Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
EMBO Rep ; 24(5): e55373, 2023 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-36943011

RESUMEN

Upon ex vivo culture, hematopoietic stem cells (HSCs) quickly lose potential and differentiate into progenitors. The identification of culture conditions that maintain the potential of HSCs ex vivo is therefore of high clinical interest. Here, we demonstrate that the potential of murine and human HSCs is maintained when cultivated for 2 days ex vivo at a pH of 6.9, in contrast to cultivation at the commonly used pH of 7.4. When cultivated at a pH of 6.9, HSCs remain smaller, less metabolically active, less proliferative and show enhanced reconstitution ability upon transplantation compared to HSC cultivated at pH 7.4. HSCs kept at pH 6.9 show an attenuated polyamine pathway. Pharmacological inhibition of the polyamine pathway in HSCs cultivated at pH 7.4 with DFMO mimics phenotypes and potential of HSCs cultivated at pH 6.9. Ex vivo exposure to a pH of 6.9 is therefore a positive regulator of HSC function by reducing polyamines. These findings might improve HSC short-term cultivation protocols for transplantation and gene therapy interventions.


Asunto(s)
Células Madre Hematopoyéticas , Humanos , Ratones , Animales , Células Madre Hematopoyéticas/metabolismo , Concentración de Iones de Hidrógeno
2.
PLoS Biol ; 16(9): e2003389, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30235201

RESUMEN

Hematopoietic stem cells (HSCs) balance self-renewal and differentiation to maintain homeostasis. With aging, the frequency of polar HSCs decreases. Cell polarity in HSCs is controlled by the activity of the small RhoGTPase cell division control protein 42 (Cdc42). Here we demonstrate-using a comprehensive set of paired daughter cell analyses that include single-cell 3D confocal imaging, single-cell transplants, single-cell RNA-seq, and single-cell transposase-accessible chromatin sequencing (ATAC-seq)-that the outcome of HSC divisions is strongly linked to the polarity status before mitosis, which is in turn determined by the level of the activity Cdc42 in stem cells. Aged apolar HSCs undergo preferentially self-renewing symmetric divisions, resulting in daughter stem cells with reduced regenerative capacity and lymphoid potential, while young polar HSCs undergo preferentially asymmetric divisions. Mathematical modeling in combination with experimental data implies a mechanistic role of the asymmetric sorting of Cdc42 in determining the potential of daughter cells via epigenetic mechanisms. Therefore, molecules that control HSC polarity might serve as modulators of the mode of stem cell division regulating the potential of daughter cells.


Asunto(s)
División Celular/genética , Senescencia Celular/genética , Epigénesis Genética , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Envejecimiento/metabolismo , Animales , División Celular Asimétrica/genética , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Agregación Celular , Linaje de la Célula/efectos de los fármacos , Polaridad Celular/efectos de los fármacos , Cromatina , Ratones Endogámicos C57BL , Transcriptoma/genética , Proteína Wnt-5a/farmacología , Proteína de Unión al GTP cdc42/metabolismo
3.
Nature ; 503(7476): 392-6, 2013 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-24141946

RESUMEN

Many organs with a high cell turnover (for example, skin, intestine and blood) are composed of short-lived cells that require continuous replenishment by somatic stem cells. Ageing results in the inability of these tissues to maintain homeostasis and it is believed that somatic stem-cell ageing is one underlying cause of tissue attrition with age or age-related diseases. Ageing of haematopoietic stem cells (HSCs) is associated with impaired haematopoiesis in the elderly. Despite a large amount of data describing the decline of HSC function on ageing, the molecular mechanisms of this process remain largely unknown, which precludes rational approaches to attenuate stem-cell ageing. Here we report an unexpected shift from canonical to non-canonical Wnt signalling in mice due to elevated expression of Wnt5a in aged HSCs, which causes stem-cell ageing. Wnt5a treatment of young HSCs induces ageing-associated stem-cell apolarity, reduction of regenerative capacity and an ageing-like myeloid-lymphoid differentiation skewing via activation of the small Rho GTPase Cdc42. Conversely, Wnt5a haploinsufficiency attenuates HSC ageing, whereas stem-cell-intrinsic reduction of Wnt5a expression results in functionally rejuvenated aged HSCs. Our data demonstrate a critical role for stem-cell-intrinsic non-canonical Wnt5a signalling in HSC ageing.


Asunto(s)
Senescencia Celular , Células Madre Hematopoyéticas/citología , Vía de Señalización Wnt , Animales , Diferenciación Celular , Polaridad Celular , Femenino , Haploinsuficiencia , Masculino , Ratones , Ratones Endogámicos C57BL , Fenotipo , Rejuvenecimiento , Proteínas Wnt/deficiencia , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , Proteína Wnt-5a , Proteína de Unión al GTP cdc42/metabolismo
5.
Life Sci Alliance ; 5(8)2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35487692

RESUMEN

Upon aging, the function of the intestinal epithelium declines with a concomitant increase in aging-related diseases. ISCs play an important role in this process. It is known that ISC clonal dynamics follow a neutral drift model. However, it is not clear whether the drift model is still valid in aged ISCs. Tracking of clonal dynamics by clonal tracing revealed that aged crypts drift into monoclonality substantially faster than young ones. However, ISC tracing experiments, in vivo and ex vivo, implied a similar clonal expansion ability of both young and aged ISCs. Single-cell RNA sequencing for 1,920 high Lgr5 ISCs from young and aged mice revealed increased heterogeneity among subgroups of aged ISCs. Genes associated with cell adhesion were down-regulated in aged ISCs. ISCs of aged mice indeed show weaker adhesion to the matrix. Simulations applying a single cell-based model of the small intestinal crypt demonstrated an accelerated clonal drift at reduced adhesion strength, implying a central role for reduced adhesion for affecting clonal dynamics upon aging.


Asunto(s)
Intestinos , Células Madre , Animales , Células Cultivadas , Íleon , Mucosa Intestinal/metabolismo , Ratones , Células Madre/metabolismo
6.
Leukemia ; 35(6): 1751-1762, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33077869

RESUMEN

Shwachman-Diamond syndrome (SDS) is a bone marrow failure (BMF) syndrome associated with an increased risk of myelodysplasia and leukemia. The molecular mechanisms of SDS are not fully understood. We report that primitive hematopoietic cells from SDS patients present with a reduced activity of the small RhoGTPase Cdc42 and concomitantly a reduced frequency of HSCs polar for polarity proteins. The level of apolarity of SDS HSCs correlated with the magnitude of HSC depletion in SDS patients. Importantly, exogenously provided Wnt5a or GDF11 that elevates the activity of Cdc42 restored polarity in SDS HSCs and increased the number of HSCs in SDS patient samples in surrogate ex vivo assays. Single cell level RNA-Seq analyses of SDS HSCs and daughter cells demonstrated that SDS HSC treated with GDF11 are transcriptionally more similar to control than to SDS HSCs. Treatment with GDF11 reverted pathways in SDS HSCs associated with rRNA processing and ribosome function, but also viral infection and immune function, p53-dependent DNA damage, spindle checkpoints, and metabolism, further implying a role of these pathways in HSC failure in SDS. Our data suggest that HSC failure in SDS is driven at least in part by low Cdc42 activity in SDS HSCs. Our data thus identify novel rationale approaches to attenuate HSCs failure in SDS.


Asunto(s)
Células de la Médula Ósea/citología , Polaridad Celular , Células Madre Hematopoyéticas/citología , Síndrome de Shwachman-Diamond/prevención & control , Proteína de Unión al GTP cdc42/metabolismo , Células de la Médula Ósea/metabolismo , Proteínas Morfogenéticas Óseas/química , Proteínas Morfogenéticas Óseas/metabolismo , Células Cultivadas , Factores de Diferenciación de Crecimiento/química , Factores de Diferenciación de Crecimiento/metabolismo , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/patología , Humanos , Pronóstico , Síndrome de Shwachman-Diamond/etiología , Síndrome de Shwachman-Diamond/metabolismo , Síndrome de Shwachman-Diamond/patología , Proteína Wnt-5a/química , Proteína Wnt-5a/metabolismo , Proteína de Unión al GTP cdc42/química
7.
J Clin Invest ; 117(11): 3283-95, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17960249

RESUMEN

The molecular pathogenesis of the myeloid leukemias that frequently occur in patients with Fanconi anemia (FA) is not well defined. Hematopoietic stem cells bearing inactivating mutations of FA complementation group C (FANCC) are genetically unstable and hypersensitive to apoptotic cytokine cues including IFN-gamma and TNF-alpha, but neoplastic stem cell clones that arise frequently in vivo are resistant to these cytokines. Reasoning that the combination of genetic instability and cytokine hypersensitivity might create an environment supporting the emergence of leukemic stem cells, we tested the leukemia-promoting effects of TNF-alpha in murine stem cells. TNF-alpha exposure initially profoundly inhibited the growth of Fancc-/- stem cells. However, longer-term exposure of these cells promoted the outgrowth of cytogenetically abnormal clones that, upon transplantation into congenic WT mice, led to acute myelogenous leukemia. TNF-alpha induced ROS-dependent genetic instability in Fancc-/- but not in WT cells. The leukemic clones were TNF-alpha resistant but retained their characteristic hypersensitivity to mitomycin C and exhibited high levels of chromosomal instability. Expression of FANCC cDNA in Fancc-/- stem cells protected them from TNF-alpha-induced clonal evolution. We conclude that TNF-alpha exposure creates an environment in which somatically mutated preleukemic stem cell clones are selected and from which unaltered TNF-alpha-hypersensitive Fancc-/- stem cells are purged.


Asunto(s)
Proliferación Celular , Proteína del Grupo de Complementación C de la Anemia de Fanconi/inmunología , Anemia de Fanconi/inmunología , Células Madre Hematopoyéticas/fisiología , Factor de Necrosis Tumoral alfa/inmunología , Animales , Apoptosis , Diferenciación Celular/fisiología , Aberraciones Cromosómicas , Anemia de Fanconi/genética , Proteína del Grupo de Complementación C de la Anemia de Fanconi/genética , Prueba de Complementación Genética , Células Madre Hematopoyéticas/citología , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Ratones , Ratones Noqueados , FN-kappa B/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Trasplante de Células Madre , Tasa de Supervivencia
8.
Aging Cell ; 19(9): e13208, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32755011

RESUMEN

Cdc42 is a small RhoGTPase regulating multiple functions in eukaryotic cells. The activity of Cdc42 is significantly elevated in several tissues of aged mice, while the Cdc42 gain-of-activity mouse model presents with a premature aging-like phenotype and with decreased lifespan. These data suggest a causal connection between elevated activity of Cdc42, aging, and reduced lifespan. Here, we demonstrate that systemic treatment of aged (75-week-old) female C57BL/6 mice with a Cdc42 activity-specific inhibitor (CASIN) for 4 consecutive days significantly extends average and maximum lifespan. Moreover, aged CASIN-treated animals displayed a youthful level of the aging-associated cytokines IL-1ß, IL-1α, and INFγ in serum and a significantly younger epigenetic clock as based on DNA methylation levels in blood cells. Overall, our data show that systemic administration of CASIN to reduce Cdc42 activity in aged mice extends murine lifespan.


Asunto(s)
Citocinas/metabolismo , Proteína de Unión al GTP cdc42/genética , Envejecimiento , Animales , Proteínas de Drosophila , Femenino , Cadenas alfa de Integrinas , Longevidad , Ratones , Ratones Endogámicos C57BL
9.
Methods Mol Biol ; 506: 423-36, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19110642

RESUMEN

Maintaining the stability of the genome is critical to cell survival and normal cell growth. Genetic modification of hematopoietic cells might bear an inherent increased risk for the accumulation of DNA mutations. It frequently requires cultivation of the cells under super-physiological oxygen levels, which can result in increased oxidative damage, as well as under super-physiological concentrations of cytokines, which might interfere with DNA-damage checkpoint activation and by this means might result in an increased mutational load. We describe here a protocol for monitoring the frequency of DNA mutations in bone marrow cells post transduction or upon selection either in vitro or in vivo based on the lacZ-plasmid (pUR288) transgenic mouse (small blue mouse) mutation indicator strain.


Asunto(s)
Genómica , Células Madre Hematopoyéticas/metabolismo , Mutación , Animales , Secuencia de Bases , Cartilla de ADN , Electroporación , Técnicas de Transferencia de Gen , Células Madre Hematopoyéticas/citología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Reacción en Cadena de la Polimerasa
10.
Cell Rep ; 18(11): 2608-2621, 2017 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-28297666

RESUMEN

Although intestinal homeostasis is maintained by intestinal stem cells (ISCs), regeneration is impaired upon aging. Here, we first uncover changes in intestinal architecture, cell number, and cell composition upon aging. Second, we identify a decline in the regenerative capacity of ISCs upon aging because of a decline in canonical Wnt signaling in ISCs. Changes in expression of Wnts are found in stem cells themselves and in their niche, including Paneth cells and mesenchyme. Third, reactivating canonical Wnt signaling enhances the function of both murine and human ISCs and, thus, ameliorates aging-associated phenotypes of ISCs in an organoid assay. Our data demonstrate a role for impaired Wnt signaling in physiological aging of ISCs and further identify potential therapeutic avenues to improve ISC regenerative potential upon aging.


Asunto(s)
Senescencia Celular , Intestino Delgado/citología , Células Madre/citología , Células Madre/metabolismo , Vía de Señalización Wnt , Animales , Biomarcadores/metabolismo , Recuento de Células , Proliferación Celular , Femenino , Ratones , Organoides/citología , Regeneración , Nicho de Células Madre
11.
Nat Med ; 18(7): 1123-9, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22729286

RESUMEN

Tissue damage induced by ionizing radiation in the hematopoietic and gastrointestinal systems is the major cause of lethality in radiological emergency scenarios and underlies some deleterious side effects in patients undergoing radiation therapy. The identification of target-specific interventions that confer radiomitigating activity is an unmet challenge. Here we identify the thrombomodulin (Thbd)-activated protein C (aPC) pathway as a new mechanism for the mitigation of total body irradiation (TBI)-induced mortality. Although the effects of the endogenous Thbd-aPC pathway were largely confined to the local microenvironment of Thbd-expressing cells, systemic administration of soluble Thbd or aPC could reproduce and augment the radioprotective effect of the endogenous Thbd-aPC pathway. Therapeutic administration of recombinant, soluble Thbd or aPC to lethally irradiated wild-type mice resulted in an accelerated recovery of hematopoietic progenitor activity in bone marrow and a mitigation of lethal TBI. Starting infusion of aPC as late as 24 h after exposure to radiation was sufficient to mitigate radiation-induced mortality in these mice. These findings suggest that pharmacologic augmentation of the activity of the Thbd-aPC pathway by recombinant Thbd or aPC might offer a rational approach to the mitigation of tissue injury and lethality caused by ionizing radiation.


Asunto(s)
Proteína C/antagonistas & inhibidores , Traumatismos por Radiación/prevención & control , Proteínas Recombinantes/farmacología , Transducción de Señal/efectos de los fármacos , Trombomodulina/antagonistas & inhibidores , Animales , Regulación de la Expresión Génica/efectos de los fármacos , Proteínas Fluorescentes Verdes/metabolismo , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/patología , Humanos , Ratones , Ratones Endogámicos C57BL , Proteína C/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Traumatismos por Radiación/genética , Traumatismos por Radiación/patología , Tolerancia a Radiación/efectos de los fármacos , Tolerancia a Radiación/genética , Receptores de Trombina , Análisis de Supervivencia , Trombomodulina/genética , Trombomodulina/metabolismo , Irradiación Corporal Total
12.
Nat Med ; 16(10): 1141-6, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20871610

RESUMEN

Mobilization of hematopoietic stem and progenitor cells (HSPCs) from bone marrow into peripheral blood by the cytokine granulocyte colony-stimulating factor (G-CSF) has become the preferred source of HSPCs for stem cell transplants. However, G-CSF fails to mobilize sufficient numbers of stem cells in up to 10% of donors, precluding autologous transplantation in those donors or substantially delaying transplant recovery time. Consequently, new regimens are needed to increase the number of stem cells in peripheral blood upon mobilization. Using a forward genetic approach in mice, we mapped the gene encoding the epidermal growth factor receptor (Egfr) to a genetic region modifying G-CSF-mediated HSPC mobilization. Amounts of EGFR in HSPCs inversely correlated with the cells' ability to be mobilized by G-CSF, implying a negative role for EGFR signaling in mobilization. In combination with G-CSF treatment, genetic reduction of EGFR activity in HSPCs (in waved-2 mutant mice) or treatment with the EGFR inhibitor erlotinib increased mobilization. Increased mobilization due to suppression of EGFR activity correlated with reduced activity of cell division control protein-42 (Cdc42), and genetic Cdc42 deficiency in vivo also enhanced G-CSF-induced mobilization. Our findings reveal a previously unknown signaling pathway regulating stem cell mobilization and provide a new pharmacological approach for improving HSPC mobilization and thereby transplantation outcomes.


Asunto(s)
Receptores ErbB/antagonistas & inhibidores , Factor Estimulante de Colonias de Granulocitos/farmacología , Movilización de Célula Madre Hematopoyética , Transducción de Señal , Animales , Factor de Crecimiento Epidérmico/farmacología , Receptores ErbB/fisiología , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Proteína de Unión al GTP cdc42/fisiología
13.
Blood ; 108(7): 2190-7, 2006 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-16741255

RESUMEN

Hematopoietic stem and progenitor cells (HSPCs) are located in the bone marrow in close association with a highly organized 3-dimensional structure formed by stroma cells, referred to as the niche. Mobilization of HSPCs from bone marrow to peripheral blood in response to granulocyte colony-stimulating factor (G-CSF) requires de-adhesion of HSPCs from the niche. The influence of aging of HSPCs on cell-stroma interactions has not been determined in detail. Using a mouse model of G-CSF-induced mobilization, we demonstrated that the ability to mobilize hematopoietic stem cells is approximately 5-fold greater in aged mice. Competitive mobilization experiments confirmed that enhanced mobilization ability was intrinsic to the stem cell. Enhanced mobilization efficiency of primitive hematopoietic cells from aged mice correlated with reduced adhesion of hematopoietic progenitor cells to stroma and with elevated levels of GTP-bound Cdc42. These results might indicate that stroma-stem cell interactions are dynamic over a lifetime and result in physiologically relevant changes in the biology of primitive hematopoietic cells with age.


Asunto(s)
Movilización de Célula Madre Hematopoyética , Células Madre Hematopoyéticas/citología , Envejecimiento , Animales , Adhesión Celular , Movimiento Celular , Factor Estimulante de Colonias de Granulocitos/metabolismo , Ratones , Ratones Endogámicos C57BL , Modelos Biológicos , Células Madre/citología , Proteína de Unión al GTP cdc42/metabolismo , Proteínas de Unión al GTP rho/metabolismo
14.
Am J Physiol Heart Circ Physiol ; 283(3): H958-65, 2002 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12181124

RESUMEN

In this study we evaluated the contractile characteristics of sarco(endo)plasmic reticulum Ca(2+)-ATPase (SERCA)1a-expressing hearts ex vivo and in vivo and in particular their response to beta-adrenergic stimulation. Analysis of isolated, work-performing hearts revealed that transgenic (TG) hearts develop much higher maximal rates of contraction and relaxation than wild-type (WT) hearts. Addition of isoproterenol only moderately increased the maximal rate of relaxation (+20%) but did not increase contractility or decrease relaxation time in TG hearts. Perfusion with varied buffer Ca(2+) concentrations indicated an altered dose response to Ca(2+). In vivo TG hearts displayed fairly higher maximal rates of contraction (+ 25%) but unchanged relaxation parameters and a blunted but significant response to dobutamine. Our study also shows that the phospholamban (PLB) level was decreased (-40%) and its phosphorylation status modified in TG hearts. This study clearly demonstrates that increases in SERCA protein level alter the beta-adrenergic response and affect the phosphorylation of PLB. Interestingly, the overall cardiac function in the live animal is only slightly enhanced, suggesting that (neuro)hormonal regulations may play an important role in controlling in vivo heart function.


Asunto(s)
Agonistas Adrenérgicos beta/farmacología , ATPasas Transportadoras de Calcio/metabolismo , Isoproterenol/farmacología , Contracción Miocárdica/efectos de los fármacos , Miocardio/metabolismo , Animales , Calcio/farmacología , Proteínas de Unión al Calcio/metabolismo , ATPasas Transportadoras de Calcio/genética , Dobutamina/farmacología , Relación Dosis-Respuesta a Droga , Femenino , Técnicas In Vitro , Masculino , Ratones , Ratones Transgénicos , Contracción Miocárdica/fisiología , Fosforilación , Receptores Adrenérgicos beta/metabolismo , Retículo Sarcoplasmático/enzimología , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico
15.
Am J Physiol Heart Circ Physiol ; 286(3): H1146-53, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-14630633

RESUMEN

We recently developed a mouse model with a single functional allele of Serca2 (Serca2+/-) that shows impaired cardiac contractility and relaxation without overt heart disease. The goal of this study was to test the hypothesis that chronic reduction in sarco(endo)plasmic reticulum Ca(2+)-ATPase (SERCA)2 levels in combination with an increased hemodynamic load will result in an accelerated pathway to heart failure. Age-matched wild-type and Serca2+/- mice were subjected to 10 wk of pressure overload via transverse aortic coarctation surgery. Cardiac hypertrophy and heart failure were assessed by echocardiography, gravimetry/histology, hemodynamics, and Western blotting analyses. Our results showed that approximately 64% of coarcted Serca2+/- mice were in heart failure compared with 0% of coarcted wild-type mice (P < 0.05). Overall, morbidity and mortality were greatly increased in Serca2+/- mice under pressure overload. Echocardiography assessment revealed a significant increase in left ventricular (LV) mass, and LV hypertrophy in coarcted Serca2+/- mice converted from a concentric to an eccentric pattern, similar to that seen in human heart failure. Coarcted Serca2+/- mice had decreased contractile/systolic and relaxation/diastolic performance and/or function compared with coarcted wild-type mice (P < 0.05), despite a similar duration and degree of pressure overload. SERCA2a protein levels were significantly reduced (>50%) in coarcted Serca2+/- mice compared with noncoarcted and coarcted wild-type mice. Our findings suggest that reduction in SERCA2 levels in combination with an increased hemodynamic load results in an accelerated pathway to heart failure.


Asunto(s)
ATPasas Transportadoras de Calcio/genética , ATPasas Transportadoras de Calcio/metabolismo , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/fisiopatología , Animales , Calcio/metabolismo , Cateterismo Cardíaco , Diástole , Ecocardiografía , Femenino , Insuficiencia Cardíaca/diagnóstico por imagen , Hipertrofia Ventricular Izquierda/diagnóstico por imagen , Hipertrofia Ventricular Izquierda/metabolismo , Hipertrofia Ventricular Izquierda/fisiopatología , Masculino , Ratones , Ratones Noqueados , Fenotipo , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico , Sístole
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA