Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 106
Filtrar
1.
Sci Rep ; 14(1): 11585, 2024 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-38773195

RESUMEN

High-altitude cerebral edema (HACE) is a severe neurological condition that can occur at high altitudes. It is characterized by the accumulation of fluid in the brain, leading to a range of symptoms, including severe headache, confusion, loss of coordination, and even coma and death. Exosomes play a crucial role in intercellular communication, and their contents have been found to change in various diseases. This study analyzed the metabolomic characteristics of blood exosomes from HACE patients compared to those from healthy controls (HCs) with the aim of identifying specific metabolites or metabolic pathways associated with the development of HACE conditions. A total of 21 HACE patients and 21 healthy controls were recruited for this study. Comprehensive metabolomic profiling of the serum exosome samples was conducted using ultraperformance liquid chromatography-tandem mass spectrometry (UPLC‒MS/MS). Additionally, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis was performed to identify the metabolic pathways affected in HACE patients. Twenty-six metabolites, including ( +)-camphoric acid, choline, adenosine, adenosine 5'-monophosphate, deoxyguanosine 5'-monophosphate, guanosine, and hypoxanthine-9-ß-D-arabinofuranoside, among others, exhibited significant changes in expression in HACE patients compared to HCs. Additionally, these differentially abundant metabolites were confirmed to be potential biomarkers for HACE. KEGG pathway enrichment analysis revealed several pathways that significantly affect energy metabolism regulation (such as purine metabolism, thermogenesis, and nucleotide metabolism), estrogen-related pathways (the estrogen signaling pathway, GnRH signaling pathway, and GnRH pathway), cyclic nucleotide signaling pathways (the cGMP-PKG signaling pathway and cAMP signaling pathway), and hormone synthesis and secretion pathways (renin secretion, parathyroid hormone synthesis, secretion and action, and aldosterone synthesis and secretion). In patients with HACE, adenosine, guanosine, and hypoxanthine-9-ß-D-arabinofuranoside were negatively correlated with height. Deoxyguanosine 5'-monophosphate is negatively correlated with weight and BMI. Additionally, LPE (18:2/0:0) and pregnanetriol were positively correlated with age. This study identified potential biomarkers for HACE and provided valuable insights into the underlying metabolic mechanisms of this disease. These findings may lead to potential targets for early diagnosis and therapeutic intervention in HACE patients.


Asunto(s)
Biomarcadores , Edema Encefálico , Exosomas , Metabolómica , Humanos , Masculino , Femenino , Adulto , Metabolómica/métodos , Edema Encefálico/sangre , Edema Encefálico/metabolismo , Edema Encefálico/etiología , Biomarcadores/sangre , Exosomas/metabolismo , Espectrometría de Masas en Tándem , Mal de Altura/sangre , Mal de Altura/metabolismo , Persona de Mediana Edad , Redes y Vías Metabólicas , Metaboloma , Estudios de Casos y Controles , Altitud
2.
Eur J Sport Sci ; 23(10): 2002-2010, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37051668

RESUMEN

Hypoxia induced intestinal barrier injury, microbial translocation, and local/systemic inflammation may contribute to high-altitude associated gastrointestinal complications or symptoms of acute mountain sickness (AMS). Therefore, we tested the hypothesis that six-hours of hypobaric hypoxia increases circulating markers of intestinal barrier injury and inflammation. A secondary aim was to determine if the changes in these markers were different between those with and without AMS. Thirteen participants were exposed to six hours of hypobaric hypoxia, simulating an altitude of 4572 m. Participants completed two 30-minute bouts of exercise during the early hours of hypoxic exposure to mimic typical activity required by those at high altitude. Pre- and post-exposure blood samples were assessed for circulating markers of intestinal barrier injury and inflammation. Data below are presented as mean ± standard deviation or median [interquartile range]. Intestinal fatty acid binding protein (Δ251 [103-410] pg•mL-1; p = 0.002, d = 0.32), lipopolysaccharide binding protein (Δ2 ± 2.4 µg•mL-1; p = 0.011; d = 0.48), tumor necrosis factor-α (Δ10.2 [3-42.2] pg•mL-1; p = 0.005; d = 0.25), interleukin-1ß (Δ1.5 [0-6.7] pg•mL-1 p = 0.042; d = 0.18), and interleukin-1 receptor agonist (Δ3.4 [0.4-5.2] pg•mL-1p = 0.002; d = 0.23) increased from pre- to post-hypoxia. Six of the 13 participants developed AMS; however, the pre- to post-hypoxia changes for each marker were not different between those with and without AMS (p > 0.05 for all indices). These data provide evidence that high altitude exposures can lead to intestinal barrier injury, which may be an important consideration for mountaineers, military personnel, wildland firefighters, and athletes who travel to high altitudes to perform physical work or exercise.


Asunto(s)
Mal de Altura , Esfuerzo Físico , Humanos , Hipoxia , Mal de Altura/complicaciones , Mal de Altura/diagnóstico , Mal de Altura/metabolismo , Altitud , Inflamación
3.
J Bone Miner Res ; 38(4): 597-614, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36680558

RESUMEN

Chronic high-altitude hypoxia induces irreversible abnormalities in various organisms. Emerging evidence indicates that hypobaric hypoxia markedly suppresses bone mass and bone strength. However, few effective means have been identified to prevent such bone deficits. Here, we assessed the potential of pulsed electromagnetic fields (PEMFs) to noninvasively resist bone deterioration induced by hypobaric hypoxia. We observed that exogenous PEMF treatment at 15 Hz and 20 Gauss (Gs) improved the cancellous and cortical bone mass, bone microstructure, and skeletal mechano-properties in rats subjected to chronic exposure of hypobaric hypoxia simulating an altitude of 4500 m for 6 weeks by primarily modulating osteoblasts and osteoblast-mediated bone-forming activity. Moreover, our results showed that whereas PEMF stimulated the functional activity of primary osteoblasts in hypoxic culture in vitro, it had negligible effects on osteoclasts and osteocytes exposed to hypoxia. Mechanistically, the primary cilium was found to function as the major electromagnetic sensor in osteoblasts exposed to hypoxia. The polycystins PC-1/PC-2 complex was identified as the primary calcium channel in the primary cilium of hypoxia-exposed osteoblastic cells responsible for the detection of external PEMF signals, and thereby translated these biophysical signals into intracellular biochemical events involving significant increase in the intracellular soluble adenylyl cyclase (sAC) expression and subsequent elevation of cyclic adenosine monophosphate (cAMP) concentration. The second messenger cAMP inhibited the transcription of oxygen homeostasis-related hypoxia-inducible factor 1-alpha (HIF-1α), and thus enhanced osteoblast differentiation and improved bone phenotype. Overall, the present study not only advances our understanding of bone physiology at high altitudes, but more importantly, proposes effective means to ameliorate high altitude-induced bone loss in a noninvasive and cost-effective manner. © 2023 American Society for Bone and Mineral Research (ASBMR).


Asunto(s)
Mal de Altura , Ratas , Animales , Mal de Altura/metabolismo , Campos Electromagnéticos , Cilios , Huesos , Hipoxia/complicaciones , Hipoxia/metabolismo , Osteoblastos/metabolismo , AMP Cíclico/metabolismo
4.
Cell Mol Biol (Noisy-le-grand) ; 68(6): 84-91, 2022 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-36227670

RESUMEN

To investigate the oxidative stress and adaptive compensation of kidneys in rats in high-altitude hypoxia environments, 20 Wistar rats (3 months) were randomly and equally grouped. The rats in the test group were fed in a low-pressure oxygen chamber, and those in the control group (controls) were fed in a normal environment. On the 5th, 10th, 20th, and 30th day, the excretion of uric acid in rats was detected by a biochemical analyzer, the level of desmin protein in rat podocytes was detected by immunohistochemistry, and the activity of Na+-K+- ATPase in rat proximal tubular epithelial cells was measured by liquid scintillation method. The results showed that with the increased time, the level of uric acid in the blood of rats in the test group increased dramatically (P<0.05). On the 30th day, the blood uric acid content of the test group was 52.33µmol/L, and that of the control group was 38.43µmol/L. The blood uric acid content in the test group was dramatically increased relative to the control group. Immunohistochemistry showed that the desmin protein in podocytes of the test group (0.14) was considerably higher than that in the control group (P<0.05). The Na+-K+- ATPase activity of proximal renal tubular epithelial cells in the test group was 611.2 pmol pi/mg protein/h, which was considerably lower than the versus control group (P<0.05). In summary, in high altitude hypoxia environment, uric acid accumulated in the body, and renal filtration and excretion ability was limited.


Asunto(s)
Mal de Altura , Podocitos , Mal de Altura/metabolismo , Animales , Desmina/metabolismo , Desmina/farmacología , Hipoxia , Túbulos Renales Proximales/metabolismo , Oxígeno/metabolismo , Podocitos/metabolismo , Ratas , Ratas Wistar , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Ácido Úrico/metabolismo , Ácido Úrico/farmacología
5.
Int J Mol Sci ; 23(20)2022 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-36293512

RESUMEN

Exposure to high altitudes generates a decrease in the partial pressure of oxygen, triggering a hypobaric hypoxic condition. This condition produces pathophysiologic alterations in an organism. In the lung, one of the principal responses to hypoxia is the development of hypoxic pulmonary vasoconstriction (HPV), which improves gas exchange. However, when HPV is exacerbated, it induces high-altitude pulmonary hypertension (HAPH). Another important illness in hypobaric hypoxia is high-altitude pulmonary edema (HAPE), which occurs under acute exposure. Several studies have shown that inflammatory processes are activated in high-altitude illnesses, highlighting the importance of the crosstalk between hypoxia and inflammation. The aim of this review is to determine the inflammatory pathways involved in hypobaric hypoxia, to investigate the key role of inflammation in lung pathologies, such as HAPH and HAPE, and to summarize different anti-inflammatory treatment approaches for these high-altitude illnesses. In conclusion, both HAPE and HAPH show an increase in inflammatory cell infiltration (macrophages and neutrophils), cytokine levels (IL-6, TNF-α and IL-1ß), chemokine levels (MCP-1), and cell adhesion molecule levels (ICAM-1 and VCAM-1), and anti-inflammatory treatments (decreasing all inflammatory components mentioned above) seem to be promising mitigation strategies for treating lung pathologies associated with high-altitude exposure.


Asunto(s)
Mal de Altura , Hipertensión Pulmonar , Infecciones por Papillomavirus , Edema Pulmonar , Humanos , Hipertensión Pulmonar/metabolismo , Molécula 1 de Adhesión Intercelular , Altitud , Edema Pulmonar/patología , Molécula 1 de Adhesión Celular Vascular , Factor de Necrosis Tumoral alfa , Interleucina-6 , Infecciones por Papillomavirus/complicaciones , Mal de Altura/metabolismo , Hipoxia/metabolismo , Edema/complicaciones , Citocinas , Inflamación/complicaciones , Oxígeno
6.
Arch Biochem Biophys ; 729: 109393, 2022 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-36084697

RESUMEN

High-altitude cerebral edema (HACE), a potentially lethal disease, is associated with a time-dependent exposure to altitude-related hypobaric hypoxia (HH) and has reportedly been associated with microglia hyperactivation. Catechins are substances with good antioxidant properties, among which (-)-epigallocatechin gallate (EGCG) may play a neuroprotective role through the inhibition of microglia overactivation; however, the function of its analog- (-)-epicatechin gallate (ECG)-requires further elucidation. The aim of the present study was to investigate whether ECG prevented HACE by inhibiting HH-activated microglia. Primary microglia exposed to lipopolysaccharide (LPS)/ATP were co-treated with EGCG, ECG, and (-)-epigallocatechin, and ECG and EGCG exerted significant anti-inflammatory and neuroprotective effects. ECG inhibited the NF-κB pathway to prevent the activation of microglia induced by 1% O2. In addition, ECG ameliorated the increase in brain water content and aquaporin 4 expression induced by HH in mice. ECG also reduced the number of Iba1+ microglia in the brain, the release of proinflammatory factors, and the recruitment of microglia to blood vessels in HH-exposed mice. The outcomes of the present study revealed that ECG alleviated hypoxic hyperactivated microglia, reduced the neuroinflammation and blood-brain barrier permeability, and prevented HACE by inhibiting NF-κB signaling.


Asunto(s)
Mal de Altura , Edema Encefálico , Fármacos Neuroprotectores , Adenosina Trifosfato/metabolismo , Mal de Altura/complicaciones , Mal de Altura/metabolismo , Animales , Antiinflamatorios/farmacología , Antioxidantes/farmacología , Acuaporina 4/metabolismo , Acuaporina 4/farmacología , Edema Encefálico/complicaciones , Edema Encefálico/tratamiento farmacológico , Edema Encefálico/prevención & control , Catequina/análogos & derivados , Hipoxia/complicaciones , Hipoxia/metabolismo , Lipopolisacáridos/farmacología , Ratones , Microglía/metabolismo , FN-kappa B/metabolismo , Fármacos Neuroprotectores/metabolismo , Agua/metabolismo
7.
Eur J Pharmacol ; 928: 175121, 2022 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-35777443

RESUMEN

High altitude cerebral edema (HACE) is a potentially life-threatening disease encountered at high altitudes. However, effective methods for HACE prophylaxis are limited. Convincing evidence confirms that oxidative stress induced by hypobaric hypoxia (HH) is one of the main factors that account for the development of HACE. 5,6,7,8-Tetrahydroxyflavone (THF), a flavone with four consecutive OH groups in ring A, exhibited excellent antioxidant activity in vitro and could attenuate HH induced injury in vivo. The aim of this study was to investigate the protective effect of THF against HACE and its underlying mechanisms. THF administration significantly suppressed HH induced oxidative stress by reducing the formation of hydrogen peroxide and malondialdehyde, by increasing the levels of glutathione and superoxide dismutase in brain tissue. Simultaneously, THF administration inhibited inflammatory responses by decreasing the levels of tumor necrosis factor-α, interleukin-1ß, and interleukin-6 in serum and brain tissue. In addition, THF administration mitigated the energy metabolism disorder induced by HACE as evidenced by decreased levels of lactic acid, lactate dehydrogenase and pyruvate kinase as well as increased ATP levels and ATPase activities. Furthermore, THF administration decreased the expression of matrix metalloproteinase-9, aquaporin 4, hypoxia-inducible factor-1α and vascular endothelial growth factor, which attenuated blood-brain barrier (BBB) disruption and brain edema. Additionally, THF administration improved HACE induced cognitive dysfunction. These results show that THF is a promising agent in the prevention and treatment of HACE.


Asunto(s)
Mal de Altura , Edema Encefálico , Flavonas , Altitud , Mal de Altura/tratamiento farmacológico , Mal de Altura/metabolismo , Mal de Altura/prevención & control , Animales , Edema Encefálico/tratamiento farmacológico , Edema Encefálico/metabolismo , Edema Encefálico/prevención & control , Flavonas/farmacología , Hipoxia/complicaciones , Hipoxia/tratamiento farmacológico , Ratas , Factor A de Crecimiento Endotelial Vascular
8.
Exp Mol Med ; 54(6): 777-787, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35672450

RESUMEN

At high altitude Andean region, hypoxia-induced excessive erythrocytosis (EE) is the defining feature of Monge's disease or chronic mountain sickness (CMS). At the same altitude, resides a population that has developed adaptive mechanism(s) to constrain this hypoxic response (non-CMS). In this study, we utilized an in vitro induced pluripotent stem cell model system to study both populations using genomic and molecular approaches. Our whole genome analysis of the two groups identified differential SNPs between the CMS and non-CMS subjects in the ARID1B region. Under hypoxia, the expression levels of ARID1B significantly increased in the non-CMS cells but decreased in the CMS cells. At the molecular level, ARID1B knockdown (KD) in non-CMS cells increased the levels of the transcriptional regulator GATA1 by 3-fold and RBC levels by 100-fold under hypoxia. ARID1B KD in non-CMS cells led to increased proliferation and EPO sensitivity by lowering p53 levels and decreasing apoptosis through GATA1 mediation. Interestingly, under hypoxia ARID1B showed an epigenetic role, altering the chromatin states of erythroid genes. Indeed, combined Real-time PCR and ATAC-Seq results showed that ARID1B modulates the expression of GATA1 and p53 and chromatin accessibility at GATA1/p53 target genes. We conclude that ARID1B is a novel erythroid regulator under hypoxia that controls various aspects of erythropoiesis in high-altitude dwellers.


Asunto(s)
Mal de Altura , Proteínas de Unión al ADN , Factores de Transcripción , Mal de Altura/genética , Mal de Altura/metabolismo , Cromatina/genética , Cromatina/metabolismo , Enfermedad Crónica , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Eritropoyesis/genética , Humanos , Hipoxia/genética , Hipoxia/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteína p53 Supresora de Tumor/genética
9.
CNS Neurosci Ther ; 28(8): 1183-1194, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35582960

RESUMEN

INTRODUCTION: Acute high-altitude hypoxia exposure causes multiple adverse neurological consequences. However, the exact mechanisms are still unclear, and there is no targeted treatment with few side effects. Excessive cerebral formaldehyde (FA) impairs numerous functions, and can be eliminated by nano-packed coenzyme Q10 (CoQ10). AIMS: In this study, we aimed to investigate whether cerebral FA was accumulated after hypobaric hypoxia exposure, and further explored the preventative effect of CoQ10 through FA elimination. RESULTS: Accumulated cerebral FA was found in C57BL/6 mice after acute high-altitude hypoxia exposure, which resulted in FA metabolic disturbance with the elevation of semicarbazide-sensitive amine oxidase, and declination of aldehyde dehydrogenase-2. Excessive FA was also found to induce neuronal ferroptosis in vivo. Excitingly, administration with CoQ10 for 3 days before acute hypobaric hypoxia reduced cerebral FA accumulation, alleviated subsequent neuronal ferroptosis, and preserved neurological functions. CONCLUSION: Cerebral FA accumulation mediates neurological deficits under acute hypobaric hypoxia, and CoQ10 supplementation may be a promising preventative strategy for visitors and sojourners at plateau.


Asunto(s)
Mal de Altura , Altitud , Mal de Altura/metabolismo , Animales , Formaldehído/toxicidad , Hipoxia/complicaciones , Hipoxia/metabolismo , Ratones , Ratones Endogámicos C57BL
10.
Neurochem Res ; 47(8): 2254-2262, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35552996

RESUMEN

We aimed to explore the protective effects and potential treatment mechanism of Epigallocatechin-3-gallate (EGCG) in an animal model of chronic exposure in a natural high-altitude hypoxia (HAH) environment. Behavioral alterations were assessed with the Morris water maze test. Iron accumulation in the hippocampus was detected by using DAB enhanced Perls' staining, MRI, qPCR and colorimetry, respectively. Oxidative stress (malondialdehyde, MDA), apoptosis (Caspase-3), and neural regeneration (brain-derived neurotrophic factor, BDNF) were detected by using ELISA and western blotting. Neural ultrastructural changes were evaluated by transmission electron microscopy (TEM). The results showed that learning and memory performance of rats decreased when exposure to HAH environment. It was followed by iron accumulation, dysfunctional iron metabolism, reduced BDNF and the upregulation of MDA and Caspase-3. TEM confirmed the ultrastructural changes in neurons and mitochondria. EGCG reduced HAH-induced cognitive impairment, iron deposition, oxidative stress, and apoptosis and promoted neuronal regeneration against chronic HAH-mediated neural injury.


Asunto(s)
Mal de Altura , Factor Neurotrófico Derivado del Encéfalo , Mal de Altura/metabolismo , Animales , Apoptosis , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Caspasa 3/metabolismo , Catequina/análogos & derivados , Cognición , Hipocampo/metabolismo , Hipoxia/tratamiento farmacológico , Hipoxia/metabolismo , Hierro/metabolismo , Aprendizaje por Laberinto , Neuronas/metabolismo , Estrés Oxidativo , Ratas , Regeneración
11.
Free Radic Biol Med ; 184: 99-113, 2022 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-35398201

RESUMEN

Chronic mountain sickness (CMS) is a high-altitude (HA) maladaptation syndrome characterised by elevated systemic oxidative-nitrosative stress (OXNOS) due to a free radical-mediated reduction in vascular nitric oxide (NO) bioavailability. To better define underlying mechanisms and vascular consequences, this study compared healthy male lowlanders (80 m, n = 10) against age/sex-matched highlanders born and bred in La Paz, Bolivia (3600 m) with (CMS+, n = 10) and without (CMS-, n = 10) CMS. Cephalic venous blood was assayed using electron paramagnetic resonance spectroscopy and reductive ozone-based chemiluminescence. Nutritional intake was assessed via dietary recall. Systemic vascular function and structure were assessed via flow-mediated dilatation, aortic pulse wave velocity and carotid intima-media thickness using duplex ultrasound and applanation tonometry. Basal systemic OXNOS was permanently elevated in highlanders (P = <0.001 vs. lowlanders) and further exaggerated in CMS+, reflected by increased hydroxyl radical spin adduct formation (P = <0.001 vs. CMS-) subsequent to liberation of free 'catalytic' iron consistent with a Fenton and/or nucleophilic addition mechanism(s). This was accompanied by elevated global protein carbonylation (P = 0.046 vs. CMS-) and corresponding reduction in plasma nitrite (P = <0.001 vs. lowlanders). Dietary intake of vitamins C and E, carotene, magnesium and retinol were lower in highlanders and especially deficient in CMS + due to reduced consumption of fruit and vegetables (P = <0.001 to 0.028 vs. lowlanders/CMS-). Systemic vascular function and structure were also impaired in highlanders (P = <0.001 to 0.040 vs. lowlanders) with more marked dysfunction observed in CMS+ (P = 0.035 to 0.043 vs. CMS-) in direct proportion to systemic OXNOS (r = -0.692 to 0.595, P = <0.001 to 0.045). Collectively, these findings suggest that lifelong exposure to iron-catalysed systemic OXNOS, compounded by a dietary deficiency of antioxidant micronutrients, likely contributes to the systemic vascular complications and increased morbidity/mortality in CMS+. TRIAL REGISTRY: ClinicalTrials.gov; No: NCT01182792; URL: www.clinicaltrials.gov.


Asunto(s)
Mal de Altura , Altitud , Mal de Altura/metabolismo , Grosor Intima-Media Carotídeo , Enfermedad Crónica , Espectroscopía de Resonancia por Spin del Electrón , Radicales Libres , Humanos , Hierro , Masculino , Análisis de la Onda del Pulso
12.
Drug Discov Today ; 26(11): 2774-2781, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34302973

RESUMEN

Cellular exposure to extreme environments leads to the expression of multiple proteins that participate in pathophysiological manifestations. Hypobaric hypoxia at high altitude (HA) generates reactive oxygen species (ROS) that can damage telomeres. Tankyrase (TNKS) belongs to multiple telomeric protein complexes and is actively involved in DNA damage repair. Although published research on TNKS indicates its possible role in cancer and other hypoxic diseases, its role in HA sicknesses remains elusive. Understanding the roles of telomeres, telomerase, and TNKS could ameliorate physiological issues experienced at HA. In addition, telomeric TNKS could be a potential biomarker in hypoxia-induced sicknesses or acclimatization. Thus, a new research avenue on TNKS linked to HA sickness might lead to the discovery of drugs for hypobaric hypoxia.


Asunto(s)
Mal de Altura/metabolismo , Altitud , Edema Encefálico/metabolismo , Hipertensión Pulmonar/metabolismo , Tanquirasas/metabolismo , Telomerasa/metabolismo , Telómero/metabolismo , Daño del ADN , Reparación del ADN , Descubrimiento de Drogas , Humanos , Factor 1 Inducible por Hipoxia/metabolismo , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo
13.
Exp Mol Med ; 53(1): 125-135, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33473144

RESUMEN

Monge's disease (chronic mountain sickness (CMS)) is a maladaptive condition caused by chronic (years) exposure to high-altitude hypoxia. One of the defining features of CMS is excessive erythrocytosis with extremely high hematocrit levels. In the Andean population, CMS prevalence is vastly different between males and females, being rare in females. Furthermore, there is a sharp increase in CMS incidence in females after menopause. In this study, we assessed the role of sex hormones (testosterone, progesterone, and estrogen) in CMS and non-CMS cells using a well-characterized in vitro erythroid platform. While we found that there was a mild (nonsignificant) increase in RBC production with testosterone, we observed that estrogen, in physiologic concentrations, reduced sharply CD235a+ cells (glycophorin A; a marker of RBC), from 56% in the untreated CMS cells to 10% in the treated CMS cells, in a stage-specific and dose-responsive manner. At the molecular level, we determined that estrogen has a direct effect on GATA1, remarkably decreasing the messenger RNA (mRNA) and protein levels of GATA1 (p < 0.01) and its target genes (Alas2, BclxL, and Epor, p < 0.001). These changes result in a significant increase in apoptosis of erythroid cells. We also demonstrate that estrogen regulates erythropoiesis in CMS patients through estrogen beta signaling and that its inhibition can diminish the effects of estrogen by significantly increasing HIF1, VEGF, and GATA1 mRNA levels. Taken altogether, our results indicate that estrogen has a major impact on the regulation of erythropoiesis, particularly under chronic hypoxic conditions, and has the potential to treat blood diseases, such as high altitude severe erythrocytosis.


Asunto(s)
Mal de Altura/sangre , Eritrocitos/efectos de los fármacos , Estrógenos/farmacología , Policitemia/metabolismo , Mal de Altura/metabolismo , Células Cultivadas , Eritrocitos/metabolismo , Estrógenos/metabolismo , Femenino , Factor de Transcripción GATA1/genética , Factor de Transcripción GATA1/metabolismo , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Masculino , Policitemia/sangre , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
14.
Eur J Appl Physiol ; 121(1): 279-285, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33052430

RESUMEN

PURPOSE: Long static or intense dynamic apnoea-like high-altitude exposure is inducing hypoxia. Adenosine is known to participate to the adaptive response to hypoxia leading to the control of heart rate, blood pressure and vasodilation. Extracellular adenosine level is controlled through the equilibrative nucleoside transporter 1 (ENT-1) and the enzyme adenosine deaminase (ADA). The aim of this study was to determine the control of adenosine blood level (ABL) via ENT-1 and ADA during apnoea-induced hypoxia in elite freedivers was similar to high-altitude adaptation. METHODS: Ten freediver champions and ten controls were studied. Biological (e.g. ENT-1, ADA, ABL, PaO2, PaCO2 and pH) and cardiovascular (e.g. heart rate, arterial pressure) parameters were measured at rest and after a submaximal dry static apnoea. RESULTS: In freedivers, ABL was higher than in control participants in basal condition and increased more in response to apnoea. Also, freedivers showed an ADA increased in response to apnoea. Finally, ENT-1 level and function were reduced for the free divers. CONCLUSION: Our results suggest in freedivers the presence of an adaptive mechanism similar to the one observed in human exposed to chronic hypoxia induced by high-altitude environment.


Asunto(s)
Adaptación Fisiológica , Adenosina/sangre , Mal de Altura/metabolismo , Contencion de la Respiración , Buceo/fisiología , Tranportador Equilibrativo 1 de Nucleósido/metabolismo , Adenosina Desaminasa/metabolismo , Adulto , Mal de Altura/fisiopatología , Presión Sanguínea , Femenino , Frecuencia Cardíaca , Humanos , Masculino , Persona de Mediana Edad
15.
Nat Commun ; 11(1): 4928, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-33004791

RESUMEN

High-altitude adaptation of Tibetans represents a remarkable case of natural selection during recent human evolution. Previous genome-wide scans found many non-coding variants under selection, suggesting a pressing need to understand the functional role of non-coding regulatory elements (REs). Here, we generate time courses of paired ATAC-seq and RNA-seq data on cultured HUVECs under hypoxic and normoxic conditions. We further develop a variant interpretation methodology (vPECA) to identify active selected REs (ASREs) and associated regulatory network. We discover three causal SNPs of EPAS1, the key adaptive gene for Tibetans. These SNPs decrease the accessibility of ASREs with weakened binding strength of relevant TFs, and cooperatively down-regulate EPAS1 expression. We further construct the downstream network of EPAS1, elucidating its roles in hypoxic response and angiogenesis. Collectively, we provide a systematic approach to interpret phenotype-associated noncoding variants in proper cell types and relevant dynamic conditions, to model their impact on gene regulation.


Asunto(s)
Aclimatación/genética , Cromatina/metabolismo , Etnicidad/genética , Redes Reguladoras de Genes , Modelos Genéticos , Altitud , Mal de Altura/etnología , Mal de Altura/genética , Mal de Altura/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Hipoxia de la Célula/genética , Células Cultivadas , Cromatina/genética , Secuenciación de Inmunoprecipitación de Cromatina , Resistencia a la Enfermedad/genética , Femenino , Regulación de la Expresión Génica , Células Endoteliales de la Vena Umbilical Humana , Humanos , Hipoxia/genética , Hipoxia/metabolismo , Oxígeno/metabolismo , Polimorfismo de Nucleótido Simple , Embarazo , Cultivo Primario de Células , RNA-Seq , Elementos Reguladores de la Transcripción/genética , Selección Genética , Tibet/etnología , Factores de Transcripción/metabolismo , Secuenciación Completa del Genoma
16.
Am J Physiol Lung Cell Mol Physiol ; 319(2): L360-L368, 2020 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-32692577

RESUMEN

Hypobaric hypoxia poses stress to sojourners traveling to high-altitude. A cascade of physiological changes occurs to cope with or adapt to hypobaric hypoxia. However, an insufficient physiological response to the hypoxic condition resulting from imbalanced vascular homeostasis pathways results in high-altitude pulmonary edema (HAPE). The present study aims to identify the implication of miRNAs associating with HAPE and adaptation. We analyzed the expression of 1,113 miRNAs in HAPE-patients (HAPE-p), HAPE-free controls (HAPE-f), and highland natives (HLs). Based on miRNA profiling and in silico analyses, miR-124-3p emerged relevantly. We observed a significant overexpression of miR-124-3p in HAPE-p. In silico analyses revealed a direct interaction of miR-124-3p with vascular homeostasis and hypoxia-associated genes NOS3 (endothelial nitric oxide synthase), Apelin, and ETS1 (V-Ets avian erythroblastosis virus E2 oncogene homolog 1). Moreover, the transcript and biolevel expression of these genes were significantly decreased in HAPE-p when compared with HAPE-f or HLs. Our in vitro analysis in human umbilical vein endothelial cells demonstrated a significant knockdown of these genes both at transcript and protein levels following miR-124-3p overexpression. Conclusively, our results showed that miR-124-3p might play a plausible role in HAPE pathophysiology by inhibiting the expression of NOS3, Apelin, and ETS1.


Asunto(s)
Mal de Altura/sangre , Mal de Altura/metabolismo , Hipertensión Pulmonar/sangre , Hipertensión Pulmonar/metabolismo , Hipoxia/sangre , Hipoxia/metabolismo , MicroARNs/sangre , Edema Pulmonar/sangre , Edema Pulmonar/metabolismo , Adaptación Fisiológica/fisiología , Adulto , Altitud , Apelina/metabolismo , Línea Celular , Femenino , Células HEK293 , Células Endoteliales de la Vena Umbilical Humana , Humanos , Masculino , Óxido Nítrico Sintasa de Tipo III/metabolismo , Proteína Proto-Oncogénica c-ets-1/metabolismo , Adulto Joven
17.
Curr Opin Hematol ; 27(3): 155-162, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32141895

RESUMEN

PURPOSE OF REVIEW: The erythrocyte is the most abundant cell type in our body, acting as both a carrier/deliverer and sensor of oxygen (O2). Erythrocyte O2 delivery capacity is finely regulated by sophisticated metabolic control. In recent years, unbiased and robust human metabolomics screening and mouse genetic studies have advanced erythroid research revealing the differential role of erythrocyte hypoxic metabolic reprogramming in normal individuals at high altitudes and patients facing hypoxia, such as sickle cell disease (SCD) and chronic kidney disease (CKD). Here we summarize recent progress and highlight potential therapeutic possibilities. RECENT FINDINGS: Initial studies showed that elevated soluble CD73 (sCD73, converts AMP to adenosine) results in increased circulating adenosine that activates the A2B adenosine receptor (ADORA2B). Signaling through this axis is co-operatively strengthened by erythrocyte-specific synthesis of sphingosine-1-phosphate (S1P). Ultimately, these mechanisms promote the generation of 2,3-bisphosphoglycerate (2,3-BPG), an erythrocyte-specific allosteric modulator that decreases haemoglobin--O2-binding affinity, and thus, induces deoxygenated sickle Hb (deoxyHbS), deoxyHbS polymerization, sickling, chronic inflammation and tissue damage in SCD. Similar to SCD, plasma adenosine and erythrocyte S1P are elevated in humans ascending to high altitude. At high altitude, these two metabolites are beneficial to induce erythrocyte metabolic reprogramming and the synthesis of 2,3-BPG, and thus, increase O2 delivery to counteract hypoxic tissue damage. Follow-up studies showed that erythrocyte equilibrative nucleoside transporter 1 (eENT1) is a key purinergic cellular component controlling plasma adenosine in humans at high altitude and mice under hypoxia and underlies the quicker and higher elevation of plasma adenosine upon re-ascent because of prior hypoxia-induced degradation of eENT1. More recent studies demonstrated the beneficial role of erythrocyte ADORA2B-mediated 2,3-BPG production in CKD. SUMMARY: Taken together, these findings revealed the differential role of erythrocyte hypoxic metabolic reprogramming in normal humans at high altitude and patients with CKD vs. SCD patients and immediately suggest differential and precision therapies to counteract hypoxia among these groups.


Asunto(s)
Mal de Altura/metabolismo , Anemia de Células Falciformes/metabolismo , Reprogramación Celular , Eritrocitos/metabolismo , Insuficiencia Renal Crónica/metabolismo , Mal de Altura/patología , Anemia de Células Falciformes/patología , Animales , Hipoxia de la Célula , Eritrocitos/patología , Humanos , Ratones , Oxígeno/metabolismo , Insuficiencia Renal Crónica/patología
18.
Biochim Biophys Acta Mol Basis Dis ; 1866(7): 165769, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32184133

RESUMEN

Brain is well known for its disproportionate oxygen consumption and high energy-budget for optimal functioning. The decrease in oxygen supply to brain, thus, necessitates rapid activation of adaptive pathways - the absence of which manifest into vivid pathological conditions. Amongst these, oxygen sensing in glio-vascular milieu and H2S-dependent compensatory increase in cerebral blood flow (CBF) is a major adaptive response. We had recently demonstrated that the levels of H2S were significantly decreased during chronic hypobaric hypoxia (HH)-induced neuro-pathological effects. The mechanistic basis of this phenomenon, however, remained to be deciphered. We, here, describe experimental evidence for marked limitation of cysteine during HH - both in animal model as well as human volunteers ascending to high altitude. We show that the preservation of brain cysteine level, employing cysteine pro-drug (N-acetyl-L-cysteine, NAC), markedly curtailed effects of HH - not only on endogenous H2S levels but also, impairment of spatial reference memory in our animal model. We, further, present multiple lines of experimental evidence that the limitation of cysteine was causally governed by physiological propensity of brain to utilize cysteine, in cystathionine beta synthase (CBS)-dependent manner, past its endogenous replenishment potential. Notably, decrease in the levels of brain cysteine manifested despite positive effect (up-regulation) of HH on endogenous cysteine maintenance pathways and thus, qualifying cysteine as a conditionally essential nutrient (CEN) during HH. In brief, our data supports an adaptive, physiological role of CBS-mediated cysteine-utilization pathway - activated to increase endogenous levels of H2S - for optimal responses of brain to hypobaric hypoxia.


Asunto(s)
Mal de Altura/metabolismo , Encéfalo/metabolismo , Cistationina betasintasa/genética , Cisteína/metabolismo , Sulfuro de Hidrógeno/metabolismo , Acetilcisteína/farmacología , Adaptación Fisiológica , Adulto , Mal de Altura/tratamiento farmacológico , Mal de Altura/genética , Mal de Altura/patología , Animales , Encéfalo/patología , Circulación Cerebrovascular/efectos de los fármacos , Circulación Cerebrovascular/genética , Cistationina betasintasa/metabolismo , Modelos Animales de Enfermedad , Metabolismo Energético/genética , Humanos , Hipoxia/tratamiento farmacológico , Hipoxia/genética , Hipoxia/metabolismo , Masculino , Consumo de Oxígeno/genética , Profármacos/farmacología , Ratas , Adulto Joven
19.
Aging (Albany NY) ; 12(5): 4247-4267, 2020 02 29.
Artículo en Inglés | MEDLINE | ID: mdl-32112644

RESUMEN

Hypoxia-related microRNAs (miRNAs) are involved in the pathogenesis of various diseases. Because potential variations in miRNA expression mediated by hypoxic lung injury at high altitude remain incompletely characterized, we used a rat model to investigate the biochemical and miRNA changes induced by high-altitude hypoxia. After 24, 48, or 72 h of hypoxic exposure, expression of VEGF/Notch pathway-related proteins were increased in rat lung tissues. Microarray screening of hypoxic lung samples revealed 57 differentially expressed miRNAs, 19 of which were related to the VEGF/Notch signaling pathway. We verified that the top downregulated miRNA (miR-203a-3p) suppresses VEGF-A translation through direct binding and also indirectly reduces Notch1, VEGFR2, and Hes1 levels, which restricts the angiogenic capacity of pulmonary microvascular endothelial cells in vitro. These findings may aid in the development of new therapeutic strategies for the prevention and treatment of hypoxic lung injury at high altitude.


Asunto(s)
Regulación hacia Abajo , Hipoxia/metabolismo , Pulmón/metabolismo , MicroARNs/metabolismo , Receptores Notch/genética , Transducción de Señal/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Mal de Altura/genética , Mal de Altura/metabolismo , Animales , Modelos Animales de Enfermedad , Hipoxia/genética , Masculino , MicroARNs/genética , Ratas , Ratas Sprague-Dawley
20.
Int J Mol Sci ; 21(1)2020 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-31906427

RESUMEN

Hypoxia, or lack of oxygen, can occur in both physiological (high altitude) and pathological conditions (respiratory diseases). In this narrative review, we introduce high altitude pulmonary edema (HAPE), acute respiratory distress syndrome (ARDS), Chronic Obstructive Pulmonary Disease (COPD), and Cystic Fibrosis (CF) as examples of maladaptation to hypoxia, and highlight some of the potential mechanisms influencing the prognosis of the affected patients. Among the specific pathways modulated in response to hypoxia, iron metabolism has been widely explored in recent years. Recent evidence emphasizes hepcidin as highly involved in the compensatory response to hypoxia in healthy subjects. A less investigated field in the adaptation to hypoxia is the sphingolipid (SPL) metabolism, especially through Ceramide and sphingosine 1 phosphate. Both individually and in concert, iron and SPL are active players of the (mal)adaptation to physiological hypoxia, which can result in the pathological HAPE. Our aim is to identify some pathways and/or markers involved in the physiological adaptation to low atmospheric pressures (high altitudes) that could be involved in pathological adaptation to hypoxia as it occurs in pulmonary inflammatory diseases. Hepcidin, Cer, S1P, and their interplay in hypoxia are raising growing interest both as prognostic factors and therapeutical targets.


Asunto(s)
Mal de Altura/metabolismo , Fibrosis Quística/metabolismo , Hipertensión Pulmonar/metabolismo , Hipoxia/fisiopatología , Hierro/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Síndrome de Dificultad Respiratoria/metabolismo , Esfingolípidos/metabolismo , Adaptación Fisiológica , Ceramidas/metabolismo , Hepcidinas/metabolismo , Humanos , Hipoxia/metabolismo , Lisofosfolípidos/metabolismo , Esfingosina/análogos & derivados , Esfingosina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA