Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 304
Filtrar
1.
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
2.
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
3.
Nature ; 541(7635): 96-101, 2017 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-28002407

RESUMEN

Monocytes and macrophages comprise a variety of subsets with diverse functions. It is thought that these cells play a crucial role in homeostasis of peripheral organs, key immunological processes and development of various diseases. Among these diseases, fibrosis is a life-threatening disease of unknown aetiology. Its pathogenesis is poorly understood, and there are few effective therapies. The development of fibrosis is associated with activation of monocytes and macrophages. However, the specific subtypes of monocytes and macrophages that are involved in fibrosis have not yet been identified. Here we show that Ceacam1+Msr1+Ly6C-F4/80-Mac1+ monocytes, which we term segregated-nucleus-containing atypical monocytes (SatM), share granulocyte characteristics, are regulated by CCAAT/enhancer binding protein ß (C/EBPß), and are critical for fibrosis. Cebpb deficiency results in a complete lack of SatM. Furthermore, the development of bleomycin-induced fibrosis, but not inflammation, was prevented in chimaeric mice with Cebpb-/- haematopoietic cells. Adoptive transfer of SatM into Cebpb-/- mice resulted in fibrosis. Notably, SatM are derived from Ly6C-FcεRI+ granulocyte/macrophage progenitors, and a newly identified SatM progenitor downstream of Ly6C-FcεRI+ granulocyte/macrophage progenitors, but not from macrophage/dendritic-cell progenitors. Our results show that SatM are critical for fibrosis and that C/EBPß licenses differentiation of SatM from their committed progenitor.


Asunto(s)
Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Células Progenitoras de Granulocitos y Macrófagos/citología , Monocitos/clasificación , Monocitos/metabolismo , Fibrosis Pulmonar/patología , Traslado Adoptivo , Animales , Antígenos CD/metabolismo , Antígenos Ly/metabolismo , Biomarcadores/metabolismo , Bleomicina/toxicidad , Proteína beta Potenciadora de Unión a CCAAT/deficiencia , Proteína beta Potenciadora de Unión a CCAAT/genética , Moléculas de Adhesión Celular/metabolismo , Diferenciación Celular , Células Dendríticas/citología , Modelos Animales de Enfermedad , Células Progenitoras de Granulocitos y Macrófagos/metabolismo , Granulocitos/citología , Granulocitos/metabolismo , Inflamación , Masculino , Ratones , Terapia Molecular Dirigida/tendencias , Monocitos/patología , Monocitos/trasplante , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/genética , Fibrosis Pulmonar/prevención & control , Receptores de IgE/metabolismo , Receptores Depuradores de Clase A/metabolismo
4.
Immunity ; 38(2): 322-35, 2013 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-23352232

RESUMEN

Dendritic cells (DCs) are crucial for mounting allergic airway inflammation, but it is unclear which subset of DCs performs this task. By using CD64 and MAR-1 staining, we reliably separated CD11b(+) monocyte-derived DCs (moDCs) from conventional DCs (cDCs) and studied antigen uptake, migration, and presentation assays of lung and lymph node (LN) DCs in response to inhaled house dust mite (HDM). Mainly CD11b(+) cDCs but not CD103(+) cDCs induced T helper 2 (Th2) cell immunity in HDM-specific T cells in vitro and asthma in vivo. Studies in Flt3l(-/-) mice, lacking all cDCs, revealed that moDCs were also sufficient to induce Th2 cell-mediated immunity but only when high-dose HDM was given. The main function of moDCs was the production of proinflammatory chemokines and allergen presentation in the lung during challenge. Thus, we have identified migratory CD11b(+) cDCs as the principal subset inducing Th2 cell-mediated immunity in the LN, whereas moDCs orchestrate allergic inflammation in the lung.


Asunto(s)
Alérgenos/inmunología , Antígenos Dermatofagoides/inmunología , Asma/inmunología , Células Dendríticas/inmunología , Inmunidad Celular , Pyroglyphidae/inmunología , Células Th2/inmunología , Administración por Inhalación , Traslado Adoptivo , Alérgenos/aislamiento & purificación , Animales , Antígenos Dermatofagoides/administración & dosificación , Antígenos Dermatofagoides/aislamiento & purificación , Antígenos Ly/genética , Antígenos Ly/inmunología , Asma/patología , Antígeno CD11b/genética , Antígeno CD11b/inmunología , Movimiento Celular , Proliferación Celular , Células Dendríticas/trasplante , Expresión Génica , Inflamación/inmunología , Inflamación/patología , Pulmón/inmunología , Pulmón/patología , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/patología , Ratones , Ratones Transgénicos , Monocitos/inmunología , Monocitos/trasplante , Receptor 1 Gatillante de la Citotoxidad Natural/genética , Receptor 1 Gatillante de la Citotoxidad Natural/inmunología , Especificidad de Órganos , Receptores de IgG/genética , Receptores de IgG/inmunología
5.
Immunity ; 36(6): 1031-46, 2012 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-22749353

RESUMEN

GM-CSF (Csf-2) is a critical cytokine for the in vitro generation of dendritic cells (DCs) and is thought to control the development of inflammatory DCs and resident CD103(+) DCs in some tissues. Here we showed that in contrast to the current understanding, Csf-2 receptor acts in the steady state to promote the survival and homeostasis of nonlymphoid tissue-resident CD103(+) and CD11b(+) DCs. Absence of Csf-2 receptor on lung DCs abrogated the induction of CD8(+) T cell immunity after immunization with particulate antigens. In contrast, Csf-2 receptor was dispensable for the differentiation and innate function of inflammatory DCs during acute injuries. Instead, inflammatory DCs required Csf-1 receptor for their development. Thus, Csf-2 is important in vaccine-induced CD8(+) T cell immunity through the regulation of nonlymphoid tissue DC homeostasis rather than control of inflammatory DCs in vivo.


Asunto(s)
Subunidad beta Común de los Receptores de Citocinas/fisiología , Células Dendríticas/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/fisiología , Inflamación/inmunología , Animales , Linfocitos T CD8-positivos/inmunología , Diferenciación Celular , Linaje de la Célula , Subunidad beta Común de los Receptores de Citocinas/antagonistas & inhibidores , Subunidad beta Común de los Receptores de Citocinas/deficiencia , Subunidad beta Común de los Receptores de Citocinas/genética , Células Dendríticas/clasificación , Células Dendríticas/citología , Encefalomielitis Autoinmune Experimental/inmunología , Endotoxemia/inmunología , Perfilación de la Expresión Génica , Factor Estimulante de Colonias de Granulocitos y Macrófagos/deficiencia , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Homeostasis , Lipopolisacáridos/toxicidad , Listeriosis/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/trasplante , Especificidad de Órganos , Infecciones por Orthomyxoviridae/inmunología , Infecciones Neumocócicas/inmunología , Quimera por Radiación , Bazo/inmunología , Tamoxifeno/farmacología
6.
Immunity ; 36(6): 1047-59, 2012 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-22749354

RESUMEN

Dendritic cells (DCs), monocytes, and/or macrophages initiate host-protective immune responses to intracellular pathogens in part through interleukin-12 (IL-12) production, although the relative contribution of tissue resident versus recruited cells has been unclear. Here, we showed that after intraperitoneal infection with Toxoplasma gondii cysts, resident mononuclear phagocytes are replaced by circulating monocytes that differentiate in situ into inflammatory DCs (moDCs) and F4/80(+) macrophages. Importantly, NK cell-derived interferon-γ (IFN-γ) was required for both the loss of resident mononuclear phagocytes and the local differentiation of monocytes into macrophages and moDCs. This newly generated moDC population and not the resident DCs (or macrophages) served as the major source of IL-12 at the site of infection. Thus, NK cell-derived IFN-γ is important in both regulating inflammatory cell dynamics and in driving the local differentiation of monocytes into the cells required for initiating the immune response to an important intracellular pathogen.


Asunto(s)
Células Dendríticas/inmunología , Interferón gamma/fisiología , Células Asesinas Naturales/inmunología , Monocitos/inmunología , Traslado Adoptivo , Animales , Antígenos Ly/análisis , Diferenciación Celular , Quimiotaxis de Leucocito , Células Dendríticas/patología , Células Dendríticas/trasplante , Genes Reporteros , Subunidad p40 de la Interleucina-12/biosíntesis , Subunidad p40 de la Interleucina-12/genética , Células Asesinas Naturales/metabolismo , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/trasplante , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Monocitos/química , Monocitos/patología , Monocitos/trasplante , Factor 88 de Diferenciación Mieloide/fisiología , Neutrófilos/inmunología , Peritonitis/inmunología , Peritonitis/parasitología , Fagocitos/clasificación , Fagocitos/inmunología , Fagocitos/patología , Receptores de Interferón/deficiencia , Receptores de Interferón/fisiología , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/genética , Organismos Libres de Patógenos Específicos , Subgrupos de Linfocitos T/inmunología , Toxoplasmosis Animal/inmunología , Receptor de Interferón gamma
7.
Circulation ; 139(5): 647-659, 2019 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-30586712

RESUMEN

BACKGROUND: The impact of gut microbiota on the regulation of host physiology has recently garnered considerable attention, particularly in key areas such as the immune system and metabolism. These areas are also crucial for the pathophysiology of and repair after myocardial infarction (MI). However, the role of the gut microbiota in the context of MI remains to be fully elucidated. METHODS: To investigate the effects of gut microbiota on cardiac repair after MI, C57BL/6J mice were treated with antibiotics 7 days before MI to deplete mouse gut microbiota. Flow cytometry was applied to examine the changes in immune cell composition in the heart. 16S rDNA sequencing was conducted as a readout for changes in gut microbial composition. Short-chain fatty acid (SCFA) species altered after antibiotic treatment were identified by high-performance liquid chromatography. Fecal reconstitution, transplantation of monocytes, or dietary SCFA or Lactobacillus probiotic supplementation was conducted to evaluate the cardioprotective effects of microbiota on the mice after MI. RESULTS: Antibiotic-treated mice displayed drastic, dose-dependent mortality after MI. We observed an association between the gut microbiota depletion and significant reductions in the proportion of myeloid cells and SCFAs, more specifically acetate, butyrate, and propionate. Infiltration of CX3CR1+ monocytes to the peri-infarct zone after MI was also reduced, suggesting impairment of repair after MI. Accordingly, the physiological status and survival of mice were significantly improved after fecal reconstitution, transplantation of monocytes, or dietary SCFA supplementation. MI was associated with a reorganization of the gut microbial community such as a reduction in Lactobacillus. Supplementing antibiotic-treated mice with a Lactobacillus probiotic before MI restored myeloid cell proportions, yielded cardioprotective effects, and shifted the balance of SCFAs toward propionate. CONCLUSIONS: Gut microbiota-derived SCFAs play an important role in maintaining host immune composition and repair capacity after MI. This suggests that manipulation of these elements may provide opportunities to modulate pathological outcome after MI and indeed human health and disease as a whole.


Asunto(s)
Antibacterianos/toxicidad , Bacterias/efectos de los fármacos , Microbioma Gastrointestinal/efectos de los fármacos , Monocitos/inmunología , Infarto del Miocardio/microbiología , Miocardio/inmunología , Animales , Bacterias/inmunología , Bacterias/metabolismo , Modelos Animales de Enfermedad , Disbiosis , Ácidos Grasos/administración & dosificación , Ácidos Grasos/metabolismo , Trasplante de Microbiota Fecal , Femenino , Interacciones Huésped-Patógeno , Lactobacillus/inmunología , Lactobacillus/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Monocitos/metabolismo , Monocitos/trasplante , Infarto del Miocardio/inmunología , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Miocardio/metabolismo , Miocardio/patología , Probióticos/administración & dosificación , Células RAW 264.7
8.
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
9.
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
10.
J Neurosci ; 38(30): 6722-6736, 2018 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-29946039

RESUMEN

Exposure to low-dose lipopolysaccharide (LPS) before cerebral ischemia is neuroprotective in stroke models, a phenomenon termed preconditioning (PC). Although it is well established that LPS-PC induces central and peripheral immune responses, the cellular mechanisms modulating ischemic injury remain unclear. Here, we investigated the role of immune cells in the brain protection afforded by PC and tested whether monocytes may be reprogrammed by ex vivo LPS exposure, thus modulating inflammatory injury after cerebral ischemia in male mice. We found that systemic injection of low-dose LPS induces a Ly6Chi monocyte response that protects the brain after transient middle cerebral artery occlusion (MCAO) in mice. Remarkably, adoptive transfer of monocytes isolated from preconditioned mice into naive mice 7 h after transient MCAO reduced brain injury. Gene expression and functional studies showed that IL-10, inducible nitric oxide synthase, and CCR2 in monocytes are essential for neuroprotection. This protective activity was elicited even if mouse or human monocytes were exposed ex vivo to LPS and then injected into male mice after stroke. Cell-tracking studies showed that protective monocytes are mobilized from the spleen and reach the brain and meninges, where they suppress postischemic inflammation and neutrophil influx into the brain parenchyma. Our findings unveil a previously unrecognized subpopulation of splenic monocytes capable of protecting the brain with an extended therapeutic window and provide the rationale for cell therapies based on the delivery of autologous or allogeneic protective monocytes in patients after ischemic stroke.SIGNIFICANCE STATEMENT Inflammation is a key component of the pathophysiology of the brain in stroke, a leading cause of death and disability with limited therapeutic options. Here, we investigate endogenous mechanisms of protection against cerebral ischemia. Using lipopolysaccharide (LPS) preconditioning (PC) as an approach to induce ischemic tolerance in mice, we found generation of neuroprotective monocytes within the spleen, from which they traffic to the brain and meninges, suppressing postischemic inflammation. Importantly, systemic LPS-PC can be mimicked by adoptive transfer of in vitro-preconditioned mouse or human monocytes at translational relevant time points after stroke. This model of neuroprotection may facilitate clinical efforts to increase the efficacy of BM mononuclear cell treatments in acute neurological diseases such as cerebral ischemia.


Asunto(s)
Precondicionamiento Isquémico/métodos , Lipopolisacáridos/farmacología , Monocitos , Neuroprotección/inmunología , Accidente Cerebrovascular , Traslado Adoptivo , Animales , Isquemia Encefálica/inmunología , Isquemia Encefálica/patología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Monocitos/efectos de los fármacos , Monocitos/inmunología , Monocitos/trasplante , Accidente Cerebrovascular/inmunología , Accidente Cerebrovascular/patología
11.
Eur J Immunol ; 48(2): 250-257, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-28980305

RESUMEN

Viruses, particularly the Epstein-Barr virus (EBV) has long been suspected to exacerbate acute arthritic symptoms. However, the cell populations that contribute to import viruses into the inflamed tissues remain to be identified. In the present study, we have investigated the role of monocytes in the transport of Murid herpesvirus 68 (MHV-68), a mouse virus closely related to EBV, using the serum transfer-induced arthritis (STIA) model. We found compelling evidence that MHV-68 infection markedly increased disease severity in NR4A1-/- mice, which are deficient for Ly6Clow monocytes. In contrast, the MHV-68-induced enhancement of joint inflammation was lessened in CCR2-/- mice, suggesting the involvement of inflammatory Ly6Chigh monocytes in viral transport. We also observed that following selective depletion of monocyte subsets by administration of clodronate, MHV-68 transport into the synovium occurs only in the presence of Ly6Chigh monocytes. Tracking of adoptively transferred Ly6Chigh GFP infected monocytes into arthritic CCR2-/- mice by two-photon intravital microscopy showed that this monocyte subset has the capacity to deliver viruses to inflamed AR joints, as confirmed by the detection of viral DNA in inflamed tissues of recipient mice. We thus conclude that Ly6Chigh monocytes import MHV-68 when they are mobilized to the inflamed arthritic joint.


Asunto(s)
Artritis Experimental/inmunología , Artritis Reumatoide/inmunología , Infecciones por Virus de Epstein-Barr/inmunología , Infecciones por Herpesviridae/inmunología , Herpesvirus Humano 4/fisiología , Monocitos/inmunología , Rhadinovirus/fisiología , Infecciones Tumorales por Virus/inmunología , Traslado Adoptivo , Animales , Antígenos Ly/metabolismo , Artritis Experimental/virología , Artritis Reumatoide/virología , Células Cultivadas , ADN Viral/análisis , Modelos Animales de Enfermedad , Infecciones por Virus de Epstein-Barr/virología , Femenino , Infecciones por Herpesviridae/virología , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/trasplante , Muridae , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , Receptores CCR2/genética , Rhadinovirus/patogenicidad , Infecciones Tumorales por Virus/virología
12.
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
13.
Exp Physiol ; 104(5): 740-754, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30790372

RESUMEN

NEW FINDINGS: What is the central question of this study? Can a single bone marrow mononuclear cell (BMMC) transplant into the subcapsular region of kidney improve cellular communication and adhesion, while restoring renal tissue cytoarchitecture and function during renovascular hypertension? What is the main finding and its importance? The BMMC transplantation restored connexin 40 expression and led to recovery of N- and E-cadherin levels within 15 days. It was observed, for the first time, that BMMC transplantation restores expression of nephrin, a component of the glomerular filtration barrier related to podocytes and the glomerular basal membrane. ABSTRACT: Stem cell therapy has emerged as a potential treatment for renal diseases owing to the regenerative potential of stem cells. However, a better understanding of the morphological and functional changes of damaged renal cells in the presence of transplanted stem cells is needed. The aim of this study was to investigate cell-cell communication and adhesion in renal parenchyma, with analysis of fibrosis, to evaluate renal morphology and function after bone marrow mononuclear cell (BMMC) transplantation in two-kidney-one-clip rats. The BMMC therapy significantly decreased blood pressure and renin expression, improved renal morphology and restored the glomerular filtration barrier, with remodelling of podocytes. In addition, there was a reduction in fibrosis, and connexin 40 and nephrin expression were significantly increased after 7 and 15 days of transplantation. Plasma creatinine, urea and total protein levels were restored, and proteinuria was reduced. Furthermore, N- and E-cadherin expression was increased soon after BMMC therapy. Green fluorescent protein-positive BMMCs were found in the renal cortex 24 and 48 h after transplantation into the renal subcapsule, and at 7 and 15 days after transplantation, these cells were observed throughout the renal medulla, indicating cellular migration. Therefore, these data suggest that transplanted BMMCs improve cell-cell communication and adhesion between damaged cells, which is accompanied by a recovery of renal morphology and function.


Asunto(s)
Trasplante de Médula Ósea/métodos , Barrera de Filtración Glomerular/patología , Hipertensión Renovascular/patología , Hipertensión Renovascular/terapia , Uniones Intercelulares/patología , Animales , Presión Sanguínea , Cadherinas/metabolismo , Comunicación Celular , Fibrosis , Riñón/patología , Corteza Renal/patología , Masculino , Monocitos/trasplante , Podocitos/patología , Ratas , Ratas Wistar , Renina/biosíntesis
14.
Cancer Sci ; 109(11): 3403-3410, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30142694

RESUMEN

Although first-line chemotherapy has a high rate of complete responses in ovarian cancer patients, the vast majority of patients present with recurrent disease that has become refractory to conventional chemotherapy. Peritoneal dissemination and malignant ascites are the hallmarks of recurrent or advanced ovarian cancer and severely reduce quality of life. Development of therapeutic measures to treat such patients is eagerly anticipated. Macrophage infiltration is observed in various types of cancer including epithelial ovarian cancer. In addition, macrophages are involved in the formation of spheroids in the malignant ascites of ovarian cancer and promote cancer growth. iPS-ML, macrophage-like myelomonocytic cells generated from human induced pluripotent stem (iPS) cells, made close contacts with ovarian cancer cells in vitro. We hypothesized that, if we inoculate iPS-ML-producing IFN-ß (iPS-ML/IFN-ß) into the peritoneal cavity of patients with ovarian cancer, IFN-ß produced by the iPS-ML/IFN-ß would efficiently act on the cancer cells to suppress cancer growth. To evaluate this hypothesis, we injected iPS-ML/IFN-ß into SCID mice bearing peritoneally disseminated human ovarian cancer cells, SKOV3. Immunohistochemical analysis of the intraperitoneal tumors detected iPS-ML/IFN-ß infiltrating into the cancer tissues. Therapy with iPS-ML/IFN-ß significantly suppressed tumor progression. In addition, dramatic reduction of cancer-related ascites was observed. Collectively, it is suggested that iPS-ML/IFN-ß therapy offers a new approach for the treatment of patients with advanced ovarian cancer.


Asunto(s)
Ascitis/terapia , Interferón beta/metabolismo , Monocitos/trasplante , Neoplasias Ováricas/terapia , Neoplasias Peritoneales/terapia , Animales , Ascitis/etiología , Línea Celular Tumoral , Técnicas de Cocultivo , Femenino , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/inmunología , Ratones , Ratones SCID , Monocitos/citología , Monocitos/inmunología , Neoplasias Ováricas/complicaciones , Neoplasias Ováricas/inmunología , Neoplasias Peritoneales/complicaciones , Neoplasias Peritoneales/inmunología , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto
15.
J Neuroinflammation ; 15(1): 47, 2018 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-29454374

RESUMEN

BACKGROUND: It is well understood that hypoxic-ischemic (HI) brain injury during the highly vulnerable perinatal period can lead to cerebral palsy, the most prevalent cause of chronic disability in children. Recently, human clinical trials have reported safety and some efficacy following treatment of cerebral palsy using umbilical cord blood (UCB) cells. UCB is made up of many different cell types, including endothelial progenitor cells (EPCs), T regulatory cells (Tregs), and monocyte-derived suppressor cells (MDSCs). How each cell type contributes individually towards reducing neuroinflammation and/or repairing brain injury is not known. In this study, we examined whether human (h) UCB, or specific UCB cell types, could reduce peripheral and cerebral inflammation, and promote brain repair, when given early after perinatal HI brain injury. METHODS: HI brain injury was induced in postnatal day (PND) 7 rat pups and cells were administered intraperitoneally on PND 8. Behavioral testing was performed 7 days post injury, and then, brains and spleens were collected for analysis. RESULTS: We found in vitro that all UCB cell types, except for EPCs, were immunomodulatory. Perinatal HI brain injury induced significant infiltration of CD4+ T cells into the injured cerebral hemisphere, and this was significantly reduced by all hUCB cell types tested. Compared to HI, UCB, Tregs, and EPCs were able to reduce motor deficits, reduce CD4+ T cell infiltration into the brain, and reduce microglial activation. In addition to the beneficial effects of UCB, EPCs also significantly reduced cortical cell death, returned CD4+ T cell infiltration to sham levels, and reduced the peripheral Th1-mediated pro-inflammatory shift. CONCLUSION: This study highlights that cells found in UCB is able to mediate neuroinflammation and is an effective neuroprotective therapy. Our study also shows that particular cells found in UCB, namely EPCs, may have an added advantage over using UCB alone. This work has the potential to progress towards tailored UCB therapies for the treatment of perinatal brain injury.


Asunto(s)
Trasplante de Células Madre de Sangre del Cordón Umbilical/métodos , Células Progenitoras Endoteliales/trasplante , Sangre Fetal/citología , Hipoxia-Isquemia Encefálica/terapia , Monocitos/trasplante , Linfocitos T Reguladores/trasplante , Animales , Animales Recién Nacidos , Células Progenitoras Endoteliales/metabolismo , Sangre Fetal/metabolismo , Sangre Fetal/trasplante , Humanos , Hipoxia-Isquemia Encefálica/metabolismo , Hipoxia-Isquemia Encefálica/patología , Inflamación/metabolismo , Inflamación/patología , Inflamación/terapia , Monocitos/metabolismo , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Linfocitos T Reguladores/metabolismo
16.
Am Heart J ; 201: 141-148, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29803986

RESUMEN

BACKGROUND: Heart failure following myocardial infarction is a common, disabling, and deadly condition. Direct injection of autologous bone marrow mononuclear cells into the myocardium may result in improved functional recovery, relieve symptoms, and improve other cardiovascular outcomes. METHODS: CardiAMP-HF is a randomized, double-blind, sham-controlled, pivotal trial designed to investigate the safety and efficacy of autologous bone marrow mononuclear cells treatment for patients with medically refractory and symptomatic ischemic cardiomyopathy. The primary end point is change in 6-minute walk distance adjusted for major adverse cardiovascular events at 12 months following treatment. Particularly novel aspects of this trial include a cell potency assay to screen subjects who have bone marrow cell characteristics that suggest a favorable response to treatment, a point-of-care treatment method, a high target dose of 200 million cells, and an efficient transcatheter intramyocardial delivery method that is associated with high cell retention. CONCLUSIONS: This novel approach may lead to a new treatment for those with ischemic heart disease suffering from medically refractory heart failure.


Asunto(s)
Trasplante de Médula Ósea/métodos , Insuficiencia Cardíaca/terapia , Monocitos/trasplante , Infarto del Miocardio/complicaciones , Volumen Sistólico/fisiología , Función Ventricular Izquierda/fisiología , Adulto , Anciano , Método Doble Ciego , Femenino , Estudios de Seguimiento , Insuficiencia Cardíaca/etiología , Insuficiencia Cardíaca/fisiopatología , Humanos , Masculino , Persona de Mediana Edad , Monocitos/citología , Infarto del Miocardio/fisiopatología , Infarto del Miocardio/terapia , Trasplante Autólogo , Resultado del Tratamiento , Adulto Joven
17.
Basic Res Cardiol ; 114(1): 1, 2018 11 12.
Artículo en Inglés | MEDLINE | ID: mdl-30443679

RESUMEN

Monocytes are involved in adverse left ventricular (LV) remodelling following myocardial infarction (MI). To provide therapeutic opportunities we aimed to identify gene transcripts in monocytes that relate to post-MI healing and evaluated intervention with the observed gene activity in a rat MI model. In 51 MI patients treated by primary percutaneous coronary intervention (PCI), the change in LV end-diastolic volume index (EDVi) from baseline to 4-month follow-up was assessed using cardiovascular magnetic resonance imaging (CMR). Circulating monocytes were collected at day 5 (Arterioscler Thromb Vasc Biol 35:1066-1070, 2015; Cell Stem Cell 16:477-487, 2015; Curr Med Chem 13:1877-1893, 2006) after primary PCI for transcriptome analysis. Transcriptional profiling and pathway analysis revealed that patients with a decreased LV EDVi showed an induction of type I interferon (IFN) signalling (type I IFN pathway: P value < 0.001; false discovery rate < 0.001). We subsequently administered 15,000 Units of IFN-α subcutaneously in a rat MI model for three consecutive days following MI. Cardiac function was measured using echocardiography and infarct size/cardiac inflammation using (immuno)-histochemical analysis. We found that IFN-α application deteriorated ventricular dilatation and increased infarct size at day 28 post-MI. Moreover, IFN-α changed the peripheral monocyte subset distribution towards the pro-inflammatory monocyte subset whereas in the myocardium, the presence of the alternative macrophage subset was increased at day 3 post-MI. Our findings suggest that induction of type I IFN signalling in human monocytes coincides with adverse LV remodelling. In rats, however, IFN-α administration deteriorated post-MI healing. These findings underscore important but also contradictory roles for the type I IFN response during cardiac healing following MI.


Asunto(s)
Interferón Tipo I/metabolismo , Monocitos/trasplante , Infarto del Miocardio/metabolismo , Infarto del Miocardio/terapia , Remodelación Ventricular , Adulto , Anciano , Animales , Trasplante de Médula Ósea/métodos , Femenino , Humanos , Interferón Tipo I/farmacología , Masculino , Persona de Mediana Edad , Monocitos/metabolismo , Infarto del Miocardio/patología , Ratas , Ratas Wistar , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Remodelación Ventricular/efectos de los fármacos , Cicatrización de Heridas/efectos de los fármacos , Cicatrización de Heridas/fisiología
18.
Arterioscler Thromb Vasc Biol ; 37(2): 258-263, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27908893

RESUMEN

OBJECTIVE: To create a model of atherosclerosis using green fluorescent protein (GFP)-targeted monocytes/macrophages, allowing analysis of both endogenous GFP+ and adoptively transferred GFP+ myeloid cells in arterial inflammation. APPROACH AND RESULTS: hCD68GFP reporter mice were crossed with ApoE-/- mice. Expression of GFP was localized to macrophages in atherosclerotic plaques and in angiotensin II-induced aortic aneurysms and correlated with galectin 3 and mCD68 expression. Flow cytometry confirmed GFP+ expression in CD11b+/CD64+, CD11c+/MHC-IIHI, and CD11b+/F4/80+ myeloid cells. Adoptive transfer of GFP+ monocytes demonstrated monocyte recruitment to both adventitia and atherosclerotic plaque, throughout the aortic root, within 72 hours. We demonstrated the biological utility of hCD68GFP monocytes by comparing the recruitment of wild-type and CCR2-/- monocytes to sites of inflammation. CONCLUSIONS: hCD68GFP/ApoE-/- mice provide a new approach to study macrophage accumulation in atherosclerotic plaque progression and to identify cells recruited from adoptively transferred monocytes.


Asunto(s)
Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/metabolismo , Aorta/metabolismo , Enfermedades de la Aorta/metabolismo , Apolipoproteínas E/deficiencia , Aterosclerosis/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Macrófagos/metabolismo , Monocitos/metabolismo , Placa Aterosclerótica , Traslado Adoptivo , Angiotensina II , Animales , Antígenos CD/genética , Antígenos de Diferenciación/metabolismo , Antígenos de Diferenciación Mielomonocítica/genética , Aorta/patología , Aneurisma de la Aorta/inducido químicamente , Aneurisma de la Aorta/genética , Aneurisma de la Aorta/metabolismo , Aneurisma de la Aorta/patología , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/patología , Apolipoproteínas E/genética , Aterosclerosis/genética , Aterosclerosis/patología , Antígeno CD11b/metabolismo , Antígeno CD11c/metabolismo , Rastreo Celular/métodos , Células Cultivadas , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Galectina 3/metabolismo , Predisposición Genética a la Enfermedad , Proteínas Fluorescentes Verdes/genética , Macrófagos/patología , Macrófagos/trasplante , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Monocitos/patología , Monocitos/trasplante , Fenotipo , Receptores de IgG/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal
19.
J Clin Apher ; 33(3): 324-330, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29232011

RESUMEN

In addition to stem cells, T-cells, natural killer cells, dendritic cells, and monocytes are also collected and infused from the autograft in patients undergoing autologous peripheral blood hematopoietic stem cell transplantation. Recent reports have shown that these autograft immune effector cells can affect the clinical outcome postautologous peripheral blood hematopoietic stem cell transplantation. In this article, I will review the clinical impact on the survival of these autograft immune effector cells conferring the concept of autologous graft versus tumor effect.


Asunto(s)
Autoinjertos/citología , Trasplante de Células Madre de Sangre Periférica/métodos , Autoinjertos/inmunología , Células Dendríticas/inmunología , Células Dendríticas/trasplante , Efecto Injerto vs Tumor , Humanos , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/trasplante , Monocitos/inmunología , Monocitos/trasplante , Linfocitos T/inmunología , Linfocitos T/trasplante , Resultado del Tratamiento
20.
Nano Lett ; 17(9): 5546-5554, 2017 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-28758755

RESUMEN

Metastasis causes high mortality of breast cancer, and the inability of drug delivery to metastatic sites remains a crucial challenge for antimetastasis therapy. Herein, we report that inflammatory monocytes loading legumain-activated nanoparticles can actively target lung metastases and initiate metastasis-specific intelligent drug release for antimetastasis therapy. The cytotoxic mertansine is conjugated to poly(styrene-co-maleic anhydride) with a legumain-sensitive peptide and self-assembled into nanoparticles (SMNs), and then loaded into inflammatory monocytes to prepare the SMNs-loaded monocytes delivery system (M-SMNs). M-SMNs would be in living state in circulation to ensure their active targeting to lung metastases, and responsively damaged at the metastatic sites upon the differentiation of monocytes into macrophages. The anticancer drugs are intelligently released from M-SMNs as free drug molecules and drug-loaded microvesicles, resulting in considerable inhibition on the proliferation, migration, and invasion activities of metastatic 4T1 breast cancer cells. Moreover, M-SMNs significantly improve the delivery to lung metastases and penetrate the metastatic tumors, thus producing a 77.8% inhibition of lung metastases. Taken together, our findings provide an intelligent biomimetic drug delivery strategy via the biological properties of inflammatory monocytes for effective antimetastasis therapy.


Asunto(s)
Antineoplásicos Fitogénicos/administración & dosificación , Neoplasias de la Mama/patología , Cisteína Endopeptidasas/administración & dosificación , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/secundario , Maitansina/administración & dosificación , Monocitos/inmunología , Invasividad Neoplásica/prevención & control , Animales , Antineoplásicos Fitogénicos/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/inmunología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cisteína Endopeptidasas/uso terapéutico , Sistemas de Liberación de Medicamentos/métodos , Liberación de Fármacos , Femenino , Humanos , Neoplasias Pulmonares/inmunología , Maitansina/uso terapéutico , Ratones , Monocitos/trasplante , Invasividad Neoplásica/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA