Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Cell Rep ; 43(4): 114103, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38607920

ABSTRACT

Hypoxia-inducible factor-1α (HIF1α) attenuates mitochondrial activity while promoting glycolysis. However, lower glycolysis is compromised in human clear cell renal cell carcinomas, in which HIF1α acts as a tumor suppressor by inhibiting cell-autonomous proliferation. Here, we find that, unexpectedly, HIF1α suppresses lower glycolysis after the glyceraldehyde 3-phosphate dehydrogenase (GAPDH) step, leading to reduced lactate secretion in different tumor cell types when cells encounter a limited pyruvate supply such as that typically found in the tumor microenvironment in vivo. This is because HIF1α-dependent attenuation of mitochondrial oxygen consumption increases the NADH/NAD+ ratio that suppresses the activity of the NADH-sensitive GAPDH glycolytic enzyme. This is manifested when pyruvate supply is limited, since pyruvate acts as an electron acceptor that prevents the increment of the NADH/NAD+ ratio. Furthermore, this anti-glycolytic function provides a molecular basis to explain how HIF1α can suppress tumor cell proliferation by increasing the NADH/NAD+ ratio.


Subject(s)
Cell Proliferation , Glycolysis , Hypoxia-Inducible Factor 1, alpha Subunit , NAD , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , NAD/metabolism , Cell Line, Tumor , Mitochondria/metabolism , Animals , Pyruvic Acid/metabolism , Lactic Acid/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Mice
2.
Commun Biol ; 6(1): 111, 2023 01 27.
Article in English | MEDLINE | ID: mdl-36707617

ABSTRACT

Fatty infiltration, the ectopic deposition of adipose tissue within skeletal muscle, is mediated via the adipogenic differentiation of fibro-adipogenic progenitors (FAPs). We used single-nuclei and single-cell RNA sequencing to characterize FAP heterogeneity in patients with fatty infiltration. We identified an MME+ FAP subpopulation which, based on ex vivo characterization as well as transplantation experiments, exhibits high adipogenic potential. MME+ FAPs are characterized by low activity of WNT, known to control adipogenic commitment, and are refractory to the inhibitory role of WNT activators. Using preclinical models for muscle damage versus fatty infiltration, we show that many MME+ FAPs undergo apoptosis during muscle regeneration and differentiate into adipocytes under pathological conditions, leading to a reduction in their abundance. Finally, we utilized the varying fat infiltration levels in human hip muscles and found less MME+ FAPs in fatty infiltrated human muscle. Altogether, we have identified the dominant adipogenic FAP subpopulation in skeletal muscle.


Subject(s)
Adipogenesis , Muscle, Skeletal , Humans , Cell Differentiation/physiology , Adipocytes
3.
J Inherit Metab Dis ; 45(2): 278-291, 2022 03.
Article in English | MEDLINE | ID: mdl-34936099

ABSTRACT

Creatine (Cr) is a nitrogenous organic acid and plays roles such as fast phosphate energy buffer to replenish ATP, osmolyte, antioxidant, neuromodulator, and as a compound with anabolic and ergogenic properties in muscle. Cr is taken from the diet or endogenously synthetized by the enzymes arginine:glycine amidinotransferase and guanidinoacetate methyltransferase, and specifically taken up by the transporter SLC6A8. Loss-of-function mutations in the genes encoding for the enzymes or the transporter cause creatine deficiency syndromes (CDS). CDS are characterized by brain Cr deficiency, intellectual disability with severe speech delay, behavioral troubles, epilepsy, and motor dysfunction. Among CDS, the X-linked Cr transporter deficiency (CTD) is the most prevalent with no efficient treatment so far. Different animal models of CTD show reduced brain Cr levels, cognitive deficiencies, and together they cover other traits similar to those of patients. However, motor function was poorly explored in CTD models, and some controversies in the phenotype exist in comparison with CTD patients. Our recently described Slc6a8Y389C knock-in rat model of CTD showed mild impaired motor function, morphological alterations in cerebellum, reduced muscular mass, Cr deficiency, and increased guanidinoacetate content in muscle, although no consistent signs of muscle atrophy. Our results indicate that such motor dysfunction co-occurred with both nervous and muscle dysfunctions, suggesting that muscle strength and performance as well as neuronal connectivity might be affected by this Cr deficiency in muscle and brain.


Subject(s)
Cerebellar Diseases , Creatine , Animals , Cerebellum/metabolism , Guanidinoacetate N-Methyltransferase/genetics , Humans , Membrane Transport Proteins , Muscles/metabolism , Muscular Atrophy , Rats , Syndrome
4.
Skelet Muscle ; 10(1): 21, 2020 07 09.
Article in English | MEDLINE | ID: mdl-32646489

ABSTRACT

BACKGROUND: Satellite cells (SCs) are required for muscle repair following injury and are involved in muscle remodeling upon muscular contractions. Exercise stimulates SC accumulation and myonuclear accretion. To what extent exercise training at different mechanical loads drive SC contribution to myonuclei however is unknown. RESULTS: By performing SC fate tracing experiments, we show that 8 weeks of voluntary wheel running increased SC contribution to myofibers in mouse plantar flexor muscles in a load-dependent, but fiber type-independent manner. Increased SC fusion however was not exclusively linked to muscle hypertrophy as wheel running without external load substantially increased SC fusion in the absence of fiber hypertrophy. Due to nuclear propagation, nuclear fluorescent fate tracing mouse models were inadequate to quantify SC contribution to myonuclei. Ultimately, by performing fate tracing at the DNA level, we show that SC contribution mirrors myonuclear accretion during exercise. CONCLUSIONS: Collectively, mechanical load during exercise independently promotes SC contribution to existing myofibers. Also, due to propagation of nuclear fluorescent reporter proteins, our data warrant caution for the use of existing reporter mouse models for the quantitative evaluation of satellite cell contribution to myonuclei.


Subject(s)
Cell Fusion , Muscle Fibers, Skeletal/cytology , Running , Satellite Cells, Skeletal Muscle/cytology , Animals , Cell Nucleus/physiology , Cells, Cultured , Mice , Mice, Inbred C57BL , Muscle Fibers, Skeletal/physiology , Satellite Cells, Skeletal Muscle/physiology
5.
Cell Metab ; 31(6): 1136-1153.e7, 2020 06 02.
Article in English | MEDLINE | ID: mdl-32492393

ABSTRACT

Endothelial cell (EC)-derived signals contribute to organ regeneration, but angiocrine metabolic communication is not described. We found that EC-specific loss of the glycolytic regulator pfkfb3 reduced ischemic hindlimb revascularization and impaired muscle regeneration. This was caused by the reduced ability of macrophages to adopt a proangiogenic and proregenerative M2-like phenotype. Mechanistically, loss of pfkfb3 reduced lactate secretion by ECs and lowered lactate levels in the ischemic muscle. Addition of lactate to pfkfb3-deficient ECs restored M2-like polarization in an MCT1-dependent fashion. Lactate shuttling by ECs enabled macrophages to promote proliferation and fusion of muscle progenitors. Moreover, VEGF production by lactate-polarized macrophages was increased, resulting in a positive feedback loop that further stimulated angiogenesis. Finally, increasing lactate levels during ischemia rescued macrophage polarization and improved muscle reperfusion and regeneration, whereas macrophage-specific mct1 deletion prevented M2-like polarization. In summary, ECs exploit glycolysis for angiocrine lactate shuttling to steer muscle regeneration from ischemia.


Subject(s)
Endothelial Cells/chemistry , Ischemia/metabolism , Lactates/pharmacology , Macrophages/drug effects , Muscle, Skeletal/drug effects , Animals , Cells, Cultured , Ischemia/pathology , Macrophage Activation/drug effects , Macrophages/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Muscle, Skeletal/metabolism
6.
Nat Commun ; 11(1): 174, 2020 01 10.
Article in English | MEDLINE | ID: mdl-31924757

ABSTRACT

mTORC1 is an important regulator of muscle mass but how it is modulated by oxygen and nutrients is not completely understood. We show that loss of the prolyl hydroxylase domain isoform 1 oxygen sensor in mice (PHD1KO) reduces muscle mass. PHD1KO muscles show impaired mTORC1 activation in response to leucine whereas mTORC1 activation by growth factors or eccentric contractions was preserved. The ability of PHD1 to promote mTORC1 activity is independent of its hydroxylation activity but is caused by decreased protein content of the leucyl tRNA synthetase (LRS) leucine sensor. Mechanistically, PHD1 interacts with and stabilizes LRS. This interaction is promoted during oxygen and amino acid depletion and protects LRS from degradation. Finally, elderly subjects have lower PHD1 levels and LRS activity in muscle from aged versus young human subjects. In conclusion, PHD1 ensures an optimal mTORC1 response to leucine after episodes of metabolic scarcity.


Subject(s)
Leucine-tRNA Ligase/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Muscles/metabolism , Procollagen-Proline Dioxygenase/metabolism , Adult , Aged , Aging/metabolism , Amino Acids/metabolism , Animals , Disease Models, Animal , Female , HEK293 Cells , Humans , Hydroxylation , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Leucine/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle Development , Muscles/pathology , Oxygen/metabolism , Procollagen-Proline Dioxygenase/genetics , Signal Transduction
7.
Front Cell Dev Biol ; 6: 100, 2018.
Article in English | MEDLINE | ID: mdl-30255018

ABSTRACT

Endothelial cells (ECs) make up the lining of our blood vessels and they ensure optimal nutrient and oxygen delivery to the parenchymal tissue. In response to oxygen and/or nutrient deprivation, ECs become activated and sprout into hypo-vascularized tissues forming new vascular networks in a process termed angiogenesis. New sprouts are led by migratory tip cells and extended through the proliferation of trailing stalk cells. Activated ECs rewire their metabolism to cope with the increased energetic and biosynthetic demands associated with migration and proliferation. Moreover, metabolic signaling pathways interact and integrate with angiogenic signaling events. These metabolic adaptations play essential roles in determining EC fate and function, and are perturbed during pathological angiogenesis, as occurs in cancer. The angiogenic switch, or the growth of new blood vessels into an expanding tumor, increases tumor growth and malignancy. Limiting tumor angiogenesis has therefore long been a goal for anticancer therapy but the traditional growth factor targeted anti-angiogenic treatments have met with limited success. In recent years however, it has become increasingly recognized that focusing on altered tumor EC metabolism provides an attractive alternative anti-angiogenic strategy. In this review, we will describe the EC metabolic signature and how changes in EC metabolism affect EC fate during physiological sprouting, as well as in the cancer setting. Then, we will discuss the potential of targeting EC metabolism as a promising approach to develop new anti-cancer therapies.

8.
Adipocyte ; 6(2): 161-166, 2017 04 03.
Article in English | MEDLINE | ID: mdl-28425849

ABSTRACT

Aging is associated with progressive visceral white adipose tissue (WAT) expansion both in human and mouse. Importantly, WAT enlargement is initiated early in life, suggesting that molecular mechanisms underlying age-dependent obesity are activated at early stages of lifetime. Our recent study found that age-dependent obesity was associated with a specific decline in mitochondrial complex IV activity, which leads to reduced fatty acid oxidation and subsequent adipocyte hypertrophy. At the molecular level, global mitochondrial complex IV inhibition was driven by hypoxia-inducible factor-1α (HIF1α)-mediated repression of some of its key subunits, including cytochrome c oxidase 5b (Cox5b). In this commentary, we compare age-dependent WAT responses with those observed in the high fat diet model of extreme obesity. Furthermore, we discuss the potential scenarios that could initiate age-dependent WAT expansion as well as the mechanisms by which HIF1α could be activated in WAT.


Subject(s)
Mitochondria/metabolism , Obesity/metabolism , Adipocytes/metabolism , Adipose Tissue, White/metabolism , Age Factors , Animals , Diet, High-Fat , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Intra-Abdominal Fat/metabolism , Lipid Metabolism , Mice , Mitochondrial Proteins/metabolism , Obesity/genetics
9.
Cell Rep ; 16(11): 2991-3002, 2016 09 13.
Article in English | MEDLINE | ID: mdl-27626667

ABSTRACT

Aging is associated with progressive white adipose tissue (WAT) enlargement initiated early in life, but the molecular mechanisms involved remain unknown. Here we show that mitochondrial complex IV (CIV) activity and assembly are already repressed in white adipocytes of middle-aged mice and involve a HIF1A-dependent decline of essential CIV components such as COX5B. At the molecular level, HIF1A binds to the Cox5b proximal promoter and represses its expression. Silencing of Cox5b decreased fatty acid oxidation and promoted intracellular lipid accumulation. Moreover, local in vivo Cox5b silencing in WAT of young mice increased the size of adipocytes, whereas restoration of COX5B expression in aging mice counteracted adipocyte enlargement. An age-dependent reduction in COX5B gene expression was also found in human visceral adipose tissue. Collectively, our findings establish a pivotal role for CIV dysfunction in progressive white adipocyte enlargement during aging, which can be restored to alleviate age-dependent WAT expansion.


Subject(s)
Aging/metabolism , Electron Transport Complex IV/metabolism , Mitochondria/metabolism , Obesity/metabolism , Adipocytes, White/metabolism , Adipose Tissue, White/metabolism , Animals , Cell Size , Epididymis/metabolism , Gene Expression Regulation , Gene Silencing , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Male , Mice , Obesity/genetics , Obesity/pathology , Promoter Regions, Genetic/genetics , Protein Binding/genetics
10.
Mol Cell ; 48(5): 681-91, 2012 Dec 14.
Article in English | MEDLINE | ID: mdl-23103253

ABSTRACT

The mammalian target of rapamycin (mTOR) pathway, which is essential for cell proliferation, is repressed in certain cell types in hypoxia. However, hypoxia-inducible factor 2α (HIF2α) can act as a proliferation-promoting factor in some biological settings. This paradoxical situation led us to study whether HIF2α has a specific effect on mTORC1 regulation. Here we show that activation of the HIF2α pathway increases mTORC1 activity by upregulating expression of the amino acid carrier SLC7A5. At the molecular level we also show that HIF2α binds to the Slc7a5 proximal promoter. Our findings identify a link between the oxygen-sensing HIF2α pathway and mTORC1 regulation, revealing the molecular basis of the tumor-promoting properties of HIF2α in von Hippel-Lindau-deficient cells. We also describe relevant physiological scenarios, including those that occur in liver and lung tissue, wherein HIF2α or low-oxygen tension drive mTORC1 activity and SLC7A5 expression.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Carcinoma, Renal Cell/metabolism , Kidney Neoplasms/metabolism , Large Neutral Amino Acid-Transporter 1/metabolism , Proteins/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Binding Sites , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Cell Hypoxia , Cell Line, Tumor , Cell Proliferation , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Large Neutral Amino Acid-Transporter 1/genetics , Liver/metabolism , Lung/metabolism , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Knockout , Mice, SCID , Multiprotein Complexes , Neoplasm Transplantation , Promoter Regions, Genetic , Proteins/genetics , RNA Interference , Signal Transduction , TOR Serine-Threonine Kinases , Time Factors , Transfection , Tumor Burden , Up-Regulation , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Von Hippel-Lindau Tumor Suppressor Protein/metabolism
11.
PLoS One ; 6(7): e22589, 2011.
Article in English | MEDLINE | ID: mdl-21811636

ABSTRACT

Von Hippel Lindau (Vhl) gene inactivation results in embryonic lethality. The consequences of its inactivation in adult mice, and of the ensuing activation of the hypoxia-inducible factors (HIFs), have been explored mainly in a tissue-specific manner. This mid-gestation lethality can be also circumvented by using a floxed Vhl allele in combination with an ubiquitous tamoxifen-inducible recombinase Cre-ER(T2). Here, we characterize a widespread reduction in Vhl gene expression in Vhl(floxed)-UBC-Cre-ER(T2) adult mice after dietary tamoxifen administration, a convenient route of administration that has yet to be fully characterized for global gene inactivation. Vhl gene inactivation rapidly resulted in a marked splenomegaly and skin erythema, accompanied by renal and hepatic induction of the erythropoietin (Epo) gene, indicative of the in vivo activation of the oxygen sensing HIF pathway. We show that acute Vhl gene inactivation also induced Epo gene expression in the heart, revealing cardiac tissue to be an extra-renal source of EPO. Indeed, primary cardiomyocytes and HL-1 cardiac cells both induce Epo gene expression when exposed to low O(2) tension in a HIF-dependent manner. Thus, as well as demonstrating the potential of dietary tamoxifen administration for gene inactivation studies in UBC-Cre-ER(T2) mouse lines, this data provides evidence of a cardiac oxygen-sensing VHL/HIF/EPO pathway in adult mice.


Subject(s)
Erythropoietin/genetics , Gene Silencing , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Myocardium/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Animals , Animals, Newborn , Body Weight/drug effects , Cells, Cultured , Diet , Erythropoietin/metabolism , Gene Expression Regulation/drug effects , Gene Silencing/drug effects , Glucose Transporter Type 1/genetics , Glucose Transporter Type 1/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Integrases/metabolism , Mice , Myocardium/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Organ Specificity/drug effects , Tamoxifen/administration & dosage , Tamoxifen/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...