Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
1.
Curr Mol Pharmacol ; 16(2): 139-146, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-35232343

RESUMEN

BACKGROUND: Vascular endothelial dysfunction (VED) significantly results in catastrophic cardiovascular diseases with multiple aetiologies. Variations in vasoactive peptides, including angiotensin II and endothelin 1, and metabolic perturbations like hyperglycaemia, altered insulin signalling, and homocysteine levels result in pathogenic signalling cascades, which ultimately lead to VED. Endoplasmic reticulum (ER) stress reduces nitric oxide availability, causes aberrant angiogenesis, and enhances oxidative stress pathways, consequently promoting endothelial dysfunction. Moreover, the renin-angiotensin system (RAS) has widely been acknowledged to impact angiogenesis, endothelial repair and inflammation. Interestingly, experimental studies at the preclinical level indicate a possible pathological link between the two pathways in the development of VED. Furthermore, pharmacological modulation of ER stress ameliorates angiotensin-II mediated VED as well as RAS intervention either through inhibition of the pressor arm or enhancement of the depressor arm of RAS, mitigating ER stress-induced endothelial dysfunction and thus emphasizing a vital crosstalk. CONCLUSION: Deciphering the pathway overlap between RAS and ER stress may open potential therapeutic avenues to combat endothelial dysfunction and associated diseases. Several studies suggest that alteration in a component of RAS may induce ER stress or induction of ER stress may modulate the RAS components. In this review, we intend to elaborate on the crosstalk of ER stress and RAS in the pathophysiology of VED.


Asunto(s)
Estrés del Retículo Endoplásmico , Endotelio Vascular , Sistema Renina-Angiotensina , Enfermedades Vasculares , Humanos , Angiotensina II/farmacología , Estrés del Retículo Endoplásmico/fisiología , Endotelio Vascular/metabolismo , Enfermedades Vasculares/metabolismo
2.
Matrix Biol ; 112: 72-89, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35964866

RESUMEN

Intrarenal extracellular matrix production or kidney fibrosis is a prevalent feature of all forms of chronic kidney disease (CKD). The transforming growth factor-beta (TGFß) is believed to be a major driver of extracellular matrix production. Nevertheless, anti-TGFß therapies have consistently failed to reduce extracellular matrix production in CKD patients indicating the need for novel therapeutic strategies. We have previously shown that necroinflammation contributes to acute kidney injury. Here, we show that chronic/persistent necroinflammation drives intrarenal extracellular matrix production during CKD. We found that renal expression of receptor-interacting protein kinase-1 (RIPK1), RIPK3, and mixed lineage kinase domain-like (MLKL) increases with the production of intrarenal extracellular matrix and declined kidney function in both humans and mice. Furthermore, we found that TGFß exposure induces the translocation of RIPK3 and MLKL to mitochondria resulting in mitochondrial dysfunction and ROS production. Mitochondrial ROS activates the serine-threonine kinase calcium/calmodulin-dependent protein kinases-II (CaMKII) that increases phosphorylation of Smad2/3 and subsequent production of alpha-smooth muscle actin (αSMA), collagen (Col) 1α1, etc. in response to TGFß during the intrarenal extracellular matrix production. Consistent with this, deficiency or knockdown of RIPK3 or MLKL as well as pharmacological inhibition of RIPK1, RIPK3, and CaMKII prevents the intrarenal extracellular matrix production in oxalate-induced CKD and unilateral ureteral obstruction (UUO). Together, RIPK1, RIPK3, MLKL, CaMKII, and Smad2/3 are molecular targets to inhibit intrarenal extracellular matrix production and preserve kidney function during CKD.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina , Insuficiencia Renal Crónica , Actinas/metabolismo , Animales , Calcio/metabolismo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Matriz Extracelular/metabolismo , Humanos , Ratones , Mitocondrias/metabolismo , Oxalatos/metabolismo , Proteínas Quinasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Insuficiencia Renal Crónica/genética , Insuficiencia Renal Crónica/metabolismo , Factores de Crecimiento Transformadores/metabolismo
4.
Antioxidants (Basel) ; 11(2)2022 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-35204131

RESUMEN

Inflammasomes are multiprotein complexes with an important role in the innate immune response. Canonical activation of inflammasomes results in caspase-1 activation and maturation of cytokines interleukin-1ß and -18. These cytokines can elicit their effects through receptor activation, both locally within a certain tissue and systemically. Animal models of kidney diseases have shown inflammasome involvement in inflammation, pyroptosis and fibrosis. In particular, the inflammasome component nucleotide-binding domain-like receptor family pyrin domain containing 3 (NLRP3) and related canonical mechanisms have been investigated. However, it has become increasingly clear that other inflammasome components are also of importance in kidney disease. Moreover, it is becoming obvious that the range of molecular interaction partners of inflammasome components in kidney diseases is wide. This review provides insights into these current areas of research, with special emphasis on the interaction of inflammasome components and redox signalling, endoplasmic reticulum stress, and mitochondrial function. We present our findings separately for acute kidney injury and chronic kidney disease. As we strictly divided the results into preclinical and clinical data, this review enables comparison of results from those complementary research specialities. However, it also reveals that knowledge gaps exist, especially in clinical acute kidney injury inflammasome research. Furthermore, patient comorbidities and treatments seem important drivers of inflammasome component alterations in human kidney disease.

5.
Cells ; 11(2)2022 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-35053307

RESUMEN

Neutrophil extracellular traps (NETs) are associated with multiple disease pathologies including sepsis, asthma, rheumatoid arthritis, cancer, systemic lupus erythematosus, acute respiratory distress syndrome, and COVID-19. NETs, being a disintegrated death form, suffered inconsistency in their identification, nomenclature, and quantifications that hindered therapeutic approaches using NETs as a target. Multiple strategies including microscopy, ELISA, immunoblotting, flow cytometry, and image-stream-based methods have exhibited drawbacks such as being subjective, non-specific, error-prone, and not being high throughput, and thus demand the development of innovative and efficient approaches for their analyses. Here, we established an imaging and computational algorithm using high content screening (HCS)-cellomics platform that aid in easy, rapid, and specific detection as well as analyses of NETs. This method employed membrane-permeable and impermeable DNA dyes in situ to identify NET-forming cells. Automated algorithm-driven single-cell analysis of change in nuclear morphology, increase in nuclear area, and change in intensities provided precise detection of NET-forming cells and eliminated user bias with other cell death modalities. Further combination with Annexin V staining in situ detected specific death pathway, e.g., apoptosis, and thus, discriminated between NETs, apoptosis, and necrosis. Our approach does not utilize fixation and permeabilization steps that disturb NETs, and thus, allows the time-dependent monitoring of NETs. Together, this specific imaging-based high throughput method for NETs analyses may provide a good platform for the discovery of potential inhibitors of NET formation and/or agents to modulate neutrophil death, e.g., NETosis-apoptosis switch, as an alternative strategy to enhance the resolution of inflammation.


Asunto(s)
Algoritmos , COVID-19/sangre , Trampas Extracelulares/metabolismo , Citometría de Flujo , Neutrófilos/metabolismo , SARS-CoV-2/metabolismo , Análisis de la Célula Individual , Humanos
6.
Pharmacol Res ; 168: 105602, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33838293

RESUMEN

Diabetes is a chronic metabolic disorder with a high rate of morbidity and mortality. Insufficient insulin secretion and insulin action are two major causes for the development of diabetes, which is characterized by a persistent increase in blood glucose level. Diet and sedentary life style play pivotal role in development of vascular complications in type 2 diabetes. Dietary modification is associated with a reprogramming of nutrient intake, which are proven to be effective for the management of diabetes and associated complications. Dietary modifications modulate various molecular key players linked with the functions of nutrient signalling, regulation of autophagy, and energy metabolism. It activates silent mating type information regulation 2 homolog1 (SIRT1) and AMP-activated protein kinase (AMPK). AMPK mainly acts as an energy sensor and inhibits autophagy repressor Mammalian target of rapamycin (mTOR) under nutritional deprivation. Under calorie restriction (CR), SIRT1 gets activated directly or indirectly and plays a central role in autophagy via the regulation of protein acetylation. Dietary modification is also effective in controlling inflammation and apoptosis by decreasing the level of pro-inflammatory cytokines like nuclear factor kappa- beta (NF-kß), tissue growth factor-beta (TGF-ß), tissue necrosis factor-alpha (TNF-α) and interleukin-6 (IL-6). It also improves glucose homeostasis and insulin secretion through beta cell regeneration. This indicates calorie intake plays a crucial role in the pathogenesis of type 2 diabetes-associated complications. The present review, emphasizes the role of dietary modifications in diabetes and associated complications.


Asunto(s)
Complicaciones de la Diabetes/dietoterapia , Diabetes Mellitus Tipo 2/dietoterapia , Animales , Restricción Calórica , Diabetes Mellitus Tipo 2/complicaciones , Cardiomiopatías Diabéticas/dietoterapia , Nefropatías Diabéticas/dietoterapia , Neuropatías Diabéticas/dietoterapia , Retinopatía Diabética/dietoterapia , Humanos
7.
Life Sci ; 271: 119193, 2021 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-33577856

RESUMEN

AIMS: Mineralization of crystalline particles and the formation of renal calculi contribute to the pathogenesis of crystal nephropathies. Several recent studies on the biology of crystal handling implicated intrarenal crystal deposition-induced necroinflammation in their pathogenesis. We hypothesized that 6,7-dihydroxycoumarin (DHC) inhibit intrarenal crystal cytotoxicity and necroinflammation, and ameliorate crystal-induced chronic kidney disease (CKD). MAIN METHODS: An unbiased high content screening coupled with fluorescence microscopy was used to identify compounds that inhibit CaOx crystal cytotoxicity. The ligand-protein interactions were identified using computational models e.g. molecular docking and molecular dynamics simulations. Furthermore, mice and rat models of oxalate-induced CKD were used for in-vivo studies. Renal injury, crystal deposition, and fibrosis were assessed by histology analysis. Western blots were used to quantify the protein expression. Data were expressed as boxplots and analyzed using one way ANOVA. KEY FINDINGS: An unbiased high-content screening in-vitro identified 6,7-DHC as a promising candidate. Further, 6,7-DHC protected human and mouse cells from calcium oxalate (CaOx) crystal-induced necroptosis in-vitro as well as mice and rats from oxalate-induced CKD in either preventive or therapeutic manner. Computational modeling demonstrated that 6,7-DHC interact with MLKL, the key protein in the necroptosis machinery, and inhibit its phosphorylation by ATP, which was evident in both in-vitro and in-vivo analyses. SIGNIFICANCE: Together, our results indicate that 6,7-DHC possesses a novel pharmacological property as a MLKL inhibitor and could serve as a lead molecule for further development of coumarin-based novel MLKL inhibitors. Furthermore, our data identify 6,7-DHC as a novel therapeutic strategy to combat crystal nephropathies.


Asunto(s)
Oxalato de Calcio/toxicidad , Cálculos Renales/tratamiento farmacológico , Cálculos Renales/metabolismo , Necroptosis/efectos de los fármacos , Proteínas Quinasas/metabolismo , Umbeliferonas/uso terapéutico , Animales , Antioxidantes/farmacología , Antioxidantes/uso terapéutico , Células HEK293 , Humanos , Cálculos Renales/inducido químicamente , Masculino , Ratones , Ratones Endogámicos C57BL , Simulación del Acoplamiento Molecular/métodos , Necroptosis/fisiología , Fosforilación/efectos de los fármacos , Fosforilación/fisiología , Proteínas Quinasas/química , Estructura Secundaria de Proteína , Ratas , Ratas Wistar , Umbeliferonas/farmacología
8.
Drug Discov Today ; 26(8): 2053-2063, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33617976

RESUMEN

Kidney fibrosis is marked by excessive extracellular matrix deposition during disease progression. Unfortunately, existing kidney function parameters do not predict the extent of kidney fibrosis. Moreover, the traditional histology methods for the assessment of kidney fibrosis require liquid and imaging biomarkers as well as needle-based biopsies, which are invasive and often associated with kidney injury. The repetitive analyses required to monitor the disease progression are therefore difficult. Hence, there is an unmet medical need for non-invasive and informative diagnostic approaches to monitor kidney fibrosis during the progression of chronic kidney disease. Here, we summarize the modern advances in diagnostic imaging techniques that have shown promise for non-invasive estimation of kidney fibrosis in pre-clinical and clinical studies.


Asunto(s)
Enfermedades Renales/diagnóstico por imagen , Riñón/diagnóstico por imagen , Insuficiencia Renal Crónica/diagnóstico por imagen , Animales , Biomarcadores/metabolismo , Progresión de la Enfermedad , Matriz Extracelular/patología , Fibrosis/diagnóstico por imagen , Humanos , Riñón/patología , Enfermedades Renales/patología , Insuficiencia Renal Crónica/patología
9.
Methods Mol Biol ; 2248: 231-241, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33185880

RESUMEN

The tumor necrosis factor (TNF) superfamily (TNFSF) members play crucial roles in the pathogenesis of acute and chronic kidney diseases. They orchestrate inflammation, cell survival, tissue repair as well as fibrosis in kidneys upon injury by engaging respective receptors on the cell membranes. Therefore, the TNFSF ligands, as well as their receptors, have gained enormous interest as putative drug targets to combat kidney diseases. It was shown that the expression profiles of TNFSF ligands differ in human and mice solid organs, as well as during acute kidney injuries and chronic kidney diseases in mice. This indicates that the mRNA expressions of TNFSF ligands highly depend on the species and nature of the injury, which needs to be given appropriate consideration while extrapolating the data between species and between different kidney diseases. The protocol presented here describes the use of real-time polymerase chain reaction (RT-PCR) to quantify the mRNA expressions of TNFSF ligands in healthy and injured murine kidneys.


Asunto(s)
Perfilación de la Expresión Génica , Enfermedades Renales/etiología , Riñón/metabolismo , Transcriptoma , Factores de Necrosis Tumoral/genética , Animales , Biomarcadores , Modelos Animales de Enfermedad , Enfermedades Renales/metabolismo , Ratones , Familia de Multigenes , Insuficiencia Renal Crónica/etiología , Insuficiencia Renal Crónica/metabolismo , Insuficiencia Renal Crónica/patología , Factores de Necrosis Tumoral/metabolismo
10.
Artículo en Inglés | MEDLINE | ID: mdl-32979084

RESUMEN

Due to a tragic increase in the incidences of diabetes globally, diabetic kidney disease (DKD) has emerged as one of the leading causes of end-stage renal diseases (ESRD). Hyperglycaemia-mediated overactivation of the renin-angiotensin-aldosterone system (RAAS) is key to the development and progression of DKD. Consequently, RAAS inhibition by angiotensin-converting enzyme inhibitors (ACEi) or angiotensin receptor blockers (ARBs) is the first-line therapy for the clinical management of DKD. However, numerous clinical and preclinical evidences suggested that RAAS inhibition can only halt the progression of the DKD to a certain extent, and they are inadequate to cure DKD completely. Recent studies have improved understanding of the complexity of the RAAS. It consists of two counter-regulatory arms, the deleterious pressor arm (ACE/angiotensin II/AT1 receptor axis) and the beneficial depressor arm (ACE2/angiotensin-(1-7)/Mas receptor axis). These advances have paved the way for the development of new therapies targeting the RAAS for better treatment of DKD. In this review, we aimed to summarise the involvement of the depressor arm of the RAAS in DKD. Moreover, in modern drug discovery and development, an advance approach is the bispecific therapeutics, targeting two independent signalling pathways. Here, we discuss available reports of these bispecific drugs involving the RAAS as well as propose potential treatments based on neurohormonal balance as credible therapeutic strategies for DKD.


Asunto(s)
Diabetes Mellitus , Nefropatías Diabéticas , Antagonistas de Receptores de Angiotensina/farmacología , Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Inhibidores de la Enzima Convertidora de Angiotensina/uso terapéutico , Nefropatías Diabéticas/tratamiento farmacológico , Humanos , Sistema Renina-Angiotensina , Vasoconstrictores/farmacología
12.
Cells ; 9(6)2020 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-32498376

RESUMEN

The COVID-19 pandemic is progressing worldwide with an alarming death toll. There is an urgent need for novel therapeutic strategies to combat potentially fatal complications. Distinctive clinical features of severe COVID-19 include acute respiratory distress syndrome, neutrophilia, and cytokine storm, along with severe inflammatory response syndrome or sepsis. Here, we propose the putative role of enhanced neutrophil infiltration and the release of neutrophil extracellular traps, complement activation and vascular thrombosis during necroinflammation in COVID-19. Furthermore, we discuss how neutrophilic inflammation contributes to the higher mortality of COVID-19 in patients with underlying co-morbidities such as diabetes and cardiovascular diseases. This perspective highlights neutrophils as a putative target for the immunopathologic complications of severely ill COVID-19 patients. Development of the novel therapeutic strategies targeting neutrophils may help reduce the overall disease fatality rate of COVID-19.


Asunto(s)
Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/patología , Trampas Extracelulares/inmunología , Neutrófilos/inmunología , Neumonía Viral/inmunología , Neumonía Viral/patología , Enzima Convertidora de Angiotensina 2 , Animales , Betacoronavirus/fisiología , COVID-19 , Enfermedades Cardiovasculares/complicaciones , Infecciones por Coronavirus/complicaciones , Infecciones por Coronavirus/tratamiento farmacológico , Complicaciones de la Diabetes/virología , Humanos , Inflamación/inmunología , Inflamación/patología , Necrosis/inmunología , Necrosis/patología , Neutrófilos/metabolismo , Pandemias , Peptidil-Dipeptidasa A/metabolismo , Neumonía Viral/complicaciones , Neumonía Viral/tratamiento farmacológico , SARS-CoV-2
13.
Life Sci ; 256: 117860, 2020 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-32534037

RESUMEN

Optimal tissue oxygenation is essential for its normal function. Suboptimal oxygenation or ischemia contributes to increased mortalities during various pathological conditions such as stroke, acute kidney injury (AKI), cardiac failure. Despite the rapid progression of renal tissue injury, the mechanism underlying renal ischemia/reperfusion injury (IRI) remains highly unclear. Experimental in vitro and in vivo models epitomizing the fundamental process is critical to the research of the pathogenesis of IRI and the development of plausible therapeutics. In this review, we describe the in vitro and in vivo models of IRI, ranges from proximal tubular cell lines to surgery-based animal models like clamping of both renal pedicles (bilateral IRI), clamping of one renal pedicle (unilateral IRI), clamping of one/or both renal arteries/or vein, or unilateral IRI with contralateral nephrectomy (uIRIx). Also, advanced technologies like three-dimensional kidney organoids, kidney-on-a-chip are explained. This review provides thoughtful information for establishing reliable and pertinent models for studying IRI-associated acute renal pathologies.


Asunto(s)
Enfermedades Renales/fisiopatología , Riñón/fisiopatología , Daño por Reperfusión/fisiopatología , Lesión Renal Aguda/fisiopatología , Animales , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Humanos , Riñón/irrigación sanguínea , Oxígeno/metabolismo , Arteria Renal/metabolismo , Reproducibilidad de los Resultados
14.
Drug Discov Today ; 25(7): 1253-1261, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32371137

RESUMEN

Fibrosis is a wound-healing process that results in tissue scarring and organ dysfunction. Several novel mechanisms of fibrogenesis have been discovered recently. In this review, we focus on the role of poly-ADP ribose polymerase (PARP) in major organ fibrosis, such as lungs, heart, liver, and kidneys. PARP is a dynamic enzyme that modulates different cellular proteins by the addition of PAR groups and mediates an array of cellular events in both normal physiological and pathophysiological states. The US Food and Drug Administration (FDA) and European Medicines Agency (EMA) recently approved several PARP inhibitors, such as olaparib, niraparib, talazoparib, and rucaparib, for the treatment of ovarian and germline BRCA-mutant breast cancers. Consequently, repurposing these drugs could provide an opportunity to counter organ fibrosis.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Fibrosis/tratamiento farmacológico , Neoplasias Ováricas/tratamiento farmacológico , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Poli(ADP-Ribosa) Polimerasas/metabolismo , Animales , Neoplasias de la Mama/metabolismo , Reposicionamiento de Medicamentos/métodos , Femenino , Fibrosis/metabolismo , Humanos , Neoplasias Ováricas/metabolismo
15.
Adv Sci (Weinh) ; 7(8): 1903337, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32328427

RESUMEN

Calcium oxalate (CaOx) crystal-induced nephropathies comprise a range of kidney disorders, for which there are no efficient pharmacological treatments. Although CaOx crystallization inhibitors have been suggested as a therapeutic modality already decades ago, limited progress has been made in the discovery of potent molecules with efficacy in animal disease models. Herein, an image-based machine learning approach to systematically screen chemically modified myo-inositol hexakisphosphate (IP6) analogues is utilized, which enables the identification of a highly active divalent inositol phosphate molecule. To date, this is the first molecule shown to completely inhibit the crystallization process in the nanomolar range, reduce crystal-cell interactions, thereby preventing CaOx-induced transcriptomic changes, and decrease renal CaOx deposition and kidney injury in a mouse model of hyperoxaluria. In conclusion, IP6 analogues based on such a scaffold may represent a new treatment option for CaOx nephropathies.

16.
Circ Res ; 126(8): e37-e52, 2020 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-32089086

RESUMEN

RATIONALE: Cholesterol crystal embolism can be a life-threatening complication of advanced atherosclerosis. Pathophysiology and molecular targets for treatment are largely unknown. OBJECTIVE: We aimed to develop a new animal model of cholesterol crystal embolism to dissect the molecular mechanisms of cholesterol crystal (CC)-driven arterial occlusion, tissue infarction, and organ failure. METHODS AND RESULTS: C57BL/6J mice were injected with CC into the left kidney artery. Primary end point was glomerular filtration rate (GFR). CC caused crystal clots occluding intrarenal arteries and a dose-dependent drop in GFR, followed by GFR recovery within 4 weeks, that is, acute kidney disease. In contrast, the extent of kidney infarction was more variable. Blocking necroptosis using mixed lineage kinase domain-like deficient mice or necrostatin-1s treatment protected from kidney infarction but not from GFR loss because arterial obstructions persisted, identifying crystal clots as a primary target to prevent organ failure. CC involved platelets, neutrophils, fibrin, and extracellular DNA. Neutrophil depletion or inhibition of the release of neutrophil extracellular traps had little effects, but platelet P2Y12 receptor antagonism with clopidogrel, fibrinolysis with urokinase, or DNA digestion with recombinant DNase I all prevented arterial occlusions, GFR loss, and kidney infarction. The window-of-opportunity was <3 hours after CC injection. However, combining Nec-1s (necrostatin-1s) prophylaxis given 1 hour before and DNase I 3 hours after CC injection completely prevented kidney failure and infarcts. In vitro, CC did not directly induce plasmatic coagulation but induced neutrophil extracellular trap formation and DNA release mainly from kidney endothelial cells, neutrophils, and few from platelets. CC induced ATP release from aggregating platelets, which increased fibrin formation in a DNase-dependent manner. CONCLUSIONS: CC embolism causes arterial obstructions and organ failure via the formation of crystal clots with fibrin, platelets, and extracellular DNA as critical components. Therefore, our model enables to unravel the pathogenesis of the CC embolism syndrome as a basis for both prophylaxis and targeted therapy.


Asunto(s)
Colesterol/toxicidad , Embolia por Colesterol/patología , Riñón/irrigación sanguínea , Riñón/patología , Insuficiencia Renal/patología , Animales , Embolia por Colesterol/inducido químicamente , Células Endoteliales/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Insuficiencia Renal/inducido químicamente
17.
J Am Soc Nephrol ; 31(4): 799-816, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32086278

RESUMEN

BACKGROUND: Hereditary deficiency of adenine phosphoribosyltransferase causes 2,8-dihydroxyadenine (2,8-DHA) nephropathy, a rare condition characterized by formation of 2,8-DHA crystals within renal tubules. Clinical relevance of rodent models of 2,8-DHA crystal nephropathy induced by excessive adenine intake is unknown. METHODS: Using animal models and patient kidney biopsies, we assessed the pathogenic sequelae of 2,8-DHA crystal-induced kidney damage. We also used knockout mice to investigate the role of TNF receptors 1 and 2 (TNFR1 and TNFR2), CD44, or alpha2-HS glycoprotein (AHSG), all of which are involved in the pathogenesis of other types of crystal-induced nephropathies. RESULTS: Adenine-enriched diet in mice induced 2,8-DHA nephropathy, leading to progressive kidney disease, characterized by crystal deposits, tubular injury, inflammation, and fibrosis. Kidney injury depended on crystal size. The smallest crystals were endocytosed by tubular epithelial cells. Crystals of variable size were excreted in urine. Large crystals obstructed whole tubules. Medium-sized crystals induced a particular reparative process that we term extratubulation. In this process, tubular cells, in coordination with macrophages, overgrew and translocated crystals into the interstitium, restoring the tubular luminal patency; this was followed by degradation of interstitial crystals by granulomatous inflammation. Patients with adenine phosphoribosyltransferase deficiency showed similar histopathological findings regarding crystal morphology, crystal clearance, and renal injury. In mice, deletion of Tnfr1 significantly reduced tubular CD44 and annexin two expression, as well as inflammation, thereby ameliorating the disease course. In contrast, genetic deletion of Tnfr2, Cd44, or Ahsg had no effect on the manifestations of 2,8-DHA nephropathy. CONCLUSIONS: Rodent models of the cellular and molecular mechanisms of 2,8-DHA nephropathy and crystal clearance have clinical relevance and offer insight into potential future targets for therapeutic interventions.


Asunto(s)
Adenina Fosforribosiltransferasa/deficiencia , Adenina/análogos & derivados , Enfermedades Renales/etiología , Enfermedades Renales/patología , Errores Innatos del Metabolismo/etiología , Errores Innatos del Metabolismo/patología , Urolitiasis/etiología , Urolitiasis/patología , Adenina/fisiología , Adenina Fosforribosiltransferasa/metabolismo , Adulto , Animales , Estudios de Cohortes , Dieta , Modelos Animales de Enfermedad , Femenino , Humanos , Lactante , Masculino , Errores Innatos del Metabolismo/metabolismo , Ratones , Persona de Mediana Edad , Urolitiasis/metabolismo
18.
Nephrol Dial Transplant ; 35(1): 86-97, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31081025

RESUMEN

BACKGROUND: Dysbiosis, bacterial translocation and systemic inflammation have been found to be associated with human and experimental forms of chronic kidney disease (CKD), but the functional contribution of the intestinal microbiota to CKD-related intestinal barrier dysfunction and CKD progression is unknown, especially in CKD secondary to hyperoxaluria and nephrocalcinosis. METHODS: C57BL/6N mice fed an oxalate-rich diet for either 10 or 20 days developed reversible or progressive kidney disease, respectively. RESULTS: Oxalate-induced CKD manifested as azotaemia, renal anaemia and hyperkalaemia. CKD was associated with persistent dysbiosis and intestinal barrier dysfunction. Local as well as systemic inflammation was evident and partially persisted despite better renal function after returning to an oxalate-free diet, indicating some innate immune memory. Eradication of the microbiota with a combination of antibiotics improved intestinal barrier function but had no effect on renal function, nephrocalcinosis, kidney remodelling and atrophy compared with control mice not receiving antibiotics. CONCLUSIONS: Together, in chronic oxalate nephropathy, the intestinal microbiota contributes to the CKD-related dysfunction of the intestinal barrier but not to the progression of nephrocalcinosis itself, as well to its related kidney atrophy and excretory dysfunction.


Asunto(s)
Disbiosis/etiología , Microbioma Gastrointestinal , Hiperoxaluria/complicaciones , Inflamación/etiología , Nefrocalcinosis/complicaciones , Insuficiencia Renal Crónica/etiología , Animales , Progresión de la Enfermedad , Disbiosis/patología , Humanos , Inflamación/patología , Intestinos/microbiología , Masculino , Ratones , Ratones Endogámicos C57BL , Insuficiencia Renal Crónica/patología
19.
FEBS J ; 287(5): 818-832, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31829497

RESUMEN

Crystals and nano- and microparticles form inside the human body from intrinsic proteins, minerals, or metabolites or enter the body as particulate matter from occupational and environmental sources. Associated tissue injuries and diseases mostly develop from cellular responses to such crystal deposits and include inflammation, cell necrosis, granuloma formation, tissue fibrosis, and stone-related obstruction of excretory organs. But how do crystals and nano- and microparticles trigger these biological processes? Which pathomechanisms are identical across different particle types, sizes, and shapes? In addition, which mechanisms are specific to the atomic or molecular structure of crystals or to specific sizes or shapes? Do specific cellular or molecular mechanisms qualify as target for therapeutic interventions? Here, we provide a guide to approach this diverse and multidisciplinary research domain. We give an overview about the clinical spectrum of crystallopathies, about shared and specific pathomechanisms as a conceptual overview before digging deeper into the specialty field of interest.


Asunto(s)
Material Particulado/metabolismo , Fibrosis/metabolismo , Fibrosis/patología , Granuloma/metabolismo , Granuloma/patología , Humanos , Inflamación/metabolismo , Inflamación/patología , Modelos Biológicos , Nanopartículas/metabolismo , Necrosis/metabolismo , Necrosis/patología
20.
Pharmacol Res ; 152: 104617, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31881272

RESUMEN

Increased interest in understanding the liver-kidney axis in health and disease during the last decade unveiled multiple recent evidence that suggested a strong association of fatty liver diseases with chronic kidney disease (CKD). Low-grade systemic inflammation is thought to be the major contributing factor to the pathogenesis of CKD associated with fatty liver. However, other contributing factors largely remained unclear, for example, gut microbiota and intestinal barrier integrity. Homeostasis of the gut microbiome is very crucial for the health of an individual. Imbalance in the gut microbiota leads to various diseases like fatty liver disease and CKD. On the contrary, disease conditions can also distinctly change gut microbiota. In this review, we propose the pathogenic role of the gut-liver-kidney axis in the development and progression of CKD associated with chronic fatty liver diseases, either non-alcoholic fatty liver disease or non-alcoholic steatohepatitis in experimental models and humans. Further, we discuss the therapeutic potential and highlight the future research directions for therapeutic targeting of the gut-liver-kidney axis.


Asunto(s)
Hígado Graso/complicaciones , Microbioma Gastrointestinal , Intestinos , Riñón , Hígado , Insuficiencia Renal Crónica/etiología , Animales , Hígado Graso/microbiología , Humanos , Insuficiencia Renal Crónica/microbiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...