AIM:To elucidate the role of vascular endothelial growth factor-165b(VEGF-165b)in blood-retinal barrier(BRB)injury in the rat acute glaucoma model.METHODS:In this study,the rat acute high intraocular pressure(HIOP)mod...AIM:To elucidate the role of vascular endothelial growth factor-165b(VEGF-165b)in blood-retinal barrier(BRB)injury in the rat acute glaucoma model.METHODS:In this study,the rat acute high intraocular pressure(HIOP)model was established before and after intravitreous injection of anti-VEGF-165b antibody.The expression of VEGF-165b and zonula occludens-1(ZO-1)in rat retina was detected by double immunofluorescence staining and Western blotting,and the breakdown of BRB was detected by Evans blue(EB)dye.RESULTS:The intact retina of rats expressed VEGF-165b and ZO-1 protein,which were mainly located in the retinal ganglion cell layer and the inner nuclear layer and were both co-expressed with vascular endothelial cell markers CD31.After acute HIOP,the expression of VEGF-165b was up-regulated;the expression of ZO-1 was down-regulated at 12h and then recovered at 3d;EB leakage increased,peaking at 12h.After intravitreous injection of anti-VEGF-165b antibody,the expression of VEGF-165b protein was no significantly changed;and the down-regulation of the expression of ZO-1 was more obvious;EB leakage became more serious,peaking at 3d.EB analysis also showed that EB leakage in the peripheral retina was greater than that in the central retina.CONCLUSION:The endogenous VEGF-165b protein may protect the BRB from acute HIOP by regulating the expression of ZO-1.The differential destruction of BRB after acute HIOP may be related to the selective loss of retinal ganglion cells.展开更多
Magnolol,a compound extracted from Magnolia officinalis,demonstrates potential efficacy in addressing metabolic dysfunction and cardiovascular diseases.Its biological activities encompass anti-inflammatory,antioxidant...Magnolol,a compound extracted from Magnolia officinalis,demonstrates potential efficacy in addressing metabolic dysfunction and cardiovascular diseases.Its biological activities encompass anti-inflammatory,antioxidant,anticoagulant,and anti-diabetic effects.Growth/differentiation factor-15(GDF-15),a member of the transforming growth factorβsuperfamily,is considered a potential therapeutic target for metabolic disorders.This study investigated the impact of magnolol on GDF-15 production and its underlying mechanism.The research examined the pharmacological effect of magnolol on GDF-15 expression in vitro and in vivo,and determined the involvement of endoplasmic reticulum(ER)stress signaling in this process.Luciferase reporter assays,chromatin immunoprecipitation,and in vitro DNA binding assays were employed to examine the regulation of GDF-15 by activating transcription factor 4(ATF4),CCAAT enhancer binding proteinγ(CEBPG),and CCCTC-binding factor(CTCF).The study also investigated the effect of magnolol and ATF4 on the activity of a putative enhancer located in the intron of the GDF-15 gene,as well as the influence of single nucleotide polymorphisms(SNPs)on magnolol and ATF4-induced transcription activity.Results demonstrated that magnolol triggers GDF-15 production in endothelial cells(ECs),hepatoma cell line G2(HepG2)and hepatoma cell line 3B(Hep3B)cell lines,and primary mouse hepatocytes.The cooperative binding of ATF4 and CEBPG upstream of the GDF-15 gene or the E1944285 enhancer located in the intron led to full-power transcription of the GDF-15 gene.SNP alleles were found to impact the magnolol and ATF4-induced transcription activity of GDF-15.In high-fat diet ApoE^(-/-)mice,administration of magnolol induced GDF-15 production and partially suppressed appetite through GDF-15.These findings suggest that magnolol regulates GDF-15 expression through priming of promoter and enhancer activity,indicating its potential as a drug for the treatment of metabolic disorders.展开更多
BACKGROUND Hypoxia-inducible factor 1α(HIF-1α)plays a crucial role in the prognosis of breast cancer,but the current evidence remains inconclusive.AIM To provide comprehensive evidence about the correlation of alter...BACKGROUND Hypoxia-inducible factor 1α(HIF-1α)plays a crucial role in the prognosis of breast cancer,but the current evidence remains inconclusive.AIM To provide comprehensive evidence about the correlation of altered HIF-1αexpression with overall survival(OS)and disease-free survival(DFS)in breast cancer patients.METHODS A systematic search was conducted in PubMed,Embase,and Web of Science databases to collect relevant articles that were published before April 8,2024.A meta-analysis was used to assess the impact of altered HIF-1αexpression on the OS and DFS of breast cancer patients.Subgroup and sensitivity analyses were also performed in this meta-analysis.RESULTS This meta-analysis included 40 studies.The average percentage of breast cancer patients with high HIF-1αexpression was 39.6%.The overall meta-analysis results demonstrated that high HIF-1αexpression is strongly linked to poor outcomes in patients of breast cancer.Compared with low HIF-1αexpression,the overall hazard ratio for OS in patients with high HIF-1αexpression was 1.47[95%confidence interval(CI):1.29-1.69],and the overall hazard ratio for DFS was 1.82(95%CI:1.56-2.12).Furthermore,both OS[1.18(95%CI:1.01-1.38)]and DFS[1.79(95%CI:1.03-3.11)]were markedly shorter in triple-negative breast cancer cases with high HIF-1αexpression.Subgroup analysis revealed that the antibody used to detect HIF-1αexpression affected only the correlation linking HIF-1αexpression to DFS in breast cancer patients(P=0.0004).Furthermore,the sensitivity analysis demonstrates that the overall conclusions of the meta-analysis were unaffected by the removal of individual studies.CONCLUSION Compared to patients with low HIF-1αexpression,those with high expression level had shorter OS and DFS.However,the prognostic significance of high HIF-1αexpression varies across molecularly stratified breast cancer cohorts needs to be further elucidated.展开更多
BACKGROUND Hypoxia in oral cancer promotes tumoral invasion by inducing epithelial-mesenchymal transition,leading to aggressive tumor progression.AIM To characterize the expression of hypoxia-inducible factor 1-alpha(...BACKGROUND Hypoxia in oral cancer promotes tumoral invasion by inducing epithelial-mesenchymal transition,leading to aggressive tumor progression.AIM To characterize the expression of hypoxia-inducible factor 1-alpha(HIF-1α)at the invasive tumor front(ITF)in comparison to tumor islands(TI)in oral squamous cell carcinoma(OSCC)and to explore its relationship with E-cadherin and Vimentin expression.METHODS Thirty-eight cases of OSCC and five cases of normal oral mucosa(NOM)were included in this study.The ITF was identified based on the region and immune expression of AE1/AE3.Immunohistochemistry was performed to assess the expression of HIF-1α,Vimentin,and E-cadherin.The immunostaining was analyzed using an immunoreactive score,and the results were illustrated using immunofluorescence.RESULTS HIF-1αexpression was significantly higher in the TI region compared to the ITF region(P=0.0134).Additionally,a significant difference was observed between TI and NOM(P=0.0115).In the ITF regions,HIF-1αexpression showed a significant correlation with Vimentin expression,with higher levels of HIF-1αassociated with increased Vimentin expression(P=0.017).CONCLUSION Based on the results of this study,HIF-1αappears to play a distinct role in OSCC tumor progression,underscoring the importance of exploring hypoxia-driven changes in cellular phenotype at the ITF of OSCC.Further research is needed to better understand their impact on OSCC prognosis.展开更多
Adult neurogenesis continuously produces new neurons critical for cognitive plasticity in adult rodents.While it is known transforming growth factor-βsignaling is important in embryonic neurogenesis,its role in postn...Adult neurogenesis continuously produces new neurons critical for cognitive plasticity in adult rodents.While it is known transforming growth factor-βsignaling is important in embryonic neurogenesis,its role in postnatal neurogenesis remains unclear.In this study,to define the precise role of transforming growth factor-βsignaling in postnatal neurogenesis at distinct stages of the neurogenic cascade both in vitro and in vivo,we developed two novel inducible and cell type-specific mouse models to specifically silence transforming growth factor-βsignaling in neural stem cells in(mGFAPcre-ALK5fl/fl-Ai9)or immature neuroblasts in(DCXcreERT2-ALK5fl/fl-Ai9).Our data showed that exogenous transforming growth factor-βtreatment led to inhibition of the proliferation of primary neural stem cells while stimulating their migration.These effects were abolished in activin-like kinase 5(ALK5)knockout primary neural stem cells.Consistent with this,inhibition of transforming growth factor-βsignaling with SB-431542 in wild-type neural stem cells stimulated proliferation while inhibited the migration of neural stem cells.Interestingly,deletion of transforming growth factor-βreceptor in neural stem cells in vivo inhibited the migration of postnatal born neurons in mGFAPcre-ALK5fl/fl-Ai9 mice,while abolishment of transforming growth factor-βsignaling in immature neuroblasts in DCXcreERT2-ALK5fl/fl-Ai9 mice did not affect the migration of these cells in the hippocampus.In summary,our data supports a dual role of transforming growth factor-βsignaling in the proliferation and migration of neural stem cells in vitro.Moreover,our data provides novel insights on cell type-specific-dependent requirements of transforming growth factor-βsignaling on neural stem cell proliferation and migration in vivo.展开更多
Pulmonary arterial hypertension(PAH)is a progressive disease marked by degeneration of the lung’s blood vessels.As the disease progresses,the resistance to blood flow in the pulmonary arteries increases,putting a str...Pulmonary arterial hypertension(PAH)is a progressive disease marked by degeneration of the lung’s blood vessels.As the disease progresses,the resistance to blood flow in the pulmonary arteries increases,putting a strain on the right side of the heart as it pumps blood through the lungs.PAH is characterized by changes in the structure of blood vessels and excessive cell growth.Untreated PAH leads to irreversible right-sided heart failure,often despite medical intervention.Patients experience a gradual decline in function until they are unable to perform daily activities.Advances in treatment have improved the prognosis for many PAH patients.Currently approved therapies target the prostacyclin,endothelin,nitric oxide,or phosphodiesterase pathways to slow the progression of the disease.To address the unmet need for effective PAH therapies,research efforts are focused on identifying new targets and developing therapies that specifically address the underlying disease mechanisms and restore vascular wall homeostasis.Among these,sotatercept,a fusion protein that targets the transforming growth factor-βsuperfamily signaling pathway,has emerged as a promising therapeutic option.In this review,we examine the available evidence from clinical trials to assess the potential of sotatercept as a treatment for PAH.展开更多
OBJECTIVE:To investigate the impact of Shenhua tablet(肾华片,SHT)on renal macrophage polarization and renal injury in mice with diabetic kidney disease(DKD)and to explore the potential mechanism involving the hypoxia-...OBJECTIVE:To investigate the impact of Shenhua tablet(肾华片,SHT)on renal macrophage polarization and renal injury in mice with diabetic kidney disease(DKD)and to explore the potential mechanism involving the hypoxia-inducible factor-1α(HIF-1α)and pyruvate kinase M2(PKM2)signaling pathway,along with the glycolysis metabolism pathway.METHODS:The animals were divided into the following groups:Model,Control,dapagliflozin,SHT low-dose,SHT medium-dose,and SHT high-dose.We assessed 24-hour urine protein(24 h-UTP)levels,urinary albuminto-creatinine ratio,and regularly monitored fasting blood glucose during the treatment period.After treatment,we examined renal tissue structure,renal function(urea nitrogen,uric acid,creatinine,cystatin C,β2-microglobulin),and glycolysis in renal macrophages.Additionally,we observed macrophage polarization in renal tissue and measured inflammatory factors(tumor necrosis factor-α,interleukin-1β,interleukin-6,interleukin-10,monocyte chemoattractant protein-1)to assess the immunoinflammatory status of the renal tissue.Finally,we investigated the expression of the HIF-1α/PKM2 signaling pathway in macrophages to explore its role in the glycolysis process.RESULTS:SHT shows a beneficial effect in treating DKD by reducing 24 h-UTP,regulating blood glucose levels,improving renal tissue structure,protecting renal function,inhibiting macrophage glycolysis,reducing macrophage transformation to the M1 state,and suppressing the expression of the HIF-1α/PKM2 signaling pathway.CONCLUSION:SHT may exert renoprotective effects by inhibiting macrophage glycolysis via the HIF-1α/PKM2 signaling pathway.This inhibition decreases macrophage M1 polarization and reduces immunoinflammatory injury in the renal tissue of DKD mice.展开更多
Objective MicroRNA-1(miR-1)aggravates myocardial ischemia–reperfusion(I/R)injury,whereas insulin-like growth factor-1(IGF-1)maintains cardiomyocyte homeostasis.In this study,the aim is to investigate whether miR-1 ca...Objective MicroRNA-1(miR-1)aggravates myocardial ischemia–reperfusion(I/R)injury,whereas insulin-like growth factor-1(IGF-1)maintains cardiomyocyte homeostasis.In this study,the aim is to investigate whether miR-1 can exacerbate I/R injury through the regulation of IGF-1.Methods The infarct area,lactate dehydrogenase,miR-1 level,and apoptosis level were examined in the Langendorff isolated rat I/R model.The hypoxia–reoxygenation model of rat cardiacmyocytes and H9c2 cells were developed to determine the levels of miR-1,IGF-1 mRNA,and IGF-1 protein.Furthermore,the dual-luciferase assay was used to verify the relationship between miR-1 and IGF-1.Results Overexpression of miR-1 increased the level of apoptosis and decreased the IGF-1 expression.However,inhibition of miR-1 expression decreased the level of apoptosis,alleviated the degree of injury,and increased the IGF-1 expression.Overexpression of IGF-1 also reduced the degree of cellular damage and level of apoptosis caused by the overexpression of miR-1.When IGF-1 was knocked down,myocardial cells displayed more severe damage and a higher apoptosis level,even with decreased levels of miR-1.Conclusion miR-1 promotes apoptosis and aggravates I/R injury by downregulating IGF-1.展开更多
It has been well documented that bone morphogenetic .proteins (BMPs), a group of proteins belonging to the transforming growth factor-β (TGFβ) superfamily, can induce bone formation, both in vivo and in vitro. B...It has been well documented that bone morphogenetic .proteins (BMPs), a group of proteins belonging to the transforming growth factor-β (TGFβ) superfamily, can induce bone formation, both in vivo and in vitro. Bone morphogenetic protein-2 (BMP2) is a potent osteoinductive factor and is being evaluated as a bone growth inducer for orthopedic applications.1 Vascular endothelial growth factor (VEGF), the best-characterized angiogenic factor,展开更多
Injuries caused by trauma and neurodegenerative diseases can damage the peripheral nervous system and cause functional deficits.Unlike in the central nervous system,damaged axons in peripheral nerves can be induced to...Injuries caused by trauma and neurodegenerative diseases can damage the peripheral nervous system and cause functional deficits.Unlike in the central nervous system,damaged axons in peripheral nerves can be induced to regenerate in response to intrinsic cues after reprogramming or in a growth-promoting microenvironment created by Schwann cells.However,axon regeneration and repair do not automatically result in the restoration of function,which is the ultimate therapeutic goal but also a major clinical challenge.Transforming growth factor(TGF)is a multifunctional cytokine that regulates various biological processes including tissue repair,embryo development,and cell growth and differentiation.There is accumulating evidence that TGF-βfamily proteins participate in peripheral nerve repair through various factors and signaling pathways by regulating the growth and transformation of Schwann cells;recruiting specific immune cells;controlling the permeability of the blood-nerve barrier,thereby stimulating axon growth;and inhibiting remyelination of regenerated axons.TGF-βhas been applied to the treatment of peripheral nerve injury in animal models.In this context,we review the functions of TGF-βin peripheral nerve regeneration and potential clinical applications.展开更多
BACKGROUND Pancreatic cancer(PC)is associated with some of the worst prognoses of all major cancers.Thymoquinone(TQ)has a long history in traditional medical practice and is known for its anti-cancer,anti-inflammatory...BACKGROUND Pancreatic cancer(PC)is associated with some of the worst prognoses of all major cancers.Thymoquinone(TQ)has a long history in traditional medical practice and is known for its anti-cancer,anti-inflammatory,anti-fibrosis and antioxidant pharmacological activities.Recent studies on hypoxia-inducible factor-1α(HIF-1α)and PC have shown that HIF-1αaffects the occurrence and development of PC in many aspects.In addition,TQ could inhibit the development of renal cancer by decreasing the expression of HIF-1α.Therefore,we speculate whether TQ affects HIF-1αexpression in PC cells and explore the mechanism.AIM To elucidate the effect of TQ in PC cells and the regulatory mechanism of HIF-1αexpression.METHODS Cell counting kit-8 assay,Transwell assay and flow cytometry were performed to detect the effects of TQ on the proliferative activity,migration and invasion ability and apoptosis of PANC-1 cells and normal pancreatic duct epithelial(hTERTHPNE)cells.Quantitative real-time polymerase chain reaction and western blot assay were performed to detect the expression of HIF-1αmRNA and protein in PC cells.The effects of TQ on the HIF-1αprotein initial expression pathway and ubiquitination degradation in PANC-1 cells were examined by western blot assay and co-immunoprecipitation.RESULTS TQ significantly inhibited proliferative activity,migration,and invasion ability and promoted apoptosis of PANC-1 cells;however,no significant effects on hTERT-HPNE cells were observed.TQ significantly reduced the mRNA and protein expression levels of HIF-1αin PANC-1,AsPC-1,and BxPC-3 cells.TQ significantly inhibited the expression of the HIF-1αinitial expression pathway(PI3K/AKT/mTOR)related proteins,and promoted the ubiquitination degradation of the HIF-1αprotein in PANC-1 cells.TQ had no effect on the hydroxylation and von Hippel Lindau protein mediated ubiquitination degradation of the HIF-1αprotein but affected the stability of the HIF-1αprotein by inhibiting the interaction between HIF-1αand HSP90,thus promoting its ubiquitination degradation.CONCLUSION The regulatory mechanism of TQ on HIF-1αprotein expression in PC cells was mainly to promote the ubiquitination degradation of the HIF-1αprotein by inhibiting the interaction between HIF-1αand HSP90;Secondly,TQ reduced the initial expression of HIF-1αprotein by inhibiting the PI3K/AKT/mTOR pathway.展开更多
基金Supported by the National Natural Science Foundation of China(No.81660217)Youth Foundation of the First Affiliated Hospital of Hainan Medical University(No.HYYFYPY201922)。
文摘AIM:To elucidate the role of vascular endothelial growth factor-165b(VEGF-165b)in blood-retinal barrier(BRB)injury in the rat acute glaucoma model.METHODS:In this study,the rat acute high intraocular pressure(HIOP)model was established before and after intravitreous injection of anti-VEGF-165b antibody.The expression of VEGF-165b and zonula occludens-1(ZO-1)in rat retina was detected by double immunofluorescence staining and Western blotting,and the breakdown of BRB was detected by Evans blue(EB)dye.RESULTS:The intact retina of rats expressed VEGF-165b and ZO-1 protein,which were mainly located in the retinal ganglion cell layer and the inner nuclear layer and were both co-expressed with vascular endothelial cell markers CD31.After acute HIOP,the expression of VEGF-165b was up-regulated;the expression of ZO-1 was down-regulated at 12h and then recovered at 3d;EB leakage increased,peaking at 12h.After intravitreous injection of anti-VEGF-165b antibody,the expression of VEGF-165b protein was no significantly changed;and the down-regulation of the expression of ZO-1 was more obvious;EB leakage became more serious,peaking at 3d.EB analysis also showed that EB leakage in the peripheral retina was greater than that in the central retina.CONCLUSION:The endogenous VEGF-165b protein may protect the BRB from acute HIOP by regulating the expression of ZO-1.The differential destruction of BRB after acute HIOP may be related to the selective loss of retinal ganglion cells.
基金supported by the National Natural Science Foundation of China(Nos.82171552 and 82170479)the Natural Science Foundation of Shanghai Ctiy(No.21ZR1457500)the Science and Technology Bureau of Shanghai Putuo District(No.ptkwws202102).
文摘Magnolol,a compound extracted from Magnolia officinalis,demonstrates potential efficacy in addressing metabolic dysfunction and cardiovascular diseases.Its biological activities encompass anti-inflammatory,antioxidant,anticoagulant,and anti-diabetic effects.Growth/differentiation factor-15(GDF-15),a member of the transforming growth factorβsuperfamily,is considered a potential therapeutic target for metabolic disorders.This study investigated the impact of magnolol on GDF-15 production and its underlying mechanism.The research examined the pharmacological effect of magnolol on GDF-15 expression in vitro and in vivo,and determined the involvement of endoplasmic reticulum(ER)stress signaling in this process.Luciferase reporter assays,chromatin immunoprecipitation,and in vitro DNA binding assays were employed to examine the regulation of GDF-15 by activating transcription factor 4(ATF4),CCAAT enhancer binding proteinγ(CEBPG),and CCCTC-binding factor(CTCF).The study also investigated the effect of magnolol and ATF4 on the activity of a putative enhancer located in the intron of the GDF-15 gene,as well as the influence of single nucleotide polymorphisms(SNPs)on magnolol and ATF4-induced transcription activity.Results demonstrated that magnolol triggers GDF-15 production in endothelial cells(ECs),hepatoma cell line G2(HepG2)and hepatoma cell line 3B(Hep3B)cell lines,and primary mouse hepatocytes.The cooperative binding of ATF4 and CEBPG upstream of the GDF-15 gene or the E1944285 enhancer located in the intron led to full-power transcription of the GDF-15 gene.SNP alleles were found to impact the magnolol and ATF4-induced transcription activity of GDF-15.In high-fat diet ApoE^(-/-)mice,administration of magnolol induced GDF-15 production and partially suppressed appetite through GDF-15.These findings suggest that magnolol regulates GDF-15 expression through priming of promoter and enhancer activity,indicating its potential as a drug for the treatment of metabolic disorders.
基金Supported by the Henan Province Medical Science and Technology Tackling Plan Joint Construction Project,No.LHGJ20220684Zhengzhou University Tianjian Advanced Biomedical Laboratory Funding Project,No.BS20240101.
文摘BACKGROUND Hypoxia-inducible factor 1α(HIF-1α)plays a crucial role in the prognosis of breast cancer,but the current evidence remains inconclusive.AIM To provide comprehensive evidence about the correlation of altered HIF-1αexpression with overall survival(OS)and disease-free survival(DFS)in breast cancer patients.METHODS A systematic search was conducted in PubMed,Embase,and Web of Science databases to collect relevant articles that were published before April 8,2024.A meta-analysis was used to assess the impact of altered HIF-1αexpression on the OS and DFS of breast cancer patients.Subgroup and sensitivity analyses were also performed in this meta-analysis.RESULTS This meta-analysis included 40 studies.The average percentage of breast cancer patients with high HIF-1αexpression was 39.6%.The overall meta-analysis results demonstrated that high HIF-1αexpression is strongly linked to poor outcomes in patients of breast cancer.Compared with low HIF-1αexpression,the overall hazard ratio for OS in patients with high HIF-1αexpression was 1.47[95%confidence interval(CI):1.29-1.69],and the overall hazard ratio for DFS was 1.82(95%CI:1.56-2.12).Furthermore,both OS[1.18(95%CI:1.01-1.38)]and DFS[1.79(95%CI:1.03-3.11)]were markedly shorter in triple-negative breast cancer cases with high HIF-1αexpression.Subgroup analysis revealed that the antibody used to detect HIF-1αexpression affected only the correlation linking HIF-1αexpression to DFS in breast cancer patients(P=0.0004).Furthermore,the sensitivity analysis demonstrates that the overall conclusions of the meta-analysis were unaffected by the removal of individual studies.CONCLUSION Compared to patients with low HIF-1αexpression,those with high expression level had shorter OS and DFS.However,the prognostic significance of high HIF-1αexpression varies across molecularly stratified breast cancer cohorts needs to be further elucidated.
基金Supported by Comisión Sectorial de Investigación Científica(CSIC-Research Group 88180)The Agencia Nacional de Investigación e Innovación/Sistema Nacional de Investigadores(ANII/SNI)The Programa de Desarrollo de las Ciencias Básicas(PEDECIBA),Uruguay.
文摘BACKGROUND Hypoxia in oral cancer promotes tumoral invasion by inducing epithelial-mesenchymal transition,leading to aggressive tumor progression.AIM To characterize the expression of hypoxia-inducible factor 1-alpha(HIF-1α)at the invasive tumor front(ITF)in comparison to tumor islands(TI)in oral squamous cell carcinoma(OSCC)and to explore its relationship with E-cadherin and Vimentin expression.METHODS Thirty-eight cases of OSCC and five cases of normal oral mucosa(NOM)were included in this study.The ITF was identified based on the region and immune expression of AE1/AE3.Immunohistochemistry was performed to assess the expression of HIF-1α,Vimentin,and E-cadherin.The immunostaining was analyzed using an immunoreactive score,and the results were illustrated using immunofluorescence.RESULTS HIF-1αexpression was significantly higher in the TI region compared to the ITF region(P=0.0134).Additionally,a significant difference was observed between TI and NOM(P=0.0115).In the ITF regions,HIF-1αexpression showed a significant correlation with Vimentin expression,with higher levels of HIF-1αassociated with increased Vimentin expression(P=0.017).CONCLUSION Based on the results of this study,HIF-1αappears to play a distinct role in OSCC tumor progression,underscoring the importance of exploring hypoxia-driven changes in cellular phenotype at the ITF of OSCC.Further research is needed to better understand their impact on OSCC prognosis.
基金supported by NIH grants,Nos.R01NS125074,R01AG083164,R01NS107365,and R21NS127177(to YL),1F31NS129204-01A1(to KW)and Albert Ryan Fellowship(to KW).
文摘Adult neurogenesis continuously produces new neurons critical for cognitive plasticity in adult rodents.While it is known transforming growth factor-βsignaling is important in embryonic neurogenesis,its role in postnatal neurogenesis remains unclear.In this study,to define the precise role of transforming growth factor-βsignaling in postnatal neurogenesis at distinct stages of the neurogenic cascade both in vitro and in vivo,we developed two novel inducible and cell type-specific mouse models to specifically silence transforming growth factor-βsignaling in neural stem cells in(mGFAPcre-ALK5fl/fl-Ai9)or immature neuroblasts in(DCXcreERT2-ALK5fl/fl-Ai9).Our data showed that exogenous transforming growth factor-βtreatment led to inhibition of the proliferation of primary neural stem cells while stimulating their migration.These effects were abolished in activin-like kinase 5(ALK5)knockout primary neural stem cells.Consistent with this,inhibition of transforming growth factor-βsignaling with SB-431542 in wild-type neural stem cells stimulated proliferation while inhibited the migration of neural stem cells.Interestingly,deletion of transforming growth factor-βreceptor in neural stem cells in vivo inhibited the migration of postnatal born neurons in mGFAPcre-ALK5fl/fl-Ai9 mice,while abolishment of transforming growth factor-βsignaling in immature neuroblasts in DCXcreERT2-ALK5fl/fl-Ai9 mice did not affect the migration of these cells in the hippocampus.In summary,our data supports a dual role of transforming growth factor-βsignaling in the proliferation and migration of neural stem cells in vitro.Moreover,our data provides novel insights on cell type-specific-dependent requirements of transforming growth factor-βsignaling on neural stem cell proliferation and migration in vivo.
文摘Pulmonary arterial hypertension(PAH)is a progressive disease marked by degeneration of the lung’s blood vessels.As the disease progresses,the resistance to blood flow in the pulmonary arteries increases,putting a strain on the right side of the heart as it pumps blood through the lungs.PAH is characterized by changes in the structure of blood vessels and excessive cell growth.Untreated PAH leads to irreversible right-sided heart failure,often despite medical intervention.Patients experience a gradual decline in function until they are unable to perform daily activities.Advances in treatment have improved the prognosis for many PAH patients.Currently approved therapies target the prostacyclin,endothelin,nitric oxide,or phosphodiesterase pathways to slow the progression of the disease.To address the unmet need for effective PAH therapies,research efforts are focused on identifying new targets and developing therapies that specifically address the underlying disease mechanisms and restore vascular wall homeostasis.Among these,sotatercept,a fusion protein that targets the transforming growth factor-βsuperfamily signaling pathway,has emerged as a promising therapeutic option.In this review,we examine the available evidence from clinical trials to assess the potential of sotatercept as a treatment for PAH.
基金National Natural Science Foundation of China:Basic Research on the Mechanism of Organ Immune Damage and the Diagnosis and Treatment of Integrated Traditional Chinese and Western Medicine(No.32141005)。
文摘OBJECTIVE:To investigate the impact of Shenhua tablet(肾华片,SHT)on renal macrophage polarization and renal injury in mice with diabetic kidney disease(DKD)and to explore the potential mechanism involving the hypoxia-inducible factor-1α(HIF-1α)and pyruvate kinase M2(PKM2)signaling pathway,along with the glycolysis metabolism pathway.METHODS:The animals were divided into the following groups:Model,Control,dapagliflozin,SHT low-dose,SHT medium-dose,and SHT high-dose.We assessed 24-hour urine protein(24 h-UTP)levels,urinary albuminto-creatinine ratio,and regularly monitored fasting blood glucose during the treatment period.After treatment,we examined renal tissue structure,renal function(urea nitrogen,uric acid,creatinine,cystatin C,β2-microglobulin),and glycolysis in renal macrophages.Additionally,we observed macrophage polarization in renal tissue and measured inflammatory factors(tumor necrosis factor-α,interleukin-1β,interleukin-6,interleukin-10,monocyte chemoattractant protein-1)to assess the immunoinflammatory status of the renal tissue.Finally,we investigated the expression of the HIF-1α/PKM2 signaling pathway in macrophages to explore its role in the glycolysis process.RESULTS:SHT shows a beneficial effect in treating DKD by reducing 24 h-UTP,regulating blood glucose levels,improving renal tissue structure,protecting renal function,inhibiting macrophage glycolysis,reducing macrophage transformation to the M1 state,and suppressing the expression of the HIF-1α/PKM2 signaling pathway.CONCLUSION:SHT may exert renoprotective effects by inhibiting macrophage glycolysis via the HIF-1α/PKM2 signaling pathway.This inhibition decreases macrophage M1 polarization and reduces immunoinflammatory injury in the renal tissue of DKD mice.
基金supported by the National Natural Science Foundation of China(81473453,81673800)the Projects of International Science and Technology Cooperation in Henan(182102410084).
文摘Objective MicroRNA-1(miR-1)aggravates myocardial ischemia–reperfusion(I/R)injury,whereas insulin-like growth factor-1(IGF-1)maintains cardiomyocyte homeostasis.In this study,the aim is to investigate whether miR-1 can exacerbate I/R injury through the regulation of IGF-1.Methods The infarct area,lactate dehydrogenase,miR-1 level,and apoptosis level were examined in the Langendorff isolated rat I/R model.The hypoxia–reoxygenation model of rat cardiacmyocytes and H9c2 cells were developed to determine the levels of miR-1,IGF-1 mRNA,and IGF-1 protein.Furthermore,the dual-luciferase assay was used to verify the relationship between miR-1 and IGF-1.Results Overexpression of miR-1 increased the level of apoptosis and decreased the IGF-1 expression.However,inhibition of miR-1 expression decreased the level of apoptosis,alleviated the degree of injury,and increased the IGF-1 expression.Overexpression of IGF-1 also reduced the degree of cellular damage and level of apoptosis caused by the overexpression of miR-1.When IGF-1 was knocked down,myocardial cells displayed more severe damage and a higher apoptosis level,even with decreased levels of miR-1.Conclusion miR-1 promotes apoptosis and aggravates I/R injury by downregulating IGF-1.
文摘It has been well documented that bone morphogenetic .proteins (BMPs), a group of proteins belonging to the transforming growth factor-β (TGFβ) superfamily, can induce bone formation, both in vivo and in vitro. Bone morphogenetic protein-2 (BMP2) is a potent osteoinductive factor and is being evaluated as a bone growth inducer for orthopedic applications.1 Vascular endothelial growth factor (VEGF), the best-characterized angiogenic factor,
基金supported by the National Natural Science Foundation of China,Nos.31971277 and 31950410551(both to DY)。
文摘Injuries caused by trauma and neurodegenerative diseases can damage the peripheral nervous system and cause functional deficits.Unlike in the central nervous system,damaged axons in peripheral nerves can be induced to regenerate in response to intrinsic cues after reprogramming or in a growth-promoting microenvironment created by Schwann cells.However,axon regeneration and repair do not automatically result in the restoration of function,which is the ultimate therapeutic goal but also a major clinical challenge.Transforming growth factor(TGF)is a multifunctional cytokine that regulates various biological processes including tissue repair,embryo development,and cell growth and differentiation.There is accumulating evidence that TGF-βfamily proteins participate in peripheral nerve repair through various factors and signaling pathways by regulating the growth and transformation of Schwann cells;recruiting specific immune cells;controlling the permeability of the blood-nerve barrier,thereby stimulating axon growth;and inhibiting remyelination of regenerated axons.TGF-βhas been applied to the treatment of peripheral nerve injury in animal models.In this context,we review the functions of TGF-βin peripheral nerve regeneration and potential clinical applications.
基金Supported by Health Commission of Qinghai Province,No.2021-wjzdx-18.
文摘BACKGROUND Pancreatic cancer(PC)is associated with some of the worst prognoses of all major cancers.Thymoquinone(TQ)has a long history in traditional medical practice and is known for its anti-cancer,anti-inflammatory,anti-fibrosis and antioxidant pharmacological activities.Recent studies on hypoxia-inducible factor-1α(HIF-1α)and PC have shown that HIF-1αaffects the occurrence and development of PC in many aspects.In addition,TQ could inhibit the development of renal cancer by decreasing the expression of HIF-1α.Therefore,we speculate whether TQ affects HIF-1αexpression in PC cells and explore the mechanism.AIM To elucidate the effect of TQ in PC cells and the regulatory mechanism of HIF-1αexpression.METHODS Cell counting kit-8 assay,Transwell assay and flow cytometry were performed to detect the effects of TQ on the proliferative activity,migration and invasion ability and apoptosis of PANC-1 cells and normal pancreatic duct epithelial(hTERTHPNE)cells.Quantitative real-time polymerase chain reaction and western blot assay were performed to detect the expression of HIF-1αmRNA and protein in PC cells.The effects of TQ on the HIF-1αprotein initial expression pathway and ubiquitination degradation in PANC-1 cells were examined by western blot assay and co-immunoprecipitation.RESULTS TQ significantly inhibited proliferative activity,migration,and invasion ability and promoted apoptosis of PANC-1 cells;however,no significant effects on hTERT-HPNE cells were observed.TQ significantly reduced the mRNA and protein expression levels of HIF-1αin PANC-1,AsPC-1,and BxPC-3 cells.TQ significantly inhibited the expression of the HIF-1αinitial expression pathway(PI3K/AKT/mTOR)related proteins,and promoted the ubiquitination degradation of the HIF-1αprotein in PANC-1 cells.TQ had no effect on the hydroxylation and von Hippel Lindau protein mediated ubiquitination degradation of the HIF-1αprotein but affected the stability of the HIF-1αprotein by inhibiting the interaction between HIF-1αand HSP90,thus promoting its ubiquitination degradation.CONCLUSION The regulatory mechanism of TQ on HIF-1αprotein expression in PC cells was mainly to promote the ubiquitination degradation of the HIF-1αprotein by inhibiting the interaction between HIF-1αand HSP90;Secondly,TQ reduced the initial expression of HIF-1αprotein by inhibiting the PI3K/AKT/mTOR pathway.