of highly activated B cells, macrophages, CD4(+)T cells, and FDCs. However, the physiological role of FDCs in HIV-1 replication remains largely unknown. We demonstrate in our current study that FDCs can efficiently activate HIV-1 replication in latently infected monocytic cells via an intercellular communication network mediated by the P-selectin/P-selectin glycoprotein ligand 1 (PSGL-1) interaction. Upon coculture with FDCs, HIV-1 replication was significantly induced in infected monocytic cell lines, primary monocytes, or macrophages. These cocultures were found to synergistically induce the expression of P-selectin in FDCs via NF-kappaB activation and its cognate receptor PSGL-1 in HIV-1-infected cells. Consistent with this observation, we find that this response is significantly blocked by antagonistic Abs against PSGL-1 and almost completely inhibited by PSGL-1 small interfering RNA. Moreover, a selective inhibitor for Syk, which is a downstream effector of PSGL-1, blocked HIV-1 replication in our cultures. We have thus elucidated a novel regulatory mechanism in which FDCs are a potent positive bystander that facilitates HIV-1 replication in adjacent infected monocytic cells via a juxtacrine signaling mechanism.


Assuntos
Comunicação Celular/imunologia , Células Dendríticas Foliculares/imunologia , HIV-1/imunologia , Macrófagos/imunologia , Glicoproteínas de Membrana/fisiologia , Monócitos/imunologia , Selectina-P/metabolismo , Replicação Viral/imunologia , Linhagem Celular Tumoral , Células Cultivadas , Técnicas de Cocultura , Células Dendríticas Foliculares/virologia , Humanos , Ligantes , Macrófagos/virologia , Glicoproteínas de Membrana/metabolismo , Monócitos/virologia , Selectina-P/fisiologia , Tonsila Palatina , Transdução de Sinais/imunologia , Ativação Viral/imunologia , Latência Viral/imunologia
2.
Int J Cancer ; 124(3): 622-9, 2009 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-18973272

RESUMO

Epstein-Barr Virus (EBV)-associated immunoblastic lymphoma occurs in immunocompromised patients such as those with AIDS or transplant recipients after primary EBV infection or reactivation of a preexisting latent EBV infection. In the present study, we evaluated the effect of ritonavir, an HIV protease inhibitor, on EBV-positive lymphoblastoid B cells in vitro and in mice model. We found that it induced cell-cycle arrest at G1-phase and apoptosis through down-regulation of cell-cycle gene cyclin D2 and antiapoptotic gene survivin. Furthermore, ritonavir suppressed transcriptional activation of NF-kappaB in these cells. Ritonavir efficiently prevented growth and infiltration of lymphoma cells in various organs of NOD/SCID/gammacnull mice at the same dose used for treatment of patients with AIDS. Our results indicate that ritonavir targets NF-kappaB activated in tumor cells and shows anti-tumor effects. These data also suggest that this compound may have promise for treatment or prevention of EBV-associated lymphoproliferative diseases that occur in immunocompromised patients.


Assuntos
Infecções por Vírus Epstein-Barr/tratamento farmacológico , Inibidores da Protease de HIV/farmacologia , Linfoma de Células B/tratamento farmacológico , NF-kappa B/efeitos dos fármacos , Ritonavir/farmacologia , Animais , Apoptose/efeitos dos fármacos , Western Blotting , Ciclo Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Ensaio de Desvio de Mobilidade Eletroforética , Infecções por Vírus Epstein-Barr/complicações , Herpesvirus Humano 4 , Humanos , Hibridização in Situ Fluorescente , Linfoma de Células B/virologia , Camundongos , NF-kappa B/metabolismo , Reação em Cadeia da Polimerase , Ativação Transcricional/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Cancer Sci ; 100(5): 807-12, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19309363

RESUMO

The Epstein-Barr virus-encoded latent membrane protein 1 is considered the Epstein-Barr virus oncogene based on its importance in Epstein-Barr virus-induced B-lymphocyte transformation. Beta-catenin is a potential oncogene, and its accumulation has been implicated in a variety of human cancers. Here, we found that beta-catenin protein was highly expressed in Epstein-Barr virus-immortalized B-cell lines compared with peripheral blood mononuclear cells from healthy donors. Beta-catenin expression in Epstein-Barr virus-immortalized B-cell line decreased following treatment with LY294002, an inhibitor of phosphatidylinositol 3-kinase. Treatment with LY294002 or knockdown of beta-catenin by small interfering RNA reduced the growth of Epstein-Barr virus-immortalized B-cell line. Transient transfection of latent membrane protein 1 expression plasmid increased beta-catenin protein expression and beta-catenin-dependent transcription. Latent membrane protein 1 deletions mutants lacking the carboxyl-terminal activating region 1 domain failed to enhance beta-catenin protein expression and beta-catenin-dependent transcriptional activity. They also failed to increase phosphorylated AKT expression. Dominant-negative AKT suppressed latent membrane protein 1-induced beta-catenin-dependent transcriptional activity. These results suggest that latent membrane protein 1 activates beta-catenin through the phosphatidylinositol 3-kinase/AKT signaling pathway. Activation of the beta-catenin pathway by Epstein-Barr virus may contribute to the lymphoproliferation characteristic of Epstein-Barr virus-infected B-cells.


Assuntos
Linfócitos B/metabolismo , Herpesvirus Humano 4/metabolismo , Transdução de Sinais , Proteínas da Matriz Viral/metabolismo , beta Catenina/metabolismo , Linfócitos B/citologia , Linhagem Celular , Proliferação de Células , Herpesvirus Humano 4/genética , Humanos , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fatores de Transcrição TCF/metabolismo , Transcrição Gênica/genética , Proteínas da Matriz Viral/genética , beta Catenina/genética
4.
Cancer Sci ; 100(4): 778-81, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19245436

RESUMO

CXC chemokine receptor-4, the receptor for stromal cell-derived factor-1alpha as well as human immunodeficiency virus type 1, belongs to the chemokine receptor family and has been shown to play a critical role in directing the migration of cancer cells to sites of metastasis as well as human immunodeficiency virus type 1 infection. We had previously reported that a duodenally absorbable CXC chemokine receptor-4 antagonist, KRH-1636, showed a potent anti-human immunodeficiency virus type 1 activity both in vivo and in vitro. In this study, we initially examined the effect of the compound and its derivatives on stromal cell-derived factor-1alpha-mediated chemotaxis of cancer cells in order to evaluate if they could be applicable as a novel inhibitor of cancer metastasis. We found that both KRH-2731 and KRH-3955 were highly potent antagonists of stromal cell-derived factor-1alpha-mediated chemotaxis, i.e. the derivatives exhibited 50% effective concentrations of less than 10 nM, for more than 1000-fold efficacy improvement over the prototype KRH-1636. We further demonstrated the greater anti-human immunodeficiency virus type 1 efficacy of the derivatives compared with the original KRH-1636. Taken together, the KRH-1636 derivatives KRH-2731 and KRH-3955 may be promising as a novel inhibitory drug for cancer metastasis as well as for human immunodeficiency virus type 1 infection.


Assuntos
Fármacos Anti-HIV/farmacologia , Quimiocina CXCL12/farmacologia , Quimiotaxia de Leucócito/efeitos dos fármacos , HIV-1/efeitos dos fármacos , Receptores CXCR4/antagonistas & inibidores , Arginina/análogos & derivados , Arginina/farmacologia , Benzilaminas/farmacologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Relação Dose-Resposta a Droga , Infecções por HIV/prevenção & controle , Humanos , Imidazóis/farmacologia , Concentração Inibidora 50 , Células Jurkat , Piridinas/farmacologia
5.
Microbes Infect ; 10(5): 471-80, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18406652

RESUMO

Latent infection of human immunodeficiency virus type 1 (HIV-1) represents a major hurdle in the treatment of acquired immunodeficiency syndrome (AIDS) patients. Statins were recently reported to suppress acute HIV-1 infection and reduce infectious virion production, but the precise mechanism of inhibition has remained elusive. Here we demonstrate that lypophilic statins suppress HIV-1 virion release from tumor necrosis factor alpha-stimulated latently infected U1 cells through inhibition of protein geranylgeranylation, but not by cholesterol depletion. Indeed, this suppression was reversed by the addition of geranylgeranylpyrophosphate, and a geranylgeranyltransferase-1 inhibitor reduced HIV-1 production. Notably, silencing of the endogenous Rab11a GTPase expression in U1 cells by RNA interference destabilized Gag and reduced virion production both in vitro and in NOD/SCID/gammac null mice. Our findings thus suggest that small GTPase proteins play an important role in HIV-1 replication, and therefore could be attractive molecular targets for anti-HIV-1 therapy.


Assuntos
HIV-1/efeitos dos fármacos , HIV-1/fisiologia , Prenilação de Proteína , Sinvastatina/farmacologia , Replicação Viral/efeitos dos fármacos , Alquil e Aril Transferases/antagonistas & inibidores , Animais , Linhagem Celular Tumoral , Proteína do Núcleo p24 do HIV/metabolismo , Infecções por HIV/metabolismo , HIV-1/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Microscopia Eletrônica de Transmissão , Fosfatos de Poli-Isoprenil/metabolismo , Precursores de Proteínas/metabolismo , Interferência de RNA , Fator de Necrose Tumoral alfa/farmacologia , Proteínas rab de Ligação ao GTP/metabolismo
6.
Microbes Infect ; 10(7): 748-56, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18538617

RESUMO

Epstein-Barr virus (EBV) causes EBV-associated lymphoproliferative diseases in patients with profound immune suppression. Most of these diseases are life-threatening and the prognosis of AIDS-associated lymphomas is extremely unfavorable. Polyclonal expansion of virus infected B-cell predisposes them to transformation. We investigated the possibility of nuclear factor kappa B (NF-kappaB) inhibition by dehydroxymethylepoxyquinomicin (DHMEQ) for the treatment and prevention of EBV-associated lymphoproliferative diseases. We examined the effect of DHMEQ on apoptosis induction in four EBV-transformed lymphoblastoid cell lines as well as peripheral blood mononuclear cells infected with EBV under immunosuppressed condition. DHMEQ inhibits NF-kappaB activation in EBV-transformed lymphoblastoid cell lines and induces apoptosis by activation of mitochondrial and membranous pathways. Using an in vivo NOD/SCIDgammac mouse model, we showed that DHMEQ has a potent inhibitory effect on the growth of lymphoblastoid cells. In addition, DHMEQ selectively purges EBV-infected cells expressing latent membrane protein (LMP) 1 from peripheral blood mononuclear cells and inhibits the outgrowth of lymphoblastoid cells. These results suggest that NF-kappaB is a molecular target for the treatment and prevention of EBV-associated lymphoproliferative diseases. As a potent NF-kappaB inhibitor, DHMEQ is a potential compound for applying this strategy in clinical medicine.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Linfócitos B/virologia , Benzamidas/farmacologia , Cicloexanonas/farmacologia , Herpesvirus Humano 4/fisiologia , Fatores Imunológicos/farmacologia , NF-kappa B/antagonistas & inibidores , Animais , Linhagem Celular Tumoral , Sobrevivência Celular , Humanos , Transtornos Linfoproliferativos/tratamento farmacológico , Camundongos , Camundongos SCID
7.
Breast Cancer ; 18(1): 64-7, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20354831

RESUMO

A suggestive case of metastatic disease from breast cancer is reported. The HER-2-positive tumor was refractory to several agents, including anti-HER-2 therapy, trastuzumab, and lapatinib. After re-induction of trastuzumab in combination with activated natural killer (NK) cell injection therapy, tumor markers decreased, and finally a synergistic effect of taxane and capecitabine led to treatment response. This case suggests that multidisciplinary therapy including an immunological approach might be a breakthrough in the treatment of refractory disease.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias da Mama/terapia , Carcinoma Ductal de Mama/terapia , Células Matadoras Naturais/transplante , Neoplasias Pulmonares/terapia , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais Humanizados , Neoplasias da Mama/imunologia , Neoplasias da Mama/patologia , Hidrocarbonetos Aromáticos com Pontes/administração & dosagem , Capecitabina , Carcinoma Ductal de Mama/imunologia , Carcinoma Ductal de Mama/secundário , Terapia Combinada , Desoxicitidina/administração & dosagem , Desoxicitidina/análogos & derivados , Resistencia a Medicamentos Antineoplásicos , Feminino , Fluoruracila/administração & dosagem , Fluoruracila/análogos & derivados , Humanos , Lapatinib , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/secundário , Pessoa de Meia-Idade , Prognóstico , Quinazolinas/administração & dosagem , Receptor ErbB-2/metabolismo , Terapia de Salvação , Taxoides/administração & dosagem , Trastuzumab
8.
Biomed Pharmacother ; 63(9): 703-6, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19269774

RESUMO

Natural killer (NK) activity of immune cells plays a central role in host defense against cancer and virus-infected cells. Natural cytotoxic activity of peripheral-blood mononuclear cells was assessed by a Calcein-AM release assay in 89 subjects. In the present study, we here demonstrated that NK activities of peripheral-blood mononuclear cells (PBMCs) from breast cancer patients were significantly lower as compared with that of healthy individuals. There were significant differences in the NK activities of PBMCs from HER2-negative breast cancer patients as compared with HER2-positive patients. Our results suggest that NK activity of PBMCs is lower in breast cancer indicating a role for immunological natural host defense mechanisms against cancer.


Assuntos
Neoplasias da Mama/imunologia , Células Matadoras Naturais/imunologia , Leucócitos Mononucleares/imunologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Feminino , Humanos , Células K562 , Pessoa de Meia-Idade
9.
Breast Cancer Res Treat ; 104(3): 267-75, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17066321

RESUMO

Natural killer (NK) cells play a central role in host defense against tumor and virus-infected cells. Direct role of NK cells in tumor growth and metastasis remains to be elucidated. We here demonstrated that NOD/SCID/gammac(null) (NOG) mice lacking T, B and NK cells inoculated with breast cancer cells were efficient in the formation of a large tumor and spontaneous organ-metastasis. In contrast, breast cancer cells produced a small tumor at inoculated site in T and B cell knock-out NOD/SCID mice with NK cells while completely failed to metastasize into various organs. Immunosupression of NOD/SCID by treatment with an anti-murine TM-beta1 antibody, which transiently abrogates NK cell activity in vivo, resulted in enhancing tumor formation and organ-metastasis in comparison with non-treated NOD/SCID mice. Activated NK cells inhibited tumor growth in vivo. The rapid and efficient engraftment of the breast cancer cells in NOG mice suggests that this new animal model could provide a unique opportunity to understand and investigate the mechanism of tumor cell growth and metastasis. Our results suggest that NK cells play an important role in cancer growth and metastasis and could be a promising immunotherapeutic strategy against cancer either alone or in combination with conventional therapy.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Células Matadoras Naturais/metabolismo , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Citometria de Fluxo , Humanos , Imunossupressores/farmacologia , Imunoterapia/métodos , Células K562 , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Metástase Neoplásica , Transplante de Neoplasias
10.
Lab Invest ; 87(4): 372-82, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17310217

RESUMO

Constitutive nuclear factor kappaB (NF-kappaB) activation characterizes Hodgkin/Reed-Sternberg (H-RS) cells. Blocking constitutive NF-kappaB has been shown to be a potential strategy to treat Hodgkin lymphoma (HL). Here, for the first time we show that although constitutive NF-kappaB level of H-RS cell lines is very high, topoisomerase inhibitors further enhance NF-kappaB activation through IkappaB kinase activation in not only H-RS cell lines with wild-type IkappaBalpha, but also in those with IkappaBalpha mutations and lacking wild-type IkappaBalpha. Thus, both constitutive and inducible NF-kappaB are potential targets to treat HL. We also present the data that indicate the involvement of IkappaBbeta in NF-kappaB induction by topoisomerase inhibitors. A new NF-kappaB inhibitor, dehydroxymethylepoxyquinomicin (DHMEQ) inhibited constitutive NF-kappaB activity and induced apoptosis of H-RS cell lines. DHMEQ also inhibited the growth of H-RS cells without significant systemic toxicity in a NOD/SCID/gammac(null) (NOG) mice model. DHMEQ and topoisomerase inhibitors revealed enhancement of apoptosis of H-RS cells by blocking inducible NF-kappaB. Results of this study suggest that both constitutive and inducible NF-kappaB are molecular targets of DHMEQ in the treatment of HL. The results also indicate that IkappaBbeta is involved in NF-kappaB activation in H-RS cells and IkappaBbeta substitutes for IkappaBalpha in H-RS cells lacking wild-type IkappaBalpha.


Assuntos
Benzamidas/farmacologia , Cicloexanonas/farmacologia , Proteínas I-kappa B/metabolismo , NF-kappa B/metabolismo , Células de Reed-Sternberg/metabolismo , Animais , Apoptose , Caspases/metabolismo , Linhagem Celular , Daunorrubicina/farmacologia , Ativação Enzimática , Etoposídeo/farmacologia , Doença de Hodgkin , Humanos , Quinase I-kappa B/metabolismo , Proteínas I-kappa B/genética , Proteínas I-kappa B/fisiologia , Leucócitos Mononucleares/metabolismo , Camundongos , Mutação , Inibidor de NF-kappaB alfa , NF-kappa B/antagonistas & inibidores , Transplante de Neoplasias , Inibidores da Topoisomerase , Transplante Heterólogo , Células Tumorais Cultivadas
11.
Cancer Sci ; 97(12): 1381-7, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16995875

RESUMO

Natural killer (NK) cells are an important component of the innate immune response against microbial infections and tumors. Direct involvement of NK cells in tumor growth and infiltration has not yet been demonstrated clearly. Primary effusion lymphoma (PEL) cells were able to produce tumors and ascites very efficiently with infiltration of cells in various organs of T-, B- and NK-cell knock-out NOD/SCID/gammac(null) (NOG) mice within 3 weeks. In contrast, PEL cells formed small tumors at inoculated sites in T- and B-cell knock-out NOD/SCID mice with NK-cells while completely failing to infiltrate into various organs. Immunosupression of NOD/SCID by treatment with an antimurine TM-beta1 antibody, which transiently abrogates NK cell activity in vivo, resulted in enhanced tumorigenicity and organ infiltration in comparison with non-treated NOD/SCID mice. Activated human NK cells inhibited tumor growth and infiltration in NOG mice. Our results suggest that NK cells play an important role in growth and infiltration of PEL cells, and activated NK cells could be a promising immunotherapeutic tool against tumor or virus-infected cells either alone or in combination with conventional therapy. The rapid and efficient engraftment of PEL cells in NOG mice also suggests that this new animal model could provide a unique opportunity to understand and investigate the mechanism of pathogenesis and malignant cell growth.


Assuntos
Síndrome da Imunodeficiência Adquirida/complicações , Linfócitos B/metabolismo , HIV-1/patogenicidade , Células Matadoras Naturais/imunologia , Linfoma/terapia , Derrame Pleural Maligno/terapia , Linfócitos T/metabolismo , Animais , Linfócitos B/imunologia , Proliferação de Células , Células Cultivadas/patologia , Células Cultivadas/transplante , Células Cultivadas/virologia , Modelos Animais de Doenças , Citometria de Fluxo , Humanos , Técnicas Imunoenzimáticas , Células Matadoras Naturais/metabolismo , Células Matadoras Naturais/transplante , Depleção Linfocítica , Linfócitos do Interstício Tumoral , Linfoma/imunologia , Linfoma/virologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Neoplasias Experimentais/etiologia , Neoplasias Experimentais/patologia , Neoplasias Experimentais/terapia , Derrame Pleural Maligno/imunologia , Derrame Pleural Maligno/virologia , Linfócitos T/imunologia
12.
Int J Cancer ; 119(4): 823-30, 2006 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-16557588

RESUMO

Human T-cell leukemia virus type-I (HTLV-I) encodes for the viral protein Tax, which is known to significantly disrupt transcriptional control of cytokines, cytokine receptors and other immuno-modulatory proteins in T cells. Specific dysregulation of these factors can alter the course and pathogenesis of infection. Soluble interleukin-6 receptor (sIL-6R) was shown to circulate at elevated levels in HTLV-I-infected patients, and high expressions of IL-6R and sIL-6R by HTLV-I-infected T cells were clinically and experimentally associated with Tax activity. To examine roles of Tax in expression of the IL-6R gene, the JPX-9 cell line was used, which is derived from Jurkat cell line expressing Tax cDNA. Over-expression of Tax enhanced IL-6R expression but not in Tax mutant JPX-9/M cell line. The clinical relevance of these observations was further demonstrated by ELISA using sera obtained from HTLV-I-infected patients. Our results revealed that sIL-6R levels were apparently elevated in HAM/TSP patients who were expressing Tax in their cells, while ATL patients' cells barely expressed Tax. HTLV-I-infected T-cell lines stimulated by IL-6/sIL-6R showed gp130-mediated STAT3 activity. IL-6/sIL-6R enhanced proliferation of HTLV-I-infected T cells in association with activation of STAT3. Consequently, Tax-mediated regulations of IL-6R and sIL-6R observed in HTLV-I-associated disorders may contribute to proliferation of HTLV-I-infected T cells through activation of inducible STAT3, and ultimately affect malignant growth and transformation of T cells by HTLV-I.


Assuntos
Regulação da Expressão Gênica , Produtos do Gene tax/metabolismo , Vírus Linfotrópico T Tipo 1 Humano/fisiologia , Receptores de Interleucina-6/metabolismo , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Linhagem Celular , Proliferação de Células , Meios de Cultura , Humanos , Interleucina-6/farmacologia , Receptores de Interleucina-6/genética , Solubilidade
13.
Med Microbiol Immunol ; 194(4): 175-80, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15654627

RESUMO

Human cells have developed innate immunity, exploiting several means to block virus infection, and viruses have evolved diverse strategies to resist these. We show here that the human immunodeficiency virus 1 (HIV-1) could neither progressively infect engrafted human leukemic T cells nor repress their growth in NOG mice. However, ED-40515(-) cells infected with HIV-1 before inoculation were found to significantly delay the onset of tumor growth and increased the survival period of NOG mice. ED-40515(-) tumor cells showed resistance to HIV-1 which was apparently correlated with the down-regulation of CD4 and CXCR4 molecules in NOG mice. Serum from three different mouse strains, including NOG, retained a suppressive effect on the CD4 molecule of ED-40515(-) cells in vitro. ED-40515(-) cells obtained from mice re-expressed CD4 and CXCR4 molecules upon in vitro culture and were again successfully infected with HIV-1. These findings indicate that HIV-1 may initially successfully delay or regress tumor growth in NOG mice, but eventually fails to do so because of the evolution of HIV-resistant cells due to a rapid down-modulation of CD4 and CXCR4. Our data also demonstrated that some unknown soluble factor(s) present in mouse serum was responsible for conferring resistance to HIV infection to human T cells.


Assuntos
Fatores Biológicos/fisiologia , Antígenos CD4/metabolismo , Linhagem Celular Transformada/virologia , Modelos Animais de Doenças , Infecções por HIV/imunologia , HIV-1 , Receptores CXCR4/metabolismo , Soro/fisiologia , Linfócitos T/imunologia , Animais , Proteínas de Transporte , Linhagem Celular Transformada/imunologia , Regulação para Baixo , Humanos , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Linfócitos T/metabolismo , Linfócitos T/virologia
14.
Cancer Sci ; 96(8): 466-73, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16108827

RESUMO

As there are very few reproducible animal models without conditioning available for the study of human B-cell-type Hodgkin's lymphoma (HL), we investigated the ability of HL cells to induce tumors using novel NOD/SCID/gammac(null) (NOG) mice. Four human Epstein-Barr virus-negative cell lines (KM-H2 and L428 originated from B cells, L540 and HDLM2 originated from T cells) were inoculated either subcutaneously in the postauricular region or intravenously in the tail of unmanipulated NOG mice. All cell lines successfully engrafted and produced tumors with infiltration of cells in various organs of all mice. Tumor cells had classical histomorphology as well as expression patterns of the tumor marker CD30, which is a cell surface antigen expressed on HL. Tumor progression in mice inoculated with B-cell-type, but not T-cell-type, HL cells correlated with an elevation in serum human interleukin-6 levels. Tumor cells from the mice also retained strong nuclear factor (NF)-kappaB DNA binding activity, and the induced NF-kappaB components were indistinguishable from those cultured in vitro. The reproducible growth behavior and preservation of characteristic features of both B-cell-type and T-cell-type HL in the mice suggest that this new xenotransplant model can provide a unique opportunity to understand and investigate the mechanism of pathogenesis and malignant cell growth, and to develop novel anticancer therapies.


Assuntos
Doença de Hodgkin/patologia , Antígeno Ki-1/genética , NF-kappa B/genética , Animais , Divisão Celular , Linhagem Celular Tumoral , Humanos , Interleucina-6/sangue , Células Jurkat , Metástase Linfática , Camundongos , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Metástase Neoplásica , Transplante Heterólogo/métodos
15.
Int J Cancer ; 114(1): 32-8, 2005 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-15523684

RESUMO

Multiple myeloma (MM) is a fatal lymphoid malignancy that is incurable with conventional modalities of chemotherapy. Strong and constitutive activation of nuclear factor kappa B (NF-kappaB) is a common characteristic of MM cells. In our study we successfully target NF-kappaB with a novel NF-kappaB inhibitor dehydroxymethylepoxyquinomycin (DHMEQ). DHMEQ completely abrogates constitutive NF-kappaB activity and induces apoptosis of MM cells, whereas control peripheral blood mononuclear cells (PBMC) are resistant to NF-kappaB inhibition and apoptosis by DHMEQ treatment. DHMEQ inhibition of NF-kappaB triggers activation of caspases 8 and 9, as well as G0/G1 cell cycle arrest accompanied by downregulation of antiapoptotic genes Bcl-XL and c-FLIP and cell cycle progression gene cyclins D1 and D2. DHMEQ-mediated inhibition of vascular endothelial growth factor (VEGF) production in MM cells raises the possibility that DHMEQ abrogates the autocrine VEGF loop and enhances its antitumor effects by inhibiting neovascularization in the bone marrow. Using an in vivo NOD/SCID/gammac(null) (NOG) mice model, we show that DHMEQ has a potent inhibitory effect on the growth of MM cells. Compared to other compounds having the potential to inhibit NF-kappaB, DHMEQ is a unique compound that blocks the translocation of NF-kappaB p65 into the nucleus and selectively targets NF-kappaB activated in tumor cells. Therefore, our study presents a new molecular target therapy in MM.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Benzamidas/farmacologia , Cicloexanonas/farmacologia , Mieloma Múltiplo/tratamento farmacológico , NF-kappa B/antagonistas & inibidores , Animais , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD , Proteínas de Ligação ao Cálcio , Caspase 3 , Caspase 8 , Caspase 9 , Caspases/metabolismo , Linhagem Celular Tumoral , Ciclina D , Ciclina D1/metabolismo , Ciclinas/metabolismo , Modelos Animais de Doenças , Regulação para Baixo , Ativação Enzimática/efeitos dos fármacos , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Glicoproteínas de Membrana , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Mieloma Múltiplo/metabolismo , Proteínas do Tecido Nervoso , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Sinaptotagmina I , Sinaptotagminas , Translocação Genética/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Proteína bcl-X
16.
Cancer Sci ; 95(7): 564-8, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15245591

RESUMO

Clinically and biologically relevant animal models are indispensable to evaluate both the pathophysiology and strategies for diagnosis and treatment of multiple myeloma (MM). We examined the tumorigenicity of MM cell lines KMM-1 and U-266 in an in vivo cell proliferation model using NOD/SCID/gammacnull (NOG) mice. Two cell lines were inoculated either subcutaneously (s.c.) in the post-auricular region or intravenously (i.v.) in the tail of NOG mice. The KMM-1 cell line produced a progressively growing large tumor with infiltration of the cells expressing human lambda-chain in various organs of all NOG mice, while the U-266 cell line failed to do so. Tumor cells grown in NOG mice maintained the original histomorphology, as well as expression patterns of tumor markers human lambda Ig light chain and VEGF. Tumor progression in mice also correlated with elevation of serum human soluble IL-6R and gp130. Tumor cells sustained a strong NF-kappaB activity in vivo and induced NF-kappaB components were indistinguishable from those in cells cultured in vitro. The rapid and efficient engraftment of the MM cell line in NOG mice suggests that this is a very useful animal model which could provide a novel system in which to clarify the mechanism of growth of cancer cells, as well as to develop new therapeutic regimens against MM.


Assuntos
Modelos Animais de Doenças , Camundongos SCID , Mieloma Múltiplo/fisiopatologia , Mieloma Múltiplo/veterinária , NF-kappa B/farmacologia , Animais , Divisão Celular , Progressão da Doença , Humanos , Camundongos , Camundongos Endogâmicos NOD , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA