Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Hepatology ; 2024 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-38975812

RESUMEN

BACKGROUND AND AIMS: Antimicrobial proteins of the REG3 family provide a first line of protection against infections and transformed cells. Their expression is inducible by inflammation, which makes their role in cancer biology less clear, since an immune- inflammatory context may preexist or coexist with cancer, as occurs in hepatocellular carcinoma (HCC). The aim of this study is to clarify the role of REG3A in liver carcinogenesis and to determine whether carbohydrate-binding functions are involved. APPROACH AND RESULTS: This study provides evidence of the suppressive role of REG3A in HCC by reducing O-GlcNAcylation in two mouse models of HCC, in vitro cell studies, and in clinical samples. REG3A expression in hepatocytes significantly reduces global O- GlcNAcylation and O-GlcNAcylation of c-MYC in preneoplastic and tumor livers and markedly inhibits HCC development in REG3A-c-MYC double transgenic mice and in mice exposed to diethylnitrosamine (DEN). REG3A modifies O-GlcNAcylation without altering the expression or activity of OGT, OGA, or GFAT. Reduced O-GlcNAcylation was consistent with decreased levels of UDP-GlcNAc in pre-cancerous and cancerous livers. This effect is linked to the ability of REG3A to bind Glc and Glc-6P, suggested by a REG3A mutant unable to bind Glc and Glc- 6P and alter O-GlcNAcylation. Importantly, cirrhotic patients with high hepatic REG3A expression had lower levels of O-GlcNAcylation and longer cancer-free survival than REG3A- negative cirrhotic livers. CONCLUSION: REG3A helps fight liver cancer by reducing O-GlcNAcylation. This study suggests a new paradigm for the regulation of O-GlcNAc signalling in cancer-related pathways through interactions with the carbohydrate-binding function of REG3A.

2.
STAR Protoc ; 5(1): 102817, 2024 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-38183655

RESUMEN

Uridine diphosphate N-acetylglucosamine (UDP-GlcNAc) is the end product of the hexosamine biosynthetic pathway and the substrate for protein O-linked N-acetylglucosaminylation (O-GlcNAcylation). Here, we present a protocol for the quantification of UDP-GlcNAc using an enzymatic microplate assay. We also detail procedures for the extraction of polar metabolites and total protein fraction for the parallel quantification of UDP-GlcNAc and the western blot analysis of O-GlcNAcylated proteins, O-linked N-acetylglucosamine transferase, and O-GlcNAcase from the same sample. For complete details on the use and execution of this protocol, please refer to Sunden et al. (2023).1 In addition, a preview article by Chatham et al. provides a useful summary of the method.2.


Asunto(s)
Proteínas , Uridina Difosfato
3.
Nucleic Acids Res ; 52(1): e6, 2024 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-38008466

RESUMEN

Enzymatic methods to quantify deoxyribonucleoside triphosphates have existed for decades. In contrast, no general enzymatic method to quantify ribonucleoside triphosphates (rNTPs), which drive almost all cellular processes and serve as precursors of RNA, exists to date. ATP can be measured with an enzymatic luminometric method employing firefly luciferase, but the quantification of other ribonucleoside mono-, di-, and triphosphates is still a challenge for a non-specialized laboratory and practically impossible without chromatography equipment. To allow feasible quantification of ribonucleoside phosphates in any laboratory with typical molecular biology and biochemistry tools, we developed a robust microplate assay based on real-time detection of the Broccoli RNA aptamer during in vitro transcription. The assay employs the bacteriophage T7 and SP6 RNA polymerases, two oligonucleotide templates encoding the 49-nucleotide Broccoli aptamer, and a high-affinity fluorogenic aptamer-binding dye to quantify each of the four canonical rNTPs. The inclusion of nucleoside mono- and diphosphate kinases in the assay reactions enabled the quantification of the mono- and diphosphate counterparts. The assay is inherently specific and tolerates concentrated tissue and cell extracts. In summary, we describe the first chromatography-free method to quantify ATP, ADP, AMP, GTP, GDP, GMP, UTP, UDP, UMP, CTP, CDP and CMP in biological samples.


Asunto(s)
Bioquímica , Ribonucleótidos , Difosfatos , Nucleótidos/química , Ribonucleótidos/análisis , Bioquímica/métodos
4.
Front Cell Dev Biol ; 11: 1257651, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37731815

RESUMEN

The mitochondrion is a major hub of cellular metabolism and involved directly or indirectly in almost all biological processes of the cell. In mitochondrial diseases, compromised respiratory electron transfer and oxidative phosphorylation (OXPHOS) lead to compensatory rewiring of metabolism with resemblance to the Warburg-like metabolic state of cancer cells. The transcription factor MYC (or c-MYC) is a major regulator of metabolic rewiring in cancer, stimulating glycolysis, nucleotide biosynthesis, and glutamine utilization, which are known or predicted to be affected also in mitochondrial diseases. Albeit not widely acknowledged thus far, several cell and mouse models of mitochondrial disease show upregulation of MYC and/or its typical transcriptional signatures. Moreover, gene expression and metabolite-level changes associated with mitochondrial integrated stress response (mt-ISR) show remarkable overlap with those of MYC overexpression. In addition to being a metabolic regulator, MYC promotes cellular proliferation and modifies the cell cycle kinetics and, especially at high expression levels, promotes replication stress and genomic instability, and sensitizes cells to apoptosis. Because cell proliferation requires energy and doubling of the cellular biomass, replicating cells should be particularly sensitive to defective OXPHOS. On the other hand, OXPHOS-defective replicating cells are predicted to be especially vulnerable to high levels of MYC as it facilitates evasion of metabolic checkpoints and accelerates cell cycle progression. Indeed, a few recent studies demonstrate cell cycle defects and nuclear DNA damage in OXPHOS deficiency. Here, we give an overview of key mitochondria-dependent metabolic pathways known to be regulated by MYC, review the current literature on MYC expression in mitochondrial diseases, and speculate how its upregulation may be triggered by OXPHOS deficiency and what implications this has for the pathogenesis of these diseases.

5.
Cell Rep Methods ; 3(7): 100518, 2023 07 24.
Artículo en Inglés | MEDLINE | ID: mdl-37533645

RESUMEN

O-linked N-acetylglucosaminylation (O-GlcNAcylation) is a ubiquitous and dynamic non-canonical glycosylation of intracellular proteins. Several branches of metabolism converge at the hexosamine biosynthetic pathway (HBP) to produce the substrate for protein O-GlcNAcylation, the uridine diphosphate N-acetylglucosamine (UDP-GlcNAc). Availability of UDP-GlcNAc is considered a key regulator of O-GlcNAcylation. Yet UDP-GlcNAc concentrations are rarely reported in studies exploring the HBP and O-GlcNAcylation, most likely because the methods to measure it are restricted to specialized chromatographic procedures. Here, we introduce an enzymatic method to quantify cellular and tissue UDP-GlcNAc. The method is based on O-GlcNAcylation of a substrate peptide by O-linked N-acetylglucosamine transferase (OGT) and subsequent immunodetection of the modification. The assay can be performed in dot-blot or microplate format. We apply it to quantify UDP-GlcNAc concentrations in several mouse tissues and cell lines. Furthermore, we show how changes in UDP-GlcNAc levels correlate with O-GlcNAcylation and the expression of OGT and O-GlcNAcase (OGA).


Asunto(s)
Pruebas de Enzimas , Proteínas , Ratones , Animales , Glicosilación , Uridina Difosfato
6.
Nat Commun ; 14(1): 2356, 2023 04 24.
Artículo en Inglés | MEDLINE | ID: mdl-37095097

RESUMEN

Accumulating evidence suggests mitochondria as key modulators of normal and premature aging, yet whether primary oxidative phosphorylation (OXPHOS) deficiency can cause progeroid disease remains unclear. Here, we show that mice with severe isolated respiratory complex III (CIII) deficiency display nuclear DNA damage, cell cycle arrest, aberrant mitoses, and cellular senescence in the affected organs such as liver and kidney, and a systemic phenotype resembling juvenile-onset progeroid syndromes. Mechanistically, CIII deficiency triggers presymptomatic cancer-like c-MYC upregulation followed by excessive anabolic metabolism and illicit cell proliferation against lack of energy and biosynthetic precursors. Transgenic alternative oxidase dampens mitochondrial integrated stress response and the c-MYC induction, suppresses the illicit proliferation, and prevents juvenile lethality despite that canonical OXPHOS-linked functions remain uncorrected. Inhibition of c-MYC with the dominant-negative Omomyc protein relieves the DNA damage in CIII-deficient hepatocytes in vivo. Our results connect primary OXPHOS deficiency to genomic instability and progeroid pathogenesis and suggest that targeting c-MYC and aberrant cell proliferation may be therapeutic in mitochondrial diseases.


Asunto(s)
Enfermedades Mitocondriales , Progeria , Ratones , Animales , Progeria/patología , Complejo III de Transporte de Electrones , Senescencia Celular/genética , Ciclo Celular
7.
Dis Model Mech ; 15(10)2022 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-36285626

RESUMEN

Isolated populations have been valuable for the discovery of rare monogenic diseases and their causative genetic variants. Finnish disease heritage (FDH) is an example of a group of hereditary monogenic disorders caused by single major, usually autosomal-recessive, variants enriched in the population due to several past genetic drift events. Interestingly, distinct subpopulations have remained in Finland and have maintained their unique genetic repertoire. Thus, FDH diseases have persisted, facilitating vigorous research on the underlying molecular mechanisms and development of treatment options. This Review summarizes the current status of FDH, including the most recently discovered FDH disorders, and introduces a set of other recently identified diseases that share common features with the traditional FDH diseases. The Review also discusses a new era for population-based studies, which combine various forms of big data to identify novel genotype-phenotype associations behind more complex conditions, as exemplified here by the FinnGen project. In addition to the pathogenic variants with an unequivocal causative role in the disease phenotype, several risk alleles that correlate with certain phenotypic features have been identified among the Finns, further emphasizing the broad value of studying genetically isolated populations.


Asunto(s)
Investigación Biomédica Traslacional , Finlandia/epidemiología , Fenotipo
8.
FEBS J ; 289(22): 6936-6958, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-34428349

RESUMEN

Coenzyme Q (CoQ, ubiquinone) is the electron-carrying lipid in the mitochondrial electron transport system (ETS). In mammals, it serves as the electron acceptor for nine mitochondrial inner membrane dehydrogenases. These include the NADH dehydrogenase (complex I, CI) and succinate dehydrogenase (complex II, CII) but also several others that are often omitted in the context of respiratory enzymes: dihydroorotate dehydrogenase, choline dehydrogenase, electron-transferring flavoprotein dehydrogenase, mitochondrial glycerol-3-phosphate dehydrogenase, proline dehydrogenases 1 and 2, and sulfide:quinone oxidoreductase. The metabolic pathways these enzymes are involved in range from amino acid and fatty acid oxidation to nucleotide biosynthesis, methylation, and hydrogen sulfide detoxification, among many others. The CoQ-linked metabolism depends on CoQ reoxidation by the mitochondrial complex III (cytochrome bc1 complex, CIII). However, the literature is surprisingly limited as for the role of the CoQ-linked metabolism in the pathogenesis of human diseases of oxidative phosphorylation (OXPHOS), in which the CoQ homeostasis is directly or indirectly affected. In this review, we give an introduction to CIII function, and an overview of the pathological consequences of CIII dysfunction in humans and mice and of the CoQ-dependent metabolic processes potentially affected in these pathological states. Finally, we discuss some experimental tools to dissect the various aspects of compromised CoQ oxidation.


Asunto(s)
Complejo III de Transporte de Electrones , Ubiquinona , Humanos , Ratones , Animales , Membranas Mitocondriales/metabolismo , Mitocondrias/metabolismo , Oxidación-Reducción , Mamíferos/metabolismo
9.
Biochim Biophys Acta Mol Basis Dis ; 1868(1): 166298, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34751152

RESUMEN

In the diagnostic work-up of a newborn infant with a metabolic crisis, lethal multiorgan failure on day six of life, and increased excretion of 3-methylglutaconic acid, we found using whole genome sequencing a homozygous SERAC1 mutation indicating MEGDHEL syndrome (3-methylglutaconic aciduria with deafness-dystonia, hepatopathy, encephalopathy, and Leigh-like syndrome). The SERAC1 protein is located at the contact site between mitochondria and the endoplasmic reticulum (ER) and is crucial for cholesterol trafficking. Our aim was to investigate the effect of the homozygous truncating mutation on mitochondrial structure and function. In the patient fibroblasts, no SERAC1 protein was detected, the mitochondrial network was severely fragmented, and the cristae morphology was altered. Filipin staining showed uneven localization of unesterified cholesterol. The calcium buffer function between cytoplasm and mitochondria was deficient. In liver mitochondria, complexes I, III, and IV were clearly decreased. In transfected COS-1 cells the mutant protein with the a 45-amino acid C-terminal truncation was distributed throughout the cell, whereas wild-type SERAC1 partially colocalized with the mitochondrial marker MT-CO1. The structural and functional mitochondrial abnormalities, caused by the loss of SERAC1, suggest that the crucial disease mechanism is disrupted interplay between the ER and mitochondria leading to decreased influx of calcium to mitochondria and secondary respiratory chain deficiency.


Asunto(s)
Hidrolasas de Éster Carboxílico/genética , Errores Innatos del Metabolismo/genética , Mitocondrias Hepáticas/genética , Enfermedades Mitocondriales/genética , Calcio/metabolismo , Colesterol/metabolismo , Retículo Endoplásmico/genética , Retículo Endoplásmico/metabolismo , Femenino , Glutaratos/metabolismo , Humanos , Recién Nacido , Masculino , Errores Innatos del Metabolismo/metabolismo , Errores Innatos del Metabolismo/patología , Mitocondrias Hepáticas/metabolismo , Mitocondrias Hepáticas/patología , Enfermedades Mitocondriales/metabolismo , Enfermedades Mitocondriales/patología , Secuenciación Completa del Genoma
10.
Nucleic Acids Res ; 48(15): e87, 2020 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-32573728

RESUMEN

Deoxyribonucleoside triphosphates (dNTPs) are vital for the biosynthesis and repair of DNA. Their cellular concentration peaks during the S phase of the cell cycle. In non-proliferating cells, dNTP concentrations are low, making their reliable quantification from tissue samples of heterogeneous cellular composition challenging. Partly because of this, the current knowledge related to the regulation of and disturbances in cellular dNTP concentrations derive mostly from cell culture experiments with little corroboration at the tissue or organismal level. Here, we fill the methodological gap by presenting a simple non-radioactive microplate assay for the quantification of dNTPs with a minimum requirement of 4-12 mg of biopsy material. In contrast to published assays, this assay is based on long synthetic single-stranded DNA templates (50-200 nucleotides), an inhibitor-resistant high-fidelity DNA polymerase, and the double-stranded-DNA-binding EvaGreen dye. The assay quantified reliably less than 50 fmol of each of the four dNTPs and discriminated well against ribonucleotides. Additionally, thermostable RNAse HII-mediated nicking of the reaction products and a subsequent shift in their melting temperature allowed near-complete elimination of the interfering ribonucleotide signal, if present. Importantly, the assay allowed measurement of minute dNTP concentrations in mouse liver, heart and skeletal muscle.


Asunto(s)
ADN Polimerasa Dirigida por ADN/genética , Desoxirribonucleótidos/aislamiento & purificación , Oligonucleótidos/genética , Animales , ADN de Cadena Simple/genética , ADN Polimerasa Dirigida por ADN/química , Desoxirribonucleótidos/genética , Ratones , Inhibidores de la Síntesis del Ácido Nucleico/química , Oligonucleótidos/síntesis química , Ribonucleasa H/genética
11.
Nat Commun ; 11(1): 322, 2020 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-31949167

RESUMEN

We previously observed an unexpected fivefold (35 vs. 200 days) difference in the survival of respiratory chain complex III (CIII) deficient Bcs1lp.S78G mice between two congenic backgrounds. Here, we identify a spontaneous homoplasmic mtDNA variant (m.G14904A, mt-Cybp.D254N), affecting the CIII subunit cytochrome b (MT-CYB), in the background with short survival. We utilize maternal inheritance of mtDNA to confirm this as the causative variant and show that it further decreases the low CIII activity in Bcs1lp.S78G tissues to below survival threshold by 35 days of age. Molecular dynamics simulations predict D254N to restrict the flexibility of MT-CYB ef loop, potentially affecting RISP dynamics. In Rhodobacter cytochrome bc1 complex the equivalent substitution causes a kinetics defect with longer occupancy of RISP head domain towards the quinol oxidation site. These findings represent a unique case of spontaneous mitonuclear epistasis and highlight the role of mtDNA variation as modifier of mitochondrial disease phenotypes.


Asunto(s)
Complejo III de Transporte de Electrones/genética , Complejo III de Transporte de Electrones/metabolismo , Epistasis Genética/genética , Errores Innatos del Metabolismo/genética , Errores Innatos del Metabolismo/metabolismo , Mitocondrias/genética , Enfermedades Mitocondriales/genética , ATPasas Asociadas con Actividades Celulares Diversas/genética , ATPasas Asociadas con Actividades Celulares Diversas/metabolismo , Animales , Grupo Citocromo b/química , Grupo Citocromo b/genética , Citocromos b , ADN Mitocondrial , Complejo III de Transporte de Electrones/química , Metabolismo Energético , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Simulación de Dinámica Molecular , Oxidación-Reducción
12.
Biochim Biophys Acta Mol Basis Dis ; 1866(1): 165573, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31672551

RESUMEN

Mice homozygous for the human GRACILE syndrome mutation (Bcs1lc.A232G) display decreased respiratory chain complex III activity, liver dysfunction, hypoglycemia, rapid loss of white adipose tissue and early death. To assess the underlying mechanism of the lipodystrophy in homozygous mice (Bcs1lp.S78G), these and wild-type control mice were subjected to a short 4-hour fast. The homozygotes had low baseline blood glucose values, but a similar decrease in response to fasting as in wild-type mice, resulting in hypoglycemia in the majority. Despite the already depleted glycogen and increased triacylglycerol content in the mutant livers, the mice responded to fasting by further depletion and increase, respectively. Increased plasma free fatty acids (FAs) upon fasting suggested normal capacity for mobilization of lipids from white adipose tissue into circulation. Strikingly, however, serum glycerol concentration was not increased concomitantly with free FAs, suggesting its rapid uptake into the liver and utilization for fuel or gluconeogenesis in the mutants. The mutant hepatocyte mitochondria were capable of responding to fasting by appropriate morphological changes, as analyzed by electron microscopy, and by increasing respiration. Mutants showed increased hepatic gene expression of major metabolic controllers typically associated with fasting response (Ppargc1a, Fgf21, Cd36) already in the fed state, suggesting a chronic starvation-like metabolic condition. Despite this, the mutant mice responded largely normally to fasting by increasing hepatic respiration and switching to FA utilization, indicating that the mechanisms driving these adaptations are not compromised by the CIII dysfunction. SUMMARY STATEMENT: Bcs1l mutant mice with severe CIII deficiency, energy deprivation and post-weaning lipolysis respond to fasting similarly to wild-type mice, suggesting largely normal systemic lipid mobilization and utilization mechanisms.


Asunto(s)
Complejo III de Transporte de Electrones/metabolismo , Ayuno/fisiología , Movilización Lipídica/fisiología , Acidosis Láctica/metabolismo , Animales , Glucemia/metabolismo , Colestasis/metabolismo , Transporte de Electrón/fisiología , Femenino , Retardo del Crecimiento Fetal/metabolismo , Gluconeogénesis/fisiología , Glucógeno/metabolismo , Hemosiderosis/metabolismo , Hepatocitos/metabolismo , Hepatocitos/fisiología , Homocigoto , Hipoglucemia/metabolismo , Hipoglucemia/fisiopatología , Hígado/metabolismo , Hígado/fisiología , Masculino , Errores Innatos del Metabolismo/metabolismo , Ratones , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Mitocondrias/fisiología , Enfermedades Mitocondriales/congénito , Enfermedades Mitocondriales/metabolismo , Aminoacidurias Renales/metabolismo , Triglicéridos/metabolismo
13.
EBioMedicine ; 42: 30-31, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30956172
14.
EMBO Mol Med ; 11(1)2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30530468

RESUMEN

Alternative oxidase (AOX) is a non-mammalian enzyme that can bypass blockade of the complex III-IV segment of the respiratory chain (RC). We crossed a Ciona intestinalis AOX transgene into RC complex III (cIII)-deficient Bcs1lp.S78G knock-in mice, displaying multiple visceral manifestations and premature death. The homozygotes expressing AOX were viable, and their median survival was extended from 210 to 590 days due to permanent prevention of lethal cardiomyopathy. AOX also prevented renal tubular atrophy and cerebral astrogliosis, but not liver disease, growth restriction, or lipodystrophy, suggesting distinct tissue-specific pathogenetic mechanisms. Assessment of reactive oxygen species (ROS) production and damage suggested that ROS were not instrumental in the rescue. Cardiac mitochondrial ultrastructure, mitochondrial respiration, and pathological transcriptome and metabolome alterations were essentially normalized by AOX, showing that the restored electron flow upstream of cIII was sufficient to prevent cardiac energetic crisis and detrimental decompensation. These findings demonstrate the value of AOX, both as a mechanistic tool and a potential therapeutic strategy, for cIII deficiencies.


Asunto(s)
Cardiomiopatías/prevención & control , Respiración de la Célula , Complejo III de Transporte de Electrones/deficiencia , Proteínas Mitocondriales/metabolismo , Oxidorreductasas/metabolismo , Proteínas de Plantas/metabolismo , Proteínas Recombinantes/metabolismo , Animales , Ciona intestinalis/enzimología , Ciona intestinalis/genética , Técnicas de Sustitución del Gen , Ratones Endogámicos C57BL , Proteínas Mitocondriales/genética , Oxidorreductasas/genética , Proteínas de Plantas/genética , Proteínas Recombinantes/genética , Análisis de Supervivencia
15.
FASEB J ; : fj201800090R, 2018 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-29782205

RESUMEN

Biosynthetic precursors of NAD+ can replenish a decreased cellular NAD+ pool and, supposedly via sirtuin (SIRT) deacetylases, improve mitochondrial function. We found decreased hepatic NAD+ concentration and downregulated biosynthesis in Bcs1lp.S78G knock-in mice with respiratory chain complex III deficiency and mitochondrial hepatopathy. Aiming at ameliorating disease progression via NAD+ repletion and improved mitochondrial function, we fed these mice nicotinamide riboside (NR), a NAD+ precursor. A targeted metabolomics verified successful administration and suggested enhanced NAD+ biosynthesis in the treated mice, although hepatic NAD+ concentration was unchanged at the end point. In contrast to our expectations, NR did not improve the hepatopathy, hepatic mitochondrial respiration, or survival of Bcs1lp.S78G mice. We linked this lack of therapeutic effect to NAD+-independent activation of SIRT-1 and -3 via AMPK and cAMP signaling related to the starvation-like metabolic state of Bcs1lp.S78G mice. In summary, we describe an unusual metabolic state with NAD+ depletion accompanied by energy deprivation signals, uncompromised SIRT function, and upregulated oxidative metabolism. Our study highlights that the knowledge of the underlying complex metabolic alterations is critical when designing therapies for mitochondrial dysfunction.-Purhonen, J., Rajendran, J., Tegelberg, S., Smolander, O.-P., Pirinen, E., Kallijärvi, J., Fellman, V. NAD+ repletion produces no therapeutic effect in mice with respiratory chain complex III deficiency and chronic energy deprivation.

16.
EMBO J ; 36(20): 3029-3045, 2017 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-28899900

RESUMEN

Expression of the Ret receptor tyrosine kinase is a defining feature of enteric neurons. Its importance is underscored by the effects of its mutation in Hirschsprung disease, leading to absence of gut innervation and severe gastrointestinal symptoms. We report a new and physiologically significant site of Ret expression in the intestine: the intestinal epithelium. Experiments in Drosophila indicate that Ret is expressed both by enteric neurons and adult intestinal epithelial progenitors, which require Ret to sustain their proliferation. Mechanistically, Ret is engaged in a positive feedback loop with Wnt/Wingless signalling, modulated by Src and Fak kinases. We find that Ret is also expressed by the developing intestinal epithelium of mice, where its expression is maintained into the adult stage in a subset of enteroendocrine/enterochromaffin cells. Mouse organoid experiments point to an intrinsic role for Ret in promoting epithelial maturation and regulating Wnt signalling. Our findings reveal evolutionary conservation of the positive Ret/Wnt signalling feedback in both developmental and homeostatic contexts. They also suggest an epithelial contribution to Ret loss-of-function disorders such as Hirschsprung disease.


Asunto(s)
Diferenciación Celular , Proliferación Celular , Células Epiteliales/fisiología , Mucosa Intestinal/fisiología , Proteínas Proto-Oncogénicas c-ret/metabolismo , Animales , Drosophila , Regulación de la Expresión Génica , Humanos , Ratones , Vía de Señalización Wnt
17.
PLoS One ; 12(5): e0176166, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28467503

RESUMEN

Glial cell line-derived neurotrophic factor (GDNF) is a ligand that activates, through co-receptor GDNF family receptor alpha-1 (GFRα1) and receptor tyrosine kinase "RET", several signaling pathways crucial in the development and sustainment of multiple neuronal populations. We decided to study whether non-mammalian orthologs of these three proteins have conserved their function: can they activate the human counterparts? Using the baculovirus expression system, we expressed and purified Danio rerio RET, and its binding partners GFRα1 and GDNF, and Drosophila melanogaster RET and two isoforms of co-receptor GDNF receptor-like. Our results report high-level insect cell expression of post-translationally modified and dimerized zebrafish RET and its binding partners. We also found that zebrafish GFRα1 and GDNF are comparably active as mammalian cell-produced ones. We also report the first measurements of the affinity of the complex to RET in solution: at least for zebrafish, the Kd for GFRα1-GDNF binding RET is 5.9 µM. Surprisingly, we also found that zebrafish GDNF as well as zebrafish GFRα1 robustly activated human RET signaling and promoted the survival of cultured mouse dopaminergic neurons with comparable efficiency to mammalian GDNF, unlike E. coli-produced human proteins. These results contradict previous studies suggesting that mammalian GFRα1 and GDNF cannot bind and activate non-mammalian RET and vice versa.


Asunto(s)
Dopamina/metabolismo , Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Neuronas/metabolismo , Proteínas Proto-Oncogénicas c-ret/metabolismo , Secuencia de Aminoácidos , Animales , Supervivencia Celular , Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial/química , Humanos , Fosforilación , Homología de Secuencia de Aminoácido , Pez Cebra
18.
Orphanet J Rare Dis ; 12(1): 73, 2017 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-28427446

RESUMEN

BACKGROUND: Mitochondrial diseases due to defective respiratory chain complex III (CIII) are relatively uncommon. The assembly of the eleven-subunit CIII is completed by the insertion of the Rieske iron-sulfur protein, a process for which BCS1L protein is indispensable. Mutations in the BCS1L gene constitute the most common diagnosed cause of CIII deficiency, and the phenotypic spectrum arising from mutations in this gene is wide. RESULTS: A case of CIII deficiency was investigated in depth to assess respiratory chain function and assembly, and brain, skeletal muscle and liver histology. Exome sequencing was performed to search for the causative mutation(s). The patient's platelets and muscle mitochondria showed respiration defects and defective assembly of CIII was detected in fibroblast mitochondria. The patient was compound heterozygous for two novel mutations in BCS1L, c.306A > T and c.399delA. In the cerebral cortex a specific pattern of astrogliosis and widespread loss of microglia was observed. Further analysis showed loss of Kupffer cells in the liver. These changes were not found in infants suffering from GRACILE syndrome, the most severe BCS1L-related disorder causing early postnatal mortality, but were partially corroborated in a knock-in mouse model of BCS1L deficiency. CONCLUSIONS: We describe two novel compound heterozygous mutations in BCS1L causing CIII deficiency. The pathogenicity of one of the mutations was unexpected and points to the importance of combining next generation sequencing with a biochemical approach when investigating these patients. We further show novel manifestations in brain, skeletal muscle and liver, including abnormality in specialized resident macrophages (microglia and Kupffer cells). These novel phenotypes forward our understanding of CIII deficiencies caused by BCS1L mutations.


Asunto(s)
Acidosis Láctica/genética , Colestasis/genética , Retardo del Crecimiento Fetal/genética , Hemosiderosis/genética , Errores Innatos del Metabolismo/genética , Enfermedades Mitocondriales/congénito , Aminoacidurias Renales/genética , Animales , Transporte de Electrón/fisiología , Complejo III de Transporte de Electrones/genética , Flavoproteínas Transportadoras de Electrones/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Ratones , Enfermedades Mitocondriales/genética , Encefalomiopatías Mitocondriales/genética , Mutación/genética
19.
Sci Rep ; 7(1): 957, 2017 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-28424480

RESUMEN

Mitochondrial disorders are among the most prevalent inborn errors of metabolism but largely lack treatments and have poor outcomes. High-fat, low-carbohydrate ketogenic diets (KDs) have shown beneficial effects in mouse models of mitochondrial myopathies, with induction of mitochondrial biogenesis as the suggested main mechanism. We fed KD to mice with respiratory chain complex III (CIII) deficiency and progressive hepatopathy due to mutated BCS1L, a CIII assembly factor. The mutant mice became persistently ketotic and tolerated the KD for up to 11 weeks. Liver disease progression was attenuated by KD as shown by delayed fibrosis, reduced cell death, inhibition of hepatic progenitor cell response and stellate cell activation, and normalization of liver enzyme activities. Despite no clear signs of increased mitochondrial biogenesis in the liver, CIII assembly and activity were improved and mitochondrial morphology in hepatocytes normalized. Induction of hepatic glutathione transferase genes and elevated total glutathione level were normalized by KD. Histological findings and transcriptome changes indicated modulation of liver macrophage populations by the mutation and the diet. These results reveal a striking beneficial hepatic response to KD in mice with mitochondrial hepatopathy and warrant further investigations of dietary modification in the management of these conditions in patients.


Asunto(s)
ATPasas Asociadas con Actividades Celulares Diversas/genética , Complejo III de Transporte de Electrones/deficiencia , Hepatopatías/dietoterapia , Miopatías Mitocondriales/complicaciones , Mutación , Animales , Células Cultivadas , Dieta Cetogénica , Modelos Animales de Enfermedad , Complejo III de Transporte de Electrones/genética , Células Estrelladas Hepáticas/citología , Humanos , Hepatopatías/etiología , Ratones , Miopatías Mitocondriales/genética , Resultado del Tratamiento
20.
PLoS One ; 11(12): e0168774, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27997587

RESUMEN

The COX7A2L (Supercomplex Assembly Factor I, SCAFI) protein has been proposed to be a mitochondrial supercomplex assembly factor required for respirasome (supercomplex containing complexes I, III, and IV) formation. In the C57BL/6 mouse strain a homozygous in-frame 6-base-pair deletion in the COX7a2l/SCAF1 gene resulting in unstable protein and suggesting loss of function was previously identified. The loss of SCAFI was shown to impede respirasome formation, a major concern for the use of C57BL mouse strains in mitochondrial research. In contradiction, another recent study suggested that supercomplex formation is independent of SCAFI isoforms. We investigated whether SCAFI isoform status affected the disease severity and supercomplex formation in the liver of Bcs1lc.232A>G knock-in mice with incomplete complex III assembly. In homozygotes (Bcs1lG/G) of mixed (C57BL/6:129/Sv) genetic background, the lifespan was similar in mice with wild-type SCAFI allele and in those homozygous (SCAFIshort/short) for the deleted SCAF1 variant (34±3 days; n = 6 vs. 32±2 days; n = 7, respectively). SCAFI heterozygosity (SCAFIlong/short) resulted in decreased SCAFI protein but respirasome assembly was unaffected. Congenic (C57BL/6) mice were of the genotype SCAFIshort/short and had no detectable SCAFI protein. In their liver mitochondria, respirasome composition was altered as compared to mixed background mice. Complex IV was mainly present as monomers and dimers, and only low amounts were found in combination with complex I and complex III or with precomplex III. The main supercomplex in the liver mitochondria of C57BL/6 mice comprised only complexes I and III. In conclusion, in liver mitochondria of C57BL/6 mice, supercomplexes had markedly reduced amount of, but were not completely depleted of, complex IV, supporting a role for COX7A2L/SCAFI in supercomplex assembly. However, the disease progression of the Bcs1l mutant mice was unrelated to SCAFI isoforms and supercomplex composition, suggesting that other genetic factors contribute to the different survival in the different genetic backgrounds.


Asunto(s)
Complejo III de Transporte de Electrones/metabolismo , Complejo IV de Transporte de Electrones/metabolismo , Mitocondrias Hepáticas/metabolismo , Proteínas Mitocondriales/metabolismo , Chaperonas Moleculares/metabolismo , Mutación , ATPasas Asociadas con Actividades Celulares Diversas , Animales , Complejo III de Transporte de Electrones/genética , Complejo IV de Transporte de Electrones/genética , Ratones , Ratones Mutantes , Mitocondrias Hepáticas/genética , Proteínas Mitocondriales/genética , Chaperonas Moleculares/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Especificidad de la Especie
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...