Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
Cancer Sci ; 113(4): 1220-1234, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35189004

RESUMO

Owing to recent advances in immunotherapies, the overall survival of patients with skin cutaneous melanoma (SKCM) has increased; however, the 5-year survival rate of metastatic patients remains poor. Skin cutaneous melanoma-upregulated genes were screened via analysis of differentially expressed genes (GSE3189 and GSE46517), and metastasis-related oncogenes were identified via weighted gene coexpression network analysis of the GSE46517 dataset. As confirmed by the Tumor Immune Estimation Resource, we found highly expressed centromere protein F (CENPF) in SKCM and its metastases. Immunostaining of human melanoma tissues demonstrated high CENPF expression. According to the Kaplan-Meier survival curve log-rank test, receiver-operating characteristic curve, and univariate and multivariate analyses, the Cancer Genome Atlas (TCGA) database suggested CENPF be a typical independent predictor of SKCM. The CIBERSORT algorithm classified the types of the immune cells from GSE46517 and showed higher proportion of CD4+ memory-activated T cells in metastatic melanoma. Single-sample gene set enrichment analysis of TCGA data confirmed the correlation between CENPF and activated CD4+ T cells. Centromere protein F was positively correlated with tumor mutational burden and CD4+ memory T cell markers (interleukin [IL]-23A, CD28, and CD62L), negatively associated with memory T cell maintenance factors (IL-7 and IL-15) by correlation analysis. Moreover, immunofluorescence showed high coexpression of CENPF and IL23A, CD4 in melanoma. Upregulated CENPF might lead to premature depletion of CD4+ memory T cells and immunosuppression. Nomogram indicated CENPF clinical predictive value for 1-, 3-, 5-, and 7-year melanoma overall survival. Therefore, CENPF plays a vital role in the progression and metastasis of melanoma and can be an effective therapeutic target.


Assuntos
Melanoma , Neoplasias Cutâneas , Biomarcadores Tumorais/metabolismo , Linfócitos T CD4-Positivos/metabolismo , Proteínas Cromossômicas não Histona , Regulação Neoplásica da Expressão Gênica , Humanos , Melanoma/patologia , Células T de Memória , Proteínas dos Microfilamentos , Prognóstico , Neoplasias Cutâneas/patologia , Melanoma Maligno Cutâneo
2.
Lab Invest ; 100(5): 751-761, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31925326

RESUMO

The skin plays a critical role in maintenance of water homeostasis. Dysfunction of the skin barrier causes not only delayed wound healing and hypertrophic scarring, but it also contributes to the development of various skin diseases. Dermatitis is a chronic inflammatory skin disorder that has several different subtypes. Skin of contact dermatitis and atopic dermatitis (AD) show epidermal barrier dysfunction. Nax is a sodium channel that regulates inflammatory gene expression in response to perturbation of barrier function of the skin. We found that in vivo knockdown of Nax using RNAi reduced hyperkeratosis and keratinocyte hyperproliferation in rabbit ear dermatitic skin. Increased infiltration of inflammatory cells (mast cells, eosinophils, T cells, and macrophages), a characteristic of dermatitis, was reduced by Nax knockdown. Upregulation of PAR-2 and thymic stromal lymphopoietin (TSLP), which induce Th2-mediated allergic responses, was inhibited by Nax knockdown. In addition, expression of COX-2, IL-1ß, IL-8, and S100A9, which are downstream genes of Nax and are involved in dermatitis pathogenesis, were also decreased by Nax knockdown. Our data show that knockdown of Nax relieved dermatitis symptoms in vivo and indicate that Nax is a novel therapeutic target for dermatitis, which currently has limited therapeutic options.


Assuntos
Dermatite Atópica , Pele , Canais de Sódio Disparados por Voltagem , Animais , Proliferação de Células/genética , Dermatite Atópica/genética , Dermatite Atópica/patologia , Dermatite Atópica/fisiopatologia , Regulação para Baixo/genética , Eosinófilos/metabolismo , Feminino , Técnicas de Silenciamento de Genes , Inflamação/genética , Inflamação/patologia , Inflamação/fisiopatologia , Queratinócitos/metabolismo , Ceratose/genética , Ceratose/patologia , Ceratose/fisiopatologia , Mastócitos/metabolismo , Coelhos , Pele/citologia , Pele/patologia , Pele/fisiopatologia , Canais de Sódio Disparados por Voltagem/genética , Canais de Sódio Disparados por Voltagem/metabolismo
3.
BMC Cancer ; 20(1): 927, 2020 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-32993558

RESUMO

BACKGROUND: Human skin cutaneous melanoma is the most common and dangerous skin tumour, but its pathogenesis is still unclear. Although some progress has been made in genetic research, no molecular indicators related to the treatment and prognosis of melanoma have been found. In various diseases, dysregulation of lncRNA is common, but its role has not been fully elucidated. In recent years, the birth of the "competitive endogenous RNA" theory has promoted our understanding of lncRNAs. METHODS: To identify the key lncRNAs in melanoma, we reconstructed a global triple network based on the "competitive endogenous RNA" theory. Gene Ontology and KEGG pathway analysis were performed using DAVID (Database for Annotation, Visualization, and Integration Discovery). Our findings were validated through qRT-PCR assays. Moreover, to determine whether the identified hub gene signature is capable of predicting the survival of cutaneous melanoma patients, a multivariate Cox regression model was performed. RESULTS: According to the "competitive endogenous RNA" theory, 898 differentially expressed mRNAs, 53 differentially expressed lncRNAs and 16 differentially expressed miRNAs were selected to reconstruct the competitive endogenous RNA network. MALAT1, LINC00943, and LINC00261 were selected as hub genes and are responsible for the tumorigenesis and prognosis of cutaneous melanoma. CONCLUSIONS: MALAT1, LINC00943, and LINC00261 may be closely related to tumorigenesis in cutaneous melanoma. In addition, MALAT1 and LINC00943 may be independent risk factors for the prognosis of patients with this condition and might become predictive molecules for the long-term treatment of melanoma and potential therapeutic targets.


Assuntos
Carcinogênese/genética , Melanoma/genética , RNA Longo não Codificante/genética , Neoplasias Cutâneas/genética , Feminino , Regulação Neoplásica da Expressão Gênica/genética , Redes Reguladoras de Genes/genética , Humanos , Estimativa de Kaplan-Meier , Masculino , MicroRNAs/genética , Prognóstico , RNA Longo não Codificante/classificação , RNA Mensageiro/genética , Melanoma Maligno Cutâneo
4.
Exp Dermatol ; 28(5): 576-584, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30903711

RESUMO

Nax is an atypical sodium channel that mediates inflammatory pathways in pathological conditions of the skin. In this study, we developed a skin inflammation model in the rabbit ear through application of imiquimod (IMQ). Knockdown of Nax using RNAi attenuated IMQ-induced skin inflammation, including skin erythema, scaling and papule formation. Histologic analysis showed that thickening and insufficient differentiation of the epidermis found in psoriasis-like skin were normalized by administration of Nax -RNAi. Excessive infiltration of inflammatory cells found in inflammatory lesions, such as mast cells, eosinophils, neutrophils, T cells and macrophages, was reduced by Nax -RNAi. Expression of S100A9, which is a downstream gene of Nax and a mediator of inflammation, was decreased by Nax -RNAi. Our results demonstrated that knockdown of Nax ameliorated IMQ-induced psoriasis-like skin inflammation in vivo. Thus, targeting of Nax may represent a potential therapeutic option for the treatment of psoriasis.


Assuntos
Dermatite/tratamento farmacológico , Imiquimode/farmacologia , Psoríase/tratamento farmacológico , Pele/efeitos dos fármacos , Canais de Sódio Disparados por Voltagem/genética , Animais , Epiderme/efeitos dos fármacos , Feminino , Técnicas de Silenciamento de Genes , Inflamação/induzido quimicamente , Psoríase/induzido quimicamente , Interferência de RNA , Coelhos , Pele/patologia
5.
BMC Cancer ; 19(1): 609, 2019 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-31226958

RESUMO

BACKGROUND: Esophageal squamous cell carcinoma (ESCC) is the major subtype of esophageal cancer with high aggressiveness and poor prognosis. There is an urgent need for understanding the molecular mechanism underlying the development and progression of ESCC. METHODS: ESCC tissues and corresponding non-neoplastic tissues were collected. The expression and function of miR-124-3p and BCAT1 in two cell lines KYSE-150 and Eca109 were determined. RESULTS: We show downregulation of miR-124-3p expression in ESCC tissues, which is highly correlated with proliferation and migration of ESCC cell lines KYSE-150 and Eca109. miR-124-3p show high correlation with TNM stage and differentiation grade. Furthermore, miR-124-3p directly targets mRNA 3'UTR region of BCAT1, which results in upregulation of BCAT1 expression as observed in ESCC tissues and cell lines. Also, our data indicates that BCAT1 high expression is strongly linked to the disease-free survival, tumor size, pathologic stage, T classification and differentiation grade. On the other hand, we clarified the upstream mechanism regulating miR-124-3p expression in ESCC, which involves in the hypermethylation-silencing regulation mediated by DNA methyltransferase 1(DNMT1), which is of high expression in ESCC tissues and cell lines in the present study. In addition, DNMT1 knockdown or inhibition of DNMT1 function contributes to downregulation of miR-124-3p and BCAT1 expression. CONCLUSIONS: Our study thus clarifies a new mechanism that DNMT1/miR-124/BCAT1 axis regulates the development and progression of ESCC.


Assuntos
DNA (Citosina-5-)-Metiltransferase 1/metabolismo , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/patologia , Carcinoma de Células Escamosas do Esôfago/metabolismo , Carcinoma de Células Escamosas do Esôfago/patologia , Transaminases/metabolismo , Regiões 3' não Traduzidas , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Sobrevivência Celular , DNA (Citosina-5-)-Metiltransferase 1/genética , Metilação de DNA , Progressão da Doença , Intervalo Livre de Doença , Feminino , Humanos , Masculino , MicroRNAs/genética , MicroRNAs/metabolismo , Pessoa de Meia-Idade , Invasividade Neoplásica/fisiopatologia , Estadiamento de Neoplasias , Transaminases/genética , Transfecção
6.
Cell Physiol Biochem ; 49(6): 2333-2347, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30261495

RESUMO

BACKGROUND/AIMS: Little is known how miR-203 is involved in epidermal stem cells (ESCs) differentiation and scar formation. METHODS: We first used luciferase assay to determine the interaction of miR-203 with the 3'-UTR in regulation of Hes1 expression. We then used flow cytometry to analyze the effects of miR-203 expression on the differentiation of ESCs to MFB by determination of CK15 ratio and α-SMA. To confirm the results of flow cytometry analysis, we used Western blot to examine the expression of α-SMA, Collagen I (Col I), and Collagen III (Col III), as well as the expression of Notch1, Jagged1, and Hes1 in ESCs after the treatment of pre-miR-203 or anti-miR-203. Finally, we examined the effects local anti-miR-203 treatment on would closure and scar formation using a mouse skin wound model. RESULTS: Pre-miR-203 treatment increased ESCs differentiation to MFB cells, as indicated by decreased CK15 ratio and increased MFB biomarkers. This phenomenon was reversed by overexpression of Hes1 in ESCs. In addition, skin incision increased expression of miR-203 in wound tissue. Local treatment of anti-miR-203 could accelerate wound closure and reduce scar formation in vivo, which was associated with increased re-epithelialization, skin attachment regeneration, and collagen reassignment. Finally, we confirmed that anti-miR-203 treatment could inhibit ESCs differentiation in vivo via increasing Hesl expression. CONCLUSION: Taken together, our results suggested that overexpression of miR-203 in ESCs after skin wound may be a critical mechanism underlying the scar formation.


Assuntos
Cicatriz/prevenção & controle , MicroRNAs/metabolismo , Fatores de Transcrição HES-1/metabolismo , Cicatrização , Regiões 3' não Traduzidas , Actinas/metabolismo , Animais , Antagomirs/metabolismo , Diferenciação Celular , Cicatriz/patologia , Células Epidérmicas , Feminino , Hiperplasia/patologia , Queratina-15/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/antagonistas & inibidores , MicroRNAs/genética , Miofibroblastos/citologia , Miofibroblastos/metabolismo , Pele/patologia , Células-Tronco/citologia , Células-Tronco/metabolismo , Fatores de Transcrição HES-1/antagonistas & inibidores , Fatores de Transcrição HES-1/genética
7.
Am J Pathol ; 186(1): 109-22, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26597884

RESUMO

The most critical function of the epidermis is to prevent water loss and maintain skin homeostasis. Disruption of the functional skin barrier causes delayed wound healing, hypertrophic scarring, and many skin diseases. Herein, we show that reduced hydration increases the expression of S100 protein family members, S100A8/S100A9, in stratified keratinocyte culture and human ex vivo skin culture. Immunohistological analyses show that S100A8/A9 are highly expressed in the epidermis of human hypertrophic scar and keloid tissues. Reduced hydration demonstrates activation of fibroblasts in the keratinocyte-fibroblast co-culture. In contrast, knockdown of S100A8 or S100A9 by RNA interference in keratinocytes failed to activate fibroblasts. Pretreatment with pharmacological blockers of S100A8/A9 receptors, Toll-like receptor 4 and receptor for advanced glycation end products, inhibits fibroblast activation induced by recombinant S100A8/A9 proteins. Moreover, we observe that local delivery of S100A8 protein results in a marked increase in hypertrophic scarring in the in vivo rabbit ear scar model. Our results indicate that hydration status promotes fibroblast activation and fibrosis by directly affecting the expression of inflammatory signaling in keratinocytes, thereby strongly suggesting S100A8/A9 to be novel targets in preventing scarring.


Assuntos
Calgranulina A/biossíntese , Calgranulina B/biossíntese , Cicatriz/metabolismo , Epiderme/patologia , Fibroblastos/metabolismo , Queratinócitos/metabolismo , Adulto , Animais , Western Blotting , Técnicas de Cocultura , Derme/patologia , Feminino , Fibrose/patologia , Técnicas de Silenciamento de Genes , Humanos , Imuno-Histoquímica , Masculino , Coelhos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Água , Adulto Jovem
8.
Exp Dermatol ; 25(8): 604-10, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-26997546

RESUMO

Hypertrophic scarring is a common dermal fibroproliferative disorder characterized by excessive collagen deposition. Prostaglandin E2 (PGE2 ), an important inflammatory product synthesized via the arachidonic acid cascade, has been shown to act as a fibroblast modulator and to possess antifibroblastic activity. However, the mechanism underlying the antifibrotic effect of PGE2 remains unclear. In this study, we explored the effects of PGE2 on TGF-ß1-treated dermal fibroblasts in terms of collagen production and to determine the regulatory pathways involved, as well as understand the antiscarring function of PGE2 in vivo. We found that PGE2 inhibited TGF-ß1-induced collagen synthesis by regulating the balance of matrix metalloproteinases (MMPs) and tissue inhibitor of metalloproteinase (TIMP). It did so by upregulating cAMP through the E prostanoid (EP)2 receptor. We determined that inhibition of the TGF-ß1/Smad pathway by PGE2 is associated with its ability to inhibit collagen synthesis. An in vivo study further confirmed that PGE2 inhibits hypertrophic scar formation by decreasing collagen production. Our results demonstrate that the novel anti-scarring function of PGE2 is achieved by balancing MMPs/TIMP expression and decreasing collagen production.


Assuntos
Cicatriz/prevenção & controle , Colágeno/biossíntese , Dinoprostona/uso terapêutico , Fibroblastos/metabolismo , Animais , Azetidinas , Colforsina , AMP Cíclico/metabolismo , Ciclo-Oxigenase 2/metabolismo , Dinoprostona/metabolismo , Dinoprostona/farmacologia , Avaliação Pré-Clínica de Medicamentos , Feminino , Fibroblastos/efeitos dos fármacos , Humanos , Isoindóis , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Coelhos , Transdução de Sinais/efeitos dos fármacos , Proteínas Smad/metabolismo , Sulfonamidas , Inibidor Tecidual de Metaloproteinase-1/metabolismo , Fator de Crescimento Transformador beta1
9.
J Cell Physiol ; 230(8): 1895-905, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25546411

RESUMO

Endothelial dysfunction is a major characteristic of diabetic vasculopathy. Protection of the vascular endothelium is an essential aspect of preventing and treating diabetic vascular complications. Although Angiopoietin-1 (Ang-1) is an important endothelial-specific protective factor, whether Ang-1 protects vascular cells undergoing advanced glycation end product (AGE) injury has not been investigated. The aim of the present study was to determine the potential effects of Ang-1 on endothelial cells after exposure to AGE. We show here that Ang-1 prevented AGE-induced vascular leakage by enhancing the adherens junctions between endothelial cells, and this process was mediated by the phosphorylation and membrane localization of VE-cadherin. Furthermore, Ang-1 also protected endothelial cells from AGE-induced death by regulating phosphatidylinositol 3-kinase (PI3K)/Akt-dependent Bad phosphorylation. Our findings suggest that the novel protective mechanisms of Ang-1 on endothelium are achieved by strengthening endothelial cell junctions and reducing endothelial cell death after AGE injury.


Assuntos
Angiopoietina-1/metabolismo , Apoptose/fisiologia , Células Endoteliais/patologia , Produtos Finais de Glicação Avançada/toxicidade , Junções Intercelulares/metabolismo , Angiopoietina-1/farmacologia , Apoptose/efeitos dos fármacos , Western Blotting , Permeabilidade Capilar/efeitos dos fármacos , Permeabilidade Capilar/fisiologia , Linhagem Celular , Imunofluorescência , Técnicas de Silenciamento de Genes , Humanos , Imunoprecipitação , Marcação In Situ das Extremidades Cortadas , Junções Intercelulares/efeitos dos fármacos
10.
Inflammation ; 2024 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-38954262

RESUMO

Long-term inflammation and impaired angiogenesis are thought to be the causes of delayed healing or nonhealing of diabetic wounds. S100A12 is an essential pro-inflammatory factor involved in inflammatory reactions and serves as a biomarker for various inflammatory diseases. However, whether high level of S100A12 exists in and affects the healing of diabetic wounds, as well as the underlying molecular mechanisms, remain unclear. In this study, we found that the serum concentration of S100A12 is significantly elevated in patients with type 2 diabetes. Exposure of stratified epidermal cells to high glucose environment led to increased expression and secretion of S100A12, resulting in impaired endothelial function by binding to the advanced glycation endproducts (RAGE) or Toll-like receptor 4 (TLR4) on endothelial cell. The transcription factor Krüpple-like Factor 5 (KLF5) is highly expressed in the epidermis under high glucose conditions, activating the transcriptional activity of the S100A12 and boost its expression. By establishing diabetic wounds model in alloxan-induced diabetic rabbit, we found that local inhibition of S100A12 significantly accelerated diabetic wound healing by promoting angiogenesis. Our results illustrated the novel endothelial-specific injury function of S100A12 in diabetic wounds and suggest that S100A12 is a potential target for the treatment of diabetic wounds.

11.
EBioMedicine ; 106: 105258, 2024 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-39068733

RESUMO

BACKGROUND: Severe burn wounds face two primary challenges: dysregulated cellular impairment functions following infection and an unbalanced wound hydration microenvironment leading to excessive inflammation and collagen deposition. These results in hypertrophic scar contraction, causing significant deformity and disability in survivors. METHODS: A three-dimensional (3D) printed double-layer hydrogel (DLH) was designed and fabricated to address the problem of scar formation after burn injury. DLH was developed using methacrylated silk fibroin (SFMA) and gelatin methacryloyl (GelMA) for the upper layer, and GelMA and hyaluronic acid methacryloyl (HAMA) for the lower layer. To combat infection, copper-epigallocatechin gallate (Cu-EGCG) was incorporated into the lower layer bioink, collectively referred to as DLS. To balance wound hydration levels, HaCaT cells were additionally encapsulated in the upper layer, designed as DLS/c. FINDINGS: DLH demonstrated suitable porosity, appropriate mechanical properties, and excellent biocompatibility. DLS exhibited potent antimicrobial properties, exerted anti-inflammatory effects by regulating macrophage polarisation, and may enhance angiogenesis through the HIF-1α/VEGF pathway. In the DLS/c group, animal studies showed significant improvements in epidermal formation, barrier function, and epidermal hydration, accompanied by reduced inflammation. In addition, Masson's trichrome and Sirius red staining revealed that the structure and ratio of dermal collagen in DLS/c resembled that of normal skin, indicating considerable potential for scarless wound healing. INTERPRETATION: This biomimetic matrix shows promise in addressing the challenges of burn wounds and aiming for scarless repair, with benefits such as anti-infection, epidermal hydration, biological induction, and optimised topological properties. FUNDING: Shown in Acknowledgements.

12.
Bioeng Transl Med ; 8(1): e10373, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36684072

RESUMO

Burn infection delays wound healing and increases the burn patient mortality. Consequently, a new dressing with antibacterial and anti-inflammatory dual properties is urgently required for wound healing. In this study, we propose a combination of methacrylate gelatin (GelMA) hydrogel system with silver nanoparticles embed in γ-cyclodextrin metal-organic frameworks (Ag@MOF) and hyaluronic acid-epigallocatechin gallate (HA-E) for the burn wound infection treatment. Ag@MOF is used as an antibacterial agent and epigallocatechin gallate (EGCG) has exhibited biological properties of anti-inflammation and antibacterial. The GelMA/HA-E/Ag@MOF hydrogel enjoys suitable physical properties and sustained release of Ag+. Meanwhile, the hydrogel has excellent biocompatibility and could promote macrophage polarization from M1 to M2. In vivo wound healing evaluations further demonstrate that the GelMA/HA-E/Ag@MOF hydrogel reduces the number of the bacterium efficiently, accelerates wound healing, promotes early angiogenesis, and regulates immune reaction. A further evaluation indicates that the noncanonical Wnt signal pathway is significantly activated in the GelMA/HA-E/Ag@MOF hydrogel treated group. In conclusion, the GelMA/HA-E/Ag@MOF hydrogel could serve as a promising multifunctional dressing for the burn wound healing.

13.
Dev Cell ; 58(13): 1139-1152.e6, 2023 07 10.
Artigo em Inglês | MEDLINE | ID: mdl-37192621

RESUMO

Pressure therapy (PT) is an effective intervention for reducing scarring, but its underlying mechanism remains largely unclear. Here, we demonstrate that human scar-derived myofibroblasts dedifferentiate into normal fibroblasts in response to PT, and we identify how SMYD3/ITGBL1 contributes to the nuclear relay of mechanical signals. In clinical specimens, reductions in SMYD3 and ITGBL1 expression levels are strongly associated with the anti-scarring effects of PT. The integrin ß1/ILK pathway is inhibited in scar-derived myofibroblasts upon PT, leading to decreased TCF-4 and subsequently to reductions in SMYD3 expression, which reduces the levels of H3K4 trimethylation (H3K4me3) and further suppresses ITGBL1 expression, resulting the dedifferentiation of myofibroblasts into fibroblasts. In animal models, blocking SMYD3 expression results in reductions of scarring, mimicking the positive effects of PT. Our results show that SMYD3 and ITGBL1 act as sensors and mediators of mechanical pressure to inhibit the progression of fibrogenesis and provide therapeutic targets for fibrotic diseases.


Assuntos
Cicatriz , Miofibroblastos , Animais , Humanos , Miofibroblastos/metabolismo , Cicatriz/patologia , Fibroblastos/metabolismo , Transdução de Sinais , Integrina beta1/metabolismo , Histona-Lisina N-Metiltransferase/metabolismo
14.
Eur J Med Res ; 28(1): 473, 2023 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-37915101

RESUMO

BACKGROUND AND PURPOSE: The efficacy and safety of tirofiban in endovascular therapy for cardioembolic ischemic stroke patients remain controversial. This study aimed to evaluate the role of intravenous tirofiban before endovascular therapy in cardioembolic stroke. METHODS: This post hoc analysis utilized data from the RESCUE BT (Endovascular Treatment With versus Without Tirofiban for Patients with Large Vessel Occlusion Stroke) trial, which was an investigator-initiated, randomized, double-blind, placebo-controlled trial. Participants were randomized to receive either tirofiban or a placebo in a 1:1 ratio before undergoing endovascular therapy. The study included patients aged 18 years or older, presenting with occlusion of the internal carotid artery or middle cerebral artery (MCA) M1/M2 within 24 h of the last known well time, and with a stroke etiology of cardioembolism. The primary efficacy outcome was global disability at 90 days, assessed using the modified Rankin Scale (mRS). The safety outcome included symptomatic intracranial hemorrhage (sICH) within 48 h and mortality within 90 days. RESULTS: A total of 406 cardioembolic stroke patients were included in this study, with 212 assigned to the tirofiban group and 194 assigned to the placebo group. Tirofiban treatment did not correlate with a favorable shift towards a lower 90-day mRS score (adjusted common odds ratio [OR], 0.91; 95% CI 0.64-1.3; p = 0.617). However, the tirofiban group had a significantly higher risk of symptomatic intracranial hemorrhage (sICH) within 48 h (adjusted OR, 3.26; 95% CI 1.4-7.57; p = 0.006) compared to the placebo group. The adjusted odds ratio (aOR) for mortality within 90 days was 1.48 (95% CI 0.88-2.52; p = 0.143). CONCLUSIONS: Tirofiban treatment was not associated with a lower level of disability and increased the incidence of sICH after endovascular therapy in cardioembolic stroke patients.


Assuntos
Isquemia Encefálica , AVC Embólico , Procedimentos Endovasculares , Acidente Vascular Cerebral , Humanos , Tirofibana/uso terapêutico , AVC Embólico/complicações , AVC Embólico/tratamento farmacológico , Isquemia Encefálica/tratamento farmacológico , Resultado do Tratamento , Acidente Vascular Cerebral/tratamento farmacológico , Acidente Vascular Cerebral/etiologia , Hemorragias Intracranianas/induzido quimicamente , Hemorragias Intracranianas/complicações , Procedimentos Endovasculares/efeitos adversos
15.
Front Endocrinol (Lausanne) ; 14: 1275612, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38107519

RESUMO

Background: The treatment of diabetic foot ulcers (DFUs) poses a challenging medical problem that has long plagued individuals with diabetes. Clinically, wounds that fail to heal for more than 12 weeks after the formation of DFUs are referred to as non-healing/chronic wounds. Among various factors contributing to the non-healing of DFUs, the impairment of skin microvascular endothelial cell function caused by high glucose plays a crucial role. Our study aimed to reveal the transcriptomic signatures of non-healing DFUs endothelial cells, providing novel intervention targets for treatment strategies. Methods: Based on the GEO dataset (GSE165816), we selected DFU-Healer, DFU-Non-healer, and healthy non-diabetic controls as research subjects. Single-cell RNA transcriptomic sequencing technology was employed to analyze the heterogeneity of endothelial cells in different skin tissue samples and identify healing-related endothelial cell subpopulations. Immunofluorescence was applied to validate the sequencing results on clinical specimens. Results: The number of endothelial cells and vascular density showed no significant differences among the three groups of skin specimens. However, endothelial cells from non-healing DFUs exhibited apparent inhibition of angiogenesis, inflammation, and immune-related signaling pathways. The expression of CCND1, ENO1, HIF1α, and SERPINE1 was significantly downregulated at the transcriptomic and histological levels. Further analysis demonstrated that healing-related endothelial cell subpopulations in non-healing DFUs has limited connection with other cell types and weaker differentiation ability. Conclusion: At the single-cell level, we uncovered the molecular and functional specificity of endothelial cells in non-healing DFUs and highlighted the importance of endothelial cell immune-mediated capability in angiogenesis and wound healing. This provides new insights for the treatment of DFUs.


Assuntos
Diabetes Mellitus , Pé Diabético , Humanos , Células Endoteliais/metabolismo , Pele/metabolismo , Cicatrização/genética , Perfilação da Expressão Gênica
16.
Front Nutr ; 9: 1018534, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36276839

RESUMO

Tomato is one of the most widely cultivated horticultural plants in the world, while the key volatile compounds of tomato fruits generally derive from fatty acid, carotenoid, phenylalanine, and branched-chain amino acid pathways. As an important endogenous signal molecule, methyl salicylate (MeSA) plays a crucial role in the fruit ripening process of plant. Recently, it has been demonstrated that MeSA can maintain the flavor quality of full ripe tomatoes after cold-storage preservation. However, few research teams attempted to investigate the effects of MeSA plus low temperature treatment on the different volatile biosynthetic pathways of tomatoes previously. Therefore, in this study, the effects of methyl salicylate pre-treatment (0.05 mM MeSA, 24 h) on the volatile profile and flavor-related key gene expressions of tomato fruits stored at 10°C were evaluated for the first time. Our results showed that the loss of volatile compounds in low temperature-treated tomato fruits could be effectively alleviated by MeSA pre-treatment. Although MeSA had no remarkable effect on the formation of carotenoid pathway- and branched-chain amino acid pathway-related volatiles in tomatoes subjected to low temperature, the content of fatty acid pathway-related volatiles (including cis-3-hexenal, hexanal, and trans-2-hexenal) in full red fruits of 10°C MeSA group was remarkably higher than that of 10°C control group. Furthermore, MeSA pre-treatment significantly up-regulated the expression of LOXC or LOXD gene in low temperature-treated fruits at breaker or full red stage, respectively. In conclusion, pre-treatment with MeSA might avoid the loss of aromatic compounds in tomato fruits stored at low temperature by activating the fatty acid pathway.

17.
Bioact Mater ; 10: 236-246, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-34901542

RESUMO

Scar contraction frequently happens in patients with deep burn injuries. Hitherto, porcine dermal extracellular matrix (dECM) has supplied microenvironments that assist in wound healing but fail to inhibit scar contraction. To overcome this drawback, we integrate dECM into three-dimensional (3D)-printed dermal analogues (PDA) to prevent scar contraction. We have developed thermally gelled, non-rheologically modified dECM powder (dECMp) inks and successfully transformed them into PDA that was endowed with a micron-scale spatial structure. The optimal crosslinked PDA exhibited desired structure, good mechanical properties as well as excellent biocompatibility. Moreover, in vivo experiments demonstrated that PDA could significantly reduced scar contraction and improved cosmetic upshots of split thickness skin grafts (STSG) than the commercially available dermal templates and STSG along. The PDA has also induced an early, intense neovascularization, and evoked a type-2-like immune response. PDA's superior beneficial effects may attribute to their desired porous structure, the well-balanced physicochemical properties, and the preserved dermis-specific ECM cues, which collectively modulated the expression of genes such as Wnt11, ATF3, and IL1ß, and influenced the crucial endogenous signalling pathways. The findings of this study suggest that PDA is a clinical translatable material that possess high potential in reducing scar contraction.

18.
J Invest Dermatol ; 141(6): 1573-1584, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33259831

RESUMO

The progression of diabetic complications does not halt despite the termination of hyperglycemia, suggesting a metabolic memory phenomenon. However, whether metabolic memory exists in and affects the healing of diabetic wounds, as well as the underlying molecular mechanisms, remain unclear. In this study, we found that wound healing was delayed, and angiogenesis was decreased in mice with diabetes despite the normalization of glycemic control. Thus, we hypothesized that transient hyperglycemic spikes may be a risk factor for diabetic wound healing. We showed that transient hyperglycemia caused persistent damage to the vascular endothelium. Transient hyperglycemia directly upregulated DNMT1 expression, leading to the hypermethylation of Ang-1 and reduced Ang-1 expression, which in turn induced long-lasting activation of NF-κB and subsequent endothelial dysfunction. An in vivo study further showed that inhibition of DNMT1 promoted angiogenesis and accelerated diabetic wound healing by regulating the Ang-1/NF-κB signaling pathway. These results highlight the dramatic and long-lasting effects of transient hyperglycemic spikes on wound healing and suggest that DNMT1 is a target for diabetic vascular complications.


Assuntos
Glicemia/metabolismo , DNA (Citosina-5-)-Metiltransferase 1/metabolismo , Diabetes Mellitus Experimental/complicações , Pé Diabético/patologia , Endotélio Vascular/patologia , Angiopoietina-1/genética , Angiopoietina-1/metabolismo , Animais , Glicemia/análise , Células Cultivadas , Metilação de DNA , Diabetes Mellitus Experimental/sangue , Diabetes Mellitus Experimental/induzido quimicamente , Diabetes Mellitus Experimental/metabolismo , Pé Diabético/sangue , Humanos , Camundongos , Neovascularização Fisiológica/genética , Transdução de Sinais/genética , Fator de Transcrição RelA/genética , Fator de Transcrição RelA/metabolismo , Cicatrização/genética
19.
Stem Cell Res Ther ; 11(1): 303, 2020 07 22.
Artigo em Inglês | MEDLINE | ID: mdl-32698863

RESUMO

The epidermis, which is the outermost layer of mammalian skin, provides an essential barrier that is essential for maintenance of life. The epidermis is a stratified epithelium, which is maintained by the proliferation of epidermal stem cells (EPSCs) at the basal layer of the epidermis. As a unique cell population characterized by self-renewal and differentiation capabilities, EPSCs ensure the maintenance of adult skin homeostasis and participate in repair of the epidermis after injury. Recently, the utilization of EPSCs for wound healing and tissue regeneration has been attracting increased attention from researchers. In addition, the advances in tissue engineering have increased the interest in applying EPSCs in tissue-engineered scaffolds to further reconstitute injured tissues. In this review, we introduce research developments related to EPSCs, including methods recently used in the culture and enrichment of EPSCs, as well as advanced tools to study EPSCs. The function and mechanism of the EPSC-dermal units in the development and homeostasis of the skin are also summarized. Finally, the potential applications of EPSCs in skin tissue engineering are discussed.


Assuntos
Células Epidérmicas , Engenharia Tecidual , Animais , Epiderme , Pele , Células-Tronco
20.
Cell Death Discov ; 6: 102, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33083022

RESUMO

Activation of the cannabinoid CB1 receptor induces neuroprotection against brain ischemia/reperfusion injury (IRI); however, the mechanism is still unknown. In this study, we used oxygen-glucose deprivation/reoxygenation (OGD/R)-induced injury in neuronal cells and middle cerebral artery occlusion (MCAO)-induced brain IRI in rats to mimic ischemic brain injury, and hypothesized that the CB1 receptor agonist arachidonyl-2-chloroethylamide (ACEA) would protect ischemic neurons by inhibiting mitochondrial fission via dynamin-related protein 1 (Drp1). We found that OGD/R injury reduced cell viability and mitochondrial function, increased lactate dehydrogenase (LDH) release, and increased cell apoptosis, and mitochondrial fission. Notably, ACEA significantly abolished the OGD/R-induced neuronal injuries described above. Similarly, ACEA significantly reversed MCAO-induced increases in brain infarct volume, neuronal apoptosis and mitochondrial fission, leading to the recovery of neurological functions. The neuroprotective effects of ACEA were obviously blocked by coadministration of the CB1 receptor antagonist AM251 or by the upregulation of Drp1 expression, indicating that ACEA alleviates brain IRI via the CB1-Drp1 pathway. Our findings suggest that the CB1 receptor links aberrant mitochondrial fission to brain IRI, providing a new therapeutic target for brain IRI treatment.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA