Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
FASEB J ; 36(5): e22246, 2022 05.
Article in English | MEDLINE | ID: mdl-35405035

ABSTRACT

Sickling is the central pathogenic process of sickle cell disease (SCD), one of the most prevalent inherited hemolytic disorders. Having no easy access to antioxidants in the cytosol, elevated levels of reactive oxygen species (ROS) residing at the plasma membrane in sickle red blood cells (sRBCs) easily oxidize membrane proteins and thus contribute to sickling. Although the ubiquitin-proteasome system (UPS) is essential to rapidly clear ROS-damaged membrane proteins and maintain cellular homeostasis, the function and regulatory mechanism of the UPS for their clearance in sRBCs remains unidentified. Elevated levels of polyubiquitinated membrane-associated proteins in human sRBCs are reported here. High throughput and untargeted proteomic analyses of membrane proteins immunoprecipitated by ubiquitin antibodies detected elevated levels of ubiquitination of a series of proteins including cytoskeletal proteins, transporters, ROS-related proteins, and UPS machinery components in sRBCs. Polyubiquitination of membrane-associated catalase was increased in sRBCs, associated with decreased catalase activity and elevated ROS. Surprisingly, shuttling of p97 (ATP-dependent valosin-containing chaperone protein), a key component of the UPS to shuttle polyubiquitinated proteins from the membrane to cytosol for proteasomal degradation, was significantly impaired, resulting in significant accumulation of p97 along with polyubiquitinated proteins in the membrane of human sRBCs. Functionally, inhibition of p97 directly promoted accumulation of polyubiquitinated membrane-associated proteins, excessive ROS levels, and sickling in response to hypoxia. Overall, we revealed that p97 dysfunction underlies impaired UPS and contributes to oxidative stress in sRBCs.


Subject(s)
Anemia, Sickle Cell , Oxidative Stress , Valosin Containing Protein , Adenosine Triphosphatases/metabolism , Catalase/metabolism , Cell Cycle Proteins/metabolism , Humans , Proteasome Endopeptidase Complex/metabolism , Proteomics , Quality Control , Reactive Oxygen Species , Ubiquitin/metabolism , Valosin Containing Protein/metabolism
2.
Cell Metab ; 34(2): 299-316.e6, 2022 02 01.
Article in English | MEDLINE | ID: mdl-35108516

ABSTRACT

Due to lack of nuclei and de novo protein synthesis, post-translational modification (PTM) is imperative for erythrocytes to regulate oxygen (O2) delivery and combat tissue hypoxia. Here, we report that erythrocyte transglutminase-2 (eTG2)-mediated PTM is essential to trigger O2 delivery by promoting bisphosphoglycerate mutase proteostasis and the Rapoport-Luebering glycolytic shunt for adaptation to hypoxia, in healthy humans ascending to high altitude and in two distinct murine models of hypoxia. In a pathological hypoxia model with chronic kidney disease (CKD), eTG2 is critical to combat renal hypoxia-induced reduction of Slc22a5 transcription and OCNT2 protein levels via HIF-1α-PPARα signaling to maintain carnitine homeostasis. Carnitine supplementation is an effective and safe therapeutic approach to counteract hypertension and progression of CKD by enhancing erythrocyte O2 delivery. Altogether, we reveal eTG2 as an erythrocyte protein stabilizer orchestrating O2 delivery and tissue adaptive metabolic reprogramming and identify carnitine-based therapy to mitigate hypoxia and CKD progression.


Subject(s)
Carnitine , Renal Insufficiency, Chronic , Animals , Carnitine/metabolism , Erythrocytes/metabolism , Erythrocytes/pathology , Homeostasis , Humans , Hypoxia/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , Oxygen/metabolism , Renal Insufficiency, Chronic/pathology , Solute Carrier Family 22 Member 5/metabolism , Transglutaminases/metabolism
3.
PLoS Biol ; 19(6): e3001239, 2021 06.
Article in English | MEDLINE | ID: mdl-34138843

ABSTRACT

Hypoxia drives aging and promotes age-related cognition and hearing functional decline. Despite the role of erythrocytes in oxygen (O2) transport, their role in the onset of aging and age-related cognitive decline and hearing loss (HL) remains undetermined. Recent studies revealed that signaling through the erythrocyte adenosine A2B receptor (ADORA2B) promotes O2 release to counteract hypoxia at high altitude. However, nothing is known about a role for erythrocyte ADORA2B in age-related functional decline. Here, we report that loss of murine erythrocyte-specific ADORA2B (eAdora2b-/-) accelerates early onset of age-related impairments in spatial learning, memory, and hearing ability. eAdora2b-/- mice display the early aging-like cellular and molecular features including the proliferation and activation of microglia and macrophages, elevation of pro-inflammatory cytokines, and attenuation of hypoxia-induced glycolytic gene expression to counteract hypoxia in the hippocampus (HIP), cortex, or cochlea. Hypoxia sufficiently accelerates early onset of cognitive and cochlear functional decline and inflammatory response in eAdora2b-/- mice. Mechanistically, erythrocyte ADORA2B-mediated activation of AMP-activated protein kinase (AMPK) and bisphosphoglycerate mutase (BPGM) promotes hypoxic and metabolic reprogramming to enhance production of 2,3-bisphosphoglycerate (2,3-BPG), an erythrocyte-specific metabolite triggering O2 delivery. Significantly, this finding led us to further discover that murine erythroblast ADORA2B and BPGM mRNA levels and erythrocyte BPGM activity are reduced during normal aging. Overall, we determined that erythrocyte ADORA2B-BPGM axis is a key component for anti-aging and anti-age-related functional decline.


