Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
J Clin Invest ; 133(20)2023 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-37843279

RESUMEN

Regulatory T cells (Tregs) are instrumental in maintaining immune tolerance and preventing destructive autoimmunity, but how heterogeneous Treg populations are established remains largely unknown. Here, we show that Zfp335 deletion in Tregs failed to differentiate into effector Tregs (eTregs) and lose Treg-suppressive function and that KO mice exhibited early-onset lethal autoimmune inflammation with unrestricted activation of conventional T cells. Single-cell RNA-Seq analyses revealed that Zfp335-deficient Tregs lacked a eTreg population and showed dramatic accumulation of a dysfunctional Treg subset. Mechanistically, Zfp335-deficient Tregs displayed reduced oxidative phosphorylation and dysfunctional mitochondrial activity. Further studies revealed that Zfp335 controlled eTreg differentiation by regulating fatty acid oxidation (FAO) through direct targeting of the FAO enzyme Hadha. Importantly, we demonstrate a positive correlation between ZNF335 and HADHA expression in human eTregs. Our findings reveal that Zfp335 controls FAO-driven eTreg differentiation to establish immune tolerance.


Asunto(s)
Tolerancia Inmunológica , Linfocitos T Reguladores , Animales , Humanos , Ratones , Autoinmunidad , Ácidos Grasos/genética , Ácidos Grasos/metabolismo , Subunidad alfa de la Proteína Trifuncional Mitocondrial/metabolismo
2.
Mol Biol Rep ; 50(2): 961-970, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36376538

RESUMEN

BACKGROUND: Nonalcoholic fatty liver disease (NAFLD) is a liver metabolic syndrome and still lacks effective treatments because the molecular mechanism underlying the development of NAFLD is not completely understood. We investigated the role of Hydroxyl CoA dehydrogenase alpha subunit (HADHA) in the pathogenesis of NAFLD. METHODS: HADHA expression was detected both in NAFLD cell and mice, and knockdown of HADHA in free fatty acids (FFA)-treated L02 or overexpression of HADHA in high fat diet (HFD)-fed mice was used to detected the influence of HADHA on hepatic steatosis, mitochondrial dysfunction, and oxidative stress by regulating of MKK3/MAPK signaling. RESULTS: Our data revealed that HADHA expression was decreased in FFA-treated L02 cells and in HFD-fed mice. Knockdown of HADHA markedly aggravated hepatic steatosis, inflammation and oxidative stress in FFA-treated L02 cells, which was associated with the activation of MKK3/MAPK signalling pathways. Moreover, oxidative stress and liver lesions were improved in NAFLD mice by upregulation of HADHA. Importantly, we demonstrated that overexpression of HADHA inhibited the expression of p-MAPK in NAFLD mice, reducing lipid accumulation and steatosis. CONCLUSION: HADHA may function as a protective factor in the progression of NAFLD by alleviating abnormal metabolism and oxidative stress by suppressing MKK3/MAPK signalling pathway activation, providing a new target for the treatment of NAFLD.


Asunto(s)
Subunidad alfa de la Proteína Trifuncional Mitocondrial , Enfermedad del Hígado Graso no Alcohólico , Animales , Ratones , Dieta Alta en Grasa/efectos adversos , Ácidos Grasos no Esterificados/metabolismo , Inflamación/metabolismo , Metabolismo de los Lípidos , Hígado/metabolismo , Ratones Endogámicos C57BL , Subunidad alfa de la Proteína Trifuncional Mitocondrial/metabolismo , Enfermedad del Hígado Graso no Alcohólico/patología , Estrés Oxidativo
3.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 53(5): 805-814, 2022 Sep.
Artículo en Chino | MEDLINE | ID: mdl-36224682

RESUMEN

Objective: To explore the effects of hydroxyacyl-CoA dehydrogenase alpha subunit (HADHA) on the migration and invasion of HTR-8/SVneo cells, a human trophoblast cell line, and its potential mechanism of action. Methods: Immunofluorescence staining was done to evaluate the expression levels of HADHA in samples of normal villi and recurrent spontaneous abortion (RSA) villi at 6-8 weeks. Lentiviral infection system was used to construct stable HTR-8/SVneo cell lines with HADHA overexpression and knockdown. Western blot, qRT-PCR, Wound-healing assay, and Transwell assay were used to determine the effect of HADHA on the migration and invasion of HTR-8/SVneo cells and the expression of relevant genes. Transcriptome sequencing and bioinformatics analysis were done to screen for the potential target genes and signaling pathways regulated by HADHA. The specific molecular mechanism of how HADHA regulates the migration and invasion of HTR-8/SVneo cells was examined by adding the inhibitor of protein kinase B (PKB/AKT). Results: HADHA was highly expressed in extravillous trophoblasts (EVT) of RSA villus samples as compared with samples from the normal control group. In HTR-8/SVneo cells overexpressing HADHA, the expression levels of migration and invasion-related genes, including HLA-G, MMP2, MMP9, and NCAD, were decreased (P<0.01,P<0.05), and the migration and invasion abilities of HTR-8/SVneo cells were weakened (P<0.05). HADHA knockdown increased the expression levels of HLA-G, MMP2, MMP9, and NCAD (P<0.01, P<0.05), and promoted the migration and invasion of HTR-8/SVneo cells (P<0.05). In addition, HADHA overexpression decreased the phosphorylation levels of PI3K and AKT (P<0.05) and inhibited the PI3K/AKT signaling pathway. HADHA knockdown activated the PI3K/AKT signaling pathway. When MK-2206, an AKT inhibitor, was added to stable HTR-8/SVneo cell lines with HADHA knockdown, the migration and invasion of the cells were significantly reduced. Conclusion: HADHA inhibits the migration and invasion of HTR-8/SVneo cells by inhibiting the PI3K/AKT signaling pathway.


Asunto(s)
Preeclampsia , Proteínas Proto-Oncogénicas c-akt , Movimiento Celular/fisiología , Coenzima A/metabolismo , Coenzima A/farmacología , Femenino , Antígenos HLA-G/metabolismo , Antígenos HLA-G/farmacología , Humanos , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Subunidad alfa de la Proteína Trifuncional Mitocondrial/metabolismo , Oxidorreductasas/metabolismo , Oxidorreductasas/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Embarazo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Trofoblastos/metabolismo
4.
Oncogene ; 41(48): 5199-5213, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36273042

RESUMEN

Cancer cells rely on heightened protein quality control mechanisms, including the ubiquitin-proteosome system that is predominantly driven by ubiquitination comprising E1, E2, and E3 trienzyme cascades. Although E3s have been extensively studied, the implication of E2s in tumorigenesis is poorly defined. Here we reveal a critical E2 in the pathogenesis of hepatocellular carcinoma (HCC). Among all of E2s, UBE2O shows the strongest association with HCC survival prognosis, and its expression is increased in HCC tumors. Accordingly, UBE2O deficiency inhibits HCC growth and metastasis both in vitro and in vivo, while its overexpression has opposite effects. Depending on both E2 and E3 enzymatic activities, UBE2O can interact with and mediate the ubiquitination and degradation of HADHA, a mitochondrial ß-oxidation enzyme, thereby modulating lipid metabolic reprogramming. HADHA is reduced in HCC tumors and inversely correlated with UBE2O levels. Importantly, HADHA acts as a tumor suppressor and primarily mediates UBE2O's function on HCC. Moreover, liver-specific deletion of Ube2o in mice are resistant to DEN-induced hepatocarcinogenesis, along with HADHA upregulation and reduced hepatic lipid accumulation. These data reveal UBE2O as a novel oncogenic driver for metabolic reprogramming and HCC development, highlighting the potential of targeting UBE2O/HADHA axis for HCC therapy.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Subunidad alfa de la Proteína Trifuncional Mitocondrial , Enzimas Ubiquitina-Conjugadoras , Animales , Ratones , Carcinoma Hepatocelular/genética , Lípidos , Neoplasias Hepáticas/genética , Subunidad alfa de la Proteína Trifuncional Mitocondrial/metabolismo , Ubiquitinación , Enzimas Ubiquitina-Conjugadoras/metabolismo
5.
Science ; 378(6618): eabj3510, 2022 10 28.
Artículo en Inglés | MEDLINE | ID: mdl-36302005

RESUMEN

Spermidine (SPD) delays age-related pathologies in various organisms. SPD supplementation overcame the impaired immunotherapy against tumors in aged mice by increasing mitochondrial function and activating CD8+ T cells. Treatment of naïve CD8+ T cells with SPD acutely enhanced fatty acid oxidation. SPD conjugated to beads bound to the mitochondrial trifunctional protein (MTP). In the MTP complex, synthesized and purified from Escherichia coli, SPD bound to the α and ß subunits of MTP with strong affinity and allosterically enhanced their enzymatic activities. T cell-specific deletion of the MTP α subunit abolished enhancement of programmed cell death protein 1 (PD-1) blockade immunotherapy by SPD, indicating that MTP is required for SPD-dependent T cell activation.


Asunto(s)
Linfocitos T CD8-positivos , Mitocondrias , Subunidad alfa de la Proteína Trifuncional Mitocondrial , Subunidad beta de la Proteína Trifuncional Mitocondrial , Neoplasias , Espermidina , Animales , Ratones , Linfocitos T CD8-positivos/inmunología , Activación de Linfocitos , Mitocondrias/metabolismo , Subunidad alfa de la Proteína Trifuncional Mitocondrial/metabolismo , Subunidad beta de la Proteína Trifuncional Mitocondrial/metabolismo , Espermidina/farmacología , Espermidina/metabolismo , Neoplasias/inmunología
6.
Nat Commun ; 13(1): 386, 2022 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-35046401

RESUMEN

Disordered hepatic glucagon response contributes to hyperglycemia in diabetes. The regulators involved in glucagon response are less understood. This work aims to investigate the roles of mitochondrial ß-oxidation enzyme HADHA and its downstream ketone bodies in hepatic glucagon response. Here we show that glucagon challenge impairs expression of HADHA. Liver-specific HADHA overexpression reversed hepatic gluconeogenesis in mice, while HADHA knockdown augmented glucagon response. Stable isotope tracing shows that HADHA promotes ketone body production via ß-oxidation. The ketone body ß-hydroxybutyrate (BHB) but not acetoacetate suppresses gluconeogenesis by selectively inhibiting HDAC7 activity via interaction with Glu543 site to facilitate FOXO1 nuclear exclusion. In HFD-fed mice, HADHA overexpression improved metabolic disorders, and these effects are abrogated by knockdown of BHB-producing enzyme. In conclusion, BHB is responsible for the inhibitory effect of HADHA on hepatic glucagon response, suggesting that HADHA activation or BHB elevation by pharmacological intervention hold promise in treating diabetes.


Asunto(s)
Ácido 3-Hidroxibutírico/biosíntesis , Glucagón/metabolismo , Hígado/metabolismo , Mitocondrias/metabolismo , Subunidad alfa de la Proteína Trifuncional Mitocondrial/metabolismo , Ácido 3-Hidroxibutírico/metabolismo , Acetilación , Animales , Glucemia/metabolismo , Dieta Alta en Grasa , Proteína Forkhead Box O1/metabolismo , Gluconeogénesis , Células HEK293 , Histona Desacetilasas/metabolismo , Humanos , Hidroxibutirato Deshidrogenasa , Marcaje Isotópico , Cuerpos Cetónicos/metabolismo , Luciferasas/metabolismo , Masculino , Ratones Endogámicos C57BL , Modelos Biológicos , Oxidación-Reducción , Unión Proteica
7.
J Cancer Res Ther ; 17(5): 1281-1285, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34850779

RESUMEN

OBJECTIVES: Wilms tumor is a common pediatric malignant tumor that accounts for approximately 95% of kidney tumors in children. The role of lipid metabolism in tumors has attracted increased attention in recent years. We examined the role of hydroxyacyl-CoA dehydrogenase trifunctional multienzyme complex subunit alpha (HADHA), a lipid metabolism enzyme, in the pathogenesis of Wilms tumor. MATERIALS AND METHODS: In a previous study, we screened Wilms tumors and adjacent normal tissues for differentially expressed proteins by mass spectrometry and verified the results by western blot analysis. The Oncomine database and quantitative reverse transcription-polymerase chain reaction were used to verify the expression of HADHA at the genetic level. Immunohistochemistry and immunofluorescence were also used to validate the differential expression of the HADHA protein. The relationship between histopathological typing, clinical pathology, and HADHA expression was analyzed in 65 paraffin-embedded specimens from pediatric Wilms tumor patients. Kaplan-Meier survival curves were used to analyze the relationship between the expression of HADHA and patient prognosis. RESULTS: HADHA was expressed at low levels in Wilms tumor tissue compared with the corresponding normal tissue. The expression of HADHA was closely associated with histopathological typing (P = 0.030). The prognostic analysis of 65 children with Wilms tumor showed that high expression of HADHA was closely associated with poor prognosis (P = 0.046). CONCLUSIONS: HADHA expression is downregulated in Wilms tumor tissues, but high expression in tumor tissues is associated with clinical stage and the prognosis of children with this tumor.


Asunto(s)
Neoplasias Renales/patología , Metabolismo de los Lípidos , Subunidad alfa de la Proteína Trifuncional Mitocondrial/metabolismo , Tumor de Wilms/patología , Biomarcadores de Tumor , Niño , Preescolar , Femenino , Estudios de Seguimiento , Humanos , Lactante , Neoplasias Renales/enzimología , Neoplasias Renales/genética , Masculino , Subunidad alfa de la Proteína Trifuncional Mitocondrial/genética , Pronóstico , Tasa de Supervivencia , Tumor de Wilms/enzimología , Tumor de Wilms/genética
8.
PLoS One ; 16(10): e0256619, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34644302

RESUMEN

Mitochondrial trifunctional protein (TFP) is a membrane-associated heterotetramer that catalyzes three of the four reactions needed to chain-shorten long-chain fatty acids inside the mitochondria. TFP is known to be heavily modified by acetyllysine and succinyllysine post-translational modifications (PTMs), many of which are targeted for reversal by the mitochondrial sirtuin deacylases SIRT3 and SIRT5. However, the functional significance of these PTMs is not clear, with some reports showing TFP gain-of-function and some showing loss-of-function upon increased acylation. Here, we mapped the known SIRT3/SIRT5-targeted lysine residues onto the recently solved TFP crystal structure which revealed that many of the target sites are involved in substrate channeling within the TFPα subunit. To test the effects of acylation on substate channeling through TFPα, we enzymatically synthesized the physiological long-chain substrate (2E)-hexadecenoyl-CoA. Assaying TFP in SIRT3 and SIRT5 knockout mouse liver and heart mitochondria with (2E)-hexadecenoyl-CoA revealed no change in enzyme activity. Finally, we investigated the effects of lysine acylation on TFP membrane binding in vitro. Acylation did not alter recombinant TFP binding to cardiolipin-containing liposomes. However, the presence of liposomes strongly abrogated the acylation reaction between succinyl-CoA and TFP lysine residues. Thus, TFP in the membrane-bound state may be protected against lysine acylation.


Asunto(s)
Ácidos Grasos/química , Mitocondrias/metabolismo , Subunidad alfa de la Proteína Trifuncional Mitocondrial/metabolismo , Sirtuina 3/metabolismo , Sirtuinas/metabolismo , Acetilación , Animales , Metabolismo Energético/fisiología , Liposomas/metabolismo , Hígado/metabolismo , Lisina/química , Ratones , Ratones Noqueados , Mitocondrias/enzimología , Subunidad alfa de la Proteína Trifuncional Mitocondrial/genética , Miocardio/metabolismo , Procesamiento Proteico-Postraduccional , Sirtuina 3/genética , Sirtuinas/genética , Ácido Succínico/química
9.
Sci Rep ; 11(1): 13297, 2021 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-34168259

RESUMEN

Pancreatic cancer stem cells (PCSCs) play a key role in the aggressiveness of pancreatic ductal adenocarcinomas (PDAC); however, little is known about their signaling and metabolic pathways. Here we show that PCSCs have specific and common proteome and lipidome modulations. PCSCs displayed downregulation of lactate dehydrogenase A chain, and upregulation of trifunctional enzyme subunit alpha. The upregulated proteins of PCSCs are mainly involved in fatty acid (FA) elongation and biosynthesis of unsaturated FAs. Accordingly, lipidomics reveals an increase in long and very long-chain unsaturated FAs, which are products of fatty acid elongase-5 predicted as a key gene. Moreover, lipidomics showed the induction in PCSCs of molecular species of cardiolipin with mixed incorporation of 16:0, 18:1, and 18:2 acyl chains. Our data indicate a crucial role of FA elongation and alteration in cardiolipin acyl chain composition in PCSCs, representing attractive therapeutic targets in PDAC.


Asunto(s)
Carcinoma Ductal Pancreático/metabolismo , Cardiolipinas/metabolismo , Subunidad alfa de la Proteína Trifuncional Mitocondrial/metabolismo , Células Madre Neoplásicas/metabolismo , Neoplasias Pancreáticas/metabolismo , Humanos , Metabolismo de los Lípidos , Lipidómica , Proteómica , Regulación hacia Arriba
10.
Mol Cell ; 80(1): 43-58.e7, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32937100

RESUMEN

Immune cell function depends on specific metabolic programs dictated by mitochondria, including nutrient oxidation, macromolecule synthesis, and post-translational modifications. Mitochondrial adaptations have been linked to acute and chronic inflammation, but the metabolic cues and precise mechanisms remain unclear. Here we reveal that histone deacetylase 3 (HDAC3) is essential for shaping mitochondrial adaptations for IL-1ß production in macrophages through non-histone deacetylation. In vivo, HDAC3 promoted lipopolysaccharide-induced acute inflammation and high-fat diet-induced chronic inflammation by enhancing NLRP3-dependent caspase-1 activation. HDAC3 configured the lipid profile in stimulated macrophages and restricted fatty acid oxidation (FAO) supported by exogenous fatty acids for mitochondria to acquire their adaptations and depolarization. Rather than affecting nuclear gene expression, HDAC3 translocated to mitochondria to deacetylate and inactivate an FAO enzyme, mitochondrial trifunctional enzyme subunit α. HDAC3 may serve as a controlling node that balances between acquiring mitochondrial adaptations and sustaining their fitness for IL-1ß-dependent inflammation.


Asunto(s)
Ácidos Grasos/metabolismo , Histona Desacetilasas/metabolismo , Inflamación/metabolismo , Interleucina-1beta/metabolismo , Mitocondrias/metabolismo , Adulto , Animales , Caspasa 1/metabolismo , Femenino , Humanos , Inflamación/patología , Metabolismo de los Lípidos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Mitocondrias/ultraestructura , Subunidad alfa de la Proteína Trifuncional Mitocondrial/metabolismo , Células Mieloides/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Oxidación-Reducción , Fosforilación Oxidativa , Adulto Joven
11.
Int J Mol Sci ; 21(16)2020 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-32824240

RESUMEN

Increased platelet activation and apoptosis are characteristic of diabetic (DM) platelets, where a Parkin-dependent mitophagy serves a major endogenous protective role. We now demonstrate that Parkin is highly expressed in both healthy platelets and diabetic platelets, compared to other mitochondria-enriched tissues such as the heart, muscle, brain, and liver. Abundance of Parkin in a small, short-lived anucleate cell suggest significance in various key processes. Through proteomics we identified 127 Parkin-interacting proteins in DM platelets and compared them to healthy controls. We assessed the 11 highest covered proteins by individual IPs and confirmed seven proteins that interacted with Parkin; VCP/p97, LAMP1, HADHA, FREMT3, PDIA, ILK, and 14-3-3. Upon further STRING analysis using GO and KEGG, interactions were divided into two broad groups: targeting platelet activation through (1) actions on mitochondria and (2) actions on integrin signaling. Parkin plays an important role in mitochondrial protection through mitophagy (VCP/p97), recruiting phagophores, and targeting lysosomes (with LAMP1). Mitochondrial ß-oxidation may also be regulated by the Parkin/HADHA interaction. Parkin may regulate platelet aggregation and activation through integrin signaling through interactions with proteins like FREMT3, PDIA, ILK, and 14-3-3. Thus, platelet Parkin may regulate the protection (mitophagy) and stress response (platelet activation) in DM platelets. This study identified new potential therapeutic targets for platelet mitochondrial dysfunction and hyperactivation in diabetes mellitus.


Asunto(s)
Plaquetas/metabolismo , Diabetes Mellitus/metabolismo , Estrés Fisiológico , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas 14-3-3/metabolismo , Animales , Células Cultivadas , Humanos , Proteínas de Membrana de los Lisosomas/metabolismo , Ratones , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Subunidad alfa de la Proteína Trifuncional Mitocondrial/metabolismo , Mitofagia , Activación Plaquetaria , Unión Proteica , Proteínas Serina-Treonina Quinasas/metabolismo , Ubiquitina-Proteína Ligasas/genética , Proteína que Contiene Valosina/metabolismo
12.
J Clin Invest ; 130(3): 1392-1404, 2020 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-31985487

RESUMEN

Mechanisms mediating the cardioprotective actions of glucagon-like peptide 1 (GLP-1) were unknown. Here, we show in both ex vivo and in vivo models of ischemic injury that treatment with GLP-1(28-36), a neutral endopeptidase-generated (NEP-generated) metabolite of GLP-1, was as cardioprotective as GLP-1 and was abolished by scrambling its amino acid sequence. GLP-1(28-36) enters human coronary artery endothelial cells (caECs) through macropinocytosis and acts directly on mouse and human coronary artery smooth muscle cells (caSMCs) and caECs, resulting in soluble adenylyl cyclase Adcy10-dependent (sAC-dependent) increases in cAMP, activation of protein kinase A, and cytoprotection from oxidative injury. GLP-1(28-36) modulates sAC by increasing intracellular ATP levels, with accompanying cAMP accumulation lost in sAC-/- cells. We identify mitochondrial trifunctional protein-α (MTPα) as a binding partner of GLP-1(28-36) and demonstrate that the ability of GLP-1(28-36) to shift substrate utilization from oxygen-consuming fatty acid metabolism toward oxygen-sparing glycolysis and glucose oxidation and to increase cAMP levels is dependent on MTPα. NEP inhibition with sacubitril blunted the ability of GLP-1 to increase cAMP levels in coronary vascular cells in vitro. GLP-1(28-36) is a small peptide that targets novel molecular (MTPα and sAC) and cellular (caSMC and caEC) mechanisms in myocardial ischemic injury.


Asunto(s)
Cardiotónicos/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Mitocondrias Cardíacas/metabolismo , Subunidad alfa de la Proteína Trifuncional Mitocondrial/metabolismo , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/prevención & control , Animales , Vasos Coronarios/metabolismo , Vasos Coronarios/patología , AMP Cíclico/genética , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Células Endoteliales/metabolismo , Células Endoteliales/patología , Péptido 1 Similar al Glucagón/genética , Humanos , Ratones , Ratones Noqueados , Mitocondrias Cardíacas/genética , Mitocondrias Cardíacas/patología , Subunidad alfa de la Proteína Trifuncional Mitocondrial/genética , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Isquemia Miocárdica/genética , Isquemia Miocárdica/patología , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Sistemas de Mensajero Secundario/genética
13.
Lab Invest ; 100(3): 353-362, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31527828

RESUMEN

Cancer cells, including malignant lymphoma cells, alter their metabolism, termed "metabolic reprograming," on initiation of malignant transformation as well as upon accumulation of genetic abnormalities. Here, to identify a novel therapeutic target involved in the metabolic changes during malignant lymphoma, we performed global analyses combined with shotgun proteomics, in silico database analysis, and clinic-pathologic analysis of nonneoplastic lymphoid tissue and malignant lymphoma tissue and verified the molecular functions in vitro. In total, 2002 proteins were detected from both samples and proteins related to fatty acid beta-oxidation (FAO) were detected more frequently in malignant lymphoma tissue. Consequently, the most frequently detected protein, the mitochondrial trifunctional enzyme subunit-alpha (HADHA), was identified as a potential target. Immunohistochemical analyses revealed that HADHA tended to be overexpressed in a high-grade subtype of malignant lymphoma tissue. Clinicopathologic study revealed that HADHA overexpression was correlated with significantly worse overall survival (P = 0.013) and was an independent prognostic predictor in diffuse large B-cell lymphoma (P = 0.027). In vitro, downregulation of HADHA negatively regulated cell growth by causing G0/G1 arrest (P = 0.0008) similar to treatment with etomoxir, an inhibitor of FAO (P = 0.032). Moreover, downregulation of HADHA increased the susceptibility to doxorubicin (P = 0.002) and etoposide (P = 0.004). Moreover, these phenotypes were confirmed in an HADHA knockout system. Thus, we provide a basis for a novel therapeutic strategy through the regulation of HADHA and FAO in patients with refractory malignant lymphoma.


Asunto(s)
Linfoma , Subunidad alfa de la Proteína Trifuncional Mitocondrial , Antineoplásicos/farmacología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Regulación hacia Abajo , Descubrimiento de Drogas , Ácidos Grasos/metabolismo , Femenino , Humanos , Tejido Linfoide/química , Tejido Linfoide/metabolismo , Linfoma/metabolismo , Linfoma/mortalidad , Linfoma/patología , Masculino , Persona de Mediana Edad , Subunidad alfa de la Proteína Trifuncional Mitocondrial/antagonistas & inhibidores , Subunidad alfa de la Proteína Trifuncional Mitocondrial/genética , Subunidad alfa de la Proteína Trifuncional Mitocondrial/metabolismo , Oxidación-Reducción , Proteoma/análisis , Proteoma/metabolismo
14.
ACS Chem Biol ; 15(1): 158-170, 2020 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-31874028

RESUMEN

Selective toxicity among cancer cells of the same lineage is a hallmark of targeted therapies. As such, identifying compounds that impair proliferation of a subset of non-small-cell lung cancer (NSCLC) cell lines represents one strategy to discover new drugs for lung cancer. Previously, phenotypic screens of 202 103 compounds led to the identification of 208 selective NSCLC toxins ( McMillan , E. A. , Cell , 2018 , 173 , 864 ). The mechanism of action for the majority of these compounds remains unknown. Here, we discovered the target for a series of quinazoline diones (QDC) that demonstrate selective toxicity among 96 NSCLC lines. Using photoreactive probes, we found that the QDC binds to both mitochondrial complex I of the electron transport chain and hydroxyacyl CoA dehydrogenase subunit alpha (HADHA), which catalyzes long-chain fatty acid oxidation. Inhibition of complex I is the on-target activity for QDC, while binding to HADHA is off-target. The sensitivity profile of the QDC across NSCLC lines correlated with the sensitivity profiles of six additional structurally distinct compounds. The antiproliferative activity of these compounds is also the consequence of binding to mitochondrial complex I, reflecting significant structural diversity among complex I inhibitors. Small molecules targeting complex I are currently in clinical development for the treatment of cancer. Our results highlight complex I as a target in NSCLC and report structurally diverse scaffolds that inhibit complex I.


Asunto(s)
Antineoplásicos/química , Carcinoma de Pulmón de Células no Pequeñas/dietoterapia , Complejo I de Transporte de Electrón/antagonistas & inhibidores , Inhibidores Enzimáticos/química , Neoplasias Pulmonares/dietoterapia , Quinazolinonas/química , 3-Hidroxiacil-CoA Deshidrogenasas/metabolismo , Antineoplásicos/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Inhibidores Enzimáticos/farmacología , Ácidos Grasos/metabolismo , Técnicas de Inactivación de Genes , Humanos , Subunidad alfa de la Proteína Trifuncional Mitocondrial/genética , Subunidad alfa de la Proteína Trifuncional Mitocondrial/metabolismo , Estructura Molecular , Terapia Molecular Dirigida , Oxidación-Reducción , Consumo de Oxígeno , Unión Proteica , Conformación Proteica , Proteómica , Quinazolinonas/farmacología , Relación Estructura-Actividad , Especificidad por Sustrato
15.
Nat Commun ; 10(1): 4671, 2019 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-31604922

RESUMEN

Mitochondrial trifunctional protein deficiency, due to mutations in hydratase subunit A (HADHA), results in sudden infant death syndrome with no cure. To reveal the disease etiology, we generated stem cell-derived cardiomyocytes from HADHA-deficient hiPSCs and accelerated their maturation via an engineered microRNA maturation cocktail that upregulated the epigenetic regulator, HOPX.  Here we report, matured HADHA mutant cardiomyocytes treated with an endogenous mixture of fatty acids manifest the disease phenotype: defective calcium dynamics and repolarization kinetics which results in a pro-arrhythmic state. Single cell RNA-seq reveals a cardiomyocyte developmental intermediate, based on metabolic gene expression. This intermediate gives rise to mature-like cardiomyocytes in control cells but, mutant cells transition to a pathological state with reduced fatty acid beta-oxidation, reduced mitochondrial proton gradient, disrupted cristae structure and defective cardiolipin remodeling. This study reveals that HADHA (tri-functional protein alpha), a monolysocardiolipin acyltransferase-like enzyme, is required for fatty acid beta-oxidation and cardiolipin remodeling, essential for functional mitochondria in human cardiomyocytes.


Asunto(s)
Cardiolipinas/metabolismo , Ácidos Grasos/metabolismo , Subunidad alfa de la Proteína Trifuncional Mitocondrial/fisiología , Miocitos Cardíacos/metabolismo , Calcio/metabolismo , Línea Celular , Electrofisiología , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Proteínas de Homeodominio/fisiología , Células Madre Embrionarias Humanas , Humanos , MicroARNs/fisiología , Mitocondrias/fisiología , Proteína Trifuncional Mitocondrial/deficiencia , Subunidad alfa de la Proteína Trifuncional Mitocondrial/genética , Subunidad alfa de la Proteína Trifuncional Mitocondrial/metabolismo , Miocitos Cardíacos/patología , Miocitos Cardíacos/fisiología , Oxidación-Reducción , Técnicas de Placa-Clamp , RNA-Seq , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , Proteínas Supresoras de Tumor/fisiología
16.
Anal Chem ; 91(17): 10937-10942, 2019 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-31393705

RESUMEN

Proteoforms, the primary effectors of biological processes, are the different forms of proteins that arise from molecular processing events such as alternative splicing and post-translational modifications. Heart diseases exhibit changes in proteoform levels, motivating the development of a deeper understanding of the heart proteoform landscape. Our recently developed two-dimensional top-down proteomics platform coupling serial size exclusion chromatography (sSEC) to reversed-phase chromatography (RPC) expanded coverage of the human heart proteome and allowed observation of high-molecular weight proteoforms. However, most of these observed proteoforms were not identified due to the difficulty in obtaining quality tandem mass spectrometry (MS2) fragmentation data for large proteoforms from complex biological mixtures on a chromatographic time scale. Herein, we sought to identify human heart proteoforms in this data set using an enhanced version of Proteoform Suite, which identifies proteoforms by intact mass alone. Specifically, we added a new feature to Proteoform Suite to determine candidate identifications for isotopically unresolved proteoforms larger than 50 kDa, enabling subsequent MS2 identification of important high-molecular weight human heart proteoforms such as lamin A (72 kDa) and trifunctional enzyme subunit α (79 kDa). With this new workflow for large proteoform identification, endogenous human cardiac myosin binding protein C (140 kDa) was identified for the first time. This study demonstrates the integration of our sSEC-RPC-MS proteomics platform with intact-mass analysis through Proteoform Suite to create a catalog of human heart proteoforms and facilitate the identification of large proteoforms in complex systems.


Asunto(s)
Proteínas Portadoras/aislamiento & purificación , Lamina Tipo A/aislamiento & purificación , Subunidad alfa de la Proteína Trifuncional Mitocondrial/aislamiento & purificación , Miocardio/química , Procesamiento Proteico-Postraduccional , Proteoma/aislamiento & purificación , Programas Informáticos , Empalme Alternativo , Secuencia de Aminoácidos , Proteínas Portadoras/química , Proteínas Portadoras/metabolismo , Cromatografía en Gel , Cromatografía de Fase Inversa , Humanos , Lamina Tipo A/química , Lamina Tipo A/metabolismo , Subunidad alfa de la Proteína Trifuncional Mitocondrial/química , Subunidad alfa de la Proteína Trifuncional Mitocondrial/metabolismo , Miocardio/metabolismo , Proteoma/química , Proteoma/metabolismo , Proteómica/métodos , Espectrometría de Masas en Tándem
17.
Cell Death Dis ; 10(2): 66, 2019 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-30683850

RESUMEN

Protein acetylation has a crucial role in energy metabolism. Here we performed the first large-scale profiling of acetylome in rat islets, showing that almost all enzymes in core metabolic pathways related to insulin secretion were acetylated. Label-free quantitative acetylome of islets in response to high glucose revealed hyperacetylation of enzymes involved in fatty acid ß-oxidation (FAO), including trifunctional enzyme subunit alpha (ECHA). Acetylation decreased the protein stability of ECHA and its ability to promote FAO. The overexpression of SIRT3, a major mitochondrial deacetylase, prevented the degradation of ECHA via decreasing its acetylation level in ß-cells. SIRT3 expression was upregulated in rat islets upon exposure to low glucose or fasting. SIRT3 overexpression in islets markedly decreased palmitate-potentiated insulin secretion, whereas islets from SIRT3 knockout mice secreted more insulin, with an opposite action on FAO. ECHA overexpression partially reversed SIRT3 deficiency-elicited insulin hypersecretion. Our study highlights the potential role of protein acetylation in insulin secretion.


Asunto(s)
Ácidos Grasos/metabolismo , Glucosa/metabolismo , Células Secretoras de Insulina/fisiología , Sirtuina 3/metabolismo , Sirtuinas/metabolismo , Acetilación , Animales , Estabilidad de Enzimas , Ayuno , Células HEK293 , Humanos , Secreción de Insulina/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Subunidad alfa de la Proteína Trifuncional Mitocondrial/metabolismo , Oxidación-Reducción , Ratas , Ratas Sprague-Dawley , Sirtuina 3/genética , Transfección
18.
J Biol Chem ; 293(46): 17676-17684, 2018 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-30323061

RESUMEN

Sirtuin 3 (SIRT3) deacetylates and activates several mitochondrial fatty acid oxidation enzymes in the liver. Here, we investigated whether the protein acetylase GCN5 general control of amino acid synthesis 5-like 1 (GCN5L1), previously shown to oppose SIRT3 activity, is involved in the regulation of hepatic fatty acid oxidation. We show that GCN5L1 abundance is significantly up-regulated in response to an acute high-fat diet (HFD). Transgenic GCN5L1 overexpression in the mouse liver increased protein acetylation levels, and proteomic detection of specific lysine residues identified numerous sites that are co-regulated by GCN5L1 and SIRT3. We analyzed several fatty acid oxidation proteins identified by the proteomic screen and found that hyperacetylation of hydroxyacyl-CoA dehydrogenase trifunctional multienzyme complex subunit α (HADHA) correlates with increased GCN5L1 levels. Stable GCN5L1 knockdown in HepG2 cells reduced HADHA acetylation and increased activities of fatty acid oxidation enzymes. Mice with a liver-specific deletion of GCN5L1 were protected from hepatic lipid accumulation following a chronic HFD and did not exhibit hyperacetylation of HADHA compared with WT controls. Finally, we found that GCN5L1-knockout mice lack HADHA that is hyperacetylated at three specific lysine residues (Lys-350, Lys-383, and Lys-406) and that acetylation at these sites is significantly associated with increased HADHA activity. We conclude that GCN5L1-mediated regulation of mitochondrial protein acetylation plays a role in hepatic metabolic homeostasis.


Asunto(s)
Ácidos Grasos/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Acetilación , Animales , Dieta Alta en Grasa/efectos adversos , Hígado Graso/prevención & control , Células Hep G2 , Humanos , Lisina/química , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Mitocondriales , Subunidad alfa de la Proteína Trifuncional Mitocondrial/metabolismo , Proteínas del Tejido Nervioso/genética , Oxidación-Reducción , Isoformas de Proteínas/metabolismo , Procesamiento Proteico-Postraduccional , Proteómica , Sirtuina 3/genética
19.
Sci Rep ; 8(1): 3254, 2018 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-29459657

RESUMEN

Children diagnosed with Long-Chain-3-Hydroxy-Acyl-CoA-Dehydrogenase-Deficiency (LCHADD) or Very-Long-Chain-3-Hydroxy-Acyl-CoA-Dehydrogenase-Deficiency (VLCADD) frequently present with hypertrophic cardiomyopathy or muscle weakness which is caused by the accumulation of fatty acid metabolites due to inactivating mutations in the mitochondrial trifunctional protein. By analyzing mitochondrial morphology we uncovered that mutations within the HADHA or the ACADVL gene not only affect fatty acid oxidation, but also cause significant changes in the DNM1L/MFN2 ratio leading to the significant accumulation of truncated and punctate mitochondria in contrast to network-like mitochondrial morphology in controls. These striking morphological abnormalities correlate with changes in OXPHOS, an imbalance in ROS levels, reduced mitochondrial respiration, reduced growth rates and significantly increased glucose uptake per cell, suggesting that HADHA and ACADVL mutations shift cellular energy household into glycolysis. Experiments using the NOX2-specific inhibitor Phox-I2 suggest that NOX2 is activated by accumulating long-chain fatty acids and generates ROS, which in turn changes mitochondrial morphology and activity. We thereby provide novel insights into the cellular energy household of cells from LCHADD/VLCADD patients and demonstrate for the first time a connection between fatty acid metabolism, mitochondrial morphology and ROS in patients with these rare genetic disorders.


Asunto(s)
Acil-CoA Deshidrogenasa de Cadena Larga/deficiencia , Cardiomiopatía Hipertrófica/patología , Mitocondrias/enzimología , Enfermedades Mitocondriales/patología , Dinámicas Mitocondriales , Subunidad alfa de la Proteína Trifuncional Mitocondrial/deficiencia , Debilidad Muscular/patología , Acil-CoA Deshidrogenasa de Cadena Larga/metabolismo , Cardiomiopatía Hipertrófica/genética , Respiración de la Célula , Dinaminas , GTP Fosfohidrolasas/análisis , Glucólisis , Humanos , Proteínas Asociadas a Microtúbulos/análisis , Mitocondrias/fisiología , Mitocondrias/ultraestructura , Enfermedades Mitocondriales/genética , Proteínas Mitocondriales/análisis , Subunidad alfa de la Proteína Trifuncional Mitocondrial/metabolismo , Debilidad Muscular/genética , Fosforilación Oxidativa
20.
Mol Biosyst ; 13(8): 1504-1511, 2017 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-28632266

RESUMEN

Xuesaitong injection (XST), which mainly consists of Panax notoginseng saponins, has been widely used for treating cardio-cerebral vascular diseases. However, the underlying mechanisms of XST associated with its cardioprotective effects are still unclear. To identify the potential target proteins of XST, two-dimensional gel electrophoresis (2-DE)-based proteomics was utilized to analyze the protein profile of myocardium in rats with myocardial ischemia/reperfusion (I/R) injury. The differentially expressed proteins were identified by matrix assisted laser desorption/ionization time-of-flight mass spectrometry. It is interesting that XST can alter the expression of 7 proteins, including pyruvate dehydrogenase E1 alpha (PDHA1), hydroxyacyl-coenzyme A dehydrogenase (HADHA), peroxiredoxin 3 (PRX3), gamma-enolase, acetyl-coenzyme A acyltransferase 2 (ACAA2), etc. Functional analysis revealed that those proteins were chiefly related to cardiac energy metabolism and oxidative stress. The cardioprotective effects of XST were further validated in H9c2 cardiac muscle cells with hypoxia/reoxygenation injury. We found that XST can promote the activity of PDH, an important enzyme related to the TCA cycle, as well as increase the intracellular content of acetyl-CoA and ATP. Moreover, XST also attenuated intracellular MDA release in H2O2-injured cardiac cells. This is the first study on the proteomic expression of XST-treated myocardium with I/R injury to reveal that the cardioprotective effects of XST may be attributed to the PDH-mediated restoration of aerobic glucose oxidation.


Asunto(s)
Fármacos Cardiovasculares/farmacología , Medicamentos Herbarios Chinos/farmacología , Metabolismo Energético/efectos de los fármacos , Regulación de la Expresión Génica , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Fármacos Neuroprotectores/farmacología , Saponinas/farmacología , Acetil-CoA C-Aciltransferasa/genética , Acetil-CoA C-Aciltransferasa/metabolismo , Animales , Línea Celular , Metabolismo Energético/genética , Perfilación de la Expresión Génica , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Masculino , Subunidad alfa de la Proteína Trifuncional Mitocondrial/genética , Subunidad alfa de la Proteína Trifuncional Mitocondrial/metabolismo , Daño por Reperfusión Miocárdica/genética , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Miocardio/metabolismo , Miocardio/patología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Estrés Oxidativo , Fosfopiruvato Hidratasa/genética , Fosfopiruvato Hidratasa/metabolismo , Piruvato Deshidrogenasa (Lipoamida)/genética , Piruvato Deshidrogenasa (Lipoamida)/metabolismo , Ratas , Ratas Sprague-Dawley , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...