14 research outputs found

    Insuffisance rénale aiguë : rÎle du métabolisme, place du facteur de transcription HNF-1b et identification de nouveaux biomarqueurs diagnostiques

    Get PDF
    L'insuffisance rĂ©nale aiguĂ« (IRA), dĂ©finie comme une altĂ©ration brutale de la fonction de filtration rĂ©nale, est un vĂ©ritable problĂšme de santĂ© publique par sa frĂ©quence et son importante morbi-mortalitĂ©. La physiopathologie de ce syndrome est encore incomplĂštement comprise mais elle implique une adaptation mĂ©tabolique insuffisante de l'Ă©pithĂ©lium tubulaire rĂ©nal dont les mĂ©canismes rĂ©gulateurs ne sont pas complĂštement connus. Ce dĂ©ficit d'adaptation est Ă  l'origine d'une souffrance cellulaire expliquant en partie les lĂ©sions tissulaires. De plus, ce syndrome est souvent ignorĂ© ou identifiĂ© trop tardivement, en raison de critĂšres diagnostiques imparfaits. Ces lacunes expliquent qu'il n'existe Ă  l'heure actuelle aucun traitement spĂ©cifique efficace. Mon travail de thĂšse s'est organisĂ© autour de ces trois problĂ©matiques : physiopathologique, thĂ©rapeutique et diagnostique. Le premier axe de mon travail, physiopathologique, a Ă©valuĂ© le rĂŽle du facteur de transcription Hepatocyte Nuclear Factor 1 ß (HNF-1ß) dans la rĂ©gulation du mĂ©tabolisme, notamment Ă©nergĂ©tique, au sein de l'Ă©pithĂ©lium tubulaire rĂ©nal. Nous avons dĂ©montrĂ© que l'invalidation de ce facteur de transcription dans une lignĂ©e tubulaire proximale murine, induit une rĂ©orientation du mĂ©tabolisme Ă©nergĂ©tique en conditions basales, avec rĂ©duction du mĂ©tabolisme oxydatif mitochondrial et augmentation de la glycolyse, sans d'autre explication que la modulation de PGC-1alpha. Ces anomalies, proches de celles induites par l'hypoxie dans les cellules sauvages, ne permettent pas d'adaptation supplĂ©mentaire des cellules invalidĂ©es Ă  un stress. L'invalidation d'Hnf1b induit Ă©galement des modifications mĂ©taboliques au-delĂ  de la production Ă©nergĂ©tique Ă  l'image de la synthĂšse des phospholipides, potentiellement en lien avec un rĂ©gulation directe de la choline kinase-alpha. Ce travail confirme un rĂŽle d'Hnf1b dans le mĂ©tabolisme cellulaire Ă©nergĂ©tique, pouvant expliquer une partie du phĂ©notype des patients souffrant d'une mutation de ce facteur mais le plaçant aussi comme un rĂ©gulateur de la rĂ©ponse Ă  l'agression en situation gĂ©nĂ©rale. Le deuxiĂšme axe, thĂ©rapeutique, s'est intĂ©ressĂ© aux anomalies de la voie de biosynthĂšse de novo du Nicotinamide AdĂ©nine DinuclĂ©otide (NAD) Ă  partir du tryptophane en contexte d'IRA. En comparant, par approche mĂ©tabolomique ciblĂ©e, la composition des urines de patients dĂ©veloppant ou non une IRA, aprĂšs chirurgie cardiaque, nous avons pu confirmer dans un nombre plus important de patients, certaines anomalies observĂ©s dans des Ă©tudes antĂ©rieures. NĂ©anmoins, Ă  l'inverse d'observations publiĂ©es, une supplĂ©mentation en nicotinamide, prĂ©curseur alternatif de la biosynthĂšse du NAD, ne permet pas de restaurer le stock intra-rĂ©nal de NAD ni d'amĂ©liorer les consĂ©quences rĂ©nales d'une ischĂ©mie-reperfusion chez la souris. Ces rĂ©sultats incitent Ă  la prudence quant Ă  l'efficacitĂ© attendue de ce traitement, en cours d'essai thĂ©rapeutique. Le troisiĂšme axe, diagnostique, s'est intĂ©ressĂ© Ă  l'identification de biomarqueurs urinaires permettant le diagnostic d'une IRA. Nous avons identifiĂ©, par approche peptidomique sur des urines prĂ©levĂ©es prĂ©cocement aprĂšs chirurgie cardiaque, de nombreux peptides diffĂ©rentiellement prĂ©sents en cas d'IRA. Leur combinaison en une signature permet un diagnostic prĂ©coce de l'AKI avec de bonnes performances (AUC = 0.79) dans divers contextes. La poursuite de ces analyses, avec utilisation de nouvelles approches (mĂ©tabolomique) ou de combinaisons d'approches (multiomics, scores clinico-biologiques), donnent dĂ©jĂ  de bons rĂ©sultats accroissant encore ce bĂ©nĂ©fice diagnostique avec mĂȘme un potentiel prĂ©dictif. Bien que prĂ©liminaire et difficilement transposable en l'Ă©tat Ă  la pratique, ce type d'approche offre des perspectives intĂ©ressantes dans le diagnostic et la prĂ©diction de cette affection. Ce travail de thĂšse, a permis de mettre en Ă©vidence de nouveaux Ă©lĂ©ments utiles concernant la physiopathologie, le traitement et le diagnostic de l'IRA.Acute Kidney Injury (AKI), defined as an abrupt decrease in glomerular filtration rate, is associated with an increased rate of death and chronic kidney disease. Its pathophysiology remains elusive but includes metabolic maladaptation of the kidney tubule cells. This maladaptation results in cell death that accounts in part for the observed tissular lesions. AKI is often missed or only detected at a late stage, also because of suboptimal diagnostic criteria. These shortcomings preclude the development of effective treatment and early detection despite years of active research. My thesis project explored the pathophysiology, therapy and diagnosis of AKI. The first part of my work addressed the role of transcription factor Hepatocyte nuclear factor 1b (HNF-1b) in the cell metabolism of tubular epithelial cells. Hnf1b invalidation in a mouse proximal tubule cell line induced a metabolic switch associated to a reduced mitochondrial oxidative metabolism and increased glycolysis recapitulating what is observed in wild-type cells during hypoxic challenge. Hnf1b invalidation also induced wider metabolic changes such as alteration of phospholipid biosynthesis. This work unraveled a new role of HNF-1b by regulating the energetic metabolism, that could underlie some kidney phenotypes observed in HNF1B patients, but also prompts to consider HNF-1b as master regulator of epithelial response to aggression in a more general context. The second part of my thesis focused on the treatment of AKI. I studied the de novo biosynthetic pathway of nicotinamide adenine dinucleotide (NAD) in AKI. Using a targeted metabolomic strategy on urine samples, we confirmed previously published abnormalities in a large cohort of 167 patients. However, contrary to published literature, nicotinamide supplementation, an alternative precursor to tryptophan for NAD biosynthesis, was not able to either restore the intra-renal NAD stock, nor improve kidney outcomes after ischemia-reperfusion injury in mice. Although currently being evaluated in various clinical trials, our results encourage to remain cautious about benefit expected from such a treatment. The diagnosis of AKI was the third concern of my work. Using urinary peptidome analysis on early postoperative samples after cardiac surgery, we identified various peptides differentially abundant between AKI and non AKI patients. Their combination into a signature allowed early diagnosis of AKI (AUROC = 0.79) in this setting, but also in non-selected patients admitted to the ICU. Preliminary data show that their combination with clinical data and potentially with other omics traits (metabolites) are promising to further increase the predictive performance. Although currently not directly applicable in the clinical setting, this kind of approach offers new perspectives to diagnose AKI. This thesis work shed light on new useful elements about the pathophysiology, treatment and diagnosis of AKI

    Insuffisance rénale aiguë : rÎle du métabolisme, place du facteur de transcription HNF-1b et identification de nouveaux biomarqueurs diagnostiques

    No full text
    Acute Kidney Injury (AKI), defined as an abrupt decrease in glomerular filtration rate, is associated with an increased rate of death and chronic kidney disease. Its pathophysiology remains elusive but includes metabolic maladaptation of the kidney tubule cells. This maladaptation results in cell death that accounts in part for the observed tissular lesions. AKI is often missed or only detected at a late stage, also because of suboptimal diagnostic criteria. These shortcomings preclude the development of effective treatment and early detection despite years of active research. My thesis project explored the pathophysiology, therapy and diagnosis of AKI. The first part of my work addressed the role of transcription factor Hepatocyte nuclear factor 1b (HNF-1b) in the cell metabolism of tubular epithelial cells. Hnf1b invalidation in a mouse proximal tubule cell line induced a metabolic switch associated to a reduced mitochondrial oxidative metabolism and increased glycolysis recapitulating what is observed in wild-type cells during hypoxic challenge. Hnf1b invalidation also induced wider metabolic changes such as alteration of phospholipid biosynthesis. This work unraveled a new role of HNF-1b by regulating the energetic metabolism, that could underlie some kidney phenotypes observed in HNF1B patients, but also prompts to consider HNF-1b as master regulator of epithelial response to aggression in a more general context. The second part of my thesis focused on the treatment of AKI. I studied the de novo biosynthetic pathway of nicotinamide adenine dinucleotide (NAD) in AKI. Using a targeted metabolomic strategy on urine samples, we confirmed previously published abnormalities in a large cohort of 167 patients. However, contrary to published literature, nicotinamide supplementation, an alternative precursor to tryptophan for NAD biosynthesis, was not able to either restore the intra-renal NAD stock, nor improve kidney outcomes after ischemia-reperfusion injury in mice. Although currently being evaluated in various clinical trials, our results encourage to remain cautious about benefit expected from such a treatment. The diagnosis of AKI was the third concern of my work. Using urinary peptidome analysis on early postoperative samples after cardiac surgery, we identified various peptides differentially abundant between AKI and non AKI patients. Their combination into a signature allowed early diagnosis of AKI (AUROC = 0.79) in this setting, but also in non-selected patients admitted to the ICU. Preliminary data show that their combination with clinical data and potentially with other omics traits (metabolites) are promising to further increase the predictive performance. Although currently not directly applicable in the clinical setting, this kind of approach offers new perspectives to diagnose AKI. This thesis work shed light on new useful elements about the pathophysiology, treatment and diagnosis of AKI.L’insuffisance rĂ©nale aigue (IRA), dĂ©finie comme une altĂ©ration brutale de la fonction de filtration rĂ©nale, est un vĂ©ritable problĂšme de santĂ© publique par sa frĂ©quence et son importante morbi-mortalitĂ©. La physiopathologie de ce syndrome est encore incomplĂštement comprise mais elle implique une adaptation mĂ©tabolique insuffisante de l’épithĂ©lium tubulaire rĂ©nal dont les mĂ©canismes rĂ©gulateurs ne sont pas complĂštement connus. Ce dĂ©ficit d’adaptation est Ă  l’origine d’une souffrance cellulaire expliquant en partie les lĂ©sions tissulaires. De plus, ce syndrome est souvent ignorĂ© ou identifiĂ© trop tardivement, en raison de critĂšres diagnostiques imparfaits. Ces lacunes expliquent qu’il n’existe Ă  l’heure actuelle aucun traitement spĂ©cifique efficace. Mon travail de thĂšse s’est organisĂ© autour de ces trois problĂ©matiques : physiopathologique, thĂ©rapeutique et diagnostique.Le premier axe de mon travail, physiopathologique, a Ă©valuĂ© le rĂŽle du facteur de transcription Hepatocyte Nuclear Factor 1 ÎČ (HNF-1ÎČ) dans la rĂ©gulation du mĂ©tabolisme, notamment Ă©nergĂ©tique, au sein de l’épithĂ©lium tubulaire rĂ©nal. Nous avons dĂ©montrĂ© que l’invalidation de ce facteur de transcription dans une lignĂ©e tubulaire proximale murine, induit une rĂ©orientation du mĂ©tabolisme Ă©nergĂ©tique en conditions basales, avec rĂ©duction du mĂ©tabolisme oxydatif mitochondrial et augmentation de la glycolyse, sans d’autre explication que la modulation de PGC-1α. Ces anomalies, proches de celles induites par l’hypoxie dans les cellules sauvages, ne permettent pas d’adaptation supplĂ©mentaire des cellules invalidĂ©es Ă  un stress. L’invalidation d’Hnf1b induit Ă©galement des modifications mĂ©taboliques au-delĂ  de la production Ă©nergĂ©tique Ă  l’image de la synthĂšse des phospholipides, potentiellement en lien avec un rĂ©gulation directe de la choline kinase-α. Ce travail confirme un rĂŽle d’Hnf1b dans le mĂ©tabolisme cellulaire Ă©nergĂ©tique, pouvant expliquer une partie du phĂ©notype des patients souffrant d’une mutation de ce facteur mais le plaçant aussi comme un rĂ©gulateur de la rĂ©ponse Ă  l’agression en situation gĂ©nĂ©rale Le deuxiĂšme axe, thĂ©rapeutique, s’est intĂ©ressĂ© aux anomalies de la voie de biosynthĂšse de novo du Nicotinamide AdĂ©nine DinuclĂ©otide (NAD) Ă  partir du tryptophane en contexte d’IRA. En comparant, par approche mĂ©tabolomique ciblĂ©e, la composition des urines de patients dĂ©veloppant ou non une IRA, aprĂšs chirurgie cardiaque, nous avons pu confirmer dans un nombre plus important de patients, certaines anomalies observĂ©s dans des Ă©tudes antĂ©rieures. NĂ©anmoins, Ă  l’inverse d’observations publiĂ©es, une supplĂ©mentation en nicotinamide, prĂ©curseur alternatif de la biosynthĂšse du NAD, ne permet pas de restaurer le stock intra-rĂ©nal de NAD ni d’amĂ©liorer les consĂ©quences rĂ©nales d’une ischĂ©mie-reperfusion chez la souris. Ces rĂ©sultats incitent Ă  la prudence quant Ă  l’efficacitĂ© attendue de ce traitement, en cours d’essai thĂ©rapeutique. Le troisiĂšme axe, diagnostique, s’est intĂ©ressĂ© Ă  l’identification de biomarqueurs urinaires permettant le diagnostic d’une IRA. Nous avons identifiĂ©, par approche peptidomique sur des urines prĂ©levĂ©es prĂ©cocement aprĂšs chirurgie cardiaque, de nombreux peptides diffĂ©rentiellement prĂ©sents en cas d’IRA. Leur combinaison en une signature permet un diagnostic prĂ©coce de l’AKI avec de bonnes performances (AUC = 0.79) dans divers contextes. La poursuite de ces analyses, avec utilisation de nouvelles approches (mĂ©tabolomique) ou de combinaisons d’approches (multiomics, scores clinico-biologiques), donnent dĂ©jĂ  de bons rĂ©sultats accroissant encore ce bĂ©nĂ©fice diagnostique avec mĂȘme un potentiel prĂ©dictif. Bien que prĂ©liminaire et difficilement transposable en l’état Ă  la pratique, ce type d’approche offre des perspectives intĂ©ressantes dans le diagnostic et la prĂ©diction de cette affection. Ce travail de thĂšse, a permis de mettre en Ă©vidence de nouveaux Ă©lĂ©ments utiles concernant la physiopathologie, le traitement et le diagnostic de l’IRA

    Acute kidney injury : role of metabolism, HNF-1b transcription factor as a master regulator and new diagnostic biomarkers identification

    No full text
    L’insuffisance rĂ©nale aigue (IRA), dĂ©finie comme une altĂ©ration brutale de la fonction de filtration rĂ©nale, est un vĂ©ritable problĂšme de santĂ© publique par sa frĂ©quence et son importante morbi-mortalitĂ©. La physiopathologie de ce syndrome est encore incomplĂštement comprise mais elle implique une adaptation mĂ©tabolique insuffisante de l’épithĂ©lium tubulaire rĂ©nal dont les mĂ©canismes rĂ©gulateurs ne sont pas complĂštement connus. Ce dĂ©ficit d’adaptation est Ă  l’origine d’une souffrance cellulaire expliquant en partie les lĂ©sions tissulaires. De plus, ce syndrome est souvent ignorĂ© ou identifiĂ© trop tardivement, en raison de critĂšres diagnostiques imparfaits. Ces lacunes expliquent qu’il n’existe Ă  l’heure actuelle aucun traitement spĂ©cifique efficace. Mon travail de thĂšse s’est organisĂ© autour de ces trois problĂ©matiques : physiopathologique, thĂ©rapeutique et diagnostique.Le premier axe de mon travail, physiopathologique, a Ă©valuĂ© le rĂŽle du facteur de transcription Hepatocyte Nuclear Factor 1 ÎČ (HNF-1ÎČ) dans la rĂ©gulation du mĂ©tabolisme, notamment Ă©nergĂ©tique, au sein de l’épithĂ©lium tubulaire rĂ©nal. Nous avons dĂ©montrĂ© que l’invalidation de ce facteur de transcription dans une lignĂ©e tubulaire proximale murine, induit une rĂ©orientation du mĂ©tabolisme Ă©nergĂ©tique en conditions basales, avec rĂ©duction du mĂ©tabolisme oxydatif mitochondrial et augmentation de la glycolyse, sans d’autre explication que la modulation de PGC-1α. Ces anomalies, proches de celles induites par l’hypoxie dans les cellules sauvages, ne permettent pas d’adaptation supplĂ©mentaire des cellules invalidĂ©es Ă  un stress. L’invalidation d’Hnf1b induit Ă©galement des modifications mĂ©taboliques au-delĂ  de la production Ă©nergĂ©tique Ă  l’image de la synthĂšse des phospholipides, potentiellement en lien avec un rĂ©gulation directe de la choline kinase-α. Ce travail confirme un rĂŽle d’Hnf1b dans le mĂ©tabolisme cellulaire Ă©nergĂ©tique, pouvant expliquer une partie du phĂ©notype des patients souffrant d’une mutation de ce facteur mais le plaçant aussi comme un rĂ©gulateur de la rĂ©ponse Ă  l’agression en situation gĂ©nĂ©rale Le deuxiĂšme axe, thĂ©rapeutique, s’est intĂ©ressĂ© aux anomalies de la voie de biosynthĂšse de novo du Nicotinamide AdĂ©nine DinuclĂ©otide (NAD) Ă  partir du tryptophane en contexte d’IRA. En comparant, par approche mĂ©tabolomique ciblĂ©e, la composition des urines de patients dĂ©veloppant ou non une IRA, aprĂšs chirurgie cardiaque, nous avons pu confirmer dans un nombre plus important de patients, certaines anomalies observĂ©s dans des Ă©tudes antĂ©rieures. NĂ©anmoins, Ă  l’inverse d’observations publiĂ©es, une supplĂ©mentation en nicotinamide, prĂ©curseur alternatif de la biosynthĂšse du NAD, ne permet pas de restaurer le stock intra-rĂ©nal de NAD ni d’amĂ©liorer les consĂ©quences rĂ©nales d’une ischĂ©mie-reperfusion chez la souris. Ces rĂ©sultats incitent Ă  la prudence quant Ă  l’efficacitĂ© attendue de ce traitement, en cours d’essai thĂ©rapeutique. Le troisiĂšme axe, diagnostique, s’est intĂ©ressĂ© Ă  l’identification de biomarqueurs urinaires permettant le diagnostic d’une IRA. Nous avons identifiĂ©, par approche peptidomique sur des urines prĂ©levĂ©es prĂ©cocement aprĂšs chirurgie cardiaque, de nombreux peptides diffĂ©rentiellement prĂ©sents en cas d’IRA. Leur combinaison en une signature permet un diagnostic prĂ©coce de l’AKI avec de bonnes performances (AUC = 0.79) dans divers contextes. La poursuite de ces analyses, avec utilisation de nouvelles approches (mĂ©tabolomique) ou de combinaisons d’approches (multiomics, scores clinico-biologiques), donnent dĂ©jĂ  de bons rĂ©sultats accroissant encore ce bĂ©nĂ©fice diagnostique avec mĂȘme un potentiel prĂ©dictif. Bien que prĂ©liminaire et difficilement transposable en l’état Ă  la pratique, ce type d’approche offre des perspectives intĂ©ressantes dans le diagnostic et la prĂ©diction de cette affection. Ce travail de thĂšse, a permis de mettre en Ă©vidence de nouveaux Ă©lĂ©ments utiles concernant la physiopathologie, le traitement et le diagnostic de l’IRA.Acute Kidney Injury (AKI), defined as an abrupt decrease in glomerular filtration rate, is associated with an increased rate of death and chronic kidney disease. Its pathophysiology remains elusive but includes metabolic maladaptation of the kidney tubule cells. This maladaptation results in cell death that accounts in part for the observed tissular lesions. AKI is often missed or only detected at a late stage, also because of suboptimal diagnostic criteria. These shortcomings preclude the development of effective treatment and early detection despite years of active research. My thesis project explored the pathophysiology, therapy and diagnosis of AKI. The first part of my work addressed the role of transcription factor Hepatocyte nuclear factor 1b (HNF-1b) in the cell metabolism of tubular epithelial cells. Hnf1b invalidation in a mouse proximal tubule cell line induced a metabolic switch associated to a reduced mitochondrial oxidative metabolism and increased glycolysis recapitulating what is observed in wild-type cells during hypoxic challenge. Hnf1b invalidation also induced wider metabolic changes such as alteration of phospholipid biosynthesis. This work unraveled a new role of HNF-1b by regulating the energetic metabolism, that could underlie some kidney phenotypes observed in HNF1B patients, but also prompts to consider HNF-1b as master regulator of epithelial response to aggression in a more general context. The second part of my thesis focused on the treatment of AKI. I studied the de novo biosynthetic pathway of nicotinamide adenine dinucleotide (NAD) in AKI. Using a targeted metabolomic strategy on urine samples, we confirmed previously published abnormalities in a large cohort of 167 patients. However, contrary to published literature, nicotinamide supplementation, an alternative precursor to tryptophan for NAD biosynthesis, was not able to either restore the intra-renal NAD stock, nor improve kidney outcomes after ischemia-reperfusion injury in mice. Although currently being evaluated in various clinical trials, our results encourage to remain cautious about benefit expected from such a treatment. The diagnosis of AKI was the third concern of my work. Using urinary peptidome analysis on early postoperative samples after cardiac surgery, we identified various peptides differentially abundant between AKI and non AKI patients. Their combination into a signature allowed early diagnosis of AKI (AUROC = 0.79) in this setting, but also in non-selected patients admitted to the ICU. Preliminary data show that their combination with clinical data and potentially with other omics traits (metabolites) are promising to further increase the predictive performance. Although currently not directly applicable in the clinical setting, this kind of approach offers new perspectives to diagnose AKI. This thesis work shed light on new useful elements about the pathophysiology, treatment and diagnosis of AKI

    Insuffisance rénale aiguë : rÎle du métabolisme, place du facteur de transcription HNF-1b et identification de nouveaux biomarqueurs diagnostiques

    Get PDF
    Acute Kidney Injury (AKI), defined as an abrupt decrease in glomerular filtration rate, is associated with an increased rate of death and chronic kidney disease. Its pathophysiology remains elusive but includes metabolic maladaptation of the kidney tubule cells. This maladaptation results in cell death that accounts in part for the observed tissular lesions. AKI is often missed or only detected at a late stage, also because of suboptimal diagnostic criteria. These shortcomings preclude the development of effective treatment and early detection despite years of active research. My thesis project explored the pathophysiology, therapy and diagnosis of AKI. The first part of my work addressed the role of transcription factor Hepatocyte nuclear factor 1b (HNF-1b) in the cell metabolism of tubular epithelial cells. Hnf1b invalidation in a mouse proximal tubule cell line induced a metabolic switch associated to a reduced mitochondrial oxidative metabolism and increased glycolysis recapitulating what is observed in wild-type cells during hypoxic challenge. Hnf1b invalidation also induced wider metabolic changes such as alteration of phospholipid biosynthesis. This work unraveled a new role of HNF-1b by regulating the energetic metabolism, that could underlie some kidney phenotypes observed in HNF1B patients, but also prompts to consider HNF-1b as master regulator of epithelial response to aggression in a more general context. The second part of my thesis focused on the treatment of AKI. I studied the de novo biosynthetic pathway of nicotinamide adenine dinucleotide (NAD) in AKI. Using a targeted metabolomic strategy on urine samples, we confirmed previously published abnormalities in a large cohort of 167 patients. However, contrary to published literature, nicotinamide supplementation, an alternative precursor to tryptophan for NAD biosynthesis, was not able to either restore the intra-renal NAD stock, nor improve kidney outcomes after ischemia-reperfusion injury in mice. Although currently being evaluated in various clinical trials, our results encourage to remain cautious about benefit expected from such a treatment. The diagnosis of AKI was the third concern of my work. Using urinary peptidome analysis on early postoperative samples after cardiac surgery, we identified various peptides differentially abundant between AKI and non AKI patients. Their combination into a signature allowed early diagnosis of AKI (AUROC = 0.79) in this setting, but also in non-selected patients admitted to the ICU. Preliminary data show that their combination with clinical data and potentially with other omics traits (metabolites) are promising to further increase the predictive performance. Although currently not directly applicable in the clinical setting, this kind of approach offers new perspectives to diagnose AKI. This thesis work shed light on new useful elements about the pathophysiology, treatment and diagnosis of AKI.L’insuffisance rĂ©nale aigue (IRA), dĂ©finie comme une altĂ©ration brutale de la fonction de filtration rĂ©nale, est un vĂ©ritable problĂšme de santĂ© publique par sa frĂ©quence et son importante morbi-mortalitĂ©. La physiopathologie de ce syndrome est encore incomplĂštement comprise mais elle implique une adaptation mĂ©tabolique insuffisante de l’épithĂ©lium tubulaire rĂ©nal dont les mĂ©canismes rĂ©gulateurs ne sont pas complĂštement connus. Ce dĂ©ficit d’adaptation est Ă  l’origine d’une souffrance cellulaire expliquant en partie les lĂ©sions tissulaires. De plus, ce syndrome est souvent ignorĂ© ou identifiĂ© trop tardivement, en raison de critĂšres diagnostiques imparfaits. Ces lacunes expliquent qu’il n’existe Ă  l’heure actuelle aucun traitement spĂ©cifique efficace. Mon travail de thĂšse s’est organisĂ© autour de ces trois problĂ©matiques : physiopathologique, thĂ©rapeutique et diagnostique.Le premier axe de mon travail, physiopathologique, a Ă©valuĂ© le rĂŽle du facteur de transcription Hepatocyte Nuclear Factor 1 ÎČ (HNF-1ÎČ) dans la rĂ©gulation du mĂ©tabolisme, notamment Ă©nergĂ©tique, au sein de l’épithĂ©lium tubulaire rĂ©nal. Nous avons dĂ©montrĂ© que l’invalidation de ce facteur de transcription dans une lignĂ©e tubulaire proximale murine, induit une rĂ©orientation du mĂ©tabolisme Ă©nergĂ©tique en conditions basales, avec rĂ©duction du mĂ©tabolisme oxydatif mitochondrial et augmentation de la glycolyse, sans d’autre explication que la modulation de PGC-1α. Ces anomalies, proches de celles induites par l’hypoxie dans les cellules sauvages, ne permettent pas d’adaptation supplĂ©mentaire des cellules invalidĂ©es Ă  un stress. L’invalidation d’Hnf1b induit Ă©galement des modifications mĂ©taboliques au-delĂ  de la production Ă©nergĂ©tique Ă  l’image de la synthĂšse des phospholipides, potentiellement en lien avec un rĂ©gulation directe de la choline kinase-α. Ce travail confirme un rĂŽle d’Hnf1b dans le mĂ©tabolisme cellulaire Ă©nergĂ©tique, pouvant expliquer une partie du phĂ©notype des patients souffrant d’une mutation de ce facteur mais le plaçant aussi comme un rĂ©gulateur de la rĂ©ponse Ă  l’agression en situation gĂ©nĂ©rale Le deuxiĂšme axe, thĂ©rapeutique, s’est intĂ©ressĂ© aux anomalies de la voie de biosynthĂšse de novo du Nicotinamide AdĂ©nine DinuclĂ©otide (NAD) Ă  partir du tryptophane en contexte d’IRA. En comparant, par approche mĂ©tabolomique ciblĂ©e, la composition des urines de patients dĂ©veloppant ou non une IRA, aprĂšs chirurgie cardiaque, nous avons pu confirmer dans un nombre plus important de patients, certaines anomalies observĂ©s dans des Ă©tudes antĂ©rieures. NĂ©anmoins, Ă  l’inverse d’observations publiĂ©es, une supplĂ©mentation en nicotinamide, prĂ©curseur alternatif de la biosynthĂšse du NAD, ne permet pas de restaurer le stock intra-rĂ©nal de NAD ni d’amĂ©liorer les consĂ©quences rĂ©nales d’une ischĂ©mie-reperfusion chez la souris. Ces rĂ©sultats incitent Ă  la prudence quant Ă  l’efficacitĂ© attendue de ce traitement, en cours d’essai thĂ©rapeutique. Le troisiĂšme axe, diagnostique, s’est intĂ©ressĂ© Ă  l’identification de biomarqueurs urinaires permettant le diagnostic d’une IRA. Nous avons identifiĂ©, par approche peptidomique sur des urines prĂ©levĂ©es prĂ©cocement aprĂšs chirurgie cardiaque, de nombreux peptides diffĂ©rentiellement prĂ©sents en cas d’IRA. Leur combinaison en une signature permet un diagnostic prĂ©coce de l’AKI avec de bonnes performances (AUC = 0.79) dans divers contextes. La poursuite de ces analyses, avec utilisation de nouvelles approches (mĂ©tabolomique) ou de combinaisons d’approches (multiomics, scores clinico-biologiques), donnent dĂ©jĂ  de bons rĂ©sultats accroissant encore ce bĂ©nĂ©fice diagnostique avec mĂȘme un potentiel prĂ©dictif. Bien que prĂ©liminaire et difficilement transposable en l’état Ă  la pratique, ce type d’approche offre des perspectives intĂ©ressantes dans le diagnostic et la prĂ©diction de cette affection. Ce travail de thĂšse, a permis de mettre en Ă©vidence de nouveaux Ă©lĂ©ments utiles concernant la physiopathologie, le traitement et le diagnostic de l’IRA

    Ibrutinib does not prevent kidney fibrosis following acute and chronic injury

    No full text
    Abstract Recent studies suggested that ibrutinib, a Bruton tyrosine kinase (BTK) inhibitor, developed for the treatment of chronic lymphocytic leukemia, may prevent NLRP3 inflammasome activation in macrophages, IL-1ÎČ secretion and subsequent development of inflammation and organ fibrosis. The role of NLRP3 has been underlined in the various causes of acute kidney injury (AKI), a pathology characterized by high morbimortality and risk of transition toward chronic kidney disease (CKD). We therefore hypothesized that the BTK-inhibitor ibrutinib could be a candidate drug for AKI treatment. Here, we observed in both an AKI model (glycerol-induced rhabdomyolysis) and a model of rapidly progressive kidney fibrosis (unilateral ureteral obstruction), that ibrutinib did not prevent inflammatory cell recruitment in the kidney and fibrosis. Moreover, ibrutinib pre-exposure led to high mortality rate owing to severer rhabdomyolysis and AKI. In vitro, ibrutinib potentiated or had no effect on the secretion of IL-1ÎČ by monocytes exposed to uromodulin or myoglobin, two danger-associated molecule patterns proteins involved in the AKI to CKD transition. According to these results, ibrutinib should not be considered a candidate drug for patients developing AKI

    The tryptophan pathway and nicotinamide supplementation in ischaemic acute kidney injury

    No full text
    International audienceAbstract Background Down-regulation of the enzymes involved in tryptophan-derived nicotinamide (NAM) adenine dinucleotide (NAD+) production was identified after acute kidney injury (AKI), leading to the hypothesis that supplementation with NAM may increase the kidney NAD+ content, rescuing tryptophan pathways and subsequently improving kidney outcomes. Methods Urinary measurement of tryptophan and kynurenin using liquid chromatography–mass spectrometry metabolomics was used in a cohort of 167 cardiac bypass surgery patients along with tests for correlation to the development of postoperative AKI. A mouse model of ischaemic AKI using ischaemia–reperfusion injury (bilateral clamping of renal arteries for 25 min) was also used. Results We identified a significant decrease in urinary tryptophan and kynurenin in patients developing AKI, irrespective of the Kidney Disease: Improving Global Outcomes (KDIGO) stage. Although a significant difference was observed, tryptophan and kynurenin moderately discriminated for the development of all AKI KDIGO stages {area under the curve [AUC] 0.82 [95% confidence interval (CI) 0.75–0.88] and 0.75 [0.68–0.83], respectively} and severe KDIGO Stages 2–3 AKI [AUC 0.71 (95% CI 0.6–0.81) and 0.66 (0.55–0.77), respectively]. Sparked by this confirmation in humans, we aimed to confirm the potential preventive effect of NAM supplementation in wild-type male and female C57BL/6 mice subjected to ischaemic AKI. NAM supplementation had no effect on renal function (blood urea nitrogen at Day 1, sinistrin–fluorescein isothiocyanate glomerular filtration rate), architecture (periodic acid–Schiff staining) and injury or inflammation (kidney injury molecule 1 and IL18 messenger RNA expression). In addition, NAM supplementation did not increase post-AKI NAD+ kidney content. Conclusion Notwithstanding the potential role of NAM supplementation in the setting of basal NAD+ deficiency, our findings in mice and the reanalysis of published data do not confirm that NAM supplementation can actually improve renal outcomes after ischaemic AKI in unselected animals and probably patients

    Hepatocyte nuclear factor‐1ÎČ shapes the energetic homeostasis of kidney tubule cells

    No full text
    International audienceEnergetic metabolism controls key steps of kidney development, homeostasis, and epithelial repair following acute kidney injury (AKI). Hepatocyte nuclear factor-1ÎČ (HNF-1ÎČ) is a master transcription factor that controls mitochondrial function in proximal tubule (PT) cells. Patients with HNF1B pathogenic variant display a wide range of kidney developmental abnormalities and progressive kidney fibrosis. Characterizing the metabolic changes in PT cells with HNF-1ÎČ deficiency may help to identify new targetable molecular hubs involved in HNF1B-related kidney phenotypes and AKI. Here, we combined 1 H-NMR-based metabolomic analysis in a murine PT cell line with CrispR/Cas9-induced Hnf1b invalidation (Hnf1b-/- ), clustering analysis, targeted metabolic assays, and datamining of published RNA-seq and ChIP-seq dataset to identify the role of HNF-1ÎČ in metabolism. Hnf1b-/- cells grown in normoxic conditions display intracellular ATP depletion, increased cytosolic lactate concentration, increased lipid droplet content, failure to use pyruvate for energetic purposes, increased levels of tricarboxylic acid (TCA) cycle intermediates and oxidized glutathione, and a reduction of TCA cycle byproducts, all features consistent with mitochondrial dysfunction and an irreversible switch toward glycolysis. Unsupervised clustering analysis showed that Hnf1b-/- cells mimic a hypoxic signature and that they cannot furthermore increase glycolysis-dependent energetic supply during hypoxic challenge. Metabolome analysis also showed alteration of phospholipid biosynthesis in Hnf1b-/- cells leading to the identification of Chka, the gene coding for choline kinase α, as a new putative target of HNF-1ÎČ. HNF-1ÎČ shapes the energetic metabolism of PT cells and HNF1B deficiency in patients could lead to a hypoxia-like metabolic state precluding further adaptation to ATP depletion following AKI

    Spectrum of Kidney Disorders Associated with T-Cell Immunoclones

    No full text
    Large granular T-cell leukemia is a clonal hematological condition often associated with autoimmune disorders. Whether small-sized T-cell clones that are otherwise asymptomatic can promote immune kidney disorders remains elusive. In this monocentric retrospective cohort in a tertiary referral center in France, we reviewed characteristics of 29 patients with T-cell clone proliferation and autoimmune kidney disorders. Next-generation sequencing of the T-cell receptor of circulating T-cells was performed in a subset of patients. The T-cell clones were detected owing to systematic screening (mean count 0.32 × 109/L, range 0.13–3.7). Strikingly, a common phenotype of acute interstitial nephropathy was observed in 22 patients (median estimated glomerular filtration rate at presentation of 22 mL/min/1.73 m2 (range 0–56)). Kidney biopsies showed polymorphic inflammatory cell infiltration (predominantly CD3+ T-cells, most of them demonstrating positive phospho-STAT3 staining) and non-necrotic granuloma in six cases. Immune-mediated glomerulopathy only or in combination with acute interstitial nephropathy was identified in eight patients. Next-generation sequencing (n = 13) identified a major T-cell clone representing more than 1% of the T-cell population in all but two patients. None had a mutation of STAT3. Twenty patients (69%) had two or more extra-kidney autoimmune diseases. Acute interstitial nephropathies were controlled with corticosteroids, cyclosporin A, or tofacitinib. Thus, we showed that small-sized T-cell clones (i.e., without lymphocytosis) undetectable without specific screening are associated with various immune kidney disorders, including a previously unrecognized phenotype characterized by severe inflammatory kidney fibrosis and lymphocytic JAK/STAT activation

    Prognostic Factors in Anti-glomerular Basement Membrane Disease: A Multicenter Study of 119 Patients

    No full text
    International audienceWe report the overall and renal outcome in a French nationwide multicenter cohort of 119 patients with anti-glomerular basement membrane (anti-GBM) disease. Sixty-four patients (54%) had an exclusive renal involvement, 7 (6%) an isolated alveolar hemorrhage and 48 (40%) a combined renal and pulmonary involvement. Initial renal replacement therapy (RRT) was required in 78% of patients; 82% received plasmapheresis, 82% cyclophosphamide, and 9% rituximab. ANCA positive (28%) patients were older (70 vs. 47 years, p < 0.0001), less frequently smokers (26 vs. 54%, p = 0.03), and had less pulmonary involvement than ANCA- patients. The 5 years overall survival was 92%. Risk factors of death (n = 11, 9.2%) were age at onset [HR 4.10 per decade (1.89–8.88) p = 0.003], hypertension [HR 19.9 (2.52–157 0.2) p = 0.005], dyslipidemia [HR 11.1 (2.72–45) p = 0.0008], and need for mechanical ventilation [HR 5.20 (1.02–26.4) p = 0.047]. The use of plasmapheresis was associated with better survival [HR 0.29 (0.08–0.98) p = 0.046]. At 3 months, 55 (46%) patients had end-stage renal disease (ESRD) vs. 37 (31%) ESRD-free and 27 (23%) unevaluable with follow-up < 3 months. ESRD patients were older, more frequently female and had a higher serum creatinine level at presentation than those without ESRD. ESRD-free survival was evaluated in patients alive without ESRD at 3 months (n = 37) using a landmark approach. In conclusion, this large French nationwide study identifies prognosis factors of renal and overall survival in anti-GBM patients
    corecore