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










Publication year range
1.
J Clin Invest ; 2024 Apr 10.
Article in English | MEDLINE | ID: mdl-38598837

ABSTRACT

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 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, 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.

2.
JCI Insight ; 6(10)2021 05 24.
Article in English | MEDLINE | ID: mdl-33848265

ABSTRACT

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.


Subject(s)
Endothelium, Vascular , Intravital Microscopy/methods , Kidney Glomerulus , Single-Cell Analysis/methods , Animals , Endothelium, Vascular/cytology , Endothelium, Vascular/diagnostic imaging , Endothelium, Vascular/physiology , Kidney Glomerulus/cytology , Kidney Glomerulus/diagnostic imaging , Kidney Glomerulus/physiology , Mice , Mice, Transgenic
3.
Am J Physiol Renal Physiol ; 320(3): F492-F504, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33491562

ABSTRACT

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.


Subject(s)
Glomerular Mesangium/ultrastructure , Juxtaglomerular Apparatus/ultrastructure , Kidney Glomerulus/ultrastructure , Kidney Tubules/ultrastructure , Animals , Cell Communication/physiology , Epithelial Cells/cytology , Epithelial Cells/ultrastructure , Glomerular Mesangium/cytology , Kidney Glomerulus/cytology , Kidney Tubules/cytology , Mice , Rabbits , Rats
4.
JCI Insight ; 5(19)2020 10 02.
Article in English | MEDLINE | ID: mdl-32870819

ABSTRACT

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.


Subject(s)
Endothelium, Vascular/immunology , Glycocalyx/metabolism , Hyaluronoglucosaminidase/administration & dosage , Kidney Glomerulus/immunology , Lupus Nephritis/prevention & control , Polysaccharide-Lyases/administration & dosage , T-Lymphocytes/immunology , Animals , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Female , Hyaluronic Acid/metabolism , Immunologic Memory/immunology , Kidney Glomerulus/metabolism , Kidney Glomerulus/pathology , Lupus Nephritis/immunology , Lupus Nephritis/metabolism , Lupus Nephritis/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocytes/metabolism , T-Lymphocytes/pathology
5.
Methods Cell Biol ; 154: 85-107, 2019.
Article in English | MEDLINE | ID: mdl-31493823

ABSTRACT

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.


Subject(s)
Epithelial Cells/ultrastructure , Intravital Microscopy/methods , Microscopy, Fluorescence, Multiphoton/methods , Nephritis/physiopathology , Podocytes/ultrastructure , Ureteral Obstruction/physiopathology , Animals , Cell Movement , Disease Models, Animal , Doxorubicin/administration & dosage , Epithelial Cells/metabolism , Gene Expression , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Intravital Microscopy/instrumentation , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , Mice, Transgenic , Microscopy, Fluorescence, Multiphoton/instrumentation , Nephritis/chemically induced , Nephritis/metabolism , Podocytes/metabolism , Single-Cell Analysis/methods , Ureteral Obstruction/metabolism , Red Fluorescent Protein
6.
Kidney Int ; 95(1): 94-107, 2019 01.
Article in English | MEDLINE | ID: mdl-30389198

ABSTRACT

Aldosterone contributes to end-organ damage in heart failure and chronic kidney disease. Mineralocorticoid-receptor inhibitors limit activation of the receptor by aldosterone and slow disease progression, but side effects, including hyperkalemia, limit their clinical use. Damage to the endothelial glycocalyx (a luminal biopolymer layer) has been implicated in the pathogenesis of endothelial dysfunction and albuminuria, but to date no one has investigated whether the glomerular endothelial glycocalyx is affected by aldosterone. In vitro, human glomerular endothelial cells exposed to 0.1 nM aldosterone and 145 mMol NaCl exhibited reduced cell surface glycocalyx components (heparan sulfate and syndecan-4) and disrupted shear sensing consistent with damage of the glycocalyx. In vivo, administration of 0.6 µg/g/d of aldosterone (subcutaneous minipump) and 1% NaCl drinking water increased glomerular matrix metalloproteinase 2 activity, reduced syndecan 4 expression, and caused albuminuria. Intravital multiphoton imaging confirmed that aldosterone caused damage of the glomerular endothelial glycocalyx and increased the glomerular sieving coefficient for albumin. Targeting matrix metalloproteinases 2 and 9 with a specific gelatinase inhibitor preserved the glycocalyx, blocked the rise in glomerular sieving coefficient, and prevented albuminuria. Together these data suggest that preservation of the glomerular endothelial glycocalyx may represent a novel strategy for limiting the pathological effects of aldosterone.


