Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Cell Rep ; 43(9): 114670, 2024 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-39213156

RESUMEN

Neutrophils from skull bone marrow (Nskull) are activated under some brain stresses, but their effects on traumatic brain injury (TBI) are lacking. Here, we find Nskull infiltrates brain tissue quickly and persistently after TBI, which is distinguished by highly and specifically expressed osteocalcin (OCN) from blood-derived neutrophils (Nblood). Reprogramming of glucose metabolism by reducing glycolysis-related enzyme glyceraldehyde 3-phosphate dehydrogenase expression is involved in the antiapoptotic and proliferative abilities of OCN-expressing Nskull. The transcription factor Fos-like 1 governs the specific gene profile of Nskull including C-C motif chemokine receptor-like 2 (CCRL2), arginase 1 (Arg1), and brain-derived neurotrophic factor (BDNF) in addition to OCN. Selective knockout of CCRL2 in Nskull demonstrates that CCRL2 mediates its recruitment, whereas high Arg1 expression is consistent with its immunosuppressive effects on Nblood, and the secretion of BDNF facilitating dendritic growth contributes to its neuroprotection. Thus, our findings provide insight into the roles of Nskull in TBI.

2.
Mil Med Res ; 11(1): 20, 2024 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-38556884

RESUMEN

BACKGROUND: Neutrophils are traditionally viewed as first responders but have a short onset of action in response to traumatic brain injury (TBI). However, the heterogeneity, multifunctionality, and time-dependent modulation of brain damage and outcome mediated by neutrophils after TBI remain poorly understood. METHODS: Using the combined single-cell transcriptomics, metabolomics, and proteomics analysis from TBI patients and the TBI mouse model, we investigate a novel neutrophil phenotype and its associated effects on TBI outcome by neurological deficit scoring and behavioral tests. We also characterized the underlying mechanisms both in vitro and in vivo through molecular simulations, signaling detections, gene expression regulation assessments [including dual-luciferase reporter and chromatin immunoprecipitation (ChIP) assays], primary cultures or co-cultures of neutrophils and oligodendrocytes, intracellular iron, and lipid hydroperoxide concentration measurements, as well as forkhead box protein O1 (FOXO1) conditional knockout mice. RESULTS: We identified that high expression of the FOXO1 protein was induced in neutrophils after TBI both in TBI patients and the TBI mouse model. Infiltration of these FOXO1high neutrophils in the brain was detected not only in the acute phase but also in the chronic phase post-TBI, aggravating acute brain inflammatory damage and promoting late TBI-induced depression. In the acute stage, FOXO1 upregulated cytoplasmic Versican (VCAN) to interact with the apoptosis regulator B-cell lymphoma-2 (BCL-2)-associated X protein (BAX), suppressing the mitochondrial translocation of BAX, which mediated the antiapoptotic effect companied with enhancing interleukin-6 (IL-6) production of FOXO1high neutrophils. In the chronic stage, the "FOXO1-transferrin receptor (TFRC)" mechanism contributes to FOXO1high neutrophil ferroptosis, disturbing the iron homeostasis of oligodendrocytes and inducing a reduction in myelin basic protein, which contributes to the progression of late depression after TBI. CONCLUSIONS: FOXO1high neutrophils represent a novel neutrophil phenotype that emerges in response to acute and chronic TBI, which provides insight into the heterogeneity, reprogramming activity, and versatility of neutrophils in TBI.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Neutrófilos , Animales , Humanos , Ratones , Proteína X Asociada a bcl-2/metabolismo , Encéfalo , Lesiones Traumáticas del Encéfalo/complicaciones , Depresión , Proteína Forkhead Box O1/metabolismo , Hierro
3.
Exp Neurol ; 374: 114691, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38224942

RESUMEN

Blood-brain barrier (BBB) impairment and glutamate release are two pathophysiological features of traumatic brain injury (TBI), contributing to secondary brain damage and neuroinflammation. However, our knowledge of BBB integrity damage and dysfunction are still limited due to the diverse and fluctuating expression of glutamate receptors after trauma. Here, we confirmed the downregulation of metabotropic glutamate receptor 5 (mGluR5) on microvascular endothelial cell within the acute phase of TBI, and the recovered mGluR5 levels on BBB was positively associated with blood perfusion and neurological recovery. In whole body mGluR5-knockout mice, BBB dysfunction and neurological deficiency were exacerbated after TBI compared with wild type mice. In terms of mechanism, the amino acid sequence 201-259 of cytoskeletal protein Alpha-actinin-1 (ACTN1) interacted with mGluR5, facilitating mGluR5 translocation from cytoplasmic compartment to plasma membrane in endothelial cells. Activation of plasma membrane mGluR5 triggers the PLC/PKCµ/c-Jun signaling pathway, leading to increased expression of the tight junction-actin cytoskeleton connecting protein zonula occludens-1 (ZO-1). Our findings uncover a novel mechanism mediated by membrane and cytoplasmic mGluR5 in endothelial cell integrity maintenance and repair, providing the potential therapeutic target for TBI treatment targeting at mGluR5 and mGluR5/ACTN1 complex in BBB.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Lesiones Encefálicas , Animales , Ratones , Barrera Hematoencefálica/metabolismo , Lesiones Encefálicas/metabolismo , Lesiones Traumáticas del Encéfalo/metabolismo , Células Endoteliales/metabolismo , Ratones Noqueados , Receptor del Glutamato Metabotropico 5/metabolismo
4.
Biochem Mol Biol Educ ; 51(5): 557-565, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37409779

RESUMEN

The development of information technology and portable devices has sparked a revolution in the field of education, facilitating access to diverse educational resources and lifelong learning. In particular, the COVID-19 pandemic has accelerated the transition from face-to-face to distance teaching, which requires online education to be provided worldwide. Biochemistry and Molecular Biology are key basic medical courses in laboratory-based science that cover complicated theories and applications. The balance between traditional and online courses, and the effectiveness of online courses, are fundamental to the teaching quality of Biochemistry and Molecular Biology. In this study, we explored the concepts, designs, and practices of a new blended online course and identified potential challenges. We hope that our experiences will provide new ideas for online teaching and promote teaching reform and the development of Medical Biochemistry and Molecular Biology education.

5.
Eur J Pharmacol ; 953: 175838, 2023 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-37307937

RESUMEN

Evidence is mounting that sinomenine and peroxisome proliferator-activated receptor ß/δ (PPARß/δ) are effective against lipopolysaccharide (LPS)-induced acute lung injury (ALI) via anti-inflammatory properties. However, it is unknown whether PPARß/δ plays a role in the protective effect of sinomenine on ALI. Here, we initially observed that preemptive administration of sinomenine markedly alleviated lung pathological changes, pulmonary edema and neutrophil infiltration, accompanied by inhibition of the expression of the pro-inflammatory cytokines Tumor necrosis factor-α (TNF-α) and Interleukin-6 (IL-6), which were largely reversed following the addition of a PPARß/δ antagonist. Subsequently, we also noticed that sinomenine upregulated adenosine A2A receptor expression in a PPARß/δ-dependent manner in LPS-stimulated bone marrow-derived macrophages (BMDMs). Further investigation indicated that PPARß/δ directly bound to the functional peroxisome proliferator responsive element (PPRE) in the adenosine A2A receptor gene promoter region to enhance the expression of the adenosine A2A receptor. Sinomenine was identified as a PPARß/δ agonist. It could bind with PPARß/δ, and promote the nuclear translocation and transcriptional activity of PPARß/δ. In addition, combined treatment with sinomenine and an adenosine A2A receptor agonist exhibited synergistic effects and better protective roles than their single use against ALI. Taken together, our results reveal that sinomenine exerts advantageous effects on ALI by activating of PPARß/δ, with the subsequent upregulation of adenosine A2A receptor expression, and provide a novel and potential therapeutic application for ALI.


Asunto(s)
Lesión Pulmonar Aguda , PPAR delta , PPAR-beta , Humanos , PPAR-beta/metabolismo , Lipopolisacáridos/farmacología , Receptor de Adenosina A2A , PPAR delta/metabolismo , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/tratamiento farmacológico , Lesión Pulmonar Aguda/genética
6.
Neurosci Bull ; 38(10): 1199-1214, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35819574

RESUMEN

The brain pericyte is a unique and indispensable part of the blood-brain barrier (BBB), and contributes to several pathological processes in traumatic brain injury (TBI). However, the cellular and molecular mechanisms by which pericytes are regulated in the damaged brain are largely unknown. Here, we show that the formation of neutrophil extracellular traps (NETs) induces the appearance of CD11b+ pericytes after TBI. These CD11b+ pericyte subsets are characterized by increased permeability and pro-inflammatory profiles compared to CD11b- pericytes. Moreover, histones from NETs by Dectin-1 facilitate CD11b induction in brain pericytes in PKC-c-Jun dependent manner, resulting in neuroinflammation and BBB dysfunction after TBI. These data indicate that neutrophil-NET-pericyte and histone-Dectin-1-CD11b are possible mechanisms for the activation and dysfunction of pericytes. Targeting NETs formation and Dectin-1 are promising means of treating TBI.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Trampas Extracelulares , Barrera Hematoencefálica/metabolismo , Encéfalo/patología , Lesiones Traumáticas del Encéfalo/metabolismo , Trampas Extracelulares/metabolismo , Histonas , Humanos , Lectinas Tipo C , Pericitos/patología
7.
Cancer Sci ; 113(6): 1955-1967, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35363928

RESUMEN

Neutrophils are the first defenders of the innate system for injury and infection. They have gradually been recognized as important participants in tumor initiation and development due to their heterogeneity and plasticity. In the tumor microenvironment (TME), neutrophils can exert antitumor and protumor functions, depending on the surroundings. Tumor cells systemically alter intracellular amino acid (AA) metabolism and extracellular AA distribution to meet their proliferation need, leading to metabolic reprogramming and TME reshaping. However, the underlying mechanisms that determine how altered AAs affect neutrophils in TME are less-explored. Here, we identified that abundant glutamate releasing from tumor cells blunted neutrophils' cell-killing effects toward tumor cells in vitro and in vivo. Mass spectrometric detection, flow cytometry, and western blot experiments proved that increased levels of pSTAT3/RAB10/ARF4, mediated by glutamate, were accompanied with immunosuppressive phenotypes of neutrophils in TME. We also discovered that riluzole, an FDA-approved glutamate release inhibitor, significantly inhibited tumor growth by restoring neutrophils' cell-killing effects and decreasing glutamate secretion from tumor cells. These findings highlight the importance of tumor-released glutamate on neutrophil transformation in TME, providing new possible cancer treatments targeting altered glutamate metabolism.


Asunto(s)
Neoplasias , Microambiente Tumoral , Apoptosis , Ácido Glutámico , Humanos , Neoplasias/patología , Neutrófilos/metabolismo
8.
Sci Adv ; 7(50): eabj4226, 2021 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-34890233

RESUMEN

In this study, we report a novel role of metabotropic glutamate receptor 4 (GRM4) in suppressing antitumor immunity. We revealed in three murine syngeneic tumor models (B16, MC38, and 3LL) that either genetic knockout (Grm4−/−) or pharmacological inhibition led to significant delay in tumor growth. Mechanistically, perturbation of GRM4 resulted in a strong antitumor immunity by promoting natural killer (NK), CD4+, and CD8+ T cells toward an activated, proliferative, and functional phenotype. Single-cell RNA sequencing and T cell receptor profiling further defined the clonal expansion and immune landscape changes in CD8+ T cells. We further showed that Grm4−/− intrinsically activated interferon-γ production in CD8+ T cells through cyclic adenosine 3',5'-monophosphate (cAMP)/cAMP response element binding protein­mediated pathway. Our study appears to be of clinical significance as a signature of NKhigh-GRM4low and CD8high-GRM4low correlated with improved survival in patients with melanoma. Targeting GRM4 represents a new approach for cancer immunotherapy.

9.
Acta Biomater ; 105: 239-252, 2020 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-31958597

RESUMEN

The unique metabolic demand of cancer cells suggests a new therapeutic strategy targeting the metabolism in cancers. V9302 is a recently reported inhibitor of ASCT2 amino acid transporter which shows promising antitumor activity by blocking glutamine uptake. However, its poor solubility in aqueous solutions and tumor cells' compensatory metabolic shift to glucose metabolism may limit the antitumor efficacy of V9302. 2-Deoxyglucose (2-DG), a derivative of glucose, has been developed as a potential antitumor agent through inhibiting glycolysis in tumor cells. In order to achieve enhanced antitumor effect by inhibiting both metabolic pathways, a 2-DG prodrug-based micellar carrier poly-(oligo ethylene glycol)-co-poly(4-((4-oxo-4-((4-vinylbenzyl)oxy)butyl)disulfaneyl)butanoic acid)-(2-deoxyglucose) (POEG-p-2DG) was developed. POEG-p-2DG well retained the pharmacological activity of 2-DG in vitro and in vivo, More importantly, POEG-p-2DG could self-assemble to form micelles that were capable of loading V9302 to achieve co-delivery of 2-DG and V9302. V9302-loaded POEG-p2DG micelles were small in sizes (~10 nm), showed a slow kinetics of drug release and demonstrated targeted delivery to tumor. In addition, V9302 loaded POEG-p-2DG micelles exhibited improved anti-tumor efficacy both in vitro and in vivo. Interestingly, 2-DG treatment further decreased the glutamine uptake when combined with V9302, likely due to inhibition of ASCT2 glycosylation. These results suggest that POEG-p2DG prodrug micelles may serve as a dual functional carrier for V9302 to achieve synergistic targeting of metabolism in cancers. STATEMENT OF SIGNIFICANCE: Unique cancer cell's metabolism profile denotes a new therapeutic strategy. V9302 is a recently reported glutamine metabolism inhibitor that shows promising antitumor activity. However, its poor waster solubility and tumor cell's compensatory metabolic network may limit its potential clinical application. 2-Deoxyglucose(2-DG) is a widely used glycolysis inhibitor. However, its clinical application is hindered by low efficacy as monotherapy. Thus, in this study, we developed a redox-sensitive, 2-DG-based prodrug polymer, as a dual-functional carrier for co-delivery of V9302 and 2-DG as a combination strategy. V9302 loaded POEG-p-2DG micelle showed significantly improved antitumor activity through synergistic targeting of both glutamine and glycolysis metabolism pathway. More interestingly, POEG-p-2DG itself further facilitates inhibition of glutamine metabolism, likely through inhibition of ASCT2 glycosylation.


Asunto(s)
Desoxiglucosa/administración & dosificación , Glutamina/metabolismo , Micelas , Neoplasias/metabolismo , Profármacos/administración & dosificación , Animales , Antineoplásicos/farmacología , Muerte Celular , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Desoxiglucosa/sangre , Desoxiglucosa/farmacocinética , Liberación de Fármacos , Sinergismo Farmacológico , Femenino , Glucosa/metabolismo , Humanos , Ratones Endogámicos BALB C , Polietilenglicoles/síntesis química , Polietilenglicoles/química , Distribución Tisular
10.
Brain Behav Immun ; 79: 114-124, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30682501

RESUMEN

Antidepressant-like effects of metabotropic glutamate receptor 5 (mGluR5) have been verified by specific antagonists or whole body knock-out (KO) mice. Previous experiments indicate that blocking mGluR5 exerts antidepressant-like effects through neuronal mechanisms, like modulating NMDA receptor activity or 5-HT system. Here we found that transplanting bone marrow from mGluR5 KO mice to WT mice could also show antidepressant-like effects, which were confirmed by sucrose preference test and tail suspension test. Furthermore, mGluR5 deficiency dramatically inhibits cytokines release from bone marrow cells, such as IL-1ß, TNF-α and IL-6, alleviating proinflammatory responses in LPS-induced depression model. In addition, inhibited cytokines could decrease the activation of brain endothelial cells in ERK-dependent manner. These data provide the evidence that blocking mGluR5 could improve depression through inhibiting peripheral immune responses, confirming the causal relationship between peripheral immune phenotype and brain behavior.


Asunto(s)
Antidepresivos/metabolismo , Depresión/metabolismo , Receptor del Glutamato Metabotropico 5/metabolismo , Animales , Antidepresivos/farmacología , Conducta Animal/efectos de los fármacos , Médula Ósea/metabolismo , Trasplante de Médula Ósea/métodos , Encéfalo/metabolismo , Citocinas/metabolismo , Depresión/tratamiento farmacológico , Depresión/etiología , Trastorno Depresivo/tratamiento farmacológico , Trastorno Depresivo/etiología , Trastorno Depresivo/metabolismo , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Lipopolisacáridos/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/metabolismo , Receptor del Glutamato Metabotropico 5/genética , Receptor del Glutamato Metabotropico 5/fisiología , Receptores de N-Metil-D-Aspartato/metabolismo
11.
J Neuroinflammation ; 15(1): 146, 2018 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-29776443

RESUMEN

Our knowledge of the pathophysiology about traumatic brain injury (TBI) is still limited. Neutrophils, as the most abundant leukocytes in circulation and the first-line transmigrated immune cells at the sites of injury, are highly involved in the initiation, development, and recovery of TBI. Nonetheless, our understanding about neutrophils in TBI is obsolete, and mounting evidences from recent studies have challenged the conventional views. This review summarizes what is known about the relationships between neutrophils and pathophysiology of TBI. In addition, discussions are made on the complex roles as well as the controversial views of neutrophils in TBI.


Asunto(s)
Lesiones Traumáticas del Encéfalo/patología , Lesiones Traumáticas del Encéfalo/fisiopatología , Neutrófilos/fisiología , Animales , Humanos
12.
Sci Rep ; 7(1): 9998, 2017 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-28855570

RESUMEN

Both brain native inflammatory cells and infiltrated peripheral white blood cells (WBCs) are primary participants in the brain inflammatory damage post-TBI. Metabotropic glutamate receptor 5 (mGluR5) has been reported to regulate microglias and astrocytes to affect inflammation after TBI, but its effect on modulating infiltrated peripheral WBCs remains unclear. In a mouse moderate TBI model, we found that mGluR5 knockout (KO) significantly reduced neutrophil infiltration and inflammatory cytokine expression in the brain at 24 hours post TBI, which was accompanied by improved neurological dysfunction. Further investigation indicated that mGluR5 KO reduced the permeability of blood-brain barrier (BBB), the entrance for neutrophils to enter brain, and markedly decreased the mRNA levels of neutrophil-associated chemokines in brain tissue, including CXCL1, CXCL2, CCL2, CCL4 and CCL5. Using brain microvascular endothelial cells (BMECs), neutrophils and a BBB model in vitro, we confirmed the inhibitory effect of mGluR5 deficiency on neutrophil infiltration and demonstrated that blockade of protein kinase C (PKC) signaling was involved in it. These results provide insight into the role of mGluR5 in the regulation of inflammation in the acute phase of TBI, which may provide novel clues for TBI therapy.


Asunto(s)
Barrera Hematoencefálica/patología , Lesiones Traumáticas del Encéfalo/patología , Infiltración Neutrófila , Receptor del Glutamato Metabotropico 5/metabolismo , Animales , Células Cultivadas , Citocinas/biosíntesis , Modelos Animales de Enfermedad , Células Endoteliales/fisiología , Ratones , Ratones Noqueados , Receptor del Glutamato Metabotropico 5/deficiencia
13.
Sci Rep ; 6: 33614, 2016 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-27647162

RESUMEN

Systemic inflammatory response syndrome (SIRS) is an overwhelming whole body inflammation caused by infectious diseases or sterile insults. Neutrophils are the dominant participants during inflammation, and their survival and death determine the initiation as well as resolution of SIRS. Apoptosis and autophagy are two fundamental cellular processes that modulating cell fate, but their correlation and regulators in neutrophils under SIRS condition have not been elucidated. In this study, we demonstrated that high dose of LPS induced both apoptosis and autophagy of neutrophils in a mouse SIRS model and LPS-stimulated neutrophils in vitro. Moreover, we found that the adenosine 2A receptor (A2AR), a known anti-inflammatory G protein-coupled receptor (GPCR), could inhibit LPS-induced neutrophil apoptosis by suppressing the LPS-induced autophagy. Activation of A2AR suppressed LPS-induced autophagy by inhibiting the ROS-JNK pathway as well as promoting GPCR ßϒ subunit-AKT signaling. The A2AR-inhibited autophagy suppressed apoptosis of neutrophils by blocking caspase8, caspase3 and PARP signaling. These findings not only increase our understandings of neutrophils' fate and function in response to systemic inflammation, but also identify a novel anti-inflammatory role of A2AR in modulating neutrophils' survival during inflammation.


Asunto(s)
Apoptosis , Autofagia , Neutrófilos/metabolismo , Receptor de Adenosina A2A/metabolismo , Síndrome de Respuesta Inflamatoria Sistémica/etiología , Síndrome de Respuesta Inflamatoria Sistémica/metabolismo , ADP Ribosa Transferasas , Animales , Apoptosis/inmunología , Autofagia/inmunología , Caspasa 3/metabolismo , Caspasa 8/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Lipopolisacáridos/inmunología , Ratones , Neutrófilos/inmunología , Neutrófilos/patología , Neutrófilos/ultraestructura , Fosforilación , Proteína Quinasa C/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal
14.
Cell Signal ; 27(10): 2026-34, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26171727

RESUMEN

Inflammation is a pathological course involved in several diseases. Both adenosine A2A receptor (A2AR) and miR-214 play important roles in regulation of inflammation. However, the internal link between them and their synergic modulation in inflammatory response has not been elucidated. In this study, we found that miR-214 and A2AR activation could downregulate the expressions of each other in murine macrophages. Comparing with the well known anti-inflammatory role of A2AR, miR-214 promoted the release of inflammatory cytokines TNF-α and IL-6. Further investigation demonstrated that miR-214 downregulated A2AR expression by directly targeting the 3'-untranslated region of A2AR mRNA. Instead of directly interacting with miR-214, A2AR activation repressed miR-214 expression by stimulating PKA signaling to suppress the nuclear translocation of NF-κB which could enhance the transcript activity of miR-214 gene promoter. Then using an LPS-induced ALI mouse model, in which inflammation is a hallmark, we confirmed their negative relationship and demonstrated that combination of miR-214 antagomir and A2AR agonist CGS21680 exerts more anti-inflammatory effect including alleviating the pathological changes, suppressing the neutrophil infiltration and the expression of inflammatory cytokines than using one of them alone. These findings for the first time uncovered a mutual suppression feedback loop between A2AR signaling and miR-214 in inflammation, which may provide new insight of inflammatory regulation and potential therapeutic significance for some inflammation-associated diseases.


Asunto(s)
MicroARNs/genética , Receptor de Adenosina A2A/metabolismo , Transducción de Señal , Regiones no Traducidas 3' , Lesión Pulmonar Aguda/inmunología , Lesión Pulmonar Aguda/metabolismo , Animales , Secuencia de Bases , Sitios de Unión , Línea Celular , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Citocinas/genética , Citocinas/metabolismo , Retroalimentación Fisiológica , Inflamación/metabolismo , Lipopolisacáridos/farmacología , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Regiones Promotoras Genéticas , Interferencia de ARN , Transcripción Genética
15.
Curr Eye Res ; 38(12): 1191-7, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24047084

RESUMEN

Pterygium is a type of benign uncontrolled growth of the conjunctive tissue that lays over the sclera. It can significantly alter visual function in advanced cases and become inflamed, leading to redness and irritation in the area. Although the exact etiology of pterygium remains uncertain, recent advances have provided important insight into the pathogenesis of pterygium. These studies indicate that tumor suppressor gene p53 and other genes associated with DNA repair, cell proliferation, migration and angiogenesis are critical for the development of pterygium. In addition, Human papillomavirus infection has been shown to be a risk factor in some populations. In this article, the current understanding of the pathogenesis of pterygium is reviewed.


Asunto(s)
Conjuntivitis Viral , Reparación del ADN/genética , Infecciones por Papillomavirus/complicaciones , Pterigion , Proteína p53 Supresora de Tumor/genética , Conjuntivitis Viral/etiología , Conjuntivitis Viral/genética , Conjuntivitis Viral/virología , Humanos , Pterigion/etiología , Pterigion/genética , Pterigion/virología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA