Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
1.
Int J Mol Sci ; 25(8)2024 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-38674031

RESUMO

Hemangioblasts give rise to endothelial progenitor cells (EPCs), which also express the cell surface markers CD133 and c-kit. They may differentiate into the outgrowth endothelial cells (OECs) that control neovascularization in the developing embryo. According to numerous studies, reduced levels of EPCs in circulation have been linked to human cardiovascular disorders. Furthermore, preeclampsia and senescence have been linked to levels of EPCs produced from cord blood. Uncertainties surround how preeclampsia affects the way EPCs function. It is reasonable to speculate that preeclampsia may have an impact on the function of fetal EPCs during the in utero period; however, the present literature suggests that maternal vasculopathies, including preeclampsia, damage fetal circulation. Additionally, the differentiation potential and general activity of EPCs may serve as an indicator of the health of the fetal vascular system as they promote neovascularization and repair during pregnancy. Thus, the purpose of this review is to compare-through the assessment of their quantity, differentiation potency, angiogenic activity, and senescence-the angiogenic function of fetal EPCs obtained from cord blood for normal and pregnancy problems (preeclampsia, gestational diabetes mellitus, and fetal growth restriction). This will shed light on the relationship between the angiogenic function of fetal EPCs and pregnancy complications, which could have an effect on the management of long-term health issues like metabolic and cardiovascular disorders in offspring with abnormal vasculature development.


Assuntos
Diabetes Gestacional , Células Progenitoras Endoteliais , Sangue Fetal , Retardo do Crescimento Fetal , Pré-Eclâmpsia , Humanos , Gravidez , Feminino , Diabetes Gestacional/metabolismo , Diabetes Gestacional/sangue , Pré-Eclâmpsia/sangue , Células Progenitoras Endoteliais/metabolismo , Sangue Fetal/citologia , Sangue Fetal/metabolismo , Retardo do Crescimento Fetal/patologia , Diferenciação Celular
2.
Int J Mol Sci ; 24(22)2023 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-38003275

RESUMO

Maternal hyperglycemia, induced by gestational diabetes mellitus (GDM), has detrimental effects on fetal vascular development, ultimately increasing the risk of cardiovascular diseases in offspring. The potential underlying mechanisms through which these complications occur are due to functional impairment and epigenetic changes in fetal endothelial progenitor cells (EPCs), which remain less defined. We confirm that intrauterine hyperglycemia leads to the impaired angiogenic function of fetal EPCs, as observed through functional assays of outgrowth endothelial cells (OECs) derived from fetal EPCs of GDM pregnancies (GDM-EPCs). Notably, PCDH10 expression is increased in OECs derived from GDM-EPCs, which is associated with the inhibition of angiogenic function in fetal EPCs. Additionally, increased PCDH10 expression is correlated with the hypomethylation of the PCDH10 promoter. Our findings demonstrate that in utero exposure to GDM can induce angiogenic dysfunction in fetal EPCs through altered gene expression and epigenetic changes, consequently increasing the susceptibility to cardiovascular diseases in the offspring of GDM mothers.


Assuntos
Doenças Cardiovasculares , Diabetes Gestacional , Células Progenitoras Endoteliais , Hiperglicemia , Gravidez , Feminino , Humanos , Diabetes Gestacional/metabolismo , Células Progenitoras Endoteliais/metabolismo , Feto/metabolismo , Hiperglicemia/metabolismo , Protocaderinas
3.
Am J Reprod Immunol ; 90(2): e13744, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37491916

RESUMO

PROBLEM: Direct interactions between macrophages and lymphatic vessels have been shown previously. In pre-eclampsia (PE), macrophages are dominantly polarized into a proinflammatory M1 phenotype and lymphangiogenesis is defective in the decidua. Here, we investigated whether decidual lymphatic endothelial cells (dLECs) affect macrophage polarization in PE. METHOD OF STUDY: THP-1 macrophages were cocultured with dLECs or cultured in the conditioned medium (CM) of dLECs. Macrophage polarization was measured using flow cytometry. Granulocyte-macrophage colony-stimulating factor (GM-CSF) expression in dLECs was measured using qRT-PCR and ELISA. The activation of nuclear translocation of nuclear factor-κ (NF-κB), an upstream signaling molecule of GM-CSF, was assessed by immunocytochemical localization of p65. Through GM-CSF knockdown and NF-κB inhibition in dLEC, we evaluated whether the GM-CSF/NF-κB pathway of PE dLEC affects decidual macrophage polarization. RESULTS: The ratio of inflammatory M1 macrophages with HLA-DR+ /CD80+ markers significantly increased following coculturing with PE dLECs or culturing in PE dLEC CM, indicating that the PE dLEC-derived soluble factor acts in a paracrine manner. GM-CSF expression was significantly upregulated in PE dLECs. Recombinant human GM-CSF induced macrophage polarization toward an M1-like phenotype, whereas its knockdown in PE dLECs suppressed it, suggesting PE dLECs induce M1 macrophage polarization by secreting GM-CSF. The NF-κB p65 significantly increased in PE dLECs compared to the control, and pretreatment with an NF-κB inhibitor significantly suppressed GM-CSF production from PE dLECs. CONCLUSIONS: In PE, dLECs expressing high levels of GM-CSF via the NF-κB-dependent pathway play a role in inducing decidual M1 macrophage polarization.


Assuntos
NF-kappa B , Pré-Eclâmpsia , Gravidez , Feminino , Humanos , NF-kappa B/metabolismo , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Pré-Eclâmpsia/metabolismo , Células Endoteliais/metabolismo , Macrófagos/metabolismo , Fator Estimulador de Colônias de Macrófagos
4.
Int J Mol Sci ; 22(8)2021 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-33921847

RESUMO

The abnormal development or disruption of the lymphatic vasculature has been implicated in metabolic and hypertensive diseases. Recent evidence suggests that the offspring exposed to preeclampsia (PE) in utero are at higher risk of long-term health problems, such as cardiovascular and metabolic diseases in adulthood, owing to in utero fetal programming. We aimed to investigate lymphangiogenic activities in the lymphatic endothelial progenitor cells (LEPCs) of the offspring of PE. Human umbilical cord blood LEPCs from pregnant women with severe PE (n = 10) and gestationally matched normal pregnancies (n = 10) were purified with anti-vascular endothelial growth factor receptor 3 (VEGFR3)/podoplanin/CD11b microbeads using a magnetic cell sorter device. LEPCs from PE displayed significantly delayed differentiation and reduced formation of lymphatic endothelial cell (LEC) colonies compared with the LEPCs from normal pregnancies. LECs differentiated from PE-derived LEPCs exhibited decreased tube formation, migration, proliferation, adhesion, wound healing, and 3D-sprouting activities as well as increased lymphatic permeability through the disorganization of VE-cadherin junctions, compared with the normal pregnancy-derived LECs. In vivo, LEPCs from PE showed significantly reduced lymphatic vessel formation compared to the LEPCs of the normal pregnancy. Gene expression analysis revealed that compared to the normal pregnancy-derived LECs, the PE-derived LECs showed a significant decrease in the expression of pro-lymphangiogenic genes (GREM1, EPHB3, VEGFA, AMOT, THSD7A, ANGPTL4, SEMA5A, FGF2, and GBX2). Collectively, our findings demonstrate, for the first time, that LEPCs from PE have reduced lymphangiogenic activities in vitro and in vivo and show the decreased expression of pro-lymphangiogenic genes. This study opens a new avenue for investigation of the molecular mechanism of LEPC differentiation and lymphangiogenesis in the offspring of PE and subsequently may impact the treatment of long-term health problems such as cardiovascular and metabolic disorders of offspring with abnormal development of lymphatic vasculature.


Assuntos
Antígeno CD11b/metabolismo , Células Progenitoras Endoteliais/metabolismo , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Adulto , Animais , Diferenciação Celular/fisiologia , Movimento Celular/fisiologia , Proliferação de Células/fisiologia , Feminino , Humanos , Vasos Linfáticos/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pré-Eclâmpsia/metabolismo , Gravidez , Cicatrização/fisiologia
5.
Stem Cells Int ; 2020: 8403192, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32587622

RESUMO

Preeclampsia is a syndrome characterized by deterioration of either the maternal condition or the fetal condition. The adverse intrauterine environment made by preeclampsia results into intrauterine growth restriction and increased risk of a variety of diseases in future life. Given the adverse environment of fetal circulation made in the preeclamptic condition, and the role of mesenchymal stem cell (MSC) as a multipotent progenitor cell, we hypothesized that MSCs derived from human umbilical cord blood (hUCB-MSCs) obtained from preeclampsia are adversely altered or affected compared with normal pregnancy. The aim of this study was to analyze the biological characteristics and compare the functional abilities and gene expression patterns of hUCB-MSCs originating from pregnant women with and without severe preeclampsia. hUCB-MSCs were isolated and cultured from 28 pregnant women with severe preeclampsia and 30 normal pregnant women. hUCB-MSCs obtained from women with preeclampsia were less proliferative and more senescent and had lower telomerase activity and higher ROS activity than cells from women with normal pregnancy. In addition, many senescence-related differentially expressed genes (DEGs) were identified by analysis of microarray gene expression profiles and significantly associated with the Gene Ontology term cell aging. In conclusion, hUCB-MSCs obtained from women with preeclampsia showed the poorly proliferative, more senescent, and decreased telomerase activity, and these characters may be related with functional impairment of MSC from preeclampsia compared with cells from normal pregnancy.

6.
Stem Cell Res Ther ; 11(1): 44, 2020 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-32014040

RESUMO

BACKGROUND: Sepsis is a global inflammatory disease that causes death. It has been reported that mesenchymal stem cell (MSC) treatment can attenuate inflammatory and septic symptoms. In this study, we investigated how interactions between neutrophils and human umbilical cord blood (hUCB)-MSCs in the liver of septic mice are involved in mitigating sepsis that is mediated by MSCs. Accordingly, we aimed to determine whether hUCB-MSC application could be an appropriate treatment for sepsis. METHODS: To induce septic condition, lipopolysaccharide (LPS) was intraperitoneally (i.p.) injected into mice 24 h after the intravenous (i.v.) injection of saline or hUCB-MSCs. To determine the effect of hUCB-MSCs on the immune response during sepsis, histologic analysis, immunoassays, and two-photon intravital imaging were performed 6 h post-LPS injection. For the survival study, mice were monitored for 6 days after LPS injection. RESULTS: The injection (i.v.) of hUCB-MSCs alleviated the severity of LPS-induced sepsis by increasing IL-10 levels (p < 0.001) and decreasing mortality (p < 0.05) in septic mice. In addition, this significantly reduced the recruitment of neutrophils (p < 0.001) to the liver. In hUCB-MSC-treated condition, we also observed several distinct patterns of dynamic interactions between neutrophils and hUCB-MSCs in the inflamed mouse liver, as well as vigorous interactions between hepatic stellate cells (HSCs or ito cells) and hUCB-MSCs. Interestingly, hUCB-MSCs that originated from humans were not recognized as foreign in the mouse body and consequently did not cause graft rejection. CONCLUSIONS: These distinct interaction patterns between innate immune cells and hUCB-MSCs demonstrated that hUCB-MSCs have beneficial effects against LPS-induced sepsis through associations with neutrophils. In addition, the immunomodulatory properties of hUCB-MSCs might enable immune evasion in the host. Taken together, our results suggest the prospects of hUCB-MSCs as a therapeutic tool to inhibit inflammation and alleviate pathological immune responses such as sepsis.


Assuntos
Sangue Fetal/metabolismo , Fígado/fisiopatologia , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/metabolismo , Neutrófilos/metabolismo , Sepse/terapia , Animais , Feminino , Humanos , Camundongos , Sepse/sangue , Sepse/mortalidade , Análise de Sobrevida
7.
Curr Eye Res ; 44(11): 1181-1188, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31204524

RESUMO

Purpose: The function of Solcoseryl in the corneal epithelium has not been fully examined. Here, we investigated the roles of Solcoseryl in the regulation of gene expression and corneal epithelial cell (CEC) activity.Materials and Methods: The effect of Solcoseryl on CEC activity was analyzed through cell migration, adhesion, proliferation, and wound healing assays. Analysis of gene expression was conducted via western blotting and quantitative reverse transcription polymerase chain reaction (PCR).Results: The results demonstrated that Solcoseryl increased the adhesion, migration, proliferation, and wound healing of CECs. Analysis of gene expression showed that Solcoseryl-stimulated CECs exhibited increased expression of mucin family genes, such as MUC1, -5AC, -7, and -16. Solcoseryl also increased the activities of the intracellular signaling molecules AKT, FAK, ERK, and Src in CECs. Using pharmacologic inhibitors of ERK and AKT, we showed that the expression of mucin genes by Solcoseryl is mediated by the activation of ERK and AKT signaling.Conclusions: Our findings demonstrate that Solcoseryl may contribute to the wound healing of CECs by enhancing their migration, adhesion, and proliferation. Additionally, our results suggest that Solcoseryl has a protective effect on ocular surfaces due to its induction of the expression of mucin genes in CECs. These findings suggest that Solcoseryl is a useful therapeutic target for patients with corneal wounds.


Assuntos
Sangue , Doenças da Córnea/terapia , Epitélio Corneano/metabolismo , Expressão Gênica , Mucinas/genética , Cicatrização/fisiologia , Animais , Western Blotting , Bovinos , Movimento Celular , Proliferação de Células , Células Cultivadas , Doenças da Córnea/metabolismo , Doenças da Córnea/patologia , Epitélio Corneano/patologia , Humanos , Mucinas/biossíntese , Soluções Oftálmicas , Transdução de Sinais
8.
Pregnancy Hypertens ; 15: 146-153, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30825912

RESUMO

OBJECTIVES: Individuals delivered from preeclamptic pregnancies exhibit a long-term increased risk of developing cardiovascular and metabolic diseases, likely caused by aberrant fetal cell reprogramming incurred in utero. The present study investigated the functional impairment and epigenetic changes exhibited by endothelial progenitor cells derived from offspring born to preeclamptic pregnancies. STUDY DESIGN: The capacity of CD133+/C-kit+/Lin- (CKL-) human umbilical cord blood endothelial progenitor cells (EPCs) derived from gestationally matched normal and preeclamptic (n = 10 each) pregnancies to differentiate to form outgrowth endothelial cells (OECs) was assessed by observing both their morphology, and the number and size of generated OECs colonies. Likewise, OECs angiogenic function was evaluated via migration, adhesion, and tube-formation assays. EPCs from preeclampsia were cultured in normal-, and preeclampsia-derived serum-conditioned media to assess the effects of environmental factors on EPC differentiation potency and OEC angiogenic function, and finally, EPCs H3K4, H3K9, and H3K27 trimethylation levels were assayed. RESULTS: The preeclampsia-derived CKL- EPCs exhibited decreased H3K4 and H3K9 trimethylation levels, significantly delayed differentiation times, and a significant reduction in both their number of generated OECs colonies, and exhibited reduced OECs migration, adhesion, and tube formation activities compared to those achieved by the normal-derived EPCs. Interestingly, the reduced differentiation potency of the preeclampsia-derived EPCs was not rescued via exposure to normal serum. CONCLUSIONS: Exposure to preeclampsia significantly and irreversibly reduced CKL- EPC differentiation potency and OEC angiogenic function, likely reflecting incurred irreversible epigenetic changes.


Assuntos
Antígeno AC133/sangue , Células Progenitoras Endoteliais/citologia , Epigênese Genética , Pré-Eclâmpsia/sangue , Adulto , Análise de Variância , Estudos de Casos e Controles , Movimento Celular , Feminino , Sangue Fetal , Humanos , Pré-Eclâmpsia/fisiopatologia , Gravidez
9.
PLoS One ; 14(2): e0211864, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30753226

RESUMO

Corneal dystrophy is an autosomal dominant disorder caused by mutations of the transforming growth factor ß-induced (TGFBI) gene on chromosome 5q31.8. This disease is therefore ideally suited for gene therapy using genome-editing technology. Here, we isolated human limbal epithelial stem cells (ABCG2+/ABCB5+ double-positive LESCs) and established a TGFBI knockout using RNA-guided clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 genome editing. An LESC clone generated with a single-guide RNA (sgRNA) targeting exon 4 of the TGFBI gene was sequenced in order to identify potential genomic insertions and deletions near the Cas9/sgRNA-target sites. A detailed analysis of the differences between wild type LESCs and the single LESC clone modified by the TGFBI-targeting sgRNA revealed two distinct mutations, an 8 bp deletion and a 14 bp deletion flanked by a single point mutation. These mutations each lead to a frameshift missense mutation and generate premature stop codons downstream in exon 4. To validate the TGFBI knockout LESC clone, we used single cell culture to isolate four individual sub-clones, each of which was found to possess both mutations present in the parent clone, indicating that the population is homogenous. Furthermore, we confirmed that TGFBI protein expression is abolished in the TGFBI knockout LESC clone using western blot analysis. Collectively, our results suggest that genome editing of TGFBI in LESCs by CRISPR/Cas9 may be useful strategy to treat corneal dystrophy.


Assuntos
Sistemas CRISPR-Cas/genética , Distrofias Hereditárias da Córnea/genética , Proteínas da Matriz Extracelular/genética , Terapia Genética , Fator de Crescimento Transformador beta/genética , Subfamília B de Transportador de Cassetes de Ligação de ATP , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/genética , Distrofias Hereditárias da Córnea/patologia , Distrofias Hereditárias da Córnea/terapia , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Extremidades/crescimento & desenvolvimento , Extremidades/patologia , Edição de Genes , Regulação da Expressão Gênica/genética , Técnicas de Inativação de Genes , Humanos , Cultura Primária de Células , RNA Guia de Cinetoplastídeos/genética , Deleção de Sequência/genética , Análise de Célula Única , Células-Tronco/metabolismo
10.
Am J Reprod Immunol ; 80(1): e12970, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29756666

RESUMO

PROBLEM: The lymphatic vasculature controls leukocytes trafficking and limits the adaptive immune response. In previous models of preeclampsia (PE), defective immune function caused by disruption of lymphangiogenesis was shown to be involved in the disease pathophysiology. Especially, the dysfunction of regulatory T cells (Treg) at the maternal-fetal interface may be one of the causes of severe PE. In particular, activation of Tregs to obtain immune tolerance requires adequate antigen presentation through the lymphatic system. We hypothesized that impaired lymphangiogenesis and imbalanced Tregs at the maternal-fetal interface are associated with the pathophysiology of severe PE. However, the current research addressing this hypothesis is limited. Therefore, to compare differences in lymphangiogenesis in severe PE and normal conditions, we aimed to examine the location of lymphatics at the maternal-fetal interface and to investigate the association between lymphangiogenesis and Tregs in severe PE. METHOD OF STUDY: We obtained entire uterus from normal pregnant mice. Placental and fetal membranes, including decidua, were obtained from 10 pregnant women with severe PE and 10 gestational age-matched controls. Immunohistochemistry for LYVE1 was used to localize the distribution of lymphatic vessels and CD4, CD25, and FOXP3 for Treg. RESULTS: LYVE1-positive vessels were present in the uterine wall of mice. LYVE1-positive lymphatic vessels were localized on the human decidua. Tubular lymphatics were abundant in the control decidua, but significantly reduced in severe PE. Furthermore, lymphatic vessel density correlated with the number of decidual Tregs. CONCLUSION: Abnormal decidual lymphangiogenesis is associated with reduced numbers of decidual Tregs in severe PE.


Assuntos
Vasos Linfáticos/imunologia , Pré-Eclâmpsia/imunologia , Linfócitos T Reguladores/imunologia , Animais , Antígenos CD4/imunologia , Membranas Extraembrionárias/imunologia , Feminino , Fatores de Transcrição Forkhead/imunologia , Idade Gestacional , Humanos , Subunidade alfa de Receptor de Interleucina-2/imunologia , Linfangiogênese/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Placenta/imunologia , Gravidez , Útero/imunologia , Proteínas de Transporte Vesicular/imunologia
11.
Stem Cells Int ; 2017: 7678637, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29230251

RESUMO

Homeostasis and regeneration of corneal epithelia are sustained by limbal epithelial stem cells (LESCs); thus, an LESC deficiency is a major cause of blindness worldwide. Despite the generally promising results of cultivated LESC transplantation, it has been limited by variations in long-term success rates, the use of xenogeneic and undefined culture components, and a scarcity of donor tissues. In this study, we identified the culture conditions required to expand LESCs in vitro and established human limbus-derived highly proliferative ABCG2+/ABCB5+ double-positive LESCs. These LESCs exhibited the LESC marker profile and differentiated into corneal epithelial cells. In addition, cultured LESCs expressed high levels of the stem cell markers Sox2, Oct4, c-Myc, and Klf4, had high telomerase activity, and had stable, normal genomes. These results suggest that our novel cultivation protocol affects the phenotype and differentiation capacity of LESCs. From the limbus, which contains a heterogenous cell population, we have derived highly proliferative ABCG2+/ABCB5+ double-positive cells with the ability to differentiate into corneal epithelial cells. This study opens a new avenue for investigation of the molecular mechanism of LESC maintenance and expansion in vitro and may impact the treatment of corneal disease, particularly corneal blindness due to an LESC deficiency.

12.
Stem Cells Int ; 2017: 4206187, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28894469

RESUMO

Ex vivo culture of human limbal epithelial cells (LECs) is used to treat limbal stem cell (LSC) deficiency, a vision loss condition, and suitable culture systems using feeder cells or serum without animal elements have been developed. This study evaluated the use of human umbilical cord or placenta mesenchymal stem cells (C-MSCs or P-MSCs, resp.) as feeder cells in an animal/serum-free coculture system with human LECs. C-/P-MSCs stimulated LEC colony formation of the stem cell markers (p63, ABCG2) and secreted known LEC clonal growth factors (keratinocyte growth factor, ß-nerve growth factor). Transforming growth factor-ß-induced protein (TGFBIp), an extracellular matrix (ECM) protein, was produced by C-/P-MSCs and resulted in an increase in p63+ ABCG2+ LEC colonies. TGFBIp-activated integrin signaling molecules (FAK, Src, and ERK) were expressed in LECs, and TGFBIp-induced LEC proliferation was effectively blocked by a FAK inhibitor. In conclusion, C-/P-MSCs enhanced LEC culture by increasing growth of the LSC population by secreting growth factors and the ECM protein TGFBIp, which is suggested to be a novel factor for promoting the growth of LECs in culture. C-/P-MSCs may be useful for the generation of animal-free culture systems for the treatment of LSC deficiency.

13.
J Pineal Res ; 63(3)2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28580641

RESUMO

Endoplasmic reticulum (ER) stress is emerging as a factor for the pathogenesis of granular corneal dystrophy type 2 (GCD2). This study was designed to investigate the molecular mechanisms underlying the protective effects of melatonin on ER stress in GCD2. Our results showed that GCD2 corneal fibroblasts were more susceptible to ER stress-induced death than were wild-type cells. Melatonin significantly inhibited GCD2 corneal cell death, caspase-3 activation, and poly (ADP-ribose) polymerase 1 cleavage caused by the ER stress inducer, tunicamycin. Under ER stress, melatonin significantly suppressed the induction of immunoglobulin heavy-chain-binding protein (BiP) and activation of inositol-requiring enzyme 1α (IRE1α), and their downstream target, alternative splicing of X-box binding protein 1(XBP1). Notably, the reduction in BiP and IRE1α by melatonin was suppressed by the ubiquitin-proteasome inhibitor, MG132, but not by the autophagy inhibitor, bafilomycin A1, indicating involvement of the ER-associated protein degradation (ERAD) system. Melatonin treatment reduced the levels of transforming growth factor-ß-induced protein (TGFBIp) significantly, and this reduction was suppressed by MG132. We also found reduced mRNA expression of the ERAD system components HRD1 and SEL1L, and a reduced level of SEL1L protein in GCD2 cells. Interestingly, melatonin treatments enhanced SEL1L levels and suppressed the inhibition of SEL1L N-glycosylation caused by tunicamycin. In conclusion, this study provides new insights into the mechanisms by which melatonin confers its protective actions during ER stress. The results also indicate that melatonin might have potential as a therapeutic agent for ER stress-related diseases including GCD2.


Assuntos
Antioxidantes/uso terapêutico , Distrofias Hereditárias da Córnea/tratamento farmacológico , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Melatonina/uso terapêutico , Antioxidantes/farmacologia , Morte Celular/efeitos dos fármacos , Células Cultivadas , Córnea/citologia , Avaliação Pré-Clínica de Medicamentos , Endorribonucleases/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Humanos , Melatonina/farmacologia , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Resposta a Proteínas não Dobradas , Proteína 1 de Ligação a X-Box/metabolismo
14.
Yonsei Med J ; 58(2): 423-431, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28120575

RESUMO

PURPOSE: Transforming growth factor-ß-induced protein (TGFBIp) is highly expressed in the cornea, and mutant TGFBIp induces corneal diseases. However, the function of TGFBIp in cornea epithelium is not fully investigated. Here, we tested the importance of TGFBIp in regulation of gene expression and corneal epithelial cell (CEC) activity. MATERIALS AND METHODS: The effect of TGFBIp on CEC activity was analyzed by cell migration, adhesion, proliferation and wound healing assay. Analysis of gene expression was examined by western blot and quantitative reverse transcription PCR. RESULTS: The results demonstrated that TGFBIp increased adhesion, migration, proliferation, and wound healing of CECs. Analysis of gene expression presented that TGFBIp-stimulated CECs exhibited increased expression of mucin family genes, such as MUC1, -4, -5AC, and -16. Furthermore, TGFBIp treatment increased the expression of MUC1, -4, -5AC, -7, and -16 in conjunctival epithelial cells. TGFBIp also increased the activity of intracellular signaling molecules ERK and AKT in CECs. Using pharmacologic inhibitors of ERK and AKT, we showed that the expression of mucin genes by TGFBIp is mediated by the activation of ERK and AKT signaling. CONCLUSION: Our findings demonstrate that the locally generated TGFBIp in the cornea may contribute to wound healing of CECs by enhancing the migration, adhesion, and proliferation of CECs. In addition, our results suggest that TGFBIp has a protective effect on ocular surfaces by inducing the expression of mucin genes in corneal and conjunctival epithelial cells. These data suggest that TGFBIp is a useful therapeutic target for patients with corneal wounds.


Assuntos
Córnea/citologia , Células Epiteliais/metabolismo , Proteínas da Matriz Extracelular/fisiologia , Mucinas/metabolismo , Fator de Crescimento Transformador beta/fisiologia , Cicatrização/fisiologia , Western Blotting , Adesão Celular , Movimento Celular/efeitos dos fármacos , Proliferação de Células , Células Cultivadas , Túnica Conjuntiva/citologia , Expressão Gênica , Humanos , Mucinas/genética , Transdução de Sinais
15.
Biochem Biophys Res Commun ; 477(4): 841-846, 2016 09 02.
Artigo em Inglês | MEDLINE | ID: mdl-27373828

RESUMO

Granular corneal dystrophy type 2 (GCD2) is caused by a point mutation (R124H) in the transforming growth factor ß-induced (TGFBI) gene. In GCD2 corneal fibroblasts, secretion of the accumulated mutant TGFBI-encoded protein (TGFBIp) is delayed via the endoplasmic reticulum (ER)/Golgi-dependent secretory pathway. However, ER stress as the pathogenic mechanism underlying GCD2 has not been fully characterized. The aim of this study was to confirm whether ER stress is linked to GCD2 pathogenesis and whether the chemical chaperone, 4-phenylbutyric acid (4-PBA), could be exploited as a therapy for GCD2. We found that the ER chaperone binding immunoglobulin protein (BiP) and the protein disulfide isomerase (PDI) were elevated in GCD2. Western bolt analysis also showed a significant increase in both the protein levels and the phosphorylation of the key ER stress kinases, inositol-requiring enzyme 1α (IRE1α) and double stranded RNA activated protein kinase (PKR)-like ER kinase, as well as in levels of their downstream targets, X box-binding protein 1 (XBP1) and activating transcription factor 4, respectively, in GCD2 corneal fibroblasts. GCD2 cells were found to be more susceptible to ER stress-induced cell death than were wild-type corneal fibroblasts. Treatment with 4-PBA considerably reduced the levels of BiP, IRE1α, and XBP1 in GCD2 cells; notably, 4-PBA treatment significantly reduced the levels of TGFBIp without change in TGFBI mRNA levels. In addition, TGFBIp levels were significantly reduced under ER stress and this reduction was considerably suppressed by the ubiquitin proteasome inhibitor MG132, indicating TGFBIp degradation via the ER-associated degradation pathway. Treatment with 4-PBA not only protected against the GCD2 cell death induced by ER stress but also significantly suppressed the MG132-mediated increase in TGFBIp levels under ER stress. Together, these results suggest that ER stress might comprise an important factor in GCD2 pathophysiology and that the effects of 4-PBA treatment might have important implications for the development of GCD2 therapeutics.


Assuntos
Córnea/fisiopatologia , Distrofias Hereditárias da Córnea/fisiopatologia , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Degradação Associada com o Retículo Endoplasmático/efeitos dos fármacos , Proteínas da Matriz Extracelular/metabolismo , Fenilbutiratos/administração & dosagem , Fator de Crescimento Transformador beta/metabolismo , Apoptose/efeitos dos fármacos , Células Cultivadas , Córnea/efeitos dos fármacos , Córnea/metabolismo , Distrofias Hereditárias da Córnea/tratamento farmacológico , Distrofias Hereditárias da Córnea/patologia , Relação Dose-Resposta a Droga , Regulação para Baixo/efeitos dos fármacos , Proteínas da Matriz Extracelular/genética , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fibroblastos/patologia , Humanos , Mutação/efeitos dos fármacos , Mutação/genética , Fator de Crescimento Transformador beta/genética
16.
Biochem Biophys Res Commun ; 472(1): 150-5, 2016 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-26915797

RESUMO

Transforming growth factor-ß (TGF-ß)-induced gene (TGFBI) protein (TGFBIp) is associated with granular corneal dystrophy type 2 (GCD2). TGFBIp levels can affect GCD2 phenotypes, but the underlying molecular mechanisms have not been fully elucidated. We investigated the involvement of microRNA (miRNA) and TGF-ß in the regulation of TGFBIp expression in corneal fibroblasts. Ectopic expression of miR-9, miR-21, and miR-181a significantly decreased TGFBIp levels. Conversely, expression of miR-21 and miR-181a was induced by TGF-ß1. Expression of miR-21 was 10-fold higher than that of miR-9 and miR-181a in corneal fibroblasts. Additionally, TGF-ß1 expression was significantly higher than that of TGF-ß2 and TGF-ß3 in corneal fibroblasts, whereas expression of all three TGF-ß forms was not significantly different between wild-type (WT) and GCD2 homozygotes (HO) corneal fibroblasts. Taken together, these data indicate that TGFBIp expression is positively regulated by TGF-ß, whereas TGF-ß-induced miR-21 and miR-181a negatively regulate TGFBIp expression. In conclusion, TGFBIp levels in corneal fibroblasts are controlled via the coordinated activity of miR-21 and miR-181a and by Smad signaling. Pharmacologic modulation of these miRNAs and TGF-ß signaling could have therapeutic potential for TGFBI-associated corneal dystrophy, including GCD2.


Assuntos
Córnea/metabolismo , Proteínas da Matriz Extracelular/genética , Proteínas da Matriz Extracelular/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Proteínas Smad/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo , Células Cultivadas , Córnea/citologia , Distrofias Hereditárias da Córnea/genética , Distrofias Hereditárias da Córnea/metabolismo , Distrofias Hereditárias da Córnea/patologia , Fibroblastos/citologia , Fibroblastos/metabolismo , Regulação da Expressão Gênica , Homozigoto , Humanos , Modelos Biológicos , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Mutação Puntual , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais
17.
Sci Rep ; 6: 20739, 2016 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-26857144

RESUMO

Metastasis is the main cause of mortality in cancer patients. Although there are many anti-cancer drugs targeting tumor growth, anti-metastatic agents are rarely developed. Angiogenesis and lymphangiogenesis are crucial for cancer progression; in particular, lymphangiogenesis is pivotal for metastasis in cancer. Here we report that lithium inhibits colon cancer metastasis by blocking lymphangiogenesis. Lithium reduces the expression of transforming growth factor-ß-induced protein (TGFBIp) in colon cancer cells by inhibiting Smad3 phosphorylation via GSK3ß inactivation. Moreover, lithium inhibits lymphatic endothelial cell migration, which is increased upon TGFBIp expression in tumor cells. Lithium had no significant effect on SW620 tumor growth in vitro and in vivo; however, it inhibited lymphangiogenesis in tumors. In tumor xenografts model, lithium was found to prevent metastasis to the lungs, liver, and lymph nodes by inhibiting TGFBIp-induced tumor lymphangiogenesis. Collectively, our findings demonstrate a novel role of lithium in the inhibition of colon cancer metastasis by blocking TGFBIp expression, and thereby TGFBIp-induced lymphangiogenesis, in primary tumors.


Assuntos
Neoplasias do Colo/tratamento farmacológico , Proteínas da Matriz Extracelular/biossíntese , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Lítio/farmacologia , Linfangiogênese/efeitos dos fármacos , Proteínas de Neoplasias/metabolismo , Fator de Crescimento Transformador beta/biossíntese , Animais , Linhagem Celular Tumoral , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Glicogênio Sintase Quinase 3 beta/metabolismo , Camundongos , Metástase Neoplásica , Fosforilação/efeitos dos fármacos , Proteína Smad3/metabolismo
18.
Prog Retin Eye Res ; 50: 67-88, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26612778

RESUMO

Transforming growth factor beta-induced (TGFBI) corneal dystrophies are a group of inherited progressive corneal diseases. Accumulation of transforming growth factor beta-induced protein (TGFBIp) is involved in the pathogenesis of TGFBI corneal dystrophies; however, the exact molecular mechanisms are not fully elucidated. In this review article, we summarize the current knowledge of TGFBI corneal dystrophies including clinical manifestations, epidemiology, most common and recently reported associated mutations for each disease, and treatment modalities. We review our current understanding of the molecular mechanisms of granular corneal dystrophy type 2 (GCD2) and studies of other TGFBI corneal dystrophies. In GCD2 corneal fibroblasts, alterations of morphological characteristics of corneal fibroblasts, increased susceptibility to intracellular oxidative stress, dysfunctional and fragmented mitochondria, defective autophagy, and alterations of cell cycle were observed. Other studies of mutated TGFBIp show changes in conformational structure, stability and proteolytic properties in lattice and granular corneal dystrophies. Future research should be directed toward elucidation of the biochemical mechanism of deposit formation, the relationship between the mutated TGFBIp and the other materials in the extracellular matrix, and the development of gene therapy and pharmaceutical agents.


Assuntos
Distrofias Hereditárias da Córnea , Fator de Crescimento Transformador beta/metabolismo , Autofagia/fisiologia , Córnea/metabolismo , Distrofias Hereditárias da Córnea/etiologia , Distrofias Hereditárias da Córnea/genética , Distrofias Hereditárias da Córnea/metabolismo , Distrofias Hereditárias da Córnea/terapia , Fibroblastos/patologia , Predisposição Genética para Doença , Humanos , Mutação , Estresse Oxidativo/fisiologia
19.
Stem Cells Int ; 2016: 7162160, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-28074098

RESUMO

Recent evidence suggests that mononuclear cells (MNCs) derived from bone marrow and cord blood can differentiate into mesenchymal stem cells (MSCs) or outgrowth endothelial cells (OECs). However, controversy exists as to whether MNCs have the pluripotent capacity to differentiate into MSCs or OECs or are a mixture of cell lineage-determined progenitors of MSCs or OECs. Here, using CD133+/C-kit+/Lin- mononuclear cells (CKL- cells) isolated from human umbilical cord blood using magnetic cell sorting, we characterized the potency of MNC differentiation. We first found that CKL- cells cultured with conditioned medium of OECs or MSCs differentiated into OECs or MSCs and this differentiation was also induced by cell-to-cell contact. When we cultured single CKL- cells on OEC- or MSC-conditioned medium, the cells differentiated morphologically and genetically into OEC- or MSC-like cells, respectively. Moreover, we confirmed that OECs or MSCs differentiated from CKL- cells had the ability to form capillary-like structures in Matrigel and differentiate into osteoblasts, chondrocytes, and adipocytes. Finally, using microarray analysis, we identified specific factors of OECs or MSCs that could potentially be involved in the differentiation fate of CKL- cells. Together, these results suggest that cord blood-derived CKL- cells possess at least bipotential differentiation capacity toward MSCs or OECs.

20.
BMC Med Genomics ; 8: 74, 2015 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-26553048

RESUMO

BACKGROUND: TGFß1-induced expression of transforming growth factor ß-induced protein (TGFBIp) and extracellular matrix (ECM) genes plays a major role in the development of granular corneal dystrophy type 2 (GCD2: also called Avellino corneal dystrophy). Although some key transcription factors are known, the epigenetic mechanisms modulating TGFBIp and ECM expression remain unclear. We examined the role of chromatin markers such as histone H3 lysine methylation (H3Kme) in TGFß1-induced TGFBIp and ECM gene expression in normal and GCD2-derived human corneal fibroblasts. METHODS: Wild-type (n = 3), GCD2-heterozygous (n = 1), and GCD2-homozygous (n = 3) primary human corneal fibroblasts were harvested from human donors and patients prepared. Microarray and gene-expression profiling, Chromatin immunoprecipitation microarray analysis, and Methylated DNA isolation assay-assisted CpG microarrays was performed in Wild-type and GCD2-homozygous human cells. RESULTS: Transcription and extracellular-secretion levels of TGFBIp were high in normal cells compared with those in GCD2-derived cells and were related to H3K4me3 levels but not to DNA methylation over the TGFBI locus. TGFß1 increased the expression of TGFBIp and the ECM-associated genes connective tissue growth factor, collagen-α2[Ι], and plasminogen activator inhibitor-1 in normal corneal fibroblasts. Increased levels of gene-activating markers (H3K4me1/3) and decreased levels of repressive markers (H3K27me3) at the promoters of those gene accompanied the changes in expression. TGFß1 also increased recruitment of the H3K4 methyltransferase MLL1 and of SET7/9 and also the binding of Smad3 to the promoters. Knockdown of both MLL1 and SET7/9 significantly blocked the TGFß1-induced gene expression and inhibited TGFß1-induced changes in promoter H3K4me1/3 levels. Those effects were very weak, however, in GCD2-derived corneal fibroblasts. CONCLUSIONS: Taken together, the results show the functional role of H3K4me in TGFß1-mediated TGFBIp and ECM gene expression in corneal fibroblasts. Pharmacologic and other therapies that regulate these modifications could have potential cornea-protective effects for granular corneal dystrophy.


Assuntos
Distrofias Hereditárias da Córnea/genética , Proteínas da Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Fibroblastos/citologia , Fibroblastos/patologia , Regulação da Expressão Gênica , Histonas/metabolismo , Fator de Crescimento Transformador beta/genética , Córnea/citologia , Córnea/patologia , Distrofias Hereditárias da Córnea/metabolismo , Distrofias Hereditárias da Córnea/patologia , Proteínas da Matriz Extracelular/metabolismo , Técnicas de Silenciamento de Genes , Histona Metiltransferases , Histona-Lisina N-Metiltransferase/deficiência , Histona-Lisina N-Metiltransferase/genética , Histona-Lisina N-Metiltransferase/metabolismo , Histonas/química , Homozigoto , Humanos , Lisina/metabolismo , Metilação , Proteína de Leucina Linfoide-Mieloide/deficiência , Proteína de Leucina Linfoide-Mieloide/genética , Proteína de Leucina Linfoide-Mieloide/metabolismo , Transporte Proteico , Proteína Smad3/metabolismo , Transcrição Gênica , Fator de Crescimento Transformador beta/metabolismo , Fator de Crescimento Transformador beta1/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA