Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 16.927
Filter
Add more filters

Publication year range
1.
Annu Rev Immunol ; 38: 341-363, 2020 04 26.
Article in English | MEDLINE | ID: mdl-31961750

ABSTRACT

Recent years have witnessed an emergence of interest in understanding metabolic changes associated with immune responses, termed immunometabolism. As oxygen is central to all aerobic metabolism, hypoxia is now recognized to contribute fundamentally to inflammatory and immune responses. Studies from a number of groups have implicated a prominent role for oxygen metabolism and hypoxia in innate immunity of healthy tissue (physiologic hypoxia) and during active inflammation (inflammatory hypoxia). This inflammatory hypoxia emanates from a combination of recruited inflammatory cells (e.g., neutrophils, eosinophils, and monocytes), high rates of oxidative metabolism, and the activation of multiple oxygen-consuming enzymes during inflammation. These localized shifts toward hypoxia have identified a prominent role for the transcription factor hypoxia-inducible factor (HIF) in the regulation of innate immunity. Such studies have provided new and enlightening insight into our basic understanding of immune mechanisms, and extensions of these findings have identified potential therapeutic targets. In this review, we summarize recent literature around the topic of innate immunity and mucosal hypoxia with a focus on transcriptional responses mediated by HIF.


Subject(s)
Hypoxia/immunology , Hypoxia/metabolism , Immunity, Innate , Animals , Dendritic Cells/immunology , Dendritic Cells/metabolism , Disease Management , Disease Susceptibility , Energy Metabolism , Gene Expression Regulation , Host-Pathogen Interactions/immunology , Humans , Hypoxia/genetics , Hypoxia-Inducible Factor 1/genetics , Hypoxia-Inducible Factor 1/metabolism , Immunomodulation , Macrophages/immunology , Macrophages/metabolism , Monocytes/immunology , Monocytes/metabolism , Signal Transduction
2.
Cell ; 187(3): 659-675.e18, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38215760

ABSTRACT

The electron transport chain (ETC) of mitochondria, bacteria, and archaea couples electron flow to proton pumping and is adapted to diverse oxygen environments. Remarkably, in mice, neurological disease due to ETC complex I dysfunction is rescued by hypoxia through unknown mechanisms. Here, we show that hypoxia rescue and hyperoxia sensitivity of complex I deficiency are evolutionarily conserved to C. elegans and are specific to mutants that compromise the electron-conducting matrix arm. We show that hypoxia rescue does not involve the hypoxia-inducible factor pathway or attenuation of reactive oxygen species. To discover the mechanism, we use C. elegans genetic screens to identify suppressor mutations in the complex I accessory subunit NDUFA6/nuo-3 that phenocopy hypoxia rescue. We show that NDUFA6/nuo-3(G60D) or hypoxia directly restores complex I forward activity, with downstream rescue of ETC flux and, in some cases, complex I levels. Additional screens identify residues within the ubiquinone binding pocket as being required for the rescue by NDUFA6/nuo-3(G60D) or hypoxia. This reveals oxygen-sensitive coupling between an accessory subunit and the quinone binding pocket of complex I that can restore forward activity in the same manner as hypoxia.


Subject(s)
Caenorhabditis elegans , Electron Transport Complex I , Hypoxia , Animals , Mice , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Electron Transport Complex I/metabolism , Hypoxia/genetics , Hypoxia/metabolism , Mitochondria/genetics , Mitochondria/metabolism , Oxygen/metabolism
3.
Cell ; 180(1): 7-8, 2020 01 09.
Article in English | MEDLINE | ID: mdl-31951521

ABSTRACT

To celebrate the 2019 Nobel Prize in Physiology or Medicine, awarded to William G. Kaelin Jr., Sir Peter J. Ratcliffe, and Gregg L. Semenza for their discoveries of how cells sense and adapt to oxygen availability, we've asked four researchers in the oxygen-sensing field what they see on the horizon after this momentous milestone.


Subject(s)
Biomedical Research/trends , Hypoxia/metabolism , Oxygen/metabolism , Awards and Prizes , Biomedical Research/history , History, 20th Century , Humans , Nobel Prize , Research Personnel
4.
Cell ; 181(3): 716-727.e11, 2020 04 30.
Article in English | MEDLINE | ID: mdl-32259488

ABSTRACT

Human cells are able to sense and adapt to variations in oxygen levels. Historically, much research in this field has focused on hypoxia-inducible factor (HIF) signaling and reactive oxygen species (ROS). Here, we perform genome-wide CRISPR growth screens at 21%, 5%, and 1% oxygen to systematically identify gene knockouts with relative fitness defects in high oxygen (213 genes) or low oxygen (109 genes), most without known connection to HIF or ROS. Knockouts of many mitochondrial pathways thought to be essential, including complex I and enzymes in Fe-S biosynthesis, grow relatively well at low oxygen and thus are buffered by hypoxia. In contrast, in certain cell types, knockout of lipid biosynthetic and peroxisomal genes causes fitness defects only in low oxygen. Our resource nominates genetic diseases whose severity may be modulated by oxygen and links hundreds of genes to oxygen homeostasis.


Subject(s)
Lipid Metabolism/genetics , Mitochondria/genetics , Oxygen/metabolism , Transcriptome/genetics , Cell Hypoxia , Genetic Testing/methods , Genome-Wide Association Study/methods , HEK293 Cells , Humans , Hypoxia/metabolism , K562 Cells , Lipid Metabolism/physiology , Lipids/genetics , Lipids/physiology , Mitochondria/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/physiology
5.
Cell ; 177(6): 1507-1521.e16, 2019 05 30.
Article in English | MEDLINE | ID: mdl-31031004

ABSTRACT

Friedreich's ataxia (FRDA) is a devastating, multisystemic disorder caused by recessive mutations in the mitochondrial protein frataxin (FXN). FXN participates in the biosynthesis of Fe-S clusters and is considered to be essential for viability. Here we report that when grown in 1% ambient O2, FXN null yeast, human cells, and nematodes are fully viable. In human cells, hypoxia restores steady-state levels of Fe-S clusters and normalizes ATF4, NRF2, and IRP2 signaling events associated with FRDA. Cellular studies and in vitro reconstitution indicate that hypoxia acts through HIF-independent mechanisms that increase bioavailable iron as well as directly activate Fe-S synthesis. In a mouse model of FRDA, breathing 11% O2 attenuates the progression of ataxia, whereas breathing 55% O2 hastens it. Our work identifies oxygen as a key environmental variable in the pathogenesis associated with FXN depletion, with important mechanistic and therapeutic implications.


Subject(s)
Hypoxia/metabolism , Iron-Binding Proteins/metabolism , Iron-Sulfur Proteins/metabolism , Activating Transcription Factor 4/metabolism , Animals , Caenorhabditis elegans/metabolism , Female , Friedreich Ataxia/metabolism , HEK293 Cells , Humans , Hypoxia/physiopathology , Iron/metabolism , Iron Regulatory Protein 2/metabolism , Iron-Binding Proteins/physiology , Iron-Sulfur Proteins/physiology , K562 Cells , Male , Mice , Mice, Knockout , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , NF-E2-Related Factor 2/metabolism , Oxidative Stress , Saccharomyces cerevisiae/metabolism , Sulfur/metabolism , Frataxin
6.
Cell ; 161(3): 595-609, 2015 Apr 23.
Article in English | MEDLINE | ID: mdl-25892225

ABSTRACT

Organisms must be able to respond to low oxygen in a number of homeostatic and pathological contexts. Regulation of hypoxic responses via the hypoxia-inducible factor (HIF) is well established, but evidence indicates that other, HIF-independent mechanisms are also involved. Here, we report a hypoxic response that depends on the accumulation of lactate, a metabolite whose production increases in hypoxic conditions. We find that the NDRG3 protein is degraded in a PHD2/VHL-dependent manner in normoxia but is protected from destruction by binding to lactate that accumulates under hypoxia. The stabilized NDRG3 protein binds c-Raf to mediate hypoxia-induced activation of Raf-ERK pathway, promoting angiogenesis and cell growth. Inhibiting cellular lactate production abolishes the NDRG3-mediated hypoxia responses. Our study, therefore, elucidates the molecular basis for lactate-induced hypoxia signaling, which can be exploited for the development of therapies targeting hypoxia-induced diseases.


Subject(s)
Hypoxia/metabolism , Lactic Acid/metabolism , Cell Hypoxia , Cell Line , Gene Expression Regulation , Humans , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Intracellular Signaling Peptides and Proteins , MAP Kinase Signaling System , Neovascularization, Pathologic/metabolism , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/metabolism , Oxygen/metabolism , Protein Binding , raf Kinases/metabolism
7.
Nature ; 631(8020): 350-359, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38926577

ABSTRACT

Insect respiration has long been thought to be solely dependent on an elaborate tracheal system without assistance from the circulatory system or immune cells1,2. Here we describe that Drosophila crystal cells-myeloid-like immune cells called haemocytes-control respiration by oxygenating Prophenoloxidase 2 (PPO2) proteins. Crystal cells direct the movement of haemocytes between the trachea of the larval body wall and the circulation to collect oxygen. Aided by copper and a neutral pH, oxygen is trapped in the crystalline structures of PPO2 in crystal cells. Conversely, PPO2 crystals can be dissolved when carbonic anhydrase lowers the intracellular pH and then reassembled into crystals in cellulo by adhering to the trachea. Physiologically, larvae lacking crystal cells or PPO2, or those expressing a copper-binding mutant of PPO2, display hypoxic responses under normoxic conditions and are susceptible to hypoxia. These hypoxic phenotypes can be rescued by hyperoxia, expression of arthropod haemocyanin or prevention of larval burrowing activity to expose their respiratory organs. Thus, we propose that insect immune cells collaborate with the tracheal system to reserve and transport oxygen through the phase transition of PPO2 crystals, facilitating internal oxygen homeostasis in a process that is comparable to vertebrate respiration.


Subject(s)
Catechol Oxidase , Drosophila Proteins , Drosophila melanogaster , Enzyme Precursors , Hemocytes , Oxygen , Phase Transition , Respiration , Animals , Female , Male , Biological Transport , Carbonic Anhydrases/metabolism , Catechol Oxidase/metabolism , Copper/metabolism , Crystallization , Drosophila melanogaster/anatomy & histology , Drosophila melanogaster/cytology , Drosophila melanogaster/enzymology , Drosophila melanogaster/immunology , Drosophila melanogaster/metabolism , Drosophila Proteins/metabolism , Enzyme Precursors/metabolism , Hemocyanins/metabolism , Hemocytes/immunology , Hemocytes/metabolism , Homeostasis , Hydrogen-Ion Concentration , Hyperoxia/metabolism , Hypoxia/metabolism , Larva/anatomy & histology , Larva/cytology , Larva/immunology , Larva/metabolism , Oxygen/metabolism
8.
Nature ; 618(7967): 974-980, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37258677

ABSTRACT

Phosphorus is a limiting nutrient that is thought to control oceanic oxygen levels to a large extent1-3. A possible increase in marine phosphorus concentrations during the Ediacaran Period (about 635-539 million years ago) has been proposed as a driver for increasing oxygen levels4-6. However, little is known about the nature and evolution of phosphorus cycling during this time4. Here we use carbonate-associated phosphate (CAP) from six globally distributed sections to reconstruct oceanic phosphorus concentrations during a large negative carbon-isotope excursion-the Shuram excursion (SE)-which co-occurred with global oceanic oxygenation7-9. Our data suggest pulsed increases in oceanic phosphorus concentrations during the falling and rising limbs of the SE. Using a quantitative biogeochemical model, we propose that this observation could be explained by carbon dioxide and phosphorus release from marine organic-matter oxidation primarily by sulfate, with further phosphorus release from carbon-dioxide-driven weathering on land. Collectively, this may have resulted in elevated organic-pyrite burial and ocean oxygenation. Our CAP data also seem to suggest equivalent oceanic phosphorus concentrations under maximum and minimum extents of ocean anoxia across the SE. This observation may reflect decoupled phosphorus and ocean anoxia cycles, as opposed to their coupled nature in the modern ocean. Our findings point to external stimuli such as sulfate weathering rather than internal oceanic phosphorus-oxygen cycling alone as a possible control on oceanic oxygenation in the Ediacaran. In turn, this may help explain the prolonged rise of atmospheric oxygen levels.


Subject(s)
Oceans and Seas , Phosphorus , Seawater , Atmosphere/chemistry , Carbon Dioxide/metabolism , Carbon Isotopes , Geologic Sediments/chemistry , History, Ancient , Hypoxia/metabolism , Oxygen/analysis , Oxygen/history , Oxygen/metabolism , Phosphorus/analysis , Phosphorus/history , Phosphorus/metabolism , Seawater/chemistry , Sulfates/metabolism , Carbonates/analysis , Carbonates/metabolism , Oxidation-Reduction
9.
Trends Biochem Sci ; 49(7): 573-582, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38599898

ABSTRACT

Investigating how cells and organisms sense and respond to O2 levels is essential to our understanding of physiology and pathology. This field has advanced considerably since the discovery of the major transcription factor family, hypoxia-inducible factor (HIF), and the enzymes that control its levels: prolyl hydroxylases (PHDs). However, with its expansion, new complexities have emerged. Herein we highlight three main areas where, in our opinion, the research community could direct some of their attention. These include non-transcriptional roles of HIFs, specificity and O2 sensitivity of 2-oxoglutarate-dependent dioxygenases (2-OGDDs), and new tools and methods to detect O2 concentrations in cells and organs. A greater understanding of these areas would answer big questions and help drive our knowledge of cellular responses to hypoxia forward.


Subject(s)
Oxygen , Humans , Animals , Oxygen/metabolism , Hypoxia/metabolism , Hypoxia-Inducible Factor 1/metabolism
10.
Physiol Rev ; 101(3): 1177-1235, 2021 07 01.
Article in English | MEDLINE | ID: mdl-33570461

ABSTRACT

The carotid body (CB) is the main peripheral chemoreceptor for arterial respiratory gases O2 and CO2 and pH, eliciting reflex ventilatory, cardiovascular, and humoral responses to maintain homeostasis. This review examines the fundamental biology underlying CB chemoreceptor function, its contribution to integrated physiological responses, and its role in maintaining health and potentiating disease. Emphasis is placed on 1) transduction mechanisms in chemoreceptor (type I) cells, highlighting the role played by the hypoxic inhibition of O2-dependent K+ channels and mitochondrial oxidative metabolism, and their modification by intracellular molecules and other ion channels; 2) synaptic mechanisms linking type I cells and petrosal nerve terminals, focusing on the role played by the main proposed transmitters and modulatory gases, and the participation of glial cells in regulation of the chemosensory process; 3) integrated reflex responses to CB activation, emphasizing that the responses differ dramatically depending on the nature of the physiological, pathological, or environmental challenges, and the interactions of the chemoreceptor reflex with other reflexes in optimizing oxygen delivery to the tissues; and 4) the contribution of enhanced CB chemosensory discharge to autonomic and cardiorespiratory pathophysiology in obstructive sleep apnea, congestive heart failure, resistant hypertension, and metabolic diseases and how modulation of enhanced CB reactivity in disease conditions may attenuate pathophysiology.


Subject(s)
Autonomic Nervous System/metabolism , Carotid Body/metabolism , Chemoreceptor Cells/metabolism , Hypoxia/metabolism , Animals , Cardiovascular System/metabolism , Humans
11.
Mol Cell ; 77(1): 95-107.e5, 2020 01 02.
Article in English | MEDLINE | ID: mdl-31628042

ABSTRACT

GTP cyclohydrolase I (GTPCH), 6-pyruvoyltetrahydropterin synthase (PTPS), and sepiapterin reductase (SR) are sequentially responsible for de novo synthesis of tetrahydrobiopterin (BH4), a known co-factor for nitric oxide synthase (NOS). The implication of BH4-biosynthesis process in tumorigenesis remains to be investigated. Here, we show that PTPS, which is highly expressed in early-stage colorectal cancer, is phosphorylated at Thr 58 by AMPK under hypoxia; this phosphorylation promotes PTPS binding to LTBP1 and subsequently drives iNOS-mediated LTBP1 S-nitrosylation through proximal-coupling BH4 production within the PTPS/iNOS/LTBP1 complex. In turn, LTBP1 S-nitrosylation results in proteasome-dependent LTBP1 protein degradation, revealing an inverse relationship between PTPS pT58 and LTBP1 stability. Physiologically, the repressive effect of PTPS on LTBP1 leads to impaired transforming growth factor ß (TGF-ß) secretion and thereby maintains tumor cell growth under hypoxia. Our findings illustrate a molecular mechanism underlying the regulation of LTBP1-TGF-ß signaling by the BH4-biosynthesis pathway and highlight the specific requirement of PTPS for tumor growth.


Subject(s)
Cell Proliferation/physiology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Hypoxia/metabolism , Latent TGF-beta Binding Proteins/metabolism , Phosphorus-Oxygen Lyases/metabolism , Animals , Cell Line , Cell Line, Tumor , HCT116 Cells , HEK293 Cells , HT29 Cells , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Nitric Oxide Synthase/metabolism , Phosphorylation/physiology , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Signal Transduction/physiology , Transforming Growth Factor beta/metabolism
12.
EMBO J ; 42(6): e112067, 2023 03 15.
Article in English | MEDLINE | ID: mdl-36808622

ABSTRACT

A role for hypoxia-inducible factors (HIFs) in hypoxia-dependent regulation of tumor cell metabolism has been thoroughly investigated and covered in reviews. However, there is limited information available regarding HIF-dependent regulation of nutrient fates in tumor and stromal cells. Tumor and stromal cells may generate nutrients necessary for function (metabolic symbiosis) or deplete nutrients resulting in possible competition between tumor cells and immune cells, a result of altered nutrient fates. HIF and nutrients in the tumor microenvironment (TME) affect stromal and immune cell metabolism in addition to intrinsic tumor cell metabolism. HIF-dependent metabolic regulation will inevitably result in the accumulation or depletion of essential metabolites in the TME. In response, various cell types in the TME will respond to these hypoxia-dependent alterations by activating HIF-dependent transcription to alter nutrient import, export, and utilization. In recent years, the concept of metabolic competition has been proposed for critical substrates, including glucose, lactate, glutamine, arginine, and tryptophan. In this review, we discuss how HIF-mediated mechanisms control nutrient sensing and availability in the TME, the competition for nutrients, and the metabolic cross-talk between tumor and stromal cells.


Subject(s)
Neoplasms , Tumor Microenvironment , Humans , Hypoxia/metabolism , Neoplasms/metabolism , Cell Hypoxia , Nutrients , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
13.
EMBO J ; 42(22): e114093, 2023 Nov 15.
Article in English | MEDLINE | ID: mdl-37902464

ABSTRACT

Owing to their capability to disrupt the oxidative protein folding environment in the endoplasmic reticulum (ER), thiol antioxidants, such as dithiothreitol (DTT), are used as ER-specific stressors. We recently showed that thiol antioxidants modulate the methionine-homocysteine cycle by upregulating an S-adenosylmethionine-dependent methyltransferase, rips-1, in Caenorhabditis elegans. However, the changes in cellular physiology induced by thiol stress that modulate the methionine-homocysteine cycle remain uncharacterized. Here, using forward genetic screens in C. elegans, we discover that thiol stress enhances rips-1 expression via the hypoxia response pathway. We demonstrate that thiol stress activates the hypoxia response pathway. The activation of the hypoxia response pathway by thiol stress is conserved in human cells. The hypoxia response pathway enhances thiol toxicity via rips-1 expression and confers protection against thiol toxicity via rips-1-independent mechanisms. Finally, we show that DTT might activate the hypoxia response pathway by producing hydrogen sulfide. Our studies reveal an intriguing interaction between thiol-mediated reductive stress and the hypoxia response pathway and challenge the current model that thiol antioxidant DTT disrupts only the ER milieu in the cell.


Subject(s)
Caenorhabditis elegans , Endoplasmic Reticulum , Animals , Humans , Caenorhabditis elegans/genetics , Endoplasmic Reticulum/metabolism , Antioxidants , Hypoxia/genetics , Hypoxia/metabolism , Homocysteine/metabolism , Methionine/metabolism , Endoplasmic Reticulum Stress
14.
Cell ; 148(3): 399-408, 2012 Feb 03.
Article in English | MEDLINE | ID: mdl-22304911

ABSTRACT

Oxygen homeostasis represents an organizing principle for understanding metazoan evolution, development, physiology, and pathobiology. The hypoxia-inducible factors (HIFs) are transcriptional activators that function as master regulators of oxygen homeostasis in all metazoan species. Rapid progress is being made in elucidating homeostatic roles of HIFs in many physiological systems, determining pathological consequences of HIF dysregulation in chronic diseases, and investigating potential targeting of HIFs for therapeutic purposes.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Biological Evolution , Oxygen/metabolism , Animals , Chronic Disease , Embryonic Development , Humans , Hypoxia/metabolism
15.
Cell ; 149(2): 295-306, 2012 Apr 13.
Article in English | MEDLINE | ID: mdl-22484060

ABSTRACT

Congenital scoliosis, a lateral curvature of the spine caused by vertebral defects, occurs in approximately 1 in 1,000 live births. Here we demonstrate that haploinsufficiency of Notch signaling pathway genes in humans can cause this congenital abnormality. We also show that in a mouse model, the combination of this genetic risk factor with an environmental condition (short-term gestational hypoxia) significantly increases the penetrance and severity of vertebral defects. We demonstrate that hypoxia disrupts FGF signaling, leading to a temporary failure of embryonic somitogenesis. Our results potentially provide a mechanism for the genesis of a host of common sporadic congenital abnormalities through gene-environment interaction.


Subject(s)
Gene-Environment Interaction , Scoliosis/embryology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Female , Haploinsufficiency , Humans , Hypoxia/metabolism , Male , Mesoderm/metabolism , Mice , Mice, Inbred C57BL , Pedigree , Penetrance , Receptors, Notch/metabolism , Scoliosis/congenital , Signal Transduction , Spine/embryology
16.
Proc Natl Acad Sci U S A ; 121(10): e2309957121, 2024 Mar 05.
Article in English | MEDLINE | ID: mdl-38422022

ABSTRACT

Hypoxia signaling influences tumor development through both cell-intrinsic and -extrinsic pathways. Inhibiting hypoxia-inducible factor (HIF) function has recently been approved as a cancer treatment strategy. Hence, it is important to understand how regulators of HIF may affect tumor growth under physiological conditions. Here we report that in aging mice factor-inhibiting HIF (FIH), one of the most studied negative regulators of HIF, is a haploinsufficient suppressor of spontaneous B cell lymphomas, particular pulmonary B cell lymphomas. FIH deficiency alters immune composition in aged mice and creates a tumor-supportive immune environment demonstrated in syngeneic mouse tumor models. Mechanistically, FIH-defective myeloid cells acquire tumor-supportive properties in response to signals secreted by cancer cells or produced in the tumor microenvironment with enhanced arginase expression and cytokine-directed migration. Together, these data demonstrate that under physiological conditions, FIH plays a key role in maintaining immune homeostasis and can suppress tumorigenesis through a cell-extrinsic pathway.


Subject(s)
Lymphoma, B-Cell , Repressor Proteins , Animals , Mice , Hypoxia/metabolism , Mixed Function Oxygenases/metabolism , Repressor Proteins/metabolism , Tumor Microenvironment
17.
Proc Natl Acad Sci U S A ; 121(8): e2303119121, 2024 Feb 20.
Article in English | MEDLINE | ID: mdl-38349880

ABSTRACT

Coupling red blood cell (RBC) supply to O2 demand is an intricate process requiring O2 sensing, generation of a stimulus, and signal transduction that alters upstream arteriolar tone. Although actively debated, this process has been theorized to be induced by hypoxia and to involve activation of endothelial inwardly rectifying K+ channels (KIR) 2.1 by elevated extracellular K+ to trigger conducted hyperpolarization via connexin40 (Cx40) gap junctions to upstream resistors. This concept was tested in resting healthy skeletal muscle of Cx40-/- and endothelial KIR2.1-/- mice using state-of-the-art live animal imaging where the local tissue O2 environment was manipulated using a custom gas chamber. Second-by-second capillary RBC flow responses were recorded as O2 was altered. A stepwise drop in PO2 at the muscle surface increased RBC supply in capillaries of control animals while elevated O2 elicited the opposite response; capillaries were confirmed to express Cx40. The RBC flow responses were rapid and tightly coupled to O2; computer simulations did not support hypoxia as a driving factor. In contrast, RBC flow responses were significantly diminished in Cx40-/- mice. Endothelial KIR2.1-/- mice, on the other hand, reacted normally to O2 changes, even when the O2 challenge was targeted to a smaller area of tissue with fewer capillaries. Conclusively, microvascular O2 responses depend on coordinated electrical signaling via Cx40 gap junctions, and endothelial KIR2.1 channels do not initiate the event. These findings reconceptualize the paradigm of blood flow regulation in skeletal muscle and how O2 triggers this process in capillaries independent of extracellular K+.


Subject(s)
Capillaries , Oxygen , Animals , Mice , Capillaries/physiology , Gap Junction alpha-5 Protein/metabolism , Gap Junctions/metabolism , Hypoxia/metabolism , Muscle, Skeletal/metabolism , Oxygen/metabolism
18.
Proc Natl Acad Sci U S A ; 121(1): e2315930120, 2024 Jan 02.
Article in English | MEDLINE | ID: mdl-38147558

ABSTRACT

Red blood cell (RBC) metabolic reprogramming upon exposure to high altitude contributes to physiological human adaptations to hypoxia, a multifaceted process critical to health and disease. To delve into the molecular underpinnings of this phenomenon, first, we performed a multi-omics analysis of RBCs from six lowlanders after exposure to high-altitude hypoxia, with longitudinal sampling at baseline, upon ascent to 5,100 m and descent to sea level. Results highlighted an association between erythrocyte levels of 2,3-bisphosphoglycerate (BPG), an allosteric regulator of hemoglobin that favors oxygen off-loading in the face of hypoxia, and expression levels of the Rhesus blood group RHCE protein. We then expanded on these findings by measuring BPG in RBCs from 13,091 blood donors from the Recipient Epidemiology and Donor Evaluation Study. These data informed a genome-wide association study using BPG levels as a quantitative trait, which identified genetic polymorphisms in the region coding for the Rhesus blood group RHCE as critical determinants of BPG levels in erythrocytes from healthy human volunteers. Mechanistically, we suggest that the Rh group complex, which participates in the exchange of ammonium with the extracellular compartment, may contribute to intracellular alkalinization, thus favoring BPG mutase activity.


Subject(s)
Altitude , Blood Group Antigens , Hypoxia , Rh-Hr Blood-Group System , Humans , 2,3-Diphosphoglycerate/metabolism , Erythrocytes/metabolism , Genome-Wide Association Study , Hypoxia/genetics , Hypoxia/metabolism , Polymorphism, Genetic , Rh-Hr Blood-Group System/genetics , Rh-Hr Blood-Group System/metabolism
19.
PLoS Genet ; 20(4): e1011232, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38669270

ABSTRACT

Animals often grow and develop in unpredictable environments where factors like food availability, temperature, and oxygen levels can fluctuate dramatically. To ensure proper sexual maturation into adulthood, juvenile animals need to adapt their growth and developmental rates to these fluctuating environmental conditions. Failure to do so can result in impaired maturation and incorrect body size. Here we describe a mechanism by which Drosophila larvae adapt their development in low oxygen (hypoxia). During normal development, larvae grow and increase in mass until they reach critical weight (CW), after which point a neuroendocrine circuit triggers the production of the steroid hormone ecdysone from the prothoracic gland (PG), which promotes maturation to the pupal stage. However, when raised in hypoxia (5% oxygen), larvae slow their growth and delay their maturation to the pupal stage. We find that, although hypoxia delays the attainment of CW, the maturation delay occurs mainly because of hypoxia acting late in development to suppress ecdysone production. This suppression operates through a distinct mechanism from nutrient deprivation, occurs independently of HIF-1 alpha and does not involve dilp8 or modulation of Ptth, the main neuropeptide that initiates ecdysone production in the PG. Instead, we find that hypoxia lowers the expression of the EGF ligand, spitz, and that the delay in maturation occurs due to reduced EGFR/ERK signaling in the PG. Our study sheds light on how animals can adjust their development rate in response to changing oxygen levels in their environment. Given that hypoxia is a feature of both normal physiology and many diseases, our findings have important implications for understanding how low oxygen levels may impact animal development in both normal and pathological situations.


Subject(s)
Drosophila Proteins , Drosophila melanogaster , Ecdysone , Epidermal Growth Factor , Larva , Signal Transduction , Animals , Ecdysone/metabolism , Larva/growth & development , Larva/genetics , Larva/metabolism , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Epidermal Growth Factor/metabolism , Epidermal Growth Factor/genetics , Drosophila melanogaster/growth & development , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Hypoxia/metabolism , Gene Expression Regulation, Developmental , ErbB Receptors/metabolism , ErbB Receptors/genetics , Oxygen/metabolism , Pupa/growth & development , Pupa/metabolism , Pupa/genetics
20.
EMBO J ; 41(23): e111239, 2022 12 01.
Article in English | MEDLINE | ID: mdl-36278281

ABSTRACT

Bone-derived mesenchymal stem cells (MSCs) reside in a hypoxic niche that maintains their differentiation potential. While hypoxia (low oxygen concentration) was reported to critically support stem cell function and osteogenesis, the molecular events triggering changes in stem cell fate decisions in response to normoxia (high oxygen concentration) remain elusive. Here, we study the impact of normoxia on mitochondrial-nuclear communication during stem cell differentiation. We show that normoxia-cultured murine MSCs undergo profound transcriptional alterations which cause irreversible osteogenesis defects. Mechanistically, high oxygen promotes chromatin compaction and histone hypo-acetylation, particularly on promoters and enhancers of osteogenic genes. Although normoxia induces metabolic rewiring resulting in elevated acetyl-CoA levels, histone hypo-acetylation occurs due to the trapping of acetyl-CoA inside mitochondria owing to decreased citrate carrier (CiC) activity. Restoring the cytosolic acetyl-CoA pool remodels the chromatin landscape and rescues the osteogenic defects. Collectively, our results demonstrate that the metabolism-chromatin-osteogenesis axis is perturbed upon exposure to high oxygen levels and identifies CiC as a novel, oxygen-sensitive regulator of the MSC function.


Subject(s)
Histones , Osteogenesis , Mice , Animals , Osteogenesis/physiology , Acetyl Coenzyme A/metabolism , Histones/metabolism , Cell Differentiation/physiology , Mitochondria/metabolism , Hypoxia/metabolism , Oxygen/metabolism , Chromatin/metabolism , Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL