Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 161
Filtrar
Mais filtros

País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Adv Exp Med Biol ; 1221: 331-349, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32274716

RESUMO

It has been speculated for many years that heparanase plays an important role in the progression of cancer due largely to the finding that its expression is weak or absent in normal tissues but generally as tumors become more aggressive heparanase expression increases. However, it is only in the last decade or so that we have begun to understand the molecular mechanism behind the sinister role that heparanase plays in cancer. In this review, we describe the many functions of heparanase in promoting the growth, angiogenesis and metastasis of multiple myeloma, a devastating cancer that localizes predominantly within the bone marrow and spreads throughout the skeletal system devouring bone and ultimately leading to death of almost all patients diagnosed with this disease. We also explore recent discoveries related to how heparanase primes exosome biogenesis and how heparanase enhances myeloma tumor chemoresistance. Discovery of these multiple tumor-promoting pathways that are driven by heparanase identified the enzyme as an ideal target for therapy, an approach recently tested in a Phase I trial in myeloma patients.


Assuntos
Glucuronidase/metabolismo , Mieloma Múltiplo/enzimologia , Mieloma Múltiplo/patologia , Progressão da Doença , Resistencia a Medicamentos Antineoplásicos , Exossomos , Glucuronidase/antagonistas & inibidores , Humanos , Mieloma Múltiplo/irrigação sanguínea , Mieloma Múltiplo/tratamento farmacológico
2.
Stem Cells ; 35(3): 739-753, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27641212

RESUMO

Mesenchymal stromal cells (MSCs) are multipotent progenitor cells and there is much interest in how MSCs contribute to the regulation of the tumor microenvironment. Whether MSCs exert a supportive or suppressive effect on tumor progression is still controversial, but is likely dependent on a variety of factors that are tumor-type dependent. Multiple myeloma (MM) is characterized by growth of malignant plasma cells in the bone marrow. It has been shown that the progression of MM is governed by MSCs, which act as a stroma of the myeloma cells. Although stroma is created via mutual communication between myeloma cells and MSCs, the mechanism is poorly understood. Here we explored the role of lysophosphatidic acid (LPA) signaling in cellular events where MSCs were converted into either MM-supportive or MM-suppressive stroma. We found that myeloma cells stimulate MSCs to produce autotaxin, an indispensable enzyme for the biosynthesis of LPA, and LPA receptor 1 (LPA1) and 3 (LPA3) transduce opposite signals to MSCs to determine the fate of MSCs. LPA3-silenced MSCs (siLPA3-MSCs) exhibited cellular senescence-related phenotypes in vitro, and significantly promoted progression of MM and tumor-related angiogenesis in vivo. In contrast, siLPA1-MSCs showed resistance to cellular senescence in vitro, and efficiently delayed progression of MM and tumor-related angiogenesis in vivo. Consistently, anti-MM effects obtained by LPA1-silencing in MSCs were completely reproduced by systemic administration of Ki6425, an LPA1 antagonist. Collectively, our results indicate that LPA signaling determines the fate of MSCs and has potential as a therapeutic target in MM. Stem Cells 2017;35:739-753.


Assuntos
Senescência Celular , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/patologia , Mieloma Múltiplo/irrigação sanguínea , Mieloma Múltiplo/patologia , Neovascularização Patológica/metabolismo , Receptores de Ácidos Lisofosfatídicos/metabolismo , Transdução de Sinais , Animais , Fibroblastos Associados a Câncer/metabolismo , Fibroblastos Associados a Câncer/patologia , Carcinogênese/patologia , Linhagem Celular Tumoral , Proliferação de Células , Transdiferenciação Celular , Progressão da Doença , Feminino , Humanos , Camundongos Endogâmicos BALB C , Camundongos Nus , Diester Fosfórico Hidrolases/metabolismo , RNA Interferente Pequeno/metabolismo , Receptores de Ácidos Lisofosfatídicos/antagonistas & inibidores , Regulação para Cima , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Int J Mol Sci ; 19(7)2018 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-30002349

RESUMO

Angiogenesis is a constant hallmark of multiple myeloma (MM) progression and involves direct production of angiogenic cytokines by plasma cells and their induction within the bone marrow microenvironment. This article summarizes the more recent literature data concerning the employment of anti-angiogenic therapeutic agents actually used in preclinical models and clinical settings for the treatment of multiple myeloma.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Medula Óssea , Mieloma Múltiplo , Neovascularização Patológica , Microambiente Tumoral/efeitos dos fármacos , Animais , Medula Óssea/metabolismo , Medula Óssea/patologia , Humanos , Mieloma Múltiplo/irrigação sanguínea , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/metabolismo , Mieloma Múltiplo/patologia , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia
4.
Angiogenesis ; 20(4): 443-462, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28840415

RESUMO

Multiple myeloma (MM) is characterized by the clonal expansion of plasma cells in the bone marrow that leads to events such as bone destruction, anaemia and renal failure. Despite the several therapeutic options available, there is still no effective cure, and the standard survival is up to 4 years. The evolution from the asymptomatic stage of monoclonal gammopathy of undetermined significance to MM and the progression of the disease itself are related to cellular and molecular alterations in the bone marrow microenvironment, including the development of the vasculature. Post-natal vasculogenesis is characterized by the recruitment to the tumour vasculature of bone marrow progenitors, known as endothelial progenitor cells (EPCs), which incorporate newly forming blood vessels and differentiate into endothelial cells. Several processes related to EPCs, such as recruitment, mobilization, adhesion and differentiation, are tightly controlled by cells and molecules in the bone marrow microenvironment. In this review, the bone marrow microenvironment and the mechanisms associated to the development of the neovasculature promoted by EPCs are discussed in detail in both a non-pathological scenario and in MM. The latest developments in therapy targeting the vasculature and EPCs in MM are also highlighted. The identification and characterization of the pathways relevant to the complex setting of MM are of utter importance to identify not only biomarkers for an early diagnosis and disease progression monitoring, but also to reveal intervention targets for more effective therapy directed to cancer cells and the endothelial mediators relevant to neovasculature development.


Assuntos
Células Progenitoras Endoteliais/metabolismo , Mieloma Múltiplo/irrigação sanguínea , Neovascularização Patológica/patologia , Animais , Medula Óssea/patologia , Microambiente Celular , Humanos , Modelos Biológicos
5.
Eur J Haematol ; 98(6): 529-541, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28208215

RESUMO

Multiple myeloma (MM) is a hematological malignancy that remains incurable, with relapse rates >90%. The main limiting factor for the effective use of chemotherapies in MM is the serious side effects caused by these drugs. The emphasis in cancer treatment has shifted from cytotoxic, non-specific chemotherapies to molecularly targeted and rationally designed therapies showing greater efficacy and fewer side effects. Traditional chemotherapy has shown several disadvantages such as lack of targeting capabilities, systemic toxicity, and side effects; low therapeutic index, as well as most anticancer drugs, has poor water solubility. Nanoparticle delivery systems (NPs) are capable of targeting large doses of chemotherapies into the target area while sparing healthy tissues, overcoming the limitations of traditional chemotherapy. Here, we review the current state of the art in nanoparticle-based strategies designed to treat MM. Many nanoparticle delivery systems have been studied for myeloma using non-targeted NPs (liposomes, polymeric NPs, and inorganic NPs), triggered NPs, as well as targeted NPs (VLA-4, ABC drug transporters, bone microenvironment targeting). The results in preclinical and clinical studies are promising; however, there remains much to be learned in the emerging field of nanomedicine in myeloma.


Assuntos
Antineoplásicos/uso terapêutico , Sistemas de Liberação de Medicamentos/métodos , Terapia de Alvo Molecular , Mieloma Múltiplo/tratamento farmacológico , Nanomedicina/métodos , Nanopartículas/uso terapêutico , Ensaios Clínicos como Assunto , Humanos , Concentração de Íons de Hidrogênio , Interações Hidrofóbicas e Hidrofílicas , Luz , Campos Magnéticos , Mieloma Múltiplo/irrigação sanguínea , Mieloma Múltiplo/patologia , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/patologia , Permeabilidade , Recidiva
6.
J Pathol ; 239(2): 162-73, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26956697

RESUMO

Multiple myeloma (MM) pathogenesis and progression largely rely on the cells and extracellular factors in the bone marrow (BM) microenvironment. Compelling studies have identified tumour exosomes as key regulators in the maintenance and education of the BM microenvironment by targeting stromal cells, immune cells, and vascular cells. However, the role of MM exosomes in the modification of the BM microenvironment and MM progression remains unclear. Here, we explored the functions of MM exosomes in angiogenesis and immunosuppression in vitro and in vivo. Murine MM exosomes carrying multiple angiogenesis-related proteins enhanced angiogenesis and directly promoted endothelial cell growth. Several pathways such as signal transducer and activator of transcription 3 (STAT3), c-Jun N-terminal kinase, and p53 were modulated by the exosomes in endothelial and BM stromal cells. These exosomes promoted the growth of myeloid-derived suppressor cells (MDSCs) in naive mice through activation of the STAT3 pathway and changed their subsets to similar phenotypes to those seen in MM-bearing mice. Moreover, MM exosomes up-regulated inducible nitric oxide synthase and enhanced the immunosuppressive capacity of BM MDSCs in vivo. Our data show that MM exosomes modulate the BM microenvironment through enhancement of angiogenesis and immunosuppression, which will further facilitate MM progression. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Assuntos
Microambiente Celular , Exossomos/imunologia , Tolerância Imunológica , Mieloma Múltiplo/imunologia , Neovascularização Patológica , Animais , Medula Óssea/imunologia , Medula Óssea/patologia , Linhagem Celular Tumoral , Proliferação de Células , Modelos Animais de Doenças , Progressão da Doença , Exossomos/patologia , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Células-Tronco Mesenquimais/imunologia , Células-Tronco Mesenquimais/patologia , Camundongos , Camundongos Endogâmicos C57BL , Mieloma Múltiplo/irrigação sanguínea , Mieloma Múltiplo/patologia , Células Supressoras Mieloides/imunologia , Células Supressoras Mieloides/patologia , Óxido Nítrico Sintase Tipo II/metabolismo , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Proteína Supressora de Tumor p53/metabolismo
7.
Br J Haematol ; 174(1): 127-35, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-26991959

RESUMO

This prospective study aimed to investigate the prognostic significance of dynamic contrast enhanced magnetic resonance imaging (DCE-MRI) as a non-invasive imaging technique delivering the quantitative parameters amplitude A (reflecting blood volume) and exchange rate constant kep (reflecting vascular permeability) in patients with asymptomatic monoclonal plasma cell diseases. We analysed DCE-MRI parameters in 33 healthy controls and 148 patients with monoclonal gammopathy of undetermined significance (MGUS) or smouldering multiple myeloma (SMM) according to the 2003 IMWG guidelines. All individuals underwent standardized DCE-MRI of the lumbar spine. Regions of interest were drawn manually on T1-weighted images encompassing the bone marrow of each of the 5 lumbar vertebrae sparing the vertebral vessel. Prognostic significance for median of amplitude A (univariate: P < 0·001, hazard ratio (HR) 2·42, multivariate P = 0·02, HR 2·7) and exchange rate constant kep (univariate P = 0·03, HR 1·92, multivariate P = 0·46, HR 1·5) for time to progression of 79 patients with SMM was found. Patients with amplitude A above the optimal cut-off point of 0·89 arbitrary units had a 2-year progression rate into symptomatic disease of 80%. In conclusion, DCE-MRI parameters are of prognostic significance for time to progression in patients with SMM but not in individuals with MGUS.


Assuntos
Imageamento por Ressonância Magnética/métodos , Microcirculação , Gamopatia Monoclonal de Significância Indeterminada/diagnóstico por imagem , Mieloma Múltiplo/diagnóstico por imagem , Adulto , Idoso , Idoso de 80 Anos ou mais , Medula Óssea/irrigação sanguínea , Medula Óssea/diagnóstico por imagem , Medula Óssea/patologia , Estudos de Casos e Controles , Progressão da Doença , Feminino , Humanos , Vértebras Lombares/irrigação sanguínea , Vértebras Lombares/diagnóstico por imagem , Masculino , Pessoa de Meia-Idade , Gamopatia Monoclonal de Significância Indeterminada/patologia , Mieloma Múltiplo/irrigação sanguínea , Mieloma Múltiplo/patologia , Prognóstico , Estudos Prospectivos , Adulto Jovem
8.
Blood ; 124(25): 3748-57, 2014 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-25320245

RESUMO

Exosomes are small endosome-derived vesicles containing a wide range of functional proteins, mRNA, and miRNA. Exosomal miRNA from cancer cells helps modulate the microenvironment. In multiple myeloma (MM), the massive proliferation of malignant plasma cells causes hypoxia. To date, the majority of in vitro hypoxia studies of cancer cells have used acute hypoxic exposure (3-24 hours). Thus, we attempted to clarify the role of MM-derived exosomes in hypoxic bone marrow by using MM cells grown continuously in vitro under chronic hypoxia (hypoxia-resistant MM [HR-MM] cells). The HR-MM cells produced more exosomes than the parental cells under normoxia or acute hypoxia conditions, and miR-135b was significantly upregulated in exosomes from HR-MM cells. Exosomal miR-135b directly suppressed its target factor-inhibiting hypoxia-inducible factor 1 (FIH-1) in endothelial cells. Finally, exosomal miR-135b from HR-MM cells enhanced endothelial tube formation under hypoxia via the HIF-FIH signaling pathway. This in vitro HR myeloma cell model will be useful for investigating MM cell-endothelial cell interactions under hypoxic conditions, which may mimic the in vivo bone marrow microenvironment. Although tumor angiogenesis is regulated by various factors, exosomal miR-135b may be a target for controlling MM angiogenesis.


Assuntos
Exossomos/genética , MicroRNAs/genética , Oxigenases de Função Mista/genética , Neovascularização Patológica/genética , Proteínas Repressoras/genética , Regiões 3' não Traduzidas/genética , Vasos Sanguíneos/crescimento & desenvolvimento , Vasos Sanguíneos/metabolismo , Western Blotting , Hipóxia Celular , Linhagem Celular Tumoral , Células Cultivadas , Exossomos/ultraestrutura , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Células Endoteliais da Veia Umbilical Humana/metabolismo , Células Endoteliais da Veia Umbilical Humana/fisiologia , Humanos , Luciferases/genética , Luciferases/metabolismo , Microscopia Eletrônica de Transmissão , Oxigenases de Função Mista/metabolismo , Mieloma Múltiplo/irrigação sanguínea , Mieloma Múltiplo/genética , Mieloma Múltiplo/patologia , Análise de Sequência com Séries de Oligonucleotídeos , Proteínas Repressoras/metabolismo
9.
Int Arch Allergy Immunol ; 168(4): 263-8, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26894886

RESUMO

BACKGROUND: Osteolytic bone disease is a major hallmark in multiple myeloma (MM) progression and affects many patients. Several inflammatory cells are involved in MM progression. Among them, mast cells (MCs) accumulated in the bone marrow (BM) microenvironment are known to play an important role in the mechanism of neovascularization. METHODS: In 52 newly diagnosed active MM patients, we measured BM MC density (MCD) using an immunohistochemical stain for tryptase, serum levels of matrix metalloproteinase-9 (MMP-9) and receptor activator of nuclear factor x03BA;B ligand (RANKL) by a solid-phase sandwich enzyme-linked immunosorbent assay, along with urine levels of N-terminal propeptide of procollagen type I (Ntx) by a competitive inhibition enzyme-linked immunosorbent assay, in various clinical stages and skeletal grades. RESULTS: MCD, RANKL and Ntx were higher in MM patients. All values increased in association with both the clinical stage and skeletal grade. Furthermore, MCD correlated positively with MMP-9, RANKL and Ntx. CONCLUSIONS: Our data suggest that MCs may contribute to osteolytic processes during MM progression. Although the major role of MCs in tumor progression is to enhance angiogenesis, it seems that they may affect MM bone disease and may secrete a plethora of mediators that may directly and indirectly have an impact on osteolysis.


Assuntos
Células da Medula Óssea/patologia , Osso e Ossos/patologia , Regulação Neoplásica da Expressão Gênica , Mastócitos/patologia , Mieloma Múltiplo/patologia , Osteólise Essencial/patologia , Idoso , Idoso de 80 Anos ou mais , Células da Medula Óssea/metabolismo , Osso e Ossos/irrigação sanguínea , Osso e Ossos/metabolismo , Estudos de Casos e Controles , Contagem de Células , Microambiente Celular , Colágeno Tipo I/genética , Colágeno Tipo I/urina , Progressão da Doença , Feminino , Humanos , Masculino , Mastócitos/metabolismo , Metaloproteinase 9 da Matriz/sangue , Metaloproteinase 9 da Matriz/genética , Pessoa de Meia-Idade , Mieloma Múltiplo/irrigação sanguínea , Mieloma Múltiplo/diagnóstico , Mieloma Múltiplo/genética , Gradação de Tumores , Estadiamento de Neoplasias , Neovascularização Patológica/sangue , Neovascularização Patológica/genética , Neovascularização Patológica/patologia , Osteólise Essencial/sangue , Osteólise Essencial/diagnóstico , Osteólise Essencial/genética , Peptídeos/genética , Peptídeos/urina , Ligante RANK/sangue , Ligante RANK/genética , Triptases/sangue , Triptases/genética
11.
J Pathol ; 229(1): 87-98, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22847671

RESUMO

Pentraxin 3 (PTX3) is a soluble pattern recognition receptor that binds with high affinity and selectivity to fibroblast growth factor-2 (FGF2), thus inhibiting its pro-angiogenic activity. Here we investigated the effects of PTX3 on monoclonal gammopathy of undetermined significance (MGUS) and multiple myeloma (MM) patient-derived bone marrow (BM) plasma cells (PCs), endothelial cells (ECs), and fibroblasts (FBs), and assessed whether PTX3 can modulate the cross-talk between PCs and those microenvironment cells. PTX3 and FGF2 expression was evaluated by ELISA. Functional studies, including cell viability, wound healing, chemotaxis, and Matrigel(®) assays, were performed on MGUS and MM ECs and FBs upon the PTX3 treatment. Through western blot PTX3-induced modulation in FGF2/FGF receptor signalling pathways was evaluated in MGUS and MM ECs and FBs through western blot. Co-cultures between MM ECs/FBs and human PC lines were used to evaluate possible PTX3 indirect effects on MM PCs. Adhesion molecules were studied by flow cytometry. PTX3 provides a direct time- and dose-dependent apoptotic effect on MM ECs and FBs, but not on either MM primary PCs or human PC lines. PTX3 inhibits migration of MM ECs and FBs in a dose-dependent manner, and impacts in vitro and in vivo FGF2-mediated MM angiogenesis. Co-cultures of PCs and ECs/FBs show that PTX3 treatment indirectly impairs PC viability and adhesion. We conclude that PTX3 is an anti-angiogenic factor in MM and behaves as a cytotoxic molecule on MM cells by inhibiting the cross-talk between PCs and ECs/FBs.


Assuntos
Células da Medula Óssea/metabolismo , Proteína C-Reativa/metabolismo , Comunicação Celular , Células Endoteliais/metabolismo , Fibroblastos/metabolismo , Mieloma Múltiplo/metabolismo , Plasmócitos/metabolismo , Componente Amiloide P Sérico/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Apoptose , Western Blotting , Células da Medula Óssea/patologia , Estudos de Casos e Controles , Adesão Celular , Moléculas de Adesão Celular/metabolismo , Linhagem Celular , Microambiente Celular , Quimiotaxia , Embrião de Galinha , Técnicas de Cocultura , Meios de Cultivo Condicionados/metabolismo , Citocinas/metabolismo , Células Endoteliais/patologia , Ensaio de Imunoadsorção Enzimática , Feminino , Fator 2 de Crescimento de Fibroblastos/metabolismo , Fibroblastos/patologia , Citometria de Fluxo , Humanos , Masculino , Pessoa de Meia-Idade , Gamopatia Monoclonal de Significância Indeterminada/metabolismo , Gamopatia Monoclonal de Significância Indeterminada/patologia , Mieloma Múltiplo/irrigação sanguínea , Mieloma Múltiplo/patologia , Neovascularização Patológica , Plasmócitos/patologia , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Transdução de Sinais , Fatores de Tempo , Células Tumorais Cultivadas
12.
Adv Exp Med Biol ; 816: 361-76, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24818730

RESUMO

Both innate and adaptive immune cells are involved in the mechanisms of endothelial cell proliferation, migration and activation, via production and release of a large spectrum of pro-angiogenic mediators, thus creating the specific microenvironment that favors increased rate of tissue vascularization. In this article, we focus on the immune cell component of the angiogenic process occurring during multiple myeloma progression. We also provide information on some anti-angiogenic properties of immune cells that may be applied for a potential pharmacological use as anti-angiogenic agents in the disease treatment.


Assuntos
Inflamação/complicações , Mieloma Múltiplo/irrigação sanguínea , Mieloma Múltiplo/etiologia , Neovascularização Patológica/imunologia , Animais , Anti-Inflamatórios/uso terapêutico , Humanos , Imunomodulação , Inflamação/patologia , Inflamação/terapia , Macrófagos/fisiologia , Mastócitos/fisiologia , Monócitos/fisiologia , Mieloma Múltiplo/terapia , Neovascularização Patológica/terapia
13.
Carcinogenesis ; 34(2): 426-35, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23104180

RESUMO

Deregulated microRNAs (miRNAs) and their roles in cancer development have attracted much attention. Two miRNAs, miR-15a and miR-16, which act as putative tumor suppressor by targeting the oncogene BCL2, have been implicated in cell cycle, apoptosis and proliferation. In this study, we investigated the possible role of miR-15a/16 in the angiogenesis of multiple myeloma (MM). Using a stem-loop quantitative reverse transcription-PCR, we analyzed miR-15a/16 expressions in bone marrow samples from newly diagnosed MM patients and a panel of MM cell lines. miRNA transfection, western blotting analysis and assay of luciferase activity were used to examine whether vascular endothelial growth factor (VEGF) is the target of miR-15a/16. The functional roles of miR-15a/16 on tumorigenesis and angiogenesis were examined by in vitro angiogenesis models and in vivo tumor xenograft model. We showed that miR-15a and miR-16 were significantly underexpressed in primary MM cells as well as in MM cell lines. The aberrant expression of miR-15a/16 was detected especially in advanced stage MM. In human MM cell lines and normal plasma cells, expression of miR-15a/16 inversely correlated with the expression of VEGF-A. Western blotting combined with the luciferase reporter assay demonstrated that VEGF-A was a direct target of miR-15a/16. Ectopic overexpression of miR-15a/16 led to decreased pro-angiogenic activity of MM cells. Finally, infection of lentivirus-miR-15a or lentivirus-miR-16 resulted in significant inhibition of tumor growth and angiogenesis in nude mice. This study suggest that miR-15a/16 could play a role in the tumorigenesis of MM at least in part by modulation of angiogenesis through targeting VEGF-A.


Assuntos
Regulação Neoplásica da Expressão Gênica , MicroRNAs/genética , Gamopatia Monoclonal de Significância Indeterminada/patologia , Mieloma Múltiplo/irrigação sanguínea , Neovascularização Patológica/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Regiões 3' não Traduzidas/genética , Animais , Apoptose , Western Blotting , Estudos de Casos e Controles , Adesão Celular , Movimento Celular , Proliferação de Células , Feminino , Humanos , Técnicas Imunoenzimáticas , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos NOD , Camundongos Nus , Camundongos SCID , Gamopatia Monoclonal de Significância Indeterminada/genética , Gamopatia Monoclonal de Significância Indeterminada/metabolismo , Mieloma Múltiplo/genética , Mieloma Múltiplo/metabolismo , Estadiamento de Neoplasias , Plasmócitos/metabolismo , Plasmócitos/patologia , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator A de Crescimento do Endotélio Vascular/genética
14.
Biochem Biophys Res Commun ; 431(4): 693-7, 2013 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-23375814

RESUMO

Vascular endothelial growth factor (VEGF) has been reported not only to induce angiogenesis within the bone marrow, but also directly stimulate the proliferation and survival of multiple myeloma cells, thus being involved in the development and progression of this second most common hematological malignancy. We, along with others, have found that pigment epithelium-derived factor (PEDF) has anti-angiogenic and anti-vasopermeability properties both in cell culture and animal models by counteracting the biological actions of VEGF. However, effects of PEDF on VEGF-exposed myeloma cells remain unknown. In this study, we examined whether and how PEDF could inhibit the VEGF-induced proliferation and survival of myeloma cells. PEDF, a glutathione peroxidase mimetic, ebselen, or an inhibitor of NADPH oxidase, diphenylene iodonium significantly inhibited the VEGF-induced reactive oxygen species (ROS) generation, increase in anti-apoptotic and growth-promoting factor, myeloid cell leukemia 1 (Mcl-1) expression, and proliferation in U266 myeloma cells. VEGF blocked apoptosis of multiple myeloma cells isolated from patients, which was prevented by PEDF. PEDF also reduced p22phox levels in VEGF-exposed U266 cells. Furthermore, overexpression of dominant-negative human Rac-1 mutant mimicked the effects of PEDF on ROS generation and Mcl-1 expression in U266 cells. Our present study suggests that PEDF could block the VEGF-induced proliferation and survival of multiple myeloma U266 cells through its anti-oxidative properties via suppression of p22phox, one of the membrane components of NADPH oxidase. Suppression of VEGF signaling by PEDF may be a novel therapeutic target for multiple myeloma.


Assuntos
Inibidores da Angiogênese/farmacologia , Antioxidantes/farmacologia , Proteínas do Olho/farmacologia , Mieloma Múltiplo/irrigação sanguínea , Fatores de Crescimento Neural/farmacologia , Serpinas/farmacologia , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Humanos , Mieloma Múltiplo/metabolismo , Proteína de Sequência 1 de Leucemia de Células Mieloides , NADPH Oxidases/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , Espécies Reativas de Oxigênio/metabolismo , Células Tumorais Cultivadas , Fator A de Crescimento do Endotélio Vascular/farmacologia , Fator A de Crescimento do Endotélio Vascular/fisiologia
15.
Blood ; 117(21): 5692-700, 2011 May 26.
Artigo em Inglês | MEDLINE | ID: mdl-21454451

RESUMO

Previous studies have established a role of vascular-disrupting agents as anti- cancer agents. Plinabulin is a novel vascular-disrupting agent that exhibits potent interruption of tumor blood flow because of the disruption of tumor vascular endothelial cells, resulting in tumor necrosis. In addition, plinabulin exerts a direct action on tumor cells, resulting in apoptosis. In the present study, we examined the anti-multiple myeloma (MM) activity of plinabulin. We show that low concentrations of plinabulin exhibit a potent antiangiogenic action on vascular endothelial cells. Importantly, plinabulin also induces apoptotic cell death in MM cell lines and tumor cells from patients with MM, associated with mitotic growth arrest. Plinabulin-induced apoptosis is mediated through activation of caspase-3, caspase-8, caspase-9, and poly(ADP-ribose) polymerase cleavage. Moreover, plinabulin triggered phosphorylation of stress response protein JNK, as a primary target, whereas blockade of JNK with a biochemical inhibitor or small interfering RNA strategy abrogated plinabulin-induced mitotic block or MM cell death. Finally, in vivo studies show that plinabulin was well tolerated and significantly inhibited tumor growth and prolonged survival in a human MM.1S plasmacytoma murine xenograft model. Our study therefore provides the rationale for clinical evaluation of plinabulin to improve patient outcome in MM.


Assuntos
Inibidores da Angiogênese/farmacologia , Apoptose/efeitos dos fármacos , Imidazóis/farmacologia , Proteína Quinase 8 Ativada por Mitógeno/metabolismo , Mieloma Múltiplo/irrigação sanguínea , Mieloma Múltiplo/tratamento farmacológico , Neovascularização Patológica/prevenção & controle , Piperazinas/farmacologia , Animais , Western Blotting , Proliferação de Células/efeitos dos fármacos , Dicetopiperazinas , Imunofluorescência , Humanos , Masculino , Camundongos , Camundongos SCID , Proteína Quinase 8 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 8 Ativada por Mitógeno/genética , RNA Interferente Pequeno/genética , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
16.
Neoplasma ; 60(1): 33-40, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23067214

RESUMO

Patients with multiple myeloma (MM) have increased bone marrow angiogenesis, but the angiogenic properties of myeloma cells and the mechanism of MM-induced angiogenesis have not been completely clarified. Notch1 signal has been identified as a critical factor in the regulation of vessel formation. However, the role of Notch1 in the angiogenesis of MM is unclear. We constitutively overexpressed active Notch1 in RPMI8226 cells to explore the effect of Notch1 signaling on cell growth and tumor angiogenesis in vivo and in vitro. We found that Notch1 overexpression promoted myeloma cells growth and increased drug resistance. Moreover, vascular endothelial growth factor (VEGF) expression was increased. Finally, our in vitro results were supported by the in vivo finding in human myeloma xenograft Nonobese diabetic/severe combined immunodeficient (NOD/SCID) models. Notch1 overexpression in MM cells resulted in up-regulation of VEGF expression, promotion of tumor growth, and increased microvessel density (MVD). Our study suggests that Notch1-induced angiogenesis is partly due to activation of VEGF pathway.


Assuntos
Proliferação de Células , Mieloma Múltiplo/patologia , Neovascularização Patológica , Receptor Notch1/metabolismo , Animais , Apoptose , Western Blotting , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Técnicas Imunoenzimáticas , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Mieloma Múltiplo/irrigação sanguínea , Mieloma Múltiplo/metabolismo , Células Tumorais Cultivadas , Fator A de Crescimento do Endotélio Vascular/metabolismo
17.
Ter Arkh ; 85(7): 98-102, 2013.
Artigo em Russo | MEDLINE | ID: mdl-24137956

RESUMO

AIM: To determine the significance of the angiogenic activity estimated from the gene expression of the vascular endothelial growth factors (VEGFs) VEGF-A, VEGF-C, and VEGF-D and their receptors VEGFR1, VEGFRls, VEGFR2, and VEGFR3 in the mononuclear cell fraction of bone marrow (BM) aspirates with tumor plasma cells predominating in different variants of the course of multiple myeloma (MM). MATERIALS AND METHODS: The gene expression of VEGF-A, VEGF-C, and VEGF-D and their receptors VEGFRI, VEGFRls, VEGFR2, and VEGFR3 was determined by reverse-transcription polymerase chain reaction (RT-PCR). RESULTS: VEGF-A, VEGF-C, VEGF-D, as well as VEGFR1, VEGFRls, VEGFR2, and VEGFR3 were expressed showing different intensities in the mononuclear cell fraction of BM aspirates with a predominance of tumor plasma cells in the patients with MM, which allowed patient groups to be identified. In the group of high gene expression of VEGFs and their receptors, the number of clusters of plasma cells and vascular endothelium in the BM aspirates and the degree of osteolysis in the skeletal bones of patients with MM were significantly higher than those in the group of low or absent gene expression. The survival in the latter group was significantly higher. CONCLUSION: The investigation could provide an estimate of angiogenic processes in MM and establish their association with clinical manifestations and cytological characteristics.


Assuntos
Expressão Gênica , Mieloma Múltiplo/genética , Neovascularização Patológica/genética , Receptores de Fatores de Crescimento do Endotélio Vascular/genética , Fatores de Crescimento do Endotélio Vascular/genética , Adulto , Idoso , Medula Óssea/metabolismo , Medula Óssea/patologia , Intervalo Livre de Doença , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Mieloma Múltiplo/irrigação sanguínea , Mieloma Múltiplo/metabolismo , Mieloma Múltiplo/patologia , Prognóstico , Reação em Cadeia da Polimerase Via Transcriptase Reversa
18.
BMC Cancer ; 12: 28, 2012 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-22260501

RESUMO

BACKGROUND: Ergosterol peroxide (EP) derived from edible mushroom has been shown to exert anti-tumor activity in several cancer cells. In the present study, anti-angiogenic activity of EP was investigated with the underlying molecular mechanisms in human multiple myeloma U266 cells. RESULTS: Despite weak cytotoxicity against U266 cells, EP suppressed phosphorylation, DNA binding activity and nuclear translocalization of signal transducer and activator of transcription 3 (STAT3) in U266 cells at nontoxic concentrations. Also, EP inhibited phosphorylation of the upstream kinases Janus kinase 2 (JAK2) and Src in a time-dependent manner. Furthermore, EP increased the expression of protein tyrosine phosphatase SHP-1 at protein and mRNA levels, and conversely silencing of the SHP-1 gene clearly blocked EP-mediated STAT3 inactivation. In addition, EP significantly decreased vascular endothelial growth factor (VEGF), one of STAT3 target genes at cellular and protein levels as well as disrupted in vitro tube formation assay. Moreover, EP significantly suppressed the growth of U266 cells inoculated in female BALB/c athymic nude mice and immunohistochemistry revealed that EP effectively reduced the expression of STAT3 and CD34 in tumor sections compared to untreated control. CONCLUSION: These findings suggest that EP can exert antitumor activity in multiple myeloma U266 cells partly with antiangiogenic activity targeting JAK2/STAT3 signaling pathway as a potent cancer preventive agent for treatment of multiple myeloma cells.


Assuntos
Antineoplásicos/farmacologia , Ergosterol/análogos & derivados , Mieloma Múltiplo/tratamento farmacológico , Proteína Tirosina Fosfatase não Receptora Tipo 6/metabolismo , Fator de Transcrição STAT3/antagonistas & inibidores , Análise de Variância , Inibidores da Angiogênese/farmacologia , Animais , Linhagem Celular Tumoral , Ergosterol/farmacologia , Feminino , Humanos , Imuno-Histoquímica , Janus Quinase 2/efeitos dos fármacos , Janus Quinase 2/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Mieloma Múltiplo/irrigação sanguínea , Mieloma Múltiplo/metabolismo , Neovascularização Patológica/prevenção & controle , Fosforilação/efeitos dos fármacos , Proteína Tirosina Fosfatase não Receptora Tipo 6/genética , RNA/efeitos dos fármacos , RNA/metabolismo , RNA Mitocondrial , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição STAT3/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
19.
Bioengineered ; 13(1): 667-683, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34852710

RESUMO

The importance of angiogenesis in multiple myeloma (MM) is unquestionable; however, to date, the success of antiangiogenic therapies has been fairly limited. Exosomal circular RNAs (circRNAs) have been proven to be pivotal players in angiogenesis in various cancers. Nevertheless, their role in MM remains unknown. Therefore, we aimed to identify differentially expressed circRNAs in peripheral blood exosomes from MM patients and explore their diagnostic and prognostic values. We screened 2,052 circRNAs with significant differential expression between MM patients and healthy controls via high-throughput sequencing. qRT-PCR confirmed that the expression of circ-ATP10A was significantly increased in MM patients. The bioinformatics analyses suggested that circ-ATP10A can act as a microRNA (miRNA) sponge and regulate the expression of downstream vascular endothelial growth factor-B (VEGFB), hypoxia-inducible factor-1alpha (HIF1A), platelet-derived growth factor subunit A (PDGFA), and fibroblast growth factor (FGF). The immunohistochemical results indicated that the circ-ATP10A level was positively correlated with the protein levels of VEGFB and marrow microvessel density (MVD) in MM patients, and the receiver operating characteristic (ROC) curve, area under the ROC curve (AUC) and Kaplan-Meier survival curve analyses confirmed it as a prognostic biomarker. Collectively, our study indicates that exosomal circ-ATP10A is a valuable prognostic biomarker in MM and may promote MM angiogenesis by targeting hsa-miR-6758-3p/hsa-miR-3977/hsa-miR-6804-3p/hsa-miR-1266-3p/hsa-miR-3620-3p and modulating their downstream mRNAs, such as VEGFB, HIF1A, PDGF, and FGF.


Assuntos
Biomarcadores Tumorais , MicroRNAs , Mieloma Múltiplo , Neovascularização Patológica , RNA Circular , RNA Mensageiro , RNA Neoplásico , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Feminino , Humanos , Masculino , MicroRNAs/genética , MicroRNAs/metabolismo , Mieloma Múltiplo/irrigação sanguínea , Mieloma Múltiplo/genética , Mieloma Múltiplo/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo , RNA Circular/genética , RNA Circular/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Neoplásico/genética , RNA Neoplásico/metabolismo
20.
Br J Haematol ; 155(4): 438-48, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21923653

RESUMO

Recent advances regarding the introduction of anti-adhesion strategies as a novel therapeutic concept in oncology hold great promise. Here we evaluated the therapeutic potential of the new-in-class-molecule selective-adhesion-molecule (SAM) inhibitor Natalizumab, a recombinant humanized IgG4 monoclonal antibody, which binds integrin-α4, in multiple myeloma (MM). Natalizumab, but not a control antibody, inhibited adhesion of MM cells to non-cellular and cellular components of the microenvironment as well as disrupted the binding of already adherent MM cells. Consequently, Natalizumab blocked both the proliferative effect of MM-bone marrow (BM) stromal cell interaction on tumour cells, and vascular endothelial growth factor (VEGF)-induced angiogenesis in the BM milieu. Moreover, Natalizumab also blocked VEGF- and insulin-like growth factor 1 (IGF-1)-induced signalling sequelae triggering MM cell migration. In agreement with our in vitro results, Natalizumab inhibited tumour growth, VEGF secretion, and angiogenesis in a human severe combined immunodeficiency murine model of human MM in the human BM microenvironment. Importantly, Natalizumab not only blocked tumour cell adhesion, but also chemosensitized MM cells to bortezomib, in an in vitro therapeutically representative human MM-stroma cell co-culture system model. Our data therefore provide the rationale for the clinical evaluation of Natalizumab, preferably in combination with novel agents (e.g. bortezomib) to enhance MM cytotoxicity and improve patient outcome.


Assuntos
Anticorpos Monoclonais Humanizados/farmacologia , Células da Medula Óssea/patologia , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/patologia , Animais , Células da Medula Óssea/efeitos dos fármacos , Adesão Celular/efeitos dos fármacos , Processos de Crescimento Celular/efeitos dos fármacos , Processos de Crescimento Celular/fisiologia , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Modelos Animais de Doenças , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/patologia , Fibronectinas/metabolismo , Humanos , Imuno-Histoquímica , Integrina alfa4/biossíntese , Masculino , Camundongos , Camundongos SCID , Mieloma Múltiplo/irrigação sanguínea , Mieloma Múltiplo/metabolismo , Natalizumab , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/patologia , Transdução de Sinais/efeitos dos fármacos , Microambiente Tumoral , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Fator A de Crescimento do Endotélio Vascular/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA