Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 368
Filtrar
1.
J Cell Mol Med ; 28(14): e18558, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-39048917

RESUMEN

Myocardial ischemia-reperfusion injury (MIRI) represents a critical pathology in acute myocardial infarction (AMI), which is characterized by high mortality and morbidity. Cardiac microvascular dysfunction contributes to MIRI, potentially culminating in heart failure (HF). Pigment epithelium-derived factor (PEDF), which belongs to the non-inhibitory serpin family, exhibits several physiological effects, including anti-angiogenesis, anti-inflammatory and antioxidant properties. Our study aims to explore the impact of PEDF and its functional peptide 34-mer on both cardiac microvascular perfusion in MIRI rats and human cardiac microvascular endothelial cells (HCMECs) injury under hypoxia reoxygenation (HR). It has been shown that MIRI is accompanied by ferroptosis in HCMECs. Furthermore, we investigated the effect of PEDF and its 34-mer, particularly regarding the Nrf2/HO-1 signalling pathway. Our results demonstrated that PEDF 34-mer significantly ameliorated cardiac microvascular dysfunction following MIRI. Additionally, they exhibited a notable suppression of ferroptosis in HCMECs, and these effects were mediated through activation of Nrf2/HO-1 signalling. These findings highlight the therapeutic potential of PEDF and 34-mer in alleviating microvascular dysfunction and MIRI. By enhancing cardiac microvascular perfusion and mitigating endothelial ferroptosis, PEDF and its derivative peptide represent promising candidates for the treatment of AMI.


Asunto(s)
Células Endoteliales , Proteínas del Ojo , Ferroptosis , Daño por Reperfusión Miocárdica , Factor 2 Relacionado con NF-E2 , Factores de Crecimiento Nervioso , Serpinas , Transducción de Señal , Serpinas/farmacología , Serpinas/metabolismo , Factores de Crecimiento Nervioso/farmacología , Factores de Crecimiento Nervioso/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Animales , Ferroptosis/efectos de los fármacos , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Daño por Reperfusión Miocárdica/patología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Humanos , Proteínas del Ojo/metabolismo , Proteínas del Ojo/farmacología , Transducción de Señal/efectos de los fármacos , Ratas , Hemo-Oxigenasa 1/metabolismo , Masculino , Ratas Sprague-Dawley , Microvasos/efectos de los fármacos , Microvasos/metabolismo , Microvasos/patología , Péptidos/farmacología
2.
J Pharmacol Sci ; 156(1): 45-48, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39068034

RESUMEN

The gene product of ocular albinism 1 (OA1)/G-protein-coupled receptor (GPR)143 is a receptor for L-3,4-dihydroxyphenylanine (l-DOPA), the most effective agent for Parkinson's disease. When overexpressed, human wild-type GPR143, but not its mutants, inhibits neurite outgrowth in PC12 cells. We investigated the downstream signaling pathway for GPR143-induced inhibition of neurite outgrowth. Nifedipine restored GPR143-induced neurite outgrowth inhibition to the level of control transfectant but did not affect outgrowth in GPR143-knockdown cells. Cilnidipine and flunarizine also suppressed the GPR143-induced inhibition, but their effects at higher concentrations still occurred even in GPR143-knockdown cells. These results suggest that GPR143 regulates neurite outgrowth via L-type calcium channel(s).


Asunto(s)
Canales de Calcio Tipo L , Proyección Neuronal , Nifedipino , Receptores Acoplados a Proteínas G , Células PC12 , Animales , Ratas , Canales de Calcio Tipo L/metabolismo , Canales de Calcio Tipo L/genética , Nifedipino/farmacología , Proyección Neuronal/efectos de los fármacos , Receptores Acoplados a Proteínas G/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/fisiología , Humanos , Proteínas del Ojo/genética , Proteínas del Ojo/metabolismo , Proteínas del Ojo/farmacología , Flunarizina/farmacología , Transducción de Señal/efectos de los fármacos , Levodopa/farmacología , Técnicas de Silenciamiento del Gen , Neuritas/efectos de los fármacos , Bloqueadores de los Canales de Calcio/farmacología , Glicoproteínas de Membrana
3.
Gene Ther ; 31(9-10): 511-523, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38961279

RESUMEN

Neovascular age-related macular degeneration (nAMD) causes severe visual impairment. Pigment epithelium-derived factor (PEDF), soluble CD59 (sCD59), and soluble fms-like tyrosine kinase-1 (sFLT-1) are potential therapeutic agents for nAMD, which target angiogenesis and the complement system. Using the AAV2/8 vector, two bi-target gene therapy agents, AAV2/8-PEDF-P2A-sCD59 and AAV2/8-sFLT-1-P2A-sCD59, were generated, and their therapeutic efficacy was investigated in laser-induced choroidal neovascularization (CNV) and Vldlr-/- mouse models. After a single injection, AAV2/8-mediated gene expression was maintained at high levels in the retina for two months. Both AAV2/8-PEDF-P2A-sCD59 and AAV2/8-sFLT-1-P2A-sCD59 significantly reduced CNV development for an extended period without side effects and provided efficacy similar to two injections of current anti-vascular endothelial growth factor monotherapy. Mechanistically, these agents suppressed the extracellular signal-regulated kinase and nuclear factor-κB pathways, resulting in anti-angiogenic activity. This study demonstrated the safety and long-lasting effects of AAV2/8-PEDF-P2A-sCD59 and AAV2/8-sFLT-1-P2A-sCD59 in CNV treatment, providing a promising therapeutic strategy for nAMD.


Asunto(s)
Neovascularización Coroidal , Dependovirus , Terapia Genética , Vectores Genéticos , Neovascularización Coroidal/terapia , Animales , Dependovirus/genética , Ratones , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Factores de Crecimiento Nervioso/genética , Factores de Crecimiento Nervioso/metabolismo , Proteínas del Ojo/genética , Proteínas del Ojo/metabolismo , Proteínas del Ojo/farmacología , Receptor 1 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Antígenos CD59/genética , Antígenos CD59/metabolismo , Ratones Endogámicos C57BL , Humanos , Modelos Animales de Enfermedad , Parvovirinae/genética , Degeneración Macular/terapia , Serpinas
4.
Aging Dis ; 15(5): 2003-2007, 2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-38421833

RESUMEN

Geographic atrophy (GA) is an advanced form of age-related macular degeneration (AMD), that starts with atrophic lesions in the outer retina that expand to cover the macula and fovea, leading to severe vision loss over time. Pigment Epithelium-Derived Factor (PEDF) has a diverse-range of properties, including its ability to promote cell survival, reduce inflammation, inhibit angiogenesis, combat oxidative stress, regulate autophagy, and stimulate anti-apoptotic pathways, making it a promising therapeutic candidate for GA. However, the relatively short half-life of PEDF protein has precluded its potential as a clinical therapy for GA since it would require frequent injections. Therefore, we describe administration of a PEDF gene, comparing and contrasting delivery routes, viral and non-viral vectors, and consider the critical challenges for PEDF as a neuroprotectant for GA.


Asunto(s)
Proteínas del Ojo , Atrofia Geográfica , Factores de Crecimiento Nervioso , Serpinas , Animales , Humanos , Proteínas del Ojo/metabolismo , Proteínas del Ojo/uso terapéutico , Proteínas del Ojo/administración & dosificación , Proteínas del Ojo/genética , Proteínas del Ojo/farmacología , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Atrofia Geográfica/tratamiento farmacológico , Atrofia Geográfica/metabolismo , Factores de Crecimiento Nervioso/administración & dosificación , Factores de Crecimiento Nervioso/uso terapéutico , Fármacos Neuroprotectores/uso terapéutico , Fármacos Neuroprotectores/administración & dosificación , Fármacos Neuroprotectores/farmacología , Serpinas/administración & dosificación , Serpinas/uso terapéutico , Serpinas/genética , Serpinas/metabolismo , Serpinas/farmacología
5.
Endocrine ; 83(1): 77-91, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37682419

RESUMEN

BACKGROUND: Accumulation of bone marrow adipose tissue (BMAT) is always seen in osteoporosis induced by estrogen deficiency. Herein, we aimed to investigate the mechanisms and consequences of this phenomenon by establishing a mouse model of osteoporosis caused by ovariectomy (OVX)-mimicked estrogen deficiency. METHODS: Micro-CT, osmium tetroxide staining, and histological analyses were performed to examine the changes in bone microstructure, BMAT and white adipose tissue (WAT) in OVX mice compared to sham mice. The osteogenesis and adipogenesis of primary bone marrow stromal cells (BMSCs) isolated from sham and OVX mice were compared in vitro. The molecular phenotypes of BMAT and WAT were determined and compared by quantitative PCR (qPCR). Bone marrow adipocyte-conditioned medium (BMA CM) was prepared from sham or OVX mice for coculture assays, and BMSCs or bone marrow monocytes/macrophages (BMMs) were isolated and subjected to osteoblast and osteoclast differentiation, respectively. Cell staining and qPCR were used to assess the effects of BMAT on bone metabolism. RESULTS: OVX-induced estrogen deficiency induced reductions in both cortical and trabecular bone mass along with an expansion of BMAT volume. At the cellular level, loss of estrogen inhibited BMSC osteogenesis and promoted BMSC adipogenesis, whereas addition of estradiol exerted the opposite effects. In response to estrogen deficiency, despite the common proinflammatory molecular phenotype observed in both fat depots, BMAT, unlike WAT, unexpectedly exhibited an increase in adipocyte differentiation and lipolytic activity as well as the maintenance of insulin sensitivity. Importantly, BMAT, but not WAT, presented increased mRNA levels of both BMP receptor inhibitors (Grem1, Chrdl1) and Rankl following OVX. In addition, treatment with BMA CM, especially from OVX mice, suppressed the osteoblast differentiation of BMSCs while favoring the osteoclast differentiation of BMMs. CONCLUSION: Our study illustrates that OVX-induced estrogen deficiency results in bone loss and BMAT expansion by triggering imbalance between the osteogenesis and adipogenesis of BMSCs. Furthermore, expanded BMAT, unlike typical WAT, may negatively regulate bone homeostasis through paracrine inhibition of osteoblast-mediated bone formation and promotion of osteoclast-mediated bone resorption.


Asunto(s)
Médula Ósea , Osteoporosis , Femenino , Ratones , Animales , Humanos , Médula Ósea/metabolismo , Tejido Adiposo/metabolismo , Osteoporosis/etiología , Osteoporosis/metabolismo , Osteogénesis , Diferenciación Celular , Estrógenos/farmacología , Ovariectomía/efectos adversos , Proteínas del Ojo/farmacología , Proteínas del Tejido Nervioso
6.
J Pharm Pharmacol ; 75(6): 746-757, 2023 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-37104852

RESUMEN

OBJECTIVES: Cardiovascular diseases are the leading cause of death worldwide, with patients having limited options for treatment. Pigment epithelium-derived factor (PEDF) is an endogenous multifunctional protein with several mechanisms of action. Recently, PEDF has emerged as a potential cardioprotective agent in response to myocardial infarction. However, PEDF is also associated with pro-apoptotic effects, complicating its role in cardioprotection. This review summarises and compares knowledge of PEDF's activity in cardiomyocytes with other cell types and draws links between them. Following this, the review offers a novel perspective of PEDF's therapeutic potential and recommends future directions to understand the clinical potential of PEDF better. KEY FINDINGS: PEDF's mechanisms as a pro-apoptotic and pro-survival protein are not well understood, despite PEDF's implication in several physiological and pathological activities. However, recent evidence suggests that PEDF may have significant cardioprotective properties mediated by key regulators dependent on cell type and context. CONCLUSIONS: While PEDF's cardioprotective activity shares some key regulators with its apoptotic activity, cellular context and molecular features likely allow manipulation of PEDF's cellular activity, highlighting the importance of further investigation into its activities and its potential to be applied as a therapeutic to mitigate damage from a range of cardiac pathologies.


Asunto(s)
Miocitos Cardíacos , Serpinas , Humanos , Miocitos Cardíacos/metabolismo , Serpinas/farmacología , Serpinas/fisiología , Proteínas del Ojo/farmacología , Proteínas del Ojo/fisiología , Factores de Crecimiento Nervioso/farmacología , Factores de Crecimiento Nervioso/fisiología
7.
Am J Respir Cell Mol Biol ; 69(1): 87-98, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37094101

RESUMEN

Pulmonary hypertension (PH) is a disease characterized by advanced pulmonary vasculature remodeling that is thought to be curable only through lung transplantation. The application of angiogenic hepatocyte growth factor (HGF) is reported to be protective in PH through its anti-vascular remodeling effect, but excessive HGF-mediated immature neovascularization is not conducive to the restoration of pulmonary perfusion because of apparent vascular leakage. As a canonical antiangiogenic molecule, pigment epithelium-derived factor (PEDF) inhibits angiogenesis and reduces vascular permeability in a variety of diseases. However, the effect of PEDF on HGF-based PH treatment remains to be determined. In this study, monocrotaline-induced PH rats and endothelial cells isolated from rat and human PH lung tissues were used. We assessed PH progression, right cardiac function, and pulmonary perfusion in HGF- and/or PEDF-treated rats with PH. Additionally, the receptor and mechanism responsible for the role of PEDF in HGF-based PH therapy were investigated. In this study, we found that HGF and PEDF jointly prevent PH development and improve right cardiac function in rats with PH. Moreover, PEDF delivery increases the pulmonary perfusion in PH lungs and inhibits immature angiogenesis and vascular endothelial (VE)-cadherin junction disintegration induced by HGF without affecting the therapeutic inhibition of pulmonary vascular remodeling by HGF. Mechanistically, PEDF targets VE growth factor receptor 2 and suppresses its phosphorylation at Y951 and Y1175 but not Y1214. Finally, VE growth factor receptor 2/VE protein tyrosine phosphatase/VE-cadherin complex formation and Akt and Erk1/2 inactivation were observed in rat and human PH lung endothelial cells. Collectively, our data indicate that PEDF additively enhances the efficacy of HGF against PH, which may provide new insights into treatment strategies for clinical PH.


Asunto(s)
Hipertensión Pulmonar , Serpinas , Ratas , Humanos , Animales , Factor de Crecimiento de Hepatocito/efectos adversos , Factor de Crecimiento de Hepatocito/metabolismo , Hipertensión Pulmonar/metabolismo , Células Endoteliales/metabolismo , Proteínas del Ojo/farmacología , Proteínas del Ojo/metabolismo , Serpinas/farmacología , Serpinas/metabolismo
8.
Biomed Pharmacother ; 151: 113113, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35598368

RESUMEN

Ocular ischemia is a vision-threatening disease, and is a medical condition associated with many ocular diseases. Anti-VEGF therapy has limitations related to its side effects and suppression of physiological revascularization. Pigment epithelium derived factor (PEDF) has anti-angiogenesis and neurotrophic neuroprotective functions and is a promising agent in the treatment of ischemia-induced retinal neurodegeneration. The purpose of this study is to investigate the effect of PEDF and anti-VEGF and the combined therapy on the ischemic rat eye model ex vivo. In this study, the PEDF protein, anti-VEGF drug (Avastin) or the combination of PEDF and Avastin were intravitreally injected immediately after eye enucleation. Then the eyes were incubated in Dulbecco's modified eagle medium (DMEM) at 4 â„ƒ for 14 h. After that the eyes were fixed immediately by formalin. VEGF, PEDF and glial fibrillary acidic protein (GFAP) were detected by immunohistochemical (IHC) staining. The IHC staining intensity was evaluated for each eye. Compared to the groups treated by vehicle, PEDF, and anti-VEGF alone, the value of staining intensity of VEGF and GFAP was significantly reduced in the retina and choroidal vessels of the PEDF/Anti-VEGF treatment group. The intravitreally injected PEDF protein can locate in the retina and the choroidal vessels. Compared to the vehicle-treatment group, both the PEDF-treatment and the PEDF/Anti-VEGF treatment groups showed significantly decreased number of TUNEL-positive nuclei, and the PEDF/Anti-VEGF treatment group had the least TUNEL-positive nuclei. Combination of PEDF and an anti-VEGF drug (Avastin) is a possible therapeutic strategy against ischemic retinal and choroidal diseases.


Asunto(s)
Proteínas del Ojo , Enfermedades de la Retina , Serpinas , Animales , Bevacizumab/farmacología , Bevacizumab/uso terapéutico , Epitelio/metabolismo , Proteínas del Ojo/metabolismo , Proteínas del Ojo/farmacología , Proteínas del Ojo/uso terapéutico , Isquemia/tratamiento farmacológico , Isquemia/prevención & control , Ratas , Retina/patología , Enfermedades de la Retina/tratamiento farmacológico , Enfermedades de la Retina/metabolismo , Serpinas/metabolismo , Serpinas/farmacología , Serpinas/uso terapéutico , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/metabolismo , Factores de Crecimiento Endotelial Vascular
9.
Cancer Gene Ther ; 29(10): 1332-1341, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35246611

RESUMEN

Ovarian cancer (OC) is one of the most dangerous gynecological malignancies with no effective treatment so far. Pigment epithelium-derived factor (PEDF) has been reported to have ideal anti-tumor effects, but its relationship with the regulation of tumor-associated macrophage polarization is currently unclear. In this study, the mRNA expression of PEDF and macrophage markers were determined in OC tissues from clinic patients and five OC (A2780, SKOV3, CAOV3, OVCAR3, and OVCA433) cell lines through quantitative reverse transcription PCR. Afterwards, tumor growth, cell proliferation and apoptosis, and macrophage polarization in OC tumor-bearing mice with PEDF overexpression were recorded and investigated. Finally, the polarization of macrophages was explored in the presence of lentiviral PEDF overexpression, adipose triglyceride lipase (ATGL) and laminin receptor (LR) knockdown, and mitogen-activated protein kinase (MAPK) pathway inhibition. Our results suggest that PEDF mRNA level is significantly decreased in OC tissues and cells and has a significant negative correlation with OC progression and the level of tumor-related macrophage markers. Furthermore, OC tumors overexpressing PEDF show suppressed growth viability and increased apoptosis rate. The fluorescence activated cell sorting (FACS) analysis reveals that PEDF can promote macrophage polarization in OC tumors towards M1 subtype. Mechanistically, we found that ATGL and extracellular-regulated kinase 1/2 (ERK1/2) signaling are involved in the regulation of macrophage polarization in OC tumors by PEDF. Taken together, these data indicate that the role of PEDF in regulating the polarization of tumor-associated macrophages may make it a potential therapeutic strategy for the treatment of OC in the future.


Asunto(s)
Neoplasias Ováricas , Serpinas , Animales , Apoptosis/genética , Línea Celular Tumoral , Proteínas del Ojo/genética , Proteínas del Ojo/metabolismo , Proteínas del Ojo/farmacología , Femenino , Humanos , Lipasa/genética , Lipasa/metabolismo , Ratones , Proteínas Quinasas Activadas por Mitógenos , Factores de Crecimiento Nervioso , Neoplasias Ováricas/genética , Neoplasias Ováricas/terapia , ARN Mensajero/metabolismo , Receptores de Laminina/metabolismo , Serpinas/genética , Serpinas/metabolismo , Serpinas/farmacología , Macrófagos Asociados a Tumores
10.
BMC Ophthalmol ; 22(1): 88, 2022 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-35193548

RESUMEN

BACKGROUND: Pigment epithelial-derived factor (PEDF), a 50 kDa secreted glycoprotein, exhibits distinct effects on a range of cell types. PEDF has been shown to inhibit vascular endothelial growth factor (VEGF)-mediated angiogenesis and widely accepted as a promising agent for treatment eye diseases related to neovascularization. A pool of short peptide fragments derived from PEDF reportedly manifests angioinhibitory activity. This study aims to determine the minimal PEDF fragment which can exert the anti-VEGF effect. METHODS: A series of shorter synthetic peptides, derived from the 34-mer (PEDF amino acid positions Asp44-Asn77), were synthesized. An MTT assay was used to evaluate the ability of the 34-mer-derived peptides to inhibit VEGF-induced proliferation of multiple myeloma RPMI8226 cells. Cell apoptosis was monitored by annexin V-FITC staining. Western blot analysis was used to detect phosphorylated kinases, including c-Jun N-terminal kinase (JNK) and p38 mitogen-activated protein kinase (MAPK), and the expression of apoptosis-associated proteins, including p53, bax and caspase-3. VEGF-mediated angiogenesis of human umbilical vein endothelial cells (HUVECs), rat aortic ring and mouse cornea were used to detect the angioinhibitory activity of the PEDF-derived peptides. RESULTS: The MTT assay showed that the anti-VEGF effect of a 7-mer (Asp64-Ser70) was 1.5-fold greater than the 34-mer. In addition, massive apoptosis (37%) was induced by 7-mer treatment. The 7-mer induced JNK phosphorylation in RPMI8226 cells. Cell apoptosis and apoptosis-associated proteins induced by the 7-mer were blocked by pharmacological inhibition of JNK, but not p38 MAPK. Moreover, the 7-mer prevented VEGF-mediated angiogenesis of endothelial cells (ECs), including tube formation, aortic EC spreading and corneal neovascularization in mice. CONCLUSIONS: This is the first study to show that the PEDF 7-mer peptide manifests anti-VEGF activity, further establishing its potential as an anti-angiogenic agent.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Proteínas del Ojo/farmacología , Factores de Crecimiento Nervioso/farmacología , Péptidos/farmacología , Serpinas/farmacología , Animales , Línea Celular Tumoral , Células Endoteliales/metabolismo , Proteínas del Ojo/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ratones , Factores de Crecimiento Nervioso/metabolismo , Ratas , Serpinas/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
11.
Biomed Pharmacother ; 142: 111951, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34333290

RESUMEN

Tissue expansion is widely used to obtain new skin tissue for repairing defects in the clinical practice of plastic surgery. One major complication can be dermal thinning during expansion, which usually leads to skin rupture. Collagen synthesis can determine dermal thickness and can be influenced by macrophage polarization during expansion. The aim of the study was to test whether pigment epithelium-derived factor (PEDF) could be a modulator of collagen synthesis in fibroblasts by regulating macrophage polarization during skin expansion. Our results showed that PEDF mRNA expression was increased in expanded human and mouse epidermis. PEDF protein levels were elevated in the subcutaneous exudates of a rat skin expansion model. Increased PEDF mRNA expression was accompanied by dermal thinning during a three-week expansion protocol. Subcutaneous injection of PEDF in vivo further resulted in dermal thinning and cell number increase of M1 macrophage in the expanded skin. PEDF also promoted macrophage polarization in vitro to the M1 subtype under hypoxic conditions. PEDF did not influence collagen gene expression in fibroblasts directly, but attenuated collagen synthesis in a macrophage-mediated manner. Additionally, blockage of PEDF receptors on macrophages with inhibitors rescued collagen synthesis in fibroblasts. Our research demonstrated PEDF elevation in expanded skin leads to dermal thinning through M1 macrophage-mediated collagen synthesis inhibition in fibroblasts. Our results could form a basis for the development of novel strategies to improve skin integrity in expanded skin by using PEDF.


Asunto(s)
Colágeno/biosíntesis , Proteínas del Ojo/metabolismo , Proteínas del Ojo/farmacología , Fibroblastos/metabolismo , Activación de Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Factores de Crecimiento Nervioso/metabolismo , Factores de Crecimiento Nervioso/farmacología , Serpinas/metabolismo , Serpinas/farmacología , Animales , Hipoxia de la Célula , Línea Celular , Colágeno/genética , Epidermis/metabolismo , Proteínas del Ojo/genética , Humanos , Hipoxia/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Modelos Animales , Factores de Crecimiento Nervioso/genética , Ratas Sprague-Dawley , Receptores de Neuropéptido/antagonistas & inhibidores , Serpinas/genética , Piel/irrigación sanguínea , Piel/efectos de los fármacos , Piel/metabolismo , Expansión de Tejido
12.
J Invest Dermatol ; 141(12): 2820-2828, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34246620

RESUMEN

Pathomechanisms in IgA pemphigus are assumed to rely on Fc-dependent cellular activation by antigen-specific IgA autoantibodies; however, models for the disease and more detailed pathophysiologic data are lacking. In this study, we aimed to establish in vitro models of disease for IgA pemphigus, allowing us to study the effects of the interaction of anti-keratinocyte IgA with cell surface FcαRs. Employing multiple in vitro assays, such as a skin cryosection assay and a human skin organ culture model, in this study, we present mechanistic data for the pathogenesis of IgA pemphigus, mediated by anti-desmoglein 3 IgA autoantibodies. Our results reveal that this disease is dependent on FcαR-mediated activation of leukocytes in the epidermis. Importantly, this cell-dependent pathology can be dose-dependently abrogated by peptide-mediated inhibition of FcαR:IgA-Fc interaction, as confirmed in an additional model for IgA-dependent disease, that is, IgA vasculitis. These data suggest that IgA pemphigus can be modeled in vitro and that IgA pemphigus and IgA vasculitis are FcαR-dependent disease entities that can be specifically targeted in these experimental systems.


Asunto(s)
Inmunoglobulina A/inmunología , Neutrófilos/fisiología , Pénfigo/etiología , Receptores Fc/antagonistas & inhibidores , Antígenos CD/fisiología , Desmogleína 3/inmunología , Proteínas del Ojo/farmacología , Humanos , Pénfigo/inmunología , Fragmentos de Péptidos/farmacología , Receptores Fc/fisiología
13.
Invest Ophthalmol Vis Sci ; 62(7): 18, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-34132748

RESUMEN

Purpose: The cornea is richly innervated by the trigeminal ganglion (TG) and its function supported by secretions from the adjacent lacrimal (LG) and meibomian glands (MG). In this study we examined how pigment epithelium-derived factor (PEDF) gene deletion affects the cornea structure and function. Methods: We used PEDF hemizygous and homozygous knockout mice to study effects of PEDF deficiency on corneal innervation assessed by beta tubulin staining, mRNA expression of trophic factors, and PEDF receptors by adjacent supporting glands, corneal sensitivity measured using a Cochet-Bonnet esthesiometer, and tear production using phenol red cotton thread wetting. Results: Loss of PEDF was accompanied by reduced corneal innervation and sensitivity, increased corneal surface injury and tear production, thinning of the corneal stroma and loss of stromal cells. PEDF mRNA was expressed in the cornea and its supporting tissues, the TG, LG, and MG. Deletion of one or both PEDF alleles resulted in decreased expression of essential trophic support in the TG, LG, and MG including nerve growth factor, brain-derived neurotrophic growth factor, and GDNF with significantly increased levels of NT-3 in the LG and decreased EGF expression in the cornea. Decreased transcription of the putative PEDF receptors, adipose triglyceride lipase, lipoprotein receptor-related protein 6, laminin receptor, PLXDC1, and PLXDC2 was also evident in the TG, LG and MG with the first three showing increased levels in corneas of the Pedf+/- and Pedf-/- mice compared to wildtype controls. Constitutive inactivation of ERK1/2 and Akt was pronounced in the TG and cornea, although their protein levels were dramatically increased in Pedf-/- mice. Conclusions: This study highlights an essential role for PEDF in corneal structure and function and confirms the reported rescue of exogenous PEDF treatment in corneal pathologies. The pleiotropic effects of PEDF deletion on multiple trophic factors, receptors and signaling molecules are strong indications that PEDF is a key coordinator of molecular mechanisms that maintain corneal function and could be exploited in therapeutic options for several ocular surface diseases.


Asunto(s)
Córnea , Enfermedades de la Córnea , Proteínas del Ojo , Factores de Crecimiento Nervioso , Serpinas , Lágrimas/fisiología , Ganglio del Trigémino , Animales , Córnea/inervación , Córnea/patología , Córnea/fisiopatología , Enfermedades de la Córnea/metabolismo , Enfermedades de la Córnea/fisiopatología , Enfermedades de la Córnea/terapia , Lesiones de la Cornea/metabolismo , Lesiones de la Cornea/fisiopatología , Proteínas del Ojo/genética , Proteínas del Ojo/farmacología , Eliminación de Gen , Humanos , Ratones , Ratones Noqueados , Factores de Crecimiento Nervioso/deficiencia , Factores de Crecimiento Nervioso/genética , Factores de Crecimiento Nervioso/metabolismo , Factores de Crecimiento Nervioso/farmacología , Inhibidores de Proteasas/farmacología , Receptores de Neuropéptido/metabolismo , Serpinas/deficiencia , Serpinas/genética , Serpinas/farmacología , Ganglio del Trigémino/metabolismo , Ganglio del Trigémino/fisiopatología , Tubulina (Proteína)/metabolismo , Percepción Visual/fisiología
14.
J Neurochem ; 159(5): 840-856, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34133756

RESUMEN

Pigment epithelium-derived factor (PEDF) is a cytoprotective protein for the retina. We hypothesize that this protein acts on neuronal survival and differentiation of photoreceptor cells in culture. The purpose of the present study was to evaluate the neurotrophic effects of PEDF and its fragments in an in vitro model of cultured primary retinal neurons that die spontaneously in the absence of trophic factors. We used Wistar albino rats. Cell death was assayed by immunofluorescence and flow cytometry through TUNEL assay, propidium iodide, mitotracker, and annexin V. Immunofluorescence of cells for visualizing rhodopsin, CRX, and antisyntaxin under confocal microscopy was performed. Neurite outgrowth was also quantified. Results show that PEDF protected photoreceptor precursors from apoptosis, preserved mitochondrial function and promoted polarization of opsin enhancing their developmental process, as well as induced neurite outgrowth in amacrine neurons. These effects were abolished by an inhibitor of the PEDF receptor or receptor-derived peptides that block ligand/receptor interactions. While all the activities were specifically conferred by short peptide fragments (17 amino acid residues) derived from the PEDF neurotrophic domain, no effects were triggered by peptides from the PEDF antiangiogenic region. The observed effects on retinal neurons imply a specific activation of the PEDF receptor by a small neurotrophic region of PEDF. Our findings support the neurotrophic PEDF peptides as neuronal guardians for the retina, highlighting their potential as promoters of retinal differentiation, and inhibitors of retinal cell death and its blinding consequences. Cover Image for this issue: https://doi.org/10.1111/jnc.15089.


Asunto(s)
Células Amacrinas/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Proteínas del Ojo/farmacología , Factores de Crecimiento Nervioso/farmacología , Proyección Neuronal/efectos de los fármacos , Neuronas/efectos de los fármacos , Células Fotorreceptoras de Vertebrados/efectos de los fármacos , Serpinas/farmacología , Células Amacrinas/fisiología , Secuencia de Aminoácidos , Animales , Diferenciación Celular/fisiología , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Células Cultivadas , Proteínas del Ojo/genética , Femenino , Masculino , Factores de Crecimiento Nervioso/genética , Proyección Neuronal/fisiología , Neuronas/fisiología , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/farmacología , Células Fotorreceptoras de Vertebrados/fisiología , Ratas , Ratas Wistar , Serpinas/genética
15.
Cell Death Dis ; 12(4): 295, 2021 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-33731707

RESUMEN

Nasopharyngeal carcinoma (NPC) is one of the most malignant tumors in southern China and Asia, and lymph node metastasis is an important cause for treatment failure. Lymphangiogenesis is a crucial step in lymphatic metastasis of NPC, while little is known about lymphangiogenesis in NPC. Similar to angiogenesis, lymphangitic neovascularization is a process of balance between pro-lymphangiogenesis and anti-lymphangiogenesis factors, but there are few studies on endogenous lymphangiogenesis inhibitors. Pigment epithelium-derived factor (PEDF) is a well-known effective endogenous angiogenesis inhibitor. However, the relationship between PEDF and lymphangiogenesis remains unknown. Our present study reveals that PEDF is lowly expressed in human NPC tissues with poor prognosis and is negatively correlated with lymphatic vessel density (LVD). Consistently, PEDF inhibits lymphangiogenesis and lymphatic metastasis of NPC in vivo experiments. Mechanistically, PEDF inhibits the proliferation, migration, and tube formation of lymphatic endothelial cells and promotes cell apoptosis. On the other hand, PEDF reduces the expression and secretion of vascular endothelial growth factor C (VEGF-C) of NPC cells through the nuclear factor-κB (NF-κB) signaling pathway. Our findings indicate that PEDF plays a vital role in lymphatic metastasis by targeting both lymphatic endothelial cells and NPC cells, and PEDF may represent a novel therapeutic target for NPC.


Asunto(s)
Proteínas del Ojo/uso terapéutico , Metástasis Linfática/tratamiento farmacológico , Carcinoma Nasofaríngeo/tratamiento farmacológico , Factores de Crecimiento Nervioso/uso terapéutico , Inhibidores de Proteasas/uso terapéutico , Serpinas/uso terapéutico , Animales , Proteínas del Ojo/farmacología , Humanos , Ratones , Factores de Crecimiento Nervioso/farmacología , Inhibidores de Proteasas/farmacología , Serpinas/farmacología , Transfección
16.
Am J Pathol ; 191(4): 720-729, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33453179

RESUMEN

Pigment epithelium-derived factor (PEDF) is a widely expressed 50-kDa glycoprotein belonging to the serine protease inhibitor family, with well-established anti-inflammatory functions. Recently, we demonstrated the immunoregulatory role played by PEDF in dry eye disease (DED) by suppressing the maturation of antigen-presenting cells at the ocular surface following exposure to the desiccating stress. In this study, we evaluated the effect of PEDF on the immunosuppressive characteristics of regulatory T cells (Tregs), which are functionally impaired in DED. In the presence of PEDF, the in vitro cultures prevented proinflammatory cytokine (associated with type 17 helper T cells)-induced loss of frequency and suppressive phenotype of Tregs derived from normal mice. Similarly, PEDF maintained the in vitro frequency and enhanced the suppressive phenotype of Tregs derived from DED mice. On systemically treating DED mice with PEDF, moderately higher frequencies and significantly enhanced suppressive function of Tregs were observed in the draining lymphoid tissues, leading to the efficacious amelioration of the disease. Our results demonstrate that PEDF promotes the suppressive capability of Tregs and attenuates their type 17 helper T-cell-mediated dysfunction in DED, thereby playing a role in the suppression of DED.


Asunto(s)
Antiinflamatorios/farmacología , Síndromes de Ojo Seco/tratamiento farmacológico , Proteínas del Ojo/farmacología , Factores de Crecimiento Nervioso/farmacología , Serpinas/farmacología , Linfocitos T Reguladores/efectos de los fármacos , Células Th17/efectos de los fármacos , Animales , Células Presentadoras de Antígenos/efectos de los fármacos , Citocinas/genética , Modelos Animales de Enfermedad , Proteínas del Ojo/metabolismo , Femenino , Ratones Endogámicos C57BL , Factores de Crecimiento Nervioso/metabolismo , Fenotipo , Serpinas/metabolismo
17.
Curr Eye Res ; 46(3): 302-308, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32862727

RESUMEN

PURPOSE: To investigate the antioxidative effect and mechanism of pigment epithelium-derived factor (PEDF) on the ocular surface damage in diabetic mice. METHODS: C57BL/6 mice were injected intraperitoneally with streptozocin to generate diabetic models and then 50 nM PEDF or artificial tears were used to treat the diabetic mice. Treatment was given three times a day for eight weeks. Corneal epithelial damage, corneal sensitivity, and tear volume were quantified by fluorescein staining, esthesiometer, and phenol red cotton thread, respectively. Animals were sacrificed at 16 weeks after diabetes and the whole globe specimens were subjected to histochemical staining. Reactive oxygen species (ROS) generation was detected by 2',7-dichlorodihydrofluorescein probe. The levels of receptor for advanced glycation end products (RAGE) and superoxide dismutase 1 (SOD1) were examined by quantitative real-time PCR and western blotting. RESULTS: Topical application of PEDF improved corneal epithelial damage, increased corneal sensitivity, and tear volume in diabetic mice. ROS levels in the cornea were significantly higher in the diabetic mice than in the normal mice. Moreover, PEDF attenuated the accumulation of ROS, decreased the expression of RAGE, and elevated SOD1 expression in the cornea. CONCLUSIONS: Topical application of PEDF can alleviate diabetes-related ocular surface damage and increase tear volume, along with the improvement of oxidative stress status.


Asunto(s)
Antioxidantes/metabolismo , Córnea/efectos de los fármacos , Enfermedades de la Córnea/tratamiento farmacológico , Diabetes Mellitus Experimental/complicaciones , Proteínas del Ojo/farmacología , Factores de Crecimiento Nervioso/farmacología , Serpinas/farmacología , Lágrimas/metabolismo , Animales , Córnea/diagnóstico por imagen , Córnea/metabolismo , Enfermedades de la Córnea/etiología , Enfermedades de la Córnea/metabolismo , Diabetes Mellitus Experimental/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Inhibidores de Proteasas/farmacología , Especies Reactivas de Oxígeno/metabolismo
18.
Biochem Pharmacol ; 183: 114339, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33189676

RESUMEN

Lung cancer is one of the leading causes of death in cancer patients. Epithelial-mesenchymal transition (EMT) plays an important role in lung cancer progression. Therefore, for lung cancer treatment, it is crucial to find substances that inhibit EMT. Ethacrynic acid (ECA) is a diuretic that inhibits cellular ion flux and exerts anticancer effects. However, the effects of ECA on EMT in lung cancer remain unclear. We examined the effects of ECA on sphingosylphosphorylcholine (SPC) or TGF-ß1-induced EMT process in A549 and H1299 cells via reverse transcription polymerase chain reaction and Western blotting. We found that ECA inhibited SPC-induced EMT and SPC-induced WNT signalling in EMT. We observed that SPC induces the expression of NDP [Norrie disease protein] and WNT-2, whereas ECA suppressed their expression. SPC-induced WNT activation, EMT, migration, and invasion were suppressed by NDP small-interfering RNA (siNDP), but NDP overexpression (pNDP) enhanced these events in A549 and H1299 cells. Accordingly, NDP expression may influence lung cancer prognosis. In summary, our results revealed that ECA inhibited SPC or TGF-ß1-induced EMT in A549 and H1299 lung cancer cells by downregulating NDP expression and inhibiting WNT activation. Therefore, ECA might be a new drug candidate for lung cancer treatment.


Asunto(s)
Transición Epitelial-Mesenquimal/efectos de los fármacos , Ácido Etacrínico/farmacología , Proteínas del Ojo/farmacología , Neoplasias Pulmonares/metabolismo , Proteínas del Tejido Nervioso/farmacología , Inhibidores del Simportador de Cloruro Sódico y Cloruro Potásico/farmacología , Vía de Señalización Wnt/efectos de los fármacos , Células A549 , Animales , Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Relación Dosis-Respuesta a Droga , Transición Epitelial-Mesenquimal/fisiología , Ácido Etacrínico/uso terapéutico , Proteínas del Ojo/antagonistas & inhibidores , Proteínas del Ojo/biosíntesis , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteínas del Tejido Nervioso/biosíntesis , ARN Interferente Pequeño/farmacología , Inhibidores del Simportador de Cloruro Sódico y Cloruro Potásico/uso terapéutico , Vía de Señalización Wnt/fisiología
19.
Int J Mol Sci ; 21(19)2020 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-33008127

RESUMEN

Here, we evaluated the effects of PEDF (pigment epithelium-derived factor) and PEDF peptides on cone-photoreceptor cell damage in a mouse model of focal LED-induced phototoxicity (LIP) in vivo. Swiss mice were dark-adapted overnight, anesthetized, and their left eyes were exposed to a blue LED placed over the cornea. Immediately after, intravitreal injection of PEDF, PEDF-peptide fragments 17-mer, 17-mer[H105A] or 17-mer[R99A] (all at 10 pmol) were administered into the left eye of each animal. BDNF (92 pmol) and bFGF (27 pmol) injections were positive controls, and vehicle negative control. After 7 days, LIP resulted in a consistent circular lesion located in the supratemporal quadrant and the number of S-cones were counted within an area centered on the lesion. Retinas treated with effectors had significantly greater S-cone numbers (PEDF (60%), 17-mer (56%), 17-mer [H105A] (57%), BDNF (64%) or bFGF (60%)) relative to their corresponding vehicle groups (≈42%). The 17-mer[R99A] with no PEDF receptor binding and no neurotrophic activity, PEDF combined with a molar excess of the PEDF receptor blocker P1 peptide, or with a PEDF-R enzymatic inhibitor had undetectable effects in S-cone survival. The findings demonstrated that the cone survival effects were mediated via interactions between the 17-mer region of the PEDF molecule and its PEDF-R receptor.


Asunto(s)
Proteínas del Ojo/farmacología , Factores de Crecimiento Nervioso/farmacología , Péptidos/farmacología , Retina/efectos de los fármacos , Células Fotorreceptoras Retinianas Conos/efectos de los fármacos , Serpinas/farmacología , Animales , Córnea/efectos de los fármacos , Córnea/crecimiento & desarrollo , Córnea/metabolismo , Dermatitis Fototóxica , Modelos Animales de Enfermedad , Proteínas del Ojo/metabolismo , Humanos , Ratones , Factores de Crecimiento Nervioso/metabolismo , Fragmentos de Péptidos/farmacología , Péptidos/genética , Fotoperiodo , Receptores de Neuropéptido/genética , Retina/crecimiento & desarrollo , Células Fotorreceptoras Retinianas Conos/metabolismo , Células Fotorreceptoras Retinianas Conos/patología , Serpinas/metabolismo
20.
Mol Vis ; 26: 641-651, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33088168

RESUMEN

Purpose: Tofacitinib is a pan-Janus kinase (JAK) inhibitor that suppresses cytokine signaling and in turn, the cells that participate in inflammatory immunopathogenic processes. We examined the capacity of tofacitinib to inhibit the induction of experimental autoimmune uveitis (EAU) and related immune responses. Methods: EAU was induced in B10.A mice with immunization with bovine interphotoreceptor retinoid-binding protein (IRBP), emulsified in complete Freund's adjuvant (CFA), and a simultaneous injection of pertussis toxin. Tofacitinib, 25 mg/kg, was administered daily, and the vehicle was used for control. EAU development was assessed by histological analysis of the mouse eyes, and related immune responses were assessed by (i) the levels of interferon (IFN)-γ and interleukin (IL)-17, secreted by spleen cells cultured with IRBP; (ii) flow cytometric analysis of intracellular expression by spleen, or eye-infiltrating CD4 or CD8 cells of IFN-γ, IL-17, and their transcription factors, T-bet and RORγt. In addition, the inflammation-related cell markers CD44 and CD62L and Ki67, a proliferation marker, were tested. The proportions of T-regulatory cells expressing FoxP3 were determined by flow cytometric intracellular staining, while levels of antibody to IRBP were measured with enzyme-linked immunosorbent assay (ELISA). Results: Treatment with tofacitinib significantly suppressed the development of EAU and reduced the levels of secreted IFN-γ, but not of IL-17. Further, treatment with tofacitinib reduced in the spleen and eye-infiltrating cells the intracellular expression of IFN-γ and its transcription factor T-bet. In contrast, treatment with tofacitinib had essentially no effect on the intracellular expression of IL-17 and its transcription factor, RORγt. The selective effect of tofacitinib treatment was particularly evident in the CD8 population. Treatment with tofacitinib also increased the population of CD44, but reduced the populations of cells producing CD62L and Ki67. Treatment with tofacitinib had no effect on the proportion of FoxP3 producing regulatory cells and on the antibody production to IRBP. Conclusions: Treatment with tofacitinib inhibited the development of EAU, reduced the production of IFN-γ, but had essentially no effect on the production of IL-17.


Asunto(s)
Ojo/metabolismo , Piperidinas/farmacología , Pirimidinas/farmacología , Células TH1/efectos de los fármacos , Células Th17/efectos de los fármacos , Uveítis/tratamiento farmacológico , Uveítis/inmunología , Animales , Antígenos CD4/sangre , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Antígenos CD8/sangre , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Ojo/efectos de los fármacos , Ojo/patología , Proteínas del Ojo/farmacología , Factores de Transcripción Forkhead/sangre , Receptores de Hialuranos/sangre , Terapia de Inmunosupresión , Interferón gamma/sangre , Interleucina-17/sangre , Antígeno Ki-67/sangre , Selectina L/sangre , Ratones , Piperidinas/administración & dosificación , Pirimidinas/administración & dosificación , Proteínas de Unión al Retinol/farmacología , Células TH1/inmunología , Células Th17/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA