Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
2.
Blood Adv ; 6(2): 672-678, 2022 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-34714910

RESUMO

Bone marrow (BM) mesenchymal stromal cells (MSCs) are abnormal in multiple myeloma (MM) and play a critical role by promoting growth, survival, and drug resistance of MM cells. We observed higher Toll-like receptor 4 (TLR4) gene expression in MM MSCs than in MSCs from healthy donors. At the clinical level, we highlighted that TLR4 expression in MM MSCs evolves in parallel with the disease stage. Thus, we reasoned that the TLR4 axis is pivotal in MM by increasing the protumor activity of MSCs. Challenging primary MSCs with TLR4 agonists increased the expression of CD54 and interleukin-6 (IL-6), 2 factors directly implicated in MM MSC-MM cell crosstalk. Then, we evaluated the therapeutic efficacy of a TLR4 antagonist combined or not with conventional treatment in vitro with MSC-MM cell coculture and in vivo with the Vk*MYC mouse model. Selective inhibition of TLR4 specifically reduced the MM MSC ability to support the growth of MM cells in an IL-6-dependent manner and delayed the development of MM in the Vk*MYC mouse model by altering the early disease phase in vivo. For the first time, we demonstrate that specific targeting of the pathological BM microenvironment via TLR4 signaling could be an innovative approach to alter MM pathology development.


Assuntos
Células-Tronco Mesenquimais , Mieloma Múltiplo , Animais , Células Cultivadas , Interleucina-6 , Células-Tronco Mesenquimais/metabolismo , Camundongos , Mieloma Múltiplo/metabolismo , Receptor 4 Toll-Like/genética , Microambiente Tumoral
3.
Stem Cells Int ; 2020: 4173578, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32215016

RESUMO

Multiple myeloma (MM) is an incurable B cell neoplasia characterized by the accumulation of tumor plasma cells within the bone marrow (BM). As a consequence, bone osteolytic lesions develop in 80% of patients and remain even after complete disease remission. We and others had demonstrated that BM-derived mesenchymal stromal cells (MSCs) are abnormal in MM and thus cannot be used for autologous treatment to repair bone damage. Adipose stromal cells (ASCs) represent an interesting alternative to MSCs for cellular therapy. Thus, in this study, we wondered whether they could be a good candidate in repairing MM bone lesions. For the first time, we present a transcriptomic, phenotypic, and functional comparison of ASCs from MM patients and healthy donors (HDs) relying on their autologous MSC counterparts. In contrast to MM MSCs, MM ASCs did not exhibit major abnormalities. However, the changes observed in MM ASCs and the supportive property of ASCs on MM cells question their putative and safety uses at an autologous or allogenic level.

4.
Stem Cells ; 38(1): 146-159, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31502731

RESUMO

Clinical-grade mesenchymal stromal cells (MSCs) can be expanded from bone marrow and adipose tissue to treat inflammatory diseases and degenerative disorders. However, the influence of their tissue of origin on their functional properties, including their immunosuppressive activity, remains unsolved. In this study, we produced paired bone marrow-derived mesenchymal stromal cell (BM-MSC) and adipose-derived stromal cell (ASC) batches from 14 healthy donors. We then compared them using transcriptomic, phenotypic, and functional analyses and validated our results on purified native MSCs to infer which differences were really endowed by tissue of origin. Cultured MSCs segregated together owing to their tissue of origin based on their gene expression profile analyzed using differential expression and weighted gene coexpression network analysis. This translated into distinct immune-related gene signatures, phenotypes, and functional cell interactions. Importantly, sorted native BM-MSCs and ASCs essentially displayed the same distinctive patterns than their in vitro-expanded counterparts. As a whole, ASCs exhibited an immune profile consistent with a stronger inhibition of immune response and a lower immunogenicity, supporting the use of adipose tissue as a valuable source for clinical applications.


Assuntos
Células-Tronco Mesenquimais/metabolismo , Transcriptoma/genética , Adulto , Diferenciação Celular , Células Cultivadas , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Fenótipo , Adulto Jovem
5.
Int J Pharm ; 524(1-2): 433-442, 2017 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-28279740

RESUMO

Controlling microarchitecture in polymer scaffolds is a priority in material design for soft tissue applications. This paper reports for the first time the elaboration of alginate foam-based scaffolds for mesenchymal stem cell (MSC) delivery and a comparative study of various surfactants on the final device performance. The use of surfactants permitted to obtain highly interconnected porous scaffolds with tunable pore size on surface and in cross-section. Their mechanical properties in compression appeared to be adapted to soft tissue engineering. Scaffold structures could sustain MSC proliferation over 14 days. Paracrine activity of scaffold-seeded MSCs varied with the scaffold structure and growth factors release was globally improved in comparison with control alginate scaffolds. Our results provide evidence that exploiting different surfactant types for alginate foam preparation could be an original method to obtain biocompatible scaffolds with tunable architecture for soft tissue engineering.


Assuntos
Alginatos/química , Materiais Biocompatíveis , Células-Tronco Mesenquimais/citologia , Engenharia Tecidual , Tecidos Suporte , Ácido Glucurônico/química , Ácidos Hexurônicos/química , Humanos , Porosidade
6.
Stem Cells Transl Med ; 5(7): 847-56, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27217345

RESUMO

UNLABELLED: : Osteoarthritis (OA) is the most widespread musculoskeletal disorder in adults. It leads to cartilage damage associated with subchondral bone changes and synovial inflammation, causing pain and disability. The present study aimed at evaluating the safety of a dose-escalation protocol of intra-articular injected adipose-derived stromal cells (ASCs) in patients with knee OA, as well as clinical efficacy as secondary endpoint. A bicentric, uncontrolled, open phase I clinical trial was conducted in France and Germany with regulatory agency approval for ASC expansion procedure in both countries. From April 2012 to December 2013, 18 consecutive patients with symptomatic and severe knee OA were treated with a single intra-articular injection of autologous ASCs. The study design consisted of three consecutive cohorts (six patients each) with dose escalation: low dose (2 × 10(6) cells), medium dose (10 × 10(6)), and high dose (50 × 10(6)). The primary outcome parameter was safety evaluated by recording adverse events throughout the trial, and secondary parameters were pain and function subscales of the Western Ontario and McMaster Universities Arthritis Index. After 6 months of follow-up, the procedure was found to be safe, and no serious adverse events were reported. Four patients experienced transient knee joint pain and swelling after local injection. Interestingly, patients treated with low-dose ASCs experienced significant improvements in pain levels and function compared with baseline. Our data suggest that the intra-articular injection of ASCs is a safe therapeutic alternative to treat severe knee OA patients. A placebo-controlled double-blind phase IIb study is being initiated to assess clinical and structural efficacy. SIGNIFICANCE: Although this phase I study included a limited number of patients without a placebo arm, it showed that local injection of autologous adipose-derived stem cells was safe and well tolerated in patients with knee osteoarthritis. This study also provides encouraging preliminary evidence of efficacy. Larger and controlled long-term studies are now mandatory to confirm whether this new strategy of cell therapy can improve pain and induce structural benefit in osteoarthritis.


Assuntos
Tecido Adiposo/citologia , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/citologia , Osteoartrite do Joelho/terapia , Tecido Adiposo/transplante , Idoso , Contagem de Células , Feminino , Humanos , Injeções Intra-Articulares , Masculino , Transplante de Células-Tronco Mesenquimais/efeitos adversos , Transplante de Células-Tronco Mesenquimais/métodos , Pessoa de Meia-Idade , Osteoartrite do Joelho/patologia , Índice de Gravidade de Doença , Resultado do Tratamento
7.
Oncotarget ; 6(30): 29034-47, 2015 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-26362269

RESUMO

The aim of this work was to understand whether the nature of breast cancer cells could modify the nature of the dialog of mesenchymal stem cells (MSCs) with cancer cells. By treating MSCs with the conditioned medium of metastatic Estrogen-receptor (ER)-negative MDA-MB-231, or non-metastatic ER-positive MCF-7 breast cancer cells, we observed that a number of chemokines were produced at higher levels by MSCs treated with MDA-MB-231 conditioned medium (CM). MDA-MB-231 cells were able to induce NF-κB signaling in MSC cells. This was shown by the use of a NF-kB chemical inhibitor or an IκB dominant negative mutant, nuclear translocation of p65 and induction of NF-κB signature. Our results suggest that MDA-MB-231 cells exert their effects on MSCs through the secretion of IL-1ß, that activates MSCs and induces the same chemokines as the MDA-MB-231CM. In addition, inhibition of IL-1ß secretion in the MDA-MB-231 cells reduces the induced production of a panel of chemokines by MSCs, as well the motility of MDA-MB-231 cells. Our data suggest that aggressive breast cancer cells secrete IL-1ß, which increases the production of chemokines by MSCs.


Assuntos
Neoplasias da Mama/metabolismo , Quimiocinas/metabolismo , Interleucina-1beta/metabolismo , Células-Tronco Mesenquimais/metabolismo , Comunicação Parácrina , Microambiente Tumoral , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Movimento Celular , Quimiocinas/genética , Meios de Cultivo Condicionados/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Proteínas I-kappa B/genética , Proteínas I-kappa B/metabolismo , Interleucina-1beta/genética , Mutação , Invasividade Neoplásica , Interferência de RNA , Transdução de Sinais , Fatores de Tempo , Fator de Transcrição RelA/metabolismo , Transfecção
8.
Tissue Eng Part A ; 21(3-4): 767-81, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25322665

RESUMO

The aim of this study was to assess the immune modulatory properties of human mesenchymal stromal cells obtained from bone marrow (BM-MSCs), fat (ASCs), and cord blood (CB-MSCs) in the presence of a hydroxyapatite and tricalcium-phosphate (HA/TCP) biomaterial as a scaffold for MSC delivery. In resting conditions, a short-term culture with HA/TCP did not modulate the anti-apoptotic and suppressive features of the various MSC types toward T, B, and NK cells; in addition, when primed with inflammatory cytokines, MSCs similarly increased their suppressive capacities in the presence or absence of HA/TCP. The long-term culture of BM-MSCs with HA/TCP induced an osteoblast-like phenotype with upregulation of OSTERIX and OSTEOCALCIN, similar to what was obtained with dexamethasone and, to a higher extent, with bone morphogenetic protein 4 (BMP-4) treatment. MSC-derived osteoblasts did not trigger immune cell activation, but were less efficient than undifferentiated MSCs in inhibiting stimulated T and NK cells. Interestingly, their suppressive machinery included not only the activation of indoleamine-2,3 dioxygenase (IDO), which plays a central role in T-cell inhibition, but also cyclooxygenase-2 (COX-2) that was not significantly involved in the immune modulatory effect of human undifferentiated MSCs. Since COX-2 is significantly involved in bone healing, its induction by HA/TCP could also contribute to the therapeutic activity of MSCs for bone tissue engineering.


Assuntos
Fosfatos de Cálcio/farmacologia , Durapatita/farmacologia , Imunomodulação/imunologia , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/imunologia , Tecidos Suporte , Substitutos Ósseos/química , Substitutos Ósseos/farmacologia , Fosfatos de Cálcio/química , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/imunologia , Células Cultivadas , Cerâmica/química , Cerâmica/farmacologia , Durapatita/química , Desenho de Equipamento , Análise de Falha de Equipamento , Humanos , Fatores Imunológicos/química , Fatores Imunológicos/farmacologia , Imunomodulação/efeitos dos fármacos , Teste de Materiais
9.
Plast Reconstr Surg ; 133(4): 815-826, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24675187

RESUMO

BACKGROUND: Adipose tissue is widely used in plastic surgery. The main obstacle is that it can be used only immediately after liposuction, while reconstruction often requires several procedures to achieve optimal results. This study aimed to develop a cryopreservation protocol directly applicable to clinical situations, allowing repetitive procedures without multiple tissue harvests. METHODS: The authors first tested scalable bags suitable for therapeutic uses. All subsequent experiments were performed in those bags. The authors evaluated in vitro, on the basis of cell viability, cell number, phenotype, and stromal cell proliferation, the efficacy of six cryopreservation media composed of an external cryoprotectant (human albumin or hydroxylethyl starch) with or without an internal cryoprotectant (dimethyl sulfoxide). Two storage temperatures (-196°C and -80°C) were tested in vitro and in vivo (subcutaneous graft in 30 nude mice) with the selected medium. RESULTS: The combination of 5% dimethyl sulfoxide and 95% hydroxylethyl yielded in vitro results that were good and the most consistent. With this cryoprotective solution, the authors observed no significant difference in vitro for a storage period of 7 days. When the storage was extended to 1 month, the cell viability was decreased by 10 percent for both storage temperatures. The in vivo experiments assessed the superiority of cryopreservation at -80°C with less graft resorption (60 percent and 70 percent, respectively, for -80°C and -196°C) and less fibrosis. CONCLUSION: The study's protocol with a chemically defined cryoprotective solution, specific scalable bags constrained in an aluminum holder, and a storage temperature of -80°C is promising for long-term adipose tissue cryopreservation.


Assuntos
Tecido Adiposo , Criopreservação/instrumentação , Criopreservação/métodos , Procedimentos de Cirurgia Plástica , Tecido Adiposo/citologia , Tecido Adiposo/transplante , Adulto , Animais , Células Cultivadas , Crioprotetores/uso terapêutico , Desenho de Equipamento , Feminino , Xenoenxertos , Humanos , Masculino , Camundongos , Camundongos Nus , Modelos Animais , Células Estromais/citologia
10.
Cytotherapy ; 16(2): 245-57, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24438903

RESUMO

BACKGROUND AIMS: Non-revascularizable critical limb ischemia (CLI) is the most severe stage of peripheral arterial disease, with no therapeutic option. Extensive preclinical studies have demonstrated that adipose-derived stroma cell (ASC) transplantation strongly improves revascularization and tissue perfusion in ischemic limbs. This study, named ACellDREAM, is the first phase I trial to evaluate the feasibility and safety of intramuscular injections of autologous ASC in non-revascularizable CLI patients. METHODS: Seven patients were consecutively enrolled, on the basis of the following criteria: (i) lower-limb rest pain or ulcer; (ii) ankle systolic oxygen pressure <50 or 70 mm Hg for non-diabetic and diabetic patients, respectively, or first-toe systolic oxygen pressure <30 mm Hg or 50 mm Hg for non-diabetic and diabetic patients, respectively; (iii) not suitable for revascularization. ASCs from abdominal fat were grown for 2 weeks and were then characterized. RESULTS: More than 200 million cells were obtained, with almost total homogeneity and no karyotype abnormality. The expressions of stemness markers Oct4 and Nanog were very low, whereas expression of telomerase was undetectable in human ASCs compared with human embryonic stem cells. ASCs (10(8)) were then intramuscularly injected into the ischemic leg of patients, with no complication, as judged by an independent committee. Trans-cutaneous oxygen pressure tended to increase in most patients. Ulcer evolution and wound healing showed improvement. CONCLUSIONS: These data demonstrate the feasibility and safety of autologous ASC transplantation in patients with objectively proven CLI not suitable for revascularization. The improved wound healing also supports a putative functional efficiency.


Assuntos
Tecido Adiposo/citologia , Células-Tronco Adultas/metabolismo , Extremidades/patologia , Isquemia/terapia , Doença Arterial Periférica/terapia , Transplante de Células-Tronco , Células Estromais/metabolismo , Adulto , Células-Tronco Adultas/citologia , Células-Tronco Adultas/transplante , Idoso , Idoso de 80 Anos ou mais , Técnicas de Cultura de Células , Células Cultivadas , Extremidades/irrigação sanguínea , Extremidades/transplante , Estudos de Viabilidade , Feminino , Proteínas de Homeodomínio/metabolismo , Humanos , Injeções Intramusculares , Masculino , Pessoa de Meia-Idade , Proteína Homeobox Nanog , Neovascularização Fisiológica , Fator 3 de Transcrição de Octâmero/metabolismo , Células Estromais/citologia , Células Estromais/transplante , Resultado do Tratamento
11.
Tissue Eng Part C Methods ; 20(3): 239-51, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23845029

RESUMO

Successful preliminary studies have encouraged a more translational phase for stem cell research. Nevertheless, advances in the culture of human bone marrow-derived mesenchymal stromal/stem cells (hBM-MSC) and osteoconductive qualities of combined biomaterials can be undermined if necessary cell transportation procedures prove unviable. We aimed at evaluating the effect of transportation conditions on cell function, including the ability to form bone in vivo, using procedures suited to clinical application. hBM-MSC expanded in current Good Manufacturing Practice (cGMP) facilities (cGMP-hBM-MSC) to numbers suitable for therapy were transported overnight within syringes and subsequently tested for viability. Scaled-down experiments mimicking shipment for 18 h at 4°C tested the influence of three different clinical-grade transportation buffers (0.9% saline alone or with 4% human serum albumin [HSA] from two independent sources) compared with cell maintenance medium. Cell viability after shipment was >80% in all cases, enabling evaluation of (1) adhesion to plastic flasks and hydroxyapatite tricalcium phosphate osteoconductive biomaterial (HA/ß-TCP 3D scaffold); (2) proliferation rate; (3) ex vivo osteogenic differentiation in contexts of 2D monolayers on plastic and 3D HA/ß-TCP scaffolds; and (4) in vivo ectopic bone formation after subcutaneous implantation of cells with HA/ß-TCP scaffold into NOD/SCID mice. Von Kossa staining was used to assess ex vivo osteogenic differentiation in 3D cultures, providing a quantifiable test of 3D biomineralization ex vivo as a rapid, cost-effective potency assay. Near-equivalent capacities for cell survival, proliferation, and osteogenic differentiation were found for all transportation buffers. Moreover, cGMP-hBM-MSC transported from a production facility under clinical-grade conditions of 4% HSA in 0.9% saline to a destination 18 h away showed prompt adhesion to HA/ß-TCP 3D scaffold and subsequent in vivo bone formation. A successfully validated transportation protocol extends the applicability of fresh stem cells involving multicentric trials for regenerative medicine.


Assuntos
Células da Medula Óssea/citologia , Regeneração Óssea , Separação Celular/métodos , Células-Tronco Mesenquimais/citologia , Meios de Transporte , Animais , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/metabolismo , Regeneração Óssea/efeitos dos fármacos , Soluções Tampão , Fosfatos de Cálcio , Adesão Celular/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Coristoma/patologia , Durapatita/farmacologia , Matriz Extracelular/efeitos dos fármacos , Matriz Extracelular/metabolismo , Humanos , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Osteogênese/efeitos dos fármacos , Preservação Biológica , Tecidos Suporte/química
12.
Acta Biomater ; 10(2): 901-11, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24211733

RESUMO

Three-dimensional (3D) scaffolds hold great potential for stem cell-based therapies. Indeed, recent results have shown that biomimetic scaffolds may enhance cell survival and promote an increase in the concentration of therapeutic cells at the injury site. The aim of this work was to engineer an original polymeric scaffold based on the respective beneficial effects of alginate and chitosan. Formulations were made from various alginate/chitosan ratios to form opposite-charge polyelectrolyte complexes (PECs). After freeze-drying, the resultant matrices presented a highly interconnected porous microstructure and mechanical properties suitable for cell culture. In vitro evaluation demonstrated their compatibility with mesenchymal stell cell (MSC) proliferation and their ability to maintain paracrine activity. Finally, the in vivo performance of seeded 3D PEC scaffolds with a polymeric ratio of 40/60 was evaluated after an acute myocardial infarction provoked in a rat model. Evaluation of cardiac function showed a significant increase in the ejection fraction, improved neovascularization, attenuated fibrosis as well as less left ventricular dilatation as compared to an animal control group. These results provide evidence that 3D PEC scaffolds prepared from alginate and chitosan offer an efficient environment for 3D culturing of MSCs and represent an innovative solution for tissue engineering.


Assuntos
Alginatos/química , Quitosana/química , Eletrólitos/química , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/citologia , Isquemia Miocárdica/terapia , Tecidos Suporte/química , Animais , Materiais Biocompatíveis/farmacologia , Adesão Celular/efeitos dos fármacos , Feminino , Fibrose , Testes de Função Cardíaca , Humanos , Fenômenos Mecânicos/efeitos dos fármacos , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Microscopia Eletrônica de Varredura , Isquemia Miocárdica/fisiopatologia , Comunicação Parácrina/efeitos dos fármacos , Próteses e Implantes , Ratos , Ratos Endogâmicos Lew
13.
J Cell Mol Med ; 18(1): 104-14, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24188055

RESUMO

Bone marrow mesenchymal stem cells (MSCs) are plastic adherent cells that can differentiate into various tissue lineages, including osteoblasts, adipocytes and chondrocytes. However, this progenitor property is not shared by all cells within the MSC population. In addition, MSCs vary in their proliferation capacity and expression of markers. Because of heterogeneity of CD146 expression in the MSC population, we compared CD146(-/Low) and CD146(High) cells under clonal conditions and after sorting of the non-clonal cell population to determine whether this expression is associated with specific functions. CD146(-/Low) and CD146(High) bone marrow MSCs did not differ in colony-forming unit-fibroblast number, osteogenic, adipogenic and chondrogenic differentiation or in vitro haematopoietic-supportive activity. However, CD146(-/Low) clones proliferated slightly but significantly faster than did CD146(High) clones. In addition, a strong expression of CD146 molecule was associated with a commitment to a vascular smooth muscle cell (VSMC) lineage characterized by a strong up-regulation of calponin-1 and SM22α expression and an ability to contract collagen matrix. Thus, within a bone marrow MSC population, certain subpopulations characterized by high expression of CD146, are committed towards a VSMC lineage.


Assuntos
Diferenciação Celular , Células-Tronco Mesenquimais/fisiologia , Miócitos de Músculo Liso/metabolismo , Antígeno CD146/metabolismo , Proliferação de Células , Separação Celular , Células Cultivadas , Fator 2 de Crescimento de Fibroblastos/fisiologia , Humanos , Dados de Sequência Molecular , Músculo Liso Vascular/citologia , Fenótipo , Transcriptoma , Fator de Crescimento Transformador beta1/fisiologia
14.
Stem Cells Transl Med ; 2(12): 946-52, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24191265

RESUMO

Growth differentiation factor 15 (GDF15) is a divergent member of the transforming growth factor ß family discovered in a broad range of cells, as indicated by the diversity of its nomenclature. However, the only tissue that expresses a high amount of GDF15 in the physiologic state is placenta. GDF15 is easily detected in blood, and its concentration varies with age. In fact, increased blood concentration of GDF15 is associated with numerous pathological conditions. However, the biological significance underlying these observations is far from clear. GDF15 could have a positive or negative role depending on the state of cells or their environment. Furthermore, study of its biology is hampered by lack of knowledge of its receptor and thus the signaling pathways that drive its action. GDF15 seems to be an integrative signal in pathologic conditions, giving information on severity of disease. Its effectiveness in classifying patients to modulate treatment remains to be shown. Development of therapeutic interventions with GDF15 or anti-GDF15 agents remains difficult until we uncover the mechanism that drives its activity.


Assuntos
Fator 15 de Diferenciação de Crescimento/metabolismo , Neoplasias/metabolismo , Transdução de Sinais , Animais , Biomarcadores Tumorais/metabolismo , Doenças Cardiovasculares/metabolismo , Eritrócitos/metabolismo , Eritropoese , Regulação da Expressão Gênica , Fator 15 de Diferenciação de Crescimento/genética , Humanos , Neoplasias/genética , Neoplasias/patologia , Prognóstico
15.
PLoS One ; 8(10): e77895, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24205017

RESUMO

Mesenchymal stromal cells are emerging as an extremely promising therapeutic agent for tissue regeneration due to their multi-potency, immune-modulation and secretome activities, but safety remains one of the main concerns, particularly when in vitro manipulation, such as cell expansion, is performed before clinical application. Indeed, it is well documented that in vitro expansion reduces replicative potential and some multi-potency and promotes cell senescence. Furthermore, during in vitro aging there is a decrease in DNA synthesis and repair efficiency thus leading to DNA damage accumulation and possibly inducing genomic instability. The European Research Project ADIPOA aims at validating an innovative cell-based therapy where autologous adipose stromal cells (ASCs) are injected in the diseased articulation to activate regeneration of the cartilage. The primary objective of this paper was to assess the safety of cultured ASCs. The maintenance of genetic integrity was evaluated during in vitro culture by karyotype and microsatellite instability analysis. In addition, RT-PCR array-based evaluation of the expression of genes related to DNA damage signaling pathways was performed. Finally, the senescence and replicative potential of cultured cells was evaluated by telomere length and telomerase activity assessment, whereas anchorage-independent clone development was tested in vitro by soft agar growth. We found that cultured ASCs do not show genetic alterations and replicative senescence during the period of observation, nor anchorage-independent growth, supporting an argument for the safety of ASCs for clinical use.


Assuntos
Tecido Adiposo/citologia , Cartilagem/citologia , Cartilagem/metabolismo , Diferenciação Celular , Regeneração/fisiologia , Células Estromais/citologia , Tecido Adiposo/metabolismo , Idoso , Biomarcadores/metabolismo , Cartilagem/lesões , Técnicas de Cultura de Células , Proliferação de Células , Ensaio de Imunoadsorção Enzimática , Feminino , Perfilação da Expressão Gênica , Instabilidade Genômica , Humanos , Masculino , Análise de Sequência com Séries de Oligonucleotídeos , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Estromais/metabolismo
16.
Stem Cell Res ; 11(2): 834-44, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23811540

RESUMO

Our work aimed at evaluating the role of adipose stem cells (ASC) on chondrocytes from osteoarthritic (OA) patients and identifying the mediators involved. We used primary chondrocytes, ASCs from different sources and bone marrow mesenchymal stromal cells (MSC) from OA donors. ASCs or MSCs were co-cultured with chondrocytes in a minimal medium and using cell culture inserts. Under these conditions, ASCs did not affect the proliferation of chondrocytes but significantly decreased camptothecin-induced apoptosis. Both MSCs and ASCs from different sources allowed chondrocytes in the cocultures maintaining a stable expression of markers specific for a mature phenotype, while expression of hypertrophic and fibrotic markers was decreased. A number of factors known to regulate the chondrocyte phenotype (IL-1ß, IL-1RA, TNF-α) and matrix remodeling (TIMP-1 and -2, MMP-1 and -9, TSP-1) were not affected. However, a significant decrease of TGF-ß1 secretion by chondrocytes and induction of HGF secretion by ASCs was observed. Addition of a neutralizing anti-HGF antibody reversed the anti-fibrotic effect of ASCs whereas hypertrophic markers were not modulated. In summary, ASCs are an interesting source of stem cells for efficiently reducing hypertrophy and dedifferentiation of chondrocytes, at least partly via the secretion of HGF. This supports the interest of using these cells in therapies for osteo-articular diseases.


Assuntos
Adipócitos/citologia , Condrócitos/metabolismo , Células-Tronco Mesenquimais/citologia , Osteoartrite/patologia , Adipócitos/metabolismo , Adulto , Apoptose/fisiologia , Diferenciação Celular/fisiologia , Células Cultivadas , Condrócitos/citologia , Feminino , Humanos , Masculino , Células-Tronco Mesenquimais/metabolismo , Pessoa de Meia-Idade , Osteoartrite/genética , Osteoartrite/metabolismo , Fenótipo
17.
Arthritis Rheum ; 65(5): 1271-81, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23613363

RESUMO

OBJECTIVE: To examine the effect of different sources of good manufacturing practice clinical grade adipose-derived mesenchymal stem cells (AD-MSCs) on inflammatory factors in osteoarthritic (OA) chondrocytes and synoviocytes. METHODS: AD-MSCs from infrapatellar Hoffa fat, subcutaneous (SC) hip fat, and SC abdominal fat were cocultured in Transwells with chondrocytes or synoviocytes. Inflammatory factors (interleukin-1ß [IL-1ß], tumor necrosis factor α, IL-6, CXCL1/growth-related oncogene α, CXCL8/IL-8, CCL2/monocyte chemotactic protein 1, CCL3/macrophage inflammatory protein 1α, and CCL5/RANTES) were evaluated by quantitative reverse transcription-polymerase chain reaction or multiplex bead-based immunoassay. The role of different immunomodulators was analyzed. RESULTS: All the inflammatory factors analyzed were down-modulated at the messenger RNA or protein level independently by all 3 AD-MSC sources or by allogeneic AD-MSCs used in coculture with chondrocytes or synoviocytes. Inflammatory factor down-modulation was observed only when AD-MSCs were cocultured with chondrocytes or synoviocytes that produced high levels of inflammatory factors, but no effect was observed in cells that produced low levels of those factors, thus highlighting a dependence of the AD-MSC effect on existing inflammation. The immunomodulators IL-10, IL-1 receptor antagonist, fibroblast growth factor 2, indoleamine 2,3-dioxygenase 1, and galectin 1 were not involved in AD-MSC effects, whereas the cyclooxygenase 2 (COX-2)/prostaglandin E2 (PGE2 ) pathway exerted a role in the mechanism of antiinflammatory AD-MSC action. CONCLUSION: The antiinflammatory effects of AD-MSCs are probably not dependent on AD-MSC adipose tissue sources and donors but rather on the inflammatory status of OA chondrocytes and synoviocytes. AD-MSCs seem to be able to sense and respond to the local environment. Even though a combination of different molecules may be involved in AD-MSC effects, the COX-2/PGE2 pathway may play a role, suggesting that AD-MSCs may be useful for therapies in osteoarticular diseases.


Assuntos
Adipócitos/citologia , Condrócitos/citologia , Dinoprostona/metabolismo , Células-Tronco Mesenquimais/citologia , Osteoartrite/patologia , Membrana Sinovial/citologia , Idoso , Biomarcadores/metabolismo , Cartilagem Articular/patologia , Células Cultivadas , Quimiocinas/genética , Quimiocinas/metabolismo , Condrócitos/metabolismo , Técnicas de Cocultura , Regulação para Baixo , Feminino , Regulação da Expressão Gênica , Humanos , Masculino , Células-Tronco Mesenquimais/metabolismo , Membrana Sinovial/metabolismo
18.
Cytotherapy ; 15(6): 641-8, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23570660

RESUMO

BACKGROUND AIMS: Adipose tissue is a rich and very convenient source of cells for regenerative medicine therapeutic approaches. However, a characterization of the population of adipose-derived stromal and stem cells (ASCs) with the greatest therapeutic potential remains unclear. Under the authority of International Federation of Adipose Therapeutics and International Society for Cellular Therapy, this paper sets out to establish minimal definitions of stromal cells both as uncultured stromal vascular fraction (SVF) and as an adherent stromal/stem cells population. METHODS: Phenotypic and functional criteria for the identification of adipose-derived cells were drawn from the literature. RESULTS: In the SVF, cells are identified phenotypically by the following markers: CD45-CD235a-CD31-CD34+. Added value may be provided by both a viability marker and the following surface antigens: CD13, CD73, CD90 and CD105. The fibroblastoid colony-forming unit assay permits the evaluation of progenitor frequency in the SVF population. In culture, ASCs retain markers in common with other mesenchymal stromal/stem cells (MSCs), including CD90, CD73, CD105, and CD44 and remain negative for CD45 and CD31. They can be distinguished from bone-marrow-derived MSCs by their positivity for CD36 and negativity for CD106. The CFU-F assay is recommended to calculate population doublings capacity of ASCs. The adipocytic, chondroblastic and osteoblastic differentiation assays serve to complete the cell identification and potency assessment in conjunction with a quantitative evaluation of the differentiation either biochemically or by reverse transcription polymerase chain reaction. CONCLUSIONS: The goal of this paper is to provide initial guidance for the scientific community working with adipose-derived cells and to facilitate development of international standards based on reproducible parameters.


Assuntos
Tecido Adiposo/citologia , Diferenciação Celular , Terapia Baseada em Transplante de Células e Tecidos , Células-Tronco/citologia , Células Estromais/citologia , Adipócitos/citologia , Antígenos CD34/metabolismo , Células Cultivadas , Citometria de Fluxo , Humanos , Células-Tronco Mesenquimais/citologia , Obesidade/patologia , Obesidade/terapia , Padrões de Referência , Medicina Regenerativa
19.
Arthritis Rheum ; 65(7): 1786-94, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23553439

RESUMO

OBJECTIVE: Mesenchymal stem cells (MSCs) represent a promising tool for cell therapy for several disorders, among them the osteoarticular diseases. For such clinical applications, intraarticular (IA) injection of MSCs may be favored for higher levels of safety and targeting of specific joints. Although the safety of intravenous (IV) administration of MSCs has been reported in a number of clinical trials, the safety and biodistribution of MSCs after IA injection have not been tested. Our objective was to assess the toxicity of clinical-grade human adipose-derived MSCs (AD-MSCs), as well as their biodistribution, after IA injection into SCID mice. METHODS: SCID mice received IA or IV administration of 10(6) human AD-MSCs. Several tissues were recovered at different time points and processed for histologic assessment or real-time polymerase chain reaction (PCR) analysis. A highly sensitive assay was used to monitor the distribution of AD-MSCs, based on amplification of human-specific Alu sequences. RESULTS: Absence of toxicity was observed after AD-MSC infusion. Alu PCR assay revealed a high sensitivity (1 human AD-MSC/10(5) murine cells), with a large linear range (1-5 × 10(4) /10(5) murine cells). Importantly, 15% of the IA-injected AD-MSCs were detectable in the joint for the first month and 1.5% of the AD-MSCs engrafted over the long term, at least 6 months. AD-MSCs were observed in the injected joints and in areas of tissue referred to as stem cell niches, such as the bone marrow, adipose tissue, and muscle. CONCLUSION: These data support the feasibility and safety of using IA delivery of human AD-MSCs in the treatment of rheumatic diseases that affect the joints.


Assuntos
Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/metabolismo , Tecido Adiposo/citologia , Animais , Células Cultivadas , Feminino , Humanos , Infusões Intravenosas , Injeções Intra-Articulares , Masculino , Transplante de Células-Tronco Mesenquimais/efeitos adversos , Camundongos , Camundongos SCID
20.
Stem Cells Dev ; 22(12): 1789-801, 2013 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-23339531

RESUMO

Clinical-grade mesenchymal stromal cells (MSCs) are usually expanded from bone marrow (BMMSCs) or adipose tissue (ADSCs) using processes mainly differing in the use of fetal calf serum (FCS) or human platelet lysate (PL). We aimed to compare immune modulatory properties of clinical-grade MSCs using a combination of fully standardized in vitro assays. BMMSCs expanded with FCS (BMMSC-FCS) or PL (BMMSC-PL), and ADSC-PL were analyzed in quantitative phenotypic and functional experiments, including their capacity to inhibit the proliferation of T, B, and NK cells. The molecular mechanisms supporting T-cell inhibition were investigated. These parameters were also evaluated after pre-stimulation of MSCs with inflammatory cytokines. BMMSC-FCS, BMMSC-PL, and ADSC-PL displayed significant differences in expression of immunosuppressive and adhesion molecules. Standardized functional assays revealed that resting MSCs inhibited proliferation of T and NK cells, but not B cells. ADSC-PL were the most potent in inhibiting T-cell growth, a property ascribed to interferon-γ-dependent indoleamine 2,3-dioxygenase activity. MSCs did not stimulate allogeneic T cell proliferation but were efficiently lysed by activated NK cells. The systematic use of quantitative and reproducible validation techniques highlights differences in immunological properties of MSCs produced using various clinical-grade processes. ADSC-PL emerge as a promising candidate for future clinical trials.


Assuntos
Tecido Adiposo/citologia , Células da Medula Óssea/citologia , Células-Tronco Mesenquimais/citologia , Tecido Adiposo/efeitos dos fármacos , Tecido Adiposo/imunologia , Animais , Linfócitos B/citologia , Linfócitos B/imunologia , Plaquetas/química , Plaquetas/imunologia , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/imunologia , Bovinos , Extratos Celulares/química , Extratos Celulares/farmacologia , Proliferação de Células , Técnicas de Cocultura , Citocinas/biossíntese , Citocinas/imunologia , Humanos , Imunomodulação , Indolamina-Pirrol 2,3,-Dioxigenase/imunologia , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Células Matadoras Naturais/citologia , Células Matadoras Naturais/imunologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/imunologia , Especificidade de Órgãos , Controle de Qualidade , Soro/química , Soro/imunologia , Linfócitos T/citologia , Linfócitos T/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...