Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
1.
Adv Biol (Weinh) ; 8(1): e2300292, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37786269

ABSTRACT

Type A aortic dissection (AD) is a catastrophic cardiovascular disease. Hypoxia-inducible factor-1α (HIF-1α) and autophagy are reported to be upregulated in the AD specimens. However, the interaction between HIF-1α and autophagy in the pathogenesis of AD remains to be explored. HIF-1α and LC3 levels are evaluated in 10 AD and 10 normal aortic specimens. MDC staining, autophagic vacuoles, and autophagic flux are detected in human aortic smooth muscle cells (HASMCs) under hypoxia treatment. CCK-8, transwell, and wound healing assay are used to identify proliferation and migration under hypoxia treatment. Furthermore, 3-MA is used to inhibit autophagy in hypoxia-treated HASMCs. This study reveals that AD tissues highly express HIF-1α and the LC3. Autophagy is induced under hypoxia in a time-dependent manner, and autophagy is positively related to HIF-1α in HASMCs. Moreover, the proliferation and migration of HASMCs are enhanced by hypoxia, whereas the knockdown of HIF-1α attenuates this effect. Additionally, inhibiting autophagy with 3-MA ameliorates hypoxia-induced proliferation and migration of HASMCs. In summary, the above results indicate that HIF-1α facilitates HASMC proliferation and migration by upregulating autophagy in a hypoxic microenvironment. Thus, inhibition of autophagy may be a novel therapeutic target for the prevention and treatment of AD.


Subject(s)
Aortic Dissection , Muscle, Smooth, Vascular , Humans , Autophagy , Cell Proliferation , Hypoxia , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/pharmacology
3.
Oxid Med Cell Longev ; 2023: 4365895, 2023.
Article in English | MEDLINE | ID: mdl-36778206

ABSTRACT

Testes produce sperms, and gamete generation relies on a proper niche environment. The disruption of hierarchical regulatory homeostasis in Leydig or Sertoli cells may evoke a sterile phenotype in humans. In this study, we recapitulated type 2 diabetes mellitus by using a high-fat diet- (HFD-) fed mouse model to identify the phenotype and potential mechanism of diabetes-induced testicular impairment. At the end of the study, blood glucose levels, testosterone structure, testicular antioxidant capacity, and testosterone level and the expression of hypoxia-inducible factor- (HIF-) 1α, apoptosis-related protein cleaved-caspase3, and autophagy-related proteins such as LC3I/II, p62, and Beclin1 were evaluated. We found that long-term HFD treatment causes the development of diabetes mellitus, implicating increased serum glucose level, cell apoptosis, and testicular atrophy (P < 0.05 vs. Ctrl). Mechanistically, the results showed enhanced expression of HIF-1α in both Sertoli and Leydig cells (P < 0.05 vs. Ctrl). Advanced glycation end products (AGEs) were demonstrated to be a potential factor leading to HIF-1α upregulation in both cell types. In Sertoli cells, high glucose treatment had minor effects on Sertoli cell autophagy. However, AGE treatment stagnated the autophagy flux and escalated cell apoptosis (P < 0.05 vs. Ctrl+Ctrl). In Leydig cells, high glucose treatment was adequate to encumber autophagy induction and enhance oxidative stress. Similarly, AGE treatment facilitated HIF-1α expression and hampered testosterone production (P < 0.05 vs. Ctrl+Ctrl). Overall, these findings highlight the dual effects of diabetes on autophagy regulation in Sertoli and Leydig cells while imposing oxidative stress in both cell types. Furthermore, the upregulation of HIF-1α, which could be triggered by AGE treatment, may negatively affect both cell types. Together, these findings will help us further understand the molecular mechanism of diabetes-induced autophagy dysregulation and testicular impairment, enriching the content of male reproductive biology in diabetic patients.


Subject(s)
Diabetes Mellitus, Type 2 , Testis , Mice , Animals , Humans , Male , Oxidative Stress , Autophagy , Testosterone , Glucose/pharmacology , Hypoxia-Inducible Factor 1, alpha Subunit/pharmacology
4.
Oxid Med Cell Longev ; 2023: 5157417, 2023.
Article in English | MEDLINE | ID: mdl-36819784

ABSTRACT

Enriched environment (EE) has been proven to be an effective intervention strategy which can improve neurofunctional recovery following cerebral ischemia/reperfusion (I/R) injury. However, it still needs further investigation for the underlying mechanisms. Recently, it has been shown that ferroptosis played an essential role in the pathophysiological development of ischemic stroke (IS). This study is aimed at investigating whether EE plays a neuroprotective role by attenuating ferroptosis after cerebral I/R injury. We used middle cerebral artery occlusion/reperfusion (MCAO/R) to build a model of cerebral I/R injury. To evaluate the effect of EE on neurological recovery, we used the modified neurological severity score (mNSS) and the Morris water maze (MWM). We used the western blot to detect the protein levels of glutathione peroxidase 4 (GPX4), hypoxia-inducible factor-1α (HIF-1α), and acyl-CoA synthetase long-chain family member 4 (ACSL4). We used the quantitative real-time PCR (qRT-PCR) to measure the mRNA levels of ACSL4 and inflammatory cytokines including tumor necrosis factor alpha (TNFα), interleukin-6 (IL-6), and interleukin 1 beta (IL-1ß). The occurrence of ferroptosis was detected by TdT-mediated dUTP nick-end labeling (TUNEL) assay, diaminobenzidine- (DAB-) enhanced Perls' staining, iron level assays, and malondialdehyde (MDA) level assays. The results verified that EE enhanced functional recovery and attenuated ferroptosis and neuroinflammation after cerebral I/R injury. EE increased the expression of HIF-1α while inhibited the expression of ACSL4. Our research indicated that EE improved functional recovery after cerebral I/R injury through attenuating ferroptosis, and this might be related to its regulation of the neuroinflammation and HIF-1α-ACSL4 pathway.


Subject(s)
Brain Ischemia , Ferroptosis , Reperfusion Injury , Humans , Brain Ischemia/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/pharmacology , Infarction, Middle Cerebral Artery , Neuroinflammatory Diseases , Reperfusion Injury/metabolism , Coenzyme A Ligases/metabolism
5.
Biol Trace Elem Res ; 201(11): 5230-5241, 2023 Nov.
Article in English | MEDLINE | ID: mdl-36710293

ABSTRACT

Fluorosis can induce neurotoxicity. Sodium butyrate (SB), a histone deacetylase inhibitor, has important research potential in correcting glucose metabolism disorders and is widely used in a variety of neurological diseases and metabolic diseases, but it is not yet known whether it plays a role in combating fluoride-induced neurotoxicity. This study aims to evaluate the effect of SB on fluoride neurotoxicity and the possible associated mechanisms. The results of HE staining and Morris water maze showed that, in mice exposed to 100 mg/L fluoride for 3 months, the hippocampal cells arranged in loosely with large cell gaps and diminished in number. One thousand milligram per kilogram per day SB treatment improved fluoride-induced neuronal cell damage and spatial learning memory impairment. Western blot results showed that the abundance of malate dehydrogenase 2 (MDH2) and pyruvate dehydrogenase (PDH) in the hippocampus of fluorosis mice was increased, the abundance of pyruvate kinase M (PKM), lactate dehydrogenase (LDH), hexokinase (HK), phosphatidylinositol 3-kinase (PI3K), phosphorylated Akt (P-AKT), and hypoxia-inducible factor 1α (HIF-1α) was inhibited, and the content of lactate and ATP was decreased. SB treatment reversed the decreased glycolysis in the hippocampus of fluorosis mice. These results suggested that SB could ameliorate fluorosis-induced neurotoxicity, which might be linked with its function in regulating glycolysis as well as inhibition of the PI3K/AKT/HIF-1α pathway. Sodium butyrate ameliorates fluorosis-induced neurotoxicity by regulating hippocampal glycolysis in vivo (created with MedPeer (www.medpeer.cn)).


Subject(s)
Proto-Oncogene Proteins c-akt , Signal Transduction , Animals , Mice , Proto-Oncogene Proteins c-akt/metabolism , Butyric Acid/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Fluorides/pharmacology , Glycolysis , Phosphatidylinositol 3-Kinase/metabolism , Lactic Acid/metabolism , Hippocampus/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/pharmacology
6.
Biol Trace Elem Res ; 201(5): 2284-2293, 2023 May.
Article in English | MEDLINE | ID: mdl-35715716

ABSTRACT

Arsenic is a common environmental pollutant that can cause damage to multiple systems and organs in the body. The lungs are particularly sensitive to arsenic exposure, and respiratory disease is thought to be the leading cause of death from arsenic poisoning. Our previous study found that human bronchial epithelial (HBE) cells treated with NaAsO2 exhibited mitochondrial dysfunction accompanied by elevated HIF-1α; however, the molecular mechanism was unclear. The aim of the current study was to confirm the role of HIF-1α in arsenic-induced mitochondrial damage. The results of this study indicated that NaAsO2 treatment induced mitochondrial ultrastructure impairment and depolarization of the mitochondrial membrane potential. Furthermore, NaAsO2 induced a significant decrease in basal respiration, maximal respiration, spare respiratory capacity, ATP (adenosine-triphosphate)-associated mitochondrial respiration and proton leakage in HBE cells (P < 0.05), while promoting an increase in ECAR (extracellular acidification rate) values. To clarify the role of HIF-1α, the effect of HIF-1α siRNA on NaAsO2-induced glycolysis in HBE cells was examined, and the results showed that HIF-1α siRNA reversed the NaAsO2-induced elevation in PKM2 (Tyr105), HIF-1α, GLUT1 and HK2 protein expression and decreased the NaAsO2-mediated glycolysis level, glycolytic capacity and glycolytic reserve. These findings suggest that targeting metabolic dysregulation has significant implications for targeting arsenic-induced lung injury and that HIF-1α is an exciting new therapeutic target for the treatment of arsenic-induced lung injury.


Subject(s)
Arsenic , Lung Injury , Humans , Arsenic/toxicity , Epithelial Cells , Lung , RNA, Small Interfering/pharmacology , Hypoxia-Inducible Factor 1, alpha Subunit/pharmacology
7.
Vet Comp Oncol ; 21(1): 12-19, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36054173

ABSTRACT

The transcription factor hypoxia-inducible factor 1α (HIF-1α) is activated in response to oxygen deficiency, and is expressed in several cancers under intratumoral hypoxic stress that arises during pathogenic processes. Hypoxic stimulation enhanced the growth potential of canine lymphoma cells by activating the HIF-1α signalling pathway in a previously reported study. The aim of this study was to establish a molecular design strategy for a novel hypoxia-targeting therapy for intestinal T-cell lymphoma (ITL) in dogs. We assessed the relationship between immunohistochemistry-based HIF-1α expression and clinical information, including signalment, tumour area, clinical signs, systemic diseases, treatment protocol, follow-up information, chemotherapy response and overall survivals (OS), using 48 tissue samples from dogs with ITL. We investigated the effects of hypoxic stimulation on the biological behaviour of cell lines from three different types of canine ITL. We assessed the effects of evofosfamide (Evo; hypoxia-activated prodrug) on cell lines cultured under hypoxic conditions. Our data showed that treatment response and overall survival might be significantly decreased in dogs with higher HIF-1α expression than in those with lower HIF-1α expression. Hypoxic culture (1% O2 , 72 h) enhanced the invasiveness of cell lines and decreased their sensitivity to CCNU, resulting in hypoxia-dependent aggressive behaviour. Sensitivity to Evo significantly increased in cell lines cultured under hypoxia compared with those cultured under normoxia, which exhibited hypoxia-dependent apoptosis. Additionally, Evo downregulated HIF-1α expression in cell lines cultured under hypoxia, suggesting that Evo might inhibit cell growth by inactivating HIF-1α-dependent cell signalling. Our results revealed the preclinical antitumor activity of Evo and provide a rationale for treatment strategies for dogs with ITL.


Subject(s)
Dog Diseases , Lymphoma, T-Cell , Dogs , Animals , Cell Hypoxia/physiology , Dog Diseases/drug therapy , Hypoxia/veterinary , Cell Line , Lymphoma, T-Cell/veterinary , Hypoxia-Inducible Factor 1, alpha Subunit/pharmacology , Cell Line, Tumor
8.
Molecules ; 27(21)2022 Oct 24.
Article in English | MEDLINE | ID: mdl-36364008

ABSTRACT

Prostate cancer (PCa) is the most common malignancy to endanger the health of male genitourinary system. Clinically, paclitaxel (PTX) (C47H51NO14), a diterpene alkaloid, is commonly used as an effective natural antineoplastic drug during the treatment of PCa. However, the mechanism and pathway involved in the function of PTX are poorly understood. In the current study, we employed the CCK-8 assay, revealing that PTX can inhibit the survival and induce the apoptosis of PC3M cells (a human prostate cancer cell line) in a concentration-dependent manner. Reactive oxygen species (ROS), as a metabolic intermediate produced by the mitochondrial respiratory chain, are highly accumulated under the PTX treatment, which results in a sharp decrease of the mitochondrial membrane potential in PC3M cells. Additionally, the migration and invasion of PC3M cells are weakened due to PTX treatment. Further analysis reveals that N-acetylcysteine (NAC), which functions as an antioxidant, not only rescues the decreased mitochondrial membrane potential induced by the abnormal ROS level, but also restores the migration and invasion of PC3M cells. In a subsequent exploration of the detailed mechanism, we found that hypoxia-inducible factor (HIF)-1α works as a downstream gene that can respond to the increased ROS in PC3M cells. Under PTX treatment, the expression levels of HIF-1α mRNA and protein are significantly increased, which stimulate the activation of JNK/caspase-3 signaling and promote the apoptosis of PC3M cells. In summary, we demonstrate that PTX regulates the expression of HIF-1α through increased ROS accumulation, thereby promoting the activation of JNK/caspase-3 pathway to induce the apoptosis of PCa cells. This study provides new insights into the mechanism of antineoplastic action of taxanes and unveils the clinical benefit of the ROS-HIF-1α signaling pathway, which may offer a potential therapeutic target to prevent the development of PCa.


Subject(s)
Antineoplastic Agents , Prostatic Neoplasms , Male , Humans , Reactive Oxygen Species/metabolism , Paclitaxel/pharmacology , Caspase 3/genetics , Caspase 3/pharmacology , Apoptosis , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/pharmacology , Antineoplastic Agents/pharmacology
9.
Toxicology ; 480: 153324, 2022 10.
Article in English | MEDLINE | ID: mdl-36115646

ABSTRACT

Trichothecene mycotoxins have a strong immunosuppressive effect, which may even escape host immune surveillance and damage the immune repair to show an "immune evasion" effect. Increasing lines of evidence have shown that hypoxia and hypoxia-inducible factors (HIFs) are key mediators of trichothecenes, and these toxins appear to be closely related to the "immune evasion" mechanisms. Therefore, we used RAW264.7 cell model to explore the association of T-2 toxins with "immune evasion" process and hypoxia, as well as their cross-linking effects induced by T-2 toxin. Our results showed that HIF-1α is an important toxicity target of T-2 toxin, which could induce intracellular hypoxia. T-2 toxin induced an "immune evasion" process by activating the PD-1/PD-L1 signaling pathway. Interestingly, when HIF-1α activation was blocked, the "immune evasion" process regulated by PD-1/PD-L1 signaling was activated, resulting in the cells damage, suggesting that hypoxia induced by T-2 toxin plays a protective role for RAW264.7 cell damage. Thus, our work shows that HIF-1α inhibits T-2 toxin-mediated "immune evasion" process by negatively regulating PD-1/PD-L1signaling. This study contributes to a better understanding of the immunotoxicity mechanism of trichothecenes.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/pharmacology , T-2 Toxin , Trichothecenes , B7-H1 Antigen/metabolism , Humans , Hypoxia , Programmed Cell Death 1 Receptor/metabolism , T-2 Toxin/toxicity , Trichothecenes/toxicity
10.
Vet Med Sci ; 8(5): 2223-2229, 2022 09.
Article in English | MEDLINE | ID: mdl-36044612

ABSTRACT

BACKGROUND: Hypoxia-inducible factor-1α (HIF-1α), vascular endothelial growth factor (VEGF) and transforming growth factor ß1 (TGF-ß1 ) are multifunctional growth factors that play an important role in follicular growth and development. However, its biological function in the follicular development of Tibetan sheep at different stages has not been described. OBJECTIVES: The purpose of this study was to investigate the effect of VEGF, TGF-ß1 and HIF-1α expression and distribution on the development of follicles of different sizes. METHODS: Immunohistochemistry (IHC), western blot (WB) and quantification real-time polymerase chain reaction (qRT-PCR) were used to detect the localisation and quantitative expression of VEGF, TGF-ß1 and HIF-1α proteins and mRNA in small- (< 3 mm), medium- (3 mm < diameter < 5 mm)-, and large- (> 5 mm) sized follicles. RESULTS: The results showed that the proteins VEGF, TGF-ß1 and HIF-1α, as well as their mRNA, were expressed in follicles. However, the expression in medium-sized follicles was significantly higher than that in large- and small-sized follicles (p <0.05). IHC also showed that the proteins VEGF, TGF-ß1 , and HIF-1α were distributed in granulosa cells (GCs) in small-, medium-, and large-sized follicles. CONCLUSIONS: This study indicates that VEGF, TGF-ß1 and HIF-1α, which operate in an autocrine or paracrine manner with the GCs, influence the follicular progressive growth, suggesting that these growth factors are closely associated with the follicular growth and development in ovarian.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit , Vascular Endothelial Growth Factor A , Animals , Female , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/pharmacology , Ovarian Follicle , RNA, Messenger/metabolism , Sheep , Tibet , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/pharmacology , Vascular Endothelial Growth Factors/metabolism , Vascular Endothelial Growth Factors/pharmacology
11.
Oral Oncol ; 131: 105940, 2022 08.
Article in English | MEDLINE | ID: mdl-35689951

ABSTRACT

BACKGROUND: This study aimed to investigate the potential role of nidogen 1 (NID1), a basement membrane component, in the growth and metastasis of salivary gland adenoid cystic carcinoma (SACC) and the underlying molecular mechanism. METHODS: High-throughput next-generation sequencing was used to compare the gene expression profiles of SACC with and without lung metastasis. Luciferase gene reporter assays were used to measure the NID1 promoter activity. BALB/c nude mice were used to establish a lung metastasis model of SACC to evaluate the prometastatic activity of NID1. ChIP and dual-luciferase reporter assays were performed to confirm the HIF-1α-binding site in the NID1 promoter. RESULTS: NID1 expression in SACC was significantly increased and associated with lung metastasis (P = 0.011). The elevated NID1 expression was a predictor of poor outcomes in patients with SACC (P < 0.05). Overexpression of NID1 promoted cancer cell migration and invasion through PI3K/AKT pathway activation and subsequent epithelial-mesenchymal transition (EMT), as indicated by the upregulation of N-cadherin and vimentin. Furthermore, in vivo live monitoring of a mouse model of lung cancer demonstrated the pro-metastatic role of NID1 in SACC cell lung metastasis. Hypoxia-inducible factor 1α (HIF-1α) upregulation via transfection of an HIF-1α-overexpressing plasmid enhanced HIF-1α binding to the NID1 promoter and the subsequent transcriptional activity and expression of NID1. CONCLUSION: HIF-1α-activated NID1 overexpression promotes SACC cell metastasis via PI3K/AKT pathway activation and EMT. Thus, NID1 could be a novel biomarker and therapeutic target for preventing metastasis and treating patients with SACC in future.


Subject(s)
Carcinoma, Adenoid Cystic , Lung Neoplasms , Salivary Gland Neoplasms , Animals , Carcinoma, Adenoid Cystic/pathology , Cell Line, Tumor , Cell Movement/genetics , Epithelial-Mesenchymal Transition/physiology , Gene Expression Regulation, Neoplastic , Hypoxia-Inducible Factor 1, alpha Subunit/pharmacology , Lung Neoplasms/secondary , Membrane Glycoproteins , Mice , Mice, Nude , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Salivary Gland Neoplasms/pathology , Salivary Glands/metabolism , Signal Transduction
12.
Am J Transplant ; 22(10): 2323-2336, 2022 10.
Article in English | MEDLINE | ID: mdl-35729702

ABSTRACT

The mammalian target of rapamycin (mTOR) pathway is frequently deregulated and has critical roles in cancer progression. mTOR inhibitor has been widely used in several kinds of cancers and is strongly recommended in patients with hepatocellular carcinoma (HCC) after liver transplantation (LT). However, the poor response to mTOR inhibitors due to resistance remains a challenge. Hypoxia-associated resistance limits the therapeutic efficacy of targeted drugs. The present study established models of HCC clinical samples and cell lines resistance to mTOR inhibitor sirolimus and screened out E2F7 as a candidate gene induced by hypoxia and promoting sirolimus resistance. E2F7 suppressed mTOR complex 1 via directly binding to the promoter of the TSC1 gene and stabilizes hypoxia-inducible factor-1α activating its downstream genes, which are responsible for E2F7-dependent mTOR inhibitor resistance. Clinically, low E2F7 expression could be an effective biomarker for recommending patients with HCC for anti-mTOR-based therapies after LT. Targeting E2F7 synergistically inhibited HCC growth with sirolimus in vivo. E2F7 is a promising target to reverse mTOR inhibition resistance. Collectively, our study points to a role for E2F7 in promoting mTOR inhibitor resistance in HCC and emphasizes its potential clinical significance in patients with HCC after LT.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Liver Transplantation , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/surgery , Cell Line, Tumor , Cell Proliferation , E2F7 Transcription Factor , Humans , Hypoxia/drug therapy , Hypoxia-Inducible Factor 1, alpha Subunit/pharmacology , Hypoxia-Inducible Factor 1, alpha Subunit/therapeutic use , Liver Neoplasms/drug therapy , Liver Neoplasms/surgery , MTOR Inhibitors , Sirolimus/pharmacology , TOR Serine-Threonine Kinases
13.
J Cardiovasc Transl Res ; 15(5): 1010-1023, 2022 10.
Article in English | MEDLINE | ID: mdl-35377130

ABSTRACT

Abnormal function of endothelial cells (ECs) is an important reason for vascular endothelial remodeling and atherosclerotic plaque formation in patients with atherosclerosis (AS). Here, we report for the first time that the vascular ECs with apoptosis resistance phenotype (ARECs) exist in peripheral blood of AS patients. Our research data showed that the switch of regulation modes between HIF-1α and Bax operated by lncRNA-ASLNC18810 is the direct cause for the formation of ARECs. When ASLNC18810 is low or missing, HIF-1α indirectly negatively regulates the Bax in post-transcription through HIF-1α/miR-559/Bax pathway which makes ECs acquire apoptosis resistance and form ARECs. The functional experiments results showed that ASLNC18810 could effectively eliminate the anti-apoptotic properties of ARECs by blocking the HIF-1α/miR559/Bax pathway and maintaining HIF-1α/Bax pathway. In a word, our study shows that ASLNC18810 has full potential to become a biological target for the prevention and treatment of atherosclerotic plaques by regulating ARECs. ASLNC18810 was significantly upregulated in ECs compared to ARECs. With high level of ASLNC18810 in ECs, ASLNC18810 binds to miR-559 as a miRNA sponge and suppresses the inhibition effect of miR-559 on Bax protein, this direct positive transcriptional regulation between HIF-1α and Bax endows the apoptotic property in ECs induced by Ox-LDL. However, with low expression of ASLNC18810 in ARECs, the post-transcriptional regulation of Bax by miR-559 dominates and the indirect negative regulation between HIF-1α and Bax endows the anti-apoptotic property of ARECs. To sum up, low ASLNC18810 expression-mediated switching of HIF-1α/Bax pathway to HIF-1α/miR-559/Bax pathway is the internal reason for ECs to obtain apoptosis resistance and the formation of ARECs under the ox-LDL induction.


Subject(s)
Atherosclerosis , MicroRNAs , Plaque, Atherosclerotic , RNA, Long Noncoding , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Endothelial Cells/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism , Atherosclerosis/genetics , Atherosclerosis/metabolism , Apoptosis , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/pharmacology
14.
J Cardiovasc Transl Res ; 15(4): 772-785, 2022 08.
Article in English | MEDLINE | ID: mdl-35194735

ABSTRACT

Cardiac ischemia-reperfusion injury usually results in acute myocardial infarction (AMI). MiRNAs have been identified as key regulators of AMI. This study was carried out to investigate the effect of miR-27-3p on cardiomyocyte injury in AMI. CCK-8 and flow cytometry assays were used to evaluate cell viability and apoptosis. The expression levels of miR-27-3p, galectin-3, and hypoxia-inducible factor-1α were measured by qRT-PCR. The relationship among miR-27-3p, galectin-3, and HIF-1α was assessed by bioinformatics analysis and luciferase assay. The effects of miR-27-3p and/or galectin-3 and HIF-1α on the inhibition of cell viability and apoptosis induced by H/R were explored. The expression levels of apoptosis-related proteins were determined by Western blot analysis. The expression levels of miR-27-3p were reduced in both ischemia-reperfusion myocardium and HL-1 cells during hypoxia. Overexpression of miR-27-3p reduced I/R-induced myocardial injury, and HIF-1α can reduce this effect. H/R reduced the expression levels of miR-27-3p in HL-1 cardiomyocytes, and HIF1-α reduced this effect, indicating that HIF1-α could regulate the expression of miR-27-3p, and galectin-3 was a target of miR-27-3p. Finally, overexpression of galectin-3 reduced the protective effect of miR-27-3p on cardiomyocyte injury. The expression levels of HIF1-α were increased, and miR-27-3p was downregulated after AMI. HIF-1α promoted myocardial protection by upregulating miR-27-3p, and downregulation of miR-27-3p promoted myocardium cell injury by targeting galectin-3.


Subject(s)
MicroRNAs , Myocardial Infarction , Reperfusion Injury , Humans , Galectin 3/genetics , Galectin 3/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/pharmacology , Reperfusion Injury/metabolism , Myocytes, Cardiac/metabolism , Apoptosis , MicroRNAs/genetics , MicroRNAs/metabolism , Myocardium/metabolism , Apoptosis Regulatory Proteins/metabolism , Myocardial Infarction/genetics , Myocardial Infarction/prevention & control , Myocardial Infarction/metabolism , Reperfusion
15.
J Biomed Nanotechnol ; 18(1): 112-119, 2022 Jan 01.
Article in English | MEDLINE | ID: mdl-35180904

ABSTRACT

Bone marrow mesenchymal stem cells (BMSC) have the ability to multi polarize with multiple tropisms and participate in tissue remodeling. This study assessed the effect of titanium dioxide nanotubes with different diameters on ossification of BMSC cells and HIF-1α expression in BMSC ossification. Titanium dioxide nanotubes with different diameters were prepared and then the following groups were set according to the size of pressure; Ti group, NT10 group, NT30 group, and NT60 group. Analysis of cell morphology was done by fluorescence microscope, while adhesion and proliferation were assessed by MTT assay. Moreover, ALP activity, collagen secretion and outer matrix mineralization and expression of HIF-1α, VEGF, and TWIST were assessed by RT-PCR and Western blot. The P3 generation of BMSC cells was successfully obtained. Three types of nanotubes were arranged regularly and contact angle showed NT60Ti>NT30>NT60 (P < 0.05). Cells from NT30 and N60 groups showed obvious expansion with pseudopodia and pseudo plates of cells. Cell adhesion showed changes in sizes of NT10>Ti>NT30>NT66. NT60 group showed lower cell proliferation and higher ALP activity and collagen secretion than the other groups. NT30 and NT60 group presented higher mineralization level, larger diameter, and higher degree of promotion. The NT30 group presented lowest content of HIF-1α (0.12 ± 0.03), VEGF (0.013 ± 0.004), and TWIST (0.014 ± 0.003). Inoculation of BMSCs on titanium dioxide nanotubes of different diameters under cyclical tensile stress environment can promote growth of BMSC cells in a diameter-dependent manner.


Subject(s)
Mesenchymal Stem Cells , Nanotubes , Bone Marrow Cells/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/pharmacology , Osteogenesis , Titanium/pharmacology
16.
J Hepatol ; 76(1): 160-173, 2022 01.
Article in English | MEDLINE | ID: mdl-34599999

ABSTRACT

BACKGROUND & AIMS: An invasive form of intestinal Entamoeba (E.) histolytica infection, which causes amoebic liver abscess, is more common in men than in women. Immunopathological mechanisms are responsible for the more severe outcome in males. Here, we used a mouse model of hepatic amoebiasis to investigate the contribution of hepatic hypoxia-inducible factor (HIF)-1α to T helper 17 (Th17)/regulatory T cell (Treg) responses in the context of the sex-specific outcome of liver damage. METHODS: C57BL/6J mice were infected intrahepatically with E. histolytica trophozoites. HIF-1α expression was determined by qPCR, flow cytometry and immunohistochemistry. Tregs and Th17 cells were analysed by immunohistochemistry and flow cytometry. Finally, male and female hepatocyte-specific Hif1α knockout mice were generated, and the effect of HIF-1α on abscess development, the cytokine milieu, and Th17/Treg differentiation was examined. RESULTS: E. histolytica infection increased hepatic HIF-1α levels, along with the elevated frequencies of hepatic Th17 and Treg cells. While the Th17 cell population was larger in male mice, Tregs characterised by increased expression of Foxp3 in female mice. Male mice displayed increased IL-6 expression, contributing to immunopathology; this increase in IL-6 expression declined upon deletion of hepatic HIF-1α. In both sexes, hepatic deletion of HIF-1α reduced the Th17 cell frequency; however, the percentage of Tregs was reduced in female mice only. CONCLUSIONS: Hepatic HIF-1α modulates the sex-specific outcome of murine E. histolytica infection. Our results suggest that in male mice, Th17 cells can be modulated by hepatic HIF-1α via IL-6, indicating marked involvement in the immunopathology underlying abscess development. Strong expression of Foxp3 by hepatic Tregs from female mice suggests a potent immunosuppressive function, leading to initiation of liver regeneration. LAY SUMMARY: Infection with the parasite Entamoeba histolytica activates immunopathological mechanisms in male mice, which lead to liver abscesses that are larger than those in female mice. In the absence of the protein HIF-1α in hepatocytes, abscess formation is reduced; moreover, the sex difference in abscess size is abolished. These results suggest that HIF-1α modulates the immune response involved in the induction of immunopathology, resulting in differential disease susceptibility in males and females.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/pharmacology , Liver Abscess, Amebic/genetics , Th17 Cells/metabolism , Animals , Disease Models, Animal , Entamoeba/drug effects , Entamoeba/pathogenicity , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Liver Abscess, Amebic/drug therapy , Mice , Mice, Inbred C57BL , Th17 Cells/microbiology
17.
Int J Mol Sci ; 22(21)2021 Oct 21.
Article in English | MEDLINE | ID: mdl-34768786

ABSTRACT

Intervertebral discs (IVDs) have poor nutrient diffusion, because the nucleus pulposus (NP) lacks direct vascular supply and likely generates adenosine triphosphate by anaerobic glycolysis. Regulation of glycolysis is mediated by hypoxia-inducible factor-1α (HIF-1α), a transcription factor that responds to local oxygen tension. Constitutively active HIF-1α (CA HIF-1α) was created by point mutation and determined the protective role of HIF-1α in IVD degeneration. Under fluoroscopy, rat caudal IVD segments were stabbed by a needle puncture, and pcDNA3- HIF-1α wild-type (WT) or pcDNA3-CA HIF-1α was transfected into NP cell lines. The constitutive activity of CA HIF-1α was analyzed using a luciferase assay after cell lysis. Next, IVD tissue samples were retrieved from five patients with degenerative lumbar spinal stenosis at the time of surgery, and NP cells were cultured. NP cells were transfected with CA HIF-1α, and relevant gene expression was measured. HIF-1α protein levels in the nucleus were significantly higher, and transcriptional activity was 10.3-fold higher in NP cells with CA HIF-1α than in those with HIF-1α WT. Gene transfer of CA HIF-1α into NP cells enhanced the expression of Glut-1, Glut-3, aggrecan, type II collagen, and Sox9. Moreover, CA HIF-1α reduced the apoptosis of NP cells induced by the Fas ligand. The HIF-1α and collagen 2 expression levels were notably increased in the NP cells of the CA HIF-1α transfected segments in histology and immunohistochemistry study. Collectively, these results suggest that activation of HIF-1α signaling pathway may play a protective role against IVD degeneration and could be used as a future therapeutic agent.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Intervertebral Disc Degeneration/prevention & control , Animals , Cell Line , Collagen Type II/metabolism , Gene Expression/genetics , Gene Expression Regulation/genetics , Glycolysis , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/pharmacology , Intervertebral Disc/pathology , Male , Nucleus Pulposus/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology
18.
Clin Transl Med ; 11(11): e577, 2021 11.
Article in English | MEDLINE | ID: mdl-34841716

ABSTRACT

Drug resistance is a major hurdle for the effectiveness of tamoxifen (TAM) to provide clinical benefit. Therefore, it is essential to identify a sensitizer that could be used to improve TAM efficacy in treating TAM-resistant breast cancer. Here, we investigated the ability of baicalein to reverse TAM resistance. We found that baicalein increased the efficacy of TAM in inhibiting proliferation and inducing apoptosis of TAM-resistant cells. It also enhanced the TAM-induced growth reduction of resistant cells from NOD/SCID mouse mammary fat pads, without causing obvious systemic toxicity. Analyses using the CellMiner tool and the Kaplan-Meier plotter database showed that HIF-1α expression was inversely correlated with TAM therapeutic response in NCI-60 cancer cells and breast cancer patients. HIF-1α expression was increased in TAM-resistant cells due to an increase in mRNA levels and reduced ubiquitin-mediated degradation. Baicalein reduced HIF-1α expression by promoting its interaction with PHD2 and pVHL, thus facilitating ubiquitin ligase-mediated proteasomal degradation and thereby suppressing the nuclear translocation, binding to the hypoxia-response element, and transcriptional activity of HIF-1α. As a result, baicalein downregulated aerobic glycolysis by restricting glucose uptake, lactate production, ATP generation, lactate/pyruvate ratio and expression of HIF-1α-targeted glycolytic genes, thereby enhancing the antiproliferative efficacy of TAM. Furthermore, baicalein interfered with HIF-1α inhibition of mitochondrial biosynthesis, which increased mitochondrial DNA content and mitochondrial numbers, restored the generation of reactive oxygen species in mitochondria, and thus enhanced the TAM-induced mitochondrial apoptotic pathway. The HIF-1α stabilizer dimethyloxallyl glycine prevented the baicalein-induced downregulation of glycolysis and mitochondrial biosynthesis and reduced the effects of baicalein on reversing TAM resistance. Our results indicate that baicalein is a promising candidate to help overcome TAM resistance by sensitizing resistant cells to TAM-induced growth inhibition and apoptosis. The mechanism underlying the effects of baicalein consists of inhibition of HIF-1α-mediated aerobic glycolysis and mitochondrial dysfunction.


Subject(s)
Breast Neoplasms/drug therapy , Flavanones/pharmacology , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Tamoxifen/pharmacology , Warburg Effect, Oncologic/drug effects , Animals , Disease Models, Animal , Drug Resistance/drug effects , Female , Flavanones/metabolism , Flavanones/therapeutic use , Hypoxia-Inducible Factor 1, alpha Subunit/pharmacology , Hypoxia-Inducible Factor 1, alpha Subunit/therapeutic use , Mice, Inbred NOD/metabolism , Mitochondrial Diseases/drug therapy , Mitochondrial Diseases/physiopathology , Real-Time Polymerase Chain Reaction/methods , Real-Time Polymerase Chain Reaction/statistics & numerical data , Tamoxifen/metabolism , Tamoxifen/therapeutic use
19.
FEBS Open Bio ; 11(5): 1465-1475, 2021 05.
Article in English | MEDLINE | ID: mdl-33773069

ABSTRACT

Farnesyltransferase inhibitors (FTIs) suppress tumor aggressiveness in several malignancies by inhibiting Ras signaling. However, treatment of cells with a low dose of the FTI tipifarnib suppresses the expression of hypoxia-inducible factor-1α (HIF-1α) and results in antitumor effects without inhibiting the Ras pathway. Although we previously reported that elevated HIF-1α expression is associated with an aggressive phenotype in gastric cancer (GC), little is known about the antitumor effects of FTIs on GC. In this study, we examined the relationship between the antitumor effects of low-dose tipifarnib and HIF-1α expression in GC cells. Under normoxic conditions, HIF-1α was expressed only in MKN45 and KATOIII cells. The inhibitory effect of tipifarnib on HIF-1α was observed in HIF-1α-positive cells. Low-dose tipifarnib had antitumor effects only on HIF-1α-positive cells both in vitro and in vivo. Furthermore, low-dose tipifarnib inactivated ras homolog enriched in brain (Rheb)/mammalian target of rapamycin (mTOR) signaling and decreased intracellular reactive oxygen species (ROS) levels in HIF-1α-positive GC cells. Our results that the antitumor effects of low-dose tipifarnib are at least partially mediated through suppression of mTOR signaling and HIF-1α expression via inhibition of Rheb farnesylation and reduction in ROS levels. These findings suggest that low-dose tipifarnib may be capable of exerting an antitumor effect that is dependent on HIF-1α expression in GC cells. Tipifarnib may have potential as a novel therapeutic agent for HIF-1α-expressing GC exhibiting an aggressive phenotype.


Subject(s)
Quinolones/pharmacology , Stomach Neoplasms/drug therapy , TOR Serine-Threonine Kinases/metabolism , Cell Line, Tumor , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/pharmacology , Quinolones/metabolism , Reactive Oxygen Species , Signal Transduction/drug effects , Stomach Neoplasms/metabolism , TOR Serine-Threonine Kinases/physiology
20.
Exp Cell Res ; 395(1): 112175, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32679233

ABSTRACT

Autophagy is a basic catabolic response that eukaryotic cells use to degrade unnecessary or dysfunctional cellular components in an orderly and regulated manner. It plays important roles in maintaining cellular homeostasis, energy homeostasis, response to environmental stimuli, and the development of cancer. In solid tumors, hypoxia induces an increased HIF-1a that activates autophagy. However, the exact mechanism by which induced HIF-1a stimulates autophagy in cancer cells remains elusive. In the present study, we confirmed that ANKRD37 is upregulated in colon cancer tissue. Moreover, the higher expression level of ANKRD37 is related to a poorer survival rate. Using RNA interference, immunoblot, and immunofluorescence, we discovered that in cancer cell line RKO, hypoxia-induced HIF-1a regulates autophagy activity by increasing ANKRD37 level. In addition, intranuclear ANKRD37 played an important role in the regulation of hypoxia-induced autophagy. The translocation of ANKRD37 into cell nuclear is required for promoting cell growth and HIF-1a induced autophagy. These findings provide new insights to understand the hypoxia regulation mechanisms and the role of autophagy in cancer development.


Subject(s)
Autophagy/drug effects , Colonic Neoplasms/genetics , Gene Expression Regulation, Neoplastic/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Autophagy/genetics , Autophagy/physiology , Cell Hypoxia/drug effects , Cell Hypoxia/physiology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/physiology , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/pharmacology , RNA Interference/drug effects , RNA Interference/physiology , Transcriptional Activation/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...