Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Blood ; 139(26): 3708-3721, 2022 06 30.
Artigo em Inglês | MEDLINE | ID: mdl-35090171

RESUMO

Pivotal clinical trials of B-cell maturation antigen-targeted chimeric antigen receptor T (CART)-cell therapy in patients with relapsed/refractory multiple myeloma (MM) resulted in remarkable initial responses, which led to a recent US Food and Drug Administration approval. Despite the success of this therapy, durable remissions continue to be low, and the predominant mechanism of resistance is loss of CART cells and inhibition by the tumor microenvironment (TME). MM is characterized by an immunosuppressive TME with an abundance of cancer-associated fibroblasts (CAFs). Using MM models, we studied the impact of CAFs on CART-cell efficacy and developed strategies to overcome CART-cell inhibition. We showed that CAFs inhibit CART-cell antitumor activity and promote MM progression. CAFs express molecules such as fibroblast activation protein and signaling lymphocyte activation molecule family-7, which are attractive immunotherapy targets. To overcome CAF-induced CART-cell inhibition, CART cells were generated targeting both MM cells and CAFs. This dual-targeting CART-cell strategy significantly improved the effector functions of CART cells. We show for the first time that dual targeting of both malignant plasma cells and the CAFs within the TME is a novel strategy to overcome resistance to CART-cell therapy in MM.


Assuntos
Fibroblastos Associados a Câncer , Mieloma Múltiplo , Medula Óssea , Fibroblastos Associados a Câncer/patologia , Terapia Baseada em Transplante de Células e Tecidos , Fibroblastos , Humanos , Imunoterapia Adotiva/métodos , Mieloma Múltiplo/patologia , Microambiente Tumoral
2.
Blood ; 133(7): 697-709, 2019 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-30463995

RESUMO

Chimeric antigen receptor T (CAR-T) cell therapy is a new pillar in cancer therapeutics; however, its application is limited by the associated toxicities. These include cytokine release syndrome (CRS) and neurotoxicity. Although the IL-6R antagonist tocilizumab is approved for treatment of CRS, there is no approved treatment of neurotoxicity associated with CD19-targeted CAR-T (CART19) cell therapy. Recent data suggest that monocytes and macrophages contribute to the development of CRS and neurotoxicity after CAR-T cell therapy. Therefore, we investigated neutralizing granulocyte-macrophage colony-stimulating factor (GM-CSF) as a potential strategy to manage CART19 cell-associated toxicities. In this study, we show that GM-CSF neutralization with lenzilumab does not inhibit CART19 cell function in vitro or in vivo. Moreover, CART19 cell proliferation was enhanced and durable control of leukemic disease was maintained better in patient-derived xenografts after GM-CSF neutralization with lenzilumab. In a patient acute lymphoblastic leukemia xenograft model of CRS and neuroinflammation (NI), GM-CSF neutralization resulted in a reduction of myeloid and T cell infiltration in the central nervous system and a significant reduction in NI and prevention of CRS. Finally, we generated GM-CSF-deficient CART19 cells through CRISPR/Cas9 disruption of GM-CSF during CAR-T cell manufacturing. These GM-CSFk/o CAR-T cells maintained normal functions and had enhanced antitumor activity in vivo, as well as improved overall survival, compared with CART19 cells. Together, these studies illuminate a novel approach to abrogate NI and CRS through GM-CSF neutralization, which may potentially enhance CAR-T cell function. Phase 2 studies with lenzilumab in combination with CART19 cell therapy are planned.


Assuntos
Citocinas/metabolismo , Fator Estimulador de Colônias de Granulócitos e Macrófagos/antagonistas & inibidores , Doenças do Sistema Imunitário/terapia , Inflamação/terapia , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Receptores de Antígenos de Linfócitos T/uso terapêutico , Receptores de Antígenos Quiméricos/imunologia , Animais , Anticorpos Neutralizantes/farmacologia , Proliferação de Células , Humanos , Doenças do Sistema Imunitário/imunologia , Doenças do Sistema Imunitário/metabolismo , Inflamação/imunologia , Inflamação/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Camundongos , Leucemia-Linfoma Linfoblástico de Células Precursoras/imunologia , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Receptores de Antígenos Quiméricos/metabolismo , Síndrome , Transplante Heterólogo , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Cancer Cell ; 10(1): 65-75, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16843266

RESUMO

Tyrosine kinases are aberrantly activated in numerous malignancies, including acute myeloid leukemia (AML). To identify tyrosine kinases activated in AML, we developed a screening strategy that rapidly identifies tyrosine-phosphorylated proteins using mass spectrometry. This allowed the identification of an activating mutation (A572V) in the JAK3 pseudokinase domain in the acute megakaryoblastic leukemia (AMKL) cell line CMK. Subsequent analysis identified two additional JAK3 alleles, V722I and P132T, in AMKL patients. JAK3(A572V), JAK3(V722I), and JAK3(P132T) each transform Ba/F3 cells to factor-independent growth, and JAK3(A572V) confers features of megakaryoblastic leukemia in a murine model. These findings illustrate the biological importance of gain-of-function JAK3 mutations in leukemogenesis and demonstrate the utility of proteomic approaches to identifying clinically relevant mutations.


Assuntos
Leucemia Experimental/genética , Leucemia Megacarioblástica Aguda/genética , Proteínas Tirosina Quinases/genética , Alelos , Animais , Apoptose/efeitos dos fármacos , Benzamidas , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Humanos , Mesilato de Imatinib , Janus Quinase 2 , Janus Quinase 3 , Células K562 , Leucemia Experimental/metabolismo , Leucemia Experimental/patologia , Leucemia Megacarioblástica Aguda/metabolismo , Leucemia Megacarioblástica Aguda/patologia , Camundongos , Camundongos Endogâmicos C57BL , Modelos Moleculares , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Fosforilação/efeitos dos fármacos , Piperazinas/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Estrutura Terciária de Proteína/genética , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Pirimidinas/farmacologia , RNA Interferente Pequeno/genética , TYK2 Quinase
4.
Eur J Haematol ; 91(6): 504-13, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23992230

RESUMO

The introduction of novel immunomodulatory drugs (IMiDs) has dramatically improved the survival of patients with multiple myeloma (MM). While it has been shown that patients with specific cytogenetic subtypes, namely t(4;14), have the best outcomes when treated with bortezomib-based regimens, the relationship between cytogenetic subtypes and response to IMiDs remains unclear. Using DNA synthesis assays, we investigated the relationship between cytogenetic subtype and lenalidomide response in a representative panel of human myeloma cell lines (HMCLs). We examined HMCL protein expression levels of the lenalidomide target cereblon (CRBN) and its downstream target interferon regulatory factor-4 (IRF4), which have previously been shown to be predictive of lenalidomide response in HMCLs. Our results reveal that lenalidomide response did not correlate with specific cytogenetic translocations. There were distinct groups of lenalidomide-responsive and non-responsive HMCLs, as defined by inhibition of cellular proliferation; notably, all of the hyperdiploid HMCLs fell into the latter category. Repeated dosing of lenalidomide significantly lowered the IC50 of the responsive HMCL ALMC-1 (IC50 = 2.6 µm vs. 0.005 µm, P < 0.0001), but did not have an effect on the IC50 of the non-responsive DP-6 HMCL (P > 0.05). Moreover, no association was found between lenalidomide responsiveness and CRBN and IRF4 expression. Our data indicate that lenalidomide sensitivity is independent of cytogenetic subtype in HMCLs. While CRBN and IRF4 have been shown to be associated with response to lenalidomide in patients, these findings do not translate back to HMCLs, which could be attributable to factors present in the bone marrow microenvironment.


Assuntos
Fatores Imunológicos/farmacologia , Fatores Reguladores de Interferon/genética , Mieloma Múltiplo/genética , Peptídeo Hidrolases/genética , Talidomida/análogos & derivados , Proteínas Adaptadoras de Transdução de Sinal , Apoptose/efeitos dos fármacos , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Aberrações Cromossômicas , Relação Dose-Resposta a Droga , Expressão Gênica , Humanos , Fatores Reguladores de Interferon/metabolismo , Lenalidomida , Mieloma Múltiplo/tratamento farmacológico , Talidomida/farmacologia , Ubiquitina-Proteína Ligases
5.
Cancer Cell ; 7(2): 117-9, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15710324

RESUMO

The Abl inhibitor imatinib is a highly effective therapy for patients with chronic myeloid leukemia. Relapses are relatively uncommon in newly diagnosed patients, but are the rule in patients with more advanced disease. Mutations in the BCR-ABL gene are the most common cause of relapse. Working from the imatinib chemical structure, a higher-affinity family member, AMN107, was designed. AMN107 is approximately 20-fold more potent than imatinib, and this translates into improved inhibitory activity against most of the common BCR-ABL mutations. The implications of these results, and the potential role this and other novel ABL inhibitors may have in treating patients with CML, are discussed.


Assuntos
Antineoplásicos/farmacologia , Leucemia Mielogênica Crônica BCR-ABL Positiva/tratamento farmacológico , Piperazinas/farmacologia , Pirimidinas/química , Pirimidinas/farmacologia , Animais , Benzamidas , Linhagem Celular Tumoral , Proteínas de Fusão bcr-abl/genética , Proteínas de Fusão bcr-abl/metabolismo , Humanos , Mesilato de Imatinib , Modelos Químicos , Mutação , Piperazinas/química , Recidiva
6.
Int J Cancer ; 131(5): E804-12, 2012 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-22120774

RESUMO

Osteosarcoma (OS) is the most frequent primary bone tumor. Despite multiagent neoadjuvant chemotherapy, patients with metastatic disease have a poor prognosis. Moreover, currently used chemotherapeutics have severe toxic side effects. Thus, novel agents with improved antimetastatic activity and reduced toxicity are needed. Taurolidine, a broad-spectrum antimicrobial, has recently been shown to have antineoplastic properties against a variety of tumors and low systemic toxicity. Consequently, we investigated in our study the antineoplastic potential of taurolidine against OS in two different mouse models. Although both OS cell lines, K7M2 and LM8, were sensitive for the compound in vitro, intraperitoneal application of taurolidine failed to inhibit primary tumor growth. Moreover, it enhanced the metastatic load in both models 1.7- to 20-fold and caused severe liver deformations and up to 40% mortality. Thus, systemic toxicity was further investigated in tumor-free mice histologically, by electron microscopy and by measurements of representative liver enzymes. Taurolidine dose-dependent fibrous thickening of the liver capsule and adhesions and atrophies of the liver lobes were comparable in healthy and tumor-bearing mice. Liver toxicity was further indicated by up to eightfold elevated levels of the liver enzymes alanine transaminase, aspartate transaminase and GLDH in the circulation. Ultrastructural analysis of affected liver tissue showed swollen mitochondria with cristolysis and numerous lipid vacuoles in the cytoplasm of hepatocytes. The findings of our study question the applicability of taurolidine for OS treatment and may suggest the need for caution regarding the widespread clinical use of taurolidine as an antineoplastic agent.


Assuntos
Antibióticos Antineoplásicos/efeitos adversos , Neoplasias Ósseas/tratamento farmacológico , Doença Hepática Induzida por Substâncias e Drogas/etiologia , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Pulmonares/tratamento farmacológico , Osteossarcoma/tratamento farmacológico , Taurina/análogos & derivados , Tiadiazinas/efeitos adversos , Animais , Neoplasias Ósseas/patologia , Feminino , Neoplasias Hepáticas/secundário , Neoplasias Pulmonares/secundário , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Osteossarcoma/patologia , Taurina/efeitos adversos , Células Tumorais Cultivadas
7.
Cancer ; 118(8): 2117-27, 2012 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-21935912

RESUMO

BACKGROUND: Metastasizing osteosarcoma has a mean 5-year survival rate of only 20% to 30%. Therefore, novel chemotherapeutics for more effective treatment of this disease are required. METHODS: The antineoplastic activity of honokiol, which was demonstrated previously in numerous malignancies, was studied in vivo in C3H mice subcutaneously injected with syngeneic ß-galactosidase bacterial gene (lacZ)-expressing LM8 osteosarcoma (LM8-lacZ) cells. In vitro cytotoxic effects of honokiol were investigated in 8 human and 2 murine osteosarcoma cell lines with different in vivo metastatic potential. RESULTS: Seven days after subcutaneous flank injection of LM8-lacZ cells, daily intraperitoneal treatment of mice with 150 mg/kg honokiol reduced the number of micrometastases in the lung by 41% and reduced the number of macrometastases in the lung and liver by 69% and 80%, respectively, compared with control. Primary tumor growth was not inhibited. In osteosarcoma cell lines, honokiol inhibited the metabolic activity with a half-maximal concentration (IC(50) ) between 8.0 µg/mL and 16 µg/mL. Cyclosporin A partially reversed the inhibition of metabolic activity in LM8-lacZ cells. Cell proliferation and wound healing migration of LM8-lacZ cells were inhibited by honokiol with an IC(50) between 5.0 µg/mL and 10 µg/mL. Higher concentrations caused rapid cell death, which was distinct from necrosis, apoptosis, or autophagy but was associated with swelling of the endoplasmic reticulum, cytoplasmic vacuolation, and morphologically altered mitochondria. CONCLUSIONS: Honokiol exhibited prominent antimetastatic activity in experimental osteosarcoma and caused rapid cell death in vitro that was unrelated to necrosis, apoptosis, or autophagy. The authors concluded that honokiol has considerable potential for the treatment of metastasizing osteosarcoma.


Assuntos
Antineoplásicos/farmacologia , Compostos de Bifenilo/farmacologia , Neoplasias Ósseas/tratamento farmacológico , Lignanas/farmacologia , Neoplasias Hepáticas/secundário , Neoplasias Pulmonares/secundário , Osteossarcoma/tratamento farmacológico , Osteossarcoma/secundário , Animais , Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Compostos de Bifenilo/uso terapêutico , Neoplasias Ósseas/patologia , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Humanos , Lignanas/uso terapêutico , Neoplasias Hepáticas/prevenção & controle , Neoplasias Pulmonares/prevenção & controle , Camundongos , Camundongos Endogâmicos C3H , Osteossarcoma/patologia
8.
Noncoding RNA ; 8(1)2022 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-35202088

RESUMO

The differentiation of B cells into antibody secreting plasma cells (PCs) is governed by a strict regulatory network that results in expression of specific transcriptomes along the activation continuum. In vitro models yielding significant numbers of PCs phenotypically identical to the in vivo state enable investigation of pathways, metabolomes, and non-coding (ncRNAs) not previously identified. The objective of our study was to characterize ncRNA expression during human B cell activation and differentiation. To achieve this, we used an in vitro system and performed RNA-seq on resting and activated B cells and PCs. Characterization of coding gene transcripts, including immunoglobulin (Ig), validated our system and also demonstrated that memory B cells preferentially differentiated into PCs. Importantly, we identified more than 980 ncRNA transcripts that are differentially expressed across the stages of activation and differentiation, some of which are known to target transcription, proliferation, cytoskeletal, autophagy and proteasome pathways. Interestingly, ncRNAs located within Ig loci may be targeting both Ig and non-Ig-related transcripts. ncRNAs associated with B cell malignancies were also identified. Taken together, this system provides a platform to study the role of specific ncRNAs in B cell differentiation and altered expression of those ncRNAs involved in B cell malignancies.

9.
Blood Adv ; 6(11): 3458-3471, 2022 06 14.
Artigo em Inglês | MEDLINE | ID: mdl-35395072

RESUMO

Identifying factors secreted by multiple myeloma (MM) cells that may contribute to MM tumor biology and progression is of the utmost importance. In this study, hepatoma-derived growth factor (HDGF) was identified as a protein present in extracellular vesicles (EVs) released from human MM cell lines (HMCLs). Investigation of the role of HDGF in MM cell biology revealed lower proliferation of HMCLs following HDGF knockdown and AKT phosphorylation following the addition of exogenous HDGF. Metabolic analysis demonstrated that HDGF enhances the already high glycolytic levels of HMCLs and significantly lowers mitochondrial respiration, indicating that HDGF may play a role in myeloma cell survival and/or act in a paracrine manner on cells in the bone marrow (BM) tumor microenvironment (ME). Indeed, HDGF polarizes macrophages to an M1-like phenotype and phenotypically alters naïve CD14+ monocytes to resemble myeloid-derived suppressor cells which are functionally suppressive. In summary, HDGF is a novel factor in MM biology and may function to both maintain MM cell viability as well as modify the tumor ME.


Assuntos
Vesículas Extracelulares , Mieloma Múltiplo , Humanos , Peptídeos e Proteínas de Sinalização Intercelular , Proteômica , Microambiente Tumoral
10.
J Histochem Cytochem ; 68(2): 99-112, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31855110

RESUMO

The ability to visualize and quantify the spatial arrangement and geographic proximity of immune cells with tumor cells provides valuable insight into the complex mechanisms underlying cancer biology and progression. Multiplexing, which involves immunofluorescence labeling and the visualization of multiple epitopes within formalin-fixed paraffin embedded tissue sections, is a methodology that is being increasingly employed. Despite the power of immunofluorescence multiplex analysis, application of this technology to bone marrow core biopsies has not yet been realized. Given our specific long term goal to identify immune cells in proximity to bone marrow malignant plasma cells in multiple myeloma patients, we describe in this study adaptation of multiplex immunofluorescence analysis to this tissue. We first identified a blocking strategy that quenched autofluorescence. We next employed a multiplex strategy that uses a simple stripping solution to remove primary and secondary antibodies prior to subsequent rounds of staining. This method was found to be highly efficient and did not significantly alter antigenicity or tissue integrity. Our studies illustrate for the first time that immunofluorescence multiplexing is achievable in bone marrow core biopsies and will provide a novel opportunity to analyze the role of the immune contexture in disease progression of the monoclonal gammopathies.


Assuntos
Medula Óssea/patologia , Imunofluorescência , Anticorpos Monoclonais/imunologia , Biópsia , Medula Óssea/imunologia , Humanos , Mieloma Múltiplo/imunologia , Mieloma Múltiplo/patologia
11.
Exp Hematol ; 57: 42-49.e1, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29030084

RESUMO

The genetic abnormalities underlying multiple myeloma (MM) are notoriously complex and intraclonal heterogeneity is a common disease feature. In the current study, we describe the establishment of a monoclonal immunoglobulin A (IgA) kappa (κ) MM cell line designated MC-B11/14. Cytogenetic and fluorescence in situ hybridization analyses of the original and relapse patient samples revealed that the MM clone was nonhyperdiploid and possessed an 11;14 chromosomal translocation. The MC-B11/14 cell line, established from the relapse sample, is tetraploid and houses the t(11;14) abnormality. Given our long-standing interest in Ig function and secretion, we next used CRISPR technology to knock out IgA heavy-chain expression in the MC-B11/14 cells to assess the biological consequences of converting this cell line to one only expressing κ light chains. As expected, secretion of intact IgA was undetectable from MC-B11/14IgA- cells. Sensitivity to pomalidomide treatment was similar between the MC-B11/14WT and MC-B11/14IgA- cells; however, MC-B11/14IgA- cells were found to be significantly more resistant to bortezomib treatment. This study describes the establishment of a new human MM cell line tool with which to study disease biology and the use of CRISPR technology to create a potentially useful model with which to study MM light-chain escape.


Assuntos
Sistemas CRISPR-Cas , Linhagem Celular Tumoral , Técnicas de Inativação de Genes , Genes de Imunoglobulinas , Imunoglobulina A/genética , Cadeias Pesadas de Imunoglobulinas/genética , Mieloma Múltiplo/patologia , Sequência de Aminoácidos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Sequência de Bases , Transplante de Medula Óssea , Bortezomib/administração & dosagem , Bortezomib/farmacologia , Linhagem Celular Tumoral/efeitos dos fármacos , Linhagem Celular Tumoral/metabolismo , Cromossomos Humanos Par 11/genética , Cromossomos Humanos Par 11/ultraestrutura , Cromossomos Humanos Par 14/genética , Cromossomos Humanos Par 14/ultraestrutura , Terapia Combinada , Evolução Fatal , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Cadeias kappa de Imunoglobulina/biossíntese , Cadeias kappa de Imunoglobulina/genética , Imunofenotipagem , Masculino , Pessoa de Meia-Idade , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/terapia , Proteínas do Mieloma/biossíntese , Proteínas do Mieloma/genética , Alinhamento de Sequência , Tetraploidia , Talidomida/análogos & derivados , Talidomida/farmacologia , Translocação Genética
12.
Cancer Res ; 65(11): 4500-5, 2005 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-15930265

RESUMO

Imatinib, a Bcr-Abl tyrosine kinase inhibitor, is a highly effective therapy for patients with chronic myelogenous leukemia (CML). Despite durable responses in most chronic phase patients, relapses have been observed and are much more prevalent in patients with advanced disease. The most common mechanism of acquired imatinib resistance has been traced to Bcr-Abl kinase domain mutations with decreased imatinib sensitivity. Thus, alternate Bcr-Abl kinase inhibitors that have activity against imatinib-resistant mutants would be useful for patients who relapse on imatinib therapy. Two such Bcr-Abl inhibitors are currently being evaluated in clinical trials: the improved potency, selective Abl inhibitor AMN107 and the highly potent dual Src/Abl inhibitor BMS-354825. In the current article, we compared imatinib, AMN107, and BMS-354825 in cellular and biochemical assays against a panel of 16 kinase domain mutants representing >90% of clinical isolates. We report that AMN107 and BMS-354825 are 20-fold and 325-fold more potent than imatinib against cells expressing wild-type Bcr-Abl and that similar improvements are maintained for all imatinib-resistant mutants tested, with the exception of T315I. Thus, both inhibitors hold promise for treating imatinib-refractory CML.


Assuntos
Piperazinas/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Proteínas Tirosina Quinases/antagonistas & inibidores , Pirimidinas/farmacologia , Tiazóis/farmacologia , Animais , Antineoplásicos/farmacologia , Benzamidas , Linhagem Celular , Dasatinibe , Proteínas de Fusão bcr-abl , Mesilato de Imatinib , Leucemia Mielogênica Crônica BCR-ABL Positiva/tratamento farmacológico , Leucemia Mielogênica Crônica BCR-ABL Positiva/enzimologia , Camundongos , Modelos Moleculares , Estrutura Terciária de Proteína , Proteínas Tirosina Quinases/genética
14.
Leuk Res ; 30(9): 1097-104, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16464493

RESUMO

STAT5 is constitutively phosphorylated in leukemic cells in approximately 70% of acute myeloid leukemia (AML) patients. To identify kinase candidates potentially responsible for STAT5 phosphorylation, we used liquid chromatography-tandem mass spectrometry (LC-MS/MS) mass spectrometry to detect phosphoproteins in AML cell lines. We established TEL-ARG and BCR-ABL fusion proteins as the mechanism underlying STAT5 phosphorylation in HT-93 and KBM-3 cells, respectively. In addition, we identified a JAK2 pseudokinase domain mutation in HEL cells and using siRNA downregulation, established JAK2 as the kinase responsible for phosphorylating STAT5. This study illustrates the benefit of LC-MS/MS mass spectrometry and siRNA for the identification of novel targets and mutations.


Assuntos
Carcinógenos , Leucemia Mieloide Aguda/genética , Proteínas de Neoplasias/genética , Carcinógenos/metabolismo , Linhagem Celular Tumoral , Cromatografia Líquida , Humanos , Leucemia Mieloide Aguda/metabolismo , Espectrometria de Massas , Mutação , Proteínas de Neoplasias/metabolismo , Fosforilação , Processamento de Proteína Pós-Traducional/genética , Proteômica
15.
Clin Cancer Res ; 11(19 Pt 1): 6987-93, 2005 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-16203792

RESUMO

PURPOSE: Chronic myeloid leukemia (CML) is effectively treated with imatinib. However, reactivation of Bcr-Abl via kinase domain mutations that reduce sensitivity to imatinib can cause relapse. As combination therapy is frequently used to prevent emergence of resistance, the combination of imatinib with an inhibitor of imatinib-resistant Bcr-Abl mutants (e.g., Src/Abl inhibitors AP23848 and BMS-354825) was investigated. EXPERIMENTAL DESIGN: To test this approach, cellular proliferation and Bcr-Abl tyrosine phosphorylation assays were done on Ba/F3 cells expressing wild-type (WT) Bcr-Abl and four common imatinib-resistant mutants (Y253F, E255K, T315I, and M351T). Colony-forming assays with primary CML cells were also done. RESULTS: Both Src/Abl inhibitors retained full inhibitory capacity when coadministered with imatinib at concentrations above typical clinical levels. For cells expressing WT Bcr-Abl or the marginally imatinib-resistant mutant M351T, inclusion of imatinib at therapeutic levels enhanced the effects of the Src/Abl inhibitors. By comparison, for the highly imatinib-resistant mutants Y253F and E255K, inclusion of imatinib at clinical levels resulted in only a slight enhancement beyond the effects of the Src/Abl inhibitors. None of the inhibitors affected Bcr-Abl T315I cells. Colony-forming assays with primary CML cells yielded analogous results. CONCLUSIONS: Our results indicate that Src/Abl inhibitors are compatible with imatinib and suggest that combined Abl inhibitor therapy is a feasible treatment strategy for patients with CML.


Assuntos
Resistencia a Medicamentos Antineoplásicos , Leucemia Mielogênica Crônica BCR-ABL Positiva/tratamento farmacológico , Piperazinas/farmacologia , Proteínas Proto-Oncogênicas c-abl/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-abl/metabolismo , Pirimidinas/farmacologia , Quinases da Família src/metabolismo , Trifosfato de Adenosina/análogos & derivados , Trifosfato de Adenosina/farmacologia , Animais , Antineoplásicos/farmacologia , Benzamidas , Proliferação de Células , Dasatinibe , Relação Dose-Resposta a Droga , Vetores Genéticos , Células-Tronco Hematopoéticas/citologia , Humanos , Mesilato de Imatinib , Immunoblotting , Concentração Inibidora 50 , Camundongos , Mutação , Fosfotirosina/química , Mutação Puntual , Inibidores de Proteínas Quinases/farmacologia , Estrutura Terciária de Proteína , Células-Tronco/metabolismo , Tiazóis/farmacologia , Fatores de Tempo
16.
Oncogene ; 23(6): 1197-205, 2004 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-14647450

RESUMO

Receptor crosstalk is an emerging and recurrent theme in cytokine and growth factor signaling; however, insight into the mechanism(s) underlying these interactions remains limited. Recently, we reported that crosstalk occurs between ErbB3 and the interferon alpha (IFN-alpha) signaling complex in the myeloma cell line KAS-6/1 and that this crosstalk contributes to the regulation of cell proliferation. In this study, we examined the mechanism underlying the transactivation of ErbB3 in the IFN-alpha growth-responsive KAS-6/1 cells. The examination of IFN-alpha receptor 1 and 2 (IFNAR1 and IFNAR2) levels revealed that the KAS-6/1 cell line overexpresses IFNAR1 relative to other myeloma cell lines that are growth arrested by IFN-alpha. Subsequent investigation of Tyk2, which is constitutively associated with IFNAR1, demonstrated that Tyk2 activation is uniquely sustained in the KAS-6/1 cell line following IFN-alpha stimulation. Interestingly, silencing of Tyk2 expression via siRNA resulted in attenuation of ErbB3 transactivation. However, inhibition of Jak1 expression also decreased IFN-alpha-induced tyrosine phosphorylation of ErbB3. Finally, siRNA downregulation of Tyk2 and Jak1 was found to decrease IFN-alpha-stimulated proliferation. These findings validate our previous report of ErbB3 involvement in IFN-alpha-induced proliferation and further suggest that both Janus kinase members, Tyk2 and Jak1, play a role in the transactivation of ErbB3 in this model system.


Assuntos
Regulação Neoplásica da Expressão Gênica/genética , Interferon-alfa/fisiologia , Proteínas Tirosina Quinases/metabolismo , Receptor Cross-Talk/fisiologia , Receptor ErbB-3/metabolismo , Receptores de Interferon/fisiologia , Transdução de Sinais/fisiologia , Sequência de Bases , Divisão Celular , Replicação do DNA , Humanos , Interferon-alfa/genética , Interferon-alfa/farmacologia , Proteínas de Membrana , Mieloma Múltiplo , RNA Interferente Pequeno/genética , Receptor de Interferon alfa e beta , Receptores de Interferon/genética , Transfecção , Células Tumorais Cultivadas
17.
Oncogene ; 22(23): 3598-607, 2003 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-12789268

RESUMO

We have previously demonstrated that the responsiveness of multiple myeloma (MM) cells to interferon-alpha (IFN-alpha) stimulation is variable, with an atypical growth response displayed by some cells. Here we report the ability of IFN-alpha to induce tyrosine phosphorylation of a 180 kDa band in the KAS-6/1 MM cell line, which is growth responsive to IFN-alpha. Further characterization demonstrated that this band corresponds to ErbB3. To our knowledge, this is the first report of ErbB3 expression in a cell type of the hematopoietic lineage. Although ErbB receptors have been shown to crosscommunicate with various other receptors, our results show for the first time that the IFN-alpha receptor can crosscommunicate with ErbB3. To address the significance of these observations, we transfected ErbB3-negative DP-6 MM cells with ErbB3 and used siRNA to silence ErbB3 in the KAS-6/1 cell line. Although IFN-alpha transactivated ErbB3 in the DP-6 transfectants, it did not confer growth responsiveness to IFN-alpha. Interestingly, silencing ErbB3 expression in the KAS-6/1 cells decreased the overall growth response to IFN-alpha and to interleukin-6. These results suggest that ErbB3 expression alone does not uniquely confer IFN-alpha growth responsiveness, but instead may amplify proliferation rates in MM cells that have acquired atypical expression of this receptor.


Assuntos
Interferon-alfa/metabolismo , Mieloma Múltiplo/metabolismo , Receptor Cross-Talk/fisiologia , Receptor ErbB-3/metabolismo , Divisão Celular/efeitos dos fármacos , Proteínas de Ligação a DNA/efeitos dos fármacos , Proteínas de Ligação a DNA/metabolismo , Regulação para Baixo , Ativação Enzimática/efeitos dos fármacos , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Interferon-alfa/farmacologia , Cinética , Substâncias Macromoleculares , Proteínas Quinases Ativadas por Mitógeno/efeitos dos fármacos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/genética , Fosforilação/efeitos dos fármacos , RNA Interferente Pequeno/farmacologia , Receptor ErbB-3/efeitos dos fármacos , Receptor ErbB-3/genética , Receptor de Interferon alfa e beta , Receptores de Interferon/metabolismo , Fator de Transcrição STAT3 , Transdução de Sinais , Transativadores/efeitos dos fármacos , Transativadores/metabolismo , Células Tumorais Cultivadas , Tirosina/metabolismo
18.
Oncotarget ; 5(14): 5686-99, 2014 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-25015330

RESUMO

Multiple myeloma (MM) is characterized by the clonal expansion of malignant plasma cells within the bone marrow. There is a growing literature that tumor cells release biologically active microvesicles (MVs) that modify both local and distant microenvironments. In this study, our goals were to determine if MM cells release MVs, and if so, begin to characterize their biologic activity. Herein we present clear evidence that not only do both patient MM cells and human MM cell lines (HMCLs) release MVs, but that these MVs stimulate MM cell growth. Of interest, MM-derived MVs were enriched with the biologically active form of CD147, a transmembrane molecule previously shown by us to be crucial for MM cell proliferation. Using MVs isolated from HMCLs stably transfected with a CD147-GFP fusion construct (CD147GFP), we observed binding and internalization of MV-derived CD147 with HMCLs. Cells with greater CD147GFP internalization proliferated at a higher rate than did cells with less CD147GFP association. Lastly, MVs obtained from CD147 downregulated HMCLs were attenuated in their ability to stimulate HMCL proliferation. In summary, this study demonstrates the significance of MV shedding and MV-mediated intercellular communication on malignant plasma cell proliferation, and identifies the role of MV-enriched CD147 in this process.


Assuntos
Basigina/biossíntese , Mieloma Múltiplo/metabolismo , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Proliferação de Células/fisiologia , Humanos , Imunofenotipagem , Mieloma Múltiplo/genética , Mieloma Múltiplo/patologia
19.
Cell Cycle ; 12(19): 3175-83, 2013 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-24013424

RESUMO

Increased use of the glycolytic pathway, even in the presence of oxygen, has recently been recognized as a key characteristic of malignant cells. However, the glycolytic phenotype results in increased lactic acid production and, in order to prevent cellular acidosis, tumor cells must increase proton efflux via upregulation of pH regulators such as proton-pumps, sodium-proton exchangers, and/or monocarboxylate transporters (MCT) (e.g., MCT1, MCT4). Interestingly, expression of MCT1 and MCT4 has been previously shown to be dependent upon expression of the transmembrane glycoprotein CD147. Recently, we demonstrated that primary patient multiple myeloma (MM) cells and human MM cell lines (HMCLs) overexpress CD147. Therefore, the goal of the current study was to specifically determine if MCT1 and MCT4 were also overexpressed in MM cells. RT-PCR analysis demonstrated both primary patient MM cells and HMCLs overexpress MCT1 and MCT4 mRNA. Notably, primary MM cells or HMCLs were found to express variable levels of MCT1 and/or MCT4 at the protein level despite CD147 expression. In those HMCLs positive for MCT1 and/or MCT4 protein expression, MCT1 and/or MCT4 were found to be associated with CD147. Specific siRNA-mediated downregulation of MCT1 but not MCT4 resulted in decreased HMCL proliferation, decreased lactate export, and increased cellular media pH. However, western blot analysis revealed that downregulation of MCT1 also downregulated CD147 and vice versa despite no effect on mRNA levels. Taken together, these data demonstrate the association between MCT1 and CD147 proteins in MM cells and importance of their association for lactate export and proliferation in MM cells.


Assuntos
Basigina/metabolismo , Ácido Láctico/metabolismo , Transportadores de Ácidos Monocarboxílicos/metabolismo , Simportadores/metabolismo , Proliferação de Células , Regulação para Baixo , Perfilação da Expressão Gênica , Humanos , Transportadores de Ácidos Monocarboxílicos/antagonistas & inibidores , Transportadores de Ácidos Monocarboxílicos/genética , Mieloma Múltiplo/metabolismo , Mieloma Múltiplo/patologia , Proteínas Musculares/antagonistas & inibidores , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Interferência de RNA , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/metabolismo , Simportadores/antagonistas & inibidores , Simportadores/genética , Células Tumorais Cultivadas
20.
PLoS One ; 8(7): e70554, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23894671

RESUMO

The biology of the malignant plasma cells (PCs) in multiple myeloma (MM) is highly influenced by the bone marrow (BM) microenvironment in which they reside. More specifically, BM stromal cells (SCs) are known to interact with MM cells to promote MM cell survival and proliferation. By contrast, it is unclear if innate immune cells within this same space also actively participate in the pathology of MM. Our study shows for the first time that eosinophils (Eos) can contribute to the biology of MM by enhancing the proliferation of some malignant PCs. We first demonstrate that PCs and Eos can be found in close proximity in the BM. In culture, Eos were found to augment MM cell proliferation that is predominantly mediated through a soluble factor(s). Fractionation of cell-free supernatants and neutralization studies demonstrated that this activity is independent of Eos-derived microparticles and a proliferation-inducing ligand (APRIL), respectively. Using a multicellular in vitro system designed to resemble the native MM niche, SCs and Eos were shown to have non-redundant roles in their support of MM cell growth. Whereas SCs induce MM cell proliferation predominantly through the secretion of IL-6, Eos stimulate growth of these malignant cells via an IL-6-independent mechanism. Taken together, our study demonstrates for the first time a role for Eos in the pathology of MM and suggests that therapeutic strategies targeting these cells may be beneficial.


Assuntos
Comunicação Celular , Eosinófilos/metabolismo , Mieloma Múltiplo/patologia , Plasmócitos/patologia , Células da Medula Óssea/patologia , Linhagem Celular Tumoral , Proliferação de Células , Eosinófilos/imunologia , Humanos , Interleucina-6/imunologia , Interleucina-6/metabolismo , Mieloma Múltiplo/imunologia , Plasmócitos/imunologia , Solubilidade , Células Estromais/patologia , Sindecana-1/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA