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










Base de datos
Intervalo de año de publicación
1.
Blood Adv ; 6(11): 3458-3471, 2022 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-35395072

RESUMEN

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.


Asunto(s)
Vesículas Extracelulares , Mieloma Múltiple , Humanos , Péptidos y Proteínas de Señalización Intercelular , Proteómica , Microambiente Tumoral
2.
Noncoding RNA ; 8(1)2022 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-35202088

RESUMEN

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.

3.
Blood ; 139(26): 3708-3721, 2022 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-35090171

RESUMEN

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.


Asunto(s)
Fibroblastos Asociados al Cáncer , Mieloma Múltiple , Médula Ósea , Fibroblastos Asociados al Cáncer/patología , Tratamiento Basado en Trasplante de Células y Tejidos , Fibroblastos , Humanos , Inmunoterapia Adoptiva/métodos , Mieloma Múltiple/patología , Microambiente Tumoral
4.
J Histochem Cytochem ; 68(2): 99-112, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31855110

RESUMEN

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.


Asunto(s)
Médula Ósea/patología , Técnica del Anticuerpo Fluorescente , Anticuerpos Monoclonales/inmunología , Biopsia , Médula Ósea/inmunología , Humanos , Mieloma Múltiple/inmunología , Mieloma Múltiple/patología
5.
Blood ; 133(7): 697-709, 2019 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-30463995

RESUMEN

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.


Asunto(s)
Citocinas/metabolismo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/antagonistas & inhibidores , Enfermedades del Sistema Inmune/terapia , Inflamación/terapia , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Receptores de Antígenos de Linfocitos T/uso terapéutico , Receptores Quiméricos de Antígenos/inmunología , Animales , Anticuerpos Neutralizantes/farmacología , Proliferación Celular , Humanos , Enfermedades del Sistema Inmune/inmunología , Enfermedades del Sistema Inmune/metabolismo , Inflamación/inmunología , Inflamación/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Ratones , Leucemia-Linfoma Linfoblástico de Células Precursoras/inmunología , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Receptores Quiméricos de Antígenos/metabolismo , Síndrome , Trasplante Heterólogo , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Exp Hematol ; 57: 42-49.e1, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29030084

RESUMEN

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.


Asunto(s)
Sistemas CRISPR-Cas , Línea Celular Tumoral , Técnicas de Inactivación de Genes , Genes de Inmunoglobulinas , Inmunoglobulina A/genética , Cadenas Pesadas de Inmunoglobulina/genética , Mieloma Múltiple/patología , Secuencia de Aminoácidos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Secuencia de Bases , Trasplante de Médula Ósea , Bortezomib/administración & dosificación , Bortezomib/farmacología , Línea Celular Tumoral/efectos de los fármacos , Línea Celular Tumoral/metabolismo , Cromosomas Humanos Par 11/genética , Cromosomas Humanos Par 11/ultraestructura , Cromosomas Humanos Par 14/genética , Cromosomas Humanos Par 14/ultraestructura , Terapia Combinada , Resultado Fatal , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Cadenas kappa de Inmunoglobulina/biosíntesis , Cadenas kappa de Inmunoglobulina/genética , Inmunofenotipificación , Masculino , Persona de Mediana Edad , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/terapia , Proteínas de Mieloma/biosíntesis , Proteínas de Mieloma/genética , Alineación de Secuencia , Tetraploidía , Talidomida/análogos & derivados , Talidomida/farmacología , Translocación Genética
7.
Oncotarget ; 5(14): 5686-99, 2014 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-25015330

RESUMEN

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.


Asunto(s)
Basigina/biosíntesis , Mieloma Múltiple/metabolismo , Línea Celular Tumoral , Membrana Celular/metabolismo , Proliferación Celular/fisiología , Humanos , Inmunofenotipificación , Mieloma Múltiple/genética , Mieloma Múltiple/patología
8.
Eur J Haematol ; 91(6): 504-13, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23992230

RESUMEN

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.


Asunto(s)
Factores Inmunológicos/farmacología , Factores Reguladores del Interferón/genética , Mieloma Múltiple/genética , Péptido Hidrolasas/genética , Talidomida/análogos & derivados , Proteínas Adaptadoras Transductoras de Señales , Apoptosis/efectos de los fármacos , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Aberraciones Cromosómicas , Relación Dosis-Respuesta a Droga , Expresión Génica , Humanos , Factores Reguladores del Interferón/metabolismo , Lenalidomida , Mieloma Múltiple/tratamiento farmacológico , Talidomida/farmacología , Ubiquitina-Proteína Ligasas
9.
Cell Cycle ; 12(19): 3175-83, 2013 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-24013424

RESUMEN

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.


Asunto(s)
Basigina/metabolismo , Ácido Láctico/metabolismo , Transportadores de Ácidos Monocarboxílicos/metabolismo , Simportadores/metabolismo , Proliferación Celular , Regulación hacia Abajo , Perfilación de la Expresión Génica , Humanos , Transportadores de Ácidos Monocarboxílicos/antagonistas & inhibidores , Transportadores de Ácidos Monocarboxílicos/genética , Mieloma Múltiple/metabolismo , Mieloma Múltiple/patología , Proteínas Musculares/antagonistas & inhibidores , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Interferencia de ARN , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , Simportadores/antagonistas & inhibidores , Simportadores/genética , Células Tumorales Cultivadas
10.
PLoS One ; 8(7): e70554, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23894671

RESUMEN

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.


Asunto(s)
Comunicación Celular , Eosinófilos/metabolismo , Mieloma Múltiple/patología , Células Plasmáticas/patología , Células de la Médula Ósea/patología , Línea Celular Tumoral , Proliferación Celular , Eosinófilos/inmunología , Humanos , Interleucina-6/inmunología , Interleucina-6/metabolismo , Mieloma Múltiple/inmunología , Células Plasmáticas/inmunología , Solubilidad , Células del Estroma/patología , Sindecano-1/metabolismo
11.
Int J Cancer ; 131(5): E804-12, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-22120774

RESUMEN

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.


Asunto(s)
Antibióticos Antineoplásicos/efectos adversos , Neoplasias Óseas/tratamiento farmacológico , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Osteosarcoma/tratamiento farmacológico , Taurina/análogos & derivados , Tiadiazinas/efectos adversos , Animales , Neoplasias Óseas/patología , Femenino , Neoplasias Hepáticas/secundario , Neoplasias Pulmonares/secundario , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Osteosarcoma/patología , Taurina/efectos adversos , Células Tumorales Cultivadas
12.
Cancer ; 118(8): 2117-27, 2012 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-21935912

RESUMEN

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.


Asunto(s)
Antineoplásicos/farmacología , Compuestos de Bifenilo/farmacología , Neoplasias Óseas/tratamiento farmacológico , Lignanos/farmacología , Neoplasias Hepáticas/secundario , Neoplasias Pulmonares/secundario , Osteosarcoma/tratamiento farmacológico , Osteosarcoma/secundario , Animales , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Compuestos de Bifenilo/uso terapéutico , Neoplasias Óseas/patología , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Humanos , Lignanos/uso terapéutico , Neoplasias Hepáticas/prevención & control , Neoplasias Pulmonares/prevención & control , Ratones , Ratones Endogámicos C3H , Osteosarcoma/patología
13.
J Orthop Res ; 29(6): 938-46, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21284029

RESUMEN

More effective treatment of patients with metastasizing osteosarcoma (OS) with a mean 5-year survival rate of <20% requires more detailed knowledge on the complex mechanisms of metastasis for the design of new drugs, which selectively target metastasizing cells. Moreover, novel diagnostic imaging technology for early detection of metastases is needed. Mouse models, which reproduce human metastasizing OS and allow visualization of single metastatic cells are instrumental for preclinical testing of new pharmaceuticals and diagnostic instruments. Here, the low metastatic Dunn cell line and its highly metastatic LM8 subline, both equipped with a constitutively expressed lacZ gene, were used to improve the well-established OS models in syngeneic C3H mice to achieve ex vivo visualization of single metastatic cells in affected organs by X-gal staining. These models, combined with a technique for in situ high quality lung tissue-maintaining perfusion revealed, as a novel finding, single metastasizing Dunn cells in lung and liver. Importantly, constitutive lacZ gene expression did not affect in vitro and in vivo tumorigenic and metastatic properties of Dunn and LM8 cells. Thus, these improved Dunn and LM8 OS mouse models will in the future serve as a benchmark for the development of new metastasis-targeting drugs and metastasis-imaging technology.


Asunto(s)
Neoplasias Óseas/patología , Operón Lac , Osteosarcoma/patología , Sarcoma Experimental/patología , Transgenes , Animales , Línea Celular Tumoral , Técnicas de Transferencia de Gen , Neoplasias Hepáticas Experimentales/secundario , Neoplasias Pulmonares/secundario , Ratones , Ratones Endogámicos C3H , Metástasis de la Neoplasia , Retroviridae , Adhesión del Tejido
15.
Blood ; 111(4): 2238-45, 2008 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-18025156

RESUMEN

Despite vast improvements in our understanding of cancer genetics, a large percentage of cancer cases present without knowledge of the causative genetic events. Tyrosine kinases are frequently implicated in the pathogenesis of numerous types of cancer, but identification and validation of tyrosine kinase targets in cancer can be a time-consuming process. We report the establishment of an efficient, functional screening assay using RNAi technology to directly assess and compare the effect of individually targeting each member of the tyrosine kinase family. We demonstrate that siRNA screening can identify tyrosine kinase targets containing activating mutations in Janus kinase (JAK) 3 (A572V) in CMK cells and c-KIT (V560G) in HMC1.1 cells. In addition, this assay identifies targets that do not contain mutations, such as JAK1 and the focal adhesion kinases (FAK), that are crucial to the survival of the cancer cells. This technique, with additional development, might eventually offer the potential to match specific therapies with individual patients based on a functional assay.


Asunto(s)
Leucemia Mieloide Aguda/tratamiento farmacológico , Proteínas Tirosina Quinasas/genética , Interferencia de ARN/fisiología , ARN Interferente Pequeño/genética , Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Biblioteca de Genes , Humanos , Janus Quinasa 1/genética , Janus Quinasa 3/genética , Células K562 , Leucemia Mieloide Aguda/enzimología , Leucemia Mieloide Aguda/genética
16.
Invest New Drugs ; 26(4): 289-97, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18071634

RESUMEN

Curcumin (diferuloylmethane), one of the main components of the Indian spice turmeric, is known to possess potent anti-inflammatory and anti-oxidant properties. In addition, curcumin has also been shown to have in vitro and in vivo efficacy against a variety of malignancies. In the current study we examined the cytotoxic effect of curcumin on seven osteosarcoma (OS) cell lines with varying degrees of in vivo metastatic potential. Curcumin inhibited the growth of all OS cell lines tested with half-maximal inhibitory concentration values ranging from 14.4 to 24.6 microM. Growth inhibition was associated with a dose dependent increase in the number of apoptotic cells and accumulation of cells in the G(2)/M phase of the cell cycle. Curcumin treatment also resulted in cleavage of caspase-3 and poly adenosine diphosphate-ribose polymerase. Moreover, curcumin treatment was associated with an increase in cellular levels of the apoptotic B-cell leukemia/lymphoma 2 (Bcl-2)-associated X protein and a decrease in cellular content of the anti-apoptotic protein Bcl-2. In addition, curcumin treatment also inhibited the migration of OS cell lines. These data indicate that the potent cytotoxic activity of curcumin on OS cell lines is mediated by induction of apoptotic processes. Thus, curcumin has potential to be a novel OS chemotherapeutic agent.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Apoptosis/efectos de los fármacos , Curcumina/farmacología , Osteosarcoma/tratamiento farmacológico , Antineoplásicos Fitogénicos/administración & dosificación , Caspasa 3/efectos de los fármacos , Caspasa 3/metabolismo , División Celular/efectos de los fármacos , Línea Celular Tumoral , Curcuma/química , Curcumina/administración & dosificación , Fase G2/efectos de los fármacos , Humanos , India , Concentración 50 Inhibidora , Poli(ADP-Ribosa) Polimerasas/efectos de los fármacos , Poli(ADP-Ribosa) Polimerasas/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/efectos de los fármacos , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteína X Asociada a bcl-2/efectos de los fármacos , Proteína X Asociada a bcl-2/metabolismo
17.
Invest New Drugs ; 25(4): 305-12, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17458504

RESUMEN

Taurolidine, the active agent of Taurolin, is a broad spectrum anti-biotic that has been used for over 15 years for the treatment of severe surgical infections. Recently, taurolidine has been shown to possess anti-neoplastic properties in vitro and in vivo against a variety of cancers including ovarian, colon and prostate. In this study we assessed the cytotoxic activity of taurolidine against human osteosarcoma (OS) cell lines and normal human bone cells. Treatment with taurolidine inhibited the growth of all ten osteosarcoma cell lines tested and taurolidine was equally potent against cell lines with and without distinct genetic defects (i.e. p53, Rb). Moreover, taurolidine-induced growth inhibition was found to be associated with a dose dependent increase in the number of apoptotic cells and apoptosis was shown to be caspase-dependent. Taurolidine treatment was also found to inhibit adhesion of OS cell lines. Compared to OS cell lines, normal bone cells in primary culture were found to be less sensitive to the cytotoxic and anti-adhesive effects of taurolidine. These data indicate that taurolidine possesses potent anti-neoplastic activity against osteosarcoma cell lines and may have potential as a novel OS chemotherapeutic agent.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis , Taurina/análogos & derivados , Tiadiazinas/farmacología , Neoplasias Óseas , Línea Celular Tumoral , Humanos , Osteosarcoma , Taurina/farmacología
18.
Blood ; 110(1): 323-33, 2007 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-17360941

RESUMEN

Activated tyrosine kinases have been frequently implicated in the pathogenesis of cancer, including acute myeloid leukemia (AML), and are validated targets for therapeutic intervention with small-molecule kinase inhibitors. To identify novel activated tyrosine kinases in AML, we used a discovery platform consisting of immunoaffinity profiling coupled to mass spectrometry that identifies large numbers of tyrosine-phosphorylated proteins, including active kinases. This method revealed the presence of an activated colony-stimulating factor 1 receptor (CSF1R) kinase in the acute megakaryoblastic leukemia (AMKL) cell line MKPL-1. Further studies using siRNA and a small-molecule inhibitor showed that CSF1R is essential for the growth and survival of MKPL-1 cells. DNA sequence analysis of cDNA generated by 5'RACE from CSF1R coding sequences identified a novel fusion of the RNA binding motif 6 (RBM6) gene to CSF1R gene generated presumably by a t(3;5)(p21;q33) translocation. Expression of the RBM6-CSF1R fusion protein conferred interleukin-3 (IL-3)-independent growth in BaF3 cells, and induces a myeloid proliferative disease (MPD) with features of megakaryoblastic leukemia in a murine transplant model. These findings identify a novel potential therapeutic target in leukemogenesis, and demonstrate the utility of phosphoproteomic strategies for discovery of tyrosine kinase alleles.


Asunto(s)
Leucemia Megacarioblástica Aguda/genética , Proteínas de Fusión Oncogénica/fisiología , Proteínas de Unión al ARN/genética , Receptor de Factor Estimulante de Colonias de Macrófagos/genética , Animales , Línea Celular , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Cromosomas Humanos Par 3 , Cromosomas Humanos Par 5 , Humanos , Leucemia Megacarioblástica Aguda/etiología , Ratones , Trasplante de Neoplasias , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/aislamiento & purificación , Proteínas Tirosina Quinasas/aislamiento & purificación , Receptor de Factor Estimulante de Colonias de Macrófagos/aislamiento & purificación , Análisis de Secuencia de ADN , Translocación Genética , Trasplante Heterólogo
19.
Blood ; 108(13): 4202-4, 2006 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-16946300

RESUMEN

The 8p11 myeloproliferative syndrome (EMS) is associated with translocations that disrupt the FGFR1 gene. To date, 8 fusion partners of FGFR1 have been identified. However, no primary leukemia cell lines were identified that contain any of these fusions. Here, we screened more than 40 acute myeloid leukemia cell lines for constitutive phosphorylation of STAT5 and applied an immunoaffinity profiling strategy to identify tyrosine-phosphorylated proteins in the KG-1 cell line. Mass spectrometry analysis of KG-1 cells revealed aberrant tyrosine phosphorylation of FGFR1. Subsequent analysis led to the identification of a fusion of the FGFR1OP2 gene to the FGFR1 gene. Small interfering RNA (siRNA) against FGFR1 specifically inhibited the growth and induced apoptosis of KG-1 cells. Thus, the KG-1 cell line provides an in vitro model for the study of FGFR1 fusions associated with leukemia and for the analysis of small molecule inhibitors against FGFR1 fusions.


Asunto(s)
Apoptosis/genética , Leucemia Mieloide Aguda/genética , Modelos Biológicos , Proteínas de Fusión Oncogénica/biosíntesis , Proteínas Proto-Oncogénicas/genética , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Línea Celular Tumoral , Humanos , Leucemia Mieloide Aguda/metabolismo , Proteínas de Fusión Oncogénica/genética , Proteínas Proto-Oncogénicas/biosíntesis , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/farmacología , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/biosíntesis , Factor de Transcripción STAT5/biosíntesis , Factor de Transcripción STAT5/genética
20.
Cancer Cell ; 10(1): 65-75, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16843266

RESUMEN

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.


Asunto(s)
Leucemia Experimental/genética , Leucemia Megacarioblástica Aguda/genética , Proteínas Tirosina Quinasas/genética , Alelos , Animales , Apoptosis/efectos de los fármacos , Benzamidas , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Humanos , Mesilato de Imatinib , Janus Quinasa 2 , Janus Quinasa 3 , Células K562 , Leucemia Experimental/metabolismo , Leucemia Experimental/patología , Leucemia Megacarioblástica Aguda/metabolismo , Leucemia Megacarioblástica Aguda/patología , Ratones , Ratones Endogámicos C57BL , Modelos Moleculares , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Fosforilación/efectos de los fármacos , Piperazinas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Estructura Terciaria de Proteína/genética , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Pirimidinas/farmacología , ARN Interferente Pequeño/genética , TYK2 Quinasa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...