Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.613
Filtrar
1.
Theranostics ; 14(9): 3509-3525, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38948065

RESUMEN

Rationale: Current treatments for ocular angiogenesis primarily focus on blocking the activity of vascular endothelial growth factor (VEGF), but unfavorable side effects and unsatisfactory efficacy remain issues. The identification of novel targets for anti-angiogenic treatment is still needed. Methods: We investigated the role of tsRNA-1599 in ocular angiogenesis using endothelial cells, a streptozotocin (STZ)-induced diabetic model, a laser-induced choroidal neovascularization model, and an oxygen-induced retinopathy model. CCK-8 assays, EdU assays, transwell assays, and matrigel assays were performed to assess the role of tsRNA-1599 in endothelial cells. Retinal digestion assays, Isolectin B4 (IB4) staining, and choroidal sprouting assays were conducted to evaluate the role of tsRNA-1599 in ocular angiogenesis. Transcriptomic analysis, metabolic analysis, RNA pull-down assays, and mass spectrometry were utilized to elucidate the mechanism underlying angiogenic effects mediated by tsRNA-1599. Results: tsRNA-1599 expression was up-regulated in experimental ocular angiogenesis models and endothelial cells in response to angiogenic stress. Silencing of tsRNA-1599 suppressed angiogenic effects in endothelial cells in vitro and inhibited pathological ocular angiogenesis in vivo. Mechanistically, tsRNA-1599 exhibited little effect on VEGF signaling but could cause reduced glycolysis and NAD+/NADH production in endothelial cells by regulating the expression of HK2 gene through interacting with YBX1, thus affecting endothelial effects. Conclusions: Targeting glycolytic reprogramming of endothelial cells by a tRNA-derived small RNA represents an exploitable therapeutic approach for ocular neovascular diseases.


Asunto(s)
Neovascularización Coroidal , Células Endoteliales , Glucólisis , Animales , Glucólisis/efectos de los fármacos , Ratones , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Neovascularización Coroidal/tratamiento farmacológico , Neovascularización Coroidal/metabolismo , Humanos , Proteína 1 de Unión a la Caja Y/metabolismo , Proteína 1 de Unión a la Caja Y/genética , Inhibidores de la Angiogénesis/farmacología , Hexoquinasa/metabolismo , Hexoquinasa/genética , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Experimental/metabolismo , Ratones Endogámicos C57BL , Masculino , Modelos Animales de Enfermedad , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/metabolismo , Neovascularización Patológica/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Retinopatía Diabética/tratamiento farmacológico , Retinopatía Diabética/metabolismo , Retinopatía Diabética/genética , Células Endoteliales de la Vena Umbilical Humana , ARN Pequeño no Traducido/genética , ARN Pequeño no Traducido/metabolismo
2.
Signal Transduct Target Ther ; 9(1): 158, 2024 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-38862461

RESUMEN

Cell membrane-camouflaged nanoparticles possess inherent advantages derived from their membrane structure and surface antigens, including prolonged circulation in the bloodstream, specific cell recognition and targeting capabilities, and potential for immunotherapy. Herein, we introduce a cell membrane biomimetic nanodrug platform termed MPB-3BP@CM NPs. Comprising microporous Prussian blue nanoparticles (MPB NPs) serving as both a photothermal sensitizer and carrier for 3-bromopyruvate (3BP), these nanoparticles are cloaked in a genetically programmable cell membrane displaying variants of signal regulatory protein α (SIRPα) with enhanced affinity to CD47. As a result, MPB-3BP@CM NPs inherit the characteristics of the original cell membrane, exhibiting an extended circulation time in the bloodstream and effectively targeting CD47 on the cytomembrane of colorectal cancer (CRC) cells. Notably, blocking CD47 with MPB-3BP@CM NPs enhances the phagocytosis of CRC cells by macrophages. Additionally, 3BP, an inhibitor of hexokinase II (HK2), suppresses glycolysis, leading to a reduction in adenosine triphosphate (ATP) levels and lactate production. Besides, it promotes the polarization of tumor-associated macrophages (TAMs) towards an anti-tumor M1 phenotype. Furthermore, integration with MPB NPs-mediated photothermal therapy (PTT) enhances the therapeutic efficacy against tumors. These advantages make MPB-3BP@CM NPs an attractive platform for the future development of innovative therapeutic approaches for CRC. Concurrently, it introduces a universal approach for engineering disease-tailored cell membranes for tumor therapy.


Asunto(s)
Antígeno CD47 , Membrana Celular , Neoplasias Colorrectales , Nanopartículas , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/terapia , Nanopartículas/química , Humanos , Antígeno CD47/genética , Ratones , Membrana Celular/metabolismo , Membrana Celular/genética , Animales , Piruvatos/química , Piruvatos/farmacología , Hexoquinasa/genética , Línea Celular Tumoral , Macrófagos/metabolismo , Macrófagos/efectos de los fármacos , Ferrocianuros
3.
Nat Commun ; 15(1): 5410, 2024 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-38926365

RESUMEN

METTL3 is the catalytic subunit of the methyltransferase complex, which mediates m6A modification to regulate gene expression. In addition, METTL3 regulates transcription in an enzymatic activity-independent manner by driving changes in high-order chromatin structure. However, how these functions of the methyltransferase complex are coordinated remains unknown. Here we show that the methyltransferase complex coordinates its enzymatic activity-dependent and independent functions to regulate cellular senescence, a state of stable cell growth arrest. Specifically, METTL3-mediated chromatin loops induce Hexokinase 2 expression through the three-dimensional chromatin organization during senescence. Elevated Hexokinase 2 expression subsequently promotes liquid-liquid phase separation, manifesting as stress granule phase separation, by driving metabolic reprogramming. This correlates with an impairment of translation of cell-cycle related mRNAs harboring polymethylated m6A sites. In summary, our results report a coordination of m6A-dependent and -independent function of the methyltransferase complex in regulating senescence through phase separation driven by metabolic reprogramming.


Asunto(s)
Senescencia Celular , Cromatina , Metiltransferasas , Gránulos de Estrés , Metiltransferasas/metabolismo , Metiltransferasas/genética , Cromatina/metabolismo , Humanos , Gránulos de Estrés/metabolismo , Gránulos de Estrés/genética , Hexoquinasa/metabolismo , Hexoquinasa/genética , ARN Mensajero/metabolismo , ARN Mensajero/genética , Adenosina/metabolismo , Adenosina/análogos & derivados , Células HEK293 , Reprogramación Metabólica , Separación de Fases
4.
Chem Biol Interact ; 397: 111062, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38763349

RESUMEN

Acute lung injury is the leading cause of paraquat (PQ) poisoning-related mortality. The mechanism by which macrophages are involved in PQ-induced acute lung injury remains unclear. In recent years, the role of metabolic reprogramming in macrophage functional transformation has received significant attention. The current study aimed to identify the role of altered macrophage glucose metabolism and molecular mechanisms in PQ poisoning-induced acute lung injury. We established a model of acute lung injury in PQ-intoxicated mice via the intraperitoneal injection of PQ. PQ exposure induces macrophage M1 polarization and promotes the release of inflammatory factors, which causes the development of acute lung injury in mice. In vitro analysis revealed that PQ altered glucose metabolism, which could be reversed by siRNA transfection to silence the expression of HK1, a key enzyme in glucose metabolism. RNA sequencing revealed that the ERK/MAPK pathway was the crucial molecular mechanism of PQ pathogenesis. Further, U0126, an ERK inhibitor, could inhibit PQ-induced HK1 activation and macrophage M1 polarization. These findings provide novel insights into the previously unrecognized mechanism of ERK/MAPK-HK1 activation in PQ poisoning.


Asunto(s)
Lesión Pulmonar Aguda , Glucosa , Hexoquinasa , Sistema de Señalización de MAP Quinasas , Macrófagos , Ratones Endogámicos C57BL , Paraquat , Animales , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/metabolismo , Lesión Pulmonar Aguda/patología , Paraquat/toxicidad , Ratones , Glucosa/metabolismo , Macrófagos/metabolismo , Macrófagos/efectos de los fármacos , Hexoquinasa/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Transducción de Señal/efectos de los fármacos , Células RAW 264.7
5.
Cytokine ; 180: 156663, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38815522

RESUMEN

BACKGROUND: Glioma represents the predominant malignant brain tumor. This investigation endeavors to elucidate the impact and prospective mechanisms of glycolysis-related lncARSR on glioma. METHODS: LncARSR level was assessed in normal glial cells and glioma cells. Cell proliferation, migration, and invasion measurements were conducted through CCK-8, wound healing, and transwell assay. Flow cytometry was utilized to measure cell apoptosis and cell cycle. Biochemical assay kits and immunoblotting were employed to measure the content of glycolysis-related indicators and protein expression, respectively. We analyzed the impact of both lncARSR knockdown and overexpression of the Signal Transducer and Activator of Transcription 3 (STAT3) on Hexokinase 2 (HK2) using dual luciferase reporter assays and Chromatin Immunoprecipitation (ChIP) experiments. Further assessment of the impact of lncARSR on glioma progression was conducted through animal experiments. RESULTS: LncARSR was expressed at elevated levels in glioma cells compared to normal glial cells. Overexpressing lncARSR enhanced proliferation, migration, invasion, and G2/M phase arrest in glioma cells and also increased glucose, lactate, ATP production, as well as the expression of HK2, PFK1, PKM2, GLUT1, and LDHA. STAT3 binding to the HK2 gene promoter was weakened following the knockdown of lncARSR. Upregulation of STAT3 reversed the suppressed functions of knocking down lncARSR on cell proliferation, migration, invasion, G2/M phase arrest, and glycolysis and counteracted its promotional effect on cell apoptosis. In vivo, knocking down lncARSR inhibits glioma growth and ki67 and PCNA expression. CONCLUSION: LncARSR promotes the development of glioma by enhancing glycolysis through the STAT3-HK2 axis.


Asunto(s)
Movimiento Celular , Proliferación Celular , Glioma , Glucólisis , Hexoquinasa , ARN Largo no Codificante , Factor de Transcripción STAT3 , Factor de Transcripción STAT3/metabolismo , Glioma/metabolismo , Glioma/patología , Glioma/genética , Hexoquinasa/metabolismo , Hexoquinasa/genética , Humanos , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Línea Celular Tumoral , Animales , Movimiento Celular/genética , Ratones , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/genética , Regulación Neoplásica de la Expresión Génica , Ratones Desnudos , Apoptosis , Transducción de Señal
6.
Diabetes Res Clin Pract ; 212: 111721, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38821414

RESUMEN

AIMS: Autoantibodies against hexokinase 1 (HK1) were recently proposed to be associated with diabetic macular edema (DME). We hypothesized that anti-HK1 autoantibodies can be used as DME markers and to predict DME onset. MATERIALS AND METHODS: Serum from patients with 1) DME, 2) diabetes mellitus (DM), 3) allergies or autoimmunities, and 4) control subjects was tested for anti-HK1 and anti-hexokinase 2 (HK2) autoantibodies by immunoblotting. Patients with DM were prospectively followed for up to nine years, and the association of anti-HK1 antibodies with new-onset DME was evaluated. The vitreous humor was also tested for autoantibodies. RESULTS: Among patients with DME, 32 % were positive for anti-HK1 autoantibodies (42 % of those with underlying type 1 DM and 31 % of those with underlying type 2 DM), and 12 % were positive for anti-HK2 autoantibodies, with only partial overlap of these two groups of patients. Anti-HK1 positive were also 7 % of patients with DM, 6 % of patients with allergies and autoimmunities, and 3 % of control subjects. The latter three groups were anti-HK2 negative. Only one of seven patients with DM who were initially anti-HK1 positive developed DME. CONCLUSIONS: Anti-HK1 autoantibodies can be used as DME markers but fail to predict DME onset.


Asunto(s)
Autoanticuerpos , Retinopatía Diabética , Hexoquinasa , Edema Macular , Humanos , Hexoquinasa/inmunología , Autoanticuerpos/sangre , Autoanticuerpos/inmunología , Retinopatía Diabética/inmunología , Retinopatía Diabética/sangre , Masculino , Femenino , Persona de Mediana Edad , Edema Macular/inmunología , Edema Macular/sangre , Anciano , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/complicaciones , Diabetes Mellitus Tipo 1/sangre , Estudios Prospectivos , Adulto , Diabetes Mellitus Tipo 2/inmunología , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/sangre , Biomarcadores/sangre
7.
Cancer Immunol Immunother ; 73(7): 122, 2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38714539

RESUMEN

Neuroblastoma (NB) is the most common and deadliest extracranial solid tumor in children. Targeting tumor-associated macrophages (TAMs) is a strategy for attenuating tumor-promoting states. The crosstalk between cancer cells and TAMs plays a pivotal role in mediating tumor progression in NB. The overexpression of Hexokinase-3 (HK3), a pivotal enzyme in glucose metabolism, has been associated with poor prognosis in NB patients. Furthermore, it correlates with the infiltration of M2-like macrophages within NB tumors, indicating its significant involvement in tumor progression. Therefore, HK3 not only directly regulates the malignant biological behaviors of tumor cells, such as proliferation, migration, and invasion, but also recruits and polarizes M2-like macrophages through the PI3K/AKT-CXCL14 axis in neuroblastoma. The secretion of lactate and histone lactylation alterations within tumor cells accompanies this interaction. Additionally, elevated expression of HK3 in M2-TAMs was found at the same time. Modulating HK3 within M2-TAMs alters the biological behavior of tumor cells, as demonstrated by our in vitro studies. This study highlights the pivotal role of HK3 in the progression of NB malignancy and its intricate regulatory network with M2-TAMs. It establishes HK3 as a promising dual-functional biomarker and therapeutic target in combating neuroblastoma.


Asunto(s)
Hexoquinasa , Neuroblastoma , Macrófagos Asociados a Tumores , Neuroblastoma/metabolismo , Neuroblastoma/patología , Humanos , Hexoquinasa/metabolismo , Macrófagos Asociados a Tumores/metabolismo , Macrófagos Asociados a Tumores/inmunología , Proliferación Celular , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Fosfatidilinositol 3-Quinasas/metabolismo , Línea Celular Tumoral , Movimiento Celular , Quimiocinas CXC/metabolismo , Animales , Microambiente Tumoral/inmunología
8.
Ecotoxicol Environ Saf ; 279: 116500, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38795416

RESUMEN

Hexavalent chromium [Cr(VI)] is one of the most common environmental contaminants due to its tremendous industrial applications, but its effects and mechanism remain to be investigated. Our previous studies showed that Cr(VI) exposure caused malignant transformation and tumorigenesis. This study showed that glycolytic proteins HK2 and LDHA levels were statistically significant changed in blood samples of Cr(VI)-exposed workers and in Cr-T cells compared to the control subjects and parental cells. HK2 and LDHA knockdown inhibited cell proliferation and angiogenesis, and higher HK2 and LDHA expression levels are associated with advanced stages and poor prognosis of lung cancer. We found that miR-218 levels were significantly decreased and miR-218 directly targeted HK2 and LDHA for inhibiting their expression. Overexpression of miR-218 inhibited glucose consumption and lactate production in Cr-T cells. Further study found that miR-218 inhibited tumor growth and angiogenesis by decreasing HK2 and LDHA expression in vivo. MiR-218 levels were negatively correlated with HK2 and LDHA expression levels and cancer development in human lung and other cancers. These results demonstrated that miR-218/HK2/LDHA pathway is vital for regulating Cr(VI)-induced carcinogenesis and human cancer development.


Asunto(s)
Carcinogénesis , Cromo , Hexoquinasa , Neoplasias Pulmonares , MicroARNs , Regulación hacia Arriba , MicroARNs/genética , Humanos , Cromo/toxicidad , Hexoquinasa/genética , Hexoquinasa/metabolismo , Carcinogénesis/inducido químicamente , Neoplasias Pulmonares/inducido químicamente , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/genética , Pronóstico , Animales , Proliferación Celular/efectos de los fármacos , L-Lactato Deshidrogenasa/metabolismo , Exposición Profesional/efectos adversos , Ratones , Isoenzimas
9.
Am J Chin Med ; 52(3): 865-884, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38790085

RESUMEN

Ovarian cancer is a common, highly lethal tumor. Herein, we reported that S-phase kinase-associated protein 2 (Skp2) is essential for the growth and aerobic glycolysis of ovarian cancer cells. Skp2 was upregulated in ovarian cancer tissues and associated with poor clinical outcomes. Using a customized natural product library screening, we found that xanthohumol inhibited aerobic glycolysis and cell viability of ovarian cancer cells. Xanthohumol facilitated the interaction between E3 ligase Cdh1 and Skp2 and promoted the Ub-K48-linked polyubiquitination of Skp2 and degradation. Cdh1 depletion reversed xanthohumol-induced Skp2 downregulation, enhancing HK2 expression and glycolysis in ovarian cancer cells. Finally, a xenograft tumor model was employed to examine the antitumor efficacy of xanthohumol in vivo. Collectively, we discovered that xanthohumol promotes the binding between Skp2 and Cdh1 to suppress the Skp2/AKT/HK2 signal pathway and exhibits potential antitumor activity for ovarian cancer cells.


Asunto(s)
Flavonoides , Glucólisis , Neoplasias Ováricas , Propiofenonas , Proteínas Quinasas Asociadas a Fase-S , Ubiquitinación , Propiofenonas/farmacología , Proteínas Quinasas Asociadas a Fase-S/metabolismo , Proteínas Quinasas Asociadas a Fase-S/genética , Flavonoides/farmacología , Femenino , Humanos , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/metabolismo , Glucólisis/efectos de los fármacos , Animales , Transducción de Señal/efectos de los fármacos , Cadherinas/metabolismo , Carcinogénesis/efectos de los fármacos , Antígenos CD/metabolismo , Hexoquinasa/metabolismo , Línea Celular Tumoral , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratones , Fitoterapia , Ratones Desnudos , Antineoplásicos Fitogénicos/farmacología
10.
J Transl Med ; 22(1): 460, 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38750462

RESUMEN

BACKGROUND: Chaperonin Containing TCP1 Subunit 6 A (CCT6A) is a prominent protein involved in the folding and stabilization of newly synthesized proteins. However, its roles and underlying mechanisms in lung adenocarcinoma (LUAD), one of the most aggressive cancers, remain elusive. METHODS: Our study utilized in vitro cell phenotype experiments to assess CCT6A's impact on the proliferation and invasion capabilities of LUAD cell lines. To delve into CCT6A's intrinsic mechanisms affecting glycolysis and proliferation in lung adenocarcinoma, we employed transcriptomic sequencing and liquid chromatography-mass spectrometry analysis. Co-immunoprecipitation (Co-IP) and chromatin immunoprecipitation (CHIP) assays were also conducted to substantiate the mechanism. RESULTS: CCT6A was found to be significantly overexpressed in LUAD and associated with a poorer prognosis. The silencing of CCT6A inhibited the proliferation and migration of LUAD cells and elevated apoptosis rates. Mechanistically, CCT6A interacted with STAT1 protein, forming a complex that enhances the stability of STAT1 by protecting it from ubiquitin-mediated degradation. This, in turn, facilitated the transcription of hexokinase 2 (HK2), a critical enzyme in aerobic glycolysis, thereby stimulating LUAD's aerobic glycolysis and progression. CONCLUSION: Our findings reveal that the CCT6A/STAT1/HK2 axis orchestrated a reprogramming of glucose metabolism and thus promoted LUAD progression. These insights position CCT6A as a promising candidate for therapeutic intervention in LUAD treatment.


Asunto(s)
Adenocarcinoma del Pulmón , Proliferación Celular , Chaperonina con TCP-1 , Progresión de la Enfermedad , Glucólisis , Hexoquinasa , Neoplasias Pulmonares , Factor de Transcripción STAT1 , Humanos , Adenocarcinoma del Pulmón/patología , Adenocarcinoma del Pulmón/metabolismo , Adenocarcinoma del Pulmón/genética , Hexoquinasa/metabolismo , Factor de Transcripción STAT1/metabolismo , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/genética , Chaperonina con TCP-1/metabolismo , Línea Celular Tumoral , Movimiento Celular , Regulación Neoplásica de la Expresión Génica , Apoptosis , Transducción de Señal , Invasividad Neoplásica
11.
Sci Rep ; 14(1): 12143, 2024 05 27.
Artículo en Inglés | MEDLINE | ID: mdl-38802677

RESUMEN

Microglia are natural immune cells in the central nervous system, and the activation of microglia is accompanied by a reprogramming of glucose metabolism. In our study, we investigated the role of long non-coding RNA taurine-upregulated gene 1 (TUG1) in regulating microglial glucose metabolism reprogramming and activation. BV2 cells were treated with Lipopolysaccharides (LPS)/Interferon-γ (IFN-γ) to establish a microglial activation model. The glycolysis inhibitor 2-Deoxy-D-glucose (2-DG) was used as a control. The expression levels of TUG1 mRNA and proinflammatory cytokines such as Interleukin-1ß (IL-1ß), Interleukin -6, and Tumor Necrosis Factor-α mRNA and anti-inflammatory cytokines such as IL-4, Arginase 1(Arg1), CD206, and Ym1 were detected by RT-qPCR. TUG1 was silenced using TUG1 siRNA and knocked out using CRISPR/Cas9. The mRNA and protein expression levels of key enzymes involved in glucose metabolism, such as Hexokinase2, Glyceraldehyde-3-phosphate dehydrogenase (GAPDH), Lactate dehydrogenase, Glucose 6 phosphate dehydrogenase, and Pyruvate dehydrogenase (PDH), were determined by RT-qPCR and Western blotting. The glycolytic rate of microglial cells was measured using Seahorse. Differential metabolites were determined by metabolomics, and pathway enrichment was performed using these differential metabolites. Our findings revealed that the expression of TUG1 was elevated in proinflammatory-activated microglia and positively correlated with the levels of inflammatory factors. The expression of anti-inflammatory cytokines such as IL-4, Arg1, CD206, and Ym1 were decreased when induced with LPS/IFN-γ. However, this decrease was reversed by the treatment with 2-DG. Silencing of GAPDH led to an increase in the expression of TUG1 and inflammatory factors. TUG1 knockout (TUG1KO) inhibited the expression of glycolytic key enzymes and promoted the expression of oxidative phosphorylation key enzymes, shifting the metabolic profile of activated microglia from glycolysis to oxidative phosphorylation. Additionally, TUG1KO reduced the accumulation of metabolites, facilitating the restoration of the tricarboxylic acid cycle and enhancing oxidative phosphorylation in microglia. Furthermore, the downregulation of TUG1 was found to reduce the expression of both proinflammatory and anti-inflammatory cytokines under normal conditions. Interestingly, when induced with LPS/IFN-γ, TUG1 downregulation showed a potentially beneficial effect on microglia in terms of inflammation. Downregulation of TUG1 expression inhibits glycolysis and facilitates the shift of microglial glucose metabolism from glycolysis to oxidative phosphorylation, promoting their transformation towards an anti-inflammatory phenotype and exerting anti-inflammatory effects in BV2.


Asunto(s)
Glucosa , Glucólisis , Lipopolisacáridos , Microglía , ARN Largo no Codificante , Microglía/metabolismo , Animales , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Glucosa/metabolismo , Ratones , Lipopolisacáridos/farmacología , Citocinas/metabolismo , Inflamación/metabolismo , Inflamación/genética , Interferón gamma/metabolismo , beta-N-Acetilhexosaminidasas/metabolismo , beta-N-Acetilhexosaminidasas/genética , Línea Celular , Receptor de Manosa , Lectinas de Unión a Manosa/metabolismo , Lectinas de Unión a Manosa/genética , Desoxiglucosa/farmacología , Interleucina-4/metabolismo , Interleucina-1beta/metabolismo , Reprogramación Metabólica , Arginasa , Hexoquinasa , Lectinas
12.
Biochemistry (Mosc) ; 89(2): 299-312, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38622097

RESUMEN

A decrease in muscle mass and its functionality (strength, endurance, and insulin sensitivity) is one of the integral signs of aging. One of the triggers of aging is an increase in the production of mitochondrial reactive oxygen species. Our study was the first to examine age-dependent changes in the production of mitochondrial reactive oxygen species related to a decrease in the proportion of mitochondria-associated hexokinase-2 in human skeletal muscle. For this purpose, a biopsy was taken from m. vastus lateralis in 10 young healthy volunteers and 70 patients (26-85 years old) with long-term primary arthrosis of the knee/hip joint. It turned out that aging (comparing different groups of patients), in contrast to inactivity/chronic inflammation (comparing young healthy people and young patients), causes a pronounced increase in peroxide production by isolated mitochondria. This correlated with the age-dependent distribution of hexokinase-2 between mitochondrial and cytosolic fractions, a decrease in the rate of coupled respiration of isolated mitochondria and respiration when stimulated with glucose (a hexokinase substrate). It is discussed that these changes may be caused by an age-dependent decrease in the content of cardiolipin, a potential regulator of the mitochondrial microcompartment containing hexokinase. The results obtained contribute to a deeper understanding of age-related pathogenetic processes in skeletal muscles and open prospects for the search for pharmacological/physiological approaches to the correction of these pathologies.


Asunto(s)
Hexoquinasa , Mitocondrias , Humanos , Adulto , Persona de Mediana Edad , Anciano , Anciano de 80 o más Años , Especies Reactivas de Oxígeno/metabolismo , Hexoquinasa/metabolismo , Músculo Esquelético/metabolismo , Envejecimiento/fisiología , Mitocondrias Musculares/metabolismo
13.
Cell Cycle ; 23(4): 435-447, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38626328

RESUMEN

The precise mechanisms underlying the inhibitory effects of SIRT3, a mitochondrial sirtuin protein, on hepatocellular carcinoma (HCC) development, as well as its impact on mitochondrial respiration, remain poorly understood. We assessed sirtuins 3 (SIRT3) levels in HCC tissues and Huh7 cells cultured under hypoxic condition. We investigated the effects of SIRT3 on cell proliferation, glycolytic metabolism, mitochondrial respiration, mitophagy, and mitochondrial biogenesis in Huh7 cells. Besides, we explored the potential mechanisms regulating SIRT3 expression in hypoxically cultured Huh7 cells. Gradual reduction in SIRT3 expressions were observed in both adjacent tumor tissues and tumor tissues. Similarly, SIRT3 expressions were diminished in Huh7 cells cultured under hypoxic condition. Forced expression of SIRT3 attenuated the growth of hypoxically cultured Huh7 cells. SIRT3 overexpression led to a decrease in extracellular acidification rate while increasing oxygen consumption rate. SIRT3 downregulated the levels of hexokinase 2 and pyruvate kinase M2. Moreover, SIRT3 enhanced mitophagy signaling, as indicated by mtKeima, and upregulated key proteins involved in various mitophagic pathways while reducing intracellular reactive oxygen species levels. Furthermore, SIRT3 increased proxisome proliferator-activated receptor-gamma coactivator 1α levels and the amount of mitochondrial DNA in Huh7 cells. Notably, ß-catenin expressions were elevated in Huh7 cells cultured under hypoxic condition. Antagonists and agonists of ß-catenin respectively upregulated and downregulated SIRT3 expressions in hypoxically cultured Huh7 cells. The modulationsof glycolysis and mitochondrial respiration represent the primary mechanism through which SIRT3, suppressed by ß-catenin, inhibits HCC cell proliferation.


Asunto(s)
Carcinoma Hepatocelular , Proliferación Celular , Glucólisis , Neoplasias Hepáticas , Mitocondrias , Sirtuina 3 , beta Catenina , Humanos , Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/metabolismo , Sirtuina 3/metabolismo , Sirtuina 3/genética , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/genética , Línea Celular Tumoral , beta Catenina/metabolismo , Mitocondrias/metabolismo , Mitofagia/efectos de los fármacos , Transducción de Señal , Hipoxia de la Célula , Hexoquinasa/metabolismo , Hexoquinasa/genética , Especies Reactivas de Oxígeno/metabolismo , Regulación Neoplásica de la Expresión Génica
14.
Cell Signal ; 119: 111183, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38636768

RESUMEN

Hepatocellular carcinoma (HCC) is one of the leading causes of cancer-related mortality worldwide, with Hepatitis B virus (HBV) infection being the leading cause. This study aims to investigate the role of HBV in HCC pathogenesis involving glucose metabolism. Long non-coding RNA (lncRNA) OIP5-AS1 was significantly downregulated in HBV-positive HCC patients, and its low expression indicated a poor prognosis. This lncRNA was primarily localized in the cytoplasm, acting as a tumor suppressor. HBV protein X (HBx) repressed OIP5-AS1 expression by inhibiting a ligand-activated transcriptional factor peroxisome proliferator-activated receptor α (PPARα). Furthermore, mechanistic studies revealed that OIP5-AS1 inhibited tumor growth by suppressing Hexokinase domain component 1 (HKDC1)-mediated glycolysis. The expression of HKDC1 could be enhanced by transcriptional factor sterol regulatory element-binding protein 1 (SREBP1). OIP5-AS1 facilitated the ubiquitination and degradation of SREBP1 to suppress HKDC1 transcription, which inhibited glycolysis. The results suggest that lncRNA OIP5-AS1 plays an anti-oncogenic role in HBV-positive HCC via the HBx/OIP5-AS1/HKDC1 axis, providing a promising diagnostic marker and therapeutic target for HBV-positive HCC patients.


Asunto(s)
Carcinoma Hepatocelular , Regulación Neoplásica de la Expresión Génica , Glucólisis , Hexoquinasa , Neoplasias Hepáticas , ARN Largo no Codificante , Transactivadores , Proteínas Reguladoras y Accesorias Virales , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Humanos , Carcinoma Hepatocelular/virología , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/virología , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Glucólisis/genética , Transactivadores/metabolismo , Transactivadores/genética , Hexoquinasa/metabolismo , Hexoquinasa/genética , Animales , Virus de la Hepatitis B , Masculino , Línea Celular Tumoral , Regulación hacia Abajo , Ratones , Ratones Desnudos , Femenino , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Ratones Endogámicos BALB C , PPAR alfa/metabolismo , PPAR alfa/genética
15.
Oncol Res ; 32(5): 899-910, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38686047

RESUMEN

Osteosarcoma is a very serious primary bone cancer with a high death rate and a dismal prognosis. Since there is no permanent therapy for this condition, it is necessary to develop a cure. Therefore, this investigation was carried out to assess the impacts and biological functions of hydroxysafflor yellow A (HYSA) in osteosarcoma cell lines (MG63). In this investigational study, MG63 cells were utilized. Microarray experiments, quantitative polymerase chain reaction (qPCR), immunofluorescent staining, extracellular acidification rate (ECAR), oxygen consumption rate (OCR), glucose consumption, lactate production, and ATP levels, proliferation assay, 5-Ethynyl-2'-deoxyuridine (EDU) staining, and Western blot were performed. In MG63 cells, HYSA lowered cell proliferation and metastasis rates, suppressed EDU cell number, and enhanced caspase-3/9 activity levels. HYSA reduced the Warburg effect and induced ferroptosis (FPT) in MG63 cells. Inhibiting ferroptosis diminished HYSA's anti-cancer activities in MG63 cells. The stimulation of the HIF-1α/SLC7A11 pathway decreased HYSA's anti-cancer activities in MG63 cells. HIF-1α is one target spot for HYSA in a model of osteosarcoma cancer (OC). HYSA altered HIF-1α's thermophoretic activity; following binding with HYSA, HIF-1α's melting point increased from ~55°C to ~60°C. HYSA significantly enhanced the thermal stability of exogenous WT HIF-1α while not affecting Mut HIF-1α, suggesting that ARG-311, GLY-312, GLN-347, and GLN-387 may be involved in the interaction between HIF-1α and HYSA. Conclusively, our study revealed that HYSA induced FPT and reduced the Warburg effect of OC through mitochondrial damage by HIF-1α/HK2/SLC7A11 pathway. HYSA is a possible therapeutic option for OC or other cancers.


Asunto(s)
Neoplasias Óseas , Proliferación Celular , Chalcona , Ferroptosis , Osteosarcoma , Quinonas , Humanos , Sistema de Transporte de Aminoácidos y+/efectos de los fármacos , Sistema de Transporte de Aminoácidos y+/metabolismo , Neoplasias Óseas/patología , Neoplasias Óseas/metabolismo , Neoplasias Óseas/tratamiento farmacológico , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Chalcona/farmacología , Chalcona/análogos & derivados , Ferroptosis/efectos de los fármacos , Subunidad alfa del Factor 1 Inducible por Hipoxia/efectos de los fármacos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Osteosarcoma/metabolismo , Osteosarcoma/patología , Osteosarcoma/tratamiento farmacológico , Quinonas/farmacología , Transducción de Señal/efectos de los fármacos , Hexoquinasa/efectos de los fármacos , Hexoquinasa/metabolismo
16.
Pathol Res Pract ; 257: 155281, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38669868

RESUMEN

BACKGROUND: Colorectal cancer (CRC) theratened thousands of people every year. Emerging evidences suggested that circular RNAs (circRNAs) were involved in CRC malignancies. However, the underlying mechanisms have yet not been revealed. METHODS: Quantitative real-time PCR (qRT-PCR) was used to determine the expression of circ_0087862 and microRNA-512-3p (miR-512-3p). Western blot was performed to measure the protein expression of hexokinase 2 (HK2), B-cell lymphoma-2 (Bcl-2), BCL2-associated X (Bax) and BCL2 antagonist/killer 1 (Bak). Moreover, 3-(4, 5-dimethyl-2-thiazolyl)-2, 5-diphenyl-2-H-tetrazolium bromide (MTT) assay, colony formation and 5-ethynyl-2'-deoxyuridine (EdU) assay were employed to assess CRC cell proliferation. Also, migration/invasion abilities and apoptosis rates were investigated by transwell assay and flow cytometry. Glucose consumption, lactate production and ATP production were detected using the corresponding kits. Dual-luciferase reporter analysis and RNA immunoprecipitation (RIP) experiments were utilized to analyze the target association of miR-512-3p and circ_0087862 or HK2. Finally, xenograft assay was carried out to analyze the function of circ_0087862 in tumor growth in vivo. RESULTS: Circ_0087862 expression was elevated in CRC tissues and cells. Circ_0087862 silencing repressed cell viabilities, proliferation, migration/invasion and glycolysis, and reinforced cell apoptosis. However, HK2 could weaken these impacts. Additionally, miR-512-3p targeted HK2, and circ_0087862 could regulate HK2 expression by miR-512-3p. Furthermore, circ_0087862 silencing decreased CRC cell xenograft tumor growth. CONCLUSION: Collectively, our data suggested that circ_0087862 knockdown impeded cell viabilities, proliferation, and glycolysis, and contributed to cell apoptosis in CRC, indicating circ_0087862 as a promising tumor promoter.


Asunto(s)
Apoptosis , Proliferación Celular , Neoplasias Colorrectales , Regulación Neoplásica de la Expresión Génica , Hexoquinasa , MicroARNs , ARN Circular , Animales , Femenino , Humanos , Masculino , Ratones , Apoptosis/genética , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/metabolismo , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica/genética , Hexoquinasa/genética , Hexoquinasa/metabolismo , Ratones Desnudos , MicroARNs/genética , MicroARNs/metabolismo , ARN Circular/genética , ARN Circular/metabolismo
17.
BMC Med Genomics ; 17(1): 112, 2024 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-38685060

RESUMEN

BACKGROUND: The Warburg effect is a hallmark characteristic of colorectal cancer (CRC). Despite extensive research, the role of long non-coding RNAs (lncRNAs) in influencing the Warburg effect remains incompletely understood. Our study aims to identify lncRNAs that may modulate the Warburg effect by functioning as competing endogenous RNAs (ceRNAs). METHODS: Utilizing bioinformatics approaches, we extracted glycolysis-associated gene data from the Kyoto Encyclopedia of Genes and Genomes (KEGG) and identified 101 glycolysis-related lncRNAs in CRC. We employed Univariable Cox regression, Least Absolute Shrinkage and Selection Operator (LASSO) regression analysis, and Multivariable Cox regression to develop a prognostic model comprising four glycolysis-linked lncRNAs. We then constructed a prognostic nomogram integrating this lncRNA model with other relevant clinical parameters. RESULTS: The prognostic efficacy of our four-lncRNA signature and its associated nomogram was validated in both training and validation cohorts. Functional assays demonstrated significant glycolysis and hexokinase II (HK2) inhibition following the silencing of RUNDC3A - AS1, a key lncRNA in our prognostic signature, highlighting its regulatory importance in the Warburg effect. CONCLUSIONS: Our research illuminates the critical role of glycolysis-centric lncRNAs in CRC. The developed prognostic model and nomogram underscore the pivotal prognostic and regulatory significance of the lncRNA RUNDC3A - AS1 in the Warburg effect in colorectal cancer.


Asunto(s)
Neoplasias Colorrectales , Progresión de la Enfermedad , Glucólisis , ARN Largo no Codificante , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Humanos , Glucólisis/genética , Pronóstico , Hexoquinasa/genética , Hexoquinasa/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Masculino , Nomogramas , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Perfilación de la Expresión Génica
18.
Acta Biomater ; 181: 308-316, 2024 06.
Artículo en Inglés | MEDLINE | ID: mdl-38570107

RESUMEN

Acute lung injury (ALI) represents a critical respiratory condition typified by rapid-onset lung inflammation, contributing to elevated morbidity and mortality rates. Central to ALI pathogenesis lies macrophage dysfunction, characterized by an overabundance of pro-inflammatory cytokines and a shift in metabolic activity towards glycolysis. This study emphasizes the crucial function of glucose metabolism in immune cell function under inflammatory conditions and identifies hexokinase 2 (HK2) as a key regulator of macrophage metabolism and inflammation. Given the limitations of HK2 inhibitors, we propose the CRISPR/Cas9 system for precise HK2 downregulation. We developed an aerosolized core-shell liposomal nanoplatform (CSNs) complexed with CaP for efficient drug loading, targeting lung macrophages. Various CSNs were synthesized to encapsulate an mRNA based CRISPR/Cas9 system (mCas9/gHK2), and their gene editing efficiency and HK2 knockout were examined at both gene and protein levels in vitro and in vivo. The CSN-mCas9/gHK2 treatment demonstrated a significant reduction in glycolysis and inflammation in macrophages. In an LPS-induced ALI mouse model, inhaled CSN-mCas9/gHK2 mitigated the proinflammatory tumor microenvironment and reprogrammed glucose metabolism in the lung, suggesting a promising strategy for ALI prevention and treatment. This study highlights the potential of combining CRISPR/Cas9 gene editing with inhalation delivery systems for effective, localized pulmonary disease treatment, underscoring the importance of targeted gene modulation and metabolic reprogramming in managing ALI. STATEMENT OF SIGNIFICANCE: This study investigates an inhalable CRISPR/Cas9 gene editing system targeting pulmonary macrophages, with the aim of modulating glucose metabolism to alleviate Acute Lung Injury (ALI). The research highlights the role of immune cell metabolism in inflammation, as evidenced by changes in macrophage glucose metabolism and a notable reduction in pulmonary edema and inflammation. Additionally, observed alterations in macrophage polarization and cytokine levels in bronchoalveolar lavage fluid suggest potential therapeutic implications. These findings not only offer insights into possible ALI treatments but also contribute to the understanding of immune cell metabolism in inflammatory diseases, which could be relevant for various inflammatory and metabolic disorders.


Asunto(s)
Lesión Pulmonar Aguda , Sistemas CRISPR-Cas , Hexoquinasa , Lesión Pulmonar Aguda/patología , Lesión Pulmonar Aguda/terapia , Animales , Ratones , Hexoquinasa/genética , Hexoquinasa/metabolismo , Ratones Endogámicos C57BL , Macrófagos/metabolismo , Macrófagos/efectos de los fármacos , Administración por Inhalación , Liposomas/química , Células RAW 264.7 , Masculino , Reprogramación Celular/efectos de los fármacos , Edición Génica , Glucólisis/efectos de los fármacos
19.
Front Biosci (Landmark Ed) ; 29(4): 160, 2024 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-38682208

RESUMEN

BACKGROUND: Nasopharyngeal carcinoma (NPC) is a malignant tumor associated with Epstein-Barr virus (EBV) infection. Chemoradiotherapy is the mainstream treatment for locally advanced NPC, and chemotherapeutic drugs are an indispensable part of NPC treatment. However, the toxic side-effects of chemotherapy drugs limit their therapeutic value, and new chemotherapy drugs are urgently needed for NPC. Silvestrol, an emerging natural plant anticancer molecule, has shown promising antitumor activity in breast cancer, melanoma, liver cancer, and other tumor types by promoting apoptosis in cancer cells to a greater extent than in normal cells. However, the effects of silvestrol on NPC and its possible molecular mechanisms have yet to be fully explored. METHODS: Cell counting kit-8 (CCK-8), cell scratch, flow cytometry, 5-ethynyl-2'-deoxyuridine (EdU), and Western blot (WB) assays were used to evaluate the effects of silvestrol on the cell viability, cell cycle, apoptosis, and migration of NPC cells. RNA sequencing (RNA-Seq) was used to study the effect of extracellular signal-regulated kinase (ERK) inhibitors on the cell transcriptome, and immunohistochemistry (IHC) to assess protein expression levels in patient specimens. RESULTS: Silvestrol inhibited cell migration and DNA replication of NPC cells, while promoting the expression of cleaved caspase-3, apoptosis, and cell cycle arrest. Furthermore, silvestrol altered the level of ERK phosphorylation. The ERK-targeted inhibitor LY3214996 attenuated silvestrol-mediated inhibition of NPC cell proliferation but not migration. Analysis of RNA-Seq data and WB were used to identify and validate the downstream regulatory targets of silvestrol. Expression of GADD45A, RAP1A, and hexokinase-II (HK2) proteins was inhibited by silvestrol and LY3214996. Finally, IHC revealed that GADD45A, RAP1A, and HK2 protein expression was more abundant in cancer tissues than in non-tumor tissues. CONCLUSIONS: Silvestrol inhibits the proliferation of NPC cells by targeting ERK phosphorylation. However, the inhibition of NPC cell migration by silvestrol was independent of the Raf-MEK-ERK pathway. RAP1A, HK2, and GADD45A may be potential targets for the action of silvestrol.


Asunto(s)
Benzofuranos , Proteinas GADD45 , Hexoquinasa , Sistema de Señalización de MAP Quinasas , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas , Proteínas de Unión al GTP rap1 , Humanos , Apoptosis/efectos de los fármacos , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/genética , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Carcinoma Nasofaríngeo/tratamiento farmacológico , Carcinoma Nasofaríngeo/genética , Carcinoma Nasofaríngeo/patología , Neoplasias Nasofaríngeas/tratamiento farmacológico , Neoplasias Nasofaríngeas/genética , Neoplasias Nasofaríngeas/patología , Hexoquinasa/genética , Hexoquinasa/metabolismo , Proteínas de Unión al GTP rap1/genética , Proteínas de Unión al GTP rap1/metabolismo , Proteinas GADD45/genética , Proteinas GADD45/metabolismo
20.
Exp Mol Med ; 56(3): 747-759, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38531963

RESUMEN

Intervertebral disc degeneration (IDD) is an important pathological basis for degenerative spinal diseases and is involved in mitophagy dysfunction. However, the molecular mechanisms underlying mitophagy regulation in IDD remain unclear. This study aimed to clarify the role of DJ-1 in regulating mitophagy during IDD pathogenesis. Here, we showed that the mitochondrial localization of DJ-1 in nucleus pulposus cells (NPCs) first increased and then decreased in response to oxidative stress. Subsequently, loss- and gain-of-function experiments revealed that overexpression of DJ-1 in NPCs inhibited oxidative stress-induced mitochondrial dysfunction and mitochondria-dependent apoptosis, whereas knockdown of DJ-1 had the opposite effect. Mechanistically, mitochondrial translocation of DJ-1 promoted the recruitment of hexokinase 2 (HK2) to damaged mitochondria by activating Akt and subsequently Parkin-dependent mitophagy to inhibit oxidative stress-induced apoptosis in NPCs. However, silencing Parkin, reducing mitochondrial recruitment of HK2, or inhibiting Akt activation suppressed DJ-1-mediated mitophagy. Furthermore, overexpression of DJ-1 ameliorated IDD in rats through HK2-mediated mitophagy. Taken together, these findings indicate that DJ-1 promotes HK2-mediated mitophagy under oxidative stress conditions to inhibit mitochondria-dependent apoptosis in NPCs and could be a therapeutic target for IDD.


Asunto(s)
Degeneración del Disco Intervertebral , Mitofagia , Proteína Desglicasa DJ-1 , Animales , Ratas , Apoptosis , Hexoquinasa/genética , Hexoquinasa/farmacología , Hexoquinasa/uso terapéutico , Degeneración del Disco Intervertebral/genética , Degeneración del Disco Intervertebral/metabolismo , Mitofagia/genética , Mitofagia/fisiología , Proteínas Proto-Oncogénicas c-akt , Ubiquitina-Proteína Ligasas/genética , Proteína Desglicasa DJ-1/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA