Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 16.604
Filtrar
1.
Exp Cell Res ; 438(2): 114061, 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38692345

RESUMEN

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.


Asunto(s)
Apoptosis , Furanos , Inflamación , Ratones Endogámicos C57BL , Infarto del Miocardio , Miocitos Cardíacos , Estrés Oxidativo , Piroptosis , Sulfonamidas , Piroptosis/efectos de los fármacos , Animales , Ratones , Apoptosis/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Sulfonamidas/farmacología , Humanos , Inflamación/metabolismo , Inflamación/patología , Inflamación/tratamiento farmacológico , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Masculino , Furanos/farmacología , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Infarto del Miocardio/tratamiento farmacológico , Indenos/farmacología , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , para-Aminobenzoatos/farmacología , Inflamasomas/metabolismo , Inflamasomas/efectos de los fármacos , Modelos Animales de Enfermedad , Miocardio/metabolismo , Miocardio/patología , Hipoxia/metabolismo , Hipoxia/complicaciones , Dipéptidos
2.
Cell Biol Toxicol ; 40(1): 32, 2024 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-38767703

RESUMEN

BACKGROUND: Recent studies have emphasized the critical role of Telocytes (TCs)-derived exosomes in organ tissue injury and repair. Our previous research showed a significant increase in ITGB1 within TCs. Pulmonary Arterial Hypertension (PAH) is marked by a loss of microvessel regeneration and progressive vascular remodeling. This study aims to investigate whether exosomes derived from ITGB1-modified TCs (ITGB1-Exo) could mitigate PAH. METHODS: We analyzed differentially expressed microRNAs (DEmiRs) in TCs using Affymetrix Genechip miRNA 4.0 arrays. Exosomes isolated from TC culture supernatants were verified through transmission electron microscopy and Nanoparticle Tracking Analysis. The impact of miR-429-3p-enriched exosomes (Exo-ITGB1) on hypoxia-induced pulmonary arterial smooth muscle cells (PASMCs) was evaluated using CCK-8, transwell assay, and inflammatory factor analysis. A four-week hypoxia-induced mouse model of PAH was constructed, and H&E staining, along with Immunofluorescence staining, were employed to assess PAH progression. RESULTS: Forty-five miRNAs exhibited significant differential expression in TCs following ITGB1 knockdown. Mus-miR-429-3p, significantly upregulated in ITGB1-overexpressing TCs and in ITGB1-modified TC-derived exosomes, was selected for further investigation. Exo-ITGB1 notably inhibited the migration, proliferation, and inflammation of PASMCs by targeting Rac1. Overexpressing Rac1 partly counteracted Exo-ITGB1's effects. In vivo administration of Exo-ITGB1 effectively reduced pulmonary vascular remodeling and inflammation. CONCLUSIONS: Our findings reveal that ITGB1-modified TC-derived exosomes exert anti-inflammatory effects and reverse vascular remodeling through the miR-429-3p/Rac1 axis. This provides potential therapeutic strategies for PAH treatment.


Asunto(s)
Exosomas , Integrina beta1 , MicroARNs , Telocitos , Proteína de Unión al GTP rac1 , MicroARNs/genética , MicroARNs/metabolismo , Animales , Exosomas/metabolismo , Exosomas/genética , Proteína de Unión al GTP rac1/metabolismo , Proteína de Unión al GTP rac1/genética , Integrina beta1/metabolismo , Integrina beta1/genética , Ratones , Telocitos/metabolismo , Masculino , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Ratones Endogámicos C57BL , Hipertensión Arterial Pulmonar/metabolismo , Hipertensión Arterial Pulmonar/genética , Hipertensión Arterial Pulmonar/patología , Hipoxia/metabolismo , Hipoxia/genética , Hipoxia/complicaciones , Proliferación Celular/genética , Movimiento Celular/genética , Humanos , Remodelación Vascular/genética , Neuropéptidos
3.
Nat Commun ; 15(1): 3970, 2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38730227

RESUMEN

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.


Asunto(s)
Mal de Altura , Altitud , Regulación de la Expresión Génica , Hipoxia , Animales , Mal de Altura/genética , Mal de Altura/metabolismo , Ovinos , Hipoxia/genética , Hipoxia/metabolismo , Humanos , Aclimatación/genética , Transcripción Genética , Análisis de la Célula Individual , Femenino , Multiómica
4.
Int J Mol Sci ; 25(9)2024 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-38731803

RESUMEN

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.


Asunto(s)
Atletas , Biomarcadores , Hipoxia , Humanos , Masculino , Hipoxia/metabolismo , Proyectos Piloto , Natación/fisiología , Adulto Joven , Miocardio/metabolismo , Mioglobina/metabolismo , Troponina I/metabolismo , Proteína 3 de Unión a Ácidos Grasos/metabolismo , Adolescente , Proteínas de Unión a Ácidos Grasos/metabolismo , Resistencia Física/fisiología , Forma MB de la Creatina-Quinasa/sangre , Forma MB de la Creatina-Quinasa/metabolismo , Adaptación Fisiológica , Altitud
5.
Int J Mol Sci ; 25(9)2024 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-38731800

RESUMEN

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.


Asunto(s)
Homeostasis , Subunidad alfa del Factor 1 Inducible por Hipoxia , Enfermedades Neurodegenerativas , Oxígeno , Enfermedad de Parkinson , Humanos , Oxígeno/metabolismo , Animales , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/patología , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/etiología , Factor 1 Inducible por Hipoxia/metabolismo , Hipoxia/metabolismo
6.
Neoplasma ; 71(2): 123-142, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38766851

RESUMEN

Lung cancer represents the leading cause of cancer-related deaths. Non-small cell lung cancer (NSCLC), the most common form of lung cancer, is a molecularly heterogeneous disease with intratumoral heterogeneity and a significant mutational burden associated with clinical outcome. Tumor microenvironment (TME) plays a fundamental role in the initiation and progression of primary de novo lung cancer and significantly influences the response of tumor cells to therapy. Hypoxia, an integral part of the tumor microenvironment and a serious clinical phenomenon, is associated with increased genetic instability and a more aggressive phenotype of NSCLC, which correlates with the risk of metastasis. Low oxygen concentration influences all components of TME including the immune microenvironment. Hypoxia-inducible pathway activated in response to low oxygen supply mediates the expression of genes important for the adaptation of tumor cells to microenvironmental changes. A highly active transmembrane hypoxia-induced metalloenzyme - carbonic anhydrase IX (CAIX), as a part of transport metabolon, contributes to the maintenance of intracellular pH within physiological values and to the acidification of the extracellular space. CAIX supports cell migration and invasion and plays an important role in NSCLC tumor tissue and pleural effusion. Due to its high expression, it also represents a potential diagnostic differential biomarker and therapeutic target in NSCLC. To test new potential targeted therapeutic compounds, suitable models are required that more faithfully simulate tumor tissue, TME components, and spatial architecture.


Asunto(s)
Antígenos de Neoplasias , Biomarcadores de Tumor , Anhidrasa Carbónica IX , Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Microambiente Tumoral , Humanos , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/diagnóstico , Carcinoma de Pulmón de Células no Pequeñas/patología , Anhidrasa Carbónica IX/metabolismo , Biomarcadores de Tumor/metabolismo , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/diagnóstico , Neoplasias Pulmonares/metabolismo , Antígenos de Neoplasias/metabolismo , Hipoxia/metabolismo
7.
Bull Exp Biol Med ; 176(5): 680-686, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38733478

RESUMEN

Morphological, molecular, and biological features of the systemic inflammatory response induced by LPS administration were assessed in adult and old male Wistar rats with high and low resistance to hypoxia. In 6 h after LPS administration, mRNA expression levels of Hif1a, Vegf, Nfkb, and level of IL-1ß protein in old rats were higher than in adult rats regardless of hypoxia tolerance. The morphometric study showed that the number of neutrophils in the interalveolar septa of the lungs was significantly higher in low-resistant adult and old rats 6 h after LPS administration. Thus, in old male Wistar rats, systemic inflammatory response is more pronounced than in adult rats and depends on the initial tolerance to hypoxia, which should be considered when developing new approaches to the therapy of systemic inflammatory response in individuals of different ages.


Asunto(s)
Subunidad alfa del Factor 1 Inducible por Hipoxia , Hipoxia , Interleucina-1beta , Ratas Wistar , Animales , Masculino , Ratas , Hipoxia/metabolismo , Hipoxia/genética , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Lipopolisacáridos/farmacología , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , FN-kappa B/metabolismo , FN-kappa B/genética , Pulmón/patología , Pulmón/metabolismo , Pulmón/efectos de los fármacos , Pulmón/inmunología , Neutrófilos/metabolismo , Neutrófilos/inmunología , Inflamación/metabolismo , Inflamación/patología , Factores de Edad , ARN Mensajero/genética , ARN Mensajero/metabolismo
8.
PLoS One ; 19(5): e0295094, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38743782

RESUMEN

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.


Asunto(s)
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Hipoxia , Animales , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Hipoxia/genética , Hipoxia/metabolismo , Sitios de Unión , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Factor 1 Inducible por Hipoxia/metabolismo , Factor 1 Inducible por Hipoxia/genética
9.
Nihon Yakurigaku Zasshi ; 159(3): 160-164, 2024.
Artículo en Japonés | MEDLINE | ID: mdl-38692880

RESUMEN

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.


Asunto(s)
Hipoxia , Humanos , Hipoxia/tratamiento farmacológico , Hipoxia/metabolismo , Animales , Desarrollo de Medicamentos , Terapia Molecular Dirigida , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo
10.
Nihon Yakurigaku Zasshi ; 159(3): 165-168, 2024.
Artículo en Japonés | MEDLINE | ID: mdl-38692881

RESUMEN

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.


Asunto(s)
Oxígeno , Canal Catiónico TRPA1 , Canales de Potencial de Receptor Transitorio , Canal Catiónico TRPA1/metabolismo , Humanos , Oxígeno/metabolismo , Animales , Canales de Potencial de Receptor Transitorio/metabolismo , Hipoxia/metabolismo , Canales de Calcio/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Cuerpo Carotídeo/metabolismo
11.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 40(4): 327-332, 2024 Apr.
Artículo en Chino | MEDLINE | ID: mdl-38710517

RESUMEN

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.


Asunto(s)
Caspasa 1 , Hipoxia , Inflamasomas , Interleucina-1beta , Hígado , Ratones Endogámicos C57BL , Especies Reactivas de Oxígeno , Animales , Masculino , Inflamasomas/metabolismo , Hipoxia/metabolismo , Hipoxia/complicaciones , Especies Reactivas de Oxígeno/metabolismo , Hígado/metabolismo , Hígado/patología , Caspasa 1/metabolismo , Interleucina-1beta/metabolismo , Ratones , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Proteínas Reguladoras de la Apoptosis/metabolismo , Alanina Transaminasa/sangre , Proteínas Adaptadoras de Señalización CARD/metabolismo , Aspartato Aminotransferasas/sangre , Hepatopatías/etiología , Hepatopatías/metabolismo , Hepatopatías/patología
12.
Function (Oxf) ; 5(3): zqae010, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38706960

RESUMEN

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.


Asunto(s)
Cuerpo Carotídeo , Ratones Noqueados , Receptores Odorantes , Tirosina 3-Monooxigenasa , Animales , Cuerpo Carotídeo/metabolismo , Tirosina 3-Monooxigenasa/metabolismo , Tirosina 3-Monooxigenasa/genética , Ratones , Hipoxia/metabolismo , Hipoxia/genética , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Masculino , Ratones Endogámicos C57BL
13.
Cancer J ; 30(3): 159-169, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38753750

RESUMEN

ABSTRACT: Imaging glucose metabolism with [18F]fluorodeoxyglucose positron emission tomography has transformed the diagnostic and treatment algorithms of numerous malignancies in clinical practice. The cancer phenotype, though, extends beyond dysregulation of this single pathway. Reprogramming of other pathways of metabolism, as well as altered perfusion and hypoxia, also typifies malignancy. These features provide other opportunities for imaging that have been developed and advanced into humans. In this review, we discuss imaging metabolism, perfusion, and hypoxia in cancer, focusing on the underlying biology to provide context. We conclude by highlighting the ability to image multiple facets of biology to better characterize cancer and guide targeted treatment.


Asunto(s)
Fluorodesoxiglucosa F18 , Neoplasias , Tomografía de Emisión de Positrones , Humanos , Fluorodesoxiglucosa F18/metabolismo , Neoplasias/diagnóstico por imagen , Neoplasias/metabolismo , Neoplasias/diagnóstico , Tomografía de Emisión de Positrones/métodos , Radiofármacos/metabolismo , Hipoxia/metabolismo , Hipoxia/diagnóstico por imagen
14.
FASEB J ; 38(10): e23671, 2024 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-38752538

RESUMEN

NLRP3 inflammasome activation has emerged as a critical initiator of inflammatory response in ischemic retinopathy. Here, we identified the effect of a potent, selective NLRP3 inhibitor, MCC950, on autophagy and apoptosis under hypoxia. Neonatal mice were exposed to hyperoxia for 5 days to establish oxygen-induced retinopathy (OIR) model. Intravitreal injection of MCC950 was given, and then autophagy and apoptosis markers were assessed. Retinal autophagy, apoptosis, and related pathways were evaluated by western blot, immunofluorescent labeling, transmission electron microscopy, and TUNEL assay. Autophagic activity in Müller glia after NLRP3 inflammasome inhibition, together with its influence on photoreceptor death, was studied using western blot, immunofluorescence staining, mRFP-GFP-LC3 adenovirus transfection, cell viability, proliferation, and apoptosis assays. Results showed that activation of NLRP3 inflammasome in Müller glia was detected in OIR model. MCC950 could improve impaired retinal autophagic flux and attenuate retinal apoptosis while it regulated the retinal AMPK/mTOR/ULK-1 pathway. Suppressed autophagy and depressed proliferation capacity resulting from hypoxia was promoted after MCC950 treatment in Müller glia. Inhibition of AMPK and ULK-1 pathway significantly interfered with the MCC950-induced autophagy activity, indicating MCC950 positively modulated autophagy through AMPK/mTOR/ULK-1 pathway in Müller cells. Furthermore, blockage of autophagy in Müller glia significantly induced apoptosis in the cocultured 661W photoreceptor cells, whereas MCC950 markedly preserved the density of photoreceptor cells. These findings substantiated the therapeutic potential of MCC950 against impaired autophagy and subsequent apoptosis under hypoxia. Such protective effect might involve the modulation of AMPK/mTOR/ULK-1 pathway. Targeting NLRP3 inflammasome in Müller glia could be beneficial for photoreceptor survival under hypoxic conditions.


Asunto(s)
Apoptosis , Autofagia , Células Ependimogliales , Furanos , Indenos , Inflamasomas , Proteína con Dominio Pirina 3 de la Familia NLR , Sulfonamidas , Animales , Autofagia/efectos de los fármacos , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Ratones , Apoptosis/efectos de los fármacos , Sulfonamidas/farmacología , Inflamasomas/metabolismo , Furanos/farmacología , Células Ependimogliales/metabolismo , Células Ependimogliales/efectos de los fármacos , Indenos/farmacología , Ratones Endogámicos C57BL , Hipoxia/metabolismo , Óxidos S-Cíclicos/farmacología , Sulfonas/farmacología , Células Fotorreceptoras de Vertebrados/metabolismo , Células Fotorreceptoras de Vertebrados/efectos de los fármacos , Células Fotorreceptoras de Vertebrados/patología , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Células Fotorreceptoras/metabolismo , Células Fotorreceptoras/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
15.
Sci Adv ; 10(20): eadj3301, 2024 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-38758780

RESUMEN

Myeloid cells are highly prevalent in glioblastoma (GBM), existing in a spectrum of phenotypic and activation states. We now have limited knowledge of the tumor microenvironment (TME) determinants that influence the localization and the functions of the diverse myeloid cell populations in GBM. Here, we have utilized orthogonal imaging mass cytometry with single-cell and spatial transcriptomic approaches to identify and map the various myeloid populations in the human GBM tumor microenvironment (TME). Our results show that different myeloid populations have distinct and reproducible compartmentalization patterns in the GBM TME that is driven by tissue hypoxia, regional chemokine signaling, and varied homotypic and heterotypic cellular interactions. We subsequently identified specific tumor subregions in GBM, based on composition of identified myeloid cell populations, that were linked to patient survival. Our results provide insight into the spatial organization of myeloid cell subpopulations in GBM, and how this is predictive of clinical outcome.


Asunto(s)
Glioblastoma , Células Mieloides , Microambiente Tumoral , Glioblastoma/patología , Glioblastoma/metabolismo , Humanos , Células Mieloides/metabolismo , Células Mieloides/patología , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/genética , Línea Celular Tumoral , Análisis de la Célula Individual , Hipoxia/metabolismo , Perfilación de la Expresión Génica
16.
Sci Rep ; 14(1): 10206, 2024 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-38702334

RESUMEN

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.


Asunto(s)
Altitud , Hipoxia , Leptina , Transducción de Señal , Leptina/metabolismo , Leptina/sangre , Animales , Ratas , Masculino , Hipoxia/metabolismo , Hipoxia/fisiopatología , Humanos , Mal de Altura/metabolismo , Mal de Altura/fisiopatología , Miocardio/metabolismo , Miocardio/patología , Adulto , Corazón/fisiopatología
17.
Front Immunol ; 15: 1372959, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38690277

RESUMEN

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.


Asunto(s)
Perfilación de la Expresión Génica , Hipertensión Pulmonar , Hipoxia , Análisis de la Célula Individual , Transcriptoma , Animales , Ratones , Hipoxia/metabolismo , Hipoxia/inmunología , Hipertensión Pulmonar/etiología , Hipertensión Pulmonar/inmunología , Hipertensión Pulmonar/genética , Macrófagos Alveolares/inmunología , Macrófagos Alveolares/metabolismo , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad , Masculino , Pulmón/inmunología , Pulmón/patología , Pulmón/metabolismo
18.
Sci Rep ; 14(1): 11387, 2024 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-38762567

RESUMEN

Identifying and controlling tumor escape mechanisms is crucial for improving cancer treatment effectiveness. Experimental studies reveal tumor hypoxia and adenosine as significant contributors to such mechanisms. Hypoxia exacerbates adenosine levels in the tumor microenvironment. Combining inhibition of these factors with dendritic cell (DC)-based immunotherapy promises improved clinical outcomes. However, challenges include understanding dynamics, optimal vaccine dosages, and timing. Mathematical models, including agent-based, diffusion, and ordinary differential equations, address these challenges. Here, we employ these models for the first time to elucidate how hypoxia and adenosine facilitate tumor escape in DC-based immunotherapy. After parameter estimation using experimental data, we optimize vaccination protocols to minimize tumor growth. Sensitivity analysis highlights adenosine's significant impact on immunotherapy efficacy. Its suppressive role impedes treatment success, but inhibiting adenosine could enhance therapy, as suggested by the model. Our findings shed light on hypoxia and adenosine-mediated tumor escape mechanisms, informing future treatment strategies. Additionally, identifiability analysis confirms accurate parameter determination using experimental data.


Asunto(s)
Adenosina , Células Dendríticas , Inmunoterapia , Escape del Tumor , Adenosina/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Humanos , Inmunoterapia/métodos , Microambiente Tumoral/inmunología , Animales , Modelos Teóricos , Neoplasias/terapia , Neoplasias/inmunología , Hipoxia Tumoral , Ratones , Hipoxia/metabolismo
19.
BMC Vet Res ; 20(1): 135, 2024 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-38570796

RESUMEN

AIMS: We investigated the effects of intraperitoneal injections of titanium dioxide nanoparticles (TiO2 NPs, 100 mg/kg) for 5 consecutive days on the developmental competence of murine oocytes. Furthermore, study the effects of TiO2 NPs on antioxidant and oxidative stress biomarkers, as well as their effects on expression of apoptotic and hypoxia inducing factor-1α (HIF1A) protein translation. Moreover, the possible ameliorating effects of intraperitoneal injections of fructose (2.75 mM/ml) was examined. MATERIALS AND METHODS: Thirty sexually mature (8-12 weeks old; ~ 25 g body weight) female mice were used for the current study. The female mice were assigned randomly to three treatment groups: Group1 (G1) mice were injected intraperitoneal (ip) with deionized water for 5 consecutive days; Group 2 (G2) mice were injected ip with TiO2 NPs (100 mg/kg BW) for 5 consecutive days; Group 3 (G3) mice were injected ip with TiO2 NPs (100 mg/kg BW + fructose (2.75 mM) for 5 consecutive days. RESULTS: Nano-titanium significantly decreased expression of GSH, GPx, and NO, expression of MDA and TAC increased. The rates of MI, MII, GVBD and degenerated oocytes were significantly less for nano-titanium treated mice, but the rate of activated oocytes was significantly greater than those in control oocytes. TiO2 NPs significantly increased expression of apoptotic genes (BAX, Caspase 3 and P53) and HIF1A. Intraperitoneal injection of fructose (2.75 mM/kg) significantly alleviated the detrimental effects of TiO2 NPs. Transmission electron microscopy indicated that fructose mitigated adverse effects of TiO2 NPs to alter the cell surface of murine oocytes. CONCLUSION: Results of this study suggest that the i/p infusion of fructose for consecutive 5 days enhances development of murine oocytes and decreases toxic effects of TiO2 NPs through positive effects on oxidative and antioxidant biomarkers in cumulus-oocyte complexes and effects to inhibit TiO2-induced increases in expression of apoptotic and hypoxia inducing factors.


Asunto(s)
Nanopartículas del Metal , Nanopartículas , Ratones , Femenino , Animales , Antioxidantes/metabolismo , Hígado/metabolismo , Estrés Oxidativo , Titanio/toxicidad , Oocitos , Hipoxia/metabolismo , Hipoxia/veterinaria , Biomarcadores/metabolismo , Nanopartículas del Metal/toxicidad
20.
Ren Fail ; 46(1): 2332492, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38584135

RESUMEN

Acute kidney injury (AKI) is associated with a high mortality rate. Pathologically, renal ischemia/reperfusion injury (RIRI) is one of the primary causes of AKI, and hypoxia-inducible factor (HIF)-1α may play a defensive role in RIRI. This study assessed the role of hypoxia-inducible factor 1α (HIF-1α)-mediated mitophagy in protection against RIRI in vitro and in vivo. The human tubular cell line HK-2 was used to assess hypoxia/reoxygenation (H/R)-induced mitophagy through different in vitro assays, including western blotting, immunofluorescence staining, terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL), and reactive oxygen species (ROS) measurement. Additionally, a rat RIRI model was established for evaluation by renal histopathology, renal Doppler ultrasound, and transmission electron microscopy to confirm the in vitro data. The selective HIF-1α inhibitor LW6 reduced H/R-induced mitophagy but increased H/R-induced apoptosis and ROS production. Moreover, H/R treatment enhanced expression of the FUN14 domain-containing 1 (FUNDC1) protein. Additionally, FUNDC1 overexpression reversed the effects of LW6 on the altered expression of light chain 3 (LC3) BII and voltage-dependent anion channels as well as blocked the effects of HIF-1α inhibition in cells. Pretreatment of the rat RIRI model with roxadustat, a novel oral HIF-1α inhibitor, led to decreased renal injury and apoptosis in vivo. In conclusion, the HIF-1α/FUNDC1 signaling pathway mediates H/R-promoted renal tubular cell mitophagy, whereas inhibition of this signaling pathway protects cells from mitophagy, thus aggravating apoptosis, and ROS production. Accordingly, roxadustat may protect against RIRI-related AKI.


Asunto(s)
Lesión Renal Aguda , Daño por Reperfusión , Animales , Humanos , Ratas , Lesión Renal Aguda/etiología , Lesión Renal Aguda/prevención & control , Lesión Renal Aguda/metabolismo , Apoptosis , Hipoxia/metabolismo , Hipoxia/patología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Isquemia , Riñón/patología , Proteínas de la Membrana/metabolismo , Proteínas Mitocondriales , Mitofagia , Especies Reactivas de Oxígeno/metabolismo , Daño por Reperfusión/metabolismo , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA