Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
J Immunol ; 184(11): 6529-36, 2010 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-20421638

RESUMEN

Src homology region 2 domain-containing phosphatase 1 (SHP-1) has been implicated as a potential cancer therapeutic target by its negative regulation of immune cell activation and the activity of the SHP-1 inhibitor sodium stibogluconate that induced IFN-gamma(+) cells for anti-tumor action. To develop more potent SHP-1-targeted anti-cancer agents, inhibitory leads were identified from a library of 34,000 drug-like compounds. Among the leads and active at low nM for recombinant SHP-1, tyrosine phosphatase inhibitor-1 (TPI-1) selectively increased SHP-1 phospho-substrates (pLck-pY394, pZap70, and pSlp76) in Jurkat T cells but had little effects on pERK1/2 or pLck-pY505 regulated by phosphatases SHP-2 or CD45, respectively. TPI-1 induced mouse splenic-IFN-gamma(+) cells in vitro, approximately 58-fold more effective than sodium stibogluconate, and increased mouse splenic-pLck-pY394 and -IFN-gamma(+) cells in vivo. TPI-1 also induced IFN-gamma(+) cells in human peripheral blood in vitro. Significantly, TPI-1 inhibited ( approximately 83%, p < 0.002) the growth of B16 melanoma tumors in mice at a tolerated oral dose in a T cell-dependent manner but had little effects on B16 cell growth in culture. TPI-1 also inhibited B16 tumor growth and prolonged tumor mice survival as a tolerated s.c. agent. TPI-1 analogs were identified with improved activities in IFN-gamma(+) cell induction and in anti-tumor actions. In particular, analog TPI-1a4 as a tolerated oral agent completely inhibited the growth of K1735 melanoma tumors and was more effective than the parental lead against MC-26 colon cancer tumors in mice. These results designate TPI-1 and the analogs as novel SHP-1 inhibitors with anti-tumor activity likely via an immune mechanism, supporting SHP-1 as a novel target for cancer treatment.


Asunto(s)
Antineoplásicos/administración & dosificación , Neoplasias Experimentales/tratamiento farmacológico , Proteína Tirosina Fosfatasa no Receptora Tipo 6/antagonistas & inhibidores , Administración Oral , Animales , Western Blotting , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Humanos , Células Jurkat , Activación de Linfocitos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL
2.
Blood Cells Mol Dis ; 44(4): 209-14, 2010 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-20226699

RESUMEN

Dual specificity protein tyrosine phosphatase PRL-2 is overexpressed in pediatric acute myeloid leukemia (AML) and is located at human chromosome 1p35, a region often rearranged or amplified in malignant lymphoma and B-cell chronic lymphocytic leukemia (B-CLL). Little is known of the significance of PRL-2 expression in hematopoietic malignancies. Herein we demonstrated that ectopic expression of PRL-2 in murine pre-B-cell line Baf3ER and mouse bone marrow cells induced key features associated with malignant progression and metastasis. PRL-2-transfected Baf3ER cells had augmented growth responses to hematopoietic growth factors Epo or IL-3 with shortened cell cycle, reduced requirement (5x) for Epo in cell survival, increased cell migration (3x), reduced cell adhesion (5x), and conversion to an immature cell morphology in association with increased expression (3x) of stem cell marker Bmi-1. When transduced into mouse bone marrow cells, PRL-2 increased Epo-induced colony formation (4x) and gave rise to larger colonies. These observations provide evidences implicating PRL-2 as a pathogenic molecule in hematopoietic malignancies and suggest its potential as a novel therapeutic target.


Asunto(s)
Eritropoyetina/farmacología , Interleucina-3/farmacología , Células Precursoras de Linfocitos B/efectos de los fármacos , Proteínas Tirosina Fosfatasas/fisiología , Animales , Células de la Médula Ósea/citología , Adhesión Celular/efectos de los fármacos , Desdiferenciación Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Línea Celular/citología , Línea Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Ensayo de Unidades Formadoras de Colonias , Epoetina alfa , Neoplasias Hematológicas/etiología , Humanos , Ratones , Ratones Endogámicos C57BL , Células Precursoras de Linfocitos B/citología , Proteínas Tirosina Fosfatasas/genética , Proteínas Recombinantes de Fusión/fisiología , Proteínas Recombinantes , Factor de Transcripción STAT5/biosíntesis , Factor de Transcripción STAT5/genética , Transducción Genética
4.
Curr Oncol Rep ; 10(2): 114-21, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18377824

RESUMEN

Protein tyrosine phosphatases (PTPases) are attractive targets for developing novel cancer therapeutics. Activated via gain-of-function point mutations or overexpression, several PTPases have been identified as critical oncogenic molecules in human malignancies that may be targeted with small chemical inhibitors as a therapeutic strategy. Tumor suppressor PTPases have also been discovered as contributing factors in cancer development that may be targeted via intervention of downstream signaling events for therapeutic purposes. In addition, PTPases have been identified as key negative regulators of cytokines or immune cells. Targeting these negative PTPases may improve the efficacy of cytokine therapy and immunotherapy, which currently have modest response rates and limited survival benefit. Inhibitors of selective PTPases have demonstrated significant preclinical antitumor activity, leading to early-phase clinical trials. Further research and development could lead to PTPase-targeted cancer therapeutics in the near future.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Neoplasias/metabolismo , Proteínas Tirosina Fosfatasas/fisiología , Antineoplásicos/farmacología , Inhibidores Enzimáticos/farmacología , Genes Supresores de Tumor , Humanos , Sistema de Señalización de MAP Quinasas , Neoplasias/enzimología , Monoéster Fosfórico Hidrolasas/fisiología , Mutación Puntual , Proteínas Tirosina Fosfatasas/metabolismo , Transducción de Señal , Resultado del Tratamiento
5.
Cancer Chemother Pharmacol ; 60(3): 341-9, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17103170

RESUMEN

BACKGROUND: Pre-clinical activity of SSG against melanoma and renal cancer has been identified recently although the drug's mechanism of action and activity against tumors of additional histological-types remain undefined. METHODS: The effects of SSG and SSG combination with other agents on DU145 human prostate carcinoma xenograft tumors in mice and on DU145 cell subpopulations of differential SSG sensitivities were evaluated. RESULTS: DU145 tumor growth was inhibited by SSG (69%), IFNalpha2 (33%) or the combination (80%) that induced complete regression of WM9 human melanoma tumors. DU145 cells in culture were also partially growth inhibited by SSG at killing doses (200-800 mug/ml) for WM9 cells, indicating a correlation of SSG inhibition of cancer cell growth in vitro and in vivo. DU145 cells formed multiple micro tumors in mice treated with SSG or SSG/IFNalpha2 in contrast to the single large tumors in the control or IFNalpha2-treated mice, suggesting the existence of an SSG-resistant subpopulation in DU145 cells. Indeed, DU145 but not WM9 cells formed colonies (approximately 4% frequency) when cultured in the presence of SSG. Single cell clone (DU145-7) isolated from DU145 cells showed SSG-resistant growth in culture, unassociated with cross-resistance to IFNalpha2 and converted to SSG-responsive cells by BSO that inhibited intracellular glutathione levels. CONCLUSIONS: These results implicate a role for direct growth inhibition in SSG anti-tumor action, provide novel insights into the mechanism of tumor resistance to the drug and suggest a therapeutic potential for SSG and its combinations with IFNalpha2 or BSO for prostate cancer that warrants further investigation.


Asunto(s)
Gluconato de Sodio Antimonio/uso terapéutico , Interferón-alfa/uso terapéutico , Neoplasias de la Próstata/tratamiento farmacológico , Animales , Gluconato de Sodio Antimonio/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , División Celular/efectos de los fármacos , Línea Celular Tumoral , Femenino , Humanos , Interferón-alfa/administración & dosificación , Masculino , Melanoma , Ratones , Ratones Desnudos , Neoplasias de la Próstata/patología , Trasplante Heterólogo
6.
Mol Cancer Ther ; 1(14): 1255-64, 2002 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12516958

RESUMEN

The PRL family oncogenic phosphatases are attractive targets for developing inhibitors as anticancer therapeutics given their potentially pathogenic role in human malignancies. Herein we demonstrate that pentamidine, an anti-protozoa drug with an unknown mechanism of action, is an inhibitor of PRLs with anticancer potential. Pentamidine at its therapeutic doses inhibited recombinant PRL phosphatases in vitro and inactivated ectopically expressed PRLs in NIH3T3 transfectants with an effective duration more than 24 h after a pulse cell treatment. The drug had in vitro growth-inhibitory activity against human cancer cell lines that express the endogenous PRLs. Pentamidine at a tolerable dose markedly inhibited the growth of WM9 human melanoma tumors in nude mice coincident with the induction of tumor cell necrosis and is capable of inactivating ectopically expressed PRL-2 in the cancer cells. These observations suggest the potential of pentamidine in anticancer therapies and may provide a basis for developing novel PTPase-targeted therapeutics.


Asunto(s)
Antineoplásicos/farmacología , Pentamidina/farmacología , Proteínas Tirosina Fosfatasas/metabolismo , Células 3T3 , Animales , Antifúngicos/farmacología , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Humanos , Ratones , Ratones Desnudos , Fosforilación , Proteínas Recombinantes/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo , Transfección , Células Tumorales Cultivadas
7.
J Interferon Cytokine Res ; 23(12): 745-56, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-14769151

RESUMEN

Interferon (IFN)-dependent cellular effects are mediated by transcriptional induction of responsive genes, collectively referred to as IFN-stimulated genes (ISGs). Which ISGs regulate the potent antiviral, antiproliferative, apoptosis-inducing, antiangiogenic, and immunologic effects of IFNs remains largely undetermined. To identify genes that might be useful for predicting or targeting apoptosis induction in response to IFNs, WM9 melanoma cells were assessed. WM9 cells had equivalent antiviral activity in response to IFN-beta and IFN-alpha2 but underwent apoptosis only in response to IFN-beta. RNA samples from WM9 cells and WM35 cells, a second melanoma cell line, treated with IFN-alpha2 or IFN-beta were assessed on oligonucleotide arrays. For 95% of genes assessed, IFN-beta was more potent than IFN-alpha2 in inducing ISG expression. Using a 22,000-gene oligonucleotide array, the largest yet reported for assessing ISG induction, approximately 910 genes were identified as induced by IFN-beta at 500 U/ml, and 260 ISGs were identified as significantly induced by IFN-beta at both 50 and 500 U/ml. Of these 260, 209 were defined as new ISGs based on the array analysis. Confirmation by Northern blot or semiquantitative or quantitative PCR was undertaken for 28, and all were confirmed. Nearly half of the 260 genes were functionally categorized as encoding growth-regulatory proteins. Of the 104 with described growth-regulatory function, 71 were induced more than three times by 500 U/ml and twice by 50 U/ml IFN-beta, and 48 of these were new ISGs. Included in this latter category were tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL), XIAP-associated factor 1 (XAF1), galectin 9, a cyclin E binding protein, amphiphysin 1, MyD88, and several ubiquitin pathway genes. The diversity of stimulated genes suggests the full therapeutic potential of IFN regulation of gene expression has yet to be realized.


Asunto(s)
Apoptosis/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Interferón-alfa/farmacología , Interferón beta/farmacología , Melanoma/genética , Melanoma/patología , Apoptosis/genética , Proteínas Reguladoras de la Apoptosis , Línea Celular Tumoral , Galectinas/genética , Perfilación de la Expresión Génica , Humanos , Cinética , Glicoproteínas de Membrana/genética , Proteínas de la Membrana/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ligando Inductor de Apoptosis Relacionado con TNF , Factor de Necrosis Tumoral alfa/genética
8.
Chin Med J (Engl) ; 115(10): 1457-9, 2002 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12490086

RESUMEN

OBJECTIVE: To confirm the existence of point mutations in the SSU rDNA variable regions of 5 Leishmania donovani (L.d.) isolates from different epidemic foci in China. METHODS: Specific SSU rDNA fragments from nuclear DNA of 7 Leishmania species/isolates were amplified by PCR and then cloned into pGEM(R)-T Easy Vectors. After that, the specific fragments were sequenced by an automated DNA sequencer. RESULTS: Sequence analysis showed that the amplified DNA fragments of 7 Leishmania species/isolates were all 392 bp in length. All 5 point mutations were located in two unique sequence blocks (UQ-I and UQ-II), and no insertions or deletions were found. The identities of comparison of Leishmania in GeneBank were more than 98%. CONCLUSION: Five point mutations exist in the SSU rDNA variable region of 5 L.d. isolates from different epidemic foci of visceral leishmaniasis (VL) in China. Sequence differences of the SSU rDNA variable region exist among L.d. isolates from different foci.


Asunto(s)
ADN Protozoario/química , ADN Ribosómico/química , Leishmania donovani/genética , Leishmaniasis Visceral/parasitología , Mutación Puntual , Animales , Humanos , Reacción en Cadena de la Polimerasa
9.
Oncotarget ; 2(12): 1155-64, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22201704

RESUMEN

Since sodium stibogluconate (SSG) inhibited phosphatases including SHP-1 and augmented anti-tumor actions of IFN-α2b in vitro and in mice, two Phase I trials of SSG/IFN-α2b combination were undertaken to evaluate safety and target inhibition. Escalating doses of SSG (200-1200 mg/m2) and fixed doses of IFN-α2b (3x106 units/m2) with or without chemotherapy (dacarbazine, vinblastine, cisplatin) were evaluated for side effects and impact on SHP-1 phospho-substrates and IFNα-stimulated-genes (ISGs) in peripheral blood in 40 patients with metastatic melanoma, soft tissue sarcomas, gastrointestinal stromal tumors, and breast or colorectal carcinomas who did not have other established treatment options. Common adverse events were bone marrow suppression, fatigue, gastrointestinal upset, and asymptomatic lipase elevation (n=13); the latter was dose related and mostly after 10d of SSG/IFN-α2b in combination. Levels of SHP-1 substrates (pSTAT1, pSTAT3, pLck and pSlp76) were increased (up to 3x) in peripheral blood cells following SSG with no potentiation by combination with IFN-α2b. Representative ISGs in peripheral blood were induced after IFN-α2b at 4 and 24 hrs with selective modulations by combination. The median time on trials was 2.3 months (10-281d) with no objective regression of disease. Alive at 1y were 17/40 (43%) patients and after 2y were 8/40 (20%) following treatment initiation. These data demonstrate that SSG impacted signal molecules consistent with PTP inhibition and was tolerated in combination with IFN-α2b. Phase II investigations of SSG could safely utilize doses of up to 1200 mg/m2 of SSG for up to 10d alone or in combination with IFN-α2b with or without chemotherapy.


Asunto(s)
Gluconato de Sodio Antimonio , Quimioterapia Combinada/efectos adversos , Quimioterapia Combinada/métodos , Interferón-alfa/uso terapéutico , Adulto , Anciano , Gluconato de Sodio Antimonio/administración & dosificación , Gluconato de Sodio Antimonio/efectos adversos , Gluconato de Sodio Antimonio/farmacocinética , Gluconato de Sodio Antimonio/uso terapéutico , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Cisplatino/farmacología , Neoplasias Colorrectales/tratamiento farmacológico , Dacarbazina/farmacología , Inhibidores Enzimáticos/uso terapéutico , Femenino , Tumores del Estroma Gastrointestinal/tratamiento farmacológico , Humanos , Interferón alfa-2 , Interferón-alfa/administración & dosificación , Masculino , Melanoma/tratamiento farmacológico , Persona de Mediana Edad , Monoéster Fosfórico Hidrolasas/antagonistas & inhibidores , Monoéster Fosfórico Hidrolasas/sangre , Proteína Tirosina Fosfatasa no Receptora Tipo 6/antagonistas & inhibidores , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/uso terapéutico , Sarcoma/tratamiento farmacológico , Vinblastina/farmacología
10.
Mol Cancer Ther ; 9(8): 2287-96, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20682647

RESUMEN

Drug resistance is a major obstacle in cancer treatments and diminishes the clinical efficacy of biological, cytotoxic, or targeted therapeutics. Being an antiapoptotic mediator of chemoresistance in breast and lung cancer cells, MKP1 phosphatase might be targeted for overcoming chemoresistance and improving therapeutic efficacy. In this work, tyrosine phosphatase inhibitor-3 (TPI-3) was identified as a novel small molecule inhibitor of MKP1 and was capable of sensitizing tumors to bio- and chemotherapeutics in mice as a tolerated oral agent. Effective against recombinant MKP1, TPI-3 selectively increased MKP1 phosphosubstrates in Jurkat cells and induced cell death via apoptosis at nanomolar concentrations. TPI-3 also increased MKP1 phosphosubstrates in WM9 human melanoma cells and synergized with biotherapeutic IFN alpha 2b in the growth inhibition of melanoma cells in vitro (combination index, <1). WM9 xenografts unresponsive to individual agents were significantly inhibited (62%, P = 0.001) in mice by a tolerated combination of oral TPI-3 (10 mg/kg, 5 d/wk) and IFN alpha 2b. MKP1 expression was detected in human melanoma cell lines and tissue samples at levels up to six times higher than those in normal or nonmalignant melanocytes. TPI-3 also interacted positively with chemotherapeutics, 5-fluorouracil/leucovorin, against MC-26 colon cancer cells in vitro and in mice. Altogether, our data show the preclinical activities of TPI-3 in overcoming cancer resistance to bio- and chemotherapeutics, implicate MKP1 as a drug-resistant molecule in melanoma, and support the targeting of MKP1 for improving cancer therapeutic efficacy.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias del Colon/tratamiento farmacológico , Inhibidores Enzimáticos/uso terapéutico , Interferón-alfa/uso terapéutico , Melanoma/tratamiento farmacológico , Tiazoles/uso terapéutico , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Neoplasias del Colon/enzimología , Neoplasias del Colon/patología , Resistencia a Antineoplásicos/efectos de los fármacos , Sinergismo Farmacológico , Fosfatasa 1 de Especificidad Dual/antagonistas & inhibidores , Fosfatasa 1 de Especificidad Dual/metabolismo , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Fluorouracilo/farmacología , Fluorouracilo/uso terapéutico , Humanos , Interferón alfa-2 , Interferón-alfa/farmacología , Melanoma/enzimología , Melanoma/patología , Ratones , Proteínas Recombinantes , Especificidad por Sustrato/efectos de los fármacos , Tiazoles/química , Tiazoles/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
11.
J Interferon Cytokine Res ; 29(8): 451-60, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19514839

RESUMEN

Sodium stibogluconate (SSG), an inhibitor of SHP-1 that negatively regulates cytokine signaling and immunity, suppressed growth of murine Renca tumors in combination with interleukin-2 (IL-2) via a T-cell-dependent mechanism. The ability of SSG to interact with IL-2 in activating primary human immune cells was evaluated herein by assessing its induction of interferon (IFN)-gamma(+) TH1 cells in human peripheral blood in vitro. The significance of IFN-gamma(+) cells was also investigated by assessing SSG/IL-2 antitumor activity in wild-type and IFN-gamma(-/-) mice. IFN-gamma(+) cells but not IL-5(+) cells were induced markedly (9.1x) in healthy peripheral blood by SSG/IL-2 in contrast to the modest induction by SSG alone (2.1x) at its clinically achievable dose (20 microg/mL) or by IL-2 (3.1x) at its C(max) of low-dose schedule (30 IU/mL). SSG at a higher dose (100 microg/mL) was less effective alone (1.5x) or in combination with IL-2 (7.8x). Peripheral IFN-gamma(+) cells were induced after 4 or 16 h treatment with SSG/IL-2 within CD4(+) and CD8(+) lymphocytes coincided with heightened CD69 expression (approximately 3-4x). SSG/IL-2 was also more effective than the single agents in inducing IFN-gamma(+) cells in the peripheral blood of melanoma patients, whose basal IFN-gamma(+) cell levels were approximately 5% of healthy controls. Renca tumor growth was inhibited by SSG/IL-2 in wild-type but not IFN-gamma(-/-) mice. These results demonstrate SSG interactions with IL-2 in vitro to activate key antitumor immune cells in peripheral blood of healthy and melanoma donors, providing further evidence for proof of concept clinical trials for effecting augmentation of IL-2 through inhibiting negative regulatory protein tyrosine phosphatases.


Asunto(s)
Antígenos CD/metabolismo , Antígenos de Diferenciación de Linfocitos T/metabolismo , Gluconato de Sodio Antimonio/farmacología , Antineoplásicos/farmacología , Inhibidores de Crecimiento/farmacología , Interferón gamma/metabolismo , Interleucina-2/metabolismo , Interleucina-5/metabolismo , Neoplasias Renales/inmunología , Leucocitos Mononucleares/inmunología , Melanoma/inmunología , Animales , Antígenos CD/genética , Antígenos de Diferenciación de Linfocitos T/genética , Gluconato de Sodio Antimonio/administración & dosificación , Antineoplásicos/administración & dosificación , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Células Cultivadas , Relación Dosis-Respuesta a Droga , Femenino , Inhibidores de Crecimiento/administración & dosificación , Humanos , Inmunofenotipificación , Interferón gamma/genética , Interferón gamma/inmunología , Interleucina-2/inmunología , Interleucina-5/genética , Interleucina-5/inmunología , Neoplasias Renales/patología , Neoplasias Renales/terapia , Lectinas Tipo C , Leucocitos Mononucleares/metabolismo , Leucocitos Mononucleares/patología , Activación de Linfocitos/efectos de los fármacos , Masculino , Melanoma/patología , Melanoma/terapia , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Trasplante de Neoplasias , Proteína Tirosina Fosfatasa no Receptora Tipo 6/antagonistas & inhibidores , Transducción de Señal
12.
Biochem Biophys Res Commun ; 342(2): 547-55, 2006 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-16487932

RESUMEN

Several lines of evidence have suggested that protein tyrosine phosphatases, including CD45 and SHP-1, regulate macrophage activation. Macrophages from mice lacking SHP-1 (motheaten mice) are hyper-responsive to many stimuli, suggesting that SHP-1 may negatively regulate macrophage activation. Herein we report that the repressible/inducible over-expression of wild-type SHP-1 in a subclone of RAW 264.7 macrophages (RAW-TT10 cells) inhibited both TNF secretion and iNOS protein accumulation in response to stimulation with lipopolysaccharide (LPS) and recombinant murine interferon-gamma and led to diminished LPS-mediated tyrosine phosphorylation of vav1. In contrast, expression of a truncated SHP-1 construct previously shown to interfere with endogenous SHP-1 function modestly augmented LPS-mediated TNF and iNOS production and did not inhibit vav1 tyrosine phosphorylation. Taken together, these data provide the first direct evidence that SHP-1 inhibits macrophage activation by LPS and suggest that this effect may be mediated in part by dephosphorylation of vav1.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/fisiología , Lipopolisacáridos/farmacología , Macrófagos/enzimología , Óxido Nítrico Sintasa de Tipo II/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo II/biosíntesis , Proteínas Tirosina Fosfatasas/fisiología , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/biosíntesis , Animales , Línea Celular , Células Cultivadas , Células Clonales , Inhibidores Enzimáticos/farmacología , Interferón gamma/farmacología , Activación de Macrófagos/inmunología , Macrófagos/inmunología , Ratones , Fosforilación , Proteína Tirosina Fosfatasa no Receptora Tipo 6 , Proteínas Proto-Oncogénicas c-vav/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-vav/metabolismo , Proteínas Recombinantes , Tetraciclina/farmacología , Transfección
13.
J Immunol ; 175(10): 7003-8, 2005 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-16272361

RESUMEN

IL-2 therapy results in 10-20% response rates in advanced renal cell carcinoma (RCC) via activating immune cells, in which the protein tyrosine phosphatase Src homology 2 domain-containing phosphatase 1 (SHP-1) is a key negative regulator. Based on finding that sodium stibogluconate (SSG) inhibited SHP-1, the anti-RCC potential and action mechanism of SSG and SSG/IL-2 in combination were investigated in a murine renal cancer model (Renca). Despite its failure to inhibit Renca cell proliferation in cultures, SSG induced 61% growth inhibition of Renca tumors in BALB/c mice coincident with an increase (2-fold) in tumor-infiltrating macrophages (Mphi). A combination of SSG and IL-2 was more effective in inhibiting tumor growth (91%) and inducing tumor-infiltrating Mphi (4-fold), whereas IL-2 alone had little effect. Mphi increases were also detected in the spleens of mice treated with SSG (3-fold) or SSG/IL-2 in combination (6-fold), suggesting a systemic Mphi expansion similar to those in SHP-deficient mice. T cell involvement in the anti-Renca tumor action of the combination was suggested by the observations that the treatment induced spleen IFN-gamma T cells in BALB/c mice, but failed to inhibit Renca tumor growth in athymic nude mice and that SSG treatment of T cells in vitro increased production of IFN-gamma capable of activating tumoricidal Mphi. The SSG and SSG/IL-2 combination treatments were tolerated in the mice. These results together demonstrate an anti-Renca tumor activity of SSG that was enhanced in combination with IL-2 and functions via a T cell-dependent mechanism with increased IFN-gamma production and expansion/activation of Mphi. Our findings suggest that SSG might improve anti-RCC efficacy of IL-2 therapy by enhancing antitumor immunity.


Asunto(s)
Gluconato de Sodio Antimonio/administración & dosificación , Carcinoma de Células Renales/tratamiento farmacológico , Interleucina-2/administración & dosificación , Neoplasias Renales/tratamiento farmacológico , Animales , Carcinoma de Células Renales/inmunología , Carcinoma de Células Renales/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Quimioterapia Combinada , Inhibidores Enzimáticos/administración & dosificación , Femenino , Factores Inmunológicos/administración & dosificación , Interferón gamma/biosíntesis , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Neoplasias Renales/inmunología , Neoplasias Renales/patología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/patología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Proteína Fosfatasa 1 , Proteína Tirosina Fosfatasa no Receptora Tipo 6 , Proteínas Tirosina Fosfatasas/antagonistas & inhibidores , Proteínas Recombinantes/administración & dosificación , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Linfocitos T/patología
14.
Blood Cells Mol Dis ; 28(1): 63-74, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-11987243

RESUMEN

SHP-1 protein tyrosine phosphatase is a critical regulator of signaling in hematopoietic cells as illustrated by the lethal hematopoietic disorders in SHP-1-deficient mice. We and others have shown in previous studies that SHP-1 regulates membrane receptor signaling: it binds via its N-terminal region SH2 domains to tyrosine phosphorylated membrane receptors to dephosphorylate key substrates in the receptor complexes. Here we demonstrate that the SHP-1 C-terminal region contains a bipartite NLS that mediates SHP-1 nuclear localization in response to cytokines. This NLS was located within amino acids 576-595 of the PTPase and, when fused by itself to EGFP, targeted the fluorescent protein into the nuclei of transiently transfected NIH3T3 fibroblasts and Bac1.2f5 macrophage cells. When positioned within SHP-1, the activity of the NSL was under tight regulation as indicated by the predominant cytoplasmic distribution of the EGFP/SHP-1 fusion protein in NIH3T3 transfectants and the exclusive cytoplasmic localization of the endogenous SHP-1 in hematopoietic cell line PBLC-1. Activation of the NLS in SHP-1 by IL-4 was demonstrated by increased nuclear localization of the EGFP/SHP-1 fusion protein in NIH3T3 transfectants and of the endogenous SHP-1 protein in PBCL-1 cells at 4, 6 and 8 h post-IL-4 stimulation. SHP-1 nuclear localization in PBCL-1 cells was also induced by IL-7 in a similar manner, suggesting it as a common event in cytokine signaling. In comparison to that of the wild-type phosphatase, an SHP-1 mutant lacking the NLS showed only approximately half of the activity in inhibiting proliferation of NIH3T3 transfectants. These results provide evidence of cytokine-regulated SHP-1 nuclear localization mediated by a bipartite NLS and suggest that SHP-1 regulates nuclear signaling in cell growth control.


Asunto(s)
Transporte Activo de Núcleo Celular , Citocinas/farmacología , Señales de Localización Nuclear/fisiología , Proteínas Tirosina Fosfatasas/metabolismo , Células 3T3 , Transporte Activo de Núcleo Celular/efectos de los fármacos , Animales , Secuencia de Bases , División Celular/efectos de los fármacos , Células Madre Hematopoyéticas/metabolismo , Interleucina-4/farmacología , Interleucina-7/farmacología , Péptidos y Proteínas de Señalización Intracelular , Ratones , Microscopía Fluorescente , Señales de Localización Nuclear/efectos de los fármacos , Proteína Tirosina Fosfatasa no Receptora Tipo 6 , Proteínas Tirosina Fosfatasas/química , Proteínas Tirosina Fosfatasas/genética , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Transfección
15.
Biochem Biophys Res Commun ; 302(4): 646-52, 2003 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-12646217

RESUMEN

The Src-homology 2 domain containing protein tyrosine phosphatase-1 (SHP-1) is involved in the pathogenesis of infection with Leishmania. Recently, we identified elongation factor-1 alpha (EF-1 alpha) from Leishmania donovani as a SHP-1 binding and activating protein [J. Biol. Chem. 277 (2002) 50190]. To characterize this apparent Leishmania virulence factor further, the cDNA encoding L. donovani EF-1 alpha was cloned and sequenced. Whereas nearly complete sequence conservation was observed amongst EF-1 alpha proteins from trypanosomatids, the deduced amino acid sequence of EF-1 alpha of L. donovani when compared to mammalian EF-1 alpha sequences showed a number of significant changes. Protein structure modeling-based upon the known crystal structure of EF-1 alpha for Saccharomyces cerevisiae-identified a hairpin loop present in mammalian EF-1 alpha and absent from the Leishmania protein which corresponded to a 12 amino acid deletion. Consistent with these structural differences, the sub-cellular distributions of L. donovani EF-1 alpha and host EF-1 alpha were strikingly different. Interestingly, infection of macrophages with L. donovani caused redistribution of host as well as pathogen EF-1 alpha. Since EF-1 alpha is essential for survival, the distinct biochemical and structural properties of Leishmania EF-1 alpha may provide a novel target for drug development.


Asunto(s)
Leishmania donovani/metabolismo , Factor 1 de Elongación Peptídica/genética , Proteínas Protozoarias/química , Proteínas Protozoarias/genética , Secuencia de Aminoácidos , Animales , Fraccionamiento Celular , Clonación Molecular , Activación Enzimática , Humanos , Péptidos y Proteínas de Señalización Intracelular , Leishmaniasis Visceral/sangre , Leishmaniasis Visceral/inmunología , Macrófagos/citología , Macrófagos/metabolismo , Macrófagos/parasitología , Modelos Moleculares , Datos de Secuencia Molecular , Peso Molecular , Factor 1 de Elongación Peptídica/química , Factor 1 de Elongación Peptídica/metabolismo , Proteína Fosfatasa 1 , Estructura Terciaria de Proteína , Proteína Tirosina Fosfatasa no Receptora Tipo 6 , Proteínas Tirosina Fosfatasas/metabolismo , Proteínas Protozoarias/metabolismo , Alineación de Secuencia
16.
J Biol Chem ; 277(51): 50190-7, 2002 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-12384497

RESUMEN

The human leishmaniasis are persistent infections of macrophages caused by protozoa of the genus Leishmania. The chronic nature of these infections is in part related to induction of macrophage deactivation, linked to activation of the Src homology 2 domain containing tyrosine phosphatase-1 (SHP-1) in infected cells. To investigate the mechanism of SHP-1 activation, lysates of Leishmania donovani promastigotes were subjected to SHP-1 affinity chromatography and proteins bound to the matrix were sequenced by mass spectrometry. This resulted in the identification of Leishmania elongation factor-1alpha (EF-1alpha) as a SHP-1-binding protein. Purified Leishmania EF-1alpha, but not host cell EF-1alpha, bound directly to SHP-1 in vitro leading to its activation. Three independent lines of evidence indicated that Leishmania EF-1alpha may be exported from the phagosome thereby enabling targeting of host SHP-1. First, cytosolic fractions prepared from macrophages infected with [(35)S]methionine-labeled organisms contained Leishmania EF-1alpha. Second, confocal, fluorescence microscopy using Leishmania-specific antisera detected Leishmania EF-1alpha in the cytosol of infected cells. Third, co-immunoprecipitation showed that Leishmania EF-1alpha was associated with SHP-1 in vivo in infected cells. Finally, introduction of purified Leishmania EF-1alpha, but not the corresponding host protein into macrophages activated SHP-1 and blocked the induction of inducible nitric-oxide synthase expression in response to interferon-gamma. Thus, Leishmania EF-1alpha is identified as a novel SHP-1-binding and activating protein that recapitulates the deactivated phenotype of infected macrophages.


Asunto(s)
Leishmania donovani/metabolismo , Macrófagos/metabolismo , Factor 1 de Elongación Peptídica/metabolismo , Proteínas Tirosina Fosfatasas/metabolismo , Animales , Línea Celular , Citosol/metabolismo , Glutatión Transferasa/metabolismo , Immunoblotting , Péptidos y Proteínas de Señalización Intracelular , Leishmania donovani/patogenicidad , Macrófagos/parasitología , Espectrometría de Masas , Ratones , Microscopía Fluorescente , Óxido Nítrico Sintasa/metabolismo , Óxido Nítrico Sintasa de Tipo II , Fenotipo , Pruebas de Precipitina , Unión Proteica , Proteína Fosfatasa 1 , Estructura Terciaria de Proteína , Proteína Tirosina Fosfatasa no Receptora Tipo 6 , Proteínas Tirosina Fosfatasas/química , Dominios Homologos src
17.
J Biol Chem ; 278(19): 16520-7, 2003 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-12615921

RESUMEN

Stat5A, a member of the signal transducers and activators of transcription (Stat) family, is activated upon a single tyrosine phosphorylation. Although much is known about the activation process, the mechanism by which the tyrosine-phosphorylated Stat5A proteins are inactivated is largely unknown. In this report, we demonstrate that down-regulation of the tyrosine-phosphorylated Stat5A was via dephosphorylation. Using tyrosine-phosphorylated peptides derived from Stat5A, we were able to purify protein-tyrosine phosphatase Shp-2 from cell lysates. Shp-2, but not Shp-1, specifically interacted with Stat5A in vivo, and the interaction was tyrosine phosphorylation-dependent. Moreover, Shp-2 was able to accelerate Stat5A dephosphorylation, and dephosphorylation of Stat5A was dramatically delayed in Shp-2-deficient cells. Therefore, we conclude that Shp-2 is a Stat5A phosphatase, which down-regulates the active Stat5A in vivo.


Asunto(s)
Proteínas de Unión al ADN/análisis , Proteínas de la Leche , Proteínas Tirosina Fosfatasas/análisis , Transactivadores/análisis , Animales , Células COS , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Regulación hacia Abajo , Péptidos y Proteínas de Señalización Intracelular , Ratones , Proteína Tirosina Fosfatasa no Receptora Tipo 11 , Proteínas Tirosina Fosfatasas/química , Proteínas Tirosina Fosfatasas/genética , Proteínas Tirosina Fosfatasas/metabolismo , Proteínas Tirosina Fosfatasas con Dominio SH2 , Factor de Transcripción STAT5 , Especificidad por Sustrato , Transactivadores/química , Transactivadores/genética , Transactivadores/metabolismo , Dominios Homologos src
18.
J Immunol ; 169(10): 5978-85, 2002 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-12421984

RESUMEN

Cancer cell resistance limits the efficacy of IFNs. In this study, we show that sodium stibogluconate (SSG) and IFN-alpha synergized to overcome IFN-alpha resistance in various human cancer cell lines in culture and eradicated IFN-alpha-refractory WM9 human melanoma tumors in nude mice with no obvious toxicity. SSG enhanced IFN-alpha-induced Stat1 tyrosine phosphorylation, inactivated intracellular SHP-1 and SHP-2 that negatively regulate IFN signaling, and induced cellular protein tyrosine phosphorylation in cancer cell lines. These effects are consistent with inactivation of phosphatases as the basis of SSG anticancer activity. Characterization of SSG by chromatography revealed that only selective compounds in SSG were effective protein tyrosine phosphatase inhibitors. These observations suggest the potential of SSG as a clinically usable protein tyrosine phosphatase inhibitor in cancer treatment and provide insights for developing phosphatase-targeted therapeutics.


Asunto(s)
Gluconato de Sodio Antimonio/farmacología , Antineoplásicos/farmacología , Interferón-alfa/farmacología , Interferón beta/farmacología , Adyuvantes Inmunológicos/farmacología , Adyuvantes Inmunológicos/uso terapéutico , Animales , Gluconato de Sodio Antimonio/uso terapéutico , Antineoplásicos/uso terapéutico , División Celular/efectos de los fármacos , Proteínas de Unión al ADN/metabolismo , Resistencia a Antineoplásicos , Sinergismo Farmacológico , Quimioterapia Combinada , Inhibidores de Crecimiento/farmacología , Inhibidores de Crecimiento/uso terapéutico , Humanos , Interferón-alfa/uso terapéutico , Péptidos y Proteínas de Señalización Intracelular , Linfoma/tratamiento farmacológico , Linfoma/metabolismo , Linfoma/patología , Melanoma/tratamiento farmacológico , Melanoma/patología , Ratones , Ratones Desnudos , Fosforilación/efectos de los fármacos , Proteína Tirosina Fosfatasa no Receptora Tipo 11 , Proteínas Tirosina Fosfatasas/antagonistas & inhibidores , Proteínas Tirosina Fosfatasas/metabolismo , Factor de Transcripción STAT1 , Transactivadores/metabolismo , Células Tumorales Cultivadas/efectos de los fármacos , Células Tumorales Cultivadas/enzimología , Células Tumorales Cultivadas/metabolismo , Células Tumorales Cultivadas/patología
19.
Blood ; 100(5): 1852-9, 2002 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-12176909

RESUMEN

Fcgamma receptor-mediated phagocytosis is a complex process involving the activation of protein tyrosine kinases, events that are potentially down-regulated by protein tyrosine phosphatases. We used the J774A.1 macrophage cell line to examine the roles played by the protein tyrosine phosphatase SHP-1 in the negative regulation of Fcgamma receptor-mediated phagocytosis. Stimulation with sensitized sheep red blood cells (sRBCs) induced tyrosine phosphorylation of CBL and association of CBL with CRKL. These events were completely or partially abrogated by PP1 or the heterologous expression of dominant-negative SYK, respectively. Heterologous expression of wild-type but not catalytically inactive SHP-1 also completely abrogated the phagocytosis of IgG-sensitized sRBCs. Most notably, overexpressed SHP-1 associates with CBL and this association led to CBL dephosphorylation, loss of the CBL-CRKL interaction, and the suppression of Rac activation. These data represent the first direct evidence that SHP-1 is involved in the regulation of Fcgamma receptor-mediated phagocytosis and suggest that activating signals mediated by SRC family kinases SYK, CBL, phosphatidyl inositol-3 (PI-3) kinase, and Rac are directly opposed by inhibitory signals through SHP-1.


Asunto(s)
Fagocitosis/fisiología , Proteínas Tirosina Fosfatasas/fisiología , Receptores de IgG/fisiología , Proteínas de Unión al GTP rac/fisiología , Animales , Línea Celular , Regulación de la Expresión Génica/fisiología , Péptidos y Proteínas de Señalización Intracelular , Macrófagos/fisiología , Ratones , Proteína Tirosina Fosfatasa no Receptora Tipo 6 , Transducción de Señal , Proteínas de Unión al GTP rac/antagonistas & inhibidores , Dominios Homologos src/fisiología
20.
J Biol Chem ; 279(5): 3245-53, 2004 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-14600148

RESUMEN

Over the past decade, a wealth of knowledge has been obtained concerning the mechanisms by which interferons (IFNs) and other cytokines activate or down-regulate immediate early genes via the Jak/Stat pathway. In contrast, little information is available on interferon-activated gene expression in naïve cells compared with cells that have been desensitized and subsequently resensitized to the actions of these cytokines. In naïve cells, the ISG54 gene is activated via IFN beta-stimulated formation of ISGF3, a heterotrimeric DNA binding complex consisting of p48 (IRF9) and tyrosine-phosphorylated Stat1 and Stat2. In contrast, in previously desensitized cells IFN beta weakly stimulates the assembly of an ISGF3-like complex that lacks Stat1, even though ISG54 mRNA induction is the same as in naïve cells. The lack of Stat1 tyrosine phosphorylation and DNA binding is due to increased activity of a protein-tyrosine phosphatase. In cells that do not express the tyrosine phosphatase Tc-PTP, the rate of Stat1 dephosphorylation is the same in naïve and previously desensitized cells. These results implicate Tc-PTP in a novel role in the regulation of type 1 interferon-stimulated gene expression.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Interferón Tipo I/farmacología , Proteínas Tirosina Fosfatasas/fisiología , Transactivadores/metabolismo , Animales , Western Blotting , Línea Celular , Núcleo Celular/metabolismo , Células Cultivadas , Cromatina/metabolismo , Citoplasma/metabolismo , Regulación hacia Abajo , Regulación de la Expresión Génica , Humanos , Inmunoglobulinas/metabolismo , Interferón Tipo I/química , Interferón beta/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Fosforilación , Pruebas de Precipitina , Unión Proteica , Proteína Tirosina Fosfatasa no Receptora Tipo 1 , Proteína Tirosina Fosfatasa no Receptora Tipo 2 , Proteínas Tirosina Fosfatasas/metabolismo , ARN/metabolismo , ARN Mensajero/metabolismo , Factor de Transcripción STAT1 , Factores de Tiempo , Tirosina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA