Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Nat Aging ; 2024 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-38689133

RESUMEN

By 2030, individuals 65 years of age or older will make up approximately 20% of the world's population1. Older individuals are at the highest risk for mortality from infections, largely due to the pro-inflammatory, dysfunctional immune response, which is collectively known as immunosenescence2. During aging, CD8+ T cells acquire an exhausted phenotype, including increased expression of inhibitory receptors, such as programmed cell death 1 (PD1), a decline in effector function and elevated expression of inflammatory factors3-7. PD1 reduces T cell receptor activity via SHP2-dependent dephosphorylation of multiple pathways; accordingly, inhibiting PD1 activity through monoclonal antibodies increases CD8+ T cell effector response in young mice8-11. Attempts to improve CD8+ T cell responses by blocking inhibitory receptors are attractive; however, they can lead to adverse immune events due to overamplification of T cell receptor signaling and T cell activation12,13. Here we investigated the effect of monoclonal anti-PD1 immunotherapy during normal microbial experience, otherwise known as exposure to dirty mice, to determine whether it either improves exhausted CD8+ T cell responses in old mice or leads to a heightened inflammatory response and increased mortality.

2.
Cell Rep ; 43(3): 113967, 2024 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-38492219

RESUMEN

Non-canonical lipolysis induced by inflammatory cytokines or Toll-like receptor ligands is required for the regulation of inflammation during endotoxemia and sepsis. Canonical lipolysis induced by catecholamines declines during aging due to factors including an expansion of lymphocytes, pro-inflammatory macrophage polarization, and an increase in chronic low-grade inflammation; however, the extent to which the non-canonical pathway of lipolysis is active and impacted by immune cells during aging remains unclear. Therefore, we aimed to define the extent to which immune cells from old mice influence non-canonical lipolysis during sepsis. We identified age-associated impairments of non-canonical lipolysis and an accumulation of dysfunctional B1 B cells in the visceral white adipose tissue (vWAT) of old mice. Lifelong deficiency of B cells results in restored non-canonical lipolysis and reductions in pro-inflammatory macrophage populations. Our study suggests that targeting the B cell-macrophage signaling axis may resolve metabolic dysfunction in aged vWAT and attenuate septic severity in older individuals.


Asunto(s)
Lipólisis , Sepsis , Animales , Ratones , Tejido Adiposo/metabolismo , Inflamación/metabolismo , Macrófagos/metabolismo , Sepsis/metabolismo , Ratones Endogámicos C57BL
3.
Immunometabolism (Cobham) ; 6(1): e00038, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38249577

RESUMEN

Fatty acid oxidation (FAO), primarily known as ß-oxidation, plays a crucial role in breaking down fatty acids within mitochondria and peroxisomes to produce cellular energy and preventing metabolic dysfunction. Myeloid cells, including macrophages, microglia, and monocytes, rely on FAO to perform essential cellular functions and uphold tissue homeostasis. As individuals age, these cells show signs of inflammaging, a condition that includes a chronic onset of low-grade inflammation and a decline in metabolic function. These lead to changes in fatty acid metabolism and a decline in FAO pathways. Recent studies have shed light on metabolic shifts occurring in macrophages and monocytes during aging, correlating with an altered tissue environment and the onset of inflammaging. This review aims to provide insights into the connection of inflammatory pathways and altered FAO in macrophages and monocytes from older organisms. We describe a model in which there is an extended activation of receptor for advanced glycation end products, nuclear factor-κB (NF-κB) and the nod-like receptor family pyrin domain containing 3 inflammasome within macrophages and monocytes. This leads to an increased level of glycolysis, and also promotes pro-inflammatory cytokine production and signaling. As a result, FAO-related enzymes such as 5' AMP-activated protein kinase and peroxisome proliferator-activated receptor-α are reduced, adding to the escalation of inflammation, accumulation of lipids, and heightened cellular stress. We examine the existing body of literature focused on changes in FAO signaling within macrophages and monocytes and their contribution to the process of inflammaging.

4.
Curr Diab Rep ; 22(11): 537-548, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36239841

RESUMEN

PURPOSE OF REVIEW: Obesity has increased worldwide recently and represents a major global health challenge. This review focuses on the obesity-associated cellular senescence in various organs and the role of these senescent cells (SnCs) in driving complications associated with obesity. Also, the ability to target SnCs pharmacologically with drugs termed senotherapeutics as a therapy for these complications is discussed. RECENT FINDINGS: Several studies have shown a positive correlation between obesity and SnC burden in organs such as adipose tissue, liver, and pancreatic-ß-cells. These SnCs produce several secretory factors which affect other cells and tissues in a paracrine manner resulting in organ dysfunction. The accumulation of SnCs in adipocytes affects their lipid storage and impairs adipogenesis. The inflammatory senescence-associated secretory phenotype (SASP) of SnCs downregulates the antioxidant capacity and mitochondrial function in tissues. Senescent hepatocytes cannot oxidize fatty acids, which leads to lipid deposition and senescence in ß-cells decrease function. These and other adverse effects of SnCs contribute to insulin resistance and type-2 diabetes. The reduction in the SnC burden genetically or pharmacologically improves the complications associated with obesity. The accumulation of SnCs with age and disease accelerates aging. Obesity is a key driver of SnC accumulation, and the complications associated with obesity can be controlled by reducing the SnC burden. Thus, senotherapeutic drugs have the potential to be an effective therapeutic option.


Asunto(s)
Antioxidantes , Senoterapéuticos , Humanos , Senescencia Celular/genética , Obesidad/complicaciones , Obesidad/tratamiento farmacológico , Ácidos Grasos , Lípidos
5.
Artículo en Inglés | MEDLINE | ID: mdl-35131468

RESUMEN

Adipose tissue is a critical organ for nutrient sensing, energy storage and maintaining metabolic health. The failure of adipose tissue homeostasis leads to metabolic disease that is seen during obesity or aging. Local metabolic processes are coordinated by interacting microenvironments that make up the complexity and heterogeneity of the adipose tissue. Catecholamine-induced lipolysis, a critical pathway in adipocytes that drives the release of stored triglyceride as free fatty acid after stimulation, is impaired during aging. The impairment of this pathway is associated with a failure to maintain a healthy body weight, core body-temperature during cold stress or mount an immune response. Along with impairments in aged adipocytes, aging is associated with an accumulation of inflammation, immune cell activation, and increased dysfunction in the nervous and lymphatic systems within the adipose tissue. Together these microenvironments support the initiation of stimulated lipolysis and the transport of free fatty acid under conditions of metabolic homeostasis. However, during aging, the defects in these cellular systems result in a reduction in ability to stimulate lipolysis. This review will focus on how the immune, nervous and lymphatic systems interact during tissue homeostasis, review areas that are impaired with aging and discuss areas of research that are currently unclear.


Asunto(s)
Ácidos Grasos no Esterificados , Lipólisis , Adipocitos/metabolismo , Tejido Adiposo/metabolismo , Ácidos Grasos no Esterificados/metabolismo
6.
Diabetes ; 71(1): 23-30, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34995348

RESUMEN

Age-related immunosenescence, defined as an increase in inflammaging and the decline of the immune system, leads to tissue dysfunction and increased risk for metabolic disease. The elderly population is expanding, leading to a heightened need for therapeutics to improve health span. With age, many alterations of the immune system are observed, including shifts in the tissue-resident immune cells, increased expression of inflammatory factors, and the accumulation of senescent cells, all of which are responsible for a chronic inflammatory loop. Adipose tissue and the immune cell activation within are of particular interest for their well-known roles in metabolic disease. Recent literature reveals that adipose tissue is an organ in which signs of initial aging occur, including immune cell activation. Aged adipose tissue reveals changes in many innate and adaptive immune cell subsets, revealing a complex interaction that contributes to inflammation, increased senescence, impaired catecholamine-induced lipolysis, and impaired insulin sensitivity. Here, we will describe current knowledge surrounding age-related changes in immune cells while relating those findings to recent discoveries regarding immune cells in aged adipose tissue.


Asunto(s)
Tejido Adiposo/patología , Microambiente Celular/fisiología , Senescencia Celular/fisiología , Inflamación/patología , Leucocitos/fisiología , Tejido Adiposo/metabolismo , Anciano , Envejecimiento/sangre , Envejecimiento/inmunología , Envejecimiento/metabolismo , Animales , Microambiente Celular/inmunología , Femenino , Humanos , Inmunosenescencia/fisiología , Inflamación/metabolismo , Macrófagos/fisiología , Masculino , Ratones
7.
Cell Metab ; 33(11): 2277-2287.e5, 2021 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-34473956

RESUMEN

Aging impairs the integrated immunometabolic responses, which have evolved to maintain core body temperature in homeotherms to survive cold stress, infections, and dietary restriction. Adipose tissue inflammation regulates the thermogenic stress response, but how adipose tissue-resident cells instigate thermogenic failure in the aged are unknown. Here, we define alterations in the adipose-resident immune system and identify that type 2 innate lymphoid cells (ILC2s) are lost in aging. Restoration of ILC2 numbers in aged mice to levels seen in adults through IL-33 supplementation failed to rescue old mice from metabolic impairment and increased cold-induced lethality. Transcriptomic analyses revealed intrinsic defects in aged ILC2, and adoptive transfer of adult ILC2s are sufficient to protect old mice against cold. Thus, the functional defects in adipose ILC2s during aging drive thermogenic failure.


Asunto(s)
Inmunidad Innata , Interleucina-33 , Tejido Adiposo , Envejecimiento , Animales , Pulmón , Linfocitos , Ratones , Ratones Endogámicos C57BL
8.
Science ; 373(6552)2021 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-34103349

RESUMEN

The COVID-19 pandemic has revealed the pronounced vulnerability of the elderly and chronically ill to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-induced morbidity and mortality. Cellular senescence contributes to inflammation, multiple chronic diseases, and age-related dysfunction, but effects on responses to viral infection are unclear. Here, we demonstrate that senescent cells (SnCs) become hyper-inflammatory in response to pathogen-associated molecular patterns (PAMPs), including SARS-CoV-2 spike protein-1, increasing expression of viral entry proteins and reducing antiviral gene expression in non-SnCs through a paracrine mechanism. Old mice acutely infected with pathogens that included a SARS-CoV-2-related mouse ß-coronavirus experienced increased senescence and inflammation, with nearly 100% mortality. Targeting SnCs by using senolytic drugs before or after pathogen exposure significantly reduced mortality, cellular senescence, and inflammatory markers and increased antiviral antibodies. Thus, reducing the SnC burden in diseased or aged individuals should enhance resilience and reduce mortality after viral infection, including that of SARS-CoV-2.


Asunto(s)
Envejecimiento , Senescencia Celular/efectos de los fármacos , Infecciones por Coronavirus/mortalidad , Flavonoles/uso terapéutico , Moléculas de Patrón Molecular Asociado a Patógenos/metabolismo , Glicoproteína de la Espiga del Coronavirus/metabolismo , Animales , COVID-19/inmunología , COVID-19/mortalidad , Línea Celular , Infecciones por Coronavirus/inmunología , Dasatinib/farmacología , Dasatinib/uso terapéutico , Femenino , Flavonoles/farmacología , Regulación de la Expresión Génica , Humanos , Lipopolisacáridos , Masculino , Ratones , Ratones Endogámicos C57BL , Virus de la Hepatitis Murina/inmunología , Quercetina/farmacología , Quercetina/uso terapéutico , Receptores de Coronavirus/genética , Receptores de Coronavirus/metabolismo , Organismos Libres de Patógenos Específicos , Tratamiento Farmacológico de COVID-19
9.
Curr Opin Pharmacol ; 57: 107-116, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33684669

RESUMEN

Aging is associated with the highest risk for morbidity and mortality to chronic or metabolic diseases, which are present in 50% of the elderly. Improving metabolic and immune function of the elderly would improve quality of life and reduce the risk for all other diseases. Tissue-resident macrophages and the NLRP3 inflammasome are established drivers of inflammaging and metabolic dysfunction. Energy-sensing signaling pathways connect sterile and metabolic inflammation with cellular senescence and tissue dysfunction. We discuss recent advances in the immunometabolism field. Common themes revealed by recent publications include the alterations in metabolic signaling (SIRTUIN, AMPK, or mTOR pathways) in aged immune cells, the impact of senescence on inflammaging and tissue dysfunction, and the age-related changes in metabolic tissues, especially adipose tissue, as an immunological organ. Promising gerotherapeutics are candidates to broadly target nutrient and energy sensing, inflammatory and senescence pathways, and have potential to improve healthspan and treat age-related diseases.


Asunto(s)
Inmunosenescencia , Tejido Adiposo , Anciano , Envejecimiento , Humanos , Inflamasomas , Inflamación , Calidad de Vida
10.
Immunity ; 54(1): 6-8, 2021 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-33440137

RESUMEN

Inflammaging is associated with chronic diseases, but tissue-specific changes in the immune system remain unknown. In this issue of Immunity, Mogilenko et al. use single-cell RNA sequencing, flow cytometry, and mass cytometry to describe age-related differences, including the accumulation of age-associated T cells that contribute to inflammaging.


Asunto(s)
Envejecimiento , Inflamación , Enfermedad Crónica , Granzimas , Humanos , Linfocitos T
11.
Cell Metab ; 30(6): 1024-1039.e6, 2019 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-31735593

RESUMEN

During aging, visceral adiposity is often associated with alterations in adipose tissue (AT) leukocytes, inflammation, and metabolic dysfunction. However, the contribution of AT B cells in immunometabolism during aging is unexplored. Here, we show that aging is associated with an expansion of a unique population of resident non-senescent aged adipose B cells (AABs) found in fat-associated lymphoid clusters (FALCs). AABs are transcriptionally distinct from splenic age-associated B cells (ABCs) and show greater expansion in female mice. Functionally, whole-body B cell depletion restores proper lipolysis and core body temperature maintenance during cold stress. Mechanistically, the age-induced FALC formation, AAB, and splenic ABC expansion is dependent on the Nlrp3 inflammasome. Furthermore, AABs express IL-1R, and inhibition of IL-1 signaling reduces their proliferation and increases lipolysis in aging. These data reveal that inhibiting Nlrp3-dependent B cell accumulation can be targeted to reverse metabolic impairment in aging AT.


Asunto(s)
Tejido Adiposo , Envejecimiento/metabolismo , Linfocitos B , Homeostasis , Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/fisiología , Tejido Adiposo/metabolismo , Tejido Adiposo/patología , Animales , Linfocitos B/metabolismo , Linfocitos B/patología , Regulación de la Temperatura Corporal , Respuesta al Choque por Frío , Femenino , Interleucina-18/metabolismo , Interleucina-1beta/metabolismo , Lipólisis , Masculino , Ratones , Receptores de Interleucina-1/metabolismo
12.
Nature ; 550(7674): 119-123, 2017 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-28953873

RESUMEN

Catecholamine-induced lipolysis, the first step in the generation of energy substrates by the hydrolysis of triglycerides, declines with age. The defect in the mobilization of free fatty acids in the elderly is accompanied by increased visceral adiposity, lower exercise capacity, failure to maintain core body temperature during cold stress, and reduced ability to survive starvation. Although catecholamine signalling in adipocytes is normal in the elderly, how lipolysis is impaired in ageing remains unknown. Here we show that adipose tissue macrophages regulate the age-related reduction in adipocyte lipolysis in mice by lowering the bioavailability of noradrenaline. Unexpectedly, unbiased whole-transcriptome analyses of adipose macrophages revealed that ageing upregulates genes that control catecholamine degradation in an NLRP3 inflammasome-dependent manner. Deletion of NLRP3 in ageing restored catecholamine-induced lipolysis by downregulating growth differentiation factor-3 (GDF3) and monoamine oxidase A (MAOA) that is known to degrade noradrenaline. Consistent with this, deletion of GDF3 in inflammasome-activated macrophages improved lipolysis by decreasing levels of MAOA and caspase-1. Furthermore, inhibition of MAOA reversed the age-related reduction in noradrenaline concentration in adipose tissue, and restored lipolysis with increased levels of the key lipolytic enzymes adipose triglyceride lipase (ATGL) and hormone sensitive lipase (HSL). Our study reveals that targeting neuro-immunometabolic signalling between the sympathetic nervous system and macrophages may offer new approaches to mitigate chronic inflammation-induced metabolic impairment and functional decline.


Asunto(s)
Adipocitos/metabolismo , Tejido Adiposo/metabolismo , Envejecimiento/metabolismo , Catecolaminas/metabolismo , Inflamasomas/metabolismo , Lipólisis , Macrófagos/metabolismo , Tejido Adiposo/citología , Tejido Adiposo/efectos de los fármacos , Envejecimiento/efectos de los fármacos , Envejecimiento/genética , Animales , Caspasa 1/metabolismo , Catecolaminas/farmacología , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Factor 3 de Diferenciación de Crecimiento/deficiencia , Factor 3 de Diferenciación de Crecimiento/genética , Factor 3 de Diferenciación de Crecimiento/metabolismo , Lipasa/metabolismo , Lipólisis/efectos de los fármacos , Lipólisis/genética , Ratones , Monoaminooxidasa/metabolismo , Inhibidores de la Monoaminooxidasa/farmacología , Proteína con Dominio Pirina 3 de la Familia NLR/deficiencia , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Norepinefrina/metabolismo , Esterol Esterasa/metabolismo
13.
Cell Rep ; 19(2): 225-234, 2017 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-28402847

RESUMEN

In concert with their phagocytic activity, macrophages are thought to regulate the host immunometabolic responses primarily via their ability to produce specific cytokines and metabolites. Here, we show that IL-4-differentiated, M2-like macrophages secrete IGF1, a hormone previously thought to be exclusively produced from liver. Ablation of IGF1 receptors from myeloid cells reduced phagocytosis, increased macrophages in adipose tissue, elevated adiposity, lowered energy expenditure, and led to insulin resistance in mice fed a high-fat diet. The investigation of adipose macrophage phenotype in obese myeloid IGF1R knockout (MIKO) mice revealed a reduction in transcripts associated with M2-like macrophage activation. Furthermore, the MIKO mice infected with helminth Nippostrongylus brasiliensis displayed delayed resolution from infection with normal insulin sensitivity. Surprisingly, cold challenge did not trigger an overt M2-like state and failed to induce tyrosine hydroxylase expression in adipose tissue macrophages of control or MIKO mice. These results show that IGF1 signaling shapes the macrophage-activation phenotype.


Asunto(s)
Resistencia a la Insulina/genética , Factor I del Crecimiento Similar a la Insulina/genética , Macrófagos/inmunología , Infecciones por Strongylida/inmunología , Tejido Adiposo/inmunología , Tejido Adiposo/metabolismo , Adiposidad , Animales , Diferenciación Celular/inmunología , Dieta Alta en Grasa , Resistencia a la Insulina/inmunología , Factor I del Crecimiento Similar a la Insulina/inmunología , Interleucina-4/inmunología , Macrófagos/metabolismo , Ratones , Ratones Noqueados , Nippostrongylus/patogenicidad , Fagocitosis/genética , Transducción de Señal/inmunología , Infecciones por Strongylida/metabolismo , Infecciones por Strongylida/parasitología
14.
Cell Rep ; 14(7): 1571-1580, 2016 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-26876170

RESUMEN

The hallmarks of age-related immune senescence are chronic inflammation, aberrant expansion of effector memory, and loss of naive T lymphocytes due in part to systemic activation of innate immune sensor NLRP3 inflammasome in myeloid lineage cells. The endogenous mechanisms that regulate inflammasome activation during aging are unknown. Here, we present evidence that growth hormone receptor (GH-R)-dependent downregulation of NLRP3 inflammasome in macrophages is linked to pro-longevity effects that maintain immune system homeostasis in aging. Deletion of GH-R prevented the macrophage-driven age-related activation of inflammasome in response to NLRP3 ligands and also increased the preservation of naive T cells, even in advanced age and with higher IFNγ secretion from effector cells. The mechanism of inflammasome inhibition is linked to autocrine somatotropic axis as ablation of IGF1R in macrophages lowered the NLRP3 inflammasome activation. Together, our findings show that functional somatotropic axis in macrophages controls inflammation, thus linking NLRP3-mediated innate immune signaling to health span and longevity.


Asunto(s)
Envejecimiento/genética , Proteínas Portadoras/genética , Inflamasomas/genética , Macrófagos/inmunología , Receptor IGF Tipo 1/genética , Receptores de Somatotropina/genética , Envejecimiento/inmunología , Animales , Comunicación Autocrina , Células de la Médula Ósea/citología , Células de la Médula Ósea/inmunología , Proteínas Portadoras/inmunología , Regulación de la Expresión Génica , Homeostasis/inmunología , Inmunidad Innata , Memoria Inmunológica , Inflamasomas/inmunología , Interferón gamma/biosíntesis , Interferón gamma/inmunología , Longevidad/genética , Longevidad/inmunología , Macrófagos/citología , Ratones , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR , Receptor IGF Tipo 1/deficiencia , Receptor IGF Tipo 1/inmunología , Receptores de Somatotropina/deficiencia , Receptores de Somatotropina/inmunología , Transducción de Señal , Bazo/citología , Bazo/inmunología , Linfocitos T/citología , Linfocitos T/inmunología
15.
J Biol Chem ; 290(49): 29402-13, 2015 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-26438821

RESUMEN

Dietary lipid overload and calorie excess during obesity is a low grade chronic inflammatory state with diminished ability to appropriately metabolize glucose or lipids. Macrophages are critical in maintaining adipose tissue homeostasis, in part by regulating lipid metabolism, energy homeostasis, and tissue remodeling. During high fat diet-induced obesity, macrophages are activated by lipid derived "danger signals" such as ceramides and palmitate and promote the adipose tissue inflammation in an Nlrp3 inflammasome-dependent manner. Given that the metabolic fate of fatty acids in macrophages is not entirely elucidated, we have hypothesized that de novo synthesis of ceramide, through the rate-limiting enzyme serine palmitoyltransferase long chain (Sptlc)-2, is required for saturated fatty acid-driven Nlrp3 inflammasome activation in macrophages. Here we report that mitochondrial targeted overexpression of catalase, which is established to mitigate oxidative stress, controls ceramide-induced Nlrp3 inflammasome activation but does not affect the ATP-mediated caspase-1 cleavage. Surprisingly, myeloid cell-specific deletion of Sptlc2 is not required for palmitate-driven Nlrp3 inflammasome activation. Furthermore, the ablation of Sptlc2 in macrophages did not impact macrophage polarization or obesity-induced adipose tissue leukocytosis. Consistent with these data, investigation of insulin resistance using hyperinsulinemic-euglycemic clamps revealed no significant differences in obese mice lacking ceramide de novo synthesis machinery in macrophages. These data suggest that alternate metabolic pathways control fatty acid-derived ceramide synthesis in macrophage and the Nlrp3 inflammasome activation in obesity.


Asunto(s)
Proteínas Portadoras/genética , Ceramidas/metabolismo , Inflamasomas/metabolismo , Resistencia a la Insulina , Macrófagos/metabolismo , Obesidad/metabolismo , Tejido Adiposo/metabolismo , Animales , Células de la Médula Ósea/citología , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Ácidos Grasos/química , Femenino , Inflamación/metabolismo , Lípidos/química , Masculino , Ratones , Ratones Transgénicos , Mitocondrias/patología , Proteína con Dominio Pirina 3 de la Familia NLR , Estrés Oxidativo , Serina C-Palmitoiltransferasa/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...