Subject(s)
Auditory Pathways/physiopathology , Cognitive Dysfunction/metabolism , Erythrocytes/metabolism , Hypoxia/metabolism , Receptor, Adenosine A2B/metabolism , 2,3-Diphosphoglycerate/metabolism , Aging/pathology , Animals , Bisphosphoglycerate Mutase/genetics , Bisphosphoglycerate Mutase/metabolism , Brain/pathology , Brain/physiopathology , Cochlea/physiopathology , Cognitive Dysfunction/complications , Cognitive Dysfunction/genetics , Cognitive Dysfunction/physiopathology , Enzyme Activation , Gene Deletion , Glycolysis , Hypoxia/complications , Hypoxia/genetics , Hypoxia/physiopathology , Inflammation/complications , Inflammation/pathology , Inflammation Mediators/metabolism , Macrophages/metabolism , Mice, Inbred C57BL , Microglia/metabolism , Microglia/pathology , Receptor, Adenosine A2B/deficiency
4.
JCI Insight ; 6(3)2021 02 08.
Article in English | MEDLINE | ID: mdl-33351786

ABSTRACT

Computational models based on recent maps of the RBC proteome suggest that mature erythrocytes may harbor targets for common drugs. This prediction is relevant to RBC storage in the blood bank, in which the impact of small molecule drugs or other xenometabolites deriving from dietary, iatrogenic, or environmental exposures ("exposome") may alter erythrocyte energy and redox metabolism and, in so doing, affect red cell storage quality and posttransfusion efficacy. To test this prediction, here we provide a comprehensive characterization of the blood donor exposome, including the detection of common prescription and over-the-counter drugs in blood units donated by 250 healthy volunteers in the Recipient Epidemiology and Donor Evaluation Study III Red Blood Cell-Omics (REDS-III RBC-Omics) Study. Based on high-throughput drug screenings of 1366 FDA-approved drugs, we report that approximately 65% of the tested drugs had an impact on erythrocyte metabolism. Machine learning models built using metabolites as predictors were able to accurately predict drugs for several drug classes/targets (bisphosphonates, anticholinergics, calcium channel blockers, adrenergics, proton pump inhibitors, antimetabolites, selective serotonin reuptake inhibitors, and mTOR), suggesting that these drugs have a direct, conserved, and substantial impact on erythrocyte metabolism. As a proof of principle, here we show that the antacid ranitidine - though rarely detected in the blood donor population - has a strong effect on RBC markers of storage quality in vitro. We thus show that supplementation of blood units stored in bags with ranitidine could - through mechanisms involving sphingosine 1-phosphate-dependent modulation of erythrocyte glycolysis and/or direct binding to hemoglobin - improve erythrocyte metabolism and storage quality.


Subject(s)
Blood Donors , Erythrocytes/drug effects , Erythrocytes/metabolism , Exposome , Nonprescription Drugs/adverse effects , Nonprescription Drugs/pharmacokinetics , Prescription Drugs/adverse effects , Prescription Drugs/pharmacokinetics , Adolescent , Adult , Aged , Animals , Energy Metabolism/drug effects , Erythrocyte Transfusion , Female , Glycolysis/drug effects , Healthy Volunteers , Hemoglobins/metabolism , High-Throughput Screening Assays , Humans , In Vitro Techniques , Machine Learning , Male , Metabolomics , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Models, Biological , Oxidation-Reduction/drug effects , Phosphotransferases (Alcohol Group Acceptor)/deficiency , Phosphotransferases (Alcohol Group Acceptor)/genetics , Ranitidine/pharmacology , Young Adult
5.
JCI Insight ; 5(10)2020 05 21.
Article in English | MEDLINE | ID: mdl-32434995

ABSTRACT

Insufficient O2 supply is frequently associated with fetal growth restriction (FGR), a leading cause of perinatal mortality and morbidity. Although the erythrocyte is the most abundant and only cell type to deliver O2 in our body, its function and regulatory mechanism in FGR remain unknown. Here, we report that genetic ablation of mouse erythrocyte equilibrative nucleoside transporter 1 (eENT1) in dams, but not placentas or fetuses, results in FGR. Unbiased high-throughput metabolic profiling coupled with in vitro and in vivo flux analyses with isotopically labeled tracers led us to discover that maternal eENT1-dependent adenosine uptake is critical in activating AMPK by controlling the AMP/ATP ratio and its downstream target, bisphosphoglycerate mutase (BPGM); in turn, BPGM mediates 2,3-BPG production, which enhances O2 delivery to maintain placental oxygenation. Mechanistically and functionally, we revealed that genetic ablation of maternal eENT1 increases placental HIF-1α; preferentially reduces placental large neutral aa transporter 1 (LAT1) expression, activity, and aa supply; and induces FGR. Translationally, we revealed that elevated HIF-1α directly reduces LAT1 gene expression in cultured human trophoblasts. We demonstrate the importance and molecular insight of maternal eENT1 in fetal growth and open up potentially new diagnostic and therapeutic possibilities for FGR.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Equilibrative Nucleoside Transporter 1/metabolism , Erythrocytes/metabolism , Fetal Development , Fetus/metabolism , Hypoxia/metabolism , Placenta/metabolism , Animals , Enzyme Activation , Female , Mice , Mice, Knockout , Pregnancy
6.
Circ Res ; 127(3): 360-375, 2020 07 17.
Article in English | MEDLINE | ID: mdl-32284030

ABSTRACT

RATIONALE: Hypoxia promotes renal damage and progression of chronic kidney disease (CKD). The erythrocyte is the only cell type for oxygen (O2) delivery. Sphingosine 1-phosphate (S1P)-a highly enriched biolipid in erythrocytes-is recently reported to be induced under high altitude in normal humans to enhance O2 delivery. However, nothing is known about erythrocyte S1P in CKD. OBJECTIVE: To investigate the function and metabolic basis of erythrocyte S1P in CKD with a goal to explore potential therapeutics. METHODS AND RESULTS: Using erythrocyte-specific SphK1 (sphingosine kinase 1; the only enzyme to produce S1P in erythrocytes) knockout mice (eSphK1-/-) in an experimental model of hypertensive CKD with Ang II (angiotensin II) infusion, we found severe renal hypoxia, hypertension, proteinuria, and fibrosis in Ang II-infused eSphk1-/- mice compared with controls. Untargeted metabolomics profiling and in vivo U-13C6 isotopically labeled glucose flux analysis revealed that SphK1 is required for channeling glucose metabolism toward glycolysis versus pentose phosphate pathway, resulting in enhanced erythroid-specific Rapoport-Luebering shunt in Ang II-infused mice. Mechanistically, increased erythrocyte S1P functioning intracellularly activates AMPK (AMP-activated protein kinase) 1α and BPGM (bisphosphoglycerate mutase) by reducing ceramide/S1P ratio and inhibiting PP2A (protein phosphatase 2A), leading to increased 2,3-bisphosphoglycerate (an erythrocyte-specific metabolite negatively regulating Hb [hemoglobin]-O2-binding affinity) production and thus more O2 delivery to counteract kidney hypoxia and progression to CKD. Preclinical studies revealed that an AMPK agonist or a PP2A inhibitor rescued the severe CKD phenotype in Ang II-infused eSphK1-/- mice and prevented development of CKD in the control mice by inducing 2,3-bisphosphoglycerate production and thus enhancing renal oxygenation. Translational research validated mouse findings in erythrocytes of hypertensive CKD patients and cultured human erythrocytes. CONCLUSIONS: Our study elucidates the beneficial role of eSphk1-S1P in hypertensive CKD by channeling glucose metabolism toward Rapoport-Luebering shunt and inducing 2,3-bisphosphoglycerate production and O2 delivery via a PP2A-AMPK1α signaling pathway. These findings reveal the metabolic and molecular basis of erythrocyte S1P in CKD and new therapeutic avenues.


Subject(s)
Cellular Reprogramming , Energy Metabolism , Erythrocytes/metabolism , Kidney/metabolism , Renal Insufficiency, Chronic/blood , Adult , Animals , Case-Control Studies , Cell Hypoxia , Disease Models, Animal , Erythrocytes/enzymology , Female , Fibrosis , Humans , Hypertension/complications , Kidney/pathology , Male , Metabolome , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Renal Insufficiency, Chronic/enzymology , Renal Insufficiency, Chronic/etiology , Renal Insufficiency, Chronic/pathology
7.
FASEB J ; 34(3): 4041-4054, 2020 03.
Article in English | MEDLINE | ID: mdl-31930569

ABSTRACT

Recent evidence indicates that elevated placental adenosine signaling contributes to preeclampsia (PE). However, the molecular basis for the chronically enhanced placental adenosine signaling in PE remains unclear. Here, we report that hypoxia-inducible factor-1α (HIF-1α) is crucial for the enhancement of placental adenosine signaling. Utilizing a pharmacologic approach to reduce placental adenosine levels, we found that enhanced adenosine underlies increased placental HIF-1α in an angiotensin receptor type 1 receptor agonistic autoantibody (AT1 -AA)-induced mouse model of PE. Knockdown of placental HIF-1α in vivo suppressed the accumulation of adenosine and increased ecto-5'-nucleotidase (CD73) and adenosine A2B receptor (ADORA2B) in the placentas of PE mouse models induced by AT1 -AA or LIGHT, a TNF superfamily cytokine (TNFSF14). Human in vitro studies using placental villous explants demonstrated that increased HIF-1α resulting from ADORA2B activation facilitates the induction of CD73, ADORA2B, and FLT-1 expression. Overall, we demonstrated that (a) elevated placental HIF-1α by AT1 -AA or LIGHT upregulates CD73 and ADORA2B expression and (b) enhanced adenosine signaling through upregulated ADORA2B induces placental HIF-1α expression, which creates a positive feedback loop that promotes FLT-1 expression leading to disease development. Our results suggest that adenosine-based therapy targeting the malicious cycle of placental adenosine signaling may elicit therapeutic effects on PE.


Subject(s)
Adenosine/metabolism , Autoantibodies/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Placenta/metabolism , Pre-Eclampsia/metabolism , RNA, Small Interfering/metabolism , Animals , Autoantibodies/genetics , Blood Pressure/genetics , Blood Pressure/physiology , Female , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Immunoblotting , Immunohistochemistry , Mice , Mice, Inbred C57BL , Pre-Eclampsia/genetics , Pregnancy , RNA, Small Interfering/genetics , Reverse Transcriptase Polymerase Chain Reaction , Vascular Endothelial Growth Factor Receptor-1/genetics , Vascular Endothelial Growth Factor Receptor-1/metabolism
8.
Nat Metab ; 1(4): 431-444, 2019 04.
Article in English | MEDLINE | ID: mdl-31984309

ABSTRACT

Tumor hypoxia is a major contributor to resistance to anti-cancer therapies. Given that the results of hypoxia-targeted therapy trials have been disappointing, a more personalized approach may be needed. Here we characterize multi-OMIC molecular features associated with tumor hypoxia and identify molecular alterations that correlate with both drug-resistant and drug-sensitive responses to anti-cancer drugs. Based on a well-established hypoxia gene expression signature, we classify about 10,000 tumor samples into hypoxia score-high and score-low groups across different cancer types from The Cancer Genome Atlas and demonstrate their prognostic associations. We then identify various types of molecular features associated with hypoxia status that correlate with drug resistance but, in some cases, also with drug sensitivity, contrasting the conventional view that hypoxia confers drug resistance. We further show that 110 out of 121 (90.9%) clinically actionable genes can be affected by hypoxia status and experimentally validate the predicted effects of hypoxia on the response to several drugs in cultured cells. Our study provides a comprehensive molecular-level understanding of tumor hypoxia and may have practical implications for clinical cancer therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Molecular Targeted Therapy , Neoplasms/drug therapy , Tumor Hypoxia/genetics , Female , Humans , Male , Neoplasms/genetics , Neoplasms/pathology , Transcriptome
9.
Blood Adv ; 2(15): 1957-1968, 2018 08 14.
Article in English | MEDLINE | ID: mdl-30097462

ABSTRACT

Although excessive plasma adenosine is detrimental in sickle cell disease (SCD), the molecular mechanism underlying elevated circulating adenosine remains unclear. Here we report that the activity of soluble CD73, an ectonucleotidase producing extracellular adenosine, was significantly elevated in a murine model of SCD and correlated with increased plasma adenosine. Mouse genetic studies demonstrated that CD73 activity contributes to excessive induction of plasma adenosine and thereby promotes sickling, hemolysis, multiorgan damage, and disease progression. Mechanistically, we showed that erythrocyte adenosine 5'-monophosphate-activated protein kinase (AMPK) was activated both in SCD patients and in the murine model of SCD. AMPK functions downstream of adenosine receptor ADORA2B signaling and contributes to sickling by regulating the production of erythrocyte 2,3-bisphosphoglycerate (2,3-BPG), a negative allosteric regulator of hemoglobin-O2 binding affinity. Preclinically, we reported that treatment of α,ß-methylene adenosine 5'-diphosphate, a potent CD73 specific inhibitor, significantly decreased sickling, hemolysis, multiorgan damage, and disease progression in the murine model of SCD. Taken together, both human and mouse studies reveal a novel molecular mechanism contributing to the pathophysiology of SCD and identify potential therapeutic strategies to treat SCD.


Subject(s)
5'-Nucleotidase , Adenosine Triphosphate/analogs & derivatives , Anemia, Sickle Cell , Erythrocytes/enzymology , 2,3-Diphosphoglycerate/metabolism , 5'-Nucleotidase/antagonists & inhibitors , 5'-Nucleotidase/genetics , 5'-Nucleotidase/metabolism , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Adenosine/metabolism , Adenosine Triphosphate/pharmacology , Anemia, Sickle Cell/drug therapy , Anemia, Sickle Cell/enzymology , Anemia, Sickle Cell/genetics , Anemia, Sickle Cell/pathology , Animals , Erythrocytes/pathology , Female , GPI-Linked Proteins/antagonists & inhibitors , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Humans , Male , Mice , Mice, Knockout , Receptor, Adenosine A2B/genetics , Receptor, Adenosine A2B/metabolism
10.
Haematologica ; 103(2): 361-372, 2018 02.
Article in English | MEDLINE | ID: mdl-29079593

ABSTRACT

Hypoxanthine catabolism in vivo is potentially dangerous as it fuels production of urate and, most importantly, hydrogen peroxide. However, it is unclear whether accumulation of intracellular and supernatant hypoxanthine in stored red blood cell units is clinically relevant for transfused recipients. Leukoreduced red blood cells from glucose-6-phosphate dehydrogenase-normal or -deficient human volunteers were stored in AS-3 under normoxic, hyperoxic, or hypoxic conditions (with oxygen saturation ranging from <3% to >95%). Red blood cells from healthy human volunteers were also collected at sea level or after 1-7 days at high altitude (>5000 m). Finally, C57BL/6J mouse red blood cells were incubated in vitro with 13C1-aspartate or 13C5-adenosine under normoxic or hypoxic conditions, with or without deoxycoformycin, a purine deaminase inhibitor. Metabolomics analyses were performed on human and mouse red blood cells stored for up to 42 or 14 days, respectively, and correlated with 24 h post-transfusion red blood cell recovery. Hypoxanthine increased in stored red blood cell units as a function of oxygen levels. Stored red blood cells from human glucose-6-phosphate dehydrogenase-deficient donors had higher levels of deaminated purines. Hypoxia in vitro and in vivo decreased purine oxidation and enhanced purine salvage reactions in human and mouse red blood cells, which was partly explained by decreased adenosine monophosphate deaminase activity. In addition, hypoxanthine levels negatively correlated with post-transfusion red blood cell recovery in mice and - preliminarily albeit significantly - in humans. In conclusion, hypoxanthine is an in vitro metabolic marker of the red blood cell storage lesion that negatively correlates with post-transfusion recovery in vivo Storage-dependent hypoxanthine accumulation is ameliorated by hypoxia-induced decreases in purine deamination reaction rates.


Subject(s)
Erythrocytes/metabolism , Hypoxanthine/blood , Hypoxia , Purines/metabolism , Animals , Blood Preservation/methods , Deamination , Erythrocyte Transfusion , Humans , Mice , Mice, Inbred C57BL
11.
Sci Rep ; 7(1): 15281, 2017 11 10.
Article in English | MEDLINE | ID: mdl-29127281

ABSTRACT

Elevated sphingosine 1-phosphate (S1P) is detrimental in Sickle Cell Disease (SCD), but the mechanistic basis remains obscure. Here, we report that increased erythrocyte S1P binds to deoxygenated sickle Hb (deoxyHbS), facilitates deoxyHbS anchoring to the membrane, induces release of membrane-bound glycolytic enzymes and in turn switches glucose flux towards glycolysis relative to the pentose phosphate pathway (PPP). Suppressed PPP causes compromised glutathione homeostasis and increased oxidative stress, while enhanced glycolysis induces production of 2,3-bisphosphoglycerate (2,3-BPG) and thus increases deoxyHbS polymerization, sickling, hemolysis and disease progression. Functional studies revealed that S1P and 2,3-BPG work synergistically to decrease both HbA and HbS oxygen binding affinity. The crystal structure at 1.9 Å resolution deciphered that S1P binds to the surface of 2,3-BPG-deoxyHbA and causes additional conformation changes to the T-state Hb. Phosphate moiety of the surface bound S1P engages in a highly positive region close to α1-heme while its aliphatic chain snakes along a shallow cavity making hydrophobic interactions in the "switch region", as well as with α2-heme like a molecular "sticky tape" with the last 3-4 carbon atoms sticking out into bulk solvent. Altogether, our findings provide functional and structural bases underlying S1P-mediated pathogenic metabolic reprogramming in SCD and novel therapeutic avenues.


Subject(s)
Anemia, Sickle Cell/metabolism , Erythrocytes, Abnormal/metabolism , Hemoglobin A/metabolism , Hemoglobin, Sickle/metabolism , Lysophospholipids/metabolism , Sphingosine/analogs & derivatives , 2,3-Diphosphoglycerate/chemistry , 2,3-Diphosphoglycerate/metabolism , Anemia, Sickle Cell/pathology , Animals , Erythrocytes, Abnormal/pathology , Female , Hemoglobin A/chemistry , Hemoglobin, Sickle/chemistry , Hemolysis , Humans , Lysophospholipids/chemistry , Male , Mice , Mice, Transgenic , Oxidative Stress , Pentose Phosphate Pathway , Sphingosine/chemistry , Sphingosine/metabolism
12.
J Appl Physiol (1985) ; 123(4): 951-956, 2017 Oct 01.
Article in English | MEDLINE | ID: mdl-28572494

ABSTRACT

Erythrocytes are vital to human adaptation under hypoxic conditions because of their abundance in number and irreplaceable function of delivering oxygen (O2). However, although multiple large-scale altitude studies investigating the overall coordination of the human body for hypoxia adaptation have been conducted, detailed research with a focus on erythrocytes was missing due to lack of proper techniques. The recently maturing metabolomics profiling technology appears to be the answer to this limitation. Metabolomics profiling provides unbiased high-throughput screening data that reveal the overall metabolic status of erythrocytes. Recent studies have exploited this new technology and provided novel insight into erythrocyte physiology and pathology. In particular, a series of studies focusing on erythrocyte purinergic signaling have reported that adenosine signaling, coupled with 5' AMP-activated protein kinase (AMPK) and the production of erythrocyte-enriched bioactive signaling lipid sphingosine 1-phosphate, regulate erythrocyte glucose metabolism for more O2 delivery. Moreover, an adenosine-dependent "erythrocyte hypoxic memory" was discovered that provides an explanation for fast acclimation upon re-ascent. These findings not only shed new light on our understanding of erythrocyte function and hypoxia adaptation, but also offer a myriad of novel therapeutic possibilities to counteract various hypoxic conditions.


Subject(s)
Adaptation, Physiological/physiology , Adenosine/metabolism , Erythrocytes/metabolism , Hypoxia/metabolism , Signal Transduction/physiology , AMP-Activated Protein Kinases/metabolism , Acclimatization , Animals , Humans , Oxygen/metabolism
13.
Hypertension ; 70(1): 209-218, 2017 07.
Article in English | MEDLINE | ID: mdl-28507174

ABSTRACT

Preeclampsia is a prevalent pregnancy hypertensive disease with both maternal and fetal morbidity and mortality. Emerging evidence indicates that global placental DNA hypomethylation is observed in patients with preeclampsia and is linked to altered gene expression and disease development. However, the molecular basis underlying placental epigenetic changes in preeclampsia remains unclear. Using 2 independent experimental models of preeclampsia, adenosine deaminase-deficient mice and a pathogenic autoantibody-induced mouse model of preeclampsia, we demonstrate that elevated placental adenosine not only induces hallmark features of preeclampsia but also causes placental DNA hypomethylation. The use of genetic approaches to express an adenosine deaminase minigene specifically in placentas, or adenosine deaminase enzyme replacement therapy, restored placental adenosine to normal levels, attenuated preeclampsia features, and abolished placental DNA hypomethylation in adenosine deaminase-deficient mice. Genetic deletion of CD73 (an ectonucleotidase that converts AMP to adenosine) prevented the elevation of placental adenosine in the autoantibody-induced preeclampsia mouse model and ameliorated preeclampsia features and placental DNA hypomethylation. Immunohistochemical studies revealed that elevated placental adenosine-mediated DNA hypomethylation predominantly occurs in spongiotrophoblasts and labyrinthine trophoblasts and that this effect is independent of A2B adenosine receptor activation in both preeclampsia models. Extending our mouse findings to humans, we used cultured human trophoblasts to demonstrate that adenosine functions intracellularly and induces DNA hypomethylation without A2B adenosine receptor activation. Altogether, both mouse and human studies reveal novel mechanisms underlying placental DNA hypomethylation and potential therapeutic approaches for preeclampsia.


Subject(s)
Adenosine Deaminase , Placenta , Pre-Eclampsia , Adenosine Deaminase/metabolism , Adenosine Deaminase/pharmacology , Animals , Autoantibodies/metabolism , Cells, Cultured , DNA Methylation , Disease Models, Animal , Enzyme Replacement Therapy/methods , Epigenomics , Female , Humans , Mice , Placenta/drug effects , Placenta/metabolism , Pre-Eclampsia/drug therapy , Pre-Eclampsia/metabolism , Pregnancy , Treatment Outcome , Trophoblasts/drug effects , Trophoblasts/metabolism
14.
Nat Commun ; 8: 14108, 2017 02 07.
Article in English | MEDLINE | ID: mdl-28169986

ABSTRACT

Faster acclimatization to high altitude upon re-ascent is seen in humans; however, the molecular basis for this enhanced adaptive response is unknown. We report that in healthy lowlanders, plasma adenosine levels are rapidly induced by initial ascent to high altitude and achieved even higher levels upon re-ascent, a feature that is positively associated with quicker acclimatization. Erythrocyte equilibrative nucleoside transporter 1 (eENT1) levels are reduced in humans at high altitude and in mice under hypoxia. eENT1 deletion allows rapid accumulation of plasma adenosine to counteract hypoxic tissue damage in mice. Adenosine signalling via erythrocyte ADORA2B induces PKA phosphorylation, ubiquitination and proteasomal degradation of eENT1. Reduced eENT1 resulting from initial hypoxia is maintained upon re-ascent in humans or re-exposure to hypoxia in mice and accounts for erythrocyte hypoxic memory and faster acclimatization. Our findings suggest that targeting identified purinergic-signalling network would enhance the hypoxia adenosine response to counteract hypoxia-induced maladaptation.


Subject(s)
Acclimatization/physiology , Adenosine/metabolism , Equilibrative Nucleoside Transporter 1/metabolism , Erythrocytes/physiology , Hypoxia/physiopathology , Receptor, Adenosine A2B/metabolism , 5'-Nucleotidase/blood , 5'-Nucleotidase/metabolism , Adenosine/blood , Adult , Altitude , Altitude Sickness/blood , Altitude Sickness/physiopathology , Animals , Cyclic AMP-Dependent Protein Kinases/metabolism , Equilibrative Nucleoside Transporter 1/blood , Equilibrative Nucleoside Transporter 1/genetics , Female , GPI-Linked Proteins/blood , GPI-Linked Proteins/metabolism , Healthy Volunteers , Humans , Hypoxia/blood , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxygen/metabolism , Phosphorylation , Receptor, Adenosine A2B/genetics , Signal Transduction/physiology , Ubiquitination , Young Adult
15.
J Proteome Res ; 15(10): 3883-3895, 2016 10 07.
Article in English | MEDLINE | ID: mdl-27646145

ABSTRACT

Red blood cells (RBCs) are key players in systemic oxygen transport. RBCs respond to in vitro hypoxia through the so-called oxygen-dependent metabolic regulation, which involves the competitive binding of deoxyhemoglobin and glycolytic enzymes to the N-terminal cytosolic domain of band 3. This mechanism promotes the accumulation of 2,3-DPG, stabilizing the deoxygenated state of hemoglobin, and cytosol acidification, triggering oxygen off-loading through the Bohr effect. Despite in vitro studies, in vivo adaptations to hypoxia have not yet been completely elucidated. Within the framework of the AltitudeOmics study, erythrocytes were collected from 21 healthy volunteers at sea level, after exposure to high altitude (5260 m) for 1, 7, and 16 days, and following reascent after 7 days at 1525 m. UHPLC-MS metabolomics results were correlated to physiological and athletic performance parameters. Immediate metabolic adaptations were noted as early as a few hours from ascending to >5000 m, and maintained for 16 days at high altitude. Consistent with the mechanisms elucidated in vitro, hypoxia promoted glycolysis and deregulated the pentose phosphate pathway, as well purine catabolism, glutathione homeostasis, arginine/nitric oxide, and sulfur/H2S metabolism. Metabolic adaptations were preserved 1 week after descent, consistently with improved physical performances in comparison to the first ascendance, suggesting a mechanism of metabolic memory.


Subject(s)
Adaptation, Physiological , Altitude Sickness/metabolism , Erythrocytes/metabolism , Acclimatization/physiology , Adult , Altitude , Altitude Sickness/physiopathology , Arginine/metabolism , Glutathione/metabolism , Glycolysis , Healthy Volunteers , Humans , Pentose Phosphate Pathway , Purines/metabolism , Sulfur/metabolism , Time Factors , Young Adult
16.
Circulation ; 134(5): 405-21, 2016 Aug 02.
Article in English | MEDLINE | ID: mdl-27482003

ABSTRACT

BACKGROUND: High altitude is a challenging condition caused by insufficient oxygen supply. Inability to adjust to hypoxia may lead to pulmonary edema, stroke, cardiovascular dysfunction, and even death. Thus, understanding the molecular basis of adaptation to high altitude may reveal novel therapeutics to counteract the detrimental consequences of hypoxia. METHODS: Using high-throughput, unbiased metabolomic profiling, we report that the metabolic pathway responsible for production of erythrocyte 2,3-bisphosphoglycerate (2,3-BPG), a negative allosteric regulator of hemoglobin-O2 binding affinity, was significantly induced in 21 healthy humans within 2 hours of arrival at 5260 m and further increased after 16 days at 5260 m. RESULTS: This finding led us to discover that plasma adenosine concentrations and soluble CD73 activity rapidly increased at high altitude and were associated with elevated erythrocyte 2,3-BPG levels and O2 releasing capacity. Mouse genetic studies demonstrated that elevated CD73 contributed to hypoxia-induced adenosine accumulation and that elevated adenosine-mediated erythrocyte A2B adenosine receptor activation was beneficial by inducing 2,3-BPG production and triggering O2 release to prevent multiple tissue hypoxia, inflammation, and pulmonary vascular leakage. Mechanistically, we demonstrated that erythrocyte AMP-activated protein kinase was activated in humans at high altitude and that AMP-activated protein kinase is a key protein functioning downstream of the A2B adenosine receptor, phosphorylating and activating BPG mutase and thus inducing 2,3-BPG production and O2 release from erythrocytes. Significantly, preclinical studies demonstrated that activation of AMP-activated protein kinase enhanced BPG mutase activation, 2,3-BPG production, and O2 release capacity in CD73-deficient mice, in erythrocyte-specific A2B adenosine receptor knockouts, and in wild-type mice and in turn reduced tissue hypoxia and inflammation. CONCLUSIONS: Together, human and mouse studies reveal novel mechanisms of hypoxia adaptation and potential therapeutic approaches for counteracting hypoxia-induced tissue damage.


Subject(s)
AMP-Activated Protein Kinases/blood , Adaptation, Physiological/physiology , Altitude Sickness/blood , Erythrocytes/metabolism , Receptor, Adenosine A2B/blood , 2,3-Diphosphoglycerate/blood , 5'-Nucleotidase/blood , 5'-Nucleotidase/deficiency , Acute Lung Injury/physiopathology , Adenosine/blood , Adult , Altitude Sickness/enzymology , Altitude Sickness/physiopathology , Animals , Bisphosphoglycerate Mutase/blood , Enzyme Activation , GPI-Linked Proteins/blood , Humans , Metabolome , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxygen/blood , Phosphorylation , Protein Processing, Post-Translational
17.
Nat Commun ; 7: 12086, 2016 07 15.
Article in English | MEDLINE | ID: mdl-27417539

ABSTRACT

Sphingosine-1-phosphate (S1P) is a bioactive signalling lipid highly enriched in mature erythrocytes, with unknown functions pertaining to erythrocyte physiology. Here by employing nonbiased high-throughput metabolomic profiling, we show that erythrocyte S1P levels rapidly increase in 21 healthy lowland volunteers at 5,260 m altitude on day 1 and continue increasing to 16 days with concurrently elevated erythrocyte sphingonisne kinase 1 (Sphk1) activity and haemoglobin (Hb) oxygen (O2) release capacity. Mouse genetic studies show that elevated erythrocyte Sphk1-induced S1P protects against tissue hypoxia by inducing O2 release. Mechanistically, we show that intracellular S1P promotes deoxygenated Hb anchoring to the membrane, enhances the release of membrane-bound glycolytic enzymes to the cytosol, induces glycolysis and thus the production of 2,3-bisphosphoglycerate (2,3-BPG), an erythrocyte-specific glycolytic intermediate, which facilitates O2 release. Altogether, we reveal S1P as an intracellular hypoxia-responsive biolipid promoting erythrocyte glycolysis, O2 delivery and thus new therapeutic opportunities to counteract tissue hypoxia.


Subject(s)
Altitude Sickness/metabolism , Erythrocytes/metabolism , Lysophospholipids/blood , Oxygen/blood , Sphingosine/analogs & derivatives , 2,3-Diphosphoglycerate/metabolism , Adaptation, Physiological , Adult , Animals , Female , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/metabolism , Glycolysis , Humans , Hypoxia/metabolism , Lysophospholipids/metabolism , Male , Mice, Inbred C57BL , Mice, Mutant Strains , Oxygen/metabolism , Phosphotransferases (Alcohol Group Acceptor)/blood , Phosphotransferases (Alcohol Group Acceptor)/genetics , Sphingosine/blood , Sphingosine/metabolism
18.
Sci Rep ; 6: 29637, 2016 07 20.
Article in English | MEDLINE | ID: mdl-27436223

ABSTRACT

Although Lands' cycle was discovered in 1958, its function and cellular regulation in membrane homeostasis under physiological and pathological conditions remain largely unknown. Nonbiased high throughput metabolomic profiling revealed that Lands' cycle was impaired leading to significantly elevated erythrocyte membrane lysophosphatidylcholine (LysoPC) content and circulating and erythrocyte arachidonic acid (AA) in mice with sickle cell disease (SCD), a prevalent hemolytic genetic disorder. Correcting imbalanced Lands' cycle by knockdown of phospholipase 2 (cPLA2) or overexpression of lysophosphatidycholine acyltransferase 1 (LPCAT1), two key enzymes of Lands' cycle in hematopoietic stem cells, reduced elevated erythrocyte membrane LysoPC content and circulating AA levels and attenuated sickling, inflammation and tissue damage in SCD chimeras. Human translational studies validated SCD mouse findings and further demonstrated that imbalanced Lands' cycle induced LysoPC production directly promotes sickling in cultured mouse and human SCD erythrocytes. Mechanistically, we revealed that hypoxia-mediated ERK activation underlies imbalanced Lands' cycle by preferentially inducing the activity of PLA2 but not LPCAT in human and mouse SCD erythrocytes. Overall, our studies have identified a pathological role of imbalanced Lands' cycle in SCD erythrocytes, novel molecular basis regulating Lands' cycle and therapeutic opportunities for the disease.


Subject(s)
Anemia, Sickle Cell/metabolism , Arachidonic Acid/blood , Erythrocytes/metabolism , Lysophosphatidylcholines/metabolism , Metabolomics/methods , 1-Acylglycerophosphocholine O-Acyltransferase/genetics , Anemia, Sickle Cell/blood , Anemia, Sickle Cell/genetics , Animals , Cell Hypoxia , Cells, Cultured , Disease Models, Animal , Female , Gene Knockdown Techniques , Group IV Phospholipases A2/genetics , Humans , Male , Mice
19.
J Biol Chem ; 291(7): 3455-67, 2016 Feb 12.
Article in English | MEDLINE | ID: mdl-26668315

ABSTRACT

The MAPK-interacting kinases 1 and 2 (MNK1 and MNK2) are activated by extracellular signal-regulated kinases 1 and 2 (ERK1/2) or p38 in response to cellular stress and extracellular stimuli that include growth factors, cytokines, and hormones. Modulation of MNK activity affects translation of mRNAs involved in the cell cycle, cancer progression, and cell survival. However, the mechanism by which MNK selectively affects translation of these mRNAs is not understood. MNK binds eukaryotic translation initiation factor 4G (eIF4G) and phosphorylates the cap-binding protein eIF4E. Using a cell-free translation system from rabbit reticulocytes programmed with mRNAs containing different 5'-ends, we show that an MNK inhibitor, CGP57380, affects translation of only those mRNAs that contain both a cap and a hairpin in the 5'-UTR. Similarly, a C-terminal fragment of human eIF4G-1, eIF4G(1357-1600), which prevents binding of MNK to intact eIF4G, reduces eIF4E phosphorylation and inhibits translation of only capped and hairpin-containing mRNAs. Analysis of proteins bound to m(7)GTP-Sepharose reveals that both CGP and eIF4G(1357-1600) decrease binding of eIF4E to eIF4G. These data suggest that MNK stimulates translation only of mRNAs containing both a cap and 5'-terminal RNA duplex via eIF4E phosphorylation, thereby enhancing the coupled cap-binding and RNA-unwinding activities of eIF4F.


Subject(s)
Eukaryotic Initiation Factor-4E/metabolism , Eukaryotic Initiation Factor-4G/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Protein Biosynthesis/drug effects , Protein Serine-Threonine Kinases/metabolism , RNA Caps/metabolism , RNA, Messenger/metabolism , Amino Acid Substitution , Animals , Cell-Free System/drug effects , Cell-Free System/enzymology , Cell-Free System/metabolism , Eukaryotic Initiation Factor-4E/chemistry , Eukaryotic Initiation Factor-4E/genetics , Eukaryotic Initiation Factor-4G/chemistry , Eukaryotic Initiation Factor-4G/genetics , Humans , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/genetics , Inverted Repeat Sequences , Mutant Proteins/antagonists & inhibitors , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Phosphorylation/drug effects , Protein Interaction Domains and Motifs , Protein Kinase Inhibitors/pharmacology , Protein Processing, Post-Translational/drug effects , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/genetics , RNA/chemistry , RNA/metabolism , RNA Caps/chemistry , RNA Folding/drug effects , RNA, Messenger/chemistry , Rabbits , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Reticulocytes/drug effects , Reticulocytes/enzymology , Reticulocytes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL