Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 50
Filtrar
1.
bioRxiv ; 2024 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-38915542

RESUMO

Angiogenesis plays a vital role for postnatal development and tissue repair following ischemia. Reactive oxygen species (ROS) generated by NADPH oxidases (NOXes) and mitochondria act as signaling molecules that promote angiogenesis in endothelial cells (ECs) which mainly relies on aerobic glycolysis for ATP production. However, the connections linking redox signaling with glycolysis are not well understood. The GTPase Drp1 is a member of the dynamin superfamily that moves from cytosol to mitochondria through posttranslational modifications to induce mitochondrial fission. The role of Drp1 in ROS-dependent VEGF signaling and angiogenesis in ECs has not been previously described. Here, we identify an unexpected function of endothelial Drp1 as a redox sensor, transmitting VEGF-induced H 2 O 2 signals to enhance glycolysis and angiogenesis. Loss of Drp1 expression in ECs inhibited VEGF-induced angiogenic responses. Mechanistically, VEGF rapidly induced the NOX4-dependent sulfenylation (CysOH) of Drp1 on Cys 644 , promoting disulfide bond formation with the metabolic kinase AMPK and subsequent sulfenylation of AMPK at Cys 299 / 304 via the mitochondrial fission-mitoROS axis. This cysteine oxidation of AMPK, in turn, enhanced glycolysis and angiogenesis. In vivo , mice with EC-specific Drp1 deficiency or CRISPR/Cas9-engineered "redox-dead" (Cys to Ala) Drp1 knock-in mutations exhibited impaired retinal angiogenesis and post-ischemic neovascularization. Our findings uncover a novel role for endothelial Drp1 in linking VEGF-induced mitochondrial redox signaling to glycolysis through a cysteine oxidation-mediated Drp1-AMPK redox relay, driving both developmental and reparative angiogenesis.

2.
J Allergy Clin Immunol ; 152(5): 1141-1152.e2, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37562753

RESUMO

BACKGROUND: Dendritic cells (DCs) are heterogeneous, comprising multiple subsets with unique functional specifications. Our previous work has demonstrated that the specific conventional type 2 DC subset, CSF1R+cDC2s, plays a critical role in sensing aeroallergens. OBJECTIVE: It remains to be understood how CSF1R+cDC2s recognize inhaled allergens. We sought to elucidate the transcriptomic programs and receptor-ligand interactions essential for function of this subset in allergen sensitization. METHODS: We applied single-cell RNA sequencing to mouse lung DCs. Conventional DC-selective knockout mouse models were employed, and mice were subjected to inhaled allergen sensitization with multiple readouts of asthma pathology. Under the clinical arm of this work, human lung transcriptomic data were integrated with mouse data, and bronchoalveolar lavage (BAL) specimens were collected from subjects undergoing allergen provocation, with samples assayed for C1q. RESULTS: We found that C1q is selectively enriched in lung CSF1R+cDC2s, but not in other lung cDC2 or cDC1 subsets. Depletion of C1q in conventional DCs significantly attenuates allergen sensing and features of asthma. Additionally, we found that C1q binds directly to human dust mite allergen, and the C1q receptor CD91 (LRP1) is required for lung CSF1R+cDC2s to recognize the C1q-allergen complex and induce allergic lung inflammation. Lastly, C1q is enriched in human BAL samples following subsegmental allergen challenge, and human RNA sequencing data demonstrate close homology between lung IGSF21+DCs and mouse CSF1R+cDC2s. CONCLUSIONS: C1q is secreted from the CSF1R+cDC2 subset among conventional DCs. Our data indicate that the C1q-LRP1 axis represents a candidate for translational therapeutics in the prevention and suppression of allergic lung inflammation.


Assuntos
Asma , Pneumonia , Animais , Humanos , Camundongos , Alérgenos/metabolismo , Asma/metabolismo , Complemento C1q/metabolismo , Células Dendríticas , Camundongos Knockout , Pneumonia/metabolismo , Receptores Proteína Tirosina Quinases , Receptores de Fator Estimulador de Colônias/metabolismo
3.
Am J Respir Crit Care Med ; 207(11): 1451-1463, 2023 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-36790376

RESUMO

Rationale: The resolution of inflammation is an active process coordinated by mediators and immune cells to restore tissue homeostasis. However, the mechanisms for resolving eosinophilic allergic lung inflammation triggered by inhaled allergens have not been fully elucidated. Objectives: Our objectives were to investigate the cellular mechanism of tissue-resident macrophages involved in the resolution process of eosinophilic lung inflammation. Methods: For the study, we used the institutional review board-approved protocol for human subsegmental bronchoprovocation with allergen, mouse models for allergic lung inflammation, and novel transgenic mice, including a conditional CCL26 knockout. The samples were analyzed using mass cytometry, single-cell RNA sequencing, and biophysical and immunological analyses. Measurements and Main Results: We compared alveolar macrophage (AM) subsets in the BAL before and after allergen provocation. In response to provocation with inhaled allergens, the subsets of AMs are dynamically changed in humans and mice. In the steady state, the AM subset expressing CX3CR1 is a relatively small fraction in bronchoalveolar space and lung tissue but drastically increases after allergen challenges. This subset presents unique patterns of gene expression compared with classical AMs, expressing high C1q family genes. CX3CR1+ macrophages are activated by airway epithelial cell-derived CCL26 via a receptor-ligand interaction. The binding of CCL26 to the CX3CR1+ receptor induces CX3CR1+ macrophages to secrete C1q, subsequently facilitating the clearance of eosinophils. Furthermore, the depletion of CX3CR1 macrophages or CCL26 in airway epithelial cells delays the resolution of allergic lung inflammation displaying prolonged tissue eosinophilia. Conclusions: These findings indicate that the CCL26-CX3CR1 pathway is pivotal in resolving eosinophilic allergic lung inflammation.


Assuntos
Alveolite Alérgica Extrínseca , Hipersensibilidade , Pneumonia , Eosinofilia Pulmonar , Humanos , Camundongos , Animais , Complemento C1q/metabolismo , Pulmão/metabolismo , Macrófagos , Alérgenos , Inflamação/metabolismo , Pneumonia/metabolismo , Quimiocina CCL26/metabolismo
4.
Angiogenesis ; 26(1): 77-96, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-35984546

RESUMO

VEGFR2 signaling in endothelial cells (ECs) is regulated by reactive oxygen species (ROS) derived from NADPH oxidases (NOXs) and mitochondria, which plays an important role in postnatal angiogenesis. However, it remains unclear how highly diffusible ROS signal enhances VEGFR2 signaling and reparative angiogenesis. Protein disulfide isomerase A1 (PDIA1) functions as an oxidoreductase depending on the redox environment. We hypothesized that PDIA1 functions as a redox sensor to enhance angiogenesis. Here we showed that PDIA1 co-immunoprecipitated with VEGFR2 or colocalized with either VEGFR2 or an early endosome marker Rab5 at the perinuclear region upon stimulation of human ECs with VEGF. PDIA1 silencing significantly reduced VEGF-induced EC migration, proliferation and spheroid sprouting via inhibiting VEGFR2 signaling. Mechanistically, VEGF stimulation rapidly increased Cys-OH formation of PDIA1 via the NOX4-mitochondrial ROS axis. Overexpression of "redox-dead" mutant PDIA1 with replacement of the active four Cys residues with Ser significantly inhibited VEGF-induced PDIA1-CysOH formation and angiogenic responses via reducing VEGFR2 phosphorylation. Pdia1+/- mice showed impaired angiogenesis in developmental retina and Matrigel plug models as well as ex vivo aortic ring sprouting model. Study using hindlimb ischemia model revealed that PDIA1 expression was markedly increased in angiogenic ECs of ischemic muscles, and that ischemia-induced limb perfusion recovery and neovascularization were impaired in EC-specific Pdia1 conditional knockout mice. These results suggest that PDIA1 can sense VEGF-induced H2O2 signal via CysOH formation to promote VEGFR2 signaling and angiogenesis in ECs, thereby enhancing postnatal angiogenesis. The oxidized PDIA1 is a potential therapeutic target for treatment of ischemic vascular diseases.


Assuntos
Células Endoteliais , Isomerases de Dissulfetos de Proteínas , Camundongos , Humanos , Animais , Células Endoteliais/metabolismo , Isomerases de Dissulfetos de Proteínas/genética , Isomerases de Dissulfetos de Proteínas/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Peróxido de Hidrogênio/metabolismo , Neovascularização Fisiológica , Oxirredução , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Isquemia/metabolismo
5.
Sci Rep ; 12(1): 16488, 2022 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-36182964

RESUMO

Blood-brain barrier (BBB) dysfunction is emerging as a key pathogenic factor in the progression of Alzheimer's disease (AD), where increased microvascular endothelial permeability has been proposed to play an important role. However, the molecular mechanisms leading to increased brain microvascular permeability in AD are not fully understood. We studied brain endothelial permeability in female APPswe/PS1∆E9 (APP/PS1) mice which constitute a transgenic mouse model of amyloid-beta (Aß) amyloidosis and found that permeability increases with aging in the areas showing the greatest amyloid plaque deposition. We performed an unbiased bulk RNA-sequencing analysis of brain endothelial cells (BECs) in female APP/PS1 transgenic mice. We observed that upregulation of interferon signaling gene expression pathways in BECs was among the most prominent transcriptomic signatures in the brain endothelium. Immunofluorescence analysis of isolated BECs from female APP/PS1 mice demonstrated higher levels of the Type I interferon-stimulated gene IFIT2. Immunoblotting of APP/PS1 BECs showed downregulation of the adherens junction protein VE-cadherin. Stimulation of human brain endothelial cells with interferon-ß decreased the levels of the adherens junction protein VE-cadherin as well as tight junction proteins Occludin and Claudin-5 and increased barrier leakiness. Depletion of the Type I interferon receptor in human brain endothelial cells prevented interferon-ß-induced VE-cadherin downregulation and restored endothelial barrier integrity. Our study suggests that Type I interferon signaling contributes to brain endothelial dysfunction in AD.


Assuntos
Doença de Alzheimer , Interferon Tipo I , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Animais , Barreira Hematoencefálica/metabolismo , Encéfalo/metabolismo , Claudina-5/metabolismo , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Endotélio/metabolismo , Feminino , Humanos , Interferon Tipo I/metabolismo , Interferon beta/metabolismo , Camundongos , Camundongos Transgênicos , Ocludina/metabolismo , Placa Amiloide/patologia , RNA/metabolismo , Receptor de Interferon alfa e beta/metabolismo , Proteínas de Junções Íntimas/metabolismo
6.
Redox Biol ; 52: 102304, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35413643

RESUMO

As essential regulators of mitochondrial quality control, mitochondrial dynamics and mitophagy play key roles in maintenance of metabolic health and cellular homeostasis. Here we show that knockdown of the membrane-inserted scaffolding and structural protein caveolin-1 (Cav-1) and expression of tyrosine 14 phospho-defective Cav-1 mutant (Y14F), as opposed to phospho-mimicking Y14D, altered mitochondrial morphology, and increased mitochondrial matrix mixing, mitochondrial fusion and fission dynamics as well as mitophagy in MDA-MB-231 triple negative breast cancer cells. Further, we found that interaction of Cav-1 with mitochondrial fusion/fission machinery Mitofusin 2 (Mfn2) and Dynamin related protein 1 (Drp1) was enhanced by Y14D mutant indicating Cav-1 Y14 phosphorylation prevented Mfn2 and Drp1 translocation to mitochondria. Moreover, limiting mitochondrial recruitment of Mfn2 diminished formation of the PINK1/Mfn2/Parkin complex required for initiation of mitophagy resulting in accumulation of damaged mitochondria and ROS (mtROS). Thus, these studies indicate that phospho-Cav-1 may be an important switch mechanism in cancer cell survival which could lead to novel strategies for complementing cancer therapies.


Assuntos
Caveolina 1 , Mitofagia , Caveolina 1/genética , Caveolina 1/metabolismo , Mitocôndrias/metabolismo , Dinâmica Mitocondrial/fisiologia , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Mitofagia/fisiologia , Espécies Reativas de Oxigênio/metabolismo
7.
Nat Cell Biol ; 24(1): 35-50, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-35027734

RESUMO

Vascular endothelial growth factor receptor type 2 (VEGFR2, also known as KDR and FLK1) signalling in endothelial cells (ECs) is essential for developmental and reparative angiogenesis. Reactive oxygen species and copper (Cu) are also involved in these processes. However, their inter-relationship is poorly understood. Evidence of the role of the endothelial Cu importer CTR1 (also known as SLC31A1) in VEGFR2 signalling and angiogenesis in vivo is lacking. Here, we show that CTR1 functions as a redox sensor to promote angiogenesis in ECs. CTR1-depleted ECs showed reduced VEGF-induced VEGFR2 signalling and angiogenic responses. Mechanistically, CTR1 was rapidly sulfenylated at Cys189 at its cytosolic C terminus after stimulation with VEGF, which induced CTR1-VEGFR2 disulfide bond formation and their co-internalization to early endosomes, driving sustained VEGFR2 signalling. In vivo, EC-specific Ctr1-deficient mice or CRISPR-Cas9-generated redox-dead Ctr1(C187A)-knockin mutant mice had impaired developmental and reparative angiogenesis. Thus, oxidation of CTR1 at Cys189 promotes VEGFR2 internalization and signalling to enhance angiogenesis. Our study uncovers an important mechanism for sensing reactive oxygen species through CTR1 to drive neovascularization.


Assuntos
Transportador de Cobre 1/metabolismo , Cobre/metabolismo , Neovascularização Fisiológica/fisiologia , Espécies Reativas de Oxigênio/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Animais , Bovinos , Linhagem Celular , Transportador de Cobre 1/genética , Cisteína/metabolismo , Feminino , Células HEK293 , Células Endoteliais da Veia Umbilical Humana , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Oxirredução , Transdução de Sinais/fisiologia
8.
Neurol Genet ; 7(6): e639, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34881353

RESUMO

OBJECTIVE: This study aimed to determine the frequency of pathogenic NOTCH3 variants among Koreans. METHODS: In this cross-sectional study, we queried for pathogenic NOTCH3 variants in 2 Korean public genome databases: the Korean Reference Genome Database (KRGDB) and the Korean Genome Project (Korea1K). In addition, we screened the 3 most common pathogenic NOTCH3 variants (p.Arg75Pro, p.Arg544Cys, and p.Arg578Cys) for 1,000 individuals on Jeju Island, where the largest number of patients with cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) have been reported in Korea. RESULTS: The pathogenic NOTCH3 variant (p.Arg544Cys) was found in 0.12% of sequences in the KRGDB, and 3 pathogenic variants (p.Arg75Pro, p.Arg182Cys, and p.Arg544Cys) were present in 0.44% of the Korea1K database. Of the 1,000 individuals on Jeju Island, we found 2 cysteine-altering NOTCH3 variants (p.Arg544Cys variant in 9 and p.Arg578Cys in 1 individual) in 1.00% of the participants (95% confidence interval: 0.48%-1.83%). The presence of cysteine-altering NOTCH3 variants was significantly associated with a history of stroke (p < 0.001). DISCUSSION: Pathogenic NOTCH3 variants are frequently found in the general Korean population. Such a high prevalence of pathogenic variants could threaten the brain health of tens of thousands to hundreds of thousands of older adults in Korea.

9.
Cells ; 9(6)2020 06 26.
Artigo em Inglês | MEDLINE | ID: mdl-32604782

RESUMO

Epithelial-to-mesenchymal transition (EMT) promulgates epithelial cell associated disease-defining characteristics in tumorigenesis and organ fibrosis. Growth factors such as epidermal growth factor and fibroblast growth factor in addition to cytokines such as transforming growth factor-ß1 (TGF-ß1) is said to play a prominent role in remodeling related pathological events of cancer progression such as invasion, metastasis, apoptosis, EMT, etc. through redox related cellular secondary messengers, in particular the reactive oxygen species (ROS). However, the signaling cascade underlying the redox mechanism and thereby the progression of EMT remains largely unknown. In this study, upon TGF-ß1 treatment, we observed an induction in NOX isoforms-NOX2 and NOX4-that have time (early and late) and cellular localization (nucleus and autophagosome co-localized) dependent effects in mediating EMT associated cell proliferation and migration through activation of the focal adhesion kinase (FAK)/SRC pathway in HeLa, human cervical cancer cells. Upon silencing NOX2/4 gene expression and using the SRC inhibitor (AZD0530), progression of TGF-ß1 induced EMT related cellular remodeling, extra cellular matrix (ECM) production, cell migration and invasion, got significantly reverted. Together, these results indicate that NOX2 and NOX4 play important, albeit distinct, roles in the activation of cytokine mediated EMT and its associated processes via tyrosine phosphorylation of the FAK/SRC pathway.


Assuntos
Quinase 1 de Adesão Focal/genética , NADPH Oxidases/metabolismo , Isoformas de Proteínas/metabolismo , Neoplasias do Colo do Útero/genética , Células Epiteliais , Transição Epitelial-Mesenquimal , Feminino , Células HeLa , Humanos , Oxirredução , Fosforilação , Transfecção
10.
J Cancer ; 11(15): 4495-4509, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32489467

RESUMO

Objective: We propose that sirtuin (SIRT) may induce a pro-apoptotic effect by deacetylating transcription factors in A549 cells: depletion of sirtuin-1 (SIRT1) induced cell cycle arrest in cisplatin-resistant A549 (A549/CADD) cells. Methods: Protein and mRNA levels of SIRT1 were investigated using western blot and RT-PCR. In A549 and A549/CADD cells, the cytotoxicity of cisplatin administration was evaluated by MTT assay, proliferation was measured by ECIS, and the cell cycle distribution was analyzed using FACS. Cells were transfected with pcDNA3.1-Myc-SIRT1 or pcDNA3.1-Myc-Control vectors to analyze the impact of SIRT-1 on cisplatin induced drug resistance. SIRT1 localization was studied using immunofluorescence analysis. In addition, immunoprecipitation and 20S proteasome activity assay were performed to examine the relationship of SIRT1 with the proteasome complex. Results: A549/CADD cells exhibited a mesenchymal-like cell characteristic. SIRT1 expression was markedly decreased in A549/CADD cells. We observed that cisplatin regulates p53 stability through the depletion of ubiquitination following SIRT1 downregulation. Furthermore, cisplatin treatment increased proteasomal activity and significantly decreased cytoplasmic SIRT1 protein levels in A549/CADD cells. Conclusion: In this study, we found SIRT1 to be depleted in A549/CADD cells and also determined the underlying resistance mechanism which may act as novel therapeutic targets in overcoming drug resistance.

11.
Adv Wound Care (New Rochelle) ; 8(4): 125-135, 2019 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-31737411

RESUMO

Objective: Recognized as pathogen-associated molecular patterns (PAMPs), ß-glucans, a naturally occurring heterogeneous group of polysaccharides, were investigated for their ability to accelerate wound healing in the form of high water-retaining hydrogel dressing. Approach: Full-thickness wounds on the dorsal side of mice created using a 5-mm biopsy punch were treated with ß-glucan-based hydrogel for 2 weeks. Standardized photographs of the wound site were taken at regular time intervals to calculate the percentage of wound closure. Tissues isolated from the wound area were subjected to histological examination and immunoblot analysis. Results: ß-Glucan-based hydrogel significantly accelerated the duration of wound healing and enhanced the development of skin appendages in the regenerated skin tissue. Increased expression of transforming growth factor-ß3 in the skin tissue isolated from the healed wound site indicated that skin regeneration rather than skin repair occurred, thereby minimizing cutaneous scarring. The expression level of cytokeratin 10 and cytokeratin 14 in the isolated skin tissue revealed that the wounds treated with hydrogel showed proper differentiation and proliferation of keratinocytes in the epidermal layer. Innovation: Immunomodulating ß-glucan (responsible for fighting infections at the wound site, and enhancing the migration and proliferation of keratinocytes and fibroblasts) in the form of a three-dimensional hydrogel membrane that retains a high water content (responsible for cooling and soothing effect around the wound site, thereby reducing pain) was prepared and analyzed for its effects on the cutaneous wound healing mechanism. Conclusion: ß-Glucan-based hydrogels are promising as wet wound dressings in the health care industry.

12.
Cell Rep ; 23(12): 3565-3578, 2018 06 19.
Artigo em Inglês | MEDLINE | ID: mdl-29924999

RESUMO

Mitochondrial dynamics are tightly controlled by fusion and fission, and their dysregulation and excess reactive oxygen species (ROS) contribute to endothelial cell (EC) dysfunction. How redox signals regulate coupling between mitochondrial dynamics and endothelial (dys)function remains unknown. Here, we identify protein disulfide isomerase A1 (PDIA1) as a thiol reductase for the mitochondrial fission protein Drp1. A biotin-labeled Cys-OH trapping probe and rescue experiments reveal that PDIA1 depletion in ECs induces sulfenylation of Drp1 at Cys644, promoting mitochondrial fragmentation and ROS elevation without inducing ER stress, which drives EC senescence. Mechanistically, PDIA1 associates with Drp1 to reduce its redox status and activity. Defective wound healing and angiogenesis in diabetic or PDIA1+/- mice are restored by EC-targeted PDIA1 or the Cys oxidation-defective mutant Drp1. Thus, this study uncovers a molecular link between PDIA1 and Drp1 oxidoreduction, which maintains normal mitochondrial dynamics and limits endothelial senescence with potential translational implications for vascular diseases associated with diabetes or aging.


Assuntos
Senescência Celular , Dinaminas/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Dinâmica Mitocondrial , Pró-Colágeno-Prolina Dioxigenase/metabolismo , Isomerases de Dissulfetos de Proteínas/metabolismo , Animais , Respiração Celular , Cisteína/metabolismo , Diabetes Mellitus Tipo 2/patologia , Estresse do Retículo Endoplasmático , Humanos , Camundongos , Mitocôndrias/metabolismo , Mutação/genética , Oxirredução , Ligação Proteica , Espécies Reativas de Oxigênio/metabolismo , Cicatrização
13.
Int J Mol Sci ; 19(4)2018 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-29641498

RESUMO

Cutaneous wound repair is an intricate process whereby the skin reprograms itself after injury. In the mid-phase of wound repair, the proliferation, migration, and differentiation of cells are the major mechanisms to lead remodeling. We investigated the effect of BMM ((1E,2E)-1,2-bis((6-bromo-2H-chromen-3-yl)methylene)hydrazine), a novel synthetic material, on the migration and viability of keratinocytes or fibroblasts using the in vitro scratch woundhealing, electric cell-substrate imedance sensing (ECIS), invasion, and MTT assays. Cell migration-related factors were analyzed using western blot, and we found that treatment with BMM stimulated the EMT pathway and focal adhesion kinase (FAK)/Src signaling. Differentiation of HaCaT keratinocyte and fibroblast cells was also stimulated by BMM and specifically, NOX2/4 contributed to the activation of fibroblasts for wound healing. Furthermore, BMM treated HaCaT keratinocyte and fibroblast-co-cultured cells increased migration and differentiation. TGF-ß and Cyr61 were also secreted to a greater extent than in single cultured cells. In vivo experiments showed that treatment with BMM promotes wound closure by promoting re-epithelialization. In this study, we demonstrated that a novel synthetic material, BMM, is capable of promoting wound healing via the stimulation of re-epithelialization in the epidermis and the activation of fibroblasts in the dermis, in particular, via the acceleration of the interaction between the epidermis and dermis.


Assuntos
Benzopiranos/farmacologia , Fibroblastos/efeitos dos fármacos , Hidrazinas/farmacologia , Reepitelização , Animais , Benzopiranos/química , Linhagem Celular , Movimento Celular , Proliferação de Células , Células Cultivadas , Proteína Rica em Cisteína 61/metabolismo , Fibroblastos/metabolismo , Fibroblastos/fisiologia , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Humanos , Hidrazinas/química , Queratinócitos/efeitos dos fármacos , Queratinócitos/metabolismo , Queratinócitos/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos ICR , NADPH Oxidases/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo , Quinases da Família src/metabolismo
14.
Am J Physiol Cell Physiol ; 312(6): C749-C764, 2017 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-28424170

RESUMO

Reactive oxygen species (ROS) derived from NADPH oxidase (NOX) and mitochondria play a critical role in growth factor-induced switch from a quiescent to an angiogenic phenotype in endothelial cells (ECs). However, how highly diffusible ROS produced from different sources can coordinate to stimulate VEGF signaling and drive the angiogenic process remains unknown. Using the cytosol- and mitochondria-targeted redox-sensitive RoGFP biosensors with real-time imaging, here we show that VEGF stimulation in human ECs rapidly increases cytosolic RoGFP oxidation within 1 min, followed by mitochondrial RoGFP oxidation within 5 min, which continues at least for 60 min. Silencing of Nox4 or Nox2 or overexpression of mitochondria-targeted catalase significantly inhibits VEGF-induced tyrosine phosphorylation of VEGF receptor type 2 (VEGFR2-pY), EC migration and proliferation at the similar extent. Exogenous hydrogen peroxide (H2O2) or overexpression of Nox4, which produces H2O2, increases mitochondrial ROS (mtROS), which is prevented by Nox2 siRNA, suggesting that Nox2 senses Nox4-derived H2O2 to promote mtROS production. Mechanistically, H2O2 increases S36 phosphorylation of p66Shc, a key mtROS regulator, which is inhibited by siNox2, but not by siNox4. Moreover, Nox2 or Nox4 knockdown or overexpression of S36 phosphorylation-defective mutant p66Shc(S36A) inhibits VEGF-induced mtROS, VEGFR2-pY, EC migration, and proliferation. In summary, Nox4-derived H2O2 in part activates Nox2 to increase mtROS via pSer36-p66Shc, thereby enhancing VEGFR2 signaling and angiogenesis in ECs. This may represent a novel feed-forward mechanism of ROS-induced ROS release orchestrated by the Nox4/Nox2/pSer36-p66Shc/mtROS axis, which drives sustained activation of angiogenesis signaling program.


Assuntos
Retroalimentação Fisiológica , Peróxido de Hidrogênio/metabolismo , Glicoproteínas de Membrana/genética , Mitocôndrias/metabolismo , NADPH Oxidases/genética , Transdução de Sinais , Técnicas Biossensoriais , Catalase/genética , Catalase/metabolismo , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Regulação da Expressão Gênica , Células Endoteliais da Veia Umbilical Humana , Humanos , Glicoproteínas de Membrana/antagonistas & inibidores , Glicoproteínas de Membrana/metabolismo , Microscopia de Fluorescência , Mitocôndrias/efeitos dos fármacos , NADPH Oxidase 2 , NADPH Oxidase 4 , NADPH Oxidases/antagonistas & inibidores , NADPH Oxidases/metabolismo , Neovascularização Fisiológica/efeitos dos fármacos , Neovascularização Fisiológica/genética , Oxirredução , Fosforilação/efeitos dos fármacos , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Proteína 1 de Transformação que Contém Domínio 2 de Homologia de Src/genética , Proteína 1 de Transformação que Contém Domínio 2 de Homologia de Src/metabolismo , Imagem com Lapso de Tempo , Fator A de Crescimento do Endotélio Vascular/farmacologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
15.
Exp Mol Med ; 49(3): e302, 2017 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-28303029

RESUMO

Keratinocyte-fibroblast interactions are critical for skin repair after injury. During the proliferative phase of wound healing, proliferation, migration and differentiation of these cells are the major mechanisms leading to tissue remodeling. We have previously reported that glycitin, a major soy isoflavone, stimulates dermal fibroblast proliferation; and the phytochemical, 4',6,7-trimethoxyisoflavone (TMF), induces migration of HaCaT keratinocyte cells. We therefore investigated whether these compounds display synergistic effects on skin cells during wound healing in vitro and in vivo. Co-treatment with TMF and glycitin synergistically promotes the proliferation and migration of both keratinocytes and dermal fibroblasts, with a 1:1 ratio of these compounds showing the greatest efficacy in our co-culture system. This keratinocyte-fibroblast interaction occurred via the secretion of TGF-ß, and the induction of differentiation and proliferation was confirmed in both indirect and direct co-culture assays. In an excisional and burn wound animal model, mice treated with a 1:1 ratio of TMF and glycitin showed faster wound closure, regeneration and scar reduction than even the positive control drug. These data indicate that two isoflavones, TMF and glycitin, act synergistically to promote wound healing and anti-scarring and could potentially be developed together as a bioactive therapeutic for wound treatment.


Assuntos
Queimaduras/tratamento farmacológico , Cicatriz/tratamento farmacológico , Fibroblastos/efeitos dos fármacos , Isoflavonas/farmacologia , Queratinócitos/efeitos dos fármacos , Animais , Movimento Celular , Proliferação de Células , Células Cultivadas , Feminino , Fibroblastos/metabolismo , Humanos , Isoflavonas/uso terapêutico , Queratinócitos/metabolismo , Linfotoxina-alfa/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR
16.
Cancer Res Treat ; 48(3): 1141-53, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26693911

RESUMO

PURPOSE: One of the features in cancer development is the migration of cancer cells to form metastatic lesions. CYR61 protein promotes migration and the epithelial-mesenchymal transition in several cancer cell types. Evidence suggests that CYR61 and dexamethasone are relevant to colorectal cancer. However, relationships between them and colorectal cancer are still unclear. Understanding the molecular mechanism of colorectal cancer progression related with CYR61 and dexamethasone, which is widely used for combination chemotherapy, is necessary for improved therapy. MATERIALS AND METHODS: We used colorectal cancer cells, HCT116, co-treated with transforming growth factor ß1 (TGF-ß1) and dexamethasone to examine the inhibitory migration effect of dexamethasone by migratory assay. Alternatively, both migratory pathways, expression of AKT and ERK, and the target factor CYR61 was also tested by co-treatment with TGF-ß1 and dexamethasone. RESULTS: We report that dexamethasone significantly inhibited TGF-ß1-induced cell migration, without affecting cell proliferation. Importantly, we observed that TGF-ß1 promoted the epithelial-mesenchymal transition process and that dexamethasone co-treatment abolished this effect. ERK and AKT signaling pathways were found to mediate TGF-ß1-induced migration, which was inhibited by dexamethasone. In addition, TGF-ß1 treatment induced CYR61 expression whereas dexamethasone reduced it. These observations were compatible with the modulation of migration observed following treatment of HCT116 cells with human recombinant CYR61 and anti-CYR61 antibody. Our results also indicated that TGF-ß1 enhanced collagen I and reduced matrix metalloproteinase 1 expression, which was reversed by dexamethasone treatment. CONCLUSION: These findings suggested that dexamethasone inhibits AKT and ERK phosphorylation, leading to decreased CYR61 expression, which in turn blocks TGF-ß1-induced migration.


Assuntos
Antineoplásicos Hormonais/farmacologia , Movimento Celular/efeitos dos fármacos , Neoplasias Colorretais/patologia , Proteína Rica em Cisteína 61/metabolismo , Dexametasona/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Fator de Crescimento Transformador beta1/metabolismo , Anticorpos/farmacologia , Caderinas , Proliferação de Células/efeitos dos fármacos , Colágeno Tipo I/metabolismo , Proteína Rica em Cisteína 61/antagonistas & inibidores , Progressão da Doença , Regulação para Baixo , Transição Epitelial-Mesenquimal/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Células HCT116 , Humanos , Metaloproteinase 1 da Matriz/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Recombinantes/metabolismo , Regulação para Cima
17.
PLoS One ; 9(6): e99654, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24959670

RESUMO

Ubiquitin C-terminal hydrolase-L1 (UCH-L1) has been proposed as one of the Parkinson's disease (PD) related genes, but the possible molecular connection between UCH-L1 and PD is not well understood. In this study, we discovered an N-terminal 11 amino acid truncated variant UCH-L1 that we called NT-UCH-L1, in mouse brain tissue as well as in NCI-H157 lung cancer and SH-SY5Y neuroblastoma cell lines. In vivo experiments and hydrogen-deuterium exchange (HDX) with tandem mass spectrometry (MS) studies showed that NT-UCH-L1 is readily aggregated and degraded, and has more flexible structure than UCH-L1. Post-translational modifications including monoubiquitination and disulfide crosslinking regulate the stability and cellular localization of NT-UCH-L1, as confirmed by mutational and proteomic studies. Stable expression of NT-UCH-L1 decreases cellular ROS levels and protects cells from H2O2, rotenone and CCCP-induced cell death. NT-UCH-L1-expressing transgenic mice are less susceptible to degeneration of nigrostriatal dopaminergic neurons seen in the MPTP mouse model of PD, in comparison to control animals. These results suggest that NT-UCH-L1 may have the potential to prevent neural damage in diseases like PD.


Assuntos
Encéfalo/enzimologia , Mitocôndrias/metabolismo , Doença de Parkinson/genética , Doença de Parkinson/patologia , Ubiquitina Tiolesterase/genética , Ubiquitina Tiolesterase/metabolismo , Animais , Encéfalo/patologia , Linhagem Celular Tumoral , Medição da Troca de Deutério , Modelos Animais de Doenças , Estabilidade Enzimática , Células HeLa , Humanos , Peróxido de Hidrogênio/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Processamento de Proteína Pós-Traducional , Ubiquitina Tiolesterase/química
18.
Cell Signal ; 26(4): 784-96, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24378533

RESUMO

The epithelial-mesenchymal transition (EMT) is a critical biological process characterized by morphological and behavioral changes in cells. The regulatory and signaling mechanisms of both developmental and pathological EMT have been investigated. Reactive oxygen species (ROS) play a role in early EMT, but the exact mechanism by which ROS are involved is unclear. We investigated ROS-mediated EMT in human HeLa cells. Transforming growth factor beta (TGF-ß) treatments lead to dramatic NADPH oxidase 2 (NOX2) inductions in HeLa cells; antioxidant treatment prevented TGF-ß-driven EMT. Over-expression of the p40phox subunit (NCF4) led to activation of the NOX2 complex and ROS production. We showed that NOX2 and NOX5 mRNA was increased, along with increased expression of several matrix metalloproteinases (MMPs) in response to NCF4 expression. Moreover, these changes were reversible upon ROS scavenging. Down-regulation of E-cadherin and up-regulation of Snail, Slug and vimentin occurred at the transcriptional level. We also showed that new EMT regulator, YB-1 is a downstream target in ROS-induced EMT. Together, these data suggest that ROS switching is necessary for increased EMT but is not required for the morphological changes that accompany EMT.


Assuntos
Transição Epitelial-Mesenquimal , NADPH Oxidases/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Antioxidantes/farmacologia , Caderinas/metabolismo , Regulação para Baixo , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Células HeLa , Humanos , Metaloproteinases da Matriz/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , NADPH Oxidase 2 , NADPH Oxidase 5 , NADPH Oxidases/química , NADPH Oxidases/genética , RNA Mensageiro/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fatores de Transcrição da Família Snail , Fatores de Transcrição/metabolismo , Fator de Crescimento Transformador beta1/farmacologia , Regulação para Cima , Vimentina/metabolismo , Proteína 1 de Ligação a Y-Box/metabolismo
19.
Phytomedicine ; 21(4): 570-7, 2014 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-24388604

RESUMO

Flavonoids are major active ingredients in plants and are considered components of food that provide medical or health benefits. They have diversified structures and have effects on human health, including wound healing induction. More than a hundred flavonoids were screened for HaCaT keratinocytes cellular migration measurements and the relationships between their structural properties and the effects promoting cellular migration were examined. Here, among flavonoids used in the previous structure-activity relationship calculations, 4',6,7-trimethoxyisoflavone (TMF) was one of the compounds showing the best activity, so that its molecular mechanism of the wound healing effect on HaCaT keratinocytes was investigated in more detail. Our data revealed that TMF increased the wound healing rate, but not the proliferation rate, in a dose-dependent manner. Treatment of keratinocytes with TMF influenced signaling pathways, affecting the phosphorylation of AKT and ERK in a time-dependent manner. TMF also induced the cell-cell adhesion protein E-cadherin, which is essential for promoting collective cell migration. Furthermore, the TMF treatment group also showed higher ROS and NOX2 transcriptional and protein levels. Correlating with matrix metalloproteinase induction by TMF, levels of extracellular matrix proteins such as collagens I and III were significantly lower in the treatment group. To confirm that the effects of TMF occur through the NOX2 pathway, we co-treated cells with TMF plus an NADPH inhibitor (DPI) or a ROS scavenger (NAC). Western blotting revealed that DPI and NAC attenuated the effect of TMF, suggesting that TMF induces ROS through the NOX2 pathway and regulates keratinocyte migration. In summary, TMF promotes wound healing through NOX2 induction, which leads to collective migration and MMP activation.


Assuntos
Movimento Celular/efeitos dos fármacos , Isoflavonas/farmacologia , Queratinócitos/efeitos dos fármacos , Glicoproteínas de Membrana/metabolismo , NADPH Oxidases/metabolismo , Cicatrização/efeitos dos fármacos , Biomarcadores/metabolismo , Avaliação Pré-Clínica de Medicamentos , Ativação Enzimática/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Metaloproteinases da Matriz/metabolismo , NADPH Oxidase 2 , Proteínas Proto-Oncogênicas c-akt/metabolismo , Espécies Reativas de Oxigênio/metabolismo
20.
Biomol NMR Assign ; 8(1): 165-8, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23595857

RESUMO

The KCNH channels are voltage-gated potassium channels that play important roles in heart and nerve cells. The N-terminal region of the KCNH channel contains a Per-Arnt-Sim (PAS) domain which is important for the channel gating through interaction with other regions of the channel. To study the solution structure of the N-terminal PAS domain of the KCNH channel from Zebrafish (zNTD), we over-expressed and purified zNTD. We report the resonance assignments for zNTD. The data will allow us to perform structural studies for this domain, which will provide insight into its structural basis for the molecular interaction with other regions of the KCNH channel.


Assuntos
Canais de Potássio Éter-A-Go-Go/química , Ressonância Magnética Nuclear Biomolecular , Proteínas de Peixe-Zebra/química , Peixe-Zebra/metabolismo , Sequência de Aminoácidos , Animais , Isótopos de Carbono , Hidrogênio , Dados de Sequência Molecular , Isótopos de Nitrogênio , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA