Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 16.618
Filter
1.
Nat Commun ; 15(1): 3970, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38730227

ABSTRACT

High-altitude hypoxia acclimatization requires whole-body physiological regulation in highland immigrants, but the underlying genetic mechanism has not been clarified. Here we use sheep as an animal model for low-to-high altitude translocation. We generate multi-omics data including whole-genome sequences, time-resolved bulk RNA-Seq, ATAC-Seq and single-cell RNA-Seq from multiple tissues as well as phenotypic data from 20 bio-indicators. We characterize transcriptional changes of all genes in each tissue, and examine multi-tissue temporal dynamics and transcriptional interactions among genes. Particularly, we identify critical functional genes regulating the short response to hypoxia in each tissue (e.g., PARG in the cerebellum and HMOX1 in the colon). We further identify TAD-constrained cis-regulatory elements, which suppress the transcriptional activity of most genes under hypoxia. Phenotypic and transcriptional evidence indicate that antenatal hypoxia could improve hypoxia tolerance in offspring. Furthermore, we provide time-series expression data of candidate genes associated with human mountain sickness (e.g., BMPR2) and high-altitude adaptation (e.g., HIF1A). Our study provides valuable resources and insights for future hypoxia-related studies in mammals.


Subject(s)
Altitude Sickness , Altitude , Gene Expression Regulation , Hypoxia , Animals , Altitude Sickness/genetics , Altitude Sickness/metabolism , Sheep , Hypoxia/genetics , Hypoxia/metabolism , Humans , Acclimatization/genetics , Transcription, Genetic , Single-Cell Analysis , Female , Multiomics
2.
Int J Mol Sci ; 25(9)2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38731803

ABSTRACT

This study explores the effects of normobaric hypoxia and intermittent hypoxic training (IHT) on the physiological condition of the cardiac muscle in swimmers. Hypoxia has been reported to elicit both beneficial and adverse changes in the cardiovascular system, but its impact on the myocardium during acute exercise and altitude/hypoxic training remains less understood. We aimed to determine how a single bout of intense interval exercise and a four-week period of high-intensity endurance training under normobaric hypoxia affect cardiac marker activity in swimmers. Sixteen young male swimmers were divided into two groups: one undergoing training in hypoxia and the other in normoxia. Cardiac markers, including troponin I and T (cTnI and cTnT), heart-type fatty acid-binding protein (H-FABP), creatine kinase-MB isoenzyme (CK-MB), and myoglobin (Mb), were analyzed to assess the myocardium's response. We found no significant differences in the physiological response of the cardiac muscle to intense physical exertion between hypoxia and normoxia. Four weeks of IHT did not alter the resting levels of cTnT, cTnI, and H-FABP, but it resulted in a noteworthy decrease in the resting concentration of CK-MB, suggesting enhanced cardiac muscle adaptation to exercise. In contrast, a reduction in resting Mb levels was observed in the control group training in normoxia. These findings suggest that IHT at moderate altitudes does not adversely affect cardiac muscle condition and may support cardiac muscle adaptation, affirming the safety and efficacy of IHT as a training method for athletes.


Subject(s)
Athletes , Biomarkers , Hypoxia , Humans , Male , Hypoxia/metabolism , Pilot Projects , Swimming/physiology , Young Adult , Myocardium/metabolism , Myoglobin/metabolism , Troponin I/metabolism , Fatty Acid Binding Protein 3/metabolism , Adolescent , Fatty Acid-Binding Proteins/metabolism , Physical Endurance/physiology , Creatine Kinase, MB Form/blood , Creatine Kinase, MB Form/metabolism , Adaptation, Physiological , Altitude
3.
Int J Mol Sci ; 25(9)2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38731800

ABSTRACT

Understanding the molecular underpinnings of neurodegeneration processes is a pressing challenge for medicine and neurobiology. Alzheimer's disease (AD) and Parkinson's disease (PD) represent the most prevalent forms of neurodegeneration. To date, a substantial body of experimental evidence has strongly implicated hypoxia in the pathogenesis of numerous neurological disorders, including AD, PD, and other age-related neurodegenerative conditions. Hypoxia-inducible factor (HIF) is a transcription factor that triggers a cell survival program in conditions of oxygen deprivation. The involvement of HIF-1α in neurodegenerative processes presents a complex and sometimes contradictory picture. This review aims to elucidate the current understanding of the interplay between hypoxia and the development of AD and PD, assess the involvement of HIF-1 in their pathogenesis, and summarize promising therapeutic approaches centered on modulating the activity of the HIF-1 complex.


Subject(s)
Homeostasis , Hypoxia-Inducible Factor 1, alpha Subunit , Neurodegenerative Diseases , Oxygen , Parkinson Disease , Humans , Oxygen/metabolism , Animals , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Parkinson Disease/metabolism , Parkinson Disease/pathology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/etiology , Hypoxia-Inducible Factor 1/metabolism , Hypoxia/metabolism
4.
Function (Oxf) ; 5(3): zqae010, 2024.
Article in English | MEDLINE | ID: mdl-38706960

ABSTRACT

The Olfr78 gene encodes a G-protein-coupled olfactory receptor that is expressed in several ectopic sites. Olfr78 is one of the most abundant mRNA species in carotid body (CB) glomus cells. These cells are the prototypical oxygen (O2) sensitive arterial chemoreceptors, which, in response to lowered O2 tension (hypoxia), activate the respiratory centers to induce hyperventilation. It has been proposed that Olfr78 is a lactate receptor and that glomus cell activation by the increase in blood lactate mediates the hypoxic ventilatory response (HVR). However, this proposal has been challenged by several groups showing that Olfr78 is not a physiologically relevant lactate receptor and that the O2-based regulation of breathing is not affected in constitutive Olfr78 knockout mice. In another study, constitutive Olfr78 knockout mice were reported to have altered systemic and CB responses to mild hypoxia. To further characterize the functional role of Olfr78 in CB glomus cells, we here generated a conditional Olfr78 knockout mouse strain and then restricted the knockout to glomus cells and other catecholaminergic cells by crossing with a tyrosine hydroxylase-specific Cre driver strain (TH-Olfr78 KO mice). We find that TH-Olfr78 KO mice have a normal HVR. Interestingly, glomus cells of TH-Olfr78 KO mice exhibit molecular and electrophysiological alterations as well as a reduced dopamine content in secretory vesicles and neurosecretory activity. These functional characteristics resemble those of CB neuroblasts in wild-type mice. We suggest that, although Olfr78 is not essential for CB O2 sensing, activation of Olfr78-dependent pathways is required for maturation of glomus cells.


Subject(s)
Carotid Body , Mice, Knockout , Receptors, Odorant , Tyrosine 3-Monooxygenase , Animals , Carotid Body/metabolism , Tyrosine 3-Monooxygenase/metabolism , Tyrosine 3-Monooxygenase/genetics , Mice , Hypoxia/metabolism , Hypoxia/genetics , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Male , Mice, Inbred C57BL
5.
Front Immunol ; 15: 1372959, 2024.
Article in English | MEDLINE | ID: mdl-38690277

ABSTRACT

Introduction: Hypoxia is a common pathological driver contributing to various forms of pulmonary vascular diseases leading to pulmonary hypertension (PH). Pulmonary interstitial macrophages (IMs) play pivotal roles in immune and vascular dysfunction, leading to inflammation, abnormal remodeling, and fibrosis in PH. However, IMs' response to hypoxia and their role in PH progression remain largely unknown. We utilized a murine model of hypoxia-induced PH to investigate the repertoire and functional profiles of IMs in response to acute and prolonged hypoxia, aiming to elucidate their contributions to PH development. Methods: We conducted single-cell transcriptomic analyses to characterize the repertoire and functional profiles of murine pulmonary IMs following exposure to hypobaric hypoxia for varying durations (0, 1, 3, 7, and 21 days). Hallmark pathways from the mouse Molecular Signatures Database were utilized to characterize the molecular function of the IM subpopulation in response to hypoxia. Results: Our analysis revealed an early acute inflammatory phase during acute hypoxia exposure (Days 1-3), which was resolved by Day 7, followed by a pro-remodeling phase during prolonged hypoxia (Days 7-21). These phases were marked by distinct subpopulations of IMs: MHCIIhiCCR2+EAR2+ cells characterized the acute inflammatory phase, while TLF+VCAM1hi cells dominated the pro-remodeling phase. The acute inflammatory phase exhibited enrichment in interferon-gamma, IL-2, and IL-6 pathways, while the pro-remodeling phase showed dysregulated chemokine production, hemoglobin clearance, and tissue repair profiles, along with activation of distinct complement pathways. Discussion: Our findings demonstrate the existence of distinct populations of pulmonary interstitial macrophages corresponding to acute and prolonged hypoxia exposure, pivotal in regulating the inflammatory and remodeling phases of PH pathogenesis. This understanding offers potential avenues for targeted interventions, tailored to specific populations and distinct phases of the disease. Moreover, further identification of triggers for pro-remodeling IMs holds promise in unveiling novel therapeutic strategies for pulmonary hypertension.


Subject(s)
Gene Expression Profiling , Hypertension, Pulmonary , Hypoxia , Single-Cell Analysis , Transcriptome , Animals , Mice , Hypoxia/metabolism , Hypoxia/immunology , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/immunology , Hypertension, Pulmonary/genetics , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Mice, Inbred C57BL , Disease Models, Animal , Male , Lung/immunology , Lung/pathology , Lung/metabolism
6.
PLoS One ; 19(5): e0295094, 2024.
Article in English | MEDLINE | ID: mdl-38743782

ABSTRACT

Oxygen is essential to all the aerobic organisms. However, during normal development, disease and homeostasis, organisms are often challenged by hypoxia (oxygen deprivation). Hypoxia-inducible transcription factors (HIFs) are master regulators of hypoxia response and are evolutionarily conserved in metazoans. The homolog of HIF in the genetic model organism C. elegans is HIF-1. In this study, we aimed to understand short-term hypoxia response to identify HIF-1 downstream genes and identify HIF-1 direct targets in C. elegans. The central research questions were: (1) which genes are differentially expressed in response to short-term hypoxia? (2) Which of these changes in gene expression are dependent upon HIF-1 function? (3) Are any of these hif-1-dependent genes essential to survival in hypoxia? (4) Which genes are the direct targets of HIF-1? We combine whole genome gene expression analyses and chromatin immunoprecipitation sequencing (ChIP-seq) experiments to address these questions. In agreement with other published studies, we report that HIF-1-dependent hypoxia-responsive genes are involved in metabolism and stress response. Some HIF-1-dependent hypoxia-responsive genes like efk-1 and phy-2 dramatically impact survival in hypoxic conditions. Genes regulated by HIF-1 and hypoxia overlap with genes responsive to hydrogen sulfide, also overlap with genes regulated by DAF-16. The genomic regions that co-immunoprecipitate with HIF-1 are strongly enriched for genes involved in stress response. Further, some of these potential HIF-1 direct targets are differentially expressed under short-term hypoxia or are differentially regulated by mutations that enhance HIF-1 activity.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans , Hypoxia , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Hypoxia/genetics , Hypoxia/metabolism , Binding Sites , Gene Expression Profiling , Gene Expression Regulation , Hypoxia-Inducible Factor 1/metabolism , Hypoxia-Inducible Factor 1/genetics
7.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 40(4): 327-332, 2024 Apr.
Article in Chinese | MEDLINE | ID: mdl-38710517

ABSTRACT

Objective To investigate the liver injury induced by chronic intermittent hypoxia (CIH) activation of NOD-like receptor pyrin domain containing protein 1 (NLRP1) inflammasome. Methods C57BL/6 male mice were randomly divided into control group and CIH group. Mice in CIH group were put into CIH chamber for molding (8 hours a day for 4 weeks). After 4 weeks of molding, liver tissue cells was observed by HE staining, and the levels of alanine aminotransferase (ALT) and aspartate aminotransferase (AST) in serum of mice were detected by kit. The levels of reactive oxygen species (ROS) in liver tissue were detected by dihydroethidine (DHE). The expression and localization of NLRP1, apoptosis speck-like protein containing a caspase activation and recruiting domain (ASC) and caspase-1 were detected by immunohistochemical staining. The protein expressions of NLRP1, ASC, caspase-1, interleukin 1ß (IL-1ß) and tumor necrosis factor α (TNF-α) were detected by Western blot analysis. The serum levels of IL-1ß and TNF-α were detected by ELISA. Results Compared with the control group, the CIH group exhibited significant pathological changes in hepatocytes. Hepatocytes showed signs of rupture and necrosis, accompanied by inflammatory cell aggregation. Furthermore, the levels of ALT, AST, ROS, IL-1ß and TNF-α were elevated, along with increased protein expressions of NLRP1, ASC, caspase-1, IL-1ß and TNF-α. Conclusion CIH causes liver injury by activating NLRP1 inflammasome.


Subject(s)
Caspase 1 , Hypoxia , Inflammasomes , Interleukin-1beta , Liver , Mice, Inbred C57BL , Reactive Oxygen Species , Animals , Male , Inflammasomes/metabolism , Hypoxia/metabolism , Hypoxia/complications , Reactive Oxygen Species/metabolism , Liver/metabolism , Liver/pathology , Caspase 1/metabolism , Interleukin-1beta/metabolism , Mice , Adaptor Proteins, Signal Transducing/metabolism , Tumor Necrosis Factor-alpha/metabolism , Apoptosis Regulatory Proteins/metabolism , Alanine Transaminase/blood , CARD Signaling Adaptor Proteins/metabolism , Aspartate Aminotransferases/blood , Liver Diseases/etiology , Liver Diseases/metabolism , Liver Diseases/pathology
8.
Sci Rep ; 14(1): 10206, 2024 05 03.
Article in English | MEDLINE | ID: mdl-38702334

ABSTRACT

Cardiovascular function and adipose metabolism were markedly influenced under high altitudes. However, the interplay between adipokines and heart under hypoxia remains to be elucidated. We aim to explore alterations of adipokines and underlying mechanisms in regulating cardiac function under high altitudes. We investigated the cardiopulmonary function and five adipokines in Antarctic expeditioners at Kunlun Station (4,087 m) for 20 days and established rats exposed to hypobaric hypoxia (5,000 m), simulating Kunlun Station. Antarctic expeditioners exhibited elevated heart rate, blood pressure, systemic vascular resistance, and decreased cardiac pumping function. Plasma creatine phosphokinase-MB (CK-MB) and platelet-endothelial cell adhesion molecule-1 (sPecam-1) increased, and leptin, resistin, and lipocalin-2 decreased. Plasma leptin significantly correlated with altered cardiac function indicators. Additionally, hypoxic rats manifested impaired left ventricular systolic and diastolic function, elevated plasma CK-MB and sPecam-1, and decreased plasma leptin. Chronic hypoxia for 14 days led to increased myocyte hypertrophy, fibrosis, apoptosis, and mitochondrial dysfunction, coupled with reduced protein levels of leptin signaling pathways in myocardial tissues. Cardiac transcriptome analysis revealed leptin was associated with downregulated genes involved in rhythm, Na+/K+ transport, and cell skeleton. In conclusion, chronic hypoxia significantly reduced leptin signaling pathways in cardiac tissues along with significant pathological changes, thus highlighting the pivotal role of leptin in regulation of cardiac function under high altitudes.


Subject(s)
Altitude , Hypoxia , Leptin , Signal Transduction , Leptin/metabolism , Leptin/blood , Animals , Rats , Male , Hypoxia/metabolism , Hypoxia/physiopathology , Humans , Altitude Sickness/metabolism , Altitude Sickness/physiopathology , Myocardium/metabolism , Myocardium/pathology , Adult , Heart/physiopathology
9.
Nihon Yakurigaku Zasshi ; 159(3): 160-164, 2024.
Article in Japanese | MEDLINE | ID: mdl-38692880

ABSTRACT

The 2019 Nobel Prize in Physiology or Medicine was awarded to Dr. William G. Kaelin Jr, Dr. Peter J. Ratcliffe, and Dr. Gregg L. Semenza for their elucidation of new physiological mechanisms "How cells sense and adapt to oxygen availability". Moreover, two different drugs, HIF-PH inhibitors and HIF-2 inhibitors were also developed based on the discovery. Interestingly, those three doctors have different backgrounds as a medical oncologist, a nephrologist, and a pediatrician, respectively. They have started the research based on their own unique perspectives and eventually merged as "the elucidation of the response mechanism of living organisms to hypoxic environments". In this review, we will explain how the translational research that has begun to solve unmet clinical needs successfully contributed to the development of innovative therapeutic drugs.


Subject(s)
Hypoxia , Humans , Hypoxia/drug therapy , Hypoxia/metabolism , Animals , Drug Development , Molecular Targeted Therapy , Basic Helix-Loop-Helix Transcription Factors/metabolism
10.
Nihon Yakurigaku Zasshi ; 159(3): 165-168, 2024.
Article in Japanese | MEDLINE | ID: mdl-38692881

ABSTRACT

Molecular oxygen suffices the ATP production required for the survival of us aerobic organisms. But it is also true that oxygen acts as a source of reactive oxygen species that elicit a spectrum of damages in living organisms. To cope with such intrinsic ambiguity of biological activity oxygen exerts, aerobic mechanisms are equipped with an exquisite adaptive system, which sensitively detects partial pressure of oxygen within the body and controls appropriate oxygen supply to the tissues. Physiological responses to hypoxia are comprised of the acute and chronic phases, in the former of which the oxygen-sensing remains controversial particularly from mechanistic points of view. Recently, we have revealed that the prominently redox-sensitive cation channel TRPA1 plays key roles in oxygen-sensing mechanisms identified in the peripheral tissues and the central nervous system. In this review, we summarize recent development of researches on oxygen-sensing mechanisms including that in the carotid body, which has been recognized as the oxygen receptor organ central to acute oxygen-sensing. We also discuss how ubiquitously the TRPA1 contributes to the mechanisms underlying the acute phase of adaptation to hypoxia.


Subject(s)
Oxygen , TRPA1 Cation Channel , Transient Receptor Potential Channels , TRPA1 Cation Channel/metabolism , Humans , Oxygen/metabolism , Animals , Transient Receptor Potential Channels/metabolism , Hypoxia/metabolism , Calcium Channels/metabolism , Nerve Tissue Proteins/metabolism , Reactive Oxygen Species/metabolism , Carotid Body/metabolism
11.
Exp Cell Res ; 438(2): 114061, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38692345

ABSTRACT

Acute myocardial infarction (AMI) is a prevalent cardiovascular disease with high morbidity and mortality rates worldwide. Pyroptosis is an inflammatory form of programmed cell death that has been linked to various pathological conditions. However, its exact contribution to the onset and progression of heart injury in AMI has not yet fully elucidated. Herein, we established mouse AMI model by ligating the left anterior descending artery and performed transcriptome analysis during the early phase of AMI. Mouse HL-1 and human AC-16 cardiomyocytes were subjected to hypoxia to simulate ischemic injury in vitro. Our results revealed a significant activation of the inflammatory response at 3 h post-ligation, as confirmed by RNA sequencing. We identified the occurrence of NLRP3 inflammasome-mediated pyroptosis in the cardiac tissues of human cases with AMI, as well as in mouse models of AMI and hypoxia-induced cardiomyocytes, using immunohistochemistry staining and Western blotting assays. Concurrently, pharmacological inhibition of NLRP3 inflammasome-mediated pyroptosis with MCC950 and VX-765 effectively decreased hypoxia-induced cardiomyocytes injury, while mitigating myocardial oxidative stress, apoptosis and inflammation caused by hypoxia. Moreover, the circulating levels of gasdermin D (GSDMD), the pyroptosis executor, were remarkably elevated in the plasma of mice with early AMI and in the supernatant of hypoxia-exposed cardiomyocytes in a time-dependent manner using ELISA and Western blotting. Furthermore, the change in circulating GSDMD positively correlated with Creatine Kinase-MB (CK-MB) in the plasma of early-stage AMI mouse. In summary, these findings indicated a critical role for NLRP3 inflammasome-mediated pyroptosis in the progression of AMI, the administration of MCC950 and VX-765 may be attractive candidate therapeutic approaches for cardiac injury caused by acute hypoxia or even AMI. Additionally, the circulating GSDMD exhibits potential as a newly diagnostic biomarker for AMI.


Subject(s)
Apoptosis , Furans , Inflammation , Mice, Inbred C57BL , Myocardial Infarction , Myocytes, Cardiac , Oxidative Stress , Pyroptosis , Sulfonamides , Pyroptosis/drug effects , Animals , Mice , Apoptosis/drug effects , Oxidative Stress/drug effects , Sulfonamides/pharmacology , Humans , Inflammation/metabolism , Inflammation/pathology , Inflammation/drug therapy , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Male , Furans/pharmacology , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/drug therapy , Indenes/pharmacology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , para-Aminobenzoates/pharmacology , Inflammasomes/metabolism , Inflammasomes/drug effects , Disease Models, Animal , Myocardium/metabolism , Myocardium/pathology , Hypoxia/metabolism , Hypoxia/complications , Dipeptides
12.
Int J Mol Sci ; 25(8)2024 Apr 14.
Article in English | MEDLINE | ID: mdl-38673925

ABSTRACT

The protective effects of hydrogen sulfide (H2S) against ischemic brain injury and its role in promoting angiogenesis have been established. However, the specific mechanism underlying these effects remains unclear. This study is designed to investigate the regulatory impact and mechanism of H2S on VEGFR2 phosphorylation. Following expression and purification, the recombinant His-VEGFR2 protein was subjected to LC-PRM/MS analysis to identify the phosphorylation sites of VEGFR2 upon NaHS treatment. Adenovirus infection was used to transfect primary rat brain artery endothelial cells (BAECs) with the Ad-VEGFR2WT, Ad-VEGFR2Y797F, and Ad-VEGFR2S799A plasmids. The expression of VEGFR2 and recombinant Flag-VEGFR2, along with Akt phosphorylation, cell proliferation, and LDH levels, was assessed. The migratory capacity and tube-forming potential of BAECs were assessed using wound healing, transwell, and tube formation assays. NaHS notably enhanced the phosphorylation of VEGFR2 at Tyr797 and Ser799 sites. These phosphorylation sites were identified as crucial for mediating the protective effects of NaHS against hypoxia-reoxygenation (H/R) injury. NaHS significantly enhanced the Akt phosphorylation, migratory capacity, and tube formation of BAECs and upregulated the expression of VEGFR2 and recombinant proteins. These findings suggest that Tyr797 and Ser799 sites of VEGFR2 serve as crucial mediators of H2S-induced pro-angiogenic effects and protection against H/R injury.


Subject(s)
Endothelial Cells , Hydrogen Sulfide , Vascular Endothelial Growth Factor Receptor-2 , Phosphorylation/drug effects , Vascular Endothelial Growth Factor Receptor-2/metabolism , Vascular Endothelial Growth Factor Receptor-2/genetics , Hydrogen Sulfide/pharmacology , Hydrogen Sulfide/metabolism , Animals , Rats , Endothelial Cells/metabolism , Endothelial Cells/drug effects , Neovascularization, Physiologic/drug effects , Cell Movement/drug effects , Rats, Sprague-Dawley , Cell Hypoxia , Cell Proliferation/drug effects , Tyrosine/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Reperfusion Injury/metabolism , Reperfusion Injury/genetics , Angiogenesis Inducing Agents/pharmacology , Angiogenesis Inducing Agents/metabolism , Serine/metabolism , Hypoxia/metabolism
13.
Free Radic Biol Med ; 219: 141-152, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38636714

ABSTRACT

Pulmonary hypertension (PH) is a devastating disease that lacks effective treatment options and is characterized by severe pulmonary vascular remodeling. Pulmonary arterial endothelial cell (PAEC) dysfunction drives the initiation and pathogenesis of pulmonary arterial hypertension. Canonical transient receptor potential (TRPC) channels, a family of Ca2+-permeable channels, play an important role in various diseases. However, the effect and mechanism of TRPCs on PH development have not been fully elucidated. Among the TRPC family members, TRPC4 expression was markedly upregulated in PAECs from hypoxia combined with SU5416 (HySu)-induced PH mice and monocrotaline (MCT)-treated PH rats, as well as in hypoxia-exposed PAECs, suggesting that TRPC4 in PAECs may participate in the occurrence and development of PH. In this study, we aimed to investigate whether TRPC4 in PAECs has an aggravating effect on PH and elucidate the molecular mechanisms. We observed that hypoxia treatment promoted PAEC apoptosis through a caspase-12/endoplasmic reticulum stress (ERS)-dependent pathway. Knockdown of TRPC4 attenuated hypoxia-induced apoptosis and caspase-3/caspase-12 activity in PAECs. Accordingly, adeno-associated virus (AAV) serotype 6-mediated pulmonary endothelial TRPC4 silencing (AAV6-Tie-shRNA-TRPC4) or TRPC4 antagonist suppressed PH progression as evidenced by reduced right ventricular systolic pressure (RVSP), pulmonary vascular remodeling, PAEC apoptosis and reactive oxygen species (ROS) production. Mechanistically, unbiased RNA sequencing (RNA-seq) suggested that TRPC4 deficiency suppressed the expression of the proapoptotic protein sushi domain containing 2 (Susd2) in hypoxia-exposed mouse PAECs. Moreover, TRPC4 activated hypoxia-induced PAEC apoptosis by promoting Susd2 expression. Therefore, inhibiting TRPC4 ameliorated PAEC apoptosis and hypoxic PH in animals by repressing Susd2 signaling, which may serve as a therapeutic target for the management of PH.


Subject(s)
Apoptosis , Endoplasmic Reticulum Stress , Endothelial Cells , Hypertension, Pulmonary , Hypoxia , TRPC Cation Channels , Animals , TRPC Cation Channels/metabolism , TRPC Cation Channels/genetics , Mice , Endothelial Cells/metabolism , Endothelial Cells/pathology , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/pathology , Hypertension, Pulmonary/genetics , Rats , Hypoxia/metabolism , Pulmonary Artery/pathology , Pulmonary Artery/metabolism , Male , Monocrotaline/toxicity , Vascular Remodeling/genetics , Disease Models, Animal , Humans , Signal Transduction , Mice, Inbred C57BL , Rats, Sprague-Dawley , Cells, Cultured , Indoles , Pyrroles
14.
Nat Commun ; 15(1): 3435, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38653959

ABSTRACT

Wound healing is an obvious clinical concern that can be hindered by inadequate angiogenesis, inflammation, and chronic hypoxia. While exosomes derived from adipose tissue-derived stem cells have shown promise in accelerating healing by carrying therapeutic growth factors and microRNAs, intracellular cargo delivery is compromised in hypoxic tissues due to activated hypoxia-induced endocytic recycling. To address this challenge, we have developed a strategy to coat oxygen nanobubbles with exosomes and incorporate them into a polyvinyl alcohol/gelatin hybrid hydrogel. This approach not only alleviates wound hypoxia but also offers an efficient means of delivering exosome-coated nanoparticles in hypoxic conditions. The self-healing properties of the hydrogel, along with its component, gelatin, aids in hemostasis, while its crosslinking bonds facilitate hydrogen peroxide decomposition, to ameliorate wound inflammation. Here, we show the potential of this multifunctional hydrogel for enhanced healing, promoting angiogenesis, facilitating exosome delivery, mitigating hypoxia, and inhibiting inflammation in a male rat full-thickness wound model.


Subject(s)
Exosomes , Hydrogels , Oxygen , Wound Healing , Exosomes/metabolism , Wound Healing/drug effects , Animals , Hydrogels/chemistry , Male , Rats , Oxygen/metabolism , Humans , Rats, Sprague-Dawley , Nanoparticles/chemistry , Polyvinyl Alcohol/chemistry , Neovascularization, Physiologic/drug effects , Gelatin/chemistry , Hypoxia/metabolism , Inflammation/metabolism
15.
Biol Sex Differ ; 15(1): 30, 2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38566248

ABSTRACT

BACKGROUND: Neonatal hypoxia ischemia (HI) related brain injury is one of the major causes of learning disabilities and memory deficits in children. In both human and animal studies, female neonate brains are less susceptible to HI than male brains. Phosphorylation of the nerve growth factor receptor TrkB has been shown to provide sex-specific neuroprotection following in vivo HI in female mice in an estrogen receptor alpha (ERα)-dependent manner. However, the molecular and cellular mechanisms conferring sex-specific neonatal neuroprotection remain incompletely understood. Here, we test whether female neonatal hippocampal neurons express autonomous neuroprotective properties and assess the ability of testosterone (T) to alter this phenotype. METHODS: We cultured sexed hippocampal neurons from ERα+/+ and ERα-/- mice and subjected them to 4 h oxygen glucose deprivation and 24 h reoxygenation (4-OGD/24-REOX). Sexed hippocampal neurons were treated either with vehicle control (VC) or the TrkB agonist 7,8-dihydroxyflavone (7,8-DHF) following in vitro ischemia. End points at 24 h REOX were TrkB phosphorylation (p-TrkB) and neuronal survival assessed by immunohistochemistry. In addition, in vitro ischemia-mediated ERα gene expression in hippocampal neurons were investigated following testosterone (T) pre-treatment and TrkB antagonist therapy via q-RTPCR. Multifactorial analysis of variance was conducted to test for significant differences between experimental conditions. RESULTS: Under normoxic conditions, administration of 3 µM 7,8-DHF resulted an ERα-dependent increase in p-TrkB immunoexpression that was higher in female, as compared to male neurons. Following 4-OGD/24-REOX, p-TrkB expression increased 20% in both male and female ERα+/+ neurons. However, with 3 µM 7,8-DHF treatment p-TrkB expression increased further in female neurons by 2.81 ± 0.79-fold and was ERα dependent. 4-OGD/24-REOX resulted in a 56% increase in cell death, but only female cells were rescued with 3 µM 7,8-DHF, again in an ERα dependent manner. Following 4-OGD/3-REOX, ERα mRNA increased ~ 3 fold in female neurons. This increase was blocked with either the TrkB antagonist ANA-12 or pre-treatment with T. Pre-treatment with T also blocked the 7,8-DHF- dependent sex-specific neuronal survival in female neurons following 4-OGD/24-REOX. CONCLUSIONS: OGD/REOX results in sex-dependent TrkB phosphorylation in female neurons that increases further with 7,8-DHF treatment. TrkB phosphorylation by 7,8-DHF increased ERα mRNA expression and promoted cell survival preferentially in female hippocampal neurons. The sex-dependent neuroprotective actions of 7,8-DHF were blocked by either ANA-12 or by T pre-treatment. These results are consistent with a model for a female-specific neuroprotective pathway in hippocampal neurons in response to hypoxia. The pathway is activated by 7,8-DHF, mediated by TrkB phosphorylation, dependent on ERα and blocked by pre-exposure to T.


Subject(s)
Estrogen Receptor alpha , Neuroprotective Agents , Child , Female , Animals , Male , Mice , Humans , Estrogen Receptor alpha/metabolism , Neuroprotection , Sex Characteristics , Testosterone/pharmacology , Testosterone/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/metabolism , Neurons/metabolism , Hippocampus/metabolism , Ischemia , Hypoxia/metabolism , RNA, Messenger/metabolism
16.
BMC Cancer ; 24(1): 509, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38654280

ABSTRACT

BACKGROUND: Glioblastoma is a malignant and aggressive type of central nevous system malignancy characterized by many distinct biological features including extensive hypoxia. Hypoxia in glioblatoma associates with complex signaling patterns including activation of several pathways such as MAPK, PI3K-AKT/mTOR and IL-6/JAK/STAT3 with the master regulator HIF-1, which in turn drive particular tumor behaviors determining, in the end, treatment outcomes and patients fate. Thus, the present study was designed to investigate the expression of selected hypoxia related factors including STAT3 in a small set of long-term surviving glioma patients. METHODS: The expression of selected hypoxia related factors including STAT3 was evaluated in a time series of formalin fixed paraffin embedded and cryopreserved glioma samples from repeatedly resected patients. In addition, comparative studies were also conducted on primary glioma cells derived from original patient samples, stabilized glioma cell lines and tumor-xenograft mice model. Obtained data were correlated with clinical findings too. RESULTS: Glioblastoma samples of the analyzed patients displayed heterogeneity in the expression of hypoxia- related and EMT markers with most interesting trend being observed in pSTAT3. This heterogeneity was subsequently confirmed in other employed models (primocultures derived from glioblastoma tissue resections, cryopreserved tumor specimens, stabilized glioblastoma cell line in vitro and in vivo) and concerned, in particular, STAT3 expression which remained stable. In addition, subsequent studies on the role of STAT3 in the context of glioblastoma hypoxia demonstrated opposing effects of its deletion on cell viability as well as the expression of hypoxia and EMT markers. CONCLUSIONS: Our results suport the importance of STAT3 expression and activity in the context of hypoxia in malignant glioblastoma long-term surviving glioma patients while emphasizing heterogeneity of biological outcomes in varying employed tumor models.


Subject(s)
Glioma , STAT3 Transcription Factor , STAT3 Transcription Factor/metabolism , Humans , Animals , Mice , Glioma/metabolism , Glioma/pathology , Glioma/genetics , Male , Female , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Brain Neoplasms/genetics , Cell Line, Tumor , Middle Aged , Biomarkers, Tumor/metabolism , Aged , Adult , Glioblastoma/metabolism , Glioblastoma/pathology , Glioblastoma/genetics , Gene Expression Regulation, Neoplastic , Hypoxia/metabolism
17.
Cells ; 13(7)2024 Apr 03.
Article in English | MEDLINE | ID: mdl-38607059

ABSTRACT

Embryonic neurogenesis can be defined as a period of prenatal development during which divisions of neural stem and progenitor cells give rise to neurons. In the central nervous system of most mammals, including humans, the majority of neocortical neurogenesis occurs before birth. It is a highly spatiotemporally organized process whose perturbations lead to cortical malformations and dysfunctions underlying neurological and psychiatric pathologies, and in which oxygen availability plays a critical role. In case of deprived oxygen conditions, known as hypoxia, the hypoxia-inducible factor (HIF) signaling pathway is activated, resulting in the selective expression of a group of genes that regulate homeostatic adaptations, including cell differentiation and survival, metabolism and angiogenesis. While a physiological degree of hypoxia is essential for proper brain development, imbalanced oxygen levels can adversely affect this process, as observed in common obstetrical pathologies such as prematurity. This review comprehensively explores and discusses the current body of knowledge regarding the role of hypoxia and the HIF pathway in embryonic neurogenesis of the mammalian cortex. Additionally, it highlights existing gaps in our understanding, presents unanswered questions, and provides avenues for future research.


Subject(s)
Neural Stem Cells , Humans , Animals , Pregnancy , Female , Neural Stem Cells/metabolism , Neurons/metabolism , Neurogenesis/genetics , Hypoxia/metabolism , Oxygen/metabolism , Mammals
18.
Methods Mol Biol ; 2803: 163-172, 2024.
Article in English | MEDLINE | ID: mdl-38676892

ABSTRACT

Pulmonary hypertension (PH) is a devastating disease, characterized by complex remodeling of the pulmonary vasculature. PH is classified into five groups based on different etiology, pathology, as well as therapy and prognosis. Animal models are essential for the study of underlying mechanisms, pathophysiology, and preclinical testing of new therapies for PH. The complexity of the disease with different clinical entities dictates the necessity for more than one animal model to resemble PH, as a single model cannot imitate the broad spectrum of human PH.Here we describe a detailed protocol for creating a rat model of PH with right ventricular (RV) failure. Furthermore, we present how to characterize it hemodynamically by invasive measurements of RV and pulmonary arterial (PA) pressures. Animals subjected to this model display severe pulmonary vascular remodeling and RV dysfunction. In this model, rats undergo a single subcutaneous injection of Sugen (SU5416, a vascular endothelial growth factor inhibitor) and are immediately exposed to chronic hypoxia in a hypoxia chamber for 3-6 weeks. This Sugen/Hypoxia rat model resembles Group 1 PH.


Subject(s)
Disease Models, Animal , Heart Failure , Hypertension, Pulmonary , Hypoxia , Animals , Hypertension, Pulmonary/physiopathology , Hypertension, Pulmonary/pathology , Hypertension, Pulmonary/etiology , Rats , Hypoxia/metabolism , Heart Failure/physiopathology , Heart Failure/etiology , Heart Failure/pathology , Pyrroles/pharmacology , Indoles/pharmacology , Ventricular Dysfunction, Right/physiopathology , Ventricular Dysfunction, Right/etiology , Hemodynamics , Pulmonary Artery/pathology , Pulmonary Artery/physiopathology , Male , Humans , Vascular Remodeling , Vascular Endothelial Growth Factor A/metabolism
19.
Zhonghua Bing Li Xue Za Zhi ; 53(5): 452-457, 2024 May 08.
Article in Chinese | MEDLINE | ID: mdl-38678325

ABSTRACT

Objective: To investigate the role of RNA m6A methylation in mediating cerebellar dysplasia through analyzing the phenotypes of the mouse cerebella and the expression of several key m6A regulators upon hypobaric hypoxia treatment. Methods: Five-day old C57/BL6 mice were exposed to hypobaric hypoxia for 9 days. The status of mouse cerebellar development was analyzed by comparing the body weights, brain weights and histological features. Immunostaining of cell-type-specific markers was performed to analyze the cerebellar morphology. Real-time PCR, Western blot and immunohistochemical staining were performed to detect the expression of key m6A regulators in the mouse cerebella. Results: Compared with the control, the body weights, brain weights and cerebellar volumes of hypobaric hypoxic mice were significantly reduced (P<0.01). The expression of specific markers in different cells, including NeuN (mature neuron), Calbindin-D28K (Purkinje cell) and GFAP (astrocyte), was decreased in hypobaric hypoxic mouse cerebella (P<0.01), accompanied with disorganized cellular structure. The expression of methyltransferase METTL3 was significantly down-regulated in the cerebella of hypobaric hypoxic mice (P<0.05). Conclusions: Hypobaric hypoxia stimulation causes mouse cerebellar dysplasia, with structural abnormalities in mature granular neurons, Purkinje cells and astrocytes. Expression of METTL3 is decreased in hypobaric hypoxic mice cerebellum compared with that of normobaric normoxic mice, suggesting that its mediated RNA m6A methylation may play an important role in hypobaric hypoxia-induced mouse cerebellar dysplasia.


Subject(s)
Calbindins , Cerebellum , DNA-Binding Proteins , Hypoxia , Methyltransferases , Mice, Inbred C57BL , Nerve Tissue Proteins , Purkinje Cells , Animals , Mice , Cerebellum/metabolism , Hypoxia/metabolism , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Purkinje Cells/metabolism , Purkinje Cells/pathology , Calbindins/metabolism , Calbindins/genetics , Methyltransferases/metabolism , Methyltransferases/genetics , Glial Fibrillary Acidic Protein/metabolism , Glial Fibrillary Acidic Protein/genetics , Astrocytes/metabolism , Down-Regulation , Methylation , Adenosine/metabolism , Adenosine/analogs & derivatives , Nervous System Malformations/metabolism , Nervous System Malformations/genetics
20.
Front Immunol ; 15: 1385907, 2024.
Article in English | MEDLINE | ID: mdl-38605960

ABSTRACT

The human intestinal tract constitutes a complex ecosystem, made up of countless gut microbiota, metabolites, and immune cells, with hypoxia being a fundamental environmental characteristic of this ecology. Under normal physiological conditions, a delicate balance exists among these complex "residents", with disruptions potentially leading to inflammatory bowel disease (IBD). The core pathology of IBD features a disrupted intestinal epithelial barrier, alongside evident immune and microecological disturbances. Central to these interconnected networks is hypoxia-inducible factor-1α (HIF-1α), which is a key regulator in gut cells for adapting to hypoxic conditions and maintaining gut homeostasis. Short-chain fatty acids (SCFAs), as pivotal gut metabolites, serve as vital mediators between the host and microbiota, and significantly influence intestinal ecosystem. Recent years have seen a surge in research on the roles and therapeutic potential of HIF-1α and SCFAs in IBD independently, yet reviews on HIF-1α-mediated SCFAs regulation of IBD under hypoxic conditions are scarce. This article summarizes evidence of the interplay and regulatory relationship between SCFAs and HIF-1α in IBD, pivotal for elucidating the disease's pathogenesis and offering promising therapeutic strategies.


Subject(s)
Fatty Acids, Volatile , Hypoxia-Inducible Factor 1, alpha Subunit , Inflammatory Bowel Diseases , Humans , Fatty Acids, Volatile/metabolism , Hypoxia/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Inflammatory Bowel Diseases/metabolism , Microbiota
SELECTION OF CITATIONS
SEARCH DETAIL
...