Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 304
Filtrar
1.
Cardiovasc Res ; 120(8): 839-854, 2024 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-38742656

RESUMEN

Cell-free tissue-engineered vascular grafts provide a promising alternative to treat cardiovascular disease, but timely endothelialization is essential for ensuring patency and proper functioning post-implantation. Recent studies from our lab showed that blood cells like monocytes (MCs) and macrophages (Mϕ) may contribute directly to cellularization and regeneration of bioengineered arteries in small and large animal models. While MCs and Mϕ are leucocytes that are part of the innate immune response, they share common developmental origins with endothelial cells (ECs) and are known to play crucial roles during vessel formation (angiogenesis) and vessel repair after inflammation/injury. They are highly plastic cells that polarize into pro-inflammatory and anti-inflammatory phenotypes upon exposure to cytokines and differentiate into other cell types, including EC-like cells, in the presence of appropriate chemical and mechanical stimuli. This review focuses on the developmental origins of MCs and ECs; the role of MCs and Mϕ in vessel repair/regeneration during inflammation/injury; and the role of chemical signalling and mechanical forces in Mϕ inflammation that mediates vascular graft regeneration. We postulate that comprehensive understanding of these mechanisms will better inform the development of strategies to coax MCs/Mϕ into endothelializing the lumen and regenerate the smooth muscle layers of cell-free bioengineered arteries and veins that are designed to treat cardiovascular diseases and perhaps the native vasculature as well.


Asunto(s)
Prótesis Vascular , Macrófagos , Monocitos , Regeneración , Ingeniería de Tejidos , Humanos , Monocitos/metabolismo , Monocitos/trasplante , Ingeniería de Tejidos/métodos , Animales , Macrófagos/metabolismo , Neovascularización Fisiológica , Fenotipo , Implantación de Prótesis Vascular/instrumentación , Implantación de Prótesis Vascular/efectos adversos , Células Endoteliales/metabolismo , Células Endoteliales/trasplante , Diseño de Prótesis , Mecanotransducción Celular
2.
JCI Insight ; 8(5)2023 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-36883565

RESUMEN

Gastrointestinal graft-versus-host disease (GvHD) is a major cause of mortality and morbidity following allogeneic bone marrow transplantation (allo-BMT). Chemerin is a chemotactic protein that recruits leukocytes to inflamed tissues by interacting with ChemR23/CMKLR1, a chemotactic receptor expressed by leukocytes, including macrophages. During acute GvHD, chemerin plasma levels were strongly increased in allo-BM-transplanted mice. The role of the chemerin/CMKLR1 axis in GvHD was investigated using Cmklr1-KO mice. WT mice transplanted with an allogeneic graft from Cmklr1-KO donors (t-KO) had worse survival and more severe GvHD. Histological analysis demonstrated that the gastrointestinal tract was the organ mostly affected by GvHD in t-KO mice. The severe colitis of t-KO mice was characterized by massive neutrophil infiltration and tissue damage associated with bacterial translocation and exacerbated inflammation. Similarly, Cmklr1-KO recipient mice showed increased intestinal pathology in both allogeneic transplant and dextran sulfate sodium-induced colitis. Notably, the adoptive transfer of WT monocytes into t-KO mice mitigated GvHD manifestations by decreasing gut inflammation and T cell activation. In patients, higher chemerin serum levels were predictive of GvHD development. Overall, these results suggest that CMKLR1/chemerin may be a protective pathway for the control of intestinal inflammation and tissue damage in GvHD.


Asunto(s)
Trasplante de Médula Ósea , Colitis , Enfermedad Injerto contra Huésped , Animales , Ratones , Traslado Adoptivo/métodos , Traslocación Bacteriana/genética , Traslocación Bacteriana/inmunología , Trasplante de Médula Ósea/efectos adversos , Quimiocinas/sangre , Quimiocinas/genética , Quimiocinas/inmunología , Colitis/sangre , Colitis/genética , Colitis/inmunología , Colitis/patología , Colitis/terapia , Enfermedad Injerto contra Huésped/sangre , Enfermedad Injerto contra Huésped/genética , Enfermedad Injerto contra Huésped/inmunología , Enfermedad Injerto contra Huésped/patología , Enfermedad Injerto contra Huésped/terapia , Inflamación/sangre , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Péptidos y Proteínas de Señalización Intercelular/sangre , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/inmunología , Monocitos/inmunología , Monocitos/trasplante , Infiltración Neutrófila/genética , Infiltración Neutrófila/inmunología , Receptores de Quimiocina/sangre , Receptores de Quimiocina/genética , Receptores de Quimiocina/inmunología , Trasplante Homólogo/efectos adversos
3.
J Immunol ; 208(3): 745-752, 2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-35031577

RESUMEN

Cystic fibrosis (CF) is an inherited life-threatening disease accompanied by repeated lung infections and multiorgan inflammation that affects tens of thousands of people worldwide. The causative gene, cystic fibrosis transmembrane conductance regulator (CFTR), is mutated in CF patients. CFTR functions in epithelial cells have traditionally been thought to cause the disease symptoms. Recent work has shown an additional defect: monocytes from CF patients show a deficiency in integrin activation and adhesion. Because monocytes play critical roles in controlling infections, defective monocyte function may contribute to CF progression. In this study, we demonstrate that monocytes from CFTRΔF508 mice (CF mice) show defective adhesion under flow. Transplanting CF mice with wild-type (WT) bone marrow after sublethal irradiation replaced most (60-80%) CF monocytes with WT monocytes, significantly improved survival, and reduced inflammation. WT/CF mixed bone marrow chimeras directly demonstrated defective CF monocyte recruitment to the bronchoalveolar lavage and the intestinal lamina propria in vivo. WT mice reconstituted with CF bone marrow also show lethality, suggesting that the CF defect in monocytes is not only necessary but also sufficient to cause disease. We also show that monocyte-specific knockout of CFTR retards weight gains and exacerbates dextran sulfate sodium-induced colitis. Our findings show that providing WT monocytes by bone marrow transfer rescues mortality in CF mice, suggesting that similar approaches may mitigate disease in CF patients.


Asunto(s)
Adhesión Celular/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Fibrosis Quística/terapia , Monocitos/inmunología , Monocitos/trasplante , Animales , Trasplante de Médula Ósea , Líquido del Lavado Bronquioalveolar/citología , Colitis/patología , Fibrosis Quística/patología , Integrinas/metabolismo , Mucosa Intestinal/citología , Mucosa Intestinal/inmunología , Ratones , Ratones Endogámicos C57BL
4.
Theranostics ; 12(2): 512-529, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34976198

RESUMEN

Rationale: Monocytes belong to the mononuclear phagocyte system and are immune responders to tissue injury and infection. There were also reports of monocytes transforming to microglia-like cells. Here we explore the roles of monocytes in microglia ontogeny and the pathogenesis of neonatal cerebral hypoxic-ischemic (HI) brain injury in mice. Methods: We used three genetic methods to track the development of monocytes, including CX3CR1GFP/+; CCR2RFP/+ reporter mice, adoptive transfer of GFP+ monocytes, and fate-mapping with CCR2-CreER mice, in neonatal mouse brains with or without lipopolysaccharide (LPS, 0.3 mg/kg)-sensitized Vannucci HI. We also used genetic (CCR2RFP/ RFP, CCR2 knockout) and pharmacological methods (RS102895, a CCR2 antagonist) to test the roles of monocytic influx in LPS/HI brain injury. Results: CCR2+ monocytes entered the late-embryonic brains via choroid plexus, but rapidly became CX3CR1+ amoeboid microglial cells (AMCs). The influx of CCR2+ monocytes declined after birth, but recurred after HI or LPS-sensitized HI (LPS/HI) brain injury, particularly in the hippocampus. The CCR2-CreER-based fate-mapping showed that CCR2+ monocytes became CD68+ TNFα+ macrophages within 4 d after LPS/HI, and maintained as TNFα+ MHCII+ macrophages or persisted as Tmem119+ Sall1+ P2RY12+ ramified microglia for at least five months after injury. Genetic deletion of the chemokine receptor CCR2 markedly diminished monocytic influx, the expression of pro- and anti-inflammatory cytokines, and brain damage. Post-LPS/HI application of RS102895 also reduced inflammatory responses and brain damage, leading to better cognitive functions. Conclusion: These results suggest that monocytes promote acute inflammatory responses and may become pathological microglia long after the neonatal LPS/HI insult. Further, blocking the influx of monocytes may be a potential therapy for neonatal brain injury.


Asunto(s)
Lesiones Encefálicas/patología , Hipoxia-Isquemia Encefálica/patología , Microglía/patología , Monocitos/inmunología , Enfermedades Neuroinflamatorias/patología , Traslado Adoptivo , Animales , Animales Recién Nacidos , Movimiento Celular , Células Cultivadas , Plexo Coroideo/citología , Plexo Coroideo/inmunología , Femenino , Inflamación/patología , Masculino , Ratones Endogámicos C57BL , Monocitos/trasplante , Enfermedades Neuroinflamatorias/inmunología , Receptores CCR2/genética , Receptores CCR2/metabolismo
5.
Arterioscler Thromb Vasc Biol ; 42(2): 175-188, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34879707

RESUMEN

OBJECTIVE: Monocytes, which play an important role in arteriogenesis, can build immunologic memory by a functional reprogramming that modifies their response to a second challenge. This process, called trained immunity, is evoked by insults that shift monocyte metabolism, increasing HIF (hypoxia-inducible factor)-1α levels. Since ischemia enhances HIF-1α, we evaluate whether ischemia can lead to a functional reprogramming of monocytes, which would contribute to arteriogenesis after hindlimb ischemia. METHODS AND RESULTS: Mice exposed to ischemia by 24 hours (24h) of femoral artery occlusion (24h trained) or sham were subjected to hindlimb ischemia one week later; the 24h trained mice showed significant improvement in blood flow recovery and arteriogenesis after hindlimb ischemia. Adoptive transfer using bone marrow-derived monocytes (BM-Mono) from 24h trained or sham donor mice, demonstrated that recipients subjected to hindlimb ischemia who received 24h ischemic-trained monocytes had remarkable blood flow recovery and arteriogenesis. Further, ischemic-trained BM-Mono had increased HIF-1α and GLUT-1 (glucose transporter-1) gene expression during femoral artery occlusion. Circulating cytokines and GLUT-1 were also upregulated during femoral artery occlusion.Transcriptomic analysis and confirmatory qPCR performed in 24h trained and sham BM-Mono revealed that among the 15 top differentially expressed genes, 4 were involved in lipid metabolism in the ischemic-trained monocytes. Lipidomic analysis confirmed that ischemia training altered the cholesterol metabolism of these monocytes. Further, several histone-modifying epigenetic enzymes measured by qPCR were altered in mouse BM-Mono exposed to 24h hypoxia. CONCLUSIONS: Ischemia training in BM-Mono leads to a unique gene profile and improves blood flow and arteriogenesis after hindlimb ischemia.


Asunto(s)
Traslado Adoptivo , Miembro Posterior/irrigación sanguínea , Isquemia/terapia , Monocitos/trasplante , Neovascularización Fisiológica , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Miembro Posterior/inmunología , Miembro Posterior/fisiopatología , Isquemia/inmunología , Isquemia/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Monocitos/inmunología
6.
Front Immunol ; 12: 783305, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34899748

RESUMEN

Cellular immunotherapies represent a promising approach for the treatment of cancer. Engineered adoptive cell therapies redirect and augment a leukocyte's inherent ability to mount an immune response by introducing novel anti-tumor capabilities and targeting moieties. A prominent example of this approach is the use of T cells engineered to express chimeric antigen receptors (CARs), which have demonstrated significant efficacy against some hematologic malignancies. Despite increasingly sophisticated strategies to harness immune cell function, efficacy against solid tumors has remained elusive for adoptive cell therapies. Amongst cell types used in immunotherapies, however, macrophages have recently emerged as prominent candidates for the treatment of solid tumors. In this review, we discuss the use of monocytes and macrophages as adoptive cell therapies. Macrophages are innate immune cells that are intrinsically equipped with broad therapeutic effector functions, including active trafficking to tumor sites, direct tumor phagocytosis, activation of the tumor microenvironment and professional antigen presentation. We focus on engineering strategies for manipulating macrophages, with a specific focus on CAR macrophages (CAR-M). We highlight CAR design for macrophages, the production of CAR-M for adoptive cell transfer, and clinical considerations for their use in treating solid malignancies. We then outline recent progress and results in applying CAR-M as immunotherapies. The recent development of engineered macrophage-based therapies holds promise as a key weapon in the immune cell therapy armamentarium.


Asunto(s)
Terapia Genética , Inmunoterapia Adoptiva , Macrófagos/trasplante , Monocitos/trasplante , Neoplasias/terapia , Receptores Quiméricos de Antígenos/genética , Animales , Humanos , Macrófagos/inmunología , Macrófagos/metabolismo , Monocitos/inmunología , Monocitos/metabolismo , Neoplasias/genética , Neoplasias/inmunología , Neoplasias/metabolismo , Fenotipo , Receptores Quiméricos de Antígenos/inmunología , Receptores Quiméricos de Antígenos/metabolismo , Microambiente Tumoral/inmunología
7.
Cardiovasc Res ; 117(5): 1358-1371, 2021 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-33038226

RESUMEN

AIMS: Prior studies have focused on the role of the kidney and vasculature in salt-induced modulation of blood pressure; however, recent data indicate that sodium accumulates in tissues and can activate immune cells. We sought to examine mechanisms by which salt causes activation of human monocytes both in vivo and in vitro. METHODS AND RESULTS: To study the effect of salt in human monocytes, monocytes were isolated from volunteers to perform several in vitro experiments. Exposure of human monocytes to elevated Na+ex vivo caused a co-ordinated response involving isolevuglandin (IsoLG)-adduct formation, acquisition of a dendritic cell (DC)-like morphology, expression of activation markers CD83 and CD16, and increased production of pro-inflammatory cytokines tumour necrosis factor-α, interleukin (IL)-6, and IL-1ß. High salt also caused a marked change in monocyte gene expression as detected by RNA sequencing and enhanced monocyte migration to the chemokine CC motif chemokine ligand 5. NADPH-oxidase inhibition attenuated monocyte activation and IsoLG-adduct formation. The increase in IsoLG-adducts correlated with risk factors including body mass index, pulse pressure. Monocytes exposed to high salt stimulated IL-17A production from autologous CD4+ and CD8+ T cells. In addition, to evaluate the effect of salt in vivo, monocytes and T cells isolated from humans were adoptively transferred to immunodeficient NSG mice. Salt feeding of humanized mice caused monocyte-dependent activation of human T cells reflected by proliferation and accumulation of T cells in the bone marrow. Moreover, we performed a cross-sectional study in 70 prehypertensive subjects. Blood was collected for flow cytometric analysis and 23Na magnetic resonance imaging was performed for tissue sodium measurements. Monocytes from humans with high skin Na+ exhibited increased IsoLG-adduct accumulation and CD83 expression. CONCLUSION: Human monocytes exhibit co-ordinated increases in parameters of activation, conversion to a DC-like phenotype and ability to activate T cells upon both in vitro and in vivo sodium exposure. The ability of monocytes to be activated by sodium is related to in vivo cardiovascular disease risk factors. We therefore propose that in addition to the kidney and vasculature, immune cells like monocytes convey salt-induced cardiovascular risk in humans.


Asunto(s)
Metabolismo de los Lípidos/efectos de los fármacos , Lípidos , Monocitos/efectos de los fármacos , NADPH Oxidasas/metabolismo , Cloruro de Sodio/farmacología , Traslado Adoptivo , Adulto , Anciano , Animales , Antígenos CD/metabolismo , Células Cultivadas , Técnicas de Cocultivo , Citocinas/metabolismo , Activación Enzimática , Femenino , Proteínas Ligadas a GPI/metabolismo , Humanos , Inmunoglobulinas/metabolismo , Mediadores de Inflamación/metabolismo , Activación de Linfocitos , Masculino , Glicoproteínas de Membrana/metabolismo , Ratones Transgénicos , Persona de Mediana Edad , Monocitos/enzimología , Monocitos/inmunología , Monocitos/trasplante , Fenotipo , Receptores de IgG/metabolismo , Cloruro de Sodio Dietético/farmacología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Antígeno CD83
8.
Pharmacol Res ; 161: 105094, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32795509

RESUMEN

Oncolytic virus (OV) immunotherapy has demonstrated to be a promising approach in cancer treatment due to tumor-specific oncolysis. However, their clinical use so far has been largely limited due to the lack of suitable delivery strategies with high efficacy. Direct 'intratumoral' injection is the way to cross the hurdles of systemic toxicity, while providing local effects. Progress in this field has enabled the development of alternative way using 'systemic' oncolytic virotherapy for producing better results. One major potential roadblock to systemic OV delivery is the low virus persistence in the face of hostile immune system. The delivery challenge is even greater when attempting to target the oncolytic viruses into the entire tumor mass, where not all tumor cells are equally exposed to exactly the same microenvironment. The microenvironment of many tumors is known to be massively infiltrated with various types of leucocytes in both primary and metastatic sites. Interestingly, this intratumoral immune cell heterogeneity exhibits a degree of organized distribution inside the tumor bed as evidenced, for example, by the hypoxic tumor microenviroment where predominantly recruits tumor-associated macrophages. Although in vivo OV delivery seems complicated and challenging, recent results are encouraging for decreasing the limitations of systemically administered oncolytic viruses and an improved efficiency of oncolytic viral therapy in targeting cancerous tissues in vitro. Here, we review the latest developments of carrier cell-based oncolytic virus delivery using tumor-infiltrating immune cells with a focus on the main features of each cellular vehicle.


Asunto(s)
Fibroblastos Asociados al Cáncer/virología , Células Asesinas Inducidas por Citocinas/virología , Inmunoterapia Adoptiva , Linfocitos Infiltrantes de Tumor/virología , Monocitos/virología , Neoplasias/terapia , Viroterapia Oncolítica , Virus Oncolíticos/patogenicidad , Linfocitos T/virología , Animales , Fibroblastos Asociados al Cáncer/inmunología , Fibroblastos Asociados al Cáncer/trasplante , Células Asesinas Inducidas por Citocinas/inmunología , Células Asesinas Inducidas por Citocinas/trasplante , Humanos , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/trasplante , Monocitos/inmunología , Monocitos/trasplante , Neoplasias/inmunología , Neoplasias/patología , Neoplasias/virología , Células Madre Neoplásicas/inmunología , Células Madre Neoplásicas/patología , Células Madre Neoplásicas/virología , Virus Oncolíticos/genética , Virus Oncolíticos/inmunología , Fenotipo , Linfocitos T/inmunología , Linfocitos T/trasplante , Hipoxia Tumoral , Microambiente Tumoral
9.
Eur J Histochem ; 64(s2)2020 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-32705858

RESUMEN

After the facial nerve axotomy (FNA), the distal end of the axon would gradually decay and disappear. Accumulated evidence shows that transplantation of bone marrow mesenchymal stem cells (BMSCs) reveals potential in the treatment of nervous system diseases or injuries. This study is aimed at investigating the therapeutic effects of co-transplantation of BMSCs and monocytes in FNA. We found that co-culture significantly elevated the CD4+/CD8+ ratio and CD4+ CD25+ T cell proportion compared with monocytes transplantation, and enhanced the differentiation of BMSCs into neurons. After the cell transplantation, the lowest apoptosis in the facial nerve nucleus was found in the co-transplantation group 2 (BMSCs:monocytes= 1:30). Moreover, the lowest expression levels of pro-inflammatory cytokines and the highest expression levels of anti-inflammatory cytokines were observed in the co-transplantation group 2 (BMSCs: monocytes= 1:30). The highest expression levels of protein in the JAK/STAT6 pathway and the SDF-1/CXCR4 axis were found in the co-transplantation group 2. BMSC/monocyte co-transplantation significantly improves the microenvironment in the facial nerve nucleus in FNA rats; therefore these findings suggest that it could promote the anti-/pro-inflammatory balance shift towards the anti-inflammatory microenvironment, alleviating survival conditions for BMSCs, regulating BMSC the chemotaxis homing, differentiation, and the section of BMSCs, and finally reducing the neuronal apoptosis. These findings might provide essential evidence for the in-hospital treatment of FNA with co-transplantation of BMSCs and monocytes.


Asunto(s)
Axotomía/efectos adversos , Traumatismos del Nervio Facial/terapia , Nervio Facial/cirugía , Núcleo Motor del Nervio Facial/citología , Trasplante de Células Madre Mesenquimatosas , Monocitos/trasplante , Animales , Caspasa 3/metabolismo , Diferenciación Celular/fisiología , Microambiente Celular/fisiología , Técnicas de Cocultivo , Masculino , Células Madre Mesenquimatosas/metabolismo , Monocitos/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Ratas Sprague-Dawley , Transducción de Señal/fisiología
11.
Cytotherapy ; 22(1): 35-43, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31902660

RESUMEN

BACKGROUND: Clinical studies have shown the efficacy of combination therapy for various malignancies. In this study, the characteristics, safety and feasibility of use of cascade-primed (CAPRI) cells for the combination treatment of non-small-cell lung cancer (NSCLC) were evaluated both in vitro and in vivo. METHODS: Sixty-five patients with stage II-IV NSCLC were recruited. Of these patients, 31 patients received CAPRI cell therapy combined with chemotherapy (CAPRI group), and the other 34 patients constituted the control group and received chemotherapy alone. This study primarily aimed to evaluate the overall survival (OS), progression-free survival (PFS), short-term responses and treatment efficacy. RESULTS: CD83, CD1a, CD80 and CD86 marker levels were significantly upregulated in CAPRI cells. Interferon-γ expression levels were highest in CD3+CD8+ cells (33.77% ± 4.40%). Furthermore, interleukin-2 levels were highest in CD3+CD56+ cells (26.73% ± 6.63%), whereas perforin expression levels were similar in CD3+CD8+ and CD3+CD56+ cells. Furthermore, CAPRI cells had a better anti-tumor potential in CD3+CD56+ cells and displayed the highest expression levels of CD107a to H460 and A549 cell lines. The 5-year OS was significantly greater in the CAPRI group than in the control group (P = 0.008), and the PFS of two groups exhibited a significant difference (P = 0.007). Median OS (48 versus 31.6 months; P = 0.004) and PFS (48 versus 36.4 months; P = 0.016) differed between these two groups. Moreover, treatment-associated toxicities were mild and well-tolerated by patients with NSCLC. CONCLUSION: CAPRI cell therapy potentially prolongs the survival of patients with NSCLC when combined with chemotherapy.


Asunto(s)
Antineoplásicos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/terapia , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Inmunoterapia Adoptiva/métodos , Neoplasias Pulmonares/terapia , Adyuvantes Inmunológicos/uso terapéutico , Adulto , Linfocitos T CD4-Positivos/trasplante , Linfocitos T CD8-positivos/trasplante , Carcinoma de Pulmón de Células no Pequeñas/mortalidad , Carcinoma de Pulmón de Células no Pequeñas/patología , Tratamiento Basado en Trasplante de Células y Tejidos/efectos adversos , Terapia Combinada , Células Dendríticas/trasplante , Femenino , Estudios de Seguimiento , Humanos , Interleucina-2/sangre , Neoplasias Pulmonares/mortalidad , Neoplasias Pulmonares/patología , Masculino , Persona de Mediana Edad , Monocitos/trasplante , Estadificación de Neoplasias , Supervivencia sin Progresión , Estudios Retrospectivos , Resultado del Tratamiento
12.
Trends Pharmacol Sci ; 41(1): 42-54, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31839374

RESUMEN

Nanoparticulate carriers, often referred to as nanoparticles (NPs), represent an important pharmacological advance for drug protection and tissue-specific drug delivery. Accessing the central nervous system (CNS), however, is a complex process regulated by mainly three brain barriers. While some leukocyte (i.e., immune cell) subsets are equipped with the adequate molecular machinery to infiltrate the CNS in physiological and/or pathological contexts, the successful delivery of NPs into the CNS remains hindered by the tightness of the brain barriers. Here, we present an overview of the three major brain barriers and the mechanisms allowing leukocytes to migrate across each of them. We subsequently review different immune-inspired and -mediated strategies to deliver NPs into the CNS. Finally, we discuss the prospect of exploiting leukocyte trafficking mechanisms for further progress.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Sistemas de Liberación de Medicamentos/métodos , Macrófagos/trasplante , Monocitos/trasplante , Nanopartículas/administración & dosificación , Animales , Barrera Hematoencefálica/inmunología , Encéfalo/irrigación sanguínea , Encéfalo/metabolismo , Movimiento Celular , Sistema Nervioso Central/metabolismo , Humanos , Leucocitos/química , Leucocitos/citología , Macrófagos/inmunología , Monocitos/inmunología , Nanopartículas/química , Nanopartículas/metabolismo
13.
Stem Cell Rev Rep ; 16(1): 198-211, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31828580

RESUMEN

BACKGROUND: Bone marrow-derived mononuclear cells (BM-MNC) consist of a heterogeneous mix of mesenchymal stem cells (MSC), hematopoietic progenitor cells (HPC), endothelial progenitor cells (EPC), monocytes, lymphocytes and pluripotent stem cells. Whereas the importance of MSC and EPC has been well documented in bone healing and regeneration studies, the role of pluripotent stem cells is still poorly understood. In the present study we evaluated if and how Very Small Embryonic Like cells (VSEL), isolated from rat BM-MNC, contribute to bone healing. METHODS: Large bone defects were made in the femurs of 38 Sprague Dawley female rats and treated with ß-TCP scaffold granules seeded with male VSEL; BM-MNC, VSEL-depleted BM-MNC or scaffold alone, and bone healing was evaluated at 8 weeks post-surgery. RESULTS: Bone healing was significantly increased in defects treated with VSEL and BM-MNC, compared to defects treated with VSEL-depleted BM-MNC. Donor cells were detected in new bone tissue, in all the defects treated with cells, and in fibrous tissue only in defects treated with VSEL-depleted BM-MNC. The number of CD68+ cells was the highest in the VSEL-depleted group, whereas the number of TRAP positive cells was the lowest in this group. CONCLUSIONS: Based on the results, we can conclude that VSEL play a role in BM-MNC induced bone formation. In our rat femur defect model, in defects treated with VSEL-depleted BM-MNC, osteoclastogenesis and bone formation were decreased, and foreign body reaction was increased.


Asunto(s)
Células Madre Adultas/trasplante , Regeneración Ósea/genética , Trasplante de Células Madre Mesenquimatosas , Células Madre Pluripotentes/trasplante , Adulto , Animales , Células Progenitoras Endoteliales/trasplante , Humanos , Monocitos/trasplante , Osteogénesis/genética , Ratas
14.
Am J Hypertens ; 33(1): 31-42, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31328772

RESUMEN

BACKGROUND: Essential hypertension is associated with increased plasma concentrations of extracellular vesicles (EVs). We aimed to determine the role of monocyte miR-27a in EVs on arterial Mas receptor expression, and its involvement in the pathogenesis of hypertension. METHODS: THP-1 cells were transfected with miR-27a mimic and miR-27a inhibitor, and EVs were collected. Mas receptor expression and endothelial nitric oxide synthase (eNOS) phosphorylation were determined by immunoblotting. Sprague-Dawley (SD) rats received EVs via tail-vein injection. Blood pressure (BP) was measured with the tail-cuff method. The vasodilatory response of mesenteric arteries was measured using a small vessel myograph. RESULTS: EVs from THP-1 cells increased rat BP by impairing Ang-(1-7)-mediated vasodilation in mesenteric arteries, which was further exaggerated by EVs from lipopolysaccharides-treated THP-1 cells. As the receptor and key signaling of Ang-(1-7), next experiments found that Mas receptor expression and eNOS phosphorylation were decreased in mesenteric arteries from EVs-treated SD rats. Screening studies found miR-27a in EVs may be involved in this process. Through transfection with miR-27a inhibitor or miR-27a mimic, we found that miR-27a downregulates Mas receptor expression in endothelial cells. Injection of EVs from miR-27a-transfected HEK-293 cells decreased Mas receptor and eNOS phosphorylation in mesenteric arteries, impaired Ang-(1-7)-mediated vasodilation and increased BP. Earlier effects were reversed using cells with downregulation of miR-27 in EVs. CONCLUSIONS: Monocyte miR-27a in EVs decreases Mas receptor expression and eNOS phosphorylation in endothelium, impairs Ang-(1-7)-mediated vasodilation, and causes hypertension. Understanding the contributions of EVs in the pathogenesis of hypertension may facilitate their use as a diagnostic biomarker.


Asunto(s)
Presión Sanguínea , Vesículas Extracelulares/metabolismo , Hipertensión/enzimología , Arterias Mesentéricas/enzimología , MicroARNs/metabolismo , Monocitos/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Modelos Animales de Enfermedad , Vesículas Extracelulares/genética , Vesículas Extracelulares/trasplante , Células HEK293 , Humanos , Hipertensión/genética , Hipertensión/fisiopatología , Masculino , Arterias Mesentéricas/fisiopatología , MicroARNs/genética , Monocitos/trasplante , Fosforilación , Proto-Oncogenes Mas , Ratas Sprague-Dawley , Transducción de Señal , Células THP-1 , Vasodilatación
15.
J Clin Invest ; 130(2): 774-788, 2020 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-31661470

RESUMEN

Efficacy of dendritic cell (DC) cancer vaccines is classically thought to depend on their antigen-presenting cell (APC) activity. Studies show, however, that DC vaccine priming of cytotoxic T lymphocytes (CTLs) requires the activity of endogenous DCs, suggesting that exogenous DCs stimulate antitumor immunity by transferring antigens (Ags) to endogenous DCs. Such Ag transfer functions are most commonly ascribed to monocytes, implying that undifferentiated monocytes would function equally well as a vaccine modality and need not be differentiated to DCs to be effective. Here, we used several murine cancer models to test the antitumor efficacy of undifferentiated monocytes loaded with protein or peptide Ag. Intravenously injected monocytes displayed antitumor activity superior to DC vaccines in several cancer models, including aggressive intracranial glioblastoma. Ag-loaded monocytes induced robust CTL responses via Ag transfer to splenic CD8+ DCs in a manner independent of monocyte APC activity. Ag transfer required cell-cell contact and the formation of connexin 43-containing gap junctions between monocytes and DCs. These findings demonstrate the existence of an efficient gap junction-mediated Ag transfer pathway between monocytes and CD8+ DCs and suggest that administration of tumor Ag-loaded undifferentiated monocytes may serve as a simple and efficacious immunotherapy for the treatment of human cancers.


Asunto(s)
Antígenos de Neoplasias/inmunología , Linfocitos T CD8-positivos/inmunología , Inmunidad Celular , Inmunoterapia , Monocitos , Neoplasias Experimentales , Animales , Ratones , Ratones Noqueados , Monocitos/inmunología , Monocitos/trasplante , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/terapia
16.
PLoS One ; 14(9): e0222912, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31569199

RESUMEN

To confirm that neoplastic monocyte-derived collagen- and fibronectin-producing fibrocytes induce bone marrow (BM) fibrosis in primary myelofibrosis (PMF), we injected PMF BM-derived fibrocyte-precursor CD14+/CD34- monocytes into the tail vein of NOD-SCID-γ (NSG) mice. PMF BM-derived CD14+/CD34- monocytes engrafted and induced a PMF-like phenotype with splenomegaly, myeloid hyperplasia with clusters of atypical megakaryocytes, persistence of the JAK2V617F mutation, and BM and spleen fibrosis. As control we used normal human BM-derived CD14+/CD34- monocytes. These monocytes also engrafted and gave rise to normal megakaryocytes that, like PMF CD14+/CD34--derived megakaryocytes, expressed HLA-ABC and human CD42b antigens. Using 2 clonogenic assays we confirmed that PMF and normal BM-derived CD14+/CD34- monocytes give rise to megakaryocyte colony-forming cells, suggesting that a subpopulation BM monocytes harbors megakaryocyte progenitor capacity. Taken together, our data suggest that PMF monocytes induce myelofibrosis-like phenotype in immunodeficient mice and that PMF and normal BM-derived CD14+/CD34- monocytes give rise to megakaryocyte progenitor cells.


Asunto(s)
Células de la Médula Ósea/inmunología , Fibroblastos/inmunología , Hiperplasia/inmunología , Huésped Inmunocomprometido , Monocitos/inmunología , Mielofibrosis Primaria/inmunología , Esplenomegalia/inmunología , Traslado Adoptivo , Animales , Antígenos CD34/genética , Antígenos CD34/inmunología , Células de la Médula Ósea/patología , Femenino , Fibroblastos/patología , Fibroblastos/trasplante , Expresión Génica , Antígenos HLA/genética , Antígenos HLA/inmunología , Humanos , Hiperplasia/etiología , Hiperplasia/genética , Hiperplasia/patología , Janus Quinasa 2/genética , Janus Quinasa 2/inmunología , Receptores de Lipopolisacáridos/genética , Receptores de Lipopolisacáridos/inmunología , Megacariocitos/inmunología , Megacariocitos/patología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Monocitos/patología , Monocitos/trasplante , Mutación , Mielofibrosis Primaria/etiología , Mielofibrosis Primaria/genética , Mielofibrosis Primaria/patología , Esplenomegalia/etiología , Esplenomegalia/genética , Esplenomegalia/patología
17.
Sci Rep ; 9(1): 7711, 2019 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-31118440

RESUMEN

Critical limb ischemia (CLI) is associated with a high risk of limb amputation. It has been shown that cell therapy is safe and has beneficial effects on ischemic clinical symptoms in patients with CLI. The aim of this study was to further investigate the outcomes of intramuscular injection of autologous bone-marrow mononuclear cells (BM-MNCs) in a long-term follow-up period in atherosclerotic peripheral arterial disease (PAD) patients who have no optional therapy. This study was a retrospective and observational study that was carried out to evaluate long-term clinical outcomes in 42 lower limbs of 30 patients with atherosclerotic PAD who underwent BM-MNC implantation. The median follow-up period was 9.25 (range, 6-16) years. The overall amputation-free rates were 73.0% at 5 years after BM-MNC implantation and 70.4% at 10 years in patients with atherosclerotic PAD. The overall amputation-free rates at 5 years and at 10 years after implantation of BM-MNCs were significantly higher in atherosclerotic PAD patients than in internal controls and historical controls. There were no significant differences in amputation rates between the internal control group and historical control group. The rate of overall survival was not significantly different between the BM-MNC implantation group and the historical control group. Implantation of autologous BM-MNCs is feasible for a long-term follow-up period in patients with CLI who have no optional therapy.


Asunto(s)
Trasplante de Médula Ósea , Isquemia/terapia , Pierna/irrigación sanguínea , Monocitos/trasplante , Anciano , Anciano de 80 o más Años , Amputación Quirúrgica , Comorbilidad , Femenino , Estudios de Seguimiento , Humanos , Inyecciones Intramusculares , Isquemia/etiología , Isquemia/cirugía , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Enfermedad Arterial Periférica/complicaciones , Supervivencia sin Progresión , Estudios Retrospectivos , Trasplante Autólogo , Resultado del Tratamiento
18.
World J Gastroenterol ; 25(13): 1560-1565, 2019 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-30983816

RESUMEN

Cellular therapy may be the solution of challenging problems in colorectal surgery such as impaired healing leading to anastomotic leakage and metastatic colorectal cancer (CRC). This review aimed to illustrate the role of cellular therapy in promotion of wound healing and management of metastatic CRC. An organized literature search for the role of cellular therapy in promotion of wound healing and management of metastatic CRC was conducted. Electronic databases including PubMed/Medline, Scopus, and Embase were queried for the search process. Two types of cellular therapy have been recognized, the mesenchymal stem cells (MSCs) and bone marrow-mononuclear cells (BM-MNCs) therapy. These cells have been shown to accelerate and promote healing of various tissue injuries in animal and human studies. In addition, experimental studies have reported that MSCs may help suppress the progression of colon cancer in rat models. This article reviews the possible mechanisms of action and clinical utility of MSCs and BM-MNCs in promotion of healing and suppression of tumor growth in light of the published literature. Cellular therapy has a potentially important role in colorectal surgery, particularly in the promotion of wound healing and management of metastatic CRC. Future directions of cellular therapy in colorectal surgery were explored which may help stimulate futures studies on the role of cellular therapy in colorectal surgery.


Asunto(s)
Neoplasias Colorrectales/terapia , Trasplante de Células Madre Mesenquimatosas , Monocitos/trasplante , Complicaciones Posoperatorias/terapia , Cicatrización de Heridas , Animales , Colectomía/efectos adversos , Colon/patología , Colon/cirugía , Neoplasias Colorrectales/patología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Humanos , Complicaciones Posoperatorias/etiología , Proctectomía/efectos adversos , Ratas , Recto/patología , Recto/cirugía , Resultado del Tratamiento
19.
Sci Rep ; 9(1): 6434, 2019 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-31015492

RESUMEN

Liver diseases are a major health problem worldwide leading to high mortality rates and causing a considerable economic burden in many countries. Cellular therapies as potential treatments for liver diseases have proven beneficial in most of the conditions. In recent years, studies involving therapy with bone marrow cells have been implemented to promote liver regeneration and to reduce hepatic fibrosis, however identifying the cell population present in the bone marrow that is responsible for hepatic improvement after therapy is still necessary. The aim of the present study was the evaluation of the therapeutic efficacy of monocytes obtained from bone marrow in fibrosis resulting from S. mansoni infection in C57BL/6 mice. Monocytes were isolated by immunomagnetic separation and administered to the infected animals. The effects of treatment were evaluated through morphometric, biochemical, immunological and molecular analyzes. Monocyte therapy promoted reduction of liver fibrosis induced by S. mansoni infection, associated with a decrease in production of inflammatory and pro-fibrogenic mediators. In addition, monocyte infusion caused downregulation of factors associated with the M1 activation profile, as well as upregulation of M2reg markers. The findings altogether reinforce the hypothesis that the predominance of M2reg macrophages, producers of immunosuppressive cytokines, may favor the improvement of hepatic fibrosis in a preclinical model, through fibrous tissue remodeling, modulation of the inflammatory response and fibrogenesis.


Asunto(s)
Traslado Adoptivo/métodos , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Cirrosis Hepática/terapia , Regeneración Hepática , Monocitos/trasplante , Schistosoma mansoni/patogenicidad , Esquistosomiasis mansoni/terapia , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/inmunología , Diferenciación Celular , Citocinas/genética , Citocinas/inmunología , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Hígado/inmunología , Hígado/parasitología , Hígado/patología , Cirrosis Hepática/inmunología , Cirrosis Hepática/parasitología , Cirrosis Hepática/patología , Macrófagos/citología , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Monocitos/citología , Monocitos/inmunología , Schistosoma mansoni/crecimiento & desarrollo , Esquistosomiasis mansoni/inmunología , Esquistosomiasis mansoni/parasitología , Esquistosomiasis mansoni/patología
20.
Cytotherapy ; 21(4): 393-415, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30871899

RESUMEN

Cell-based therapies are a rapidly developing area of regenerative medicine as dynamic treatments that execute therapeutic functions multimodally. Monocytes and macrophages, as innate immune cells that control inflammation and tissue repair, are increasing popular clinical candidates due to their spectrum of functionality. In this article, we review the role of monocytes and macrophages specifically in inflammatory and degenerative disease pathology and the evidence supporting the use of these cells as an effective therapeutic strategy. We compare current strategies of exogenously polarized monocyte/macrophage therapies regarding dosage, delivery and processing to identify outcomes, advances and challenges to their clinical use. Monocytes/macrophages hold the potential to be a promising therapeutic avenue but understanding and optimization of disease-specific efficacy is needed to accelerate their clinical use.


Asunto(s)
Enfermedad , Inflamación/terapia , Macrófagos/trasplante , Monocitos/trasplante , Animales , Humanos , Inflamación/patología , Medicina Regenerativa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA