Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Cell Stem Cell ; 31(8): 1145-1161.e15, 2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-38772377

RESUMEN

Aging generally predisposes stem cells to functional decline, impairing tissue homeostasis. Here, we report that hematopoietic stem cells (HSCs) acquire metabolic resilience that promotes cell survival. High-resolution real-time ATP analysis with glucose tracing and metabolic flux analysis revealed that old HSCs reprogram their metabolism to activate the pentose phosphate pathway (PPP), becoming more resistant to oxidative stress and less dependent on glycolytic ATP production at steady state. As a result, old HSCs can survive without glycolysis, adapting to the physiological cytokine environment in bone marrow. Mechanistically, old HSCs enhance mitochondrial complex II metabolism during stress to promote ATP production. Furthermore, increased succinate dehydrogenase assembly factor 1 (SDHAF1) in old HSCs, induced by physiological low-concentration thrombopoietin (TPO) exposure, enables rapid mitochondrial ATP production upon metabolic stress, thereby improving survival. This study provides insight into the acquisition of resilience through metabolic reprogramming in old HSCs and its molecular basis to ameliorate age-related hematopoietic abnormalities.


Asunto(s)
Adenosina Trifosfato , Células Madre Hematopoyéticas , Mitocondrias , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/citología , Animales , Mitocondrias/metabolismo , Adenosina Trifosfato/metabolismo , Ratones , Senescencia Celular , Ratones Endogámicos C57BL , Glucólisis , Envejecimiento/metabolismo , Estrés Oxidativo
2.
Elife ; 122024 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-38573813

RESUMEN

Metabolic pathways are plastic and rapidly change in response to stress or perturbation. Current metabolic profiling techniques require lysis of many cells, complicating the tracking of metabolic changes over time after stress in rare cells such as hematopoietic stem cells (HSCs). Here, we aimed to identify the key metabolic enzymes that define differences in glycolytic metabolism between steady-state and stress conditions in murine HSCs and elucidate their regulatory mechanisms. Through quantitative 13C metabolic flux analysis of glucose metabolism using high-sensitivity glucose tracing and mathematical modeling, we found that HSCs activate the glycolytic rate-limiting enzyme phosphofructokinase (PFK) during proliferation and oxidative phosphorylation (OXPHOS) inhibition. Real-time measurement of ATP levels in single HSCs demonstrated that proliferative stress or OXPHOS inhibition led to accelerated glycolysis via increased activity of PFKFB3, the enzyme regulating an allosteric PFK activator, within seconds to meet ATP requirements. Furthermore, varying stresses differentially activated PFKFB3 via PRMT1-dependent methylation during proliferative stress and via AMPK-dependent phosphorylation during OXPHOS inhibition. Overexpression of Pfkfb3 induced HSC proliferation and promoted differentiated cell production, whereas inhibition or loss of Pfkfb3 suppressed them. This study reveals the flexible and multilayered regulation of HSC glycolytic metabolism to sustain hematopoiesis under stress and provides techniques to better understand the physiological metabolism of rare hematopoietic cells.


Asunto(s)
Glucólisis , Fosfofructoquinasa-2 , Animales , Ratones , Adenosina Trifosfato/metabolismo , Anaerobiosis , Hematopoyesis , Células Madre Hematopoyéticas/metabolismo , Fosforilación Oxidativa , Fosfofructoquinasa-2/genética , Fosfofructoquinasa-2/metabolismo , Monoéster Fosfórico Hidrolasas/metabolismo
3.
Exp Hematol ; 112-113: 44-59.e6, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35907584

RESUMEN

Bone marrow (BM) innervation regulates the mobilization of hematopoietic stem and progenitor cells (HSPCs) from BM and stress hematopoiesis either by acting directly on HSPCs or by altering the niche function of mesenchymal and endothelial cells. However, the spatial distribution of BM innervation across bone regions is yet to be fully elucidated. Thus, we aimed to characterize the distribution of sympathetic and nociceptive nerves in each bone and BM region using three-dimensional quantitative microscopy. We discovered that sympathetic and nociceptive nerves were the major fibers throughout the BM. Compared with other femoral regions, central parts of the femoral BM were more densely innervated by both sympathetic and nociceptive nerves. Each region of the sternum was similarly innervated by sympathetic and nociceptive nerves. Further, the majority of sympathetic and nociceptive nerves in the BM ran parallel with arteries and arterioles, whereas the degree varied according to the bone type or BM region. In conclusion, this study provides spatial, topological, and functional information on BM innervation in a quantitative manner and illustrates that sympathetic and nociceptive nerves are two major components in BM innervation, mostly associated with arteries and arterioles.


Asunto(s)
Células de la Médula Ósea , Médula Ósea , Animales , Médula Ósea/metabolismo , Células de la Médula Ósea/metabolismo , Células Endoteliales/metabolismo , Células Madre Hematopoyéticas/metabolismo , Ratones , Nocicepción
4.
J Biol Chem ; 296: 100563, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33745970

RESUMEN

Hematopoietic stem cells (HSCs) and their progeny sustain lifetime hematopoiesis. Aging alters HSC function, number, and composition and increases risk of hematological malignancies, but how these changes occur in HSCs remains unclear. Signaling via p38 mitogen-activated kinase (p38MAPK) has been proposed as a candidate mechanism underlying induction of HSC aging. Here, using genetic models of both chronological and premature aging, we describe a multimodal role for p38α, the major p38MAPK isozyme in hematopoiesis, in HSC aging. We report that p38α regulates differentiation bias and sustains transplantation capacity of HSCs in the early phase of chronological aging. However, p38α decreased HSC transplantation capacity in the late progression phase of chronological aging. Furthermore, codeletion of p38α in mice deficient in ataxia-telangiectasia mutated, a model of premature aging, exacerbated aging-related HSC phenotypes seen in ataxia-telangiectasia mutated single-mutant mice. Overall, these studies provide new insight into multiple functions of p38MAPK, which both promotes and suppresses HSC aging context dependently.


Asunto(s)
Envejecimiento/patología , Diferenciación Celular , Senescencia Celular , Células Madre Hematopoyéticas/metabolismo , Proteína Quinasa 14 Activada por Mitógenos/fisiología , Envejecimiento/metabolismo , Animales , Proteínas de la Ataxia Telangiectasia Mutada/fisiología , Proliferación Celular , Femenino , Hematopoyesis , Células Madre Hematopoyéticas/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Especies Reactivas de Oxígeno/metabolismo
5.
Sci Rep ; 10(1): 6422, 2020 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-32286470

RESUMEN

Physiological regulation of blood flow in bone marrow is important to maintain oxygen and glucose supplies but also the physiological hypoxic state of the hematopoietic stem cell (HSC) niche. However, regulatory mechanisms underlying microcirculation in the bone marrow (BM) niche remain unclear. Here, we identify vessels functioning in control of blood flow in bone marrow and assess their contractility. To evaluate contractile potential of Alexa Fluor 633 (AF633; an arterial marker)-positive vessels, we performed immunohistochemistry for α-smooth muscle actin (α-SMA) and found it expressed around AF633+ vessels in the femoral and calvarial marrow. To validate AF633+ vessel contractility, we developed a simple system to locally administer vasoactive agents that penetrate BM through transcalvarial vessels. After exposure of the calvarial surface to FITC-dextran (70 kDa), FITC intensity in calvarial bone marrow gradually increased. When we evaluated the effect of transcalvarial administration (TCA) of norepinephrine (NE) on vascular tone of AF633+ arteries and behavior of transplanted blood cells, NE administration decreased artery diameter and transendothelial migration of transplanted cells, suggesting that adrenergic signaling regulates the HSC niche microcirculation and blood cell migration into the BM via effects on BMarteries. We conclude that TCA is a useful tool for bone marrow research.


Asunto(s)
Médula Ósea/irrigación sanguínea , Médula Ósea/diagnóstico por imagen , Microscopía Intravital , Animales , Arterias/diagnóstico por imagen , Arterias/efectos de los fármacos , Arterias/fisiología , Vías de Administración de Medicamentos , Antígenos Comunes de Leucocito/metabolismo , Linfocitos/citología , Linfocitos/efectos de los fármacos , Masculino , Ratones Endogámicos C57BL , Norepinefrina/farmacología , Cráneo/diagnóstico por imagen , Bibliotecas de Moléculas Pequeñas/administración & dosificación , Migración Transendotelial y Transepitelial/efectos de los fármacos , Vasoconstricción
6.
Cell ; 178(5): 1072-1087.e14, 2019 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-31442401

RESUMEN

Nutritional status potentially influences immune responses; however, how nutritional signals regulate cellular dynamics and functionality remains obscure. Herein, we report that temporary fasting drastically reduces the number of lymphocytes by ∼50% in Peyer's patches (PPs), the inductive site of the gut immune response. Subsequent refeeding seemingly restored the number of lymphocytes, but whose cellular composition was conspicuously altered. A large portion of germinal center and IgA+ B cells were lost via apoptosis during fasting. Meanwhile, naive B cells migrated from PPs to the bone marrow during fasting and then back to PPs during refeeding when stromal cells sensed nutritional signals and upregulated CXCL13 expression to recruit naive B cells. Furthermore, temporal fasting before oral immunization with ovalbumin abolished the induction of antigen-specific IgA, failed to induce oral tolerance, and eventually exacerbated food antigen-induced diarrhea. Thus, nutritional signals are critical in maintaining gut immune homeostasis.


Asunto(s)
Linfocitos B/fisiología , Inmunidad Mucosa , Animales , Antígenos/inmunología , Linfocitos B/inmunología , Linfocitos B/metabolismo , Médula Ósea/inmunología , Médula Ósea/metabolismo , Quimiocina CXCL13/genética , Quimiocina CXCL13/metabolismo , Ayuno , Regulación de la Expresión Génica , Glucólisis , Inmunoglobulina A/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Estado Nutricional , Ovalbúmina/inmunología , Ganglios Linfáticos Agregados/inmunología , Ganglios Linfáticos Agregados/metabolismo , Ganglios Linfáticos Agregados/patología , Receptores CXCR5/genética , Receptores CXCR5/metabolismo , Transducción de Señal , Células del Estroma/citología , Células del Estroma/metabolismo , Serina-Treonina Quinasas TOR/metabolismo
7.
Cell Rep ; 28(1): 145-158.e9, 2019 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-31269436

RESUMEN

Hematopoietic stem cells (HSCs) maintain lifelong hematopoiesis by remaining quiescent in the bone marrow niche. Recapitulation of a quiescent state in culture has not been achieved, as cells rapidly proliferate and differentiate in vitro. After exhaustive analysis of different environmental factor combinations and concentrations as a way to mimic physiological conditions, we were able to maintain engraftable quiescent HSCs for 1 month in culture under very low cytokine concentrations, hypoxia, and very high fatty acid levels. Exogenous fatty acids were required likely due to suppression of intrinsic fatty acid synthesis by hypoxia and low cytokine conditions. By contrast, high cytokine concentrations or normoxia induced HSC proliferation and differentiation. Our culture system provides a means to evaluate properties of steady-state HSCs and test effects of defined factors in vitro under near-physiological conditions.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Citocinas/farmacología , Ácidos Grasos/farmacología , Hematopoyesis/efectos de los fármacos , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/efectos de los fármacos , Animales , Apoptosis , Médula Ósea/metabolismo , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Hipoxia de la Célula/fisiología , Proliferación Celular/efectos de los fármacos , Proliferación Celular/fisiología , Colesterol/farmacología , Ontología de Genes , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/metabolismo , Humanos , Insulina/farmacología , Ratones , Ratones Endogámicos C57BL , Análisis de Secuencia por Matrices de Oligonucleótidos , Análisis de la Célula Individual , Factor de Células Madre/farmacología , Nicho de Células Madre/efectos de los fármacos , Nicho de Células Madre/fisiología
8.
J Clin Biochem Nutr ; 63(1): 70-79, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-30087547

RESUMEN

Carbon monoxide-generating heme oxygenase-2 is expressed in neurons and plays a crucial role for regulating hypoxic vasodilation through mechanisms unlocking carbon monoxide-dependent inhibition of H2S-generating cystathionine ß-synthase expressed in astrocytes. This study aims to examine whether heme oxygenase-2 plays a protective role in mice against stroke. Focal ischemia was induced by middle cerebral artery occlusion. Regional differences in metabolites among ipsilateral and contralateral hemispheres were analysed by quantitative imaging mass spectrometry equipped with an image-processing platform to optimize comparison of local metabolite contents among different animals. Under normoxia, blood flow velocity in precapillary arterioles were significantly elevated in heme oxygenase-2-null mice vs controls, while metabolic intermediates of central carbon metabolism and glutamate synthesis were elevated in the brain of heme oxygenase-2-null mice, suggesting greater metabolic demands to induce hyperemia in these mice. In response to focal ischemia, heme oxygenase-2-null mice exhibited greater regions of ischemic core that coincide with notable decreases in energy metabolism in the contralateral hemisphere as well as in penumbra. In conclusion, these findings suggest that heme oxygenase-2 is involved in mechanisms by which not only protects against compromised energy metabolism of the ipsilateral hemisphere but also ameliorates transhemispheric diaschisis of the contralateral hemisphere in ischemic brain.

9.
Nat Commun ; 9(1): 1561, 2018 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-29674746

RESUMEN

Gold deposition with diagonal angle towards boehmite-based nanostructure creates random arrays of horse-bean-shaped nanostructures named gold-nanofève (GNF). GNF generates many electromagnetic hotspots as surface-enhanced Raman spectroscopy (SERS) excitation sources, and enables large-area visualization of molecular vibration fingerprints of metabolites in human cancer xenografts in livers of immunodeficient mice with sufficient sensitivity and uniformity. Differential screening of GNF-SERS signals in tumours and those in parenchyma demarcated tumour boundaries in liver tissues. Furthermore, GNF-SERS combined with quantum chemical calculation identified cysteine-derived glutathione and hypotaurine (HT) as tumour-dominant and parenchyma-dominant metabolites, respectively. CD44 knockdown in cancer diminished glutathione, but not HT in tumours. Mechanisms whereby tumours sustained HT under CD44-knockdown conditions include upregulation of PHGDH, PSAT1 and PSPH that drove glycolysis-dependent activation of serine/glycine-cleavage systems to provide one-methyl group for HT synthesis. HT was rapidly converted into taurine in cancer cells, suggesting that HT is a robust anti-oxidant for their survival under glutathione-suppressed conditions.


Asunto(s)
Antioxidantes/química , Antioxidantes/metabolismo , Oro/química , Nanopartículas del Metal/química , Neoplasias/metabolismo , Espectrometría Raman/métodos , Taurina/análogos & derivados , Animales , Femenino , Glutatión/metabolismo , Oro/metabolismo , Humanos , Hígado/química , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones SCID , Neoplasias/genética , Espectrometría Raman/instrumentación , Taurina/química , Taurina/metabolismo , Transaminasas/genética , Transaminasas/metabolismo
10.
Rinsho Ketsueki ; 58(10): 1844-1850, 2017.
Artículo en Japonés | MEDLINE | ID: mdl-28978823

RESUMEN

During steady-state conditions, hematopoietic stem cells (HSCs) maintain a quiescent status in the cell cycle. Upon infection or inflammation, bone marrow HSCs begin proliferating and generating differentiated hematopoietic cells via multi-lineage differentiation and self-renewal; this effect is partially due to the alteration of their surrounding microenvironment or niche. In addition, recent studies have revealed that the bone marrow niche critically contributes to abnormal hematopoiesis, including leukemogenesis. In this review, we discuss the recent advances in our understanding of HSC/niche functions and the regulatory machineries employed during homeostasis, stress hematopoiesis, or disease conditions.


Asunto(s)
Hematopoyesis , Nicho de Células Madre , Animales , Células Madre Hematopoyéticas , Humanos , Leucemia , Neovascularización Patológica
11.
Front Cell Dev Biol ; 5: 62, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28660186

RESUMEN

Continuous generation of blood cells over an organism's lifetime is supported by hematopoietic stem/progenitor cells (HSPCs) capable of producing all hematopoietic cell subtypes. Adult mammalian HSPCs are localized to bone marrow and regulated by their neighboring microenvironment, or "niche." Because interactions of HSPCs with their niches are highly dynamic and complex, the recent development of imaging technologies provides a powerful new tool to understand stem cell/niche biology. In this review, we discuss recent advances in our understanding of dynamic HSPC/niche interactions during development, homeostasis, disease states or aging with a focus on studies advanced by imaging analysis. We also summarize methods to visualize HSPCs and niche cells in vivo, including use of HSPC reporter mice and chemical probes. Findings emerging from these investigations could suggest novel therapies for diseases and aging.

12.
Cell Stem Cell ; 19(2): 192-204, 2016 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-27345838

RESUMEN

Hematopoietic stem cells (HSCs) maintain quiescence by activating specific metabolic pathways, including glycolysis. We do not yet have a clear understanding of how this metabolic activity changes during stress hematopoiesis, such as bone marrow transplantation. Here, we report a critical role for the p38MAPK family isoform p38α in initiating hematopoietic stem and progenitor cell (HSPC) proliferation during stress hematopoiesis in mice. We found that p38MAPK is immediately phosphorylated in HSPCs after a hematological stress, preceding increased HSPC cycling. Conditional deletion of p38α led to defective recovery from hematological stress and a delay in initiation of HSPC proliferation. Mechanistically, p38α signaling increases expression of inosine-5'-monophosphate dehydrogenase 2 in HSPCs, leading to altered levels of amino acids and purine-related metabolites and changes in cell-cycle progression in vitro and in vivo. Our studies have therefore uncovered a p38α-mediated pathway that alters HSPC metabolism to respond to stress and promote recovery.


Asunto(s)
Ciclo Celular , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Purinas/metabolismo , Estrés Fisiológico , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Apoptosis , Proliferación Celular , Activación Enzimática , Hematopoyesis , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/enzimología , IMP Deshidrogenasa/antagonistas & inhibidores , IMP Deshidrogenasa/metabolismo , Ratones Endogámicos C57BL , Factor de Transcripción Asociado a Microftalmía/metabolismo , Mutación/genética , Fenotipo , Especies Reactivas de Oxígeno/metabolismo
13.
Acta Neurochir (Wien) ; 158(6): 1057-67, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27040552

RESUMEN

BACKGROUND: It is believed that increased intracranial pressure immediately after subarachnoid hemorrhage (SAH) causes extensive brain ischemia and results in worsening clinical status. Arterial flow to the cerebral surfaces is clinically well maintained during clipping surgery regardless of the severity of the World Federation of Neurological Societies grade after SAH. To explore what kinds of changes occur in the cortical microcirculation, not at the cerebral surface, we examined cortical microcirculation after SAH using two-photon laser scanning microscopy (TPLSM). METHODS: SAH was induced in mice with an endovascular perforation model. Following continuous injection of rhodamine 6G, velocities of labeled platelets and leukocytes and unlabeled red blood cells (RBCs) were measured in the cortical capillaries 60 min after SAH with a line-scan method using TPLSM, and the data were compared to a sham group and P-selectin monoclonal antibody-treated group. RESULTS: Velocities of leukocytes, platelets, and RBCs in capillaries decreased significantly 60 min after SAH. Rolling and adherent leukocytes suddenly prevented other blood cells from flowing in the capillaries. Flowing blood cells also decreased significantly in each capillary after SAH. This no-reflow phenomenon induced by plugging leukocytes was often observed in the SAH group but not in the sham group. The decreased velocities of blood cells were reversed by pretreatment with the monoclonal antibody of P-selection, an adhesion molecule expressed on the surfaces of both endothelial cells and platelets. CONCLUSIONS: SAH caused sudden worsening of cortical microcirculation at the onset. Leukocyte plugging in capillaries is one of the reasons why cortical microcirculation is aggravated after SAH.


Asunto(s)
Circulación Cerebrovascular , Leucocitos/patología , Microcirculación , Hemorragia Subaracnoidea/fisiopatología , Animales , Velocidad del Flujo Sanguíneo , Masculino , Ratones , Hemorragia Subaracnoidea/sangre
14.
Cancer Res ; 76(10): 2954-63, 2016 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-26980765

RESUMEN

Extracellular free amino acids contribute to the interaction between a tumor and its microenvironment through effects on cellular metabolism and malignant behavior. System xc(-) is composed of xCT and CD98hc subunits and functions as a plasma membrane antiporter for the uptake of extracellular cystine in exchange for intracellular glutamate. Here, we show that the EGFR interacts with xCT and thereby promotes its cell surface expression and function in human glioma cells. EGFR-expressing glioma cells manifested both enhanced antioxidant capacity as a result of increased cystine uptake, as well as increased glutamate, which promotes matrix invasion. Imaging mass spectrometry also revealed that brain tumors formed in mice by human glioma cells stably overexpressing EGFR contained higher levels of reduced glutathione compared with those formed by parental cells. Targeted inhibition of xCT suppressed the EGFR-dependent enhancement of antioxidant capacity in glioma cells, as well as tumor growth and invasiveness. Our findings establish a new functional role for EGFR in promoting the malignant potential of glioma cells through interaction with xCT at the cell surface. Cancer Res; 76(10); 2954-63. ©2016 AACR.


Asunto(s)
Sistema de Transporte de Aminoácidos y+/metabolismo , Neoplasias Encefálicas/patología , Receptores ErbB/metabolismo , Regulación Neoplásica de la Expresión Génica , Glioma/patología , Animales , Antioxidantes/metabolismo , Apoptosis , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/metabolismo , Membrana Celular/metabolismo , Movimiento Celular , Proliferación Celular , Cistina/metabolismo , Glioma/tratamiento farmacológico , Glioma/metabolismo , Ácido Glutámico/metabolismo , Humanos , Ratones , Ratones Endogámicos NOD , Ratones Desnudos , Ratones SCID , Especies Reactivas de Oxígeno/metabolismo , Sulfasalazina/farmacología , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Pflugers Arch ; 468(1): 13-22, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26490456

RESUMEN

Bone marrow, the site of hematopoiesis throughout adulthood, is a physiologically hypoxic organ. Thus, various biological oxygen sensors and their signaling cascades play a pivotal role in hematopoietic systems in the bone marrow under both physiologic and pathologic conditions. Hypoxia-inducible factors (HIFs) are hypoxic stress sensor proteins that are stabilized under homeostatic or stress-induced hypoxia. In the hypoxic bone marrow, HIFs play crucial roles in hematopoietic stem cells (HSCs) and in the cells of the HSC niche. The signals downstream of the HIFs maintain HSC quiescence, survival, and metabolic homeostasis through both cell-autonomous and non-cell-autonomous mechanisms. Leukemic stem cells (LSCs) hijack these delicate hypoxia-sensing mechanisms to sustain their self-renewal potential, promoting disease progression and drug resistance even under normoxic conditions. This review focuses on HIF-mediated oxygen-sensing mechanisms of adult HSCs and LSCs and their niche cells in the hypoxic bone marrow.

16.
Respir Physiol Neurobiol ; 184(2): 139-48, 2012 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-22516267

RESUMEN

It has been recognized that gaseous molecules and their signaling cascades play a vital role in alterations of metabolic systems in physiologic and pathologic conditions. Contrary to this awareness, detailed mechanisms whereby gases exert their actions, in particular in vivo, have been unclear because of several reasons. Gaseous signaling involves diverse reactions with metal centers of metalloproteins and thiol modification of cysteine residues of proteins. Both the multiplicity of gas targets and the technical limitations in accessing local gas concentrations make dissection of exact actions of any gas mediator a challenge. However, a series of advanced technologies now offer ways to explore gas-responsive regulatory processes in vivo. Imaging mass spectrometry combined with quantitative metabolomics by capillary-electrophoresis/mass spectrometry reveals spatio-temporal profiles of many metabolites. Comparing the metabolic footprinting of murine samples with a targeted deletion of a specific gas-producing enzyme makes it possible to determine sites of actions of the gas. In this review, we intend to elaborate on the ideas how small gaseous molecules interact with metabolic systems to control organ functions such as cerebral vascular tone and energy metabolism in vivo.


Asunto(s)
Gases , Metabolismo/fisiología , Transducción de Señal/fisiología , Animales , Monóxido de Carbono/fisiología , Cistationina betasintasa/fisiología , Humanos , Sulfuro de Hidrógeno , Hipoxia/fisiopatología , Metabolómica , Modelos Moleculares
17.
J Mol Med (Berl) ; 90(3): 245-54, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22331189

RESUMEN

Carbon monoxide (CO) is a gaseous product generated by heme oxygenase (HO), which oxidatively degrades heme. While the stress-inducible HO-1 has well been recognized as an anti-oxidative defense mechanism under stress conditions, recent studies suggest that cancer cells utilize the reaction for their survival. HO-2, the constitutive isozyme, also plays protective roles as a tonic regulator for neurovascular function. Although protective roles of the enzyme reaction and CO have extensively been studied, little information is available on the molecular mechanisms by which the gas exerts its biological actions. Recent studies using metabolomics revealed that CO inhibits cystathionine ß-synthase (CBS), which generates H(2)S, another gaseous mediator. The CO-dependent CBS inhibition may impact on the remethylation cycle and related metabolic pathways including the methionine salvage pathway and polyamine synthesis. This review focuses on the gas-responsive regulation of metabolic systems, particularly the remethylation and transsulfuration pathways, and their putative implications for cancer and ischemic diseases.


Asunto(s)
Monóxido de Carbono/metabolismo , Metilación/efectos de los fármacos , Isquemia Miocárdica/fisiopatología , Neoplasias/fisiopatología , Azufre/metabolismo , Monóxido de Carbono/farmacología , Monóxido de Carbono/fisiología , Cistationina betasintasa/metabolismo , Hemo Oxigenasa (Desciclizante)/metabolismo , Humanos , Sulfuro de Hidrógeno/metabolismo
18.
Proc Natl Acad Sci U S A ; 109(4): 1293-8, 2012 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-22232681

RESUMEN

Enhancement of cerebral blood flow by hypoxia is critical for brain function, but signaling systems underlying its regulation have been unclear. We report a pathway mediating hypoxia-induced cerebral vasodilation in studies monitoring vascular disposition in cerebellar slices and in intact mouse brains using two-photon intravital laser scanning microscopy. In this cascade, hypoxia elicits cerebral vasodilation via the coordinate actions of H(2)S formed by cystathionine ß-synthase (CBS) and CO generated by heme oxygenase (HO)-2. Hypoxia diminishes CO generation by HO-2, an oxygen sensor. The constitutive CO physiologically inhibits CBS, and hypoxia leads to increased levels of H(2)S that mediate the vasodilation of precapillary arterioles. Mice with targeted deletion of HO-2 or CBS display impaired vascular responses to hypoxia. Thus, in intact adult brain cerebral cortex of HO-2-null mice, imaging mass spectrometry reveals an impaired ability to maintain ATP levels on hypoxia.


Asunto(s)
Monóxido de Carbono/metabolismo , Cerebro/irrigación sanguínea , Sulfuro de Hidrógeno/metabolismo , Hipoxia/fisiopatología , Microcirculación/fisiología , Flujo Sanguíneo Regional/fisiología , Vasodilatación/fisiología , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico , Adenosina Trifosfato/metabolismo , Análisis de Varianza , Animales , Western Blotting , Cistationina betasintasa/metabolismo , Cartilla de ADN/genética , Hemo Oxigenasa (Desciclizante)/genética , Hemo Oxigenasa (Desciclizante)/metabolismo , Inmunohistoquímica , Espectrometría de Masas , Ratones , Microscopía Confocal
19.
Antioxid Redox Signal ; 13(8): 1157-67, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20486758

RESUMEN

Local responses of energy metabolism during brain ischemia are too heterogeneous to decipher redox distribution between anoxic core and adjacent salvageable regions such as penumbra. Imaging mass spectrometry combined by capillary electrophoresis/mass spectrometry providing quantitative metabolomics revealed spatio-temporal changes in adenylates and NADH in a mouse middle-cerebral artery occlusion model. Unlike the core where ATP decreased, the penumbra displayed paradoxical elevation of ATP despite the constrained blood supply. It is noteworthy that the NADH elevation in the ischemic region is clearly demarcated by the ATP-depleting core. Results suggest that metabolism in ischemic penumbra does not respond passively to compromised circulation, but actively compensates energy charges.


Asunto(s)
Adenosina Trifosfato/metabolismo , Isquemia Encefálica/metabolismo , Animales , Isquemia Encefálica/diagnóstico , Isquemia Encefálica/fisiopatología , Corteza Cerebral/irrigación sanguínea , Corteza Cerebral/fisiopatología , Modelos Animales de Enfermedad , Electroforesis Capilar , Infarto de la Arteria Cerebral Media/metabolismo , Infarto de la Arteria Cerebral Media/fisiopatología , Espectrometría de Masas , Ratones , Oxidación-Reducción , Transducción de Señal
20.
J Infect Chemother ; 10(1): 46-8, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-14991518

RESUMEN

We report a case of infection of a mastoid cavity after mastoidectomy had been performed for chronic otitis media with cholesteatoma. The infection was caused by Staphylococcus intermedius after a pet dog had licked the patient's ears. Bacterial strains from the dog's saliva and the otorrhea in the patient were confirmed to be identical by pulsed-field gel electrophoresis analysis. The possibility of an oral transmission route of S. intermedius from pets to humans should be noted.


Asunto(s)
Mastoiditis/microbiología , Infecciones Estafilocócicas/microbiología , Infecciones Estafilocócicas/transmisión , Staphylococcus/genética , Zoonosis , Animales , ADN Bacteriano/análisis , Diagnóstico Diferencial , Perros , Electroforesis en Gel de Campo Pulsado , Femenino , Humanos , Persona de Mediana Edad , Saliva/microbiología , Staphylococcus/aislamiento & purificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA