Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Cells ; 10(10)2021 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-34685707

RESUMEN

BACKGROUND: Systemic sclerosis (SSc) is a severe autoimmune disease for which mesenchymal stromal cells (MSCs)-based therapy was reported to reduce SSc-related symptoms in pre-clinical studies. Recently, extracellular vesicles released by MSCs (MSC-EVs) were shown to mediate most of their therapeutic effect. Here, we aimed at improving their efficacy by increasing the MSC-EV dose or by IFNγ-priming of MSCs. METHODS: small size (ssEVs) and large size EVs (lsEVs) were recovered from murine MSCs that were pre-activated using 1 or 20 ng/mL of IFNγ. In the HOCl-induced model of SSc, mice were treated with EVs at day 21 and sacrificed at day 42. Lung and skin samples were collected for histological and molecular analyses. RESULTS: increasing the dose of MSC-EVs did not add benefit to the dose previously reported to be efficient in SSc. By contrast, IFNγ pre-activation improved MSC-EVs-based treatment, essentially in the lungs. Low doses of IFNγ decreased the expression of fibrotic markers, while high doses improved remodeling and anti-inflammatory markers. IFNγ pre-activation upregulated iNos, IL1ra and Il6 in MSCs and ssEVs and the PGE2 protein in lsEVs. CONCLUSION: IFNγ-pre-activation improved the therapeutic effect of MSC-EVs preferentially in the lungs of SSc mice by modulating anti-inflammatory and anti-fibrotic markers.


Asunto(s)
Vesículas Extracelulares/metabolismo , Interferón gamma/farmacología , Células Madre Mesenquimatosas/metabolismo , Fibrosis Pulmonar/complicaciones , Fibrosis Pulmonar/terapia , Esclerodermia Sistémica/complicaciones , Esclerodermia Sistémica/patología , Animales , Antiinflamatorios/farmacología , Vesículas Extracelulares/efectos de los fármacos , Vesículas Extracelulares/genética , Terapia de Inmunosupresión , Células Madre Mesenquimatosas/efectos de los fármacos , Ratones Endogámicos C57BL , MicroARNs/genética , MicroARNs/metabolismo , Esclerodermia Sistémica/inducido químicamente , Piel/patología , Regulación hacia Arriba/efectos de los fármacos
2.
Autoimmun Rev ; 20(12): 102987, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34718160

RESUMEN

The relationship between cancer and primary Sjögren's syndrome (pSS) is uncertain. While the increased risk of hematological malignancies is well-known, data on the comparative incidence of solid neoplasms is conflicting. This study aimed to explore the associations between cancer and pSS. This nationwide population-based retrospective study from the French health insurance database (PMSI) evaluated patients hospitalized with new-onset pSS from 2011 to 2018 against age- and sex-matched hospitalized controls (1:10). The incidence of hematological malignancies and solid neoplasms was compared between the two groups. Mortality and multiple cancer incidence were also evaluated. Adjusted Hazard Ratios (aHR) calculations included confounding factors, such as low socioeconomic status. Among 25,661 hospitalized patients with pSS versus 252,543 matched patients (median follow-up of 3.96 years), we observed a higher incidence rate of lymphomas (aHR, 1.97 [95% CI, 1.59-2.43]), Waldenström macroglobulinemia (aHR, 10.8 [6.5-18.0]), and leukemia (aHR, 1.61 [1.1-2.4]). Thyroid cancer incidence was higher (aHR, 1.7 [1.1-2.8]), whereas bladder and breast cancer incidences were lower (aHR, 0.58 [0.37-0.89] and 0.60 [0.49-0.74], respectively). pSS patients with breast cancer exhibited a lower mortality rate. A limitation was that the database only encompasses hospitalized patients, and immunological and histological details are not listed. We confirmed the increased risk of hematological malignancies and thyroid cancers among patients with pSS. The lower risk of breast cancer suggests a role of hormonal factors and raises questions of the concept of immune surveillance within breast tissue. Epidemiological and translational studies are required to elucidate the relationships between pSS and cancer.


Asunto(s)
Neoplasias , Síndrome de Sjögren , Hospitalización , Humanos , Incidencia , Estudios Retrospectivos , Factores de Riesgo , Síndrome de Sjögren/complicaciones , Síndrome de Sjögren/epidemiología
3.
Int J Mol Sci ; 22(13)2021 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-34202139

RESUMEN

Systemic sclerosis (SSc) is a complex disorder resulting from dysregulated interactions between the three main pathophysiological axes: fibrosis, immune dysfunction, and vasculopathy, with no specific treatment available to date. Adipose tissue-derived mesenchymal stromal cells (ASCs) and their extracellular vesicles (EVs) have proved efficacy in pre-clinical murine models of SSc. However, their precise action mechanism is still not fully understood. Because of the lack of availability of fibroblasts isolated from SSc patients (SSc-Fb), our aim was to determine whether a TGFß1-induced model of human myofibroblasts (Tß-Fb) could reproduce the characteristics of SSc-Fb and be used to evaluate the anti-fibrotic function of ASCs and their EVs. We found out that Tß-Fb displayed the main morphological and molecular features of SSc-Fb, including the enlarged hypertrophic morphology and expression of several markers associated with the myofibroblastic phenotype. Using this model, we showed that ASCs were able to regulate the expression of most myofibroblastic markers on Tß-Fb and SSc-Fb, but only when pre-stimulated with TGFß1. Of interest, ASC-derived EVs were more effective than parental cells for improving the myofibroblastic phenotype. In conclusion, we provided evidence that Tß-Fb are a relevant model to mimic the main characteristics of SSc fibroblasts and investigate the mechanism of action of ASCs. We further reported that ASC-EVs are more effective than parental cells suggesting that the TGFß1-induced pro-fibrotic environment may alter the function of ASCs.


Asunto(s)
Tejido Adiposo/citología , Vesículas Extracelulares/metabolismo , Células Madre Mesenquimatosas/metabolismo , Esclerodermia Sistémica/etiología , Esclerodermia Sistémica/metabolismo , Animales , Biomarcadores , Comunicación Celular , Susceptibilidad a Enfermedades , Fibroblastos/metabolismo , Fibrosis , Humanos , Inmunofenotipificación , Células Madre Mesenquimatosas/citología , Ratones , Miofibroblastos/metabolismo , Esclerodermia Sistémica/patología , Factor de Crecimiento Transformador beta1/metabolismo
4.
J Autoimmun ; 121: 102660, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34020253

RESUMEN

Systemic sclerosis (SSc) is a potentially lethal disease with no curative treatment. Mesenchymal stromal cells (MSCs) have proved efficacy in SSc but no data is available on MSC-derived extracellular vesicles (EVs) in this multi-organ fibrosis disease. Small size (ssEVs) and large size EVs (lsEVs) were isolated from murine MSCs or human adipose tissue-derived MSCs (ASCs). Control antagomiR (Ct) or antagomiR-29a-3p (A29a) were transfected in MSCs and ASCs before EV production. EVs were injected in the HOCl-induced SSc model at day 21 and euthanasized at day 42. We found that both ssEVs and lsEVs were effective to slow-down the course of the disease. All disease parameters improved in skin and lungs. Interestingly, down-regulating miR-29a-3p in MSCs totally abolished therapeutic efficacy. Besides, we demonstrated a similar efficacy of human ASC-EVs and importantly, EVs from A29a-transfected ASCs failed to improve skin fibrosis. We identified Dnmt3a, Pdgfrbb, Bcl2, Bcl-xl as target genes of miR-29a-3p whose regulation was associated with skin fibrosis improvement. Our study highlights the therapeutic role of miR-29a-3p in SSc and the importance of regulating methylation and apoptosis.


Asunto(s)
Vesículas Extracelulares/trasplante , Células Madre Mesenquimatosas/inmunología , MicroARNs/metabolismo , Esclerodermia Sistémica/terapia , Animales , Apoptosis/genética , Apoptosis/inmunología , Metilación de ADN/inmunología , ADN Metiltransferasa 3A/genética , ADN Metiltransferasa 3A/metabolismo , Modelos Animales de Enfermedad , Vesículas Extracelulares/metabolismo , Femenino , Regulación de la Expresión Génica/inmunología , Humanos , Ácido Hipocloroso/administración & dosificación , Ácido Hipocloroso/toxicidad , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Ratones , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Esclerodermia Sistémica/inducido químicamente , Esclerodermia Sistémica/inmunología
5.
Artículo en Inglés | MEDLINE | ID: mdl-33015001

RESUMEN

Extracellular vesicles (EVs), including exosomes and microvesicles, derived from mesenchymal stem/stromal cells (MSCs) exert similar effects as their parental cells, and are of interest for various therapeutic applications. EVs can act through uptake by the target cells followed by release of their cargo inside the cytoplasm, or through interaction of membrane-bound ligands with receptors expressed on target cells to stimulate downstream intracellular pathways. EV-based therapeutics may be directly used as substitutes of intact cells or after modification for targeted drug delivery. However, for the development of EV-based therapeutics, several production, isolation, and characterization requirements have to be met and the quality of the final product has to be tested before its clinical implementation. In this review, we discuss the challenges associated with the development of EV-based therapeutics and the regulatory specifications for their successful clinical translation.

6.
Front Cell Dev Biol ; 8: 107, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32154253

RESUMEN

Aging is associated with high prevalence of chronic degenerative diseases that take a large part of the increasing burden of morbidities in a growing demographic of elderly people. Aging is a complex process that involves cell autonomous and cell non-autonomous mechanisms where senescence plays an important role. Senescence is characterized by the loss of proliferative potential, resistance to cell death by apoptosis and expression of a senescence-associated secretory phenotype (SASP). SASP includes pro-inflammatory cytokines and chemokines, tissue-damaging proteases, growth factors; all contributing to tissue microenvironment alteration and loss of tissue homeostasis. Emerging evidence suggests that the changes in the number and composition of extracellular vesicles (EVs) released by senescent cells contribute to the adverse effects of senescence in aging. In addition, age-related alterations in mesenchymal stem/stromal cells (MSCs) have been associated to dysregulated functions. The loss of functional stem cells necessary to maintain tissue homeostasis likely directly contributes to aging. In this review, we will focus on the characteristics and role of EVs isolated from senescent MSCs, the potential effect of MSC-derived EVs in aging and discuss their therapeutic potential to improve age-related diseases.

7.
Biomaterials ; 226: 119544, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31648137

RESUMEN

Mesenchymal stem/stromal cells (MSCs) are of interest in the context of osteoarthritis (OA) therapy. We previously demonstrated that TGFß-induced gene product-h3 (TGFBI/BIGH3) is downregulated in human MSCs (hMSCs) from patients with OA, suggesting a possible link with their impaired regenerative potential. In this study, we investigated TGFBI contribution to MSC-based therapy in OA models. First, we showed that co-culture with murine MSCs (mMSCs) partly restored the expression of anabolic markers and decreased expression of catabolic markers in OA-like chondrocytes only upon priming by TGFß3. Moreover, TGFß3-primed hMSCs not only modulated the expression of anabolic and catabolic markers, but also decreased inflammatory factors. Then, we found that upon TGFBI silencing, mMSCs partly lost their inductive effect on chondrocyte anabolic markers. Injection of hMSCs in which TGFBI was silenced did not protect mice from OA development. Finally, we showed that MSC chondroprotection was attributed to the presence of TGFBI mRNA and protein in extracellular vesicles. Our findings suggest that TGFBI is a chondroprotective factor released by MSCs and an anabolic regulator of cartilage homeostasis.


Asunto(s)
Cartílago Articular , Vesículas Extracelulares , Células Madre Mesenquimatosas , Osteoartritis , Animales , Células Cultivadas , Condrocitos , Técnicas de Cocultivo , Humanos , Ratones , Osteoartritis/terapia
8.
Front Immunol ; 9: 2571, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30455706

RESUMEN

Objectives: Skin fibrosis is the hallmark of systemic sclerosis (SSc) a rare intractable disease with unmet medical need. We previously reported the anti-fibrotic potential of mesenchymal stem cells (MSCs) in a murine model of SSc. This model, based on daily intra-dermal injections of hypochlorite (HOCl) during 6 weeks, is an inducible model of the disease. Herein, we aimed at characterizing the development of skin fibrosis in HOCl-induced SSc (HOCl-SSc), and evaluating the impact of MSC infusion during the fibrogenesis process. Methods: After HOCl-SSc induction in BALB/c mice, clinical, histological and biological parameters were measured after 3 weeks (d21) and 6 weeks (d42) of HOCl challenge, and 3 weeks after HOCl discontinuation (d63). Treated-mice received infusions of 2.5 × 105 MSCs 3 weeks before sacrifice (d0, d21, d42). Results: HOCl injections induced a two-step process of fibrosis development: first, an 'early inflammatory phase', characterized at d21 by highly proliferative infiltrates of myofibroblasts, T-lymphocytes and macrophages. Second, a phase of 'established matrix fibrosis', characterized at d42 by less inflammation, but strong collagen deposition and followed by a third phase of 'spontaneous tissue remodeling' after HOCl discontinuation. This phase was characterized by partial fibrosis receding, due to enhanced MMP1/TIMP1 balance. MSC treatment reduced skin thickness in the three phases of fibrogenesis, exerting more specialized mechanisms: immunosuppression, abrogation of myofibroblast activation, or further enhancing tissue remodeling, depending on the injection time-point. Conclusion: HOCl-SSc mimics three fibrotic phenotypes of scleroderma, all positively impacted by MSC therapy, demonstrating the great plasticity of MSC, a promising cure for SSc.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Colágeno/metabolismo , Fibrosis/terapia , Trasplante de Células Madre Mesenquimatosas/métodos , Esclerodermia Sistémica/terapia , Enfermedades de la Piel/terapia , Animales , Modelos Animales de Enfermedad , Femenino , Ácido Hipocloroso , Células Madre Mesenquimatosas/citología , Ratones , Ratones Endogámicos BALB C , Estrés Oxidativo/fisiología , Piel/patología , Enfermedades de la Piel/inducido químicamente
9.
Front Immunol ; 9: 2938, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30619298

RESUMEN

Systemic sclerosis (SSc) is a rare autoimmune disease, which is potentially lethal. The physiopathology of the disease is still incompletely elucidated although the role of fibroblasts, endothelial cells (ECs), immune cells. and the environment (i.e., oxidative stress) has been demonstrated. This is an intractable disease with an urgent need to provide better therapeutic options to patients. Mesenchymal stem cells (MSCs) represent a promising therapeutic approach thanks to the number of trophic and pleiotropic properties they exert. Among these, MSCs display anti-fibrotic, angiogenic, and immunomodulatory capacities that might be of interest in the treatment of SSc by acting on different processes that are dysregulated in the disease. In the recent years, the therapeutic effectiveness of MSCs has been demonstrated in different preclinical animal models and is being investigated in phase I clinical trials. Both allogenic and autologous transplantation of MSCs isolated from bone marrow or adipose tissue is being evaluated. The rationale for using allogenic MSCs in SSc, as well as in other autoimmune diseases, is based on the possibility that autologous MSCs might be altered in these diseases. In SSc, reports from the literature are controversial. Nevertheless, the role of the oxidative environment and of the crosstalk with neighboring cells (fibroblasts and ECs) on the functional properties of MSCs has been reported. Here, we review the preclinical and clinical data reporting the interest of MSC-based treatment in SSc and question the use of autologous or allogeneic MSCs in perspective of clinical applications.


Asunto(s)
Enfermedades Autoinmunes/terapia , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/inmunología , Esclerodermia Sistémica/terapia , Animales , Enfermedades Autoinmunes/inmunología , Diferenciación Celular , Proliferación Celular , Humanos , Células Madre Mesenquimatosas/citología , Esclerodermia Sistémica/inmunología , Trasplante Autólogo , Trasplante Homólogo
10.
Front Immunol ; 9: 3056, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30622540

RESUMEN

Objectives: Fibrosis is a hallmark of systemic sclerosis (SSc), an intractable disease where innovative strategies are still being sought. Among novel anti-fibrotic approaches, mesenchymal stromal/stem cell (MSC)-based therapy appears promising. Previously, we reported anti-fibrotic effects of MSC in an experimental model of SSc, through various mechanisms (tissue remodeling, immunomodulation, anti-oxidant defense). Since immunomodulation is a pivotal mechanism for MSC therapeutic effects, we investigated the specific role of critical molecules associated with MSC immunosuppressive properties and hypothesized that MSC defective for these molecules would be less effective in reducing fibrosis in SSc. Methods: SSc was induced by 6-week daily intradermal injections of hypochlorite (HOCl) in mice. MSC were isolated from the bone marrow of wild type mice (WT) or mice knockout for IL1RA, IL6, or iNOS (IL1RA-/-, IL6-/-, or iNOS-/- MSC, respectively). Treated-mice received 2.5 × 105 MSC intravenous infusion at d21. Skin thickness, histological and biological parameters were evaluated in skin and blood at d42. Results: IL1RA-/- and IL6-/- MSC exerted similar anti-fibrotic properties as WT MSC, with a reduction of skin thickness together with less collagen deposition. Conversely, iNOS-/- MSC did not exert anti-fibrotic functions as shown by a similar skin thickness progression as non-treated HOCl-SSc mice. Compared with WT MSC, iNOS-/- MSC kept some immunosuppressive and tissue remodeling properties, but lost their capacity to reduce oxidative stress in HOCl-SSc mice. Conclusion: Our study highlights the crucial role of iNOS, whose activity is required for the anti-fibrotic properties of MSC in experimental SSc, with a special emphasis on NO-related anti-oxidant functions.


Asunto(s)
Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/inmunología , Óxido Nítrico Sintasa de Tipo II/metabolismo , Esclerodermia Sistémica/terapia , Piel/patología , Animales , Modelos Animales de Enfermedad , Femenino , Fibrosis , Humanos , Ácido Hipocloroso/administración & dosificación , Ácido Hipocloroso/toxicidad , Inyecciones Intradérmicas , Proteína Antagonista del Receptor de Interleucina 1/genética , Interleucina-6/genética , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/inmunología , Estrés Oxidativo/inmunología , Esclerodermia Sistémica/inducido químicamente , Esclerodermia Sistémica/inmunología , Esclerodermia Sistémica/patología , Piel/efectos de los fármacos , Piel/inmunología , Resultado del Tratamiento
11.
Front Immunol ; 9: 3112, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30687318

RESUMEN

Systemic sclerosis (SSc) is an autoimmune connective tissue disorder, characterized by multisystem involvement, vasculopathy, and fibrosis. An increased risk of malignancy is observed in SSc (including breast and lung cancers), and in a subgroup of patients with specific autoantibodies (i.e., anti-RNA polymerase III and related autoantibodies), SSc could be a paraneoplastic syndrome and might be directly related to an immune response against cancer. Herein, we reviewed the literature, focusing on the most recent articles, and shed light onto the potential relationship between cancer and scleroderma regarding temporal and immunological dimensions.


Asunto(s)
Autoanticuerpos/inmunología , Enfermedades Autoinmunes/inmunología , Neoplasias/inmunología , Síndromes Paraneoplásicos/inmunología , Esclerodermia Sistémica/inmunología , Enfermedades Autoinmunes/complicaciones , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/patología , Fibrosis , Humanos , Neoplasias/complicaciones , Neoplasias/genética , Síndromes Paraneoplásicos/complicaciones , Síndromes Paraneoplásicos/genética , ARN Polimerasa III/inmunología , Esclerodermia Sistémica/complicaciones , Esclerodermia Sistémica/genética , Esclerodermia Sistémica/patología , Piel/inmunología , Piel/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...