Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
1.
J Surg Res ; 288: 329-340, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37060859

RESUMEN

INTRODUCTION: Peripheral nerve injuries have been associated with increased healthcare costs and decreased patients' quality of life. Aging represents one factor that slows the speed of peripheral nervous system (PNS) regeneration. Since cellular homeostasis imbalance associated with aging lead to an increased failure in nerve regeneration in mammals of advanced age, this systematic review aims to determine the main molecular and cellular mechanisms involved in peripheral nerve regeneration in aged murine models after a peripheral nerve injuries. METHODS: Following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines, a literature search of 4 databases was conducted in July 2022 for studies comparing the peripheral nerve regeneration capability between young and aged murine models. RESULTS: After the initial search yielded 744 publications, ten articles fulfilled the inclusion criteria. These studies show that age-related changes such as chronic inflammatory state, delayed macrophages' response to injury, dysfunctional Schwann Cells (SCs), and microenvironment alterations cause a reduction in the regenerative capability of the PNS in murine models. Furthermore, identifying altered gene expression patterns of SC after nerve damage can contribute to the understanding of physiological modifications produced by aging. CONCLUSIONS: The interaction between macrophages and SC plays a crucial role in the nerve regeneration of aged models. Therefore, studies aimed at developing new and promising therapies for nerve regeneration should focus on these cellular groups to enhance the regenerative capabilities of the PNS in elderly populations.


Asunto(s)
Traumatismos de los Nervios Periféricos , Humanos , Animales , Ratones , Anciano , Traumatismos de los Nervios Periféricos/terapia , Calidad de Vida , Nervios Periféricos , Envejecimiento , Regeneración Nerviosa , Mamíferos
2.
J Neurosci ; 41(41): 8644-8667, 2021 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-34493542

RESUMEN

Western-style diets cause disruptions in myelinating cells and astrocytes within the mouse CNS. Increased CD38 expression is present in the cuprizone and experimental autoimmune encephalomyelitis models of demyelination and CD38 is the main nicotinamide adenine dinucleotide (NAD+)-depleting enzyme in the CNS. Altered NAD+ metabolism is linked to both high fat consumption and multiple sclerosis (MS). Here, we identify increased CD38 expression in the male mouse spinal cord following chronic high fat consumption, after focal toxin [lysolecithin (LL)]-mediated demyelinating injury, and in reactive astrocytes within active MS lesions. We demonstrate that CD38 catalytically inactive mice are substantially protected from high fat-induced NAD+ depletion, oligodendrocyte loss, oxidative damage, and astrogliosis. A CD38 inhibitor, 78c, increased NAD+ and attenuated neuroinflammatory changes induced by saturated fat applied to astrocyte cultures. Conditioned media from saturated fat-exposed astrocytes applied to oligodendrocyte cultures impaired myelin protein production, suggesting astrocyte-driven indirect mechanisms of oligodendrogliopathy. In cerebellar organotypic slice cultures subject to LL-demyelination, saturated fat impaired signs of remyelination effects that were mitigated by concomitant 78c treatment. Significantly, oral 78c increased counts of oligodendrocytes and remyelinated axons after focal LL-induced spinal cord demyelination. Using a RiboTag approach, we identified a unique in vivo brain astrocyte translatome profile induced by 78c-mediated CD38 inhibition in mice, including decreased expression of proinflammatory astrocyte markers and increased growth factors. Our findings suggest that a high-fat diet impairs oligodendrocyte survival and differentiation through astrocyte-linked mechanisms mediated by the NAD+ase CD38 and highlights CD38 inhibitors as potential therapeutic candidates to improve myelin regeneration.SIGNIFICANCE STATEMENT Myelin disturbances and oligodendrocyte loss can leave axons vulnerable, leading to permanent neurologic deficits. The results of this study suggest that metabolic disturbances, triggered by consumption of a diet high in fat, promote oligodendrogliopathy and impair myelin regeneration through astrocyte-linked indirect nicotinamide adenine dinucleotide (NAD+)-dependent mechanisms. We demonstrate that restoring NAD+ levels via genetic inactivation of CD38 can overcome these effects. Moreover, we show that therapeutic inactivation of CD38 can enhance myelin regeneration. Together, these findings point to a new metabolic targeting strategy positioned to improve disease course in multiple sclerosis and other conditions in which the integrity of myelin is a key concern.


Asunto(s)
ADP-Ribosil Ciclasa 1/metabolismo , Astrocitos/metabolismo , Glicoproteínas de Membrana/metabolismo , Vaina de Mielina/metabolismo , NAD+ Nucleosidasa/fisiología , Regeneración Nerviosa/fisiología , Remielinización/fisiología , ADP-Ribosil Ciclasa 1/antagonistas & inhibidores , ADP-Ribosil Ciclasa 1/genética , Animales , Cerebelo/metabolismo , Dieta Alta en Grasa/efectos adversos , Masculino , Glicoproteínas de Membrana/antagonistas & inhibidores , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Vaina de Mielina/genética , Técnicas de Cultivo de Órganos
3.
J Mol Cell Cardiol ; 166: 11-22, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35114253

RESUMEN

CD38 enzymatic activity regulates NAD+ and cADPR levels in mammalian tissues, and therefore has a prominent role in cellular metabolism and calcium homeostasis. Consequently, it is reasonable to hypothesize about its involvement in cardiovascular physiology as well as in heart related pathological conditions. AIM: To investigate the role of CD38 in cardiovascular performance, and its involvement in cardiac electrophysiology and calcium-handling. METHODS AND RESULTS: When submitted to a treadmill exhaustion test, a way of evaluating cardiovascular performance, adult male CD38KO mice showed better exercise capacity. This benefit was also obtained in genetically modified mice with catalytically inactive (CI) CD38 and in WT mice treated with antibody 68 (Ab68) which blocks CD38 activity. Hearts from these 3 groups (CD38KO, CD38CI and Ab68) showed increased NAD+ levels. When CD38KO mice were treated with FK866 which inhibits NAD+ biosynthesis, exercise capacity as well as NAD+ in heart tissue decreased to WT levels. Electrocardiograms of conscious unrestrained CD38KO and CD38CI mice showed lower basal heart rates and higher heart rate variability than WT mice. Although inactivation of CD38 in mice resulted in increased SERCA2a expression in the heart, the frequency of spontaneous calcium release from the sarcoplasmic reticulum under stressful conditions (high extracellular calcium concentration) was lower in CD38KO ventricular myocytes. When mice were challenged with caffeine-epinephrine, CD38KO mice had a lower incidence of bidirectional ventricular tachycardia when compared to WT ones. CONCLUSION: CD38 inhibition improves exercise performance by regulating NAD+ homeostasis. CD38 is involved in cardiovascular function since its genetic ablation decreases basal heart rate, increases heart rate variability and alters calcium handling in a way that protects mice from developing catecholamine induced ventricular arrhythmias.


Asunto(s)
ADP-Ribosil Ciclasa 1/metabolismo , Calcio , Glicoproteínas de Membrana/metabolismo , NAD , ADP-Ribosil Ciclasa 1/genética , Animales , Arritmias Cardíacas/etiología , Arritmias Cardíacas/metabolismo , Calcio/metabolismo , Catecolaminas/metabolismo , Tolerancia al Ejercicio , Frecuencia Cardíaca , Masculino , Mamíferos/metabolismo , Ratones , Miocitos Cardíacos/metabolismo , NAD/metabolismo
4.
Am J Physiol Cell Physiol ; 322(3): C521-C545, 2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-35138178

RESUMEN

Nicotinamide adenine dinucleotide (NAD) acts as a cofactor in several oxidation-reduction (redox) reactions and is a substrate for a number of nonredox enzymes. NAD is fundamental to a variety of cellular processes including energy metabolism, cell signaling, and epigenetics. NAD homeostasis appears to be of paramount importance to health span and longevity, and its dysregulation is associated with multiple diseases. NAD metabolism is dynamic and maintained by synthesis and degradation. The enzyme CD38, one of the main NAD-consuming enzymes, is a key component of NAD homeostasis. The majority of CD38 is localized in the plasma membrane with its catalytic domain facing the extracellular environment, likely for the purpose of controlling systemic levels of NAD. Several cell types express CD38, but its expression predominates on endothelial cells and immune cells capable of infiltrating organs and tissues. Here we review potential roles of CD38 in health and disease and postulate ways in which CD38 dysregulation causes changes in NAD homeostasis and contributes to the pathophysiology of multiple conditions. Indeed, in animal models the development of infectious diseases, autoimmune disorders, fibrosis, metabolic diseases, and age-associated diseases including cancer, heart disease, and neurodegeneration are associated with altered CD38 enzymatic activity. Many of these conditions are modified in CD38-deficient mice or by blocking CD38 NADase activity. In diseases in which CD38 appears to play a role, CD38-dependent NAD decline is often a common denominator of pathophysiology. Thus, understanding dysregulation of NAD homeostasis by CD38 may open new avenues for the treatment of human diseases.


Asunto(s)
Glicósido Hidrolasas , NAD , ADP-Ribosil Ciclasa 1/genética , ADP-Ribosil Ciclasa 1/metabolismo , Animales , Células Endoteliales/metabolismo , Ratones , NAD/metabolismo , NAD+ Nucleosidasa/metabolismo
5.
J Am Soc Nephrol ; 27(5): 1437-47, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26538633

RESUMEN

Autosomal dominant polycystic kidney disease (ADPKD) is a genetic disorder characterized by the accumulation of kidney cysts that ultimately leads to loss of renal function and kidney failure. At present, the treatment for ADPKD is largely supportive. Multiple studies have focused on pharmacologic approaches to slow the development of the cystic disease; however, little is known about the role of nutrition and dietary manipulation in PKD. Here, we show that food restriction (FR) effectively slows the course of the disease in mouse models of ADPKD. Mild to moderate (10%-40%) FR reduced cyst area, renal fibrosis, inflammation, and injury in a dose-dependent manner. Molecular and biochemical studies in these mice indicate that FR ameliorates ADPKD through a mechanism involving suppression of the mammalian target of the rapamycin pathway and activation of the liver kinase B1/AMP-activated protein kinase pathway. Our data suggest that dietary interventions such as FR, or treatment that mimics the effects of such interventions, may be potential and novel preventive and therapeutic options for patients with ADPKD.


Asunto(s)
Alimentos , Riñón Poliquístico Autosómico Dominante/dietoterapia , Riñón Poliquístico Autosómico Dominante/metabolismo , Animales , Biomarcadores , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones , Transducción de Señal
6.
J Biol Chem ; 289(9): 5518-27, 2014 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-24415752

RESUMEN

Liver gluconeogenesis is essential to provide energy to glycolytic tissues during fasting periods. However, aberrant up-regulation of this metabolic pathway contributes to the progression of glucose intolerance in individuals with diabetes. Phosphoenolpyruvate carboxykinase (PEPCK) expression plays a critical role in the modulation of gluconeogenesis. Several pathways contribute to the regulation of PEPCK, including the nuclear receptor Rev-erbα and the histone deacetylase SIRT1. Deleted in breast cancer 1 (DBC1) is a nuclear protein that binds to and regulates both Rev-erbα and SIRT1 and, therefore, is a candidate to participate in the regulation of PEPCK. In this work, we provide evidence that DBC1 regulates glucose metabolism and the expression of PEPCK. We show that DBC1 levels decrease early in the fasting state. Also, DBC1 KO mice display higher gluconeogenesis in a normal and a high-fat diet. DBC1 absence leads to an increase in PEPCK mRNA and protein expression. Conversely, overexpression of DBC1 results in a decrease in PEPCK mRNA and protein levels. DBC1 regulates the levels of Rev-erbα, and manipulation of Rev-erbα activity or levels prevents the effect of DBC1 on PEPCK. In addition, Rev-erbα levels decrease in the first hours of fasting. Finally, knockdown of the deacetylase SIRT1 eliminates the effect of DBC1 knockdown on Rev-erbα levels and PEPCK expression, suggesting that the mechanism of PEPCK regulation is, at least in part, dependent on the activity of this enzyme. Our results point to DBC1 as a novel regulator of gluconeogenesis.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Gluconeogénesis/fisiología , Glucosa/biosíntesis , Hígado/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Grasas de la Dieta/farmacología , Ayuno/metabolismo , Regulación Enzimológica de la Expresión Génica/fisiología , Glucosa/genética , Células Hep G2 , Humanos , Hígado/citología , Ratones , Ratones Noqueados , Miembro 1 del Grupo D de la Subfamilia 1 de Receptores Nucleares/genética , Miembro 1 del Grupo D de la Subfamilia 1 de Receptores Nucleares/metabolismo , Fosfoenolpiruvato Carboxiquinasa (ATP)/biosíntesis , Fosfoenolpiruvato Carboxiquinasa (ATP)/genética , Sirtuina 1/genética , Sirtuina 1/metabolismo
7.
Biochem J ; 451(3): 453-61, 2013 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-23398316

RESUMEN

The nuclear receptor Rev-erbα has been implicated as a major regulator of the circadian clock and integrates circadian rhythm and metabolism. Rev-erbα controls circadian oscillations of several clock genes and Rev-erbα protein degradation is important for maintenance of the circadian oscillations and also for adipocyte differentiation. Elucidating the mechanisms that regulate Rev-erbα stability is essential for our understanding of these processes. In the present paper, we report that the protein DBC1 (Deleted in Breast Cancer 1) is a novel regulator of Rev-erbα. Rev-erbα and DBC1 interact in cells and in vivo, and DBC1 modulates the Rev-erbα repressor function. Depletion of DBC1 by siRNA (small interfering RNA) in cells or in DBC1-KO (knockout) mice produced a marked decrease in Rev-erbα protein levels, but not in mRNA levels. In contrast, DBC1 overexpression significantly enhanced Rev-erbα protein stability by preventing its ubiquitination and degradation. The regulation of Rev-erbα protein levels and function by DBC1 depends on both the N-terminal and C-terminal domains of DBC1. More importantly, in cells depleted of DBC1, there was a dramatic decrease in circadian oscillations of both Rev-erbα and BMAL1. In summary, our data identify DBC1 as an important regulator of the circadian receptor Rev-erbα and proposes that Rev-erbα could be involved in mediating some of the physiological effects of DBC1.


Asunto(s)
Relojes Circadianos/genética , Ritmo Circadiano/genética , Miembro 1 del Grupo D de la Subfamilia 1 de Receptores Nucleares/genética , ARN Mensajero/genética , Factores de Transcripción ARNTL/genética , Factores de Transcripción ARNTL/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Línea Celular , Regulación de la Expresión Génica , Humanos , Ratones , Ratones Noqueados , Miembro 1 del Grupo D de la Subfamilia 1 de Receptores Nucleares/metabolismo , Plásmidos , Estabilidad Proteica , Estructura Terciaria de Proteína , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética , Transducción de Señal , Transfección
8.
Nat Aging ; 2024 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-39044033

RESUMEN

How hematopoietic stem cells (HSCs) maintain metabolic homeostasis to support tissue repair and regeneration throughout the lifespan is elusive. Here, we show that CD38, an NAD+-dependent metabolic enzyme, promotes HSC proliferation by inducing mitochondrial Ca2+ influx and mitochondrial metabolism in young mice. Conversely, aberrant CD38 upregulation during aging is a driver of HSC deterioration in aged mice due to dysregulated NAD+ metabolism and compromised mitochondrial stress management. The mitochondrial calcium uniporter, a mediator of mitochondrial Ca2+ influx, also supports HSC proliferation in young mice yet drives HSC decline in aged mice. Pharmacological inactivation of CD38 reverses HSC aging and the pathophysiological changes of the aging hematopoietic system in aged mice. Together, our study highlights an NAD+ metabolic checkpoint that balances mitochondrial activation to support HSC proliferation and mitochondrial stress management to enhance HSC self-renewal throughout the lifespan, and links aberrant Ca2+ signaling to HSC aging.

9.
Cardiovasc Res ; 120(3): 286-300, 2024 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-38271281

RESUMEN

AIMS: Doxorubicin (DXR) is a chemotherapeutic agent that causes dose-dependent cardiotoxicity. Recently, it has been proposed that the NADase CD38 may play a role in doxorubicin-induced cardiotoxicity (DIC). CD38 is the main NAD+-catabolizing enzyme in mammalian tissues. Interestingly, in the heart, CD38 is mostly expressed as an ecto-enzyme that can be targeted by specific inhibitory antibodies. The goal of the present study is to characterize the role of CD38 ecto-enzymatic activity in cardiac metabolism and the development of DIC. METHODS AND RESULTS: Using both a transgenic animal model and a non-cytotoxic enzymatic anti-CD38 antibody, we investigated the role of CD38 and its ecto-NADase activity in DIC in pre-clinical models. First, we observed that DIC was prevented in the CD38 catalytically inactive (CD38-CI) transgenic mice. Both left ventricular systolic function and exercise capacity were decreased in wild-type but not in CD38-CI mice treated with DXR. Second, blocking CD38-NADase activity with the specific antibody 68 (Ab68) likewise protected mice against DIC and decreased DXR-related mortality by 50%. A reduction of DXR-induced mitochondrial dysfunction, energy deficiency, and inflammation gene expression were identified as the main mechanisms mediating the protective effects. CONCLUSION: NAD+-preserving strategies by inactivation of CD38 via a genetic or a pharmacological-based approach improve cardiac energetics and reduce cardiac inflammation and dysfunction otherwise seen in an acute DXR cardiotoxicity model.


Asunto(s)
NAD+ Nucleosidasa , NAD , Ratones , Animales , NAD+ Nucleosidasa/metabolismo , ADP-Ribosil Ciclasa 1/genética , ADP-Ribosil Ciclasa 1/metabolismo , NAD/metabolismo , Cardiotoxicidad , Ratones Transgénicos , Doxorrubicina/toxicidad , Inflamación , Mamíferos/metabolismo
10.
J Biol Chem ; 287(28): 23489-501, 2012 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-22553202

RESUMEN

The NAD(+)-dependent deacetylase SIRT1 is a key regulator of several aspects of metabolism and aging. SIRT1 activation is beneficial for several human diseases, including metabolic syndrome, diabetes, obesity, liver steatosis, and Alzheimer disease. We have recently shown that the protein deleted in breast cancer 1 (DBC1) is a key regulator of SIRT1 activity in vivo. Furthermore, SIRT1 and DBC1 form a dynamic complex that is regulated by the energetic state of the organism. Understanding how the interaction between SIRT1 and DBC1 is regulated is therefore essential to design strategies aimed to activate SIRT1. Here, we investigated which pathways can lead to the dissociation of SIRT1 and DBC1 and consequently to SIRT1 activation. We observed that PKA activation leads to a fast and transient activation of SIRT1 that is DBC1-dependent. In fact, an increase in cAMP/PKA activity resulted in the dissociation of SIRT1 and DBC1 in an AMP-activated protein kinase (AMPK)-dependent manner. Pharmacological AMPK activation led to SIRT1 activation by a DBC1-dependent mechanism. Indeed, we found that AMPK activators promote SIRT1-DBC1 dissociation in cells, resulting in an increase in SIRT1 activity. In addition, we observed that the SIRT1 activation promoted by PKA and AMPK occurs without changes in the intracellular levels of NAD(+). We propose that PKA and AMPK can acutely activate SIRT1 by inducing dissociation of SIRT1 from its endogenous inhibitor DBC1. Our experiments provide new insight on the in vivo mechanism of SIRT1 regulation and a new avenue for the development of pharmacological SIRT1 activators targeted at the dissociation of the SIRT1-DBC1 complex.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Sirtuina 1/metabolismo , Acrilamidas/farmacología , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/fisiología , Secuencia de Aminoácidos , Animales , Western Blotting , Carbazoles/farmacología , Línea Celular Tumoral , Células Cultivadas , Activación Enzimática/efectos de los fármacos , Células HEK293 , Células Hep G2 , Humanos , Ratones , Ratones Noqueados , Modelos Biológicos , Mutación , NAD/metabolismo , Niacinamida/farmacología , Fosforilación/efectos de los fármacos , Piperidinas/farmacología , Interferencia de ARN , Resveratrol , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Sirtuina 1/antagonistas & inhibidores , Sirtuina 1/genética , Estilbenos/farmacología
11.
Sci Rep ; 13(1): 22085, 2023 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-38086958

RESUMEN

The functionally pleiotropic ectoenzyme CD38 is a glycohydrolase widely expressed on immune and non-hematopoietic cells. By converting NAD+ to ADP-ribose and nicotinamide, CD38 governs organismal NAD+ homeostasis and the activity of NAD+-dependent cellular enzymes. CD38 has emerged as a major driver of age-related NAD+ decline underlying adverse metabolic states, frailty and reduced health span. CD38 is upregulated in systemic sclerosis (SSc), a chronic disease characterized by fibrosis in multiple organs. We sought to test the hypothesis that inhibition of the CD38 ecto-enzymatic activity using a heavy-chain monoclonal antibody Ab68 will, via augmenting organismal NAD+, prevent fibrosis in a mouse model of SSc characterized by NAD+ depletion. Here we show that treatment of mice with a non-cytotoxic heavy-chain antibody that selectively inhibits CD38 ectoenzyme resulted in NAD+ boosting that was associated with significant protection from fibrosis in multiple organs. These findings suggest that targeted inhibition of CD38 ecto-enzymatic activity could be a potential pharmacological approach for SSc fibrosis treatment.


Asunto(s)
Antígenos CD , Antígenos de Diferenciación , Ratones , Animales , ADP-Ribosil Ciclasa 1/metabolismo , Antígenos CD/metabolismo , Antígenos de Diferenciación/metabolismo , NAD+ Nucleosidasa/metabolismo , NAD/metabolismo , ADP-Ribosil Ciclasa , Glicoproteínas de Membrana/metabolismo , Glicósido Hidrolasas , Fibrosis
12.
J Clin Transl Res ; 8(5): 382-389, 2022 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-36518551

RESUMEN

Background and Aim: Although a natural phenomenon, aging is a degenerative condition that promotes cellular malfunction and subsequent organ and body dysfunction. According to the World Health Organization, the elderly are the fastest growing age group worldwide. A 2012 population report stated that 43.1 million adults of 65 years or older lived in the United States, which is expected to jump to 83.7 million in 2050, placing an additional burden on an already stretched health-care network. Elderly patients broadly impact our health-care system, as reported in a 2014 wound report. 8.2 million patients were diagnosed with at least one type of wound, with patients 75 years or older making up most of the diagnoses. Aging affects all stages of the wound healing cascade. Although wound healing is downregulated in the elderly, scarce information exists regarding the effects of aging and flap survival in this group. Therefore, this study aims to report the impact of age on the survival of flaps in murine models. We hypothesize that increased aged animals will have decreased flap survival. Methods: A systematic review was performed on February 1, 2022, following the Preferred Reporting Items for Systematic Reviews and Meta-Analysis. We searched for full-text articles written in English, consisting of experimental murine models that compared flap survival between aged and young animals, in the following databases: PubMed, Scopus, CINAHL, and Web of Science. The terms "mice" OR "rats" AND "surgical flaps" AND "aging" guided our search. Models affected by chronic diseases were excluded from the study. Results: Out of the 208 articles found by our search, seven were included according to our inclusion and exclusion criteria. Five studies used rats as experimental models, while the remaining two used mice. Local flaps were done in five studies, and two performed free flaps, transferring them from young and aged animals to young controls. Five articles reported lower flap survival in elder groups when exposed to ischemic insults. Three papers reported a deficiency in angiogenesis, vasculogenesis, and vascular reactivity as plausible causes for lack of survival, with one author correlating and verifying their results in human subjects. Although one article reported a lack of statistical power, they perceived a trend similar to the previous studies. Finally, one article reported inconclusive and variable results. Conclusion: Evidence suggests that a lack of angiogenic and vasculogenic response in conjunction with decreased vascular reactivity are responsible for the diminished survival of flaps in the elder. Therapeutic means to boost the angiogenic, vasculogenic, and vascular reactivity response to improve patient outcomes require further research to understand the time course and mechanisms of flap survival in the elderly. Relevance for Patients: All humans will feel the effects of aging one way or another. However, we can all agree that aging affects our basic biological processes, which negatively affects macroscopic appearance. One of the essential aspects downregulated in the elderly is their ability to respond to tissue injury and hypoxia, creating non-favorable circumstances for wound healing. Furthermore, to manage these non-healing wounds, flaps are raised to create a covering for these defects. However, age also impacts the ability of these flaps to survive, augmenting the problem and entering a vicious circle. To improve outcomes, we must focus our future research on understanding the basic principles of how aging affects the survival of flaps in elderly population.

13.
Aging Cell ; 21(4): e13589, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35263032

RESUMEN

Nicotinamide adenine dinucleotide (NAD) levels decline during aging, contributing to physical and metabolic dysfunction. The NADase CD38 plays a key role in age-related NAD decline. Whether the inhibition of CD38 increases lifespan is not known. Here, we show that the CD38 inhibitor 78c increases lifespan and healthspan of naturally aged mice. In addition to a 10% increase in median survival, 78c improved exercise performance, endurance, and metabolic function in mice. The effects of 78c were different between sexes. Our study is the first to investigate the effect of CD38 inhibition in naturally aged animals.


Asunto(s)
Longevidad , NAD , ADP-Ribosil Ciclasa 1/metabolismo , Envejecimiento/metabolismo , Animales , Ratones , NAD/metabolismo , NAD+ Nucleosidasa/metabolismo
14.
Front Immunol ; 13: 840246, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35281060

RESUMEN

Nicotinamide adenine dinucleotide (NAD) metabolism plays an important role in the regulation of immune function. However, a complete picture of how NAD, its metabolites, precursors, and metabolizing enzymes work together in regulating immune function and inflammatory diseases is still not fully understood. Surprisingly, few studies have compared the effect of different forms of vitamin B3 on cellular functions. Therefore, we investigated the role of NAD boosting in the regulation of macrophage activation and function using different NAD precursors supplementation. We compared nicotinamide mononucleotide (NMN), nicotinamide riboside (NR), and nicotinamide (NAM) supplementation, with the recently described potent NAD precursor NRH. Our results show that only NRH supplementation strongly increased NAD+ levels in both bone marrow-derived and THP-1 macrophages. Importantly, NRH supplementation activated a pro-inflammatory phenotype in resting macrophages, inducing gene expression of several cytokines, chemokines, and enzymes. NRH also potentiated the effect of lipopolysaccharide (LPS) on macrophage activation and cytokine gene expression, suggesting that potent NAD+ precursors can promote inflammation in macrophages. The effect of NRH in NAD+ boosting and gene expression was blocked by inhibitors of adenosine kinase, equilibrative nucleoside transporters (ENT), and IκB kinase (IKK). Interestingly, the IKK inhibitor, BMS-345541, blocked the mRNA expression of several enzymes and transporters involved in the NAD boosting effect of NRH, indicating that IKK is also a regulator of NAD metabolism. In conclusion, NAD precursors such as NRH may be important tools to understand the role of NAD and NADH metabolism in the inflammatory process of other immune cells, and to reprogram immune cells to a pro-inflammatory phenotype, such as the M2 to M1 switch in macrophage reprogramming, in the cancer microenvironment.


Asunto(s)
NAD , Niacinamida , Citocinas , Glicósidos , Macrófagos/metabolismo , NAD/metabolismo , Niacinamida/análogos & derivados , Niacinamida/farmacología , Fenotipo
15.
Front Endocrinol (Lausanne) ; 13: 896356, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35600581

RESUMEN

Advanced paternal age has increasingly been recognized as a risk factor for male fertility and progeny health. While underlying causes are not well understood, aging is associated with a continuous decline of blood and tissue NAD+ levels, as well as a decline of testicular functions. The important basic question to what extent ageing-related NAD+ decline is functionally linked to decreased male fertility has been difficult to address due to the pleiotropic effects of aging, and the lack of a suitable animal model in which NAD+ levels can be lowered experimentally in chronologically young adult males. We therefore developed a transgenic mouse model of acquired niacin dependency (ANDY), in which NAD+ levels can be experimentally lowered using a niacin-deficient, chemically defined diet. Using ANDY mice, this report demonstrates for the first time that decreasing body-wide NAD+ levels in young adult mice, including in the testes, to levels that match or exceed the natural NAD+ decline observed in old mice, results in the disruption of spermatogenesis with small testis sizes and reduced sperm counts. ANDY mice are dependent on dietary vitamin B3 (niacin) for NAD+ synthesis, similar to humans. NAD+-deficiency the animals develop on a niacin-free diet is reversed by niacin supplementation. Providing niacin to NAD+-depleted ANDY mice fully rescued spermatogenesis and restored normal testis weight in the animals. The results suggest that NAD+ is important for proper spermatogenesis and that its declining levels during aging are functionally linked to declining spermatogenesis and male fertility. Functions of NAD+ in retinoic acid synthesis, which is an essential testicular signaling pathway regulating spermatogonial proliferation and differentiation, may offer a plausible mechanism for the hypospermatogenesis observed in NAD+-deficient mice.


Asunto(s)
Niacina , Envejecimiento , Animales , Masculino , Ratones , Ratones Transgénicos , NAD/metabolismo , NAD/farmacología , Niacina/metabolismo , Niacina/farmacología , Espermatogénesis
16.
iScience ; 25(11): 105431, 2022 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-36388973

RESUMEN

In mammals, nicotinamide (NAM) is the primary NAD precursor available in circulation, a signaling molecule, and a precursor for methyl-nicotinamide (M-NAM) synthesis. However, our knowledge about how the body regulates tissue NAM levels is still limited. Here we demonstrate that dietary vitamin B3 partially regulates plasma NAM and NAM-derived metabolites, but not their tissue levels. We found that NAD de novo synthesis from tryptophan contributes to plasma and tissue NAM, likely by providing substrates for NAD-degrading enzymes. We also demonstrate that tissue NAM is mainly generated by endogenous metabolism and that the NADase CD38 is the main enzyme that produces tissue NAM. Tissue-specific CD38-floxed mice revealed that CD38 activity on endothelial and immune cells is the major contributor to tissue steady-state levels of NAM in tissues like spleen and heart. Our findings uncover the presence of different pools of NAM in the body and a central role for CD38 in regulating tissue NAM levels.

17.
EMBO Mol Med ; 14(5): e12860, 2022 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-35298089

RESUMEN

Duchenne muscular dystrophy (DMD) is characterized by progressive muscle degeneration. Two important deleterious features are a Ca2+ dysregulation linked to Ca2+ influxes associated with ryanodine receptor hyperactivation, and a muscular nicotinamide adenine dinucleotide (NAD+ ) deficit. Here, we identified that deletion in mdx mice of CD38, a NAD+ glycohydrolase-producing modulators of Ca2+ signaling, led to a fully restored heart function and structure, with skeletal muscle performance improvements, associated with a reduction in inflammation and senescence markers. Muscle NAD+ levels were also fully restored, while the levels of the two main products of CD38, nicotinamide and ADP-ribose, were reduced, in heart, diaphragm, and limb. In cardiomyocytes from mdx/CD38-/- mice, the pathological spontaneous Ca2+ activity was reduced, as well as in myotubes from DMD patients treated with isatuximab (SARCLISA® ) a monoclonal anti-CD38 antibody. Finally, treatment of mdx and utrophin-dystrophin-deficient (mdx/utr-/- ) mice with CD38 inhibitors resulted in improved skeletal muscle performances. Thus, we demonstrate that CD38 actively contributes to DMD physiopathology. We propose that a selective anti-CD38 therapeutic intervention could be highly relevant to develop for DMD patients.


Asunto(s)
Distrofia Muscular de Duchenne , ADP-Ribosil Ciclasa 1 , Animales , Humanos , Ratones , Ratones Endogámicos mdx , Músculo Esquelético , Distrofia Muscular de Duchenne/genética , Miocitos Cardíacos/patología , NAD/genética , NAD/uso terapéutico , NAD+ Nucleosidasa/genética , Fenotipo
18.
J Biol Chem ; 285(52): 40830-7, 2010 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-21030595

RESUMEN

HDAC3 is a member of the class I histone deacetylase family that regulates gene expression by deacetylation of histones and non-histone proteins. HDAC3 activity has been shown to be modulated by interaction with the co-repressors NCoR and SMRT. Here, we present evidence that the nuclear protein DBC1 is an endogenous inhibitor of HDAC3. DBC1 has been previously identified as a regulator of some nuclear receptors, the methyltransferase SUV39H1, and the NAD-dependent deacetylase SIRT1. Furthermore, DBC1 has been shown to influence transcription regulation and apoptosis, and it may also act as a tumor suppressor. We found that DBC1 interacts and specifically inhibits the deacetylase HDAC3. This interaction depends on the N terminus of DBC1 and the C terminus of HDAC3. Expression of DBC1 not only inhibited HDAC3 activity but also altered its subcellular distribution. In addition, knockdown of endogenous DBC1 in cells and knock-out in mouse tissues increased HDAC3 deacetylase activity. Together, these results identify DBC1 as a new regulator of HDAC3 and demonstrate that DBC1 is a negative regulator of two key distinct deacetylases, SIRT1 and HDAC3. These findings may lead to a better understanding of the biological roles of DBC1 and HDAC3 in metabolic diseases and cancer.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Histona Desacetilasas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Apoptosis/genética , Células HEK293 , Histona Desacetilasas/genética , Humanos , Metiltransferasas/genética , Metiltransferasas/metabolismo , Ratones , Ratones Noqueados , Células 3T3 NIH , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Sirtuina 1/genética , Sirtuina 1/metabolismo , Transcripción Genética
19.
Cell Metab ; 33(6): 1076-1087, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33930322

RESUMEN

NAD(H) and NADP(H) have traditionally been viewed as co-factors (or co-enzymes) involved in a myriad of oxidation-reduction reactions including the electron transport in the mitochondria. However, NAD pathway metabolites have many other important functions, including roles in signaling pathways, post-translational modifications, epigenetic changes, and regulation of RNA stability and function via NAD-capping of RNA. Non-oxidative reactions ultimately lead to the net catabolism of these nucleotides, indicating that NAD metabolism is an extremely dynamic process. In fact, recent studies have clearly demonstrated that NAD has a half-life in the order of minutes in some tissues. Several evolving concepts on the metabolism, transport, and roles of these NAD pathway metabolites in disease states such as cancer, neurodegeneration, and aging have emerged in just the last few years. In this perspective, we discuss key recent discoveries and changing concepts in NAD metabolism and biology that are reshaping the field. In addition, we will pose some open questions in NAD biology, including why NAD metabolism is so fast and dynamic in some tissues, how NAD and its precursors are transported to cells and organelles, and how NAD metabolism is integrated with inflammation and senescence. Resolving these questions will lead to significant advancements in the field.


Asunto(s)
Mitocondrias/metabolismo , NADP/metabolismo , NAD/metabolismo , Animales , Metabolismo Energético , Humanos
20.
iScience ; 24(1): 101902, 2021 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-33385109

RESUMEN

The processes underlying synchronous multiple organ fibrosis in systemic sclerosis (SSc) remain poorly understood. Age-related pathologies are associated with organismal decline in nicotinamide adenine dinucleotide (NAD+) that is due to dysregulation of NAD+ homeostasis and involves the NADase CD38. We now show that CD38 is upregulated in patients with diffuse cutaneous SSc, and CD38 levels in the skin associate with molecular fibrosis signatures, as well as clinical fibrosis scores, while expression of key NAD+-synthesizing enzymes is unaltered. Boosting NAD+ via genetic or pharmacological CD38 targeting or NAD+ precursor supplementation protected mice from skin, lung, and peritoneal fibrosis. In mechanistic experiments, CD38 was found to reduce NAD+ levels and sirtuin activity to augment cellular fibrotic responses, while inhibiting CD38 had the opposite effect. Thus, we identify CD38 upregulation and resulting disrupted NAD+ homeostasis as a fundamental mechanism driving fibrosis in SSc, suggesting that CD38 might represent a novel therapeutic target.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA