Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 204
Filtrar
1.
Apoptosis ; 29(9-10): 1600-1618, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39110356

RESUMEN

High-altitude pulmonary edema (HAPE) is a fatal threat for sojourners who ascend rapidly without sufficient acclimatization. Acclimatized sojourners and adapted natives are both insensitive to HAPE but have different physiological traits and molecular bases. In this study, based on GSE52209, the gene expression profiles of HAPE patients were compared with those of acclimatized sojourners and adapted natives, with the common and divergent differentially expressed genes (DEGs) and their hub genes identified, respectively. Bioinformatic methodologies for functional enrichment analysis, immune infiltration, diagnostic model construction, competing endogenous RNA (ceRNA) analysis and drug prediction were performed to detect potential biological functions and molecular mechanisms. Next, an array of in vivo experiments in a HAPE rat model and in vitro experiments in HUVECs were conducted to verify the results of the bioinformatic analysis. The enriched pathways of DEGs and immune landscapes for HAPE were significantly different between sojourners and natives, and the common DEGs were enriched mainly in the pathways of development and immunity. Nomograms revealed that the upregulation of TNF-α and downregulation of RPLP0 exhibited high diagnostic efficiency for HAPE in both sojourners and natives, which was further validated in the HAPE rat model. The addition of TNF-α and RPLP0 knockdown activated apoptosis signaling in endothelial cells (ECs) and enhanced endothelial permeability. In conclusion, TNF-α and RPLP0 are shared biomarkers and molecular bases for HAPE susceptibility during the acclimatization/adaptation/maladaptation processes in sojourners and natives, inspiring new ideas for predicting and treating HAPE.


Asunto(s)
Mal de Altura , Apoptosis , Células Endoteliales , Proteínas Ribosómicas , Factor de Necrosis Tumoral alfa , Animales , Humanos , Masculino , Ratas , Altitud , Mal de Altura/genética , Mal de Altura/metabolismo , Mal de Altura/patología , Apoptosis/genética , Células Endoteliales/metabolismo , Células Endoteliales/patología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Hipertensión Pulmonar/genética , Hipertensión Pulmonar/metabolismo , Hipertensión Pulmonar/patología , Ratas Sprague-Dawley , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo , Proteínas Ribosómicas/genética , Proteínas Ribosómicas/metabolismo
2.
Invest Ophthalmol Vis Sci ; 65(10): 19, 2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-39133471

RESUMEN

Purpose: High altitude retinopathy (HAR) is a retinal functional disorder caused by inadequate adaptation after exposure to high altitude. However, the cellular and molecular mechanisms underlying retinal dysfunction remain elusive. Retinal ganglion cell (RGC) injury is the most important pathological basis for most retinal and optic nerve diseases. Studies focusing on RGC injury after high-altitude exposure (HAE) are scanty. Therefore, the present study sought to explore both functional and morphological alterations of RGCs after HAE. Methods: A mouse model of acute hypobaric hypoxia was established by mimicking the conditions of a high altitude of 5000 m. After HAE for 2, 4, 6, 10, 24, and 72 hours, the functional and morphological alterations of RGCs were assessed using retinal hematoxylin and eosin (H&E) sections, retinal whole mounts, transmission electron microscopy (TEM), and the photopic negative response (PhNR) of the electroretinogram. Results: Compared with the control group, the thickness of the ganglion cell layer and retinal nerve fiber layer increased significantly, RGC loss remained significant, and the amplitudes of a-wave, b-wave, and PhNR were significantly reduced after HAE. In addition, RGCs and their axons exhibited an abnormal ultrastructure after HAE, including nuclear membrane abnormalities, uneven distribution of chromatin in the nucleus, decreased cytoplasmic electron density, widening and vacuolization of the gap between axons, loosening and disorder of myelin sheath structure, widening of the gap between myelin sheath and axon membrane, decreased axoplasmic density, unclear microtubule and nerve fiber structure, and abnormal mitochondrial structure (mostly swollen, with widened membrane gaps and reduced cristae and vacuolization). Conclusions: The study findings confirm that the morphology and function of RGCs are damaged after HAE. These findings lay the foundation for further study of the specific molecular mechanisms of HAR and promote the effective prevention.


Asunto(s)
Modelos Animales de Enfermedad , Electrorretinografía , Ratones Endogámicos C57BL , Microscopía Electrónica de Transmisión , Células Ganglionares de la Retina , Animales , Células Ganglionares de la Retina/patología , Células Ganglionares de la Retina/ultraestructura , Ratones , Masculino , Mal de Altura/fisiopatología , Mal de Altura/patología , Enfermedades de la Retina/fisiopatología , Enfermedades de la Retina/etiología , Enfermedades de la Retina/patología , Altitud , Enfermedad Aguda
3.
Cell Commun Signal ; 22(1): 374, 2024 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-39054523

RESUMEN

BACKGROUND: High-altitude cerebral edema (HACE) is considered an end-stage acute mountain sickness (AMS) that typically occurs in people after rapid ascent to 2500 m or more. While hypoxia is a fundamental feature of the pathophysiological mechanism of HACE, emerging evidence suggests that inflammation serves as a key risk factor in the occurrence and development of this disease. However, little is known about the molecular mechanism underlying their crosstalk. METHODS: A mouse HACE model was established by combination treatment with hypobaric hypoxia exposure and lipopolysaccharides (LPS) stimulation. Lactylated-proteomic analysis of microglia was performed to reveal the global profile of protein lactylation. Molecular modeling was applied to evaluate the 3-D modeling structures. A combination of experimental approaches, including western blotting, quantitative real-time reverse transcriptionpolymerase chain reaction (qRT-PCR), and enzyme-linked immunosorbent assay (ELISA), confocal microscopy and RNA interference, were used to explore the underlying molecular mechanisms. RESULTS: We found that hypoxia exposure increased the lactate concentration and lactylation in mouse HACE model. Moreover, hypoxia aggravated the microglial neuroinflammatory response in a lactate-dependent manner. Global profiling of protein lactylation has shown that a large quantity of lysine-lactylated proteins are induced by hypoxia and preferentially occur in protein complexes, such as the NuRD complex, ribosome biogenesis complex, spliceosome complex, and DNA replication complex. The molecular modeling data indicated that lactylation could affect the 3-D theoretical structure and increase the solvent accessible surface area of HDAC1, MTA1 and Gatad2b, the core members of the NuRD complex. Further analysis by knockdown or selectively inhibition indicated that the NuRD complex is involved in hypoxia-mediated aggravation of inflammation. CONCLUSIONS: These results revealed a comprehensive profile of protein lactylation in microglia and suggested that protein lysine lactylation plays an important role in the regulation of protein function and subsequently contributes to the neuroinflammatory response under hypoxic conditions.


Asunto(s)
Edema Encefálico , Microglía , Microglía/metabolismo , Microglía/patología , Animales , Edema Encefálico/metabolismo , Edema Encefálico/patología , Ratones , Mal de Altura/metabolismo , Mal de Altura/patología , Masculino , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad , Lipopolisacáridos/farmacología , Altitud , Proteómica
4.
Respir Physiol Neurobiol ; 327: 104286, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38825093

RESUMEN

High-altitude environments present extreme conditions characterized by low barometric pressure and oxygen deficiency, which can disrupt brain functioning and cause edema formation. The objective of the present study is to investigate several biomolecule expressions and their role in the development of High Altitude Cerebral Edema in a rat model. Specifically, the study focuses on analyzing the changes in total arginase, nitric oxide, and lipid peroxidation (MDA) levels in the brain following acute hypobaric hypoxic exposure (7620 m, SO2=8.1 %, for 24 h) along with the histopathological assessment. The histological examination revealed increased TNF-α activity, and an elevated number of mast cells in the brain, mainly in the hippocampus and cerebral cortex. The research findings demonstrated that acute hypobaric hypoxic causes increased levels of apoptotic cells, shrinkage, and swelling of neurons, accompanied by the formation of protein aggregation in the brain parenchyma. Additionally, the level of nitric oxide and MDA was found to have increased (p<0.0001), however, the level of arginase decreased indicating active lipid peroxidation and redox imbalance in the brain. This study provides insights into the pathogenesis of HACE by evaluating some biomolecules that play a pivotal role in the inflammatory response and the redox landscape in the brain. The findings could have significant implications for understanding the neuronal dysfunction and the pathological mechanisms underlying HACE development.


Asunto(s)
Mal de Altura , Edema Encefálico , Estrés Oxidativo , Animales , Edema Encefálico/metabolismo , Edema Encefálico/etiología , Edema Encefálico/patología , Estrés Oxidativo/fisiología , Masculino , Mal de Altura/metabolismo , Mal de Altura/patología , Ratas , Modelos Animales de Enfermedad , Peroxidación de Lípido/fisiología , Encéfalo/metabolismo , Encéfalo/patología , Óxido Nítrico/metabolismo , Ratas Wistar , Enfermedades Neuroinflamatorias/metabolismo , Arginasa/metabolismo
5.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 54(6): 1269-1275, 2023 Nov 20.
Artículo en Chino | MEDLINE | ID: mdl-38162056

RESUMEN

Objective: To establish an animal model of high-altitude cerebral edema (HACE), to explore the altitude and oxygen partial pressure conditions that can lead to obvious clinical manifestations of HACE, and to lay the foundation for further research of the pathogenic mechanisms and intervention strategies of HACE. Methods: Male BALB/c mice of 8 weeks old were randomly assigned to Control and HACE groups. The Control group (n=10) was treated with normobaric and normoxic conditions, while the HACE groups were placed in hypobaric hypoxic (HH) chambers for the durations of 6 h, 12 h, 24 h, 48 h and 72 h, respectively, receiving treatments of simulated HH conditions at the altitudes of 4000 m (n=10 for each group receiving different durations of HH treatment), 5000 m (n=10 for each group receiving different durations of HH treatment), and 6000 m (n=10 for each group receiving different durations of HH treatment). HE staining was performed to observe the morphological changes of the brain tissue and the appropriate simulated altitude conditions were selected accordingly for the construction and evaluation of the best HACE model. The HACE model was evaluated in the following ways, the mouse brain was weighed and the cerebral edema was measured accordingly, Evans blue (EB) was injected to determine the permeability of the blood-brain barrier (BBB), and the cell apoptosis was determined by immunofluorescence staining. Results: There were no deaths in the groups treated with the HH conditions of the altitudes of 4000 m and 5000 m, while the mortality in the 6000 m altitude treatment groups was 12.2%. HE staining showed no significant changes in brain morphology or structure in the group receiving HH treatment for the altitude of 4000 m. A small amount of brain cell edema was observed in the groups receiving 48 h and 72 h of HH treatment for the altitude of 5000 m. The groups receiving HH treatment for the altitude of 6000 m demonstrated the most prominent modeling effect. HE staining showed increased volume and swelling of brain cells in all the 6000 m groups, especially in the 24 h, 48 h and 72 h treatment groups. In all the 6000 m groups, cell arrangement disorder, gap enlargement, and nuclear contraction were observed. Evaluation of the modeling effect demonstrated that, in the HACE mice model constructed with the HH conditions for the altitude of 6000 m, cerebral edema and EB permeability increased after 12 h HH treatment and there was no obvious apoptosis in the modeling groups receiving different durations of treatment. Conclusion: The HACE model can be established effectively by simulating conditions at the altitude of 6000 m (the atmospheric pressure being 47.19 kPa and the oxygen partial pressure being 9.73 kPa) with a HH chamber.


Asunto(s)
Mal de Altura , Edema Encefálico , Ratones , Animales , Masculino , Altitud , Edema Encefálico/etiología , Mal de Altura/metabolismo , Mal de Altura/patología , Encéfalo/metabolismo , Hipoxia/patología , Modelos Animales de Enfermedad , Oxígeno
6.
J Nepal Health Res Counc ; 20(2): 354-360, 2022 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-36550712

RESUMEN

BACKGROUND: Trekkers in high altitude of Himalayas could lead to Acute Mountain Sickness and High Altitude Cerebral Edema. This study was conducted to evaluate magnetic resonance imaging findings among the clinically suspected High Altitude Cerebral Edema patients rescued from high altitudes in Nepal Himalayas. METHODS: 49 patients with clinically suspected High Altitude Cerebral Edema were retrospectively evaluated in this cross-sectional study who were sent for a brain magnetic resonance imaging. They were categorized in 3 groups according to the magnetic resonance imaging features in this study. RESULTS: There was a slight male preponderance. 6 patients (12.25%) had magnetic resonance imaging findings highly suggestive of High Altitude Cerebral Edema. 5 patients had T2 high signal intensity and restricted diffusion in the splenium of corpus callosum of which 3 had features of microhemorrhage. One patient with normal brain morphology and intensity in T1, T2, and FLAIR images showed innumerable variable-sized microhemorrhages in Susceptibility Weighted Imaging. 14 of patients showed various T2 and FLAIR white matter high signal intensity without restricted diffusion. And one patient had features of subacute lacunar infarcts. 28 patients (57.14 %) showed no abnormal signal changes in the magnetic resonance imaging scan. CONCLUSIONS: Typical magnetic resonance imaging features of cytotoxic edema in corpus callosum and microhemorrhage in the patients with High Altitude Cerebral Edema further support the findings in other similar studies. T2 white matter hyperintensities in deep, subcortical or periventricular location and lacunar infarcts could be seen in High Altitude Cerebral Edema. Normal magnetic resonance imaging of the brain is not infrequent.


Asunto(s)
Mal de Altura , Edema Encefálico , Accidente Vascular Cerebral Lacunar , Humanos , Masculino , Mal de Altura/diagnóstico por imagen , Mal de Altura/patología , Edema Encefálico/diagnóstico por imagen , Edema Encefálico/etiología , Edema Encefálico/patología , Altitud , Estudios Retrospectivos , Estudios Transversales , Nepal , Imagen por Resonancia Magnética
7.
Eur J Neurol ; 29(10): 3112-3116, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35726171

RESUMEN

BACKGROUND AND PURPOSE: Animal studies suggest that exposure to severe ambient hypoxia for several days may have beneficial long-term effects on neurodegenerative diseases. Because, the acute risks of exposing human beings to prolonged severe hypoxia on brain structure and function are uncertain, we conducted a pilot study in healthy persons. METHODS: We included two professional mountaineers (participants A and B) in a 35-day study comprising an acclimatization period and 14 consecutive days with oxygen concentrations between 8% and 8.8%. They underwent cerebral magnetic resonance imaging at seven time points and a cognitive test battery covering a spectrum of cognitive domains at 27 time points. We analysed blood neuron specific enolase and neurofilament light chain levels before, during, and after hypoxia. RESULTS: In hypoxia, white matter volumes increased (maximum: A, 4.3% ± 0.9%; B, 4.5% ± 1.9%) whilst gray matter volumes (A, -1.5% ± 0.8%; B, -2.5% ± 0.9%) and cerebrospinal fluid volumes (A, -2.7% ± 2.4%; B, -5.9% ± 8.2%) decreased. Furthermore, the number (A, 11-17; B, 26-126) and volumes (A, 140%; B, 285%) of white matter hyperintensities increased in hypoxia but had returned to baseline after a 3.5-month recovery phase. Diffusion weighted imaging of the white matter indicated cytotoxic edema formation. We did not observe changes in cognitive performance or biochemical brain injury markers. DISCUSSION: In highly selected healthy individuals, severe sustained normobaric hypoxia over 2 weeks elicited reversible changes in brain morphology without clinically relevant changes in cognitive function or brain injury markers. The finding may pave the way for future translational studies assessing the therapeutic potential of hypoxia in neurodegenerative diseases.


Asunto(s)
Mal de Altura , Lesiones Encefálicas , Mal de Altura/diagnóstico por imagen , Mal de Altura/etiología , Mal de Altura/patología , Animales , Biomarcadores , Encéfalo/diagnóstico por imagen , Encéfalo/patología , Lesiones Encefálicas/complicaciones , Lesiones Encefálicas/patología , Humanos , Hipoxia/complicaciones , Hipoxia/patología , Imagen por Resonancia Magnética , Proyectos Piloto
8.
Front Endocrinol (Lausanne) ; 13: 831369, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35222286

RESUMEN

Exposure to hypobaric hypoxia at high altitude puts mountaineers at risk of acute mountain sickness. The carbonic anhydrase inhibitor acetazolamide is used to accelerate acclimatization, when it is not feasible to make a controlled and slow ascend. Studies in rodents have suggested that exposure to hypobaric hypoxia deteriorates bone integrity and reduces bone strength. The study investigated the effect of treatment with acetazolamide and the bisphosphonate, zoledronate, on the skeletal effects of exposure to hypobaric hypoxia. Eighty 16-week-old female RjOrl : SWISS mice were divided into five groups: 1. Baseline; 2. Normobaric; 3. Hypobaric hypoxia; 4. Hypobaric hypoxia + acetazolamide, and 5. Hypobaric hypoxia + zoledronate. Acetazolamide was administered in the drinking water (62 mg/kg/day) for four weeks, and zoledronate (100 µg/kg) was administered as a single subcutaneous injection at study start. Exposure to hypobaric hypoxia significantly increased lung wet weight and decreased femoral cortical thickness. Trabecular bone was spared from the detrimental effects of hypobaric hypoxia, although a trend towards reduced bone volume fraction was found at the L4 vertebral body. Treatment with acetazolamide did not have any negative skeletal effects, but could not mitigate the altitude-induced bone loss. Zoledronate was able to prevent the altitude-induced reduction in cortical thickness. In conclusion, simulated high altitude affected primarily cortical bone, whereas trabecular bone was spared. Only treatment with zoledronate prevented the altitude-induced cortical bone loss. The study provides preclinical support for future studies of zoledronate as a potential pharmacological countermeasure for altitude-related bone loss.


Asunto(s)
Acetazolamida/uso terapéutico , Mal de Altura , Altitud , Hueso Esponjoso/efectos de los fármacos , Hueso Cortical/efectos de los fármacos , Ácido Zoledrónico/uso terapéutico , Absorciometría de Fotón , Mal de Altura/patología , Mal de Altura/fisiopatología , Animales , Densidad Ósea , Hueso Esponjoso/patología , Hueso Cortical/patología , Femenino , Ratones , Músculo Cuádriceps/patología
9.
Mol Med Rep ; 25(2)2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34913080

RESUMEN

The aim of the present study was to investigate the changes in lung histomorphology and oxidative stress, as well as the expression of interleukin (IL)­17C and other inflammatory factors during acute mountain sickness (AMS) in male Sprague­Dawley rats and to explore the underlying mechanism. Rats were randomly divided into a control group (0 h) and three hypoxia stress groups, exposed to low­pressure oxygen storage at a simulated altitude of 6,000 m for 24, 48 and 72 h, respectively. Morphological changes in lung tissue were observed by hematoxylin and eosin staining under light microscopy and transmission electron microscopy. The expression of inflammatory factors IL­17C, nuclear factor­κB (NF­κB), IL­1ß, IL­6 and tumor necrosis factor­α (TNF­α) in lung tissue was assessed by RNA sequencing and verified by reverse transcription­quantitative PCR (RT­qPCR) and western blotting (WB). Superoxide dismutase (SOD) and glutathione peroxidase (GSH­Px) enzyme activity and malondialdehyde (MDA) expression were also measured. Experimental groups were compared to the control group following 24, 48 and 72 h of hypoxic stress. Lung tissue suffered from different degrees of injury, and the damage was the most severe after 48 h of hypoxic stress. RNA sequencing data from the lung tissue of rats from each group suggested that the expression of IL­17C, NF­κB, IL­1ß, IL­6, and TNF­α increased significantly after hypoxic stress. RT­qPCR and WB demonstrated that the expression of IL­17C and NF­κB increased significantly after hypoxia lasting 48 and 72 h. IL­1ß expression increased significantly after hypoxia stress lasting 24 and 48 h, and the expressions of TNF­α and IL­6 increased significantly after hypoxia stress lasting 24, 48 and 72 h (P<0.01). The enzyme activity of SOD and GSH­Px decreased significantly after lasting 24, 48 and 72 h of hypoxia (P<0.01), and MDA increased significantly after hypoxic stress lasting 48 and 72 h (P<0.01). In conclusion, under hypoxic stress, rats quickly initiate oxidative stress and immune responses. However, with prolonged hypoxic stress time, excessive oxidative stress can further stimulate the immune system in vivo, and release a large quantity of inflammatory factors accumulating in the body. This, in turn, may lead to the occurrence of inflammatory storms and further damage the lung tissue resulting in AMS.


Asunto(s)
Mal de Altura/inmunología , Mediadores de Inflamación/metabolismo , Pulmón/patología , Mal de Altura/patología , Animales , Modelos Animales de Enfermedad , Humanos , Mediadores de Inflamación/análisis , Pulmón/inmunología , Masculino , Estrés Oxidativo/inmunología , RNA-Seq , Ratas , Ratas Sprague-Dawley
10.
Nat Commun ; 12(1): 5987, 2021 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-34645793

RESUMEN

Following prolonged exposure to hypoxic conditions, for example, due to ascent to high altitude, stroke, or traumatic brain injury, cerebral edema can develop. The exact nature and genesis of hypoxia-induced edema in healthy individuals remain unresolved. We examined the effects of prolonged, normobaric hypoxia, induced by 16 h of exposure to simulated high altitude, on healthy brains using proton, dynamic contrast enhanced, and sodium MRI. This dual approach allowed us to directly measure key factors in the development of hypoxia-induced brain edema: (1) Sodium signals as a surrogate of the distribution of electrolytes within the cerebral tissue and (2) Ktrans as a marker of blood-brain-barrier integrity. The measurements point toward an accumulation of sodium ions in extra- but not in intracellular space in combination with an intact endothelium. Both findings in combination are indicative of ionic extracellular edema, a subtype of cerebral edema that was only recently specified as an intermittent, yet distinct stage between cytotoxic and vasogenic edemas. In sum, here a combination of imaging techniques demonstrates the development of ionic edemas following prolonged normobaric hypoxia in agreement with cascadic models of edema formation.


Asunto(s)
Mal de Altura/patología , Edema Encefálico/patología , Encéfalo/patología , Hipoxia/patología , Adulto , Mal de Altura/diagnóstico por imagen , Mal de Altura/metabolismo , Barrera Hematoencefálica/metabolismo , Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Edema Encefálico/diagnóstico por imagen , Edema Encefálico/metabolismo , Estudios de Cohortes , Femenino , Humanos , Hipoxia/diagnóstico por imagen , Hipoxia/metabolismo , Imagen por Resonancia Magnética , Masculino , Tamaño de los Órganos , Sodio/metabolismo
11.
Anal Biochem ; 631: 114342, 2021 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-34419454

RESUMEN

18ß-Glycyrrhetinic acid (GA) is the triterpenoid aglycone component of glycyrrhizic acid, a natural product of traditional Chinese medicine, and has been proven to possess a variety of pharmacological effects. The protection function and the mechanism of GA on rats with high-altitude pulmonary hypertension (HAPH) are studied using proton nuclear magnetic resonance (1H NMR) metabonomics technology and biochemical analysis. An HAPH model is established, and 60 male rats are randomly divided into the following groups: Control(normal saline, 0.4 mL/100 g), model (normal saline, 0.4 mL/100 g), Nifedipine (nifedipine, 2.7 mg/kg), and high-, medium-, and low-dose GA groups (100, 50, and 25 mg/kg GA designated as GA.H, GA.M, and GA.L, respectively). Serum biochemical indicators of rats in each group are measured, and pathological changes in the pulmonary artery are observed. 1H NMR metabonomics technology is used for serum analysis. Results show that GA can significantly reduce pulmonary arterial pressure and malondialdehyde levels and increase the glutathione peroxidase and superoxide dismutase activities in HAPH rats. Pathological results show that GA can alleviate pulmonary artery injuries of HAPH rats. Metabolomics analytical findings show that GA can alleviate the metabolic disorder of HAPH rats through anti-oxidation and anti-inflammatory effects, improve their bodies' ability to resist hypoxia, and restore various metabolic pathways (energy metabolism, amino acid metabolism, and lipid metabolism). GA has potential therapeutic effects on HAPH rats, but its target needs to be further studied.


Asunto(s)
Mal de Altura/prevención & control , Ácido Glicirretínico/análogos & derivados , Hipertensión Pulmonar/prevención & control , Espectroscopía de Resonancia Magnética/métodos , Metabolómica/métodos , Sustancias Protectoras/farmacología , Mal de Altura/etiología , Mal de Altura/patología , Animales , Análisis Químico de la Sangre/métodos , Modelos Animales de Enfermedad , Metabolismo Energético/efectos de los fármacos , Ácido Glicirretínico/administración & dosificación , Ácido Glicirretínico/farmacología , Hipertensión Pulmonar/etiología , Hipertensión Pulmonar/patología , Metabolismo de los Lípidos/efectos de los fármacos , Masculino , Malondialdehído/sangre , Análisis Multivariante , Sustancias Protectoras/administración & dosificación , Protones , Arteria Pulmonar/efectos de los fármacos , Arteria Pulmonar/ultraestructura , Ratas Sprague-Dawley , Superóxido Dismutasa/sangre
12.
Biomed Pharmacother ; 139: 111669, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34243609

RESUMEN

High-altitude pulmonary edema (HAPE) is a life-threatening disease occurs in hypobaric hypoxia (HH) environment, which could be treated by Dexamethasone, but might cause side-effects. Potentilla anserina L polysaccharide (PAP) holds promising physiological and pharmacological properties which could be beneficial for HAPE treatment. In our study, the anti-hypoxia effect of PAP was firstly investigated through anti-normobaric hypoxia test and anti-acute hypoxia test. Then we established a model of HAPE and measured the lung water content, pathological changes and MDA, NO, SOD, GSH concentrations in lung tissues. We also evaluated the protein and mRNA levels of pro-inflammatory cytokines (IL-1ß, IL-6, TNF-α, VEGF, NF-κB and HIF-1α) by ELISA kits, RT-PCR and Western blotting. As expected, PAP could dramatically reduce the lung water content, alleviate lung tissue injury, and inhibit MDA and NO production, it also promote SOD activity and GSH expression. In addition, it has been found that PAP blocked the NF-κB and HIF-1α signaling pathway activation, inhibited the generation of downstream pro-inflammatory cytokines. Therefore, PAP provides great potential in HAPE treatment mainly through suppression of oxidative stress and inflammatory suppression.


Asunto(s)
Mal de Altura/tratamiento farmacológico , Hipertensión Pulmonar/tratamiento farmacológico , Hipoxia/patología , Pulmón/efectos de los fármacos , Polisacáridos/farmacología , Potentilla/química , Mal de Altura/patología , Animales , Citocinas/metabolismo , Femenino , Hipertensión Pulmonar/patología , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Estrés Oxidativo/efectos de los fármacos , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Transducción de Señal/efectos de los fármacos
13.
Am J Med Sci ; 362(2): 154-160, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34083021

RESUMEN

BACKGROUND: We aimed to review records from 429 patients with high altitude pulmonary edema (HAPE) to identify some of the salient characteristics associated with this condition. MATERIALS AND METHODS: General information and clinical symptoms, along with laboratory test results from HAPE patients were collected and analyzed. Blood assay results and imaging at admission were compared with those at discharge. Results from routine blood assays were compared among three subgroups of these patients that were generated based upon the duration of their hypoxia exposure. RESULTS: Of these 429 HAPE patients, 9.32% also showed high altitude cerebral edema (HACE). White blood cell and neutrophil counts, as well as levels of alanine aminotransferase and aspartate aminotransferase, uric acid, lactic dehydrogenase and creatine kinase were all increased in HAPE patients, with further increases observed in those with HAPE combined with HACE. Levels of white blood cells, neutrophils, lymphocytes, and hemoglobin concentrations in HAPE patients at admission were significantly higher than that obtained at discharge. White blood cell and neutrophil counts were lower in patients who developed HAPE after a duration of 7 days of high altitude exposure as compared with those who developed the condition within 1 or 3 days. CONCLUSIONS: A combination of HAPE and HACE was present in 9.32% of the patients with HAPE. HAPE was more prevalent in males. Hepatocytes and the myocardium were likely sites of damage in patients with HAPE, with more severe damage observed in the patients with HAPE/HACE. White blood cell and neutrophil counts were significantly higher than normal ranges and these levels were negatively correlated with the duration of hypoxia exposure.


Asunto(s)
Mal de Altura/patología , Edema Pulmonar/etiología , Edema Pulmonar/patología , Adolescente , Adulto , Anciano , Edema Encefálico/etiología , Edema Encefálico/patología , China , Femenino , Humanos , Masculino , Persona de Mediana Edad , Estudios Retrospectivos , Adulto Joven
14.
Respir Res ; 22(1): 52, 2021 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-33579290

RESUMEN

BACKGROUND: Chinese Yunnan Province, located in the Yunnan-Guizhou Plateau, is a famous tourist paradise where acute high-altitude illness common occurs among lowland people visitors due to non-acclimatization to the acute hypobaric hypoxia (AHH) conditions. Traditional Chinese medicine, such as Qi-Long-Tian (QLT) formula, has shown effectiveness and safety in the treatment of acute high-altitude diseases. The aim of this study was to clarify the therapeutic mechanisms of this traditional formula using a rat model in a simulated plateau environment. METHODS: Following testing, lung tissue samples were evaluated by hematoxylin-eosin staining and for biochemical characteristics. mRNA-Seq was used to compare differentially expressed genes in control rats, and in rats exposed to AHH and AHH with QLT treatment. RESULTS: Inflammation-related effectors induced following QLT treatment for AHH included MMP9 and TIMP1, and involved several phosphorylation signaling pathways implicated in AHH pathogenesis such as PI3K/AKT and MAPK signaling. CONCLUSION: This study provides insights into the major signaling pathways induced by AHH and in the protective mechanisms involved in QLT formula activity.


Asunto(s)
Mal de Altura/tratamiento farmacológico , Mal de Altura/metabolismo , Altitud , Medicamentos Herbarios Chinos/uso terapéutico , Mediadores de Inflamación/metabolismo , Pulmón/metabolismo , Mal de Altura/patología , Animales , Medicamentos Herbarios Chinos/aislamiento & purificación , Medicamentos Herbarios Chinos/farmacología , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Inflamación/patología , Mediadores de Inflamación/antagonistas & inhibidores , Pulmón/efectos de los fármacos , Pulmón/patología , Masculino , Ratas , Ratas Wistar
15.
Am J Med ; 133(12): e706-e715, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32479750

RESUMEN

BACKGROUND: Acetazolamide is the most common medication used for acute mountain sickness prevention, with speculation that a reduced dose may be as efficacious as standard dosing with fewer side effects. METHODS: This double-blind, randomized, controlled noninferiority trial compared acetazolamide 62.5 mg twice daily to the standard dose acetazolamide 125 mg twice daily starting the evening prior to ascent from 1240 m (4100 ft) to 3810 m (12,570 ft) over 4 hours. The primary outcome was acute mountain sickness incidence (ie, headache, Lake Louise Questionnaire ≥3, and another symptom). RESULTS: A total of 106 participants were analyzed, with 51 (48%) randomized to 125 mg and 55 (52%) to 62.5 mg, with a combined acute mountain sickness incidence of 53 (50%) and mean severity of 3 (± 2.1). The 62.5-mg group failed to fall within the prespecified 26% noninferiority margin for acute mountain sickness incidence (62.5 mg = 30 [55%] vs 125 mg = 23 [45%], 95% confidence interval [CI] -11% to 30%). Participants in the 62.5-mg group had a higher risk of acute mountain sickness (odds ratio = 1.5, 95% CI 0.7-3.2) and moderate acute mountain sickness (odds ratio = 1.8, 95% CI 0.6-5.9), with a number needed to harm (NNH) of 9, with a number needed to treat (NNT) in the 125-mg group of 4.8. Increased acute mountain sickness incidence and symptom severity corresponded to lower weight-based and body mass index dosing, with similar side effects between groups. CONCLUSION: Acetazolamide 62.5 mg twice daily failed to demonstrate equal effectiveness to 125 mg twice daily for prevention of acute mountain sickness. With increased risk and no demonstrable symptomatic or physiologic benefits, acetazolamide 62.5 mg twice daily should not be recommended for acute mountain sickness prevention.


Asunto(s)
Acetazolamida/administración & dosificación , Acetazolamida/uso terapéutico , Mal de Altura/prevención & control , Inhibidores de Anhidrasa Carbónica/administración & dosificación , Inhibidores de Anhidrasa Carbónica/uso terapéutico , Mal de Altura/patología , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Humanos , Oportunidad Relativa , Factores de Riesgo , Índice de Severidad de la Enfermedad , Encuestas y Cuestionarios
17.
Blood Cells Mol Dis ; 84: 102446, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32470757

RESUMEN

Excessive erythrocytosis (EE) is a characteristic of chronic mountain sickness (CMS). Currently, the pathogenesis of CMS remains unclear. This study was intended to investigate the role of EPAS1 in the proliferation of erythroblasts in CMS. Changes of HIF-1α and EPAS1/HIF-2α in the bone marrow erythroblasts of 21 patients with CMS and 14 control subjects residing at the same altitudes were determined by RT-qPCR and western blotting. We also developed a lentiviral vector, Lv-EPAS1/sh-EPAS1, to over-express/silence EPAS1 in K562 cells. Cells cycle and proliferation were detected by flow cytometry. Transcriptome analyses were carried out on Illumina. CMS patients showed a higher expression of EPAS1/HIF-2α in the bone marrow erythroblasts than those of controls. Variations in EPAS1 expression in CMS patients were positively correlated with RBC levels, and negatively correlated with SaO2. Over-expressing of EPAS1 in K562 cells accelerated the erythroid cells cycle progression and promoted the erythroid cells proliferation-and vice versa. Transcriptome data indicated that proliferation-related DEGs were significantly enriched in EPAS1 overexpression/silencing K562 cells. Our results suggest that EPAS1 might participate in the pathogenesis of EE by regulating the proliferation of erythroblasts.


Asunto(s)
Mal de Altura/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Eritroblastos/patología , Adulto , Mal de Altura/genética , Mal de Altura/patología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/análisis , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Ciclo Celular , Línea Celular , Proliferación Celular , Enfermedad Crónica , Eritroblastos/citología , Eritroblastos/metabolismo , Humanos , Persona de Mediana Edad , Transcriptoma , Regulación hacia Arriba
18.
Blood Cells Mol Dis ; 82: 102421, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32171843

RESUMEN

Venous thrombo-embolism (VTE) is multi-factorial disease involving several genetic and acquired risk factors responsible for its onset. It may occur spontaneously upon climbing at High Altitude (HA). Several studies demonstrated that hypoxic conditions prevailing at HA pose an independent risk factor for VTE; however, molecular mechanism remains unknown. Present study aims to identify genes associated with HA-induced VTE pathophysiology using real time TaqMan Low-Density Array (TLDA) of known candidate genes. Gene expression of total 93 genes were studied and analyzed in patients of VTE from HA (HA-VTE) and from sea level (SL-VTE) in comparison to respective controls. Both HA-VTE and SL-VTE patients showed up-regulation of 37 genes involved in blood coagulation cascade, clot formation, platelet formation, endothelial response, angiogenesis, cell adhesion and calcium channel activity. Seven genes including ACE, EREG, C8A, DLG2, USF1, F2 and PCDHA7 were up-regulated in both HA-controls and VTE patients (both HA-VTE and SL-VTE) indicating their role during VTE event and also upon HA exposure. Ten genes; CDH18, FGA, EDNBR, GATA2, MAPK9, BCAR1, FRK, F11, PCDHA1 and ST8SIA4 were uniquely up-regulated in HA-VTE. The differentially expressed genes from the present study could be determining factors for HA-VTE susceptibility and provide insights into VTE occurrence at HA.


Asunto(s)
Mal de Altura , Coagulación Sanguínea , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Análisis de Secuencia por Matrices de Oligonucleótidos , Tromboembolia Venosa , Adulto , Altitud , Mal de Altura/sangre , Mal de Altura/complicaciones , Mal de Altura/patología , Femenino , Humanos , Masculino , Tromboembolia Venosa/sangre , Tromboembolia Venosa/genética , Tromboembolia Venosa/patología
19.
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
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...