Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Blood ; 138(17): 1554-1569, 2021 10 28.
Artículo en Inglés | MEDLINE | ID: mdl-34077954

RESUMEN

Trained immunity (TI) is a proinflammatory program induced in monocyte/macrophages upon sensing of specific pathogens and is characterized by immunometabolic and epigenetic changes that enhance cytokine production. Maladaptive activation of TI (ie, in the absence of infection) may result in detrimental inflammation and development of disease; however, the exact role and extent of inappropriate activation of TI in the pathogenesis of human diseases is undetermined. In this study, we uncovered the oncogene-induced, maladaptive induction of TI in the pathogenesis of a human inflammatory myeloid neoplasm (Erdheim-Chester disease, [ECD]), characterized by the BRAFV600E oncogenic mutation in monocyte/macrophages and excess cytokine production. Mechanistically, myeloid cells expressing BRAFV600E exhibit all molecular features of TI: activation of the AKT/mammalian target of rapamycin signaling axis; increased glycolysis, glutaminolysis, and cholesterol synthesis; epigenetic changes on promoters of genes encoding cytokines; and enhanced cytokine production leading to hyperinflammatory responses. In patients with ECD, effective therapeutic strategies combat this maladaptive TI phenotype; in addition, pharmacologic inhibition of immunometabolic changes underlying TI (ie, glycolysis) effectively dampens cytokine production by myeloid cells. This study revealed the deleterious potential of inappropriate activation of TI in the pathogenesis of human inflammatory myeloid neoplasms and the opportunity for inhibition of TI in conditions characterized by maladaptive myeloid-driven inflammation.


Asunto(s)
Enfermedad de Erdheim-Chester/genética , Inflamación/genética , Proteínas Proto-Oncogénicas B-raf/genética , Células Cultivadas , Epigénesis Genética , Enfermedad de Erdheim-Chester/inmunología , Enfermedad de Erdheim-Chester/patología , Humanos , Inmunidad , Inflamación/inmunología , Inflamación/patología , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/patología , Oncogenes , Mutación Puntual , Proteínas Proto-Oncogénicas B-raf/inmunología
2.
FASEB J ; 34(5): 6493-6507, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32239723

RESUMEN

Autosomal dominant polycystic kidney disease (ADPKD) is a common monogenic disorder, characterized by bilateral renal cyst formation. Multiple pathways are de-regulated in cystic epithelia offering good opportunities for therapy. Others and we have previously reported that metabolic reprogramming, including alterations of the TCA cycle, are prominent features of ADPKD. Several lines of evidence suggest that mitochondrial impairment might be responsible for the metabolic alterations. Here, we performed morphologic and morphometric evaluation of mitochondria by TEM in an orthologous mouse model of PKD caused by mutations in the Pkd1 gene (Ksp-Cre;Pkd1flox/- ). Furthermore, we measured mitochondrial respiration by COX and SDH enzymatic activity in situ. We found several alterations including reduced mitochondrial mass, altered structure and fragmentation of the mitochondrial network in cystic epithelia of Ksp-Cre;Pkd1flox/- mice. At the molecular level, we found reduced expression of the pro-fusion proteins OPA1 and MFN1 and up-regulation of the pro-fission protein DRP1. Importantly, administration of Mdivi-1, which interferes with DRP1 rescuing mitochondrial fragmentation, significantly reduced kidney/body weight, cyst formation, and improved renal function in Ksp-Cre;Pkd1flox/- mice. Our data indicate that impaired mitochondrial structure and function play a role in disease progression, and that their improvement can significantly modify the course of the disease.


Asunto(s)
Quistes/patología , Modelos Animales de Enfermedad , Mitocondrias/patología , Enfermedades Renales Poliquísticas/patología , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora/fisiología , Animales , Proliferación Celular , Quistes/genética , Quistes/metabolismo , Progresión de la Enfermedad , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/genética , Mitocondrias/metabolismo , Enfermedades Renales Poliquísticas/genética , Enfermedades Renales Poliquísticas/metabolismo
3.
Hum Mol Genet ; 21(17): 3858-70, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-22678058

RESUMEN

The mitochondrial protein AFG3L2 forms homo-oligomeric and hetero-oligomeric complexes with paraplegin in the inner mitochondrial membrane, named m-AAA proteases. These complexes are in charge of quality control of misfolded proteins and participate in the regulation of OPA1 proteolytic cleavage, required for mitochondrial fusion. Mutations in AFG3L2 cause spinocerebellar ataxia type 28 and a complex neurodegenerative syndrome of childhood. In this study, we demonstrated that the loss of AFG3L2 in mouse embryonic fibroblasts (MEFs) reduces mitochondrial Ca(2+) uptake capacity. This defect is neither a consequence of global alteration in cellular Ca(2+) homeostasis nor of the reduced driving force for Ca(2+) internalization within mitochondria, since cytosolic Ca(2+) transients and mitochondrial membrane potential remain unaffected. Moreover, experiments in permeabilized cells revealed unaltered mitochondrial Ca(2+) uptake speed in Afg3l2(-/-) cells, indicating the presence of functional Ca(2+) uptake machinery. Our results show that the defective Ca(2+) handling in Afg3l2(-/-) cells is caused by fragmentation of the mitochondrial network, secondary to respiratory dysfunction and the consequent processing of OPA1. This leaves a number of mitochondria devoid of connections to the ER and thus without Ca(2+) elevations, hampering the proper Ca(2+) diffusion along the mitochondrial network. The recovery of mitochondrial fragmentation in Afg3l2(-/-) MEFs by overexpression of OPA1 rescues the impaired mitochondrial Ca(2+) buffering, but fails to restore respiration. By linking mitochondrial morphology and Ca(2+) homeostasis, these findings shed new light in the molecular mechanisms underlining neurodegeneration caused by AFG3L2 mutations.


Asunto(s)
Proteasas ATP-Dependientes/deficiencia , Proteasas ATP-Dependientes/metabolismo , Calcio/metabolismo , Retículo Endoplásmico/metabolismo , Mitocondrias/metabolismo , ATPasas Asociadas con Actividades Celulares Diversas , Animales , Respiración de la Célula , Embrión de Mamíferos/citología , Fibroblastos/metabolismo , Fibroblastos/patología , GTP Fosfohidrolasas/metabolismo , Potencial de la Membrana Mitocondrial , Ratones , Modelos Biológicos
4.
EMBO Mol Med ; 16(6): 1379-1403, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38684863

RESUMEN

Polycystic kidney disease (PKD) is a genetic disorder characterized by bilateral cyst formation. We showed that PKD cells and kidneys display metabolic alterations, including the Warburg effect and glutaminolysis, sustained in vitro by the enzyme asparagine synthetase (ASNS). Here, we used antisense oligonucleotides (ASO) against Asns in orthologous and slowly progressive PKD murine models and show that treatment leads to a drastic reduction of total kidney volume (measured by MRI) and a prominent rescue of renal function in the mouse. Mechanistically, the upregulation of an ATF4-ASNS axis in PKD is driven by the amino acid response (AAR) branch of the integrated stress response (ISR). Metabolic profiling of PKD or control kidneys treated with Asns-ASO or Scr-ASO revealed major changes in the mutants, several of which are rescued by Asns silencing in vivo. Indeed, ASNS drives glutamine-dependent de novo pyrimidine synthesis and proliferation in cystic epithelia. Notably, while several metabolic pathways were completely corrected by Asns-ASO, glycolysis was only partially restored. Accordingly, combining the glycolytic inhibitor 2DG with Asns-ASO further improved efficacy. Our studies identify a new therapeutic target and novel metabolic vulnerabilities in PKD.


Asunto(s)
Aspartatoamoníaco Ligasa , Modelos Animales de Enfermedad , Enfermedades Renales Poliquísticas , Animales , Humanos , Ratones , Aspartatoamoníaco Ligasa/metabolismo , Aspartatoamoníaco Ligasa/genética , Aspartatoamoníaco Ligasa/antagonistas & inhibidores , Progresión de la Enfermedad , Riñón/patología , Riñón/metabolismo , Oligonucleótidos Antisentido/farmacología , Oligonucleótidos Antisentido/uso terapéutico , Enfermedades Renales Poliquísticas/metabolismo , Enfermedades Renales Poliquísticas/tratamiento farmacológico , Enfermedades Renales Poliquísticas/patología , Enfermedades Renales Poliquísticas/genética
5.
Nat Metab ; 5(3): 385-397, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36879119

RESUMEN

Depriving cells of nutrients triggers an energetic crisis, which is resolved by metabolic rewiring and organelle reorganization. Primary cilia are microtubule-based organelles at the cell surface, capable of integrating multiple metabolic and signalling cues, but their precise sensory function is not fully understood. Here we show that primary cilia respond to nutrient availability and adjust their length via glutamine-mediated anaplerosis facilitated by asparagine synthetase (ASNS). Nutrient deprivation causes cilia elongation, mediated by reduced mitochondrial function, ATP availability and AMPK activation independently of mTORC1. Of note, glutamine removal and replenishment is necessary and sufficient to induce ciliary elongation or retraction, respectively, under nutrient stress conditions both in vivo and in vitro by restoring mitochondrial anaplerosis via ASNS-dependent glutamate generation. Ift88-mutant cells lacking cilia show reduced glutamine-dependent mitochondrial anaplerosis during metabolic stress, due to reduced expression and activity of ASNS at the base of cilia. Our data indicate a role for cilia in responding to, and possibly sensing, cellular glutamine levels via ASNS during metabolic stress.


Asunto(s)
Aspartatoamoníaco Ligasa , Glutamina , Glutamina/metabolismo , Aspartatoamoníaco Ligasa/metabolismo , Cilios/metabolismo , Transducción de Señal
6.
EMBO Mol Med ; 15(6): e16910, 2023 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-37158102

RESUMEN

MYC is a key oncogenic driver in multiple tumor types, but concomitantly endows cancer cells with a series of vulnerabilities that provide opportunities for targeted pharmacological intervention. For example, drugs that suppress mitochondrial respiration selectively kill MYC-overexpressing cells. Here, we unravel the mechanistic basis for this synthetic lethal interaction and exploit it to improve the anticancer effects of the respiratory complex I inhibitor IACS-010759. In a B-lymphoid cell line, ectopic MYC activity and treatment with IACS-010759 added up to induce oxidative stress, with consequent depletion of reduced glutathione and lethal disruption of redox homeostasis. This effect could be enhanced either with inhibitors of NADPH production through the pentose phosphate pathway, or with ascorbate (vitamin C), known to act as a pro-oxidant at high doses. In these conditions, ascorbate synergized with IACS-010759 to kill MYC-overexpressing cells in vitro and reinforced its therapeutic action against human B-cell lymphoma xenografts. Hence, complex I inhibition and high-dose ascorbate might improve the outcome of patients affected by high-grade lymphomas and potentially other MYC-driven cancers.


Asunto(s)
Linfoma de Células B , Linfoma , Humanos , Línea Celular Tumoral , Linfoma/tratamiento farmacológico , Linfoma/metabolismo , Linfoma/patología , Linfoma de Células B/tratamiento farmacológico , Estrés Oxidativo , Proteínas Proto-Oncogénicas c-myc/metabolismo
7.
Nat Commun ; 14(1): 6513, 2023 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-37845212

RESUMEN

Fibrocystin/Polyductin (FPC), encoded by PKHD1, is associated with autosomal recessive polycystic kidney disease (ARPKD), yet its precise role in cystogenesis remains unclear. Here we show that FPC undergoes complex proteolytic processing in developing kidneys, generating three soluble C-terminal fragments (ICDs). Notably, ICD15, contains a novel mitochondrial targeting sequence at its N-terminus, facilitating its translocation into mitochondria. This enhances mitochondrial respiration in renal epithelial cells, partially restoring impaired mitochondrial function caused by FPC loss. FPC inactivation leads to abnormal ultrastructural morphology of mitochondria in kidney tubules without cyst formation. Moreover, FPC inactivation significantly exacerbates renal cystogenesis and triggers severe pancreatic cystogenesis in a Pkd1 mouse mutant Pkd1V/V in which cleavage of Pkd1-encoded Polycystin-1 at the GPCR Proteolysis Site is blocked. Deleting ICD15 enhances renal cystogenesis without inducing pancreatic cysts in Pkd1V/V mice. These findings reveal a direct link between FPC and a mitochondrial pathway through ICD15 cleavage, crucial for cystogenesis mechanisms.


Asunto(s)
Quiste Pancreático , Riñón Poliquístico Autosómico Recesivo , Ratones , Animales , Receptores de Superficie Celular/metabolismo , Riñón/metabolismo , Riñón Poliquístico Autosómico Recesivo/metabolismo , Canales Catiónicos TRPP/genética , Canales Catiónicos TRPP/metabolismo , Túbulos Renales/metabolismo
8.
Mol Oncol ; 16(5): 1132-1152, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34632715

RESUMEN

Multiple molecular features, such as activation of specific oncogenes (e.g., MYC, BCL2) or a variety of gene expression signatures, have been associated with disease course in diffuse large B-cell lymphoma (DLBCL), although their relationships and implications for targeted therapy remain to be fully unraveled. We report that MYC activity is closely correlated with-and most likely a driver of-gene signatures related to oxidative phosphorylation (OxPhos) in DLBCL, pointing to OxPhos enzymes, in particular mitochondrial electron transport chain (ETC) complexes, as possible therapeutic targets in high-grade MYC-associated lymphomas. In our experiments, indeed, MYC sensitized B cells to the ETC complex I inhibitor IACS-010759. Mechanistically, IACS-010759 triggered the integrated stress response (ISR) pathway, driven by the transcription factors ATF4 and CHOP, which engaged the intrinsic apoptosis pathway and lowered the apoptotic threshold in MYC-overexpressing cells. In line with these findings, the BCL2-inhibitory compound venetoclax synergized with IACS-010759 against double-hit lymphoma (DHL), a high-grade malignancy with concurrent activation of MYC and BCL2. In BCL2-negative lymphoma cells, instead, killing by IACS-010759 was potentiated by the Mcl-1 inhibitor S63845. Thus, combining an OxPhos inhibitor with select BH3-mimetic drugs provides a novel therapeutic principle against aggressive, MYC-associated DLBCL variants.


Asunto(s)
Linfoma de Células B Grandes Difuso , Proteínas Proto-Oncogénicas c-myc , Humanos , Linfoma de Células B Grandes Difuso/tratamiento farmacológico , Linfoma de Células B Grandes Difuso/genética , Oncogenes , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Respiración
9.
Biochim Biophys Acta Mol Basis Dis ; 1868(6): 166371, 2022 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-35218894

RESUMEN

Cardiovascular manifestations account for marked morbi-mortality in autosomal dominant polycystic kidney disease (ADPKD). Pkd1- and Pkd2-deficient mice develop cardiac dysfunction, however the underlying mechanisms remain largely unclear. It is unknown whether impairment of polycystin-1 cleavage at the G-protein-coupled receptor proteolysis site, a significant ADPKD mutational mechanism, is involved in this process. We analyzed the impact of polycystin-1 cleavage on heart metabolism using Pkd1V/V mice, a model unable to cleave this protein and with early cardiac dysfunction. Pkd1V/V hearts showed lower levels of glucose and amino acids and higher lipid levels than wild-types, as well as downregulation of p-AMPK, p-ACCß, CPT1B-Cpt1b, Ppara, Nppa and Acta1. These findings suggested decreased fatty acid ß-oxidation, which was confirmed by lower oxygen consumption by Pkd1V/V isolated mitochondria using palmitoyl-CoA. Pkd1V/V hearts also presented increased oxygen consumption in response to glucose, suggesting that alternative substrates may be used to generate energy. Pkd1V/V hearts displayed a higher density of decreased-size mitochondria, a finding associated with lower MFN1, Parkin and BNIP3 expression. These derangements were correlated with increased apoptosis and inflammation but not hypertrophy. Notably, Pkd1V/V neonate cardiomyocytes also displayed shifts in oxygen consumption and p-AMPK downregulation, suggesting that, at least partially, the metabolic alterations are not induced by kidney dysfunction. Our findings reveal that disruption of polycystin-1 cleavage leads to cardiac metabolic rewiring in mice, expanding the understanding of heart dysfunction associated with Pkd1 deficiency and likely with human ADPKD.


Asunto(s)
Riñón Poliquístico Autosómico Dominante , Canales Catiónicos TRPP , Animales , Corazón , Ratones , Mitocondrias/metabolismo , Mutación , Riñón Poliquístico Autosómico Dominante/genética , Riñón Poliquístico Autosómico Dominante/metabolismo , Canales Catiónicos TRPP/genética , Canales Catiónicos TRPP/metabolismo
10.
Cell Rep ; 35(1): 108955, 2021 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-33826894

RESUMEN

Trained immunity (TI) is a de facto innate immune memory program induced in monocytes/macrophages by exposure to pathogens or vaccines, which evolved as protection against infections. TI is characterized by immunometabolic changes and histone post-translational modifications, which enhance production of pro-inflammatory cytokines. As aberrant activation of TI is implicated in inflammatory diseases, tight regulation is critical; however, the mechanisms responsible for this modulation remain elusive. Interleukin-37 (IL-37) is an anti-inflammatory cytokine that curbs inflammation and modulates metabolic pathways. In this study, we show that administration of recombinant IL-37 abrogates the protective effects of TI in vivo, as revealed by reduced host pro-inflammatory responses and survival to disseminated candidiasis. Mechanistically, IL-37 reverses the immunometabolic changes and histone post-translational modifications characteristic of TI in monocytes, thus suppressing cytokine production in response to infection. IL-37 thereby emerges as an inhibitor of TI and as a potential therapeutic target in immune-mediated pathologies.


Asunto(s)
Antiinflamatorios/farmacología , Inmunidad Innata , Interleucina-1/farmacología , Animales , Candidiasis/genética , Candidiasis/inmunología , Candidiasis/microbiología , Epigénesis Genética/efectos de los fármacos , Glucólisis/efectos de los fármacos , Glucólisis/genética , Interacciones Huésped-Patógeno/efectos de los fármacos , Interacciones Huésped-Patógeno/inmunología , Humanos , Inmunidad Innata/efectos de los fármacos , Masculino , Ratones Endogámicos C57BL , Neutrófilos/efectos de los fármacos , Neutrófilos/metabolismo
11.
Cell Signal ; 67: 109495, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31816397

RESUMEN

Autosomal Dominant Polycystic Kidney Disease (ADPKD) is a slowly progressive disease characterized by the relentless growth of renal cysts throughout the life of affected individuals. Early evidence suggested that the epithelia lining the cysts share neoplastic features, leading to the definition of PKD as a "neoplasm in disguise". Recent work from our and other laboratories has identified a profound metabolic reprogramming in PKD, similar to the one reported in cancer and consistent with the reported increased proliferation. Multiple lines of evidence suggest that aerobic glycolysis (a Warburg-like effect) is present in the disease, along with other metabolic dysfunctions such as an increase in the pentose phosphate pathway, in glutamine anaplerosis and fatty acid biosynthesis, while fatty acid oxidation and oxidative phosphorylation (OXPHOS) are decreased. In addition to glutamine, other amino acid-related pathways appear altered, including asparagine and arginine. The precise origin of the metabolic alterations is not entirely clear, but two hypotheses can be formulated, not mutually exclusive. First, the polycystins have been recently shown to regulate directly mitochondrial function and structure either by regulating Ca2+ uptake in mitochondria at the Mitochondria Associated Membranes (MAMs) of the Endoplasmic Reticulum, or by a direct translocation of a small fragment of the protein into the matrix of mitochondria. One alternative possibility is that metabolic and mitochondrial dysfunctions in ADPKD are secondary to the de-regulation of proliferation, driven by the multiple signaling pathways identified in the disease, which include mTORC1 and AMPK among the most relevant. While the precise mechanisms underlying these novel alterations identified in ADPKD will need further investigation, it is evident that they offer a great opportunity for novel interventions in the disease.


Asunto(s)
Mitocondrias/metabolismo , Enfermedades Renales Poliquísticas/metabolismo , Animales , Epigénesis Genética , Humanos , Metabolismo de los Lípidos/genética , Modelos Biológicos , Enfermedades Renales Poliquísticas/genética , Enfermedades Renales Poliquísticas/terapia , Transducción de Señal/genética
12.
EBioMedicine ; 61: 103050, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-33045469

RESUMEN

BACKGROUND: Mutations of the mitochondrial protein paraplegin cause hereditary spastic paraplegia type 7 (SPG7), a so-far untreatable degenerative disease of the upper motoneuron with still undefined pathomechanism. The intermittent mitochondrial permeability transition pore (mPTP) opening, called flickering, is an essential process that operates to maintain mitochondrial homeostasis by reducing intra-matrix Ca2+ and reactive oxygen species (ROS) concentration, and is critical for efficient synaptic function. METHODS: We use a fluorescence-based approach to measure mPTP flickering in living cells and biochemical and molecular biology techniques to dissect the pathogenic mechanism of SPG7. In the SPG7 animal model we evaluate the potential improvement of the motor defect, neuroinflammation and neurodegeneration by means of an mPTP inducer, the benzodiazepine Bz-423. FINDINGS: We demonstrate that paraplegin is required for efficient transient opening of the mPTP, that is impaired in both SPG7 patients-derived fibroblasts and primary neurons from Spg7-/- mice. We show that dysregulation of mPTP opening at the pre-synaptic terminal impairs neurotransmitter release leading to ineffective synaptic transmission. Lack of paraplegin impairs mPTP flickering by a mechanism involving increased expression and activity of sirtuin3, which promotes deacetylation of cyclophilin D, thus hampering mPTP opening. Pharmacological treatment with Bz-423, which bypasses the activity of CypD, normalizes synaptic transmission and rescues the motor impairment of the SPG7 mouse model. INTERPRETATION: mPTP targeting opens a new avenue for the potential therapy of this form of spastic paraplegia. FUNDING: Telethon Foundation grant (TGMGCSBX16TT); Dept. of Defense, US Army, grant W81XWH-18-1-0001.


Asunto(s)
ATPasas Asociadas con Actividades Celulares Diversas/genética , Metaloendopeptidasas/genética , Poro de Transición de la Permeabilidad Mitocondrial/metabolismo , Paraplejía Espástica Hereditaria/genética , Paraplejía Espástica Hereditaria/metabolismo , ATPasas Asociadas con Actividades Celulares Diversas/metabolismo , Animales , Apoptosis/genética , Transporte Biológico , Calcio/metabolismo , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Edición Génica , Células HEK293 , Humanos , Potencial de la Membrana Mitocondrial , Metaloendopeptidasas/metabolismo , Ratones , Ratones Noqueados , Mitocondrias/genética , Mitocondrias/metabolismo , Mutación , Neuronas/metabolismo , Fenotipo , Especies Reactivas de Oxígeno/metabolismo , Sirtuina 3/metabolismo , Médula Espinal/metabolismo , Médula Espinal/patología , Vesículas Sinápticas/metabolismo
13.
J Neurosci ; 28(11): 2827-36, 2008 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-18337413

RESUMEN

The mitochondrial metalloprotease AFG3L2 assembles with the homologous protein paraplegin to form a supracomplex in charge of the essential protein quality control within mitochondria. Mutations of paraplegin cause a specific axonal degeneration of the upper motoneuron and, therefore, hereditary spastic paraplegia. Here we present two Afg3l2 murine models: a newly developed null and a spontaneous mutant that we found carrier of a missense mutation. Contrasting with the mild and late onset axonal degeneration of paraplegin-deficient mouse, Afg3l2 models display a marked impairment of axonal development with delayed myelination and poor axonal radial growth leading to lethality at P16. The increased severity of the Afg3l2 mutants is explained by two main molecular features that differentiate AFG3L2 from paraplegin: its higher neuronal expression and its versatile ability to support both hetero-oligomerization and homo-oligomerization. Our data assign to AFG3L2 a crucial role by linking mitochondrial metabolism and axonal development. Moreover, we propose AFG3L2 as an excellent candidate for motoneuron and cerebellar diseases with early onset unknown etiology.


Asunto(s)
Adenosina Trifosfatasas/biosíntesis , Axones/enzimología , Mitocondrias/enzimología , Proteínas Mitocondriales/biosíntesis , Proteasas ATP-Dependientes , ATPasas Asociadas con Actividades Celulares Diversas , Adenosina Trifosfatasas/genética , Secuencia de Aminoácidos , Animales , Animales Recién Nacidos , Axones/patología , Axones/fisiología , Ratones , Ratones Mutantes , Mitocondrias/genética , Mitocondrias/patología , Proteínas Mitocondriales/genética , Datos de Secuencia Molecular
14.
J Cell Biol ; 163(4): 777-87, 2003 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-14623864

RESUMEN

Mmutations in paraplegin, a putative mitochondrial metallopeptidase of the AAA family, cause an autosomal recessive form of hereditary spastic paraplegia (HSP). Here, we analyze the function of paraplegin at the cellular level and characterize the phenotypic defects of HSP patients' cells lacking this protein. We demonstrate that paraplegin coassembles with a homologous protein, AFG3L2, in the mitochondrial inner membrane. These two proteins form a high molecular mass complex, which we show to be aberrant in HSP fibroblasts. The loss of this complex causes a reduced complex I activity in mitochondria and an increased sensitivity to oxidant stress, which can both be rescued by exogenous expression of wild-type paraplegin. Furthermore, complementation studies in yeast demonstrate functional conservation of the human paraplegin-AFG3L2 complex with the yeast m-AAA protease and assign proteolytic activity to this structure. These results shed new light on the molecular pathogenesis of HSP and functionally link AFG3L2 to this neurodegenerative disease.


Asunto(s)
Complejo I de Transporte de Electrón/metabolismo , Metaloendopeptidasas/deficiencia , Metaloendopeptidasas/metabolismo , Mitocondrias/enzimología , Paraplejía Espástica Hereditaria/enzimología , Proteasas ATP-Dependientes , ATPasas Asociadas con Actividades Celulares Diversas , Adenosina Trifosfatasas/genética , Adenosina Trifosfatasas/metabolismo , Respiración de la Célula/genética , Células Cultivadas , Regulación hacia Abajo/genética , Complejo I de Transporte de Electrón/deficiencia , Fibroblastos , Humanos , Membranas Intracelulares/metabolismo , Sustancias Macromoleculares , Metaloendopeptidasas/genética , Estrés Oxidativo/genética , Filogenia , Especies Reactivas de Oxígeno/farmacología , Saccharomyces cerevisiae/enzimología , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Homología de Secuencia de Ácido Nucleico , Paraplejía Espástica Hereditaria/genética
16.
Cardiovasc Res ; 101(3): 492-502, 2014 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-24302629

RESUMEN

AIMS: Nitric oxide (NO) plays a key role in vascular homeostasis and is produced by endothelial NO synthase (eNOS), encoded by NOS3 gene. We previously reported the genetic association between NOS3 rs753482-A>C polymorphism on intron 19 and coronary artery disease (CAD). In the attempt of conferring functional implication to the rs753482-A>C polymorphism, we investigated its influence on transcript maturation. METHODS AND RESULTS: A transcript variant skipping exons 20-21 is prevalent in carriers of the rs753482-C allele and is translated in a novel truncated form of eNOS. The truncated eNOS displays increased basal NO production, is insensitive to calcium stimulation, and, upon heterodimerization with the full-length eNOS protein, exerts a dominant-negative effect on NO production. CAD patients and healthy subjects' carriers of the rs753482-C genotype are characterized by increased NO basal levels in peripheral blood and platelets, and negatively respond to oral glucose load by failing to increase NO synthesis following insulin wave. Furthermore, forearm vasodilation after reactive hyperaemia is dramatically impaired in rs753482-C carriers. CONCLUSIONS: We demonstrated that subjects carrying the rs753482-C genotype express a novel stable truncated form of eNOS with altered enzymatic activity that influences NO production and endothelial function. These findings open to new intriguing perspectives to several diseases involving vascular response to NO.


Asunto(s)
Enfermedad de la Arteria Coronaria/genética , Endotelio Vascular/enzimología , Frecuencia de los Genes/genética , Óxido Nítrico Sintasa de Tipo III/genética , Polimorfismo Genético/genética , Adulto , Anciano , Enfermedad de la Arteria Coronaria/enzimología , Frecuencia de los Genes/fisiología , Predisposición Genética a la Enfermedad , Pruebas Genéticas/métodos , Genotipo , Humanos , Persona de Mediana Edad , Vasodilatación/fisiología
17.
Am J Physiol Endocrinol Metab ; 294(5): E978-86, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18349107

RESUMEN

Little is known about the association of endothelial nitric oxide synthase (NOS3) gene polymorphisms and the presence of insulin resistance and the early evolution of atherosclerosis in nondiabetic subjects with cardiovascular disease (CAD) and stent implantation. The present study was performed in an attempt to better understand whether metabolic, endothelial, and angiographic findings characteristic of subjects with cardiovascular disease and in-stent restenosis are related to NOS3 variants. This is a case-control study performed from 2002 to 2006. All subjects admitted to the study were recruited in the Nord-Centre of Italy, most from Milan and its surrounding towns. Measures of glucose tolerance, insulin sensitivity, markers of endothelial dysfunction, forearm vasodilation, and adipokine levels were determined and associated to the frequency of two single-nucleotide polymorphisms of NOS3, i.e., Glu298Asp (rs1799983, G/T) and rs753482 (intron 18 A/C). A total of 747 subjects, not known to have diabetes, were evaluated: 333 subjects had asymptomatic CAD, 106 subjects had unstable angina and were evaluated for in-stent restenosis 6 mo after stent placement, and 308 were control subjects. The presence of TT and CC minor alleles was significantly greater in case groups compared with control subjects. At phenotypic level, subjects with the polymorphisms were characterized by hyperinsulinemia and reduced reactive hyperemia, whereas increased leptin and decreased adiponectin levels were present in subjects with restenosis in the presence of reduced minimal lumen diameter and length of stenosis almost doubled. Hyperinsulinemia, endothelial dysfunction, and a more atherogenic profile seem to be peculiar features of subjects with asymptomatic CAD and restenosis carrying NOS3 gene variants.


Asunto(s)
Adiponectina/metabolismo , Oclusión de Injerto Vascular/genética , Hiperinsulinismo/metabolismo , Leptina/metabolismo , Óxido Nítrico Sintasa de Tipo III/genética , Anciano , Aterosclerosis/genética , Aterosclerosis/patología , Glucemia/metabolismo , Angiografía Coronaria , Enfermedad de la Arteria Coronaria/genética , Enfermedad de la Arteria Coronaria/metabolismo , ADN/biosíntesis , ADN/genética , Complicaciones de la Diabetes/patología , Femenino , Antebrazo/irrigación sanguínea , Frecuencia de los Genes , Genotipo , Prueba de Tolerancia a la Glucosa , Hemoglobina Glucada/metabolismo , Haplotipos , Humanos , Lípidos/sangre , Masculino , Persona de Mediana Edad , Polimorfismo Genético/genética , Polimorfismo Genético/fisiología , Flujo Sanguíneo Regional/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA