Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.565
Filtrar
1.
Cell Immunol ; 401-402: 104840, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38880071

RESUMEN

Sepsis is characterized by an exacerbated inflammatory response, driven by the overproduction of cytokines, a phenomenon known as a cytokine storm. This condition is further compounded by the extensive infiltration of M1 macrophages and the pyroptosis of these cells, leading to immune paralysis. To counteract this, we sought to transition M1 macrophages into the M2 phenotype and safeguard them from pyroptosis. For this purpose, we employed ectodermal mesenchymal stem cells (EMSCs) sourced from the nasal mucosa to examine their impact on both macrophages and septic animal models. The co-culture protocol involving LPS-stimulated rat bone marrow macrophages and EMSCs was employed to examine the paracrine influence of EMSCs on macrophages. The intravenous administration of EMSCs was utilized to observe the enhancement in the survival rate of septic rat models and the protection of associated organs. The findings indicated that EMSCs facilitated M2 polarization of macrophages, which were stimulated by LPS, and significantly diminished levels of pro-inflammatory cytokines and NLRP3. Furthermore, EMSCs notably restored the mitochondrial membrane potential (MMP) of macrophages through paracrine action, eliminated excess reactive oxygen species (ROS), and inhibited macrophage pyroptosis. Additionally, the systemic integration of EMSCs substantially reduced injuries to multiple organs and preserved the fundamental functions of the heart, liver, and kidney in CLP rats, thereby extending their survival.


Asunto(s)
Macrófagos , Células Madre Mesenquimatosas , Mucosa Nasal , Piroptosis , Sepsis , Animales , Células Madre Mesenquimatosas/inmunología , Células Madre Mesenquimatosas/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Ratas , Mucosa Nasal/inmunología , Mucosa Nasal/citología , Sepsis/inmunología , Masculino , Ratas Sprague-Dawley , Trasplante de Células Madre Mesenquimatosas/métodos , Lipopolisacáridos , Citocinas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Modelos Animales de Enfermedad , Técnicas de Cocultivo , Potencial de la Membrana Mitocondrial , Células Cultivadas
2.
Nat Commun ; 15(1): 5176, 2024 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-38890279

RESUMEN

The longevity of grafts remains a major challenge in allogeneic transplantation due to immune rejection. Systemic immunosuppression can impair graft function and can also cause severe adverse effects. Here, we report a local immuno-protective strategy to enhance post-transplant persistence of allografts using a mesenchymal stem cell membrane-derived vesicle (MMV)-crosslinked hydrogel (MMV-Gel). MMVs are engineered to upregulate expression of Fas ligand (FasL) and programmed death ligand 1 (PD-L1). The MMVs are retained within the hydrogel by crosslinking. The immuno-protective microenvironment of the hydrogel protects allografts by presenting FasL and PD-L1. The binding of these ligands to T effector cells, the dominant contributors to graft destruction and rejection, results in apoptosis of T effector cells and generation of regulatory T cells. We demonstrate that implantation with MMV-Gel prolongs the survival and function of grafts in mouse models of allogeneic pancreatic islet cells and skin transplantation.


Asunto(s)
Proteína Ligando Fas , Hidrogeles , Trasplante de Islotes Pancreáticos , Ratones Endogámicos C57BL , Trasplante de Piel , Linfocitos T Reguladores , Trasplante Homólogo , Animales , Hidrogeles/química , Ratones , Proteína Ligando Fas/metabolismo , Proteína Ligando Fas/inmunología , Linfocitos T Reguladores/inmunología , Trasplante de Islotes Pancreáticos/métodos , Trasplante de Piel/métodos , Antígeno B7-H1/metabolismo , Antígeno B7-H1/inmunología , Células Madre Mesenquimatosas/inmunología , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/efectos de los fármacos , Ratones Endogámicos BALB C , Supervivencia de Injerto/efectos de los fármacos , Supervivencia de Injerto/inmunología , Rechazo de Injerto/prevención & control , Rechazo de Injerto/inmunología , Humanos , Masculino , Apoptosis/efectos de los fármacos
3.
Adv Immunol ; 162: 23-58, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38866438

RESUMEN

The intestine represents the most complex cellular network in the whole body. It is constantly faced with multiple types of immunostimulatory agents encompassing from food antigen, gut microbiome, metabolic waste products, and dead cell debris. Within the intestine, most T cells are found in three primary compartments: the organized gut-associated lymphoid tissue, the lamina propria, and the epithelium. The well-orchestrated epithelial-immune-microbial interaction is critically important for the precise immune response. The main role of intestinal mesenchymal stromal cells is to support a structural framework within the gut wall. However, recent evidence from stromal cell studies indicates that they also possess significant immunomodulatory functions, such as maintaining intestinal tolerance via the expression of PDL1/2 and MHC-II molecules, and promoting the development of CD103+ dendritic cells, and IgA+ plasma cells, thereby enhancing intestinal homeostasis. In this review, we will summarize the current understanding of CD8+ T cells and stromal cells alongside the intestinal tract and discuss the reciprocal interactions between T subsets and mesenchymal stromal cell populations. We will focus on how the tissue residency, migration, and function of CD8+ T cells could be potentially regulated by mesenchymal stromal cell populations and explore the molecular mediators, such as TGF-ß, IL-33, and MHC-II molecules that might influence these processes. Finally, we discuss the potential pathophysiological impact of such interaction in intestine hemostasis as well as diseases of inflammation, infection, and malignancies.


Asunto(s)
Linfocitos T CD8-positivos , Homeostasis , Células Madre Mesenquimatosas , Humanos , Células Madre Mesenquimatosas/inmunología , Animales , Linfocitos T CD8-positivos/inmunología , Mucosa Intestinal/inmunología , Comunicación Celular/inmunología , Intestinos/inmunología
4.
Biomolecules ; 14(6)2024 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-38927060

RESUMEN

Plasmacytoid dendritic cells (pDCs) are vital players in antiviral immune responses because of their high levels of IFN-α secretion. However, this attribute has also implicated them as critical factors behind the immunopathogenesis of inflammatory diseases, and no currently available therapy can efficiently inhibit pDCs' aberrant activation. Mesenchymal stromal cells (MSCs) possess stromal immunomodulatory functionality, regulating immune cell activation through several mechanisms, including the adenosinergic (CD39/CD73/adenosine) pathway. The IFN-γ preconditioning of bone marrow MSCs improves their inhibitory properties for therapy applications; however, isolating human gingival tissue-derived MSCs (hGMSCs) is more accessible. These cells have shown better immunomodulatory effects, yet the outcome of IFN-γ preconditioning and its impact on the adenosinergic pathway has not been evaluated. This study first validated the immunoregulatory properties of primary-cultured hGMSCs, and the results showed that IFN-γ preconditioning strengthens CD39/CD73 coexpression, adenosine production, and the regulatory properties of hGMSC, which were confirmed by describing for the first time their ability to reduce pDC activation and their IFN-α secretion and to increase the frequency of CD73+ pDC. In addition, when CD73's enzymatic activity was neutralized in hGMSCs, adenosine production and the IFN-γ preconditioning effect were restrained. This evidence might be applied to design hGMSCs- and adenosine-based immunotherapeutic strategies for treating inflammatory disorders that are associated with pDC overactivation.


Asunto(s)
5'-Nucleotidasa , Adenosina , Células Dendríticas , Encía , Interferón gamma , Células Madre Mesenquimatosas , Humanos , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/inmunología , Células Madre Mesenquimatosas/citología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Adenosina/metabolismo , Interferón gamma/metabolismo , Encía/citología , 5'-Nucleotidasa/metabolismo , Células Cultivadas , Apirasa/metabolismo , Proteínas Ligadas a GPI
6.
Biochemistry (Mosc) ; 89(5): 883-903, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38880649

RESUMEN

Immune system and bone marrow stromal cells play an important role in maintaining normal hematopoiesis. Lymphoid neoplasia disturbs not only development of immune cells, but other immune response mechanisms as well. Multipotent mesenchymal stromal cells (MSCs) of the bone marrow are involved in immune response regulation through both intercellular interactions and secretion of various cytokines. In hematological malignancies, the bone marrow stromal microenvironment, including MSCs, is altered. Aim of this study was to describe the differences of MSCs' immunological function in the patients with acute lymphoblastic leukemia (ALL) and diffuse large B-cell lymphoma (DLBCL). In ALL, malignant cells arise from the early precursor cells localized in bone marrow, while in DLBCL they arise from more differentiated B-cells. In this study, only the DLBCL patients without bone marrow involvement were included. Growth parameters, surface marker expression, genes of interest expression, and secretion pattern of bone marrow MSCs from the patients with ALL and DLBCL at the onset of the disease and in remission were studied. MSCs from the healthy donors of corresponding ages were used as controls. It has been shown that concentration of MSCs in the bone marrow of the patients with ALL is reduced at the onset of the disease and is restored upon reaching remission; in the patients with DLBCL this parameter does not change. Proliferative capacity of MSCs did not change in the patients with ALL; however, the cells of the DLBCL patients both at the onset and in remission proliferated significantly faster than those from the donors. Expression of the membrane surface markers and expression of the genes important for differentiation, immunological status maintenance, and cytokine secretion differed significantly in the MSCs of the patients from those of the healthy donors and depended on nosology of the disease. Secretomes of the MSCs varied greatly; a number of proteins associated with immune response regulation, differentiation, and maintenance of hematopoietic stem cells were depleted in the secretomes of the cells from the patients. Lymphoid neoplasia leads to dramatic changes in the functional immunological status of MSCs.


Asunto(s)
Linfoma de Células B Grandes Difuso , Células Madre Mesenquimatosas , Leucemia-Linfoma Linfoblástico de Células Precursoras , Humanos , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/inmunología , Leucemia-Linfoma Linfoblástico de Células Precursoras/inmunología , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Linfoma de Células B Grandes Difuso/inmunología , Linfoma de Células B Grandes Difuso/patología , Linfoma de Células B Grandes Difuso/metabolismo , Masculino , Adulto , Femenino , Persona de Mediana Edad , Células de la Médula Ósea/inmunología , Proliferación Celular , Adulto Joven
8.
Front Immunol ; 15: 1361596, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38690266

RESUMEN

Mesenchymal stromal/stem cells (MSCs), which are distributed in many tissues including bone marrow, have been reported to play a critical role in tumor development. While bone marrow, the primary site for hematopoiesis, is important for establishing the immune system, whether MSCs in the bone marrow can promote tumor growth via influencing hematopoiesis remains unclear. We observed that the numbers of MSCs and neutrophils were increased in bone marrow in tumor-bearing mice. Moreover, co-culture assay showed that MSCs strongly protected neutrophils from apoptosis and induced their maturation. G-CSF and GM-CSF have been well-documented to be associated with neutrophil formation. We found a remarkably increased level of G-CSF, but not GM-CSF, in the supernatant of MSCs and the serum of tumor-bearing mice. The G-CSF expression can be enhanced with inflammatory cytokines (IFNγ and TNFα) stimulation. Furthermore, we found that IFNγ and TNFα-treated MSCs enhanced their capability of promoting neutrophil survival and maturation. Our results indicate that MSCs display robustly protective effects on neutrophils to contribute to tumor growth in bone niches.


Asunto(s)
Citocinas , Células Madre Mesenquimatosas , Neutrófilos , Animales , Células Madre Mesenquimatosas/inmunología , Células Madre Mesenquimatosas/metabolismo , Neutrófilos/inmunología , Neutrófilos/metabolismo , Ratones , Citocinas/metabolismo , Ratones Endogámicos C57BL , Técnicas de Cocultivo , Factor Estimulante de Colonias de Granulocitos/metabolismo , Apoptosis , Factor de Necrosis Tumoral alfa/metabolismo , Línea Celular Tumoral , Neoplasias/inmunología , Neoplasias/patología
9.
Nat Med ; 30(6): 1556-1558, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38778211

RESUMEN

The first completed clinical trial of induced pluripotent stem cell (iPS cell)-derived cells was conducted in 15 participants with steroid-resistant acute graft-versus-host disease. After intravenous infusion of mesenchymal stromal cells (CYP-001 derived from a clone of human iPS cells), we reported the safety, tolerability and efficacy within the primary evaluation period at day 100. We now report results at the 2-year follow-up: 9 of 15 (60%) participants survived, which compares favorably with previously reported outcomes in studies of steroid-resistant acute graft-versus-host disease. Causes of death were complications commonly observed in recipients of allogeneic hematopoietic stem cell transplantation, and not considered by the investigators to be related to CYP-001 treatment. There were no serious adverse events, tumors or other safety concerns related to CYP-001. In conclusion, systemic delivery of iPS cell-derived cells was safe and well tolerated over 2 years of follow-up, with sustained outcomes up to 2 years after the first infusion. ClinicalTrials.gov registration: NCT02923375 .


Asunto(s)
Enfermedad Injerto contra Huésped , Células Madre Pluripotentes Inducidas , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Humanos , Células Madre Pluripotentes Inducidas/trasplante , Trasplante de Células Madre Mesenquimatosas/efectos adversos , Células Madre Mesenquimatosas/inmunología , Masculino , Adulto , Femenino , Persona de Mediana Edad , Esteroides/uso terapéutico , Resultado del Tratamiento , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Estudios de Seguimiento , Enfermedad Aguda , Resistencia a Medicamentos , Adulto Joven , Trasplante Homólogo/efectos adversos
10.
Expert Rev Anti Infect Ther ; 22(6): 435-451, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38804866

RESUMEN

INTRODUCTION: The emergence of antiparasitic drug resistance poses a concerning threat to animals and humans. Mesenchymal Stem Cells (MSCs) have been widely used to treat infections in humans, pets, and livestock. Although this is an emerging field of study, the current review outlines possible mechanisms and examines potential synergism in combination therapies and the possible harmful effects of such an approach. AREAS COVERED: The present study delved into the latest pre-clinical research on utilizing MSCs to treat parasitic infections. As per investigations, the introduction of MSCs to patients grappling with parasitic diseases like schistosomiasis, malaria, cystic echinococcosis, toxoplasmosis, leishmaniasis, and trypanosomiasis has shown a reduction in parasite prevalence. This intervention also alters the levels of both pro- and anti-inflammatory cytokines. Furthermore, the combined administration of MSCs and antiparasitic drugs has demonstrated enhanced efficacy in combating parasites and modulating the immune response. EXPERT OPINION: Mesenchymal stem cells are a potential solution for addressing parasitic drug resistance. This is mainly because of their remarkable immunomodulatory abilities, which can potentially help combat parasites' resistance to drugs.


Asunto(s)
Resistencia a Medicamentos , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Enfermedades Parasitarias , Humanos , Animales , Enfermedades Parasitarias/inmunología , Enfermedades Parasitarias/tratamiento farmacológico , Células Madre Mesenquimatosas/inmunología , Antiparasitarios/farmacología , Antiparasitarios/administración & dosificación , Terapia Combinada , Inmunomodulación/efectos de los fármacos , Antiinflamatorios/farmacología , Antiinflamatorios/administración & dosificación , Citocinas/metabolismo , Citocinas/inmunología
11.
PLoS One ; 19(5): e0304713, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38820477

RESUMEN

Argentatins are secondary metabolites synthesized by guayule (Parthenium argentatum A. Gray) with numerous potential medical applications. In addition to inhibiting insect growth, they are endowed with several pharmacological properties including antimicrobial and antitumorigenic activity. However, their potential as immunomodulators remains unexplored. The aim of the present study was to investigate whether argentatins can modulate the function of the immune system. Human mesenchymal stem cells were treated with argentatins and the production of several anti- and proinflammatory cytokines was evaluated. The effect of argentatins on the polarization of CD4+ T-lymphocytes and macrophages was also assessed. Results demonstrated that argentatins can modulate the production of proinflammatory cytokines and the polarization of cellular phenotypes, including Th2 lymphocytes and M1 macrophages. These findings suggest that argentatins are promising therapeutic agents in autoimmune or allergic diseases, and open new perspectives for the investigation of argentatins in immune response and in the development of more targeted and effective immunomodulatory therapies.


Asunto(s)
Citocinas , Humanos , Citocinas/metabolismo , Células Madre Mesenquimatosas/inmunología , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Macrófagos/inmunología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Factores Inmunológicos/farmacología , Agentes Inmunomoduladores/farmacología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/efectos de los fármacos
12.
Cell Biol Int ; 48(8): 1124-1137, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38741520

RESUMEN

The most common central nervous system (CNS) inflammatory disease is multiple sclerosis (MS), modeled using experimental autoimmune encephalomyelitis (EAE). Mesenchymal stem cells (MSCs) exhibit potent immunomodulatory capabilities, including the suppression of immune cell functions and anti-inflammatory cytokine production. Female C57BL/6 mice (8-10 weeks old) were divided into three groups: 1. Control, 2. Allogeneic MSCs (ALO) treatment, and 3. Syngeneic MSCs (SYN) treatment. To induce EAE, myelin oligodendrocyte glycoprotein was injected subcutaneously with complete Freund's adjuvant, followed by intraperitoneal pertussis toxin. On Days 6 and 12 postimmunization, the treatment groups received intraperitoneal injections of 2 × 106 MSCs. Daily clinical and weight assessments were performed, and on Day 25, the mice were euthanized. At the end of the period, brain histological analysis was conducted to quantify lymphocyte infiltration. T-cell characteristics were determined using enzyme-linked immunosorbent assay and Real-time polymerase chain reaction (RT-PCR). The assessment of transcription factor expression levels in the CNS was also performed using RT-PCR. Compared to the control group, both the allogeneic (ALO) and syngeneic (SYN) groups demonstrated significantly reduced disease progression. The maximum clinical scores for the control, ALO, and SYN groups were 4.4 ± 0.1, 2.4 ± 0.2, and 2.1 ± 0.2, respectively (ALO and SYN vs. Control: p < .001). In comparison to the control group, histological studies demonstrated that the allogeneic and syngeneic groups had less lymphocytic infiltration (ALO: 1.4 ± 0.1, SYN: 1.2 ± 0.2, and control: 2.8 ± 0.15; p < .001) and demyelination (ALO: 1.2 ± 0.15, SYN: 1.1 ± 0.1 and control: 2.9 ± 0.1, p < .001). ALO and SYN groups had lower expression of Th1 and Th17 cytokines and transcription factors (IFN-γ: 0.067, 0.051; STAT4: 0.189, 0.162; T-bet: 0.175, 0.163; IL-17: 0.074, 0.061; STAT3: 0.271, 0.253; ROR-γt: 0.163, 0.149, respectively) compared to the control group on Day 25 following EAE induction. Additionally, ALO and SYN groups compared to the control group, expressed more Th2 and Treg cytokines and transcription factors (IL-4: 4.25, 4.63; STAT6: 2.78, 2.96; GATA3: 2.91, 3.08; IL-27: 2.32, 2.46, IL-33: 2.71, 2.85; TGF-ß: 4.8, 5.05; IL-10: 4.71, 4.93; CTLA-4: 7.72, 7.95; PD1: 4.12,4.35; Foxp3: 3.82,4.08, respectively). This research demonstrated that MSCs possess the potential to be a therapeutic option for MS and related CNS inflammatory disorders. Their immunomodulatory properties, coupled with the observed reductions in disease severity, lymphocytic infiltration, and demyelination, indicate that MSCs could play a crucial role in altering the course of MS by mitigating inflammatory immune responses and promoting regulatory immune processes. These findings open up new possibilities for the development of MSC-based therapies for MS, and further investigation and clinical trials may be warranted to explore their efficacy and safety in human patients.


Asunto(s)
Encefalomielitis Autoinmune Experimental , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Ratones Endogámicos C57BL , Linfocitos T Reguladores , Células TH1 , Células Th17 , Células Th2 , Animales , Encefalomielitis Autoinmune Experimental/terapia , Encefalomielitis Autoinmune Experimental/inmunología , Femenino , Células Madre Mesenquimatosas/inmunología , Células Madre Mesenquimatosas/metabolismo , Trasplante de Células Madre Mesenquimatosas/métodos , Células TH1/inmunología , Ratones , Células Th17/inmunología , Linfocitos T Reguladores/inmunología , Células Th2/inmunología , Tejido Adiposo/citología , Citocinas/metabolismo
13.
Int Immunopharmacol ; 135: 112315, 2024 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-38805908

RESUMEN

Exosomes generated from mesenchymal stem cells (MSCs) are thought to be a unique therapeutic strategy for several autoimmune deficiency illnesses. The purpose of this study was to elucidate the protective effects of human umbilical cord mesenchymal stem cell-derived exosomes (hUCMSC-Exo) on CD4+ T cells dysfunction during graft-versus-host disease (GVHD) and to identify the underlying processes involved. Here, we showed that hUCMSC-Exo treatment can effectively attenuate GVHD injury by alleviating redox metabolism disorders and inflammatory cytokine bursts in CD4+ T cells. Furthermore, hUCMSC-Exo ameliorate ER stress and ATF6/CHOP signaling-mediated apoptosis in CD4+ T cells and promote the development of CD4+IL-10+ T cells during GVHD. Moreover, downregulating miR-16-5p in hUCMSC-Exo impaired their ability to prevent CD4+ T cells apoptosis and weakened their ability to promote the differentiation of CD4+IL-10+ T cells. Collectively, the obtained data suggested that hUCMSC-Exo suppress ATF6/CHOP signaling-mediated ER stress and apoptosis in CD4+ T cells, enhance the differentiation of CD4+IL-10+ T cells, and reverse the imbalance of immune homeostasis in the GVHD process by transferring miR-16-5p. Our study provided further evidence that GVHD patients can benefit from hUCMSC-Exo-mediated therapy.


Asunto(s)
Factor de Transcripción Activador 6 , Linfocitos T CD4-Positivos , Estrés del Retículo Endoplásmico , Exosomas , Enfermedad Injerto contra Huésped , Células Madre Mesenquimatosas , MicroARNs , Transducción de Señal , Factor de Transcripción CHOP , MicroARNs/metabolismo , MicroARNs/genética , Exosomas/metabolismo , Estrés del Retículo Endoplásmico/inmunología , Enfermedad Injerto contra Huésped/inmunología , Enfermedad Injerto contra Huésped/prevención & control , Humanos , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Animales , Factor de Transcripción Activador 6/metabolismo , Factor de Transcripción Activador 6/genética , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/inmunología , Factor de Transcripción CHOP/metabolismo , Factor de Transcripción CHOP/genética , Apoptosis , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Cordón Umbilical/citología , Células Cultivadas
14.
Kaohsiung J Med Sci ; 40(6): 520-529, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38712483

RESUMEN

Autoimmune disease is characterized by the proliferation of harmful immune cells, inducing tissue inflammation and ultimately causing organ damage. Current treatments often lack specificity, necessitating high doses, prolonged usage, and high recurrence rates. Therefore, the identification of innovative and safe therapeutic strategies is urgently required. Recent preclinical studies and clinical trials on inflammatory and autoimmune diseases have evidenced the immunosuppressive properties of mesenchymal stromal cells (MSCs). Studies have demonstrated that extracellular vesicles (EV) derived from MSCs can mitigate abnormal autoinflammation while maintaining safety within the diseased microenvironment. This study conducted a systematic review to elucidate the crucial role of MSC-EVs in alleviating autoimmune diseases, particularly focusing on their impact on the underlying mechanisms of autoimmune conditions such as rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), and inflammatory bowel disease (IBD). By specifically examining the regulatory functions of microRNAs (miRNAs) derived from MSC-EVs, the comprehensive study aimed to enhance the understanding related to disease mechanisms and identify potential diagnostic markers and therapeutic targets for these diseases.


Asunto(s)
Enfermedades Autoinmunes , Vesículas Extracelulares , Células Madre Mesenquimatosas , MicroARNs , Humanos , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/inmunología , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/trasplante , Enfermedades Autoinmunes/terapia , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/patología , MicroARNs/genética , MicroARNs/metabolismo , Lupus Eritematoso Sistémico/terapia , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/patología , Artritis Reumatoide/terapia , Artritis Reumatoide/inmunología , Artritis Reumatoide/metabolismo , Artritis Reumatoide/patología , Animales , Enfermedades Inflamatorias del Intestino/terapia , Enfermedades Inflamatorias del Intestino/metabolismo , Enfermedades Inflamatorias del Intestino/inmunología , Enfermedades Inflamatorias del Intestino/patología , Inmunomodulación
15.
FASEB J ; 38(10): e23683, 2024 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-38758184

RESUMEN

Mesenchymal stromal cells (MSCs) have been shown to modulate the function of various subsets of T cells such as naïve CD4+ T cells and IFNγ+CD4+ Th1 cells; however, mechanisms underlying this regulation have not been fully deciphered. Our in vitro culture assays demonstrate that MSCs suppress the activation and function of CD4+ T cells by secreting interleukin 11, and neutralization of IL11 abrogates MSC-mediated suppression of CD4+ T cell function. Moreover, delayed-type, exogenous supplementation of IL11 significantly suppressed IFNγ+ expression by Th1 cells. Th1 and CD8+ cells play central roles in T cell-mediated tissue damage. Using a murine model of hypersensitivity response to study T cell-mediated tissue damage, we show that silencing IL11 in MSCs significantly abates the capacity of MSCs to suppress the generation of IFNγ-secreting CD4+ and CD8+ cells, failing to prevent T cell-mediated tissue inflammation and tissue damage.


Asunto(s)
Linfocitos T CD8-positivos , Interferón gamma , Interleucina-11 , Células Madre Mesenquimatosas , Ratones Endogámicos C57BL , Células TH1 , Animales , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/inmunología , Células TH1/inmunología , Ratones , Interleucina-11/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Interferón gamma/metabolismo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Células Cultivadas , Femenino
16.
Aging Dis ; 15(3): 965-976, 2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38722791

RESUMEN

Emerging from several decades of extensive research, key genetic elements and biochemical mechanisms implicated in neuroinflammation have been delineated, contributing substantially to our understanding of neurodegenerative diseases (NDDs). In this minireview, we discuss data predominantly from the past three years, highlighting the pivotal roles and mechanisms of the two principal cell types implicated in neuroinflammation. The review also underscores the extended process of peripheral inflammation that predates symptomatic onset, the critical influence of neuroinflammation, and their dynamic interplay in the pathogenesis of NDDs. Confronting these complex challenges, we introduce compelling evidence supporting the use of mesenchymal stem cell-based cell-free therapy. This therapeutic strategy includes the regulation of microglia and astrocytes, modulation of peripheral nerve cell inflammation, and targeted anti-inflammatory interventions specifically designed for NDDs, while also discussing engineering and safety considerations. This innovative therapeutic approach intricately modulates the immune system across the peripheral and nervous systems, with an emphasis on achieving superior penetration and targeted delivery. The insights offered by this review have significant implications for the better understanding and management of neuroinflammation.


Asunto(s)
Células Madre Mesenquimatosas , Enfermedades Neurodegenerativas , Enfermedades Neuroinflamatorias , Animales , Humanos , Astrocitos/metabolismo , Inflamación/terapia , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/inmunología , Células Madre Mesenquimatosas/metabolismo , Microglía/metabolismo , Microglía/inmunología , Enfermedades Neurodegenerativas/terapia , Enfermedades Neurodegenerativas/inmunología , Enfermedades Neuroinflamatorias/terapia , Enfermedades Neuroinflamatorias/inmunología
17.
Stem Cell Res Ther ; 15(1): 154, 2024 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-38816862

RESUMEN

BACKGROUND: Mesenchymal stromal cells (MSCs) isolated from the periodontal ligament (hPDL-MSCs) have a high therapeutic potential, presumably due to their immunomodulatory properties. The interaction between hPDL-MSCs and immune cells is reciprocal and executed by diverse cytokine-triggered paracrine and direct cell-to-cell contact mechanisms. For the first time, this study aimed to directly compare the contribution of various mechanisms on this reciprocal interaction using different in vitro co-culture models at different inflammatory milieus. METHODS: Three co-culture models were used: indirect with 0.4 µm-pored insert, and direct with or without insert. After five days of co-culturing mitogen-activated CD4+ T lymphocytes with untreated, interleukin (IL)-1ß, or tumor necrosis factor (TNF)-α- treated hPDL-MSCs, the CD4+ T lymphocyte proliferation, viability, and cytokine secretion were investigated. The gene expression of soluble and membrane-bound immunomediators was investigated in the co-cultured hPDL-MSCs. RESULTS: Untreated hPDL-MSCs decreased the CD4+ T lymphocyte proliferation and viability more effectively in the direct co-culture models. The direct co-culture model without inserts showed a strikingly higher CD4+ T lymphocyte cell death rate. Adding IL-1ß to the co-culture models resulted in substantial CD4+ T lymphocyte response alterations, whereas adding TNF resulted in only moderate effects. The most changes in CD4+ T lymphocyte parameters upon the addition of IL-1ß or TNF-α in a direct co-culture model without insert were qualitatively different from those observed in two other models. Additionally, the co-culture models caused variability in the immunomediator gene expression in untreated and cytokine-triggered hPDL-MSCs. CONCLUSION: These results suggest that both paracrine and cell-to-cell contact mechanisms contribute to the reciprocal interaction between hPDL-MSCs and CD4+ T lymphocytes. The inflammatory environment affects each of these mechanisms, which depends on the type of cytokines used for the activation of MSCs' immunomodulatory activities. This fact should be considered by comparing the outcomes of the different models.


Asunto(s)
Linfocitos T CD4-Positivos , Técnicas de Cocultivo , Células Madre Mesenquimatosas , Comunicación Paracrina , Ligamento Periodontal , Humanos , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/inmunología , Ligamento Periodontal/citología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/inmunología , Inmunomodulación , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Comunicación Celular , Interleucina-1beta/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Citocinas/metabolismo
18.
Stem Cell Res Ther ; 15(1): 149, 2024 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-38783393

RESUMEN

BACKGROUND: Autoimmune uveitis is an inflammatory disease triggered by an aberrant immune response. Mesenchymal stem cell-derived small extracellular vesicles (MSC-sEVs) are emerging as potential therapeutic agents for this condition. CD73, an ectoenzyme present on MSC-sEVs, is involved in mitigating inflammation by converting extracellular adenosine monophosphate into adenosine. We hypothesize that the inhibitory effect of MSC-sEVs on experimental autoimmune uveitis (EAU) could be partially attributed to the surface expression of CD73. METHODS: To investigate novel therapeutic approaches for autoimmune uveitis, we performed lentiviral transduction to overexpress CD73 on the surface of MSC-sEVs, yielding CD73-enriched MSC-sEVs (sEVs-CD73). Mice with interphotoreceptor retinoid-binding protein (IRBP)-induced EAU were grouped randomly and treated with 50 µg MSC-sEVs, vector infected MSC-sEVs, sEVs-CD73 or PBS via single tail vein injection. We evaluated the clinical and histological features of the induced mice and analyzed the proportion and functional capabilities of T helper cells. Furthermore, T-cells were co-cultured with various MSC-sEVs in vitro, and we quantified the resulting inflammatory response to assess the potential therapeutic benefits of sEVs-CD73. RESULTS: Compared to MSC-sEVs, sEVs-CD73 significantly alleviates EAU, leading to reduced inflammation and diminished tissue damage. Treatment with sEVs-CD73 results in a decreased proportion of Th1 cells in the spleen, draining lymph nodes, and eyes, accompanied by an increased proportion of regulatory T-cells (Treg cells). In vitro assays further reveal that sEVs-CD73 inhibits T-cell proliferation, suppresses Th1 cells differentiation, and enhances Treg cells proportion. CONCLUSION: Over-expression of CD73 on MSC-sEVs enhances their immunosuppressive effects in EAU, indicating that sEVs-CD73 has the potential as an efficient immunotherapeutic agent for autoimmune uveitis.


Asunto(s)
5'-Nucleotidasa , Enfermedades Autoinmunes , Vesículas Extracelulares , Células Madre Mesenquimatosas , Uveítis , Animales , Uveítis/patología , Uveítis/terapia , Uveítis/metabolismo , Uveítis/inmunología , 5'-Nucleotidasa/metabolismo , 5'-Nucleotidasa/genética , Vesículas Extracelulares/metabolismo , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/inmunología , Ratones , Enfermedades Autoinmunes/terapia , Enfermedades Autoinmunes/patología , Enfermedades Autoinmunes/inmunología , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad , Femenino , Proteínas de Unión al Retinol , Humanos
19.
Int Immunopharmacol ; 134: 112171, 2024 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-38701539

RESUMEN

Prostate cancer represents the second most prevalent form of cancer found in males, and stands as the fifth primary contributor to cancer-induced mortality on a global scale. Research has shown that transplanted mesenchymal stem cells (MSCs) can migrate by homing to tumor sites in the body. In prostate cancer, researchers have explored the fact that MSC-based therapies (including genetically modified delivery vehicles or vectors) and MSC-derived exosomes are emerging as attractive options to improve the efficacy and safety of traditional cancer therapies. In addition, researchers have reported new insights into the application of extracellular vesicle (EV)-MSC therapy as a novel treatment option that could provide a more effective and targeted approach to prostate cancer treatment. Moreover, the new generation of exosomes, which contain biologically functional molecules as signal transducers between cells, can simultaneously deliver different therapeutic agents and induce an anti-tumor phenotype in immune cells and their recruitment to the tumor site. The results of the current research on the use of MSCs in the treatment of prostate cancer may be helpful to researchers and clinicians working in this field. Nevertheless, it is crucial to emphasize that although dual-role MSCs show promise as a therapeutic modality for managing prostate cancer, further investigation is imperative to comprehensively grasp their safety and effectiveness. Ongoing clinical trials are being conducted to assess the viability of MSCs in the management of prostate cancer. The results of these trials will help determine the viability of this approach. Based on the current literature, engineered MSCs-EV offer great potential for application in targeted tumor therapy.


Asunto(s)
Exosomas , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Neoplasias de la Próstata , Humanos , Masculino , Exosomas/metabolismo , Exosomas/trasplante , Neoplasias de la Próstata/terapia , Células Madre Mesenquimatosas/inmunología , Animales , Trasplante de Células Madre Mesenquimatosas/métodos
20.
Cells ; 13(10)2024 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-38786082

RESUMEN

Lung transplantation results are compromised by ischemia-reperfusion injury and alloimmune responses. Ex vivo lung perfusion (EVLP) is used to assess marginal donor lungs before transplantation but is also an excellent platform to apply novel therapeutics. We investigated donor lung immunomodulation using genetically engineered mesenchymal stromal cells with augmented production of human anti-inflammatory hIL-10 (MSCsIL-10). Pig lungs were placed on EVLP for 6 h and randomized to control (n = 7), intravascular delivery of 20 × 106 (n = 5, low dose) or 40 × 106 human MSCs IL-10 (n = 6, high dose). Subsequently, single-lung transplantation was performed, and recipient pigs were monitored for 3 days. hIL-10 secretion was measured during EVLP and after transplantation, and immunological effects were assessed by cytokine profile, T and myeloid cell characterization and mixed lymphocyte reaction. MSCIL-10 therapy rapidly increased hIL-10 during EVLP and resulted in transient hIL-10 elevation after lung transplantation. MSCIL-10 delivery did not affect lung function but was associated with dose-related immunomodulatory effects, with the low dose resulting in a beneficial decrease in apoptosis and lower macrophage activation, but the high MSCIL-10 dose resulting in inflammation and cytotoxic CD8+ T cell activation. MSCIL-10 therapy during EVLP results in a rapid and transient perioperative hIL-10 increase and has a therapeutic window for its immunomodulatory effects.


Asunto(s)
Inmunomodulación , Interleucina-10 , Trasplante de Pulmón , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Trasplante de Pulmón/métodos , Animales , Interleucina-10/metabolismo , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/inmunología , Células Madre Mesenquimatosas/citología , Porcinos , Trasplante de Células Madre Mesenquimatosas/métodos , Humanos , Ingeniería Genética , Pulmón/metabolismo , Pulmón/patología , Pulmón/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...