Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Ann Med ; 54(1): 1047-1057, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36799362

RESUMO

Introduction: Bispecific antibody (BiAb)-armed activated T cells (BATs) comprise an adoptive T cell therapy platform for treating cancer. Arming activated T cells (ATC) with anti-CD3 x anti-tumour associated antigen (TAA) BiAbs converts ATC into non-major histocompatibility complex (MHC)-restricted anti-tumour cytotoxic T lymphocytes (CTLs). Binding of target antigens via the BiAb bridge enables specific anti-tumour cytotoxicity, Th1 cytokines release, and T cell proliferation. Clinical trials in breast, prostate, and pancreatic cancer using BATs armed with chemically heteroconjugated BiAbs demonstrated safety, feasibility, induction of anti-tumour immune responses and potential increases in overall survival (OS).Objectives: The primary objective of this study was to develop a recombinant BiAb that confers enhanced anti-tumour activity of BATs against a broad range of solid tumours.Methods: A recombinant anti-epidermal growth factor receptor (EGFR) x anti-CD3 (OKT3) BiAb (rEGFRBi) was designed and expressed in CHO cells, used to arm ATC (rEGFR-BATs), and tested for specific cytotoxicity against breast, pancreatic and prostate cancers and glioblastoma.Results: rEGFR-BATs exhibit remarkably enhanced specific cytotoxicity and T1 cytokine secretion against a wide range of solid tumour cell lines vs. their respective chemically-heteroconjugated BATs.Conclusion: rEGFR-BATs may provide a "universal" T cell therapy for treating a wide range of solid tumours. KEY MESSAGEA (Gly4Ser)6 linker between the variable light and heavy chains of an scFv fused to the N-terminus of a heavy chain antibody confers unexpected stability to the heavy chain fusion protein and supports the efficient expression of the bispecific antibody.Arming of activated T cells with the rEGFRBi greatly enhances the relative cytotoxicity and Th1 cytokine secretion of theT cells relative to a chemically heteroconjugated BiAbs.rEGFR-BATs are promising candidates for the treatment of a broad range of solid tumours.


Assuntos
Anticorpos Biespecíficos , Neoplasias , Animais , Cricetinae , Humanos , Masculino , Anticorpos Biespecíficos/uso terapêutico , Anticorpos Biespecíficos/farmacologia , Cricetulus , Citocinas , Linfócitos T/metabolismo , Neoplasias/imunologia , Neoplasias/terapia
2.
Rejuvenation Res ; 24(4): 319-324, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34314252

RESUMO

Inflammaging, the increase of proinflammatory processes with increasing age, has multiple mechanisms from increasing numbers of senescent cells secreting cytokines to changes in metabolic processes. Alterations of oxygen metabolism with aging, especially decreased levels of O2 with age resulting from endocrine and cardiovascular dysfunction as well as desensitization of cellular response to hypoxia, may exacerbate inflammaging, which in turn creates further oxygen metabolic dysfunction. During aging, decline in levels of erythrocyte 2,3-bisphosphoglycerate (2,3-BPG), BPG mutase, and adenosine A2B receptor, a key adenosine signaling receptor that can augment 2,3-BPG expression, may fail to protect sensitive brain tissue from subtly reduced O2 levels, in turn resulting in increased numbers of activated microglia and secretion of proinflammatory cytokines, ultimately promoting inflammaging and senescence of endothelial cells. Interventions to restore O2 levels directly or via increasing 2,3-BPG may help promote cognitive health in old age, but significant work to quantify the degree of reduced O2 during aging in mammals, and especially humans, needs to be pursued.


Assuntos
Hipóxia Celular , Disfunção Cognitiva , Células Endoteliais , 2,3-Difosfoglicerato , Envelhecimento , Animais , Disfunção Cognitiva/fisiopatologia , Eritrócitos , Humanos
3.
Rejuvenation Res ; 23(6): 527-532, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33256568

RESUMO

SUMOylation, a conserved protein post-translational modification that performs multiple functions including regulation of nuclear transport and transcription, is implicated in numerous biological processes including aging. RNAi knockdown of the sole Small Ubiquitin-like MOdifier (SUMO) gene, smo-1, in Caenorhabditis elegans shortened lifespan, whereas overexpression in the intestine modestly increased lifespan. Smo-1 is required for mitochondrial fission in a tissue-specific manner. Fission, in turn, is needed for mitophagy to maintain mitochondrial homeostasis during aging. SUMOlyation affects DAuer Formation (DAF)-16, which can be directly SUMOylated, and SKN-1, the homolog of mammalian Nrf2. These regulators play key roles in maintaining mitochondrial homeostasis. However, given the modest effect of overexpressing smo-1 on lifespan enhancement and potential interference with other genes that can promote increased lifespan, caution is advised in the translation of this study based on C. elegans. Although inhibitors of SUMOlyation have been developed for cancer and activators also have been identified, broad-acting biochemical pathway modifiers such as SUMO are often suboptimal drug targets and may not be as promising for antiaging applications as they first appear.


Assuntos
Longevidade , Mitofagia , Proteína SUMO-1/metabolismo , Sumoilação , Animais , Caenorhabditis elegans/citologia , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Humanos , Ubiquitina/metabolismo
4.
Rejuvenation Res ; 23(5): 434-438, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32977738

RESUMO

Ferroptosis is a recently characterized cell death phenotype resulting from iron-catalyzed peroxidation of polyunsaturated fatty acid phospholipids. Increased dysfunctional iron metabolism is thought to lead to increased levels of iron and ferroptosis, which in turn leads to cell and organismal death at least in the nematode Caenorhabditis elegans. Drugs that block lipid peroxidation or scavenge intracellular iron extend healthspan and lifespan in C. elegans independently of other mechanisms such as the daf-1/daf-16 (insulin/insulin-like growth factor 1 [IGF-1]) pathway, but unlike many aging mechanisms do not alter temporal scaling across the life cycle of C. elegans, but rather act at specific late points in the organism's life history, temporarily blocking execution of critical dysfunction that results in listless worms. As such, inhibition of ferroptosis may be a means to extend healthspan and treat frailty and possibly neurodegenerative diseases that have a reported role for iron dyshomeostasis. However, a significant effort to understand ferroptosis in the context of mammalian and human biology is necessary. For example, some tumors block ferroptosis to survive. The constraints of balancing iron metabolism are significant and will require careful consideration in any drug development program.


Assuntos
Envelhecimento , Ferroptose , Fragilidade , Animais , Caenorhabditis elegans , Morte Celular , Humanos
5.
Rejuvenation Res ; 23(4): 349-352, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32718231

RESUMO

Proinflammatory alterations of white adipose tissue (WAT) with increasing age play an important role in mammalian aging. WAT produced eotaxin-1 (CCL11-C-C motif chemokine ligand 11) and monocyte chemoattractant protein 1 (MCP-1) (CCL2) are elevated in old mammals. Obese and old adipose tissues produce excessive proinflammatory cytokines such as interleukin (IL)-6, CCL2, and IL-1-beta that contribute to inflammaging. WAT-based inflammaging involves an altered homeostatic equilibrium between proinflammatory cells such as activated type 1 macrophages, B cells (high IgJ) and T cells, and anti-inflammatory eosinophils and Tregs. Specifically, young and lean individuals exhibit a high eosinophil-to-macrophage ratio with an enrichment of alternative activated tissue macrophages that is reduced in the WAT of aging mice. Eosinophils from young animals adoptively transferred to old mice, home to WAT and reverse many of the immunoinflammatory signatures associated with aging. Whether eosinophil-based therapies for inflammaging could be created remains an open question.


Assuntos
Tecido Adiposo Branco/imunologia , Tecido Adiposo/imunologia , Anti-Inflamatórios/farmacologia , Antinematódeos/farmacologia , Eosinófilos/imunologia , Helmintos/imunologia , Macrófagos/imunologia , Animais , Anti-Inflamatórios/imunologia , Antinematódeos/imunologia , Citocinas/metabolismo , Humanos
6.
Rejuvenation Res ; 23(3): 262-265, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32484032

RESUMO

Exercise has long been known to extend health and lifespan in humans and other mammals. However, typically exercise is thought to slow the loss of function that accompanies aging. Brett et al. have now shown that exercise restores functional competency to regenerate muscle stem cells (MuSCs) in mice as well as restore a significant portion of the transcriptional signature associated with young MuSCs. The mechanism involves the likely induction of plasma-borne factors that upregulate cell cycle regulator cyclin D1, which otherwise decreases with increasing age. Cyclin D1, in turn, through its noncanonical attenuation of TGF-beta/Smad3 signaling, helps maintain the regenerative capacity of MuSCs, which is lost as TGF-beta signaling increases with age. Interestingly, elevated levels of some proinflammatory regulators including NF-κB, TNF-alpha, and interleukin 6 (IL-6) are also reduced by exercise or ectopic expression of cyclin D1. Importantly, the rejuvenation is not complete, as Notch signaling, which also decreases with age, remains at old levels and the rejuvenative effect is not permanent: wearing off in ∼2 weeks after cessation of exercise. Understanding the limitations of the rejuvenative effect of exercise on MuSCs at the molecular level, including changes in the epigenome such as altered DNA methylation age, will be critical in developing more significant rejuvenative therapies including some for aged people wherein morbidities limit exercise.


Assuntos
Exercício Físico/fisiologia , Músculo Esquelético/fisiologia , Condicionamento Físico Animal/fisiologia , Rejuvenescimento/fisiologia , Células-Tronco/fisiologia , Envelhecimento/fisiologia , Animais , Ciclina D1/fisiologia , Fragilidade/fisiopatologia , Fragilidade/terapia , Humanos , Camundongos , Músculo Esquelético/citologia , NF-kappa B/metabolismo , Regeneração/fisiologia , Transdução de Sinais/fisiologia , Células-Tronco/citologia , Fator de Necrose Tumoral alfa/metabolismo
7.
Rejuvenation Res ; 22(3): 261-266, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-31140365

RESUMO

During human aging, decrease of NAD+ levels is associated with potentially reversible dysfunction in the liver, kidney, skeletal and cardiac muscle, endothelial cells, and neurons. At the same time, the number of senescent cells, associated with damage or stress that secretes proinflammatory factors (SASP or senescence-associated secretory phenotype), increases with age in many key tissues, including the kidneys, lungs, blood vessels, and brain. Senescent cells are believed to contribute to numerous age-associated pathologies and their elimination by senolytic regimens appears to help in numerous preclinical aging-associated disease models, including those for atherosclerosis, idiopathic pulmonary fibrosis, diabetes, and osteoarthritis. A recent report links these processes, such that decreased NAD+ levels associated with aging may attenuate the SASP potentially reducing its pathological effect. Conversely, increasing NAD+ levels by supplementation or genetic manipulation, which may benefit tissue homeostasis, also may worsen SASP and encourage tumorigenesis at least in mouse models of cancer. Taken together, these findings suggest a fundamental trade-off in treating aging-related diseases with drugs or supplements that increase NAD+. Even more interesting is a report that senescent cells can induce CD38 on macrophages and endothelial cells. In turn, increased CD38 expression is believed to be the key modulator of lowered NAD+ levels with aging in mammals. So, accumulation of senescent cells may itself be a root cause of decreased NAD+, which in turn could promote dysfunction. On the contrary, the lower NAD+ levels may attenuate SASP, decreasing the pathological influence of senescence. The elimination of most senescent cells by senolysis before initiating NAD+ therapies may be beneficial and increase safety, and in the best-case scenario reduce the need for NAD+ supplementation.


Assuntos
Envelhecimento/metabolismo , Senescência Celular , NAD/metabolismo , Animais , Humanos , Inflamação/patologia , NAD/química
8.
Rejuvenation Res ; 22(2): 171-174, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30943847

RESUMO

Nitric oxide signals through several distinct mechanisms, including interaction with the heme group of guanylyl cyclase enzymes resulting in modulation of cGMP levels in the vascular endothelium. Alternatively, reactive nitrogen oxide species can bind cysteine residues in target proteins forming S-nitrosothiols. S-nitrosylation is recognized as an important post-translational modification of dozens of proteins, which plays a key role in cellular homeostasis, metabolism, and various disease states. By denitrosylating target proteins, S-nitrosoglutathione reductase (GSNOR/ADH5) plays a pivotal role in the regulation of protein S-nitrosylation. GSNOR expression is reduced in primary senescent cells that accumulate during aging in rodents and humans. Reduced GSNOR activity is accompanied by mitochondrial nitrosative stress, characterized by elevated S-nitrosylation of Drp1 and Parkin with the downstream effect of impaired mitophagy. The mechanism involves epigenetic downregulation of GSNOR by the ten-eleven translocation 1 protein. Conflicting recent reports show that GSNOR levels change with age in mice and humans. One report found that GSNOR levels decreased in peripheral blood mononuclear cell and brains of young to middle-aged mice. However, another report more convincingly showed that there was a significant increase in the hippocampal expression of GSNOR in both old humans and mice. Increased GSNOR in old mice resulted in loss of synaptic plasticity and reduced long-term potentiation memory, in part, by reducing calmodulin kinase IIa activation, which is known to increase the number of AMPA glutamate receptors near synapses. GSNOR levels may be a key biochemical hallmark of aging, but subject to the Goldilocks principle such that its levels need to be maintained in a narrow range according to context, making it a problematic therapeutic target in aging as opposing changes in expression or activity would be needed to stimulate mitophagy in senescence and synaptic plasticity in aging brains.


Assuntos
Envelhecimento/metabolismo , Senescência Celular , Aldeído Oxirredutases/metabolismo , Animais , Humanos , Potenciação de Longa Duração , Metilação , Camundongos , Plasticidade Neuronal , Nitrosação
9.
Rejuvenation Res ; 21(4): 369-373, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-30058454

RESUMO

Mitochondrial-derived peptides (MDPs), encoded by mitochondrial DNA, play a cytoprotective role by helping preserve mitochondrial function and cell viability under stressful conditions. Humanin and its homologs and MOTS-c are two of several MDPs hypothesized to have antiaging activity based on correlative studies. For example, humanin plasma levels are inversely correlated with growth hormone and insulin-like growth factor 1 expression, which may promote accelerated aging. Humanin has been shown to protect cells from beta amyloid toxicity and preserve endothelial cell function in a mouse model of atherosclerosis. Furthermore, both humanin and MOTS-c improve insulin sensitivity in mouse models of type 2 diabetes. Recently it was reported that a potent analogue of humanin blocks cardiac fibrosis in aging mice. Although it has been hypothesized that MDPs might have senolytic activity, in a recent report humanin and MOTS-c both exacerbate the senescence-associated-secretory-phenotype (SASP) in senescent cells by stimulating the secretion of IL-6, IL-1ß, IL-8, IL-10 and tumor necrosis factor α. It appears that the cytoprotective activity of the MDPs may be permissive for increased expression of a set of proinflammatory cytokines. Given the potential benefits of MDPs in many of the same diseases associated with the presence of senescent cells, a combination of senolytic and MDP-based treatments may be additive or synergistic. The MDPs would protect normal cells, whereas senescent cells would be eliminated by the senolytic therapy. It is even possible that MDPs by increasing the SASP phenotype would make the senescent cells more apt to be cleared by the immune system or more sensitive to senolytics. In contrast, if the MDPs actually cytoprotect the senescent cells, then the treatment can be performed serially with the senolytic used first.


Assuntos
Senescência Celular , Mitocôndrias/metabolismo , Peptídeos/farmacologia , Animais , Senescência Celular/efeitos dos fármacos , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/farmacologia , Fenótipo
10.
Rejuvenation Res ; 21(3): 278-282, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29781380

RESUMO

The United States has the most expensive healthcare system worldwide. Yet measures of health span and life expectancy are well below the major industrialized nations. With the U.S. population aged 65 years and older projected to double by mid-century, a healthcare crisis is looming. Within this context, huge interest and investment have emerged in technologies and drugs to address aging with an expected benefit to health span. The thesis being that such basic interventions will reduce morbidity caused by many chronic diseases wherein biological age itself is the major risk factor. In the light of limited progress to date, a recent study out of the Harvard School of Public Health is quite refreshing: less than half dozen lifestyle interventions can greatly increase health span. Perhaps these are familiar: cessation of smoking, ≥30 minutes of moderate daily exercise, high-quality diet (limited processed food), modest alcohol intake, and maintenance of an optimal body mass index of 18.5-24.9 kg/m2. From age 50 years, women engaging in all of these behaviors versus those who do zero can expect to have a life expectancy of 43.1 additional years (an extra 14 years) with men gaining 37.6 years (an extra 12.2 years). A regimen to extend life expectancy is at hand. However, there is room for optimization by including the effects of sleep, intermittent fasting, and/or caloric restriction. Moreover, the extension of life expectancy by adherence to a healthy lifestyle revises the health span threshold for antiaging treatments under development and should provide a better set of controls for clinical trials investigating novel treatments of aging.


Assuntos
Doença Crônica , Dieta , Promoção da Saúde/métodos , Expectativa de Vida , Longevidade , Idoso , Envelhecimento , Índice de Massa Corporal , Restrição Calórica , Epigênese Genética , Exercício Físico , Jejum , Feminino , Humanos , Estilo de Vida , Masculino , Pessoa de Meia-Idade , Fatores de Risco , Fumar , Fatores de Tempo , Estados Unidos
11.
Rejuvenation Res ; 20(6): 525-529, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29179649

RESUMO

Frailty is a medical syndrome associated with advancing age characterized by reduced functional reserve, strength, endurance, and susceptibility to infection associated with high morbidity, hospitalization, and death. Nonspecific interventions to improve the healthspan of affected patients include physical therapy, exercise, improved nutrition, etc. Among the hallmarks of aging, depletion of stem cells with resultant compromise of regeneration and repair of tissues informs a rational stem cell-based replacement strategy. This hypothesis has been evaluated in a randomized, double-blind, placebo-controlled clinical trial utilizing human allogeneic mesenchymal stem cells (allo-hMSCs), a facile, scalable stem cell replacement therapy. Intravenous infusion of 100 or 200 million allo-hMSCs was deemed safe in aged frail individuals. However, modest improvement outcomes were limited to the lower dose, a finding that remains difficult to explain. Future studies are definitely warranted given the magnitude of this increasingly important medical syndrome.


Assuntos
Fragilidade/terapia , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/citologia , Animais , Modelos Animais de Doenças , Exossomos/metabolismo , Humanos , Imunomodulação , Pesquisa Translacional Biomédica
12.
Rejuvenation Res ; 20(5): 430-436, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28942711

RESUMO

The epigenome, which controls cell identity and function, is not maintained with 100% fidelity in somatic animal cells. Errors in the maintenance of the epigenome lead to epigenetic drift, an important hallmark of aging. Numerous studies have described DNA methylation clocks that correlate epigenetic drift with increasing age. The question of how significant a role epigenetic drift plays in creating the phenotypes associated with aging remains open. A recent study describes a new DNA methylation clock that can be slowed by caloric restriction (CR) in a way that correlates with the degree of lifespan and healthspan extension conferred by CR, suggesting that epigenetic drift itself is a determinant of mammalian lifespan. Genetic transplantation using genomic editing of DNA methylation homeostatic genes from long-lived to short-lived species is one way to potentially demonstrate a causative role for DNA methylation. Whether the DNA methylation clock be reset to youthful state, eliminating the effects of epigenetic drift without requiring a pluripotent cell intermediate is a critical question with profound implications for the development of aging therapeutics. Methods that transiently erase the DNA methylation pattern of somatic cells may be developed that reset this aging hallmark with potentially profound effects on lifespan, if DNA methylation-based epigenetic drift really plays a primary role in aging.


Assuntos
Epigênese Genética , Longevidade/genética , Mamíferos/genética , Animais , Metilação de DNA/genética , Humanos
13.
Rejuvenation Res ; 20(3): 244-247, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28537485

RESUMO

NAD+ levels decline with age in diverse animals from Caenorhabditis elegans to mice. Raising NAD+ levels by dietary supplementation with NAD+ precursors, nicotinamide riboside (NR) or nicotinamide mononucleotide (NMN), improves mitochondrial function and muscle and neural and melanocyte stem cell function in mice, as well as increases murine life span. Decreased NAD+ levels with age reduce SIRT1 function and reduce the mitochondrial unfolded protein response, which can be overcome by NR supplementation. Decreased NAD+ levels cause NAD+-binding protein DBC1 to form a complex with PARP1, inhibiting poly(adenosine diphosphate-ribose) polymerase (PARP) catalytic activity. Old mice have increased amounts of DBC1-PARP1 complexes, lower PARP activity, increased DNA damage, and reduced nonhomologous end joining and homologous recombination repair. DBC1-PARP1 complexes in old mice can be broken by increasing NAD+ levels through treatment with NMN, reducing DNA damage and restoring PARP activity to youthful levels. The mechanism of declining NAD+ levels and its fundamental importance to aging are yet to be elucidated. There is a correlation of PARP activity with mammalian life span that suggests that NAD+/SIRT1/PARP1 may be more significant than the modest effects on life span observed for NR supplementation in old mice. The NAD+/PARP1/SIRT1 axis may link NAD+ levels and DNA damage with the apparent epigenomic DNA methylation clocks that have been described.


Assuntos
Envelhecimento/metabolismo , NAD/metabolismo , Poli(ADP-Ribose) Polimerases/metabolismo , Transdução de Sinais , Sirtuína 1/metabolismo , Animais , Dano ao DNA , Humanos , Camundongos
14.
Oncotarget ; 8(24): 39356-39366, 2017 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-28454118

RESUMO

Therapeutic antibodies are effective for tumor immunotherapy and exhibit prominent clinical effects. All approved antibody therapeutics utilize IgG as the molecular format. Antibody-dependent cell-mediated cytotoxicity (ADCC) is a key mechanism for tumor cell killing by antibodies. For IgG antibodies, ADCC depends on FcγR-expressing cells, such as natural killer (NK) cells. However, in patients with a high tumor burden, antibody therapeutics may lose efficacy owing to exhaustion of FcγR-expressing effector cells as well as the inhibitory effects of certain FcγRs on effector cells. To achieve more potent effector functions, we engineered an anti-CD20 antibody to contain both IgG Fc and IgA Fc domains. These engineered antibodies interacted with both IgG and IgA Fc receptors (FcγR and FcαR) and recruited a broader range of effector cells, including monocytes, macrophages, neutrophils, and NK cells, thereby enhancing antibody-dependent cellular phagocytosis. Using transgenic mice expressing the FcαRI (CD89) in macrophages, we demonstrated that recombinant antibodies bearing the chimeric IgG and IgA Fc exhibited potent in vivo antitumor activity. Additionally, in a short-term peritoneal model using CD20-transfected LLC target cells, the in vivo cytotoxic activity of hybrid recombinant antibodies was mediated by macrophages with significant reduction in the absence of FcαRI. Our findings supported targeting of FcαRI on monocytes and macrophages for improved tumor immunotherapy.


Assuntos
Antígenos CD/metabolismo , Macrófagos/metabolismo , Monócitos/metabolismo , Neoplasias/metabolismo , Receptores Fc/metabolismo , Receptores de IgG/metabolismo , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacologia , Citotoxicidade Celular Dependente de Anticorpos/imunologia , Biomarcadores , Linhagem Celular Tumoral , Modelos Animais de Doenças , Humanos , Imunoterapia , Macrófagos/imunologia , Camundongos , Monócitos/imunologia , Neoplasias/imunologia , Neoplasias/patologia , Neoplasias/terapia , Ensaios Antitumorais Modelo de Xenoenxerto
15.
Rejuvenation Res ; 20(2): 146-150, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28314379

RESUMO

Epigenetic variation with age is one of the most important hallmarks of aging. Resetting or repairing the epigenome of aging cells in intact animals may rejuvenate the cells and perhaps the entire organism. In fact, differentiated adult cells, which by definition have undergone some epigenetic changes, are capable of being rejuvenated and reprogrammed to create pluripotent stem cells and viable cloned animals. Apparently, such reprogramming is capable of completely resetting the epigenome. However, attempts to fully reprogram differentiated cells in adult animals have failed in part because reprogramming leads to the formation of teratomas. A preliminary method to partially reprogram adult cells in mature Hutchinson-Gilford Progeria Syndrome (HGPS) mice by transient induction of the Yamanaka factors OSKM (Oct4/Sox2/Klf4/c-Myc) appears to ameliorate aging-like phenotypes in HGPS mice, and promote youthful regenerative capability in middle-aged wild-type individuals exposed to beta cell and muscle cell-specific toxins. However, whatever epigenetic repair is induced by transient reprogramming does not endure and may be due to the induction of key homeostatic regulators instead. Some of the effect of transient reprogramming may result from increased proliferation and enhanced function of adult stem cells. Partial reprogramming may point the way to new antiaging and proregenerative therapeutics. Redifferentiation of cells into their preexisting phenotype with simultaneous epigenomic rejuvenation is an interesting variation that also should be pursued. However, discovery of methods to more precisely repair the epigenome is the most likely avenue to the development of powerful new antiaging agents.


Assuntos
Reprogramação Celular/genética , Epigênese Genética , Rejuvenescimento/fisiologia , Animais , Humanos , Fator 4 Semelhante a Kruppel , Medicina Regenerativa
16.
Rejuvenation Res ; 19(5): 423-429, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27612523

RESUMO

The loss of muscle stem cell (MuSC) numbers and function in the elderly results in a dramatic delay or incomplete repair of muscle following injury or surgery. Prolonged immobility can exacerbate the loss of muscle mass with increased morbidity of affected patients. Stem cells and their niche cooperate to regulate the activation, self-renewal, differentiation, and return to quiescence of MuSCs. Extracellular matrix fibronectin (FN) and MuSC ß1-integrin have been identified as critical factors in the dysfunction of aging muscle. Reduced amounts and/or function of ß1-integrin and fibronectin are critical factors in the decline in MuSC regeneration and homeostasis with aging. Replacement of fibronectin and/or stimulation of ß1-integrin may provide a novel means to augment the decline in MuSC function with age.

17.
Rejuvenation Res ; 19(4): 337-40, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27378359

RESUMO

Until recently, the mechanism of adaptive thermogenesis was ascribed to the expression of uncoupling protein 1 (UCP1) in brown and beige adipocytes. UCP1 is known to catalyze a proton leak of the inner mitochondrial membrane, resulting in uncoupled oxidative metabolism with no production of adenosine triphosphate and increased energy expenditure. Thus increasing brown and beige adipose tissue with augmented UCP1 expression is a viable target for obesity-related disorders. Recent work demonstrates an UCP1-independent pathway to uncouple mitochondrial respiration. A secreted enzyme, PM20D1, enriched in UCP1+ adipocytes, exhibits catalytic and hydrolytic activity to reversibly form N-acyl amino acids. N-acyl amino acids act as endogenous uncouplers of mitochondrial respiration at physiological concentrations. Administration of PM20D1 or its products, N-acyl amino acids, to diet-induced obese mice improves glucose tolerance by increasing energy expenditure. In short-term studies, treated animals exhibit no toxicity while experiencing 10% weight loss primarily of adipose tissue. Further study of this metabolic pathway may identify novel therapies for diabesity, the disease state associated with diabetes and obesity.


Assuntos
Tecido Adiposo/enzimologia , Amidoidrolases/metabolismo , Respiração Celular , Dieta , Metabolismo Energético , Mitocôndrias/enzimologia , Obesidade/prevenção & controle , Redução de Peso , Aminoácidos/metabolismo , Animais , Modelos Animais de Doenças , Humanos , Camundongos , Obesidade/enzimologia , Obesidade/etiologia , Obesidade/fisiopatologia , Fosforilação Oxidativa , Proteína Desacopladora 1/metabolismo
18.
Rejuvenation Res ; 19(1): 90-3, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26837468

RESUMO

Although the thymus plays a key role in T cell maturation in mammals, it begins to atrophy and involute at sexual maturity. The diminished thymic microenvironment is thought to contribute to reduced adaptive immune function during aging, leading to the increased likelihood of infectious diseases and cancer. Caloric restriction or ectopic expression of the pro-longevity growth factor fibroblast growth factor 21 has been reported to maintain the thymus in aging mice. Moreover, forced expression of the transcription factor FoxN1 has been shown to rejuvenate thymuses from old mice almost completely, restoring their youthful state. These results open the way for development of potential drugs to restore immune function in the elderly.


Assuntos
Fatores de Crescimento de Fibroblastos/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Regeneração , Timo/fisiologia , Animais , Camundongos
19.
Rejuvenation Res ; 18(5): 479-83, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26414604

RESUMO

Reduced telomere length with increasing age in dividing cells has been implicated in contributing to the pathologies of human aging, which include cardiovascular and metabolic disorders, through induction of cellular senescence. Telomere shortening results from the absence of telomerase, an enzyme required to maintain telomere length. Telomerase reverse transcriptase (TERT), the protein subunit of telomerase, is expressed only transiently in a subset of adult somatic cells, which include stem cells and smooth muscle cells. A recent report from Xiong and colleagues demonstrates a pivotal role for the transcription co-factor peroxisome proliferator-activated receptor γ co-activator-1α (PGC-1α) in maintaining TERT expression and preventing vascular senescence and atherosclerosis in mice. Ablation of PGC-1α reduced TERT expression and increased DNA damage and reactive oxygen species (ROS), resulting in shortened telomeres and vascular senescence. In the ApoE(-/-) mouse model of atherosclerosis, forced expression of PGC-1α increased expression of TERT, extended telomeres, and reversed genomic DNA damage, vascular senescence, and the development of atherosclerotic plaques. Alpha lipoic acid (ALA) stimulated expression of PGC-1α and TERT and reversed DNA damage, vascular senescence, and atherosclerosis, similarly to ectopic expression of PGC-1α. ALA stimulated cyclic adenosine monophosphate (cAMP) signaling, which in turn activated the cAMP response element-binding protein (CREB), a co-factor for PGC-1α expression. The possibility that ALA might induce TERT to extend telomeres in human cells suggests that ALA may be useful in treating atherosclerosis and other aging-related diseases. However, further investigation is needed to identify whether ALA induces TERT in human cells, which cell types are susceptible, and whether such changes have clinical significance.


Assuntos
Aterosclerose/genética , Vasos Sanguíneos/crescimento & desenvolvimento , Dano ao DNA , Encurtamento do Telômero , Fatores de Transcrição/genética , Animais
20.
Rejuvenation Res ; 18(4): 371-5, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26160351

RESUMO

Epigenomic change and stem cell exhaustion are two of the hallmarks of aging. Accumulation of molecular damage is thought to underlie aging, but the precise molecular composition of the damage remains controversial. That some aging phenotypes, especially those that result from impaired stem cell function, are reversible suggest that such "damage" is repairable. Evidence is accumulating that dysfunction in aging stem cells results from increasing, albeit, subtle disorganization of the epigenome over time. Zhang et al. (2015) report that decreasing levels of WRN, Werner's syndrome (WS) helicase, with increasing age results in loss of heterochromatin marks in mesenchymal stem cells (MSCs) and correlates with an increased rate of cellular senescence. Although WRN plays a role in DNA repair, WRN exerted its effects on aging via maintaining heterochromatin, evidenced by reduced levels of interacting chromatin regulators heterochromatin protein 1α (HP1α), suppressor of variegation 3-9 homolog 1 (SUV39H1), and lamina-associated polypeptide 2ß (LAP2ß) as well as modified histone H3K9me3. Reducing expression of chromatin modeling co-factors SUV39H1 or HP1α in wild-type MSCs recapitulates the phenotype of WRN deficiency, resulting in reduced H3K9me3 levels and increased senescence without induction of markers of DNA damage, suggesting that chromatin disorganization and not DNA damage is responsible for the pathology of WS during aging in animals. Ectopic expression of HP1α restored H3K9me3 levels and repressed senescence in WRN-deficient MSCs. That HP1α can also suppress senescence in Hutchinson-Gilford progeria syndrome (HGPS) and extend life span in flies when over-expressed suggests that HP1α and H3K9me3 play conserved roles in maintenance of cell state. H3K9me3 levels are dynamic and expected to be potentially responsive to manipulation by extrinsic factors. Recent reports that migration inhibitory factor (MIF) or periodic fasting rejuvenate old MSCs provide the opportunity to link intrinsic and extrinsic mechanisms of aging in novel and potentially medically important ways and may lead to anti-aging treatments that reorganize the epigenome to rejuvenate cells and tissues.


Assuntos
Envelhecimento/genética , Biomarcadores/metabolismo , Epigênese Genética , Histonas/metabolismo , Lisina/metabolismo , Células-Tronco/metabolismo , Animais , Homólogo 5 da Proteína Cromobox , Humanos , Metilação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA