Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Science ; 384(6694): 401-403, 2024 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-38662837

RESUMO

Highlights from the Science family of journals.

2.
4.
Science ; 383(6682): 491-493, 2024 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-38300998

RESUMO

Highlights from the Science family of journals.

6.
Nat Aging ; 3(2): 157-161, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36873708

RESUMO

Mitochondrial dysfunction plays a central role in aging but the exact biological causes are still being determined. Here, we show that optogenetically increasing mitochondrial membrane potential during adulthood using a light-activated proton pump improves age-associated phenotypes and extends lifespan in C. elegans. Our findings provide direct causal evidence that rescuing the age-related decline in mitochondrial membrane potential is sufficient to slow the rate of aging and extend healthspan and lifespan.


Assuntos
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Animais , Caenorhabditis elegans/genética , Longevidade/genética , Proteínas de Caenorhabditis elegans/genética , Potencial da Membrana Mitocondrial , Optogenética , Rejuvenescimento
7.
Geroscience ; 45(3): 1573-1581, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36877298

RESUMO

Dietary restriction (DR) increases lifespan in many organisms, but its underlying mechanisms are not fully understood. Mitochondria play a central role in metabolic regulation and are known to undergo changes in structure and function in response to DR. Mitochondrial membrane potential (Δψm) is the driving force for ATP production and mitochondrial outputs that integrate many cellular signals. One such signal regulated by Δψm is nutrient-status sensing. Here, we tested the hypothesis that DR promotes longevity through preserved Δψm during adulthood. Using the nematode Caenorhabditis elegans, we find that Δψm declines with age relatively early in the lifespan, and this decline is attenuated by DR. Pharmacologic depletion of Δψm blocked the longevity and health benefits of DR. Genetic perturbation of Δψm and mitochondrial ATP availability similarly prevented lifespan extension from DR. Taken together, this study provides further evidence that appropriate regulation of Δψm is a critical factor for health and longevity in response to DR.


Assuntos
Proteínas de Caenorhabditis elegans , Animais , Proteínas de Caenorhabditis elegans/genética , Longevidade/genética , Potencial da Membrana Mitocondrial , Restrição Calórica , Caenorhabditis elegans/genética , Trifosfato de Adenosina/metabolismo
8.
Int J Biochem Cell Biol ; 157: 106391, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36806357

RESUMO

In vivo control over metabolism is at the cutting edge of biomedical research. The particulars of mitochondrial function are especially important to understand in vivo to progress metabolic therapies that will be relevant for diseases of aging. Understanding the differences between how mitochondria function in vitro versus in vivo will be a necessary challenge to overcome to achieve mitochondrial medicine. In this article we outline how discoveries in invertebrate models will be informative for understanding the basic biology of mitochondria to streamline translation to mammals and eventually to humans. Further, we highlight examples of how what is known about mitochondria in vitro is translatable to in vivo models and, in some cases, to human diseases.


Assuntos
Metabolismo Energético , Mitocôndrias , Animais , Humanos , Mitocôndrias/metabolismo , Envelhecimento/metabolismo , Mamíferos
9.
Nat Commun ; 13(1): 2403, 2022 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-35504873

RESUMO

C. elegans react to metabolic distress caused by mismatches in oxygen and energy status via distinct behavioral responses. At the molecular level, these responses are coordinated by under-characterized, redox-sensitive processes, thought to initiate in mitochondria. Complex I of the electron transport chain is a major site of reactive oxygen species (ROS) production and is canonically associated with oxidative damage following hypoxic exposure. Here, we use a combination of optogenetics and CRISPR/Cas9-mediated genome editing to exert spatiotemporal control over ROS production. We demonstrate a photo-locomotory remodeling of avoidance behavior by local ROS production due to the reversible oxidation of a single thiol on the complex I subunit NDUF-2.1. Reversible thiol oxidation at this site is necessary and sufficient for the behavioral response to hypoxia, does not respond to ROS produced at more distal sites, and protects against lethal hypoxic exposure. Molecular modeling suggests that oxidation at this thiol residue alters the ability for NDUF-2.1 to coordinate electron transfer to coenzyme Q by destabilizing the Q-binding pocket, causing decreased complex I activity. Overall, site-specific ROS production regulates behavioral responses and these findings provide a mechanistic target to suppress the detrimental effects of hypoxia.


Assuntos
Caenorhabditis elegans , Compostos de Sulfidrila , Animais , Aprendizagem da Esquiva , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Complexo I de Transporte de Elétrons/genética , Complexo I de Transporte de Elétrons/metabolismo , Hipóxia , Espécies Reativas de Oxigênio/metabolismo
10.
MicroPubl Biol ; 20212021.
Artigo em Inglês | MEDLINE | ID: mdl-34532702

RESUMO

Mitochondria are ATP-producing organelles that also signal throughout the cell. Mitochondrial protein homeostasis is regulated through membrane potential-dependent protein import and quality control signaling. The mitochondrial unfolded protein response (UPRmt) is a specific program that responds to imbalances in nuclear and mitochondrial gene expression. Mounting evidence suggests that the electrochemical gradient that powers mitochondrial function, the mitochondrial membrane potential (Δψm), is a core regulator of the UPRmt. Here we tested this notion directly by pharmacologically dissipating Δψm and monitoring UPRmt activation. We found that chemical dissipation of Δψm using FCCP indeed activated UPRmt dose-dependently in C. elegans assayed by the HSP-60::GFP reporter strain.

11.
Front Immunol ; 12: 666231, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34149701

RESUMO

Although cancer immunotherapy is effective against hematological malignancies, it is less effective against solid tumors due in part to significant metabolic challenges present in the tumor microenvironment (TME), where infiltrated CD8+ T cells face fierce competition with cancer cells for limited nutrients. Strong metabolic suppression in the TME is often associated with impaired T cell recruitment to the tumor site and hyporesponsive effector function via T cell exhaustion. Increasing evidence suggests that mitochondria play a key role in CD8+ T cell activation, effector function, and persistence in tumors. In this study, we showed that there was an increase in overall mitochondrial function, including mitochondrial mass and membrane potential, during both mouse and human CD8+ T cell activation. CD8+ T cell mitochondrial membrane potential was closely correlated with granzyme B and IFN-γ production, demonstrating the significance of mitochondria in effector T cell function. Additionally, activated CD8+ T cells that migrate on ICAM-1 and CXCL12 consumed significantly more oxygen than stationary CD8+ T cells. Inhibition of mitochondrial respiration decreased the velocity of CD8+ T cell migration, indicating the importance of mitochondrial metabolism in CD8+ T cell migration. Remote optical stimulation of CD8+ T cells that express our newly developed "OptoMito-On" successfully enhanced mitochondrial ATP production and improved overall CD8+ T cell migration and effector function. Our study provides new insight into the effect of the mitochondrial membrane potential on CD8+ T cell effector function and demonstrates the development of a novel optogenetic technique to remotely control T cell metabolism and effector function at the target tumor site with outstanding specificity and temporospatial resolution.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/efeitos da radiação , Optogenética/métodos , Trifosfato de Adenosina/metabolismo , Animais , Linfócitos T CD8-Positivos/metabolismo , Movimento Celular/efeitos da radiação , Citocinas/metabolismo , Citoesqueleto/metabolismo , Citoesqueleto/efeitos da radiação , Humanos , Imunoterapia , Ativação Linfocitária/efeitos da radiação , Potencial da Membrana Mitocondrial/genética , Potencial da Membrana Mitocondrial/efeitos da radiação , Camundongos , Mitocôndrias/metabolismo , Mitocôndrias/efeitos da radiação , Neoplasias/imunologia , Neoplasias/terapia
12.
Geroscience ; 43(4): 1591-1604, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33864592

RESUMO

Mitochondria are organelles that provide energy to cells through ATP production. Mitochondrial dysfunction has long been postulated to mediate cellular declines that drive biological aging. Many well-characterized hallmarks of aging may involve underlying energetic defects that stem from loss of mitochondrial function with age. Why and how mitochondrial function declines with age is an open question and one that has been difficult to answer. Mitochondria are powered by an electrochemical gradient across the inner mitochondrial membrane known as the protonmotive force (PMF). This gradient decreases with age in several experimental models. However, it is unclear if a diminished PMF is a cause or a consequence of aging. Herein, we briefly review and define mitochondrial function, we summarize how PMF changes with age in several models, and we highlight recent studies that implicate PMF in aging biology. We also identify barriers that must be addressed for the field to progress. Emerging technology permits more precise in vivo study of mitochondria that will allow better understanding of cause and effect in metabolic models of aging. Once cause and effect can be discerned more precisely, energetics approaches to combat aging may be developed to prevent or reverse functional decline.


Assuntos
Mitocôndrias
13.
Autophagy ; 17(11): 3389-3401, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-33416042

RESUMO

Mitochondrial quality control (MQC) balances organelle adaptation and elimination, and mechanistic crosstalk between the underlying molecular processes affects subsequent stress outcomes. FUNDC1 (FUN14 domain containing 1) is a mammalian mitophagy receptor that responds to hypoxia-reoxygenation (HR) stress. Here, we provide evidence that FNDC-1 is the C. elegans ortholog of FUNDC1, and that its loss protects against injury in a worm model of HR. This protection depends upon ATFS-1, a transcription factor that is central to the mitochondrial unfolded protein response (UPRmt). Global mRNA and metabolite profiling suggest that atfs-1-dependent stress responses and metabolic remodeling occur in response to the loss of fndc-1. These data support a role for FNDC-1 in non-hypoxic MQC, and further suggest that these changes are prophylactic in relation to subsequent HR. Our results highlight functional coordination between mitochondrial adaptation and elimination that organizes stress responses and metabolic rewiring to protect against HR injury.Abbreviations: AL: autolysosome; AP: autophagosome; FUNDC1: FUN14 domain containing 1; HR: hypoxia-reperfusion; IR: ischemia-reperfusion; lof: loss of function; MQC: mitochondrial quality control; PCA: principle component analysis; PPP: pentonse phosphate pathway; proK (proteinase K);UPRmt: mitochondrial unfolded protein response; RNAi: RNA interference.


Assuntos
Proteínas de Caenorhabditis elegans/fisiologia , Caenorhabditis elegans/fisiologia , Proteínas Mitocondriais/fisiologia , Mitofagia/fisiologia , Fatores de Transcrição/fisiologia , Animais , Animais Geneticamente Modificados , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Genes de Helmintos , Hipóxia/genética , Hipóxia/fisiopatologia , Mutação com Perda de Função , Proteínas de Membrana/genética , Proteínas de Membrana/fisiologia , Proteínas Mitocondriais/genética , Mitofagia/genética , Traumatismo por Reperfusão/genética , Traumatismo por Reperfusão/fisiopatologia , Fatores de Transcrição/genética
14.
FASEB J ; 34(12): 16333-16347, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33058299

RESUMO

Organisms adapt to their environment through coordinated changes in mitochondrial function and metabolism. The mitochondrial protonmotive force (PMF) is an electrochemical gradient that powers ATP synthesis and adjusts metabolism to energetic demands via cellular signaling. It is unknown how or where transient PMF changes are sensed and signaled due to the lack of precise spatiotemporal control in vivo. We addressed this by expressing a light-activated proton pump in mitochondria to spatiotemporally "turn off" mitochondrial function through PMF dissipation in tissues with light. We applied our construct-mitochondria-OFF (mtOFF)-to understand how metabolic status impacts hypoxia resistance, a response that relies on mitochondrial function. Activation of mtOFF induced starvation-like behavior mediated by AMP-activated protein kinase (AMPK). We found prophylactic mtOFF activation increased survival following hypoxia, and that protection relied on neuronal AMPK. Our study links spatiotemporal control of mitochondrial PMF to cellular metabolic changes that mediate behavior and stress resistance.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Caenorhabditis elegans/metabolismo , Metabolismo Energético/fisiologia , Hipóxia/metabolismo , Mitocôndrias/metabolismo , Neurônios/metabolismo , Animais , Bombas de Próton/metabolismo
15.
FEBS J ; 287(21): 4544-4556, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32459870

RESUMO

Developing new technologies to study metabolism is increasingly important as metabolic disease prevalence increases. Mitochondria control cellular metabolism and dynamic changes in mitochondrial function are associated with metabolic abnormalities in cardiovascular disease, cancer, and obesity. However, a lack of precise and reversible methods to control mitochondrial function has prevented moving from association to causation. Recent advances in optogenetics have addressed this challenge, and mitochondrial function can now be precisely controlled in vivo using light. A class of genetically encoded, light-activated membrane channels and pumps has addressed mechanistic questions that promise to provide new insights into how cellular metabolism downstream of mitochondrial function contributes to disease. Here, we highlight emerging reagents-mitochondria-targeted light-activated cation channels or proton pumps-to decrease or increase mitochondrial activity upon light exposure, a technique we refer to as mitochondrial light switches, or mtSWITCH . The mtSWITCH technique is broadly applicable, as energy availability and metabolic signaling are conserved aspects of cellular function and health. Here, we outline the use of these tools in diverse cellular models of disease. We review the molecular details of each optogenetic tool, summarize the results obtained with each, and outline best practices for using optogenetic approaches to control mitochondrial function and downstream metabolism.


Assuntos
Luz , Mitocôndrias/efeitos da radiação , Optogenética/métodos , Transdução de Sinais/efeitos da radiação , Animais , Humanos , Concentração de Íons de Hidrogênio/efeitos da radiação , Potencial da Membrana Mitocondrial/efeitos da radiação , Mitocôndrias/metabolismo , Força Próton-Motriz/efeitos da radiação , Espécies Reativas de Oxigênio/metabolismo
16.
EMBO Rep ; 21(4): e49113, 2020 04 03.
Artigo em Inglês | MEDLINE | ID: mdl-32043300

RESUMO

Mitochondrial respiration generates an electrochemical proton gradient across the mitochondrial inner membrane called protonmotive force (PMF) to drive diverse functions and synthesize ATP. Current techniques to manipulate the PMF are limited to its dissipation; yet, there is no precise and reversible method to increase the PMF. To address this issue, we aimed to use an optogenetic approach and engineered a mitochondria-targeted light-activated proton pump that we name mitochondria-ON (mtON) to selectively increase the PMF in Caenorhabditis elegans. Here we show that mtON photoactivation increases the PMF in a dose-dependent manner, supports ATP synthesis, increases resistance to mitochondrial toxins, and modulates energy-sensing behavior. Moreover, transient mtON activation during hypoxic preconditioning prevents the well-characterized adaptive response of hypoxia resistance. Our results show that optogenetic manipulation of the PMF is a powerful tool to modulate metabolism and cell signaling.


Assuntos
Mitocôndrias , Optogenética , Trifosfato de Adenosina , Animais , Caenorhabditis elegans/genética , Hipóxia , Mitocôndrias/genética , Prótons
17.
Antioxidants (Basel) ; 8(8)2019 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-31390791

RESUMO

Mitochondrial reactive oxygen species (ROS) can be either detrimental or beneficial depending on the amount, duration, and location of their production. Mitochondrial complex I is a component of the electron transport chain and transfers electrons from NADH to ubiquinone. Complex I is also a source of ROS production. Under certain thermodynamic conditions, electron transfer can reverse direction and reduce oxygen at complex I to generate ROS. Conditions that favor this reverse electron transport (RET) include highly reduced ubiquinone pools, high mitochondrial membrane potential, and accumulated metabolic substrates. Historically, complex I RET was associated with pathological conditions, causing oxidative stress. However, recent evidence suggests that ROS generation by complex I RET contributes to signaling events in cells and organisms. Collectively, these studies demonstrate that the impact of complex I RET, either beneficial or detrimental, can be determined by the timing and quantity of ROS production. In this article we review the role of site-specific ROS production at complex I in the contexts of pathology and physiologic signaling.

18.
G3 (Bethesda) ; 9(8): 2629-2635, 2019 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-31186306

RESUMO

The introduction of exogenous genes in single-copy at precise genomic locations is a powerful tool that has been widely used in the model organism Caenorhabditis elegans Here, we have streamlined the process by creating a rapid, cloning-free method of single-copy transgene insertion we call Mos1 element-mediated CRISPR integration (mmCRISPi). The protocol combines the impact of Mos1 mediated single-copy gene insertion (mosSCI) with the ease of CRISPR/Cas9 mediated gene editing, allowing in vivo construction of transgenes from linear DNA fragments integrated at defined loci in the C. elegans genome. This approach was validated by defining its efficiency at different integration sites in the genome and by testing transgene insert size. The mmCRISPi method benefits from in vivo recombination of overlapping PCR fragments, allowing researchers to mix-and-match between promoters, protein-coding sequences, and 3' untranslated regions, all inserted in a single step at a defined Mos1 loci.


Assuntos
Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Proteínas de Ligação a DNA/metabolismo , Transgenes , Transposases/metabolismo , Animais , Animais Geneticamente Modificados , Sistemas CRISPR-Cas , Edição de Genes , Engenharia Genética
19.
Antioxid Redox Signal ; 31(9): 591-593, 2019 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-31084372

RESUMO

The cell maintains a balance between the production and removal of reactive oxygen species (ROS). Changes in ROS levels can impact many cellular functions, and dysregulation contributes to pathologies. How a specific cellular environment or microdomain influences the ROS-generating systems and biological impact of ROS remains an active area of research. This Forum highlights the complexity of ROS microdomains and their contributions to health and disease. Novel technologies to measure or generate ROS in defined regions are important developments in the spatial control of ROS. Using these advances, the articles herein demonstrate how site-specific redox environments influence cellular function and pathology. Antioxid. Redox Signal. 31, 591-593.


Assuntos
Oxirredução , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Suscetibilidade a Doenças , Homeostase , Humanos , Espaço Intracelular/metabolismo , Mitocôndrias/metabolismo
20.
Antioxid Redox Signal ; 31(9): 594-607, 2019 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-30887829

RESUMO

Aims: How mitochondrial reactive oxygen species (ROS) impact physiological function may depend on the quantity of ROS generated or removed, and the subcellular microdomain in which this occurs. However, pharmacological tools currently available to alter ROS production in vivo lack precise spatial and temporal control. Results: We used CRISPR/Cas9 to fuse the light-sensitive ROS-generating protein, SuperNova to the C-terminus of mitochondrial complex II succinate dehydrogenase subunits B (SDHB-1::SuperNova) and C (SDHC-1::SuperNova) in Caenorhabditis elegans to localize SuperNova to the matrix-side of the inner mitochondrial membrane, and to the intermembrane space (IMS), respectively. The presence of the SuperNova protein did not impact complex II activity, mitochondrial respiration, or C. elegans development rate under dark conditions. ROS production by SuperNova protein in vitro in the form of superoxide (O2˙-) was both specific and proportional to total light irradiance in the 540-590 nm spectra, and was unaffected by varying the buffer pH to resemble the mitochondrial matrix or IMS environments. We then determined using SuperNova whether stoichiometric ROS generation in the mitochondrial matrix or IMS had distinct effects on redox signaling in vivo. Phosphorylation of PMK-1 (a p38 MAPK homolog) and transcriptional activity of SKN-1 (an Nrf2 homolog) were each dependent on both the site and duration of ROS production, with matrix-generated ROS having more prominent effects. Furthermore, matrix- but not IMS-generated ROS attenuated susceptibility to simulated ischemia reperfusion injury in C. elegans. Innovation and Conclusion: Overall, these data demonstrate that the physiological output of ROS depends on the microdomain in which it is produced. Antioxid. Redox Signal. 31, 594-607.


Assuntos
Caenorhabditis elegans/metabolismo , Complexo II de Transporte de Elétrons/metabolismo , Microdomínios da Membrana/metabolismo , Mitocôndrias/metabolismo , Oxirredução , Estresse Oxidativo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Animais , Proteínas Recombinantes de Fusão , Traumatismo por Reperfusão/etiologia , Traumatismo por Reperfusão/metabolismo , Superóxidos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...