Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Dis Model Mech ; 16(5)2023 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-36374036

RESUMO

Nicotinamide adenine dinucleotide (NAD) is a key metabolite synthesised from vitamin B3 or tryptophan. Disruption of genes encoding NAD synthesis enzymes reduces NAD levels and causes congenital NAD deficiency disorder (CNDD), characterised by multiple congenital malformations. SLC6A19 (encoding B0AT1, a neutral amino acid transporter), represents the main transporter for free tryptophan in the intestine and kidney. Here, we tested whether Slc6a19 heterozygosity in mice limits the tryptophan available for NAD synthesis during pregnancy and causes adverse pregnancy outcomes. Pregnant Slc6a19+/- mice were fed diets depleted of vitamin B3, so that tryptophan was the source of NAD during gestation. This perturbed the NAD metabolome in pregnant Slc6a19+/- females, resulting in reduced NAD levels and increased rates of embryo loss. Surviving embryos were small and exhibited specific combinations of CNDD-associated malformations. Our results show that genes not directly involved in NAD synthesis can affect NAD metabolism and cause CNDD. They also suggest that human female carriers of a SLC6A19 loss-of-function allele might be susceptible to adverse pregnancy outcomes unless sufficient NAD precursor amounts are available during gestation. This article has an associated First Person interview with the first author of the paper.


Assuntos
Sistemas de Transporte de Aminoácidos Neutros , Anormalidades Congênitas , NAD , Animais , Feminino , Camundongos , Gravidez , Sistemas de Transporte de Aminoácidos Neutros/genética , Sistemas de Transporte de Aminoácidos Neutros/metabolismo , Heterozigoto , Rim/metabolismo , NAD/deficiência , Niacinamida , Triptofano/genética , Triptofano/metabolismo , Anormalidades Congênitas/genética
2.
PLoS Pathog ; 17(8): e1009791, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34370789

RESUMO

In many Gram-positive bacteria, the redox-sensing transcriptional repressor Rex controls central carbon and energy metabolism by sensing the intra cellular balance between the reduced and oxidized forms of nicotinamide adenine dinucleotide; the NADH/NAD+ ratio. Here, we report high-resolution crystal structures and characterization of a Rex ortholog (Gbs1167) in the opportunistic pathogen, Streptococcus agalactiae, also known as group B streptococcus (GBS). We present structures of Rex bound to NAD+ and to a DNA operator which are the first structures of a Rex-family member from a pathogenic bacterium. The structures reveal the molecular basis of DNA binding and the conformation alterations between the free NAD+ complex and DNA-bound form of Rex. Transcriptomic analysis revealed that GBS Rex controls not only central metabolism, but also expression of the monocistronic rex gene as well as virulence gene expression. Rex enhances GBS virulence after disseminated infection in mice. Mechanistically, NAD+ stabilizes Rex as a repressor in the absence of NADH. However, GBS Rex is unique compared to Rex regulators previously characterized because of its sensing mechanism: we show that it primarily responds to NAD+ levels (or growth rate) rather than to the NADH/NAD+ ratio. These results indicate that Rex plays a key role in GBS pathogenicity by modulating virulence factor gene expression and carbon metabolism to harvest nutrients from the host.


Assuntos
Proteínas de Bactérias/genética , Produtos do Gene rex/genética , NAD/deficiência , Regulon , Infecções Estreptocócicas/microbiologia , Streptococcus agalactiae/patogenicidade , Virulência , Animais , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Cristalografia por Raios X , Feminino , Perfilação da Expressão Gênica , Produtos do Gene rex/química , Produtos do Gene rex/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Ligação Proteica , Conformação Proteica , Infecções Estreptocócicas/metabolismo
3.
Biol Reprod ; 105(2): 371-380, 2021 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-34056649

RESUMO

Polycystic ovary syndrome (PCOS) is a prevalent heterogeneous endocrine disorder characterized by ovulation dysfunction, androgen excess, ovarian polycystic changes, insulin resistance, and infertility. Although underlying mechanisms for PCOS are still unknown, inflammation and mitochondrial dysfunction in granulosa cells (GCs) of PCOS patients have been reported. Here, we found that Nicotinamide Adenine Dinucleotide (NAD+) levels in GCs of PCOS patients was significantly decreased when compared with controls. Also, we found that higher expression of inflammation factors, increased reactive oxygen species (ROS) accumulation, lower adenosine triphosphate (ATP) generation, and decreased mitochondrial membrane potential, as well as abnormal mitochondrial dynamics in GCs of PCOS patients. In addition, the NAD+ levels were decreased after activation of inflammation in human granulosa-like tumor cell line (KGN) treated by Lipopolysaccharide (LPS). However, supplementation of nicotinamide riboside (NR), a NAD+ precursor, could largely restore the NAD+ content, reduce ROS levels and improve mitochondrial function demonstrated by increased mitochondrial membrane potential and ATP generation in LPS-treated KGN cells. Our data suggested that inflammation decreased NAD+ levels in GCs of PCOS patients, while supplementation of NR could restore NAD+ levels and alleviated mitochondrial dysfunction in GCs of PCOS patients.


Assuntos
Células da Granulosa/patologia , Inflamação/metabolismo , NAD/deficiência , Síndrome do Ovário Policístico/fisiopatologia , Adulto , Linhagem Celular Tumoral , Feminino , Humanos , Mitocôndrias/patologia , Adulto Jovem
5.
Mol Genet Metab ; 133(1): 83-93, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33752971

RESUMO

Leigh syndrome is a severe mitochondrial neurodegenerative disease with no effective treatment. In the Ndufs4-/- mouse model of Leigh syndrome, continuously breathing 11% O2 (hypoxia) prevents neurodegeneration and leads to a dramatic extension (~5-fold) in lifespan. We investigated the effect of hypoxia on the brain metabolism of Ndufs4-/- mice by studying blood gas tensions and metabolite levels in simultaneously sampled arterial and cerebral internal jugular venous (IJV) blood. Relatively healthy Ndufs4-/- and wildtype (WT) mice breathing air until postnatal age ~38 d were compared to Ndufs4-/- and WT mice breathing air until ~38 days old followed by 4-weeks of breathing 11% O2. Compared to WT control mice, Ndufs4-/- mice breathing air have reduced brain O2 consumption as evidenced by an elevated partial pressure of O2 in IJV blood (PijvO2) despite a normal PO2 in arterial blood, and higher lactate/pyruvate (L/P) ratios in IJV plasma revealed by metabolic profiling. In Ndufs4-/- mice, hypoxia treatment normalized the cerebral venous PijvO2 and L/P ratios, and decreased levels of nicotinate in IJV plasma. Brain concentrations of nicotinamide adenine dinucleotide (NAD+) were lower in Ndufs4-/- mice breathing air than in WT mice, but preserved at WT levels with hypoxia treatment. Although mild hypoxia (17% O2) has been shown to be an ineffective therapy for Ndufs4-/- mice, we find that when combined with nicotinic acid supplementation it provides a modest improvement in neurodegeneration and lifespan. Therapies targeting both brain hyperoxia and NAD+ deficiency may hold promise for treating Leigh syndrome.


Assuntos
Encéfalo/metabolismo , Complexo I de Transporte de Elétrons/genética , Doença de Leigh/metabolismo , NAD/genética , Oxigênio/metabolismo , Animais , Encéfalo/patologia , Hipóxia Celular/fisiologia , Modelos Animais de Doenças , Complexo I de Transporte de Elétrons/metabolismo , Humanos , Doença de Leigh/genética , Doença de Leigh/terapia , Metabolômica , Camundongos , Mitocôndrias , NAD/deficiência , Doenças Neurodegenerativas , Respiração/genética
6.
Proc Natl Acad Sci U S A ; 118(8)2021 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-33597293

RESUMO

Emerging evidence suggests that intratumoral interferon (IFN) signaling can trigger targetable vulnerabilities. A hallmark of pancreatic ductal adenocarcinoma (PDAC) is its extensively reprogrammed metabolic network, in which nicotinamide adenine dinucleotide (NAD) and its reduced form, NADH, are critical cofactors. Here, we show that IFN signaling, present in a subset of PDAC tumors, substantially lowers NAD(H) levels through up-regulating the expression of NAD-consuming enzymes PARP9, PARP10, and PARP14. Their individual contributions to this mechanism in PDAC have not been previously delineated. Nicotinamide phosphoribosyltransferase (NAMPT) is the rate-limiting enzyme in the NAD salvage pathway, a dominant source of NAD in cancer cells. We found that IFN-induced NAD consumption increased dependence upon NAMPT for its role in recycling NAM to salvage NAD pools, thus sensitizing PDAC cells to pharmacologic NAMPT inhibition. Their combination decreased PDAC cell proliferation and invasion in vitro and suppressed orthotopic tumor growth and liver metastases in vivo.


Assuntos
Biomarcadores Tumorais/metabolismo , Carcinoma Ductal Pancreático/patologia , Citocinas/antagonistas & inibidores , Regulação Neoplásica da Expressão Gênica , Interferon Tipo I/metabolismo , NAD/deficiência , Nicotinamida Fosforribosiltransferase/antagonistas & inibidores , Neoplasias Pancreáticas/patologia , Animais , Apoptose , Biomarcadores Tumorais/genética , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/metabolismo , Proliferação de Células , Citocinas/genética , Citocinas/metabolismo , Humanos , Interferon Tipo I/genética , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Nicotinamida Fosforribosiltransferase/genética , Nicotinamida Fosforribosiltransferase/metabolismo , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Poli(ADP-Ribose) Polimerases/genética , Poli(ADP-Ribose) Polimerases/metabolismo , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Transdução de Sinais , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Cancer Res ; 80(22): 5024-5034, 2020 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-32998997

RESUMO

The aggressive primary brain tumor glioblastoma (GBM) is characterized by aberrant metabolism that fuels its malignant phenotype. Diverse genetic subtypes of malignant glioma are sensitive to selective inhibition of the NAD+ salvage pathway enzyme nicotinamide phosphoribosyltransferase (NAMPT). However, the potential impact of NAD+ depletion on the brain tumor microenvironment has not been elaborated. In addition, systemic toxicity of NAMPT inhibition remains a significant concern. Here we show that microparticle-mediated intratumoral delivery of NAMPT inhibitor GMX1778 induces specific immunologic changes in the tumor microenvironment of murine GBM, characterized by upregulation of immune checkpoint PD-L1, recruitment of CD3+, CD4+, and CD8+ T cells, and reduction of M2-polarized immunosuppressive macrophages. NAD+ depletion and autophagy induced by NAMPT inhibitors mediated the upregulation of PD-L1 transcripts and cell surface protein levels in GBM cells. NAMPT inhibitor modulation of the tumor immune microenvironment was therefore combined with PD-1 checkpoint blockade in vivo, significantly increasing the survival of GBM-bearing animals. Thus, the therapeutic impacts of NAMPT inhibition extended beyond neoplastic cells, shaping surrounding immune effectors. Microparticle delivery and release of NAMPT inhibitor at the tumor site offers a safe and robust means to alter an immune tumor microenvironment that could potentiate checkpoint immunotherapy for glioblastoma. SIGNIFICANCE: Microparticle-mediated local inhibition of NAMPT modulates the tumor immune microenvironment and acts cooperatively with anti-PD-1 checkpoint blockade, offering a combination immunotherapy strategy for the treatment of GBM.


Assuntos
Antígeno B7-H1/metabolismo , Neoplasias Encefálicas/terapia , Cianetos/administração & dosagem , Citocinas/antagonistas & inibidores , Glioblastoma/terapia , Guanidinas/administração & dosagem , NAD/efeitos dos fármacos , Nicotinamida Fosforribosiltransferase/antagonistas & inibidores , Microambiente Tumoral/efeitos dos fármacos , Acrilamidas/administração & dosagem , Animais , Autofagia , Antígeno B7-H1/antagonistas & inibidores , Antígeno B7-H1/genética , Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/mortalidade , Linfócitos T CD4-Positivos , Linfócitos T CD8-Positivos , Movimento Celular , Cianetos/efeitos adversos , Preparações de Ação Retardada , Portadores de Fármacos/síntese química , Glioblastoma/imunologia , Glioblastoma/metabolismo , Glioblastoma/mortalidade , Guanidinas/efeitos adversos , Humanos , Injeções Intralesionais , Macrófagos/efeitos dos fármacos , Proteínas de Membrana/metabolismo , Camundongos , NAD/análise , NAD/deficiência , Piperidinas/administração & dosagem , Polímeros/síntese química , RNA Mensageiro/metabolismo , Transdução de Sinais , Microambiente Tumoral/imunologia , Regulação para Cima/efeitos dos fármacos
8.
Med Hypotheses ; 144: 110044, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32758884

RESUMO

The SARS-CoV-2 hyperinflammatory response is associated with high mortality. This hypothesis suggests that a deficiency of nicotinamide adenine dinucleotide (NAD+) may be the primary factor related to the SARS-Cov-2 disease spectrum and the risk for mortality, as subclinical nutritional deficiencies may be unmasked by any significant increase in oxidative stress. NAD+ levels decline with age and are also reduced in conditions associated with oxidative stress as occurs with hypertension, diabetes and obesity. These groups have also been observed to have high mortality following infection with COVID-19. Further consumption of NAD+ in a pre-existent depleted state is more likely to cause progression to the hyperinflammatory stage of the disease through its limiting effects on the production of SIRT1. This provides a unifying hypothesis as to why these groups are at high risk of mortality and suggests that nutritional support with NAD+ and SIRT1 activators, could minimise disease severity if administered prophylactically and or therapeutically. The significance of this, if proven, has far-reaching consequences in the management of COVID-19 especially in third world countries, where resources and finances are limited.


Assuntos
COVID-19/imunologia , Diabetes Mellitus Tipo 2/complicações , NAD/deficiência , Obesidade/complicações , Sirtuína 1/imunologia , Proteína ADAM17/imunologia , ADP-Ribosil Ciclase 1/imunologia , Fatores Etários , Idoso , Envelhecimento , COVID-19/mortalidade , Diabetes Mellitus Tipo 2/imunologia , Progressão da Doença , Suscetibilidade a Doenças , Humanos , Inflamação , Glicoproteínas de Membrana/imunologia , NAD/química , Obesidade/imunologia , Estresse Oxidativo , Ligação Proteica , Replicação Viral , Zinco/química
9.
Eur J Pharmacol ; 879: 173158, 2020 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-32360833

RESUMO

Nicotinamide adenine nucleotide (NAD) is a small ubiquitous hydrophilic cofactor that participates in several aspects of cellular metabolism. As a coenzyme it has an essential role in the regulation of energetic metabolism, but it is also a cosubstrate for enzymes that regulate fundamental biological processes such as transcriptional regulation, signaling and DNA repairing among others. The fluctuation and oxidative state of NAD levels regulate the activity of these enzymes, which is translated into marked effects on cellular function. While alterations in NAD homeostasis are a common feature of different conditions and age-associated diseases, in general, increased NAD levels have been associated with beneficial health effects. Due to its therapeutic potential, the interest in this molecule has been renewed, and the regulation of NAD metabolism has become an attractive target for drug discovery. In fact, different approaches to replenish or increase NAD levels have been tested, including enhancement of biosynthesis and inhibition of NAD breakdown. Despite further research is needed, this review provides an overview and update on NAD metabolism, including the therapeutic potential of its regulation, as well as pharmacokinetics, safety, precautions and formulation challenges of NAD supplementation.


Assuntos
NAD/metabolismo , Animais , Dieta , Suplementos Nutricionais , Humanos , NAD/deficiência
10.
Cell Metab ; 31(6): 1078-1090.e5, 2020 06 02.
Artigo em Inglês | MEDLINE | ID: mdl-32386566

RESUMO

NAD+ is a redox-active metabolite, the depletion of which has been proposed to promote aging and degenerative diseases in rodents. However, whether NAD+ depletion occurs in patients with degenerative disorders and whether NAD+ repletion improves their symptoms has remained open. Here, we report systemic NAD+ deficiency in adult-onset mitochondrial myopathy patients. We administered an increasing dose of NAD+-booster niacin, a vitamin B3 form (to 750-1,000 mg/day; clinicaltrials.govNCT03973203) for patients and their matched controls for 10 or 4 months, respectively. Blood NAD+ increased in all subjects, up to 8-fold, and muscle NAD+ of patients reached the level of their controls. Some patients showed anemia tendency, while muscle strength and mitochondrial biogenesis increased in all subjects. In patients, muscle metabolome shifted toward controls and liver fat decreased even 50%. Our evidence indicates that blood analysis is useful in identifying NAD+ deficiency and points niacin to be an efficient NAD+ booster for treating mitochondrial myopathy.


Assuntos
Miopatias Mitocondriais/metabolismo , Músculos/metabolismo , NAD/metabolismo , Niacina/metabolismo , Adolescente , Adulto , Idoso , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Miopatias Mitocondriais/patologia , Músculos/patologia , NAD/deficiência , Adulto Jovem
11.
Proc Natl Acad Sci U S A ; 117(7): 3738-3747, 2020 02 18.
Artigo em Inglês | MEDLINE | ID: mdl-32015132

RESUMO

Causes for miscarriages and congenital malformations can be genetic, environmental, or a combination of both. Genetic variants, hypoxia, malnutrition, or other factors individually may not affect embryo development, however, they may do so collectively. Biallelic loss-of-function variants in HAAO or KYNU, two genes of the nicotinamide adenine dinucleotide (NAD) synthesis pathway, are causative of congenital malformation and miscarriage in humans and mice. The variants affect normal embryonic development by disrupting the synthesis of NAD, a key factor in multiple biological processes, from its dietary precursor tryptophan, resulting in NAD deficiency. This study demonstrates that congenital malformations caused by NAD deficiency can occur independent of genetic disruption of NAD biosynthesis. C57BL/6J wild-type mice had offspring exhibiting similar malformations when their supply of the NAD precursors tryptophan and vitamin B3 in the diet was restricted during pregnancy. When the dietary undersupply was combined with a maternal heterozygous variant in Haao, which alone does not cause NAD deficiency or malformations, the incidence of embryo loss and malformations was significantly higher, suggesting a gene-environment interaction. Maternal and embryonic NAD levels were deficient. Mild hypoxia as an additional factor exacerbated the embryo outcome. Our data show that NAD deficiency as a cause of embryo loss and congenital malformation is not restricted to the rare cases of biallelic mutations in NAD synthesis pathway genes. Instead, monoallelic genetic variants and environmental factors can result in similar outcomes. The results expand our understanding of the causes of congenital malformations and the importance of sufficient NAD precursor consumption during pregnancy.


Assuntos
Aborto Espontâneo/genética , Anormalidades Congênitas/genética , Interação Gene-Ambiente , NAD/deficiência , Aborto Espontâneo/metabolismo , Animais , Anormalidades Congênitas/metabolismo , Embrião de Mamíferos/metabolismo , Feminino , Humanos , Masculino , Fenômenos Fisiológicos da Nutrição Materna , Camundongos , Camundongos Endogâmicos C57BL , Gravidez
12.
Am J Hum Genet ; 106(1): 129-136, 2020 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-31883644

RESUMO

Birth defects occur in up to 3% of all live births and are the leading cause of infant death. Here we present five individuals from four unrelated families, individuals who share similar phenotypes with disease-causal bi-allelic variants in NADSYN1, encoding NAD synthetase 1, the final enzyme of the nicotinamide adenine dinucleotide (NAD) de novo synthesis pathway. Defects range from the isolated absence of both kidneys to multiple malformations of the vertebrae, heart, limbs, and kidney, and no affected individual survived for more than three months postnatally. NAD is an essential coenzyme for numerous cellular processes. Bi-allelic loss-of-function mutations in genes required for the de novo synthesis of NAD were previously identified in individuals with multiple congenital abnormalities affecting the heart, kidney, vertebrae, and limbs. Functional assessments of NADSYN1 missense variants, through a combination of yeast complementation and enzymatic assays, show impaired enzymatic activity and severely reduced NAD levels. Thus, NADSYN1 represents an additional gene required for NAD synthesis during embryogenesis, and NADSYN1 has bi-allelic missense variants that cause NAD deficiency-dependent malformations. Our findings expand the genotypic spectrum of congenital NAD deficiency disorders and further implicate mutation of additional genes involved in de novo NAD synthesis as potential causes of complex birth defects.


Assuntos
Carbono-Nitrogênio Ligases com Glutamina como Doadora de N-Amida/genética , Anormalidades Congênitas/etiologia , Insuficiência de Múltiplos Órgãos/etiologia , Mutação de Sentido Incorreto , NAD/deficiência , Alelos , Sequência de Aminoácidos , Anormalidades Congênitas/patologia , Feminino , Genótipo , Idade Gestacional , Humanos , Lactente , Recém-Nascido , Masculino , Insuficiência de Múltiplos Órgãos/patologia , Linhagem , Fenótipo , Gravidez , Homologia de Sequência
13.
Int J Mol Sci ; 20(23)2019 Nov 26.
Artigo em Inglês | MEDLINE | ID: mdl-31779194

RESUMO

Nicotinamide (NAM) is an amide form of vitamin B3 and the precursor of nicotinamide adenine dinucleotide (NAD+), an essential co-enzyme of redox reactions for adenosine triphosphate (ATP) production and for other metabolic processes. As NAD+ status is critical in maintaining cellular energy, vitamin B3 deficiency mainly affects tissues that need high cellular energy causing pellagra and skin sun sensitivity. In animal models, NAD+ deficiency leads to UV sensitivity of the skin, impairs DNA damage response, and increases genomic instability and cancer incidence. Furthermore, NAD+ depletion is associated with human skin aging and cancer. NAM prevents the UV-induced ATP depletion boosting cellular energy and enhances DNA repair activity in vitro and in vivo. Moreover, NAM reduces skin cancer incidence and prevents the immune-suppressive effects of UV in mice. Thus, NAM is involved in the maintenance of genomic stability and may have beneficial effects against skin aging changes and tumor development. Clinical studies showed that topical use of NAM reduces cutaneous aging. Furthermore, oral NAM administration reduces the level of UV-mediated immunosuppression and lowers the rate of non-melanoma skin cancers in high-risk patients. Therefore, NAM replenishment strategy may be a promising approach for skin cancer chemoprevention.


Assuntos
Instabilidade Genômica , Niacinamida/deficiência , Envelhecimento da Pele/efeitos dos fármacos , Neoplasias Cutâneas/genética , Animais , Metabolismo Energético/efeitos dos fármacos , Humanos , Terapia de Imunossupressão , NAD/deficiência , Niacinamida/administração & dosagem , Niacinamida/farmacologia , Neoplasias Cutâneas/prevenção & controle , Raios Ultravioleta/efeitos adversos
14.
Geroscience ; 41(5): 533-542, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31679124

RESUMO

Adjustment of cerebral blood flow (CBF) to neuronal activity via neurovascular coupling (NVC) plays an important role in the maintenance of healthy cognitive function. Strong evidence demonstrates that age-related cerebromicrovascular endothelial dysfunction and consequential impairment of NVC responses contribute importantly to cognitive decline. Recent studies demonstrate that NAD+ availability decreases with age in the vasculature and that supplemental NAD+ precursors can ameliorate cerebrovascular dysfunction, rescuing NVC responses and improving cognitive performance in aged mice. The mechanisms underlying the age-related decline in [NAD+] in cells of the neurovascular unit are likely multifaceted and may include increased utilization of NAD+ by activated poly (ADP-ribose) polymerase (PARP-1). The present study was designed to test the hypothesis that inhibition of PARP-1 activity may confer protective effects on neurovascular function in aging, similar to the recently demonstrated protective effects of treatment with the NAD+ precursor nicotinamide mononucleotide (NMN). To test this hypothesis, 24-month-old C57BL/6 mice were treated with PJ-34, a potent PARP inhibitor, for 2 weeks. NVC was assessed by measuring CBF responses (laser speckle contrast imaging) in the somatosensory whisker barrel cortex evoked by contralateral whisker stimulation. We found that NVC responses were significantly impaired in aged mice. Treatment with PJ-34 improved NVC responses by increasing endothelial NO-mediated vasodilation, which was associated with significantly improved spatial working memory. PJ-34 treatment also improved endothelium-dependent acetylcholine-induced relaxation of aorta rings. Thus, PARP-1 activation, likely by decreasing NAD+ availability, contributes to age-related endothelial dysfunction and neurovascular uncoupling, exacerbating cognitive decline. The cerebromicrovascular protective effects of pharmacological inhibition of PARP-1 highlight the preventive and therapeutic potential of treatments that restore NAD+ homeostasis as effective interventions in patients at risk for vascular cognitive impairment (VCI).


Assuntos
Cognição/efeitos dos fármacos , Endotélio Vascular/efeitos dos fármacos , Acoplamento Neurovascular/efeitos dos fármacos , Fenantrenos/farmacologia , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia , Envelhecimento/fisiologia , Animais , Circulação Cerebrovascular/fisiologia , Modelos Animais de Doenças , Memória de Curto Prazo/fisiologia , Camundongos Endogâmicos C57BL , Microcirculação/fisiologia , NAD/deficiência , NAD/metabolismo , Óxido Nítrico/metabolismo , Vasodilatação/fisiologia
15.
Am J Physiol Heart Circ Physiol ; 316(6): H1253-H1266, 2019 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-30875255

RESUMO

Age-related alterations in endothelium and the resulting vascular dysfunction critically contribute to a range of pathological conditions associated with old age. To develop therapies rationally that improve vascular health and thereby increase health span and life span in older adults, it will be essential to understand the cellular and molecular mechanisms contributing to vascular aging. Preclinical studies in model organisms demonstrate that NAD+ availability decreases with age in multiple tissues and that supplemental NAD+ precursors can ameliorate many age-related cellular impairments. Here, we provide a comprehensive overview of NAD+-dependent pathways [including the NAD+-using silent information regulator-2-like enzymes and poly(ADP-ribose) polymerase enzymes] and the potential consequences of endothelial NAD+ deficiency in vascular aging. The multifaceted vasoprotective effects of treatments that reverse the age-related decline in cellular NAD+ levels, as well as their potential limitations, are discussed. The preventive and therapeutic potential of NAD+ intermediates as effective, clinically relevant interventions in older adults at risk for ischemic heart disease, vascular cognitive impairment, and other common geriatric conditions and diseases that involve vascular pathologies (e.g., sarcopenia, frailty) are critically discussed. We propose that NAD+ precursors [e.g., nicotinamide (Nam) riboside, Nam mononucleotide, niacin] should be considered as critical components of combination therapies to slow the vascular aging process and increase cardiovascular health span.


Assuntos
Envelhecimento/metabolismo , Endotélio Vascular/metabolismo , NAD/deficiência , Doenças Vasculares/metabolismo , Fatores Etários , Envelhecimento/patologia , Animais , Senescência Celular , Endotélio Vascular/patologia , Endotélio Vascular/fisiopatologia , Metabolismo Energético , Humanos , Estresse Oxidativo , Transdução de Sinais , Doenças Vasculares/patologia , Doenças Vasculares/fisiopatologia
16.
Antioxid Redox Signal ; 30(6): 890-905, 2019 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-29295624

RESUMO

SIGNIFICANCE: Increasing evidence has indicated critical roles of nicotinamide adenine dinucleotide, oxidized form (NAD+) in various biological functions. NAD+ deficiency has been found in models of a number of diseases such as cerebral ischemia, myocardial ischemia, and diabetes, and in models of aging. Applications of NAD+ or other approaches that can restore NAD+ levels are highly protective in these models of diseases and aging. NAD+ produces its beneficial effects by targeting at multiple pathological pathways, including attenuating mitochondrial alterations, DNA damage, and oxidative stress, by modulating such enzymes as sirtuins, glyceraldehyde-3-phosphate dehydrogenase, and AP endonuclease. These findings have suggested great therapeutic and nutritional potential of NAD+ for diseases and senescence. Recent Advances: Approaches that can restore NAD+ levels are highly protective in the models of such diseases as glaucoma. The NAD+ deficiency in the diseases and aging results from not only poly(ADP-ribose) polymerase-1 (PARP-1) activation but also decreased nicotinamide phosphoribosyltransferase (Nampt) activity and increased CD38 activity. Significant biological effects of extracellular NAD+ have been found. Increasing evidence has suggested that NAD+ deficiency is a common central pathological factor in a number of diseases and aging. Critical Issues and Future Directions: Future studies are required for solidly establishing the concept that "NAD+ deficiency is a common central pathological factor in a number of disease and aging." It is also necessary to further investigate the mechanisms underlying the NAD+ deficiency in the diseases and aging. Preclinical and clinical studies should be conducted to determine the therapeutic potential of NAD+ for the diseases and aging.


Assuntos
Envelhecimento/metabolismo , Isquemia Encefálica/metabolismo , Diabetes Mellitus/metabolismo , Isquemia Miocárdica/metabolismo , NAD/deficiência , NAD/uso terapêutico , Envelhecimento/efeitos dos fármacos , Envelhecimento/patologia , Animais , Isquemia Encefálica/tratamento farmacológico , Isquemia Encefálica/patologia , Diabetes Mellitus/tratamento farmacológico , Diabetes Mellitus/patologia , Humanos , Isquemia Miocárdica/tratamento farmacológico , Isquemia Miocárdica/patologia , NAD/metabolismo , NAD/farmacologia , Transdução de Sinais/efeitos dos fármacos
17.
Cell Rep ; 24(2): 429-440, 2018 07 10.
Artigo em Inglês | MEDLINE | ID: mdl-29996103

RESUMO

Mycobacterium tuberculosis (Mtb) kills infected macrophages by inhibiting apoptosis and promoting necrosis. The tuberculosis necrotizing toxin (TNT) is a secreted nicotinamide adenine dinucleotide (NAD+) glycohydrolase that induces necrosis in infected macrophages. Here, we show that NAD+ depletion by TNT activates RIPK3 and MLKL, key mediators of necroptosis. Notably, Mtb bypasses the canonical necroptosis pathway since neither TNF-α nor RIPK1 are required for macrophage death. Macrophage necroptosis is associated with depolarized mitochondria and impaired ATP synthesis, known hallmarks of Mtb-induced cell death. These results identify TNT as the main trigger of necroptosis in Mtb-infected macrophages. Surprisingly, NAD+ depletion itself was sufficient to trigger necroptosis in a RIPK3- and MLKL-dependent manner by inhibiting the NAD+ salvage pathway in THP-1 cells or by TNT expression in Jurkat T cells. These findings suggest avenues for host-directed therapies to treat tuberculosis and other infectious and age-related diseases in which NAD+ deficiency is a pathological factor.


Assuntos
Apoptose , Macrófagos/patologia , Mycobacterium tuberculosis/metabolismo , NAD/deficiência , Animais , Apoptose/efeitos dos fármacos , Toxinas Bacterianas/toxicidade , Biocatálise/efeitos dos fármacos , Citoproteção/efeitos dos fármacos , Humanos , Células Jurkat , Macrófagos/efeitos dos fármacos , Macrófagos/microbiologia , Camundongos Endogâmicos C57BL , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/patologia , Modelos Biológicos , Mycobacterium tuberculosis/efeitos dos fármacos , NAD/metabolismo , NAD+ Nucleosidase/metabolismo , Necrose , Niacinamida/farmacologia , Proteínas Quinases/metabolismo , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo , Células THP-1 , Fator de Necrose Tumoral alfa/metabolismo
18.
N Engl J Med ; 377(6): 544-552, 2017 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-28792876

RESUMO

BACKGROUND: Congenital malformations can be manifested as combinations of phenotypes that co-occur more often than expected by chance. In many such cases, it has proved difficult to identify a genetic cause. We sought the genetic cause of cardiac, vertebral, and renal defects, among others, in unrelated patients. METHODS: We used genomic sequencing to identify potentially pathogenic gene variants in families in which a person had multiple congenital malformations. We tested the function of the variant by using assays of in vitro enzyme activity and by quantifying metabolites in patient plasma. We engineered mouse models with similar variants using the CRISPR (clustered regularly interspaced short palindromic repeats)-Cas9 system. RESULTS: Variants were identified in two genes that encode enzymes of the kynurenine pathway, 3-hydroxyanthranilic acid 3,4-dioxygenase (HAAO) and kynureninase (KYNU). Three patients carried homozygous variants predicting loss-of-function changes in the HAAO or KYNU proteins (HAAO p.D162*, HAAO p.W186*, or KYNU p.V57Efs*21). Another patient carried heterozygous KYNU variants (p.Y156* and p.F349Kfs*4). The mutant enzymes had greatly reduced activity in vitro. Nicotinamide adenine dinucleotide (NAD) is synthesized de novo from tryptophan through the kynurenine pathway. The patients had reduced levels of circulating NAD. Defects similar to those in the patients developed in the embryos of Haao-null or Kynu-null mice owing to NAD deficiency. In null mice, the prevention of NAD deficiency during gestation averted defects. CONCLUSIONS: Disruption of NAD synthesis caused a deficiency of NAD and congenital malformations in humans and mice. Niacin supplementation during gestation prevented the malformations in mice. (Funded by the National Health and Medical Research Council of Australia and others.).


Assuntos
3-Hidroxiantranilato 3,4-Dioxigenase/genética , Anormalidades Congênitas/genética , Suplementos Nutricionais , Hidrolases/genética , NAD/deficiência , Niacina/uso terapêutico , 3-Hidroxiantranilato 3,4-Dioxigenase/metabolismo , Canal Anal/anormalidades , Animais , Anormalidades Congênitas/prevenção & controle , Modelos Animais de Doenças , Esôfago/anormalidades , Feminino , Cardiopatias Congênitas/genética , Cardiopatias Congênitas/prevenção & controle , Humanos , Hidrolases/metabolismo , Rim/anormalidades , Deformidades Congênitas dos Membros/genética , Deformidades Congênitas dos Membros/prevenção & controle , Masculino , Camundongos , Camundongos Knockout , Mutação , NAD/biossíntese , NAD/genética , Análise de Sequência de DNA , Coluna Vertebral/anormalidades , Traqueia/anormalidades
20.
Trends Cancer ; 3(8): 593-610, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28780936

RESUMO

The phenomenon of aging has gained widespread attention in recent times. Although significant advances have been made to better understand aging and its related pathologies including cancer, there is not yet a clear mechanism explaining why diseases and cancer are inherent parts of the aging process. Finding a unifying equation that could bridge aging and its related diseases would allow therapeutic development and solve an immense human health problem to live longer and better. In this review, we discuss NAD+ reduction as the central mechanism that may connect aging to its related pathologies and cancer. NAD+ boosters would ensure and ameliorate health quality during aging.


Assuntos
Envelhecimento/metabolismo , Longevidade , NAD/deficiência , Neoplasias/metabolismo , Complexo Vitamínico B/uso terapêutico , Envelhecimento/sangue , Humanos , Incidência , Expectativa de Vida/tendências , NAD/biossíntese , NAD/sangue , Neoplasias/sangue , Neoplasias/tratamento farmacológico , Neoplasias/epidemiologia , Niacinamida/análogos & derivados , Niacinamida/uso terapêutico , Mononucleotídeo de Nicotinamida/uso terapêutico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...