Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 123
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
JCI Insight ; 2024 Sep 03.
Article de Anglais | MEDLINE | ID: mdl-39226116

RÉSUMÉ

Dual endothelin-1 (ET-1) and angiotensin II (AngII) receptor antagonism with sparsentan has strong antiproteinuric actions via multiple potential mechanisms that are more pronounced, or additive compared to current standard of care using angiotensin receptor blockers (ARB). Considering the many actions of ET-1 and AngII on multiple cell types, this study aimed to determine glomeruloprotective mechanisms of sparsentan compared to the ARB losartan by direct visualization of its effects in the intact kidney in focal segmental glomerulosclerosis (FSGS) using intravital multiphoton microscopy. In both healthy and FSGS models, sparsentan treatment increased afferent/efferent arteriole diameters, increased or preserved blood flow and single nephron glomerular filtration rate, attenuated acute ET-1+AngII-induced increases in podocyte calcium, reduced proteinuria, preserved podocyte number, increased both endothelial and renin lineage cells and clones in vasculature, glomeruli and tubules, restored glomerular endothelial glycocalyx, attenuated mitochondrial stress and immune cell homing. These effects were either not observed or of smaller magnitude with losartan. The pleiotropic nephroprotective effects of sparsentan included improved hemodynamics, podocyte and endothelial cell functions, and tissue repair. Compared to losartan, sparsentan was more effective in the sustained preservation of kidney structure and function, which underscores the importance of the ET-1 component in FSGS pathogenesis and therapy.

2.
Kidney Int ; 106(3): 362-364, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39174197

RÉSUMÉ

Yamamoto et al. developed an exciting technical advance to examine intracellular adenosine triphosphate levels with single-cell resolution in intact living kidney tissue, including in tubular and vascular segments that lie deep under the kidney surface. The work is a significant advance on prior in vivo biosensor studies, and it allows for mechanistic investigation of alterations in cell metabolism, kidney disease pathobiology, and the effects of drug treatments on energy sources in different kidney cell types.


Sujet(s)
Adénosine triphosphate , Techniques de biocapteur , Rein , Adénosine triphosphate/métabolisme , Adénosine triphosphate/analyse , Techniques de biocapteur/méthodes , Humains , Rein/anatomopathologie , Rein/métabolisme , Animaux , Analyse sur cellule unique/méthodes , Métabolisme énergétique/effets des médicaments et des substances chimiques , Maladies du rein/anatomopathologie , Maladies du rein/métabolisme , Maladies du rein/diagnostic
3.
J Am Heart Assoc ; 13(10): e033998, 2024 May 21.
Article de Anglais | MEDLINE | ID: mdl-38726925

RÉSUMÉ

BACKGROUND: The vasoconstrictor effects of angiotensin II via type 1 angiotensin II receptors in vascular smooth muscle cells are well established, but the direct effects of angiotensin II on vascular endothelial cells (VECs) in vivo and the mechanisms how VECs may mitigate angiotensin II-mediated vasoconstriction are not fully understood. The present study aimed to explore the molecular mechanisms and pathophysiological relevance of the direct actions of angiotensin II on VECs in kidney and brain microvessels in vivo. METHODS AND RESULTS: Changes in VEC intracellular calcium ([Ca2+]i) and nitric oxide (NO) production were visualized by intravital multiphoton microscopy of cadherin 5-Salsa6f mice or the endothelial uptake of NO-sensitive dye 4-amino-5-methylamino-2',7'-difluorofluorescein diacetate, respectively. Kidney fibrosis by unilateral ureteral obstruction and Ready-to-use adeno-associated virus expressing Mouse Renin 1 gene (Ren1-AAV) hypertension were used as disease models. Acute systemic angiotensin II injections triggered >4-fold increases in VEC [Ca2+]i in brain and kidney resistance arterioles and capillaries that were blocked by pretreatment with the type 1 angiotensin II receptor inhibitor losartan, but not by the type 2 angiotensin II receptor inhibitor PD123319. VEC responded to acute angiotensin II by increased NO production as indicated by >1.5-fold increase in 4-amino-5-methylamino-2',7'-difluorofluorescein diacetate fluorescence intensity. In mice with kidney fibrosis or hypertension, the angiotensin II-induced VEC [Ca2+]i and NO responses were significantly reduced, which was associated with more robust vasoconstrictions, VEC shedding, and microthrombi formation. CONCLUSIONS: The present study directly visualized angiotensin II-induced increases in VEC [Ca2+]i and NO production that serve to counterbalance agonist-induced vasoconstriction and maintain residual organ blood flow. These direct and endothelium-specific angiotensin II effects were blunted in disease conditions and linked to endothelial dysfunction and the development of vascular pathologies.


Sujet(s)
Angiotensine-II , Encéphale , Calcium , Hypertension artérielle , Rein , Microvaisseaux , Monoxyde d'azote , Vasoconstriction , Animaux , Souris , Angiotensine-II/pharmacologie , Encéphale/métabolisme , Encéphale/vascularisation , Calcium/métabolisme , Signalisation calcique/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine , Cellules endothéliales/métabolisme , Cellules endothéliales/effets des médicaments et des substances chimiques , Hypertension artérielle/métabolisme , Hypertension artérielle/physiopathologie , Hypertension artérielle/traitement médicamenteux , Rein/vascularisation , Rein/métabolisme , Souris de lignée C57BL , Microvaisseaux/métabolisme , Microvaisseaux/effets des médicaments et des substances chimiques , Microvaisseaux/anatomopathologie , Monoxyde d'azote/métabolisme , Vasoconstriction/effets des médicaments et des substances chimiques
4.
PNAS Nexus ; 3(5): pgae187, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38807632

RÉSUMÉ

Chronic and genetic kidney diseases such as autosomal dominant polycystic kidney disease (ADPKD) have few therapeutic options, and clinical trials testing small molecule drugs have been unfavorable due to low kidney bioavailability and adverse side effects. Although nanoparticles can be designed to deliver drugs directly to the diseased site, there are no kidney-targeted nanomedicines clinically available, and most FDA-approved nanoparticles are administered intravenously which is not ideal for chronic diseases. To meet these challenges of chronic diseases, we developed a biomaterials-based strategy using chitosan particles (CP) for oral delivery of therapeutic, kidney-targeting peptide amphiphile micelles (KMs). We hypothesized that encapsuling KMs into CP would enhance the bioavailability of KMs upon oral administration given the high stability of chitosan in acidic conditions and mucoadhesive properties enabling absorption within the intestines. To test this, we evaluated the mechanism of KM access to the kidneys via intravital imaging and investigated the KM biodistribution in a porcine model. Next, we loaded KMs carrying the ADPKD drug metformin into CP (KM-CP-met) and measured in vitro therapeutic effect. Upon oral administration in vivo, KM-CP-met showed significantly greater bioavailability and accumulation in the kidneys as compared to KM only or free drug. As such, KM-CP-met treatment in ADPKD mice (Pkd1fl/fl;Pax8-rtTA;Tet-O-Cre which develops the disease over 120 days and mimics the slow development of ADPKD) showed enhanced therapeutic efficacy without affecting safety despite repeated treatment. Herein, we demonstrate the potential of KM-CP as a nanomedicine strategy for oral delivery for the long-term treatment of chronic kidney diseases.

5.
J Clin Invest ; 134(11)2024 Apr 10.
Article de Anglais | MEDLINE | ID: mdl-38598837

RÉSUMÉ

Tissue regeneration is limited in several organs, including the kidney, contributing to the high prevalence of kidney disease globally. However, evolutionary and physiological adaptive responses and the presence of renal progenitor cells suggest an existing remodeling capacity. This study uncovered endogenous tissue remodeling mechanisms in the kidney that were activated by the loss of body fluid and salt and regulated by a unique niche of a minority renal cell type called the macula densa (MD). Here, we identified neuronal differentiation features of MD cells that sense the local and systemic environment and secrete angiogenic, growth, and extracellular matrix remodeling factors, cytokines and chemokines, and control resident progenitor cells. Serial intravital imaging, MD nerve growth factor receptor and Wnt mouse models, and transcriptome analysis revealed cellular and molecular mechanisms of these MD functions. Human and therapeutic translation studies illustrated the clinical potential of MD factors, including CCN1, as a urinary biomarker and therapeutic target in chronic kidney disease. The concept that a neuronally differentiated key sensory and regulatory cell type responding to organ-specific physiological inputs controls local progenitors to remodel or repair tissues may be applicable to other organs and diverse tissue-regenerative therapeutic strategies.


Sujet(s)
Différenciation cellulaire , Régénération , Animaux , Souris , Humains , Rein/métabolisme , Neurones/métabolisme , Neurones/anatomopathologie , Insuffisance rénale chronique/anatomopathologie , Insuffisance rénale chronique/métabolisme , Insuffisance rénale chronique/physiopathologie , Insuffisance rénale chronique/génétique , Mâle
6.
Hypertension ; 81(3): 447-455, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-37671571

RÉSUMÉ

Excess dietary salt (NaCl) intake is strongly correlated with cardiovascular disease and is a major contributing factor to the pathogenesis of hypertension. NaCl-sensitive hypertension is a multisystem disorder that involves renal dysfunction, vascular abnormalities, and neurogenically-mediated increases in peripheral resistance. Despite a major research focus on organ systems and these effector mechanisms causing NaCl-induced increases in arterial blood pressure, relatively less research has been directed at elucidating how NaCl is sensed by various tissues to elicit these downstream effects. The purpose of this review is to discuss how the brain, kidney, and gastrointestinal tract sense NaCl including key cell types, the role of NaCl versus osmolality, and the underlying molecular and electrochemical mechanisms.


Sujet(s)
Hypertension artérielle , Chlorure de sodium alimentaire , Humains , Chlorure de sodium alimentaire/métabolisme , Chlorure de sodium/métabolisme , Pression sanguine , Rein/métabolisme , Encéphale/métabolisme
7.
Sci Rep ; 12(1): 10623, 2022 06 23.
Article de Anglais | MEDLINE | ID: mdl-35739142

RÉSUMÉ

Primary open angle glaucoma (POAG) features an optic neuropathy, elevated aqueous humor (AH) TGFß2, and major risk factors of central corneal thickness (CCT), increasing age and intraocular pressure (IOP). We examined Tight skin (Tsk) mice to see if mutation of fibrillin-1, a repository for latent TGFß, is associated with characteristics of human POAG. We measured: CCT by ocular coherence tomography (OCT); IOP; retinal ganglion cell (RGC) and optic nerve axon counts by microscopic techniques; visual electrophysiologic scotopic threshold responses (STR) and pattern electroretinogram (PERG); and AH TGFß2 levels and activity by ELISA and MINK epithelial cell-based assays respectively. Tsk mice had open anterior chamber angles and compared with age-matched wild type (WT) mice: 23% thinner CCT (p < 0.003); IOP that was higher (p < 0.0001), more asymmetric (p = 0.047), rose with age (p = 0.04) and had a POAG-like frequency distribution. Tsk mice also had RGCs that were fewer (p < 0.04), declined with age (p = 0.0003) and showed increased apoptosis and glial activity; fewer optic nerve axons (p = 0.02); abnormal axons and glia; reduced STR (p < 0.002) and PERG (p < 0.007) visual responses; and higher AH TGFß2 levels (p = 0.0002) and activity (p = 1E-11) especially with age. Tsk mice showed defining features of POAG, implicating aberrant fibrillin-1 homeostasis as a pathogenic contributor to emergence of a POAG phenotype.


Sujet(s)
Humeur aqueuse , Fibrilline-1 , Glaucome à angle ouvert , Animaux , Humeur aqueuse/métabolisme , Fibrilline-1/génétique , Fibrilline-1/métabolisme , Glaucome à angle ouvert/anatomopathologie , Humains , Pression intraoculaire , Souris , Cellules ganglionnaires rétiniennes/anatomopathologie , Tonométrie oculaire , Facteur de croissance transformant bêta-2
8.
JCI Insight ; 7(1)2022 01 11.
Article de Anglais | MEDLINE | ID: mdl-34793332

RÉSUMÉ

Alport syndrome (AS) is a genetic disorder caused by mutations in type IV collagen that lead to defective glomerular basement membrane, glomerular filtration barrier (GFB) damage, and progressive chronic kidney disease. While the genetic basis of AS is well known, the molecular and cellular mechanistic details of disease pathogenesis have been elusive, hindering the development of mechanism-based therapies. Here, we performed intravital multiphoton imaging of the local kidney tissue microenvironment in a X-linked AS mouse model to directly visualize the major drivers of AS pathology. Severely distended glomerular capillaries and aneurysms were found accompanied by numerous microthrombi, increased glomerular endothelial surface layer (glycocalyx) and immune cell homing, GFB albumin leakage, glomerulosclerosis, and interstitial fibrosis by 5 months of age, with an intermediate phenotype at 2 months. Renal histology in mouse or patient tissues largely failed to detect capillary aberrations. Treatment of AS mice with hyaluronidase or the ACE inhibitor enalapril reduced the excess glomerular endothelial glycocalyx and blocked immune cell homing and GFB albumin leakage. This study identified central roles of glomerular mechanical forces and endothelial and immune cell activation early in AS, which could be therapeutically targeted to reduce mechanical strain and local tissue inflammation and improve kidney function.


Sujet(s)
Vaisseaux capillaires , Microscopie intravitale , Glomérule rénal , Néphropathie familiale avec surdité , Animaux , Vaisseaux capillaires/imagerie diagnostique , Vaisseaux capillaires/immunologie , Vaisseaux capillaires/anatomopathologie , Microenvironnement cellulaire/physiologie , Modèles animaux de maladie humaine , Humains , Glomérule rénal/vascularisation , Glomérule rénal/imagerie diagnostique , Glomérule rénal/immunologie , Glomérule rénal/anatomopathologie , Mâle , Souris , Néphropathie familiale avec surdité/imagerie diagnostique , Néphropathie familiale avec surdité/anatomopathologie
9.
Front Med (Lausanne) ; 8: 765356, 2021.
Article de Anglais | MEDLINE | ID: mdl-34722598

RÉSUMÉ

In the past two decades, intravital imaging using multiphoton microscopy has provided numerous new visual and mechanistic insights into glomerular biology and disease processes including the function of glomerular endothelial cells (GEnC), podocytes, and the development of proteinuria. Although glomerular endothelial injury is known to precede podocyte damage in several renal diseases, the primary role of GEnCs in proteinuria development received much less attention compared to the vast field of podocyte pathobiology. Consequently, our knowledge of GEnC mechanisms in glomerular diseases is still emerging. This review highlights new visual clues on molecular and cellular mechanisms of GEnCs and their crosstalk with podocytes and immune cells that were acquired recently by the application of multiphoton imaging of the intact glomerular microenvironment in various proteinuric disease models. New mechanisms of glomerular tissue remodeling and regeneration are discussed based on results of tracking the fate and function of individual GEnCs using serial intravital multiphoton imaging over several days and weeks. The three main topics of this review include (i) the role of endothelial injury and microthrombi in podocyte detachment and albumin leakage via hemodynamic and mechanical forces, (ii) the alterations of the endothelial surface layer (glycocalyx) and its interactions with circulating immune cells in lupus nephritis, and (iii) the structural and functional remodeling and regeneration of GEnCs in hypertension, diabetes, and other experimental injury conditions. By the comprehensive visual portrayal of GEnCs and the many other contributing glomerular cell types, this review emphasizes the complexity of pathogenic mechanisms that result in proteinuria development.

10.
Am J Physiol Renal Physiol ; 321(6): F689-F704, 2021 12 01.
Article de Anglais | MEDLINE | ID: mdl-34693742

RÉSUMÉ

Macula densa (MD) cells, a chief sensory cell type in the nephron, are endowed with unique microanatomic features including a high density of protein synthetic organelles and secretory vesicles in basal cell processes ("maculapodia") that suggest a so far unknown high rate of MD protein synthesis. This study aimed to explore the rate and regulation of MD protein synthesis and their effects on glomerular function using novel transgenic mouse models, newly established fluorescence cell biology techniques, and intravital microscopy. Sox2-tdTomato kidney tissue sections and an O-propargyl puromycin incorporation-based fluorescence imaging assay showed that MD cells have the highest level of protein synthesis within the kidney cortex followed by intercalated cells and podocytes. Genetic gain of function of mammalian target of rapamycin (mTOR) signaling specifically in MD cells (in MD-mTORgof mice) or their physiological activation by low-salt diet resulted in further significant increases in the synthesis of MD proteins. Specifically, these included both classic and recently identified MD-specific proteins such as cyclooxygenase 2, microsomal prostaglandin E2 synthase 1, and pappalysin 2. Intravital imaging of the kidney using multiphoton microscopy showed significant increases in afferent and efferent arteriole and glomerular capillary diameters and blood flow in MD-mTORgof mice coupled with an elevated glomerular filtration rate. The presently identified high rate of MD protein synthesis that is regulated by mTOR signaling is a novel component of the physiological activation and glomerular hemodynamic regulatory functions of MD cells that remains to be fully characterized.NEW & NOTEWORTHY This study discovered the high rate of protein synthesis in macula densa (MD) cells by applying direct imaging techniques with single cell resolution. Physiological activation and mammalian target of rapamycin signaling played important regulatory roles in this process. This new feature is a novel component of the tubuloglomerular cross talk and glomerular hemodynamic regulatory functions of MD cells. Future work is needed to elucidate the nature and (patho)physiological role of the specific proteins synthesized by MD cells.


Sujet(s)
Appareil juxtaglomérulaire/métabolisme , Biosynthèse des protéines , Animaux , Communication autocrine , Régime pauvre en sel , Débit de filtration glomérulaire , Protéines à fluorescence verte/génétique , Protéines à fluorescence verte/métabolisme , Humains , Microscopie intravitale , Appareil juxtaglomérulaire/cytologie , Protéines luminescentes/génétique , Protéines luminescentes/métabolisme , Souris de lignée C57BL , Souris transgéniques , Microscopie de fluorescence multiphotonique , Nitric oxide synthase type I/génétique , Nitric oxide synthase type I/métabolisme , Communication paracrine , Rénine/métabolisme , Transduction du signal , Sodium alimentaire/métabolisme , Sérine-thréonine kinases TOR/génétique , Sérine-thréonine kinases TOR/métabolisme , Protéine-2 du complexe de la sclérose tubéreuse/génétique , Protéine-2 du complexe de la sclérose tubéreuse/métabolisme ,
11.
Nat Commun ; 12(1): 2595, 2021 05 10.
Article de Anglais | MEDLINE | ID: mdl-33972536

RÉSUMÉ

Tissue regeneration is a process that recapitulates and restores organ structure and function. Although previous studies have demonstrated wound-induced hair neogenesis (WIHN) in laboratory mice (Mus), the regeneration is limited to the center of the wound unlike those observed in African spiny (Acomys) mice. Tissue mechanics have been implicated as an integral part of tissue morphogenesis. Here, we use the WIHN model to investigate the mechanical and molecular responses of laboratory and African spiny mice, and report these models demonstrate opposing trends in spatiotemporal morphogenetic field formation with association to wound stiffness landscapes. Transcriptome analysis and K14-Cre-Twist1 transgenic mice show the Twist1 pathway acts as a mediator for both epidermal-dermal interactions and a competence factor for periodic patterning, differing from those used in development. We propose a Turing model based on tissue stiffness that supports a two-scale tissue mechanics process: (1) establishing a morphogenetic field within the wound bed (mm scale) and (2) symmetry breaking of the epidermis and forming periodically arranged hair primordia within the morphogenetic field (µm scale). Thus, we delineate distinct chemo-mechanical events in building a Turing morphogenesis-competent field during WIHN of laboratory and African spiny mice and identify its evo-devo advantages with perspectives for regenerative medicine.


Sujet(s)
Épiderme/anatomie et histologie , Épiderme/métabolisme , Follicule pileux/métabolisme , Morphogenèse/physiologie , Régénération/physiologie , Protéine-1 apparentée à Twist/métabolisme , Cicatrisation de plaie/physiologie , Animaux , Épiderme/physiologie , Analyse de profil d'expression de gènes , Follicule pileux/anatomie et histologie , Follicule pileux/physiologie , Immunohistochimie , Souris , Souris de lignée C57BL , Souris transgéniques , Analyse sur microréseau , Microscopie à force atomique , Modèles psychologiques , Morphogenèse/génétique , Murinae , RNA-Seq , Régénération/génétique , Médecine régénérative , Transduction du signal/génétique , Transduction du signal/physiologie , Analyse spatio-temporelle , Protéine-1 apparentée à Twist/génétique , Cicatrisation de plaie/génétique
12.
JCI Insight ; 6(10)2021 05 24.
Article de Anglais | MEDLINE | ID: mdl-33848265

RÉSUMÉ

Endothelial cells are important in the maintenance of healthy blood vessels and in the development of vascular diseases. However, the origin and dynamics of endothelial precursors and remodeling at the single-cell level have been difficult to study in vivo owing to technical limitations. Therefore, we aimed to develop a direct visual approach to track the fate and function of single endothelial cells over several days and weeks in the same vascular bed in vivo using multiphoton microscopy (MPM) of transgenic Cdh5-Confetti mice and the kidney glomerulus as a model. Individual cells of the vascular endothelial lineage were identified and tracked owing to their unique color combination, based on the random expression of cyan/green/yellow/red fluorescent proteins. Experimental hypertension, hyperglycemia, and laser-induced endothelial cell ablation rapidly increased the number of new glomerular endothelial cells that appeared in clusters of the same color, suggesting clonal cell remodeling by local precursors at the vascular pole. Furthermore, intravital MPM allowed the detection of distinct structural and functional alterations of proliferating endothelial cells. No circulating Cdh5-Confetti+ cells were found in the renal cortex. Moreover, the heart, lung, and kidneys showed more significant clonal endothelial cell expansion compared with the brain, pancreas, liver, and spleen. In summary, we have demonstrated that serial MPM of Cdh5-Confetti mice in vivo is a powerful technical advance to study endothelial remodeling and repair in the kidney and other organs under physiological and disease conditions.


Sujet(s)
Endothélium vasculaire , Microscopie intravitale/méthodes , Glomérule rénal , Analyse sur cellule unique/méthodes , Animaux , Endothélium vasculaire/cytologie , Endothélium vasculaire/imagerie diagnostique , Endothélium vasculaire/physiologie , Glomérule rénal/cytologie , Glomérule rénal/imagerie diagnostique , Glomérule rénal/physiologie , Souris , Souris transgéniques
13.
Am J Physiol Renal Physiol ; 320(3): F492-F504, 2021 03 01.
Article de Anglais | MEDLINE | ID: mdl-33491562

RÉSUMÉ

Although macula densa (MD) cells are chief regulatory cells in the nephron with unique microanatomical features, they have been difficult to study in full detail due to their inaccessibility and limitations in earlier microscopy techniques. The present study used a new mouse model with a comprehensive imaging approach to visualize so far unexplored microanatomical features of MD cells, their regulation, and functional relevance. MD-GFP mice with conditional and partial induction of green fluorescent protein (GFP) expression, which specifically and intensely illuminated only single MD cells, were used with fluorescence microscopy of fixed tissue and live MD cells in vitro and in vivo with complementary electron microscopy of the rat, rabbit, and human kidney. An elaborate network of major and minor cell processes, here named maculapodia, were found at the cell base, projecting toward other MD cells and the glomerular vascular pole. The extent of maculapodia showed upregulation by low dietary salt intake and the female sex. Time-lapse imaging of maculapodia revealed highly dynamic features including rapid outgrowth and an extensive vesicular transport system. Electron microscopy of rat, rabbit, and human kidneys and three-dimensional volume reconstruction in optically cleared whole-mount MD-GFP mouse kidneys further confirmed the presence and projections of maculapodia into the extraglomerular mesangium and afferent and efferent arterioles. The newly identified dynamic and secretory features of MD cells suggest the presence of novel functional and molecular pathways of cell-to-cell communication in the juxtaglomerular apparatus between MD cells and between MD and other target cells.NEW & NOTEWORTHY This study illuminated a physiologically regulated dense network of basal cell major and minor processes (maculapodia) in macula densa (MD) cells. The newly identified dynamic and secretory features of these microanatomical structures suggest the presence of novel functional and molecular pathways of cell-to-cell communication in the juxtaglomerular apparatus between MD and other target cells. Detailed characterization of the function and molecular details of MD cell intercellular communications and their role in physiology and disease warrant further studies.


Sujet(s)
Mésangium glomérulaire/ultrastructure , Appareil juxtaglomérulaire/ultrastructure , Glomérule rénal/ultrastructure , Tubules rénaux/ultrastructure , Animaux , Communication cellulaire/physiologie , Cellules épithéliales/cytologie , Cellules épithéliales/ultrastructure , Mésangium glomérulaire/cytologie , Glomérule rénal/cytologie , Tubules rénaux/cytologie , Souris , Lapins , Rats
14.
Physiol Int ; 2021 Dec 16.
Article de Anglais | MEDLINE | ID: mdl-34978536

RÉSUMÉ

Podocyte calcium (Ca2+) signaling plays important roles in the (patho)physiology of the glomerular filtration barrier. Overactivation of podocyte transient receptor potential canonical (TRPC) channels including TRPC6 and purinergic signaling via P2 receptors that are known mechanosensors can increase podocyte intracellular Ca2+ levels ([Ca2+]i) and cause cell injury, proteinuria and glomerular disease including in diabetes. However, important mechanistic details of the trigger and activation of these pathways in vivo in the intact glomerular environment are lacking. Here we show direct visual evidence that podocytes can sense mechanical overload (increased glomerular capillary pressure) and metabolic alterations (increased plasma glucose) via TRPC6 and purinergic receptors including P2Y2. Multiphoton microscopy of podocyte [Ca2+]i was performed in vivo using wild-type and TRPC6 or P2Y2 knockout (KO) mice expressing the calcium reporter GCaMP3/5 only in podocytes and in vitro using freshly dissected microperfused glomeruli. Single-nephron intra-glomerular capillary pressure elevations induced by obstructing the efferent arteriole lumen with laser-induced microthrombus in vivo and by a micropipette in vitro triggered >2-fold increases in podocyte [Ca2+]i. These responses were blocked in TRPC6 and P2Y2 KO mice. Acute elevations of plasma glucose caused >4-fold increases in podocyte [Ca2+]i that were abolished by pharmacological inhibition of TRPC6 or P2 receptors using SAR7334 or suramin treatment, respectively. This study established the role of Ca2+ signaling via TRPC6 channels and P2 receptors in mechanical and metabolic sensing of podocytes in vivo, which are promising therapeutic targets in conditions with high intra-glomerular capillary pressure and plasma glucose, such as diabetic and hypertensive nephropathy.

15.
JCI Insight ; 5(19)2020 10 02.
Article de Anglais | MEDLINE | ID: mdl-32870819

RÉSUMÉ

Lupus nephritis (LN) is a major organ complication and cause of morbidity and mortality in patients with systemic lupus erythematosus (SLE). There is an unmet medical need for developing more efficient and specific, mechanism-based therapies, which depends on improved understanding of the underlying LN pathogenesis. Here we present direct visual evidence from high-power intravital imaging of the local kidney tissue microenvironment in mouse models showing that activated memory T cells originated in immune organs and the LN-specific robust accumulation of the glomerular endothelial glycocalyx played central roles in LN development. The glomerular homing of T cells was mediated via the direct binding of their CD44 to the hyaluronic acid (HA) component of the endothelial glycocalyx, and glycocalyx-degrading enzymes efficiently disrupted homing. Short-course treatment with either hyaluronidase or heparinase III provided long-term organ protection as evidenced by vastly improved albuminuria and survival rate. This glycocalyx/HA/memory T cell interaction is present in multiple SLE-affected organs and may be therapeutically targeted for SLE complications, including LN.


Sujet(s)
Endothélium vasculaire/immunologie , Glycocalyx/métabolisme , Hyaluronoglucosaminidase/administration et posologie , Glomérule rénal/immunologie , Glomérulonéphrite lupique/prévention et contrôle , Polysaccharide-lyases/administration et posologie , Lymphocytes T/immunologie , Animaux , Endothélium vasculaire/métabolisme , Endothélium vasculaire/anatomopathologie , Femelle , Acide hyaluronique/métabolisme , Mémoire immunologique/immunologie , Glomérule rénal/métabolisme , Glomérule rénal/anatomopathologie , Glomérulonéphrite lupique/immunologie , Glomérulonéphrite lupique/métabolisme , Glomérulonéphrite lupique/anatomopathologie , Mâle , Souris , Souris de lignée C57BL , Souris knockout , Lymphocytes T/métabolisme , Lymphocytes T/anatomopathologie
16.
J Am Soc Nephrol ; 31(7): 1555-1568, 2020 07.
Article de Anglais | MEDLINE | ID: mdl-32487560

RÉSUMÉ

BACKGROUND: The physiologic role of renomedullary interstitial cells, which are uniquely and abundantly found in the renal inner medulla, is largely unknown. Endothelin A receptors regulate multiple aspects of renomedullary interstitial cell function in vitro. METHODS: To assess the effect of targeting renomedullary interstitial cell endothelin A receptors in vivo, we generated a mouse knockout model with inducible disruption of renomedullary interstitial cell endothelin A receptors at 3 months of age. RESULTS: BP and renal function were similar between endothelin A receptor knockout and control mice during normal and reduced sodium or water intake. In contrast, on a high-salt diet, compared with control mice, the knockout mice had reduced BP; increased urinary sodium, potassium, water, and endothelin-1 excretion; increased urinary nitrite/nitrate excretion associated with increased noncollecting duct nitric oxide synthase-1 expression; increased PGE2 excretion associated with increased collecting duct cyclooxygenase-1 expression; and reduced inner medullary epithelial sodium channel expression. Water-loaded endothelin A receptor knockout mice, compared with control mice, had markedly enhanced urine volume and reduced urine osmolality associated with increased urinary endothelin-1 and PGE2 excretion, increased cyclooxygenase-2 protein expression, and decreased inner medullary aquaporin-2 protein content. No evidence of endothelin-1-induced renomedullary interstitial cell contraction was observed. CONCLUSIONS: Disruption of renomedullary interstitial cell endothelin A receptors reduces BP and increases salt and water excretion associated with enhanced production of intrinsic renal natriuretic and diuretic factors. These studies indicate that renomedullary interstitial cells can modulate BP and renal function under physiologic conditions.


Sujet(s)
Pression sanguine , Médulla rénale/physiologie , Récepteur de type A de l'endothéline/physiologie , Aldostérone/sang , Animaux , Arginine vasopressine/urine , Calcium/métabolisme , Diurèse/effets des médicaments et des substances chimiques , Endothéline-1/pharmacologie , Endothéline-1/urine , Canaux sodium épithéliaux/métabolisme , Femelle , Génotype , Débit de filtration glomérulaire , Acide hyaluronique/métabolisme , Médulla rénale/cytologie , Médulla rénale/métabolisme , Mâle , Souris , Souris knockout , Modèles animaux , Natriurèse/effets des médicaments et des substances chimiques , Nitrates/urine , Nitrites/urine , Potassium/urine , ARN messager/métabolisme , Récepteur de type A de l'endothéline/génétique , Récepteur de type A de l'endothéline/métabolisme , Modulateurs sélectifs des récepteurs des oestrogènes/pharmacologie , Sodium/urine , Chlorure de sodium alimentaire/administration et posologie , Tamoxifène/pharmacologie , Eau/administration et posologie , Eau/métabolisme
17.
Methods Mol Biol ; 2150: 243, 2020.
Article de Anglais | MEDLINE | ID: mdl-32072481

RÉSUMÉ

The original version of this chapter was inadvertently published without a proper acknowledgement. The authors informed to insert the following acknowledgement in this chapter.

18.
Methods Mol Biol ; 2150: 25-44, 2020.
Article de Anglais | MEDLINE | ID: mdl-31087287

RÉSUMÉ

Intravital multiphoton microscopy of the kidney is a powerful technique to study alterations in tissue morphology and function simultaneously in the living animal and represents a dynamic and developing research tool in the field. Recent technological advances include serial intravital multiphoton microscopy of the same kidney regions over several weeks and combined with ex vivo histology for cellular biomarker expression of the same cells, which had been subject to serial imaging before. Thus, serial intravital multiphoton microscopy followed by ex vivo histology provides unique tools to perform long-term cell fate tracing of the same renal cells during physiological and pathophysiological conditions, thereby allowing the detection of structural changes of the same renal cells over time. Examples include renal cell migration and proliferation while linking these events to local functional alterations and eventually to the expression of distinct cellular biomarkers. Here, we provide a detailed step-by-step protocol to facilitate serial intravital multiphoton microscopy for long-term in vivo tracking of renal cells and subsequent ex vivo histology for immunohistological staining of the same cells in the fixed tissue.


Sujet(s)
Suivi cellulaire/méthodes , Microscopie intravitale/méthodes , Rein/cytologie , Rein/imagerie diagnostique , Abdomen/imagerie diagnostique , Animaux , Colorants fluorescents/composition chimique , Injections , Rein/chirurgie , Souris
19.
Methods Cell Biol ; 154: 85-107, 2019.
Article de Anglais | MEDLINE | ID: mdl-31493823

RÉSUMÉ

Fluorescence microscopy techniques are powerful tools to study tissue dynamics, cellular function and biology both in vivo and in vitro. These tools allow for functional assessment and quantification along with qualitative analysis, thus providing a comprehensive understanding of various cellular processes under normal physiological and disease conditions. The main focus of this chapter is the recently developed method of serial intravital multiphoton microscopy that has helped shed light on the dynamic alterations of the spatial distribution and fate of single renal cells or cell populations and their migration patterns in the same tissue region over several days in response to various stimuli within the living kidney. This technique is very useful for studying in vivo the molecular and cellular mechanisms of tissue remodeling and repair after injury. In addition, complementary in vitro imaging tools are also described and discussed, like tissue clearing techniques and protein synthesis measurement in tissues in situ that provide an in depth assessment of changes at the cellular level. Thus, these novel fluorescence techniques can be effectively leveraged for different tissue types, experimental conditions as well as disease models to improve our understanding of renal cell biology.


Sujet(s)
Cellules épithéliales/ultrastructure , Microscopie intravitale/méthodes , Microscopie de fluorescence multiphotonique/méthodes , Néphrite/physiopathologie , Podocytes/ultrastructure , Obstruction urétérale/physiopathologie , Animaux , Mouvement cellulaire , Modèles animaux de maladie humaine , Doxorubicine/administration et posologie , Cellules épithéliales/métabolisme , Expression des gènes , Gènes rapporteurs , Protéines à fluorescence verte/génétique , Protéines à fluorescence verte/métabolisme , Microscopie intravitale/instrumentation , Protéines luminescentes/génétique , Protéines luminescentes/métabolisme , Souris , Souris transgéniques , Microscopie de fluorescence multiphotonique/instrumentation , Néphrite/induit chimiquement , Néphrite/métabolisme , Podocytes/métabolisme , Analyse sur cellule unique/méthodes , Obstruction urétérale/métabolisme ,
20.
Dev Cell ; 50(1): 102-116.e6, 2019 07 01.
Article de Anglais | MEDLINE | ID: mdl-31265809

RÉSUMÉ

The renal corpuscle of the kidney comprises a glomerular vasculature embraced by podocytes and supported by mesangial myofibroblasts, which ensure plasma filtration at the podocyte-generated slit diaphragm. With a spectrum of podocyte-expressed gene mutations causing chronic disease, an enhanced understanding of podocyte development and function to create relevant in vitro podocyte models is a clinical imperative. To characterize podocyte development, scRNA-seq was performed on human fetal kidneys, identifying distinct transcriptional signatures accompanying the differentiation of functional podocytes from progenitors. Interestingly, organoid-generated podocytes exhibited highly similar, progressive transcriptional profiles despite an absence of the vasculature, although abnormal gene expression was pinpointed in late podocytes. On transplantation into mice, organoid-derived podocytes recruited the host vasculature and partially corrected transcriptional profiles. Thus, human podocyte development is mostly intrinsically regulated and vascular interactions refine maturation. These studies support the application of organoid-derived podocytes to model disease and to restore or replace normal kidney functions.


Sujet(s)
Différenciation cellulaire , Régulation de l'expression des gènes au cours du développement , Cellules souches pluripotentes induites/cytologie , Glomérule rénal/cytologie , Organoïdes/cytologie , Podocytes/cytologie , Analyse sur cellule unique/méthodes , Cellules cultivées , Femelle , Humains , Cellules souches pluripotentes induites/métabolisme , Glomérule rénal/métabolisme , Organoïdes/métabolisme , Podocytes/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE