1
|
Farouk SM, Basha WAA, Emam MA, Metwally E. Differential expression of epithelial and smooth muscle lineage-specific markers of metanephros in one-humped camel foetuses. Anat Histol Embryol 2024; 53:e12985. [PMID: 37814965 DOI: 10.1111/ahe.12985] [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: 06/23/2023] [Revised: 09/28/2023] [Accepted: 10/02/2023] [Indexed: 10/11/2023]
Abstract
The development of the metanephros in one-humped camels involves a complex series of interactions between epithelial and mesenchymal cells. As a result, there is a synchronized differentiation process of stromal, vascular and epithelial cell types during glomerulogenesis, angiogenesis and tubulogenesis. In the current work, the metanephros of camel foetuses were divided into four stages where kidneys from each stage were processed and immunoassayed, followed by quantitative analysis to determine target protein intensities throughout metanephrogenesis in the camel. This study demonstrated robust expression of α-smooth muscle actin (α-SMA) in the glomerular mesangium, as well as in interlobular and glomerular arterioles during the earlier stages of development. However, in the late stages, α-SMA expression became more localized around the blood capillaries in both the cortex and medulla. Strong expression of CD34 was observed in the immature glomerular and peritubular endothelial cells within the subcapsular zone, as well as in the glomerular, proximal tubular and distal tubular epithelium of stage one foetuses, although its expression gradually diminished with foetal maturation. The expression pattern of osteopontin was prominently observed in the distal convoluted tubules throughout all stages, however, no expression was detected in the proximal tubules, glomeruli and arterioles. E-cadherin was detected in the developing renal tubular epithelial cells but not in the glomeruli. In conclusion, this study reveals the spatiotemporal distribution of key proteins, including α-SMA, CD34, Osteopontin and E-cadherin, which play a crucial role in metanephrogenesis in camel foetuses.
Collapse
Affiliation(s)
- Sameh M Farouk
- Cytology and Histology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, Egypt
| | - Walaa A A Basha
- Anatomy and Embryology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, Egypt
| | - Mahmoud A Emam
- Histology Department, Faculty of Veterinary Medicine, Benha University, Benha, Egypt
| | - Elsayed Metwally
- Cytology and Histology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, Egypt
| |
Collapse
|
2
|
Vinit N, Bessières B, Spaggiari E, Heidet L, Gubler MC, Dreux S, Attie-Bitach T, Blanc T, Ville Y. Pathological and sonographic review of early isolated severe lower urinary tract obstruction and implications for prenatal treatment. ULTRASOUND IN OBSTETRICS & GYNECOLOGY : THE OFFICIAL JOURNAL OF THE INTERNATIONAL SOCIETY OF ULTRASOUND IN OBSTETRICS AND GYNECOLOGY 2022; 59:513-521. [PMID: 34182598 DOI: 10.1002/uog.23718] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 06/05/2021] [Accepted: 06/14/2021] [Indexed: 06/10/2023]
Abstract
OBJECTIVE To identify favorable renal histology in fetuses with early severe lower urinary tract obstruction (LUTO) and determine the best timing and selection criteria for prenatal surgery. METHODS This multicenter, retrospective study included male fetuses with severe LUTO which died before 24 weeks of gestation during the period January 2000 to December 2018. Age-matched controls were used as reference standard for renal histology. Prenatal ultrasound features and fetal serum and/or urine β2microglobulin level were retrieved and kidney histology slides (hematein-eosin-safran and α-smooth-muscle-actin (αSMA) immunostaining) were prepared and reviewed. αSMA-positive staining of the blastema is due to its aberrant differentiation into myofibroblastic cells. Cases were sorted into histopathologic groups (favorable or unfavorable) according to the blastema's morphology and αSMA labeling and the data of these groups were compared. RESULTS Included in the study were 74 fetuses with a median gestational age at outcome of 17 + 6 (range, 13 + 0 to 23 + 5) weeks. Parenchymal organization was preserved in 48% of the kidneys. A blastema was present in 90% of the kidneys, but it was morphologically normal in only 9% and αSMA-negative in only 1% of them. Most (82%) fetuses had an unfavorable prognosis, and 36% of fetuses died ≤ 18 weeks and had severe renal lesions detected on histology (early unfavorable prognosis). A favorable renal prognosis was associated with an earlier gestational age (P = 0.001). Fetuses with LUTO had a significantly lower number of mature glomeruli (P < 0.001) compared with controls. However, there was no significant difference in the number of glomeruli generations between the early-unfavorable-prognosis group (≤ 18 weeks) and the group with a favorable prognosis (P = 0.19). A comparison of prenatal ultrasound features and biochemical markers between groups could not identify any prenatal selection criteria. CONCLUSIONS Before 18 weeks, around 30% of fetuses with severe LUTO still have potential for kidney development. Identification of these cases would enable them to be targeted for prenatal therapy. © 2021 International Society of Ultrasound in Obstetrics and Gynecology.
Collapse
Affiliation(s)
- N Vinit
- Department of Pediatric Surgery and Urology, Necker-Enfants Malades Hospital, APHP, Paris, France
- EA FETUS 7328-PACT, Imagine Institute, Paris University, Paris, France
| | - B Bessières
- EA FETUS 7328-PACT, Imagine Institute, Paris University, Paris, France
- Department of Histology, Embryology and Cytogenetics, Fetal Pathology Unit, Necker-Enfants Malades Hospital, APHP, Paris, France
| | - E Spaggiari
- EA FETUS 7328-PACT, Imagine Institute, Paris University, Paris, France
- Department of Histology, Embryology and Cytogenetics, Fetal Pathology Unit, Necker-Enfants Malades Hospital, APHP, Paris, France
- Department of Obstetrics, Fetal Medicine and Surgery, Necker-Enfants Malades Hospital, APHP, Paris, France
| | - L Heidet
- Department of Pediatric Nephrology, Reference Center for Inherited Renal Disease (MARHEA), Necker-Enfants Malades Hospital, APHP, Paris, France
- INSERM UMR 1163, Laboratory of Inherited Kidney Diseases, Imagine Institute, University of Paris, Paris, France
| | - M-C Gubler
- INSERM UMR 1163, Laboratory of Inherited Kidney Diseases, Imagine Institute, University of Paris, Paris, France
| | - S Dreux
- Department of Biochemistry-Hormonology, Robert Debré Hospital, APHP, Paris, France
| | - T Attie-Bitach
- EA FETUS 7328-PACT, Imagine Institute, Paris University, Paris, France
- Department of Histology, Embryology and Cytogenetics, Fetal Pathology Unit, Necker-Enfants Malades Hospital, APHP, Paris, France
- Université Paris Cité, Paris, France
| | - T Blanc
- Department of Pediatric Surgery and Urology, Necker-Enfants Malades Hospital, APHP, Paris, France
- Université Paris Cité, Paris, France
- INSERM U1151-CNRS UMR 8253, Paris University, Paris, France
| | - Y Ville
- EA FETUS 7328-PACT, Imagine Institute, Paris University, Paris, France
- Department of Obstetrics, Fetal Medicine and Surgery, Necker-Enfants Malades Hospital, APHP, Paris, France
- Université Paris Cité, Paris, France
| |
Collapse
|
3
|
Hudkins KL, Li X, Holland AL, Swaminathan S, Alpers CE. Regression of diabetic nephropathy by treatment with empagliflozin in BTBR ob/ob mice. Nephrol Dial Transplant 2021; 37:847-859. [PMID: 34865099 DOI: 10.1093/ndt/gfab330] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND The SGLT2 inhibitor empagliflozin lowers blood glucose via reduced tubular reabsorption of filtered glucose and is an important new therapy for diabetic nephropathy (DN). This study tested whether treatment with empagliflozin would ameliorate proteinuria and the pathologic alterations of DN including podocyte number and integrity in the leptin deficient BTBR ob/ob mouse model of DN. METHODS Study cohorts included wild type BTBR mice, untreated diabetic BTBR ob/ob mice, and mice treated with empagliflozin for six weeks after development of established DN at 18 weeks of age. RESULTS Hyperglycemia, proteinuria, serum creatinine, accumulation of mesangial matrix and the extent of mesangiolysis were reversed with empagliflozin treatment. Treatment with empagliflozin resulted in increased podocyte number and podocyte density, improvement in the degree of podocyte foot process effacement and parietal epithelial cell activation. SGLT2 inhibition reduced renal oxidative stress, measured by urinary excretion of markers of RNA/DNA damage and in situ demonstration of decreased carbonyl oxidation. There was no discernable difference in accumulations of advanced glycation endproducts by immunohistochemistry. CONCLUSION The structural improvements seen in BTBR ob/ob mice treated with empagliflozin provide insight into potential long term benefits for humans with DN, for whom there is no comparable biopsy information to identify structural changes effected by SGLT2 inhibition. The findings suggest SGLT2 inhibition may ameliorate diabetic nephropathy through glucose lowering-dependent and -independent mechanisms that lead to podocyte restoration and delay or reversal of the disease progress.
Collapse
Affiliation(s)
- Kelly L Hudkins
- Department of Pathology, University of Washington, Seattle WA, USA
| | - Xianwu Li
- Department of Pathology, University of Washington, Seattle WA, USA
| | | | | | - Charles E Alpers
- Department of Pathology, University of Washington, Seattle WA, USA
| |
Collapse
|
4
|
Raming R, Cordasic N, Kirchner P, Ekici AB, Fahlbusch FB, Woelfle J, Hilgers KF, Hartner A, Menendez-Castro C. Neonatal nephron loss during active nephrogenesis results in altered expression of renal developmental genes and markers of kidney injury. Physiol Genomics 2021; 53:509-517. [PMID: 34704838 DOI: 10.1152/physiolgenomics.00059.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Preterm neonates are at a high risk for nephron loss under adverse clinical conditions. Renal damage potentially collides with postnatal nephrogenesis. Recent animal studies suggest that nephron loss within this vulnerable phase leads to renal damage later in life. Nephrogenic pathways are commonly reactivated after kidney injury supporting renal regeneration. We hypothesized that nephron loss during nephrogenesis affects renal development, which, in turn, impairs tissue repair after secondary injury. Neonates prior to 36 wk of gestation show an active nephrogenesis. In rats, nephrogenesis is ongoing until day 10 after birth. Mimicking the situation of severe nephron loss during nephrogenesis, male pups were uninephrectomized at day 1 of life (UNXd1). A second group of males was uninephrectomized at postnatal day 14 (UNXd14), after terminated nephrogenesis. Age-matched controls were sham operated. Three days after uninephrectomy transcriptional changes in the right kidney were analyzed by RNA-sequencing, followed by functional pathway analysis. In UNXd1, 1,182 genes were differentially regulated, but only 143 genes showed a regulation both in UNXd1 and UNXd14. The functional groups "renal development" and "kidney injury" were among the most differentially regulated groups and revealed distinctive alterations. Reduced expression of candidate genes concerning renal development (Bmp7, Gdnf, Pdgf-B, Wt1) and injury (nephrin, podocin, Tgf-β1) were detected. The downregulation of Bmp7 and Gdnf persisted until day 28. In UNXd14, Six2 was upregulated and Pax2 was downregulated. We conclude that nephron loss during nephrogenesis affects renal development and induces a specific regulation of genes that might hinder tissue repair after secondary kidney injury.
Collapse
Affiliation(s)
- Roman Raming
- Department of Pediatrics and Adolescent Medicine, University Hospital of Erlangen, Erlangen, Germany
| | - Nada Cordasic
- Department of Nephrology and Hypertension, University Hospital of Erlangen, Erlangen, Germany
| | - Philipp Kirchner
- Institute of Human Genetics, University Hospital of Erlangen, Erlangen, Germany
| | - Arif B Ekici
- Institute of Human Genetics, University Hospital of Erlangen, Erlangen, Germany
| | - Fabian B Fahlbusch
- Department of Pediatrics and Adolescent Medicine, University Hospital of Erlangen, Erlangen, Germany
| | - Joachim Woelfle
- Department of Pediatrics and Adolescent Medicine, University Hospital of Erlangen, Erlangen, Germany
| | - Karl F Hilgers
- Department of Nephrology and Hypertension, University Hospital of Erlangen, Erlangen, Germany
| | - Andrea Hartner
- Department of Pediatrics and Adolescent Medicine, University Hospital of Erlangen, Erlangen, Germany
| | - Carlos Menendez-Castro
- Department of Pediatrics and Adolescent Medicine, University Hospital of Erlangen, Erlangen, Germany
| |
Collapse
|
5
|
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: 1.0] [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
|
6
|
Abstract
The postnatal kidney is predominantly composed of nephron epithelia with the interstitial components representing a small proportion of the final organ, except in the diseased state. This is in stark contrast to the developing organ, which arises from the mesoderm and comprises an expansive stromal population with distinct regional gene expression. In many organs, the identity and ultimate function of an epithelium is tightly regulated by the surrounding stroma during development. However, although the presence of a renal stromal stem cell population has been demonstrated, the focus has been on understanding the process of nephrogenesis whereas the role of distinct stromal components during kidney morphogenesis is less clear. In this Review, we consider what is known about the role of the stroma of the developing kidney in nephrogenesis, where these cells come from as well as their heterogeneity, and reflect on how this information may improve human kidney organoid models.
Collapse
Affiliation(s)
- Sean B. Wilson
- Murdoch Children's Research Institute, Parkville, VIC 3052, Australia
- Department of Paediatrics, The University of Melbourne, Melbourne, VIC 3000, Australia
| | - Melissa H. Little
- Murdoch Children's Research Institute, Parkville, VIC 3052, Australia
- Department of Paediatrics, The University of Melbourne, Melbourne, VIC 3000, Australia
- Department of Anatomy and Neuroscience, The University of Melbourne, Melbourne, VIC 3000, Australia
| |
Collapse
|
7
|
Geis L, Boudriot FF, Wagner C. Connexin mRNA distribution in adult mouse kidneys. Pflugers Arch 2021; 473:1737-1747. [PMID: 34365513 PMCID: PMC8528753 DOI: 10.1007/s00424-021-02608-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 07/18/2021] [Accepted: 07/22/2021] [Indexed: 11/25/2022]
Abstract
Kidneys are thought to express eight different connexin isoforms (i.e., Cx 26, 30, 32, 37, 40, 43, 45, and 46), which form either hemichannels or gap junctions serving to intercellular communication and functional synchronization. Proper function of connexins has already been shown to be crucial for regulation of renal hemodynamics and renin secretion, and there is also growing evidence for connexins to play a role in pathologic conditions such as renal fibrosis or diabetic nephropathy. Therefore, exact intrarenal localization of the different connexin isoforms gains particular interest. Until now intrarenal expression of connexins has mainly been examined by immunohistochemistry, which in part generated conflicting results depending on antibodies and fixation protocols used. In this work, we used fluorescent RNAscope as an alternative technical approach to localize renal connexin mRNAs in healthy mouse kidneys. Addition of RNAscope probes for cell type specific mRNAs was used to assign connexin mRNA signals to specific cell types. We hereby found Cx26 mRNA strongly expressed in proximal tubules, Cx30 mRNA was selectively detected in the urothelium, and Cx32 mRNA was found in proximal tubules and to a lesser extent also in collecting ducts. Cx37 mRNA was mainly associated with vascular endothelium, Cx40 mRNA was largely found in glomerular mesangial and less in vascular endothelial cells, Cx43 mRNA was sparsely expressed by interstitial cells of all kidney zones, and Cx45 mRNA was predominantly found in smooth muscle cell layers of both blood vessels and ureter as well as in mesangial and interstitial (fibroblastic) cells. Cx46 mRNA could not be detected. In summary our results essentially confirm previous data on connexin expression in the renal vasculature and in glomeruli. In addition, they demonstrate strong connexin gene expression in proximal tubules, and they suggest significant connexin expression in resident tubulointerstitial cells.
Collapse
Affiliation(s)
- Lisa Geis
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany.
| | | | - Charlotte Wagner
- Institute of Physiology, University of Regensburg, Regensburg, Germany
| |
Collapse
|
8
|
Abstract
Complex multicellular life in mammals relies on functional cooperation of different organs for the survival of the whole organism. The kidneys play a critical part in this process through the maintenance of fluid volume and composition homeostasis, which enables other organs to fulfil their tasks. The renal endothelium exhibits phenotypic and molecular traits that distinguish it from endothelia of other organs. Moreover, the adult kidney vasculature comprises diverse populations of mostly quiescent, but not metabolically inactive, endothelial cells (ECs) that reside within the kidney glomeruli, cortex and medulla. Each of these populations supports specific functions, for example, in the filtration of blood plasma, the reabsorption and secretion of water and solutes, and the concentration of urine. Transcriptional profiling of these diverse EC populations suggests they have adapted to local microenvironmental conditions (hypoxia, shear stress, hyperosmolarity), enabling them to support kidney functions. Exposure of ECs to microenvironment-derived angiogenic factors affects their metabolism, and sustains kidney development and homeostasis, whereas EC-derived angiocrine factors preserve distinct microenvironment niches. In the context of kidney disease, renal ECs show alteration in their metabolism and phenotype in response to pathological changes in the local microenvironment, further promoting kidney dysfunction. Understanding the diversity and specialization of kidney ECs could provide new avenues for the treatment of kidney diseases and kidney regeneration.
Collapse
|
9
|
Terasaki M, Brunson JC, Sardi J. Analysis of the three dimensional structure of the kidney glomerulus capillary network. Sci Rep 2020; 10:20334. [PMID: 33230129 PMCID: PMC7683536 DOI: 10.1038/s41598-020-77211-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 11/05/2020] [Indexed: 12/30/2022] Open
Abstract
The capillary network of the kidney glomerulus filters small molecules from the blood. The glomerular 3D structure should help to understand its function, but it is poorly characterized. We therefore devised a new approach in which an automated tape collecting microtome (ATUM) was used to collect 0.5 μm thick serial sections from fixed mouse kidneys. The sections were imaged by scanning electron microscopy at ~ 50 nm/pixel resolution. With this approach, 12 glomeruli were reconstructed at an x–y–z resolution ~ 10 × higher than that of paraffin sections. We found a previously undescribed no-cross zone between afferent and efferent branches on the vascular pole side; connections here would allow blood to exit without being adequately filtered. The capillary diameters throughout the glomerulus appeared to correspond with the amount of blood flow within them. The shortest path (minimum number of branches to travel from afferent to efferent arterioles) is relatively independent of glomerular size and is present primarily on the vascular pole size. This suggests that new branches and longer paths form on the urinary pole side. Network analysis indicates that the glomerular network does not form by repetitive longitudinal splitting of capillaries. Thus the 3D structure of the glomerular capillary network provides useful information with which to understand glomerular function. Other tissue structures in the body may benefit from this new three dimensional approach.
Collapse
Affiliation(s)
- Mark Terasaki
- Department of Cell Biology, University of Connecticut Health Center, Farmington, CT, 06030, USA.
| | - Jason Cory Brunson
- Laboratory for Systems Medicine, University of Florida Health, Gainesville, FL, 32610, USA
| | - Justin Sardi
- Department of Cell Biology, University of Connecticut Health Center, Farmington, CT, 06030, USA
| |
Collapse
|
10
|
Hudkins KL, Wietecha TA, Steegh F, Alpers CE. Beneficial effect on podocyte number in experimental diabetic nephropathy resulting from combined atrasentan and RAAS inhibition therapy. Am J Physiol Renal Physiol 2020; 318:F1295-F1305. [PMID: 32249614 DOI: 10.1152/ajprenal.00498.2019] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Podocyte loss and proteinuria are both key features of human diabetic nephropathy (DN). The leptin-deficient BTBR mouse strain with the ob/ob mutation develops progressive weight gain, type 2 diabetes, and diabetic nephropathy that has many features of advanced human DN, including increased mesangial matrix, mesangiolysis, podocyte loss, and proteinuria. Selective antagonism of the endothelin-1 type A receptor (ETAR) by atrasentan treatment in combination with renin-angiotensin-aldosterone system inhibition with losartan has been shown to have the therapeutic benefit of lowering proteinuria in patients with DN, but the underlying mechanism for this benefit is not well understood. Using a similar therapeutic approach in diabetic BTBR ob/ob mice, this treatment regimen significantly increased glomerular podocyte number compared with diabetic BTBR ob/ob controls and suggested that parietal epithelial cells were a source for podocyte restoration. Atrasentan treatment alone also increased podocyte number but to a lesser degree. Mice treated with atrasentan demonstrated a reduction in proteinuria, matching the functional improvement reported in humans. This is a first demonstration that treatment with the highly selective ETAR antagonist atrasentan can lead to restoration of the diminished podocyte number characteristic of DN in humans and thereby underlies the reduction in proteinuria in patients with diabetes undergoing similar treatment. The benefit of ETAR antagonism in DN extended to a decrease in mesangial matrix as measured by a reduction in accumulations of collagen type IV in both the atrasentan and atrasentan + losartan-treated groups compared with untreated controls.
Collapse
Affiliation(s)
- Kelly L Hudkins
- Department of Pathology, University of Washington, Seattle, Washington
| | - Tomasz A Wietecha
- Department of Pathology, University of Washington, Seattle, Washington
| | - Floor Steegh
- Department of Pathology, University of Washington, Seattle, Washington
| | - Charles E Alpers
- Department of Pathology, University of Washington, Seattle, Washington
| |
Collapse
|
11
|
Buhl EM, Djudjaj S, Klinkhammer BM, Ermert K, Puelles VG, Lindenmeyer MT, Cohen CD, He C, Borkham‐Kamphorst E, Weiskirchen R, Denecke B, Trairatphisan P, Saez‐Rodriguez J, Huber TB, Olson LE, Floege J, Boor P. Dysregulated mesenchymal PDGFR-β drives kidney fibrosis. EMBO Mol Med 2020; 12:e11021. [PMID: 31943786 PMCID: PMC7059015 DOI: 10.15252/emmm.201911021] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 12/08/2019] [Accepted: 12/09/2019] [Indexed: 12/21/2022] Open
Abstract
Kidney fibrosis is characterized by expansion and activation of platelet-derived growth factor receptor-β (PDGFR-β)-positive mesenchymal cells. To study the consequences of PDGFR-β activation, we developed a model of primary renal fibrosis using transgenic mice with PDGFR-β activation specifically in renal mesenchymal cells, driving their pathological proliferation and phenotypic switch toward myofibroblasts. This resulted in progressive mesangioproliferative glomerulonephritis, mesangial sclerosis, and interstitial fibrosis with progressive anemia due to loss of erythropoietin production by fibroblasts. Fibrosis induced secondary tubular epithelial injury at later stages, coinciding with microinflammation, and aggravated the progression of hypertensive and obstructive nephropathy. Inhibition of PDGFR activation reversed fibrosis more effectively in the tubulointerstitium compared to glomeruli. Gene expression signatures in mice with PDGFR-β activation resembled those found in patients. In conclusion, PDGFR-β activation alone is sufficient to induce progressive renal fibrosis and failure, mimicking key aspects of chronic kidney disease in humans. Our data provide direct proof that fibrosis per se can drive chronic organ damage and establish a model of primary fibrosis allowing specific studies targeting fibrosis progression and regression.
Collapse
Affiliation(s)
- Eva M Buhl
- Institute of PathologyRWTH University of AachenAachenGermany
- Division of NephrologyRWTH University of AachenAachenGermany
- Electron Microscopy FacilityRWTH University of AachenAachenGermany
| | - Sonja Djudjaj
- Institute of PathologyRWTH University of AachenAachenGermany
| | | | - Katja Ermert
- Institute of PathologyRWTH University of AachenAachenGermany
| | - Victor G Puelles
- Division of NephrologyRWTH University of AachenAachenGermany
- III. Department of MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
- Department of NephrologyMonash Health, and Center for Inflammatory DiseasesMonash UniversityMelbourneVic.Australia
| | - Maja T Lindenmeyer
- III. Department of MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Clemens D Cohen
- Nephrological CenterMedical Clinic and Policlinic IVUniversity of MunichMunichGermany
| | - Chaoyong He
- Cardiovascular Biology ProgramOklahoma Medical Research FoundationOklahoma CityOKUSA
- State Key Laboratory of Natural MedicinesDepartment of PharmacologyChina Pharmaceutical UniversityNanjingChina
| | - Erawan Borkham‐Kamphorst
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical ChemistryRWTH University of AachenAachenGermany
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical ChemistryRWTH University of AachenAachenGermany
| | - Bernd Denecke
- Interdisciplinary Center for Clinical Research (IZKF)RWTH University of AachenAachenGermany
| | - Panuwat Trairatphisan
- Faculty of MedicineInstitute for Computational BiomedicineHeidelberg University, and Heidelberg University HospitalHeidelbergGermany
| | - Julio Saez‐Rodriguez
- Faculty of MedicineInstitute for Computational BiomedicineHeidelberg University, and Heidelberg University HospitalHeidelbergGermany
| | - Tobias B Huber
- III. Department of MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Lorin E Olson
- Cardiovascular Biology ProgramOklahoma Medical Research FoundationOklahoma CityOKUSA
| | - Jürgen Floege
- Division of NephrologyRWTH University of AachenAachenGermany
| | - Peter Boor
- Institute of PathologyRWTH University of AachenAachenGermany
- Division of NephrologyRWTH University of AachenAachenGermany
| |
Collapse
|
12
|
Abstract
Chronic kidney disease (CKD) is a devastating condition that is reaching epidemic levels owing to the increasing prevalence of diabetes mellitus, hypertension and obesity, as well as ageing of the population. Regardless of the underlying aetiology, CKD is slowly progressive and leads to irreversible nephron loss, end-stage renal disease and/or premature death. Factors that contribute to CKD progression include parenchymal cell loss, chronic inflammation, fibrosis and reduced regenerative capacity of the kidney. Current therapies have limited effectiveness and only delay disease progression, underscoring the need to develop novel therapeutic approaches to either stop or reverse progression. Preclinical studies have identified several approaches that reduce fibrosis in experimental models, including targeting cytokines, transcription factors, developmental and signalling pathways and epigenetic modulators, particularly microRNAs. Some of these nephroprotective strategies are now being tested in clinical trials. Lessons learned from the failure of clinical studies of transforming growth factor β1 (TGFβ1) blockade underscore the need for alternative approaches to CKD therapy, as strategies that target a single pathogenic process may result in unexpected negative effects on simultaneously occurring processes. Additional promising avenues include preventing tubular cell injury and anti-fibrotic therapies that target activated myofibroblasts, the main collagen-producing cells.
Collapse
|
13
|
Minuth W. In Search of Imprints Left by the Impairment of Nephrogenesis. Cells Tissues Organs 2019; 207:69-82. [DOI: 10.1159/000504085] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 05/23/2019] [Indexed: 11/19/2022] Open
Abstract
Clinical aspects dealing with the impairment of nephrogenesis in preterm and low birth weight babies were intensely researched. In this context it was shown that quite different noxae can harm nephron formation, and that the morphological damage in the fetal kidney is rather complex. Some pathological findings show that the impairment leads to changes in developing glomeruli that are restricted to the maturation zone of the outer cortex in the fetal human kidney. Other data show also imprints on the stages of nephron anlage including the niche, the pretubular aggregate, the renal vesicle, and comma- and S-shaped bodies located in the overlying nephrogenic zone of the rodent and human kidneys. During our investigations it was noticed that the stages of nephron anlage in the fetal human kidney during the phase of late gestation have not been described in detail. To contribute, these stages were recorded along with corresponding images. The initial nephron formation in the rodent kidney served as a reference. Finally, the known imprints left by the impairment in both specimens were listed and discussed. In sum, the relatively paucity of data on nephron formation in the fetal human kidney during the late phase of gestation is a call to start with intense research so that concepts for a therapeutic prolongation of nephrogenesis can be designed.
Collapse
|
14
|
Chen Y, Wang N, Yuan Q, Qin J, Hu G, Li Q, Tao L, Xie Y, Peng Z. The Protective Effect of Fluorofenidone against Cyclosporine A-Induced Nephrotoxicity. Kidney Blood Press Res 2019; 44:656-668. [PMID: 31387101 DOI: 10.1159/000500924] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 05/10/2019] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND/AIMS Cyclosporine A (CsA) is an immunosuppressant drug that is used during organ transplants. However, its utility is limited by its nephrotoxic potential. This study aimed to investigate whether fluorofenidone (AKF-PD) could provide protection against CsA-induced nephrotoxicity. METHODS Eighty-five male Sprague-Dawley rats were divided into 5 groups: drug solvent, CsA, CsA with AKF-PD (250, 500 mg/kg/day), and CsA with pirfenidone (PFD, 250 mg/kg/day). Tubulointerstitial injury index, extracellular matrix (ECM) deposition, expression of type I and IV collagen, transforming growth factor (TGF)-β1, platelet-derived growth factor (PDGF), Fas ligand (FASL), cleaved-caspase-3, cleaved-poly(ADP-ribose) polymerase (PARP)-1, and the number of transferase-mediated nick end-labeling (TUNEL)-positive renal tubule cells were determined. In addition, levels of TGF-β1, FASL, cleaved-caspase-3, cleaved-PARP-1, and number of annexin V-positive cells were determined in rat proximal tubular epithelial cells (NRK-52E) treated with CsA (20 μmol/L), AKF-PD (400 μg/mL), PFD (400 μg/mL), and GW788388 (5 μmol/L). RESULTS AKF-PD (250, 500 mg/kg/day) significantly reduced tubulointerstitial injury, ECM deposition, expression of type I and IV collagen, TGF-β1, PDGF, FASL, cleaved-caspase-3, cleaved-PARP-1, and number of TUNEL-positive renal tubule cells in the CsA-treated kidneys. In addition, AKF-PD (400 μg/mL) significantly decreased TGF-β1, FASL, cleaved-caspase-3, and PARP-1 expression in NRK-52E cells and further reduced the number of annexin V-positive cells. CONCLUSION AKF-PD protect kidney from fibrosis and apoptosis in CsA-induced kidney injury.
Collapse
Affiliation(s)
- Yang Chen
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China
| | - Nasui Wang
- Division of Endocrinology and Metabolism, Department of Medicine, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Qiongjing Yuan
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China
| | - Jiao Qin
- Department of Nephrology, Changsha Central Hospital, Changsha, China
| | - Gaoyun Hu
- Department of Medicinal Chemistry, Xiangya School of Pharmacy, Central South University, Changsha, China
| | - Qianbin Li
- Department of Medicinal Chemistry, Xiangya School of Pharmacy, Central South University, Changsha, China
| | - Lijian Tao
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China
| | - Yanyun Xie
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China
| | - Zhangzhe Peng
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China,
| |
Collapse
|
15
|
Liao WQ, Cui SY, Ouyang Q, Mei Y, Cai GY, Fu B, Ma Q, Bai XY, Li QG, Chen XM. Modulation of Macrophage Polarization by Human Glomerular Mesangial Cells in Response to the Stimuli in Renal Microenvironment. J Interferon Cytokine Res 2018; 38:566-577. [PMID: 30523751 DOI: 10.1089/jir.2018.0093] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Mesangial cell (MC) activation and macrophage infiltration are 2 major events closely related with each other in mesangial proliferative glomerulonephritis. In the anti-Thy 1 nephritis model, macrophages mediate the damage and also the expansion of mesangium through secreting various inflammatory factors; however, in glomerular microenvironment how MCs affect macrophage activity in the presence of various stimuli have not yet been understood. In the present study, we found that resting human MCs (HMCs) constitutively expressed chemokine [C-C motif] ligand 2 (CCL-2) and interleukin (IL)-6 and induced M2 polarization of macrophages in the coculture system. HMC proliferation and migration and expression of IL-6, CCL-2, and macrophage colony-stimulating factor in HMCs were enhanced after platelet-derived growth factor (PDGF)-BB stimulation, among which CCL-2 was responsible for inducing the M2 polarization of macrophages. Furthermore, PDGF-BB-stimulated HMCs alleviated the classical activation of macrophages and drove more intensified M2 polarization of macrophages than resting HMCs did. However, lipopolysaccharide and interferon-γ (IFN-γ) stimulated HMCs maintained the M1 phenotype of cocultured macrophages. In conclusion, MCs actively participated in glomerular inflammation through influencing macrophage polarization. The interplay between MCs and infiltrated macrophages is finely modulated by secretory factors such as PDGF-BB and IFN-γ in response to the renal inflammatory microenvironment.
Collapse
Affiliation(s)
- Wu-Qiong Liao
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory for Kidney Diseases, Beijing, China.,School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Shao-Yuan Cui
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory for Kidney Diseases, Beijing, China
| | - Qing Ouyang
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory for Kidney Diseases, Beijing, China
| | - Yan Mei
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory for Kidney Diseases, Beijing, China
| | - Guang-Yan Cai
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory for Kidney Diseases, Beijing, China
| | - Bo Fu
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory for Kidney Diseases, Beijing, China
| | - Qian Ma
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory for Kidney Diseases, Beijing, China
| | - Xue-Yuan Bai
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory for Kidney Diseases, Beijing, China
| | - Qing-Gang Li
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory for Kidney Diseases, Beijing, China
| | - Xiang-Mei Chen
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory for Kidney Diseases, Beijing, China
| |
Collapse
|
16
|
Abstract
Kidney diseases including acute kidney injury and chronic kidney disease are among the largest health issues worldwide. Dialysis and kidney transplantation can replace a significant portion of renal function, however these treatments still have limitations. To overcome these shortcomings, a variety of innovative efforts have been introduced, including cell-based therapies. During the past decades, advances have been made in the stem cell and developmental biology, and tissue engineering. As part of such efforts, studies on renal cell therapy and artificial kidney developments have been conducted, and multiple therapeutic interventions have shown promise in the pre-clinical and clinical settings. More recently, therapeutic cell-secreting secretomes have emerged as a potential alternative to cell-based approaches. This approach involves the use of renotropic factors, such as growth factors and cytokines, that are produced by cells and these factors have shown effectiveness in facilitating kidney function recovery. This review focuses on the renotropic functions of bioactive compounds that provide protective and regenerative effects for kidney tissue repair, based on the available data in the literature.
Collapse
Affiliation(s)
- Kang Su Cho
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
- Department of Urology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - In Kap Ko
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - James J Yoo
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA.
| |
Collapse
|
17
|
Development of new method to enrich human iPSC-derived renal progenitors using cell surface markers. Sci Rep 2018; 8:6375. [PMID: 29686294 PMCID: PMC5913312 DOI: 10.1038/s41598-018-24714-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 04/05/2018] [Indexed: 01/02/2023] Open
Abstract
Cell therapy using renal progenitors differentiated from human embryonic stem cells (hESCs) or induced pluripotent stem cells (hiPSCs) has the potential to significantly reduce the number of patients receiving dialysis therapy. However, the differentiation cultures may contain undifferentiated or undesired cell types that cause unwanted side effects, such as neoplastic formation, when transplanted into a body. Moreover, the hESCs/iPSCs are often genetically modified in order to isolate the derived renal progenitors, hampering clinical applications. To establish an isolation method for renal progenitors induced from hESCs/iPSCs without genetic modifications, we screened antibodies against cell surface markers. We identified the combination of four markers, CD9−CD140a+CD140b+CD271+, which could enrich OSR1+SIX2+ renal progenitors. Furthermore, these isolated cells ameliorated renal injury in an acute kidney injury (AKI) mouse model when used for cell therapy. These cells could contribute to the development of hiPSC-based cell therapy and disease modeling against kidney diseases.
Collapse
|
18
|
França-Silva N, Reis NG, Santos PF, Balbi APC. Diabetes and pregnancy in Wistar rats: renal effects for mothers in the postpartum period. J Dev Orig Health Dis 2018; 9:77-86. [PMID: 28805180 DOI: 10.1017/s2040174417000605] [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] [Indexed: 11/07/2022]
Abstract
In this study, diabetes mellitus (DM) was induced in Wistar rats during pregnancy and maintained in the postpartum period (PP) and we evaluated systolic blood pressure (SBP), glomerular filtration rate (GFR) and renal immunohistochemical and morphometric studies from different groups: G1 (non-pregnant control rats), G2 (non-pregnant diabetic rats), G3 (control mothers) and G4 (diabetic mothers). We found that there were no differences in relation to SBP, but there was a tendency for reduction in GFR from G4 compared with the other groups (G). There was increased total kidney weight/body weight ratio of G4 compared with other G. There were increase in glomerular tuft area in G3 and G4 compared with G1 and G2. G2 and G4 showed even higher percentage of cortical collagen. G3 showed increased glomerular proliferating cells compared with G1 and G2, while in G4 this number was smaller than G3. Cell proliferation was higher in the tubulointerstitial (TBI) compartment from G4. Glomerular and TBI α-smooth muscle actin expression was increased in G4 compared with other G. The glomerular p-p38 expression showed a pattern similar to proliferation cell nuclear antigen, with a reduction of p-p38 in G4 relative to other G. The immunoreactivity of p-JNK was higher in both the glomeruli and TBI compartment in G4 compared with G1, G2 and G3. The DM induced during pregnancy and maintained in the PP resulted in renal structural and functional changes to mothers. In addition, altered mitogen-activated protein kinase expression in association with these changes may play an important role in renal damage observed in the present investigation.
Collapse
Affiliation(s)
- N França-Silva
- 1Area of Physiological Sciences,Federal University of Uberlândia,Uberlândia,Brazil
| | - N G Reis
- 2Physiological Department, Faculty of Medicine,University of São Paulo,Ribeirão Preto,Brazil
| | - P F Santos
- 3Department of Biomedicine,Federal University of Uberlândia,Uberlândia,Brazil
| | - A P C Balbi
- 1Area of Physiological Sciences,Federal University of Uberlândia,Uberlândia,Brazil
| |
Collapse
|
19
|
Patel M, Velagapudi C, Burns H, Doss R, Lee MJ, Mariappan MM, Wagner B, Arar M, Barnes VL, Abboud HE, Barnes JL. Mouse Metanephric Mesenchymal Cell-Derived Angioblasts Undergo Vasculogenesis in Three-Dimensional Culture. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 188:768-784. [PMID: 29269120 DOI: 10.1016/j.ajpath.2017.10.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 09/25/2017] [Accepted: 10/30/2017] [Indexed: 10/18/2022]
Abstract
In vitro models for the investigation of renal vascular development are limited. We previously showed that isolated metanephric mesenchymal (MM) and ureteric bud (UB) cells grown in three-dimensional (3D) matrices formed organoids that consisted of primitive vascular structures surrounding a polarized epithelium. Here, we examined the potential of two principal effectors of vasculogenesis, vascular endothelial growth factor A (VEGF-A), and platelet-derived growth factor B chain (PDGF-BB), to stimulate MM cell differentiation. The results showed that MM cells possess angioblast characteristics by expressing phenotypic markers for endothelial and mesenchymal cells. UB cells synthesize VEGF-A and PDGF-BB proteins and RNA, whereas the MM cells express the respective cognate receptors, supporting their role in directional induction of vasculogenesis. VEGF-A stimulated proliferation of MM cells in monolayer and in 3D sponges but did not affect MM cell migration, organization, or vasculogenesis. However, PDGF-BB stimulated MM cell proliferation, migration, and vasculogenesis in monolayer and organization of the cells into primitive capillary-like assemblies in 3D sea sponge scaffolds in vitro. A role for PDGF-BB in vasculogenesis in the 3D MM/UB co-culture system was validated by direct interference with PDGF-BB or PDGF receptor-β cell interactions to implicate PDGF-BB as a primary effector of MM cell vasculogenesis. Thus, MM cells resemble early renal angioblasts that may provide an ideal platform for the investigation of renal vasculogenesis in vitro.
Collapse
Affiliation(s)
- Mandakini Patel
- Department of Medicine, Division of Nephrology, Audie Murphy Memorial Veterans Administration Hospital, South Texas Veterans Health Care System, San Antonio, Texas
| | - Chakradhar Velagapudi
- Department of Medicine, Division of Nephrology, Audie Murphy Memorial Veterans Administration Hospital, South Texas Veterans Health Care System, San Antonio, Texas
| | | | | | | | | | - Brent Wagner
- Department of Medicine, Division of Nephrology, Audie Murphy Memorial Veterans Administration Hospital, South Texas Veterans Health Care System, San Antonio, Texas; The Medical Research Service, Audie Murphy Memorial Veterans Administration Hospital, South Texas Veterans Health Care System, San Antonio, Texas
| | - Mazen Arar
- Department of Pediatrics, The University of Texas Health Science Center, San Antonio, Texas
| | | | - Hanna E Abboud
- Department of Medicine, Division of Nephrology, Audie Murphy Memorial Veterans Administration Hospital, South Texas Veterans Health Care System, San Antonio, Texas; The Medical Research Service, Audie Murphy Memorial Veterans Administration Hospital, South Texas Veterans Health Care System, San Antonio, Texas
| | - Jeffrey L Barnes
- Department of Medicine, Division of Nephrology, Audie Murphy Memorial Veterans Administration Hospital, South Texas Veterans Health Care System, San Antonio, Texas; Probetex, Inc., San Antonio, Texas; The Medical Research Service, Audie Murphy Memorial Veterans Administration Hospital, South Texas Veterans Health Care System, San Antonio, Texas.
| |
Collapse
|
20
|
Chun J, Chung H, Wang X, Barry R, Taheri ZM, Platnich JM, Ahmed SB, Trpkov K, Hemmelgarn B, Benediktsson H, James MT, Muruve DA. NLRP3 Localizes to the Tubular Epithelium in Human Kidney and Correlates With Outcome in IgA Nephropathy. Sci Rep 2016; 6:24667. [PMID: 27093923 PMCID: PMC4837396 DOI: 10.1038/srep24667] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 04/01/2016] [Indexed: 01/03/2023] Open
Abstract
Nod-like receptor pyrin domain-containing-3 (NLRP3) has been implicated in the pathogenesis of experimental renal injury, yet its characterization in human kidney disease remains largely unexplored. NLRP3 expression was evaluated in human kidney biopsies, primary renal tubular cells (HPTC) and correlated to disease outcomes in patients with IgA nephropathy (IgAN). NLRP3 localized to renal tubules in normal human kidney tissue and to mitochondria within HPTC by immunohistochemistry and immunofluorescence microscopy. Compared to control kidneys, NLRP3 gene expression was increased in biopsies of patients with IgAN. While NLRP3 expression in IgAN was detected in glomeruli, it remained largely confined to the tubular epithelial compartment. In vitro NLRP3 mRNA and protein expression were transiently induced in HPTC by TGF-β1 but subsequently diminished over time as cells lost their epithelial phenotype in a process regulated by transcription and ubiquitin-mediated degradation. Consistent with the in vitro data, low NLRP3 mRNA expression in kidney biopsies was associated with a linear trend of higher risk of composite endpoint of doubling serum creatinine and end stage renal disease in patients with IgAN. Taken together, these data show that NLRP3 is primarily a kidney tubule-expressed protein that decreases in abundance in progressive IgAN.
Collapse
Affiliation(s)
- Justin Chun
- Department of Medicine, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Hyunjae Chung
- Department of Medicine, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Xiangyu Wang
- Department of Medicine, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Rebecca Barry
- Department of Medicine, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Zohreh Mohammad Taheri
- Department of Pathology and Laboratory Medicine, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Jaye M Platnich
- Department of Medicine, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Sofia B Ahmed
- Department of Medicine, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Kiril Trpkov
- Department of Pathology and Laboratory Medicine, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Brenda Hemmelgarn
- Department of Medicine, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Hallgrimur Benediktsson
- Department of Pathology and Laboratory Medicine, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Matthew T James
- Department of Medicine, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Daniel A Muruve
- Department of Medicine, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
21
|
França-Silva N, Oliveira NDG, Balbi APC. Morphofunctional renal alterations in rats induced by intrauterine hyperglycemic environment. Arch Med Sci 2016; 12:243-51. [PMID: 27186167 PMCID: PMC4848350 DOI: 10.5114/aoms.2015.48220] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Accepted: 11/28/2014] [Indexed: 01/28/2023] Open
Abstract
INTRODUCTION The renal development of rats begins in intrauterine life, finishing by 15 days after birth. Diabetes and other diseases during pregnancy can cause systemic changes in the offspring. We evaluated the structural and functional renal alterations of the offspring from diabetic mothers. MATERIAL AND METHODS Pregnant rats were separated and 1, 7, 30 and 90 days-old (DO) pups were divided into groups according to the treatment that the mothers received: G1: control, G2: untreated diabetic and G3: insulin-treated diabetic. The kidneys from offspring at 1, 7 and 30 DO were removed for immunohistochemical and histological studies. Furthermore, blood and urine samples were collected from animals at 30 DO to determine the glomerular filtration rate (GFR) by creatinine clearance, and the animals at 90 DO were subjected to blood pressure measurement by plethysmography. RESULTS Our results show an increase of PCNA(+) glomerular cells at 7 DO and a reduction in 30 DO animals as well as increased α-smooth muscle actin (α-SMA) tubulointerstitial expression at 1 and 7 DO in animals from G2, when compared with controls. The adult offspring from G2 showed reduced GFR and increased blood pressure. CONCLUSIONS Maternal diabetes may have induced programming of renal damage in offspring of hyperglycemic mothers, which may have contributed to the impairment of renal function.
Collapse
Affiliation(s)
- Nathane França-Silva
- Department of Physiological Sciences, Federal University of Uberlândia, Uberlândia, Brazil
| | | | - Ana Paula Coelho Balbi
- Department of Physiological Sciences, Federal University of Uberlândia, Uberlândia, Brazil
| |
Collapse
|
22
|
Noskovičová N, Petřek M, Eickelberg O, Heinzelmann K. Platelet-Derived Growth Factor Signaling in the Lung. From Lung Development and Disease to Clinical Studies. Am J Respir Cell Mol Biol 2015; 52:263-84. [DOI: 10.1165/rcmb.2014-0294tr] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
23
|
Kok HM, Falke LL, Goldschmeding R, Nguyen TQ. Targeting CTGF, EGF and PDGF pathways to prevent progression of kidney disease. Nat Rev Nephrol 2014; 10:700-11. [PMID: 25311535 DOI: 10.1038/nrneph.2014.184] [Citation(s) in RCA: 128] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Chronic kidney disease (CKD) is a major health and economic burden with a rising incidence. During progression of CKD, the sustained release of proinflammatory and profibrotic cytokines and growth factors leads to an excessive accumulation of extracellular matrix. Transforming growth factor β (TGF-β) and angiotensin II are considered to be the two main driving forces in fibrotic development. Blockade of the renin-angiotensin-aldosterone system has become the mainstay therapy for preservation of kidney function, but this treatment is not sufficient to prevent progression of fibrosis and CKD. Several factors that induce fibrosis have been identified, not only by TGF-β-dependent mechanisms, but also by TGF-β-independent mechanisms. Among these factors are the (partially) TGF-β-independent profibrotic pathways involving connective tissue growth factor, epidermal growth factor and platelet-derived growth factor and their receptors. In this Review, we discuss the specific roles of these pathways, their interactions and preclinical evidence supporting their qualification as additional targets for novel antifibrotic therapies.
Collapse
Affiliation(s)
- Helena M Kok
- Department of Pathology, H04.312, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, Netherlands
| | - Lucas L Falke
- Department of Pathology, H04.312, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, Netherlands
| | - Roel Goldschmeding
- Department of Pathology, H04.312, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, Netherlands
| | - Tri Q Nguyen
- Department of Pathology, H04.312, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, Netherlands
| |
Collapse
|
24
|
Pichaiwong W, Hudkins KL, Wietecha T, Nguyen TQ, Tachaudomdach C, Li W, Askari B, Kobayashi T, O'Brien KD, Pippin JW, Shankland SJ, Alpers CE. Reversibility of structural and functional damage in a model of advanced diabetic nephropathy. J Am Soc Nephrol 2013; 24:1088-102. [PMID: 23641056 DOI: 10.1681/asn.2012050445] [Citation(s) in RCA: 133] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
The reversibility of diabetic nephropathy remains controversial. Here, we tested whether replacing leptin could reverse the advanced diabetic nephropathy modeled by the leptin-deficient BTBR ob/ob mouse. Leptin replacement, but not inhibition of the renin-angiotensin-aldosterone system (RAAS), resulted in near-complete reversal of both structural (mesangial matrix expansion, mesangiolysis, basement membrane thickening, podocyte loss) and functional (proteinuria, accumulation of reactive oxygen species) measures of advanced diabetic nephropathy. Immunohistochemical labeling with the podocyte markers Wilms tumor 1 and p57 identified parietal epithelial cells as a possible source of regenerating podocytes. Thus, the leptin-deficient BTBR ob/ob mouse provides a model of advanced but reversible diabetic nephropathy for further study. These results also suggest that restoration of lost podocytes is possible but is not induced by RAAS inhibition, possibly explaining the limited efficacy of RAAS inhibitors in promoting repair of diabetic nephropathy.
Collapse
|
25
|
Bautista E, Arcos M, Jimenez-Alvarez L, García-Sancho MC, Vázquez ME, Peña E, Higuera A, Ramírez G, Fernández-Plata R, Cruz-Lagunas A, García-Moreno SA, Urrea F, Ramírez R, Correa-Rotter R, Pérez-Padilla JR, Zúñiga J. Angiogenic and inflammatory markers in acute respiratory distress syndrome and renal injury associated to A/H1N1 virus infection. Exp Mol Pathol 2013; 94:486-92. [PMID: 23542734 DOI: 10.1016/j.yexmp.2013.03.007] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Accepted: 03/20/2013] [Indexed: 12/11/2022]
Abstract
Acute kidney injury (AKI) is often associated to acute respiratory distress syndrome (ARDS) due to influenza A/H1N1 virus infection. The profile of angiogenic and inflammatory factors in ARDS patients may be relevant for AKI. We analyzed the serum levels of several angiogenic factors, cytokines, and chemokines in 32 patients with A/H1N1 virus infection (17 with ARDS/AKI and 15 ARDS patients who did not developed AKI) and in 18 healthy controls. Significantly higher levels of VEGF, MCP-1, IL-6, IL-8 and IP-10 in ARDS/AKI patients were detected. Adjusting by confusing variables, levels of MCP-1 ≥150 pg/mL (OR=12.0, p=0.04) and VEGF ≥225 pg/mL (OR=6.4, p=0.03) were associated with the development of AKI in ARDS patients. Higher levels of MCP-1 and IP-10 were significantly associated with a higher risk of death in patients with ARDS (hazard ratio (HR)=10.0, p=0.02; HR=25.5, p=0.03, respectively) even taking into account AKI. Patients with influenza A/H1N1 infection and ARDS/AKI have an over-production of MCP-1, VEGF and IP-10 possibly contributing to kidney injury and are associated to a higher risk of death.
Collapse
Affiliation(s)
- Edgar Bautista
- Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Tlalpan 4502, Tlalpan, 14080, Mexico City, Mexico.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Sarrab RM, Lennon R, Ni L, Wherlock MD, Welsh GI, Saleem MA. Establishment of conditionally immortalized human glomerular mesangial cells in culture, with unique migratory properties. Am J Physiol Renal Physiol 2011; 301:F1131-8. [PMID: 21653636 DOI: 10.1152/ajprenal.00589.2010] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The aim of this study was to establish an immortalized human mesangial cell line similar to mesangial cells in vivo for use as a tool for understanding glomerular cell function. Mesangial cells were isolated from glomerular outgrowths from a normal human kidney, then retrovirally transfected with a temperature-sensitive SV40T antigen+human telomerase (hTERT). Mesangial cells exhibited features of compact cells with small bodies in a confluent monolayer at 33°C, but the cell shape changed to flat and stellate after 5 days in growth-restrictive conditions (37°C). Western blot and immunofluorescence analysis showed that podocyte markers (nephrin, CD2AP, podocin, Wilms' tumor-1) and an endothelial-specific molecule (VE-cadherin) were not detectable in this cell line, whereas markers characteristic of mesangial cells (α-SMA, fibronectin, and PDGFβ-R) were strongly expressed. In migration assays, a significant reduction in wound surface was observed in podocyte and endothelial cells as soon as 12 h (75 and 62%, respectively) and complete wound closure after 24 h. In contrast, no significant change was observed in mesangial cells after 12 h, and even after 48 h the wounds were not completely closed. Until now, conditionally immortalized podocyte and endothelial cell lines derived from mice and humans have been described, and this has greatly boosted research on glomerular physiology and pathology. We have established the first conditionally immortalized human glomerular mesangial cell line, which will be an important adjunct in studies of representative glomerular cells, as well as in coculture studies. Unexpectedly, mesangial cells' ability to migrate seems to be slower than for other glomerular cells, suggesting this line will demonstrate functional properties distinct from previously available mesangial cell cultures. This conditionally immortalized human mesangial cell line represents a new tool for the study of human mesangial cell biology in vitro.
Collapse
Affiliation(s)
- Ramadan M Sarrab
- Children's Renal Unit and Academic Renal Unit, University of Bristol, Southmead Hospital, Bristol, United Kingdom
| | | | | | | | | | | |
Collapse
|
27
|
Huang HP, Hong CL, Kao CY, Lin SW, Lin SR, Wu HL, Shi GY, You LR, Wu CL, Yu IS. Gene targeting and expression analysis of mouse Tem1/endosialin using a lacZ reporter. Gene Expr Patterns 2011; 11:316-26. [PMID: 21402174 DOI: 10.1016/j.gep.2011.03.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2010] [Revised: 02/27/2011] [Accepted: 03/05/2011] [Indexed: 01/25/2023]
Abstract
TEM1 (endosialin) expression is increased in the stroma and tumor vasculature of several common human cancers. The exact physiological role of TEM1 is still unknown since Tem1-deficient mice are viable and show only a lower rate of abdominal site-specific tumor invasion in tumor transplantation experiments. Previous studies have reported Tem1 expression in mouse embryos and adults, but did not determine the timing or location of the earliest expression, and did not examine all organ systems. Using the highly sensitive Bluo-Gal staining method for detecting temporal and spatial Tem1-lacZ activity in lacZ knock-in (+/lacZ) mice, we found that Tem1 gene expression was initially detectable in the dorsal aortic wall, the heart, the umbilical vessels, the first branchial arch, and the cephalic mesenchyme at E9.5. From E10.5 to E14.5, Tem1 gene expression was additionally seen mainly in the genital tubercle, the mesonephros, the whisker follicles, the mesenchymal tissues around the eye, and the lung. Remarkably, the kidney expressed abundant Tem1-lacZ starting from E16.5. Postnatally, Tem1 expression decreased in most organs but elevated expression persisted in the renal glomerulus and the uterus, where the expression pattern varied at different estrous cycle stages. Co-localization studies indicated that most vimentin-positive cells co-expressed Tem1-lacZ, while a large portion of CD31- or desmin-positive cells were also positive for Tem1-lacZ. Taken together, our observations suggest that Tem1 is expressed throughout embryonic and adult development in several types of mesenchymal cells closely related to blood vessels.
Collapse
Affiliation(s)
- Hsiang-Po Huang
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Hudkins KL, Pichaiwong W, Wietecha T, Kowalewska J, Banas MC, Spencer MW, Mühlfeld A, Koelling M, Pippin JW, Shankland SJ, Askari B, Rabaglia ME, Keller MP, Attie AD, Alpers CE. BTBR Ob/Ob mutant mice model progressive diabetic nephropathy. J Am Soc Nephrol 2010; 21:1533-42. [PMID: 20634301 DOI: 10.1681/asn.2009121290] [Citation(s) in RCA: 180] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
There remains a need for robust mouse models of diabetic nephropathy (DN) that mimic key features of advanced human DN. The recently developed mouse strain BTBR with the ob/ob leptin-deficiency mutation develops severe type 2 diabetes, hypercholesterolemia, elevated triglycerides, and insulin resistance, but the renal phenotype has not been characterized. Here, we show that these obese, diabetic mice rapidly develop morphologic renal lesions characteristic of both early and advanced human DN. BTBR ob/ob mice developed progressive proteinuria beginning at 4 weeks. Glomerular hypertrophy and accumulation of mesangial matrix, characteristic of early DN, were present by 8 weeks, and glomerular lesions similar to those of advanced human DN were present by 20 weeks. By 22 weeks, we observed an approximately 20% increase in basement membrane thickness and a >50% increase in mesangial matrix. Diffuse mesangial sclerosis (focally approaching nodular glomerulosclerosis), focal arteriolar hyalinosis, mesangiolysis, and focal mild interstitial fibrosis were present. Loss of podocytes was present early and persisted. In summary, BTBR ob/ob mice develop a constellation of abnormalities that closely resemble advanced human DN more rapidly than most other murine models, making this strain particularly attractive for testing therapeutic interventions.
Collapse
Affiliation(s)
- Kelly L Hudkins
- Department of Pathology, University of Washington, Seattle, Washington, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Ito Y, Goldschmeding R, Kasuga H, Claessen N, Nakayama M, Yuzawa Y, Sawai A, Matsuo S, Weening JJ, Aten J. Expression patterns of connective tissue growth factor and of TGF-beta isoforms during glomerular injury recapitulate glomerulogenesis. Am J Physiol Renal Physiol 2010; 299:F545-58. [PMID: 20576680 DOI: 10.1152/ajprenal.00120.2009] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Transforming growth factor (TGF)-beta(1), -beta(2), and -beta(3) are involved in control of wound repair and development of fibrosis. Connective tissue growth factor (CTGF) expression is stimulated by all TGF-beta isoforms and is abundant in glomerulosclerosis and other fibrotic disorders. CTGF is hypothesized to mediate profibrotic effects of TGF-beta(1) or to facilitate interaction of TGF-beta(1) with its receptor, but its interactions with TGF-beta isoforms in nonpathological conditions are unexplored so far. Tissue repair and remodeling may recapitulate gene transcription at play in organogenesis. To further delineate the relationship between CTGF and TGF-beta, we compared expression patterns of CTGF and TGF-beta isoforms in rat and human glomerulogenesis and in various human glomerulopathies. CTGF mRNA was present in the immediate precursors of glomerular visceral and parietal epithelial cells in the comma- and S-shaped stages, but not in earlier stages of nephron development. During the capillary loop and maturing glomerular stages and simultaneous with the presence of TGF-beta(1), -beta(2), and -beta(3) protein, CTGF mRNA expression was maximal and present only in differentiating glomerular epithelial cells. CTGF protein was also present on precursors of mesangium and glomerular endothelium, suggesting possible paracrine interaction. Concomitant with the presence of TGF-beta(2) and -beta(3) protein, and in the absence of TGF-beta(1), CTGF mRNA and protein expression was restricted to podocytes in normal adult glomeruli. However, TGF-beta(1) and CTGF were again coexpressed, often with TGF-beta(2) and -beta(3), in particular in podocytes in proliferative glomerulonephritis and also in mesangial cells in diabetic nephropathy and IgA nephropathy (IgA NP). Coordinated expression of TGF-beta isoforms and of CTGF may be involved in normal glomerulogenesis and possibly in maintenance of glomerular structure and function at adult age. Prolonged overexpression of TGF-beta(1) and CTGF is associated with development of severe glomerulonephritis and glomerulosclerosis.
Collapse
Affiliation(s)
- Yasuhiko Ito
- Department of Pathology, Academic Medical Center, University of Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Abrass CK, Hansen KM, Patton BL. Laminin alpha4-null mutant mice develop chronic kidney disease with persistent overexpression of platelet-derived growth factor. THE AMERICAN JOURNAL OF PATHOLOGY 2009; 176:839-49. [PMID: 20035058 DOI: 10.2353/ajpath.2010.090570] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Each extracellular matrix compartment in the kidney has a unique composition, with regional specificity in the expression of various laminin isoforms. Although null mutations in the majority of laminin chains lead to specific developmental abnormalities in the kidney, Lama4-/- mice have progressive glomerular and tubulointerstitial fibrosis. These mice have a significant increase in expression of platelet-derived growth factor (PDGF)-BB, PDGF-DD, and PDGF receptor beta in association with immature glomerular and peritubular capillaries. In addition, mesangial cell exposure to alpha4-containing laminins, but not other isoforms, results in down-regulation of PDGF receptor mRNA and protein, suggesting a direct effect of LN411/LN421 on vessel maturation. Given the known role of overexpression of PDGF-BB and PDGF-DD on glomerular and tubulointerstitial fibrosis, these data suggest that failure of laminin alpha4-mediated down-regulation of PDGF activity contributes to the progressive renal lesions in this animal model. Given the recent demonstration that individuals with laminin alpha4 mutations develop cardiomyopathy, these findings may be relevant to kidney disease in humans.
Collapse
Affiliation(s)
- Christine K Abrass
- Primary and Specialty Care Medicine, Department of Veterans Affairs Puget Sound Health Care System, Seattle, Washington, USA.
| | | | | |
Collapse
|
31
|
Abrahamson DR. Development of kidney glomerular endothelial cells and their role in basement membrane assembly. Organogenesis 2009; 5:275-87. [PMID: 19568349 PMCID: PMC2659369 DOI: 10.4161/org.7577] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2008] [Accepted: 12/03/2008] [Indexed: 01/07/2023] Open
Abstract
Data showing that the embryonic day 12 (E12) mouse kidney contains its own pool of endothelial progenitor cells is presented. Mechanisms that regulate metanephric endothelial recruitment and differentiation, including the hypoxia-inducible transcription factors and vascular endothelial growth factor/vascular endothelial growth factor receptor signaling system, are also discussed. Finally, evidence that glomerular endothelial cells contribute importantly to assembly of the glomerular basement membrane (GBM), especially the laminin component, is reviewed. Together, this forum offers insights on blood vessel development in general, and formation of the glomerular capillary in particular, which inarguably is among the most unique vascular structures in the body.
Collapse
Affiliation(s)
- Dale R Abrahamson
- Department of Anatomy and Cell Biology; University of Kansas Medical Center; Kansas City, Kansas USA
| |
Collapse
|
32
|
Quaggin SE, Kreidberg JA. Development of the renal glomerulus: good neighbors and good fences. Development 2008; 135:609-20. [PMID: 18184729 DOI: 10.1242/dev.001081] [Citation(s) in RCA: 178] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The glomerulus of the mammalian kidney is an intricate structure that contains an unusual filtration barrier that retains higher molecular weight proteins and blood cells in the circulation. Recent studies have changed our conception of the glomerulus from a relatively static structure to a dynamic one, whose integrity depends on signaling between the three major cell lineages: podocytes, endothelial and mesangial cells. Research into the signaling pathways that control glomerular development and then maintain glomerular integrity and function has recently identified several genes, such as the nephrin and Wilms' tumor 1 genes, that are mutated in human kidney disease.
Collapse
Affiliation(s)
- Susan E Quaggin
- Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Ontario M5G 1X5, Canada
| | | |
Collapse
|
33
|
Floege J, Eitner F, Alpers CE. A New Look at Platelet-Derived Growth Factor in Renal Disease. J Am Soc Nephrol 2007; 19:12-23. [DOI: 10.1681/asn.2007050532] [Citation(s) in RCA: 235] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
|
34
|
Wagner B, Ricono JM, Gorin Y, Block K, Arar M, Riley D, Choudhury GG, Abboud HE. Mitogenic signaling via platelet-derived growth factor beta in metanephric mesenchymal cells. J Am Soc Nephrol 2007; 18:2903-11. [PMID: 17942966 DOI: 10.1681/asn.2006111229] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Mice deficient in either platelet-derived growth factor (PDGF) B chain or PDGF receptor (PDGFR) beta lack mesangial cells. PDGF stimulates proliferation and migration of metanephric mesenchymal cells, from which mesangial cells are derived. Binding of PDGF to PDGFR-beta induces autophosphorylation at specific tyrosine residues and activates various effector proteins, including phosphatidylinositol-3-kinase (PI3-K). This study explored the role of PI 3-K and reactive oxygen species (ROS) in PDGF-mediated signaling using cells established from wild-type and PDGFR-beta -/- metanephric blastemas at 11.5 days post-conception. PDGF-induced effects that were dependent on PI3-K activation were determined using PDGFR-beta -/- cells made to express "add-back" mutant PDGFR-beta capable of binding PI3-K. We found that PDGF is mitogenic for mesenchymal cells expressing PDGFR-beta, and PI3-K is an important regulator of PDGF-induced DNA synthesis. Activation of ERK1/2 is partially dependent on PI3-K, and both the PI3-K and MEK-ERK1/2 pathways contribute to PI3-K-dependent mitogenesis. In addition, PDGF-induced DNA synthesis in wild-type cells was found to be dependent on ROS that are generated downstream of PI3-K activation. Using antisense oligonucleotides and small interfering RNA, we determined that the NAD(P)H oxidase Nox4 produces these ROS that activate Akt and the MEK-ERK1/2 mitogenic cascade. In conclusion, the present study demonstrates Nox4 involvement in PDGF-induced DNA synthesis in metanephric mesenchymal cells and provides the first evidence that PDGF-induced PI3-K activity enhances production of ROS by Nox4.
Collapse
Affiliation(s)
- Brent Wagner
- Division of Nephrology, Department of Medicine, University of Texas Health Science Center, San Antonio, Texas 78229-3900, USA.
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Bertelli E, Regoli M, Fonzi L, Occhini R, Mannucci S, Ermini L, Toti P. Nestin expression in adult and developing human kidney. J Histochem Cytochem 2007; 55:411-21. [PMID: 17210924 DOI: 10.1369/jhc.6a7058.2007] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Nestin is considered a marker of neurogenic and myogenic precursor cells. Its arrangement is regulated by cyclin-dependent kinase 5 (CDK5), which is expressed in murine podocytes. We investigated nestin expression in human adult and fetal kidney as well as CDK5 presence in adult human podocytes. Confocal microscopy demonstrated that adult glomeruli display nestin immunoreactivity in vimentin-expressing cells with the podocyte morphology and not in cells bearing the endothelial marker CD31. Glomerular nestin-positive cells were CDK5 immunoreactive as well. Western blotting of the intermediate filament-enriched cytoskeletal fraction and coimmunoprecipitation of nestin with anti-CDK5 antibodies confirmed these results. Nestin was also detected in developing glomeruli within immature podocytes and a few other cells. Confocal microscopy of experiments conducted with antibodies against nestin and endothelial markers demonstrated that endothelial cells belonging to capillaries invading the lower cleft of S-shaped bodies and the immature glomeruli were nestin immunoreactive. Similar experiments carried out with antibodies raised against nestin and alpha-smooth muscle actin showed that the first mesangial cells that populate the developing glomeruli expressed nestin. In conclusion, nestin is expressed in the human kidney from the first steps of glomerulogenesis within podocytes, mesangial, and endothelial cells. This expression, restricted to podocytes in mature glomeruli, appears associated with CDK5.
Collapse
Affiliation(s)
- Eugenio Bertelli
- Dept. of Pharmacology Giorgio Segre, Section of Anatomy, University of Siena, Via Aldo Moro 4, I-53100 Siena, Italy.
| | | | | | | | | | | | | |
Collapse
|
36
|
Hassanain HH, Gregg D, Marcelo ML, Zweier JL, Souza HP, Selvakumar B, Ma Q, Moustafa-Bayoumi M, Binkley PF, Flavahan NA, Morris M, Dong C, Goldschmidt-Clermont PJ. Hypertension caused by transgenic overexpression of Rac1. Antioxid Redox Signal 2007; 9:91-100. [PMID: 17115888 DOI: 10.1089/ars.2007.9.91] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Reactive oxygen species, including superoxide, are important mediators of the pathophysiology of hypertension. In the vasculature, superoxide antagonizes nitric oxide (NO*), resulting in increased vascular tone. The GTP binding protein Rac regulates a wide variety of cellular functions, including the activation of NADPH oxidase, the major source of O2*-in the blood vessel wall. An hypothesis is that Rac1 may act as an important regulator of vascular O2*- production, contributing to the balance between O2*- and NO* and maintaining consequent homeostasis of blood pressure. To alter the activity of vascular NADPH oxidase, the authors developed a transgenic animal model that overexpresses the human cDNA of the constitutively active mutant of Rac1 (RacCA) in smooth muscle cells using the smooth muscle +/--actin promoter. The RacCA transgenic had excessive amounts of O2*- in the vessel wall that, which led to heightened production of peroxynitrite, as detected by increased protein nitration and reduced NO* levels. RacCA mice developed moderate hypertension, which was corrected by N-acetyl-L-cysteine (NAC). RacCA transgenic mice also developed left ventricular hypertrophy as a secondary effect of pressure overload. The data suggest that Rac1 is a critical regulator of the redox state of blood vessels and homeostasis of blood pressure.
Collapse
Affiliation(s)
- Hamdy H Hassanain
- Department of Anesthesiology, Dorothy M. Davis Heart and Lung Institute, The Ohio State University, Columbus, Ohio 43210, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Alvarez RH, Kantarjian HM, Cortes JE. Biology of platelet-derived growth factor and its involvement in disease. Mayo Clin Proc 2006; 81:1241-57. [PMID: 16970222 DOI: 10.4065/81.9.1241] [Citation(s) in RCA: 245] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Platelet-derived growth factor (PDGF) is mainly believed to be an important mitogen for connective tissue, especially for fibroblasts that serve in wound healing. However, PDGF also has important roles during embryonal development, and its overexpression has been linked to different types of fibrotic disorders and malignancies. Platelet-derived growth factor is synthesized by many different cell types, and its expression is broad. Its synthesis is in response to external stimuli, such as exposure to low oxygen tension, thrombin, or stimulation by other cytokines and growth factors. In addition, PDGF may function in autocrine stimulation of tumor cells, regulation of interstitial fluid pressure, and angiogenesis. Recently, several drugs were developed that are potent inhibitors of the tyrosine kinase activity of PDGF receptors. Thus, it is important to understand the physiology of PDGF and its receptors and the role of PDGF in different diseases. This review summarizes the physiologic activity of PDGF, the expression of PDGF during embryonal development, and the roles of PDGF expression in nonmalignant disease and in different tumors.
Collapse
Affiliation(s)
- Ricardo H Alvarez
- Department of Internal Medicine, The University of Texas at Houston Medical School, Houston, USA
| | | | | |
Collapse
|
38
|
Eng E, Holgren C, Hubchak S, Naaz P, Schnaper HW. Hypoxia regulates PDGF-B interactions between glomerular capillary endothelial and mesangial cells. Kidney Int 2005; 68:695-703. [PMID: 16014047 DOI: 10.1111/j.1523-1755.2005.00448.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
BACKGROUND Platelet-derived growth factor (PDGF)-B regulates mesangial cell and vessel development during embryogenesis, and contributes to the pathogenesis of adult renal and vascular diseases. Endothelial cell PDGF-B exerts paracrine effects on mesangial cells, but its regulation is not well defined. We examined the impact of hypoxia on PDGF-B-mediated interactions between glomerular endothelial and mesangial cells, a condition of potential relevance in developing, and diseased adult, kidneys. METHODS Glomerular endothelial or mesangial cells were subjected to hypoxia and responses compared to normoxic cells. Endothelial PDGF-B was studied by Northern and Western analysis. Mesangial proliferative responses to PDGF-B were assessed by (3)H-thymidine incorporation, and migration by a modified Boyden chamber assay. Hypoxia-induced changes in receptor specific binding capacity were studied by saturation binding assays. RESULTS Hypoxia stimulated increases in endothelial PDGF-B mRNA and protein. In normoxic mesangial cells, PDGF-B stimulated dose-dependent proliferation, but the proliferative response of hypoxic cells was two to three times greater. Exogenous PDGF-B also caused prompter migration in hypoxic mesangial cells. Mesangial cells were treated with endothelial cell-conditioned medium. More cells migrated when hypoxic cells were stimulated with hypoxic conditioned medium, than when normoxic cells were stimulated with normoxic conditioned medium. Preincubating conditioned medium with PDGF-B neutralizing antibody greatly decreased the chemoattractant activity. Binding studies demonstrated increased specific binding capacity in hypoxic cells. CONCLUSION Hypoxia enhances PDGF-B paracrine interactions between glomerular endothelial and mesangial cells. These hypoxia-regulated interactions may be important during glomerulogenesis in fetal life and during the pathogenesis of adult glomerular disease.
Collapse
Affiliation(s)
- Eudora Eng
- Division of Nephrology and Hypertension, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA.
| | | | | | | | | |
Collapse
|
39
|
Abstract
Platelet-derived growth factor (PDGF) isoforms play a major role in stimulating the replication, survival, and migration of myofibroblasts during the pathogenesis of fibrotic diseases. During fibrogenesis, PDGF is secreted by a variety of cell types as a response to injury, and many pro-inflammatory cytokines mediate their mitogenic effects via the autocrine release of PDGF. PDGF action is determined by the relative expression of PDGF alpha-receptors (PDGFRalpha) and beta-receptors (PDGFRbeta) on the surface of myofibroblasts. These receptors are induced during fibrogenesis, thereby amplifying biological responses to PDGF isoforms. PDGF action is also modulated by extracellular binding proteins and matrix molecules. This review summarizes the literature on the role of PDGF and its receptors in the development of fibrosis in a variety of organ systems, including lung, liver, kidney, and skin.
Collapse
Affiliation(s)
- James C Bonner
- National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA.
| |
Collapse
|
40
|
Schrijvers BF, De Vriese AS, Flyvbjerg A. From hyperglycemia to diabetic kidney disease: the role of metabolic, hemodynamic, intracellular factors and growth factors/cytokines. Endocr Rev 2004; 25:971-1010. [PMID: 15583025 DOI: 10.1210/er.2003-0018] [Citation(s) in RCA: 237] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
At present, diabetic kidney disease affects about 15-25% of type 1 and 30-40% of type 2 diabetic patients. Several decades of extensive research has elucidated various pathways to be implicated in the development of diabetic kidney disease. This review focuses on the metabolic factors beyond blood glucose that are involved in the pathogenesis of diabetic kidney disease, i.e., advanced glycation end-products and the aldose reductase system. Furthermore, the contribution of hemodynamic factors, the renin-angiotensin system, the endothelin system, and the nitric oxide system, as well as the prominent role of the intracellular signaling molecule protein kinase C are discussed. Finally, the respective roles of TGF-beta, GH and IGFs, vascular endothelial growth factor, and platelet-derived growth factor are covered. The complex interplay between these different pathways will be highlighted. A brief introduction to each system and description of its expression in the normal kidney is followed by in vitro, experimental, and clinical evidence addressing the role of the system in diabetic kidney disease. Finally, well-known and potential therapeutic strategies targeting each system are discussed, ending with an overall conclusion.
Collapse
Affiliation(s)
- Bieke F Schrijvers
- Medical Department M/Medical Research Laboratories, Clinical Institute, Aarhus University Hospital, Nørrebrogade 44, DK-8000 Aarhus C, Denmark
| | | | | |
Collapse
|
41
|
Bjarnegård M, Enge M, Norlin J, Gustafsdottir S, Fredriksson S, Abramsson A, Takemoto M, Gustafsson E, Fässler R, Betsholtz C. Endothelium-specific ablation of PDGFB leads to pericyte loss and glomerular, cardiac and placental abnormalities. Development 2004; 131:1847-57. [PMID: 15084468 DOI: 10.1242/dev.01080] [Citation(s) in RCA: 258] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Platelet-derived growth factor-B (PDGFB) is necessary for normal cardiovascular development, but the relative importance of different cellular sources of PDGFB has not been established. Using Cre-lox techniques, we show here that genetic ablation of Pdgfb in endothelial cells leads to impaired recruitment of pericytes to blood vessels. The endothelium-restricted Pdgfb knockout mutants also developed organ defects including cardiac, placental and renal abnormalities. These defects were similar to those observed in Pdgfb null mice. However, in marked contrast to the embryonic lethality of Pdgfb null mutants, the endothelium-specific mutants survived into adulthood with persistent pathological changes, including brain microhemorrhages, focal astrogliosis, and kidney glomerulus abnormalities. This spectrum of pathological changes is reminiscent of diabetic microangiopathy, suggesting that the endothelium-restricted Pdgfb knockouts may serve as models for some of the pathogenic events of vascular complications to diabetes.
Collapse
Affiliation(s)
- Mattias Bjarnegård
- Department of Medical Biochemistry, Göteborg University, PO Box 440, SE 405 30 Göteborg, Sweden
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Suzuki A, Iwatani H, Ito T, Imai E, Okabe M, Nakamura H, Isaka Y, Yamato M, Hori M. Platelet-derived growth factor plays a critical role to convert bone marrow cells into glomerular mesangial-like cells. Kidney Int 2004; 65:15-24. [PMID: 14675032 DOI: 10.1111/j.1523-1755.2004.00379.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
BACKGROUND Despite increasing interest in bone marrow-derived stem cells, little is known about critical factors that determine their fates both in vitro and in vivo. Recently, we have reported that bone marrow is a reservoir for glomerular mesangial cells in rats. To find a key factor responsible for the differentiation of bone marrow-derived cells into mesangial cells, we established a new culture system of rat bone marrow, which is based on serial replating and differential attachment to collagen types I and IV. METHODS Bone marrow cells that did not adhere to collagen type I within 24 hours were transferred to collagen type IV-coated dishes. Then, the cells attached to collagen type IV in the following 24 hours were maintained in the presence of 2% horse serum, 200 ng/mL of platelet-derived growth factor (PDGF)-BB, and 1 micromol/L of all-trans retinoic acid. In vivo effect of PDGF-B was also examined by introducing human PDGF-B gene into glomeruli. RESULTS After cultivation under the above condition for 7 days, approximately 14% of cells expressed Thy-1 and desmin, both of which are markers for rat mesangial cells. Thy-1++/desmin+ cells were stellate-shaped, and contracted in response to angiotensin II. When human PDGF-B gene was overexpressed in the glomeruli of chimeric rats whose bone marrow was transplanted from enhanced green florescent protein (EGFP) transgenic rats, the number of EGFP+ mesangial cells increased. This effect was canceled by prior introduction of a neutralizing molecule that is composed of PDGF receptor-beta ligand binding site and IgG-Fc. CONCLUSION These results indicate that PDGF-B plays a critical role to direct bone marrow-derived cells toward mesangial-like cells both in vitro and in vivo.
Collapse
Affiliation(s)
- Akira Suzuki
- Department of Internal Medicine and Therapeutics, Osaka University School of Medicine, Suita, Osaka, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Abstract
ABSTRACT. Among the genes strongly induced by hypoxia-inducible factors (HIF) and highly expressed during kidney microvascular development is vascular endothelial growth factor, which encodes a potent endothelial mitogen and chemoattractant critical for embryonic vasculogenesis and angiogenesis. In developing kidney, glomerular podocytes are particularly rich sources of vascular endothelial growth factor, which probably serves to attract endothelial precursors into vascular clefts of immature glomeruli, promote their mitosis and differentiation into glomerular endothelial cells, and assist with maintenance of their highly differentiated state through maturation. This article summarizes the structure, function, and expression of HIF and discusses HIF target genes expressed during kidney vascular development. Furthermore, it is speculated that different HIF heterodimers are stabilized in different cell populations, which may lead to cell-selective induction of HIF target genes important for renal vasculogenesis/angiogenesis. E-mail: dabrahamson@kumc.edu
Collapse
Affiliation(s)
- Paul B Freeburg
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, Kansas 66160, USA.
| | | |
Collapse
|
44
|
Basciani S, Mariani S, Arizzi M, Brama M, Ricci A, Betsholtz C, Bondjers C, Ricci G, Catizone A, Galdieri M, Spera G, Gnessi L. Expression of platelet-derived growth factor (PDGF) in the epididymis and analysis of the epididymal development in PDGF-A, PDGF-B, and PDGF receptor beta deficient mice. Biol Reprod 2003; 70:168-77. [PMID: 14522834 DOI: 10.1095/biolreprod.103.019232] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
The platelet-derived growth factor (PDGF) family of ligands and receptors play a pivotal role in the development of various organs. The critical importance of the PDGF-mediated signaling during embryonic development and adult physiology of the kidney and the common mesonephric origin of the epididymis and kidney prompted us to investigate the immunohistochemical localization of PDGF A- and B-chain and PDGF receptor (PDGFR) alpha- and beta-subunit in rat and mouse epididymis, the expression profiles of the corresponding mRNAs, and the consequences of a loss-of-function mutation at the PDGF-A, PDGF-B, and PDGFR-beta loci on mouse epididymis phenotypic appearance. Prenatally, PDGF-A and PDGFR-alpha immunohistochemical staining was seen in both species, whereas PDGF-B and PDGFR-beta were absent. The cellular localization of PDGF-A within the epithelium and the alpha-receptor in the mesenchyme in either mouse or rat before birth suggests that the PDGF-A/PDGFR-alpha system might be involved in the epididymal epithelial-mesenchymal interaction during the fetal period of life. Postnatally, PDGF A- and B-ligand and PDGFR alpha- and beta-subunit were confined in the epithelium. The identity of PDGF and PDGFR proteins were further confirmed by immunoblotting. In line with the immunohistochemical studies, PDGF-A and PDGFR-alpha mRNAs were seen by reverse transcription-polymerase chain reaction in rat and mouse tissue before birth, whereas PDGF-B and PDGFR-beta were almost not detectable. During the first days of life, PDGF-B and PDGFR-beta genes started to appear, and the overall trend in mRNA expression throughout postnatal development showed that the transcripts levels for PDGF-A, PDGF-B, PDGFR-beta, and PDGFR-alpha were constant with the only exception of a progressive decrease of PDGFR-alpha in adult rats. The PDGF-A null mutation strongly influenced the epididymal phenotype starting from puberty; only fetal PDGF-B and PDGFR-beta -/- mice were available, and no differences were seen in the epididymis of these animals, compared with wild-type littermates. Taken together, these data indicate that the PDGF system is highly expressed in the epididymis and suggest that PDGF could be involved in the maintenance of morphological structure and functional control of this organ.
Collapse
Affiliation(s)
- Sabrina Basciani
- Department of Medical Pathophysiology, University of Rome "La Sapienza," Policlinico Umberto I, 00161 Rome, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
|
46
|
Ricono JM, Xu YC, Arar M, Jin DC, Barnes JL, Abboud HE. Morphological insights into the origin of glomerular endothelial and mesangial cells and their precursors. J Histochem Cytochem 2003; 51:141-50. [PMID: 12533522 DOI: 10.1177/002215540305100202] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Glomerular endothelial and mesangial cells may originate from the metanephric mesenchyme. We used the MAb Thy1.1, a mesangial cell marker in the adult rat kidney, and rat endothelial cell markers MAb RECA-1, MAb PECAM-1 (CD31), and MAb Flk-1 as potential markers to characterize the spatial and temporal distribution of mesangial and endothelial cell precursors during nephrogenesis in the rat. At early stages of glomerulogenesis, RECA-1- and Thy1.1-positive cells were detected in the metanephric blastema at 14 days post conception (dpc) embryos and 15 dpc, respectively, with Thy1.1 expression in cells surrounding the ureteric bud. At 17 and 18 dpc, both RECA-1- and Thy1.1-positive cells were found in the cleft of the S-shaped bodies and in the capillary loops of maturing glomeruli. Double staining for BrdU, a marker of proliferation, and for RECA-1 or BrdU and Thy1.1 also localize in the cleft of S-shaped bodies and in glomerular capillary loops at later stages of development. PDGFRbeta co-localizes in cells expressing endothelial or mesangial markers. The data suggest that endothelial and mesangial cell precursors share common markers during the course of glomerulogenesis and that full differentiation of these cells occurs at late stages of glomerular maturation. Thy1.1- and RECA-1-positive cells may be derived from the metanephric blastemal cells at early stages of kidney development. A subpopulation of these Thy1.1- or RECA-1-positive cells may be precursors that can migrate into the cleft of comma and S-shaped bodies and proliferate in situ to form glomerular capillary tufts.
Collapse
Affiliation(s)
- Jill M Ricono
- Department of Medicine, University of Texas Health Science Center, San Antonio 78229, USA
| | | | | | | | | | | |
Collapse
|
47
|
Alpers CE, Hudkins KL, Segerer S, Sage EH, Pichler R, Couser WG, Johnson RJ, Bassuk JA. Localization of SPARC in developing, mature, and chronically injured human allograft kidneys. Kidney Int 2002; 62:2073-86. [PMID: 12427131 DOI: 10.1046/j.1523-1755.2002.00680.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
BACKGROUND The matricellular protein SPARC (secreted protein acidic and rich in cysteine) is expressed during development, tissue remodeling and repair. It functions as an endogenous inhibitor of cell proliferation, regulates angiogenesis, regulates cell adhesion to extracellular matrix, binds cytokines such as platelet derived growth factor and stimulates transforming growth factor-beta (TGF-beta) production. This study describes the expression of SPARC during human renal development, in normal kidneys and during renal allograft rejection. METHODS A total of 60 renal specimens, including normal areas from tumor nephrectomies (N = 24), fetal kidneys (N = 27) and explanted renal allografts (N = 9), were included in the study. SPARC protein was localized by immunohistochemistry using two different antibodies. On consecutive sections SPARC mRNA was detected by in situ hybridization. RESULTS In the normal adult kidney SPARC protein was expressed by visceral and parietal epithelial cells, collecting duct epithelium (CD), urothelium, smooth muscle cells of muscular arteries and focally in interstitial cells. During renal development immature glomeruli demonstrated a polarized SPARC expression in visceral epithelial cells at their surface abutting the capillary basement membranes. In the fully differentiated glomeruli the expression pattern mirrored that of the adult kidney. Furthermore, SPARC was abundantly expressed by derivatives of the ureteric bud, and smooth muscle cells of arterial walls. During chronic allograft rejection SPARC is expressed in neointimal arterial smooth muscle cells, infiltrating inflammatory cells as well as by interstitial myofibroblasts in areas of interstitial fibrosis. SPARC mRNA synthesis detected by in situ hybridization mirrored these protein expression patterns. CONCLUSION These studies co-localize SPARC to several sites of renal injury previously shown to be sites of PDGF B-chain expression and/or activity. We speculate that SPARC could function as an accessory molecule in chronic PDGF-mediated sclerosing interstitial and vascular injury. SPARC localization to glomerular epithelial cells corresponds to similar findings in rodents, and may reflect its role in cell adhesion and /or regulation of cell shape.
Collapse
Affiliation(s)
- Charles E Alpers
- Department of Pathology, University of Washington School of Medicine, Seattle Washington, USA.
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Changsirikulchai S, Hudkins KL, Goodpaster TA, Volpone J, Topouzis S, Gilbertson DG, Alpers CE. Platelet-derived growth factor-D expression in developing and mature human kidneys. Kidney Int 2002; 62:2043-54. [PMID: 12427128 DOI: 10.1046/j.1523-1755.2002.00662.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
BACKGROUND Platelet-derived growth factor (PDGF) is a family of growth regulatory molecules composed of sulfide-bonded dimeric structures. Two well-studied PDGF peptides (PDGF-A and PDGF-B) have been shown to mediate a wide range of biological effects. PDGF-D is a newly recognized member of the PDGF family. Initial studies of the PDGF-D gene found its expression in cells of the vascular wall, suggesting that it could participate in vascular development and pathology. However, its localization in human kidney tissues has never been studied. METHODS PDGF-D expression in fetal (N = 30) and adult (N = 25) human kidney tissues was examined by immunohistochemistry using an affinity-purified antibody raised to human PDGF-D. Antibody absorption with the immunizing peptide was employed to confirm the specificity of this antibody. PDGF-D protein and gene expression in human kidneys also were demonstrated by Western blotting and reverse transcription-polymerase chain reaction (RT-PCR). RESULTS In the developing kidney, PDGF-D was first expressed by epithelial cells of comma- and S-shaped structures of the developing nephron, and most consistently in the visceral epithelial cells in the later stages of glomerular differentiation. In addition, PDGF-D could be found in mesenchymal, presumptively fibroblast cells in the interstitium of developing renal pelvis and in fetal smooth muscle cells in arterial vessels. In the adult normal kidney, PDGF-D was expressed by the visceral epithelial cells. There was persistent expression in arterial smooth muscle cells as well as in some neointimal smooth muscle cells of arteriosclerotic vessels, and expression in smooth muscle cells of vasa rectae in the medulla. PDGF-D could be identified at the basolateral membrane of some injured tubules in areas of chronic tubulointerstitial injury routinely encountered in aging kidneys. Western blotting of homogenates of adult kidneys demonstrated monospecific bands at 50 kD corresponding to previously established size parameter for this protein. RT-PCR of human kidney RNA resulted in a 918 basepair band, the sequence of which corresponded to human PDGF-D (Genbank number AF336376). CONCLUSIONS To our knowledge, these are the first studies to localize PDGF-D in human kidneys and suggest that PDGF-D may have a role in kidney development. PDGF-D was shown to bind to PDGF beta receptor, which localizes to mesangial cells, parietal epithelial cells, and interstitial fibroblasts, suggesting potential paracrine interactions between those cells and the visceral epithelium.
Collapse
|
49
|
Nakamura H, Isaka Y, Tsujie M, Rupprecht HD, Akagi Y, Ueda N, Imai E, Hori M. Introduction of DNA enzyme for Egr-1 into tubulointerstitial fibroblasts by electroporation reduced interstitial alpha-smooth muscle actin expression and fibrosis in unilateral ureteral obstruction (UUO) rats. Gene Ther 2002; 9:495-502. [PMID: 11948374 DOI: 10.1038/sj.gt.3301681] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2001] [Accepted: 01/18/2002] [Indexed: 11/09/2022]
Abstract
The phenotypic alteration of interstitial fibroblasts into 'myofibroblasts', acquiring characteristics of both fibroblasts and smooth muscle cells is a key event in the formation of tubulointerstitial fibrosis. The up-regulation of the early growth response gene 1 (Egr-1) preceded the increased interstitial expression of alpha-smooth muscle actin (alphaSMA), a marker of phenotypic changes, in obstructed kidney, a model of interstitial fibrosis. To target Egr-1 expression in the interstitium of obstructed kidneys, we introduced a DNA enzyme for Egr-1 (ED5) or scrambled DNA (SCR) into interstitial fibroblasts by electroporation-mediated gene transfer. Northern blot analysis confirmed an increase in the cortical mRNA expression of Egr-1 in the obstructed kidneys from untreated or SCR-treated rats, while ED5 transfection blocked Egr-1 expression with a concomitant reduction in TGF-beta, alphaSMA and type I collagen mRNA expression. Consequently, ED5 inhibited interstitial fibrosis. In conclusion, electroporation-mediated retrograde gene transfer can be an ideal vehicle into interstitial fibroblasts, and molecular intervention of Egr-1 in the interstitium may become a new therapeutic strategy for interstitial fibrosis.
Collapse
Affiliation(s)
- H Nakamura
- Department of Internal Medicine and Therapeutics, Osaka University Graduate School of Medicine, Suita, Japan
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Magro G, Perris R, Romeo R, Marcello M, Lopes M, Vasquez E, Grasso S. Comparative immunohistochemical analysis of the expression of cytokeratins, vimentin and alpha-smooth muscle actin in human foetal mesonephros and metanephros. THE HISTOCHEMICAL JOURNAL 2001; 33:221-6. [PMID: 11550803 DOI: 10.1023/a:1017950425012] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The human mesonephros is currently regarded as a simplified version of the foetal metanephros, primarily due to the close morphological resemblance between these two structures. The aim of the present study was to define whether human mesonephric and foetal metanephric nephrons share immunophenotypical traits in their corresponding structures (glomeruli, proximal and distal tubules). For this purpose we first investigated immunohistochemically the overall expression and topographical distribution of cytokeratins 7, 8, 18, 19, and 20, vimentin and alpha-smooth muscle actin in mature mesonephric nephrons and compared the results with those obtained in maturing-stage foetal metanephric nephrons. No expression of cytokeratins 7 and 20 was found. Cytokeratins 8, 18, and 19 and vimentin showed a restricted and basically coincident expression along the different components of both mesonephric and metanephric nephrons. These findings indicate that the intermediate filament protein profile of human mature mesonephric nephrons closely recapitulates that observed in developing metanephros and thereby strengthens the concept that human mesonephros, a transient ontogenic structure, is largely similar to the foetal metanephros. The sole difference between human mesonephros and foetal metanephros was the divergent expression of alpha-smooth muscle actin. This protein exhibited an increasingly accentuated mesangial expression paralleling the morphological maturation of metanephric glomerulus, whereas it was absent from the mesonephric one. This would suggest that the mesangial cells in these two renal structures have a different function during the foetal life.
Collapse
Affiliation(s)
- G Magro
- Institute of Anatomic Pathology, University of Catania, Italy
| | | | | | | | | | | | | |
Collapse
|