Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 258
Filtrar
1.
Cells ; 10(7)2021 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-34359844

RESUMEN

Neutrophils are first-line responders of the innate immune system. Following myocardial infarction (MI), neutrophils are quickly recruited to the ischemic region, where they initiate the inflammatory response, aiming at cleaning up dead cell debris. However, excessive accumulation and/or delayed removal of neutrophils are deleterious. Neutrophils can promote myocardial injury by releasing reactive oxygen species, granular components, and pro-inflammatory mediators. More recent studies have revealed that neutrophils are able to form extracellular traps (NETs) and produce extracellular vesicles (EVs) to aggravate inflammation and cardiac injury. On the contrary, there is growing evidence showing that neutrophils also exert anti-inflammatory, pro-angiogenic, and pro-reparative effects, thus facilitating inflammation resolution and cardiac repair. In this review, we summarize the current knowledge on neutrophils' detrimental roles, highlighting the role of recently recognized NETs and EVs, followed by a discussion of their beneficial effects and molecular mechanisms in post-MI cardiac remodeling. In addition, emerging concepts about neutrophil diversity and their modulation of adaptive immunity are discussed.


Asunto(s)
Inmunidad Adaptativa , Trampas Extracelulares/inmunología , Mediadores de Inflamación/inmunología , Infarto del Miocardio/inmunología , Infiltración Neutrófila/inmunología , Neutrófilos/inmunología , Animales , Citocinas/genética , Citocinas/inmunología , Modelos Animales de Enfermedad , Trampas Extracelulares/genética , Vesículas Extracelulares/genética , Vesículas Extracelulares/inmunología , Regulación de la Expresión Génica , Humanos , Mediadores de Inflamación/metabolismo , Macrófagos/inmunología , Macrófagos/patología , Ratones , Infarto del Miocardio/genética , Infarto del Miocardio/patología , Miocardio/inmunología , Miocardio/patología , Neovascularización Fisiológica/genética , Neovascularización Fisiológica/inmunología , Infiltración Neutrófila/genética , Neutrófilos/patología , Especies Reactivas de Oxígeno/inmunología , Especies Reactivas de Oxígeno/metabolismo
3.
PLoS One ; 16(5): e0251121, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33983985

RESUMEN

Several angiogenesis-dependent diseases, including age-related macular degeneration and infantile hemangioma, display differential prevalence among Black, as compared to White individuals. Although socioeconomic status and genetic architecture have been suggested as explaining these differences, we have recently shown that pigment production per se might be involved. For example, we have shown that the extracellular protein fibromodulin is a pro-angiogenic factor highly secreted by melanocytes in White but not Black individuals. Still, additional pigment-dependent angiogenic factors and their molecular mechanisms remain to be identified. Understanding the contribution of pigmentation to angiogenesis in health and disease is essential for precision medicine of angiogenesis-dependent diseases with racial disparity. Toward that goal, we compared the transcriptomes of Black and White individuals in three tissues with angiogenic activity, namely artery, whole blood, and skin. We identified several differentially expressed angiogenesis pathways, including artery morphogenesis, regulation of endothelial cell chemotaxis, and cellular response to vascular endothelial growth factor stimulus. We then demonstrated that the expression of key genes in these pathways is directly modulated by the degree of pigmentation. We further identified the precise pigment production pathway controlling the expression of these genes, namely melanocortin 1 receptor (MC1R) signaling. These results demonstrate pigment-mediated regulation of angiogenesis-related pathways and their driver genes across human tissues.


Asunto(s)
Melanocitos/metabolismo , Neovascularización Fisiológica/genética , Neovascularización Fisiológica/fisiología , Animales , Arterias/metabolismo , Población Negra/genética , Sangre/metabolismo , Bases de Datos Genéticas , Femenino , Expresión Génica/genética , Regulación de la Expresión Génica/genética , Humanos , Masculino , Melanocitos/fisiología , Ratones , Ratones Endogámicos C57BL , Neovascularización Fisiológica/inmunología , Especificidad de Órganos/genética , Receptor de Melanocortina Tipo 1/genética , Receptor de Melanocortina Tipo 1/metabolismo , Piel/metabolismo , Transcriptoma/genética , Población Blanca/genética
4.
J Reprod Immunol ; 145: 103322, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33887508

RESUMEN

The underlying mechanism of preeclampsia by which an angiogenic imbalance results in systemic vascular endothelial dysfunction remains unclear. Complement activation directly induces endothelial dysfunction and is known to be involved in preeclampsia; nevertheless, the association between complement activation and angiogenic imbalance has not been established. This study aimed to evaluate whether angiogenic imbalance affects the expression and secretion of inhibitory complement factor H (CFH) in endothelial cells, resulting in complement activation and systemic vascular endothelial dysfunction. Viability of human umbilical vein endothelial cells (HUVECs) was assessed upon CFH knockdown by targeted-siRNA, and were incubated with complement factors. HUVECs were also treated with placental growth factor (PlGF) and/or soluble fms-like tyrosine kinase 1 (sFlt1), and CFH expression and secretion were measured. These cells were evaluated by cell viability assay and cell surface complement activation was quantified by immunocytochemical assessment of C5b-9 deposition. HUVECs transfected with CFH-siRNA had significantly lower viability than that of control cells. Moreover, the expression and secretion of CFH were significantly increased upon PlGF treatment compared with PlGF + sFlt1 combo. HUVECs treated with PlGF had less C5b-9 deposition and higher viability than HUVECs treated with PlGF + sFlt1. In summary, CFH was found to be essential for endothelial cell survival by inhibiting complement activation. An angiogenic imbalance, including decreased PlGF and increased sFlt1, suppresses CFH expression and secretion, resulting in complement activation on the surface of endothelial cells and systemic vascular endothelial dysfunction.


Asunto(s)
Activación de Complemento , Preeclampsia/inmunología , Estudios de Casos y Controles , Supervivencia Celular/inmunología , Células Cultivadas , Factor H de Complemento/metabolismo , Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Endotelio Vascular/patología , Femenino , Células Endoteliales de la Vena Umbilical Humana , Humanos , Neovascularización Fisiológica/inmunología , Placenta/irrigación sanguínea , Placenta/inmunología , Placenta/patología , Factor de Crecimiento Placentario/metabolismo , Preeclampsia/patología , Embarazo , Cultivo Primario de Células , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo
5.
Cells ; 10(3)2021 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-33799966

RESUMEN

The novel coronavirus severe acute respiratory syndrome-CoV-2 (SARS-CoV-2) is responsible for COVID-19 infection. The COVID-19 pandemic represents one of the worst global threats in the 21st century since World War II. This pandemic has led to a worldwide economic recession and crisis due to lockdown. Biomedical researchers, pharmaceutical companies, and premier institutes throughout the world are claiming that new clinical trials are in progress. During the severe phase of this disease, mechanical ventilators are used to assist in the management of outcomes; however, their use can lead to the development of pneumonia. In this context, mesenchymal stem cell (MSC)-derived exosomes can serve as an immunomodulation treatment for COVID-19 patients. Exosomes possess anti-inflammatory, pro-angiogenic, and immunomodulatory properties that can be explored in an effort to improve the outcomes of SARS-CoV-2-infected patients. Currently, only one ongoing clinical trial (NCT04276987) is specifically exploring the use of MSC-derived exosomes as a therapy to treat SARS-CoV-2-associated pneumonia. The purpose of this review is to provide insights of using exosomes derived from mesenchymal stem cells in management of the co-morbidities associated with SARS-CoV-2-infected persons in direction of improving their health outcome. There is limited knowledge of using exosomes in SARS-CoV-2; the clinicians and researchers should exploit exosomes as therapeutic regime.


Asunto(s)
COVID-19/terapia , Exosomas/metabolismo , Vesículas Extracelulares/metabolismo , Inmunomodulación , Células Madre Mesenquimatosas/metabolismo , Neumonía Viral/terapia , COVID-19/complicaciones , COVID-19/metabolismo , COVID-19/patología , Citocinas/metabolismo , Citocinas/farmacología , Exosomas/química , Exosomas/genética , Humanos , Inflamación/inmunología , Inflamación/terapia , Inflamación/virología , Células Madre Mesenquimatosas/inmunología , Neovascularización Fisiológica/inmunología , Neumonía Viral/complicaciones , Neumonía Viral/virología , Infecciones del Sistema Respiratorio/complicaciones , Infecciones del Sistema Respiratorio/terapia , Infecciones del Sistema Respiratorio/virología
6.
Int J Mol Sci ; 22(5)2021 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-33669136

RESUMEN

The transplantation of various immune cell types are promising approaches for the treatment of ischemic cardiovascular disease including myocardial infarction (MI) and peripheral arterial disease (PAD). Major limitation of these so-called Advanced Therapy Medicinal Products (ATMPs) is the ischemic microenvironment affecting cell homeostasis and limiting the demanded effect of the transplanted cell products. Accordingly, different clinical and experimental strategies have been evolved to overcome these obstacles. Here, we give a short review of the different experimental and clinical strategies to solve these issues due to ischemic cardiovascular disease.


Asunto(s)
Trasplante de Células/métodos , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Células Madre Hematopoyéticas/metabolismo , Isquemia/terapia , Infarto del Miocardio/terapia , Neovascularización Fisiológica/efectos de los fármacos , Enfermedad Arterial Periférica/terapia , Animales , Enfermedades Cardiovasculares/terapia , Hipoxia de la Célula/fisiología , Trasplante de Células/instrumentación , Tratamiento Basado en Trasplante de Células y Tejidos/instrumentación , Microambiente Celular/efectos de los fármacos , Humanos , Péptidos y Proteínas de Señalización Intercelular/farmacología , Péptidos y Proteínas de Señalización Intercelular/uso terapéutico , MicroARNs/genética , MicroARNs/metabolismo , Infarto del Miocardio/inmunología , Neovascularización Fisiológica/inmunología , Enfermedad Arterial Periférica/inmunología
7.
Int J Mol Sci ; 22(4)2021 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-33670600

RESUMEN

Rheumatoid arthritis (RA) is an autoimmune disease characterized by chronic inflammation of the synovial joints. Inflammation, new blood vessel formation (angiogenesis) and bone resorption (osteoclastogenesis) are three key processes involved in the joint damage and deformities of arthritis. Various gut microbiota-derived metabolites are implicated in RA pathogenesis. However, there is barely any information about the impact of two such metabolites, indole-3-aldehyde (IAld) and indole-3-acetic acid (I3AA), on arthritis-related processes. We conducted a comparative analysis of IAld and I3AA using established cell-based models to understand how they might influence RA pathogenesis. Although structurally similar, the bioactivities of these two metabolites were profoundly different. IAld but not I3AA, inhibited the expression of pro-inflammatory cytokines (IL-1ß and IL-6) in RAW 264.7 (RAW) cells stimulated with heat-killed M. tuberculosis sonicate (Mtb) and lipopolysaccharide (LPS). IAld also exhibited pro-angiogenic activity and pro-osteoclastogenic activity. In contrast, I3AA exhibited anti-angiogenic activity on endothelial cell tube formation but had no effect on osteoclastogenesis. Both IAld and I3AA have been proposed as aryl hydrocarbon receptor (AhR) agonists. Use of CH-223191, an inhibitor of the AhR, suppressed the anti-angiogenic activity of I3AA but failed to mitigate the effects of IAld. Further investigation of the anti-inflammatory activities of IAld and I3AA in LPS-treated RAW cells indicated that inhibition of MyD88-dependent activation of NF-κB and MAPK pathways was not likely involved. Our results suggest that the relative bioavailability of these indole derivatives may differentially impact RA progression and possibly other diseases that share similar cellular processes.


Asunto(s)
Artritis Reumatoide/inmunología , Enfermedades Autoinmunes/inmunología , Citocinas/inmunología , Ácidos Indolacéticos/inmunología , Indoles/inmunología , Microbiota/inmunología , Animales , Artritis Reumatoide/metabolismo , Enfermedades Autoinmunes/metabolismo , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/inmunología , Células Cultivadas , Citocinas/metabolismo , Calor , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/inmunología , Células Endoteliales de la Vena Umbilical Humana/fisiología , Humanos , Ácidos Indolacéticos/metabolismo , Ácidos Indolacéticos/farmacología , Indoles/metabolismo , Indoles/farmacología , Lipopolisacáridos/inmunología , Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Mycobacterium tuberculosis/inmunología , Mycobacterium tuberculosis/metabolismo , Neovascularización Fisiológica/efectos de los fármacos , Neovascularización Fisiológica/inmunología , Osteoclastos/citología , Osteoclastos/efectos de los fármacos , Osteoclastos/inmunología , Células RAW 264.7
8.
Front Immunol ; 12: 784098, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34975872

RESUMEN

Following a cerebral ischemic event, substantial alterations in both cellular and molecular activities occur due to ischemia-induced cerebral pathology. Mounting evidence indicates that the robust recruitment of immune cells plays a central role in the acute stage of stroke. Infiltrating peripheral immune cells and resident microglia mediate neuronal cell death and blood-brain barrier disruption by releasing inflammation-associated molecules. Nevertheless, profound immunological effects in the context of the subacute and chronic recovery phase of stroke have received little attention. Early attempts to curtail the infiltration of immune cells were effective in mitigating brain injury in experimental stroke studies but failed to exert beneficial effects in clinical trials. Neural tissue damage repair processes include angiogenesis, neurogenesis, and synaptic remodeling, etc. Post-stroke inflammatory cells can adopt divergent phenotypes that influence the aforementioned biological processes in both endothelial and neural stem cells by either alleviating acute inflammatory responses or secreting a variety of growth factors, which are substantially involved in the process of angiogenesis and neurogenesis. To better understand the multiple roles of immune cells in neural tissue repair processes post stroke, we review what is known and unknown regarding the role of immune cells in angiogenesis, neurogenesis, and neuronal remodeling. A comprehensive understanding of these inflammatory mechanisms may help identify potential targets for the development of novel immunoregulatory therapeutic strategies that ameliorate complications and improve functional rehabilitation after stroke.


Asunto(s)
Accidente Cerebrovascular Isquémico/inmunología , Neovascularización Fisiológica/inmunología , Enfermedades Neuroinflamatorias/inmunología , Plasticidad Neuronal/inmunología , Animales , Barrera Hematoencefálica/inmunología , Barrera Hematoencefálica/patología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Células Endoteliales/inmunología , Células Endoteliales/metabolismo , Humanos , Mediadores de Inflamación/metabolismo , Accidente Cerebrovascular Isquémico/patología , Linfocitos/inmunología , Linfocitos/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Microglía/inmunología , Microglía/metabolismo , Células-Madre Neurales/inmunología , Células-Madre Neurales/metabolismo , Enfermedades Neuroinflamatorias/patología , Recuperación de la Función/inmunología
9.
J Immunol ; 205(10): 2557-2565, 2020 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-33168598

RESUMEN

Regulatory T cells (Tregs) are crucial mediators of immune homeostasis. They regulate immune response by suppressing inflammation and promoting self-tolerance. In addition to their immunoregulatory role, a growing body of evidence highlights the dynamic role of Tregs in angiogenesis, the process of forming new blood vessels. Although angiogenesis is critically important for normal tissue regeneration, it is also a hallmark of pathological processes, including malignancy and chronic inflammation. Interestingly, the role of Tregs in angiogenesis has been shown to be highly tissue- and context-specific and as a result can yield either pro- or antiangiogenic effects. For these reasons, there is considerable interest in determining the molecular underpinnings of Treg-mediated modulation of angiogenesis in different disease states. The present review summarizes the role of Tregs in angiogenesis and mechanisms by which Tregs regulate angiogenesis and discusses how these mechanisms differ in homeostatic and pathological settings.


Asunto(s)
Neoplasias/inmunología , Neovascularización Patológica/inmunología , Neovascularización Fisiológica/inmunología , Linfocitos T Reguladores/inmunología , Animales , Humanos , Inflamación/inmunología , Modelos Animales , Neoplasias/irrigación sanguínea , Transducción de Señal/inmunología , Factor A de Crecimiento Endotelial Vascular/metabolismo
10.
Nanoscale ; 12(31): 16474-16488, 2020 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-32743625

RESUMEN

The immunomodulatory capability of biomaterials is of paramount importance for successful material-mediated bone regeneration. Particularly, the design of surface nano-topography can be leveraged to instruct immune reactions, yet the understanding of such "nano-morphology effect" is still very limited. Herein, highly ordered nano-concave pit (denoted as NCPit) and nano-convex dot (denoted as NCDot) microarrays with two different sizes were successfully constructed on a 316LSS surface via anodization and subsequently immersion-coating treatment, respectively. We, for the first time, comparatively investigated the interactions of NCPit and NCDot microarrays with RAW264.7 macrophages and their immunomodulatory impacts on osteogenesis and angiogenesis of human bone mesenchymal stem cells (hBMSCs) and human umbilical vein endothelial cells (HUVECs). NCDot microarrays induced macrophages towards M2 polarization with the higher expression level of anti-inflammatory markers (IL-10 and CD 206) and the lower level of pro-inflammatory markers (TNF-α, IL-1ß, IL-6 and CD 86) than those of the corresponding NCPit microarrays. During the process, the expressions of osteogenesis-related genes (Runx2, OPN and OCN) of hBMSCs, and angiogenesis-related genes (eNOS, HIF-1α, KDR and VEGF) of HUVECs were significantly upregulated by the NCDot microarray-modulating immune microenvironment of macrophages, and finally stimulated osteogenesis and angiogenesis. Thus, the prepared NCDot arrays were able to significantly promote osteo-/angiogenic activity by generating a more suitable immune microenvironment than NCPit arrays, offering substantial evidence for designing immunomodulatory biomaterials with specific microstructures and optimal bioactivity.


Asunto(s)
Materiales Biocompatibles Revestidos/química , Inmunomodulación , Neovascularización Fisiológica/inmunología , Osteogénesis/inmunología , Animales , Diferenciación Celular , Células Cultivadas , Células Endoteliales de la Vena Umbilical Humana/citología , Humanos , Macrófagos/citología , Macrófagos/inmunología , Células Madre Mesenquimatosas/citología , Ratones , Células RAW 264.7 , Propiedades de Superficie
11.
Inflamm Res ; 69(11): 1103-1110, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32808153

RESUMEN

Multiple sclerosis (MS) is an autoimmune disease, characterized by multiple demyelination of axons in both white and gray matter in the Central Nervous System (CNS). There is increasing evidence to support the notion that angiogenesis and chronic inflammation are mutually related. Different immune cells, including monocytes-macrophages, lymphocytes, neutrophils, mast cells (MCs) and dendritic cells are able to secrete an array of angiogenic cytokines, which promote growth, migration, and activation of endothelial cells. MCs play various roles in MS pathogenesis, influencing the innate immune response in peripheral tissues and in CNS. The aim of this review article is to discuss the role of MCs in MS pathogenesis with particular reference to the involvement of these inflammatory cells in the angiogenic processes occurring during MS.


Asunto(s)
Mastocitos/inmunología , Esclerosis Múltiple/inmunología , Neovascularización Patológica/inmunología , Neovascularización Fisiológica/inmunología , Animales , Humanos , Esclerosis Múltiple/tratamiento farmacológico
12.
Proc Natl Acad Sci U S A ; 117(25): 14354-14364, 2020 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-32513697

RESUMEN

Ischemic injury initiates a sterile inflammatory response that ultimately participates in the repair and recovery of tissue perfusion. Macrophages are required for perfusion recovery during ischemia, in part because they produce growth factors that aid in vascular remodeling. The input signals governing this pro-revascularization phenotype remain of interest. Here we found that hindlimb ischemia increases levels of resolvin D1 (RvD1), an inflammation-resolving lipid mediator that targets macrophages via its receptor, ALX/FPR2. Exogenous RvD1 enhances perfusion recovery during ischemia, and mice deficient in Alx/Fpr2 have an endogenous defect in this process. Mechanistically, RNA sequencing revealed that RvD1 induces a transcriptional program in macrophages characteristic of a pro-revascularization phenotype. Vascularization of ischemic skeletal muscle, as well as cutaneous wounds, is impaired in mice with myeloid-specific deficiency of Alx/Fpr2, and this is associated with altered expression of pro-revascularization genes in skeletal muscle and macrophages isolated from skeletal muscle. Collectively, these results uncover a role of ALX/FPR2 in revascularization that may be amenable to therapeutic targeting in diseases associated with altered tissue perfusion and repair.


Asunto(s)
Ácidos Docosahexaenoicos/metabolismo , Isquemia/inmunología , Neovascularización Fisiológica/inmunología , Receptores de Formil Péptido/metabolismo , Receptores de Lipoxina/metabolismo , Cicatrización de Heridas/inmunología , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Técnicas de Inactivación de Genes , Humanos , Isquemia/patología , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Ratones , Ratones Noqueados , Músculo Esquelético/irrigación sanguínea , Músculo Esquelético/inmunología , Músculo Esquelético/patología , Cultivo Primario de Células , RNA-Seq , Receptores de Formil Péptido/genética , Receptores de Lipoxina/genética , Transducción de Señal/inmunología , Piel/irrigación sanguínea , Piel/inmunología , Piel/lesiones , Piel/patología , Transcripción Genética/inmunología
13.
J Immunol ; 205(1): 102-112, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32434940

RESUMEN

To maintain homeostasis, macrophages must be capable of assuming either an inflammatory or an anti-inflammatory phenotype. To better understand the latter, we stimulated human macrophages in vitro with TLR ligands in the presence of high-density immune complexes (IC). This combination of stimuli resulted in a broad suppression of inflammatory mediators and an upregulation of molecules involved in tissue remodeling and angiogenesis. Transcriptomic analysis of TLR stimulation in the presence of IC predicted the downstream activation of AKT and the inhibition of GSK3. Consequently, we pretreated LPS-stimulated human macrophages with small molecule inhibitors of GSK3 to partially phenocopy the regulatory effects of stimulation in the presence of IC. The upregulation of DC-STAMP and matrix metalloproteases was observed on these cells and may represent potential biomarkers for this regulatory activation state. To demonstrate the presence of these anti-inflammatory, growth-promoting macrophages in a human infectious disease, biopsies from patients with leprosy (Hanseniasis) were analyzed. The lepromatous form of this disease is characterized by hypergammaglobulinemia and defective cell-mediated immunity. Lesions in lepromatous leprosy contained macrophages with a regulatory phenotype expressing higher levels of DC-STAMP and lower levels of IL-12, relative to macrophages in tuberculoid leprosy lesions. Therefore, we propose that increased signaling by FcγR cross-linking on TLR-stimulated macrophages can paradoxically promote the resolution of inflammation and initiate processes critical to tissue growth and repair. It can also contribute to infectious disease progression.


Asunto(s)
Complejo Antígeno-Anticuerpo/metabolismo , Lepra Lepromatosa/inmunología , Lepra Tuberculoide/inmunología , Macrófagos/inmunología , Biopsia , Diferenciación Celular/inmunología , Línea Celular , Progresión de la Enfermedad , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3/metabolismo , Humanos , Lepra Lepromatosa/patología , Lepra Tuberculoide/patología , Activación de Macrófagos , Macrófagos/metabolismo , Masculino , Persona de Mediana Edad , Neovascularización Fisiológica/inmunología , Proteínas Proto-Oncogénicas c-akt/metabolismo , RNA-Seq , Receptores de IgG/metabolismo , Transducción de Señal/genética , Transducción de Señal/inmunología , Piel/citología , Piel/inmunología , Piel/patología , Receptores Toll-Like/metabolismo , Adulto Joven
14.
ACS Appl Mater Interfaces ; 12(14): 16058-16075, 2020 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-32182418

RESUMEN

Recent studies show that biomaterials are capable of regulating immune responses to induce a favorable osteogenic microenvironment and promote osteogenesis and angiogenesis. In this study, we investigated the effects of zinc silicate/nanohydroxyapatite/collagen (ZS/HA/Col) scaffolds on bone regeneration and angiogenesis and explored the related mechanism. We demonstrate that 10ZS/HA/Col scaffolds significantly enhanced bone regeneration and angiogenesis in vivo compared with HA/Col scaffolds. ZS/HA/Col scaffolds increased tartrate-resistant acid phosphatase (TRAP)-positive cells, nestin-positive bone marrow stromal cells (BMSCs) and CD31-positive neovessels, and expression of osteogenesis (Bmp-2 and Osterix) and angiogenesis-related (Vegf-α and Cd31) genes increased in nascent bone. ZS/HA/Col scaffolds with 10 wt % ZS activated the p38 signaling pathway in monocytes. The monocytes subsequently differentiated into TRAP+ cells and expressed higher levels of the cytokines SDF-1, TGF-ß1, VEGF-α, and PDGF-BB, which recruited BMSCs and endothelial cells (ECs) to the defect areas. Blocking the p38 pathway in monocytes reduced TRAP+ differentiation and cytokine secretion and resulted in a decrease in BMSC and EC homing and angiogenesis. Overall, these findings demonstrate that 10ZS/HA/Col scaffolds modulate monocytes and, thereby, create a favorable osteogenic microenvironment that promotes BMSC migration and differentiation and vessel formation by activating the p38 signaling pathway.


Asunto(s)
Regeneración Ósea/efectos de los fármacos , Colágeno/química , Durapatita/química , Nanopartículas/química , Silicatos/química , Compuestos de Zinc/química , Materiales Biocompatibles/química , Materiales Biocompatibles/farmacología , Diferenciación Celular/efectos de los fármacos , Quimiocina CXCL12/genética , Colágeno/síntesis química , Colágeno/farmacología , Durapatita/síntesis química , Durapatita/farmacología , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Humanos , Inmunidad/efectos de los fármacos , Células Madre Mesenquimatosas/efectos de los fármacos , Neovascularización Fisiológica/efectos de los fármacos , Neovascularización Fisiológica/inmunología , Nestina/genética , Osteogénesis/efectos de los fármacos , Osteogénesis/inmunología , Impresión Tridimensional , Silicatos/síntesis química , Silicatos/farmacología , Fosfatasa Ácida Tartratorresistente/química , Andamios del Tejido/química , Compuestos de Zinc/síntesis química , Compuestos de Zinc/farmacología
15.
J Surg Res ; 249: 145-155, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31958599

RESUMEN

BACKGROUND: Emerging evidence has linked autophagy to skin wound healing; however, the underlying cellular and molecular mechanisms remain poorly understood. The present study was designed to determine the role of autophagy in endothelial cell (EC)-mediated skin wound healing in mice. METHODS: Autophagy-related gene (Atg7) in mouse ECs was inactivated by the Cre-loxP system under the control of an EC-specific VE-Cadherin (Cdh5) promoter (Atg7EC-/- mice). Full-thickness skin wounds were created on the dorsum of wild-type (WT), Cdh5-Cre+, floxed Atg7 (Atg7F/F), and Atg7EC-/- mice. Autophagic activity was determined by autophagic flux assay in the primary culture of ECs isolated from these mice. The wound re-epithelialization and angiogenesis was examined by histological analyses. The angiogenic activity of ECs was evaluated by tube formation assay in vitro. EC proliferation was examined by a cell count CCK-8 kit. EC-originated intercellular communication with dermal fibroblasts and keratinocytes was assessed by measuring the effect of EC conditional medium on the growth of keratinocytes and fibroblasts. The levels of VEGF, EGF, bFGF in EC conditional medium were measured by ELISA. RESULTS: Autophagy deficiency in ECs markedly enhanced the re-epithelialization and the wound closure during skin wound healing. However, it has minimal impact on angiogenesis in the wounded skin. Notably, autophagy deficiency in ECs did not affect their proliferation and migration or angiogenic activity per se but enhanced the EC conditional medium-induced proliferation and migration of keratinocytes and fibroblasts. CONCLUSIONS: These results demonstrate for the first time an inhibitory role of autophagy in the EC-originated paracrine regulation of skin wound healing.


Asunto(s)
Proteína 7 Relacionada con la Autofagia/genética , Autofagia/genética , Células Endoteliales/inmunología , Herida Quirúrgica/inmunología , Cicatrización de Heridas/inmunología , Animales , Autofagia/inmunología , Comunicación Celular/genética , Comunicación Celular/inmunología , Movimiento Celular/genética , Proliferación Celular/genética , Células Cultivadas , Medios de Cultivo Condicionados/metabolismo , Modelos Animales de Enfermedad , Endotelio Vascular/citología , Femenino , Fibroblastos , Humanos , Queratinocitos , Masculino , Ratones , Ratones Noqueados , Miocardio/citología , Neovascularización Fisiológica/genética , Neovascularización Fisiológica/inmunología , Comunicación Paracrina/genética , Comunicación Paracrina/inmunología , Cultivo Primario de Células , Piel/irrigación sanguínea , Piel/lesiones
16.
Cells ; 9(2)2020 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-31991811

RESUMEN

: Introduction: Antibody treatment with anti-thymocyte globulin (ATG) has been shown to be cardioprotective. We aimed to evaluate which single anti-T-cell epitope antibody alters chemokine expression at a level similar to ATG and identified CD3, which is a T-cell co-receptor mediating T-cell activation. Based on these results, the effects of anti-CD3 antibody treatment on angiogenesis and cardioprotection were tested in vitro and in vivo. METHODS: Concentrations of IL-8 and MCP-1 in supernatants of human peripheral blood mononuclear cell (PBMC) cultures following distinct antibody treatments were evaluated by Enzyme-linked Immunosorbent Assay (ELISA). In vivo, anti-CD3 antibodies or vehicle were injected intravenously in rats subjected to acute myocardial infarction (AMI). Chemotaxis and angiogenesis were evaluated using tube and migration assays. Intracellular pathways were assessed using Western blot. Extracellular vesicles (EVs) were quantitatively evaluated using fluorescence-activated cell scanning, exoELISA, and nanoparticle tracking analysis. Also, microRNA profiles were determined by next-generation sequencing. RESULTS: Only PBMC stimulation with anti-CD3 antibody led to IL-8 and MCP-1 changes in secretion, similar to ATG. In a rat model of AMI, systemic treatment with an anti-CD3 antibody markedly reduced infarct scar size (27.8% (Inter-quartile range; IQR 16.2-34.9) vs. 12.6% (IQR 8.3-27.2); p < 0.01). The secretomes of anti-CD3 treated PBMC neither induced cardioprotective pathways in cardiomyocytes nor pro-angiogenic mechanisms in human umbilical vein endothelial cell (HUVECs) in vitro. While EVs quantities remained unchanged, PBMC incubation with an anti-CD3 antibody led to alterations in EVs miRNA expression. CONCLUSION: Treatment with an anti-CD3 antibody led to decreased scar size in a rat model of AMI. Whereas cardioprotective and pro-angiogenetic pathways were unaltered by anti-CD3 treatment, qualitative changes in the EVs miRNA expression could be observed, which might be causal for the observed cardioprotective phenotype. We provide evidence that EVs are a potential cardioprotective treatment target. Our findings will also provide the basis for a more detailed analysis of putatively relevant miRNA candidates.


Asunto(s)
Complejo CD3/inmunología , Cicatriz/tratamiento farmacológico , Leucocitos Mononucleares/efectos de los fármacos , MicroARNs/metabolismo , Infarto del Miocardio/tratamiento farmacológico , Infarto del Miocardio/inmunología , Neovascularización Fisiológica/efectos de los fármacos , Animales , Anticuerpos/inmunología , Anticuerpos/uso terapéutico , Suero Antilinfocítico/inmunología , Suero Antilinfocítico/uso terapéutico , Cardiotónicos/inmunología , Quimiocina CCL2/metabolismo , Cicatriz/inmunología , Cicatriz/prevención & control , Modelos Animales de Enfermedad , Exosomas/efectos de los fármacos , Exosomas/inmunología , Exosomas/metabolismo , Vesículas Extracelulares/efectos de los fármacos , Vesículas Extracelulares/metabolismo , Secuenciación de Nucleótidos de Alto Rendimiento , Células Endoteliales de la Vena Umbilical Humana , Humanos , Interleucina-8/metabolismo , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Masculino , MicroARNs/genética , Neovascularización Fisiológica/inmunología , Proteoma/metabolismo , Ratas , Ratas Sprague-Dawley
17.
J Innate Immun ; 12(3): 248-256, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31574508

RESUMEN

Mononuclear phagocytes, such as macrophages and microglia, are key regulators of organ homeostasis including vascularization processes. Here, we investigated the role of the suppressor of cytokine signaling 3 (SOCS3) in myeloid cells as a regulator of mononuclear phagocyte function and their interaction with endothelial cells in the context of sprouting angiogenesis. As compared to SOCS3-sufficient counterparts, SOCS3-deficient microglia and macrophages displayed an increased phagocytic activity toward primary apoptotic endothelial cells, which was associated with an enhanced expression of the opsonin growth arrest-specific 6 (Gas6), a major prophagocytic molecule. Furthermore, we found that myeloid SOCS3 deficiency significantly reduced angiogenesis in an ex vivo mouse aortic ring assay, which could be reversed by the inhibition of the Gas6 receptor Mer. Together, SOCS3 in myeloid cells regulates the Gas6/Mer-dependent phagocytosis of endothelial cells, and thereby angiogenesis-related processes. Our findings provide novel insights into the complex crosstalk between mononuclear phagocytes and endothelial cells, and may therefore provide a new platform for the development of new antiangiogenic therapies.


Asunto(s)
Apoptosis/inmunología , Células Endoteliales/inmunología , Células Mieloides/inmunología , Neovascularización Fisiológica/inmunología , Proteína 3 Supresora de la Señalización de Citocinas/deficiencia , Animales , Apoptosis/genética , Ratones , Ratones Transgénicos , Neovascularización Fisiológica/genética , Fagocitosis , Proteína 3 Supresora de la Señalización de Citocinas/inmunología
18.
Biomed Pharmacother ; 123: 109783, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31855737

RESUMEN

Acetaminophen (APAP) overdose has become the most common cause of drug-induced acute liver failure. Angiogenesis and redox homeostasis play an important role in liver protection and repair of APAP-induced acute liver injury (AILI). Hypoxia inducible factor-1 (HIF-1) is a transcription factor that plays a crucial role in regulating the expression of genes associated with angiogenesis, redox homeostasis and energy balance. Prolyl hydroxylase 2 (PHD2) predominantly hydroxylates proline residues in HIF-1α to promote its degradation. In our previous study, we reported an intrabody against PHD2 (ER-INP) that enhances angiogenesis by blocking PHD2 activity to increase HIF-1α abundance and activity. The present study was designed to explore the role and possible mechanisms of ER-INP in AILI in mice. Mice were pretreated intravenously with ER-INP before intraperitoneal injection of APAP to induce AILI. The results showed that pretreatment with ER-INP dramatically decreased the high ALT and AST activities and significantly ameliorated the centrilobular necrosis induced by APAP administration. ER-INP expression promoted angiogenesis in vivo by upregulating the mRNA and protein levels of HIF-1α target genes. Meanwhile, ER-INP pretreatment restored redox homeostasis, verified by reinforcement of PRDX4 activity and suppression of GSH depletion. This study demonstrated that ER-INP protects against AILI in part by increasing angiogenesis and maintaining redox homeostasis. These results indicate that ER-INP may provide a potential liver protection strategy against AILI in the future.


Asunto(s)
Acetaminofén/envenenamiento , Anticuerpos/inmunología , Enfermedad Hepática Inducida por Sustancias y Drogas/prevención & control , Prolina Dioxigenasas del Factor Inducible por Hipoxia/inmunología , Animales , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Enfermedad Hepática Inducida por Sustancias y Drogas/inmunología , Células HEK293 , Homeostasis/inmunología , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Neovascularización Fisiológica/inmunología , Oxidación-Reducción/efectos de los fármacos , Células RAW 264.7
19.
Front Immunol ; 10: 1445, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31293594

RESUMEN

Mast cells are classically recognized as cells that cause IgE-mediated allergic reactions. However, their ability to store and secrete vascular endothelial growth factor (VEGF) suggests a role in vascular development and tumorigenesis. The current study sought to determine if other angiogenesis-related factors, in addition to VEGF, were also secreted by human tissue-derived mast cells. Using proteome array analysis and ELISA, we found that human skin-derived mast cells spontaneously secrete CXCL16, DPPIV, Endothelin-1, GM-CSF, IL-8, MCP-1, Pentraxin 3, Serpin E1, Serpin F1, TIMP-1, Thrombospondin-1, and uPA. We identified three groups based on their dependency for stem cell factor (SCF), which is required for mast cell survival: Endothelin-1, GM-CSF, IL-8, MCP-1, and VEGF (dependent); Pentraxin 3, Serpin E1, Serpin F1, TIMP-1, and Thrombospondin-1 (partly dependent); and CXCL16, DPPIV, and uPA (independent). Crosslinking of FcεRI with multivalent antigen enhanced the secretion of GM-CSF, Serpin E1, IL-8, and VEGF, and induced Amphiregulin and MMP-8 expression. Interestingly, FcεRI signals inhibited the spontaneous secretion of CXCL16, Endothelin-1, Serpin F1, Thrombospondin-1, MCP-1 and Pentraxin-3. Furthermore, IL-6, which we previously showed could induce VEGF, significantly enhanced MCP-1 secretion. Overall, this study identified several angiogenesis-related proteins that, in addition to VEGF, are spontaneously secreted at high concentrations from human skin-derived mast cells. These findings provide further evidence supporting an intrinsic role for mast cells in blood vessel formation.


Asunto(s)
Inductores de la Angiogénesis/inmunología , Citocinas/inmunología , Mastocitos/inmunología , Neovascularización Fisiológica/inmunología , Piel/inmunología , Células Cultivadas , Humanos , Mastocitos/citología , Receptores de IgE/inmunología , Piel/citología
20.
Sci Immunol ; 4(37)2019 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-31300479

RESUMEN

Barriers between circulation and the central nervous system (CNS) play a key role in the development and modulation of CNS immune responses. Structural variations in the vasculature traversing different anatomical regions within the CNS strongly influence where and how CNS immune responses first develop. Here, we provide an overview of cerebrovascular anatomy, focusing on the blood-CNS interface and how anatomical variations influence steady-state immunology in the compartment. We then discuss how CNS vasculature is affected by and influences the development of different pathophysiological states, such as CNS autoimmune disease, cerebrovascular injury, cerebral ischemia, and infection.


Asunto(s)
Anatomía , Sistema Nervioso Central/inmunología , Neovascularización Fisiológica/inmunología , Animales , Arterias Cerebrales/inmunología , Circulación Cerebrovascular/inmunología , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...