Subject(s)
Albuminuria/pathology , Aldosterone/metabolism , Glycocalyx/pathology , Renal Insufficiency, Chronic/pathology , Albuminuria/urine , Animals , Cell Line , Disease Models, Animal , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/pathology , Glycocalyx/drug effects , Heparitin Sulfate/metabolism , Humans , Kidney Glomerulus/cytology , Kidney Glomerulus/drug effects , Kidney Glomerulus/pathology , Male , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinase Inhibitors/pharmacology , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Renal Insufficiency, Chronic/urine , Sodium Chloride/pharmacology , Syndecan-4/metabolism
7.
Am J Physiol Renal Physiol ; 315(3): F521-F534, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29667908

ABSTRACT

The prorenin receptor (PRR) was originally proposed to be a member of the renin-angiotensin system (RAS); however, recent work questioned their association. The present paper describes a functional link between the PRR and RAS in the renal juxtaglomerular apparatus (JGA), a classic anatomical site of the RAS. PRR expression was found in the sensory cells of the JGA, the macula densa (MD), and immunohistochemistry-localized PRR to the MD basolateral cell membrane in mouse, rat, and human kidneys. MD cell PRR activation led to MAP kinase ERK1/2 signaling and stimulation of PGE2 release, the classic pathway of MD-mediated renin release. Exogenous renin or prorenin added to the in vitro microperfused JGA-induced acute renin release, which was inhibited by removing the MD or by the administration of a PRR decoy peptide. To test the function of MD PRR in vivo, we established a new mouse model with inducible conditional knockout (cKO) of the PRR in MD cells based on neural nitric oxide synthase-driven Cre-lox recombination. Deletion of the MD PRR significantly reduced blood pressure and plasma renin. Challenging the RAS by low-salt diet + captopril treatment caused further significant reductions in blood pressure, renal renin, cyclooxygenase-2, and microsomal PGE synthase expression in cKO vs. wild-type mice. These results suggest that the MD PRR is essential in a novel JGA short-loop feedback mechanism, which is integrated within the classic MD mechanism to control renin synthesis and release and to maintain blood pressure.


Subject(s)
Blood Pressure , Juxtaglomerular Apparatus/enzymology , Proton-Translocating ATPases/metabolism , Receptors, Cell Surface/metabolism , Renin-Angiotensin System , Renin/metabolism , Vacuolar Proton-Translocating ATPases/metabolism , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , Biosensing Techniques , Blood Pressure/drug effects , Captopril/pharmacology , Cyclooxygenase 2/metabolism , Diet, Sodium-Restricted , Dinoprostone/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , HEK293 Cells , Humans , Juxtaglomerular Apparatus/drug effects , Male , Mice, Inbred C57BL , Mice, Knockout , Prostaglandin-E Synthases/metabolism , Proton-Translocating ATPases/deficiency , Proton-Translocating ATPases/genetics , Rats, Sprague-Dawley , Receptors, Cell Surface/deficiency , Receptors, Cell Surface/genetics , Renin-Angiotensin System/drug effects , Secretory Pathway , Signal Transduction , Vacuolar Proton-Translocating ATPases/genetics , Prorenin Receptor
8.
Front Physiol ; 9: 1765, 2018.
Article in English | MEDLINE | ID: mdl-30618788

ABSTRACT

Chronic lithium administration for the treatment of bipolar disorder leads to nephrogenic diabetes insipidus (NDI), characterized by polyuria, natriuresis, kaliuresis, and collecting duct remodeling and cell proliferation among other features. Previously, using a 2-week lithium-induced NDI model, we reported that P2Y2 receptor (R) knockout mice are significantly resistant to polyuria, natriuresis, kaliuresis, and decrease in AQP2 protein abundance in the kidney relative to wild type mice. Here we show this protection is long-lasting, and is also associated with significant amelioration of lithium-induced collecting duct remodeling and cell proliferation. Age-matched wild type and knockout mice were fed regular (n = 5/genotype) or lithium-added (40 mmol/kg chow; n = 10/genotype) diet for 5 months and euthanized. Water intake, urine output and osmolality were monitored once in every month. Salt blocks were provided to mice on lithium-diet to prevent sodium loss. At the end of 5 months mice were euthanized and serum and kidney samples were analyzed. There was a steady increase in lithium-induced polyuria, natriuresis and kaliuresis in wild type mice over the 5-month period. Increases in these urinary parameters were very low in lithium-fed knockout mice, resulting in significantly widening differences between the wild type and knockout mice. Terminal AQP2 and NKCC2 protein abundances in the kidney were significantly higher in lithium-fed knockout vs. wild type mice. There were no significant differences in terminal serum lithium or sodium levels between the wild type and knockout mice. Confocal immunofluorescence microscopy revealed that lithium-induced marked remodeling of collecting duct with significantly increased proportion of [H+]-ATPase-positive intercalated cells and decreased proportion of AQP2-positive principal cells in the wild type, but not in knockout mice. Lithium-induced collecting duct cell proliferation (indicated by Ki67 labeling), was significantly lower in knockout vs. wild type mice. This is the first piece of evidence that purinergic signaling is potentially involved in lithium-induced collecting duct remodeling and cell proliferation. Our results demonstrate that genetic deletion of P2Y2-R protects against the key structural and functional alterations in Li-induced NDI, and underscore the potential utility of targeting this receptor for the treatment of NDI in bipolar patients on chronic lithium therapy.

9.
Purinergic Signal ; 13(2): 239-248, 2017 06.
Article in English | MEDLINE | ID: mdl-28233082

ABSTRACT

Previously, we localized ADP-activated P2Y12 receptor (R) in rodent kidney and showed that its blockade by clopidogrel bisulfate (CLPD) attenuates lithium (Li)-induced nephrogenic diabetes insipidus (NDI). Here, we evaluated the effect of prasugrel (PRSG) administration on Li-induced NDI in mice. Both CLPD and PRSG belong to the thienopyridine class of ADP receptor antagonists. Groups of age-matched adult male B6D2 mice (N = 5/group) were fed either regular rodent chow (CNT), or with added LiCl (40 mmol/kg chow) or PRSG in drinking water (10 mg/kg bw/day) or a combination of LiCl and PRSG for 14 days and then euthanized. Water intake and urine output were determined and blood and kidney tissues were collected and analyzed. PRSG administration completely suppressed Li-induced polydipsia and polyuria and significantly prevented Li-induced decreases in AQP2 protein abundance in renal cortex and medulla. However, PRSG either alone or in combination with Li did not have a significant effect on the protein abundances of NKCC2 or NCC in the cortex and/or medulla. Immunofluorescence microscopy revealed that PRSG administration prevented Li-induced alterations in cellular disposition of AQP2 protein in medullary collecting ducts. Serum Li, Na, and osmolality were not affected by the administration of PRSG. Similar to CLPD, PRSG administration had no effect on Li-induced increase in urinary Na excretion. However, unlike CLPD, PRSG did not augment Li-induced increase in urinary arginine vasopressin (AVP) excretion. Taken together, these data suggest that the pharmacological inhibition of P2Y12-R by the thienopyridine group of drugs may potentially offer therapeutic benefits in Li-induced NDI.


Subject(s)
Diabetes Insipidus, Nephrogenic/chemically induced , Kidney/drug effects , Lithium Chloride/toxicity , Prasugrel Hydrochloride/pharmacology , Purinergic P2Y Receptor Antagonists/pharmacology , Animals , Male , Mice
10.
Semin Nephrol ; 36(3): 220-36, 2016 05.
Article in English | MEDLINE | ID: mdl-27339387

ABSTRACT

Kidney cell death plays a key role in the progression of life-threatening renal diseases, such as acute kidney injury and chronic kidney disease. Injured and dying epithelial and endothelial cells take part in complex communication with the innate immune system, which drives the progression of cell death and the decrease in renal function. To improve our understanding of kidney cell death dynamics and its impact on renal disease, a study approach is needed that facilitates the visualization of renal function and morphology in real time. Intravital multiphoton microscopy of the kidney has been used for more than a decade and made substantial contributions to our understanding of kidney physiology and pathophysiology. It is a unique tool that relates renal structure and function in a time- and spatial-dependent manner. Basic renal function, such as microvascular blood flow regulation and glomerular filtration, can be determined in real time and homeostatic alterations, which are linked inevitably to cell death and can be depicted down to the subcellular level. This review provides an overview of the available techniques to study kidney dysfunction and inflammation in terms of cell death in vivo, and addresses how this novel approach can be used to improve our understanding of cell death dynamics in renal disease.


Subject(s)
Acute Kidney Injury/pathology , Cell Death , Kidney/pathology , Renal Insufficiency, Chronic/pathology , Cell Adhesion , Endothelial Cells/pathology , Humans , Intravital Microscopy , Kidney Tubules, Proximal/pathology , Leukocyte Rolling , Leukocytes/pathology , Microscopy, Fluorescence, Multiphoton
11.
Physiol Rep ; 4(5)2016 Mar.
Article in English | MEDLINE | ID: mdl-26997629

ABSTRACT

A previously healthy 7-year-old male presented with hypertensive emergency, hypokalemia, and elevated plasma renin activity and aldosterone levels. There was no evidence of virilization or cushingoid features. MRI of the abdomen revealed a large (5 × 5 × 3 cm) peripherally enhancing, heterogeneous mass arising from the left adrenal gland. The patient was treated for a suspected pheochromocytoma. However, his blood pressure was not responsive to alpha-blockade. Blood pressure was controlled with a calcium channel blocker and an angiotensin-converting enzyme (ACE) inhibitor. A complete surgical resection of the mass was performed. Postoperatively, his blood pressure normalized and he did not require antihypertensives. On pathological examination, the tumor tissue stained negative for chromogranin and positive for renin. The final diagnosis was renin-secreting adrenal corticoadenoma, an extremely rare adrenal tumor not previously reported in a pediatric patient. Malignant hypertension due to a renin-secreting tumor may need to be distinguished from a pheochromocytoma if alpha-adrenergic blockade is ineffective.


Subject(s)
Adrenal Cortex Neoplasms/diagnosis , Adrenocortical Adenoma/diagnosis , Hypertension, Malignant/diagnosis , Renin/metabolism , Adrenal Cortex Neoplasms/complications , Adrenal Cortex Neoplasms/surgery , Adrenocortical Adenoma/complications , Adrenocortical Adenoma/surgery , Child , Humans , Hypertension, Malignant/complications , Hypertension, Malignant/surgery , Male
12.
Curr Opin Nephrol Hypertens ; 25(3): 168-73, 2016 May.
Article in English | MEDLINE | ID: mdl-27008595

ABSTRACT

PURPOSE OF REVIEW: The review aims to provide a brief summary and evaluation of the current state of research that uses multiphoton fluorescence microscopy for intravital kidney imaging. RECENT FINDINGS: Direct visualization of the glomerular filter, proximal and distal tubule segments, and the renal vasculature in the living, intact kidney in zebrafish, mouse, and rat models with high temporal and spatial resolution provided new insights into the function of the normal and diseased kidney. New technical developments in fluorescence excitation and detection, in combination with transgenic animal models for cell function and fate mapping, and serial imaging of the same glomerulus in the same animal over several days further advanced the field of nephrology research, and the understanding of disease mechanisms. SUMMARY: Intravital multiphoton imaging has solved many critical technical barriers in kidney research and allowed the dynamic portrayal of the structure and function of various renal cell types in vivo. It has become a widely used research technique, with significant past achievements, and tremendous potential for future development and applications for the study and better understanding of kidney diseases.


Subject(s)
Intravital Microscopy , Kidney Diseases/diagnosis , Kidney/pathology , Microscopy, Fluorescence, Multiphoton , Animals , Epithelial Cells/metabolism , Epithelial Cells/pathology , Humans , Kidney/blood supply , Kidney Diseases/pathology , Microscopy, Fluorescence, Multiphoton/methods , Models, Animal
13.
Purinergic Signal ; 11(4): 507-18, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26386699

ABSTRACT

Lithium (Li) administration causes deranged expression and function of renal aquaporins and sodium channels/transporters resulting in nephrogenic diabetes insipidus (NDI). Extracellular nucleotides (ATP/ADP/UTP), via P2 receptors, regulate these transport functions. We tested whether clopidogrel bisulfate (CLPD), an antagonist of ADP-activated P2Y(12) receptor, would affect Li-induced alterations in renal aquaporins and sodium channels/transporters. Adult mice were treated for 14 days with CLPD and/or Li and euthanized. Urine and kidneys were collected for analysis. When administered with Li, CLPD ameliorated polyuria, attenuated the rise in urine prostaglandin E2 (PGE2), and resulted in significantly higher urinary arginine vasopressin (AVP) and aldosterone levels as compared to Li treatment alone. However, urine sodium excretion remained elevated. Semi-quantitative immunoblotting revealed that CLPD alone increased renal aquaporin 2 (AQP2), Na-K-2Cl cotransporter (NKCC2), Na-Cl cotransporter (NCC), and the subunits of the epithelial Na channel (ENaC) in medulla by 25-130 %. When combined with Li, CLPD prevented downregulation of AQP2, Na-K-ATPase, and NKCC2 but was less effective against downregulation of cortical α- or γ-ENaC (70 kDa band). Thus, CLPD primarily attenuated Li-induced downregulation of proteins involved in water conservation (AVP-sensitive), with modest effects on aldosterone-sensitive proteins potentially explaining sustained natriuresis. Confocal immunofluorescence microscopy revealed strong labeling for P2Y(12)-R in proximal tubule brush border and blood vessels in the cortex and less intense labeling in medullary thick ascending limb and the collecting ducts. Therefore, there is the potential for CLPD to be directly acting at the tubule sites to mediate these effects. In conclusion, P2Y(12)-R may represent a novel therapeutic target for Li-induced NDI.


Subject(s)
Body Water/metabolism , Kidney/metabolism , Lithium/pharmacology , Platelet Aggregation Inhibitors/pharmacology , Sodium Channels/metabolism , Ticlopidine/analogs & derivatives , Aldosterone/urine , Animals , Aquaporin 2/metabolism , Aquaporins/metabolism , Arginine Vasopressin/urine , Clopidogrel , Dinoprostone/urine , Epithelial Sodium Channels/metabolism , Kidney/drug effects , Male , Mice , Polyuria/chemically induced , Polyuria/drug therapy , Receptors, Purinergic P2Y12/drug effects , Receptors, Purinergic P2Y12/metabolism , Sodium Channels/drug effects , Solute Carrier Family 12, Member 1/metabolism , Ticlopidine/pharmacology
14.
Kidney Int ; 88(1): 44-51, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25738253

ABSTRACT

Intravital imaging using multiphoton microscopy (MPM) has become an increasingly popular and widely used experimental technique in kidney research over the past few years. MPM allows deep optical sectioning of the intact, living kidney tissue with submicron resolution, which is unparalleled among intravital imaging approaches. MPM has solved a long-standing critical technical barrier in renal research to study several complex and inaccessible cell types and anatomical structures in vivo in their native environment. Comprehensive and quantitative kidney structure and function MPM studies helped our better understanding of the cellular and molecular mechanisms of the healthy and diseased kidney. This review summarizes recent in vivo MPM studies with a focus on the glomerulus and the filtration barrier, although select, glomerulus-related renal vascular and tubular functions are also mentioned. The latest applications of serial MPM of the same glomerulus in vivo, in the intact kidney over several days, during the progression of glomerular disease are discussed. This visual approach, in combination with genetically encoded fluorescent markers of cell lineage, has helped track the fate and function (e.g., cell calcium changes) of single podocytes during the development of glomerular pathologies, and provided visual proof for the highly dynamic, rather than static, nature of the glomerular environment. Future intravital imaging applications have the promise to further push the limits of optical microscopy, and to advance our understanding of the mechanisms of kidney injury. Also, MPM will help to study new mechanisms of tissue repair and regeneration, a cutting-edge area of kidney research.


Subject(s)
Glomerular Filtration Barrier/anatomy & histology , Glomerular Filtration Barrier/physiology , Intravital Microscopy/methods , Kidney Diseases/pathology , Microscopy, Fluorescence, Multiphoton , Animals , Calcium/metabolism , Cell Movement , Kidney Diseases/physiopathology , Mice , Podocytes/physiology , Rats , Time-Lapse Imaging , Zebrafish
15.
J Clin Invest ; 124(5): 2050-8, 2014 May.
Article in English | MEDLINE | ID: mdl-24713653

ABSTRACT

Intracellular calcium ([Ca²âº]i) signaling mediates physiological and pathological processes in multiple organs, including the renal podocyte; however, in vivo podocyte [Ca²âº]i dynamics are not fully understood. Here we developed an imaging approach that uses multiphoton microscopy (MPM) to directly visualize podocyte [Ca²âº]i dynamics within the intact kidneys of live mice expressing a fluorescent calcium indicator only in these cells. [Ca²âº]i was at a low steady-state level in control podocytes, while Ang II infusion caused a minor elevation. Experimental focal podocyte injury triggered a robust and sustained elevation of podocyte [Ca²âº]i around the injury site and promoted cell-to-cell propagating podocyte [Ca²âº]i waves along capillary loops. [Ca²âº]i wave propagation was ameliorated by inhibitors of purinergic [Ca²âº]i signaling as well as in animals lacking the P2Y2 purinergic receptor. Increased podocyte [Ca²âº]i resulted in contraction of the glomerular tuft and increased capillary albumin permeability. In preclinical models of renal fibrosis and glomerulosclerosis, high podocyte [Ca²âº]i correlated with increased cell motility. Our findings provide a visual demonstration of the in vivo importance of podocyte [Ca²âº]i in glomerular pathology and suggest that purinergic [Ca²âº]i signaling is a robust and key pathogenic mechanism in podocyte injury. This in vivo imaging approach will allow future detailed investigation of the molecular and cellular mechanisms of glomerular disease in the intact living kidney.


Subject(s)
Calcium Signaling , Calcium/metabolism , Cell Movement , Glomerulosclerosis, Focal Segmental/metabolism , Glomerulosclerosis, Focal Segmental/pathology , Podocytes , Animals , Fibrosis/genetics , Fibrosis/metabolism , Fibrosis/pathology , Glomerulosclerosis, Focal Segmental/genetics , Humans , Mice , Mice, Transgenic , Microscopy, Fluorescence, Multiphoton/methods , Podocytes/metabolism , Podocytes/pathology , Receptors, Purinergic P2Y2/genetics , Receptors, Purinergic P2Y2/metabolism
16.
J Clin Invest ; 123(5): 2011-23, 2013 May.
Article in English | MEDLINE | ID: mdl-23619363

ABSTRACT

Activation of the intrarenal renin-angiotensin system (RAS) can elicit hypertension independently from the systemic RAS. However, the precise mechanisms by which intrarenal Ang II increases blood pressure have never been identified. To this end, we studied the responses of mice specifically lacking kidney angiotensin-converting enzyme (ACE) to experimental hypertension. Here, we show that the absence of kidney ACE substantially blunts the hypertension induced by Ang II infusion (a model of high serum Ang II) or by nitric oxide synthesis inhibition (a model of low serum Ang II). Moreover, the renal responses to high serum Ang II observed in wild-type mice, including intrarenal Ang II accumulation, sodium and water retention, and activation of ion transporters in the loop of Henle (NKCC2) and distal nephron (NCC, ENaC, and pendrin) as well as the transporter activating kinases SPAK and OSR1, were effectively prevented in mice that lack kidney ACE. These findings demonstrate that ACE metabolism plays a fundamental role in the responses of the kidney to hypertensive stimuli. In particular, renal ACE activity is required to increase local Ang II, to stimulate sodium transport in loop of Henle and the distal nephron, and to induce hypertension.


Subject(s)
Hypertension/metabolism , Kidney/metabolism , Peptidyl-Dipeptidase A/metabolism , Angiotensin II/metabolism , Animals , Kidney/embryology , Liver/metabolism , Loop of Henle/metabolism , Male , Mice , NG-Nitroarginine Methyl Ester/pharmacology , Protein Serine-Threonine Kinases/metabolism , Receptors, Drug/metabolism , Renin-Angiotensin System , Sodium/metabolism , Sodium-Potassium-Chloride Symporters/metabolism , Solute Carrier Family 12, Member 1 , Solute Carrier Family 12, Member 3 , Symporters/metabolism , Water/metabolism
17.
Am J Physiol Cell Physiol ; 304(2): C147-63, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23114965

ABSTRACT

The renal distal tubule Na-Cl cotransporter (NCC) reabsorbs <10% of the filtered Na(+) but is a key control point for blood pressure regulation by angiotensin II (ANG II), angiotensin-converting enzyme inhibitors (ACEI), and thiazide diuretics. This study aimed to determine whether NCC phosphorylation (NCCp) was regulated by acute (20-30 min) treatment with the ACEI captopril (12 µg/min × 20 min) or by a sub-pressor dose of ANG II (20 ng·kg(-1)·min(-1)) in Inactin-anesthetized rats. By immuno-EM, NCCp was detected exclusively in or adjacent to apical plama membranes (APM) in controls and after ACEI or ANG II treatment, while NCC total was detected in both APM and subapical cytoplasmic vesicles (SCV) in all conditions. In renal homogenates, neither ACEI nor ANG II treatment altered NCCp abundance, assayed by immunoblot. However, by density gradient fractionation we identified a pool of low-density APM in which NCCp decreased 50% in response to captopril and was restored during ANG II infusion, and another pool of higher-density APM that responded reciprocally, indicative of regulated redistribution between two APM pools. In both pools, NCCp was preferentially localized to Triton-soluble membranes. Blue Native gel electrophoresis established that APM NCCp localized to ~700 kDa complexes (containing γ-adducin) while unphosphorylated NCC in intracellular membranes primarily localized to ~400 kDa complexes: there was no evidence for native monomeric or dimeric NCC or NCCp. In summary, this study demonstrates that phosphorylated NCC, localized to multimeric complexes in the APM, redistributes in a regulated manner within the APM in response to ACEI and ANG II.


Subject(s)
Angiotensin II/pharmacology , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Kidney Tubules, Distal/metabolism , Sodium Chloride Symporters/metabolism , Animals , Calmodulin-Binding Proteins/analysis , Captopril/pharmacology , Kidney Tubules, Distal/cytology , Kidney Tubules, Distal/drug effects , Male , Phosphorylation , Rats , Rats, Sprague-Dawley , Sodium Chloride Symporter Inhibitors/pharmacology
18.
Pflugers Arch ; 465(1): 53-8, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22729752

ABSTRACT

One emerging topic in renin-angiotensin system (RAS) research is the direct local control of renin synthesis and release by endogenous metabolic intermediates. During the past few years, our laboratory has characterized the localization and signaling of the novel metabolic receptor GPR91 in the normal and diabetic kidney and established GPR91 as a new, direct link between high glucose and RAS activation in diabetes. GPR91 (also called SUCNR1) binds tricarboxylic acid (TCA) cycle intermediate succinate which can rapidly accumulate in the local tissue environment when energy supply and demand are out of balance. In a variety of physiological and pathological conditions associated with metabolic stress, succinate signaling via GPR91 appears to be an important mediator or modulator of renin secretion. This review summarizes our current knowledge on the control of renin release by molecules of endogenous metabolic pathways with the main focus on succinate/GPR91.


Subject(s)
Renin/metabolism , Stress, Physiological , Animals , Diabetes Mellitus, Type 2/metabolism , Humans , Receptors, G-Protein-Coupled/metabolism , Renin-Angiotensin System , Succinic Acid/metabolism
19.
Cell Metab ; 13(4): 469-475, 2011 Apr 06.
Article in English | MEDLINE | ID: mdl-21459331

ABSTRACT

Hypertension affects more than 1.5 billion people worldwide but the precise cause of elevated blood pressure (BP) cannot be determined in most affected individuals. Nonetheless, blockade of the renin-angiotensin system (RAS) lowers BP in the majority of patients with hypertension. Despite its apparent role in hypertension pathogenesis, the key cellular targets of the RAS that control BP have not been clearly identified. Here we demonstrate that RAS actions in the epithelium of the proximal tubule have a critical and nonredundant role in determining the level of BP. Abrogation of AT(1) angiotensin receptor signaling in the proximal tubule alone is sufficient to lower BP, despite intact vascular responses. Elimination of this pathway reduces proximal fluid reabsorption and alters expression of key sodium transporters, modifying pressure-natriuresis and providing substantial protection against hypertension. Thus, effectively targeting epithelial functions of the proximal tubule of the kidney should be a useful therapeutic strategy in hypertension.


Subject(s)
Blood Pressure/physiology , Kidney Tubules, Proximal/metabolism , Receptor, Angiotensin, Type 1/metabolism , Animals , Hypertension/pathology , Mice , Receptor, Angiotensin, Type 1/genetics , Renin-Angiotensin System/drug effects , Signal Transduction , Sodium/metabolism , Sodium-Hydrogen Exchanger 3 , Sodium-Hydrogen Exchangers/metabolism , Sodium-Potassium-Chloride Symporters/metabolism , Solute Carrier Family 12, Member 1
20.
Am J Physiol Renal Physiol ; 298(1): F177-86, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19864301

ABSTRACT

Angiotensin II (ANG II) stimulates proximal tubule (PT) sodium and water reabsorption. We showed that treating rats acutely with the angiotensin-converting enzyme inhibitor captopril decreases PT salt and water reabsorption and provokes rapid redistribution of the Na(+)/H(+) exchanger isoform 3 (NHE3), Na(+)/Pi cotransporter 2 (NaPi2), and associated proteins out of the microvilli. The aim of the present study was to determine whether acute ANG II infusion increases the abundance of PT NHE3, NaPi2, and associated proteins in the microvilli available for reabsorbing NaCl. Male Sprague-Dawley rats were infused with a dose of captopril (12 microg/min for 20 min) that increased PT flow rate approximately 20% with no change in blood pressure (BP) or glomerular filtration rate (GFR). When ANG II (20 ng x kg(-1) x min(-1) for 20 min) was added to the captopril infusate, PT volume flow rate returned to baseline without changing BP or GFR. After captopril, NHE3 was localized to the base of the microvilli and NaPi2 to subapical cytoplasmic vesicles; after 20 min ANG II, both NHE3 and NaPi2 redistributed into the microvilli, assayed by confocal microscopy and density gradient fractionation. Additional PT proteins that redistributed into low-density microvilli-enriched membranes in response to ANG II included myosin VI, DPPIV, NHERF-1, ezrin, megalin, vacuolar H(+)-ATPase, aminopeptidase N, and clathrin. In summary, in response to 20 min ANG II in the absence of a change in BP or GFR, multiple proteins traffic into the PT brush-border microvilli where they likely contribute to the rapid increase in PT salt and water reabsorption.


Subject(s)
Angiotensin II/physiology , Kidney Tubules, Proximal/metabolism , Sodium-Hydrogen Exchangers/metabolism , Sodium-Phosphate Cotransporter Proteins, Type II/metabolism , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , Blood Pressure/drug effects , Captopril/pharmacology , Clathrin/metabolism , Cytoskeletal Proteins/metabolism , Glomerular Filtration Rate/drug effects , Low Density Lipoprotein Receptor-Related Protein-2/metabolism , Male , Microvilli/drug effects , Microvilli/metabolism , Models, Animal , Myosin Heavy Chains/metabolism , Rats , Rats, Sprague-Dawley , Sodium Chloride/metabolism , Sodium-Hydrogen Exchanger 3
SELECTION OF CITATIONS
SEARCH DETAIL
...