Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Redox Biol ; 67: 102932, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37883842

RESUMEN

The NDUFS4 knockout (KO) mouse phenotype resembles the human Complex I deficiency Leigh Syndrome. The irreversible succination of protein thiols by fumarate is increased in select regions of the NDUFS4 KO brain affected by neurodegeneration. We report that dihydrolipoyllysine-residue succinyltransferase (DLST), a component of the α-ketoglutarate dehydrogenase complex (KGDHC) of the tricarboxylic acid (TCA) cycle, is succinated in the affected regions of the NDUFS4 KO brain. Succination of DLST reduced KGDHC activity in the brainstem (BS) and olfactory bulb (OB) of KO mice. The defective production of KGDHC derived succinyl-CoA resulted in decreased mitochondrial substrate level phosphorylation (SLP), further aggravating the existing oxidative phosphorylation (OXPHOS) ATP deficit. Protein succinylation, an acylation modification that requires succinyl-CoA, was reduced in the KO mice. Modeling succination of a cysteine in the spatial vicinity of the DLST active site or introduction of succinomimetic mutations recapitulates these metabolic deficits. Our data demonstrate that the biochemical deficit extends beyond impaired Complex I assembly and OXPHOS deficiency, functionally impairing select components of the TCA cycle to drive metabolic perturbations in affected neurons.


Asunto(s)
Ciclo del Ácido Cítrico , Complejo Cetoglutarato Deshidrogenasa , Ratones , Animales , Humanos , Complejo Cetoglutarato Deshidrogenasa/genética , Complejo Cetoglutarato Deshidrogenasa/metabolismo , Mitocondrias/genética , Mitocondrias/metabolismo , Ratones Noqueados , Fosforilación Oxidativa , Adenosina Trifosfato/metabolismo
2.
Semin Immunol ; 70: 101845, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37806032

RESUMEN

The gasdermin family of proteins are central effectors of the inflammatory, lytic cell death modality known as pyroptosis. Characterized in 2015, the most well-studied member gasdermin D can be proteolyzed, typically by caspases, to generate an active pore-forming N-terminal domain. At least well-studied three pharmacological inhibitors (necrosulfonamide, disulfiram, dimethyl fumarate) since 2018 have been shown to affect gasdermin D activity either through modulation of processing or interference with pore formation. A multitude of murine in vivo studies have since followed. Here, we discuss the current state of research surrounding these three inhibitors, caveats to their use, and a set of guiding principles that researchers should consider when pursuing further studies of gasdermin D inhibition.


Asunto(s)
Gasderminas , Animales , Humanos , Ratones , Caspasas/metabolismo , Gasderminas/química , Inflamasomas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de Neoplasias/química , Proteínas de Neoplasias/metabolismo , Piroptosis
3.
Breast Cancer Res Treat ; 201(3): 561-570, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37477798

RESUMEN

PURPOSE: The oncogenic factor ZNF217 promotes aggressive estrogen receptor (ER)+breast cancer disease suggesting that its inhibition may be useful in the clinic. Unfortunately, no direct pharmacological inhibitor is available. Dimethyl fumarate (DMF) exhibits anti-breast cancer activities, in vitro and in pre-clinical in vivo models. Its therapeutic benefits stem from covalent modification of cellular thiols such as protein cysteines, but the full profile of molecular targets mediating its anti-breast cancer effects remains to be determined. METHODS: ER+breast cancer cells were treated with DMF followed by cysteine-directed proteomics. Cells with modulated ZNF217 levels were used to probe the efficacy of DMF. RESULTS: Covalent modification of ZNF217 by DMF identified by proteomics was confirmed by using a DMF-chemical probe. Inhibition of ZNF217's transcriptional activity by DMF was evident on reported ZNF217-target genes. ZNF217 as an oncogene has been shown to enhance stem-like properties, survival, proliferation, and invasion. Consistent with ZNF217 inhibition, DMF was more effective at blocking these ZNF217-driven phenotypes in cells with elevated ZNF217 expression. Furthermore, partial knockdown of ZNF217 led to a reduction in DMF's efficacy. DMF's in vivo activity was evaluated in a xenograft model of MCF-7 HER2 cells that have elevated expression of ZNF217 and DMF treatment resulted in significant inhibition of tumor growth. CONCLUSION: These data indicate that DMF's anti-breast cancer activities in the ER+HER2+models, at least in part, are due to inhibition of ZNF217. DMF is identified as a new covalent inhibitor of ZNF217.


Asunto(s)
Neoplasias de la Mama , Humanos , Femenino , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Dimetilfumarato/farmacología , Dimetilfumarato/uso terapéutico , Receptores de Estrógenos , Transactivadores/genética , Transactivadores/metabolismo , Transactivadores/uso terapéutico , Células MCF-7
4.
Cell Metab ; 35(6): 961-978.e10, 2023 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-37178684

RESUMEN

Metabolic alterations in the microenvironment significantly modulate tumor immunosensitivity, but the underlying mechanisms remain obscure. Here, we report that tumors depleted of fumarate hydratase (FH) exhibit inhibition of functional CD8+ T cell activation, expansion, and efficacy, with enhanced malignant proliferative capacity. Mechanistically, FH depletion in tumor cells accumulates fumarate in the tumor interstitial fluid, and increased fumarate can directly succinate ZAP70 at C96 and C102 and abrogate its activity in infiltrating CD8+ T cells, resulting in suppressed CD8+ T cell activation and anti-tumor immune responses in vitro and in vivo. Additionally, fumarate depletion by increasing FH expression strongly enhances the anti-tumor efficacy of anti-CD19 CAR T cells. Thus, these findings demonstrate a role for fumarate in controlling TCR signaling and suggest that fumarate accumulation in the tumor microenvironment (TME) is a metabolic barrier to CD8+ T cell anti-tumor function. And potentially, fumarate depletion could be an important strategy for tumor immunotherapy.


Asunto(s)
Linfocitos T CD8-positivos , Neoplasias , Humanos , Fumaratos/farmacología , Fumaratos/metabolismo , Microambiente Tumoral , Neoplasias/metabolismo , Transducción de Señal
5.
Hum Pathol ; 134: 102-113, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36581128

RESUMEN

Fumarate hydratase (FH)-deficient renal cell carcinoma (RCC) is an aggressive, rare genetic disease affecting the kidney and other organ systems. We constructed a specialized multi-institutional cohort of 20 primary FH-deficient RCC cases with aims of characterizing a new commercially available antibody, S-(2-succino)-cysteine (2SC). Herein, we present our findings on the biomarker characterization and performance of 2SC expression by immunohistochemistry (IHC) in FH-deficient RCC and other common and rare RCC subtypes. Morphological assessment revealed characteristic cytomorphologic features and a majority (55%) of FH-deficient RCC had mixed architectural growth patterns. We observed predominantly diffuse and strong cytoplasmic staining with limited nuclear positivity for 2SC staining on IHC. Receiver operating characteristic curves (ROC) for 2SC identified the threshold IHC score (cutoff) as 90, with the sensitivity and specificity being 100% and 91%, respectively. The findings of the present study along with the prior evidence in literature encourage utilization of 2SC as a positive marker along with the loss of FH expression by anti-FH IHC staining as a negative marker, in clinical and/or pathologic scenarios when considering FH-deficient RCC in the differential diagnosis. FH-/2SC+ may serve as a comprehensive IHC panel in identifying such cases and excluding morphologically similar entities.


Asunto(s)
Carcinoma de Células Renales , Neoplasias Renales , Leiomiomatosis , Neoplasias Uterinas , Humanos , Femenino , Carcinoma de Células Renales/patología , Cisteína , Fumarato Hidratasa , Leiomiomatosis/genética , Neoplasias Renales/patología , Biomarcadores de Tumor/genética , Neoplasias Uterinas/patología
6.
J Biol Chem ; 298(12): 102639, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36309089

RESUMEN

Succination is the spontaneous reaction between the respiratory intermediate fumarate and cellular thiols that forms stable S-(2-succino)-adducts such as S-(2-succino)cysteine (2SC). 2SC is a biomarker for conditions associated with elevated fumarate levels, including diabetes, obesity, and certain cancers, and succination likely contributes to disease progression. Bacillus subtilis has a yxe operon-encoded breakdown pathway for 2SC that involves three distinct enzymatic conversions. The first step is N-acetylation of 2SC by YxeL to form N-acetyl-2SC (2SNAC). YxeK catalyzes the oxygenation of 2SNAC, resulting in its breakdown to oxaloacetate and N-acetylcysteine, which is deacetylated by YxeP to give cysteine. The monooxygenase YxeK is key to the pathway but is rare, with close homologs occurring infrequently in prokaryote and fungal genomes. The existence of additional 2SC breakdown pathways was not known prior to this study. Here, we used comparative genomics to identify a S-(2-succino) lyase (2SL) that replaces yxeK in some yxe gene clusters. 2SL genes from Enterococcus italicus and Dickeya dadantii complement B. subtilis yxeK mutants. We also determined that recombinant 2SL enzymes efficiently break down 2SNAC into fumarate and N-acetylcysteine, can perform the reverse reaction, and have minor activity against 2SC and other small molecule thiols. The strong preferences both YxeK and 2SL enzymes have for 2SNAC indicate that 2SC acetylation is a conserved breakdown step. The identification of a second naturally occurring 2SC breakdown pathway underscores the importance of 2SC catabolism and defines a general strategy for 2SC breakdown involving acetylation, breakdown, and deacetylation.


Asunto(s)
Cisteína , Liasas , Cisteína/metabolismo , Acetilcisteína , Compuestos de Sulfhidrilo , Fumaratos/metabolismo
7.
Mol Cell ; 82(7): 1249-1260.e7, 2022 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-35216667

RESUMEN

Fumarate is an oncometabolite. However, the mechanism underlying fumarate-exerted tumorigenesis remains unclear. Here, utilizing human type2 papillary renal cell carcinoma (PRCC2) as a model, we show that fumarate accumulates in cells deficient in fumarate hydratase (FH) and inhibits PTEN to activate PI3K/AKT signaling. Mechanistically, fumarate directly reacts with PTEN at cysteine 211 (C211) to form S-(2-succino)-cysteine. Succinated C211 occludes tethering of PTEN with the cellular membrane, thereby diminishing its inhibitory effect on the PI3K/AKT pathway. Functionally, re-expressing wild-type FH or PTEN C211S phenocopies an AKT inhibitor in suppressing tumor growth and sensitizing PRCC2 to sunitinib. Analysis of clinical specimens indicates that PTEN C211 succination levels are positively correlated with AKT activation in PRCC2. Collectively, these findings elucidate a non-metabolic, oncogenic role of fumarate in PRCC2 via direct post-translational modification of PTEN and further reveal potential stratification strategies for patients with FH loss by combinatorial AKTi and sunitinib therapy.


Asunto(s)
Carcinoma Papilar , Carcinoma de Células Renales , Fumaratos , Neoplasias Renales , Fosfohidrolasa PTEN , Carcinogénesis , Carcinoma Papilar/tratamiento farmacológico , Carcinoma Papilar/enzimología , Carcinoma Papilar/genética , Carcinoma Papilar/metabolismo , Carcinoma de Células Renales/tratamiento farmacológico , Carcinoma de Células Renales/enzimología , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/metabolismo , Cisteína/metabolismo , Resistencia a Antineoplásicos , Fumarato Hidratasa/genética , Fumarato Hidratasa/metabolismo , Fumaratos/farmacología , Humanos , Neoplasias Renales/tratamiento farmacológico , Neoplasias Renales/enzimología , Neoplasias Renales/genética , Neoplasias Renales/metabolismo , Fosfohidrolasa PTEN/antagonistas & inhibidores , Fosfohidrolasa PTEN/genética , Fosfatidilinositol 3-Quinasas/genética , Proteínas Proto-Oncogénicas c-akt/genética , Sunitinib/farmacología
8.
Semin Cancer Biol ; 61: 158-166, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31689495

RESUMEN

Hereditary Leiomyomatosis and Renal Cell Cancer (HLRCC) is an autosomal dominant hereditary cancer syndrome with incomplete penetrance. It is caused by a germline amorphic allele of the FH gene, which encodes the TCA cycle enzyme, fumarate hydratase (FH). HLRCC patients are genetically predisposed to develop skin leiomyomas, uterine fibroids, and the aggressive kidney cancer of type 2 papillary morphology. Loss-of-heterozygocity at the FH locus that cause a complete loss of FH enzymatic function is always detected in these tumor tissues. Molecular pathway elucidation, genomic studies, and systematic genetics screens reported over the last two decades have identified several FH-inactivation driven pathways alterations, as well as rationally conceived treatment strategies that specifically target FH-/- tumor cells. These treatment strategies include ferroptosis induction, oxidative stress promotion, and metabolic alteration. As the fundamental biology of HLRCC continues to be uncovered, these treatment strategies continue to be refined and may one day lead to a strategy to prevent disease onset among HLRCC patients. With a more complete picture of HLRCC biology, the safe translation of experimental treatment strategies into clinical practice is achievable in the foreseeable future.


Asunto(s)
Leiomiomatosis/etiología , Síndromes Neoplásicos Hereditarios/etiología , Neoplasias Cutáneas/etiología , Neoplasias Uterinas/etiología , Biomarcadores de Tumor , Manejo de la Enfermedad , Susceptibilidad a Enfermedades , Fumarato Hidratasa/genética , Genes Supresores de Tumor , Predisposición Genética a la Enfermedad , Pruebas Genéticas , Genómica/métodos , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Leiomiomatosis/diagnóstico , Leiomiomatosis/metabolismo , Leiomiomatosis/terapia , Mutación , Síndromes Neoplásicos Hereditarios/diagnóstico , Síndromes Neoplásicos Hereditarios/metabolismo , Síndromes Neoplásicos Hereditarios/terapia , Proteoma , Transducción de Señal , Neoplasias Cutáneas/diagnóstico , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/terapia , Investigación Biomédica Traslacional , Neoplasias Uterinas/diagnóstico , Neoplasias Uterinas/metabolismo , Neoplasias Uterinas/terapia
9.
Cell Metab ; 29(2): 430-442.e4, 2019 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-30415925

RESUMEN

Diabetes is a bihormonal disorder resulting from combined insulin and glucagon secretion defects. Mice lacking fumarase (Fh1) in their ß cells (Fh1ßKO mice) develop progressive hyperglycemia and dysregulated glucagon secretion similar to that seen in diabetic patients (too much at high glucose and too little at low glucose). The glucagon secretion defects are corrected by low concentrations of tolbutamide and prevented by the sodium-glucose transport (SGLT) inhibitor phlorizin. These data link hyperglycemia, intracellular Na+ accumulation, and acidification to impaired mitochondrial metabolism, reduced ATP production, and dysregulated glucagon secretion. Protein succination, reflecting reduced activity of fumarase, is observed in α cells from hyperglycemic Fh1ßKO and ß-V59M gain-of-function KATP channel mice, diabetic Goto-Kakizaki rats, and patients with type 2 diabetes. Succination is also observed in renal tubular cells and cardiomyocytes from hyperglycemic Fh1ßKO mice, suggesting that the model can be extended to other SGLT-expressing cells and may explain part of the spectrum of diabetic complications.


Asunto(s)
Adenosina Trifosfato/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Células Secretoras de Glucagón/metabolismo , Glucagón/metabolismo , Hiperglucemia/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Animales , Línea Celular , Células Secretoras de Glucagón/citología , Humanos , Células Secretoras de Insulina/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Canales de Potasio/metabolismo , Ratas , Ratas Wistar , Sodio/metabolismo
10.
Mol Cell Proteomics ; 18(3): 504-519, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30587509

RESUMEN

The fumarate ester dimethyl fumarate (DMF) has been introduced recently as a treatment for relapsing remitting multiple sclerosis (RRMS), a chronic inflammatory condition that results in neuronal demyelination and axonal loss. DMF is known to act by depleting intracellular glutathione and modifying thiols on Keap1 protein, resulting in the stabilization of the transcription factor Nrf2, which in turn induces the expression of antioxidant response element genes. We have previously shown that DMF reacts with a wide range of protein thiols, suggesting that the complete mechanisms of action of DMF are unknown. Here, we investigated other intracellular thiol residues that may also be irreversibly modified by DMF in neurons and astrocytes. Using mass spectrometry, we identified 24 novel proteins that were modified by DMF in neurons and astrocytes, including cofilin-1, tubulin and collapsin response mediator protein 2 (CRMP2). Using an in vitro functional assay, we demonstrated that DMF-modified cofilin-1 loses its activity and generates less monomeric actin, potentially inhibiting its cytoskeletal remodeling activity, which could be beneficial in the modulation of myelination during RRMS. DMF modification of tubulin did not significantly impact axonal lysosomal trafficking. We found that the oxygen consumption rate of N1E-115 neurons and the levels of proteins related to mitochondrial energy production were only slightly affected by the highest doses of DMF, confirming that DMF treatment does not impair cellular respiratory function. In summary, our work provides new insights into the mechanisms supporting the neuroprotective and remyelination benefits associated with DMF treatment in addition to the antioxidant response by Nrf2.


Asunto(s)
Astrocitos/metabolismo , Cisteína/efectos de los fármacos , Dimetilfumarato/farmacología , Factor 2 Relacionado con NF-E2/metabolismo , Neuronas/metabolismo , Células 3T3-L1 , Animales , Astrocitos/citología , Astrocitos/efectos de los fármacos , Células Cultivadas , Cofilina 1/química , Cofilina 1/metabolismo , Péptidos y Proteínas de Señalización Intercelular , Espectrometría de Masas , Ratones , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/metabolismo , Neuronas/citología , Neuronas/efectos de los fármacos , Ratas , Tubulina (Proteína)/química , Tubulina (Proteína)/metabolismo
11.
J Biol Chem ; 293(21): 8255-8263, 2018 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-29626092

RESUMEN

Cellular thiols such as cysteine spontaneously and readily react with the respiratory intermediate fumarate, resulting in the formation of stable S-(2-succino)-adducts. Fumarate-mediated succination of thiols increases in certain tumors and in response to glucotoxicity associated with diabetes. Therefore, S-(2-succino)-adducts such as S-(2-succino)cysteine (2SC) are considered oncometabolites and biomarkers for human disease. No disposal routes for S-(2-succino)-compounds have been reported prior to this study. Here, we show that Bacillus subtilis metabolizes 2SC to cysteine using a pathway encoded by the yxe operon. The first step is N-acetylation of 2SC followed by an oxygenation that we propose results in the release of oxaloacetate and N-acetylcysteine, which is deacetylated to give cysteine. Knockouts of the genes predicted to mediate each step in the pathway lose the ability to grow on 2SC as the sulfur source and accumulate the expected upstream metabolite(s). We further show that N-acetylation of 2SC relieves toxicity. This is the first demonstration of a metabolic disposal route for any S-(2-succino)-compound, paving the way toward the identification of corresponding pathways in other species.


Asunto(s)
Bacillus subtilis/metabolismo , Cisteína/análogos & derivados , Fumaratos/metabolismo , Metabolómica , Neoplasias/patología , Operón , Acetilación , Bacillus subtilis/genética , Cisteína/metabolismo , Neoplasias/genética , Transducción de Señal
12.
Amino Acids ; 50(5): 647-652, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29508062

RESUMEN

Fumarate adduction to cysteines has been implicated in the pathogenesis of several disorders. Its role, however, still remains elusive, and the need of predictive methods has not yet been met. The reactivity of cysteines found in fumarate-sensitive proteins was predicted when the collected data for eight network-type features were analyzed using classification models. Therefore, methods for evaluating the likelihood of a cysteine site to be modified by fumarate could be developed by combining concepts of network theory and machine learning.


Asunto(s)
Cisteína/química , Bases de Datos de Proteínas , Fumaratos/química , Proteínas/química , Análisis de Secuencia de Proteína , Proteínas/genética
13.
Cell Rep ; 21(4): 1036-1047, 2017 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-29069586

RESUMEN

Fumarate hydratase (FH) is an enzyme of the tricarboxylic acid (TCA) cycle mutated in hereditary and sporadic cancers. Despite recent advances in understanding its role in tumorigenesis, the effects of FH loss on mitochondrial metabolism are still unclear. Here, we used mouse and human cell lines to assess mitochondrial function of FH-deficient cells. We found that human and mouse FH-deficient cells exhibit decreased respiration, accompanied by a varying degree of dysfunction of respiratory chain (RC) complex I and II. Moreover, we show that fumarate induces succination of key components of the iron-sulfur cluster biogenesis family of proteins, leading to defects in the biogenesis of iron-sulfur clusters that affect complex I function. We also demonstrate that suppression of complex II activity is caused by product inhibition due to fumarate accumulation. Overall, our work provides evidence that the loss of a single TCA cycle enzyme is sufficient to cause combined RC activity dysfunction.


Asunto(s)
Complejo II de Transporte de Electrones/metabolismo , Complejo I de Transporte de Electrón/metabolismo , Fumarato Hidratasa/metabolismo , Animales , Línea Celular Tumoral , Respiración de la Célula , Fumarato Hidratasa/deficiencia , Fumarato Hidratasa/genética , Fumaratos/metabolismo , Humanos , Proteínas Hierro-Azufre/metabolismo , Ratones
14.
Cell Rep ; 20(13): 3135-3148, 2017 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-28954230

RESUMEN

We explored the role of the Krebs cycle enzyme fumarate hydratase (FH) in glucose-stimulated insulin secretion (GSIS). Mice lacking Fh1 in pancreatic ß cells (Fh1ßKO mice) appear normal for 6-8 weeks but then develop progressive glucose intolerance and diabetes. Glucose tolerance is rescued by expression of mitochondrial or cytosolic FH but not by deletion of Hif1α or Nrf2. Progressive hyperglycemia in Fh1ßKO mice led to dysregulated metabolism in ß cells, a decrease in glucose-induced ATP production, electrical activity, cytoplasmic [Ca2+]i elevation, and GSIS. Fh1 loss resulted in elevated intracellular fumarate, promoting succination of critical cysteines in GAPDH, GMPR, and PARK 7/DJ-1 and cytoplasmic acidification. Intracellular fumarate levels were increased in islets exposed to high glucose and in islets from human donors with type 2 diabetes (T2D). The impaired GSIS in islets from diabetic Fh1ßKO mice was ameliorated after culture under normoglycemic conditions. These studies highlight the role of FH and dysregulated mitochondrial metabolism in T2D.


Asunto(s)
Diabetes Mellitus Tipo 2/genética , Fumarato Hidratasa/deficiencia , Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Animales , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Ratones
15.
Cell Chem Biol ; 24(3): 306-315, 2017 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-28219662

RESUMEN

Enzymes of central carbon metabolism are essential mediators of Mycobacterium tuberculosis (Mtb) physiology and pathogenicity, but are often perceived to lack sufficient species selectivity to be pursued as potential drug targets. Fumarase (Fum) is an enzyme of the canonical tricarboxylic acid cycle and is dispensable in many organisms. Transposon mutagenesis studies in Mtb, however, indicate that Fum is required for optimal growth. Here, we report the generation and characterization of a genetically engineered Mtb strain in which Fum expression is conditionally regulated. This revealed that Fum deficiency is bactericidal in vitro and during both the acute and chronic phases of mouse infection. This essentiality is linked to marked accumulations of fumarate resulting in protein and metabolite succination, a covalent modification of cysteine thiol residues. These results identify Mtb Fum as a potentially species-specific drug target whose inactivation may kill Mtb through a covalently irreversible form of metabolic toxicity.


Asunto(s)
Proteínas Bacterianas/genética , Fumarato Hidratasa/genética , Mycobacterium tuberculosis/genética , Animales , Proteínas Bacterianas/metabolismo , Cromatografía Líquida de Alta Presión , Ciclo del Ácido Cítrico , Cisteína/química , Femenino , Fumarato Hidratasa/deficiencia , Fumarato Hidratasa/metabolismo , Fumaratos/análisis , Fumaratos/metabolismo , Ratones , Ratones Endogámicos C57BL , Mycobacterium tuberculosis/enzimología , Mycobacterium tuberculosis/patogenicidad , Estrés Oxidativo , Péptidos/análisis , Péptidos/química , Procesamiento Proteico-Postraduccional , Espectrometría de Masas en Tándem
16.
Biochim Biophys Acta ; 1864(2): 211-8, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26589354

RESUMEN

The adduction of fumaric acid to the sulfhydryl group of certain cysteine (Cys) residues in proteins via a Michael-like reaction leads to the formation of S-(2-succino)cysteine (2SC) sites. Although its role remains to be fully understood, this post-translational Cys modification (protein succination) has been implicated in the pathogenesis of diabetes/obesity and fumarate hydratase-related diseases. In this study, theoretical approaches to address sequence- and 3D-structure-based features possibly underlying the specificity of protein succination have been applied to perform the first analysis of the available data on the succinate proteome. A total of 182 succinated proteins, 205 modifiable, and 1750 non-modifiable sites have been examined. The rate of 2SC sites per protein ranged from 1 to 3, and the overall relative abundance of modifiable sites was 10.8%. Modifiable and non-modifiable sites were not distinguishable when the hydrophobicity of the Cys-flaking peptides, the acid dissociation constant value of the sulfhydryl groups, and the secondary structure of the Cys-containing segments were compared. By contrast, significant differences were determined when the accessibility of the sulphur atoms and the amino acid composition of the Cys-flaking peptides were analysed. Based on these findings, a sequence-based score function has been evaluated as a descriptor for Cys residues. In conclusion, our results indicate that modifiable and non-modifiable sites form heterogeneous subsets when features often discussed to describe Cys reactivity are examined. However, they also suggest that some differences exist, which may constitute the baseline for further investigations aimed at the development of predictive methods for 2SC sites in proteins.


Asunto(s)
Cisteína/análogos & derivados , Procesamiento Proteico-Postraduccional/genética , Proteínas/química , Proteoma , Aminoácidos/química , Aminoácidos/genética , Biología Computacional , Cisteína/química , Cisteína/genética , Fumaratos/química , Humanos , Modelos Teóricos , Conformación Molecular , Proteínas/genética , Análisis de Secuencia de Proteína , Succinatos/química
17.
Mass Spectrom Rev ; 33(2): 98-109, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24115015

RESUMEN

The post-translational modifications (PTMs) of cysteine residues include oxidation, S-glutathionylation, S-nitrosylation, and succination, all of which modify protein function or turnover in response to a changing intracellular redox environment. Succination is a chemical modification of cysteine in proteins by the Krebs cycle intermediate, fumarate, yielding S-(2-succino)cysteine (2SC). Intracellular fumarate concentration and succination of proteins are increased by hyperpolarization of the inner mitochondrial membrane, in concert with mitochondrial, endoplasmic reticulum (ER) and oxidative stress in 3T3 adipocytes grown in high glucose medium and in adipose tissue in obesity and diabetes in mice. Increased succination of proteins is also detected in the kidney of a fumarase deficient conditional knock-out mouse which develops renal cysts. A wide range of proteins are subject to succination, including enzymes, adipokines, cytoskeletal proteins, and ER chaperones with functional cysteine residues. There is also some overlap between succinated and glutathionylated proteins, suggesting that the same low pKa thiols are targeted by both. Succination of adipocyte proteins in diabetes increases as a result of nutrient excess derived mitochondrial stress and this is inhibited by uncouplers, which discharge the mitochondrial membrane potential (ΔΨm) and relieve the electron transport chain. 2SC therefore serves as a biomarker of mitochondrial stress or dysfunction in chronic diseases, such as obesity, diabetes, and cancer, and recent studies suggest that succination is a mechanistic link between mitochondrial dysfunction, oxidative and ER stress, and cellular progression toward apoptosis. In this article, we review the history of the succinated proteome and the challenges associated with measuring this non-enzymatic PTM of proteins by proteomics approaches.


Asunto(s)
Cisteína/análogos & derivados , Cisteína/metabolismo , Fumaratos/metabolismo , Proteoma/química , Proteoma/metabolismo , Animales , Ciclo del Ácido Cítrico , Cisteína/análisis , Diabetes Mellitus/metabolismo , Humanos , Mitocondrias/metabolismo , Neoplasias/metabolismo , Estrés Oxidativo
18.
Front Oncol ; 2: 85, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22866264

RESUMEN

The drive to understand how altered cellular metabolism and cancer are linked has caused a paradigm shift in the focus of cancer research. The discovery of a mutated metabolic enzyme, isocitrate dehydrogenase 1, that leads to accumulation of the oncometabolite 2-hydroxyglutarate, provided significant direct evidence that dysfunctional metabolism plays an important role in oncogenesis. Striking parallels exist with the Krebs cycle enzyme fumarate hydratase (FH), a tumor suppressor, whose mutation is associated with the development of leiomyomata, renal cysts, and tumors. Loss of FH enzymatic activity results in accumulation of intracellular fumarate which has been proposed to act as a competitive inhibitor of 2-oxoglutarate-dependent oxygenases including the hypoxia-inducible factor (HIF) hydroxylases, thus activating oncogenic HIF pathways. Interestingly, our studies have questioned the role of HIF and have highlighted other candidate mechanisms, in particular the non-enzymatic modification of cysteine residues (succination) that could lead to disruption or loss of protein functions, dysfunctional cell metabolism and cell signaling. Here, we discuss the evidence for proposing fumarate as an onco-metabolite.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA