Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
1.
Blood ; 141(21): 2615-2628, 2023 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-36735903

RESUMEN

Recent investigations have improved our understanding of the molecular aberrations supporting Waldenström macroglobulinemia (WM) biology; however, whether the immune microenvironment contributes to WM pathogenesis remains unanswered. First, we showed how a transgenic murine model of human-like lymphoplasmacytic lymphoma/WM exhibits an increased number of regulatory T cells (Tregs) relative to control mice. These findings were translated into the WM clinical setting, in which the transcriptomic profiling of Tregs derived from patients with WM unveiled a peculiar WM-devoted messenger RNA signature, with significant enrichment for genes related to nuclear factor κB-mediated tumor necrosis factor α signaling, MAPK, and PI3K/AKT, which was paralleled by a different Treg functional phenotype. We demonstrated significantly higher Treg induction, expansion, and proliferation triggered by WM cells, compared with their normal cellular counterpart; with a more profound effect within the context of CXCR4C1013G-mutated WM cells. By investigating the B-cell-to-T-cell cross talk at single-cell level, we identified the CD40/CD40-ligand as a potentially relevant axis that supports WM cell-Tregs interaction. Our findings demonstrate the existence of a Treg-mediated immunosuppressive phenotype in WM, which can be therapeutically reversed by blocking the CD40L/CD40 axis to inhibit WM cell growth.


Asunto(s)
Linfoma de Células B , Macroglobulinemia de Waldenström , Humanos , Animales , Ratones , Macroglobulinemia de Waldenström/patología , Ligando de CD40/genética , Fosfatidilinositol 3-Quinasas , Ligandos , Transducción de Señal , Linfoma de Células B/complicaciones , Microambiente Tumoral
2.
Radiology ; 306(3): e220542, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36255307

RESUMEN

Background Large studies on the diagnostic performance of CT-derived myocardial extracellular volume fraction (ECV) for detecting cardiac amyloidosis are lacking. A simple and practical index as a surrogate for CT ECV would be clinically useful. Purpose To compare the diagnostic performances between CT-derived myocardial ECV and myocardium-to-lumen signal ratio for the detection of cardiac amyloidosis in a large patient sample. Materials and Methods This retrospective study included patients who underwent CT ECV analysis because of suspected heart failure or cardiomyopathy between January 2018 and July 2021. CT ECV was quantified using routine pre-transcatheter aortic valve replacement planning cardiac CT, pre-atrial fibrillation ablation planning cardiac CT, or coronary CT angiography with the addition of unenhanced and delayed phase cardiac CT scans. The diagnostic performances of CT ECV and myocardium-to-lumen signal ratio in delayed phase cardiac CT (a simplified index not requiring unenhanced CT and hematocrit) for detecting cardiac amyloidosis were evaluated using the area under the receiver operating characteristic curve (AUC), sensitivity, and specificity. Results Of 552 patients (mean age, 69 years ± 14 [SD]; 295 men), 41 had cardiac amyloidosis. The sensitivity of CT ECV for amyloidosis was 90% (37 of 41 patients [95% CI: 77, 97]), with a specificity of 92% (472 of 511 patients [95% CI: 90, 95]) and optimal ECV cutoff value of 37% (AUC, 0.97 [95% CI: 0.96, 0.99]). The sensitivity of myocardium-to-lumen signal ratio was 88% (36 of 41 patients [95% CI: 74, 96]), with a specificity of 92% (469 of 511 patients [95% CI: 89, 94]) and optimal myocardium-to-lumen signal ratio cutoff value of 0.87 (AUC, 0.96 [95% CI: 0.94, 0.97]; P = .27 for comparison with ECV). Conclusion CT-derived myocardial extracellular volume fraction and myocardium-to-lumen signal ratio showed comparable and excellent diagnostic performance in detecting cardiac amyloidosis in a large patient sample. © RSNA, 2022 Online supplemental material is available for this article. See also the editorial by Williams in this issue.


Asunto(s)
Amiloidosis , Cardiomiopatías , Masculino , Humanos , Anciano , Estudios Retrospectivos , Miocardio , Amiloidosis/diagnóstico por imagen , Cardiomiopatías/diagnóstico por imagen , Tomografía Computarizada por Rayos X/métodos , Valor Predictivo de las Pruebas , Imagen por Resonancia Cinemagnética/métodos
3.
Blood ; 137(17): 2360-2372, 2021 04 29.
Artículo en Inglés | MEDLINE | ID: mdl-33150374

RESUMEN

Clonal evolution drives tumor progression, dissemination, and relapse in multiple myeloma (MM), with most patients dying of relapsed disease. This multistage process requires tumor cells to enter the circulation, extravasate, and colonize distant bone marrow (BM) sites. Here, we developed a fluorescent or DNA-barcode clone-tracking system on MM PrEDiCT (progression through evolution and dissemination of clonal tumor cells) xenograft mouse model to study clonal behavior within the BM microenvironment. We showed that only the few clones that successfully adapt to the BM microenvironment can enter the circulation and colonize distant BM sites. RNA sequencing of primary and distant-site MM tumor cells revealed a progression signature sequentially activated along human MM progression and significantly associated with overall survival when evaluated against patient data sets. A total of 28 genes were then computationally predicted to be master regulators (MRs) of MM progression. HMGA1 and PA2G4 were validated in vivo using CRISPR-Cas9 in the PrEDiCT model and were shown to be significantly depleted in distant BM sites, indicating their role in MM progression and dissemination. Loss of HMGA1 and PA2G4 also compromised the proliferation, migration, and adhesion abilities of MM cells in vitro. Overall, our model successfully recapitulates key characteristics of human MM disease progression and identified potential new therapeutic targets for MM.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Biomarcadores de Tumor/metabolismo , Modelos Animales de Enfermedad , Regulación Neoplásica de la Expresión Génica , Proteína HMGA1a/metabolismo , Mieloma Múltiple/patología , Recurrencia Local de Neoplasia/patología , Proteínas de Unión al ARN/metabolismo , Proteínas Adaptadoras Transductoras de Señales/antagonistas & inhibidores , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Apoptosis , Biomarcadores de Tumor/genética , Médula Ósea/metabolismo , Médula Ósea/patología , Sistemas CRISPR-Cas , Adhesión Celular , Movimiento Celular , Proliferación Celular , Evolución Clonal , Progresión de la Enfermedad , Femenino , Proteína HMGA1a/antagonistas & inhibidores , Proteína HMGA1a/genética , Humanos , Ratones , Ratones SCID , Mieloma Múltiple/genética , Mieloma Múltiple/metabolismo , Recurrencia Local de Neoplasia/genética , Recurrencia Local de Neoplasia/metabolismo , Pronóstico , Proteínas de Unión al ARN/antagonistas & inhibidores , Proteínas de Unión al ARN/genética , Tasa de Supervivencia , Células Tumorales Cultivadas
4.
Anal Chem ; 94(29): 10329-10336, 2022 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-35817413

RESUMEN

Recent advances in single-cell proteomics highlight the promise of sensitive analyses in limited cell populations. However, technical challenges remain for sample recovery, throughput, and versatility. Here, we first report a water droplet-in-oil digestion (WinO) method based on carboxyl-coated beads and phase transfer surfactants for proteomic analysis using limited sample amounts. This method was developed to minimize the contact area between the sample solution and the container to reduce the loss of proteins and peptides by adsorption. This method increased protein and peptide recovery 10-fold. The proteome profiles obtained from 100 cells using the WinO method highly correlated with those from 10,000 cells using the in-solution digestion method. We successfully applied the WinO method to single-cell proteomics and quantified 462 proteins. Using the WinO method, samples can be easily prepared in a multi-well plate, making it a widely applicable and suitable method for single-cell proteomics.


Asunto(s)
Proteoma , Proteómica , Digestión , Péptidos/análisis , Proteoma/análisis , Proteómica/métodos , Agua
5.
Biochem Biophys Res Commun ; 587: 153-159, 2022 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-34875534

RESUMEN

Pyruvate dehydrogenase kinase 1 (PDK1) is a Ser/Thr kinase that inactivates mitochondrial pyruvate dehydrogenase (PDH), leading to switch of glucose metabolism from mitochondrial oxidation to aerobic glycolysis. We previously reported that PDK1 inhibition is a potent therapeutic strategy in multiple myeloma (MM). However, availability of PDK1 inhibitors, which are effective at low concentrations, are limited at present, making PDK1 inhibition difficult to apply in the clinic. In the present study, we examined the efficacy and mechanism of action of JX06, a novel PDK1 inhibitor, against MM cells. We confirmed that PDK1 is highly expressed in normal plasma cells and MM cells using publicly available gene expression datasets. JX06 suppressed cell growth and induced apoptosis against MM cells from approximately 0.5 µM JX06 treatment reduced PDH phosphorylation, suggesting that JX06 is indeed inhibiting PDK1. Intracellular metabolite analysis revealed that JX06 treatment reduced metabolites associated with glucose metabolism of MM cells. Additionally, JX06 in combination with a well-known proteasome inhibitor, bortezomib, significantly increased MM cell death, which raises the possibility of combination use of JX06 with proteasome inhibitors in the clinic. These findings demonstrate that PDK1 can be potentially targeted by JX06 in MM through glycolysis inhibition, leading to a novel therapeutic strategy in MM.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Disulfiram/análogos & derivados , Inhibidores Enzimáticos/farmacología , Glucólisis/efectos de los fármacos , Morfolinas/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica , Apoptosis/genética , Bortezomib/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Conjuntos de Datos como Asunto , Disulfiram/farmacología , Sinergismo Farmacológico , Regulación Neoplásica de la Expresión Génica , Glucólisis/genética , Humanos , Cetona Oxidorreductasas/genética , Cetona Oxidorreductasas/metabolismo , Mitocondrias/efectos de los fármacos , Mitocondrias/enzimología , Mitocondrias/patología , Terapia Molecular Dirigida , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/enzimología , Mieloma Múltiple/genética , Mieloma Múltiple/patología , Fosforilación/efectos de los fármacos , Células Plasmáticas/efectos de los fármacos , Células Plasmáticas/enzimología , Células Plasmáticas/patología , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora/antagonistas & inhibidores , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora/metabolismo
6.
Rinsho Ketsueki ; 62(4): 299-304, 2021.
Artículo en Japonés | MEDLINE | ID: mdl-33967155

RESUMEN

Regulatory T cells (Tregs) are a subset of CD4+ T lymphocytes that suppress the functions of antigen-presenting cells and effector T cells, characterized by the expression of transcription factor forkhead box P3 (FOXP3). Recent studies have reported an increase in the number of Tregs in the bone marrow (BM) of multiple myeloma (MM) patients. However, the role and mechanisms of Treg accumulation in the BM of MM patients remain debatable. Here, we present our data demonstrating the significance of Tregs in the context of MM disease progression. Using the transplantable MM mouse model, we observed a significant increase in Tregs in the BM of MM-injected mice from the early disease stage. We observed extended survival in MM-injected mice with Treg depletion than in mice without Treg depletion, demonstrating direct in vivo evidence that Tregs enhance disease progression in MM. It is noteworthy that type 1 interferon (IFN) signaling is activated in MM-associated Tregs. By using type 1 IFN receptor blocking antibody treatment and type 1 IFN receptor knockout Tregs, we demonstrated a significant decrease in MM-associated Treg proliferation, which was associated with longer survival in MM-injected mice. Thus, we have demonstrated that Tregs play a significant role in MM progression; the function and homeostasis of Tregs are regulated by type 1 IFN secreted in the BM microenvironment.


Asunto(s)
Mieloma Múltiple , Linfocitos T Reguladores , Animales , Factores de Transcripción Forkhead/metabolismo , Homeostasis , Humanos , Ratones , Transducción de Señal , Microambiente Tumoral
7.
Rinsho Ketsueki ; 62(3): 196-199, 2021.
Artículo en Japonés | MEDLINE | ID: mdl-33828015

RESUMEN

We report the case of a 79-year-old man with chronic lymphocytic leukemia (CLL) with IgM-kappa type monoclonal gammopathy according to immunophenotypes and a negative result for MYD88 L265P mutation of leukemic cells. Abnormal lymphocytes and IgM increased under observation, and he experienced paresthesia. The diagnosis of IgM-type M protein associated peripheral neuropathy was confirmed by nerve conduction test, and negativity of myelin-associated glycoprotein and glycolipid antibodies. He was placed on intravenous immunoglobulin (IVIg) in combination with ibrutinib. His symptoms dramatically subsided and did not recur. Treatment with IVIg and ibrutinib may be useful for the rare complication of peripheral neuropathy with CLL.


Asunto(s)
Leucemia Linfocítica Crónica de Células B , Enfermedades del Sistema Nervioso Periférico , Adenina/análogos & derivados , Anciano , Humanos , Inmunoglobulina M , Inmunoglobulinas Intravenosas/uso terapéutico , Leucemia Linfocítica Crónica de Células B/complicaciones , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Masculino , Enfermedades del Sistema Nervioso Periférico/inducido químicamente , Enfermedades del Sistema Nervioso Periférico/tratamiento farmacológico , Piperidinas
8.
Immunol Rev ; 263(1): 160-72, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25510276

RESUMEN

Multiple myeloma (MM) is characterized by clonal expansion of malignant plasma cells in the bone marrow (BM). Despite the significant advances in treatment, MM is still a fatal malignancy. This is mainly due to the supportive role of the BM microenvironment in differentiation, migration, proliferation, survival, and drug resistance of the malignant plasma cells. The BM microenvironment is composed of a cellular compartment (stromal cells, osteoblasts, osteoclasts, endothelial cells, and immune cells) and a non-cellular compartment. In this review, we discuss the interaction between the malignant plasma cell and the BM microenvironment and the strategy to target them.


Asunto(s)
Médula Ósea/efectos de los fármacos , Trasplante de Células Madre Hematopoyéticas , Gammopatía Monoclonal de Relevancia Indeterminada/terapia , Mieloma Múltiple/terapia , Células Plasmáticas/fisiología , Microambiente Tumoral/efectos de los fármacos , Inhibidores de la Angiogénesis/uso terapéutico , Animales , Médula Ósea/inmunología , Ácidos Borónicos/uso terapéutico , Bortezomib , Humanos , Lenalidomida , Mieloma Múltiple/mortalidad , Pirazinas/uso terapéutico , Análisis de Supervivencia , Talidomida/análogos & derivados , Talidomida/uso terapéutico , Resultado del Tratamiento
9.
Rinsho Ketsueki ; 60(10): 1443-1448, 2019.
Artículo en Japonés | MEDLINE | ID: mdl-31695005

RESUMEN

Coagulation factor XIII is a fibrin-stabilizing factor that leads to the crosslinking of fibrin when activated by thrombin. Acquired factor XIII inhibitor is caused when antibodies are generated against factor XIII, reducing its activity. Here we report a case of acquired factor XIII inhibitor. Although prednisolone was administered, factor XIII activity was not recovered. Interestingly, the activity normalized following the onset of multiple myeloma. The presence of inhibitors was evaluated in the patient's plasma by absorption tests and enzyme-linked immunosorbent assay. Immunoglobulin G inhibitors of factor XIII were present at admission, but later decreased with the onset of the IgA-λ-type myeloma. Thus, it is possible that the level of factor XIII inhibitors and polyclonal immunoglobulins could have been suppressed by the progression of myeloma, resulting in the normalization of factor XIII activity.


Asunto(s)
Deficiencia del Factor XIII , Inmunoglobulina A , Mieloma Múltiple , Remisión Espontánea , Factor XIII/inmunología , Humanos
10.
Biochem Biophys Res Commun ; 507(1-4): 246-252, 2018 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-30420285

RESUMEN

The sialic glycoprotein, MUC1, is known to be involved in the pathogenesis of various types of cancers. KL-6 is one of the surface antigens of MUC1 and also a marker of interstitial pneumonitis. A fraction of patients with myeloma (3.9%) have elevated serum KL-6 levels without any evidence of interstitial pneumonitis and their myeloma cells have high MUC1 expression. We established a myeloma cell line designated EMM1 from a patient with multiple myeloma accompanied with elevated serum KL-6. EMM1 cells expressed high levels of MUC1 compared with other myeloma cell lines. Knockdown of MUC1 in EMM1 cells induced cell cycle arrest during S phase and apoptosis, suggesting that the MUC1 expression is involved in accelerated growth of EMM1 cells. RNA-seq analysis suggests that MUC1 expression activates k-ras and TNFα-induced NFκB pathways in EMM1 cells. We injected EMM1 cells subcutaneously into Rag2-/-Jak3-/- Balb/c mice to establish a mouse xenograft model. These mice had aggressive tumor growth that was accompanied by high serum KL-6 levels. In addition, MUC1 knockdown in EMM1 cells led to inhibited tumor growth. These findings demonstrate that MUC1 serves as a potential target for developing drugs for treatment of patients with KL-6+ myeloma, and EMM1 cells and EMM1-engrafted mice are useful tools for the development of such novel agents.


Asunto(s)
Mucina-1/metabolismo , Mieloma Múltiple/metabolismo , Mieloma Múltiple/patología , Animales , Puntos de Control del Ciclo Celular , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Persona de Mediana Edad , Mucina-1/sangre , Mieloma Múltiple/sangre , Invasividad Neoplásica , Fase S , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Blood ; 127(21): 2598-606, 2016 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-26903547

RESUMEN

Familial aggregation of Waldenström macroglobulinemia (WM) cases, and the clustering of B-cell lymphoproliferative disorders among first-degree relatives of WM patients, has been reported. Nevertheless, the possible contribution of inherited susceptibility to familial WM remains unrevealed. We performed whole exome sequencing on germ line DNA obtained from 4 family members in which coinheritance for WM was documented in 3 of them, and screened additional independent 246 cases by using gene-specific mutation sequencing. Among the shared germ line variants, LAPTM5(c403t) and HCLS1(g496a) were the most recurrent, being present in 3/3 affected members of the index family, detected in 8% of the unrelated familial cases, and present in 0.5% of the nonfamilial cases and in <0.05 of a control population. LAPTM5 and HCLS1 appeared as relevant WM candidate genes that characterized familial WM individuals and were also functionally relevant to the tumor clone. These findings highlight potentially novel contributors for the genetic predisposition to familial WM and indicate that LAPTM5(c403t) and HCLS1(g496a) may represent predisposition alleles in patients with familial WM.


Asunto(s)
Proteínas Sanguíneas/genética , Exoma , Predisposición Genética a la Enfermedad , Mutación de Línea Germinal , Proteínas de la Membrana/genética , Macroglobulinemia de Waldenström/genética , Proteínas Adaptadoras Transductoras de Señales , Familia , Femenino , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Masculino
12.
Rinsho Ketsueki ; 59(6): 695-697, 2018.
Artículo en Japonés | MEDLINE | ID: mdl-29973445

RESUMEN

A 76-year-old woman presented to our hospital with leukocytosis and abnormal lymphocytes. M protein of the immunoglobulin G (IgG) type was detected using immunoelectrophoresis. A bone marrow biopsy revealed infiltration of small mature lymphocytes, lymphoplasmacytoid cells with Dutcher bodies, grape cells, and Russell bodies. The MYD88 L265P mutation was detected in the abnormal peripheral lymphocytes, and a diagnosis of lymphoplasmacytoid lymphoma was established. MYD88 L265P mutation analysis is useful for making a diagnosis of non-IgM lymphoplasmacytoid lymphoma because it enables the differentiation from other low-grade B-cell malignancies.


Asunto(s)
Inmunoglobulina G , Factor 88 de Diferenciación Mieloide/genética , Paraproteínas , Macroglobulinemia de Waldenström/diagnóstico , Anciano , Análisis Mutacional de ADN , Femenino , Humanos , Mutación , Macroglobulinemia de Waldenström/genética
14.
Am J Hematol ; 92(8): E138-E145, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28474779

RESUMEN

The tumor suppressors B-lymphocyte-induced maturation protein-1 (BLIMP-1) and p53 play a crucial role in B-cell lymphomas, and their inactivation contributes to the pathogenesis of a wide spectrum of lymphoid malignancies, including diffuse large B-cell lymphomas (DLBCLs). Patients with activated B-cell-like (ABC) DLBCL may present with loss of BLIMP-1, c-Myc over-expression, decreased p53, and poor prognosis. Nevertheless, there is a lack of in vivo models recapitulating the biology of high-grade ABC DLBCL. We therefore aimed to develop an in vivo model aiming to recapitulate the phenotype observed in this cohort of patients. A Cre-Lox approach was used to achieve inactivation of both p53 and BLIMP-1 in murine B-cells. Contextual ablation of BLIMP-1 and p53 led to development of IgM-positive B-cell lymphoma with an aggressive phenotype, supported by c-Myc up-regulation, and accumulation of somatic mutations, as demonstrated by whole exome sequencing. Sensitivity of B-tumor cells to BTK inhibition was demonstrated. This model mirrors what reported in patients with ABC DLBLC, and therefore represents a novel model for studying the biology of ABC-DLBCL harboring the dual loss of BLIMP-1/p53 and c-Myc over-expression.


Asunto(s)
Linfocitos B/metabolismo , Linfocitos B/patología , Transformación Celular Neoplásica/genética , Proteínas Represoras/deficiencia , Proteína p53 Supresora de Tumor/deficiencia , Animales , Linfocitos B/efectos de los fármacos , Biomarcadores , Evolución Clonal/genética , Modelos Animales de Enfermedad , Expresión Génica , Regulación Neoplásica de la Expresión Génica , Genes myc , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Ratones , Ratones Transgénicos , Factor 1 de Unión al Dominio 1 de Regulación Positiva , Inhibidores de Proteínas Quinasas/farmacología
16.
Curr Osteoporos Rep ; 15(5): 499-506, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28889371

RESUMEN

PURPOSE OF THE REVIEW: Herein we dissect mechanisms behind the dissemination of cancer cells from primary tumor site to the bone marrow, which are necessary for metastasis development, with a specific focus on multiple myeloma. RECENT FINDINGS: The ability of tumor cells to invade vessels and reach the systemic circulation is a fundamental process for metastasis development; however, the interaction between clonal cells and the surrounding microenvironment is equally important for supporting colonization, survival, and growth in the secondary sites of dissemination. The intrinsic propensity of tumor cells to recognize a favorable milieu where to establish secondary growth is the basis of the "seed and soil" theory. This theory assumes that certain tumor cells (the "seeds") have a specific affinity for the milieu of certain organs (the "soil"). Recent literature has highlighted the important contributions of the vascular niche to the hospitable "soil" within the bone marrow. In this review, we discuss the crucial role of stromal cells and endothelial cells in supporting primary growth, homing, and metastasis to the bone marrow, in the context of multiple myeloma, a plasma cell malignancy with the unique propensity to primarily grow and metastasize to the bone marrow.


Asunto(s)
Médula Ósea/irrigación sanguínea , Neoplasias Óseas/secundario , Tejido Conectivo/irrigación sanguínea , Células Endoteliales/metabolismo , Células Madre Mesenquimatosas/metabolismo , Mieloma Múltiple/patología , Médula Ósea/metabolismo , Tejido Conectivo/metabolismo , Células Endoteliales/citología , Humanos , Células Madre Mesenquimatosas/citología , Mieloma Múltiple/metabolismo , Metástasis de la Neoplasia , Microambiente Tumoral
17.
Curr Opin Hematol ; 23(4): 426-33, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27101529

RESUMEN

PURPOSE OF REVIEW: Multiple myeloma is a plasma cell malignancy evolving in the bone marrow and leading to end organ damage such as bone lesions, cytopenias, and kidney failure. This review delineates recent advances in the molecular mechanisms leading to tumor progression in multiple myeloma. Two different aspects enable tumor expansion: cell autonomous through genomic alterations in the tumor clone and noncell autonomous deregulations in the bone marrow tumor microenvironment. These alterations provide the framework for the continuous progression of multiple myeloma from early precursor conditions such as monoclonal gammopathy of undetermined significance and smoldering multiple myeloma to overt multiple myeloma. RECENT FINDINGS: In this review, we discuss recent findings in the genomic alterations that occur in the tumor clone such as somatic genomic mutations, copy number variation and chromosomal translocation, and delineate noncell autonomous deregulations in which tumor cells take advantage of a permissive microenvironment to further proliferate. The latter compartment includes interaction with bone marrow stromal cells, osteoblasts, osteoclasts, and immune escape. SUMMARY: Understanding the mechanisms that lead tumor progression from early stages to overt multiple myeloma could guide to more effective therapies and therefore prevent disease progression.


Asunto(s)
Variación Genética , Mieloma Múltiple/genética , Mieloma Múltiple/patología , Lesiones Precancerosas , Microambiente Tumoral , Médula Ósea/metabolismo , Médula Ósea/patología , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Evolución Clonal , Progresión de la Enfermedad , Células Endoteliales/metabolismo , Humanos , Gammopatía Monoclonal de Relevancia Indeterminada/genética , Gammopatía Monoclonal de Relevancia Indeterminada/metabolismo , Gammopatía Monoclonal de Relevancia Indeterminada/patología , Mieloma Múltiple/inmunología , Mieloma Múltiple/metabolismo , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología
18.
Cancer Treat Res ; 169: 63-102, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27696259

RESUMEN

Unprecedented advances in multiple myeloma (MM) therapy during the last 15 years are predominantly based on our increasing understanding of the pathophysiologic role of the bone marrow (BM) microenvironment. Indeed, new treatment paradigms, which incorporate thalidomide, immunomodulatory drugs (IMiDs), and proteasome inhibitors, target the tumor cell as well as its BM microenvironment. Ongoing translational research aims to understand in more detail how disordered BM-niche functions contribute to MM pathogenesis and to identify additional derived targeting agents. One of the most exciting advances in the field of MM treatment is the emergence of immune therapies including elotuzumab, daratumumab, the immune checkpoint inhibitors, Bispecific T-cell engagers (BiTes), and Chimeric antigen receptor (CAR)-T cells. This chapter will review our knowledge on the pathophysiology of the BM microenvironment and discuss derived novel agents that hold promise to further improve outcome in MM.


Asunto(s)
Mieloma Múltiple/patología , Mieloma Múltiple/fisiopatología , Microambiente Tumoral , Animales , Médula Ósea/patología , Médula Ósea/fisiopatología , Humanos
19.
Cancer Treat Res ; 169: 35-49, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27696257

RESUMEN

Multiple myeloma is characterized by clonal proliferation of plasma cells within the bone marrow resulting in anemia, lytic bone lesions, hypercalcemia, and renal impairment. Despite advanced in our understanding of this complex disease in recent years, it is still considered an incurable malignancy. This is, in part, due to the highly heterogenous genomic and phenotypic nature of the disease, which is to date incompletely understood. It is clear that a deeper level of knowledge of the biological events underlying the development of these diseases is needed to identify new targets and generate effective novel therapies. MicroRNAs (miRNAs), which are single strand, 20-nucleotide, noncoding RNA's, are key regulators of gene expression and have been reported to exert transcriptional control in multiple myeloma. miRNAs are now recognized to play a role in many key areas such as cellular proliferation, differentiation, apoptosis and stress response. Substantial advances have been made in recent years in terms of our understanding of the biological role of miRNAs in a diverse range of hematological and solid malignancues, In multiple myeloma these advances have yielded new information of prognostic and diagnostic relevance which have helped to shed light on epigenetic regulation in this disease.


Asunto(s)
Epigénesis Genética , Mieloma Múltiple/genética , Humanos
20.
Blood ; 121(6): 962-70, 2013 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-23212521

RESUMEN

PU.1 has previously been shown to be down-regulated in classical Hodgkin lymphoma (cHL) cells via promoter methylation. We performed bisulfite sequencing and proved that the promoter region and the -17 kb upstream regulatory element of the PU.1 gene were highly methylated. To evaluate whether down-regulation of PU.1 is essential for the growth of cHL cells, we conditionally expressed PU.1 in 2 cHL cell lines, L428 and KM-H2. Overexpression of PU.1 induced complete growth arrest and apoptosis in both cell lines. Furthermore, in a Hodgkin lymphoma tumor xenograft model using L428 and KM-H2 cell lines, overexpression of PU.1 led to tumor regression or stable disease. Lentiviral transduction of PU.1 into primary cHL cells also induced apoptosis. DNA microarray analysis revealed that among genes related to cell cycle and apoptosis, p21 (CDKN1A) was highly up-regulated in L428 cells after PU.1 induction. Stable knockdown of p21 rescued PU.1-induced growth arrest in L428 cells, suggesting that the growth arrest and apoptosis observed are at least partially dependent on p21 up-regulation. These data strongly suggest that PU.1 is a potent tumor suppressor in cHL and that induction of PU.1 with demethylation agents and/or histone deacetylase inhibitors is worth exploring as a possible therapeutic option for patients with cHL.


Asunto(s)
Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Enfermedad de Hodgkin/genética , Proteínas Proto-Oncogénicas/genética , Transactivadores/genética , Animales , Apoptosis/genética , Secuencia de Bases , Western Blotting , Línea Celular Tumoral , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Metilación de ADN , Regulación hacia Abajo , Enfermedad de Hodgkin/metabolismo , Enfermedad de Hodgkin/patología , Humanos , Lentivirus/genética , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Datos de Secuencia Molecular , Análisis de Secuencia por Matrices de Oligonucleótidos , Regiones Promotoras Genéticas/genética , Proteínas Proto-Oncogénicas/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transactivadores/metabolismo , Transducción Genética , Células Tumorales Cultivadas , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA