Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 111
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 182(3): 641-654.e20, 2020 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-32615085

RESUMEN

Targeting glycolysis has been considered therapeutically intractable owing to its essential housekeeping role. However, the context-dependent requirement for individual glycolytic steps has not been fully explored. We show that CRISPR-mediated targeting of glycolysis in T cells in mice results in global loss of Th17 cells, whereas deficiency of the glycolytic enzyme glucose phosphate isomerase (Gpi1) selectively eliminates inflammatory encephalitogenic and colitogenic Th17 cells, without substantially affecting homeostatic microbiota-specific Th17 cells. In homeostatic Th17 cells, partial blockade of glycolysis upon Gpi1 inactivation was compensated by pentose phosphate pathway flux and increased mitochondrial respiration. In contrast, inflammatory Th17 cells experience a hypoxic microenvironment known to limit mitochondrial respiration, which is incompatible with loss of Gpi1. Our study suggests that inhibiting glycolysis by targeting Gpi1 could be an effective therapeutic strategy with minimum toxicity for Th17-mediated autoimmune diseases, and, more generally, that metabolic redundancies can be exploited for selective targeting of disease processes.


Asunto(s)
Diferenciación Celular/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Glucosa-6-Fosfato Isomerasa/metabolismo , Glucólisis/genética , Fosforilación Oxidativa , Vía de Pentosa Fosfato/fisiología , Células Th17/metabolismo , Animales , Hipoxia de la Célula/genética , Hipoxia de la Célula/inmunología , Quimera/genética , Cromatografía de Gases , Cromatografía Liquida , Infecciones por Clostridium/inmunología , Citocinas/deficiencia , Citocinas/genética , Citocinas/metabolismo , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/metabolismo , Glucosa-6-Fosfato Isomerasa/genética , Gliceraldehído-3-Fosfato Deshidrogenasa (Fosforilante)/genética , Gliceraldehído-3-Fosfato Deshidrogenasa (Fosforilante)/metabolismo , Glucólisis/inmunología , Homeostasis/genética , Homeostasis/inmunología , Inflamación/genética , Inflamación/inmunología , Espectrometría de Masas , Ratones , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Membrana Mucosa/inmunología , Membrana Mucosa/metabolismo , Membrana Mucosa/microbiología , Vía de Pentosa Fosfato/genética , Vía de Pentosa Fosfato/inmunología , RNA-Seq , Análisis de la Célula Individual , Células Th17/inmunología , Células Th17/patología
2.
Proc Natl Acad Sci U S A ; 121(20): e2318119121, 2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38709930

RESUMEN

Brain metastasis of advanced breast cancer often results in deleterious consequences. Metastases to the brain lead to significant challenges in treatment options, as the blood-brain barrier (BBB) prevents conventional therapy. Thus, we hypothesized that creation of a nanoparticle (NP) that distributes to both primary tumor site and across the BBB for secondary brain tumor can be extremely beneficial. Here, we report a simple targeting strategy to attack both the primary breast and secondary brain tumors utilizing a single NP platform. The nature of these mitochondrion-targeted, BBB-penetrating NPs allow for simultaneous targeting and drug delivery to the hyperpolarized mitochondrial membrane of the extracranial primary tumor site in addition to tumors at the brain. By utilizing a combination of such dual anatomical distributing NPs loaded with therapeutics, we demonstrate a proof-of-concept idea to combat the increased metabolic plasticity of brain metastases by lowering two major energy sources, oxidative phosphorylation (OXPHOS) and glycolysis. By utilizing complementary studies and genomic analyses, we demonstrate the utility of a chemotherapeutic prodrug to decrease OXPHOS and glycolysis by pairing with a NP loaded with pyruvate dehydrogenase kinase 1 inhibitor. Decreasing glycolysis aims to combat the metabolic flexibility of both primary and secondary tumors for therapeutic outcome. We also address the in vivo safety parameters by addressing peripheral neuropathy and neurobehavior outcomes. Our results also demonstrate that this combination therapeutic approach utilizes mitochondrial genome targeting strategy to overcome DNA repair-based chemoresistance mechanisms.


Asunto(s)
Barrera Hematoencefálica , Neoplasias Encefálicas , Neoplasias de la Mama , Nanopartículas , Fosforilación Oxidativa , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/secundario , Neoplasias Encefálicas/patología , Animales , Humanos , Femenino , Nanopartículas/química , Ratones , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/efectos de los fármacos , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Fosforilación Oxidativa/efectos de los fármacos , Línea Celular Tumoral , Mitocondrias/metabolismo , Mitocondrias/efectos de los fármacos , Sistemas de Liberación de Medicamentos/métodos , Glucólisis/efectos de los fármacos , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Profármacos/farmacología , Profármacos/uso terapéutico
3.
Proc Natl Acad Sci U S A ; 120(21): e2217826120, 2023 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-37192160

RESUMEN

Molecular classification of gastric cancer (GC) identified a subgroup of patients showing chemoresistance and poor prognosis, termed SEM (Stem-like/Epithelial-to-mesenchymal transition/Mesenchymal) type in this study. Here, we show that SEM-type GC exhibits a distinct metabolic profile characterized by high glutaminase (GLS) levels. Unexpectedly, SEM-type GC cells are resistant to glutaminolysis inhibition. We show that under glutamine starvation, SEM-type GC cells up-regulate the 3 phosphoglycerate dehydrogenase (PHGDH)-mediated mitochondrial folate cycle pathway to produce NADPH as a reactive oxygen species scavenger for survival. This metabolic plasticity is associated with globally open chromatin structure in SEM-type GC cells, with ATF4/CEBPB identified as transcriptional drivers of the PHGDH-driven salvage pathway. Single-nucleus transcriptome analysis of patient-derived SEM-type GC organoids revealed intratumoral heterogeneity, with stemness-high subpopulations displaying high GLS expression, a resistance to GLS inhibition, and ATF4/CEBPB activation. Notably, coinhibition of GLS and PHGDH successfully eliminated stemness-high cancer cells. Together, these results provide insight into the metabolic plasticity of aggressive GC cells and suggest a treatment strategy for chemoresistant GC patients.


Asunto(s)
Fosfoglicerato-Deshidrogenasa , Neoplasias Gástricas , Humanos , Fosfoglicerato-Deshidrogenasa/genética , Fosfoglicerato-Deshidrogenasa/metabolismo , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/genética , Línea Celular Tumoral , Glutamina/metabolismo , Nutrientes
4.
EMBO Rep ; 24(12): e57440, 2023 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-37885348

RESUMEN

Embryogenesis is highly dependent on maternally loaded materials, particularly those used for energy production. Different environmental conditions and genetic backgrounds shape embryogenesis. The robustness of embryogenesis in response to extrinsic and intrinsic changes remains incompletely understood. By analyzing the levels of two major nutrients, glycogen and neutral lipids, we discovered stage-dependent usage of these two nutrients along with mitochondrial morphology changes during Caenorhabditis elegans embryogenesis. ATGL, the rate-limiting lipase in cellular lipolysis, is expressed and required in the hypodermis to regulate mitochondrial function and support embryogenesis. The embryonic lethality of atgl-1 mutants can be suppressed by reducing sinh-1/age-1-akt signaling, likely through modulating glucose metabolism to maintain sustainable glucose consumption. The embryonic lethality of atgl-1(xd314) is also affected by parental nutrition. Parental glucose and oleic acid supplements promote glycogen storage in atgl-1(xd314) embryos to compensate for the impaired lipolysis. The rescue by parental vitamin B12 supplement is likely through enhancing mitochondrial function in atgl-1 mutants. These findings reveal that metabolic plasticity contributes to the robustness of C. elegans embryogenesis.


Asunto(s)
Caenorhabditis elegans , Lipólisis , Animales , Caenorhabditis elegans/metabolismo , Lipólisis/genética , Lipasa/genética , Glucosa/metabolismo , Glucógeno/metabolismo
5.
Am J Physiol Endocrinol Metab ; 326(3): E366-E381, 2024 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-38197792

RESUMEN

Mammalian oocytes develop and mature in a mutually dependent relationship with surrounding cumulus cells. The oocyte actively regulates cumulus cell differentiation and function by secreting soluble paracrine oocyte-secreted factors (OSFs). We characterized the molecular mechanisms by which two model OSFs, cumulin and BMP15, regulate oocyte maturation and cumulus-oocyte cooperativity. Exposure to these OSFs during mouse oocyte maturation in vitro altered the proteomic and multispectral autofluorescence profiles of both the oocyte and cumulus cells. In oocytes, cumulin significantly upregulated proteins involved in nuclear function. In cumulus cells, both OSFs elicited marked upregulation of a variety of metabolic processes (mostly anabolic), including lipid, nucleotide, and carbohydrate metabolism, whereas mitochondrial metabolic processes were downregulated. The mitochondrial changes were validated by functional assays confirming altered mitochondrial morphology, respiration, and content while maintaining ATP homeostasis. Collectively, these data demonstrate that cumulin and BMP15 remodel cumulus cell metabolism, instructing them to upregulate their anabolic metabolic processes, while routine cellular functions are minimized in the oocyte during maturation, in preparation for ensuing embryonic development.NEW & NOTEWORTHY Oocyte-secreted factors (OSFs) promote oocyte and cumulus cell cooperativity by altering the molecular composition of both cell types. OSFs downregulate protein catabolic processes and upregulate processes associated with DNA binding, translation, and ribosome assembly in oocytes. In cumulus cells, OSFs alter mitochondrial number, morphology, and function, and enhance metabolic plasticity by upregulating anabolic pathways. Hence, the oocyte via OSFs, instructs cumulus cells to increase metabolic processes on its behalf, thereby subduing oocyte metabolism.


Asunto(s)
Células del Cúmulo , Proteómica , Embarazo , Femenino , Animales , Ratones , Células del Cúmulo/metabolismo , Oocitos/metabolismo , Comunicación Celular , Desarrollo Embrionario , Técnicas de Maduración In Vitro de los Oocitos , Mamíferos
6.
Development ; 148(11)2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-34117889

RESUMEN

The intimate relationships between cell fate and metabolism have long been recognized, but a mechanistic understanding of how metabolic pathways are dynamically regulated during development and disease, how they interact with signalling pathways, and how they affect differential gene expression is only emerging now. We summarize the key findings and the major themes that emerged from the virtual Keystone Symposium 'Metabolic Decisions in Development and Disease' held in March 2021.


Asunto(s)
Enfermedad , Crecimiento y Desarrollo , Redes y Vías Metabólicas , Animales , Diferenciación Celular , Microbioma Gastrointestinal , Expresión Génica , Humanos , Transducción de Señal
7.
J Exp Biol ; 227(6)2024 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-38380562

RESUMEN

From bacteria to metazoans, higher density populations have lower per capita metabolic rates than lower density populations. The negative covariance between population density and metabolic rate is thought to represent a form of adaptive metabolic plasticity. A relationship between density and metabolism was actually first noted 100 years ago, and was focused on spermatozoa; even then, it was postulated that adaptive plasticity drove this pattern. Since then, contemporary studies of sperm metabolism specifically assume that sperm concentration has no effect on metabolism and that sperm metabolic rates show no adaptive plasticity. We did a systematic review to estimate the relationship between sperm aerobic metabolism and sperm concentration, for 198 estimates spanning 49 species, from protostomes to humans from 88 studies. We found strong evidence that per capita metabolic rates are concentration dependent: both within and among species, sperm have lower metabolisms in dense ejaculates, but increase their metabolism when diluted. On average, a 10-fold decrease in sperm concentration increased per capita metabolic rate by 35%. Metabolic plasticity in sperm appears to be an adaptive response, whereby sperm maximize their chances of encountering eggs.


Asunto(s)
Espermatozoides , Masculino , Espermatozoides/metabolismo , Espermatozoides/fisiología , Animales , Humanos , Densidad de Población , Metabolismo Energético , Recuento de Espermatozoides
8.
J Theor Biol ; 590: 111854, 2024 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-38763324

RESUMEN

Marine mixotrophs combine phagotrophy and phototrophy to acquire the resources they need for growth. Metabolic plasticity, the ability for individuals to dynamically alter their relative investment between different metabolic processes, allows mixotrophs to efficiently exploit variable environmental conditions. Different mixotrophs may vary in how quickly they respond to environmental stimuli, with slow-responding mixotrophs exhibiting a significant lag between a change in the environment and the resulting change metabolic strategy. In this study, we develop a model of mixotroph metabolic strategy and explore how the rate of the plastic response affects the seasonality, competitive fitness, and biogeochemical role of mixotroph populations. Fast-responding mixotrophs are characterized by more efficient resource use and higher average growth rates than slow-responding mixotrophs because any lag in the plastic response following a change in environmental conditions creates a mismatch between the mixotroph's metabolic requirements and their resource acquisition. However, this mismatch also results in increased storage of unused resources that support growth under future nutrient-limited conditions. As a result of this trade-off, mixotroph biomass and productivity are maximized at intermediate plastic response rates. Furthermore, the trade-off represents a mechanism for coexistence between fast-responding and slow-responding mixotrophs. In mixed communities, fast-responding mixotrophs are numerically dominant, but slow-responding mixotrophs persist at low abundance due to the provisioning effect that emerges as a result of their less efficient resource acquisition strategy. In addition to increased competitive ability, fast-responding mixotrophs are, on average, more autotrophic than slow-responding mixotrophs. Notably, these trade-offs associated with mixotroph response rate arise without including an explicit physiological cost associated with plasticity, a conclusion that may provide insight into evolutionary constraints of metabolic plasticity in mixotrophic organisms. When an explicit cost is added to the model, it alters the competitive relationships between fast- and slow-responding mixotrophs. Faster plastic response rates are favored by lower physiological costs as well as higher amplitude seasonal cycles.


Asunto(s)
Modelos Biológicos , Biomasa , Adaptación Fisiológica , Estaciones del Año , Ecosistema
9.
Semin Cancer Biol ; 85: 196-208, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-34500075

RESUMEN

Autophagy, a lysosomal catabolic process, involves degradation of cellular materials, protein aggregate, and dysfunctional organelles to maintain cellular homeostasis. Strikingly, autophagy exhibits a dual-sided role in cancer; on the one hand, it promotes clearance of transformed cells and inhibits tumorigenesis, while cytoprotective autophagy has a role in sustaining cancer. The autophagy signaling in the tumor microenvironment (TME) during cancer growth and therapy is not adequately understood. The review highlights the role of autophagy signaling pathways to support cancer growth and progression in adaptation to the oxidative and hypoxic context of TME. Furthermore, autophagy contributes to regulating the metabolic switch for generating sufficient levels of high-energy metabolites, including amino acids, ketones, glutamine, and free fatty acids for cancer cell survival. Interestingly, autophagy has a critical role in modulating the tumor-associated fibroblast resulting in different cytokines and paracrine signaling mediated angiogenesis and invasion of pre-metastatic niches to secondary tumor sites. Moreover, autophagy promotes immune evasion to inhibit antitumor immunity, and autophagy inhibitors enhance response to immunotherapy with infiltration of immune cells to the TME niche. Furthermore, autophagy in TME maintains and supports the survival of cancer stem cells resulting in chemoresistance and therapy recurrence. Presently, drug repurposing has enabled the use of lysosomal inhibitor-based antimalarial drugs like chloroquine and hydroxychloroquine as clinically available autophagy inhibitors in cancer therapy. We focus on the recent developments of multiple autophagy modulators from pre-clinical trials and the challenges in developing autophagy-based cancer therapy.


Asunto(s)
Neoplasias , Microambiente Tumoral , Humanos , Autofagia , Neoplasias/patología , Transducción de Señal , Neovascularización Patológica/metabolismo
10.
Mycoses ; 66(5): 430-440, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-36564594

RESUMEN

BACKGROUND: The Trichosporonaceae family comprises a large number of basidiomycetes widely distributed in nature. Some of its members, especially Trichosporon asahii, have the ability to cause human infections. This ability is related to a series of virulence factors, which include lytic enzymes production, biofilm formation, resistance to oxidising agents, melanin and glucuronoxylomannan in the cell wall, metabolic plasticity and phenotypic switching. The last two are poorly addressed within human pathogenic Trichosporonaceae. OBJECTIVE: These factors were herein studied to contribute with the knowledge of these emerging pathogens and to uncover mechanisms that would explain the higher frequency of T. asahii in human infections. METHODS: We included 79 clinical isolates phenotypically identified as Trichosporon spp. and performed their molecular identification. Lactate and N-acetyl glucosamine were the carbon sources of metabolic plasticity studies. Morphologically altered colonies after subcultures and incubation at 37°C indicated phenotypic switching. RESULTS AND CONCLUSION: The predominant species was T. asahii (n = 65), followed by Trichosporon inkin (n = 4), Apiotrichum montevideense (n = 3), Trichosporon japonicum (n = 2), Trichosporon faecale (n = 2), Cutaneotrichosporon debeurmannianum (n = 1), Trichosporon ovoides (n = 1) and Cutaneotrichosporon arboriforme (n = 1). T. asahii isolates had statistically higher growth on lactate and N-acetylglucosamine and on glucose during the first 72 h of culture. T. asahii, T. inkin and T. japonicum isolates were able to perform phenotypic switching. These results expand the virulence knowledge of Trichosporonaceae members and point for a role for metabolic plasticity and phenotypic switching on the trichosporonosis pathogenesis.


Asunto(s)
Basidiomycota , Trichosporon , Tricosporonosis , Humanos , Antifúngicos , Trichosporon/genética , Virulencia , Adaptación Fisiológica , Lactatos
11.
Int J Mol Sci ; 24(8)2023 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-37108242

RESUMEN

One important feature of tumour development is the regulatory role of metabolic plasticity in maintaining the balance of mitochondrial oxidative phosphorylation and glycolysis in cancer cells. In recent years, the transition and/or function of metabolic phenotypes between mitochondrial oxidative phosphorylation and glycolysis in tumour cells have been extensively studied. In this review, we aimed to elucidate the characteristics of metabolic plasticity (emphasizing their effects, such as immune escape, angiogenesis migration, invasiveness, heterogeneity, adhesion, and phenotypic properties of cancers, among others) on tumour progression, including the initiation and progression phases. Thus, this article provides an overall understanding of the influence of abnormal metabolic remodeling on malignant proliferation and pathophysiological changes in carcinoma.


Asunto(s)
Carcinoma , Fosforilación Oxidativa , Humanos , Glucólisis , Mitocondrias/metabolismo , Transformación Celular Neoplásica/metabolismo , Carcinoma/metabolismo
12.
Int J Mol Sci ; 24(13)2023 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-37446195

RESUMEN

Like other chronic viral infections, HIV-1 persistence inhibits the development of antigen-specific memory T-cells, resulting in the exhaustion of the immune response and chronic inflammation. Autophagy is a major lysosome-dependent mechanism of intracellular large-target degradation such as lipid and protein aggregates, damaged organelles, and intracellular pathogens. Although it is known that autophagy may target HIV-1 for elimination, knowledge of its function as a metabolic contributor in such viral infection is only in its infancy. Recent data show that elite controllers (EC), who are HIV-1-infected subjects with natural and long-term antigen (Ag)-specific T-cell protection against the virus, are characterized by distinct metabolic autophagy-dependent features in their T-cells compared to other people living with HIV-1 (PLWH). Despite durable viral control with antiretroviral therapy (ART), HIV-1-specific immune dysfunction does not normalize in non-controller PLWH. Therefore, the hypothesis of inducing autophagy to strengthen their Ag-specific T-cell immunity against HIV-1 starts to be an enticing concept. The aim of this review is to critically analyze promises and potential limitations of pharmacological and dietary interventions to activate autophagy in an attempt to rescue Ag-specific T-cell protection among PLWH.


Asunto(s)
Infecciones por VIH , VIH-1 , Humanos , VIH-1/fisiología , Linfocitos T CD4-Positivos , Linfocitos T CD8-positivos , Inflamación/metabolismo
13.
Int J Mol Sci ; 24(9)2023 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-37175952

RESUMEN

Phosphoglucomutase 1 (PGM1) is a key enzyme for the regulation of energy metabolism from glycogen and glycolysis, as it catalyzes the interconversion of glucose 1-phosphate and glucose 6-phosphate. PGM1 deficiency is an autosomal recessive disorder characterized by a highly heterogenous clinical spectrum, including hypoglycemia, cleft palate, liver dysfunction, growth delay, exercise intolerance, and dilated cardiomyopathy. Abnormal protein glycosylation has been observed in this disease. Oral supplementation with D-galactose efficiently restores protein glycosylation by replenishing the lacking pool of UDP-galactose, and rescues some symptoms, such as hypoglycemia, hepatopathy, and growth delay. However, D-galactose effects on skeletal muscle and heart symptoms remain unclear. In this study, we established an in vitro muscle model for PGM1 deficiency to investigate the role of PGM1 and the effect of D-galactose on nucleotide sugars and energy metabolism. Genome-editing of C2C12 myoblasts via CRISPR/Cas9 resulted in Pgm1 (mouse homologue of human PGM1, according to updated nomenclature) knockout clones, which showed impaired maturation to myotubes. No difference was found for steady-state levels of nucleotide sugars, while dynamic flux analysis based on 13C6-galactose suggested a block in the use of galactose for energy production in knockout myoblasts. Subsequent analyses revealed a lower basal respiration and mitochondrial ATP production capacity in the knockout myoblasts and myotubes, which were not restored by D-galactose. In conclusion, an in vitro mouse muscle cell model has been established to study the muscle-specific metabolic mechanisms in PGM1 deficiency, which suggested that galactose was unable to restore the reduced energy production capacity.


Asunto(s)
Hipoglucemia , Fosfoglucomutasa , Animales , Ratones , Galactosa/farmacología , Glucosa , Homeostasis , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/metabolismo , Nucleótidos , Fosfatos , Fosfoglucomutasa/genética , Fosfoglucomutasa/metabolismo
14.
Cancer Metastasis Rev ; 40(4): 989-1033, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-35029792

RESUMEN

Despite advancements in cancer management, tumor relapse and metastasis are associated with poor outcomes in many cancers. Over the past decade, oncogene-driven carcinogenesis, dysregulated cellular signaling networks, dynamic changes in the tissue microenvironment, epithelial-mesenchymal transitions, protein expression within regulatory pathways, and their part in tumor progression are described in several studies. However, the complexity of metabolic enzyme expression is considerably under evaluated. Alterations in cellular metabolism determine the individual phenotype and behavior of cells, which is a well-recognized hallmark of cancer progression, especially in the adaptation mechanisms underlying therapy resistance. In metabolic symbiosis, cells compete, communicate, and even feed each other, supervised by tumor cells. Metabolic reprogramming forms a unique fingerprint for each tumor tissue, depending on the cellular content and genetic, epigenetic, and microenvironmental alterations of the developing cancer. Based on its sensing and effector functions, the mechanistic target of rapamycin (mTOR) kinase is considered the master regulator of metabolic adaptation. Moreover, mTOR kinase hyperactivity is associated with poor prognosis in various tumor types. In situ metabolic phenotyping in recent studies highlights the importance of metabolic plasticity, mTOR hyperactivity, and their role in tumor progression. In this review, we update recent developments in metabolic phenotyping of the cancer ecosystem, metabolic symbiosis, and plasticity which could provide new research directions in tumor biology. In addition, we suggest pathomorphological and analytical studies relating to metabolic alterations, mTOR activity, and their associations which are necessary to improve understanding of tumor heterogeneity and expand the therapeutic management of cancer.


Asunto(s)
Ecosistema , Neoplasias , Carcinogénesis/metabolismo , Humanos , Neoplasias/patología , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Microambiente Tumoral
15.
Proc Biol Sci ; 289(1975): 20220427, 2022 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-35611529

RESUMEN

Many species up- or downregulate their resting metabolic rate (RMR) when they encounter favourable or unfavourable feeding conditions, respectively. This is thought to promote faster growth when food is abundant and conserve energy reserves when food is scarce. The time it takes to express metabolic plasticity remain little studied. Here, we develop a conceptual model showing how rapid or slow metabolic plasticity alter growth trajectories in response to changes in food supply. We test predictions from the model in a food manipulation experiment with young-of-the-year northern pike, Esox lucius, a species that experience drastic changes in food supply in nature. We find that metabolic plasticity is expressed gradually over several weeks in this species. Rapid changes in food supply thus caused apparent trait-environment mismatches that persisted for at least five weeks. Contrary to predictions, pike grew faster at high food levels when they had previously experienced low food levels and downregulated their RMR. This was not owing to increases in food intake but probably reflected that low RMRs increased the energetic scope for growth when feeding conditions improved. This highlights the important but complex effects of metabolic plasticity on growth dynamics under variable resource levels on ecologically relevant time scales.


Asunto(s)
Metabolismo Basal , Esocidae , Animales , Metabolismo Energético , Esocidae/metabolismo , Abastecimiento de Alimentos
16.
Drug Resist Updat ; 59: 100797, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34955385

RESUMEN

Despite an increasing arsenal of anticancer therapies, many patients continue to have poor outcomes due to the therapeutic failures and tumor relapses. Indeed, the clinical efficacy of anticancer therapies is markedly limited by intrinsic and/or acquired resistance mechanisms that can occur in any tumor type and with any treatment. Thus, there is an urgent clinical need to implement fundamental changes in the tumor treatment paradigm by the development of new experimental strategies that can help to predict the occurrence of clinical drug resistance and to identify alternative therapeutic options. Apart from mutation-driven resistance mechanisms, tumor microenvironment (TME) conditions generate an intratumoral phenotypic heterogeneity that supports disease progression and dismal outcomes. Tumor cell metabolism is a prototypical example of dynamic, heterogeneous, and adaptive phenotypic trait, resulting from the combination of intrinsic [(epi)genetic changes, tissue of origin and differentiation dependency] and extrinsic (oxygen and nutrient availability, metabolic interactions within the TME) factors, enabling cancer cells to survive, metastasize and develop resistance to anticancer therapies. In this review, we summarize the current knowledge regarding metabolism-based mechanisms conferring adaptive resistance to chemo-, radio-and immunotherapies as well as targeted therapies. Furthermore, we report the role of TME-mediated intratumoral metabolic heterogeneity in therapy resistance and how adaptations in amino acid, glucose, and lipid metabolism support the growth of therapy-resistant cancers and/or cellular subpopulations. We also report the intricate interplay between tumor signaling and metabolic pathways in cancer cells and discuss how manipulating key metabolic enzymes and/or providing dietary changes may help to eradicate relapse-sustaining cancer cells. Finally, in the current era of personalized medicine, we describe the strategies that may be applied to implement metabolic profiling for tumor imaging, biomarker identification, selection of tailored treatments and monitoring therapy response during the clinical management of cancer patients.


Asunto(s)
Neoplasias , Microambiente Tumoral , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Humanos , Inmunoterapia/métodos , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Medicina de Precisión
17.
Int J Mol Sci ; 23(20)2022 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-36293260

RESUMEN

Abnormal glucose metabolism is central to neurodegeneration, and considerable evidence suggests that abnormalities in key enzymes of the tricarboxylic acid (TCA) cycle underlie the metabolic deficits. Significant recent advances in the role of metabolism in cancer provide new insight that facilitates our understanding of the role of metabolism in neurodegeneration. Research indicates that the rate-limiting step of the TCA cycle, the α-ketoglutarate dehydrogenase complex (KGDHC) and its substrate alpha ketoglutarate (KG), serve as a signaling hub that regulates multiple cellular processes: (1) is the rate-limiting step of the TCA cycle, (2) is sensitive to reactive oxygen species (ROS) and produces ROS, (3) determines whether KG is used for energy or synthesis of compounds to support growth, (4) regulates the cellular responses to hypoxia, (5) controls the post-translational modification of hundreds of cell proteins in the mitochondria, cytosol, and nucleus through succinylation, (6) controls critical aspects of transcription, (7) modulates protein signaling within cells, and (8) modulates cellular calcium. The primary focus of this review is to understand how reductions in KGDHC are translated to pathologically important changes that underlie both neurodegeneration and cancer. An understanding of each role is necessary to develop new therapeutic strategies to treat neurodegenerative disease.


Asunto(s)
Complejo Cetoglutarato Deshidrogenasa , Enfermedades Neurodegenerativas , Humanos , Complejo Cetoglutarato Deshidrogenasa/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Calcio/metabolismo , Ácidos Cetoglutáricos , Glucosa , Ácidos Tricarboxílicos
18.
Semin Cancer Biol ; 66: 45-58, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-31351198

RESUMEN

Mitophagy is an evolutionarily conserved cellular process which selectively eliminates dysfunctional mitochondria by targeting them to the autophagosome for degradation. Dysregulated mitophagy results in the accumulation of damaged mitochondria, which plays an important role in carcinogenesis and tumor progression. The role of mitophagy receptors and adaptors including PINK1, Parkin, BNIP3, BNIP3L/NIX, and p62/SQSTM1, and the signaling pathways that govern mitophagy are impaired in cancer. Furthermore, the contribution of mitophagy in regulating the metabolic switch may establish a balance between aerobic glycolysis and oxidative phosphorylation for cancer cell survival. Moreover, ROS-driven mitophagy achieves different goals depending on the stage of tumorigenesis. Mitophagy promotes plasticity in the cancer stem cell through the metabolic reconfiguration for better adaption to the tumor microenvironment. In addition, the present review sheds some light on the role of mitophagy in stemness and differentiation during the transition of cell's fate, which could have a crucial role in cancer progression and metastasis. In conclusion, this review deals with the detailed molecular mechanisms underlying mitophagy, along with highlighting the dual role of mitophagy in different aspects of cancer, suggesting it as a possible target in the mitophagy-modulated cancer therapy.


Asunto(s)
Mitofagia/fisiología , Neoplasias/patología , Animales , Diferenciación Celular/fisiología , Humanos , Mitocondrias/patología , Células Madre Neoplásicas/patología , Transducción de Señal/fisiología
19.
Infect Immun ; 89(12): e0013521, 2021 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-34491791

RESUMEN

Coxiella burnetii, the causative agent of query (Q) fever in humans, is an obligate intracellular bacterium. C. burnetii can naturally infect a broad range of host organisms (e.g., mammals and arthropods) and cell types. This amphotropic nature of C. burnetii, in combination with its ability to utilize both glycolytic and gluconeogenic carbon sources, suggests that the pathogen relies on metabolic plasticity to replicate in nutritionally diverse intracellular environments. To test the significance of metabolic plasticity in C. burnetii host cell colonization, C. burnetii intracellular replication in seven distinct cell lines was compared between a metabolically competent parental strain and a mutant, CbΔpckA, unable to undergo gluconeogenesis. Both the parental strain and CbΔpckA mutant exhibited host cell-dependent infection phenotypes, which were influenced by alterations to host glycolytic or gluconeogenic substrate availability. Because the nutritional environment directly impacts host cell physiology, our analysis was extended to investigate the response of C. burnetii replication in mammalian host cells cultivated in a novel physiological medium based on the nutrient composition of mammalian interstitial fluid, interstitial fluid-modeled medium (IFmM). An infection model based on IFmM resulted in exacerbation of a replication defect exhibited by the CbΔpckA mutant in specific cell lines. The CbΔpckA mutant was also attenuated during infection of an animal host. Overall, the study underscores that gluconeogenic capacity aids C. burnetii amphotropism and that the amphotropic nature of C. burnetii should be considered when resolving virulence mechanisms in this pathogen.


Asunto(s)
Coxiella burnetii/fisiología , Metabolismo Energético , Interacciones Huésped-Patógeno , Fiebre Q/metabolismo , Fiebre Q/microbiología , Susceptibilidad a Enfermedades , Gluconeogénesis , Glucólisis , Humanos , Virulencia/genética , Factores de Virulencia/genética
20.
BMC Cancer ; 21(1): 1026, 2021 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-34525987

RESUMEN

BACKGROUND: Current treatment methods for patients with triple-negative breast cancer (TNBC) are very limited, and the prognosis of TNBC is relatively poor. It has been reported that glucose transporter 1 (GLUT1) is overexpressed in breast cancer cells; however, its association with the prognosis is mostly unclear. Moreover, retinoblastoma gene 1 (RB1) might be used as a biomarker for the sensitivity of breast cancer cells to GLUT1 inhibitors, which brought us to the hypothesis that there might be a close correlation between the expression of GLUT1-4 and the expression of RB1. METHODS: In this study, we systematically analyzed the co-expression of GLUT1-4 and the influence of GLUT1-4 gene expression on the prognosis of breast cancer using data mining methods. We also explored possible relationships between GLUT1-4 and RB1 expression in breast cancer tissues. We used public databases such as ONCOMINE, GEPIA, LinkedOmics, and COEXPEDIA. RESULTS: According to the results, the mRNA expression of SLC2A1 was significantly higher in breast cancer, while the expression levels of SLC2A2-4 were downregulated. The results also indicate that GLUT1 expression does not have significant influence on the overall survival of patients with breast cancer. The mRNA expression of SLC2A1 and RB1 is significantly correlated, which means that tissues with high RB1 mRNA expression might have relatively higher mRNA expression of SLC2A1; however, further study analyzing their roles in the expression regulation pathways with human samples is needed to verify the hypothesis. CONCLUSIONS: The mRNA expression of SLC2A1 was significantly higher in breast cancer. The overall survival of breast cancer patients wasn't significantly correlated with GLUT1-4 expression. The mRNA expression of SLC2A1 and RB1 is significantly correlated according to the analysis conducted in LinkedOmics. It provides reference for future possible individualized treatment of TNBC using GLUT1 inhibitors, especially in patients with higher mRNA expression of RB1. Further study analyzing the roles of these two genes in the regulation pathways is needed.


Asunto(s)
Transportador de Glucosa de Tipo 1/metabolismo , Transportador de Glucosa de Tipo 2/metabolismo , Transportador de Glucosa de Tipo 3/metabolismo , Transportador de Glucosa de Tipo 4/metabolismo , Proteínas de Unión a Retinoblastoma/metabolismo , Neoplasias de la Mama Triple Negativas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Bases de Datos Genéticas , Conjuntos de Datos como Asunto , Regulación hacia Abajo , Femenino , Genes de Retinoblastoma , Transportador de Glucosa de Tipo 1/genética , Transportador de Glucosa de Tipo 2/genética , Transportador de Glucosa de Tipo 3/genética , Transportador de Glucosa de Tipo 4/genética , Humanos , Pronóstico , ARN Mensajero/metabolismo , Proteínas de Unión a Retinoblastoma/genética , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/mortalidad , Ubiquitina-Proteína Ligasas/genética , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA