Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Stem Cells ; 37(8): 1083-1094, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30977188

RESUMO

The general population is chronically exposed to multiple environmental contaminants such as pesticides. We have previously demonstrated that human mesenchymal stem cells (MSCs) exposed in vitro to low doses of a mixture of seven common pesticides showed a permanent phenotype modification with a specific induction of an oxidative stress-related senescence. Pesticide mixture also induced a shift in MSC differentiation toward adipogenesis. Thus, we hypothesized that common combination of pesticides may induce a premature cellular aging of adult MSCs. Our goal was to evaluate if the prolonged exposure to pesticide mixture could accelerate aging-related markers and in particular deteriorate the immunosuppressive properties of MSCs. MSCs exposed to pesticide mixture, under long-term culture and obtained from aging donor, were compared by bulk RNA sequencing analysis. Aging, senescence, and immunomodulatory markers were compared. The protein expression of cellular aging-associated metabolic markers and immune function of MSCs were analyzed. Functional analysis of the secretome impacts on immunomodulatory properties of MSCs was realized after 21 days' exposure to pesticide mixture. The RNA sequencing analysis of MSCs exposed to pesticide showed some similarities with cells from prolonged culture, but also with the MSCs of an aged donor. Changes in the metabolic markers MDH1, GOT and SIRT3, as well as an alteration in the modulation of active T cells and modifications in cytokine production are all associated with cellular aging. A modified functional profile was found with similarities to aging process. Stem Cells 2019;37:1083-1094.


Assuntos
Envelhecimento , Antígenos de Diferenciação/metabolismo , Senescência Celular/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Praguicidas/efeitos adversos , Adulto , Idoso , Feminino , Humanos , Masculino , Células-Tronco Mesenquimais/patologia , Praguicidas/farmacologia
2.
Int J Mol Sci ; 20(22)2019 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-31717935

RESUMO

BACKGROUND: Considering the positive or negative potential effects of adipocytes, depending on their lipid composition, on breast tumor progression, it is important to evaluate whether adipose tissue (AT) harvesting procedures, including epinephrine infiltration, may influence breast cancer progression. METHODS: Culture medium conditioned with epinephrine-infiltrated adipose tissue was tested on human Michigan Cancer Foundation-7 (MCF7) breast cancer cells, cultured in monolayer or in oncospheres. Lipid composition was evaluated depending on epinephrine-infiltration for five patients. Epinephrine-infiltrated adipose tissue (EI-AT) or corresponding conditioned medium (EI-CM) were injected into orthotopic breast carcinoma induced in athymic mouse. RESULTS: EI-CM significantly increased the proliferation rate of MCF7 cells Moreover EI-CM induced an output of the quiescent state of MCF7 cells, but it could be either an activator or inhibitor of the epithelial mesenchymal transition as indicated by gene expression changes. EI-CM presented a significantly higher lipid total weight compared with the conditioned medium obtained from non-infiltrated-AT of paired-patients. In vivo, neither the EI-CM or EI-AT injection significantly promoted MCF7-induced tumor growth. CONCLUSIONS: Even though conditioned media are widely used to mimic the secretome of cells or tissues, they may produce different effects on tumor progression, which may explain some of the discrepancy observed between in vitro, preclinical and clinical data using AT samples.


Assuntos
Tecido Adiposo/metabolismo , Proliferação de Células/efeitos dos fármacos , Epinefrina/farmacologia , Metabolismo dos Lipídeos , Neoplasias Mamárias Experimentais/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto/métodos , Animais , Meios de Cultivo Condicionados/farmacologia , Feminino , Humanos , Células MCF-7 , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Nus , Ensaios Antitumorais Modelo de Xenoenxerto/normas
3.
Blood ; 128(24): 2808-2818, 2016 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-27697772

RESUMO

Mantle cell lymphoma (MCL) accumulates in lymphoid organs, but disseminates early on in extranodal tissues. Although proliferation remains located in lymphoid organs only, suggesting a major role of the tumor ecosystem, few studies have assessed MCL microenvironment. We therefore cocultured primary circulating MCL cells from 21 patients several weeks ex vivo with stromal or lymphoid-like (CD40L) cells to determine which interactions could support their proliferation. We showed that coculture with lymphoid-like cells, but not stromal cells, induced cell-cycle progression, which was amplified by MCL-specific cytokines (insulin-like growth factor-1, B-cell activating factor, interleukin-6, interleukin-10). Of interest, we showed that our model recapitulated the MCL in situ molecular signatures (ie, proliferation, NF-κB, and survival signatures). We further demonstrated that proliferating MCL harbored an imbalance in Bcl-2 family expression, leading to a consequent loss of mitochondrial priming. Of interest, this loss of priming was overcome by the type II anti-CD20 antibody obinutuzumab, which counteracted Bcl-xL induction through NF-κB inhibition. Finally, we showed that the mitochondrial priming directly correlated with the sensitivity toward venetoclax and alkylating drugs. By identifying the microenvironment as the major support for proliferation and drug resistance in MCL, our results highlight a selective approach to target the lymphoma niche.


Assuntos
Linfoma de Célula do Manto/patologia , Linfoma de Célula do Manto/terapia , Terapia de Alvo Molecular , Microambiente Tumoral , Idoso , Idoso de 80 Anos ou mais , Anticorpos Monoclonais Humanizados/farmacologia , Antígenos CD20/imunologia , Ligante de CD40/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Técnicas de Cocultura , Regulação para Baixo/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Feminino , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Tecido Linfoide/patologia , Masculino , Mesoderma/patologia , Pessoa de Meia-Idade , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , NF-kappa B/metabolismo , Microambiente Tumoral/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos , Proteína bcl-X/metabolismo
4.
Stem Cells ; 35(3): 800-811, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27860054

RESUMO

Humans are chronically exposed to multiple environmental pollutants such as pesticides with no significant evidence about the safety of such poly-exposures. We exposed mesenchymal stem cells (MSC) to very low doses of mixture of seven pesticides frequently detected in food samples for 21 days in vitro. We observed a permanent phenotype modification with a specific induction of an oxidative stress-related senescence. Pesticide mixture also induced a shift in MSC differentiation towards adipogenesis but did not initiate a tumorigenic transformation. In modified MSC in which a premalignant phenotype was induced, the exposure to pesticide mixture promoted tumorigenic phenotype both in vitro and in vivo after cell implantation, in all nude mice. Our results suggest that a common combination of pesticides can induce a premature ageing of adult MSC, and as such could accelerate age-related diseases. Exposure to pesticide mixture may also promote the tumorigenic transformation in a predisposed stromal environment. Abstract Video Link: https://youtu.be/mfSVPTol-Gk Stem Cells 2017;35:800-811.


Assuntos
Carcinogênese/patologia , Células-Tronco Mesenquimais/patologia , Praguicidas/toxicidade , Lesões Pré-Cancerosas/patologia , Adipogenia/efeitos dos fármacos , Animais , Carcinogênese/efeitos dos fármacos , Carcinogênese/metabolismo , Diferenciação Celular/efeitos dos fármacos , Respiração Celular , Senescência Celular , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Feminino , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fibroblastos/patologia , Humanos , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Camundongos Nus , Fenótipo , Lesões Pré-Cancerosas/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Estresse Fisiológico , Proteína Supressora de Tumor p53/metabolismo
5.
Calcif Tissue Int ; 103(6): 653-662, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30076439

RESUMO

Osteogenesis imperfecta (OI) is a rare heritable skeletal dysplasia mainly caused by type I collagen abnormalities and characterized by bone fragility and susceptibility to fracture. Over 85% of the patients carry dominant mutations in the genes encoding for the collagen type I α1 and α2 chains. Failure of bone union and/or presence of hyperplastic callus formation after fracture were described in OI patients. Here we used the Col1a2+/G610C mouse, carrying in heterozygosis the α2(I)-G610C substitution, to investigate the healing process of an OI bone. Tibiae of 2-month-old Col1a2+/G610C and wild-type littermates were fractured and the healing process was followed at 2, 3, and 5 weeks after injury from fibrous cartilaginous tissue formation to its bone replacement by radiography, micro-computed tomography (µCT), histological and biochemical approaches. In presence of similar fracture types, in Col1a2+/G610C mice an impairment in the early phase of bone repair was detected compared to wild-type littermates. Smaller callus area, callus bone surface, and bone volume associated to higher percentage of cartilage and lower percentage of bone were evident in Col1a2+/G610C at 2 weeks post fracture (wpf) and no change by 3 wpf. Furthermore, the biochemical analysis of collagen extracted from callus 2 wpf revealed in mutants an increased amount of type II collagen, typical of cartilage, with respect to type I, characteristic of bone. This is the first report of a delay in OI bone fracture repair at the modeling phase.


Assuntos
Colágeno Tipo I/genética , Consolidação da Fratura/genética , Osteogênese Imperfeita/genética , Osteogênese Imperfeita/patologia , Animais , Modelos Animais de Doenças , Camundongos , Mutação
6.
Int J Mol Sci ; 19(3)2018 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-29494553

RESUMO

Osteosarcoma (OS) is suspected to originate from dysfunctional mesenchymal stromal/stem cells (MSC). We sought to identify OS-derived cells (OSDC) with potential cancer stem cell (CSC) properties by comparing OSDC to MSC derived from bone marrow of patients. This study included in vitro characterization with sphere forming assays, differentiation assays, cytogenetic analysis, and in vivo investigations of their tumorigenicity and tumor supportive capacities. Primary cell lines were isolated from nine high-grade OS samples. All primary cell lines demonstrated stromal cell characteristics. Compared to MSC, OSDC presented a higher ability to form sphere clones, indicating a potential CSC phenotype, and were more efficient at differentiation towards osteoblasts. None of the OSDC displayed the complex chromosome rearrangements typical of high grade OS and none of them induced tumors in immunodeficient mice. However, two OSDC demonstrated focused genomic abnormalities. Three out of seven, and six out of seven OSDC showed a supportive role on local tumor development, and on metastatic progression to the lungs, respectively, when co-injected with OS cells in nude mice. The observation of OS-associated stromal cells with rare genetic abnormalities and with the capacity to sustain tumor progression may have implications for future tumor treatments.


Assuntos
Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/patologia , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Neoplásicas/metabolismo , Osteossarcoma/metabolismo , Osteossarcoma/patologia , Microambiente Tumoral , Adolescente , Adulto , Biomarcadores , Medula Óssea/patologia , Linhagem Celular Tumoral , Células Cultivadas , Técnicas de Cocultura , Feminino , Humanos , Imunofenotipagem , Cariótipo , Masculino , Células-Tronco Mesenquimais/patologia , Gradação de Tumores , Metástase Neoplásica , Estadiamento de Neoplasias , Células-Tronco Neoplásicas/patologia , Adulto Jovem
7.
Ann Rheum Dis ; 76(7): 1304-1312, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28288964

RESUMO

OBJECTIVES: Interleukin (IL)-38 is a newly characterised cytokine that belongs to the IL-1 family. This cytokine is expressed in the rheumatoid arthritis (RA) synovial tissue and IL-38 deficient mice have exacerbated arthritis. Here, we analysed the effect of IL-38 overexpression in the joints of arthritic mice, in human macrophages and synovial fibroblasts in vitro. METHODS: Articular injections of an adeno-associated virus (AAV) 2/8 encoding IL-38 were performed in collagen-induced arthritis (CIA), K/BxN serum transfer-induced arthritis (STIA) and antigen-induced arthritis (AIA) in mice. The effect of IL-38 overexpression was evaluated through clinical scores, immunohistochemistry, microCT, Luminex and RT-qPCR analysis. THP-1 macrophages were transduced with a lentiviral vector to overexpress IL-38. RESULTS: Clinical inflammatory scores were significantly decreased after AAV IL-38 injection in joints of mice with CIA and STIA, but not AIA. This decrease was accompanied by reduced macrophage infiltration and a decreased expression of Th17 cytokines (IL-17, IL-23, IL-22) and TNFα. However, IL-38 overexpression had no effect on cartilage or bone destruction. In vitro, the THP-1 monocytic cell line expressed less IL-6, TNFα and IL-23 after IL-38 overexpression. Conditioned media from these cells, containing released IL-38, also exert an anti-inflammatory effect on human primary macrophages and synovial fibroblasts from patients with RA. CONCLUSIONS: This study shows for the first time that IL-38 overexpression attenuates the severity of experimental arthritis. IL-38 may exert its anti-inflammatory effects by decreasing the production of proinflammatory cytokines by macrophages and synovial fibroblasts. This effect can lead to the development of novel treatment strategies in arthritis.


Assuntos
Artrite Experimental/imunologia , Artrite Reumatoide/imunologia , Fibroblastos/imunologia , Interleucinas/imunologia , Macrófagos/imunologia , Animais , Artrite Experimental/genética , Western Blotting , Osso e Ossos/diagnóstico por imagem , Cartilagem Articular/diagnóstico por imagem , Linhagem Celular , Meios de Cultivo Condicionados , Ensaio de Imunoadsorção Enzimática , Humanos , Imuno-Histoquímica , Técnicas In Vitro , Interleucina-17/genética , Interleucina-17/imunologia , Interleucina-23/genética , Interleucina-23/imunologia , Interleucina-6/genética , Interleucina-6/imunologia , Interleucinas/genética , Camundongos , Reação em Cadeia da Polimerase em Tempo Real , Membrana Sinovial/citologia , Células Th17/imunologia , Transcriptoma , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/imunologia , Microtomografia por Raio-X , Interleucina 22
8.
Clin Sci (Lond) ; 127(5): 277-93, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24827940

RESUMO

Aseptic loosening as a result of wear debris is considered to be the main cause of long-term implant failure in orthopaedic surgery and improved biomaterials for bearing surfaces decreases significantly the release of micrometric wear particles. Increasingly, in-depth knowledge of osteoimmunology highlights the role of nanoparticles and ions released from some of these new bearing couples, opening up a new era in the comprehension of aseptic loosening. Mouse models have been essential in the progress made in the early comprehension of pathophysiology and in testing new therapeutic agents for particle-induced osteolysis. However, despite this encouraging progress, there is still no valid clinical alternative to revision surgery. The present review provides an update of the most commonly used bearing couples, the current concepts regarding particle-cell interactions and the approaches used to study the biology of periprosthetic osteolysis. It also discusses the contribution and future challenges of mouse models for successful translation of the preclinical progress into clinical applications.


Assuntos
Artroplastia de Substituição/efeitos adversos , Macrófagos/fisiologia , Osteólise/etiologia , Animais , Fenômenos Biomecânicos , Cerâmica/efeitos adversos , Ensaios Clínicos como Assunto , Modelos Animais de Doenças , Falha de Equipamento , Humanos , Inflamação/fisiopatologia , Camundongos , Nanopartículas/efeitos adversos , Osteólise/fisiopatologia , Tamanho da Partícula , Polietilenos/efeitos adversos , Polietilenos/química , Polimetil Metacrilato/efeitos adversos , Reoperação , Pesquisa Translacional Biomédica
9.
Sci Rep ; 12(1): 1488, 2022 01 27.
Artigo em Inglês | MEDLINE | ID: mdl-35087119

RESUMO

Gastric cancer (GC) is the third cause of cancer-related mortality worldwide and is often diagnosed at advanced stages of the disease. This makes the development of more comprehensive models and efficient treatments crucial. One option is based on repurposing already marketed drugs as adjuvants to chemotherapy. Accordingly, we have previously developed the combination of docetaxel and the cholesterol-lowering drug, lovastatin, as a powerful trigger of HGT-1 human GC cells' apoptosis using 2D cultures. Because 3D models, known as spheroids, are getting recognized as possibly better suited than 2Ds in toxicological research, we aimed to investigate the efficacy of this drug combination with such a model. We established monocellular spheroids from two human (GC) cell lines, HGT-1 and AGS, and bicellular spheroids from these cells mixed with cancer-associated fibroblasts. With these, we surveyed drug-induced cytotoxicity with MTT assays. In addition, we used the Incucyte live imaging and analysis system to follow spheroid growth and apoptosis. Taken together, our results showed that the lovastatin + docetaxel combination was an efficient strategy to eliminate GC cells grown in 2D or 3D cultures, lending further support in favor of repurposing lovastatin as an adjuvant to taxane-based anticancer treatment.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Microscopia Intravital/métodos , Esferoides Celulares/efeitos dos fármacos , Neoplasias Gástricas/tratamento farmacológico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Apoptose/efeitos dos fármacos , Fibroblastos Associados a Câncer , Técnicas de Cultura de Células em Três Dimensões , Linhagem Celular Tumoral , Docetaxel/farmacologia , Docetaxel/uso terapêutico , Reposicionamento de Medicamentos , Ensaios de Seleção de Medicamentos Antitumorais/métodos , Humanos , Lovastatina/farmacologia , Lovastatina/uso terapêutico , Neoplasias Gástricas/patologia
10.
Biomater Sci ; 10(2): 485-498, 2022 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-34904143

RESUMO

Bone marrow is a complex and dynamic microenvironment that provides essential cues to resident cells. We developed a standardized three-dimensional (3D) model to decipher mechanisms that control human cells during hematological and non-hematological processes. Our simple 3D-model is constituted of a biphasic calcium phosphate-based scaffold and human cell lines to ensure a high reproducibility. We obtained a minimal well-organized bone marrow-like structure in which various cell types and secreted extracellular matrix can be observed and characterized by in situ imaging or following viable cell retrieval. The complexity of the system can be increased and customized, with each cellular component being independently modulated according to the issue investigated. Introduction of pathological elements in this 3D-system accurately reproduced changes observed in patient bone marrow. Hence, we have developed a handy and flexible standardized microphysiological system that mimics human bone marrow, allowing histological analysis and functional assays on collected cells.


Assuntos
Medula Óssea , Osso e Ossos , Células da Medula Óssea , Matriz Extracelular , Humanos , Reprodutibilidade dos Testes
11.
Eur Cell Mater ; 22: 84-96; discussion 96, 2011 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-21870339

RESUMO

Titanium implants are widely used in orthopaedic and dental surgery. Surface properties play a major role in cell and tissue interactions. The adhesion and differentiation of mesenchymal stem cells were studied as a function of nanostructures. Titanium surfaces with nanopores 30, 150 and 300 nm in diameter were prepared by physical vapour deposition. PCR arrays indicated that the expression of integrins was modulated by the nanopore size. Human Mesenchymal Stem Cells (hMSCs) exhibited more branched cell morphology on Ti30 than on other surfaces. Ti30 and Ti150 induced osteoblastic differentiation while Ti300 had a limited effect. Overall, nanopores of 30 nm may promote early osteoblastic differentiation and, consequently, rapid osseointegration of titanium implants.


Assuntos
Células-Tronco Mesenquimais/citologia , Osteogênese/efeitos dos fármacos , Próteses e Implantes , Titânio/farmacologia , Adesão Celular/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Forma Celular/efeitos dos fármacos , Humanos , Integrinas/análise , Integrinas/genética , Células-Tronco Mesenquimais/efeitos dos fármacos , Nanoporos , Osteoblastos/citologia , Tamanho da Partícula , Propriedades de Superfície
12.
Sci Rep ; 11(1): 12144, 2021 06 09.
Artigo em Inglês | MEDLINE | ID: mdl-34108508

RESUMO

In bone regeneration induced by the combination of mesenchymal stromal cells (MSCs) and calcium-phosphate (CaP) materials, osteoclasts emerge as a pivotal cell linking inflammation and bone formation. Favorable outcomes are observed despite short-term engraftments of implanted MSCs, highlighting their major paracrine function and the possible implication of cell death in modulating their secretions. In this work, we focused on the communication from MSCs towards osteoclasts-like cells in vitro. MSCs seeded on a CaP biomaterial or undergoing induced apoptosis produced a conditioned media favoring the development of osteoclasts from human CD14+ monocytes. On the contrary, MSCs' apoptotic secretion inhibited the development of inflammatory multinucleated giant cells formed after IL-4 stimulation. Components of MSCs' secretome before and after apoptotic stress were compared using mass spectrometry-based quantitative proteomics and a complementary immunoassay for major cytokines. CXCR-1 and CXCR-2 ligands, primarily IL-8/CXCL-8 but also the growth-regulated proteins CXCL-1, -2 or -3, were suggested as the major players of MSCs' pro-osteoclastic effect. These findings support the hypothesis that osteoclasts are key players in bone regeneration and suggest that apoptosis plays an important role in MSCs' effectiveness.


Assuntos
Apoptose , Células da Medula Óssea/citologia , Diferenciação Celular , Células Gigantes/patologia , Células-Tronco Mesenquimais/citologia , Osteoclastos/citologia , Osteogênese , Células da Medula Óssea/fisiologia , Proliferação de Células , Citocinas , Células Gigantes/metabolismo , Humanos , Células-Tronco Mesenquimais/fisiologia , Osteoclastos/fisiologia
13.
Front Immunol ; 12: 766275, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34858421

RESUMO

Knowledge about the hematopoietic niche has evolved considerably in recent years, in particular through in vitro analyzes, mouse models and the use of xenografts. Its complexity in the human bone marrow, in particular in a context of hematological malignancy, is more difficult to decipher by these strategies and could benefit from the knowledge acquired on the niches of solid tumors. Indeed, some common features can be suspected, since the bone marrow is a frequent site of solid tumor metastases. Recent research on solid tumors has provided very interesting information on the interactions between tumoral cells and their microenvironment, composed notably of mesenchymal, endothelial and immune cells. This review thus focuses on recent discoveries on tumor niches that could help in understanding hematopoietic niches, with special attention to 4 particular points: i) the heterogeneity of carcinoma/cancer-associated fibroblasts (CAFs) and mesenchymal stem/stromal cells (MSCs), ii) niche cytokines and chemokines, iii) the energy/oxidative metabolism and communication, especially mitochondrial transfer, and iv) the vascular niche through angiogenesis and endothelial plasticity. This review highlights actors and/or pathways of the microenvironment broadly involved in cancer processes. This opens avenues for innovative therapeutic opportunities targeting not only cancer stem cells but also their regulatory tumor niche(s), in order to improve current antitumor therapies.


Assuntos
Microambiente Tumoral , Animais , Fibroblastos Associados a Câncer , Comunicação Celular , Citocinas , Endotélio Vascular , Humanos , Células-Tronco Mesenquimais , Neoplasias , Neovascularização Patológica
14.
Haematologica ; 95(11): 1973-6, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20634494

RESUMO

Deletion of the 17p13 chromosomal region [del(17p)] is associated with a poor outcome in multiple myeloma. Most of the studies have targeted the TP53 gene for deletion analyses, although no study showed that this gene is the deletion target. In order to address this issue, we sequenced the TP53 gene in 92 patients with multiple myeloma at diagnosis, 54 with a del(17p) and 38 lacking del(17p). At least one mutation was found in 20 patients, all of them presenting a del(17p). The analysis of the mutation location showed that virtually all of them occurred in highly conserved domains involved in the DNA-protein interactions. In conclusion, we showed that 37% of the myeloma patients with del(17p) present a TP53 mutation versus 0% in patients lacking the del(17p). The prognostic significance of these mutations remains to be evaluated.


Assuntos
Deleção de Genes , Mieloma Múltiplo/genética , Mieloma Múltiplo/mortalidade , Proteína Supressora de Tumor p53/genética , Deleção Cromossômica , Cromossomos Humanos Par 17 , Intervalo Livre de Doença , Feminino , Humanos , Masculino , Fatores de Risco , Síndrome de Smith-Magenis , Taxa de Sobrevida
15.
Cells ; 9(4)2020 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-32326444

RESUMO

Osteosarcomas are the most frequent primary bone sarcomas, affecting mainly children, adolescents, and young adults, and with a second peak of incidence in elderly individuals. The current therapeutic management, a combined regimen of poly-chemotherapy and surgery, still remains largely insufficient, as patient survival has not improved in recent decades. Osteosarcomas are very heterogeneous tumors, both at the intra- and inter-tumor level, with no identified driver mutation. Consequently, efforts to improve treatments using targeted therapies have faced this lack of specific osteosarcoma targets. Nevertheless, these tumors are inextricably linked to their local microenvironment, composed of bone, stromal, vascular and immune cells and the osteosarcoma microenvironment is now considered to be essential and supportive for growth and dissemination. This review describes the different actors of the osteosarcoma microenvironment and gives an overview of the past, current, and future strategies of therapy targeting this complex ecosystem, with a focus on the role of extracellular vesicles and on the emergence of multi-kinase inhibitors.


Assuntos
Terapia de Alvo Molecular , Osteossarcoma/patologia , Osteossarcoma/terapia , Microambiente Tumoral , Animais , Remodelação Óssea , Humanos , Sistema Imunitário/patologia , Células-Tronco Mesenquimais/patologia , Osteossarcoma/imunologia , Osteossarcoma/fisiopatologia
16.
Matrix Biol Plus ; 6-7: 100028, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-33543025

RESUMO

Classical osteogenesis imperfecta (OI) is an inherited rare brittle bone disease caused by dominant mutations in the COL1A1 or COL1A2 genes, encoding for the α chains of collagen type I. The definitive cure for the disease will require a gene therapy approach, aimed to correct or suppress the mutant allele. Interestingly, individuals lacking α2(I) chain and synthetizing collagen α1(I)3 homotrimers do not show bone phenotype, making appealing a bone specific COL1A2 silencing approach for OI therapy. To this aim, three different Col1a2-silencing RNAs (siRNAs), -3554, -3825 and -4125, selected at the 3'-end of the murine Col1a2 transcript were tested in vitro and in vivo. In murine embryonic fibroblasts Col1a2-siRNA-3554 was able to efficiently and specifically target the Col1a2 mRNA and to strongly reduce α2(I) chain expression. Its efficiency and specificity were also demonstrated in primary murine osteoblasts, whose mineralization was preserved. The efficiency of Col1a2-siRNA-3554 was proved also in vivo. Biphasic calcium phosphate implants loaded with murine mesenchymal stem cells were intramuscularly transplanted in nude mice and injected with Col1a2-siRNA-3554 three times a week for three weeks. Collagen α2 silencing was demonstrated both at mRNA and protein level and Masson's Trichrome staining confirmed the presence of newly formed collagen matrix. Our data pave the way for further investigation of Col1a2 silencing and siRNA delivery to the bone tissue as a possible strategy for OI therapy.

17.
Cancer Res ; 67(11): 5418-24, 2007 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-17545623

RESUMO

Targeting the ubiquitin-proteasome pathway has emerged as a potent anticancer strategy. Bortezomib, a specific proteasome inhibitor, has been approved for the treatment of relapsed or refractory multiple myeloma. Multiple myeloma cell survival is highly dependent on Mcl-1 antiapoptotic molecules. In a recent study, proteasome inhibitors induced Mcl-1 accumulation that slowed down their proapoptotic effects. Consequently, we investigated the role of Bcl-2 family members in bortezomib-induced apoptosis. We found that bortezomib induced apoptosis in five of seven human myeloma cell lines (HMCL). Bortezomib-induced apoptosis was associated with Mcl-1 cleavage regardless of Mcl-1L accumulation. Furthermore, RNA interference mediated Mcl-1 decrease and sensitized RPMI-8226 HMCL to bortezomib, highlighting the contribution of Mcl-1 in bortezomib-induced apoptosis. Interestingly, an important induction of Noxa was found in all sensitive HMCL both at protein and mRNA level. Concomitant to Mcl-1 cleavage and Noxa induction, we also found caspase-3, caspase-8, and caspase-9 activation. Under bortezomib treatment, Mcl-1L/Noxa complexes were highly increased, Mcl-1/Bak complexes were disrupted, and there was an accumulation of free Noxa. Finally, we observed a dissociation of Mcl-1/Bim complexes that may be due to a displacement of Bim induced by Noxa. Thus, in myeloma cells, the mechanistic basis for bortezomib sensitivity can be explained mainly by the model in which the sensitizer Noxa can displace Bim, a BH3-only activator, from Mcl-1, thus leading to Bax/Bak activation.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Ácidos Borônicos/farmacologia , Mieloma Múltiplo/tratamento farmacológico , Proteínas de Neoplasias/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Pirazinas/farmacologia , Apoptose/fisiologia , Proteínas Reguladoras de Apoptose/metabolismo , Proteína 11 Semelhante a Bcl-2 , Bortezomib , Linhagem Celular Tumoral , Regulação para Baixo , Humanos , Proteínas de Membrana/metabolismo , Mieloma Múltiplo/genética , Mieloma Múltiplo/metabolismo , Mieloma Múltiplo/patologia , Proteína de Sequência 1 de Leucemia de Células Mieloides , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , Proteínas Proto-Oncogênicas c-bcl-2/genética , Interferência de RNA , RNA Interferente Pequeno/genética , Regulação para Cima/efeitos dos fármacos , Proteína Killer-Antagonista Homóloga a bcl-2/metabolismo
18.
Cancer Res ; 67(15): 7308-18, 2007 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-17671200

RESUMO

Osteosarcoma is the most frequent primary bone tumor that develops mainly in the young, the median age of diagnosis being 18 years. Despite improvement in osteosarcoma treatment, survival rate is only 30% at 5 years for patients with pulmonary metastases at diagnosis. This warrants exploration of new therapeutic options, and among them, osteoprotegerin (OPG), a naturally occurring protein that inhibits bone resorption, is very promising in blocking the vicious cycle between bone resorption and tumor proliferation that takes place during tumor development in bone site. As OPG binds and inhibits the activity of tumor necrosis factor-related apoptosis-inducing ligand, the truncated form of murine OPG 1-194 was used. The cDNA encoding OPG was administered by gene transfer using replication-defective adenoviral vector or was associated with an amphiphilic polymer in two models of rodent osteosarcoma. In both models, OPG gene transfer was effective in preventing the formation of osteolytic lesions associated with osteosarcoma development, in reducing the tumor incidence and the local tumor growth, leading to a 4-fold augmentation of mice survival 28 days postimplantation. On the contrary, OPG did not prevent the development of pulmonary metastasis alone, suggesting that bone environment is necessary for OPG therapeutic efficacy. Because OPG has no direct activity on osteosarcoma cells in vitro (cell binding, cell proliferation, apoptosis, or cell cycle distribution), we show that OPG exerts indirect inhibitory effect on tumor progression through the inhibition of RANKL whose production is enhanced in bone tumor environment, leading to osteolysis inhibition as reflected by osteoclast number decrease.


Assuntos
Reabsorção Óssea , Proliferação de Células , Terapia Genética , Vetores Genéticos/uso terapêutico , Osteólise/prevenção & controle , Osteoprotegerina/genética , Osteossarcoma/complicações , Adenoviridae/genética , Animais , Apoptose , Densidade Óssea , Neoplasias Ósseas/complicações , Caspases/metabolismo , Ciclo Celular , Ensaio de Imunoadsorção Enzimática , Vetores Genéticos/genética , Masculino , Camundongos , Camundongos Endogâmicos C3H , Mioblastos/citologia , Mioblastos/metabolismo , Osteólise/etiologia , Osteólise/patologia , Osteoprotegerina/uso terapêutico , Ligante RANK/metabolismo , Ratos , Transdução Genética , Ensaios Antitumorais Modelo de Xenoenxerto
19.
Front Immunol ; 10: 663, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31001270

RESUMO

A wide variety of biomaterials have been developed as both stabilizing structures for the injured bone and inducers of bone neoformation. They differ in chemical composition, shape, porosity, and mechanical properties. The most extensively employed and studied subset of bioceramics are calcium phosphate materials (CaPs). These materials, when transplanted alongside mesenchymal stem cells (MSCs), lead to ectopic (intramuscular and subcutaneous) and orthotopic bone formation in preclinical studies, and effective fracture healing in clinical trials. Human MSC transplantation in pre-clinical and clinical trials reveals very low engraftment in spite of successful clinical outcomes and their therapeutic actions are thought to be primarily through paracrine mechanisms. The beneficial role of transplanted MSC could rely on their strong immunomodulatory effect since, even without long-term engraftment, they have the ability to alter both the innate and adaptive immune response which is critical to facilitate new bone formation. This study presents the current knowledge of the immune response to the implantation of CaP biomaterials alone or in combination with MSC. In particular the central role of monocyte-derived cells, both macrophages and osteoclasts, in MSC-CaP mediated bone formation is emphasized. Biomaterial properties, such as macroporosity and surface microstructure, dictate the host response, and the ultimate bone healing cascade. Understanding intercellular communications throughout the inflammation, its resolution and the bone regeneration phase, is crucial to improve the current therapeutic strategies or develop new approaches.


Assuntos
Materiais Biocompatíveis/farmacologia , Regeneração Óssea , Fosfatos de Cálcio/farmacologia , Imunomodulação/efeitos dos fármacos , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/imunologia , Osteogênese , Animais , Regeneração Óssea/efeitos dos fármacos , Regeneração Óssea/imunologia , Humanos , Osteogênese/efeitos dos fármacos , Osteogênese/imunologia
20.
J Clin Med ; 8(10)2019 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-31561454

RESUMO

Calcific tendonitis is a frequent cause of chronic shoulder pain. Its cause is currently poorly known. The objectives of this study were to better characterize the cells and mechanisms involved in depositing apatite crystals in human tendons. Histologic sections of cadaveric calcified tendons were analyzed, and human calcific deposits from patients undergoing lavage of their calcification were obtained to perform infrared spectroscopy and mass spectrometry-based proteomic characterizations. In vitro, the mineralization ability of human rotator cuff cells from osteoarthritis donors was assessed by alizarin red or Von Kossa staining. Calcifications were amorphous areas surrounded by a fibrocartilaginous metaplasia containing hypertrophic chondrocyte-like cells that expressed tissue non-specific alkaline phosphatase (TNAP) and ectonucleotide pyrophosphatase/phosphodiesterase 1 (ENPP1), which are two key enzymes of the mineralization process. Calcific deposits were composed of apatite crystals associated with proteins involved in bone and cartilage development and endochondral bone growth. In vitro, tenocyte-like cells extracted from the rotator cuff were able to mineralize in osteogenic cultures, and expressed TNAP, type X COLLAGEN, and MMP13, which are hypertrophic chondrocytes markers. The use of a TNAP inhibitor significantly prevented mineral deposits. We provide evidence that tenocytes have a propensity to differentiate into hypertrophic chondrocyte-like cells to produce TNAP-dependent calcium deposits. We believe that these results may pave the way to identifying regulating factors that might represent valuable targets in calcific tendonitis.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA