Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 334
Filtrar
1.
Cells ; 10(9)2021 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-34571989

RESUMEN

Hypoxia and inflammation are frequently co-incidental features of the tissue microenvironment in a wide range of inflammatory diseases. While the impact of hypoxia on inflammatory pathways in immune cells has been well characterized, less is known about how inflammatory stimuli such as cytokines impact upon the canonical hypoxia-inducible factor (HIF) pathway, the master regulator of the cellular response to hypoxia. In this review, we discuss what is known about the impact of two major pro-inflammatory cytokines, tumor necrosis factor-α (TNF-α) and interleukin-1ß (IL-1ß), on the regulation of HIF-dependent signaling at sites of inflammation. We report extensive evidence for these cytokines directly impacting upon HIF signaling through the regulation of HIF at transcriptional and post-translational levels. We conclude that multi-level crosstalk between inflammatory and hypoxic signaling pathways plays an important role in shaping the nature and degree of inflammation occurring at hypoxic sites.


Asunto(s)
Factor 1 Inducible por Hipoxia/metabolismo , Interleucina-1beta/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Citocinas/metabolismo , Expresión Génica/genética , Regulación de la Expresión Génica/genética , Humanos , Hipoxia/fisiopatología , Factor 1 Inducible por Hipoxia/fisiología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/fisiología , Inflamación/fisiopatología , Interleucina-1beta/fisiología , ARN Mensajero/metabolismo , Transducción de Señal/genética , Activación Transcripcional , Factor de Necrosis Tumoral alfa/fisiología
2.
Chin J Nat Med ; 19(7): 521-527, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34247775

RESUMEN

Hypoxia-inducible factor 1 (HIF-1), as a main transcriptional regulator of metabolic adaptation to changes in the oxygen environment, participates in many physiological and pathological processes in the body, and is closely related to the pathogenesis of many diseases. This review outlines the mechanisms of HIF-1 activation, its signaling pathways, natural inhibitors, and its roles in diseases. This article can provide new insights in the diagnosis and treatment of human diseases, and recent progress on the development of HIF-1 inhibitors.


Asunto(s)
Factor 1 Inducible por Hipoxia , Transducción de Señal , Enfermedad , Humanos , Factor 1 Inducible por Hipoxia/fisiología , Oxígeno
3.
Nat Rev Rheumatol ; 17(7): 426-439, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34083809

RESUMEN

The physiologically hypoxic intervertebral disc and cartilage rely on the hypoxia-inducible factor (HIF) family of transcription factors to mediate cellular responses to changes in oxygen tension. During homeostatic development, oxygen-dependent prolyl hydroxylases, circadian clock proteins and metabolic intermediates control the activities of HIF1 and HIF2 in these tissues. Mechanistically, HIF1 is the master regulator of glycolytic metabolism and cytosolic lactate levels. In addition, HIF1 regulates mitochondrial metabolism by promoting flux through the tricarboxylic acid cycle, inhibiting downsteam oxidative phosphorylation and controlling mitochondrial health through modulation of the mitophagic pathway. Accumulation of metabolic intermediates from HIF-dependent processes contribute to intracellular pH regulation in the disc and cartilage. Namely, to prevent changes in intracellular pH that could lead to cell death, HIF1 orchestrates a bicarbonate buffering system in the disc, controlled by carbonic anhydrase 9 (CA9) and CA12, sodium bicarbonate cotransporters and an intracellular H+/lactate efflux mechanism. In contrast to HIF1, the role of HIF2 remains elusive; in disorders of the disc and cartilage, its function has been linked to both anabolic and catabolic pathways. The current knowledge of hypoxic cell metabolism and regulation of HIF1 activity provides a strong basis for the development of future therapies designed to repair the degenerative disc.


Asunto(s)
Factor 1 Inducible por Hipoxia/fisiología , Disco Intervertebral/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/fisiología , Humanos , Hipoxia/metabolismo , Factor 1 Inducible por Hipoxia/metabolismo , Disco Intervertebral/fisiología
4.
Front Immunol ; 12: 684085, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34149725

RESUMEN

Glucocorticoid-induced (GC) and hypoxia-induced transcriptional responses play an important role in tissue homeostasis and in the regulation of cellular responses to stress and inflammation. Evidence exists that there is an important crosstalk between both GC and hypoxia effects. Hypoxia is a pathophysiological condition to which cells respond quickly in order to prevent metabolic shutdown and death. The hypoxia inducible factors (HIFs) are the master regulators of oxygen homeostasis and are responsible for the ability of cells to cope with low oxygen levels. Maladaptive responses of HIFs contribute to a variety of pathological conditions including acute mountain sickness (AMS), inflammation and neonatal hypoxia-induced brain injury. Synthetic GCs which are analogous to the naturally occurring steroid hormones (cortisol in humans, corticosterone in rodents), have been used for decades as anti-inflammatory drugs for treating pathological conditions which are linked to hypoxia (i.e. asthma, ischemic injury). In this review, we investigate the crosstalk between the glucocorticoid receptor (GR), and HIFs. We discuss possible mechanisms by which GR and HIF influence one another, in vitro and in vivo, and the therapeutic effects of GCs on HIF-mediated diseases.


Asunto(s)
Glucocorticoides/farmacología , Hidrocortisona/farmacología , Factor 1 Inducible por Hipoxia/fisiología , Hipoxia/metabolismo , Receptor Cross-Talk/fisiología , Animales , Glucocorticoides/metabolismo , Homeostasis , Humanos , Hidrocortisona/metabolismo , Factor 1 Inducible por Hipoxia/metabolismo , Inflamación/metabolismo , Receptor Cross-Talk/efectos de los fármacos , Receptores de Glucocorticoides/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
5.
Artículo en Inglés | WPRIM (Pacífico Occidental) | ID: wpr-888780

RESUMEN

Hypoxia-inducible factor 1 (HIF-1), as a main transcriptional regulator of metabolic adaptation to changes in the oxygen environment, participates in many physiological and pathological processes in the body, and is closely related to the pathogenesis of many diseases. This review outlines the mechanisms of HIF-1 activation, its signaling pathways, natural inhibitors, and its roles in diseases. This article can provide new insights in the diagnosis and treatment of human diseases, and recent progress on the development of HIF-1 inhibitors.


Asunto(s)
Humanos , Enfermedad , Factor 1 Inducible por Hipoxia/fisiología , Oxígeno , Transducción de Señal
6.
Int J Mol Sci ; 21(17)2020 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-32846951

RESUMEN

Oral squamous cell carcinoma (OSCC) is the 10th most frequent human malignancy and is thus a global burden. Despite some progress in diagnosis and therapy, patients' overall survival rate, between 40 and 55%, has stagnated over the last four decades. Since the tumor node metastasis (TNM) system is not precise enough to predict the disease outcome, additive factors for diagnosis, prognosis, prediction and therapy resistance are urgently needed for OSCC. One promising candidate is the hypoxia inducible factor-1 (HIF-1), which functions as an early regulator of tumor aggressiveness and is a key promoter of energy adaptation. Other parameters comprise the composition of the tumor microenvironment, which determines the availability of nutrients and oxygen. In our opinion, these general processes are linked in the pathogenesis of OSCC. Based on this assumption, the review will summarize the major features of the HIF system-induced activities, its target proteins and related pathways of nutrient utilization and metabolism that are essential for the initiation, progression and therapeutic stratification of OSCC.


Asunto(s)
Carcinoma de Células Escamosas/metabolismo , Factor 1 Inducible por Hipoxia/fisiología , Neoplasias de la Boca/metabolismo , Animales , Carcinoma de Células Escamosas/diagnóstico , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patología , Progresión de la Enfermedad , Humanos , Neoplasias de la Boca/diagnóstico , Neoplasias de la Boca/genética , Neoplasias de la Boca/patología , Pronóstico , Microambiente Tumoral
7.
J Clin Invest ; 130(10): 5042-5051, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32730232

RESUMEN

Intermittent hypoxia (IH) is a hallmark manifestation of obstructive sleep apnea (OSA), a widespread disorder of breathing. This Review focuses on the role of hypoxia-inducible factors (HIFs) in hypertension, type 2 diabetes (T2D), and cognitive decline in experimental models of IH patterned after O2 profiles seen in OSA. IH increases HIF-1α and decreases HIF-2α protein levels. Dysregulated HIFs increase reactive oxygen species (ROS) through HIF-1-dependent activation of pro-oxidant enzyme genes in addition to reduced transcription of antioxidant genes by HIF-2. ROS in turn activate chemoreflex and suppress baroreflex, thereby stimulating the sympathetic nervous system and causing hypertension. We also discuss how increased ROS generation by HIF-1 contributes to IH-induced insulin resistance and T2D as well as disrupted NMDA receptor signaling in the hippocampus, resulting in cognitive decline.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/fisiología , Factor 1 Inducible por Hipoxia/fisiología , Apnea Obstructiva del Sueño/fisiopatología , Animales , Barorreflejo/fisiología , Cuerpo Carotídeo/fisiopatología , Disfunción Cognitiva/etiología , Disfunción Cognitiva/fisiopatología , Diabetes Mellitus Tipo 2/etiología , Diabetes Mellitus Tipo 2/fisiopatología , Modelos Animales de Enfermedad , Epigénesis Genética , Humanos , Hipertensión/etiología , Hipertensión/fisiopatología , Hipoxia/complicaciones , Hipoxia/fisiopatología , Resistencia a la Insulina/fisiología , Ratones , Modelos Biológicos , Ratas , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Apnea Obstructiva del Sueño/etiología
8.
PLoS Genet ; 16(5): e1008757, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32379754

RESUMEN

In the last decades in vitro studies highlighted the potential for crosstalk between Hypoxia-Inducible Factor-(HIF) and glucocorticoid-(GC) signalling pathways. However, how this interplay precisely occurs in vivo is still debated. Here, we use zebrafish larvae (Danio rerio) to elucidate how and to what degree hypoxic signalling affects the endogenous glucocorticoid pathway and vice versa, in vivo. Firstly, our results demonstrate that in the presence of upregulated HIF signalling, both glucocorticoid receptor (Gr) responsiveness and endogenous cortisol levels are repressed in 5 days post fertilisation larvae. In addition, despite HIF activity being low at normoxia, our data show that it already impedes both glucocorticoid activity and levels. Secondly, we further analysed the in vivo contribution of glucocorticoids to HIF activity. Interestingly, our results show that both glucocorticoid receptor (GR) and mineralocorticoid receptor (MR) play a key role in enhancing it. Finally, we found indications that glucocorticoids promote HIF signalling via multiple routes. Cumulatively, our findings allowed us to suggest a model for how this crosstalk occurs in vivo.


Asunto(s)
Glucocorticoides/farmacología , Factor 1 Inducible por Hipoxia/fisiología , Receptor Cross-Talk/fisiología , Pez Cebra , Animales , Animales Modificados Genéticamente , Translocador Nuclear del Receptor de Aril Hidrocarburo/genética , Embrión no Mamífero , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/genética , Glucocorticoides/metabolismo , Factor 1 Inducible por Hipoxia/metabolismo , Larva/genética , Larva/metabolismo , Receptor Cross-Talk/efectos de los fármacos , Receptores de Glucocorticoides/metabolismo , Receptores de Glucocorticoides/fisiología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Proteínas Supresoras de Tumor/genética , Pez Cebra/embriología , Pez Cebra/genética , Pez Cebra/crecimiento & desarrollo , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética
9.
Cancer Res ; 79(20): 5218-5232, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31488423

RESUMEN

Cancer cells rely on mitochondrial functions to regulate key survival and death signals. How cancer cells regulate mitochondrial autophagy (mitophagy) in the tumor microenvironment as well as utilize mitophagy as a survival signal is still not well understood. Here, we elucidate a key survival mechanism of mitochondrial NIX-mediated mitophagy within the hypoxic region of glioblastoma, the most malignant brain tumor. NIX was overexpressed in the pseudopalisading cells that envelop the hypoxic-necrotic regions, and mitochondrial NIX expression was robust in patient-derived glioblastoma tumor tissues and glioblastoma stem cells. NIX was required for hypoxia and oxidative stress-induced mitophagy through NFE2L2/NRF2 transactivation. Silencing NIX impaired mitochondrial reactive oxygen species clearance, cancer stem cell maintenance, and HIF/mTOR/RHEB signaling pathways under hypoxia, resulting in suppression of glioblastoma survival in vitro and in vivo. Clinical significance of these findings was validated by the compelling association between NIX expression and poor outcome for patients with glioblastoma. Taken together, our findings indicate that the NIX-mediated mitophagic pathway may represent a key therapeutic target for solid tumors, including glioblastoma. SIGNIFICANCE: NIX-mediated mitophagy regulates tumor survival in the hypoxic niche of glioblastoma microenvironment, providing a potential therapeutic target for glioblastoma.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/79/20/5218/F1.large.jpg.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Hipoxia de la Célula/fisiología , Glioblastoma/metabolismo , Proteínas de la Membrana/fisiología , Mitocondrias/metabolismo , Mitofagia/fisiología , Proteínas de Neoplasias/fisiología , Proteínas Proto-Oncogénicas/fisiología , Proteínas Supresoras de Tumor/fisiología , Animales , Neoplasias Encefálicas/patología , Glioblastoma/patología , Glioma/metabolismo , Glioma/patología , Xenoinjertos , Humanos , Factor 1 Inducible por Hipoxia/fisiología , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos BALB C , Ratones SCID , Factor 2 Relacionado con NF-E2/metabolismo , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/genética , Células Madre Neoplásicas/metabolismo , Estrés Oxidativo , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/genética , Interferencia de ARN , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/farmacología , Proteína Homóloga de Ras Enriquecida en el Cerebro/fisiología , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/fisiología , Serina-Treonina Quinasas TOR/fisiología , Microambiente Tumoral , Proteínas Supresoras de Tumor/antagonistas & inhibidores , Proteínas Supresoras de Tumor/genética
10.
J Obstet Gynaecol Res ; 45(10): 1967-1974, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31373134

RESUMEN

Angiogenesis is a very important process that helps establish and maintain the normal structure and function of the corpus luteum (CL). Early luteal development can be considered a kind of physiological injury with an inflammatory response; therefore, the inflammatory response may play an important role in the luteal angiogenesis. The inflammatory response is companied by activated leukocytes and their mediators. For luteal tissue, numerous activated leukocytes such as macrophages, neutrophils and eosinophils are present in the early luteal phase and are widely involved in neovascularization. The objective of this review is to describe the role of the inflammatory factors in the angiogenesis and to discuss their mechanism. Knowledge of action and mechanism of these inflammatory factors on angiogenic activity will be beneficial for the understanding of luteal function.


Asunto(s)
Cuerpo Lúteo/inmunología , Neovascularización Fisiológica , Animales , Femenino , Factores de Crecimiento de Fibroblastos/fisiología , Humanos , Factor 1 Inducible por Hipoxia/fisiología , Factor A de Crecimiento Endotelial Vascular/fisiología
11.
Sci Rep ; 9(1): 10599, 2019 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-31332228

RESUMEN

The transcription factor HIF-1 induces the expression of genes that are essential for cell survival and oxygen homeostasis in hypoxic conditions. The prolyl isomerase Pin1 plays a role in the regulation of HIF-1α. However, the mechanism by which Pin1 controls HIF-1α remains controversial. Surprisingly, we here show that a PIN1 transcript downregulates HIF-1α as a long non-coding RNA. Pin1-silencing siRNAs augmented the hypoxia-induced expression of HIF-1α, thereby upregulating the expression of HIF-1 target genes. However, the overexpression of Pin1 protein did not inhibit the hypoxic expression of HIF-1α. Pin1 restoration in Pin1-depleted cells also failed to reverse the induction of HIF-1α by Pin1 knockdown. Unexpectedly, HIF-1α was found to be induced by both siRNAs for PIN1 transcript variants 1/2 and that for PIN1 transcript variants 2/3, indicating that the PIN1 transcript variant 2 (PIN1-v2) is responsible for HIF-1α induction. Mechanistically, PIN1-v2, which is classified as a long non-coding RNA due to early termination of translation, was evaluated to inhibit the transcription of HIF1A gene. In conclusion, PIN1-v2 may function in balancing the HIF-1-driven gene expression under hypoxia.


Asunto(s)
Factor 1 Inducible por Hipoxia/metabolismo , Hipoxia/metabolismo , Peptidilprolil Isomerasa de Interacción con NIMA/metabolismo , ARN Largo no Codificante/metabolismo , Western Blotting , Línea Celular , Inmunoprecipitación de Cromatina , Humanos , Factor 1 Inducible por Hipoxia/fisiología , Peptidilprolil Isomerasa de Interacción con NIMA/fisiología , ARN Largo no Codificante/fisiología , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
12.
Neuromolecular Med ; 21(4): 414-431, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-30911877

RESUMEN

The blood-brain barrier (BBB) plays an important role in brain homeostasis. Hypoxia/ischemia constitutes an important stress factor involved in several neurological disorders by inducing the disruption of the BBB, ultimately leading to cerebral edema formation. Yet, our current understanding of the cellular and molecular mechanisms underlying the BBB disruption following cerebral hypoxia/ischemia remains limited. Stem cell-based models of the human BBB present some potentials to address such issues. Yet, such models have not been validated in regard of its ability to respond to hypoxia/ischemia as existing models. In this study, we investigated the cellular response of two iPSC-derived brain microvascular endothelial cell (BMEC) monolayers to respond to oxygen-glucose deprivation (OGD) stress, using two induced pluripotent stem cells (iPSC) lines. iPSC-derived BMECs responded to prolonged (24 h) and acute (6 h) OGD by showing a decrease in the barrier function and a decrease in tight junction complexes. Such iPSC-derived BMECs responded to OGD stress via a partial activation of the HIF-1 pathway, whereas treatment with anti-angiogenic pharmacological inhibitors (sorafenib, sunitinib) during reoxygenation worsened the barrier function. Taken together, our results suggest such models can respond to hypoxia/ischemia similarly to existing in vitro models and support the possible use of this model as a screening platform for identifying novel drug candidates capable to restore the barrier function following hypoxic/ischemic injury.


Asunto(s)
Barrera Hematoencefálica/fisiología , Células Endoteliales/efectos de los fármacos , Factor 1 Inducible por Hipoxia/fisiología , Hipoxia-Isquemia Encefálica/fisiopatología , Daño por Reperfusión/fisiopatología , Transducción de Señal/fisiología , Astrocitos/citología , Astrocitos/efectos de los fármacos , Diferenciación Celular , Hipoxia de la Célula , Línea Celular Transformada , Células Cultivadas , Claudina-5/fisiología , Técnicas de Cocultivo , Células Endoteliales/metabolismo , Femenino , Glucosa/farmacología , Humanos , Prolina Dioxigenasas del Factor Inducible por Hipoxia/antagonistas & inhibidores , Células Madre Pluripotentes Inducidas/citología , Células-Madre Neurales/citología , Neuronas/citología , Neuronas/efectos de los fármacos , Oxígeno/farmacología , Uniones Estrechas , Factor A de Crecimiento Endotelial Vascular/metabolismo
13.
Sheng Li Xue Bao ; 70(5): 511-520, 2018 Oct 25.
Artículo en Chino | MEDLINE | ID: mdl-30377690

RESUMEN

Hypoxic exposure activates hypoxia inducible factors (HIFs) to up-regulate the expression of its target genes. These genes encode glucose metabolism related proteins, such as glucose transporters (GLUTs) and glycolysis related enzymes, including lactate dehydrogenase A (LDHA) and aldolase A (ALDA). Therefore, HIFs participate in oxygenolysis of glucose and play an important role in mediating hypoxia response and weight loss. Exercise training influences fatty acid metabolism, insulin sensitivity and body energy balance through activating peroxisome proliferator-activated receptors (PPARs), which plays an active role in losing weight. In addition, hypoxic exposure or exercise training can activate energy sensor 5'-AMP activated protein kinase (AMPK) in cells and promote oxidation of glucose and fatty acid and weight loss. It has been shown that hypoxic training exerts a better effects on controlling weight, compared with either hypoxic exposure or exercise training alone. This paper reviewed synergistic interactions among HIFs, PPARs and AMPK under hypoxic training and proposed possible mechanisms of hypoxic training-induced weight loss via AMPK-HIFs axis or AMPK-PPARs axis, thus providing theoretical guidance for application of hypoxic training in weight control.


Asunto(s)
Proteínas Quinasas Activadas por AMP/fisiología , Factor 1 Inducible por Hipoxia/fisiología , Hipoxia , Receptores Activados del Proliferador del Peroxisoma/fisiología , Pérdida de Peso , Animales , Peso Corporal , Metabolismo Energético , Ácidos Grasos , Fructosa-Bifosfato Aldolasa/fisiología , Glucosa , Proteínas Facilitadoras del Transporte de la Glucosa/fisiología , Humanos , Resistencia a la Insulina , Isoenzimas/fisiología , L-Lactato Deshidrogenasa/fisiología , Lactato Deshidrogenasa 5 , Metabolismo de los Lípidos , Oxidación-Reducción , Regulación hacia Arriba
14.
Pharmacol Res ; 137: 159-169, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30315965

RESUMEN

Hypoxia-inducible factor-1 (HIF-1) is a transcription factor that consists of two subunits, the HIF-1α and HIF-1ß (ARNT). Under hypoxic conditions, HIF-1 is an adaptive system that regulates the transcription of multiple genes associated with growth, angiogenesis, proliferation, glucose transport, metabolism, pH regulation and cell death. However, aberrant HIF-1 activation contributes to the pathophysiology of several human diseases such as cancer, ischemic cardiovascular disorders, and pulmonary and kidney diseases. A growing body of evidence indicates that curcumin, a natural bioactive compound of turmeric root, significantly targets both HIF-1 subunits, but is more potent against HIF-1α. In this review, we have summarized the knowledge about the pharmacological effects of curcumin on HIF-1 and the related molecular mechanisms that may be effective candidates for the development of multi-targeted therapy for several human diseases.


Asunto(s)
Curcumina/farmacología , Factor 1 Inducible por Hipoxia/fisiología , Animales , Humanos , Factor 1 Inducible por Hipoxia/química , Cirrosis Hepática/metabolismo , Enfermedades Metabólicas/metabolismo , Neoplasias/metabolismo , Remodelación Vascular
15.
Praxis (Bern 1994) ; 107(21): 1155-1159, 2018.
Artículo en Alemán | MEDLINE | ID: mdl-30326811

RESUMEN

Is Oxygen Deficiency Always Harmful? Abstract. The role of the cardiovascular circulation is to supply tissue with oxygen and nutrients. Oxygen deficiency (hypoxia) is considered life-threatening, since cells die, either through apoptotic or necrotic processes. Tissue tries to counteract this by means of evolutionary signalling pathways, such as the nuclear hypoxia-inducible factor, which protects the tissue by promoting cell survival strategies and simultaneously intervening in angiogenesis, haematogenesis and metabolic processes. Recent findings indicate that these conserved signalling pathways can also function as therapeutic approaches in wound healing of bones and skin, as well as in the regeneration of tissues, e.g. in the liver, and in the hematopoietic system.


Asunto(s)
Apoptosis/fisiología , Factor 1 Inducible por Hipoxia/fisiología , Hipoxia/fisiopatología , Necrosis/fisiopatología , Supervivencia Celular/fisiología , Metabolismo Energético/fisiología , Humanos , Neovascularización Fisiológica/fisiología , Regeneración/fisiología
16.
Med. intensiva (Madr., Ed. impr.) ; 42(6): 380-390, ago.-sept. 2018. tab, graf
Artículo en Español | IBECS | ID: ibc-178650

RESUMEN

La hipoxia de la altitud (hipoxia hipobárica) no deja de ser una hipoxia celular similar a la que presentan los enfermos críticos. Estudiar a los alpinistas expuestos a la hipoxia extrema ofrece la ventaja de que es una población relativamente homogénea y sana, en contraste con la población heterogénea y generalmente menos saludable que suele observarse en las Unidades de Cuidados Críticos. El conocimiento de la fisiología y la enfermedad de la altitud abren caminos para comprender en qué medida afecta la hipoxia a los pacientes críticos. Los cambios comparables en la biogénesis mitocondrial entre ambos grupos pueden reflejar respuestas adaptativas similares y sugieren intervenciones terapéuticas basadas en la protección o estimulación de la biogénesis mitocondrial. El predominio del alelo homocigótico de inserción (II) de la enzima de conversión de la angiotensina está presente tanto en las ascensiones exitosas sin oxígeno por encima de los 8.000 m como en la supervivencia de algunas enfermedades de los enfermos críticos


High altitude sickness (hypobaric hypoxia) is a form of cellular hypoxia similar to that suffered by critically ill patients. The study of mountaineers exposed to extreme hypoxia offers the advantage of involving a relatively homogeneous and healthy population compared to those typically found in Intensive Care Units (ICUs), which are heterogeneous and generally less healthy. Knowledge of altitude physiology and pathology allows us to understanding how hypoxia affects critical patients. Comparable changes in mitochondrial biogenesis between both groups may reflect similar adaptive responses and suggest therapeutic interventions based on the protection or stimulation of such mitochondrial biogenesis. Predominance of the homozygous insertion (II) allele of the angiotensin-converting enzyme gene is present in both individuals who perform successful ascensions without oxygen above 8000 m and in critical patients who overcome certain disease conditions


Asunto(s)
Humanos , Mal de Altura/fisiopatología , Enfermedad Crítica/terapia , Hipoxia de la Célula , Factor 1 Inducible por Hipoxia/fisiología
17.
Rev. esp. cardiol. (Ed. impr.) ; 71(6): 440-449, jun. 2018. ilus, graf
Artículo en Español | IBECS | ID: ibc-178556

RESUMEN

Introducción y objetivos: El objetivo es estudiar la angiogénesis coronaria inducida por el suero coronario y la implicación del factor inducible por hipoxia-1A (FIH-1A) en la reparación de la obstrucción microvascular (OMV) tras un infarto agudo de miocardio (IAM). Métodos: Se indujo un IAM en cerdos mediante oclusión coronaria durante 90 min. Se dividió a los animales entre 1 grupo de control y 4 grupos con IAM: sin reperfusión y 1 min, 1 semana y 1 mes tras la reperfusión. Se cuantificaron la OMV y la densidad microvascular. Se determinó la capacidad angiogénica del suero coronario mediante una prueba de tubulogénesis in vitro. Se determinaron las concentraciones circulantes del FIH-1A, su implicación en el miocardio infartado y el efecto del bloqueo del FIH-1A in vitro. Resultados: En la zona infartada, la microvascularización disminuye tras 90 min de isquemia, la OMV aparece tras 1 min de reperfusión, es máxima 1 semana tras la reperfusión y se resuelve en 1 mes. La capacidad angiogénica del suero aumenta durante la isquemia y 1 min tras la reperfusión (32 ± 4 y 41 ± 9 frente a 3 ± 3 tubos en el grupo de control; p < 0,01). Las concentraciones circulantes del FIH-1A se incrementan durante la isquemia (5 min de isquemia: 273 ± 52 frente a 148 ± 48 pg/ml de los controles; p < 0,01) y se hallan en la microvasculatura de todos los grupos con IAM (sin reperfusión frente a control, el 67 ± 5% frente al 15 ± 17%; p < 0,01). El bloqueo del FIH-1A in vitro reduce la capacidad angiogénica inducida por el suero. Conclusiones: El suero coronario activa la neoangiogénesis desde antes de la reperfusión, y el FIH-1A podría tener un papel crucial. La capacidad angiogénica del suero coronario puede contribuir a la comprensión de la fisiopatología tras el IAM


Introduction and objectives: Microvascular obstruction (MVO) exerts deleterious effects following acute myocardial infarction (AMI). We investigated coronary angiogenesis induced by coronary serum and the role of hypoxia-inducible factor-1A (HIF-1A) in MVO repair. Methods: Myocardial infarction was induced in swine by transitory 90-minute coronary occlusion. The pigs were divided into a control group and 4 AMI groups: no reperfusion, 1 minute, 1 week and 1 month after reperfusion. Microvascular obstruction and microvessel density were quantified. The proangiogenic effect of coronary serum drawn from coronary sinus on endothelial cells was evaluated using an in vitro tubulogenesis assay. Circulating and myocardial HIF-1A levels and the effect of in vitro blockade of HIF-1A was assessed. Results: Compared with control myocardium, microvessel density decreased at 90-minute ischemia, and MVO first occurred at 1 minute after reperfusion. Both peaked at 1 week and almost completely resolved at 1 month. Coronary serum exerted a neoangiogenic effect on coronary endothelial cells in vitro, peaking at ischemia and 1 minute postreperfusion (32 ± 4 and 41 ± 9 tubes vs control: 3 ± 3 tubes; P < .01). Hypoxia-inducible factor-1A increased in serum during ischemia (5-minute ischemia: 273 ± 52 pg/mL vs control: 148 ± 48 pg/mL; P < .01) being present on microvessels of all AMI groups (no reperfusion: 67% ± 5% vs control: 15% ± 17%; P < .01). In vitro blockade of HIF-1A reduced the angiogenic response induced by serum. Conclusions: Coronary serum represents a potent neoangiogenic stimulus even before reperfusion; HIF-1A might be crucial. Coronary neoangiogenesis induced by coronary serum can contribute to understanding the pathophysiology of AMI


Asunto(s)
Animales , Suero/fisiología , Neovascularización Fisiológica/fisiología , Factor 1 Inducible por Hipoxia/fisiología , Infarto del Miocardio/fisiopatología , Microvasos/fisiología , Modelos Animales de Enfermedad
18.
Med Intensiva (Engl Ed) ; 42(6): 380-390, 2018.
Artículo en Inglés, Español | MEDLINE | ID: mdl-28919307

RESUMEN

High altitude sickness (hypobaric hypoxia) is a form of cellular hypoxia similar to that suffered by critically ill patients. The study of mountaineers exposed to extreme hypoxia offers the advantage of involving a relatively homogeneous and healthy population compared to those typically found in Intensive Care Units (ICUs), which are heterogeneous and generally less healthy. Knowledge of altitude physiology and pathology allows us to understanding how hypoxia affects critical patients. Comparable changes in mitochondrial biogenesis between both groups may reflect similar adaptive responses and suggest therapeutic interventions based on the protection or stimulation of such mitochondrial biogenesis. Predominance of the homozygous insertion (II) allele of the angiotensin-converting enzyme gene is present in both individuals who perform successful ascensions without oxygen above 8000 m and in critical patients who overcome certain disease conditions.


Asunto(s)
Mal de Altura/fisiopatología , Enfermedad Crítica/terapia , Hipoxia de la Célula , Humanos , Factor 1 Inducible por Hipoxia/fisiología
19.
Am J Chin Med ; 45(8): 1683-1708, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29121798

RESUMEN

This study evaluated the effects of Angelica sinensis extract [Dang Gui (DG)] administered before 60[Formula: see text]min of middle cerebral artery occlusion followed by 3[Formula: see text]d of reperfusion and investigated the involvement of mitogen-activated protein kinase (MAPK)/hypoxia-inducible factor (HIF)-1[Formula: see text] signaling in the cortical ischemic penumbra. DG was intraperitoneally administered at a dose of 0.25[Formula: see text]g/kg (DG-0.25g), 0.5[Formula: see text]g/kg (DG-0.5g), or 1[Formula: see text]g/kg (DG-1g) 30[Formula: see text]min before the onset of cerebral ischemia. Our study results revealed that DG-0.5g and DG-1g pretreatment effectively attenuated cerebral infarct and improved neurological deficits. DG-0.5g and DG-1g pretreatment significantly downregulated glial fibrillary acidic protein (GFAP), cytochrome c, and cleaved caspase-3 expression and upregulated phospho-p38 MAPK (p-p38 MAPK)/p38 MAPK, phospho-cAMP response element-binding protein (p-CREB)/CREB, cytosolic and mitochondrial phospho-Bad (p-Bad)/Bad ratios, and HIF-1[Formula: see text], vascular endothelial growth factor-A (VEGF-A), phospho-90 kDa ribosomal S6 kinase (p-p90RSK), and von Willebrand factor (vWF) expression in the cortical ischemic penumbra. Pretreatment with SB203580, a p38 MAPK inhibitor, dramatically abrogated the upregulating effects of DG-1g on p-p38 MAPK/p38 MAPK, p-CREB/CREB, and p-Bad/Bad ratios and HIF-1[Formula: see text], VEGF-A, and vWF expression and the downregulating effects of DG-1g on GFAP, cytochrome c, cleaved caspase-3, and cerebral infarction. DG-0.5g and DG-1g pretreatment provided neuroprotective effects against astrocyte-mediated cerebral infarction by activating angiogenic and anti-apoptotic signaling. Moreover, the angiogenic and anti-apoptotic effects of DG pretreatment can be attributed to the activation of p38 MAPK/HIF-1[Formula: see text]/VEGF-A/vWF signaling and p38 MAPK/HIF-1[Formula: see text]/VEGF-A/p-Bad-related regulation of cytochrome c/caspase-3 signaling, respectively, in the cortical ischemic penumbra 3[Formula: see text]d after reperfusion.


Asunto(s)
Angelica sinensis/química , Inductores de la Angiogénesis , Apoptosis/efectos de los fármacos , Isquemia Encefálica/tratamiento farmacológico , Medicamentos Herbarios Chinos/administración & dosificación , Medicamentos Herbarios Chinos/farmacología , Factor 1 Inducible por Hipoxia/metabolismo , Fitoterapia , Daño por Reperfusión/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Isquemia Encefálica/patología , Relación Dosis-Respuesta a Droga , Medicamentos Herbarios Chinos/aislamiento & purificación , Factor 1 Inducible por Hipoxia/fisiología , Infusiones Parenterales , Masculino , Ratas Sprague-Dawley , Daño por Reperfusión/patología , Factor A de Crecimiento Endotelial Vascular/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología
20.
Ann Am Thorac Soc ; 14(Supplement_3): S233-S236, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28945477

RESUMEN

Mucosal tissues represent surfaces that are exposed to the outside world and provide a conduit for internal and external communication. Tissues such as the intestine and the lung are lined by layer(s) of epithelial cells that, when organized in three dimensions, provide a critical barrier to the flux of luminal contents. This selective barrier is provided through the regulated expression of junctional proteins and mucins. Tissue oxygen metabolism is central to the maintenance of homeostasis in the mucosa. In some organs (e.g., the colon), low baseline Po2 determines tissue metabolism and results in basal expression of the transcription factor, hypoxia-inducible factor (HIF), which is enhanced after ischemia/inflammation. Recent studies have indicated that HIF contributes fundamentally to the expression of barrier-related genes and in the regulation of barrier-adaptive responses within the mucosa. Here, we briefly review recent literature on the topic of hypoxia and HIF regulation of barrier in mucosal health and during disease.


Asunto(s)
Células Epiteliales/fisiología , Factor 1 Inducible por Hipoxia/fisiología , Membrana Mucosa/fisiología , Uniones Adherentes/fisiología , Animales , Homeostasis/fisiología , Humanos , Oxígeno/metabolismo , Uniones Estrechas/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA