Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 109
Filter
1.
Free Radic Biol Med ; 222: 85-105, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38838921

ABSTRACT

Fibrosis is the tissue scarring characterized by excess deposition of extracellular matrix (ECM) proteins, mainly collagens. A fibrotic response can take place in any tissue of the body and is the result of an imbalanced reaction to inflammation and wound healing. Metabolism has emerged as a major driver of fibrotic diseases. While glycolytic shifts appear to be a key metabolic switch in activated stromal ECM-producing cells, several other cell types such as immune cells, whose functions are intricately connected to their metabolic characteristics, form a complex network of pro-fibrotic cellular crosstalk. This review purports to clarify shared and particular cellular responses and mechanisms across organs and etiologies. We discuss the impact of the cell-type specific metabolic reprogramming in fibrotic diseases in both experimental and human pathology settings, providing a rationale for new therapeutic interventions based on metabolism-targeted antifibrotic agents.

2.
J Clin Med ; 13(9)2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38730996

ABSTRACT

Background: Rapidly progressive osteoarthritis of the hip (RPOH) is a rare syndrome that involves the femoral head and acetabulum. Methods: We analyzed the incidence of RPOH in 2022. The inclusion criteria included a clinical history of pain for 1-6 months and a decrease in joint space of > 2 mm within one year or a decrease in joint space by 50% in that time accompanied by femoral and/or acetabular bone destruction. Exclusion: There are no other destructive arthropathies and no evolutionary radiological image sequence. Results: There were 15 patients, 16 hips, an incidence around 3.17%, a 1:1 laterality ratio, and 1 bilateral affected. The mean average age is 77.35 years. The male/female ratio is 1:2. The average BMI is 31.2. The time of the onset of the symptoms to the patient's diagnosis is 5 months. The functionally modified Harris scale (MHS) had an average score of 30 points. They had surgery hip arthroplasty with a cementless cup in all cases, a revision cup in one of them, and a double mobility cup in the other, with the stem cemented three times. There were no post-surgical complications. Functionally was achieved at 3 months. The average MHS is 70 at 12 months. The average MHS is 85. Conclusions: RPOH is an idiopathic entity characterized by great clinical involvement and rapid radiological evolution. It is most common in women around 77 years of age. The bone quality requires surgical alternatives to implants, and it has good functional recovery post-surgery.

3.
Redox Biol ; 68: 102957, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37977043

ABSTRACT

Progressive respiratory failure is the primary cause of death in the coronavirus disease 2019 (COVID-19) pandemic. It is the final outcome of the acute respiratory distress syndrome (ARDS), characterized by an initial exacerbated inflammatory response, metabolic derangement and ultimate tissue scarring. A positive balance of cellular energy may result crucial for the recovery of clinical COVID-19. Hence, we asked if two key pathways involved in cellular energy generation, AMP-activated protein kinase (AMPK)/acetyl-CoA carboxylase (ACC) signaling and fatty acid oxidation (FAO) could be beneficial. We tested the drugs metformin (AMPK activator) and baicalin (CPT1A activator) in different experimental models mimicking COVID-19 associated inflammation in lung and kidney. We also studied two different cohorts of COVID-19 patients that had been previously treated with metformin. These drugs ameliorated lung damage in an ARDS animal model, while activation of AMPK/ACC signaling increased mitochondrial function and decreased TGF-ß-induced fibrosis, apoptosis and inflammation markers in lung epithelial cells. Similar results were observed with two indole derivatives, IND6 and IND8 with AMPK activating capacity. Consistently, a reduced time of hospitalization and need of intensive care was observed in COVID-19 patients previously exposed to metformin. Baicalin also mitigated the activation of pro-inflammatory bone marrow-derived macrophages (BMDMs) and reduced kidney fibrosis in two animal models of kidney injury, another key target of COVID-19. In human epithelial lung and kidney cells, both drugs improved mitochondrial function and prevented TGF-ß-induced renal epithelial cell dedifferentiation. Our results support that favoring cellular energy production through enhanced FAO may prove useful in the prevention of COVID-19-induced lung and renal damage.


Subject(s)
COVID-19 , Metformin , Respiratory Distress Syndrome , Animals , Humans , Metformin/pharmacology , Metformin/therapeutic use , AMP-Activated Protein Kinases/metabolism , Kidney/metabolism , Lung/metabolism , Inflammation/drug therapy , Transforming Growth Factor beta , Fibrosis , Fatty Acids
4.
Life Sci Alliance ; 6(10)2023 10.
Article in English | MEDLINE | ID: mdl-37487638

ABSTRACT

Tubulointerstitial fibrosis is the common pathological substrate for many etiologies leading to chronic kidney disease. Although perturbations in the circadian rhythm have been associated with renal disease, the role of the molecular clock in the pathogenesis of fibrosis remains incompletely understood. We investigated the relationship between the molecular clock and renal damage in experimental models of injury and fibrosis (unilateral ureteral obstruction, folic acid, and adenine nephrotoxicity), using genetically modified mice with selective deficiencies of the clock components Bmal1, Clock, and Cry We found that the molecular clock pathway was enriched in damaged tubular epithelial cells with marked metabolic alterations. In human tubular epithelial cells, TGFß significantly altered the expression of clock components. Although Clock played a role in the macrophage-mediated inflammatory response, the combined absence of Cry1 and Cry2 was critical for the recruitment of neutrophils, correlating with a worsening of fibrosis and with a major shift in the expression of metabolism-related genes. These results support that renal damage disrupts the kidney peripheral molecular clock, which in turn promotes metabolic derangement linked to inflammatory and fibrotic responses.


Subject(s)
Adenine , Kidney , Humans , Animals , Mice , Circadian Rhythm , Epithelial Cells , Macrophages
5.
Front Mol Biosci ; 10: 1161036, 2023.
Article in English | MEDLINE | ID: mdl-37377862

ABSTRACT

Background: Chronic kidney disease (CKD) is characterized by the progressive and irreversible deterioration of kidney function and structure with the appearance of renal fibrosis. A significant decrease in mitochondrial metabolism, specifically a reduction in fatty acid oxidation (FAO) in tubular cells, is observed in tubulointerstitial fibrosis, whereas FAO enhancement provides protection. Untargeted metabolomics offers the potential to provide a comprehensive analysis of the renal metabolome in the context of kidney injury. Methodology: Renal tissue from a carnitine palmitoyl transferase 1a (Cpt1a) overexpressing mouse model, which displays enhanced FAO in the renal tubule, subjected to folic acid nephropathy (FAN) was studied through a multiplatform untargeted metabolomics approach based on LC-MS, CE-MS and GC-MS analysis to achieve the highest coverage of the metabolome and lipidome affected by fibrosis. The expression of genes related to the biochemical routes showing significant changes was also evaluated. Results: By combining different tools for signal processing, statistical analysis and feature annotation, we were able to identify variations in 194 metabolites and lipids involved in many metabolic routes: TCA cycle, polyamines, one-carbon metabolism, amino acid metabolism, purine metabolism, FAO, glycerolipids and glycerophospholipids synthesis and degradation, glycosphingolipids interconversion, and sterol metabolism. We found several metabolites strongly altered by FAN, with no reversion induced by Cpt1a overexpression (v.g. citric acid), whereas other metabolites were influenced by CPT1A-induced FAO (v.g. glycine-betaine). Conclusion: It was implemented a successful multiplatform metabolomics approach for renal tissue analysis. Profound metabolic changes accompany CKD-associated fibrosis, some associated with tubular FAO failure. These results highlight the importance of addressing the crosstalk between metabolism and fibrosis when undertaking studies attempting to elucidate the mechanism of CKD progression.

6.
Immunity ; 56(3): 516-530.e9, 2023 03 14.
Article in English | MEDLINE | ID: mdl-36738738

ABSTRACT

In vitro studies have associated oxidative phosphorylation (OXPHOS) with anti-inflammatory macrophages, whereas pro-inflammatory macrophages rely on glycolysis. However, the metabolic needs of macrophages in tissues (TMFs) to fulfill their homeostatic activities are incompletely understood. Here, we identified OXPHOS as the highest discriminating process among TMFs from different organs in homeostasis by analysis of RNA-seq data in both humans and mice. Impairing OXPHOS in TMFs via Tfam deletion differentially affected TMF populations. Tfam deletion resulted in reduction of alveolar macrophages (AMs) due to impaired lipid-handling capacity, leading to increased cholesterol content and cellular stress, causing cell-cycle arrest in vivo. In obesity, Tfam depletion selectively ablated pro-inflammatory lipid-handling white adipose tissue macrophages (WAT-MFs), thus preventing insulin resistance and hepatosteatosis. Hence, OXPHOS, rather than glycolysis, distinguishes TMF populations and is critical for the maintenance of TMFs with a high lipid-handling activity, including pro-inflammatory WAT-MFs. This could provide a selective therapeutic targeting tool.


Subject(s)
Inflammation , Oxidative Phosphorylation , Humans , Mice , Animals , Inflammation/metabolism , Macrophages/metabolism , Homeostasis , Lipids , Adipose Tissue/metabolism
7.
Liver Int ; 42(3): 651-662, 2022 03.
Article in English | MEDLINE | ID: mdl-35050550

ABSTRACT

BACKGROUND AND AIMS: PTTG1 is almost undetectable in adult livers but is highly expressed in hepatocarcinoma. While little is known about its involvement in liver fibrosis, PTTG1 expression is associated with DLK1. We assessed the role of the PTTG1/DLK1 pathway in fibrosis progression and the potential therapeutic effect of PTTG1 silencing in fibrosis. METHODS: Pttg1 and Dlk1 were studied in liver and isolated cell populations of control and fibrotic rats and in human liver biopsies. The fibrotic molecular signature was analysed in Pttg1-/- and Pttg1+/+ fibrotic mice. Finally, Pttg1 silencing was evaluated in rats as a novel antifibrotic therapy. RESULTS: Pttg1 and Dlk1 mRNA selectively increased in fibrotic rats paralleling fibrosis progression. Serum DLK1 concentrations correlated with hepatic collagen content and systemic and portal haemodynamics. Human cirrhotic livers showed greater PTTG1 and DLK1 transcript abundance than non-cirrhotic, and reduced collagen was observed in Pttg1 Pttg1-/- mice. The liver fibrotic molecular signature revealed lower expression of genes related to extracellular matrix remodelling including Mmp8 and 9 and Timp4 and greater eotaxin and Mmp13 than fibrotic Pttg1+/+ mice. Finally, interfering Pttg1 resulted in reduced liver fibrotic area, lower α-Sma and decreased portal pressure than fibrotic animals. Furthermore, Pttg1 silencing decreased the transcription of Dlk1, collagens I and III, Pdgfrß, Tgfrß, Timp1, Timp2 and Mmp2. CONCLUSIONS: Pttg1/Dlk1 are selectively overexpressed in the cirrhotic liver and participate in ECM turnover regulation. Pttg1 disruption decreases Dlk1 transcription and attenuates collagen deposition. PTTG1/DLK1 signalling is a novel pathway for targeting the progression of liver fibrosis.


Subject(s)
Calcium-Binding Proteins , Intercellular Signaling Peptides and Proteins , Membrane Proteins , Pituitary Neoplasms , Securin , Animals , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Fibrosis , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Liver/pathology , Liver Cirrhosis/pathology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Oncogenes , Pituitary Neoplasms/metabolism , Pituitary Neoplasms/pathology , Rats , Securin/genetics , Securin/metabolism
8.
Am J Kidney Dis ; 80(2): 251-263, 2022 08.
Article in English | MEDLINE | ID: mdl-34999158

ABSTRACT

Kidney fibrosis is a hallmark of chronic kidney disease (CKD) and a potential therapeutic target. However, there are conceptual and practical challenges to directly targeting kidney fibrosis. Whether fibrosis is mainly a cause or a consequence of CKD progression has been disputed. It is unclear whether specifically targeting fibrosis is feasible in clinical practice because most drugs that decrease fibrosis in preclinical models target additional and often multiple pathogenic pathways (eg, renin-angiotensin-aldosterone system blockade). Moreover, tools to assess whole-kidney fibrosis in routine clinical practice are lacking. Pirfenidone, a drug used for idiopathic pulmonary fibrosis, is undergoing a phase 2 trial for kidney fibrosis. Other drugs in use or being tested for idiopathic pulmonary fibrosis (eg, nintedanib, PRM-151, epigallocatechin gallate) are also potential candidates to treat kidney fibrosis. Novel therapeutic approaches may include antagomirs (eg, lademirsen) or drugs targeting interleukin 11 or NKD2 (WNT signaling pathway inhibitor). Reversing the dysfunctional tubular cell metabolism that leads to kidney fibrosis offers additional therapeutic opportunities. However, any future drug targeting fibrosis of the kidneys should demonstrate added benefit to a standard of care that combines renin-angiotensin system with mineralocorticoid receptor (eg, finerenone) blockade or with sodium/glucose cotransporter 2 inhibitors.


Subject(s)
Idiopathic Pulmonary Fibrosis , Renal Insufficiency, Chronic , Adaptor Proteins, Signal Transducing , Antifibrotic Agents , Calcium-Binding Proteins , Fibrosis , Humans , Idiopathic Pulmonary Fibrosis/complications , Renal Insufficiency, Chronic/etiology , Renin-Angiotensin System
9.
Kidney Int ; 100(6): 1165-1167, 2021 12.
Article in English | MEDLINE | ID: mdl-34802556

ABSTRACT

The metabolic impairment of kidney tubular cells is a key mechanism underlying the pathophysiology of renal fibrosis. In particular, a drastic reduction in fatty acid oxidation is essentially responsible for the global energy failure occurring in the tubulointerstitial compartment. Piret et al. propose a novel transcriptional regulatory mechanism involving the decrease in the expression of Krüppel-like factor 15 in proximal tubular cells after kidney injury, which results in a major derangement of fatty acid oxidation.


Subject(s)
Kidney Diseases , Kidney Tubules, Proximal , Fibrosis , Humans , Kidney/pathology , Kidney Diseases/pathology , Kidney Tubules, Proximal/metabolism , Lipid Metabolism
10.
Life Sci Alliance ; 4(8)2021 08.
Article in English | MEDLINE | ID: mdl-34183444

ABSTRACT

As neurons age, they show a decrease in their ability to degrade proteins and membranes. Because undegraded material is a source of toxic products, defects in degradation are associated with reduced cell function and survival. However, there are very few dead neurons in the aging brain, suggesting the action of compensatory mechanisms. We show in this work that ageing neurons in culture show large multivesicular bodies (MVBs) filled with intralumenal vesicles (ILVs) and secrete more small extracellular vesicles than younger neurons. We also show that the high number of ILVs is the consequence of the accumulation of cholesterol in MVBs, which in turn is due to decreased levels of the cholesterol extruding protein NPC1. NPC1 down-regulation is the consequence of a combination of upregulation of the NPC1 repressor microRNA 33, and increased degradation, due to Akt-mTOR targeting of NPC1 to the phagosome. Although releasing more exosomes can be beneficial to old neurons, other cells, neighbouring and distant, can be negatively affected by the waste material they contain.


Subject(s)
Cholesterol/metabolism , Exosomes/metabolism , MicroRNAs/genetics , Multivesicular Bodies/metabolism , Neurons/cytology , Niemann-Pick C1 Protein/genetics , Animals , Cell Line , Cellular Senescence , Down-Regulation , HEK293 Cells , Humans , Mice , Neurons/metabolism , Primary Cell Culture , Rats , Signal Transduction
11.
J Clin Invest ; 131(5)2021 03 01.
Article in English | MEDLINE | ID: mdl-33465052

ABSTRACT

Chronic kidney disease (CKD) remains a major epidemiological, clinical, and biomedical challenge. During CKD, renal tubular epithelial cells (TECs) present a persistent inflammatory and profibrotic response. Fatty acid oxidation (FAO), the main source of energy for TECs, is reduced in kidney fibrosis and contributes to its pathogenesis. To determine whether gain of function in FAO (FAO-GOF) could protect from fibrosis, we generated a conditional transgenic mouse model with overexpression of the fatty acid shuttling enzyme carnitine palmitoyl-transferase 1A (CPT1A) in TECs. Cpt1a-knockin (CPT1A-KI) mice subjected to 3 models of renal fibrosis (unilateral ureteral obstruction, folic acid nephropathy [FAN], and adenine-induced nephrotoxicity) exhibited decreased expression of fibrotic markers, a blunted proinflammatory response, and reduced epithelial cell damage and macrophage influx. Protection from fibrosis was also observed when Cpt1a overexpression was induced after FAN. FAO-GOF restored oxidative metabolism and mitochondrial number and enhanced bioenergetics, increasing palmitate oxidation and ATP levels, changes that were also recapitulated in TECs exposed to profibrotic stimuli. Studies in patients showed decreased CPT1 levels and increased accumulation of short- and middle-chain acylcarnitines, reflecting impaired FAO in human CKD. We propose that strategies based on FAO-GOF may constitute powerful alternatives to combat fibrosis inherent to CKD.


Subject(s)
Carnitine O-Palmitoyltransferase/biosynthesis , Gene Expression Regulation, Enzymologic , Kidney Tubules/enzymology , Renal Insufficiency, Chronic/prevention & control , Animals , Carnitine O-Palmitoyltransferase/genetics , Disease Models, Animal , Fatty Acids/genetics , Fatty Acids/metabolism , Fibrosis , Kidney Tubules/pathology , Mice , Mice, Knockout , Mice, Transgenic , Renal Insufficiency, Chronic/enzymology , Renal Insufficiency, Chronic/genetics
12.
Redox Biol ; 40: 101851, 2021 04.
Article in English | MEDLINE | ID: mdl-33465566

ABSTRACT

Excessive accumulation of extracellular matrix (ECM) is the hallmark of fibrotic diseases. In the kidney, it is the final common pathway of prevalent diseases, leading to chronic renal failure. While cytokines such as TGF-ß play a fundamental role in myofibroblast transformation, recent work has shown that mitochondrial dysfunction and defective fatty acid oxidation (FAO), which compromise the main source of energy for renal tubular epithelial cells, have been proposed to be fundamental contributors to the development and progression of kidney fibrosis. MicroRNAs (miRNAs), which regulate gene expression post-transcriptionally, have been reported to control renal fibrogenesis. To identify miRNAs involved in the metabolic derangement of renal fibrosis, we performed a miRNA array screen in the mouse model of unilateral ureteral obstruction (UUO). MiR-150-5p and miR-495-3p were selected for their link to human pathology, their role in mitochondrial metabolism and their targeting of the fatty acid shuttling enzyme CPT1A. We found a 2- and 4-fold upregulation of miR-150-5p and miR-495-5p, respectively, in both the UUO and the folic acid induced nephropathy (FAN) models, while TGF-ß1 upregulated their expressions in the human renal tubular epithelial cell line HKC-8. These miRNAs synergized with TGF-ß regarding its pro-fibrotic effect by enhancing the fibrosis-associated markers Acta2, Col1α1 and Fn1. Bioenergetics studies showed a reduction of FAO-associated oxygen consumption rate (OCR) in HKC-8 cells in the presence of both miRNAs. Consistently, expression levels of their mitochondrial-related target genes CPT1A, PGC1α and the mitochondrial transcription factor A (TFAM), were reduced by half in renal epithelial cells exposed to these miRNAs. By contrast, we did not detect changes in mitochondrial mass and transmembrane potential (ΔÑ°m) or mitochondrial superoxide radical anion production. Our data support that miR-150 and miR-495 may contribute to renal fibrogenesis by aggravating the metabolic failure critically involved in tubular epithelial cells, ultimately leading to fibrosis.


Subject(s)
MicroRNAs , Ureteral Obstruction , Fibrosis , Humans , Kidney/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Oxidative Stress , Transforming Growth Factor beta1/metabolism , Ureteral Obstruction/pathology
13.
FASEB J ; 35(1): e21213, 2021 01.
Article in English | MEDLINE | ID: mdl-33368614

ABSTRACT

Preclinical studies have demonstrated that activation of the NOTCH pathway plays a key role in the pathogenesis of kidney damage. There is currently no information on the role of the Delta-like homologue 1 (DLK1), a NOTCH inhibitor, in the regulation of renal damage. Here, we investigated the contribution of DLK1 to experimental renal damage and the underlying molecular mechanisms. Using a Dlk1-null mouse model in the experimental renal damage of unilateral ureteral obstruction, we found activation of NOTCH, as shown by increased nuclear translocation of the NOTCH1 intracellular domain, and upregulation of Dlk2/hey-1 expression compared to wild-type (WT) littermates. NOTCH1 over-activation in Dlk1-null injured kidneys was associated with a higher inflammatory response, characterized by infiltration of inflammatory cells, mainly CD4/IL17A + lymphocytes, and activation of the Th17 immune response. Furthermore, pharmacological NOTCH blockade inhibited the transcription factors controlling Th17 differentiation and gene expression of the Th17 effector cytokine IL-17A and other related-inflammatory factors, linked to a diminution of inflammation in the injured kidneys. We propose that the non-canonical NOTCH ligand DLK1 acts as a NOTCH antagonist in renal injury regulating the Th17-mediated inflammatory response.


Subject(s)
Calcium-Binding Proteins/deficiency , Gene Deletion , Immunity, Cellular , Kidney Diseases/immunology , Kidney/immunology , Th17 Cells/immunology , Animals , Calcium-Binding Proteins/immunology , Cell Cycle Proteins/genetics , Cell Cycle Proteins/immunology , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/immunology , Kidney/pathology , Kidney Diseases/genetics , Kidney Diseases/pathology , Mice , Th17 Cells/pathology , Ureteral Obstruction/genetics , Ureteral Obstruction/immunology , Ureteral Obstruction/pathology
14.
Nefrología (Madrid) ; 40(5): 491-498, sept.-oct. 2020. ilus
Article in Spanish | IBECS | ID: ibc-199029

ABSTRACT

Los microRNA (miRNA) son ARN endógenos de pequeño tamaño que regulan la expresión génica a través de la represión postranscripcional de sus ARN mensajeros diana. El estudio de los cambios en la expresión de ciertos miRNA en el riñón ha proporcionado evidencias sobre su papel patogénico y potencial terapéutico en nefrología. En esta revisión proponemos un abordaje de nanotecnología basado en la unión de análogos o inhibidores de miRNA formados por ácidos peptidonucleicos (PNA) a péptidos con una estructura transmembrana que es sensible a pH bajo, denominada pHLIP (del inglés pH [low] insertion peptide), apoyándonos en el concepto de que el pH acídico del microambiente tubular renal puede facilitar la concentración y la distribución del complejo pHLIP-PNA en este órgano. En este contexto hemos demostrado por primera vez que la administración dirigida de inhibidores de miR-33 con el sistema pHLIP previene eficazmente del desarrollo de fibrosis renal, abriendo las puertas de esta tecnología a nuevas estrategias de diagnóstico y tratamiento de enfermedades renales


MicroRNAs (miRNAs) are small endogenous RNAs that regulate gene expression through post-transcriptional repression of their target messenger RNAs. A study of changes in expression of certain miRNAs in the kidney has supplied evidence on their pathogenic role and therapeutic potential in nephrology. This review proposes a nanotechnology approach based on the binding of analogs or inhibitors of miRNAs formed by peptide nucleic acids (PNAs) to peptides with a transmembrane structure sensitive to a low pH, called pHLIPs (pH [low] insertion peptides). The review draws on the concept that an acidic pH in the microenvironment of the renal tubule may facilitate concentration and distribution of the pHLIP-PNA complex in this organ. In this context, we have demonstrated for the first time that targeted administration of miR-33 inhibitors with the pHLIP system effectively prevents the development of renal fibrosis, thus opening up this technology to new strategies for diagnosis and treatment of kidney diseases


Subject(s)
Humans , Animals , Male , MicroRNAs/antagonists & inhibitors , MicroRNAs/metabolism , Peptide Nucleic Acids/administration & dosage , Kidney Diseases/therapy , Ribonucleotides/antagonists & inhibitors , Peptide Nucleic Acids/therapeutic use , Nanotechnology/methods , Acidosis, Renal Tubular , Kidney Diseases/diagnosis
15.
Nefrologia (Engl Ed) ; 40(5): 491-498, 2020.
Article in English, Spanish | MEDLINE | ID: mdl-32693933

ABSTRACT

MicroRNAs (miRNAs) are small endogenous RNAs that regulate gene expression through post-transcriptional repression of their target messenger RNAs. A study of changes in expression of certain miRNAs in the kidney has supplied evidence on their pathogenic role and therapeutic potential in nephrology. This review proposes a nanotechnology approach based on the binding of analogs or inhibitors of miRNAs formed by peptide nucleic acids (PNAs) to peptides with a transmembrane structure sensitive to a low pH, called pHLIPs (pH [low] insertion peptides). The review draws on the concept that an acidic pH in the microenvironment of the renal tubule may facilitate concentration and distribution of the pHLIP-PNA complex in this organ. In this context, we have demonstrated for the first time that targeted administration of miR-33 inhibitors with the pHLIP system effectively prevents the development of renal fibrosis, thus opening up this technology to new strategies for diagnosis and treatment of kidney diseases.


Subject(s)
Kidney , Membrane Proteins/physiology , MicroRNAs/physiology , Animals , Humans
16.
Redox Biol ; 37: 101580, 2020 10.
Article in English | MEDLINE | ID: mdl-32723695

ABSTRACT

Environmental risk factors, including physicochemical agents, noise and mental stress, have a considerable impact on human health. This environmental exposure may lead to epigenetic reprogramming, including changes in non-coding RNAs (ncRNAs) signatures, which can contribute to the pathophysiology state. Oxidative stress is one of the results of this environmental disturbance by modifying cellular processes such as apoptosis, signal transduction cascades, and DNA repair mechanisms. In this review, we delineate environmental risk factors and their influence on (ncRNAs) in connection to disease. We focus on well-studied miRNAs and analyze the novel roles of long-non-coding-RNAs (lncRNAs). We discuss commonly regulated lncRNAs after exposure to different stressors, such as UV, heavy metals and pesticides among others, and the potential role of these lncRNA as exposure biomarkers, epigenetic regulators and potential therapeutic targets to diminish the deleterious secondary response to environmental agents.


Subject(s)
Environmental Health , MicroRNAs , RNA, Long Noncoding , Humans , MicroRNAs/genetics , RNA, Long Noncoding/genetics , RNA, Untranslated/genetics , Signal Transduction
17.
Anal Bioanal Chem ; 412(24): 6391-6405, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32285184

ABSTRACT

Despite the recent advances in the standardization of untargeted metabolomics workflows, there is still a lack of attention to specific data treatment strategies that require deep knowledge of the biological problem and need to be applied after a well-thought out process to understand the effect of the practice. One of those strategies is data normalization. Data-driven assumptions are critical especially addressing unwanted variation present in the biological model as it can be the case in heterogeneous tissues, cells with different sizes or biofluids with different concentrations. Chronic kidney disease (CKD) is a widespread disorder affecting kidney structure and function. Animal models are being developed to be able to get valuable insights into the etiopathogenesis of the condition and effect of the treatments. Moreover, diagnosis and disease staging still require defining appropriate biomarkers. Untargeted metabolomics has the potential to deal with those challenges. Renal fibrosis is one of the consequences of kidney injury which greatly affects the concentration of metabolites in the same quantity of sample. To overcome this challenge, several data normalization strategies have been applied, following a multilevel normalization method with the overall aim of focussing on the relevant biological information and reducing the influence of disturbing factors. A comprehensive evaluation of the performance of the normalization strategies, both on methods assessing the intragroup variation and on the impact on differential analysis, is provided. Finally, we present evidence of the importance of biological-model-driven guided normalization methods and discuss multiple criteria that need to be taken into consideration to obtain robust and reliable data. Special concern is transmitted on the misleading conclusions that might be the consequence of inappropriate data pre-treatment solutions applied for untargeted methods. Graphical abstract.


Subject(s)
Kidney/metabolism , Metabolomics/methods , Renal Insufficiency, Chronic/metabolism , Animals , Discriminant Analysis , Disease Models, Animal , Humans , Least-Squares Analysis , Male , Metabolome , Mice, Inbred C57BL , Mice, Transgenic
18.
Nat Rev Nephrol ; 16(5): 269-288, 2020 05.
Article in English | MEDLINE | ID: mdl-32060481

ABSTRACT

Chronic kidney disease (CKD) is a devastating condition that is reaching epidemic levels owing to the increasing prevalence of diabetes mellitus, hypertension and obesity, as well as ageing of the population. Regardless of the underlying aetiology, CKD is slowly progressive and leads to irreversible nephron loss, end-stage renal disease and/or premature death. Factors that contribute to CKD progression include parenchymal cell loss, chronic inflammation, fibrosis and reduced regenerative capacity of the kidney. Current therapies have limited effectiveness and only delay disease progression, underscoring the need to develop novel therapeutic approaches to either stop or reverse progression. Preclinical studies have identified several approaches that reduce fibrosis in experimental models, including targeting cytokines, transcription factors, developmental and signalling pathways and epigenetic modulators, particularly microRNAs. Some of these nephroprotective strategies are now being tested in clinical trials. Lessons learned from the failure of clinical studies of transforming growth factor ß1 (TGFß1) blockade underscore the need for alternative approaches to CKD therapy, as strategies that target a single pathogenic process may result in unexpected negative effects on simultaneously occurring processes. Additional promising avenues include preventing tubular cell injury and anti-fibrotic therapies that target activated myofibroblasts, the main collagen-producing cells.


Subject(s)
Renal Insufficiency, Chronic/complications , Renal Insufficiency, Chronic/therapy , Disease Progression , Humans , Renal Insufficiency, Chronic/pathology
19.
FASEB J ; 34(1): 410-431, 2020 01.
Article in English | MEDLINE | ID: mdl-31914684

ABSTRACT

MicroRNAs (miRNAs) regulate gene expression posttranscriptionally and control biological processes (BPs), including fibrogenesis. Kidney fibrosis remains a clinical challenge and miRNAs may represent a valid therapeutic avenue. We show that miR-9-5p protected from renal fibrosis in the mouse model of unilateral ureteral obstruction (UUO). This was reflected in reduced expression of pro-fibrotic markers, decreased number of infiltrating monocytes/macrophages, and diminished tubular epithelial cell injury and transforming growth factor-beta 1 (TGF-ß1)-dependent de-differentiation in human kidney proximal tubular (HKC-8) cells. RNA-sequencing (RNA-Seq) studies in the UUO model revealed that treatment with miR-9-5p prevented the downregulation of genes related to key metabolic pathways, including mitochondrial function, oxidative phosphorylation (OXPHOS), fatty acid oxidation (FAO), and glycolysis. Studies in human tubular epithelial cells demonstrated that miR-9-5p impeded TGF-ß1-induced bioenergetics derangement. The expression of the FAO-related axis peroxisome proliferator-activated receptor gamma coactivator 1 alpha (PGC-1α)-peroxisome proliferator-activated receptor alpha (PPARα) was reduced by UUO, although preserved by the administration of miR-9-5p. We found that in mice null for the mitochondrial master regulator PGC-1α, miR-9-5p was unable to promote a protective effect in the UUO model. We propose that miR-9-5p elicits a protective response to chronic kidney injury and renal fibrosis by inducing reprogramming of the metabolic derangement and mitochondrial dysfunction affecting tubular epithelial cells.


Subject(s)
Cellular Reprogramming , Fibrosis/prevention & control , Gene Expression Regulation , Kidney Diseases/prevention & control , MicroRNAs/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/physiology , Ureteral Obstruction/prevention & control , Animals , Fibrosis/genetics , Fibrosis/metabolism , Fibrosis/pathology , Humans , Kidney Diseases/genetics , Kidney Diseases/metabolism , Kidney Diseases/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Transcriptome , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism , Ureteral Obstruction/genetics , Ureteral Obstruction/metabolism , Ureteral Obstruction/pathology
20.
JCI Insight ; 4(22)2019 11 14.
Article in English | MEDLINE | ID: mdl-31613798

ABSTRACT

Previous work has reported the important links between cellular bioenergetics and the development of chronic kidney disease, highlighting the potential for targeting metabolic functions to regulate disease progression. More recently, it has been shown that alterations in fatty acid oxidation (FAO) can have an important impact on the progression of kidney disease. In this work, we demonstrate that loss of miR-33, an important regulator of lipid metabolism, can partially prevent the repression of FAO in fibrotic kidneys and reduce lipid accumulation. These changes were associated with a dramatic reduction in the extent of fibrosis induced in 2 mouse models of kidney disease. These effects were not related to changes in circulating leukocytes because bone marrow transplants from miR-33-deficient animals did not have a similar impact on disease progression. Most important, targeted delivery of miR-33 peptide nucleic acid inhibitors to the kidney and other acidic microenvironments was accomplished using pH low insertion peptides as a carrier. This was effective at both increasing the expression of factors involved in FAO and reducing the development of fibrosis. Together, these findings suggest that miR-33 may be an attractive therapeutic target for the treatment of chronic kidney disease.


Subject(s)
Kidney Diseases , MicroRNAs , Animals , Fatty Acids/metabolism , Fibrosis/metabolism , Fibrosis/prevention & control , Kidney Diseases/metabolism , Kidney Diseases/prevention & control , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , MicroRNAs/metabolism , Oxidation-Reduction
SELECTION OF CITATIONS
SEARCH DETAIL
...