Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Physiol Rev ; 102(2): 859-892, 2022 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-34486392

RESUMEN

Globin proteins exist in every cell type of the vasculature, from erythrocytes to endothelial cells, vascular smooth muscle cells, and peripheral nerve cells. Many globin subtypes are also expressed in muscle tissues (including cardiac and skeletal muscle), in other organ-specific cell types, and in cells of the central nervous system (CNS). The ability of each of these globins to interact with molecular oxygen (O2) and nitric oxide (NO) is preserved across these contexts. Endothelial α-globin is an example of extraerythrocytic globin expression. Other globins, including myoglobin, cytoglobin, and neuroglobin, are observed in other vascular tissues. Myoglobin is observed primarily in skeletal muscle and smooth muscle cells surrounding the aorta or other large arteries. Cytoglobin is found in vascular smooth muscle but can also be expressed in nonvascular cell types, especially in oxidative stress conditions after ischemic insult. Neuroglobin was first observed in neuronal cells, and its expression appears to be restricted mainly to the CNS and the peripheral nervous system. Brain and CNS neurons expressing neuroglobin are positioned close to many arteries within the brain parenchyma and can control smooth muscle contraction and thus tissue perfusion and vascular reactivity. Overall, reactions between NO and globin heme iron contribute to vascular homeostasis by regulating vasodilatory NO signals and scavenging reactive species in cells of the mammalian vascular system. Here, we discuss how globin proteins affect vascular physiology, with a focus on NO biology, and offer perspectives for future study of these functions.


Asunto(s)
Fenómenos Fisiológicos Cardiovasculares , Citoglobina/metabolismo , Células Endoteliales/metabolismo , Globinas/metabolismo , Animales , Humanos , Mioglobina/metabolismo , Neuroglobina/metabolismo
2.
Nature ; 595(7866): 295-302, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34079130

RESUMEN

Sickle cell disease (SCD) is caused by a mutation in the ß-globin gene HBB1. We used a custom adenine base editor (ABE8e-NRCH)2,3 to convert the SCD allele (HBBS) into Makassar ß-globin (HBBG), a non-pathogenic variant4,5. Ex vivo delivery of mRNA encoding the base editor with a targeting guide RNA into haematopoietic stem and progenitor cells (HSPCs) from patients with SCD resulted in 80% conversion of HBBS to HBBG. Sixteen weeks after transplantation of edited human HSPCs into immunodeficient mice, the frequency of HBBG was 68% and hypoxia-induced sickling of bone marrow reticulocytes had decreased fivefold, indicating durable gene editing. To assess the physiological effects of HBBS base editing, we delivered ABE8e-NRCH and guide RNA into HSPCs from a humanized SCD mouse6 and then transplanted these cells into irradiated mice. After sixteen weeks, Makassar ß-globin represented 79% of ß-globin protein in blood, and hypoxia-induced sickling was reduced threefold. Mice that received base-edited HSPCs showed near-normal haematological parameters and reduced splenic pathology compared to mice that received unedited cells. Secondary transplantation of edited bone marrow confirmed that the gene editing was durable in long-term haematopoietic stem cells and showed that HBBS-to-HBBG editing of 20% or more is sufficient for phenotypic rescue. Base editing of human HSPCs avoided the p53 activation and larger deletions that have been observed following Cas9 nuclease treatment. These findings point towards a one-time autologous treatment for SCD that eliminates pathogenic HBBS, generates benign HBBG, and minimizes the undesired consequences of double-strand DNA breaks.


Asunto(s)
Adenina/metabolismo , Anemia de Células Falciformes/genética , Anemia de Células Falciformes/terapia , Edición Génica , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/metabolismo , Globinas beta/genética , Animales , Antígenos CD34/metabolismo , Proteína 9 Asociada a CRISPR/metabolismo , Modelos Animales de Enfermedad , Femenino , Terapia Genética , Genoma Humano/genética , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/patología , Humanos , Masculino , Ratones
3.
Blood ; 142(10): 918-932, 2023 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-37339583

RESUMEN

Most cells can eliminate unstable or misfolded proteins through quality control mechanisms. In the inherited red blood cell disorder ß-thalassemia, mutations in the ß-globin gene (HBB) lead to a reduction in the corresponding protein and the accumulation of cytotoxic free α-globin, which causes maturation arrest and apoptosis of erythroid precursors and reductions in the lifespan of circulating red blood cells. We showed previously that excess α-globin is eliminated by Unc-51-like autophagy activating kinase 1 (ULK1)-dependent autophagy and that stimulating this pathway by systemic mammalian target of rapamycin complex 1 (mTORC1) inhibition alleviates ß-thalassemia pathologies. We show here that disrupting the bicistronic microRNA gene miR-144/451 alleviates ß-thalassemia by reducing mTORC1 activity and stimulating ULK1-mediated autophagy of free α-globin through 2 mechanisms. Loss of miR-451 upregulated its target messenger RNA, Cab39, which encodes a cofactor for LKB1, a serine-threonine kinase that phosphorylates and activates the central metabolic sensor adenosine monophosphate-activated protein kinase (AMPK). The resultant enhancement of LKB1 activity stimulated AMPK and its downstream effects, including repression of mTORC1 and direct activation of ULK1. In addition, loss of miR-144/451 inhibited the expression of erythroblast transferrin receptor 1, causing intracellular iron restriction, which has been shown to inhibit mTORC1, reduce free α-globin precipitates, and improve hematological indices in ß-thalassemia. The beneficial effects of miR-144/451 loss in ß-thalassemia were inhibited by the disruption of Cab39 or Ulk1 genes. Together, our findings link the severity of ß-thalassemia to a highly expressed erythroid microRNA locus and a fundamental, metabolically regulated protein quality control pathway that is amenable to therapeutic manipulation.


Asunto(s)
MicroARNs , Talasemia beta , Humanos , Talasemia beta/terapia , Proteínas Quinasas Activadas por AMP/metabolismo , Globinas alfa , Autofagia/genética , MicroARNs/genética , Diana Mecanicista del Complejo 1 de la Rapamicina , Homólogo de la Proteína 1 Relacionada con la Autofagia/genética , Homólogo de la Proteína 1 Relacionada con la Autofagia/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética
4.
Neurobiol Dis ; 159: 105483, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34400304

RESUMEN

Mitochondrial diseases are among the most prevalent groups of inherited neurological disorders, affecting up to 1 in 5000 adults. Despite the progress achieved on the identification of gene mutations causing mitochondrial pathologies, they cannot be cured so far. Harlequin mice, a relevant model of mitochondrial pathology due to apoptosis inducing factor depletion, suffer from progressive disappearance of retinal ganglion cells leading to optic neuropathy. In our previous work, we showed that administering adeno-associated virus encompassing the coding sequences for neuroglobin, (a neuroprotective molecule belonging to the globin family) or apoptosis-inducing factor, before neurodegeneration onset, prevented retinal ganglion cell loss and preserved visual function. One of the challenges to develop an effective treatment for optic neuropathies is to consider that by the time patients become aware of their handicap, a large amount of nerve fibers has already disappeared. Gene therapy was performed in Harlequin mice aged between 4 and 5 months with either a neuroglobin or an apoptosis-inducing factor vector to determine whether the increased abundance of either one of these proteins in retinas could preserve visual function at this advanced stage of the disease. We demonstrated that gene therapy, by preserving the connectivity of transduced retinal ganglion cells and optic nerve bioenergetics, results in the enhancement of visual cortex activity, ultimately rescuing visual impairment. This study demonstrates that: (a) An increased abundance of neuroglobin functionally overcomes apoptosis-inducing factor absence in Harlequin mouse retinas at a late stage of neuronal degeneration; (b) The beneficial effect for visual function could be mediated by neuroglobin localization to the mitochondria, thus contributing to the maintenance of the organelle homeostasis.


Asunto(s)
Factor Inductor de la Apoptosis/genética , Complejo IV de Transporte de Electrones/metabolismo , Complejo I de Transporte de Electrón/metabolismo , Neuroglobina/genética , Atrofia Óptica/metabolismo , Nervio Óptico/metabolismo , Células Ganglionares de la Retina/metabolismo , Agudeza Visual/genética , Corteza Visual/metabolismo , Animales , Progresión de la Enfermedad , Terapia Genética , Ratones , Atrofia Óptica/patología , Atrofia Óptica/fisiopatología , Nervio Óptico/patología , Nervio Óptico/fisiopatología , Células Ganglionares de la Retina/patología , Corteza Visual/patología , Vías Visuales
5.
Blood ; 133(23): 2518-2528, 2019 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-30971389

RESUMEN

The microRNA (miRNA) locus miR-144/451 is abundantly expressed in erythrocyte precursors, facilitating their terminal maturation and protecting against oxidant stress. However, the full repertoire of erythroid miR-144/451 target messenger RNAs (mRNAs) and associated cellular pathways is unknown. In general, the numbers of mRNAs predicted to be targeted by an miRNA vary greatly from hundreds to thousands, and are dependent on experimental approaches. To comprehensively and accurately identify erythroid miR-144/451 target mRNAs, we compared gene knockout and wild-type fetal liver erythroblasts by RNA sequencing, quantitative proteomics, and RNA immunoprecipitation of Argonaute (Ago), a component of the RNA-induced silencing complex that binds miRNAs complexed to their target mRNAs. Argonaute bound ∼1400 erythroblast mRNAs in a miR-144/451-dependent manner, accounting for one-third of all Ago-bound mRNAs. However, only ∼100 mRNAs were stabilized after miR-144/451 loss. Thus, miR-144 and miR-451 deregulate <10% of mRNAs that they bind, a characteristic that likely applies generally to other miRNAs. Using stringent selection criteria, we identified 53 novel miR-144/451 target mRNAs. One of these, Cox10, facilitates the assembly of mitochondrial electron transport complex IV. Loss of miR-144/451 caused increased Cox10 mRNA and protein, accumulation of complex IV, and increased mitochondrial membrane potential with no change in mitochondrial mass. Thus, miR-144/451 represses mitochondrial respiration during erythropoiesis by inhibiting the production of Cox10.


Asunto(s)
Transferasas Alquil y Aril/biosíntesis , Eritropoyesis/genética , Regulación de la Expresión Génica/genética , Proteínas de la Membrana/biosíntesis , MicroARNs/genética , Transferasas Alquil y Aril/genética , Animales , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados
6.
Haematologica ; 103(3): 406-416, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29269522

RESUMEN

The microRNAs miR-144 and -451 are encoded by a bicistronic gene that is strongly induced during red blood cell formation (erythropoiesis). Ablation of the miR-144/451 gene in mice causes mild anemia under baseline conditions. Here we show that miR-144/451-/- erythroblasts exhibit increased apoptosis during recovery from acute anemia. Mechanistically, miR-144/451 depletion increases the expression of the miR-451 target mRNA Cab39, which encodes a co-factor for the serine-threonine kinase LKB1. During erythropoietic stress, miR-144/451-/- erythroblasts exhibit abnormally increased Cab39 protein, which activates LKB1 and its downstream AMPK/mTOR effector pathway. Suppression of this pathway via drugs or shRNAs enhances survival of the mutant erythroblasts. Thus, miR-144/451 facilitates recovery from acute anemia by repressing Cab39/AMPK/mTOR. Our findings suggest that miR-144/451 is a key protector of erythroblasts during pathological states associated with dramatically increased erythropoietic demand, including acute blood loss and hemolytic anemia.


Asunto(s)
Anemia/sangre , Células Eritroides/citología , MicroARNs/fisiología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Enfermedad Aguda , Animales , Proteínas de Unión al Calcio/genética , Supervivencia Celular , Eritropoyesis , Redes y Vías Metabólicas , Ratones , ARN Mensajero , Serina-Treonina Quinasas TOR/metabolismo
7.
Mol Ther ; 22(6): 1096-1109, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24622090

RESUMEN

Neuroglobin (NGB) is considered as an endogenous neuroprotective molecule against stroke, since the protein alleviates the adverse effects of hypoxic and ischemic insults. We previously demonstrated the functional link between NGB and mitochondria since it is required for respiratory chain function. Thus, here, we evaluated the relevance of this effect in the Harlequin (Hq) mouse strain, which exhibits retinal ganglion cell (RGC) loss and optic atrophy due to a respiratory chain complex I (CI) defect. A twofold decrease of NGB amounts was observed in Hq retinas. We constructed a recombinant adeno-associated virus which combines to the mouse NGB open reading frame, its 5' and 3'UTR, for guarantying mRNA stability and translation capacity. The vector was administrated intravitreally to Hq mice and NGB expression was stable for up to 7 months without negative effect on retinal architecture or function. On the contrary, RGCs and their axons were substantially preserved from degeneration; consequently, CI activity in optic nerves was protected conferring improvements in vision. Hence, we established that NGB prevents respiratory chain impairment, therefore, protecting visual function otherwise compromised by mitochondrial energetic failure.


Asunto(s)
Complejo I de Transporte de Electrón/deficiencia , Globinas/genética , Proteínas del Tejido Nervioso/genética , Atrofia Óptica/prevención & control , Atrofia Óptica/terapia , Células Ganglionares de la Retina/metabolismo , Animales , Axones/metabolismo , Axones/patología , Dependovirus/genética , Modelos Animales de Enfermedad , Terapia Genética , Vectores Genéticos/administración & dosificación , Gliosis/patología , Gliosis/prevención & control , Globinas/metabolismo , Ratones , Proteínas del Tejido Nervioso/metabolismo , Neuroglobina , Atrofia Óptica/genética , Atrofia Óptica/patología , Células Ganglionares de la Retina/patología
8.
J Biol Chem ; 288(10): 6957-67, 2013 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-23288852

RESUMEN

Neuroglobins, previously thought to be restricted to vertebrate neurons, were detected in the brain of a photosymbiotic acoel, Symsagittifera roscoffensis, and in neurosensory cells of the jellyfish Clytia hemisphaerica. For the neuroglobin of S. roscoffensis, a member of a lineage that originated either at the base of the bilateria or of the deuterostome clade, we report the ligand binding properties, crystal structure at 2.3 Å, and brain immunocytochemical pattern. We also describe in situ hybridizations of two neuroglobins specifically expressed in differentiating nematocytes (neurosensory cells) and in statocytes (ciliated mechanosensory cells) of C. hemisphaerica, a member of the early branching animal phylum cnidaria. In silico searches using these neuroglobins as queries revealed the presence of previously unidentified neuroglobin-like sequences in most metazoan lineages. Because neural systems are almost ubiquitous in metazoa, the constitutive expression of neuroglobin-like proteins strongly supports the notion of an intimate association of neuroglobins with the evolution of animal neural systems and hints at the preservation of a vitally important function. Neuroglobins were probably recruited in the first protoneurons in early metazoans from globin precursors. Neuroglobins were identified in choanoflagellates, sponges, and placozoans and were conserved during nervous system evolution. Because the origin of neuroglobins predates the other metazoan globins, it is likely that neuroglobin gene duplication followed by co-option and subfunctionalization led to the emergence of globin families in protostomes and deuterostomes (i.e. convergent evolution).


Asunto(s)
Globinas/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Sistema Nervioso/metabolismo , Precursores de Proteínas/metabolismo , Secuencia de Aminoácidos , Animales , Cristalografía por Rayos X , Evolución Molecular , Perfilación de la Expresión Génica , Variación Genética , Globinas/química , Globinas/genética , Hidrozoos/genética , Hidrozoos/metabolismo , Hibridación in Situ , Modelos Moleculares , Datos de Secuencia Molecular , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/genética , Sistema Nervioso/citología , Neuroglobina , Oxígeno/química , Oxígeno/metabolismo , Filogenia , Platelmintos/genética , Platelmintos/metabolismo , Unión Proteica , Precursores de Proteínas/genética , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Homología de Secuencia de Aminoácido
9.
Biochim Biophys Acta ; 1834(9): 1772-8, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23639750

RESUMEN

Neuroglobin is a member of the globin superfamily proposed to be only expressed in neurons and involved in neuronal protection from hypoxia or oxidative stress. A significant fraction of the protein localizes within the mitochondria and is directly associated with mitochondrial metabolism and integrity. The retina is the site of the highest concentration for neuroglobin and has been reported to be up to 100-fold higher than in the brain. Since neuroglobin was especially abundant in retinal ganglion cell layer, we investigated its abundance in optic nerves. Remarkably in optic nerves, neuroglobin is observed, as expected, in retinal ganglion cell axon profiles but also astrocyte processes, in physiological conditions, possess high levels of the protein. Neuroglobin mRNA and protein levels are ~10-fold higher in optic nerves than in retinas, indicating an important accumulation of neuroglobin in these support cells. Additionally, neuroglobin levels increase in Müller cells during reactive gliosis in response to eye injury. This suggests the pivotal role of neuroglobin in retinal glia involved in neuronal support and/or healing. This article is part of a Special Issue entitled: Oxygen Binding and Sensing Proteins.


Asunto(s)
Gliosis/metabolismo , Globinas/metabolismo , Cristalino/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Nervio Óptico/metabolismo , Células Ganglionares de la Retina/metabolismo , Vías Visuales/metabolismo , Animales , Western Blotting , Gliosis/patología , Globinas/genética , Cristalino/lesiones , Cristalino/patología , Masculino , Proteínas del Tejido Nervioso/genética , Neuroglía/citología , Neuroglía/metabolismo , Neuroglobina , Nervio Óptico/citología , ARN Mensajero/genética , Ratas , Ratas Long-Evans , Reacción en Cadena en Tiempo Real de la Polimerasa , Células Ganglionares de la Retina/citología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Vías Visuales/patología
10.
Acta Crystallogr D Biol Crystallogr ; 70(Pt 4): 1005-14, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24699645

RESUMEN

Neuroglobin plays an important function in the supply of oxygen in nervous tissues. In human neuroglobin, a cysteine at position 46 in the loop connecting the C and D helices of the globin fold is presumed to form an intramolecular disulfide bond with Cys55. Rupture of this disulfide bridge stabilizes bi-histidyl haem hexacoordination, causing an overall decrease in the affinity for oxygen. Here, the first X-ray structure of wild-type human neuroglobin is reported at 1.74 Šresolution. This structure provides a direct observation of two distinct conformations of the CD region containing the intramolecular disulfide link and highlights internal cavities that could be involved in ligand migration and/or are necessary to enable the conformational transition between the low and high oxygen-affinity states following S-S bond formation.


Asunto(s)
Disulfuros/química , Globinas/química , Proteínas del Tejido Nervioso/química , Oxígeno/química , Cristalografía por Rayos X , Disulfuros/metabolismo , Globinas/metabolismo , Humanos , Modelos Moleculares , Proteínas del Tejido Nervioso/metabolismo , Neuroglobina , Oxígeno/metabolismo , Estructura Cuaternaria de Proteína , Estructura Terciaria de Proteína , Homología Estructural de Proteína
11.
Biochim Biophys Acta ; 1823(12): 2261-73, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23036890

RESUMEN

Neuroglobin is a member of the globin superfamily expressed in vertebrate brain and retina. The protein is thought to be involved in neuronal protection from hypoxia or oxidative stress and could represent a key element of Alzheimer disease pathogenesis. Our aim was to determine whether neuroglobin could be directly associated with mitochondrial metabolism and integrity. We identified three different forms of neuroglobin in the retina, varying in their apparent molecular masses; all forms are abundant in mitochondrial fractions. This indicates that a significant fraction of the protein localizes within the organelle either in the matrix or in the matrix side of the inner membrane. Since neuroglobin was especially abundant in the ganglion cell layer, we transduced retinal ganglion cells with an anti-neuroglobin short hairpin RNA using in vivo electroporation. Neuroglobin knockdown leads to reduced activities of respiratory chain complexes I and III, degeneration of retinal ganglion cells, and impairment of visual function. The deleterious effect on cell survival was confirmed in primary retinal ganglion cells subjected to inhibition of neuroglobin expression. Hence, neuroglobin should be considered as a novel mitochondrial protein involved in respiratory chain function which is essential for retinal ganglion cell integrity.


Asunto(s)
Transporte de Electrón/fisiología , Globinas/metabolismo , Mitocondrias/metabolismo , Membranas Mitocondriales/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Células Ganglionares de la Retina/fisiología , Animales , Western Blotting , Células Cultivadas , Angiografía con Fluoresceína , Globinas/antagonistas & inhibidores , Globinas/genética , Masculino , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteínas del Tejido Nervioso/genética , Neuroglobina , Neuronas/citología , Nervio Óptico/citología , Nervio Óptico/metabolismo , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Ratas , Ratas Long-Evans , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
12.
Brain ; 135(Pt 1): 35-52, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22120150

RESUMEN

The Harlequin mutant mouse, characterized by loss of function of apoptosis-inducing factor, represents a reliable genetic model that resembles pathologies caused by human mitochondrial complex I deficiency. Therefore, we extensively characterized the retinal morphology and function of Harlequin mice during the course of neuronal cell death leading to blindness, with the aim of preventing optic atrophy. Retinas and optic nerves from these mice showed an isolated respiratory chain complex I defect correlated with retinal ganglion cell loss, optic atrophy, glial and microglial cell activation. All of these changes led to irreversible vision loss. In control mice, retinas AIF1 messenger RNA was 2.3-fold more abundant than AIF2, both messenger RNAs being sorted to the mitochondrial surface. In Harlequin mouse retinas, there was a 96% decrease of both AIF1 and AIF2 messenger RNA steady-state levels. We attained substantial and long-lasting protection of retinal ganglion cell and optic nerve integrity, the preservation of complex I function in optic nerves, as well as the prevention of glial and microglial responses after intravitreal administration of an AAV2 vector containing the full-length open reading frame and the 3' untranslated region of the AIF1 gene. Therefore, we demonstrate that gene therapy for mitochondrial diseases due to mutations in nuclear DNA can be achieved, so long as the 'therapeutic gene' permits the accurate cellular localization of the corresponding messenger RNA.


Asunto(s)
Factor Inductor de la Apoptosis/genética , Regulación hacia Abajo , Terapia Genética , Atrofia Óptica/terapia , Animales , Factor Inductor de la Apoptosis/metabolismo , Modelos Animales de Enfermedad , Ratones , Atrofia Óptica/genética , Atrofia Óptica/patología , Nervio Óptico/metabolismo , Nervio Óptico/patología , Retina/metabolismo , Retina/patología
13.
Antioxidants (Basel) ; 11(1)2022 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-35052663

RESUMEN

Interest in the structure, function, and evolutionary relations of circulating and intracellular globins dates back more than 60 years to the first determination of the three-dimensional structure of these proteins. Non-erythrocytic globins have been implicated in circulatory control through reactions that couple nitric oxide (NO) signaling with cellular oxygen availability and redox status. Small artery endothelial cells (ECs) express free α-globin, which causes vasoconstriction by degrading NO. This reaction converts reduced (Fe2+) α-globin to the oxidized (Fe3+) form, which is unstable, cytotoxic, and unable to degrade NO. Therefore, (Fe3+) α-globin must be stabilized and recycled to (Fe2+) α-globin to reinitiate the catalytic cycle. The molecular chaperone α-hemoglobin-stabilizing protein (AHSP) binds (Fe3+) α-globin to inhibit its degradation and facilitate its reduction. The mechanisms that reduce (Fe3+) α-globin in ECs are unknown, although endothelial nitric oxide synthase (eNOS) and cytochrome b5 reductase (CyB5R3) with cytochrome b5 type A (CyB5a) can reduce (Fe3+) α-globin in solution. Here, we examine the expression and cellular localization of eNOS, CyB5a, and CyB5R3 in mouse arterial ECs and show that α-globin can be reduced by either of two independent redox systems, CyB5R3/CyB5a and eNOS. Together, our findings provide new insights into the regulation of blood vessel contractility.

14.
Nat Commun ; 13(1): 6405, 2022 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-36302779

RESUMEN

Resistance artery vasodilation in response to hypoxia is essential for matching tissue oxygen and demand. In hypoxia, erythrocytic hemoglobin tetramers produce nitric oxide through nitrite reduction. We hypothesized that the alpha subunit of hemoglobin expressed in endothelium also facilitates nitrite reduction proximal to smooth muscle. Here, we create two mouse strains to test this: an endothelial-specific alpha globin knockout (EC Hba1Δ/Δ) and another with an alpha globin allele mutated to prevent alpha globin's inhibitory interaction with endothelial nitric oxide synthase (Hba1WT/Δ36-39). The EC Hba1Δ/Δ mice had significantly decreased exercise capacity and intracellular nitrite consumption in hypoxic conditions, an effect absent in Hba1WT/Δ36-39 mice. Hypoxia-induced vasodilation is significantly decreased in arteries from EC Hba1Δ/Δ, but not Hba1WT/Δ36-39 mice. Hypoxia also does not lower blood pressure in EC Hba1Δ/Δ mice. We conclude the presence of alpha globin in resistance artery endothelium acts as a nitrite reductase providing local nitric oxide in response to hypoxia.


Asunto(s)
Óxido Nítrico , Nitrito Reductasas , Ratones , Animales , Nitrito Reductasas/genética , Nitrito Reductasas/farmacología , Óxido Nítrico/farmacología , Nitritos , Globinas alfa/genética , Hipoxia , Endotelio Vascular , Hemoglobinas/genética , Vasodilatación/fisiología
15.
J Biol Chem ; 284(52): 36146-36159, 2009 Dec 25.
Artículo en Inglés | MEDLINE | ID: mdl-19864414

RESUMEN

Dos from Escherichia coli is a bacterial gas sensor protein comprising a heme-containing gas sensor domain and a phosphodiesterase catalytic domain. Using a combination of static light scattering and gel filtration experiments, we established that, as are many other sensor proteins, the full-length protein is dimeric. The full-length dimer (association constant <10 nm) is more stable than the dimeric heme domain (association constant approximately 1 mum), and the dimer interface presumably includes both sensor and catalytic domains. Ultrafast spectroscopic studies showed little influence of the catalytic domain on kinetic processes in the direct vicinity of the heme. By contrast, the properties of ligand (CO and O(2)) binding to the heme in the sensor domain, occurring on a microsecond to second time scale, were found to be influenced by (i) the presence of the catalytic domain, (ii) the dimerization state, and in dimers, (iii) the ligation state of the other subunit. These results imply allosteric interactions within dimers. Steady-state titrations demonstrated marked cooperativity in oxygen binding to both the full-length protein and the isolated heme domain, a feature not reported to date for any dimeric sensor protein. Analysis of a variety of time-resolved experiments showed that Met-95 plays a major role in the intradimer interactions. The intrinsic binding and dissociation rates of Met-95 to the heme were modulated approximately 10-fold by intradimer and sensor-catalytic domain interactions. Dimerization effects were also observed for cyanide binding to the ferric heme domains, suggesting a similar role for Met-95 in ferric proteins.


Asunto(s)
Monóxido de Carbono/química , Proteínas de Escherichia coli/química , Escherichia coli/enzimología , Hemo/química , Oxígeno/química , Hidrolasas Diéster Fosfóricas/química , Multimerización de Proteína/fisiología , Monóxido de Carbono/metabolismo , Proteínas de Escherichia coli/metabolismo , Hemo/metabolismo , Ligandos , Oxígeno/metabolismo , Hidrolasas Diéster Fosfóricas/metabolismo , Estructura Cuaternaria de Proteína/fisiología , Estructura Terciaria de Proteína/fisiología
16.
Acta Crystallogr D Biol Crystallogr ; 65(Pt 4): 388-92, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19307722

RESUMEN

The purification, crystallization and successful structure determination by molecular replacement of wild-type human brain neuroglobin at 1.8 A resolution is reported. The apparent space group was orthorhombic C222(1), but the real space group was monoclinic P2(1), which resulted from twinning. Indeed, the unit-cell parameters, a = 31.2, b = 139.1, c = 31.2 A, beta = 102 degrees , display a fortuitously close to c and twinning by the operator l, -k, h occurs. Twinning was not evident from the initial analysis of intensity distribution, but pseudo-merohedral twinning was revealed by the Padilla and Yeates test based on local intensity differences. A twinning fraction of 0.5 was determined in SHELXL, indicating a perfect hemihedrally twinned crystal. To date, this type of twinning has been reported in more than ten structures, which makes it quite a common case in proteins.


Asunto(s)
Globinas/química , Proteínas del Tejido Nervioso/química , Química Encefálica , Cristalización , Cristalografía por Rayos X , Cistina/química , Humanos , Modelos Moleculares , Neuroglobina , Oxígeno/metabolismo , Conformación Proteica
18.
Sci Transl Med ; 11(506)2019 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-31434755

RESUMEN

In ß-thalassemia, accumulated free α-globin forms intracellular precipitates that impair erythroid cell maturation and viability. Protein quality control systems mitigate ß-thalassemia pathophysiology by degrading toxic free α-globin, although the associated mechanisms are poorly understood. We show that loss of the autophagy-activating Unc-51-like kinase 1 (Ulk1) gene in ß-thalassemic mice reduces autophagic clearance of α-globin in red blood cell precursors and exacerbates disease phenotypes, whereas inactivation of the canonical autophagy-related 5 (Atg5) gene has relatively minor effects. Systemic treatment with the mTORC1 inhibitor rapamycin reduces α-globin precipitates and lessens pathologies in ß-thalassemic mice via an ULK1-dependent pathway. Similarly, rapamycin reduces free α-globin accumulation in erythroblasts derived from CD34+ cells of ß-thalassemic individuals. Our findings define a drug-regulatable pathway for ameliorating ß-thalassemia.


Asunto(s)
Homólogo de la Proteína 1 Relacionada con la Autofagia/metabolismo , Autofagia , Globinas alfa/metabolismo , Talasemia beta/enzimología , Talasemia beta/patología , Animales , Antígenos CD34/metabolismo , Autofagia/efectos de los fármacos , Proteína 5 Relacionada con la Autofagia/metabolismo , Progresión de la Enfermedad , Activación Enzimática/efectos de los fármacos , Eliminación de Gen , Hematopoyesis/efectos de los fármacos , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/metabolismo , Humanos , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Ratones , Fenotipo , Reticulocitos/efectos de los fármacos , Reticulocitos/metabolismo , Reticulocitos/ultraestructura , Sirolimus/farmacología
19.
Biochemistry ; 47(21): 5718-23, 2008 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-18454557

RESUMEN

A survey is presented of picosecond kinetics of heme-residue bond formation after photolysis of histidine, methionine, or cysteine, in a broad range of ferrous six-coordinate heme proteins. These include human neuroglobin, a bacterial heme-binding superoxide dismutase (SOD), plant cytochrome b 559, the insect nuclear receptor E75, horse heart cytochrome c and the heme domain of the bacterial sensor protein Dos. We demonstrate that the fastest and dominant phase of binding of amino acid residues to domed heme invariably takes place with a time constant in the narrow range of 5-7 ps. Remarkably, this is also the case in the heme-binding SOD, where the heme is solvent-exposed. We reason that this fast phase corresponds to barrierless formation of the heme-residue bond from a configuration close to the bound state. Only in proteins where functional ligand exchange occurs, additional slower rebinding takes place on the time scale of tens of picoseconds after residue dissociation. We propose that the presence of these slower phases reflects flexibility in the heme environment that allows external ligands (O2, CO, NO, . . .) to functionally replace the internal residue after thermal dissociation of the heme-residue bond.


Asunto(s)
Hemo/química , Hemoproteínas/química , Ligandos , Superóxido Dismutasa/química , Animales , Bioquímica/métodos , Citocromos c/química , Drosophila , Escherichia coli/metabolismo , Globinas/química , Haemophilus ducreyi/metabolismo , Caballos , Humanos , Proteínas del Tejido Nervioso/química , Neuroglobina , Oxígeno/química , Spinacia oleracea/metabolismo
20.
Methods Enzymol ; 436: 341-57, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18237642

RESUMEN

Neuroglobin and cytoglobin, members of the globin family, are present in vertebrate cells at very low concentrations. As the function of both proteins is still a matter of debate, it is very important to be able to produce and purify these proteins, and in general all members of the globin family, to homogeneity. For this purpose, this chapter describes the expression of neuro- and cytoglobin by E. coli and its preparative purification. These proteins are then used in crystallization experiments. Also an analytical purification strategy is discussed in detail.


Asunto(s)
Globinas/genética , Globinas/aislamiento & purificación , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/aislamiento & purificación , Resinas Acrílicas , Cromatografía en Agarosa , Cromatografía por Intercambio Iónico , Clonación Molecular , Cristalización , Citoglobina , Electroforesis en Gel de Poliacrilamida , Escherichia coli/genética , Expresión Génica , Vectores Genéticos , Globinas/química , Humanos , Mutación , Proteínas del Tejido Nervioso/química , Neuroglobina , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Sefarosa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA