Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Front Immunol ; 12: 684657, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34489935

RESUMEN

Pulmonary arterial hypertension (PAH) is a chronic, incurable condition characterized by pulmonary vascular remodeling, perivascular inflammation, and right heart failure. Regulatory T cells (Tregs) stave off autoimmunity, and there is increasing evidence for their compromised activity in the inflammatory milieu of PAH. Abnormal Treg function is strongly correlated with a predisposition to PAH in animals and patients. Athymic Treg-depleted rats treated with SU5416, an agent causing pulmonary vascular injury, develop PAH, which is prevented by infusing missing CD4+CD25highFOXP3+ Tregs. Abnormal Treg activity may also explain why PAH disproportionately affects women more than men. This mini review focuses on the role of Tregs in PAH with a special view to sexual dimorphism and the future promise of Treg therapy.


Asunto(s)
Hipertensión Arterial Pulmonar/inmunología , Hipertensión Arterial Pulmonar/prevención & control , Linfocitos T Reguladores/inmunología , Lesiones del Sistema Vascular/inmunología , Lesiones del Sistema Vascular/prevención & control , Animales , Autoinmunidad , Endotelio Vascular/inmunología , Endotelio Vascular/patología , Humanos , Indoles/efectos adversos , Hipertensión Arterial Pulmonar/patología , Pirroles/efectos adversos , Ratas , Caracteres Sexuales , Lesiones del Sistema Vascular/patología
2.
Immunology ; 164(2): 372-385, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34077562

RESUMEN

Plasmacytoid dendritic cells (pDCs) play a key role in the initiation and amplification of systemic lupus erythematosus (SLE)-associated vascular injury. In this study, we found that dsDNA induced dose- and time-dependent increase in IFN-α and Toll-like receptor 7 (TLR7), TLR9 and IRF7 expression in pDCs. Co-cultured circulating endothelial cells (ECs) with activated pDCs significantly decreased proliferation, tube formation and migration in ECs. The elevated level of cellular IFN-α increased cell adhesion, promoted cell apoptosis, induced cell senescence and arrested cells at G0/G1 phase of endothelial progenitor cells (EPCs). Additionally, the co-culture system activated MAPK and inactivated PI3K. Pristane was used to establish a in vivo SLE-like mouse model. Importantly, we showed that INF-α-neutralizing antibody (IFN-α-NA) rescued all the changes induced by IFN-α in vitro and prevented vascular injury in pristane-induced SLE model in vivo. In conclusion, we confirmed that activated pDCs promoted vascular damage and the dysfunction of ECs/EPCs via IFN-α production. IFN-α-neutralizing antibody may be a clinical implication for preventing vascular injury. PI3K signalling and AMPK signalling were associated with SLE-associated vascular functions.


Asunto(s)
Proteínas Quinasas Activadas por AMP/inmunología , Anticuerpos Neutralizantes/inmunología , Inflamación/inmunología , Interferón-alfa/inmunología , Lupus Eritematoso Sistémico/inmunología , Fosfatidilinositol 3-Quinasas/inmunología , Lesiones del Sistema Vascular/inmunología , Animales , Células Cultivadas , Células Dendríticas/inmunología , Células Endoteliales/inmunología , Femenino , Mediadores de Inflamación/inmunología , Ratones , Ratones Endogámicos C57BL , Receptor Toll-Like 7/inmunología
3.
J Leukoc Biol ; 109(1): 35-47, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33242368

RESUMEN

The SARS-CoV-2 pandemic has led to hundreds of thousands of deaths and billions of dollars in economic damage. The immune response elicited from this virus is poorly understood. An alarming number of cases have arisen where COVID-19 patients develop complications on top of the symptoms already associated with SARS, such as thrombosis, injuries of vascular system, kidney, and liver, as well as Kawasaki disease. In this review, a bioinformatics approach was used to elucidate the immune response triggered by SARS-CoV-2 infection in primary human lung epithelial and transformed human lung alveolar. Additionally, examined the potential mechanism behind several complications that have been associated with COVID-19 and determined that a specific cytokine storm is leading to excessive neutrophil recruitment. These neutrophils are directly leading to thrombosis, organ damage, and complement activation via neutrophil extracellular trap release.


Asunto(s)
COVID-19/inmunología , SARS-CoV-2/inmunología , Transducción de Señal/inmunología , Trombosis/inmunología , Lesiones del Sistema Vascular/inmunología , COVID-19/patología , Citocinas/inmunología , Humanos , Síndrome Mucocutáneo Linfonodular/inmunología , Síndrome Mucocutáneo Linfonodular/patología , Síndrome Mucocutáneo Linfonodular/virología , Alveolos Pulmonares/inmunología , Alveolos Pulmonares/patología , Alveolos Pulmonares/virología , Trombosis/patología , Trombosis/virología , Lesiones del Sistema Vascular/patología , Lesiones del Sistema Vascular/virología
4.
Front Immunol ; 11: 575890, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33042158

RESUMEN

Thrombomodulin (TM), which is predominantly expressed on the endothelium, plays an important role in maintaining vascular homeostasis by regulating the coagulation system. Intravascular injury and inflammation are complicated physiological processes that are induced by injured endothelium-mediated pro-coagulant signaling, necrotic endothelial- and blood cell-derived damage-associated molecular patterns (DAMPs), and DAMP-mediated inflammation. During the hypercoagulable state after endothelial injury, TM is released into the intravascular space by proteolytic cleavage of the endothelium component. Recombinant TM (rTM) is clinically applied to patients with disseminated intravascular coagulation, resulting in protection from tissue injury. Recent studies have revealed that rTM functions as an inflammatory regulator beyond hemostasis through various molecular mechanisms. More specifically, rTM neutralizes DAMPs, including histones and high mobility group box 1 (HMGB1), suppresses excessive activation of the complement system, physiologically protects the endothelium, and influences both innate and acquired immunity. Neutrophil extracellular traps (NETs) promote immunothrombosis by orchestrating platelets to enclose infectious invaders as part of the innate immune system, but excessive immunothrombosis can cause intravascular injury. However, rTM can directly and indirectly regulate NET formation. Furthermore, rTM interacts with mediators of acquired immunity to resolve vascular inflammation. So far, rTM has shown good efficacy in suppressing inflammation in various experimental models, including thrombotic microangiopathy, sterile inflammatory disorders, autoimmune diseases, and sepsis. Thus, rTM has the potential to become a novel tool to regulate intravascular injury via pleiotropic effects.


Asunto(s)
Coagulación Sanguínea , Coagulación Intravascular Diseminada/metabolismo , Endotelio Vascular/metabolismo , Inflamación/metabolismo , Trombomodulina/metabolismo , Trombosis/metabolismo , Lesiones del Sistema Vascular/metabolismo , Animales , Antiinflamatorios/uso terapéutico , Plaquetas/metabolismo , Coagulación Intravascular Diseminada/sangre , Coagulación Intravascular Diseminada/inmunología , Endotelio Vascular/inmunología , Trampas Extracelulares/metabolismo , Humanos , Inflamación/sangre , Inflamación/tratamiento farmacológico , Inflamación/inmunología , Mediadores de Inflamación/metabolismo , Transducción de Señal , Trombomodulina/uso terapéutico , Trombosis/sangre , Trombosis/inmunología , Lesiones del Sistema Vascular/sangre , Lesiones del Sistema Vascular/inmunología
5.
Mol Immunol ; 118: 142-152, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31884386

RESUMEN

Antibody-mediated rejection (AbMR) adversely affects long-term graft survival in kidney transplantation. Currently, the diagnosis of AbMR requires a kidney biopsy, and detection of complement C4d deposition in the allograft is one of the diagnostic criteria. Complement activation also generates several soluble fragments which could potentially provide non-invasive biomarkers of the process. Furthermore, microvesicles released into the plasma from injured cells can serve as biomarkers of vascular injury. To explore whether soluble complement fragments or complement fragments bound to endothelial microvesicles can be used to non-invasively detect AbMR, we developed an in vitro model in which human endothelial cells were exposed to anti-HLA antibodies and complement sufficient serum. We found that complement fragments C4a and sC5b-9 were increased in the supernatants of cells exposed to complement-sufficient serum compared to cells treated complement-deficient serum. Furthermore, complement activation on the cell surface was associated with the release of microvesicles bearing C4 and C3 fragments. We next measured these analytes in plasma from kidney transplant recipients with biopsy-proven acute AbMR (n = 9) and compared the results with those from transplant recipients who also had impaired allograft function but who did not have AbMR (n = 30). Consistent with the in vitro results, complement fragments C4a and Ba were increased in plasma from patients with AbMR compared to control subjects (P < 0.001 and P < 0.01, respectively). Endothelial microvesicle counts were not increased in patients with AbMR, however, and the number of microvesicles with C4 and C3 bound to the surface was actually lower compared to control subjects (both P < 0.05). Our results suggest that plasma complement activation fragments may be useful as non-invasive biomarkers of antibody-mediated complement activation within the allograft. Complement-opsonized endothelial microvesicles are decreased in patients with AbMR, possibly due to enhanced clearance of microvesicles opsonized with C3 and C4 fragments.


Asunto(s)
Anticuerpos/inmunología , Proteínas del Sistema Complemento/inmunología , Células Endoteliales/inmunología , Lesiones del Sistema Vascular/inmunología , Adulto , Aloinjertos/inmunología , Biomarcadores/sangre , Biopsia , Células Cultivadas , Activación de Complemento/inmunología , Femenino , Rechazo de Injerto/inmunología , Humanos , Riñón/inmunología , Trasplante de Riñón/métodos , Masculino , Persona de Mediana Edad , Trasplante Homólogo/métodos
6.
Mucosal Immunol ; 13(2): 357-370, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31776431

RESUMEN

Parasitic helminths cause significant damage as they migrate through host tissues to complete their life cycle. While chronic helminth infections are characterized by a well-described Type 2 immune response, the early, tissue-invasive stages are not well understood. Here we investigate the immune pathways activated during the early stages of Heligmosomoides polygyrus bakeri (Hpb), a natural parasitic roundworm of mice. In contrast to the Type 2 immune response present at later stages of infection, a robust Type 1 immune signature including IFNg production was dominant at the time of parasite invasion and granuloma formation. This early response was associated with an accumulation of activated Natural Killer (NK) cells, with no increase of other innate lymphoid cell populations. Parabiosis and confocal microscopy studies indicated that NK cells were recruited from circulation to the small intestine, where they surrounded parasitic larvae. NK cell recruitment required IFNγ receptor signaling, but was independent of CXCR3 expression. The depletion of tissue-infiltrating NK cells altered neither worm burden nor parasite fitness, but increased vascular injury, suggesting a role for NK cells in mediating tissue protection. Together, these data identify an unexpected role for NK cells in promoting disease tolerance during the invasive stage of an enteric helminth infection.


Asunto(s)
Tracto Gastrointestinal/inmunología , Vigilancia Inmunológica , Intestinos/inmunología , Células Asesinas Naturales/inmunología , Nematospiroides dubius/fisiología , Infecciones por Strongylida/inmunología , Células TH1/metabolismo , Lesiones del Sistema Vascular/inmunología , Animales , Movimiento Celular , Femenino , Inmunidad Innata , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Parabiosis , Receptores CXCR/metabolismo , Receptores de Interferón/genética , Receptores de Interferón/metabolismo , Transducción de Señal , Proteínas de Dominio T Box/metabolismo , Células TH1/inmunología , Receptor de Interferón gamma
7.
Hum Immunol ; 80(8): 608-613, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31029511

RESUMEN

Ischemic, immunologic or pharmacological stressors can induce vascular injury and endothelial apoptosis in organ donors, in transplant candidates due to the impact of end stage organ failure on the vasculature, and in association with peri-transplantation events. Vascular injury may shape innate and adaptive immune responses, leading to dysregulation in the balance between tolerance and immunoreactivity to vascular-derived antigens. Mounting evidence shows that the early stages of apoptosis, characterized by the absence of membrane permeabilization, are prone to trigger various modes of intercellular communication allowing neoantigen production, exposure, or both. In this review, we present the evidence for the release of LG3, an immunogenic fragment of perlecan, as a consequence of caspase-3 dependent vascular apoptosis leading to the genesis of anti-LG3 autoantibodies and the consequences of these autoantibodies in native and transplanted kidneys.


Asunto(s)
Autoanticuerpos/metabolismo , Células Endoteliales/fisiología , Rechazo de Injerto/inmunología , Proteoglicanos de Heparán Sulfato/inmunología , Trasplante de Riñón , Lesiones del Sistema Vascular/inmunología , Inmunidad Adaptativa , Animales , Apoptosis , Autoinmunidad , Caspasa 3/metabolismo , Humanos , Inmunidad Innata
8.
Med Sci Monit ; 24: 8322-8332, 2018 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-30451213

RESUMEN

BACKGROUND This study investigated the distribution and features of natural killer T (NKT) cells in the peripheral blood of diabetic patients, and their regulatory roles on vascular endothelial cells. MATERIAL AND METHODS Peripheral lymphocytes were isolated from diabetic patients. NKT cell distribution, proportion, and surface and intracellular markers were detected with flow cytometry. Peripheral blood-derived NKT cells were isolated and co-cultured with human umbilical vein endothelial cells (HUVECs). Proliferation and migration of HUVECs were assessed with the CCK-8 assay and the Transwell chamber assay. RESULTS The ratios of CD3-CD56+ NK and CD3+CD56+ NKT cells in the peripheral blood of patients with type II diabetes were significantly elevated. The expression levels of NKp30, NKG2D, and NKp44 on the surface were increased in the CD3+CD56+ NKT cells, while the expression levels of NKG2A and 158b were significantly downregulated. The expression level of granzymes in the peripheral blood-derived NKT cells were not changed in patients with type II diabetes, but the expression levels of IFNg and IL-4 were significantly increased. However, after co-culture with NKT cells derived from the peripheral blood of diabetic patients, the proliferation and migration of HUVECs were significantly inhibited, and was restored by treatment with IL-4 antibody. In addition, the IL-4 stimulus inhibited the proliferation and migration of HUVECs. ls were not changed in patients with type II diabetes, while the expression levels of IFNγ and IL-4 were significantly increased. However, after co-cultured with NKT cells derived from the peripheral blood of diabetic patients, the proliferation and migration of HUVECs were significantly inhibited, which could significantly restored by the treatment of IL-4 antibody. In addition, the IL-4 stimulus could down-regulate the proliferation and migration of HUVECs.  CONCLUSIONS Peripheral blood NKT cells are increased and activated in diabetes. NKT cells inhibit the proliferation and migration of HUVECs by secreting IL-4, thereby inducing vascular injuries.


Asunto(s)
Diabetes Mellitus Tipo 2/inmunología , Células T Asesinas Naturales/inmunología , Células T Asesinas Naturales/fisiología , Adulto , Complejo CD3/inmunología , Estudios de Casos y Controles , Diabetes Mellitus Tipo 2/complicaciones , Femenino , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Interleucina-4/inmunología , Masculino , Persona de Mediana Edad , Células T Asesinas Naturales/metabolismo , Lesiones del Sistema Vascular/inmunología
9.
Front Immunol ; 9: 1938, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30210495

RESUMEN

Given the importance of high-mobility group box 1 (HMGB1) and 5-lipoxygenase (5-LO) signaling in vascular inflammation, we investigated the role of leukotriene signaling in monocytes on monocyte-to-macrophage differentiation (MMD) induced by HMGB1, and on vascular inflammation and subsequent intimal hyperplasia in a mouse model of wire-injured femoral artery. In cultured primary bone marrow-derived cells (BMDCs) stimulated with HMGB1, the number of cells with macrophage-like morphology was markedly increased in association with an increased expression of CD11b/Mac-1, which were attenuated in cells pre-treated with Zileuton, a 5-LO inhibitor as well as in 5-LO-deficient BMDCs. Of various leukotriene receptor inhibitors examined, which included leukotriene B4 receptors (BLTRs) and cysteinyl leukotriene receptors (cysLTRs), the BLTR1 inhibitor (U75302) exclusively suppressed MMD induction by HMGB1. The importance of BLTR1 in HMGB1-induced MMD was also observed in BMDCs isolated from BLTR1-deficient mice and BMDCs transfected with BLTR1 siRNA. Although leukotriene B4 (LTB4) had minimal direct effects on MMD in control and 5-LO-deficient BMDCs, MMD attenuation by HMGB1 in 5-LO-deficient BMDCs was significantly reversed by exogenous LTB4, but not in BLTR1-deficient BMDCs, suggesting that LTB4/BLTR1-mediated priming of monocytes is a prerequisite of HMGB1-induced MMD. In vivo, both macrophage infiltration and intimal hyperplasia in our wire-injured femoral artery were markedly attenuated in BLTR1-deficient mice as compared with wild-type controls, but these effects were reversed in BLTR1-deficient mice transplanted with monocytes from control mice. These results suggest that BLTR1 in monocytes is a pivotal player in MMD with subsequent macrophage infiltration into neointima, leading to vascular remodeling after vascular injury.


Asunto(s)
Alcoholes Grasos/farmacología , Arteria Femoral , Glicoles/farmacología , Monocitos , Receptores de Leucotrieno B4 , Remodelación Vascular , Lesiones del Sistema Vascular , Vasculitis , Animales , Arteria Femoral/inmunología , Arteria Femoral/lesiones , Arteria Femoral/patología , Hiperplasia , Macrófagos/inmunología , Macrófagos/patología , Ratones , Ratones Noqueados , Monocitos/inmunología , Monocitos/patología , Receptores de Leucotrieno B4/antagonistas & inhibidores , Receptores de Leucotrieno B4/genética , Receptores de Leucotrieno B4/inmunología , Remodelación Vascular/efectos de los fármacos , Remodelación Vascular/genética , Remodelación Vascular/inmunología , Lesiones del Sistema Vascular/tratamiento farmacológico , Lesiones del Sistema Vascular/genética , Lesiones del Sistema Vascular/inmunología , Lesiones del Sistema Vascular/patología , Vasculitis/tratamiento farmacológico , Vasculitis/genética , Vasculitis/inmunología , Vasculitis/patología
10.
Clin Immunol ; 194: 26-33, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29936303

RESUMEN

AIMS: Adaptive immunity is critical in vascular remodelling following arterial injury. We hypothesized that acute changes in T cells at a percutaneous transluminal angioplasty (PTA) site could serve as an index of their potential interaction with the injured vascular wall. METHODS AND RESULTS: T cell subsets were characterised in 45 patients with Rutherford 3-4 peripheral artery disease (PAD) undergoing PTA. Direct angioplasty catheter blood sampling was performed before and immediately after the procedure. PTA was associated with an acute reduction of α/ß-TcR CD8+ T cells. Further characterisation revealed significant reduction in pro-atherosclerotic CD28nullCD57+ T cells, effector (CD45RA+CCR7-) and effector memory (CD45RA-CCR7-) cells, in addition to cells bearing activation (CD69, CD38) and tissue homing/adhesion markers (CD38, CCR5). CONCLUSIONS: The acute reduction observed here is likely due to the adhesion of cells to the injured vascular wall, suggesting that immunosenescent, activated effector CD8+ cells have a role in the early vascular injury immune response following PTA in PAD patients.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Enfermedad Arterial Periférica/inmunología , Subgrupos de Linfocitos T/inmunología , Lesiones del Sistema Vascular/inmunología , Anciano , Antígenos CD/inmunología , Femenino , Humanos , Memoria Inmunológica/inmunología , Antígenos Comunes de Leucocito/inmunología , Masculino , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología
11.
J Invest Dermatol ; 138(10): 2195-2204, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29605671

RESUMEN

Aryl hydrocarbon receptor (AhR), which was discovered as a receptor for environmental concomitants, plays an important role widely in the immune system. In this study, we assessed AhR involvement in immune-complex-mediated vascular injury by examining the reverse-passive Arthus reaction in AhR heterozygous knockout (AhR+/-) mice. In the cutaneous Arthus reaction, dermal edema was severer in AhR+/- mice than in wild-type mice. The number of infiltrating neutrophils and mRNA expression levels of CXC chemokine ligand 1 and IL-6 were also increased in AhR+/- mice. Similarly, in the peritoneal Arthus reaction, infiltration of neutrophils was increased in AhR+/- mice. Peritoneal macrophages from AhR+/- mice expressed higher levels of Fcγ receptor III and produced higher levels of CXC chemokine ligand 1 and IL-6 after immune complex treatment. In addition, AhR occupied the promoter regions of Fcγ receptor III gene in peritoneal macrophages in a ligand-dependent manner. Depletion of macrophages reduced the cutaneous Arthus reaction in AhR+/- mice, and adoptive transfer of AhR+/- mice macrophages into wild-type mice exacerbated the peritoneal Arthus reaction. Furthermore, AhR expression was decreased and Fcγ receptor III expression was increased in CD14+ monocytes in peripheral blood from patients with immune-complex-mediated vasculitis compared with those from healthy controls. These results suggest that downregulation of AhR is associated with the exacerbation of immune-complex-mediated vascular injury.


Asunto(s)
Macrófagos/metabolismo , Receptores de IgG/biosíntesis , Regulación hacia Arriba , Lesiones del Sistema Vascular/inmunología , Adulto , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Macrófagos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Receptores de Hidrocarburo de Aril/deficiencia , Lesiones del Sistema Vascular/metabolismo , Lesiones del Sistema Vascular/patología
12.
PLoS Comput Biol ; 14(2): e1005876, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29447154

RESUMEN

Sepsis, a manifestation of the body's inflammatory response to injury and infection, has a mortality rate of between 28%-50% and affects approximately 1 million patients annually in the United States. Currently, there are no therapies targeting the cellular/molecular processes driving sepsis that have demonstrated the ability to control this disease process in the clinical setting. We propose that this is in great part due to the considerable heterogeneity of the clinical trajectories that constitute clinical "sepsis," and that determining how this system can be controlled back into a state of health requires the application of concepts drawn from the field of dynamical systems. In this work, we consider the human immune system to be a random dynamical system, and investigate its potential controllability using an agent-based model of the innate immune response (the Innate Immune Response ABM or IIRABM) as a surrogate, proxy system. Simulation experiments with the IIRABM provide an explanation as to why single/limited cytokine perturbations at a single, or small number of, time points is unlikely to significantly improve the mortality rate of sepsis. We then use genetic algorithms (GA) to explore and characterize multi-targeted control strategies for the random dynamical immune system that guide it from a persistent, non-recovering inflammatory state (functionally equivalent to the clinical states of systemic inflammatory response syndrome (SIRS) or sepsis) to a state of health. We train the GA on a single parameter set with multiple stochastic replicates, and show that while the calculated results show good generalizability, more advanced strategies are needed to achieve the goal of adaptive personalized medicine. This work evaluating the extent of interventions needed to control a simplified surrogate model of sepsis provides insight into the scope of the clinical challenge, and can serve as a guide on the path towards true "precision control" of sepsis.


Asunto(s)
Citocinas/metabolismo , Inmunidad Innata , Sepsis/fisiopatología , Síndrome de Respuesta Inflamatoria Sistémica/fisiopatología , Algoritmos , Sangre/metabolismo , Ensayos Clínicos como Asunto , Biología Computacional , Simulación por Computador , Endotelio Vascular/metabolismo , Humanos , Inflamación/fisiopatología , Modelos Biológicos , Modelos Estadísticos , Mortalidad , Oxígeno/metabolismo , Probabilidad , Lenguajes de Programación , Sepsis/complicaciones , Procesos Estocásticos , Estados Unidos , Lesiones del Sistema Vascular/inmunología
13.
Drug Deliv Transl Res ; 8(4): 945-953, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28656488

RESUMEN

The innate immunity system plays a critical role in vascular repair and restenosis development. Liposomes encapsulating bisphosphonates (LipBPs), but not free BPs, suppress neointima formation following vascular injury mediated in part by monocytes. The objective of this study was to elucidate the role of monocyte subpopulations on vascular healing following LipBP treatment. The potency- and dose-dependent treatment effect of clodronate (CLOD) and alendronate (ALN) liposomes on restenosis inhibition, total monocyte depletion, and monocytes subpopulation was studied. Rats subjected to carotid injury were treated by a single IV injection of LipBPs at the time of injury. Low- and high-dose LipALN treatment (3 and 10 mg/kg, respectively) resulted in a dose-dependent effect on restenosis development after 30 days. Both doses of LipALN resulted in a dose-dependent inhibition of restenosis, but only high dose of LipALN depleted monocytes (-60.1 ± 4.4%, 48 h post injury). Although LipCLOD treatment (at an equivalent potency to 3 mg/kg alendronate) significantly reduced monocyte levels (72.1 ± 6%), no restenosis inhibition was observed. The major finding of this study is the correlation found between monocyte subclasses and restenosis inhibition. Non-classical monocyte (NCM) levels were found higher in LipALN-treated rats, but lower in LipCLOD-treated rats, 24 h after injury and treatment. We suggest that the inhibition of circulating monocyte subpopulations is the predominant mechanism by which LipBPs prevent restenosis. The effect of LipBP treatment on the monocyte subpopulation correlates with the dose and potency of LipBPs.


Asunto(s)
Alendronato/administración & dosificación , Traumatismos de las Arterias Carótidas/tratamiento farmacológico , Ácido Clodrónico/administración & dosificación , Reestenosis Coronaria/prevención & control , Monocitos/inmunología , Lesiones del Sistema Vascular/tratamiento farmacológico , Animales , Traumatismos de las Arterias Carótidas/inmunología , Liposomas , Masculino , Ratas , Lesiones del Sistema Vascular/inmunología
14.
Mol Med Rep ; 17(1): 1389-1395, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29115487

RESUMEN

Heat shock protein family A member 12B (HSPA12B) is a heat shock protein primarily expressed in endothelial cells. Our previous study showed that it was protective against endothelial injury induced by lipopolysaccharide (LPS). The present study was performed to investigate whether micro (mi)RNA was involved in HSPA12B expression in endothelial cells challenged by LPS. We first screened the miRNA candidates potentially related to HSPA12B by bioinformatics analysis. Then the mimics of the miRNA candidates were transfected into human umbilical vein endothelial cells (HUVECs) to investigate the miRNAs that negatively regulated HSPA12B expression. The miRNA expression was also determined in LPS­stimulated HUVECs. Dual luciferase activity assay was performed to confirm the relationship between the candidate miRNA and HSPA12B. Role of nuclear factor (NF)­κB in the miRNA expression was investigated by using its inhibitor. Finally, the role of the miRNA on LPS induced injury was investigated. Eleven miRNAs were screened by bioinformatics analysis and 4 of them could inhibit HSPA12B expression at both mRNA and protein levels. Among the 4 miRNA candidates, only miR­4505 was highly expressed in HUVECs stimulated by LPS. Luciferase analysis showed that miR­4505 directly interacted with the 3'untranslated region of HSPA12B. LPS­induced upregulation of miR­4505 was blocked by NF­κB inhibitor. Transfection with miR­4505 mimics reduced the transendothelial electrical resistance and vascular endothelial­cadherin expression. The scratch test demonstrated that miR­4505 inhibited endothelial migration capacity. In conclusion, miR­4505 downregulates the expression of HSPA12B and aggravates the LPS­induced vascular endothelial cell injury.


Asunto(s)
Proteínas HSP70 de Choque Térmico/genética , Células Endoteliales de la Vena Umbilical Humana/metabolismo , MicroARNs/fisiología , Regiones no Traducidas 3' , Secuencia de Bases , Sitios de Unión , Células Cultivadas , Endotelio Vascular/inmunología , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Expresión Génica , Proteínas HSP70 de Choque Térmico/metabolismo , Células Endoteliales de la Vena Umbilical Humana/inmunología , Humanos , Lipopolisacáridos/farmacología , MicroARNs/genética , Interferencia de ARN/inmunología , Regulación hacia Arriba , Lesiones del Sistema Vascular/inmunología , Lesiones del Sistema Vascular/metabolismo
15.
Transplant Proc ; 49(6): 1467-1476, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28736025

RESUMEN

INTRODUCTION: Vascular progenitor cells contribute to repair of injured vasculature. In this study, we aimed to investigate the role of bone marrow-derived cells in the intimal formation after arterial injury. METHODS AND RESULTS: Balloon injury of the femoral artery of wild-type mice was followed by local delivery of bone marrow-derived cells from GFP transgenic mice. The arteries were collected 1, 4, 7, and 14 days after injury and studied for morphology, localization, and phenotypes of delivered cells. Bone marrow-derived cells were present in the intima only at the early stages of arterial injury and expressed endothelial progenitor cell markers (CD31, CD34, and VEGFR-2). In the areas where intima was thicker, bone marrow-derived cells differentiated to intimal smooth muscle cells but they did not fuse with intimal cells. Delivery of CD34+ cells contributed to a 1.5-fold inhibition of intimal hyperplasia. CONCLUSION: Bone marrow-derived endothelial cells differentiated but did not fuse with vascular smooth muscle cells at the early stages of intimal formation and contributed to intimal hyperplasia.


Asunto(s)
Antígenos CD34/inmunología , Células de la Médula Ósea/fisiología , Trasplante de Médula Ósea , Lesiones del Sistema Vascular/terapia , Animales , Células de la Médula Ósea/inmunología , Diferenciación Celular , Células Progenitoras Endoteliales/fisiología , Arteria Femoral/lesiones , Hiperplasia , Masculino , Ratones , Ratones Transgénicos , Miocitos del Músculo Liso/fisiología , Células Madre/fisiología , Túnica Íntima/lesiones , Lesiones del Sistema Vascular/inmunología
16.
Sci Rep ; 6: 35387, 2016 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-27759053

RESUMEN

Percutaneous coronary intervention is widely adopted to treat patients with coronary artery disease. However, restenosis remains an unsolved clinical problem after vascular interventions. The role of the systemic and local immune response in the development of restenosis is not fully understood. Hence, the aim of the current study was to investigate the role of the human immune system on subsequent neointima formation elicited by vascular injury in a humanized mouse model. Immunodeficient NOD.Cg-PrkdcscidIL2rgtm1Wjl(NSG) mice were reconstituted with human (h)PBMCs immediately after both carotid wire and femoral cuff injury were induced in order to identify how differences in the severity of injury influenced endothelial regeneration, neointima formation, and homing of human inflammatory and progenitor cells. In contrast to non-reconstituted mice, hPBMC reconstitution reduced neointima formation after femoral cuff injury whereas hPBMCs promoted neointima formation after carotid wire injury 4 weeks after induction of injury. Neointimal endothelium and smooth muscle cells in the injured arteries were of mouse origin. Our results indicate that the immune system may differentially respond to arterial injury depending on the severity of injury, which may also be influenced by the intrinsic properties of the arteries themselves, resulting in either minimal or aggravated neointima formation.


Asunto(s)
Traumatismos de las Arterias Carótidas/inmunología , Arteria Femoral/inmunología , Oclusión de Injerto Vascular/inmunología , Leucocitos Mononucleares/inmunología , Lesiones del Sistema Vascular/inmunología , Animales , Traumatismos de las Arterias Carótidas/parasitología , Traumatismos de las Arterias Carótidas/terapia , Modelos Animales de Enfermedad , Arteria Femoral/lesiones , Arteria Femoral/trasplante , Oclusión de Injerto Vascular/fisiopatología , Humanos , Leucocitos Mononucleares/trasplante , Ratones , Ratones SCID/inmunología , Ratones SCID/lesiones , Miocitos del Músculo Liso/inmunología , Miocitos del Músculo Liso/patología , Neointima/inmunología , Neointima/fisiopatología , Lesiones del Sistema Vascular/fisiopatología , Lesiones del Sistema Vascular/terapia
17.
J Vasc Surg ; 64(4): 1084-92, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27666446

RESUMEN

OBJECTIVE: Vascular restenosis remains a major obstacle to long-term success after vascular intervention. Circulating progenitor cells have been implicated in restenosis, and yet it has remained unclear if these cells, particularly nonendothelial progenitors, have an active role in this pathologic process. We hypothesized that circulating CD34(+)/c-kit(+) progenitors would increase after vascular injury, mirrored by changes in the injury signal, stromal cell-derived factor 1α (sdf1α). We further postulated that an antibody-based depletion would mitigate progenitor surge and, in turn, reduce restenosis in a murine model. METHODS: C57BL6 mice underwent wire injury of the femoral artery and were compared with mice with sham surgery and vessel ligation by flow cytometry as well as by sdf1α enzyme-linked immunosorbent assay of peripheral blood. Next, injured C57BL6 mice treated with a depleting antibody toward the progenitor marker sca-1 or with an isotype control were compared in terms of sdf1α as well as enumeration of progenitors. At 28 days, restenosis was quantified between sca-1- and isotype-treated animals. RESULTS: Wire injury generated an increase in sdf1α as well as a surge of CD34(+)/c-kit(+) progenitors relative to nonsurgical controls (P = .005). Treatment with sca-1 antibody ablated the peripheral surge compared with isotype-treated, injured animals (P = .02), and sca progenitor depletion reduced the 28-day intima to media ratio in a statistically significant fashion compared with either nontreated (P = .04) or isotype-treated (P = .036) animals. CONCLUSIONS: Our study has demonstrated that sca-1 antibody reduces both progenitor surge and vascular restenosis after endoluminal vascular injury in a murine model. This suggests that circulating progenitors play an active role in restenotic disease.


Asunto(s)
Anticuerpos/farmacología , Antígenos CD34/metabolismo , Arteria Femoral/efectos de los fármacos , Células Madre Hematopoyéticas/efectos de los fármacos , Proteínas de la Membrana/antagonistas & inhibidores , Neointima , Proteínas Proto-Oncogénicas c-kit/metabolismo , Lesiones del Sistema Vascular/tratamiento farmacológico , Animales , Antígenos Ly/inmunología , Antígenos Ly/metabolismo , Constricción Patológica , Modelos Animales de Enfermedad , Arteria Femoral/inmunología , Arteria Femoral/lesiones , Arteria Femoral/metabolismo , Células Madre Hematopoyéticas/inmunología , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/patología , Hiperplasia , Proteínas de la Membrana/inmunología , Proteínas de la Membrana/metabolismo , Ratones Endogámicos C57BL , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Lesiones del Sistema Vascular/inmunología , Lesiones del Sistema Vascular/metabolismo , Lesiones del Sistema Vascular/patología
18.
Acta Anaesthesiol Taiwan ; 53(1): 23-8, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25837846

RESUMEN

Cytokines are key modulators of inflammatory responses, and play an important role in the defense and repair mechanisms following trauma. After traumatic injury, an immuno-inflammatory response is initiated immediately, and cytokines rapidly appear and function as a regulator of immunity. In pathologic conditions, imbalanced cytokines may provide systemic inflammatory responses or immunosuppression. Expression of perioperative cytokines vary by different intensities of surgical trauma and types of anesthesia and anesthetic agents. Inflammatory cytokines play important roles in postoperative organ dysfunction including central nervous system, cardiovascular, lung, liver, and kidney injury. Inhibition of cytokines could protect against traumatic injury in some circumstances, therefore cytokine inhibitors or antagonists might have the potential for reducing postoperative tissue/organ dysfunction. Cytokines are also involved in wound healing and post-traumatic pain. Application of cytokines for the improvement of surgical wound healing has been reported. Anesthesia-related immune response adjustment might reduce perioperative morbidity because it reduces proinflammatory cytokine expression; however, the overall effects of anesthetics on postoperative immune-inflammatory responses needs to be further investigated.


Asunto(s)
Citocinas/fisiología , Inflamación/inmunología , Lesión Renal Aguda/inmunología , Lesión Pulmonar Aguda/inmunología , Lesiones Traumáticas del Encéfalo/inmunología , Humanos , Periodo Perioperatorio , Lesiones del Sistema Vascular/inmunología , Cicatrización de Heridas
19.
Thromb Haemost ; 113(6): 1224-35, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25880990

RESUMEN

Leukocytes recruitment to thrombi supports an intimate cellular interaction leading to the enhancement of pro-coagulant functions and pro-inflammatory responses at site of vascular injury. Recent observations of neutrophil extracellular traps (NETs) formation and its mutual reactions with platelet thrombi adds more clinical interest to the growing body of knowledge in the field of platelet-leukocyte cross-talk. However, having considered thrombus as a barrier between leukocytes and injured endothelium, the full inflammatory roles of these cells during thrombosis is still ill defined. The most recent observation of neutrophils migration into the thrombi is a phenomenon that highlights the inflammatory functions of leukocytes at the site of injury. It has been hypothesised that leukocytes migration might be associated with the conveyance of highly reactive pro-inflammatory and/or pro-coagulant mediators to sites of vascular injury. In addition, the evidence of neutrophils migration into arterial thrombi following traumatic and ischaemia-reperfusion injury highlights the already described role of these cells in atherosclerosis. Regardless of the mechanisms behind leukocyte migration, whether these migrated cells benefit normal homeostasis by their involvement in wound healing and vascular rebuilding or they increase unwilling inflammatory responses, could be of interest for future researches that provide new insight into biological importance of leukocyte recruitment to thrombi.


Asunto(s)
Plaquetas/metabolismo , Vasos Sanguíneos/metabolismo , Quimiotaxis de Leucocito , Infiltración Neutrófila , Neutrófilos/metabolismo , Trombosis/metabolismo , Lesiones del Sistema Vascular/metabolismo , Animales , Coagulación Sanguínea , Plaquetas/inmunología , Vasos Sanguíneos/inmunología , Vasos Sanguíneos/patología , Adhesión Celular , Trampas Extracelulares/metabolismo , Humanos , Mediadores de Inflamación/metabolismo , Rodamiento de Leucocito , Neutrófilos/inmunología , Selectina-P/metabolismo , Activación Plaquetaria , Transducción de Señal , Trombosis/sangre , Trombosis/inmunología , Trombosis/patología , Remodelación Vascular , Lesiones del Sistema Vascular/sangre , Lesiones del Sistema Vascular/inmunología , Lesiones del Sistema Vascular/patología , Cicatrización de Heridas
20.
PLoS One ; 10(2): e0117616, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25692290

RESUMEN

Citrus fruits are thought to have inhibitory effects on oxidative stress, thereby attenuating the onset and progression of cancer and cardiovascular disease; however, there are few reports assessing their effect on vascular remodeling. Here, we investigated the effect of drinking the juice of two different citrus fruits on vascular neointima formation using a cuff-induced vascular injury mouse model. Male C57BL6 mice were divided into five groups as follows: 1) Control (water) (C), 2) 10% Citrus unshiu (CU) juice (CU10), 3) 40% CU juice (CU40), 4) 10% Citrus iyo (CI) juice (CI10), and 5) 40% CI juice (CI40). After drinking them for 2 weeks from 8 weeks of age, cuff injury was induced by polyethylene cuff placement around the femoral artery. Neointima formation was significantly attenuated in CU40, CI10 and CI40 compared with C; however, no remarkable preventive effect was observed in CU10. The increases in levels of various inflammatory markers including cytokines such as monocyte chemotactic protein-1, interleukin-6 (IL-6), IL-1ß, and tumor necrosis factor-α in response to vascular injury did not differ significantly between C, CU10 and CI10. The increases in cell proliferation and superoxide anion production were markedly attenuated in CI10, but not in CU10 compared with C. The increase in phosphorylated ERK expression was markedly attenuated both in CU10 and CI10 without significant difference between CU10 and CI10. Accumulation of immune cells did not differ between CU10 and CI10. These results indicate that drinking citrus fruit juice attenuates vascular remodeling partly via a reduction of oxidative stress. Interestingly, the preventive efficacy on neointima formation was stronger in CI than in CU at least in part due to more prominent inhibitory effects on oxidative stress by CI.


Asunto(s)
Bebidas , Citrus/química , Ingestión de Líquidos , Frutas/química , Lesiones del Sistema Vascular/etiología , Lesiones del Sistema Vascular/fisiopatología , Animales , Proliferación Celular/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Neointima/prevención & control , Superóxidos/metabolismo , Remodelación Vascular , Lesiones del Sistema Vascular/inmunología , Lesiones del Sistema Vascular/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...