Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 133
Filtrar
1.
Lett Appl Microbiol ; 75(4): 785-795, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34821400

RESUMO

Microbial fuel cell (MFC) is an emerging technology which has been immensely investigated for wastewater treatment along with electricity generation. In the present study, the treatment efficiency of MFC was investigated for hydrocarbon containing wastewater by optimizing various parameters of MFC. Mediator-less MFC (1·2 l) was constructed, and its performance was compared with mediated MFC with Escherichia coli as a biocatalyst. MFC with electrode having biofilm proved to be better compared with MFC inoculated with suspended cells. Analysis of increasing surface area of electrode by increasing their numbers indicated increase in COD reduction from 55 to 75%. Catholyte volume was optimized to be 750 ml. Sodium benzoate (0·721 g l-1 ) and actual common effluent treatment plant (CETP) wastewater as anolyte produced 0·8 and 0·6 V voltage and 89 and 50% COD reduction, respectively, when a novel consortium of four bacterial strains were used. Twenty MFC systems with the developed consortium when electrically connected in series-parallel connection were able to generate 2·3 V and 0·5 mA current. This is the first report demonstrating the application of CETP wastewater in the MFC system, which shows potential of the system towards degradation of complex organic components present in industrial wastewater.


Assuntos
Fontes de Energia Bioelétrica , Purificação da Água , Biodegradação Ambiental , Fontes de Energia Bioelétrica/microbiologia , Eletricidade , Eletrodos , Benzoato de Sódio , Águas Residuárias
2.
N Engl J Med ; 365(15): 1384-95, 2011 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-21995386

RESUMO

BACKGROUND: Myelodysplastic syndromes are a diverse and common group of chronic hematologic cancers. The identification of new genetic lesions could facilitate new diagnostic and therapeutic strategies. METHODS: We used massively parallel sequencing technology to identify somatically acquired point mutations across all protein-coding exons in the genome in 9 patients with low-grade myelodysplasia. Targeted resequencing of the gene encoding RNA splicing factor 3B, subunit 1 (SF3B1), was also performed in a cohort of 2087 patients with myeloid or other cancers. RESULTS: We identified 64 point mutations in the 9 patients. Recurrent somatically acquired mutations were identified in SF3B1. Follow-up revealed SF3B1 mutations in 72 of 354 patients (20%) with myelodysplastic syndromes, with particularly high frequency among patients whose disease was characterized by ring sideroblasts (53 of 82 [65%]). The gene was also mutated in 1 to 5% of patients with a variety of other tumor types. The observed mutations were less deleterious than was expected on the basis of chance, suggesting that the mutated protein retains structural integrity with altered function. SF3B1 mutations were associated with down-regulation of key gene networks, including core mitochondrial pathways. Clinically, patients with SF3B1 mutations had fewer cytopenias and longer event-free survival than patients without SF3B1 mutations. CONCLUSIONS: Mutations in SF3B1 implicate abnormalities of messenger RNA splicing in the pathogenesis of myelodysplastic syndromes. (Funded by the Wellcome Trust and others.).


Assuntos
Síndromes Mielodisplásicas/genética , Fosfoproteínas/genética , Mutação Puntual , Ribonucleoproteína Nuclear Pequena U2/genética , Eritrócitos/patologia , Perfilação da Expressão Gênica , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Fenótipo , Fatores de Processamento de RNA
3.
Semin Hematol ; 60(2): 118-124, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-37099031

RESUMO

Recent advances in the understanding of Waldenström macroglobulinemia (WM) biology have impacted the development of effective novel agents and improved our knowledge of how the genomic background of WM may influence selection of therapy. Consensus Panel 7 (CP7) of the 11th International Workshop on WM was convened to examine the current generation of completed and ongoing clinical trials involving novel agents, consider updated data on WM genomics, and make recommendations on the design and prioritization of future clinical trials. CP7 considers limited duration and novel-novel agent combinations to be the priority for the next generation of clinical trials. Evaluation of MYD88, CXCR4 and TP53 at baseline in the context of clinical trials is crucial. The common chemoimmunotherapy backbones, bendamustine-rituximab (BR) and dexamethasone, rituximab and cyclophosphamide (DRC), may be considered standard-of-care for the frontline comparative studies. Key unanswered questions include the definition of frailty in WM; the importance of attaining a very good partial response or better (≥VGPR), within stipulated time frame, in determining survival outcomes; and the optimal treatment of WM populations with special needs.


Assuntos
Macroglobulinemia de Waldenstrom , Humanos , Macroglobulinemia de Waldenstrom/tratamento farmacológico , Macroglobulinemia de Waldenstrom/genética , Rituximab/uso terapêutico , Consenso , Ciclofosfamida/uso terapêutico , Cloridrato de Bendamustina/uso terapêutico
4.
Science ; 293(5532): 1133-6, 2001 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-11498590

RESUMO

Dynamic control of interferon-beta (IFN-beta) gene expression requires the regulated assembly and disassembly of the enhanceosome, a higher-order nucleoprotein complex formed in response to virus infection. The enhanceosome activates transcription by recruiting the histone acetyltransferase proteins CREB binding protein (CBP) and p300/CBP-associated factors (PCAF)/GCN5, which, in addition to modifying histones, acetylate HMGI(Y), the architectural component required for enhanceosome assembly. We show that the accurate execution of the IFN-beta transcriptional switch depends on the ordered acetylation of the high-mobility group I protein HMGI(Y) by PCAF/GCN5 and CBP, which acetylate HMGI(Y) at distinct lysine residues on endogenous promoters. Whereas acetylation of HMGI(Y) by CBP at lysine-65 destabilizes the enhanceosome, acetylation of HMGI(Y) by PCAF/GCN5 at lysine-71 potentiates transcription by stabilizing the enhanceosome and preventing acetylation by CBP.


Assuntos
Elementos Facilitadores Genéticos , Regulação da Expressão Gênica , Proteínas de Grupo de Alta Mobilidade/metabolismo , Interferon beta/genética , Proteínas de Saccharomyces cerevisiae , Fatores de Transcrição/metabolismo , Ativação Transcricional , Acetilação , Acetiltransferases/metabolismo , Sequência de Aminoácidos , Proteína de Ligação a CREB , Proteínas de Ciclo Celular , Proteína HMGA1a , Células HeLa , Proteínas de Grupo de Alta Mobilidade/química , Histona Acetiltransferases , Histonas/metabolismo , Humanos , Lisina/metabolismo , Dados de Sequência Molecular , Mutação , Proteínas Nucleares/metabolismo , Regiões Promotoras Genéticas , Ligação Proteica , Proteínas Recombinantes/metabolismo , Respirovirus/fisiologia , Transativadores/metabolismo , Fatores de Transcrição/química , Transfecção , Fatores de Transcrição de p300-CBP
5.
J Transl Sci ; 5(2)2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30873294

RESUMO

A variety of factors, whether extracellular (mutagens/carcinogens and viruses in the environment, chronic inflammation and radiation associated with the environment and/or electronic devices/machines) and/or intracellular (oxidative metabolites of food, oxidative stress due to inflammation, acid production, replication stress, DNA replication/repair errors, and certain hormones, cytokines, growth factors), pose a constant threat to the genomic integrity of a living cell. However, in the normal cellular environment multiple biological pathways including DNA repair, cell cycle, apoptosis and the immune system work in a precise, regulated (tightly controlled), timely and concerted manner to ensure genomic integrity, stability and proper functioning of a cell. If damage to DNA takes place, it is efficiently and accurately repaired by the DNA repair systems. Homologous recombination (HR) which utilizes either a homologous chromosome (in G1 phase) or a sister chromatid (in G2) as a template to repair the damage, is known to be the most precise repair system. HR in G2 which utilizes a sister chromatid as a template is also called an error free repair system. If DNA damage in a cell is so extensive that it overwhelms the repair system/s, the cell is eliminated by apoptosis. Thus, multiple pathways ensure that genome of a cell is intact and stable. However, constant exposure to DNA damage and/or dysregulation of DNA repair mechanism/s poses a risk of mutation and cancer. Oncogenesis, which seems to be a multistep process, is associated with acquisition of a number of genomic changes that enable a normal cell to progress from benign to malignant transformation. Transformed/cancer cells are recognized and killed by the immune system. However, the ongoing acquisition of new genomic changes enables cancer cells to survive/escape immune attack, evolve into a more aggressive phenotype, and eventually develop resistance to therapy. Although DNA repair (especially the HR) and the immune system play unique roles in preserving genomic integrity of a cell, they can also contribute to DNA damage, genomic instability and oncogenesis. The purpose of this article is to highlight the roles of DNA repair (especially HR) and the immune system in genomic evolution, with special focus on gastrointestinal cancer.

6.
Oncogene ; 26(16): 2374-80, 2007 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-17016430

RESUMO

Bcl-2 or Bcl-X(L) confers resistance to chemotherapy in multiple myeloma (MM). Here we characterized the effects of ABT-737, a potent small-molecule inhibitor of antiapoptotic proteins Bcl-2, Bcl-X(L) and Bcl-w with markedly higher affinity than previously reported compounds, on human MM cells. ABT-737 induces apoptosis in MM cells, including those resistant to conventional therapy. Examination of purified patient MM cells demonstrated similar results, without significant toxicity against normal peripheral blood mononuclear cells and MM bone marrow stromal cells. Importantly, ABT-737 decreases the viability of bortezomib-, dexamethasone-(Dex) and thalidomide-refractory patient MM cells. Additionally, ABT-737 abrogates MM cell growth triggered by interleukin-6 or insulin-like growth factor-1. Mechanistic studies show that ABT-737-induced apoptosis is associated with activation of caspase-8, caspase-9 and caspase-3, followed by poly(ADP-ribose) polymerase cleavage. Combining ABT-737 with proteasome inhibitor bortezomib, melphalan or dexamethasone induces additive anti-MM activity. Taken together, our study provides the rationale for clinical protocols evaluating ABT-737, alone and together with botezomib, mephalan or dexamethasone, to enhance MM cell killing, overcome drug resistance conferred by Bcl-2 and improve patient outcome in MM.


Assuntos
Antineoplásicos/farmacologia , Proteínas Reguladoras de Apoptose/antagonistas & inibidores , Compostos de Bifenilo/farmacologia , Mieloma Múltiplo/tratamento farmacológico , Nitrofenóis/farmacologia , Proteínas Proto-Oncogênicas c-bcl-2/antagonistas & inibidores , Sulfonamidas/farmacologia , Proteína bcl-X/antagonistas & inibidores , Antineoplásicos/toxicidade , Células da Medula Óssea/efeitos dos fármacos , Ácidos Borônicos/toxicidade , Bortezomib , Divisão Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Humanos , Mieloma Múltiplo/patologia , Piperazinas/farmacologia , Pirazinas/toxicidade , Células Tumorais Cultivadas
7.
Leukemia ; 32(3): 752-764, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29089645

RESUMO

X-box binding protein 1 (XBP1), CD138 (Syndecan-1) and CS1 (SLAMF7) are highly expressed antigens in cancers including multiple myeloma (MM). Here, we identify and characterize immunogenic HLA-A24 peptides derived from these antigens for potential vaccination therapy of HLA-A24+ patients with MM. The identified immunogenic HLA-A24-specific XBP1 unspliced (UN)185-193 (I S P W I L A V L), XBP1 spliced (SP)223-231 (V Y P E G P S S L), CD138265-273 (I F A V C L V G F) and CS1240-248 (L F V L G L F L W) peptides induced antigen-specific CTL with anti-MM activity in an HLA-A24 restricted manner. Furthermore, a cocktail containing the four HLA-A24 peptides evoked MM-specific CTL with distinct phenotypic profiles (CD28, CD40L, 41BB, CD38, CD69) and anti-tumor activities, evidenced by perforin upregulation, CD107a degranulation (cytotoxicity) and Th1-type cytokines (IFN-γ/IL-2/TNF-α) production in response to HLA-A24+ MM cells. The multipeptide-specific CTL included antigen-specific memory CD8+ T cells expressing both T-cell activation (CD38, CD69) and immune checkpoints antigens (CTLA, PD-1, LAG-3, TIM-3). These results provide the framework for a multipeptide vaccination therapy to induce tumor-specific CTL in HLA-A24-positive patients with myeloma and other cancers expressing these antigens.


Assuntos
ADP-Ribosil Ciclase 1/imunologia , Antígeno HLA-A24/imunologia , Mieloma Múltiplo/imunologia , Peptídeos/imunologia , Especificidade do Receptor de Antígeno de Linfócitos T/imunologia , Linfócitos T Citotóxicos/imunologia , Proteína 1 de Ligação a X-Box/imunologia , ADP-Ribosil Ciclase 1/química , ADP-Ribosil Ciclase 1/metabolismo , Sequência de Aminoácidos , Biomarcadores , Linhagem Celular Tumoral , Citocinas/metabolismo , Citotoxicidade Imunológica , Epitopos de Linfócito T/química , Epitopos de Linfócito T/imunologia , Epitopos de Linfócito T/metabolismo , Antígeno HLA-A24/genética , Antígeno HLA-A24/metabolismo , Humanos , Memória Imunológica , Peptídeos e Proteínas de Sinalização Intercelular , Ativação Linfocitária/imunologia , Mieloma Múltiplo/genética , Mieloma Múltiplo/metabolismo , Peptídeos/química , Peptídeos/metabolismo , Fenótipo , Ligação Proteica , Linfócitos T Citotóxicos/metabolismo , Proteína 1 de Ligação a X-Box/química , Proteína 1 de Ligação a X-Box/metabolismo
8.
Leukemia ; 32(4): 996-1002, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29158558

RESUMO

Arginine methyltransferases critically regulate cellular homeostasis by modulating the functional outcome of their substrates. The protein arginine methyltransferase 5 (PRMT5) is an enzyme involved in growth and survival pathways promoting tumorigenesis. However, little is known about the biologic function of PRMT5 and its therapeutic potential in multiple myeloma (MM). In the present study, we identified and validated PRMT5 as a new therapeutic target in MM. PRMT5 is overexpressed in patient MM cells and associated with decreased progression-free survival and overall survival. Either genetic knockdown or pharmacological inhibition of PRMT5 with the inhibitor EPZ015666 significantly inhibited growth of both cell lines and patient MM cells. Furthermore, PRMT5 inhibition abrogated NF-κB signaling. Interestingly, mass spectrometry identified a tripartite motif-containing protein 21 TRIM21 as a new PRMT5-partner, and we delineated a TRIM21-dependent mechanism of NF-κB inhibition. Importantly, oral administration of EPZ015666 significantly decreased MM growth in a humanized murine model of MM. These data both demonstrate the oncogenic role and prognostic relevance of PRMT5 in MM pathogenesis, and provide the rationale for novel therapies targeting PRMT5 to improve patient outcome.


Assuntos
Mieloma Múltiplo/metabolismo , Mieloma Múltiplo/patologia , Proteína-Arginina N-Metiltransferases/metabolismo , Animais , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/fisiologia , Humanos , Isoquinolinas/farmacologia , NF-kappa B/metabolismo , Prognóstico , Pirimidinas/farmacologia , Ribonucleoproteínas/metabolismo , Transdução de Sinais/efeitos dos fármacos
9.
Leukemia ; 32(4): 1003-1015, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29158557

RESUMO

Dendritic cells (DCs) have a key role in regulating tumor immunity, tumor cell growth and drug resistance. We hypothesized that multiple myeloma (MM) cells might recruit and reprogram DCs to a tumor-permissive phenotype by changes within their microRNA (miRNA) network. By analyzing six different miRNA-profiling data sets, miR-29b was identified as the only miRNA upregulated in normal mature DCs and significantly downregulated in tumor-associated DCs. This finding was validated in primary DCs co-cultured in vitro with MM cell lines and in primary bone marrow DCs from MM patients. In DCs co-cultured with MM cells, enforced expression of miR-29b counteracted pro-inflammatory pathways, including signal transducer and activator of transcription 3 and nuclear factor-κB, and cytokine/chemokine signaling networks, which correlated with patients' adverse prognosis and development of bone disease. Moreover, miR-29b downregulated interleukin-23 in vitro and in the SCID-synth-hu in vivo model, and antagonized a Th17 inflammatory response. All together, these effects translated into strong anti-proliferative activity and reduction of genomic instability of MM cells. Our study demonstrates that MM reprograms the DCs functional phenotype by downregulating miR-29b whose reconstitution impairs DCs ability to sustain MM cell growth and survival. These results underscore miR-29b as an innovative and attractive candidate for miRNA-based immune therapy of MM.


Assuntos
Células Dendríticas/patologia , Inflamação/genética , MicroRNAs/genética , Mieloma Múltiplo/genética , Mieloma Múltiplo/patologia , Animais , Medula Óssea/patologia , Linhagem Celular Tumoral , Proliferação de Células/genética , Regulação para Baixo/genética , Feminino , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Camundongos , Camundongos SCID , NF-kappa B/genética , Fator de Transcrição STAT3/genética , Regulação para Cima/genética
10.
Leukemia ; 32(1): 111-119, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28588253

RESUMO

Despite the development of novel drugs, alkylating agents remain an important component of therapy in multiple myeloma (MM). DNA repair processes contribute towards sensitivity to alkylating agents and therefore we here evaluate the role of nucleotide excision repair (NER), which is involved in the removal of bulky adducts and DNA crosslinks in MM. We first evaluated NER activity using a novel functional assay and observed a heterogeneous NER efficiency in MM cell lines and patient samples. Using next-generation sequencing data, we identified that expression of the canonical NER gene, excision repair cross-complementation group 3 (ERCC3), significantly impacted the outcome in newly diagnosed MM patients treated with alkylating agents. Next, using small RNA interference, stable knockdown and overexpression, and small-molecule inhibitors targeting xeroderma pigmentosum complementation group B (XPB), the DNA helicase encoded by ERCC3, we demonstrate that NER inhibition significantly increases sensitivity and overcomes resistance to alkylating agents in MM. Moreover, inhibiting XPB leads to the dual inhibition of NER and transcription and is particularly efficient in myeloma cells. Altogether, we show that NER impacts alkylating agents sensitivity in myeloma cells and identify ERCC3 as a potential therapeutic target in MM.


Assuntos
Reparo do DNA/genética , Mieloma Múltiplo/genética , Linhagem Celular Tumoral , DNA Helicases/genética , Proteínas de Ligação a DNA/genética , Humanos , Transcrição Gênica/genética , Xeroderma Pigmentoso/genética
11.
Leukemia ; 32(3): 744-751, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-28924240

RESUMO

14-3-3 proteins are a family of master regulators of intracellular signaling, yet their impact on proteasome function is unknown. We demonstrate that 14-3-3ζ binds the 11S proteasome activator, limiting proteasome assembly and cellular capacity for protein degradation. To define the functional impact of 14-3-3ζ proteasomal binding in myeloma cells, silencing and overexpression experiments are performed. We find that downregulation of 14-3-3ζ impairs myeloma cell growth and confers resistance to clinically used proteasome inhibitors. In a large cohort of newly diagnosed myeloma patients, elevated expression of 14-3-3ζ is associated with high risk myeloma genetic subtypes and worse prognosis overall. Our work demonstrates the important role of 14-3-3ζ in regulating proteasome function, myeloma cell growth and sensitivity to therapeutics, and suggests regulation of 14-3-3ζ as a new approach in myeloma therapy.


Assuntos
Proteínas 14-3-3/metabolismo , Antineoplásicos/farmacologia , Resistencia a Medicamentos Antineoplásicos , Complexo de Endopeptidases do Proteassoma/metabolismo , Inibidores de Proteassoma/farmacologia , Proteínas 14-3-3/genética , Linhagem Celular Tumoral , Humanos , Mieloma Múltiplo/genética , Mieloma Múltiplo/metabolismo , Mieloma Múltiplo/mortalidade , Mieloma Múltiplo/patologia , Ligação Proteica , Proteólise , Transdução de Sinais/efeitos dos fármacos
12.
J Clin Invest ; 85(1): 300-4, 1990 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-2104881

RESUMO

To study the proliferative response of hematopoietic cells to growth factors at the molecular level, we developed a cell-free system for growth factor-dependent initiation of genomic DNA replication. Nuclei were isolated from the IL-3-dependent cell line NFS/N1-H7 after a 10-h period of IL-3 deprivation. Cytosolic and membrane-containing subcellular fractions were prepared from proliferating NFS/N1-H7 cells. Nuclei from the nonproliferating cells (+/- IL-3) showed essentially no incorporation of [3H]thymidine during a 16-h incubation with a mixture of unlabeled GTP, ATP, UTP, CTP, dGTP, dATP, dCTP, and [3H]dTTP. When the combination of IL-3, a cytosolic fraction, and a membrane-containing fraction from proliferating cells was added to nuclei from nonproliferating cells, a burst of [3H]thymidine incorporation into DNA began after a 12-h lag period, attained a maximal rate at 16 h, and reached a level of 860 pmol thymidine/10(6) nuclei at 24 h (corresponding to replication of approximately 56% total mouse genomic DNA). This DNA synthesis was inhibited approximately 90% by the specific DNA polymerase alpha inhibitor aphidicolin. Deletion of a single cellular component or IL-3 from the system resulted in a marked reduction of DNA replication (-membrane, 80 +/- 4%; -cytosol, 90% +/- 4%; -IL-3, 74 +/- 7% inhibition). This model requires a growth factor (IL-3), a sedimentable cell fraction containing its receptor and possibly additional membrane-associated components, and a cytosolic fraction. It appears to recapitulate the molecular events required for progression from early G1 to S phase of the cell cycle induced by IL-3 binding to its receptor.


Assuntos
Núcleo Celular/metabolismo , Replicação do DNA/efeitos dos fármacos , Interleucina-3/farmacologia , Animais , Afidicolina , Bromodesoxiuridina/metabolismo , Ciclo Celular/efeitos dos fármacos , Linhagem Celular , Núcleo Celular/efeitos dos fármacos , Sistema Livre de Células , Diterpenos/farmacologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Células-Tronco Hematopoéticas/metabolismo , Cinética , Camundongos , Moldes Genéticos
13.
Mol Cell Biol ; 20(20): 7572-82, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11003653

RESUMO

We have previously described a SWI/SNF-related protein complex (PYR complex) that is restricted to definitive (adult-type) hematopoietic cells and that specifically binds DNA sequences containing long stretches of pyrimidines. Deletion of an intergenic DNA-binding site for this complex from a human beta-globin locus construct results in delayed human gamma- to beta-globin switching in transgenic mice, suggesting that the PYR complex acts to facilitate the switch. We now show that PYR complex DNA-binding activity also copurifies with subunits of a second type of chromatin-remodeling complex, nucleosome-remodeling deacetylase (NuRD), that has been shown to have both nucleosome-remodeling and histone deacetylase activities. Gel supershift assays using antibodies to the ATPase-helicase subunit of the NuRD complex, Mi-2 (CHD4), confirm that Mi-2 is a component of the PYR complex. In addition, we show that the hematopoietic cell-restricted zinc finger protein Ikaros copurifies with PYR complex DNA-binding activity and that antibodies to Ikaros also supershift the complex. We also show that NuRD and SWI/SNF components coimmunopurify with each other as well as with Ikaros. Competition gel shift experiments using partially purified PYR complex and recombinant Ikaros protein indicate that Ikaros functions as a DNA-binding subunit of the PYR complex. Our results suggest that Ikaros targets two types of chromatin-remodeling factors-activators (SWI/SNF) and repressors (NuRD)-in a single complex (PYR complex) to the beta-globin locus in adult erythroid cells. At the time of the switch from fetal to adult globin production, the PYR complex is assembled and may function to repress gamma-globin gene expression and facilitate gamma- to beta-globin switching.


Assuntos
Autoantígenos , Cromatina/química , Cromatina/metabolismo , Proteínas de Ligação a DNA/metabolismo , Leucemia Eritroblástica Aguda/metabolismo , Fatores de Transcrição/metabolismo , Adenosina Trifosfatases/metabolismo , Envelhecimento/fisiologia , Animais , Cromatina/genética , DNA/genética , DNA/metabolismo , DNA Helicases/metabolismo , Regulação da Expressão Gênica , Globinas/genética , Histona Desacetilases/metabolismo , Histonas/química , Histonas/metabolismo , Humanos , Fator de Transcrição Ikaros , Leucemia Eritroblástica Aguda/patologia , Substâncias Macromoleculares , Complexo Mi-2 de Remodelação de Nucleossomo e Desacetilase , Camundongos , Camundongos Transgênicos , Proteínas Nucleares/metabolismo , Testes de Precipitina , Ligação Proteica , Complexo Correpressor Histona Desacetilase e Sin3 , Especificidade por Substrato , Células Tumorais Cultivadas , Dedos de Zinco
14.
Oncogene ; 36(40): 5631-5638, 2017 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-28581522

RESUMO

Proteasome inhibition is an effective therapy for multiple myeloma (MM) patients; however, the emergence of drug resistance is common. Novel therapeutic strategies to overcome proteasome inhibitor resistance are needed. In this study, we examined whether targeting deubiquitylating (DUB) enzymes upstream of 20S proteasome overcomes proteasome inhibitor resistance. Gene expression analysis, immunohistochemical studies of MM patient bone marrow, reverse transcription-PCR and protein analysis show that Rpn11/POH1, a DUB enzyme upstream of 20S proteasome, is more highly expressed in patient MM cells than in normal plasma cells. Importantly, Rpn11 expression directly correlates with poor patient survival. Loss-of-function studies show that Rpn11-siRNA knockdown decreases MM cell viability. Pharmacological inhibition of Rpn11 with O-phenanthroline (OPA) blocks cellular proteasome function, induces apoptosis in MM cells and overcomes resistance to proteasome inhibitor bortezomib. Mechanistically, Rpn11 inhibition in MM cells activates caspase cascade and endoplasmic stress response signaling. Human MM xenograft model studies demonstrate that OPA treatment reduces progression of tumor growth and prolongs survival in mice. Finally, blockade of Rpn11 increases the cytotoxic activity of anti-MM agents lenalidomide, pomalidomide or dexamethasone. Overall, our preclinical data provide the rationale for targeting DUB enzyme Rpn11 upstream of 20S proteasome to enhance cytotoxicity and overcome proteasome inhibitor resistance in MM.


Assuntos
Antineoplásicos/uso terapêutico , Bortezomib/uso terapêutico , Mieloma Múltiplo/tratamento farmacológico , Inibidores de Proteassoma/uso terapêutico , Transativadores/antagonistas & inibidores , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células , Resistencia a Medicamentos Antineoplásicos , Humanos , Camundongos , Mieloma Múltiplo/enzimologia , Fenantrolinas/farmacologia , Prognóstico , Complexo de Endopeptidases do Proteassoma , Ubiquitinação , Ensaios Antitumorais Modelo de Xenoenxerto
15.
Nat Commun ; 8(1): 1893, 2017 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-29196615

RESUMO

Multiple myeloma is a plasma cell malignancy characterized by recurrent IgH translocations and well described genomic heterogeneity. Although transcriptome profiles in multiple myeloma has been described, landscape of expressed fusion genes and their clinical impact remains unknown. To provide a comprehensive and detailed fusion gene cartography and suggest new mechanisms of tumorigenesis in multiple myeloma, we performed RNA sequencing in a cohort of 255 newly diagnosed and homogeneously treated multiple myeloma patients with long follow-up. Here, we report that patients have on average 5.5 expressed fusion genes. Kappa and lambda light chains and IgH genes are main partners in a third of all fusion genes. We also identify recurrent fusion genes that significantly impact both progression-free and overall survival and may act as drivers of the disease. Lastly, we find a correlation between the number of fusions, the age of patients and the clinical outcome, strongly suggesting that genomic instability drives prognosis of the disease.


Assuntos
Fusão Gênica , Mieloma Múltiplo/genética , Idoso , Feminino , Humanos , Cadeias Pesadas de Imunoglobulinas/genética , Cadeias kappa de Imunoglobulina/genética , Cadeias lambda de Imunoglobulina/genética , Masculino , Pessoa de Meia-Idade , Análise de Sequência de RNA , Translocação Genética
16.
Leukemia ; 19(7): 1253-61, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15858615

RESUMO

Thalidomide alone or in combination with steroids has significant activity in multiple myeloma (MM). However, given its teratogenic potential, analogs have been synthesized, retaining the anti-MM activity without these side effects. We examined the anti-MM activity of two thalidomide analogs, CPS11 and CPS49. Direct cytotoxicity of the drugs on myeloma cell lines and patient myeloma cells was examined using thymidine uptake. Tumor cell apoptosis was evaluated by flow cytometry as well as Western blotting for caspase and PARP cleavage. Cellular signaling events were examined by immunoblotting for phosphorylated proteins. Both drugs inhibit proliferation of several MM cell lines sensitive and resistant to conventional therapies. They decrease secretion of IL-6, IGF, and VEGF by marrow stromal cells. Importantly, they inhibit proliferation of MM cells adherent to stromal cells. These drugs induce caspase-mediated apoptosis in MM cell lines, as well as patient MM cells. They inhibit the PI3K/Akt and JAK/STAT (signal transducers and activators of transcription) pathways in MM cells and are antiangiogenic in matrigel-based assays. CPS11 and CPS49 have potent antimyeloma activity and can overcome protective effects of the tumor microenvironment. They have potent antiangiogenic activity and direct effect on bone marrow stroma. These encouraging preclinical data provide the basis for further evaluation in the clinic.


Assuntos
Inibidores da Angiogênese/farmacologia , Antineoplásicos/farmacologia , Proliferação de Células/efeitos dos fármacos , Mieloma Múltiplo/tratamento farmacológico , Talidomida/análogos & derivados , Talidomida/farmacologia , Apoptose/efeitos dos fármacos , Adesão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , DNA/biossíntese , DNA/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Células Estromais/efeitos dos fármacos
17.
Cancer Res ; 54(7): 1657-9, 1994 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-8137277

RESUMO

Immunotherapy with lymphokine-activated killer (LAK) cells and interleukin 2 is one of the newer treatment modalities for cancer. This raises important questions concerning synergism or suppressive effects of other existing treatment modalities on adoptive immunotherapy with LAK cells. A tumor model with H4IIe hepatoma cells grown on each flank of ACI rats was developed to evaluate the effect of external beam irradiation of tumors on the subsequent concentration of LAK cells in these tumors. Tumors on one side were irradiated at 6, 12, or 16 Gy prior to injection of [3H]thymidine-labeled LAK cells. The effect of irradiation was measured as the ratio of 3H recovered in the unirradiated tumor compared to that in the irradiated tumor in the same animal as a function of dose and time after irradiation. This ratio was significantly greater than 1.0 for a radiation dose of 12 Gy (2.35 +/- 0.51) measured 2 days after irradiation, indicating a reduction in LAK cell numbers in the irradiated tumor. This reduction in LAK cell number persists up to at least 4 days following radiation exposure. A similar experiment using 125I-labeled interleukin 2 showed equal distribution in the irradiated and unirradiated tumors. Our data demonstrates that the concentration of LAK cells is markedly reduced by prior radiation, in contradistinction to increased uptake of immunoglobulins in irradiated tumors, as shown by others. If a similar reduction is observed for longer duration after radiation exposure, it might suggest a clinically important interaction between prior radiation exposure and adoptive immunotherapy.


Assuntos
Interleucina-2/farmacocinética , Radioisótopos do Iodo/farmacocinética , Células Matadoras Ativadas por Linfocina , Neoplasias Hepáticas Experimentais/radioterapia , Neoplasias Hepáticas Experimentais/terapia , Animais , Terapia Combinada , Imunoterapia Adotiva , Dosagem Radioterapêutica , Ratos , Ratos Endogâmicos ACI , Proteínas Recombinantes/farmacocinética , Baço/imunologia , Timidina/metabolismo , Distribuição Tecidual , Trítio
18.
Cancer Res ; 59(15): 3592-5, 1999 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-10446967

RESUMO

Adeno-associated virus (AAV) is a nonpathogenic, single-stranded DNA virus belonging to the parvoviridae family. Onco-suppressive properties of AAV against adenovirus, a DNA tumor virus, have been well documented. Rep78, a major regulatory protein of AAV, is believed to be responsible for its antioncogenic properties. Most DNA tumor viruses disturb the cell cycle pathways by essentially abrogating the functions of p53. Here we present evidence that AAV acts as an antiproliferative agent against adenovirus by protecting the adenoviral-mediated degradation of p53 as confirmed by both Western blot analysis and immunoprecipitation analysis with anti-p53 antibody. Coimmunoprecipitation experiments revealed that the AAV Rep78 is physically bound to p53 in vivo. Furthermore, the binding of purified p53 to the AAV Rep78 affinity column confirms their interaction. These results document for the first time that the antiproliferative effects of AAV against adenovirus are mediated, at least in part, by the interaction of AAV Rep78 with p53.


Assuntos
Adenoviridae/fisiologia , Proteínas E1B de Adenovirus/antagonistas & inibidores , Proteínas de Ligação a DNA/metabolismo , Dependovirus/fisiologia , Proteína Supressora de Tumor p53/metabolismo , Proteínas Virais/metabolismo , Divisão Celular , Linhagem Celular , Proteínas de Ligação a DNA/genética , Fibroblastos/virologia , Células HeLa/virologia , Humanos , Substâncias Macromoleculares , Splicing de RNA , Ubiquitinas/fisiologia , Proteínas Virais/genética
19.
Cancer Res ; 61(11): 4583-90, 2001 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-11389094

RESUMO

The high mobility group (HMG) proteins (HMGA1a, HMGA1b, and HMGA2) bind to DNA and interact with various transcriptional factors. Therefore, they play an important role in chromatin organization. HMGA protein expression is low in normal adult tissues, but abundant during embryonic development and in several experimental and human tumors. Blockage of HMGA expression inhibits the transformation of rat thyroid PC Cl 3 cells treated with oncogene-carrying retroviruses, thus implicating HMGA in rat thyroid transformation. To better understand the role of HMGA and to establish whether its up-regulated expression is sufficient to induce the transformed phenotype, we generated PC Cl 3 cells that overexpress the protein. We demonstrate that HMGA1b protein overexpression does not transform normal rat thyroid PC Cl 3 cells, but it deregulates their cell cycle: cells enter S-phase earlier and the G(2)-M transition is delayed. HMGA1-overexpressing cells undergo apoptosis through a pathway involving caspase-3 activation, probably consequent to the conflict between mitogenic pressure and the inability to proceed through the cell cycle. Using various HMGA1b gene mutations, we found that the third AT-hook domain and the acetylation site K60 are the protein regions required for induction of apoptosis in PC Cl 3 cells. In conclusion, although HMGA1 protein overexpression is associated with the malignant phenotype of rat and human thyroid cells, it does not transform normal thyroid cells in culture but leads them to programmed cell death.


Assuntos
Apoptose/fisiologia , Proteínas de Grupo de Alta Mobilidade/biossíntese , Glândula Tireoide/citologia , Glândula Tireoide/metabolismo , Animais , Proteína Quinase CDC2/metabolismo , Ciclo Celular/fisiologia , Divisão Celular/fisiologia , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Ciclinas/biossíntese , DNA Complementar/genética , Expressão Gênica , Proteínas de Grupo de Alta Mobilidade/genética , Mapeamento de Peptídeos , Isoformas de Proteínas , Ratos , Transfecção
20.
Leukemia ; 30(5): 1071-8, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26669975

RESUMO

Multiple myeloma (MM) is a plasma cell neoplasm with significant molecular heterogeneity. Gene expression profiling (GEP) has contributed significantly to our understanding of the underlying biology and has led to several prognostic gene signatures. However, the best way to apply these GEP signatures in clinical practice is unclear. In this study, we investigated the integration of proven prognostic signatures for improved patient risk stratification. Three publicly available MM GEP data sets that encompass newly diagnosed as well as relapsed patients were analyzed using standardized estimation of nine prognostic MM signature indices and simulations of signature index combinations. Cox regression analysis was used to assess the performance of simulated combination indices. Taking the average of multiple GEP signature indices was a simple but highly effective way of integrating multiple GEP signatures. Furthermore, although adding more signatures in general improved performance substantially, we identified a core signature combination, EMC92+HZDCD, as the top-performing prognostic signature combination across all data sets. In this study, we provided a rationale for gene signature integration and a practical strategy to choose an optimal risk score estimation in the presence of multiple prognostic signatures.


Assuntos
Regulação Neoplásica da Expressão Gênica , Mieloma Múltiplo/genética , Transcriptoma , Feminino , Humanos , Masculino , Prognóstico , Proteínas/genética , Recidiva , Análise de Regressão , Medição de Risco
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa