1
|
Yalcın B, Onder GO, Goktepe O, Suna PA, Mat OC, Koseoglu E, Cetindag E, Baran M, Bitgen N, Öz Gergı N Ö, Yay A. Enhanced kidney damage induced by increasing nonylphenol doses: impact on autophagy-related proteins and proinflammatory cytokines in rats. Toxicol Mech Methods 2024; 34:867-876. [PMID: 38769906 DOI: 10.1080/15376516.2024.2358348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 05/05/2024] [Accepted: 05/05/2024] [Indexed: 05/22/2024]
Abstract
Nonylphenol (NP) is an organic pollutant and endocrine disruptor chemical that has harmful effects on the environment and living organisms. This study looked at whether kidney tissues subjected to increasing doses of nonylphenol generated alterations in histopathologic, pro-inflammatory, and autophagic markers. Fifty rats were divided into five groups of ten each: group I: healthy group, II: control (corn oil), group III: 25 μl/kg NP, group IV: 50 μl/kg NP, group V: 75 μl/kg NP. The kidney tissue samples were obtained for histopathological, immunohistochemical, and biochemical analyses. The histological deteriorations observed in all NP groups included tubular epithelial cell degeneration, inflammation areas, and hemorrhage. The immunohistochemical investigations showed that NP significantly elevated the autophagy markers (Beclin-1, LC3A/B, p62), pro-inflammatory cytokines (TNF-α, IL-6), HIF-1α, and eNOS in group III, IV and V compared with group I and II. The biochemical analysis also revealed that pro-inflammatory cytokines (TNF-α, IL-1β, and IL-6) increased in correlation with the NP doses, but only IL-1β reached statistical significance in NP treated rats kidney tissue. The biochemical findings have been confirmed by the histological studies. The damage to renal tissue caused by NP exposure may worsen it by increasing inflammatory and autophagic markers.
Collapse
Affiliation(s)
- Betul Yalcın
- Department of Histology and Embryology, Adıyaman University, Adıyaman, Turkey
| | - Gozde Ozge Onder
- Department of Histology and Embryology, Erciyes University, Kayseri, Turkey
- Genome and Stem Cell Center (GENKOK), Erciyes University, Kayseri, Turkey
| | - Ozge Goktepe
- Department of Histology and Embryology, Erciyes University, Kayseri, Turkey
- Genome and Stem Cell Center (GENKOK), Erciyes University, Kayseri, Turkey
| | - Pınar Alisan Suna
- Department of Histology and Embryology, Erciyes University, Kayseri, Turkey
| | - Ozge Cengiz Mat
- Department of Histology and Embryology, Erciyes University, Kayseri, Turkey
| | - Eda Koseoglu
- Department of Histology and Embryology, Erciyes University, Kayseri, Turkey
| | - Emre Cetindag
- Department of Histology and Embryology, Erciyes University, Kayseri, Turkey
| | - Munevver Baran
- Department of Pharmacy Basic Science, Erciyes University, Kayseri, Turkey
| | - Nazmiye Bitgen
- Genome and Stem Cell Center (GENKOK), Erciyes University, Kayseri, Turkey
- Department of Medical Biology, Erciyes University, Kayseri, Turkey
| | - Özlem Öz Gergı N
- Department of Surgical Medicine Science, Anesthesiology and Reanimation, Erciyes University, Kayseri, Turkey
| | - Arzu Yay
- Department of Histology and Embryology, Erciyes University, Kayseri, Turkey
- Genome and Stem Cell Center (GENKOK), Erciyes University, Kayseri, Turkey
| |
Collapse
|
2
|
Park H, Shin JA, Lim J, Lee S, Ahn JH, Kang JL, Choi YH. Increased Caveolin-2 Expression in Brain Endothelial Cells Promotes Age-Related Neuroinflammation. Mol Cells 2022; 45:950-962. [PMID: 36572563 PMCID: PMC9794556 DOI: 10.14348/molcells.2022.0045] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 08/30/2022] [Accepted: 10/12/2022] [Indexed: 12/28/2022] Open
Abstract
Aging is a major risk factor for common neurodegenerative diseases. Although multiple molecular, cellular, structural, and functional changes occur in the brain during aging, the involvement of caveolin-2 (Cav-2) in brain ageing remains unknown. We investigated Cav-2 expression in brains of aged mice and its effects on endothelial cells. The human umbilical vein endothelial cells (HUVECs) showed decreased THP-1 adhesion and infiltration when treated with Cav-2 siRNA compared to control siRNA. In contrast, Cav-2 overexpression increased THP-1 adhesion and infiltration in HUVECs. Increased expression of Cav-2 and iba-1 was observed in brains of old mice. Moreover, there were fewer iba-1-positive cells in the brains of aged Cav-2 knockout (KO) mice than of wild-type aged mice. The levels of several chemokines were higher in brains of aged wild-type mice than in young wild-type mice; moreover, chemokine levels were significantly lower in brains of young mice as well as aged Cav-2 KO mice than in their wild-type counterparts. Expression of PECAM1 and VE-cadherin proteins increased in brains of old wild-type mice but was barely detected in brains of young wild-type and Cav-2 KO mice. Collectively, our results suggest that Cav-2 expression increases in the endothelial cells of aged brain, and promotes leukocyte infiltration and age-associated neuroinflammation.
Collapse
Affiliation(s)
- Hyunju Park
- Department of Physiology, Inflammation-Cancer Microenvironment Research Center, Seoul 07804, Korea
| | - Jung A Shin
- Department of Anatomy, Ewha Womans University College of Medicine, Seoul 07804, Korea
| | - Jiwoo Lim
- Department of Physiology, Inflammation-Cancer Microenvironment Research Center, Seoul 07804, Korea
| | - Seulgi Lee
- Department of Physiology, Inflammation-Cancer Microenvironment Research Center, Seoul 07804, Korea
| | - Jung-Hyuck Ahn
- Department of Biochemistry, Ewha Womans University College of Medicine, Seoul 07804, Korea
| | - Jihee Lee Kang
- Department of Physiology, Inflammation-Cancer Microenvironment Research Center, Seoul 07804, Korea
| | - Youn-Hee Choi
- Department of Physiology, Inflammation-Cancer Microenvironment Research Center, Seoul 07804, Korea
| |
Collapse
|
3
|
Hypertension and renal disease programming: focus on the early postnatal period. Clin Sci (Lond) 2022; 136:1303-1339. [PMID: 36073779 DOI: 10.1042/cs20220293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 08/18/2022] [Accepted: 08/25/2022] [Indexed: 11/17/2022]
Abstract
The developmental origin of hypertension and renal disease is a concept highly supported by strong evidence coming from both human and animal studies. During development there are periods in which the organs are more vulnerable to stressors. Such periods of susceptibility are also called 'sensitive windows of exposure'. It was shown that as earlier an adverse event occurs; the greater are the consequences for health impairment. However, evidence show that the postnatal period is also quite important for hypertension and renal disease programming, especially in rodents because they complete nephrogenesis postnatally, and it is also important during preterm human birth. Considering that the developing kidney is vulnerable to early-life stressors, renal programming is a key element in the developmental programming of hypertension and renal disease. The purpose of this review is to highlight the great number of studies, most of them performed in animal models, showing the broad range of stressors involved in hypertension and renal disease programming, with a particular focus on the stressors that occur during the early postnatal period. These stressors mainly include undernutrition or specific nutritional deficits, chronic behavioral stress, exposure to environmental chemicals, and pharmacological treatments that affect some important factors involved in renal physiology. We also discuss the common molecular mechanisms that are activated by the mentioned stressors and that promote the appearance of these adult diseases, with a brief description on some reprogramming strategies, which is a relatively new and promising field to treat or to prevent these diseases.
Collapse
|
4
|
Agarwal S, Sudhini YR, Polat OK, Reiser J, Altintas MM. Renal cell markers: lighthouses for managing renal diseases. Am J Physiol Renal Physiol 2021; 321:F715-F739. [PMID: 34632812 DOI: 10.1152/ajprenal.00182.2021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Kidneys, one of the vital organs in our body, are responsible for maintaining whole body homeostasis. The complexity of renal function (e.g., filtration, reabsorption, fluid and electrolyte regulation, and urine production) demands diversity not only at the level of cell types but also in their overall distribution and structural framework within the kidney. To gain an in depth molecular-level understanding of the renal system, it is imperative to discern the components of kidney and the types of cells residing in each of the subregions. Recent developments in labeling, tracing, and imaging techniques have enabled us to mark, monitor, and identify these cells in vivo with high efficiency in a minimally invasive manner. In this review, we summarize different cell types, specific markers that are uniquely associated with those cell types, and their distribution in the kidney, which altogether make kidneys so special and different. Cellular sorting based on the presence of certain proteins on the cell surface allowed for the assignment of multiple markers for each cell type. However, different studies using different techniques have found contradictions in cell type-specific markers. Thus, the term "cell marker" might be imprecise and suboptimal, leading to uncertainty when interpreting the data. Therefore, we strongly believe that there is an unmet need to define the best cell markers for a cell type. Although the compendium of renal-selective marker proteins presented in this review is a resource that may be useful to researchers, we acknowledge that the list may not be necessarily exhaustive.
Collapse
Affiliation(s)
- Shivangi Agarwal
- Department of Internal Medicine, Rush University, Chicago, Illinois
| | | | - Onur K Polat
- Department of Internal Medicine, Rush University, Chicago, Illinois
| | - Jochen Reiser
- Department of Internal Medicine, Rush University, Chicago, Illinois
| | | |
Collapse
|
5
|
Pou Casellas C, Jansen K, Rookmaaker MB, Clevers H, Verhaar MC, Masereeuw R. Regulation of Solute Carriers OCT2 and OAT1/3 in the Kidney: A Phylogenetic, Ontogenetic and Cell Dynamic Perspective. Physiol Rev 2021; 102:993-1024. [PMID: 34486394 DOI: 10.1152/physrev.00009.2021] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Over the course of more than 500 million years, the kidneys have undergone a remarkable evolution from primitive nephric tubes to intricate filtration-reabsorption systems that maintain homeostasis and remove metabolic end products from the body. The evolutionarily conserved solute carriers Organic Cation Transporter 2 (OCT2), and Organic Anion Transporters 1 and 3 (OAT1/3) coordinate the active secretion of a broad range of endogenous and exogenous substances, many of which accumulate in the blood of patients with kidney failure despite dialysis. Harnessing OCT2 and OAT1/3 through functional preservation or regeneration could alleviate the progression of kidney disease. Additionally, it would improve current in vitro test models that lose their expression in culture. With this review, we explore OCT2 and OAT1/3 regulation using different perspectives: phylogenetic, ontogenetic and cell dynamic. Our aim is to identify possible molecular targets to both help prevent or compensate for the loss of transport activity in patients with kidney disease, and to enable endogenous OCT2 and OAT1/3 induction in vitro in order to develop better models for drug development.
Collapse
Affiliation(s)
- Carla Pou Casellas
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, Netherlands.,Hubrecht Institute - Royal Netherlands Academy of Arts and Sciences, Utrecht, The Netherlands
| | - Katja Jansen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Maarten B Rookmaaker
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, Netherlands
| | - Hans Clevers
- Hubrecht Institute - Royal Netherlands Academy of Arts and Sciences, Utrecht, The Netherlands
| | - Marianne C Verhaar
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, Netherlands
| | - Rosalinde Masereeuw
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
6
|
Mercanoglu G, Semen O. Nitric oxide mediated the effects of nebivolol in cardiorenal syndrome. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2019; 22:1314-1324. [PMID: 32128097 PMCID: PMC7038421 DOI: 10.22038/ijbms.2019.37400.8927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 05/14/2019] [Indexed: 11/14/2022]
Abstract
OBJECTIVES Despite several proposed mechanisms for the pathophysiology of cardiorenal syndrome (CRS), the exact mechanism remains unclear. Nitrosative stress has been argued as a key mechanism recently. Nebivolol is a beta-blocker with nitric oxide (NO)-releasing effect. In the present study, NO-mediated effects of two different treatment regimes of nebivolol in CRS were studied. MATERIALS AND METHODS Rats were divided into: sham-operated (sham-control), myocardial infarction (MI)-induced, (MI-control) early nebivolol-treated (MI-neb1) and late nebivolol-treated (Mı-neb2) groups. The effects of nebivolol were assessed both in the early and late period of MI by histologic, hemodynamic and biologic studies. RESULTS Developed MI model was in line with the heart failure with preserved ejection fraction. Focal and total tubular damage findings were observed in MI-control group both in early and late period of MI. In parallel, subclinical functional damage was transformed into chronic renal dysfunction in this group. Increased inducible nitric oxide synthase (iNOS) and endothelial NOS (eNOS) together with decreased neuronal NOS (nNOS) levels were in parallel with the increased inflammation and nitrosative stress biomarkers. Nebivolol effectively prevented both subclinical and clinical nephropathy. There was no statistical difference between the nebivolol treatment regimes. CONCLUSION The beneficial effects of nebivolol were closely related to the reduction of nitrosative damages as well as hemodynamic alterations. The NO-mediated effects were: prevention of nitrosative damage by decreasing iNOS, preservation of nNOS in order to maintain glomerular filtration rate (GFR), and restoration of eNOS in the late period of MI. On contrary to our previous work, early nebivolol administration had a similar effect with delayed administration of nebivolol on CRS.
Collapse
Affiliation(s)
- Guldem Mercanoglu
- University of Health Sciences, Faculty of Pharmacy, Department of Pharmacology, Istanbul, Turkey
| | - Onder Semen
- Istanbul University, Istanbul Medical Faculty, Department of Pathology, Istanbul, Turkey
| |
Collapse
|
7
|
Sheldon RD, Meers GM, Morris EM, Linden MA, Cunningham RP, Ibdah JA, Thyfault JP, Laughlin MH, Rector RS. eNOS deletion impairs mitochondrial quality control and exacerbates Western diet-induced NASH. Am J Physiol Endocrinol Metab 2019; 317:E605-E616. [PMID: 31361543 PMCID: PMC6842915 DOI: 10.1152/ajpendo.00096.2019] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Dysregulated mitochondrial quality control leads to mitochondrial functional impairments that are central to the development and progression of hepatic steatosis to nonalcoholic steatohepatitis (NASH). Here, we identify hepatocellular localized endothelial nitric oxide synthase (eNOS) as a novel master regulator of mitochondrial quality control. Mice lacking eNOS were more susceptible to Western diet-induced hepatic inflammation and fibrosis in conjunction with decreased markers of mitochondrial biogenesis and turnover. The hepatocyte-specific influence was verified via magnetic activated cell sorting purified primary hepatocytes and in vitro siRNA-induced knockdown of eNOS. Hepatic mitochondria from eNOS knockout mice revealed decreased markers of mitochondrial biogenesis (PPARγ coactivator-1α, mitochondrial transcription factor A) and autophagy/mitophagy [BCL-2-interacting protein-3 (BNIP3), 1A/1B light chain 3B (LC3)], suggesting decreased mitochondrial turnover rate. eNOS knockout in primary hepatocytes exhibited reduced fatty acid oxidation capacity and were unable to mount a normal BNIP3 response to a mitophagic challenge compared with wild-type mice. Finally, we demonstrate that eNOS is required in primary hepatocytes to induce activation of the stress-responsive transcription factor nuclear factor erythroid 2-related factor 2 (NRF2). Thus, our data demonstrate that eNOS is an important regulator of hepatic mitochondrial content and function and NASH susceptibility.
Collapse
Affiliation(s)
- Ryan D Sheldon
- Research Service, Harry S Truman Memorial Veterans Medical Center, Columbia, Missouri
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
| | - Grace M Meers
- Research Service, Harry S Truman Memorial Veterans Medical Center, Columbia, Missouri
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
| | - E Matthew Morris
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas
| | - Melissa A Linden
- Research Service, Harry S Truman Memorial Veterans Medical Center, Columbia, Missouri
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
| | - Rory P Cunningham
- Research Service, Harry S Truman Memorial Veterans Medical Center, Columbia, Missouri
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
| | - Jamal A Ibdah
- Research Service, Harry S Truman Memorial Veterans Medical Center, Columbia, Missouri
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Missouri, Columbia, Missouri
| | - John P Thyfault
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas
- Kansas City Veterans Affairs Medical Center, Kansas City, Missouri
| | - M Harold Laughlin
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri
| | - R Scott Rector
- Research Service, Harry S Truman Memorial Veterans Medical Center, Columbia, Missouri
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Missouri, Columbia, Missouri
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
| |
Collapse
|
8
|
Xue H, Geurts AM, Usa K, Wang F, Lin Y, Phillips J, Henderson L, Baker MA, Tian Z, Liang M. Fumarase Overexpression Abolishes Hypertension Attributable to endothelial NO synthase Haploinsufficiency in Dahl Salt-Sensitive Rats. Hypertension 2019; 74:313-322. [PMID: 31230549 DOI: 10.1161/hypertensionaha.119.12723] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Human blood pressure salt sensitivity is associated with changes in urinary metabolites related to fumarase (Fh) and nitric oxide (NO) metabolism, and fumarase promotes NO production through an arginine regeneration pathway. We examined the role of the fumarase-NO pathway in the development of hypertension using genetically engineered rat models. Dahl salt-sensitive (SS) rats with heterozygous mutation of eNOS (endothelial NO synthase or Nos3; SS-Nos3+/-) were bred with SS rats with a hemizygous Fh transgene. SS-Nos3+/- rats without the Fh transgene (SS-Nos3+/-/Fh0/0) developed substantial hypertension with a mean arterial pressure of 134.2±3.7 mm Hg on a 0.4% NaCl diet and 178.0±3.5 mm Hg after 14 days on a 4% NaCl diet. Mean arterial pressure decreased remarkably to 123.1±1.4 mm Hg on 0.4% NaCl, and 143.3±1.5 mm Hg on 4% NaCl in SS-Nos3+/- rats with a Fh transgene (SS-Nos3+/-/Fh0/1), and proteinuria, renal fibrosis, and tubular casts were attenuated in SS-Nos3+/-/Fh0/1 rats compared with SS-Nos3+/-/Fh0/0 rats. eNOS protein abundance decreased in rats with the Nos3 heterozygous mutation, which was not influenced by Fh overexpression in rats on the 0.4% NaCl diet. However, the decrease in NO metabolite in the renal outer medulla of SS-Nos3+/-/Fh0/0 rats on the 0.4% NaCl diet was reversed in SS-Nos3+/-/Fh0/1 rats, and levels of L-arginine, but not the other 12 amino acids analyzed, were significantly higher in SS-Nos3+/-/Fh0/1 rats than in SS-Nos3+/+/Fh0/0 rats. In conclusion, fumarase has potent effects in restoring NO production and blunting the development of hypertension attributable to eNOS haploinsufficiency.
Collapse
Affiliation(s)
- Hong Xue
- From the Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China (H.X.).,Center of Systems Molecular Medicine, Department of Physiology (H.X., A.M.G., K.U., F.W., Y.L., J.P., L.H., M.A.B., M.L.)
| | - Aron M Geurts
- Center of Systems Molecular Medicine, Department of Physiology (H.X., A.M.G., K.U., F.W., Y.L., J.P., L.H., M.A.B., M.L.).,Genomic Sciences and Precision Medicine Center Medical College of Wisconsin, Milwaukee (A.M.G.)
| | - Kristie Usa
- Center of Systems Molecular Medicine, Department of Physiology (H.X., A.M.G., K.U., F.W., Y.L., J.P., L.H., M.A.B., M.L.)
| | - Feng Wang
- Center of Systems Molecular Medicine, Department of Physiology (H.X., A.M.G., K.U., F.W., Y.L., J.P., L.H., M.A.B., M.L.).,Department of Nephrology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, China (F.W., Y.L.)
| | - Yingying Lin
- Center of Systems Molecular Medicine, Department of Physiology (H.X., A.M.G., K.U., F.W., Y.L., J.P., L.H., M.A.B., M.L.).,Department of Nephrology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, China (F.W., Y.L.)
| | - Jenifer Phillips
- Center of Systems Molecular Medicine, Department of Physiology (H.X., A.M.G., K.U., F.W., Y.L., J.P., L.H., M.A.B., M.L.)
| | - Lisa Henderson
- Center of Systems Molecular Medicine, Department of Physiology (H.X., A.M.G., K.U., F.W., Y.L., J.P., L.H., M.A.B., M.L.)
| | - Maria Angeles Baker
- Center of Systems Molecular Medicine, Department of Physiology (H.X., A.M.G., K.U., F.W., Y.L., J.P., L.H., M.A.B., M.L.)
| | - Zhongmin Tian
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, China (Z.T.)
| | - Mingyu Liang
- Center of Systems Molecular Medicine, Department of Physiology (H.X., A.M.G., K.U., F.W., Y.L., J.P., L.H., M.A.B., M.L.)
| |
Collapse
|
9
|
Chisada S, Sugiyama A. Renal lesions in leptin receptor-deficient medaka ( Oryzias latipes). J Toxicol Pathol 2019; 32:297-303. [PMID: 31719758 PMCID: PMC6831499 DOI: 10.1293/tox.2019-0021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 05/11/2019] [Indexed: 12/13/2022] Open
Abstract
The aim of this study was to elucidate the renal lesions of leptin receptor-deficient
medaka showing hyperglycemia and hypoinsulinemia and to evaluate the usefulness of the
medaka as a model of diabetic nephropathy. Leptin receptor-deficient medaka at 20 and 30
weeks of age showed hyperglycemia and hypoinsulinemia; they also showed a higher level of
plasma creatinine than the control medaka. Histopathologically, dilation of glomerular
capillary lumina and of afferent/efferent arterioles was observed in leptin
receptor-deficient medaka at 20 weeks of age, and then glomerular enlargement with cell
proliferation and matrix expansion, formation of fibrin cap-like lesions, glomerular
atrophy with Bowman’s capsule dilation, and renal tubule dilation were observed at 30
weeks of age. These histopathological characteristics of leptin receptor-deficient medaka
were similar to the characteristics of kidney lesions of human and rodent models of type
II diabetes mellitus, making leptin receptor-deficient medaka a useful model of diabetic
nephropathy.
Collapse
Affiliation(s)
- Shinichi Chisada
- Department of Hygiene and Public Health, Kyorin University School of Medicine, 6-20-2 Shinkawa, Mitaka-shi, Tokyo 181-8611, Japan
| | - Akihiko Sugiyama
- Faculty of Veterinary Medicine, Okayama University of Science, 1-3 Ikoinooka, Imabari-shi, Ehime 794-8555, Japan
| |
Collapse
|
10
|
Natarajan M, Habib SL, Reddick RL, Delma CR, Manickam K, Prihoda TJ, Werner SL, Mohan S. Endothelial cell-specific overexpression of endothelial nitric oxide synthase in Ins2Akita mice exacerbates diabetic nephropathy. J Diabetes Complications 2019; 33:23-32. [PMID: 30424931 PMCID: PMC6344355 DOI: 10.1016/j.jdiacomp.2018.10.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 08/13/2018] [Accepted: 10/07/2018] [Indexed: 10/28/2022]
Abstract
Previous studies demonstrated that global deficiency of eNOS in diabetic mice exacerbated renal lesions and that overexpression of eNOS may protect against tissue injury. Our study revealed for the first time overexpression of eNOS leads to disease progression rather than protection. Transgenic mice selectively expressing eNOS in endothelial cells (eNOSTg) were cross bred with Ins2Akita type-1 (AK) diabetic mice to generate eNOS overexpressing eNOSTg/AK mice. Wild type, eNOSTg, AK and eNOSTg/AK mice were assessed for kidney function and blood glucose levels. Remarkably, overexpressing eNOSTg mice showed evidence of unpredicted glomerular injury with segmental mesangiolysis and occasional microaneurysms. Notably, in eNOSTg/AK mice overexpression of eNOS led to increased glomerular/endothelial injury that was associated with increased superoxide levels and renal dysfunction. Results indicate for the first time that overexpressing eNOS in endothelial cells cannot ameliorate diabetic lesions, but paradoxically leads to progression of nephropathy likely due to eNOS uncoupling and superoxide upsurge. This novel finding has a significant impact on current therapeutic strategies to improve endothelial function and prevent progression of diabetic renal disease. Further, the eNOSTg/AK model developed in this study has significant translational potentials for elucidating the underlying mechanism implicated in the deflected function of eNOS in diabetic nephropathy.
Collapse
Affiliation(s)
- Mohan Natarajan
- Department of Pathology and Laboratory Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Samy L Habib
- Geriatric Research Education and Clinical Center, South Texas Veterans Healthcare System and Cell Systems & Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Robert L Reddick
- Department of Pathology and Laboratory Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Caroline R Delma
- Department of Pathology and Laboratory Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Krishnan Manickam
- Department of Pathology and Laboratory Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Thomas J Prihoda
- Department of Pathology and Laboratory Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Sherry L Werner
- Department of Pathology and Laboratory Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Sumathy Mohan
- Department of Pathology and Laboratory Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.
| |
Collapse
|
11
|
Key features of the nephrogenic zone in the fetal human kidney—hardly known but relevant for the detection of first traces impairing nephrogenesis. Cell Tissue Res 2018; 375:589-603. [DOI: 10.1007/s00441-018-2937-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 09/21/2018] [Indexed: 01/09/2023]
|
12
|
Minuth WW. Concepts for a therapeutic prolongation of nephrogenesis in preterm and low-birth-weight babies must correspond to structural-functional properties in the nephrogenic zone. Mol Cell Pediatr 2017; 4:12. [PMID: 29218481 PMCID: PMC5721096 DOI: 10.1186/s40348-017-0078-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 11/20/2017] [Indexed: 12/30/2022] Open
Abstract
Numerous investigations are dealing with anlage of the mammalian kidney and primary development of nephrons. However, only few information is available about the last steps in kidney development leading at birth to a downregulation of morphogen activity in the nephrogenic zone and to a loss of stem cell niches aligned beyond the organ capsule. Surprisingly, these natural changes in the developmental program display similarities to processes occurring in the kidneys of preterm and low-birth-weight babies. Although those babies are born at a time with a principally intact nephrogenic zone and active niches, a high proportion of them suffers on impairment of nephrogenesis resulting in oligonephropathy, formation of atypical glomeruli, and immaturity of parenchyma. The setting points out that up to date not identified noxae in the nephrogenic zone hamper primary steps of parenchyma development. In this situation, a possible therapeutic aim is to prolong nephrogenesis by medications. However, actual data provide information that administration of drugs is problematic due to an unexpectedly complex microanatomy of the nephrogenic zone, in niches so far not considered textured extracellular matrix and peculiar contacts between mesenchymal cell projections and epithelial stem cells via tunneling nanotubes. Thus, it remains to be figured out whether disturbance of morphogen signaling altered synthesis of extracellular matrix, disturbed cell-to-cell contacts, or modified interstitial fluid impair nephrogenic activity. Due to most unanswered questions, search for eligible drugs prolonging nephrogenesis and their reliable administration is a special challenge for the future.
Collapse
Affiliation(s)
- Will W Minuth
- Institute of Anatomy, University of Regensburg, 93053, Regensburg, Germany.
| |
Collapse
|
13
|
Effects of Nitric Oxide on Renal Proximal Tubular Na + Transport. BIOMED RESEARCH INTERNATIONAL 2017; 2017:6871081. [PMID: 29181400 PMCID: PMC5664255 DOI: 10.1155/2017/6871081] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 09/14/2017] [Indexed: 12/28/2022]
Abstract
Nitric oxide (NO) has a wide variety of physiological functions in the kidney. Besides the regulatory effects in intrarenal haemodynamics and glomerular microcirculation, in vivo studies reported the diuretic and natriuretic effects of NO. However, opposite results showing the stimulatory effect of NO on Na+ reabsorption in the proximal tubule led to an intense debate on its physiological roles. Animal studies have showed the biphasic effect of angiotensin II (Ang II) and the overall inhibitory effect of NO on the activity of proximal tubular Na+ transporters, the apical Na+/H+ exchanger isoform 3, basolateral Na+/K+ ATPase, and the Na+/HCO3− cotransporter. However, whether these effects could be reproduced in humans remained unclear. Notably, our recent functional analysis of isolated proximal tubules demonstrated that Ang II dose-dependently stimulated human proximal tubular Na+ transport through the NO/guanosine 3′,5′-cyclic monophosphate (cGMP) pathway, confirming the human-specific regulation of proximal tubular transport via NO and Ang II. Of particular importance for this newly identified pathway is its possibility of being a human-specific therapeutic target for hypertension. In this review, we focus on NO-mediated regulation of proximal tubular Na+ transport, with emphasis on the interaction with individual Na+ transporters and the crosstalk with Ang II signalling.
Collapse
|
14
|
Gao Y, Stuart D, Pollock JS, Takahishi T, Kohan DE. Collecting duct-specific knockout of nitric oxide synthase 3 impairs water excretion in a sex-dependent manner. Am J Physiol Renal Physiol 2016; 311:F1074-F1083. [PMID: 27707708 DOI: 10.1152/ajprenal.00494.2016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 09/29/2016] [Indexed: 12/21/2022] Open
Abstract
Nitric oxide (NO) inhibits collecting duct (CD) Na+ and water reabsorption. Mice with CD-specific knockout (KO) of NO synthase 1 (NOS1) have salt-sensitive hypertension. In contrast, the role of NOS3 in CD salt and water reabsorption is unknown. Mice with CD NOS3 KO were generated with loxP-flanked exons 9-12 (encodes the calmodulin binding site) of the NOS3 gene and the aquaporin-2 promoter-Cre transgene. There were no differences between control and CD NOS3 KO mice, irrespective of sex, in food intake, water intake, urine volume, urinary Na+ or K+ excretion, plasma renin concentration, blood pressure, or pulse during 7 days of normal (0.3%), high (3.17%), or low (0.03%) Na+ intake. Blood pressure was similar between genotypes during DOCA-high salt. CD NOS3 KO did not alter urine volume or urine osmolality after water deprivation. In contrast, CD NOS3 KO male, but not female, mice had lower urine volume and higher urine osmolality over the course of 7 days of water loading compared with control mice. Male, but not female, CD NOS3 KO mice had reduced urinary nitrite+nitrate excretion compared with controls after 7 days of water loading. Urine AVP and AVP-stimulated cAMP accumulation in isolated inner medullary CD were similar between genotypes. Western analysis did not reveal a significant effect of CD NOS3 KO on renal aquaporin expression. In summary, these data suggest that CD NOS3 may be involved in the diuretic response to a water load in a sex-specific manner; the mechanism of this effect remains to be determined.
Collapse
Affiliation(s)
- Yang Gao
- Division of Nephrology, University of Utah Health Sciences Center, Salt Lake City, Utah
| | - Deborah Stuart
- Division of Nephrology, University of Utah Health Sciences Center, Salt Lake City, Utah
| | - Jennifer S Pollock
- Division of Nephrology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Takamune Takahishi
- Division of Nephrology, Vanderbilt University School of Medicine, Nashville, Tennessee; and
| | - Donald E Kohan
- Division of Nephrology, University of Utah Health Sciences Center, Salt Lake City, Utah; .,George E. Whalen Department of Veterans Affairs Medical Center, Salt Lake City, Utah
| |
Collapse
|
15
|
Cheng H, Harris RC. Renal endothelial dysfunction in diabetic nephropathy. Cardiovasc Hematol Disord Drug Targets 2015; 14:22-33. [PMID: 24720460 DOI: 10.2174/1871529x14666140401110841] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2013] [Revised: 03/21/2014] [Accepted: 03/26/2014] [Indexed: 12/24/2022]
Abstract
Endothelial dysfunction has been posited to play an important role in the pathogenesis of diabetic nephropathy (DN). Due to the heterogeneity of endothelial cells (ECs), it is difficult to generalize about endothelial responses to diabetic stimuli. At present, there are limited techniques fordirectly measuring EC function in vivo, so diagnosis of endothelial disorders still largely depends on indirect assessment of mediators arising from EC injury. In the kidney microcirculation, both afferent and efferent arteries, arterioles and glomerular endothelial cells (GEnC) have all been implicated as targets of diabetic injury. Both hyperglycemia per se, as well as the metabolic consequences of glucose dysregulation, are thought to lead to endothelial cell dysfunction. In this regard, endothelial nitric oxide synthase (eNOS) plays a central role in EC dysfunction. Impaired eNOS activity can occur at numerous levels, including enzyme uncoupling, post-translational modifications, internalization and decreased expression. Reduced nitric oxide (NO) bioavailability exacerbates oxidative stress, further promoting endothelial dysfunction and injury. The injured ECs may then function as active signal transducers of metabolic, hemodynamic and inflammatory factors that modify the function and morphology of the vessel wall and interact with adjacent cells, which may activate a cascade of inflammatory and proliferative and profibrotic responses in progressive DN. Both pharmacological approaches and potential regenerative therapies hold promise for restoration of impaired endothelial cells in diabetic nephropathy.
Collapse
Affiliation(s)
| | - Raymond C Harris
- Division of Nephrology, S3223 MCN, Vanderbilt University School of Medicine, and Nashville Veterans Affairs Hospital, Nashville, TN 37232, USA.
| |
Collapse
|
16
|
Takahashi T, Harris RC. Role of endothelial nitric oxide synthase in diabetic nephropathy: lessons from diabetic eNOS knockout mice. J Diabetes Res 2014; 2014:590541. [PMID: 25371905 PMCID: PMC4211249 DOI: 10.1155/2014/590541] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2014] [Accepted: 09/08/2014] [Indexed: 12/29/2022] Open
Abstract
Diabetic nephropathy (DN) is the leading cause of end-stage renal disease in many countries. The animal models that recapitulate human DN undoubtedly facilitate our understanding of this disease and promote the development of new diagnostic markers and therapeutic interventions. Based on the clinical evidence showing the association of eNOS dysfunction with advanced DN, we and others have created diabetic mice that lack eNOS expression and shown that eNOS-deficient diabetic mice exhibit advanced nephropathic changes with distinct features of progressive DN, including pronounced albuminuria, nodular glomerulosclerosis, mesangiolysis, and arteriolar hyalinosis. These studies clearly defined a critical role of eNOS in DN and developed a robust animal model of this disease, which enables us to study the pathogenic mechanisms of progressive DN. Further, recent studies with this animal model have explored the novel mechanisms by which eNOS deficiency causes advanced DN and provided many new insights into the pathogenesis of DN. Therefore, here we summarize the findings obtained with this animal model and discuss the roles of eNOS in DN, unresolved issues, and future investigations of this animal model study.
Collapse
Affiliation(s)
- Takamune Takahashi
- Division of Nephrology and Hypertension, Vanderbilt University School of Medicine, S-3223, Medical Center North, Nashville, TN 37232, USA
| | - Raymond C. Harris
- Division of Nephrology and Hypertension, Vanderbilt University School of Medicine, S-3223, Medical Center North, Nashville, TN 37232, USA
| |
Collapse
|
17
|
Han KH, Jung JY, Chung KY, Kim H, Kim J. Nitric oxide synthesis in the adult and developing kidney. Electrolyte Blood Press 2014; 4:1-7. [PMID: 24459479 PMCID: PMC3894539 DOI: 10.5049/ebp.2006.4.1.1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Nitric oxide (NO) is synthesized within the adult and developing kidney and plays a critical role in the regulation of renal hemodynamics and tubule function. In the adult kidney, the regulation of NO synthesis is very cell type specific and subject to distinct control mechanisms of NO synthase (NOS) isoforms. Endothelial NOS (eNOS) is expressed in the endothelial cells of glomeruli, peritubular capillaries, and vascular bundles. Neuronal NOS (nNOS) is expressed in the tubular epithelial cells of the macula densa and inner medullary collecting duct. Furthermore, in the immature kidney, the expression of eNOS and nNOS shows unique patterns distinct from that is observed in the adult. This review will summarize the localization and presumable function of NOS isoforms in the adult and developing kidney.
Collapse
Affiliation(s)
- Ki-Hwan Han
- Department of Anatomy, College of Medicine, Ewha Womans University, Seoul, Korea
| | - Ju-Young Jung
- Department of Anatomy, College of Veterinary Medicine, Chungnam National University, Daejeon, Korea
| | - Ku-Yong Chung
- Department of Surgery, College of Medicine, Ewha Womans University, Seoul, Korea
| | - Hyang Kim
- Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University, School of Medicine, Seoul, Korea
| | - Jin Kim
- Department of Anatomy and MRC for Cell Death Disease Research Center, College of Medicine, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
18
|
Vinturache AE, Smith FG. Angiotensin receptors modulate the renal hemodynamic effects of nitric oxide in conscious newborn lambs. Physiol Rep 2014; 2:e12027. [PMID: 24872358 PMCID: PMC4098750 DOI: 10.14814/phy2.12027] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Revised: 04/28/2014] [Accepted: 05/01/2014] [Indexed: 12/20/2022] Open
Abstract
This study aimed to elucidate the roles of both angiotensin II (ANG II) receptors - type 1 (AT1Rs) and type 2 (AT2Rs) - separately and together in influencing hemodynamic effects of endogenously produced nitric oxide (NO) during postnatal development. In conscious, chronically instrumented lambs aged ~1 week (8 ± 1 days, N = 8) and ~6 weeks (41 ± 2 days, N = 8), systolic, diastolic, and mean arterial pressure (SAP, DAP, MAP) and venous pressure (MVP), renal blood flow (RBF), and renal vascular resistance (RVR) were measured in response to the l-arginine analog, l-NAME after pretreatment with either the AT1R antagonist, ZD 7155, the AT2R antagonist, PD 123319, or both antagonists. The increase in SAP, DAP, and MAP by l-NAME was not altered by either ATR antagonist in either age group. The increase in RBF after l-NAME was, however, altered by both ATR antagonists in an age-dependent manner, which was mediated predominantly through AT2Rs in newborn lambs. These findings reveal that there is an age-dependent interaction between the renin-angiotensin (RAS) and the NO pathway in regulating renal but not systemic hemodynamics through both ATRs, whereas AT2Rs appear to be important in the renal hemodynamic effects of NO early in life.
Collapse
Affiliation(s)
- Angela E. Vinturache
- The Alberta Children's Hospital Research Institute for Child and Maternal Health, University of Calgary, Calgary, Alberta, Canada
- The Department of Physiology & Pharmacology, University of Calgary, Calgary, Alberta, Canada
| | - Francine G. Smith
- The Alberta Children's Hospital Research Institute for Child and Maternal Health, University of Calgary, Calgary, Alberta, Canada
- The Department of Physiology & Pharmacology, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
19
|
Larsen T, Mose FH, Bech JN, Pedersen EB. Effect of paricalcitol on renin and albuminuria in non-diabetic stage III-IV chronic kidney disease: a randomized placebo-controlled trial. BMC Nephrol 2013; 14:163. [PMID: 23889806 PMCID: PMC3729723 DOI: 10.1186/1471-2369-14-163] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2013] [Accepted: 07/19/2013] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Vitamin D receptor activators reduce albuminuria, and may improve survival in chronic kidney disease (CKD). Animal studies suggest that these pleiotropic effects of vitamin D may be mediated by suppression of renin. However, randomized trials in humans have yet to establish this relationship. METHODS In a randomized, placebo-controlled, double-blinded crossover study, the effect of oral paricalcitol (2 μg/day) was investigated in 26 patients with non-diabetic, albuminuric stage III-IV CKD. After treatment, plasma concentrations of renin (PRC), angiotensin II (AngII) and aldosterone (Aldo) were measured. GFR was determined by 51Cr-EDTA clearance. Assessment of renal NO dependency was performed by infusion of NG-monomethyl-L-arginine (L-NMMA). Albumin excretion rate (AER) was analyzed in 24-h urine and during 51Cr-EDTA clearance. RESULTS Paricalcitol did not alter plasma levels of renin, AngII, Aldo, or urinary excretion of sodium and potassium. A modest reduction of borderline significance was observed in AER, and paricalcitol abrogated the albuminuric response to L-NMMA. CONCLUSIONS In this randomized, placebo-controlled trial paricalcitol only marginally decreased AER and did not alter circulating levels of renin, AngII or Aldo. The abrogation of the rise in albumin excretion by paricalcitol during NOS blockade may indicate that favourable modulation of renal NO dependency could be involved in mediating reno-protection and survival benefits in CKD. TRIAL REGISTRATION ClinicalTrials.gov identifier: NCT01136564.
Collapse
|
20
|
Lee SY, Han SM, Kim JE, Chung KY, Han KH. Expression of E-cadherin in pig kidney. J Vet Sci 2013; 14:381-6. [PMID: 23820247 PMCID: PMC3885730 DOI: 10.4142/jvs.2013.14.4.381] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2012] [Accepted: 12/24/2012] [Indexed: 12/24/2022] Open
Abstract
E-cadherin is a cell adhesion molecule that plays an important role in maintaining renal epithelial polarity and integrity. The purpose of this study was to determine the exact cellular localization of E-cadherin in pig kidney. Kidney tissues from pigs were processed for light and electron microscopy immunocytochemistry, and immunoblot analysis. E-cadhedrin bands of the same size were detected by immunoblot of samples from rat and pig kidneys. In pig kidney, strong E-cadherin expression was observed in the basolateral plasma membrane of the tubular epithelial cells. E-cadherin immunolabeling was not detected in glomeruli or blood vessels of pig kidney. Double-labeling results demonstrated that E-cadherin was expressed in the calbindin D28k-positive distal convoluted tubule and H(+)-ATPase- positive collecting duct, but not in the aquaporin 1-positive, N-cadherin-positive proximal tubule. In contrast to rat, E-cadherin immunoreactivity was not expressed at detectable levels in the Tamm-Horsfall protein-positive thick ascending limb of pig kidney. Immunoelectron microscopy confirmed that E-cadherin was localized in both the lateral membranes and basal infoldings of the collecting duct. These results suggest that E-cadherin may be a critical adhesion molecule in the distal convoluted tubule and collecting duct cells of pig kidney.
Collapse
Affiliation(s)
- Su-Youn Lee
- Departments of Anatomy, Ewha Womans University School of Medicine, Seoul 158-710, Korea
| | | | | | | | | |
Collapse
|
21
|
Silicic acid in drinking water prevents age-related alterations in the endothelium-dependent vascular relaxation modulating eNOS and AQP1 expression in experimental mice: an immunohistochemical study. Acta Histochem 2013. [PMID: 23177919 DOI: 10.1016/j.acthis.2012.10.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The maintenance of endothelial integrity is of great importance in coping with age-related vascular alterations. Endothelium-derived nitric oxide is one of the various vasoactive substances able to regulate vascular tone and homeostasis, and whose decrease is known to be related with senescence in endothelial cells. There are reports on the efficacy of silicon, especially as silicic acid, in protecting vascular integrity during age-related vascular diseases. The aim of this study was to evaluate the ability of supplementation of silicic acid in drinking water in the maintenance of vascular health in a mouse model of early physiological aging. In particular, we evaluated the relationship between Si supplementation and endothelial nitric oxide synthase (eNOS) expression, taking into account also the aquaporin-1 (AQP-1) isoform that, as recently reported, seems to be involved in nitric oxide transport across cell membranes. Our results showed that silicic acid supplementation increased both eNOS and AQP-1 expression, suggesting that silicic acid modulation of endothelial nitric oxide synthase and aquaporin-1 could represent a potential strategy against age-related vascular senescence.
Collapse
|
22
|
Masoud MS, Anwar SS, Afzal MZ, Mehmood A, Khan SN, Riazuddin S. Pre-conditioned mesenchymal stem cells ameliorate renal ischemic injury in rats by augmented survival and engraftment. J Transl Med 2012; 10:243. [PMID: 23217165 PMCID: PMC3543338 DOI: 10.1186/1479-5876-10-243] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2012] [Accepted: 11/28/2012] [Indexed: 01/09/2023] Open
Abstract
Background Ischemia is the major cause of acute kidney injury (AKI), associated with high mortality and morbidity. Mesenchymal stem cells (MSCs) have multilineage differentiation potential and can be a potent therapeutic option for the cure of AKI. Methods MSCs were cultured in four groups SNAP (S-nitroso N-acetyl penicillamine), SNAP + Methylene Blue (MB), MB and a control for in vitro analysis. Cultured MSCs were pre-conditioned with either SNAP (100 μM) or MB (1 μM) or both for 6 hours. Renal ischemia was induced in four groups (as in in vitro study) of rats by clamping the left renal padicle for 45 minutes and then different pre-conditioned stem cells were transplanted. Results We report that pre-conditioning of MSCs with SNAP enhances their proliferation, survival and engraftment in ischemic kidney. Rat MSCs pre-conditioned with SNAP decreased cell apoptosis and increased proliferation and cytoprotective genes’ expression in vitro. Our in vivo data showed enhanced survival and engraftment, proliferation, reduction in fibrosis, significant improvement in renal function and higher expression of pro-survival and pro-angiogenic factors in ischemic renal tissue in SNAP pre-conditioned group of animals. Cytoprotective effects of SNAP pre-conditioning were abrogated by MB, an inhibitor of nitric oxide synthase (NOS) and guanylate cyclase. Conclusion The results of these studies demonstrate that SNAP pre-conditioning might be useful to enhance therapeutic potential of MSCs in attenuating renal ischemia reperfusion injury.
Collapse
Affiliation(s)
- Muhammad Shareef Masoud
- National Centre of Excellence in Molecular Biology, University of the Punjab, Lahore, Pakistan.
| | | | | | | | | | | |
Collapse
|
23
|
Abstract
According to the Developmental Origins of Health and Disease hypothesis intrauterine or postnatal adaptations to the environment causes morphologic, physiologic or metabolic changes that influence health later in life. These adaptations seem to be carried out through structural, functional and epigenetic modifications. Multiple animal models of cardiovascular programming have been developed, and a brief overview of well-known models and mechanisms is presented. However, developmental programming also offers a novel approach to prevent cardiovascular and related diseases through so-called Reprogramming: administration of appropriate or inhibition of deleterious perinatal factors in induced or genetic models ameliorated undesirable development that otherwise would inevitably have lead to more severe hypertension, cardiovascular and renal disease. A comprehensive overview of these studies suggests that, in analogy to what has been previously recognised in programming, many quite different reprogramming interventions all have similar protective effects. Whether this is due to common final epigenetic pathways remains to be shown.
Collapse
Affiliation(s)
- Manuel S Santos
- Department of Nephrology and Hypertension, Laboratory of Renal and Vascular Biology, University Medical Center Utrecht, Utrecht, The Netherlands.
| | | |
Collapse
|
24
|
Jamal A, Man HSJ, Marsden PA. Gene regulation in the vascular endothelium: why epigenetics is important for the kidney. Semin Nephrol 2012; 32:176-84. [PMID: 22617766 DOI: 10.1016/j.semnephrol.2012.02.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
We now appreciate that the vascular endothelium plays a crucial role in regulating normal blood vessel physiology in the kidney. The gene products responsible are commonly expressed exclusively, or preferentially, in this cell type. However, despite the importance of regulated gene expression in the vascular endothelium, relatively little is known about the mechanisms that restrict endothelial-specific gene expression to this cell type. Even less is known about how gene expression might be restricted to endothelial cells of discrete regions of the kidney, such as the glomerulus or vasa recta. Although significant progress has been made toward understanding the regulation of endothelial genes through cis/trans paradigms, it has become apparent that additional mechanisms also must be operative. Classic models of transcription in vascular endothelial cells, specifically the cis/trans paradigm, have limitations. For instance, how does the environment have chronic effects on gene expression in endothelial cells after weeks or years? When an endothelial cell divides, how is this information transmitted to daughter cells? Chromatin-based mechanisms, including cell-specific DNA methylation patterns and post-translational histone modifications, recently were shown to play important roles in gene expression. This review investigates the involvement of epigenetic regulatory mechanisms in vascular endothelial cell-specific gene expression using endothelial nitric oxide synthase as a prototypical model.
Collapse
Affiliation(s)
- Alisha Jamal
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | | | | |
Collapse
|
25
|
Rodebaugh J, Sekulic M, Davies W, Montgomery S, Khraibi A, Solhaug MJ, Ratliff BB. Neuronal nitric oxide synthase, nNOS, regulates renal hemodynamics in the postnatal developing piglet. Pediatr Res 2012; 71:144-9. [PMID: 22258124 DOI: 10.1038/pr.2011.23] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
INTRODUCTION Nitric oxide (NO) vasodilation critically modulates renal hemodynamics in the neonate compared with the adult. Based on the postnatal expression pattern of renal neuronal nitric oxide synthase (nNOS), the hypothesis was that nNOS is the major NOS isoform regulating renal hemodynamics in the immature, but not mature, kidney. RESULTS NOS inhibitors did not alter mean arterial pressure (MAP) in either group. Intrarenal S-methyl-L-thiocitrulline (L-SMTC) in newborns significantly reduced renal blood flow (RBF) 38 ± 4%, glomerular filtration rate (GFR) 42 ± 6%, and increased renal vascular resistance (RVR) 37 ± 7%, whereas intrarenal L-nitro-arginine methyl ester (L-NAME) affected RBF, GFR, and RVR equivalent to L-SMTC treatment. When L-NAME was administered after L-SMTC treatment, newborn renal hemodynamic changes were not further altered from what was observed when L-SMTC was administered alone. In contrast, in the adult, only intrarenal L-NAME, and not L-SMTC, affected renal hemodynamic responses. DISCUSSION In conclusion, these studies demonstrate that nNOS is an important regulator of renal hemodynamics in the newborn kidney, but not in the adult. METHODS Experiments compared renal hemodynamic responses with intrarenal infusion of L-NAME, an inhibitor of all NOS isoforms, with the selective nNOS inhibitor L-SMTC in the newborn piglet and the adult pig.
Collapse
Affiliation(s)
- Justin Rodebaugh
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, Virginia, USA
| | | | | | | | | | | | | |
Collapse
|
26
|
Lee SY, Shin JA, Kwon HM, Weiner ID, Han KH. Renal ischemia-reperfusion injury causes intercalated cell-specific disruption of occludin in the collecting duct. Histochem Cell Biol 2011; 136:637-47. [PMID: 22048282 PMCID: PMC3214267 DOI: 10.1007/s00418-011-0881-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/23/2011] [Indexed: 11/30/2022]
Abstract
Renal ischemic events open tight junctions and disrupt epithelial polarity. The purpose of this study was to examine the effects of ischemia–reperfusion (IR) injury on expression and distribution of the tight junction proteins, occludin and ZO-1, in the rat kidney. IR injury was induced by clamping both renal pedicles for 30 min and animals were killed at 6 h after the reperfusion. IR injury decreased blood bicarbonate level, but did not persistently alter pH, Na+, K+, or Cl−. In control kidneys, occludin immunoreactivity was intense in the tight junctions in the thick ascending limb, distal convoluted tubule, and collecting duct, moderate in the thin limbs of the loop of Henle, and was not detected in the proximal tubule, glomerulus, and blood vessels. ZO-1 was expressed in the same sites in which occludin was expressed, and additionally was also expressed in the proximal tubule, glomerulus, and vascular endothelial cells. IR kidneys exhibited damaged renal tubular epithelial cells in both proximal tubule and collecting duct segments in the outer medulla. In the collecting duct, the response of intercalated cells and principal cells differed. Following IR injury, intercalated cells, but not principal cells, lost their normal epithelial polarity and were frequently extruded into the tubule lumen. Occludin, instead of being localized to tight junctions, was localized diffusely in the cytoplasm in intercalated cells of IR kidneys. Principal cells, in contrast, were not detectably affected and neither occludin nor ZO-1 expression were altered in response to IR injury. The normal localization of ZO-1 expression to tight junction sites in both the proximal tubule and collecting duct was altered in response to IR, and, instead, ZO-1 expression was present diffusely in the cytoplasm. IR injury did not alter detectably either occludin or ZO-1 localization to the tight junction of the thick ascending limb cells. The abundance of total occludin protein by immunoblot analysis was not changed with IR injury. These results demonstrate that renal IR injury causes tight junction disruptions in both the proximal tubule and the collecting duct, and that altered distribution of the tight junction protein, occludin, may play a critical role in the collecting duct dysfunction which IR induces.
Collapse
Affiliation(s)
- Su-Youn Lee
- Department of Anatomy, Ewha Womans University School of Medicine, 911-1 Mok-6-dong, Yangcheon-ku, Seoul, 158-710 Korea
| | - Jung-A Shin
- Department of Anatomy, Ewha Womans University School of Medicine, 911-1 Mok-6-dong, Yangcheon-ku, Seoul, 158-710 Korea
| | - H. Moo Kwon
- Division of Nephrology, University of Maryland School of Medicine, Baltimore, MD USA
| | - I. David Weiner
- Division of Nephrology, University of Florida College of Medicine, Gainesville, FL USA
- Nephrology Section, NF/SGVHS, Gainesville, FL USA
| | - Ki-Hwan Han
- Department of Anatomy, Ewha Womans University School of Medicine, 911-1 Mok-6-dong, Yangcheon-ku, Seoul, 158-710 Korea
| |
Collapse
|
27
|
Finch JL, Suarez EB, Husain K, Ferder L, Cardema MC, Glenn DJ, Gardner DG, Liapis H, Slatopolsky E. Effect of combining an ACE inhibitor and a VDR activator on glomerulosclerosis, proteinuria, and renal oxidative stress in uremic rats. Am J Physiol Renal Physiol 2011; 302:F141-9. [PMID: 21957179 DOI: 10.1152/ajprenal.00293.2011] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Angiotensin-converting enzyme (ACE) inhibitors ameliorate the progression of renal disease. In combination with vitamin D receptor activators, they provide additional benefits. In the present study, uremic (U) rats were treated as follows: U+vehicle (UC), U+enalapril (UE; 25 mg/l in drinking water), U+paricalcitol (UP; 0.8 μg/kg ip, 3 × wk), or U+enalapril+paricalcitol (UEP). Despite hypertension in UP rats, proteinuria decreased by 32% vs. UC rats. Enalapril alone, or in combination with paricalcitol, further decreased proteinuria (≈70%). Glomerulosclerosis and interstitial infiltration increased in UC rats. Paricalcitol and enalapril inhibited this. The increase in cardiac atrial natriuretic peptide (ANP) seen in UC rats was significantly decreased by paricalcitol. Enalapril produced a more dramatic reduction in ANP. Renal oxidative stress plays a critical role in inflammation and progression of sclerosis. The marked increase in p22(phox), a subunit of NADPH oxidase, and decrease in endothelial nitric oxide synthase were inhibited in all treated groups. Cotreatment with both compounds inhibited the uremia-induced increase in proinflammatory inducible nitric oxide synthase (iNOS) and glutathione peroxidase activity better than either compound alone. Glutathione reductase was also increased in UE and UP rats vs. UC. Kidney 4-hydroxynonenal was significantly increased in the UC group compared with the normal group. Combined treatment with both compounds significantly blunted this increase, P < 0.05, while either compound alone had no effect. Additionally, the expression of Mn-SOD was increased and CuZn-SOD decreased by uremia. This was ameliorated in all treatment groups. Cotreatment with enalapril and paricalcitol had an additive effect in increasing CuZn-SOD expression. In conclusion, like enalapril, paricalcitol alone can improve proteinuria, glomerulosclerosis, and interstitial infiltration and reduce renal oxidative stress. The effects of paricalcitol may be amplified when an ACE inhibitor is added since cotreatment with both compounds seems to have an additive effect on ameliorating uremia-induced changes in iNOS and CuZn-SOD expression, peroxidase activity, and renal histomorphometry.
Collapse
Affiliation(s)
- Jane L Finch
- Renal Div., Box 8126, Washington Univ. School of Medicine, St. Louis, MO 63110, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Martins AR, Zanella CAB, Zucchi FCR, Dombroski TCD, Costa ET, Guethe LM, Oliveira AO, Donatti ALF, Neder L, Chimelli L, De Nucci G, Lee-Ho P, Murad F. Immunolocalization of nitric oxide synthase isoforms in human archival and rat tissues, and cultured cells. J Neurosci Methods 2011; 198:16-22. [PMID: 21420432 DOI: 10.1016/j.jneumeth.2011.02.024] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2010] [Revised: 02/16/2011] [Accepted: 02/24/2011] [Indexed: 12/21/2022]
Abstract
Nitric oxide (NO) exerts important physiological and pathological roles in humans. The study of NO requires the immunolocalization of its synthesizing enzymes, neuronal, endothelial and inducible NO synthases (NOS). NOS are labile to formalin-fixation and paraffin-embedding, which are used to prepare human archival tissues. This lability has made NOS immunohistochemical studies difficult, and a detailed protocol is not yet available. We describe here a protocol for the immunolocalization of NOS isoforms in human archival cerebellum and non-nervous tissues, and in rat tissues and cultured cells. Neuronal NOS antigenicity in human archival and rat nervous tissue sections was microwave-retrieved in 50 mM Tris-HCl buffer, pH 9.5, for 20 min at 900 W. Neuronal NOS was expressed in stellate, basket, Purkinje and granule cells in human and rat cerebellum. Archival and frozen human cerebellar sections showed the same neuronal NOS staining pattern. Archival cerebellar sections not subjected to antigen retrieval stained weakly. Antigenicity of inducible NOS in human lung was best retrieved in 10 mM sodium citrate buffer, pH 6.0, for 15 min at 900 W. Inflammatory cells in a human lung tuberculoma were strongly stained by anti-inducible NOS antibody. Anti-endothelial NOS strongly stained kidney glomeruli. Cultured PC12 cells were strongly stained by anti-neuronal NOS without antigen retrieving. The present immunohistochemistry protocol is easy to perform, timeless, and suitable for the localization of NOS isoforms in nervous and non-nervous tissues, in human archival and rat tissues. It has been extensively used in our laboratory, and is also appropriate for other antigens.
Collapse
Affiliation(s)
- Antonio R Martins
- Institute of Biological Sciences, Federal University of Triângulo Mineiro, Uberaba, MG, Brazil.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Effects of low protein intake on the development of the remaining kidney in subtotally nephrectomized immature rats: expression of inducible and endothelial NO synthase. Med Mol Morphol 2010; 43:116-22. [PMID: 20683700 DOI: 10.1007/s00795-009-0485-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2009] [Accepted: 11/16/2009] [Indexed: 10/19/2022]
Abstract
We examined the effects of low protein intake on the development of the remaining kidney in subtotally (5/6) nephrectomized immature rats. Three-week-old rats were kept on a diet containing either 12% protein (Lp rats) or 18% protein (Np rats) for 4 or 8 weeks after subtotal nephrectomy (SUNx). In Western blot analysis, the endothelial NO synthase (eNOS) protein expression of the Lp rats was significantly higher than that of the Np rats at 4 weeks after SUNx. Immunohistochemically, more inducible NO synthase (iNOS)-positive cells were observed in the Np rats than in the Lp rats 4 weeks after SUNx in the distal tubules. In semiquantitative RT-PCR, the expression of renin mRNA was significantly lower in the Lp rats than in the Np rats at 4 and 8 weeks after SUNx. These findings reveal that protein restriction is effective in preventing renal failure of immature rats and that the changes in the expression levels of renin, eNOS, and iNOS is involved in the process of this prevention.
Collapse
|
30
|
Ratliff B, Rodebaugh J, Sekulic M, Dong KW, Solhaug M. Nitric oxide synthase and renin-angiotensin gene expression and NOS function in the postnatal renal resistance vasculature. Pediatr Nephrol 2009; 24:355-65. [PMID: 18836750 DOI: 10.1007/s00467-008-0977-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2007] [Revised: 07/15/2008] [Accepted: 07/16/2008] [Indexed: 10/21/2022]
Abstract
Nitric oxide (NO), produced by nitric oxide synthase (NOS), critically counteracts angiotensin-II-enhanced vascular resistance in the immature kidney, perhaps due to the developmental regulation of NOS expression and function in the postnatal preglomerular resistance vessels (PRV). Our experiments measured the messenger ribonucleic acid (mRNA) gene expression of neuronal NOS (nNOS), endothelial NOS (eNOS), and components of the renin-angiotensin system (renin, AT1 and AT2 receptors), by real-time RT-PCR, as well as NOS enzymatic activity by citrulline assay in PRVs (afferent, interlobular, and arcuate arterioles) obtained from swine ages newborn, 7 and 21 days, and adult. NOS enzymatic activity was upregulated in PRVs immediately after birth but decreased to adult levels with maturation. Neuronal NOS, renin, and AT2 receptor expression in PRVs were upregulated in the newborn and decreased with age to lowest levels in the adult. In contrast, eNOS and AT1 receptor expression were downregulated at birth but increased to the highest levels in the adult. Upregulated NOS enzymatic activity in newborn PRVs supports the critical neonatal role for NO renal vascular vasodilation. Upregulated nNOS gene expression, concomitant with downregulated eNOS gene expression in neonatal PRVs, suggests that the nNOS isoform may be responsible for counteracting angiotensin II increased vascular resistance in immature porcine PRVs.
Collapse
Affiliation(s)
- Brian Ratliff
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, VA, 23501-1980, USA
| | | | | | | | | |
Collapse
|
31
|
Nakayama T, Sato W, Kosugi T, Zhang L, Campbell-Thompson M, Yoshimura A, Croker BP, Johnson RJ, Nakagawa T. Endothelial injury due to eNOS deficiency accelerates the progression of chronic renal disease in the mouse. Am J Physiol Renal Physiol 2008; 296:F317-27. [PMID: 19036847 DOI: 10.1152/ajprenal.90450.2008] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The vascular endothelium expresses endothelial nitric oxide synthase (eNOS) that generates nitric oxide (NO) to help maintain vascular integrity due to its anti-inflammatory, antiproliferative, and antithrombogenic effects. Pharmacological blockade of NO production has been shown to exacerbate renal injury in chronic renal disease and induces endothelial cell loss. However, pharmacological inhibition of NO nonspecifically blocks other types of NOS and therefore does not define the specific role of eNOS in kidney disease. We hypothesized that a lack of endothelial eNOS can induce a loss of glomerular and peritubular capillary endothelium and exacerbate renal injury in progressive renal disease. We tested out this hypothesis using remnant kidney (RK) in eNOS knockout (eNOS KO) mice. Systolic blood pressure was significantly higher, and renal function was worse in RK-eNOS KO mice compared with those in RK-C57BL6 mice. eNOS deficiency resulted in more severe glomerulosclerosis, mesangiolysis, and tubular damage. Glomerular and tubular macrophage infiltration and collagen deposition were also greater in RK-eNOS KO mice. Renal injuries in the RK-eNOS KO mice were accompanied by a greater loss of endothelial cells that was shown to be due to both a decrease in endothelial cell proliferation and an increase in apoptosis. A lack of eNOS accelerates both glomerular and tubulointerstitial injury with a loss of glomerular capillaries and peritubular capillaries. Impaired endothelial function is likely a direct risk factor for renal disease.
Collapse
Affiliation(s)
- Takahiro Nakayama
- Division of Nephrology, Univ. of Colorado Denver, P.O. Box C281, Aurora, CO 80045, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Cunha AR, Aguila MB, Mandarim-de-Lacerda CA. Effects of early postnatal hyperglycaemia on renal cortex maturity, endothelial nitric oxide synthase expression and nephron deficit in mice. Int J Exp Pathol 2008; 89:284-91. [PMID: 18715472 DOI: 10.1111/j.1365-2613.2008.00593.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The influence of hyperglycaemia on nephrogenesis on Swiss mice pups treated with streptozotocin (STZ) (40 mg/kg, i.p.) was studied after birth, at 7 and 21 days. Kidneys were prepared for light microscopy, immunohistochemistry and stereology. In 7-day-old pups, both immature and mature glomeruli were evaluated separately. Proliferating cell nuclear antigen (PCNA) and endothelial nitric oxide synthase (eNOS) immunostaining were performed and quantified. At age 7 days, the immature-to-mature glomeruli ratio (IMGR) was significantly higher in the STZ group than in the control group. There was no difference in the number of glomeruli between the STZ and control groups; however, the number of glomeruli increased by more than 20% in the control group until 21 days of age, but not in the STZ group. STZ pups showed numerous PCNA-positive nuclei mainly in tubular cells, but not control pups. At 21 days, eNOS expression in the outer layer of glomerular endothelial nuclei was strong in control pups, but weaker in STZ pups. Treatment with STZ during the early neonatal period disturbs the normal nephrogenesis occurring at this stage of the rodent's life and causes retardation in renal cortical maturity, as indicated by the increase in both PCNA expression and IMGR, and reduction in eNOS expression.
Collapse
Affiliation(s)
- Ana Rosa Cunha
- Laboratory of Morphometry and Cardiovascular Morphology, Biomedical Center, Institute of Biology, State University of Rio de Janeiro, Brazil
| | | | | |
Collapse
|
33
|
Ratliff B, Rodebaugh J, Sekulic M, Solhaug M. Glomerular eNOS gene expression during postnatal maturation and AT1 receptor inhibition. Pediatr Nephrol 2007; 22:1135-42. [PMID: 17437130 DOI: 10.1007/s00467-007-0489-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2006] [Revised: 02/11/2007] [Accepted: 02/28/2007] [Indexed: 11/29/2022]
Abstract
Glomerular maturation increases from immature superficial to advanced juxtamedullary nephrons, while nephrogenesis continues postnatally in porcine kidneys. Endothelial NOS, eNOS, shows significant postnatal renal developmental regulation, perhaps mediated by Angiotensin II (AII). The objective was to compare eNOS mRNA gene expression between superficial and juxtamedullary glomeruli obtained from piglets and adult pigs utilizing laser capture microdissection during basal conditions and, to determine the role of the AII AT1 receptor, AT1, after chronic AT1 inhibition (AT1X) with candesartan. Superficial glomerular eNOS expression was lowest in newborns (NB) and at 7 days, and was highest in 14, 21 day old piglets and adults. Juxtamedullary glomerular eNOS, while similar in NB, 14, 21 day and adult, dipped to the lowest level at 7 days. Juxtamedullary glomerular eNOS expression in the NB was 7 fold greater than in superficial glomeruli. AT1X did not change eNOS expression in adult glomeruli. AT1X significantly reduced NB eNOS expression in both superficial, 90+/-10%, and juxtamedullary glomeruli, 89+/-5% respectively. In conclusion, eNOS gene expression demonstrates significant differences between NB superficial and juxtamedullary glomeruli, significant postnatal developmental regulation of both glomerular locations, and this expression may be mediated in the NB by AII via the AT1 receptor.
Collapse
Affiliation(s)
- Brian Ratliff
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, VA 23501-1980, USA
| | | | | | | |
Collapse
|
34
|
Abstract
Endothelial cells, which form the inner cellular lining of blood vessels and lymphatics, display remarkable heterogeneity in structure and function. This is the second of a 2-part review on the phenotypic heterogeneity of blood vessel endothelial cells. The first part discusses the scope, the underlying mechanisms, and the diagnostic and therapeutic implications of phenotypic heterogeneity. Here, these principles are applied to an understanding of organ-specific phenotypes in representative vascular beds including arteries and veins, heart, lung, liver, and kidney. The goal is to underscore the importance of site-specific properties of the endothelium in mediating homeostasis and focal vascular pathology, while at the same time emphasizing the value of approaching the endothelium as an integrated system.
Collapse
Affiliation(s)
- William C Aird
- Division of Molecular and Vascular Medicine, Department of Medicine, and Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Mass 02215, USA.
| |
Collapse
|
35
|
Forbes MS, Thornhill BA, Park MH, Chevalier RL. Lack of endothelial nitric-oxide synthase leads to progressive focal renal injury. THE AMERICAN JOURNAL OF PATHOLOGY 2007; 170:87-99. [PMID: 17200185 PMCID: PMC1762694 DOI: 10.2353/ajpath.2007.060610] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Because endothelial nitric-oxide synthase (eNOS) is generally considered protective against renal injury, we examined eNOS knockout mice for kidney pathology. In 80% of the adults examined, the renal surface was marked by distinct indented scars containing crowded small glomeruli but lacking attached tubules. Although vasculature was intact in the scars, Bowman's space was dilated and glomerular tufts were degenerated. The atubular glomeruli were embedded in a dense interstitial matrix composed of cells positive for fibroblast (FSP-1) or macrophage (F4/80) markers, degenerated proximal tubules and collecting ducts, and diffuse fibrotic deposits. Surrounding regions of kidney contained mostly normal-appearing tubules, but enlarged or sclerotic glomeruli were also present. In neonatal animals, apoptosis and necrosis were concentrated in tubules within focal parenchymal zones, with narrowing of the glomerulotubular "neck." In summary, targeted deletion of eNOS in mice leads to progressive focal renal abnormalities, including glomerular hypoplasia, and tubular cell death, leading to separation of glomeruli from tubules and tubular disruption. These abnormalities begin developing during the normal up-regulation of eNOS in the maturing kidney and are similar to those of a variety of chronic renal disorders. Endogenous renal eNOS production therefore seems critical for the maintenance of nephron maturation and integrity.
Collapse
Affiliation(s)
- Michael S Forbes
- Department of Pediatrics, University of Virginia, Box 800386, Charlottesville, VA 22908, USA
| | | | | | | |
Collapse
|
36
|
Noh H, Kim JS, Han KH, Lee GT, Song JS, Chung SH, Jeon JS, Ha H, Lee HB. Oxidative stress during peritoneal dialysis: implications in functional and structural changes in the membrane. Kidney Int 2006; 69:2022-8. [PMID: 16641917 DOI: 10.1038/sj.ki.5001506] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Progressive peritoneal fibrosis, membrane hyperpermeability, and ultrafiltration failure have been observed in patients on long-term peritoneal dialysis (PD). The present study tested the hypothesis that reactive oxygen species (ROS) generated by conventional PD solution (PDS) mediate functional and structural alterations of peritoneal membrane in vivo. Sprague-Dawley rats were randomized to control, PDS, PDS with an antioxidant, and PDS with an angiotensin II (Ang II) receptor blocker. Commercial PDS containing 3.86% glucose (20-30 ml) with or without N-acetylcystein (NAC) 10 mM or losartan 5 mg/kg was administered intraperitoneally twice a day for 12 weeks. Control rats received sham injection. Rats treated with PDS had significantly lower drain volume and D(4)/D(0) glucose, but higher D(4)/P(4) creatinine and increased membrane thickness and endothelial NOS (eNOS) expression compared to control rats. Omental transforming growth factor (TGF)-beta1, vascular endothelial growth factor (VEGF), collagen I, and heat-shock protein (hsp) 47 expression and lipid peroxide levels and dialysate VEGF and Ang II concentrations were significantly increased in rats treated with PDS compared to control. All of these changes were prevented by both NAC and losartan. In conclusion, the present study demonstrates that ROS generated by conventional PDS are, in large part, responsible for peritoneal fibrosis and membrane hyperpermeability. We suggest that antioxidants or Ang II receptor blockers may allow better preservation of the structural and functional integrity of the peritoneal membrane during long-term PD.
Collapse
Affiliation(s)
- H Noh
- Hyonam Kidney Laboratory, Soon Chun Hyang University, Seoul, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Schreuder M, Delemarre-van de Waal H, van Wijk A. Consequences of Intrauterine Growth Restriction for the Kidney. Kidney Blood Press Res 2006; 29:108-25. [PMID: 16837795 DOI: 10.1159/000094538] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Low birth weight due to intrauterine growth restriction is associated with various diseases in adulthood, such as hypertension, cardiovascular disease, insulin resistance and end-stage renal disease. The purpose of this review is to describe the effects of intrauterine growth restriction on the kidney. Nephrogenesis requires a fine balance of many factors that can be disturbed by intrauterine growth restriction, leading to a low nephron endowment. The compensatory hyperfiltration in the remaining nephrons results in glomerular and systemic hypertension. Hyperfiltration is attributed to several factors, including the renin-angiotensin system (RAS), insulin-like growth factor (IGF-I) and nitric oxide. Data from human and animal studies are presented, and suggest a faltering IGF-I and an inhibited RAS in intrauterine growth restriction. Hyperfiltration makes the kidney more vulnerable during additional kidney disease, and is associated with glomerular damage and kidney failure in the long run. Animal studies have provided a possible therapy with blockage of the RAS at an early stage in order to prevent the compensatory glomerular hyperfiltration, but this is far from being applicable to humans. Research is needed to further unravel the effect of intrauterine growth restriction on the kidney.
Collapse
Affiliation(s)
- Michiel Schreuder
- Department of Pediatric Nephrology, VU University Medical Center, Amsterdam, The Netherlands.
| | | | | |
Collapse
|
38
|
Han KH, Lee UY, Jang YS, Cho YM, Jang YM, Hwang IA, Ghee JY, Lim SW, Kim WY, Yang CW, Kim J, Kwon OJ. Differential regulation of B/K protein expression in proximal and distal tubules of rat kidneys with ischemia-reperfusion injury. Am J Physiol Renal Physiol 2006; 292:F100-6. [PMID: 16896191 DOI: 10.1152/ajprenal.00009.2006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Brain/kidney (B/K) protein is a novel double C2-like-domain protein that is highly expressed in rat brain and kidney, but its cellular localization and functional role in the kidney are still undetermined. We examined the cellular localization of B/K protein in the rat kidney under normal and ischemic conditions. Ischemia-reperfusion (I/R) injury was induced by clamping both renal arteries for 45 min, and animals were killed at 1 and 6 h and 1, 2, 3, 5, 7, 14, and 28 days after the reperfusion. Kidney tissues were processed for immunohistochemistry and immunoblot analyses using rabbit anti-B/K polyclonal antibodies. In control kidneys, B/K protein was expressed primarily in distal tubules including the thick ascending limb, distal convoluted and connecting tubules, and collecting duct. Notably, B/K protein was also expressed in the straight portion (S3 segment), but not in the S1 or S2, of proximal tubules, and podocytes of the glomerulus. In rat kidneys with I/R injury, expression of B/K protein was differentially regulated according to the anatomic location. In distal tubules, overall expression of B/K protein was markedly decreased. On the other hand, I/R injury significantly increased B/K protein expression in the S3 segment of the outer medulla as well as in the rat proximal tubular epithelial cell line NRK-52E in vitro. Furthermore, B/K protein was strongly expressed in many exfoliated cells in the lumen and urine. These findings suggest that B/K protein is closely associated with cell death in proximal tubules, which are vulnerable to I/R injury in the kidney.
Collapse
Affiliation(s)
- Ki-Hwan Han
- Department of Anatomy, College of Medicine, Ewha Womans University, Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|