Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 47
1.
Nat Prod Bioprospect ; 14(1): 24, 2024 Apr 01.
Article En | MEDLINE | ID: mdl-38556609

Although non-alcoholic fatty liver disease (NAFLD) presents as an intricate condition characterized by a growing prevalence, the often-recommended lifestyle interventions mostly lack high-level evidence of efficacy and there are currently no effective drugs proposed for this indication. The present review delves into NAFLD pathology, its diverse underlying physiopathological mechanisms and the available in vitro, in vivo, and clinical evidence regarding the use of natural compounds for its management, through three pivotal targets (oxidative stress, cellular inflammation, and insulin resistance). The promising perspectives that natural compounds offer for NAFLD management underscore the need for additional clinical and lifestyle intervention trials. Encouraging further research will contribute to establishing more robust evidence and practical recommendations tailored to patients with varying NAFLD grades.

2.
Int J Mol Sci ; 25(6)2024 Mar 15.
Article En | MEDLINE | ID: mdl-38542301

FacioScapuloHumeral muscular Dystrophy (FSHD) is one of the most prevalent inherited muscle disorders and is linked to the inappropriate expression of the DUX4 transcription factor in skeletal muscles. The deregulated molecular network causing FSHD muscle dysfunction and pathology is not well understood. It has been shown that the hypoxia response factor HIF1α is critically disturbed in FSHD and has a major role in DUX4-induced cell death. In this study, we further explored the relationship between DUX4 and HIF1α. We found that the DUX4 and HIF1α link differed according to the stage of myogenic differentiation and was conserved between human and mouse muscle. Furthermore, we found that HIF1α knockdown in a mouse model of DUX4 local expression exacerbated DUX4-mediated muscle fibrosis. Our data indicate that the suggested role of HIF1α in DUX4 toxicity is complex and that targeting HIF1α might be challenging in the context of FSHD therapeutic approaches.


Muscular Dystrophy, Facioscapulohumeral , Animals , Humans , Mice , Cell Differentiation/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Muscle Cells/metabolism , Muscle, Skeletal/metabolism , Muscular Dystrophy, Facioscapulohumeral/genetics , Muscular Dystrophy, Facioscapulohumeral/metabolism
3.
Semin Dial ; 37(2): 145-152, 2024.
Article En | MEDLINE | ID: mdl-37718617

INTRODUCTION: COVID-19 vaccine was demonstrated to be effective in dialysis patients, but boosters are mandatory due to a rapid waning of anti-spike antibodies. A vaccination strategy based on immunologic response might be useful to maintain a favorable risk-benefit balance in this vulnerable population. METHODS: CoviDial is an observational prospective study enrolling 121 dialysis patients to receive a 3-dose mRNA-1273 vaccine according to a uniform schedule. At baseline, months 1, 3, 6, 9, and 12, anti-spike antibodies against four epitopes (S1, S2, ECD-S1 + S2, RBD) were monitored with a multiplex immunodot enzymatic assay. Potential correlation between initial serologic response and subsequent COVID-19 infection was then assessed. RESULTS: Overall, 96.2% and 96.8% of patients had anti-RBD antibodies at 3 and 12 months, respectively. All antibodies titers significantly decreased at month 6 compared to month 3. Booster vaccine induced a robust serologic response at month 9, but with a waning 3 months later, particularly for anti-S2 (37.2 ± 3.3 vs. 61.3 ± 3.0, p < 0.0001) and anti-S1 + S2 antibodies (68.4 ± 3.3 vs. 88.4 ± 2.3, p = 0.0015). Fifteen patients were later tested positive for SARS-CoV-2. At month 3, mean titers of anti-RBD, anti-S1 + S2, and anti-S2 antibodies were lower in the subsequent SARS-CoV-2 infected cohort (71.57 ± 9.01 vs. 85.79 ± 2.61, p = 0.0131; 41.07 ± 7.96 vs. 61.68 ± 3.56, p = 0.0237; 13.79 ± 5.03 vs. 39.70 ± 3.86, p = 0.0096; respectively). CONCLUSION: Three doses of mRNA-1273 vaccine induce a robust but time-limited immunologic response in dialysis patients. Lower anti-spike antibodies titers after initial vaccination are associated with a higher risk to subsequently contract SARS-CoV-2, even beyond 6 months.


COVID-19 Vaccines , COVID-19 , Humans , 2019-nCoV Vaccine mRNA-1273 , Renal Dialysis , Prospective Studies , COVID-19/prevention & control , SARS-CoV-2 , Vaccination
4.
Skelet Muscle ; 13(1): 21, 2023 12 16.
Article En | MEDLINE | ID: mdl-38104132

BACKGROUND: Hypoxia is known to modify skeletal muscle biological functions and muscle regeneration. However, the mechanisms underlying the effects of hypoxia on human myoblast differentiation remain unclear. The hypoxic response pathway is of particular interest in patients with hereditary muscular dystrophies since many present respiratory impairment and muscle regeneration defects. For example, an altered hypoxia response characterizes the muscles of patients with facioscapulohumeral dystrophy (FSHD). METHODS: We examined the impact of hypoxia on the differentiation of human immortalized myoblasts (LHCN-M2) cultured in normoxia (PO2: 21%) or hypoxia (PO2: 1%). Cells were grown in proliferation (myoblasts) or differentiation medium for 2 (myocytes) or 4 days (myotubes). We evaluated proliferation rate by EdU incorporation, used myogenin-positive nuclei as a differentiation marker for myocytes, and determined the fusion index and myosin heavy chain-positive area in myotubes. The contribution of HIF1α was studied by gain (CoCl2) and loss (siRNAs) of function experiments. We further examined hypoxia in LHCN-M2-iDUX4 myoblasts with inducible expression of DUX4, the transcription factor underlying FSHD pathology. RESULTS: We found that the hypoxic response did not impact myoblast proliferation but activated precocious myogenic differentiation and that HIF1α was critical for this process. Hypoxia also enhanced the late differentiation of human myocytes, but in an HIF1α-independent manner. Interestingly, the impact of hypoxia on muscle cell proliferation was influenced by dexamethasone. In the FSHD pathological context, DUX4 suppressed HIF1α-mediated precocious muscle differentiation. CONCLUSION: Hypoxia stimulates myogenic differentiation in healthy myoblasts, with HIF1α-dependent early steps. In FSHD, DUX4-HIF1α interplay indicates a novel mechanism by which DUX4 could interfere with HIF1α function in the myogenic program and therefore with FSHD muscle performance and regeneration.


Homeodomain Proteins , Hypoxia-Inducible Factor 1, alpha Subunit , Muscular Dystrophy, Facioscapulohumeral , Humans , Cell Differentiation , Gene Expression Regulation , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/metabolism , Muscular Dystrophy, Facioscapulohumeral/metabolism , Myoblasts/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
5.
Biol Sex Differ ; 14(1): 63, 2023 09 28.
Article En | MEDLINE | ID: mdl-37770988

BACKGROUND: Sex differences have been observed in the development of obesity-related complications in patients, as well as in animal models. Accumulating evidence suggests that sex-dependent regulation of lipid metabolism contributes to sex-specific physiopathology. Lipid accumulation in the renal tissue has been shown to play a major role in the pathogenesis of obesity-induced kidney injury. Unlike in males, the physiopathology of the disease has been poorly described in females, particularly regarding the lipid metabolism adaptation. METHODS: Here, we compared the lipid profile changes in the kidneys of female and male mice fed a high-fat diet (HFD) or low-fat diet (LFD) by lipidomics and correlated them with pathophysiological changes. RESULTS: We showed that HFD-fed female mice were protected from insulin resistance and hepatic steatosis compared to males, despite similar body weight gains. Females were particularly protected from renal dysfunction, oxidative stress, and tubular lipid accumulation. Both HFD-fed male and female mice presented dyslipidemia, but lipidomic analysis highlighted differential renal lipid profiles. While both sexes presented similar neutral lipid accumulation with obesity, only males showed increased levels of ceramides and phospholipids. Remarkably, protection against renal lipotoxicity in females was associated with enhanced renal adiponectin and AMP-activated protein kinase (AMPK) signaling. Circulating adiponectin and its renal receptor levels were significantly lower in obese males, but were maintained in females. This observation correlated with the maintained basal AMPK activity in obese female mice compared to males. CONCLUSIONS: Collectively, our findings suggest that female mice are protected from obesity-induced renal dysfunction and lipotoxicity associated with enhanced adiponectin and AMPK signaling compared to males.


Obesity-related complications can differ between men and women due to sex-specific differences in how fats are handled. Here, we studied the effects of high-fat diet on the kidneys of male and female mice. We found that despite gaining similar weight, obese female mice were better protected against insulin resistance, liver fat accumulation, and kidney damage caused by obesity than males. In particular, female mice were protected against lipid accumulation in the kidneys. We further analyzed the lipid profile in the kidneys of both male and female mice and observed differences in the amount and nature of the accumulated lipids. Male mice had increased levels of specific lipids, which may contribute to their higher risk of kidney damage. In contrast, female mice showed better lipid metabolism adaptation, which helped to protect their kidneys. This study also revealed an association between higher levels of the protein hormone adiponectin and higher activity of the cellular energy master regulator protein AMPK in obese females. These proteins may help prevent obesity-induced kidney damage. In obese males, these protective proteins are reduced and are associated with kidney damage. In conclusion, this study suggests that female mice are naturally shielded from obesity-induced kidney damage and lipid accumulation in the kidneys. Obesity in males is associated with the presence of potentially toxic lipids and dysregulated renal metabolism. Understanding these sex-related differences in obesity-related complications could lead to better management and treatment of kidney problems in both men and women.


Adiponectin , Kidney Diseases , Animals , Female , Male , Mice , AMP-Activated Protein Kinases/metabolism , Kidney/metabolism , Kidney Diseases/etiology , Lipidomics , Lipids , Obesity/metabolism , Sex Characteristics
6.
Skelet Muscle ; 13(1): 5, 2023 03 07.
Article En | MEDLINE | ID: mdl-36882853

BACKGROUND: We have previously demonstrated that double homeobox 4 centromeric (DUX4C) encoded for a functional DUX4c protein upregulated in dystrophic skeletal muscles. Based on gain- and loss-of-function studies we have proposed DUX4c involvement in muscle regeneration. Here, we provide further evidence for such a role in skeletal muscles from patients affected with facioscapulohumeral muscular dystrophy (FSHD). METHODS: DUX4c was studied at RNA and protein levels in FSHD muscle cell cultures and biopsies. Its protein partners were co-purified and identified by mass spectrometry. Endogenous DUX4c was detected in FSHD muscle sections with either its partners or regeneration markers using co-immunofluorescence or in situ proximity ligation assay. RESULTS: We identified new alternatively spliced DUX4C transcripts and confirmed DUX4c immunodetection in rare FSHD muscle cells in primary culture. DUX4c was detected in nuclei, cytoplasm or at cell-cell contacts between myocytes and interacted sporadically with specific RNA-binding proteins involved, a.o., in muscle differentiation, repair, and mass maintenance. In FSHD muscle sections, DUX4c was found in fibers with unusual shape or central/delocalized nuclei (a regeneration feature) staining for developmental myosin heavy chain, MYOD or presenting intense desmin labeling. Some couples of myocytes/fibers locally exhibited peripheral DUX4c-positive areas that were very close to each other, but in distinct cells. MYOD or intense desmin staining at these locations suggested an imminent muscle cell fusion. We further demonstrated DUX4c interaction with its major protein partner, C1qBP, inside myocytes/myofibers that presented features of regeneration. On adjacent muscle sections, we could unexpectedly detect DUX4 (the FSHD causal protein) and its interaction with C1qBP in fusing myocytes/fibers. CONCLUSIONS: DUX4c upregulation in FSHD muscles suggests it contributes not only to the pathology but also, based on its protein partners and specific markers, to attempts at muscle regeneration. The presence of both DUX4 and DUX4c in regenerating FSHD muscle cells suggests DUX4 could compete with normal DUX4c functions, thus explaining why skeletal muscle is particularly sensitive to DUX4 toxicity. Caution should be exerted with therapeutic agents aiming for DUX4 suppression because they might also repress the highly similar DUX4c and interfere with its physiological role.


Homeodomain Proteins , Muscular Dystrophy, Facioscapulohumeral , RNA-Binding Proteins , Transcription Factors , Humans , Carrier Proteins , Cytoplasm , Desmin , Homeodomain Proteins/genetics , Mitochondrial Proteins , Muscle Fibers, Skeletal , Muscular Dystrophy, Facioscapulohumeral/genetics , Transcription Factors/genetics , RNA-Binding Proteins/genetics
7.
J Drug Target ; 30(9): 948-960, 2022 11.
Article En | MEDLINE | ID: mdl-35723066

Proprotein convertase substilisin/kexin 9 (PCSK9) inhibitors (PCSK9i) revolutionised the lipid-lowering therapy. However, a risk of type 2 diabetes mellitus (T2DM) is evoked under PCSK9i therapy. In this review, we summarise the current knowledge on the link of PCSK9 with T2DM. A significant correlation was found between PCSK9 and insulin, homeostasis model assessment (HOMA) of insulin resistance and glycated haemoglobin. PCSK9 is also involved in inflammation. PCSK9 loss-of-function variants increased T2DM risk by altering insulin secretion. Local pancreatic low PCSK9 regulates ß-cell LDLR expression which in turn promotes intracellular cholesterol accumulation and hampers insulin secretion. Nevertheless, the association of PCSK9 loss-of-function variants and T2DM is inconsistent. InsLeu and R46L polymorphisms were associated with T2DM, low HOMA for ß-cell function and impaired fasting glucose, while the C679X polymorphism was associated with low fasting glucose in Black South African people. Hence, we assume that the impact of these variants on glucose homeostasis may vary depending on the genetic background of the studied populations and the type of effect caused by those genetic variants on the PCSK9 protein. Accordingly, these factors should be considered when choosing a genetic variant of PCSK9 to assess the impact of long-term use of PCSK9i on glucose homeostasis.


Diabetes Mellitus, Type 2 , Proprotein Convertase 9 , Cholesterol , Diabetes Mellitus, Type 2/drug therapy , Glucose , Glycated Hemoglobin/analysis , Homeostasis , Humans , Insulin , Proprotein Convertase 9/genetics , Proprotein Convertases/genetics
8.
Front Med (Lausanne) ; 9: 822870, 2022.
Article En | MEDLINE | ID: mdl-35602498

Aristolochic acid nephropathy (AAN) is a progressive tubulointerstitial nephritis caused by the intake of aristolochic acids (AA) contained in Chinese herbal remedies or contaminated food. AAN is characterized by tubular atrophy and interstitial fibrosis, characterizing advanced kidney disease. It is established that sustained or recurrent acute kidney injury (AKI) episodes contribute to the progression of CKD. Therefore, the study of underlying mechanisms of AA-induced nephrotoxicity could be useful in understanding the complex AKI-to-CKD transition. We developed a translational approach of AKI-to-CKD transition by reproducing human AAN in rodent models. Indeed, in such models, an early phase of acute tubular necrosis was rapidly followed by a massive interstitial recruitment of activated monocytes/macrophages followed by cytotoxic T lymphocytes, resulting in a transient AKI episode. A later chronic phase was then observed with progressive tubular atrophy related to dedifferentiation and necrosis of tubular epithelial cells. The accumulation of vimentin and αSMA-positive cells expressing TGFß in interstitial areas suggested an increase in resident fibroblasts and their activation into myofibroblasts resulting in collagen deposition and CKD. In addition, we identified 4 major actors in the AKI-to-CKD transition: (1) the tubular epithelial cells, (2) the endothelial cells of the interstitial capillary network, (3) the inflammatory infiltrate, and (4) the myofibroblasts. This review provides the most comprehensive and informative data we were able to collect and examines the pending questions.

9.
Redox Biol ; 51: 102251, 2022 05.
Article En | MEDLINE | ID: mdl-35248827

Facioscapulohumeral muscular dystrophy (FSHD) is characterised by descending skeletal muscle weakness and wasting. FSHD is caused by mis-expression of the transcription factor DUX4, which is linked to oxidative stress, a condition especially detrimental to skeletal muscle with its high metabolic activity and energy demands. Oxidative damage characterises FSHD and recent work suggests metabolic dysfunction and perturbed hypoxia signalling as novel pathomechanisms. However, redox biology of FSHD remains poorly understood, and integrating the complex dynamics of DUX4-induced metabolic changes is lacking. Here we pinpoint the kinetic involvement of altered mitochondrial ROS metabolism and impaired mitochondrial function in aetiology of oxidative stress in FSHD. Transcriptomic analysis in FSHD muscle biopsies reveals strong enrichment for pathways involved in mitochondrial complex I assembly, nitrogen metabolism, oxidative stress response and hypoxia signalling. We found elevated mitochondrial ROS (mitoROS) levels correlate with increases in steady-state mitochondrial membrane potential in FSHD myogenic cells. DUX4 triggers mitochondrial membrane polarisation prior to oxidative stress generation and apoptosis through mitoROS, and affects mitochondrial health through lipid peroxidation. We identify complex I as the primary target for DUX4-induced mitochondrial dysfunction, with strong correlation between complex I-linked respiration and cellular oxygenation/hypoxia signalling activity in environmental hypoxia. Thus, FSHD myogenesis is uniquely susceptible to hypoxia-induced oxidative stress as a consequence of metabolic mis-adaptation. Importantly, mitochondria-targeted antioxidants rescue FSHD pathology more effectively than conventional antioxidants, highlighting the central involvement of disturbed mitochondrial ROS metabolism. This work provides a pathomechanistic model by which DUX4-induced changes in oxidative metabolism impair muscle function in FSHD, amplified when metabolic adaptation to varying O2 tension is required.


Muscular Dystrophy, Facioscapulohumeral , Antioxidants/metabolism , Homeodomain Proteins/metabolism , Humans , Hypoxia/metabolism , Mitochondria/metabolism , Muscle, Skeletal/metabolism , Muscular Dystrophy, Facioscapulohumeral/genetics , Muscular Dystrophy, Facioscapulohumeral/metabolism , Muscular Dystrophy, Facioscapulohumeral/pathology , Oxidative Stress , Reactive Oxygen Species/metabolism
10.
Int J Mol Sci ; 22(13)2021 Jul 05.
Article En | MEDLINE | ID: mdl-34281273

Muscular dystrophies (MDs) are a group of inherited degenerative muscle disorders characterized by a progressive skeletal muscle wasting. Respiratory impairments and subsequent hypoxemia are encountered in a significant subgroup of patients in almost all MD forms. In response to hypoxic stress, compensatory mechanisms are activated especially through Hypoxia-Inducible Factor 1 α (HIF-1α). In healthy muscle, hypoxia and HIF-1α activation are known to affect oxidative stress balance and metabolism. Recent evidence has also highlighted HIF-1α as a regulator of myogenesis and satellite cell function. However, the impact of HIF-1α pathway modifications in MDs remains to be investigated. Multifactorial pathological mechanisms could lead to HIF-1α activation in patient skeletal muscles. In addition to the genetic defect per se, respiratory failure or blood vessel alterations could modify hypoxia response pathways. Here, we will discuss the current knowledge about the hypoxia response pathway alterations in MDs and address whether such changes could influence MD pathophysiology.


Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia/etiology , Hypoxia/metabolism , Muscular Dystrophies/complications , Muscular Dystrophies/metabolism , Animals , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/drug effects , Ischemia/etiology , Models, Biological , Muscle Development , Muscle, Skeletal/blood supply , Muscle, Skeletal/metabolism , Muscular Dystrophies/genetics , Oxidative Stress , Regeneration , Signal Transduction
11.
Biomolecules ; 11(5)2021 04 29.
Article En | MEDLINE | ID: mdl-33947124

(1) Background: White adipose tissue (WAT) is a dynamic and plastic tissue showing high sensitivity to carbohydrate supply. In such a context, the WAT may accordingly modulate its mitochondrial metabolic activity. We previously demonstrated that a partial replacement of glucose by galactose in a culture medium of 3T3-L1 cells leads to a poorer adipogenic yield and improved global mitochondrial health. In the present study, we investigate key mitochondrial metabolic actors reflecting mitochondrial adaptation in response to different carbohydrate supplies. (2) Methods: The metabolome of 3T3-L1 cells was investigated during the differentiation process using different glucose/galactose ratios and by a targeted approach using 1H-NMR (Proton nuclear magnetic resonance) spectroscopy; (3) Results: Our findings indicate a reduction of adipogenic and metabolic overload markers under the low glucose/galactose condition. In addition, a remodeling of the mitochondrial function triggers the secretion of metabolites with signaling and systemic energetical homeostasis functions. Finally, this study also sheds light on a new way to consider the mitochondrial metabolic function by considering noncarbohydrates related pathways reflecting both healthier cellular and mitochondrial adaptation mechanisms; (4) Conclusions: Different carbohydrates supplies induce deep mitochondrial metabolic and function adaptations leading to overall adipocytes function and profile remodeling during the adipogenesis.


Adipogenesis , Culture Media/chemistry , Metabolomics/methods , Mitochondria/metabolism , 3T3-L1 Cells , Animals , Carbohydrate Metabolism , Cell Culture Techniques , Cell Differentiation , Galactose/chemistry , Glucose/chemistry , Mice , Proton Magnetic Resonance Spectroscopy
12.
Nutrients ; 12(10)2020 Sep 29.
Article En | MEDLINE | ID: mdl-33003504

Obesity is an alarming yet increasing phenomenon worldwide, and more effective obesity management strategies have become essential. In addition to the numerous anti-adipogenic treatments promising a restauration of a healthy white adipose tissue (WAT) function, numerous studies reported on the critical role of nutritional parameters in obesity development. In a metabolic disorder context, a better control of nutrient intake is a key step in slowing down adipogenesis and therefore obesity. Of interest, the effect on WAT remodeling deserves deeper investigations. Among the different actors of WAT plasticity, the mitochondrial network plays a central role due to its dynamics and essential cellular functions. Hence, the present in vitro study, conducted on the 3T3-L1 cell line, aimed at evaluating the incidence of modulating the carbohydrates intake on adipogenesis through an integrated assessment of mitochondrial structure, dynamics, and functions-correlated changes. For this purpose, our experimental strategy was to compare the occurrence of adipogenesis in 3T3-L1 cells cultured either in a high-glucose (HG) medium (25 mM) or in a low-glucose (LG) medium (5 mM) supplemented with equivalent galactose (GAL) levels (20 mM). The present LG-GAL condition was associated, in differentiating adipocytes, to a reduced lipid droplet network, lower expressions of early and late adipogenic genes and proteins, an increased mitochondrial network with higher biogenesis marker expression, an equilibrium in the mitochondrial fusion/fission pattern, and a decreased expression of mitochondrial metabolic overload protein markers. Therefore, those main findings show a clear effect of modulating glucose accessibility on 3T3-L1 adipogenesis through a combined effect of adipogenesis modulation and overall improvement of the mitochondrial health status. This nutritional approach offers promising opportunities in the control and prevention of obesity.


Adipogenesis/drug effects , Dietary Carbohydrates/pharmacokinetics , Eating/physiology , Mitochondria/drug effects , 3T3-L1 Cells , Adipocytes/drug effects , Adipose Tissue, White/metabolism , Animals , Biological Availability , Galactose/pharmacokinetics , Glucose/pharmacokinetics , Mice
13.
Int J Mol Sci ; 21(21)2020 Oct 27.
Article En | MEDLINE | ID: mdl-33121167

Chronic kidney disease (CKD) is prevalent in 9.1% of the global population and is a significant public health problem associated with increased morbidity and mortality. CKD is associated with highly prevalent physiological and metabolic disturbances such as hypertension, obesity, insulin resistance, cardiovascular disease, and aging, which are also risk factors for CKD pathogenesis and progression. Podocytes and proximal tubular cells of the kidney strongly express AMP-activated protein kinase (AMPK). AMPK plays essential roles in glucose and lipid metabolism, cell survival, growth, and inflammation. Thus, metabolic disease-induced renal diseases like obesity-related and diabetic chronic kidney disease demonstrate dysregulated AMPK in the kidney. Activating AMPK ameliorates the pathological and phenotypical features of both diseases. As a metabolic sensor, AMPK regulates active tubular transport and helps renal cells to survive low energy states. AMPK also exerts a key role in mitochondrial homeostasis and is known to regulate autophagy in mammalian cells. While the nutrient-sensing role of AMPK is critical in determining the fate of renal cells, the role of AMPK in kidney autophagy and mitochondrial quality control leading to pathology in metabolic disease-related CKD is not very clear and needs further investigation. This review highlights the crucial role of AMPK in renal cell dysfunction associated with metabolic diseases and aims to expand therapeutic strategies by understanding the molecular and cellular processes underlying CKD.


AMP-Activated Protein Kinases/metabolism , Metabolic Diseases/complications , Renal Insufficiency, Chronic/metabolism , Disease Progression , Gene Expression Regulation , Humans , Lipid Metabolism , Metabolic Diseases/metabolism , Mitochondria/metabolism , Renal Insufficiency, Chronic/etiology
14.
Sci Rep ; 10(1): 11301, 2020 07 09.
Article En | MEDLINE | ID: mdl-32647247

Intramuscular injection and electroporation of naked plasmid DNA (IMEP) has emerged as a potential alternative to viral vector injection for transgene expression into skeletal muscles. In this study, IMEP was used to express the DUX4 gene into mouse tibialis anterior muscle. DUX4 is normally expressed in germ cells and early embryo, and silenced in adult muscle cells where its pathological reactivation leads to Facioscapulohumeral muscular dystrophy. DUX4 encodes a potent transcription factor causing a large deregulation cascade. Its high toxicity but sporadic expression constitutes major issues for testing emerging therapeutics. The IMEP method appeared as a convenient technique to locally express DUX4 in mouse muscles. Histological analyses revealed well delineated muscle lesions 1-week after DUX4 IMEP. We have therefore developed a convenient outcome measure by quantification of the damaged muscle area using color thresholding. This method was used to characterize lesion distribution and to assess plasmid recirculation and dose-response. DUX4 expression and activity were confirmed at the mRNA and protein levels and through a quantification of target gene expression. Finally, this study gives a proof of concept of IMEP model usefulness for the rapid screening of therapeutic strategies, as demonstrated using antisense oligonucleotides against DUX4 mRNA.


Disease Models, Animal , Homeodomain Proteins/metabolism , Muscle, Skeletal/metabolism , Muscular Dystrophy, Facioscapulohumeral/metabolism , Animals , Electroporation , Female , Gene Expression , Homeodomain Proteins/genetics , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Muscular Dystrophy, Facioscapulohumeral/pathology
15.
Toxicol In Vitro ; 65: 104811, 2020 Jun.
Article En | MEDLINE | ID: mdl-32119997

Aristolochic acid nephropathy (AAN) is characterized by interstitial fibrosis, proximal tubular atrophy, and hypoxia. A correlation between a reduced peritubular capillary density and the severity of fibrosis has been demonstrated. As calcium, redox and energetic homeostasis are crucial in maintaining endothelial cell function and survival, we aimed to investigate AA-induced disturbances involved in endothelial cell injury. Our results showed a cytotoxic effect of AA on EAhy926 endothelial cells. Exposure of aortic rings to AA impaired vascular relaxation to Acetylcholine (ACh). Increased levels of intracellular reactive oxygen species (ROS) were observed in cells exposed to AA. Pre-treatment with antioxidant N-acetyl cysteine inhibited AA-induced cell death. Superoxide dismutase resulted in restoring ACh-induced relaxation. An increase in intracellular calcium level ([Ca2+]i) was observed on endothelial cells. Calcium chelators BAPTA-AM or APB, a specific inhibitor of IP3R, improved cell viability. Moreover, AA exposure led to reduced AMP-activated protein kinase (AMPK) expression. AICAR, an activator of AMPK, improved the viability of AA-intoxicated cells and inhibited the rise of cytosolic [Ca2+]i levels. This study provides evidence that AA exposure increases ROS generation, disrupts calcium homeostasis and decreases AMPK activity. It also suggests that significant damage observed in endothelial cells may enhance microcirculation defects, worsening hypoxia and tubulointerstitial lesions.


Aristolochic Acids/pharmacology , Endothelial Cells/drug effects , Endothelium, Vascular/drug effects , AMP-Activated Protein Kinases/genetics , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/physiology , Calcium/metabolism , Cell Line , Endothelial Cells/metabolism , Endothelial Cells/pathology , Endothelium, Vascular/metabolism , Endothelium, Vascular/physiology , Humans , Male , Rats, Wistar , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism
16.
Int J Mol Sci ; 22(1)2020 Dec 31.
Article En | MEDLINE | ID: mdl-33396267

Exercise training is now recognized as an interesting therapeutic strategy in managing obesity and its related disorders. However, there is still a lack of knowledge about its impact on obesity-induced chronic kidney disease (CKD). Here, we investigated the effects of a delayed protocol of endurance exercise training (EET) as well as the underlying mechanism in obese mice presenting CKD. Mice fed a high-fat diet (HFD) or a low-fat diet (LFD) for 12 weeks were subsequently submitted to an 8-weeks EET protocol. Delayed treatment with EET in obese mice prevented body weight gain associated with a reduced calorie intake. EET intervention counteracted obesity-related disorders including glucose intolerance, insulin resistance, dyslipidaemia and hepatic steatosis. Moreover, our data demonstrated for the first time the beneficial effects of EET on obesity-induced CKD as evidenced by an improvement of obesity-related glomerulopathy, tubulo-interstitial fibrosis, inflammation and oxidative stress. EET also prevented renal lipid depositions in the proximal tubule. These results were associated with an improvement of the AMPK pathway by EET in renal tissue. AMPK-mediated phosphorylation of ACC and ULK-1 were particularly enhanced leading to increased fatty acid oxidation and autophagy improvement with EET in obese mice.


AMP-Activated Protein Kinases/metabolism , Autophagy , Diet, High-Fat/adverse effects , Obesity/complications , Physical Conditioning, Animal , Renal Insufficiency, Chronic/prevention & control , AMP-Activated Protein Kinases/genetics , Animals , Glucose Intolerance , Lipid Metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Phosphorylation , Renal Insufficiency, Chronic/etiology , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/pathology
17.
J Lipid Res ; 60(5): 937-952, 2019 05.
Article En | MEDLINE | ID: mdl-30862696

High-fat diet (HFD) causes renal lipotoxicity that is ameliorated with AMP-activated protein kinase (AMPK) activation. Although bioactive eicosanoids increase with HFD and are essential in regulation of renal disease, their role in the inflammatory response to HFD-induced kidney disease and their modulation by AMPK activation remain unexplored. In a mouse model, we explored the effects of HFD on eicosanoid synthesis and the role of AMPK activation in ameliorating these changes. We used targeted lipidomic profiling with quantitative MS to determine PUFA and eicosanoid content in kidneys, urine, and renal arterial and venous circulation. HFD increased phospholipase expression as well as the total and free pro-inflammatory arachidonic acid (AA) and anti-inflammatory DHA in kidneys. Consistent with the parent PUFA levels, the AA- and DHA-derived lipoxygenase (LOX), cytochrome P450, and nonenzymatic degradation (NE) metabolites increased in kidneys with HFD, while EPA-derived LOX and NE metabolites decreased. Conversely, treatment with 5-aminoimidazole-4-carboxamide-1-ß-D-furanosyl 5'-monophosphate (AICAR), an AMPK activator, reduced the free AA and DHA content and the DHA-derived metabolites in kidney. Interestingly, kidney and circulating AA, AA metabolites, EPA-derived LOX, and NE metabolites are increased with HFD; whereas, DHA metabolites are increased in kidney in contrast to their decreased circulating levels with HFD. Together, these changes showcase HFD-induced pro- and anti-inflammatory eicosanoid dysregulation and highlight the role of AMPK in correcting HFD-induced dysregulated eicosanoid pathways.


AMP-Activated Protein Kinases/metabolism , Diet, High-Fat/adverse effects , Eicosanoids/metabolism , Kidney Diseases/metabolism , Animals , Kidney Diseases/chemically induced , Male , Mice , Mice, Inbred C57BL
18.
Exp Physiol ; 103(1): 125-140, 2018 01 01.
Article En | MEDLINE | ID: mdl-28944982

NEW FINDINGS: What is the central question of this study? The metabolic pathways regulating the effects of obesity on the kidney remain unknown. We sought to determine whether inducible nitric oxide synthase (iNOS) is involved in the underlying mechanisms of high-fat diet-induced kidney disease using a specific iNOS inhibitor, N6-(1-iminoethyl)-l-lysine hydrochloride (L-NIL). What is the main finding and its importance? We did not demonstrate an upregulation of iNOS renal expression after high caloric intake, suggesting that iNOS might not be a crucial player in the development of obesity-induced kidney disease. Although L-NIL treatment clearly ameliorated systemic metabolic parameters, the effect on loss of renal function, impairment of tubular integrity, oxidative stress and inflammation appeared to be more moderate. Central obesity is related to caloric excess, promoting deleterious cellular responses in targeted organs. Nitric oxide (NO) has been determined as a key player in the pathogenesis of metabolic diseases. Here, we investigated the implication of inducible NO synthase (iNOS) in the development of obesity-induced kidney disease. C57Bl/6 male mice were randomized to a low-fat diet (LFD) or a high-fat diet (HFD) and treated with N6-(1-iminoethyl)-l-lysine hydrochloride (L-NIL), a specific iNOS inhibitor, for 16 weeks. Mice fed an HFD exhibited a significant increase in body weight, fasting blood glucose and plasma concentrations of non-esterified fatty acids, triglyceride and insulin. Inhibition of iNOS prevented these changes in mice fed an HFD. Interestingly, the significant increase in albuminuria and mesangial matrix expansion were not ameliorated with L-NIL, whereas a significant decrease in proteinuria, N-acetyl-ß-d-glucosaminidase excretion and renal triglyceride content were found, suggesting that iNOS inhibition is more suitable for tubular function than glomerular function. The urinary concentration of hydrogen peroxide, a stable product of reactive oxygen species production, that was found to be increased in mice fed an HFD, was significantly reduced with L-NIL. Finally, despite a moderate effect of L-NIL on inflammatory processes in the kidney, we demonstrated a positive impact of this treatment on adipocyte hypertrophy and on adipose tissue inflammation. These results suggest that inhibition of iNOS leads to a moderate beneficial effect on kidney function in mice fed an HFD. Further studies are needed for better understanding of the role of iNOS in obesity-induced kidney disease.


Diet, High-Fat/adverse effects , Kidney Diseases/enzymology , Kidney Diseases/pathology , Kidney/pathology , Kidney/physiology , Nitric Oxide Synthase Type II/antagonists & inhibitors , Animals , Enzyme Inhibitors/pharmacology , Kidney/drug effects , Kidney Diseases/prevention & control , Male , Mice , Mice, Inbred C57BL , Nitric Oxide Synthase Type II/physiology
19.
PLoS One ; 12(8): e0183604, 2017.
Article En | MEDLINE | ID: mdl-28832640

Aristolochic Acid (AA) nephropathy (AAN) is a progressive tubulointerstitial nephritis characterized by an early phase of acute kidney injury (AKI) leading to chronic kidney disease (CKD). The reduced nitric oxide (NO) bioavailability reported in AAN might contribute to renal function impairment and progression of the disease. We previously demonstrated that L-arginine (L-Arg) supplementation is protective in AA-induced AKI. Since the severity of AKI may be considered a strong predictor of progression to CKD, the present study aims to assess the potential benefit of L-Arg supplementation during the transition from the acute phase to the chronic phase of AAN. C57BL/6J male mice were randomly subjected to daily i.p. injections of vehicle or AA for 4 days. To determine whether renal AA-induced injuries were linked to reduced NO production, L-Arg was added to drinking water from 7 days before starting i.p. injections, until the end of the protocol. Mice were euthanized 5, 10 and 20 days after vehicle or AA administration. AA-treated mice displayed marked renal injury and reduced NO bioavailability, while histopathological features of AAN were reproduced, including interstitial cell infiltration and tubulointerstitial fibrosis. L-Arg treatment restored renal NO bioavailability and reduced the severity of AA-induced injury, inflammation and fibrosis. We concluded that reduced renal NO bioavailability contributes to the processes underlying AAN. Furthermore, L-Arg shows nephroprotective effects by decreasing the severity of acute-to-chronic transition in experimental AAN and might represent a potential therapeutic tool in the future.


Aristolochic Acids/toxicity , Kidney Diseases/metabolism , Nitric Oxide/metabolism , Animals , Arginine/administration & dosage , Biological Availability , Inflammation/prevention & control , Kidney Diseases/chemically induced , Mice
20.
Int J Mol Sci ; 18(2)2017 Jan 29.
Article En | MEDLINE | ID: mdl-28146082

The term "aristolochic acid nephropathy" (AAN) is used to include any form of toxic interstitial nephropathy that is caused either by ingestion of plants containing aristolochic acids (AA) as part of traditional phytotherapies (formerly known as "Chinese herbs nephropathy"), or by the environmental contaminants in food (Balkan endemic nephropathy). It is frequently associated with urothelial malignancies. Although products containing AA have been banned in most of countries, AAN cases remain regularly reported all over the world. Moreover, AAN incidence is probably highly underestimated given the presence of AA in traditional herbal remedies worldwide and the weak awareness of the disease. During these two past decades, animal models for AAN have been developed to investigate underlying molecular and cellular mechanisms involved in AAN pathogenesis. Indeed, a more-in-depth understanding of these processes is essential to develop therapeutic strategies aimed to reduce the global and underestimated burden of this disease. In this regard, our purpose was to build a broad overview of what is currently known about AAN. To achieve this goal, we aimed to summarize the latest data available about underlying pathophysiological mechanisms leading to AAN development with a particular emphasis on the imbalance between vasoactive factors as well as a focus on the vascular events often not considered in AAN.


Aristolochic Acids/adverse effects , Drugs, Chinese Herbal/adverse effects , Nephritis, Interstitial/etiology , Animals , Aristolochic Acids/chemistry , Aristolochic Acids/metabolism , Balkan Nephropathy/diagnosis , Balkan Nephropathy/epidemiology , Balkan Nephropathy/etiology , Biopsy , Cell Transformation, Neoplastic/chemically induced , Drugs, Chinese Herbal/chemistry , Drugs, Chinese Herbal/metabolism , Fibrosis , Humans , Kidney Neoplasms/etiology , Nephritis, Interstitial/diagnosis , Nephritis, Interstitial/epidemiology , Oxidative Stress
...