Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Exp Cell Res ; 395(2): 112203, 2020 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-32738344

RESUMO

Cellular senescence is defined by irreversible cell-cycle arrest and is an evolutionarily conserved hallmark of aging. In this study, we generate senescent microglial cells via exposure to the chemotherapy drug doxorubicin. Compared to control cells, doxorubicin-treated microglia exhibited an altered morphology characterized by an enlarged cell size, a flattened appearance, and the development of prominent filaments. Senescent cells harbored elevated levels of senescence associated-ß-galactosidase, p16Ink4a, and γ-H2AX. Senescent microglia were also less efficient at internalizing amyloid ß and pHrodo bioparticles. A detailed proteomic analysis using SWATH-MS identified 201 proteins that were significantly downregulated and 127 that were significantly upregulated in doxorubicin-treated microglia. Proteins involved in processes such as protein synthesis, RNA damage and repair, and protein degradation were largely downregulated while those compromising the integrity of the cell were predominantly upregulated. Various proteins involved in proteasomal processing were among the most significantly downregulated in senescent cells. Relevant to the deleterious senescence-associated secretory phenotype, senescent cells secreted higher levels of the inflammatory cytokines IL-6, IL-8, TNF-α, and GRO-α. Our data suggest that symptoms of brain aging and age-related neurodegenerative disease may be partially caused by defective phagocytosis, impaired proteasomal processing, and elevated cytokine secretion of senescent microglia.


Assuntos
Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Citocinas/metabolismo , Doxorrubicina/farmacologia , Microglia/efeitos dos fármacos , Doenças Neurodegenerativas/tratamento farmacológico , Envelhecimento/metabolismo , Peptídeos beta-Amiloides/efeitos dos fármacos , Peptídeos beta-Amiloides/metabolismo , Comunicação Celular/fisiologia , Senescência Celular/efeitos dos fármacos , Senescência Celular/fisiologia , Citocinas/efeitos dos fármacos , Doxorrubicina/metabolismo , Expressão Gênica/efeitos dos fármacos , Humanos , Microglia/metabolismo , Doenças Neurodegenerativas/metabolismo
2.
BMC Biotechnol ; 16(1): 85, 2016 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-27903244

RESUMO

BACKGROUND: Preservation of human skin fibroblasts and keratinocytes is essential for the creation of skin tissue banks. For successful cryopreservation of cells, selection of an appropriate cryoprotectant agent (CPA) is imperative. The aim of this study was to identify CPAs that minimize toxic effects and allow for the preservation of human fibroblasts and keratinocytes in suspension and in monolayers. RESULTS: We cryopreserved human fibroblasts and keratinocytes with different CPAs and compared them to fresh, unfrozen cells. Cells were frozen in the presence and absence of hydroxyethyl starch (HES) or dimethyl sulfoxide (DMSO), the latter of which is a commonly used CPA known to exert toxic effects on cells. Cell numbers were counted immediately post-thaw as well as three days after thawing. Cellular structures were analyzed and counted by labeling nuclei, mitochondria, and actin filaments. We found that successful cryopreservation of suspended or adherent keratinocytes can be accomplished with a 10% HES or a 5% HES, 5% DMSO solution. Cell viability of fibroblasts cryopreserved in suspension was maintained with 10% HES or 5% HES, 5% DMSO solutions. Adherent, cryopreserved fibroblasts were successfully maintained with a 5% HES, 5% DMSO solution. CONCLUSION: We conclude that skin tissue cells can be effectively cryopreserved by substituting all or a portion of DMSO with HES. Given that DMSO is the most commonly used CPA and is believed to be more toxic than HES, these findings are of clinical significance for tissue-based replacement therapies. Therapies that require the use of keratinocyte and fibroblast cells, such as those aimed at treating skin wounds or skin burns, may be optimized by substituting a portion or all of DMSO with HES during cryopreservation protocols.


Assuntos
Criopreservação/métodos , Fibroblastos/efeitos dos fármacos , Fibroblastos/fisiologia , Derivados de Hidroxietil Amido/farmacologia , Queratinócitos/efeitos dos fármacos , Queratinócitos/fisiologia , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Células Cultivadas , Crioprotetores/química , Crioprotetores/farmacologia , Fibroblastos/citologia , Humanos , Queratinócitos/citologia , Masculino , Pele/citologia , Pele/efeitos dos fármacos , Fenômenos Fisiológicos da Pele/efeitos dos fármacos
3.
Exp Cell Res ; 338(1): 97-104, 2015 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-26335540

RESUMO

Mesenchymal stem cells (MSCs) are non-immunogenic, multipotent cells with at least trilineage differentiation potential. They promote wound healing, improve regeneration of injured tissue, and mediate numerous other health effects. MSCs migrate to sites of injury and stimulate repair either through direct differentiation or indirectly through the stimulation of endogenous repair mechanisms. Using the in vitro scratch assay, we show that the inflammatory cytokines, chemokines, and growth factors TNF-α, SDF-1, PDGF, and bFGF enhance migration of rat MSCs under normoxic conditions, while TNF-α, IFN-γ, PDGF, and bFGF promote MSC migration under hypoxic conditions. This indicates that the oxygen concentration affects how MSCs will migrate in response to specific factors and, consistent with this, differential expression of cytokines was observed under hypoxic versus normoxic conditions. Using the transwell migration assay, we find that TNF-α, IFN-γ, bFGF, IGF-1, PDGF, and SDF-1 significantly increase transmigration of rat MSCs compared to unstimulated medium. MSCs derived from aged rats exhibited comparable migration to MSCs derived from young rats under hypoxic and normoxic conditions, even after application with specific factors. Similarly, migration in MSCs from aged, human donors did not statistically differ compared to migration in MSCs derived from human umbilical cord tissue or younger donors.


Assuntos
Movimento Celular , Citocinas/fisiologia , Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Células-Tronco Mesenquimais/fisiologia , Envelhecimento , Animais , Diferenciação Celular , Hipóxia Celular , Células Cultivadas , Senescência Celular , Humanos , Oxigênio/fisiologia , Ratos Sprague-Dawley
4.
Hum Mol Genet ; 22(23): 4688-97, 2013 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-23825107

RESUMO

Mutations in bestrophin-1 (Best1) cause Best vitelliform macular dystrophy (BVMD), a dominantly inherited retinal degenerative disease. Best1 is a homo-oligomeric anion channel localized to the basolateral surface of retinal pigment epithelial (RPE) cells. A number of Best1 mutants mislocalize in Madin-Darby canine kidney (MDCK) cells. However, many proteins traffic differently in MDCK and RPE cells, and MDCK cells do not express endogenous Best1. Thus, effects of Best1 mutations on localization in MDCK cells may not translate to RPE cells. To determine whether BVMD causing mutations affect Best1 localization, we compared localization and oligomerization of Best1 with Best1 mutants V9M, W93C, and R218C. In MDCK cells, Best1 and Best1(R218C) were basolaterally localized. Best1(W93C) and Best1(V9M) accumulated in cells. In cultured fetal human retinal pigment epithelium cells (fhRPE) expressing endogenous Best1, Best1(R218C) and Best1(W93C) were basolateral. Best1(V9M) was intracellular. All three mutants exhibited similar fluorescence resonance energy transfer (FRET) efficiencies to, and co-immunoprecipitated with Best1, indicating unimpaired oligomerization. When human Best1 was expressed in RPE in mouse eyes it was basolaterally localized. However, Best1(V9M) accumulated in intracellular compartments in mouse RPE. Co-expression of Best1 and Best1(W93C) in MDCK cells resulted in basolateral localization of both Best1 and Best1(W93C), but co-expression of Best1 with Best1(V9M) resulted in mislocalization of both proteins. We conclude that different mutations in Best1 cause differential effects on its localization and that this effect varies with the presence or absence of wild-type (WT) Best1. Furthermore, MDCK cells can substitute for RPE when examining the effects of BVMD causing mutations on Best1 localization if co-expressed with WT Best1.


Assuntos
Canais de Cloreto/metabolismo , Proteínas do Olho/metabolismo , Canais Iônicos/metabolismo , Distrofia Macular Viteliforme/patologia , Animais , Bestrofinas , Sinalização do Cálcio , Membrana Celular/metabolismo , Células Cultivadas , Canais de Cloreto/genética , Olho/metabolismo , Proteínas do Olho/genética , Regulação da Expressão Gênica , Humanos , Canais Iônicos/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Mutagênese Sítio-Dirigida , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Mutação de Sentido Incorreto , Transporte Proteico/genética , Distrofia Macular Viteliforme/genética
5.
Mol Vis ; 21: 347-59, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25878489

RESUMO

PURPOSE: Mutations in BEST1, encoding Bestrophin-1 (Best1), cause Best vitelliform macular dystrophy (BVMD) and other inherited retinal degenerative diseases. Best1 is an integral membrane protein localized to the basolateral plasma membrane of the retinal pigment epithelium (RPE). Data from numerous in vitro and in vivo models have demonstrated that Best1 regulates intracellular Ca2+ levels. Although it is known from in vitro and crystal structure data that Best1 is also a calcium-activated anion channel, evidence for Best1 functioning as a channel in human RPE is lacking. To assess Best1-associated channel activity in the RPE, we examined the transepithelial electrical properties of fetal human RPE (fhRPE) cells, which express endogenous Best1. METHODS: Using adenovirus-mediated gene transfer, we overexpressed Best1 and the BVMD mutant Best1W93C in fhRPE cells and assessed resting transepithelial potential (TEP), transepithelial resistance, short circuit current (Isc), and intracellular Ca2+ levels. Cl- currents were directly measured in transfected HEK293 cells using whole-cell patch clamp. RESULTS: Best1W93C showed ablated Cl- currents and, when co-expressed, suppressed the channel activity of Best1 in HEK293 cells. In fhRPE, overexpression of Best1 increased TEP and Isc, while Best1W93C diminished TEP and Isc. Substitution of Cl- in the bath media resulted in a significant reduction of Isc in monolayers overexpressing Best1, but no significant Isc change in monolayers expressing Best1W93C. We removed Ca2+ as a limit on transepithelial electrical properties by treating cells with ionomycin, and found that changes in Isc and TEP for monolayers expressing Best1 were absent in monolayers expressing Best1W93C. Similarly, inhibition of calcium-activated anion channels with niflumic acid reduced both Isc and TEP of control and Best1 monolayers, but did not notably affect Best1W93C monolayers. Stimulation with extracellular ATP induced an increase in TEP in control monolayers that was greater than that observed in those expressing Best1(W93C). Examination of [Ca2+]i following ATP stimulation demonstrated that the expression of Best1W93C impaired intracellular Ca2+ signaling. CONCLUSIONS: These data indicate that Best1 activity strongly influences electrophysiology and Ca2+ signaling in RPE cells, and that a common BVMD mutation disrupts both of these parameters. Our findings support the hypothesis that Best1 functions as an anion channel in human RPE.


Assuntos
Sinalização do Cálcio , Cálcio/metabolismo , Membrana Celular/metabolismo , Canais de Cloreto/metabolismo , Células Epiteliais/metabolismo , Proteínas do Olho/metabolismo , Epitélio Pigmentado da Retina/metabolismo , Trifosfato de Adenosina/farmacologia , Adenovírus Humanos/genética , Bestrofinas , Membrana Celular/efeitos dos fármacos , Canais de Cloreto/genética , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Proteínas do Olho/genética , Feto , Expressão Gênica , Vetores Genéticos , Células HEK293 , Humanos , Transporte de Íons/efeitos dos fármacos , Ionomicina/farmacologia , Potenciais da Membrana/efeitos dos fármacos , Mutação , Ácido Niflúmico/farmacologia , Técnicas de Patch-Clamp , Cultura Primária de Células , Epitélio Pigmentado da Retina/citologia , Epitélio Pigmentado da Retina/efeitos dos fármacos , Transfecção , Distrofia Macular Viteliforme/genética , Distrofia Macular Viteliforme/metabolismo , Distrofia Macular Viteliforme/patologia
6.
Proc Natl Acad Sci U S A ; 109(4): 1199-203, 2012 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-22223659

RESUMO

Body temperature is maintained in a narrow range in mammals, primarily controlled by sweating. In humans, the dynamic thermoregulatory organ, comprised of 2-4 million sweat glands distributed over the body, can secrete up to 4 L of sweat per day, thereby making it possible to withstand high temperatures and endure prolonged physical stress (e.g., long-distance running). The genetic basis for sweat gland function, however, is largely unknown. We find that the forkhead transcription factor, FoxA1, is required to generate mouse sweating capacity. Despite continued sweat gland morphogenesis, ablation of FoxA1 in mice results in absolute anihidrosis (lack of sweating). This inability to sweat is accompanied by down-regulation of the Na-K-Cl cotransporter 1 (Nkcc1) and the Ca(2+)-activated anion channel Bestrophin 2 (Best2), as well as glycoprotein accumulation in gland lumens and ducts. Furthermore, Best2-deficient mice display comparable anhidrosis and glycoprotein accumulation. These findings link earlier observations that both sodium/potassium/chloride exchange and Ca(2+) are required for sweat production. FoxA1 is inferred to regulate two corresponding features of sweat secretion. One feature, via Best2, catalyzes a bicarbonate gradient that could help to drive calcium-associated ionic transport; the other, requiring Nkcc1, facilitates monovalent ion exchange into sweat. These mechanistic components can be pharmaceutical targets to defend against hyperthermia and alleviate defective thermoregulation in the elderly, and may provide a model relevant to more complex secretory processes.


Assuntos
Regulação da Temperatura Corporal/fisiologia , Canais de Cloreto/metabolismo , Proteínas do Olho/metabolismo , Regulação da Expressão Gênica/fisiologia , Fator 3-alfa Nuclear de Hepatócito/metabolismo , Simportadores de Cloreto de Sódio-Potássio/metabolismo , Sudorese/fisiologia , Análise de Variância , Animais , Bestrofinas , Western Blotting , Cruzamentos Genéticos , Primers do DNA/genética , Imunofluorescência , Galactosídeos , Perfilação da Expressão Gênica , Genótipo , Fator 3-alfa Nuclear de Hepatócito/genética , Indóis , Camundongos , Modelos Biológicos , Reação em Cadeia da Polimerase em Tempo Real , Membro 2 da Família 12 de Carreador de Soluto , Sudorese/genética
7.
Exp Eye Res ; 121: 74-85, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24560797

RESUMO

BEST1 encodes Bestrophin-1 (Best1), a homo-oligomeric, integral membrane protein localized to the basolateral plasma membrane of the retinal pigment epithelium. Mutations in BEST1 cause five distinct retinal degenerative diseases, including adult vitelliform macular dystrophy (AVMD), autosomal recessive bestrophinopathy (ARB), autosomal dominant vitreoretinochoroidopathy (ADVIRC), and retinitis pigmentosa (RP). The mechanisms underlying these diseases and why mutations cause one disease over another are, for the most part, unknown. To gain insights into these four diseases, we expressed 28 Best1 mutants fused to YFP in polarized MDCK monolayers and, via confocal microscopy and immunofluorescence, live-cell FRET, and reciprocal co-immunoprecipitation experiments, screened these mutants for defects in localization and oligomerization. All 28 mutants exhibited comparable FRET efficiencies to and co-immunoprecipitated with WT Best1, indicating unimpaired oligomerization. RP- and ADVIRC-associated mutants were properly localized to the basolateral plasma membrane of cells, while two AVMD and most ARB mutants were mislocalized. When co-expressed, all mislocalized mutants caused mislocalization of WT Best1 to intracellular compartments. Our current and past results indicate that mislocalization of Best1 is not an absolute feature of any individual bestrophinopathy, occurring in AVMD, BVMD, and ARB. Furthermore, some ARB mutants that do not also cause dominant disease cause mislocalization of Best1, indicating that mislocalization is not a cause of disease, and that absence of Best1 activity from the plasma membrane is tolerated. Lastly, we find that the ARB truncation mutants L174Qfs*57 and R200X can form oligomers with WT Best1, indicating that the first ∼174 amino acids of Best1 are sufficient for oligomerization to occur.


Assuntos
Canais de Cloreto/genética , Canais de Cloreto/metabolismo , Oftalmopatias Hereditárias/genética , Proteínas do Olho/genética , Proteínas do Olho/metabolismo , Mutação de Sentido Incorreto , Multimerização Proteica/fisiologia , Doenças Retinianas/genética , Adenoviridae/genética , Animais , Proteínas de Bactérias/metabolismo , Bestrofinas , Western Blotting , Doenças da Coroide/genética , Doenças da Coroide/metabolismo , Cães , Eletrofisiologia , Oftalmopatias Hereditárias/metabolismo , Transferência Ressonante de Energia de Fluorescência , Corantes Fluorescentes/metabolismo , Expressão Gênica , Proteínas de Fluorescência Verde/metabolismo , Células HEK293 , Humanos , Proteínas Luminescentes/metabolismo , Células Madin Darby de Rim Canino/metabolismo , Microscopia Confocal , Técnicas de Patch-Clamp , Degeneração Retiniana/genética , Degeneração Retiniana/metabolismo , Doenças Retinianas/metabolismo , Retinose Pigmentar/genética , Retinose Pigmentar/metabolismo , Transfecção , Distrofia Macular Viteliforme/genética , Distrofia Macular Viteliforme/metabolismo
8.
Front Nutr ; 11: 1359176, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38476603

RESUMO

NAD+, a pivotal coenzyme central to metabolism, exhibits a characteristic decline with age. In mice, NAD+ levels can be elevated via treatment with apigenin, a natural flavonoid that inhibits the NAD+-consuming glycoprotein CD38. In animal models, apigenin positively impacts both sleep and longevity. For example, apigenin improves learning and memory in older mice, reduces tumor proliferation in a mouse xenograft model of triple-negative breast cancer, and induces sedative effects in mice and rats. Moreover, apigenin elongates survival in fly models of neurodegenerative disease and apigenin glycosides increase lifespan in worms. Apigenin's therapeutic potential is underscored by human clinical studies using chamomile extract, which contains apigenin as an active ingredient. Collectively, chamomile extract has been reported to alleviate anxiety, improve mood, and relieve pain. Furthermore, dietary apigenin intake positively correlates with sleep quality in a large cohort of adults. Apigenin's electron-rich flavonoid structure gives it strong bonding capacity to diverse molecular structures across receptors and enzymes. The effects of apigenin extend beyond CD38 inhibition, encompassing agonistic and antagonistic modulation of various targets, including GABA and inflammatory pathways. Cumulatively, a large body of evidence positions apigenin as a unique molecule capable of influencing both aging and sleep. Further studies are warranted to better understand apigenin's nuanced mechanisms and clinical potential.

9.
Geroscience ; 46(3): 3429-3443, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38441802

RESUMO

Epigenetic aging clocks are computational models that predict age using DNA methylation information. Initially, first-generation clocks were developed to make predictions using CpGs that change with age. Over time, next-generation clocks were created using CpGs that relate to both age and health. Since existing next-generation clocks were constructed in blood, we sought to develop a next-generation clock optimized for prediction in cheek swabs, which are non-invasive and easy to collect. To do this, we collected MethylationEPIC data as well as lifestyle and health information from 8045 diverse adults. Using a novel simulated annealing approach that allowed us to incorporate lifestyle and health factors into training as well as a combination of CpG filtering, CpG clustering, and clock ensembling, we constructed CheekAge, an epigenetic aging clock that has a strong correlation with age, displays high test-retest reproducibility across replicates, and significantly associates with a plethora of lifestyle and health factors, such as BMI, smoking status, and alcohol intake. We validated CheekAge in an internal dataset and multiple publicly available datasets, including samples from patients with progeria or meningioma. In addition to exploring the underlying biology of the data and clock, we provide a free online tool that allows users to mine our methylomic data and predict epigenetic age.


Assuntos
Envelhecimento , Epigênese Genética , Humanos , Reprodutibilidade dos Testes , Ilhas de CpG , Envelhecimento/genética , Estilo de Vida
10.
Ageing Res Rev ; 87: 101922, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37004845

RESUMO

The restriction of calories, branched-chain amino acids, and methionine have all been shown to extend lifespan in model organisms. Recently, glycine was found to boost longevity in genetically heterogenous mice. This simple amino acid similarly extends lifespan in rats and improves health in mammalian models of age-related disease. While compelling data indicate that glycine is a pro-longevity molecule, divergent mechanisms may underlie its effects on aging. Glycine is abundant in collagen, a building block for glutathione, a precursor to creatine, and an acceptor for the enzyme glycine N-methyltransferase (GNMT). A review of the literature strongly implicates GNMT, which clears methionine from the body by taking a methyl group from S-adenosyl-L-methionine and methylating glycine to form sarcosine. In flies, Gnmt is required for reduced insulin/insulin-like growth factor 1 signaling and dietary restriction to fully extend lifespan. The geroprotector spermidine requires Gnmt to upregulate autophagy genes and boost longevity. Moreover, the overexpression of Gnmt is sufficient to extend lifespan and reduce methionine levels. Sarcosine, or methylglycine, declines with age in multiple species and is capable of inducing autophagy both in vitro and in vivo. Taken all together, existing evidence suggests that glycine prolongs life by mimicking methionine restriction and activating autophagy.


Assuntos
Glicina , Sarcosina , Ratos , Animais , Camundongos , Humanos , Glicina/metabolismo , Envelhecimento/metabolismo , Metionina/metabolismo , Longevidade , Glicina N-Metiltransferase/genética , Glicina N-Metiltransferase/metabolismo , Racemetionina , Mamíferos/metabolismo
11.
Ageing Res Rev ; 81: 101721, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36029998

RESUMO

Alzheimer's disease (AD) is an incredibly complex and presently incurable age-related brain disorder. To better understand this debilitating disease, we collated and performed a meta-analysis on publicly available RNA-Seq, microarray, proteomics, and microRNA samples derived from AD patients and non-AD controls. 4089 samples originating from brain tissues and blood remained after applying quality filters. Since disease progression in AD correlates with age, we stratified this large dataset into three different age groups: < 75 years, 75-84 years, and ≥ 85 years. The RNA-Seq, microarray, and proteomics datasets were then combined into different integrated datasets. Ensemble machine learning was employed to identify genes and proteins that can accurately classify samples as either AD or control. These predictive inputs were then subjected to network-based enrichment analyses. The ability of genes/proteins associated with different pathways in the Molecular Signatures Database to diagnose AD was also tested. We separately identified microRNAs that can be used to make an AD diagnosis and subjected the predicted gene targets of the most predictive microRNAs to an enrichment analysis. The following key themes emerged from our machine learning and bioinformatics analyses: cell death, cellular senescence, energy metabolism, genomic integrity, glia, immune system, metal ion homeostasis, oxidative stress, proteostasis, and synaptic function. Many of the results demonstrated unique age-specificity. For example, terms highlighting cellular senescence only emerged in the earliest and intermediate age ranges while the majority of results relevant to cell death appeared in the youngest patients. Existing literature corroborates the importance of these hallmarks in AD.


Assuntos
Doença de Alzheimer , MicroRNAs , Fatores Etários , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/diagnóstico , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Encéfalo/metabolismo , Humanos , Aprendizado de Máquina , MicroRNAs/genética , MicroRNAs/metabolismo
12.
Aging Cell ; 21(8): e13664, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35778957

RESUMO

Although chronological age correlates with various age-related diseases and conditions, it does not adequately reflect an individual's functional capacity, well-being, or mortality risk. In contrast, biological age provides information about overall health and indicates how rapidly or slowly a person is aging. Estimates of biological age are thought to be provided by aging clocks, which are computational models (e.g., elastic net) that use a set of inputs (e.g., DNA methylation sites) to make a prediction. In the past decade, aging clock studies have shown that several age-related diseases, social variables, and mental health conditions associate with an increase in predicted biological age relative to chronological age. This phenomenon of age acceleration is linked to a higher risk of premature mortality. More recent research has demonstrated that predicted biological age is sensitive to specific interventions. Human trials have reported that caloric restriction, a plant-based diet, lifestyle changes involving exercise, a drug regime including metformin, and vitamin D3 supplementation are all capable of slowing down or reversing an aging clock. Non-interventional studies have connected high-quality sleep, physical activity, a healthy diet, and other factors to age deceleration. Specific molecules have been associated with the reduction or reversal of predicted biological age, such as the antihypertensive drug doxazosin or the metabolite alpha-ketoglutarate. Although rigorous clinical trials are needed to validate these initial findings, existing data suggest that aging clocks are malleable in humans. Additional research is warranted to better understand these computational models and the clinical significance of lowering or reversing their outputs.


Assuntos
Metilação de DNA , Epigênese Genética , Envelhecimento/genética , Restrição Calórica , Metilação de DNA/genética , Humanos , Estilo de Vida
13.
iScience ; 25(11): 105304, 2022 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-36304118

RESUMO

Epigenetic aging clocks are computational models that use DNA methylation sites to predict age. Since cheek swabs are non-invasive and painless, collecting DNA from buccal tissue is highly desirable. Here, we review 11 existing clocks that have been applied to buccal tissue. Two of these were exclusively trained on adults and, while moderately accurate, have not been used to capture health-relevant differences in epigenetic age. Using 130 common CpGs utilized by two or more existing buccal clocks, we generate a proof-of-concept predictor in an adult methylomic dataset. In addition to accurately estimating age (r = 0.95 and mean absolute error = 3.88 years), this clock predicted that Down syndrome subjects were significantly older relative to controls. A literature and database review of CpG-associated genes identified numerous genes (e.g., CLOCK, ELOVL2, and VGF) and molecules (e.g., alpha-linolenic acid, glycine, and spermidine) reported to influence lifespan and/or age-related disease in model organisms.

14.
Aging Cell ; 20(1): e13280, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33336875

RESUMO

Aging in humans is an incredibly complex biological process that leads to increased susceptibility to various diseases. Understanding which genes are associated with healthy aging can provide valuable insights into aging mechanisms and possible avenues for therapeutics to prolong healthy life. However, modeling this complex biological process requires an enormous collection of high-quality data along with cutting-edge computational methods. Here, we have compiled a large meta-analysis of gene expression data from RNA-Seq experiments available from the Sequence Read Archive. We began by reprocessing more than 6000 raw samples-including mapping, filtering, normalization, and batch correction-to generate 3060 high-quality samples spanning a large age range and multiple different tissues. We then used standard differential expression analyses and machine learning approaches to model and predict aging across the dataset, achieving an R2 value of 0.96 and a root-mean-square error of 3.22 years. These models allow us to explore aging across health status, sex, and tissue and provide novel insights into possible aging processes. We also explore how preprocessing parameters affect predictions and highlight the reproducibility limits of these machine learning models. Finally, we develop an online tool for predicting the ages of human transcriptomic samples given raw gene expression counts. Together, this study provides valuable resources and insights into the transcriptomics of human aging.


Assuntos
Aprendizado de Máquina/normas , Transcriptoma/genética , Envelhecimento , Nível de Saúde , Humanos , Fatores Sexuais
15.
Ageing Res Rev ; 70: 101404, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34242807

RESUMO

Machine learning models capable of predicting age given a set of inputs are referred to as aging clocks. We recently developed an aging clock that utilizes 491 plasma protein inputs, has an exceptional accuracy, and is capable of measuring biological age. Here, we demonstrate that this clock is extremely predictive (r = 0.95) when used to measure age in a novel plasma proteomic dataset derived from 370 human subjects aged 18-69 years. Over-representation analyses of the proteins that make up this clock in the Gene Ontology and Reactome databases predominantly implicated innate and adaptive immune system processes. Immunological drugs and various age-related diseases were enriched in the DrugBank and GLAD4U databases. By performing an extensive literature review, we find that at least 269 (54.8 %) of these inputs regulate lifespan and/or induce changes relevant to age-related disease when manipulated in an animal model. We also show that, in a large plasma proteomic dataset, the majority (57.2 %) of measurable clock proteins significantly change their expression level with human age. Different subsets of proteins were overlapped with distinct epigenetic, transcriptomic, and proteomic aging clocks. These findings indicate that the inputs of this age predictor likely represent a rich source of anti-aging drug targets.


Assuntos
Envelhecimento , Preparações Farmacêuticas , Idoso , Animais , Metilação de DNA , Epigênese Genética , Epigenômica , Humanos , Longevidade , Proteômica
16.
Rejuvenation Res ; 24(5): 377-389, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34486398

RESUMO

In our recent transcriptomic meta-analysis, we used random forest machine learning to accurately predict age in human blood, bone, brain, heart, and retina tissues given gene inputs. Although each tissue-specific model utilized a unique number of genes for age prediction, we found that the following six genes were prioritized in all five tissues: CHI3L2, CIDEC, FCGR3A, RPS4Y1, SLC11A1, and VTCN1. Since being selected for age prediction in multiple tissues is unique, we decided to explore these pan-tissue clock genes in greater detail. In the present study, we began by performing over-representation and network topology-based enrichment analyses in the Gene Ontology Biological Process database. These analyses revealed that the immunological terms "response to protozoan," "immune response," and "positive regulation of immune system process" were significantly enriched by these clock inputs. Expression analyses in mouse and human tissues identified that these inputs are frequently upregulated or downregulated with age. A detailed literature search showed that all six genes had noteworthy connections to age-related disease. For example, mice deficient in Cidec are protected against various metabolic defects, while suppressing VTCN1 inhibits age-related cancers in mouse models. Using a large multitissue transcriptomic dataset, we additionally generate a novel, minimalistic aging clock that can predict human age using just these six genes as inputs. Taken all together, these six genes are connected to diverse aspects of aging.


Assuntos
Envelhecimento , Aprendizado de Máquina , Envelhecimento/genética , Animais , Perfilação da Expressão Gênica , Ontologia Genética , Sistema Imunitário , Camundongos
17.
Rejuvenation Res ; 23(2): 107-121, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-31426688

RESUMO

Compelling evidence suggests that lipid metabolism, which plays critical roles in fat storage, cell membrane maintenance, and cell signaling, is intricately linked to aging. Lipid hydrolases are important enzymes that catalyze the hydrolysis of more complex lipids into simpler lipids. Diverse interventions targeting lipid hydrolases can prolong or shorten life in model organisms. For example, the genetic removal of or RNAi knockdown against a phospholipase can reduce lifespan in Caenorhabditis elegans, Drosophila melanogaster, and Mus musculus. The removal of lysosomal acid lipase results in premature death in mice, while its overexpression in nematodes generates lean, long-lived individuals. The overexpression or inhibition of diacylglycerol lipase leads to enhanced or reduced longevity, respectively, in both worms and flies. Lifespan can also be extended by knocking down triacylglycerol lipases in yeast, overexpressing fatty acid amide hydrolase in worms, or removing hepatic lipase in a mouse model of coronary disease. Conversely, flies lacking the triacylglycerol lipase Brummer are obese and short lived. Linking sphingolipids and aging, removing the sphingomyelinase inositol phosphosphingolipid phospholipase shortens chronological lifespan in Saccharomyces cerevisiae, while inhibiting an acid sphingomyelinase in worms or inactivating alkaline ceramidase in flies extends lifespan. The clinical potential of manipulating these enzymes is highlighted by the FDA-approved obesity drug orlistat, which is an inhibitor of pancreatic and hepatic lipases that induces weight loss and improves insulin/glucose homeostasis. Additional research is warranted to better understand how these lipid hydrolases impact aging and to determine if clinical interventions targeting them are capable of improving human healthspan.


Assuntos
Caenorhabditis elegans/crescimento & desenvolvimento , Drosophila melanogaster/crescimento & desenvolvimento , Lipase/metabolismo , Metabolismo dos Lipídeos , Longevidade , Animais , Caenorhabditis elegans/enzimologia , Drosophila melanogaster/enzimologia , Humanos , Hidrólise , Lipase/antagonistas & inibidores
18.
Aging Cell ; 19(11): e13256, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33031577

RESUMO

We previously identified 529 proteins that had been reported by multiple different studies to change their expression level with age in human plasma. In the present study, we measured the q-value and age coefficient of these proteins in a plasma proteomic dataset derived from 4263 individuals. A bioinformatics enrichment analysis of proteins that significantly trend toward increased expression with age strongly implicated diverse inflammatory processes. A literature search revealed that at least 64 of these 529 proteins are capable of regulating life span in an animal model. Nine of these proteins (AKT2, GDF11, GDF15, GHR, NAMPT, PAPPA, PLAU, PTEN, and SHC1) significantly extend life span when manipulated in mice or fish. By performing machine-learning modeling in a plasma proteomic dataset derived from 3301 individuals, we discover an ultra-predictive aging clock comprised of 491 protein entries. The Pearson correlation for this clock was 0.98 in the learning set and 0.96 in the test set while the median absolute error was 1.84 years in the learning set and 2.44 years in the test set. Using this clock, we demonstrate that aerobic-exercised trained individuals have a younger predicted age than physically sedentary subjects. By testing clocks associated with 1565 different Reactome pathways, we also show that proteins associated with signal transduction or the immune system are especially capable of predicting human age. We additionally generate a multitude of age predictors that reflect different aspects of aging. For example, a clock comprised of proteins that regulate life span in animal models accurately predicts age.


Assuntos
Proteínas Sanguíneas/metabolismo , Mineração de Dados/métodos , Longevidade/fisiologia , Aprendizado de Máquina/normas , Adolescente , Adulto , Idoso , Envelhecimento , Animais , Modelos Animais de Doenças , Humanos , Camundongos , Pessoa de Meia-Idade , Adulto Jovem
19.
Ageing Res Rev ; 60: 101070, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32311500

RESUMO

The development of clinical interventions that significantly improve human healthspan requires robust markers of biological age as well as thoughtful therapeutic targets. To promote these goals, we performed a systematic review and analysis of human aging and proteomics studies. The systematic review includes 36 different proteomics analyses, each of which identified proteins that significantly changed with age. We discovered 1,128 proteins that had been reported by at least two or more analyses and 32 proteins that had been reported by five or more analyses. Each of these 32 proteins has known connections relevant to aging and age-related disease. GDF15, for example, extends both lifespan and healthspan when overexpressed in mice and is additionally required for the anti-diabetic drug metformin to exert beneficial effects on body weight and energy balance. Bioinformatic enrichment analyses of our 1,128 commonly identified proteins heavily implicated processes relevant to inflammation, the extracellular matrix, and gene regulation. We additionally propose a novel proteomic aging clock comprised of proteins that were reported to change with age in plasma in three or more different studies. Using a large patient cohort comprised of 3,301 subjects (aged 18-76 years), we demonstrate that this clock is able to accurately predict human age.


Assuntos
Envelhecimento , Proteômica , Idoso , Animais , Biomarcadores , Regulação da Expressão Gênica , Humanos , Longevidade , Camundongos
20.
PLoS One ; 15(7): e0234986, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32634135

RESUMO

Glioblastoma is a common, malignant brain tumor whose disease incidence increases with age. Glioblastoma stem-like cells (GSCs) are thought to contribute to cancer therapy resistance and to be responsible for tumor initiation, maintenance, and recurrence. This study utilizes both SNP array and gene expression profiling to better understand GSCs and their relation to malignant disease. Peripheral blood and primary glioblastoma tumor tissue were obtained from patients, the latter of which was used to generate GSCs as well as a CD133pos./CD15pos. subpopulation. The stem cell features of GSCs were confirmed via the immunofluorescent expression of Nestin, SOX2, and CD133. Both tumor tissue and the isolated primary cells shared unique abnormal genomic characteristics, including a gain of chromosome 7 as well as either a partial or complete loss of chromosome 10. Individual genomic differences were also observed, including the loss of chromosome 4 and segmental uniparental disomy of 9p24.3→p21.3 in GSCs. Gene expression profiling revealed 418 genes upregulated in tumor tissue vs. CD133pos./CD15pos. cells and 44 genes upregulated in CD133pos./CD15pos. cells vs. tumor tissue. Pathway analyses demonstrated that upregulated genes in CD133pos./CD15pos. cells are relevant to cell cycle processes and cancerogenesis. In summary, we detected previously undescribed genomic and gene expression differences when comparing tumor tissue and isolated stem-like subpopulations.


Assuntos
Glioblastoma/patologia , Células-Tronco Neoplásicas/patologia , Antígeno AC133/análise , Separação Celular/métodos , Células Cultivadas , Perfilação da Expressão Gênica , Humanos , Antígenos CD15/análise , Polimorfismo de Nucleotídeo Único/genética , Manejo de Espécimes , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA