Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
PLoS Biol ; 19(6): e3001239, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34138843

RESUMEN

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.


Asunto(s)
Vías Auditivas/fisiopatología , Disfunción Cognitiva/metabolismo , Eritrocitos/metabolismo , Hipoxia/metabolismo , Receptor de Adenosina A2B/metabolismo , 2,3-Difosfoglicerato/metabolismo , Envejecimiento/patología , Animales , Bisfosfoglicerato Mutasa/genética , Bisfosfoglicerato Mutasa/metabolismo , Encéfalo/patología , Encéfalo/fisiopatología , Cóclea/fisiopatología , Disfunción Cognitiva/complicaciones , Disfunción Cognitiva/genética , Disfunción Cognitiva/fisiopatología , Activación Enzimática , Eliminación de Gen , Glucólisis , Hipoxia/complicaciones , Hipoxia/genética , Hipoxia/fisiopatología , Inflamación/complicaciones , Inflamación/patología , Mediadores de Inflamación/metabolismo , Macrófagos/metabolismo , Ratones Endogámicos C57BL , Microglía/metabolismo , Microglía/patología , Receptor de Adenosina A2B/deficiencia
2.
FASEB J ; 36(5): e22246, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35405035

RESUMEN

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.


Asunto(s)
Anemia de Células Falciformes , Estrés Oxidativo , Proteína que Contiene Valosina , Adenosina Trifosfatasas/metabolismo , Catalasa/metabolismo , Proteínas de Ciclo Celular/metabolismo , Humanos , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteómica , Control de Calidad , Especies Reactivas de Oxígeno , Ubiquitina/metabolismo , Proteína que Contiene Valosina/metabolismo
3.
Circ Res ; 127(3): 360-375, 2020 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-32284030

RESUMEN

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.


Asunto(s)
Reprogramación Celular , Metabolismo Energético , Eritrocitos/metabolismo , Riñón/metabolismo , Insuficiencia Renal Crónica/sangre , Adulto , Animales , Estudios de Casos y Controles , Hipoxia de la Célula , Modelos Animales de Enfermedad , Eritrocitos/enzimología , Femenino , Fibrosis , Humanos , Hipertensión/complicaciones , Riñón/patología , Masculino , Metaboloma , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Insuficiencia Renal Crónica/enzimología , Insuficiencia Renal Crónica/etiología , Insuficiencia Renal Crónica/patología
4.
FASEB J ; 34(3): 4041-4054, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31930569

RESUMEN

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.


Asunto(s)
Adenosina/metabolismo , Autoanticuerpos/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Placenta/metabolismo , Preeclampsia/metabolismo , ARN Interferente Pequeño/metabolismo , Animales , Autoanticuerpos/genética , Presión Sanguínea/genética , Presión Sanguínea/fisiología , Femenino , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Immunoblotting , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Preeclampsia/genética , Embarazo , ARN Interferente Pequeño/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Receptor 1 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo
5.
Haematologica ; 103(2): 361-372, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29079593

RESUMEN

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.


Asunto(s)
Eritrocitos/metabolismo , Hipoxantina/sangre , Hipoxia , Purinas/metabolismo , Animales , Conservación de la Sangre/métodos , Desaminación , Transfusión de Eritrocitos , Humanos , Ratones , Ratones Endogámicos C57BL
6.
J Biol Chem ; 291(7): 3455-67, 2016 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-26668315

RESUMEN

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.


Asunto(s)
Factor 4E Eucariótico de Iniciación/metabolismo , Factor 4G Eucariótico de Iniciación/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Biosíntesis de Proteínas/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/metabolismo , Caperuzas de ARN/metabolismo , ARN Mensajero/metabolismo , Sustitución de Aminoácidos , Animales , Sistema Libre de Células/efectos de los fármacos , Sistema Libre de Células/enzimología , Sistema Libre de Células/metabolismo , Factor 4E Eucariótico de Iniciación/química , Factor 4E Eucariótico de Iniciación/genética , Factor 4G Eucariótico de Iniciación/química , Factor 4G Eucariótico de Iniciación/genética , Humanos , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Péptidos y Proteínas de Señalización Intracelular/química , Péptidos y Proteínas de Señalización Intracelular/genética , Secuencias Invertidas Repetidas , Proteínas Mutantes/antagonistas & inhibidores , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Fragmentos de Péptidos/antagonistas & inhibidores , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Fosforilación/efectos de los fármacos , Dominios y Motivos de Interacción de Proteínas , Inhibidores de Proteínas Quinasas/farmacología , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/química , Proteínas Serina-Treonina Quinasas/genética , ARN/química , ARN/metabolismo , Caperuzas de ARN/química , Pliegue del ARN/efectos de los fármacos , ARN Mensajero/química , Conejos , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Reticulocitos/efectos de los fármacos , Reticulocitos/enzimología , Reticulocitos/metabolismo
7.
Circulation ; 134(5): 405-21, 2016 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-27482003

RESUMEN

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.


Asunto(s)
Proteínas Quinasas Activadas por AMP/sangre , Adaptación Fisiológica/fisiología , Mal de Altura/sangre , Eritrocitos/metabolismo , Receptor de Adenosina A2B/sangre , 2,3-Difosfoglicerato/sangre , 5'-Nucleotidasa/sangre , 5'-Nucleotidasa/deficiencia , Lesión Pulmonar Aguda/fisiopatología , Adenosina/sangre , Adulto , Mal de Altura/enzimología , Mal de Altura/fisiopatología , Animales , Bisfosfoglicerato Mutasa/sangre , Activación Enzimática , Proteínas Ligadas a GPI/sangre , Humanos , Metaboloma , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Oxígeno/sangre , Fosforilación , Procesamiento Proteico-Postraduccional
8.
Blood ; 125(10): 1643-52, 2015 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-25587035

RESUMEN

Erythrocyte possesses high sphingosine kinase 1 (SphK1) activity and is the major cell type supplying plasma sphingosine-1-phosphate, a signaling lipid regulating multiple physiological and pathological functions. Recent studies revealed that erythrocyte SphK1 activity is upregulated in sickle cell disease (SCD) and contributes to sickling and disease progression. However, how erythrocyte SphK1 activity is regulated remains unknown. Here we report that adenosine induces SphK1 activity in human and mouse sickle and normal erythrocytes in vitro. Next, using 4 adenosine receptor-deficient mice and pharmacological approaches, we determined that the A2B adenosine receptor (ADORA2B) is essential for adenosine-induced SphK1 activity in human and mouse normal and sickle erythrocytes in vitro. Subsequently, we provide in vivo genetic evidence that adenosine deaminase (ADA) deficiency leads to excess plasma adenosine and elevated erythrocyte SphK1 activity. Lowering adenosine by ADA enzyme therapy or genetic deletion of ADORA2B significantly reduced excess adenosine-induced erythrocyte SphK1 activity in ADA-deficient mice. Finally, we revealed that protein kinase A-mediated extracellular signal-regulated kinase 1/2 activation functioning downstream of ADORA2B underlies adenosine-induced erythrocyte SphK1 activity. Overall, our findings reveal a novel signaling network regulating erythrocyte SphK1 and highlight innovative mechanisms regulating SphK1 activity in normal and SCD.


Asunto(s)
Adenosina/sangre , Anemia de Células Falciformes/sangre , Anemia de Células Falciformes/enzimología , Eritrocitos Anormales/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/sangre , Receptor de Adenosina A2B/sangre , Adenosina Desaminasa/sangre , Adenosina Desaminasa/deficiencia , Adenosina Desaminasa/genética , Adenosina-5'-(N-etilcarboxamida)/farmacología , Agammaglobulinemia/sangre , Agammaglobulinemia/enzimología , Agammaglobulinemia/genética , Anemia de Células Falciformes/genética , Animales , Células Cultivadas , Proteínas Quinasas Dependientes de AMP Cíclico/sangre , Eritrocitos/efectos de los fármacos , Eritrocitos/enzimología , Eritrocitos/metabolismo , Eritrocitos Anormales/efectos de los fármacos , Eritrocitos Anormales/enzimología , Hemoglobina Falciforme/genética , Hemoglobina Falciforme/metabolismo , Humanos , Sistema de Señalización de MAP Quinasas , Ratones , Ratones Noqueados , Ratones Transgénicos , Modelos Biológicos , Receptor de Adenosina A2B/deficiencia , Receptor de Adenosina A2B/genética , Inmunodeficiencia Combinada Grave/sangre , Inmunodeficiencia Combinada Grave/enzimología , Inmunodeficiencia Combinada Grave/genética , Transducción de Señal
9.
J Proteome Res ; 15(10): 3883-3895, 2016 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-27646145

RESUMEN

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.


Asunto(s)
Adaptación Fisiológica , Mal de Altura/metabolismo , Eritrocitos/metabolismo , Aclimatación/fisiología , Adulto , Altitud , Mal de Altura/fisiopatología , Arginina/metabolismo , Glutatión/metabolismo , Glucólisis , Voluntarios Sanos , Humanos , Vía de Pentosa Fosfato , Purinas/metabolismo , Azufre/metabolismo , Factores de Tiempo , Adulto Joven
10.
Circulation ; 131(8): 730-41, 2015 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-25538227

RESUMEN

BACKGROUND: Preeclampsia is a prevalent hypertensive disorder of pregnancy and a leading cause of maternal and neonatal morbidity and mortality worldwide. This pathogenic condition is speculated to be caused by placental abnormalities that contribute to the maternal syndrome. However, the specific factors and signaling pathways that lead to impaired placentas and maternal disease development remain elusive. METHODS AND RESULTS: Using 2 independent animal models of preeclampsia (genetically engineered pregnant mice with elevated adenosine exclusively in placentas and a pathogenic autoantibody-induced preeclampsia mouse model), we demonstrated that chronically elevated placental adenosine was sufficient to induce hallmark features of preeclampsia, including hypertension, proteinuria, small fetuses, and impaired placental vasculature. Genetic and pharmacological approaches revealed that elevated placental adenosine coupled with excessive A2B adenosine receptor (ADORA2B) signaling contributed to the development of these features of preeclampsia. Mechanistically, we provided both human and mouse evidence that elevated placental CD73 is a key enzyme causing increased placental adenosine, thereby contributing to preeclampsia. CONCLUSIONS: We determined that elevated placental adenosine signaling is a previously unrecognized pathogenic factor for preeclampsia. Moreover, our findings revealed the molecular basis underlying the elevation of placental adenosine and the detrimental role of excess placental adenosine in the pathophysiology of preeclampsia, and thereby, we highlight novel therapeutic targets.


Asunto(s)
Adenosina/metabolismo , Placenta/metabolismo , Preeclampsia/etiología , Preeclampsia/fisiopatología , Transducción de Señal/fisiología , Regulación hacia Arriba/fisiología , 5'-Nucleotidasa/metabolismo , Adenosina Desaminasa/deficiencia , Adenosina Desaminasa/genética , Adenosina Desaminasa/metabolismo , Adulto , Animales , Autoanticuerpos/efectos adversos , Modelos Animales de Enfermedad , Femenino , Retardo del Crecimiento Fetal/etiología , Retardo del Crecimiento Fetal/fisiopatología , Eliminación de Gen , Humanos , Ratones Noqueados , Preeclampsia/inducido químicamente , Embarazo , Receptor de Adenosina A2B/genética , Receptor de Adenosina A2B/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo
11.
J Cell Sci ; 123(Pt 13): 2228-37, 2010 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-20530576

RESUMEN

Caenorhabditis elegans expresses five family members of the translation initiation factor eIF4E whose individual physiological roles are only partially understood. We report a specific role for IFE-2 in a conserved temperature-sensitive meiotic process. ife-2 deletion mutants have severe temperature-sensitive chromosome-segregation defects. Mutant germ cells contain the normal six bivalents at diakinesis at 20 degrees C but 12 univalents at 25 degrees C, indicating a defect in crossover formation. Analysis of chromosome pairing in ife-2 mutants at the permissive and restrictive temperatures reveals no defects. The presence of RAD-51-marked early recombination intermediates and 12 well condensed univalents indicate that IFE-2 is not essential for formation of meiotic double-strand breaks or their repair through homologous recombination but is required for crossover formation. However, RAD-51 foci in ife-2 mutants persist into inappropriately late stages of meiotic prophase at 25 degrees C, similar to mutants defective in MSH-4/HIM-14 and MSH-5, which stabilize a critical intermediate in crossover formation. In wild-type worms, mRNAs for msh-4/him-14 and msh-5 shift from free messenger ribonucleoproteins to polysomes at 25 degrees C but not in ife-2 mutants, suggesting that IFE-2 translationally upregulates synthesis of MSH-4/HIM-14 and MSH-5 at elevated temperatures to stabilize Holliday junctions. This is confirmed by an IFE-2-dependent increase in MSH-5 protein levels.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/metabolismo , Proteínas de Unión al ADN/metabolismo , Factor 4E Eucariótico de Iniciación/metabolismo , Meiosis/fisiología , Biosíntesis de Proteínas , ARN Mensajero/metabolismo , Animales , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Intercambio Genético , ADN/genética , ADN/metabolismo , ADN/efectos de la radiación , Roturas del ADN de Doble Cadena , Proteínas de Unión al ADN/genética , Factor 4E Eucariótico de Iniciación/genética , Femenino , Calor , Masculino , Mutación , Oogénesis/fisiología , Fenotipo , ARN Mensajero/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Espermatogénesis/fisiología
12.
Cell Metab ; 34(2): 299-316.e6, 2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-35108516

RESUMEN

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.


Asunto(s)
Carnitina , Insuficiencia Renal Crónica , Animales , Carnitina/metabolismo , Eritrocitos/metabolismo , Eritrocitos/patología , Homeostasis , Humanos , Hipoxia/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Ratones , Oxígeno/metabolismo , Insuficiencia Renal Crónica/patología , Miembro 5 de la Familia 22 de Transportadores de Solutos/metabolismo , Transglutaminasas/metabolismo
14.
Exp Mol Pathol ; 90(2): 215-25, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21236253

RESUMEN

Hypoxia confers resistance to chemoradiation therapy and promotes metastasis in head and neck squamous cell carcinomas (HNSCC). We investigated the effects of hypoxia in tumor phenotype using immunocompetent murine HNSCC models. Balb/c mice were injected intraorally with murine squamous cell carcinoma cells LY-2 and B4B8. Intratumoral hypoxia fraction was evaluated by the immunohistochemical detection of hypoxic probe pimonidazole and carbonic anhydrase IX (CAIX). Tumor cell apoptosis and autophagy in hypoxic areas of these tumors were examined immunohistochemically. Hypoxia-induced apoptotic and autophagic responses in vitro were examined by treating LY2 cells with CoCl(2). B4B8 tumors exhibited a non-aggressive phenotype characterized by its slow growth rate and the lack of metastatic spread. LY2 tumors demonstrated an aggressive phenotype characterized by rapid growth rate with regional and distant metastasis. Intratumoral hypoxia fraction in B4B8 tumors was significantly lower than in LY2 tumors. The hypoxic areas in B4B8 tumors exhibited increased apoptosis rate than that of LY2 tumors. In contrast, the hypoxic areas in LY2 tumors revealed autophagy. The induction of hypoxia in vitro elicited autophagy and not apoptosis in LY2 cells. The induction of autophagy coupled with blockage of apoptosis in hypoxic areas promotes tumor cell survival and confers aggressive phenotype in immunocompetent murine HNSCC models.


Asunto(s)
Autofagia , Carcinoma de Células Escamosas/inmunología , Carcinoma de Células Escamosas/patología , Neoplasias de Cabeza y Cuello/inmunología , Neoplasias de Cabeza y Cuello/patología , Inmunocompetencia , Animales , Apoptosis , Proteínas Reguladoras de la Apoptosis/metabolismo , Beclina-1 , Carcinoma de Células Escamosas/enzimología , Caspasa 3/metabolismo , Hipoxia de la Célula , Línea Celular Tumoral , Proliferación Celular , Modelos Animales de Enfermedad , Neoplasias de Cabeza y Cuello/enzimología , Inmunohistoquímica , Ratones , Neoplasias de la Boca/patología , Metástasis de la Neoplasia , Fenotipo , Regulación hacia Arriba
15.
JCI Insight ; 6(3)2021 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-33351786

RESUMEN

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.


Asunto(s)
Donantes de Sangre , Eritrocitos/efectos de los fármacos , Eritrocitos/metabolismo , Exposoma , Medicamentos sin Prescripción/efectos adversos , Medicamentos sin Prescripción/farmacocinética , Medicamentos bajo Prescripción/efectos adversos , Medicamentos bajo Prescripción/farmacocinética , Adolescente , Adulto , Anciano , Animales , Metabolismo Energético/efectos de los fármacos , Transfusión de Eritrocitos , Femenino , Glucólisis/efectos de los fármacos , Voluntarios Sanos , Hemoglobinas/metabolismo , Ensayos Analíticos de Alto Rendimiento , Humanos , Técnicas In Vitro , Aprendizaje Automático , Masculino , Metabolómica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Modelos Biológicos , Oxidación-Reducción/efectos de los fármacos , Fosfotransferasas (Aceptor de Grupo Alcohol)/deficiencia , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Ranitidina/farmacología , Adulto Joven
16.
JCI Insight ; 5(10)2020 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-32434995

RESUMEN

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.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Tranportador Equilibrativo 1 de Nucleósido/metabolismo , Eritrocitos/metabolismo , Desarrollo Fetal , Feto/metabolismo , Hipoxia/metabolismo , Placenta/metabolismo , Animales , Activación Enzimática , Femenino , Ratones , Ratones Noqueados , Embarazo
17.
Nat Metab ; 1(4): 431-444, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-31984309

RESUMEN

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.


Asunto(s)
Antineoplásicos/uso terapéutico , Terapia Molecular Dirigida , Neoplasias/tratamiento farmacológico , Hipoxia Tumoral/genética , Femenino , Humanos , Masculino , Neoplasias/genética , Neoplasias/patología , Transcriptoma
18.
Blood Adv ; 2(15): 1957-1968, 2018 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-30097462

RESUMEN

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.


Asunto(s)
5'-Nucleotidasa , Adenosina Trifosfato/análogos & derivados , Anemia de Células Falciformes , Eritrocitos/enzimología , 2,3-Difosfoglicerato/metabolismo , 5'-Nucleotidasa/antagonistas & inhibidores , 5'-Nucleotidasa/genética , 5'-Nucleotidasa/metabolismo , Proteínas Quinasas Activadas por AMP/genética , Proteínas Quinasas Activadas por AMP/metabolismo , Adenosina/metabolismo , Adenosina Trifosfato/farmacología , Anemia de Células Falciformes/tratamiento farmacológico , Anemia de Células Falciformes/enzimología , Anemia de Células Falciformes/genética , Anemia de Células Falciformes/patología , Animales , Eritrocitos/patología , Femenino , Proteínas Ligadas a GPI/antagonistas & inhibidores , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/metabolismo , Humanos , Masculino , Ratones , Ratones Noqueados , Receptor de Adenosina A2B/genética , Receptor de Adenosina A2B/metabolismo
19.
Hypertension ; 70(1): 209-218, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28507174

RESUMEN

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.


Asunto(s)
Adenosina Desaminasa , Placenta , Preeclampsia , Adenosina Desaminasa/metabolismo , Adenosina Desaminasa/farmacología , Animales , Autoanticuerpos/metabolismo , Células Cultivadas , Metilación de ADN , Modelos Animales de Enfermedad , Terapia de Reemplazo Enzimático/métodos , Epigenómica , Femenino , Humanos , Ratones , Placenta/efectos de los fármacos , Placenta/metabolismo , Preeclampsia/tratamiento farmacológico , Preeclampsia/metabolismo , Embarazo , Resultado del Tratamiento , Trofoblastos/efectos de los fármacos , Trofoblastos/metabolismo
20.
J Appl Physiol (1985) ; 123(4): 951-956, 2017 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-28572494

RESUMEN

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.


Asunto(s)
Adaptación Fisiológica/fisiología , Adenosina/metabolismo , Eritrocitos/metabolismo , Hipoxia/metabolismo , Transducción de Señal/fisiología , Proteínas Quinasas Activadas por AMP/metabolismo , Aclimatación , Animales , Humanos , Oxígeno/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA