Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 128
Filtrar
1.
Nature ; 621(7980): 849-856, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37730993

RESUMEN

Protective immunity against pathogens or cancer is mediated by the activation and clonal expansion of antigen-specific naive T cells into effector T cells. To sustain their rapid proliferation and effector functions, naive T cells switch their quiescent metabolism to an anabolic metabolism through increased levels of aerobic glycolysis, but also through mitochondrial metabolism and oxidative phosphorylation, generating energy and signalling molecules1-3. However, how that metabolic rewiring drives and defines the differentiation of T cells remains unclear. Here we show that proliferating effector CD8+ T cells reductively carboxylate glutamine through the mitochondrial enzyme isocitrate dehydrogenase 2 (IDH2). Notably, deletion of the gene encoding IDH2 does not impair the proliferation of T cells nor their effector function, but promotes the differentiation of memory CD8+ T cells. Accordingly, inhibiting IDH2 during ex vivo manufacturing of chimeric antigen receptor (CAR) T cells induces features of memory T cells and enhances antitumour activity in melanoma, leukaemia and multiple myeloma. Mechanistically, inhibition of IDH2 activates compensating metabolic pathways that cause a disequilibrium in metabolites regulating histone-modifying enzymes, and this maintains chromatin accessibility at genes that are required for the differentiation of memory T cells. These findings show that reductive carboxylation in CD8+ T cells is dispensable for their effector response and proliferation, but that it mainly produces a pattern of metabolites that epigenetically locks CD8+ T cells into a terminal effector differentiation program. Blocking this metabolic route allows the increased formation of memory T cells, which could be exploited to optimize the therapeutic efficacy of CAR T cells.


Asunto(s)
Linfocitos T CD8-positivos , Activación de Linfocitos , Diferenciación Celular/genética , Ciclo del Ácido Cítrico , Fosforilación Oxidativa , Memoria Inmunológica/genética
2.
Cell ; 154(3): 651-63, 2013 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-23911327

RESUMEN

Vessel sprouting by migrating tip and proliferating stalk endothelial cells (ECs) is controlled by genetic signals (such as Notch), but it is unknown whether metabolism also regulates this process. Here, we show that ECs relied on glycolysis rather than on oxidative phosphorylation for ATP production and that loss of the glycolytic activator PFKFB3 in ECs impaired vessel formation. Mechanistically, PFKFB3 not only regulated EC proliferation but also controlled the formation of filopodia/lamellipodia and directional migration, in part by compartmentalizing with F-actin in motile protrusions. Mosaic in vitro and in vivo sprouting assays further revealed that PFKFB3 overexpression overruled the pro-stalk activity of Notch, whereas PFKFB3 deficiency impaired tip cell formation upon Notch blockade, implying that glycolysis regulates vessel branching.


Asunto(s)
Células Endoteliales/metabolismo , Glucólisis , Neovascularización Fisiológica , Fosfofructoquinasa-2/metabolismo , Animales , Línea Celular Tumoral , Células Cultivadas , Células Endoteliales/citología , Femenino , Eliminación de Gen , Silenciador del Gen , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Fosfofructoquinasa-2/genética , Seudópodos/metabolismo , Pez Cebra
3.
Nature ; 587(7835): 626-631, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33116312

RESUMEN

Muscle regeneration is sustained by infiltrating macrophages and the consequent activation of satellite cells1-4. Macrophages and satellite cells communicate in different ways1-5, but their metabolic interplay has not been investigated. Here we show, in a mouse model, that muscle injuries and ageing are characterized by intra-tissue restrictions of glutamine. Low levels of glutamine endow macrophages with the metabolic ability to secrete glutamine via enhanced glutamine synthetase (GS) activity, at the expense of glutamine oxidation mediated by glutamate dehydrogenase 1 (GLUD1). Glud1-knockout macrophages display constitutively high GS activity, which prevents glutamine shortages. The uptake of macrophage-derived glutamine by satellite cells through the glutamine transporter SLC1A5 activates mTOR and promotes the proliferation and differentiation of satellite cells. Consequently, macrophage-specific deletion or pharmacological inhibition of GLUD1 improves muscle regeneration and functional recovery in response to acute injury, ischaemia or ageing. Conversely, SLC1A5 blockade in satellite cells or GS inactivation in macrophages negatively affects satellite cell functions and muscle regeneration. These results highlight the metabolic crosstalk between satellite cells and macrophages, in which macrophage-derived glutamine sustains the functions of satellite cells. Thus, the targeting of GLUD1 may offer therapeutic opportunities for the regeneration of injured or aged muscles.


Asunto(s)
Glutamina/metabolismo , Macrófagos/metabolismo , Músculo Esquelético/metabolismo , Regeneración , Células Satélite del Músculo Esquelético/metabolismo , Envejecimiento/metabolismo , Sistema de Transporte de Aminoácidos ASC/antagonistas & inhibidores , Sistema de Transporte de Aminoácidos ASC/metabolismo , Animales , Diferenciación Celular , Proliferación Celular , Femenino , Glutamato Deshidrogenasa/deficiencia , Glutamato Deshidrogenasa/genética , Glutamato Deshidrogenasa/metabolismo , Glutamato-Amoníaco Ligasa/antagonistas & inhibidores , Glutamato-Amoníaco Ligasa/metabolismo , Macrófagos/enzimología , Masculino , Ratones , Antígenos de Histocompatibilidad Menor/metabolismo , Músculo Esquelético/citología , Músculo Esquelético/lesiones , Músculo Esquelético/patología , Oxidación-Reducción , Células Satélite del Músculo Esquelético/citología , Serina-Treonina Quinasas TOR
4.
Nature ; 580(7801): 130-135, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32238926

RESUMEN

Caspase-dependent apoptosis accounts for approximately 90% of homeostatic cell turnover in the body1, and regulates inflammation, cell proliferation, and tissue regeneration2-4. How apoptotic cells mediate such diverse effects is not fully understood. Here we profiled the apoptotic metabolite secretome and determined its effects on the tissue neighbourhood. We show that apoptotic lymphocytes and macrophages release specific metabolites, while retaining their membrane integrity. A subset of these metabolites is also shared across different primary cells and cell lines after the induction of apoptosis by different stimuli. Mechanistically, the apoptotic metabolite secretome is not simply due to passive emptying of cellular contents and instead is a regulated process. Caspase-mediated opening of pannexin 1 channels at the plasma membrane facilitated the release of a select subset of metabolites. In addition, certain metabolic pathways continued to remain active during apoptosis, with the release of only select metabolites from a given pathway. Functionally, the apoptotic metabolite secretome induced specific gene programs in healthy neighbouring cells, including suppression of inflammation, cell proliferation, and wound healing. Furthermore, a cocktail of apoptotic metabolites reduced disease severity in mouse models of inflammatory arthritis and lung-graft rejection. These data advance the concept that apoptotic cells are not inert cells waiting for removal, but instead release metabolites as 'good-bye' signals to actively modulate outcomes in tissues.


Asunto(s)
Apoptosis/fisiología , Microambiente Celular , Sistemas de Mensajero Secundario/fisiología , Animales , Artritis , Caspasas/metabolismo , Línea Celular , Proliferación Celular/genética , Supervivencia Celular/genética , Conexinas/metabolismo , Modelos Animales de Enfermedad , Rechazo de Injerto , Humanos , Inflamación/genética , Trasplante de Pulmón , Linfocitos/enzimología , Linfocitos/metabolismo , Macrófagos/enzimología , Macrófagos/metabolismo , Ratones , Proteínas del Tejido Nervioso/metabolismo , Fagocitos/metabolismo , Cicatrización de Heridas/genética
5.
Nature ; 578(7795): 419-424, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31996848

RESUMEN

ATP13A2 (PARK9) is a late endolysosomal transporter that is genetically implicated in a spectrum of neurodegenerative disorders, including Kufor-Rakeb syndrome-a parkinsonism with dementia1-and early-onset Parkinson's disease2. ATP13A2 offers protection against genetic and environmental risk factors of Parkinson's disease, whereas loss of ATP13A2 compromises lysosomes3. However, the transport function of ATP13A2 in lysosomes remains unclear. Here we establish ATP13A2 as a lysosomal polyamine exporter that shows the highest affinity for spermine among the polyamines examined. Polyamines stimulate the activity of purified ATP13A2, whereas ATP13A2 mutants that are implicated in disease are functionally impaired to a degree that correlates with the disease phenotype. ATP13A2 promotes the cellular uptake of polyamines by endocytosis and transports them into the cytosol, highlighting a role for endolysosomes in the uptake of polyamines into cells. At high concentrations polyamines induce cell toxicity, which is exacerbated by ATP13A2 loss due to lysosomal dysfunction, lysosomal rupture and cathepsin B activation. This phenotype is recapitulated in neurons and nematodes with impaired expression of ATP13A2 or its orthologues. We present defective lysosomal polyamine export as a mechanism for lysosome-dependent cell death that may be implicated in neurodegeneration, and shed light on the molecular identity of the mammalian polyamine transport system.


Asunto(s)
Lisosomas/metabolismo , Poliaminas/metabolismo , ATPasas de Translocación de Protón/deficiencia , ATPasas de Translocación de Protón/genética , Animales , Biocatálisis , Transporte Biológico , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Catepsina B/metabolismo , Citosol/metabolismo , Modelos Animales de Enfermedad , Endocitosis , Humanos , Lisosomas/patología , Ratones , Mutación , Neuronas/metabolismo , Fenotipo , Poliaminas/toxicidad , ATPasas de Translocación de Protón/metabolismo , Espermidina/metabolismo , Espermina/metabolismo
6.
Hepatology ; 2024 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-38761406

RESUMEN

BACKGROUND AND AIMS: Acute-on-chronic liver failure (ACLF) is a complication of cirrhosis characterized by multiple organ failure and high short-term mortality. The pathophysiology of ACLF involves elevated systemic inflammation leading to organ failure, along with immune dysfunction that heightens susceptibility to bacterial infections. However, it is unclear how these aspects are associated with recovery and nonrecovery in ACLF. APPROACH AND RESULTS: Here, we mapped the single-cell transcriptome of circulating immune cells from patients with ACLF and acute decompensated (AD) cirrhosis and healthy individuals. We further interrogate how these findings, as well as immunometabolic and functional profiles, associate with ACLF-recovery (ACLF-R) or nonrecovery (ACLF-NR). Our analysis unveiled 2 distinct states of classical monocytes (cMons). Hereto, ACLF-R cMons were characterized by transcripts associated with immune and stress tolerance, including anti-inflammatory genes such as RETN and LGALS1 . Additional metabolomic and functional validation experiments implicated an elevated oxidative phosphorylation metabolic program as well as an impaired ACLF-R cMon functionality. Interestingly, we observed a common stress-induced tolerant state, oxidative phosphorylation program, and blunted activation among lymphoid populations in patients with ACLF-R. Conversely, ACLF-NR cMon featured elevated expression of inflammatory and stress response genes such as VIM , LGALS2 , and TREM1 , along with blunted metabolic activity and increased functionality. CONCLUSIONS: This study identifies distinct immunometabolic cellular states that contribute to disease outcomes in patients with ACLF. Our findings provide valuable insights into the pathogenesis of ACLF, shedding light on factors driving either recovery or nonrecovery phenotypes, which may be harnessed as potential therapeutic targets in the future.

7.
Nature ; 565(7740): 511-515, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30651640

RESUMEN

Endochondral ossification, an important process in vertebrate bone formation, is highly dependent on correct functioning of growth plate chondrocytes1. Proliferation of these cells determines longitudinal bone growth and the matrix deposited provides a scaffold for future bone formation. However, these two energy-dependent anabolic processes occur in an avascular environment1,2. In addition, the centre of the expanding growth plate becomes hypoxic, and local activation of the hypoxia-inducible transcription factor HIF-1α is necessary for chondrocyte survival by unidentified cell-intrinsic mechanisms3-6. It is unknown whether there is a requirement for restriction of HIF-1α signalling in the other regions of the growth plate and whether chondrocyte metabolism controls cell function. Here we show that prolonged HIF-1α signalling in chondrocytes leads to skeletal dysplasia by interfering with cellular bioenergetics and biosynthesis. Decreased glucose oxidation results in an energy deficit, which limits proliferation, activates the unfolded protein response and reduces collagen synthesis. However, enhanced glutamine flux increases α-ketoglutarate levels, which in turn increases proline and lysine hydroxylation on collagen. This metabolically regulated collagen modification renders the cartilaginous matrix more resistant to protease-mediated degradation and thereby increases bone mass. Thus, inappropriate HIF-1α signalling results in skeletal dysplasia caused by collagen overmodification, an effect that may also contribute to other diseases involving the extracellular matrix such as cancer and fibrosis.


Asunto(s)
Enfermedades Óseas/metabolismo , Enfermedades Óseas/patología , Condrocitos/metabolismo , Colágeno/biosíntesis , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Animales , Cartílago/metabolismo , Matriz Extracelular/metabolismo , Glucosa/metabolismo , Glutamina/metabolismo , Placa de Crecimiento/metabolismo , Hidroxilación , Prolina Dioxigenasas del Factor Inducible por Hipoxia/deficiencia , Prolina Dioxigenasas del Factor Inducible por Hipoxia/genética , Ácidos Cetoglutáricos/metabolismo , Lisina/metabolismo , Masculino , Ratones , Osteogénesis , Oxidación-Reducción , Prolina/metabolismo
8.
Mol Genet Metab ; 142(1): 108434, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38489976

RESUMEN

Congenital disorders of glycosylation (CDG) are a large family of rare disorders affecting the different glycosylation pathways. Defective glycosylation can affect any organ, with varying symptoms among the different CDG. Even between individuals with the same CDG there is quite variable severity. Associating specific symptoms to deficiencies of certain glycoproteins or glycolipids is thus a challenging task. In this review, we focus on the glycosphingolipid (GSL) synthesis pathway, which is still rather unexplored in the context of CDG, and outline the functions of the main GSLs, including gangliosides, and their role in the central nervous system. We provide an overview of GSL studies that have been performed in CDG and show that abnormal GSL levels are not only observed in CDG directly affecting GSL synthesis, but also in better known CDG, such as PMM2-CDG. We highlight the importance of studying GSLs in CDG in order to better understand the pathophysiology of these disorders.


Asunto(s)
Trastornos Congénitos de Glicosilación , Glicoesfingolípidos , Humanos , Trastornos Congénitos de Glicosilación/genética , Trastornos Congénitos de Glicosilación/metabolismo , Trastornos Congénitos de Glicosilación/patología , Glicoesfingolípidos/metabolismo , Glicosilación , Animales , Gangliósidos/metabolismo , Gangliósidos/deficiencia
10.
EMBO Rep ; 23(1): e53083, 2022 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-34699114

RESUMEN

Here, we investigate the impact of hypoxia on the hepatic response of glucocorticoid receptor (GR) to dexamethasone (DEX) in mice via RNA-sequencing. Hypoxia causes three types of reprogramming of GR: (i) much weaker induction of classical GR-responsive genes by DEX in hypoxia, (ii) a number of genes is induced by DEX specifically in hypoxia, and (iii) hypoxia induces a group of genes via activation of the hypothalamic-pituitary-adrenal (HPA) axis. Transcriptional profiles are reflected by changed GR DNA-binding as measured by ChIP sequencing. The HPA axis is induced by hypothalamic HIF1α and HIF2α activation and leads to GR-dependent lipolysis and ketogenesis. Acute inflammation, induced by lipopolysaccharide, is prevented by DEX in normoxia but not during hypoxia, and this is attributed to HPA axis activation by hypoxia. We unfold new physiological pathways that have consequences for patients suffering from GC resistance.


Asunto(s)
Glucocorticoides , Receptores de Glucocorticoides , Animales , Dexametasona/metabolismo , Dexametasona/farmacología , Glucocorticoides/metabolismo , Glucocorticoides/farmacología , Humanos , Sistema Hipotálamo-Hipofisario/metabolismo , Hipoxia/genética , Hipoxia/metabolismo , Ratones , Sistema Hipófiso-Suprarrenal/metabolismo , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo
11.
Nature ; 558(7711): 605-609, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29925953

RESUMEN

Reprogramming of mRNA translation has a key role in cancer development and drug resistance 1 . However, the molecular mechanisms that are involved in this process remain poorly understood. Wobble tRNA modifications are required for specific codon decoding during translation2,3. Here we show, in humans, that the enzymes that catalyse modifications of wobble uridine 34 (U34) tRNA (U34 enzymes) are key players of the protein synthesis rewiring that is induced by the transformation driven by the BRAF V600E oncogene and by resistance to targeted therapy in melanoma. We show that BRAF V600E -expressing melanoma cells are dependent on U34 enzymes for survival, and that concurrent inhibition of MAPK signalling and ELP3 or CTU1 and/or CTU2 synergizes to kill melanoma cells. Activation of the PI3K signalling pathway, one of the most common mechanisms of acquired resistance to MAPK therapeutic agents, markedly increases the expression of U34 enzymes. Mechanistically, U34 enzymes promote glycolysis in melanoma cells through the direct, codon-dependent, regulation of the translation of HIF1A mRNA and the maintenance of high levels of HIF1α protein. Therefore, the acquired resistance to anti-BRAF therapy is associated with high levels of U34 enzymes and HIF1α. Together, these results demonstrate that U34 enzymes promote the survival and resistance to therapy of melanoma cells by regulating specific mRNA translation.


Asunto(s)
Codón/genética , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Melanoma/tratamiento farmacológico , Melanoma/genética , Biosíntesis de Proteínas , Animales , Proteínas Portadoras/química , Proteínas Portadoras/metabolismo , Línea Celular Tumoral , Codón/efectos de los fármacos , Femenino , Humanos , Masculino , Diana Mecanicista del Complejo 2 de la Rapamicina/metabolismo , Melanoma/patología , Melanoma Experimental/tratamiento farmacológico , Melanoma Experimental/genética , Melanoma Experimental/patología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Fosforilación , Biosíntesis de Proteínas/efectos de los fármacos , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Proteínas Proto-Oncogénicas B-raf/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN de Transferencia/química , ARN de Transferencia/genética , ARN de Transferencia/metabolismo , Transducción de Señal , Factores de Elongación Transcripcional , Uridina/química , Uridina/genética , Uridina/metabolismo , Vemurafenib/farmacología , Vemurafenib/uso terapéutico , Pez Cebra/genética
12.
Nature ; 561(7721): 63-69, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30158707

RESUMEN

Glutamine synthetase, encoded by the gene GLUL, is an enzyme that converts glutamate and ammonia to glutamine. It is expressed by endothelial cells, but surprisingly shows negligible glutamine-synthesizing activity in these cells at physiological glutamine levels. Here we show in mice that genetic deletion of Glul in endothelial cells impairs vessel sprouting during vascular development, whereas pharmacological blockade of glutamine synthetase suppresses angiogenesis in ocular and inflammatory skin disease while only minimally affecting healthy adult quiescent endothelial cells. This relies on the inhibition of endothelial cell migration but not proliferation. Mechanistically we show that in human umbilical vein endothelial cells GLUL knockdown reduces membrane localization and activation of the GTPase RHOJ while activating other Rho GTPases and Rho kinase, thereby inducing actin stress fibres and impeding endothelial cell motility. Inhibition of Rho kinase rescues the defect in endothelial cell migration that is induced by GLUL knockdown. Notably, glutamine synthetase palmitoylates itself and interacts with RHOJ to sustain RHOJ palmitoylation, membrane localization and activation. These findings reveal that, in addition to the known formation of glutamine, the enzyme glutamine synthetase shows unknown activity in endothelial cell migration during pathological angiogenesis through RHOJ palmitoylation.


Asunto(s)
Células Endoteliales/enzimología , Células Endoteliales/patología , Glutamato-Amoníaco Ligasa/metabolismo , Glutamina/biosíntesis , Neovascularización Patológica , Actinas/metabolismo , Animales , Movimiento Celular , Células Endoteliales/metabolismo , Femenino , Glutamato-Amoníaco Ligasa/deficiencia , Glutamato-Amoníaco Ligasa/genética , Glutamato-Amoníaco Ligasa/fisiología , Células HEK293 , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/enzimología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Lipoilación , Ratones , Ácido Palmítico/metabolismo , Procesamiento Proteico-Postraduccional , Fibras de Estrés/metabolismo , Proteínas de Unión al GTP rho/química , Proteínas de Unión al GTP rho/metabolismo , Quinasas Asociadas a rho/metabolismo
13.
Am J Respir Crit Care Med ; 207(8): 998-1011, 2023 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-36724365

RESUMEN

Rationale: Chronic obstructive pulmonary disease (COPD) is a disease characterized by persistent airway inflammation and disordered macrophage function. The extent to which alterations in macrophage bioenergetics contribute to impaired antioxidant responses and disease pathogenesis has yet to be fully delineated. Objectives: Through the study of COPD alveolar macrophages (AMs) and peripheral monocyte-derived macrophages (MDMs), we sought to establish if intrinsic defects in core metabolic processes drive macrophage dysfunction and redox imbalance. Methods: AMs and MDMs from donors with COPD and healthy donors underwent functional, metabolic, and transcriptional profiling. Measurements and Main Results: We observed that AMs and MDMs from donors with COPD display a critical depletion in glycolytic- and mitochondrial respiration-derived energy reserves and an overreliance on glycolysis as a source for ATP, resulting in reduced energy status. Defects in oxidative metabolism extend to an impaired redox balance associated with defective expression of the NADPH-generating enzyme, ME1 (malic enzyme 1), a known target of the antioxidant transcription factor NRF2 (nuclear factor erythroid 2-related factor 2). Consequently, selective activation of NRF2 resets the COPD transcriptome, resulting in increased generation of TCA cycle intermediaries, improved energetic status, favorable redox balance, and recovery of macrophage function. Conclusions: In COPD, an inherent loss of metabolic plasticity leads to metabolic exhaustion and reduced redox capacity, which can be rescued by activation of the NRF2 pathway. Targeting these defects, via NRF2 augmentation, may therefore present an attractive therapeutic strategy for the treatment of the aberrant airway inflammation described in COPD.


Asunto(s)
Factor 2 Relacionado con NF-E2 , Enfermedad Pulmonar Obstructiva Crónica , Humanos , Macrófagos/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Estrés Oxidativo , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/fisiopatología , Malato Deshidrogenasa/metabolismo
14.
Ann Surg ; 2023 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-37870241

RESUMEN

OBJECTIVE: To investigate if ischemia alters donor kidney metabolism and whether these changes associate with organ function. SUMMARY BACKGROUND DATA: An unmet need in kidney transplantation is the ability to predict post-transplant organ function before transplantation. Key to such viability testing is a profound understanding of the organ's complex biochemistry and how ischemia, inevitable during the transplantation process, influences this. METHODS: First, metabolic changes in glucose, lactate and 20 amino acids induced by no, 1h of warm, or 22h of cold ischemia were investigated during 4h perfusion of pig kidneys with autologous whole blood (n=6/group), simulating the ischemia-reperfusion phase of transplantation. Next, we confirmed similar metabolic changes during normothermic preservation of pig (n=3/group; n=4 for cold ischemia) and discarded human kidneys (n=6) perfused with a red-blood cell based perfusate. RESULTS: At 2h of perfusion with autologous whole blood, abundances of 17/20 amino acids were significantly different between groups, reflecting the type of ischemia. Amino acid changes at 15 min and 2h of perfusion correlated with future kidney function during perfusion. Similar metabolic patterns were observed during perfusion preservation of pig and discarded human donor kidneys, suggesting an opportunity to assess kidney viability before transplantation. CONCLUSIONS: Perfusate metabolite changes during normothermic kidney perfusion represent a unique non-invasive opportunity to assess graft viability. These findings now need validation in transplant studies.

15.
Nature ; 542(7639): 49-54, 2017 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-28024299

RESUMEN

Lymphatic vessels are lined by lymphatic endothelial cells (LECs), and are critical for health. However, the role of metabolism in lymphatic development has not yet been elucidated. Here we report that in transgenic mouse models, LEC-specific loss of CPT1A, a rate-controlling enzyme in fatty acid ß-oxidation, impairs lymphatic development. LECs use fatty acid ß-oxidation to proliferate and for epigenetic regulation of lymphatic marker expression during LEC differentiation. Mechanistically, the transcription factor PROX1 upregulates CPT1A expression, which increases acetyl coenzyme A production dependent on fatty acid ß-oxidation. Acetyl coenzyme A is used by the histone acetyltransferase p300 to acetylate histones at lymphangiogenic genes. PROX1-p300 interaction facilitates preferential histone acetylation at PROX1-target genes. Through this metabolism-dependent mechanism, PROX1 mediates epigenetic changes that promote lymphangiogenesis. Notably, blockade of CPT1 enzymes inhibits injury-induced lymphangiogenesis, and replenishing acetyl coenzyme A by supplementing acetate rescues this process in vivo.


Asunto(s)
Ácidos Grasos/química , Ácidos Grasos/metabolismo , Linfangiogénesis , Vasos Linfáticos/citología , Vasos Linfáticos/metabolismo , Acetatos/farmacología , Acetilcoenzima A/metabolismo , Acetilación/efectos de los fármacos , Animales , Carnitina O-Palmitoiltransferasa/antagonistas & inhibidores , Carnitina O-Palmitoiltransferasa/genética , Carnitina O-Palmitoiltransferasa/metabolismo , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Epigénesis Genética , Femenino , Histonas/metabolismo , Proteínas de Homeodominio/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Linfangiogénesis/efectos de los fármacos , Linfangiogénesis/genética , Vasos Linfáticos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Oxidación-Reducción/efectos de los fármacos , Biosíntesis de Proteínas , Transcripción Genética , Proteínas Supresoras de Tumor/metabolismo , Arterias Umbilicales/citología , Regulación hacia Arriba
16.
Proc Natl Acad Sci U S A ; 117(49): 31198-31207, 2020 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-33229544

RESUMEN

Recessive loss-of-function mutations in ATP13A2 (PARK9) are associated with a spectrum of neurodegenerative disorders, including Parkinson's disease (PD). We recently revealed that the late endo-lysosomal transporter ATP13A2 pumps polyamines like spermine into the cytosol, whereas ATP13A2 dysfunction causes lysosomal polyamine accumulation and rupture. Here, we investigate how ATP13A2 provides protection against mitochondrial toxins such as rotenone, an environmental PD risk factor. Rotenone promoted mitochondrial-generated superoxide (MitoROS), which was exacerbated by ATP13A2 deficiency in SH-SY5Y cells and patient-derived fibroblasts, disturbing mitochondrial functionality and inducing toxicity and cell death. Moreover, ATP13A2 knockdown induced an ATF4-CHOP-dependent stress response following rotenone exposure. MitoROS and ATF4-CHOP were blocked by MitoTEMPO, a mitochondrial antioxidant, suggesting that the impact of ATP13A2 on MitoROS may relate to the antioxidant properties of spermine. Pharmacological inhibition of intracellular polyamine synthesis with α-difluoromethylornithine (DFMO) also increased MitoROS and ATF4 when ATP13A2 was deficient. The polyamine transport activity of ATP13A2 was required for lowering rotenone/DFMO-induced MitoROS, whereas exogenous spermine quenched rotenone-induced MitoROS via ATP13A2. Interestingly, fluorescently labeled spermine uptake in the mitochondria dropped as a consequence of ATP13A2 transport deficiency. Our cellular observations were recapitulated in vivo, in a Caenorhabditis elegans strain deficient in the ATP13A2 ortholog catp-6 These animals exhibited a basal elevated MitoROS level, mitochondrial dysfunction, and enhanced stress response regulated by atfs-1, the C. elegans ortholog of ATF4, causing hypersensitivity to rotenone, which was reversible with MitoTEMPO. Together, our study reveals a conserved cell protective pathway that counters mitochondrial oxidative stress via ATP13A2-mediated lysosomal spermine export.


Asunto(s)
Factor de Transcripción Activador 4/genética , Adenosina Trifosfatasas/genética , Proteínas de Caenorhabditis elegans/genética , Mitocondrias/genética , ATPasas de Translocación de Protón/genética , Factores de Transcripción/genética , Animales , Caenorhabditis elegans , Eflornitina/farmacología , Fibroblastos/efectos de los fármacos , Lisosomas/genética , Lisosomas/metabolismo , Mitocondrias/efectos de los fármacos , Mitocondrias/patología , Mutación/genética , Estrés Oxidativo/efectos de los fármacos , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/patología , Poliaminas/metabolismo , Rotenona/farmacología , Espermina/metabolismo , Factor de Transcripción CHOP/genética
17.
Am J Hum Genet ; 104(5): 835-846, 2019 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-30982613

RESUMEN

Phosphoglucomutase 1 (PGM1) encodes the metabolic enzyme that interconverts glucose-6-P and glucose-1-P. Mutations in PGM1 cause impairment in glycogen metabolism and glycosylation, the latter manifesting as a congenital disorder of glycosylation (CDG). This unique metabolic defect leads to abnormal N-glycan synthesis in the endoplasmic reticulum (ER) and the Golgi apparatus (GA). On the basis of the decreased galactosylation in glycan chains, galactose was administered to individuals with PGM1-CDG and was shown to markedly reverse most disease-related laboratory abnormalities. The disease and treatment mechanisms, however, have remained largely elusive. Here, we confirm the clinical benefit of galactose supplementation in PGM1-CDG-affected individuals and obtain significant insights into the functional and biochemical regulation of glycosylation. We report here that, by using tracer-based metabolomics, we found that galactose treatment of PGM1-CDG fibroblasts metabolically re-wires their sugar metabolism, and as such replenishes the depleted levels of galactose-1-P, as well as the levels of UDP-glucose and UDP-galactose, the nucleotide sugars that are required for ER- and GA-linked glycosylation, respectively. To this end, we further show that the galactose in UDP-galactose is incorporated into mature, de novo glycans. Our results also allude to the potential of monosaccharide therapy for several other CDG.


Asunto(s)
Trastornos Congénitos de Glicosilación/metabolismo , Fibroblastos/metabolismo , Galactosa/administración & dosificación , Fosfoglucomutasa/deficiencia , Uridina Difosfato Galactosa/metabolismo , Uridina Difosfato Glucosa/metabolismo , Células Cultivadas , Estudios de Cohortes , Trastornos Congénitos de Glicosilación/tratamiento farmacológico , Trastornos Congénitos de Glicosilación/patología , Fibroblastos/efectos de los fármacos , Fibroblastos/patología , Glicosilación , Humanos
18.
Genet Med ; 24(4): 894-904, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35042660

RESUMEN

PURPOSE: TRAPPC9 deficiency is an autosomal recessive disorder mainly associated with intellectual disability (ID), microcephaly, and obesity. Previously, TRAPPC9 deficiency has not been associated with biochemical abnormalities. METHODS: Exome sequencing was performed in 3 individuals with ID and dysmorphic features. N-Glycosylation analyses were performed in the patients' blood samples to test for possible congenital disorder of glycosylation (CDG). TRAPPC9 gene, TRAPPC9 protein expression, and N-glycosylation markers were assessed in patient fibroblasts. Complementation with wild-type TRAPPC9 and immunofluorescence studies to assess TRAPPC9 expression and localization were performed. The metabolic consequences of TRAPPC9 deficiency were evaluated using tracer metabolomics. RESULTS: All 3 patients carried biallelic missense variants in TRAPPC9 and presented with an N-glycosylation defect in blood, consistent with CDG type I. Extensive investigations in patient fibroblasts corroborated TRAPPC9 deficiency and an N-glycosylation defect. Tracer metabolomics revealed global metabolic changes with several affected glycosylation-related metabolites. CONCLUSION: We identified 3 TRAPPC9 deficient patients presenting with ID, dysmorphic features, and abnormal glycosylation. On the basis of our findings, we propose that TRAPPC9 deficiency could lead to a CDG (TRAPPC9-CDG). The finding of abnormal glycosylation in these patients is highly relevant for diagnosis, further elucidation of the pathophysiology, and management of the disease.


Asunto(s)
Trastornos Congénitos de Glicosilación , Discapacidad Intelectual , Microcefalia , Trastornos Congénitos de Glicosilación/genética , Glicosilación , Humanos , Discapacidad Intelectual/complicaciones , Discapacidad Intelectual/genética , Microcefalia/genética , Mutación Missense
19.
Ann Neurol ; 90(6): 887-900, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34652821

RESUMEN

OBJECTIVE: Epalrestat, an aldose reductase inhibitor increases phosphomannomutase (PMM) enzyme activity in a PMM2-congenital disorders of glycosylation (CDG) worm model. Epalrestat also decreases sorbitol level in diabetic neuropathy. We evaluated the genetic, biochemical, and clinical characteristics, including the Nijmegen Progression CDG Rating Scale (NPCRS), urine polyol levels and fibroblast glycoproteomics in patients with PMM2-CDG. METHODS: We performed PMM enzyme measurements, multiplexed proteomics, and glycoproteomics in PMM2-deficient fibroblasts before and after epalrestat treatment. Safety and efficacy of 0.8 mg/kg/day oral epalrestat were studied in a child with PMM2-CDG for 12 months. RESULTS: PMM enzyme activity increased post-epalrestat treatment. Compared with controls, 24% of glycopeptides had reduced abundance in PMM2-deficient fibroblasts, 46% of which improved upon treatment. Total protein N-glycosylation improved upon epalrestat treatment bringing overall glycosylation toward the control fibroblasts' glycosylation profile. Sorbitol levels were increased in the urine of 74% of patients with PMM2-CDG and correlated with the presence of peripheral neuropathy, and CDG severity rating scale. In the child with PMM2-CDG on epalrestat treatment, ataxia scores improved together with significant growth improvement. Urinary sorbitol levels nearly normalized in 3 months and blood transferrin glycosylation normalized in 6 months. INTERPRETATION: Epalrestat improved PMM enzyme activity, N-glycosylation, and glycosylation biomarkers in vitro. Leveraging cellular glycoproteome assessment, we provided a systems-level view of treatment efficacy and discovered potential novel biosignatures of therapy response. Epalrestat was well-tolerated and led to significant clinical improvements in the first pediatric patient with PMM2-CDG treated with epalrestat. We also propose urinary sorbitol as a novel biomarker for disease severity and treatment response in future clinical trials in PMM2-CDG. ANN NEUROL 20219999:n/a-n/a.


Asunto(s)
Trastornos Congénitos de Glicosilación/diagnóstico , Inhibidores Enzimáticos/uso terapéutico , Fosfotransferasas (Fosfomutasas)/deficiencia , Rodanina/análogos & derivados , Sorbitol/orina , Tiazolidinas/uso terapéutico , Adolescente , Adulto , Anciano , Biomarcadores/orina , Niño , Preescolar , Trastornos Congénitos de Glicosilación/tratamiento farmacológico , Trastornos Congénitos de Glicosilación/orina , Femenino , Glicosilación , Humanos , Lactante , Masculino , Persona de Mediana Edad , Gravedad del Paciente , Fosfotransferasas (Fosfomutasas)/orina , Pronóstico , Rodanina/uso terapéutico , Adulto Joven
20.
Diabetes Obes Metab ; 24(11): 2263-2272, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35801343

RESUMEN

AIM: To investigate cardiac signalling pathways connecting substrate utilization with left ventricular remodelling in a murine pressure overload model. METHODS: Cardiac hypertrophy was induced by transverse aortic constriction surgery in 20-week-old C57BL/6J mice treated with or without the sodium-glucose co-transporter 2 (SGLT2) inhibitor ertugliflozin (225 mg kg-1 chow diet) for 10 weeks. RESULTS: Ertugliflozin improved left ventricular function and reduced myocardial fibrosis. This occurred simultaneously with a fasting-like response characterized by improved glucose tolerance and increased ketone body concentrations. While cardiac insulin signalling was reduced in response to SGLT2 inhibition, AMP-activated protein kinase (AMPK) signalling was increased with induction of the fatty acid transporter cluster of differentiation 36 and phosphorylation of acetyl-CoA carboxylase (ACC). Further, enzymes responsible for ketone body catabolism (ß-hydroxybutyrate dehydrogenase, succinyl-CoA:3-oxoacid-CoA transferase and acetyl-CoA acetyltransferase 1) were induced by SGLT2 inhibition. Ertugliflozin led to more cardiac abundance of fatty acids, tricarboxylic acid cycle metabolites and ATP. Downstream mechanistic target of rapamycin (mTOR) pathway, relevant for protein synthesis, cardiac hypertrophy and adverse cardiac remodelling, was reduced by SGLT2 inhibition, with alleviation of endoplasmic reticulum (ER) stress and unfolded protein response (UPR) providing a potential mechanism for abundant reduced left ventricular apoptosis and fibrosis. CONCLUSION: SGLT2 inhibition reduced left ventricular fibrosis in a murine model of cardiac hypertrophy. Mechanistically, this was associated with reduced cardiac insulin and increased AMPK signalling as a potential mechanism for less cardiac mTOR activation with alleviation of downstream ER stress, UPR and apoptosis.


Asunto(s)
Insulinas , Inhibidores del Cotransportador de Sodio-Glucosa 2 , Proteínas Quinasas Activadas por AMP/metabolismo , Acetil-CoA C-Acetiltransferasa/metabolismo , Acetil-CoA Carboxilasa/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Apoptosis , Compuestos Bicíclicos Heterocíclicos con Puentes , Cardiomegalia/metabolismo , Cardiomegalia/patología , Coenzima A Transferasas/metabolismo , Estrés del Retículo Endoplásmico , Ácidos Grasos/metabolismo , Fibrosis , Glucosa/metabolismo , Hidroxibutirato Deshidrogenasa/metabolismo , Cetoácidos/metabolismo , Cetonas/metabolismo , Ratones , Ratones Endogámicos C57BL , Miocitos Cardíacos/metabolismo , Sirolimus/metabolismo , Sodio/metabolismo , Transportador 2 de Sodio-Glucosa/metabolismo , Inhibidores del Cotransportador de Sodio-Glucosa 2/farmacología , Inhibidores del Cotransportador de Sodio-Glucosa 2/uso terapéutico , Serina-Treonina Quinasas TOR/metabolismo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda