1
|
Chow MBCY, Yildiz V, Biederman L, Dasgupta A, Satoskar AA, Chow A, Nadasdy T, Brodsky SV. Proteinuria and proximal tubular epithelial cells: correlation between immunofluorescence, histology, and degree of proteinuria. FRONTIERS IN NEPHROLOGY 2024; 4:1469388. [PMID: 39544696 PMCID: PMC11560906 DOI: 10.3389/fneph.2024.1469388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 10/08/2024] [Indexed: 11/17/2024]
Abstract
Proteins are filtered from the blood through the glomerular filtration barrier. Filtered proteins are reabsorbed by proximal tubular epithelial cells (PTECs), which have been shown to possess the ability to regulate protein reabsorption. Histologically, these reabsorbed proteins are seen as tubular protein reabsorption droplets (TPRDs). Experimental studies indicate that PTECs play an important role in regulating proteinuria but the correlations between TPRD and the degree of proteinuria in human kidney biopsies have not been investigated in detail. Consecutive native kidney biopsies with non-proliferative glomerular disease performed at the OSUWMC for a 1-year period were analyzed. Cases with acute glomerular diseases and inadequate biopsies were excluded. The staining intensity and the percentage of TPRDs, as well as other morphologic parameters, were assessed. A total of 109 kidney biopsies were included in the study. A reverse correlation was identified between the percentage of albumin TPRDs and proteinuria (p = 0.047). There were positive correlations between proteinuria and the staining intensity for IgG TPRDs (p = 0.05) and the degree of acute tubular necrosis (ATN) (p = 0.015). In patients with no ATN, positive correlations between proteinuria and albumin and IgG TPRDs were seen, whereas in patients with ATN, these correlations were lost. A positive correlation was seen between proteinuria and chronic kidney injury. A strong correlation was noted between the degree of proteinuria and podocyte foot process effacement. Our data indicate that PTECs regulate proteinuria by absorbing proteins from the urine filtrate. Therefore, based on the human renal biopsy material, our study confirms that well-functioning renal PTECs play an important role in the regulation of proteinuria.
Collapse
Affiliation(s)
- Maria Bernadette CY Chow
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Deparment of Pathology, North District Hospital, Hong Kong, Hong Kong SAR, China
| | - Vedat Yildiz
- Center for Biostatistics, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Laura Biederman
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Department of Pathology, Nationwide Children Hospital, Columbus, OH, United States
| | - Alana Dasgupta
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Anjali A. Satoskar
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Aaron Chow
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Tibor Nadasdy
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Sergey V. Brodsky
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| |
Collapse
|
2
|
Rodrigues MC, Oliveira LBF, Vieira MAR, Caruso-Neves C, Peruchetti DB. Receptor-mediated endocytosis in kidney cells during physiological and pathological conditions. CURRENT TOPICS IN MEMBRANES 2024; 93:1-25. [PMID: 39181576 DOI: 10.1016/bs.ctm.2024.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Mammalian cell membranes are very dynamic where they respond to several environmental stimuli by rearranging the membrane composition by basic biological processes, including endocytosis. In this context, receptor-mediated endocytosis, either clathrin-dependent or caveolae-dependent, is involved in different physiological and pathological conditions. In the last years, an important amount of evidence has been reported that kidney function involves the modulation of different types of endocytosis, including renal protein handling. In addition, the dysfunction of the endocytic machinery is involved with the development of proteinuria as well as glomerular and tubular injuries observed in kidney diseases associated with hypertension, diabetes, and others. In this present review, we will discuss the mechanisms underlying the receptor-mediated endocytosis in different glomerular cells and proximal tubule epithelial cells as well as their modulation by different factors during physiological and pathological conditions. These findings could help to expand the current understanding regarding renal protein handling as well as identify possible new therapeutic targets to halt the progression of kidney disease.
Collapse
Affiliation(s)
- Mariana C Rodrigues
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Laura B F Oliveira
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Maria Aparecida R Vieira
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Celso Caruso-Neves
- Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil; Rio de Janeiro Innovation Network in Nanosystems for Health-NanoSAUDE/FAPERJ, Rio de Janeiro, RJ, Brazil; Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa, INCT-Regenera, Conselho Nacional de Desenvolvimento Científico e Tecnológico/MCTI, Rio de Janeiro, RJ, Brazil
| | - Diogo B Peruchetti
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil; Instituto Nacional de Ciência e Tecnologia em Nanobiofarmacêutica, INCT-NANOBiofar, Conselho Nacional de Desenvolvimento Científico e Tecnológico/MCTI, Belo Horizonte, MG, Brazil.
| |
Collapse
|
3
|
Huang Y, Ning X, Ahrari S, Cai Q, Rajora N, Saxena R, Yu M, Zheng J. Physiological principles underlying the kidney targeting of renal nanomedicines. Nat Rev Nephrol 2024; 20:354-370. [PMID: 38409369 DOI: 10.1038/s41581-024-00819-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/09/2024] [Indexed: 02/28/2024]
Abstract
Kidney disease affects more than 10% of the global population and is associated with considerable morbidity and mortality, highlighting a need for new therapeutic options. Engineered nanoparticles for the treatment of kidney diseases (renal nanomedicines) represent one such option, enabling the delivery of targeted therapeutics to specific regions of the kidney. Although they are underdeveloped compared with nanomedicines for diseases such as cancer, findings from preclinical studies suggest that renal nanomedicines may hold promise. However, the physiological principles that govern the in vivo transport and interactions of renal nanomedicines differ from those of cancer nanomedicines, and thus a comprehensive understanding of these principles is needed to design nanomedicines that effectively and specifically target the kidney while ensuring biosafety in their future clinical translation. Herein, we summarize the current understanding of factors that influence the glomerular filtration, tubular uptake, tubular secretion and extrusion of nanoparticles, including size and charge dependency, and the role of specific transporters and processes such as endocytosis. We also describe how the transport and uptake of nanoparticles is altered by kidney disease and discuss strategic approaches by which nanoparticles may be harnessed for the detection and treatment of a variety of kidney diseases.
Collapse
Affiliation(s)
- Yingyu Huang
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, Richardson, TX, USA
| | - Xuhui Ning
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, Richardson, TX, USA
| | - Samira Ahrari
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, Richardson, TX, USA
| | - Qi Cai
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Nilum Rajora
- Department of Internal Medicine, Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ramesh Saxena
- Department of Internal Medicine, Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Mengxiao Yu
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, Richardson, TX, USA.
| | - Jie Zheng
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, Richardson, TX, USA.
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
4
|
Kmochová T, Kidd KO, Orr A, Hnízda A, Hartmannová H, Hodaňová K, Vyleťal P, Naušová K, Brinsa V, Trešlová H, Sovová J, Barešová V, Svojšová K, Vrbacká A, Stránecký V, Robins VC, Taylor A, Martin L, Rivas-Chavez A, Payne R, Bleyer HA, Williams A, Rennke HG, Weins A, Short PJ, Agrawal V, Storsley LJ, Waikar SS, McPhail ED, Dasari S, Leung N, Hewlett T, Yorke J, Gaston D, Geldenhuys L, Samuels M, Levine AP, West M, Hůlková H, Pompach P, Novák P, Weinberg RB, Bedard K, Živná M, Sikora J, Bleyer AJ, Kmoch S. Autosomal dominant ApoA4 mutations present as tubulointerstitial kidney disease with medullary amyloidosis. Kidney Int 2024; 105:799-811. [PMID: 38096951 DOI: 10.1016/j.kint.2023.11.021] [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: 04/13/2023] [Revised: 11/03/2023] [Accepted: 11/10/2023] [Indexed: 01/21/2024]
Abstract
Sporadic cases of apolipoprotein A-IV medullary amyloidosis have been reported. Here we describe five families found to have autosomal dominant medullary amyloidosis due to two different pathogenic APOA4 variants. A large family with autosomal dominant chronic kidney disease (CKD) and bland urinary sediment underwent whole genome sequencing with identification of a chr11:116692578 G>C (hg19) variant encoding the missense mutation p.L66V of the ApoA4 protein. We identified two other distantly related families from our registry with the same variant and two other distantly related families with a chr11:116693454 C>T (hg19) variant encoding the missense mutation p.D33N. Both mutations are unique to affected families, evolutionarily conserved and predicted to expand the amyloidogenic hotspot in the ApoA4 structure. Clinically affected individuals suffered from CKD with a bland urinary sediment and a mean age for kidney failure of 64.5 years. Genotyping identified 48 genetically affected individuals; 44 individuals had an estimated glomerular filtration rate (eGFR) under 60 ml/min/1.73 m2, including all 25 individuals with kidney failure. Significantly, 11 of 14 genetically unaffected individuals had an eGFR over 60 ml/min/1.73 m2. Fifteen genetically affected individuals presented with higher plasma ApoA4 concentrations. Kidney pathologic specimens from four individuals revealed amyloid deposits limited to the medulla, with the mutated ApoA4 identified by mass-spectrometry as the predominant amyloid constituent in all three available biopsies. Thus, ApoA4 mutations can cause autosomal dominant medullary amyloidosis, with marked amyloid deposition limited to the kidney medulla and presenting with autosomal dominant CKD with a bland urinary sediment. Diagnosis relies on a careful family history, APOA4 sequencing and pathologic studies.
Collapse
Affiliation(s)
- Tereza Kmochová
- Research Unit for Rare Diseases, Department of Pediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Kendrah O Kidd
- Research Unit for Rare Diseases, Department of Pediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University, Prague, Czech Republic; Section on Nephrology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Andrew Orr
- Department of Ophthalmology and Visual Sciences, Dalhousie University, Halifax, Nova Scotia, Canada; Department of Pathology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Aleš Hnízda
- Research Unit for Rare Diseases, Department of Pediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Hana Hartmannová
- Research Unit for Rare Diseases, Department of Pediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Kateřina Hodaňová
- Research Unit for Rare Diseases, Department of Pediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Petr Vyleťal
- Research Unit for Rare Diseases, Department of Pediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Karolína Naušová
- Research Unit for Rare Diseases, Department of Pediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Vítězslav Brinsa
- Research Unit for Rare Diseases, Department of Pediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Helena Trešlová
- Research Unit for Rare Diseases, Department of Pediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Jana Sovová
- Research Unit for Rare Diseases, Department of Pediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Veronika Barešová
- Research Unit for Rare Diseases, Department of Pediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Klára Svojšová
- Research Unit for Rare Diseases, Department of Pediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Alena Vrbacká
- Research Unit for Rare Diseases, Department of Pediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Viktor Stránecký
- Research Unit for Rare Diseases, Department of Pediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Victoria C Robins
- Section on Nephrology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Abbigail Taylor
- Section on Nephrology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Lauren Martin
- Section on Nephrology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Ana Rivas-Chavez
- Section on Nephrology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Riley Payne
- Section on Nephrology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Heidi A Bleyer
- Section on Nephrology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Adrienne Williams
- Section on Nephrology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Helmut G Rennke
- Pathology Department, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Astrid Weins
- Pathology Department, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Varun Agrawal
- Division of Nephrology and Hypertension, Larner College of Medicine, University of Vermont, Burlington, Vermont, USA
| | - Leroy J Storsley
- Department of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Sushrut S Waikar
- Section of Nephrology, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts, USA
| | - Ellen D McPhail
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Surendra Dasari
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota, USA
| | - Nelson Leung
- Division of Nephrology and Hypertension, Division of Hematology, Mayo Clinic, Rochester, Minnesota, USA
| | - Tom Hewlett
- Division of Nephrology, Department of Medicine, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Jake Yorke
- Department of Pathology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Daniel Gaston
- Department of Pathology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Laurette Geldenhuys
- Department of Pathology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Mark Samuels
- Department of Medicine Université de Montréal, Montreal, Quebec, Canada; Department of Biochemistry, Université de Montréal, Montreal, Quebec, Canada; Centre de Recherche du CHU Ste-Justine, Montreal, Quebec, Canada
| | - Adam P Levine
- Research Department of Pathology, University College London, London, UK
| | - Michael West
- Division of Nephrology, Department of Medicine, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Helena Hůlková
- Research Unit for Rare Diseases, Department of Pediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University, Prague, Czech Republic; Institute of Pathology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Petr Pompach
- Institute of Microbiology of the Czech Academy of Sciences, Vestec, Czech Republic
| | - Petr Novák
- Institute of Microbiology of the Czech Academy of Sciences, Vestec, Czech Republic
| | - Richard B Weinberg
- Section on Gastroenterology, Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA; Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Karen Bedard
- Department of Pathology and Laboratory Medicine, Izaak Walton Killam Hospital, Halifax Nova Scotia, Canada
| | - Martina Živná
- Research Unit for Rare Diseases, Department of Pediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University, Prague, Czech Republic; Section on Nephrology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Jakub Sikora
- Research Unit for Rare Diseases, Department of Pediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University, Prague, Czech Republic; Institute of Pathology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Anthony J Bleyer
- Research Unit for Rare Diseases, Department of Pediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University, Prague, Czech Republic; Section on Nephrology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA.
| | - Stanislav Kmoch
- Research Unit for Rare Diseases, Department of Pediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University, Prague, Czech Republic; Section on Nephrology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| |
Collapse
|
5
|
Tanner GA. The Glomerular Sieving Coefficient of Albumin Is Really Very Low. Nephron Clin Pract 2024; 148:584-586. [PMID: 39186923 DOI: 10.1159/000538281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 03/07/2024] [Indexed: 08/28/2024] Open
Affiliation(s)
- George A Tanner
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine Indianapolis, Indianapolis, Indiana, USA
| |
Collapse
|
6
|
Satarug S, Vesey DA, Gobe GC, Phelps KR. The pathogenesis of albuminuria in cadmium nephropathy. Curr Res Toxicol 2023; 6:100140. [PMID: 38116328 PMCID: PMC10726218 DOI: 10.1016/j.crtox.2023.100140] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 11/16/2023] [Accepted: 11/30/2023] [Indexed: 12/21/2023] Open
Abstract
Background Urinary cadmium excretion (ECd) rises with renal tissue content of the metal. Whereas glomerulopathies are sometimes associated with massive albuminuria, tubular accumulation of Cd typically causes modest albuminuria. Since β2-microglobulinuria (Eβ2M) is an established marker of proximal tubular dysfunction, we hypothesized that a comparison of albuminuria (Ealb) to Eβ2M in Cd-exposed subjects would provide evidence of similar mishandling of both proteins. Methods To depict excretion rates per functional nephron, ECd, Ealb, and Eβ2M were normalized to creatinine clearance (Ccr), a surrogate for the glomerular filtration rate (GFR). Estimation of GFR itself (eGFR) was accomplished with CKD-EPI formulas (2009). Linear and logistic regression analyses were performed to relate Ealb/Ccr, Eβ2M/Ccr, and eGFR to several independent variables. Simple linear regressions of eGFR, Ealb/Ccr, and Eβ2M/Ccr on ECd/Ccr were examined before and after adjustment of dependent variables for age. All regressions were performed after log-transformation of ratios and standardization of all variables. Increments in Ealb/Ccr and Eβ2M/Ccr and decrements in eGFR were quantified through four quartiles of ECd/Ccr. Results As age or ECd/Ccr rose, Ealb/Ccr and Eβ2M/Ccr also rose, and eGFR fell. In linear regressions, slopes relating Ealb/Ccr and Eβ2M/Ccr to ECd/Ccr were similar. After adjustment of dependent variables for age, coefficients of determination (R2) for all regressions rose by a multiple, and slopes approached unity. Ealb/Ccr and Eβ2M/Ccr were similarly associated with each other. Mean Ealb/Ccr and Eβ2M/Ccr rose and mean eGFR fell in stepwise fashion through quartiles of ECd/Ccr. Whereas Eβ2M/Ccr did not vary with blood pressure, Ealb/Ccr rose in association with hypertension in two of the four quartiles. Conclusions Our data indicate that Cd in renal tissue affected tubular reabsorption of albumin and β2M similarly in a large cohort of exposed subjects. The results suggest that Cd reduced receptor-mediated endocytosis and subsequent lysosomal degradation of each protein by a shared mechanism.
Collapse
Affiliation(s)
- Soisungwan Satarug
- Centre for Kidney Disease Research, Translational Research Institute, Brisbane, Australia
| | - David A. Vesey
- Centre for Kidney Disease Research, Translational Research Institute, Brisbane, Australia
- Department of Kidney and Transplant Services, Princess Alexandra Hospital, Brisbane, Australia
| | - Glenda C. Gobe
- Centre for Kidney Disease Research, Translational Research Institute, Brisbane, Australia
- School of Biomedical Sciences, The University of Queensland, Brisbane, Australia
- NHMRC Centre of Research Excellence for CKD QLD, UQ Health Sciences, Royal Brisbane and Women’s Hospital, Brisbane, Australia
| | - Kenneth R. Phelps
- Stratton Veterans Affairs Medical Center and Albany Medical College, Albany, NY, USA
| |
Collapse
|
7
|
Molitoris BA, Dunn KW, Sandoval RM. Proximal tubule role in albumin homeostasis: controversy, species differences, and the contributions of intravital microscopy. Kidney Int 2023; 104:1065-1069. [PMID: 37981429 DOI: 10.1016/j.kint.2023.05.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 05/23/2023] [Accepted: 05/30/2023] [Indexed: 11/21/2023]
Affiliation(s)
- Bruce A Molitoris
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA.
| | - Kenneth W Dunn
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Ruben M Sandoval
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
8
|
Claudio P, Gabriella M. Nephrotic syndrome: pathophysiology and consequences. J Nephrol 2023; 36:2179-2190. [PMID: 37466816 DOI: 10.1007/s40620-023-01697-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 05/30/2023] [Indexed: 07/20/2023]
Abstract
In patients with kidney disease, nephrotic syndrome can lead to several complications including progressive kidney dysfunction. Proteinuria may lead to the formation of cellular or fibrous crescents with reciprocal development of rapidly progressive glomerulonephritis or focal glomerulosclerosis. Proteinuria may also cause overload and dysfunction of tubular epithelial cells, eventually resulting in tubular atrophy and interstitial fibrosis. Hypoalbuminemia is usually associated with increased risk of mortality and kidney dysfunction. Dyslipidemia may increase the risk of atherosclerotic complications, cause podocyte dysfunction and contribute to vascular thrombosis. Urinary loss of anticoagulants and overproduction of coagulation factors may facilitate a hypercoagulable state. Edema, hypogammaglobulinemia, loss of complement factors, and immunosuppressive therapy can favor infection. Treatment of these complications may reduce their impact on the severity of NS. Nephrotic syndrome is a kidney disorder that can worsen the quality of life and increase the risk of kidney disease progression.
Collapse
Affiliation(s)
| | - Moroni Gabriella
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072, Pieve Emanuele, Milan, Italy
- Nephrology and Dialysis Division, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089, Rozzano, Milan, Italy
| |
Collapse
|
9
|
Dylewski JF, Haddad G, Blaine J. Exploiting the neonatal Fc receptor to treat kidney disease. Kidney Int 2023; 105:S0085-2538(23)00747-0. [PMID: 39491149 PMCID: PMC11068363 DOI: 10.1016/j.kint.2023.09.024] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 09/11/2023] [Accepted: 09/25/2023] [Indexed: 05/07/2024]
Abstract
The neonatal Fc receptor (FcRn) was initially discovered as the receptor that allowed passive immunity in newborns by transporting maternal IgG through the placenta and enterocytes. Since its initial discovery, FcRn has been found to exist throughout all stages of life and in many different cell types. Beyond passive immunity, FcRn is necessary for intrinsic albumin and IgG recycling and is important for antigen processing and presentation. Given its multiple important roles, FcRn has been utilized in many disease treatments including a new class of agents that were developed to inhibit FcRn for treatment of a variety of autoimmune diseases. Certain cell populations within the kidney also express high levels of this receptor. Specifically, podocytes, proximal tubule epithelial cells, and vascular endothelial cells have been found to utilize FcRn. In this review, we summarize what is known about FcRn and its function within the kidney. We also discuss how FcRn has been used for therapeutic benefit, including how newer FcRn inhibiting agents are being used to treat autoimmune diseases. Lastly, we will discuss what renal diseases may respond to FcRn inhibitors and how further work studying FcRn within the kidney may lead to therapies for kidney diseases.
Collapse
Affiliation(s)
- James F Dylewski
- Division of Nephrology, Denver Health Medical Center, Denver, CO, USA; Division of Renal Disease and Hypertension, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, USA.
| | - George Haddad
- Division of Renal Disease and Hypertension, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| | - Judith Blaine
- Division of Renal Disease and Hypertension, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| |
Collapse
|
10
|
Haeri H, Eisermann J, Schimm H, Büscher A, Hoyer P, Hinderberger D. Profound Changes in Functional Structure and Dynamics of Serum Albumin in Children with Nephrotic Syndrome: An Exploratory Research Study. J Med Chem 2023; 66:12115-12129. [PMID: 37648246 PMCID: PMC10510392 DOI: 10.1021/acs.jmedchem.3c00680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Indexed: 09/01/2023]
Abstract
Patients with nephrotic syndrome (NS) suffer from urinary loss of albumin. As a cause, previous studies focused on the glomerular filter rather than analyzing the molecular properties of albumin itself. Later one was initiated by clinical observations indicating unexplained molecular alterations of human serum albumin (HSA) in an NS pediatric patient. Therefore, we examined serum from eight pediatric patients with steroid-sensitive and -resistant NS and compared it with serum from healthy subjects as well as commercial HSA. We used dynamic and electrophoretic light scattering to characterize the protein size and effective surface charge and electron paramagnetic resonance spectroscopy to measure the local environment and binding dynamics of up to seven fatty acids associated with HSA. Our findings suggest that pronounced differences in binding behavior and surface charge of HSA could enhance their filtration through the GBM, leading to direct toxicity of HSA to podocytes.
Collapse
Affiliation(s)
- Haleh
H. Haeri
- Physical
Chemistry, Complex Self-Organizing Systems, Martin Luther University
Halle-Wittenberg, Institute of Chemistry, Von Danckelmann-Platz 4, 06120 Halle (Saale), Germany
| | - Jana Eisermann
- Department
of Chemistry, Molecular Sciences Research Hub, Imperial College London, 82 Wood Ln, W12 0BZ London, U.K.
| | - Heike Schimm
- Physical
Chemistry, Complex Self-Organizing Systems, Martin Luther University
Halle-Wittenberg, Institute of Chemistry, Von Danckelmann-Platz 4, 06120 Halle (Saale), Germany
| | - Anja Büscher
- Universitätsklinikum
Essen (AöR), Klinik für Kinderheilkunde II, Zentrum
für Kinder- und Jugendmedizin, Hufelandstraße 55, D-45147 Essen, Germany
| | - Peter Hoyer
- Universitätsklinikum
Essen (AöR), Klinik für Kinderheilkunde II, Zentrum
für Kinder- und Jugendmedizin, Hufelandstraße 55, D-45147 Essen, Germany
| | - Dariush Hinderberger
- Physical
Chemistry, Complex Self-Organizing Systems, Martin Luther University
Halle-Wittenberg, Institute of Chemistry, Von Danckelmann-Platz 4, 06120 Halle (Saale), Germany
| |
Collapse
|
11
|
Shimada S, Hoffmann BR, Yang C, Kurth T, Greene AS, Liang M, Dash RK, Cowley AW. Metabolic Responses of Normal Rat Kidneys to a High Salt Intake. FUNCTION 2023; 4:zqad031. [PMID: 37575482 PMCID: PMC10413938 DOI: 10.1093/function/zqad031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 06/06/2023] [Accepted: 06/12/2023] [Indexed: 08/15/2023] Open
Abstract
In this study, novel methods were developed, which allowed continuous (24/7) measurement of arterial blood pressure and renal blood flow in freely moving rats and the intermittent collection of arterial and renal venous blood to estimate kidney metabolic fluxes of O2 and metabolites. Specifically, the study determined the effects of a high salt (HS; 4.0% NaCl) diet upon whole kidney O2 consumption and arterial and renal venous plasma metabolomic profiles of normal Sprague-Dawley rats. A separate group of rats was studied to determine changes in the cortex and outer medulla tissue metabolomic and mRNAseq profiles before and following the switch from a 0.4% to 4.0% NaCl diet. In addition, targeted mRNA expression analysis of cortical segments was performed. Significant changes in the metabolomic and transcriptomic profiles occurred with feeding of the HS diet. A progressive increase of kidney O2 consumption was found despite a reduction in expression of most of the mRNA encoding enzymes of TCA cycle. A novel finding was the increased expression of glycolysis-related genes in Cx and isolated proximal tubular segments in response to an HS diet, consistent with increased release of pyruvate and lactate from the kidney to the renal venous blood. Data suggests that aerobic glycolysis (eg, Warburg effect) may contribute to energy production under these circumstances. The study provides evidence that kidney metabolism responds to an HS diet enabling enhanced energy production while protecting from oxidative stress and injury. Metabolomic and transcriptomic analysis of kidneys of Sprague-Dawley rats fed a high salt diet.
Collapse
Affiliation(s)
- Satoshi Shimada
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Brian R Hoffmann
- Mass Spectrometry and Protein Chemistry, Protein Sciences, The Jackson Laboratory, Bar Harbor, ME 04609, USA
| | - Chun Yang
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Theresa Kurth
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Andrew S Greene
- Mass Spectrometry and Protein Chemistry, Protein Sciences, The Jackson Laboratory, Bar Harbor, ME 04609, USA
| | - Mingyu Liang
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Ranjan K Dash
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Biomedical Engineering, Medical College of Wisconsin and Marquette University, Milwaukee, WI 53226, USA
| | - Allen W Cowley
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
12
|
Saleh MA, Shaaban AA, Talaat IM, Elmougy A, Adra SF, Ahmad F, Qaisar R, Elmoselhi AB, Abu-Gharbieh E, El-Huneidi W, Eladl MA, Shehatou G, Kafl HE. RhoA/ROCK inhibition attenuates endothelin-1-induced elevated glomerular permeability to albumin, inflammation, and fibrosis. Life Sci 2023; 323:121687. [PMID: 37030613 DOI: 10.1016/j.lfs.2023.121687] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/25/2023] [Accepted: 04/05/2023] [Indexed: 04/10/2023]
Abstract
Endothelin-1 (ET-1) contributes to the development of kidney diseases. However, the underlying molecular mechanism is largely undefined. Here we sought to investigate the potential role of ET-1 receptors, ETA and ETB in the regulation of increased glomerular permeability and underlying signaling pathways post-ET-1 infusion. Male Sprague-Dawley rats were infused with ET-1 (2 pmol/kg per minute, i.v.) for four weeks, and the effect on glomerular permeability to albumin (Palb) and albuminuria was measured. The selective ROCK-1/2 inhibitor, Y-27632, was administered to a separate group of rats to determine its effect on ET-1-induced Palb and albuminuria. The role of ETA and ETB receptors in regulating RhoA/ROCK activity was determined by incubating isolated glomeruli from normal rats with ET-1 and with selective ETA and ETB receptor antagonists. ET-1 infusion for four weeks significantly elevated Palb and albuminuria. Y-27632 significantly reduced the elevation of Palb and albuminuria. The activities of both RhoA and ROCK-1/2 were increased by ET-1 infusion. Selective ETB receptor antagonism had no effect on the elevated activity of both RhoA and ROCK-1/2 enzymes. Selective ETA receptor and combined ETA/ETB receptors blockade restored the activity of RhoA and ROCK-1/2 to normal levels. In addition, chronic ET-1 infusion increased the levels of glomerular inflammatory and fibrotic markers. These effects were all attenuated in rats following ROCK-1/2 inhibition. These observations suggest that ET-1 contributes to increased albuminuria, inflammation, and fibrosis by modulating the activity of the ETA-RhoA/ROCK-1/2 pathway. Selective ETA receptor blockade may represent a potential therapeutic strategy to limit glomerular injury and albuminuria in kidney disease.
Collapse
Affiliation(s)
- Mohamed A Saleh
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates; Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, United Arab Emirates; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt.
| | - Ahmed A Shaaban
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt; Department of Pharmacology and Biochemistry, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa City 35712, Egypt
| | - Iman M Talaat
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates; Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, United Arab Emirates; Pathology Department, Faculty of Medicine, Alexandria University, Alexandria 21526, Egypt
| | - Atef Elmougy
- Pediatric Nephrology Unit, Mansoura University Children's Hospital, Mansoura University, Mansoura 35516, Egypt
| | - Saryia F Adra
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Firdos Ahmad
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates; Department of Biomedical Sciences, College of Health Sciences, Abu Dhabi University, Abu Dhabi 59911, United Arab Emirates
| | - Rizwan Qaisar
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Adel B Elmoselhi
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Eman Abu-Gharbieh
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates; Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Waseem El-Huneidi
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, United Arab Emirates; Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Mohamed A Eladl
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - George Shehatou
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt; Department of Pharmacology and Biochemistry, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa City 35712, Egypt
| | - Hoda E Kafl
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| |
Collapse
|
13
|
Shimada S, Hoffmann BR, Yang C, Kurth T, Greene AS, Liang M, Dash RK, Cowley AW. Metabolic responses of normal rat kidneys to a high salt intake. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.18.524636. [PMID: 36711564 PMCID: PMC9882299 DOI: 10.1101/2023.01.18.524636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
In the present study, novel methods were developed which allowed continuous (24/7) measurement of blood pressure (BP) and renal blood flow (RBF) in freely moving rats and the intermittent collection of arterial and renal venous blood to estimate kidney metabolic fluxes of O 2 and metabolites. The study determined the effects of a high salt (HS) diet upon whole kidney O 2 consumption and the metabolomic profiles of normal Sprague Dawley (SD) rats. A separate group of rats was studied to determine changes in the cortex (Cx) and outer medulla (OM) tissue metabolomic and mRNAseq profiles before and following the switch from a 0.4% to a 4.0% NaCl diet. Significant changes in the metabolomic and transcriptomic profiles occurred with feeding of the HS diet. A progressive increase of kidney O 2 consumption was found despite a reduction in expression of most of the mRNA encoding enzymes of TCA cycle. Increased glycolysis was evident with the elevation of mRNA expression encoding key glycolytic enzymes and release of pyruvate and lactate from the kidney in the renal venous blood. Glycolytic production of NADH is used in either the production of lactate or oxidized via the malate aspartate shuttle. Aerobic glycolysis (e.g., Warburg-effect) may account for the needed increase in cellular energy. The study provides evidence that kidney metabolism responds to a HS diet enabling enhanced energy production while protecting from oxidate stress and injury.
Collapse
|
14
|
Romero-Guevara R, Nicolaou O, Petracca B, Shaheed S, Sutton C, Frangou E, Afami M, Kyriacou K, Ioannides A, Xinaris C. Patient-derived podocyte spheroids reveal new insights into the etiopathogenesis of Alport syndrome. Front Cell Dev Biol 2023; 11:1111424. [PMID: 36936689 PMCID: PMC10018139 DOI: 10.3389/fcell.2023.1111424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 02/17/2023] [Indexed: 03/06/2023] Open
Abstract
Alport syndrome (AS) is a rare disease characterized by defective glomerular basement membranes, caused by mutations in COL4A3, COL4A4, and COL4A5, which synthesize collagen type IV. Patients present with progressive proteinuria, hematuria and podocyte loss. There is currently no cure for Alport syndrome, and this is mainly due to its complex and variable pathogenesis, as well as the lack of models that can faithfully mimic the human phenotype. Here we have developed a novel human culture model of Alport syndrome and used it to study the effects of different mutations on podocyte development and biology. First, we established a differentiation protocol that allowed us to generate podocyte spheroids from patient-derived human induced pluripotent stem cells (hiPSCs). We have then carried out discovery proteomics and demonstrated that a total of 178 proteins were differentially expressed between Alport (AS1 and AS3) and control (LT) podocytes. GO analysis indicated alterations in several metabolic pathways, such as oxidative phosphorylation, RNA maturation, chromatin condensation, and proliferation. Although functional assays showed no changes in lactate production and mitochondrial potential compared to healthy controls, immunofluorescence and electron microscopy analysis showed key morphological changes related to the phenotypical maturation of Alport podocytes. Moreover, the studied mutations led to persistent proliferation, increased reactive oxygen species (ROS) production and the concomitant expression of peroxisome proliferator-activated receptors α and γ (PPARα and PPARγ) in podocytes. These data on patient-derived podocytes provide evidence that collagen mutations, in addition to playing a central role in the defective development of the glomerular filtration barrier, cause significant alterations in podocyte development and metabolism very early in development, even before the formation of the filtering apparatus. In conclusion, our study provides a new methodological platform for the differentiation of podocytes and to study human podocytopathies in a personalized manner, and reveals new insights into the etiopathogenesis and pathobiology of Alport syndrome.
Collapse
Affiliation(s)
- Ricardo Romero-Guevara
- Department of Basic and Clinical Sciences, University of Nicosia Medical School, Nicosia, Cyprus
| | - Orthodoxia Nicolaou
- Department of Cancer Genetics, Therapeutics, and Ultrastructural Pathology, Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Benedetta Petracca
- Laboratory of Organ Regeneration, Department of Molecular Medicine, Institute of Pharmacological Research “Mario Negri”, Bergamo, Italy
| | - Sadr Shaheed
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
| | - Christopher Sutton
- School of Chemistry and Bioscience, University of Bradford, Bradford, United Kingdom
| | - Eleni Frangou
- Department of Basic and Clinical Sciences, University of Nicosia Medical School, Nicosia, Cyprus
- Department of Nephrology, Limassol General Hospital, Nicosia, Cyprus
| | - Marina Afami
- Department of Basic and Clinical Sciences, University of Nicosia Medical School, Nicosia, Cyprus
| | - Kyriacos Kyriacou
- Department of Cancer Genetics, Therapeutics, and Ultrastructural Pathology, Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Adonis Ioannides
- Department of Basic and Clinical Sciences, University of Nicosia Medical School, Nicosia, Cyprus
| | - Christodoulos Xinaris
- Department of Basic and Clinical Sciences, University of Nicosia Medical School, Nicosia, Cyprus
- Laboratory of Organ Regeneration, Department of Molecular Medicine, Institute of Pharmacological Research “Mario Negri”, Bergamo, Italy
- *Correspondence: Christodoulos Xinaris, ,
| |
Collapse
|
15
|
Dahlke E, Anan Y, Klie LM, Hartkopf AE, Theilig F. Megalin Orchestrates FcRn Endocytosis and Trafficking. Cells 2022; 12:cells12010053. [PMID: 36611847 PMCID: PMC9818419 DOI: 10.3390/cells12010053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 12/09/2022] [Accepted: 12/20/2022] [Indexed: 12/24/2022] Open
Abstract
The neonatal Fc receptor (FcRn) is highly expressed in the renal proximal tubule and is important for the reclamation of albumin by cellular transcytosis to prevent its loss in the urine. The initial event of this transcellular transport mechanism is the endocytosis of albumin by the apical scavenger receptors megalin and cubilin. An interaction of megalin and FcRn was postulated, however, evidence is still missing. Similarly, the intracellular trafficking of FcRn remains unknown and shall be identified in our study. Using a Venus-based bimolecular fluorescence complementation system, we detected an interaction between megalin and FcRn in the endosomal compartment, which significantly increased with the induction of endocytosis using albumin or lactoglobulin as a ligand. The interaction between megalin and FcRn occurred at a neutral and acidic pH between the extracellular domains of both proteins. Amnionless, another transmembrane acceptor of cubilin, revealed no interaction with FcRn. With the induction of endocytosis by albumin or lactoglobulin, super resolution microscopy demonstrated a redistribution of megalin and FcRn into clathrin vesicles and early endosomes. This trafficking into clathrin vesicles was impaired in megalin-deficient cells upon albumin-induced endocytosis, supporting the role of megalin in FcRn redistribution. Our results indicate that megalin and FcRn specifically bind and interact within their extracellular domains. The availability of megalin is necessary for the redistribution of FcRn. Megalin, therefore, orchestrates FcRn endocytosis and intracellular trafficking as an early event intranscytosis.
Collapse
|
16
|
Podocyturia: an earlier biomarker of cardiovascular outcomes. Sci Rep 2022; 12:21563. [PMID: 36513746 PMCID: PMC9747803 DOI: 10.1038/s41598-022-26162-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 12/12/2022] [Indexed: 12/15/2022] Open
Abstract
Urinary podocin and nephrin mRNAs (podocyturia), as candidate biomarkers of endothelial/podocyte injury, were measured by quantitative PCR in Type II diabetics with normal albumin excretion rates (AER) at baseline, at 3-4 years, and at 7 years. Development of cardiovascular disease (CVD) was collected as outcome. Visit 1 podocyturia was significantly higher in subjects who subsequently developed CVD versus those who did not. Visit 1 AER terciles exhibited similar time to CVD, in contrast with stepwise and substantial increases in CVD events predicted by Visit 1 podocyturia terciles. Covariate-adjusted hazard ratios were highest for podocin, intermediate for nephrin mRNAs, and lowest for AER. Podocyturia was also measured in patients with and without significant coronary obstruction, and in 480 normoalbuminuric subjects at the enrolment visit to the Multi-Ethnic Study of Atherosclerosis (MESA). Podocyturia > 3 × 106 copies was associated with presence of obstructive coronary artery disease. In the MESA population, Visit 1 podocyturia was significantly higher in men, subjects with elevated BMI, and those with Type II DM. Conclusions: Podocyturia may be an earlier predictor of cardiovascular events than moderate albuminuria; it is significantly higher in patients with obstructive coronary artery disease, and in subjects with established risk factors for CVD.
Collapse
|
17
|
Scheele CLGJ, Herrmann D, Yamashita E, Celso CL, Jenne CN, Oktay MH, Entenberg D, Friedl P, Weigert R, Meijboom FLB, Ishii M, Timpson P, van Rheenen J. Multiphoton intravital microscopy of rodents. NATURE REVIEWS. METHODS PRIMERS 2022; 2:89. [PMID: 37621948 PMCID: PMC10449057 DOI: 10.1038/s43586-022-00168-w] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 09/12/2022] [Indexed: 08/26/2023]
Abstract
Tissues are heterogeneous with respect to cellular and non-cellular components and in the dynamic interactions between these elements. To study the behaviour and fate of individual cells in these complex tissues, intravital microscopy (IVM) techniques such as multiphoton microscopy have been developed to visualize intact and live tissues at cellular and subcellular resolution. IVM experiments have revealed unique insights into the dynamic interplay between different cell types and their local environment, and how this drives morphogenesis and homeostasis of tissues, inflammation and immune responses, and the development of various diseases. This Primer introduces researchers to IVM technologies, with a focus on multiphoton microscopy of rodents, and discusses challenges, solutions and practical tips on how to perform IVM. To illustrate the unique potential of IVM, several examples of results are highlighted. Finally, we discuss data reproducibility and how to handle big imaging data sets.
Collapse
Affiliation(s)
- Colinda L. G. J. Scheele
- Laboratory for Intravital Imaging and Dynamics of Tumor Progression, VIB Center for Cancer Biology, KU Leuven, Leuven, Belgium
- Department of Oncology, KU Leuven, Leuven, Belgium
| | - David Herrmann
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Cancer Department, Sydney, New South Wales, Australia
- St. Vincent’s Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, New South Wales, Australia
| | - Erika Yamashita
- Department of Immunology and Cell Biology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Osaka, Japan
- WPI-Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Laboratory of Bioimaging and Drug Discovery, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | - Cristina Lo Celso
- Department of Life Sciences and Centre for Hematology, Imperial College London, London, UK
- Sir Francis Crick Institute, London, UK
| | - Craig N. Jenne
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Maja H. Oktay
- Department of Pathology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA
- Integrated Imaging Program, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA
| | - David Entenberg
- Department of Pathology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA
- Integrated Imaging Program, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA
| | - Peter Friedl
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, Nijmegen, Netherlands
- David H. Koch Center for Applied Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Roberto Weigert
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Franck L. B. Meijboom
- Department of Population Health Sciences, Sustainable Animal Stewardship, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
- Faculty of Humanities, Ethics Institute, Utrecht University, Utrecht, Netherlands
| | - Masaru Ishii
- Department of Immunology and Cell Biology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Osaka, Japan
- WPI-Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Laboratory of Bioimaging and Drug Discovery, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | - Paul Timpson
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Cancer Department, Sydney, New South Wales, Australia
- St. Vincent’s Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, New South Wales, Australia
| | - Jacco van Rheenen
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, Netherlands
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, Netherlands
| |
Collapse
|
18
|
Comper WD, Vuchkova J, McCarthy KJ. New insights into proteinuria/albuminuria. Front Physiol 2022; 13:991756. [PMID: 36225307 PMCID: PMC9548894 DOI: 10.3389/fphys.2022.991756] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 08/29/2022] [Indexed: 11/13/2022] Open
Abstract
The fractional clearance of proteins as measured in healthy human subjects increases 10,000–100,000- fold when studied in nephrotic patients. This remarkable increase cannot be accounted for by extracellular biophysical mechanisms centered at the glomerular filtration barrier. Rather, it is the nephron and its combination of filtration and cellular uptake that can provide a plausible explanation of these fractional clearance changes. The nephron has two regions that critically determine the level proteinuria/albuminuria. Glomerular filtration of plasma proteins is primarily a size selective event that is basically unchanged in acquired and genetic kidney disease. The glomerular concepts of ‘charge selectivity’ and of ‘large pores’, previously used to explain proteinuria, are now recognized to be flawed and non-existent. Filtered proteins then encounter downstream two protein receptors of the Park and Maack type associated with the proximal tubular cell. The high capacity receptor is thought to retrieve the majority of filtered proteins and return them to the blood supply. Inhibition/saturation of this pathway in kidney disease may create the nephrotic condition and hypoproteinemia/hypoalbuminemia. Inhibitors of this pathway (possibly podocyte derived) are still to be identified. A relatively small proportion of the filtered protein is directed towards a high affinity, low capacity receptor that guides the protein to undergo lysosomal degradation. Proteinuria in normoproteinemic states is derived by inhibition of this pathway, such as in diabetes. The combination of glomerular sieving, and the degradation and retrieval pathways can quantitatively account for the changes in fractional clearance of proteins in the nephrotic condition. Finally, the general retrieval of filtered protein by the proximal tubular cell focuses on the teleological importance of this cell as this retrieval represents the third pillar of retrieval that this cell participates in (it also retrieves water and salt).
Collapse
Affiliation(s)
- Wayne D. Comper
- Salaqua Diagnostics Inc, New York, NY, United States
- *Correspondence: Wayne D. Comper,
| | | | - Kevin J. McCarthy
- Department of Cellular Biology and Anatomy, LSU Health Sciences Center, Shreveport, LA, United States
| |
Collapse
|
19
|
Heyman SN, Raz I, Dwyer JP, Weinberg Sibony R, Lewis JB, Abassi Z. Diabetic Proteinuria Revisited: Updated Physiologic Perspectives. Cells 2022; 11:2917. [PMID: 36139492 PMCID: PMC9496872 DOI: 10.3390/cells11182917] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/11/2022] [Accepted: 09/13/2022] [Indexed: 11/16/2022] Open
Abstract
Albuminuria, a hallmark of diabetic nephropathy, reflects not only injury and dysfunction of the filtration apparatus, but is also affected by altered glomerular hemodynamics and hyperfiltration, as well as by the inability of renal tubular cells to fully retrieve filtered albumin. Albuminuria further plays a role in the progression of diabetic nephropathy, and the suppression of glomerular albumin leak is a key factor in its prevention. Although microalbuminuria is a classic manifestation of diabetic nephropathy, often progressing to macroalbuminuria or overt proteinuria over time, it does not always precede renal function loss in diabetes. The various components leading to diabetic albuminuria and their associations are herein reviewed, and the physiologic rationale and efficacy of therapeutic interventions that reduce glomerular hyperfiltration and proteinuria are discussed. With these perspectives, we propose that these measures should be initiated early, before microalbuminuria develops, as substantial renal injury may already be present in the absence of proteinuria. We further advocate that the inhibition of the renin-angiotensin axis or of sodium-glucose co-transport likely permits the administration of a normal recommended or even high-protein diet, highly desirable for sarcopenic diabetic patients.
Collapse
Affiliation(s)
- Samuel N. Heyman
- Department of Medicine, Hadassah Hebrew University Hospital, Mt. Scopus, Jerusalem 9765422, Israel
- Division of Geriatrics, Herzog Hospital, Jerusalem 9765422, Israel
| | - Itamar Raz
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9765422, Israel
- Diabetes Unit, Department of Endocrinology and Metabolism, Hadassah Medical Center, Jerusalem 9124001, Israel
| | - Jamie P. Dwyer
- Clinical and Translational Science Institute, University of Utah Health, Salt Lake City, UT 84112, USA
| | | | - Julia B. Lewis
- Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Departments of Medicine and Nephrology, Vanderbilt University Medical Center, Nashville, TN 37011, USA
| | - Zaid Abassi
- Department of Physiology and Biophysics, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 3200003, Israel
- Department of Laboratory Medicine, Rambam Health Care Campus, Haifa 3109601, Israel
| |
Collapse
|
20
|
Lidberg KA, Muthusamy S, Adil M, Mahadeo A, Yang J, Patel RS, Wang L, Bammler TK, Reichel J, Yeung CK, Himmelfarb J, Kelly EJ, Akilesh S. Serum Protein Exposure Activates a Core Regulatory Program Driving Human Proximal Tubule Injury. J Am Soc Nephrol 2022; 33:949-965. [PMID: 35197326 PMCID: PMC9063895 DOI: 10.1681/asn.2021060751] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Accepted: 02/06/2022] [Indexed: 11/03/2022] Open
Abstract
BACKGROUND The kidneys efficiently filter waste products while retaining serum proteins in the circulation. However, numerous diseases compromise this barrier function, resulting in spillage of serum proteins into the urine (proteinuria). Some studies of glomerular filtration suggest that tubules may be physiologically exposed to nephrotic-range protein levels. Therefore, whether serum components can directly injure the downstream tubular portions of the kidney, which in turn can lead to inflammation and fibrosis, remains controversial. METHODS We tested the effects of serum protein exposure in human kidney tubule microphysiologic systems and with orthogonal epigenomic approaches since animal models cannot directly assess the effect of serum components on tubules. RESULTS Serum, but not its major protein component albumin, induced tubular injury and secretion of proinflammatory cytokines. Epigenomic comparison of serum-injured tubules and intact kidney tissue revealed canonical stress-inducible regulation of injury-induced genes. Concordant transcriptional changes in microdissected tubulointerstitium were also observed in an independent cohort of patients with proteinuric kidney disease. CONCLUSIONS Our results demonstrate a causal role for serum proteins in tubular injury and identify regulatory mechanisms and novel pathways for intervention.
Collapse
Affiliation(s)
- Kevin A. Lidberg
- Department of Pharmaceutics, University of Washington, Seattle, Washington
| | - Selvaraj Muthusamy
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington
| | - Mohamed Adil
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington
| | - Anish Mahadeo
- Department of Pharmaceutics, University of Washington, Seattle, Washington
| | - Jade Yang
- Department of Pharmaceutics, University of Washington, Seattle, Washington
| | | | - Lu Wang
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington
| | - Theo K. Bammler
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington
| | - Jonathan Reichel
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington
| | - Catherine K. Yeung
- Department of Pharmacy, University of Washington, Seattle, Washington
- Kidney Research Institute, Seattle, Washington
| | - Jonathan Himmelfarb
- Kidney Research Institute, Seattle, Washington
- Nephrology Division, Department of Medicine, University of Washington, Seattle, Washington
| | - Edward J. Kelly
- Department of Pharmaceutics, University of Washington, Seattle, Washington
- Kidney Research Institute, Seattle, Washington
| | - Shreeram Akilesh
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington
- Kidney Research Institute, Seattle, Washington
| |
Collapse
|
21
|
Molitoris BA, Sandoval RM, Yadav SPS, Wagner MC. Albumin Uptake and Processing by the Proximal Tubule: Physiologic, Pathologic and Therapeutic Implications. Physiol Rev 2022; 102:1625-1667. [PMID: 35378997 PMCID: PMC9255719 DOI: 10.1152/physrev.00014.2021] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
For nearly 50 years the proximal tubule (PT) has been known to reabsorb, process, and either catabolize or transcytose albumin from the glomerular filtrate. Innovative techniques and approaches have provided insights into these processes. Several genetic diseases, nonselective PT cell defects, chronic kidney disease (CKD), and acute PT injury lead to significant albuminuria, reaching nephrotic range. Albumin is also known to stimulate PT injury cascades. Thus, the mechanisms of albumin reabsorption, catabolism, and transcytosis are being reexamined with the use of techniques that allow for novel molecular and cellular discoveries. Megalin, a scavenger receptor, cubilin, amnionless, and Dab2 form a nonselective multireceptor complex that mediates albumin binding and uptake and directs proteins for lysosomal degradation after endocytosis. Albumin transcytosis is mediated by a pH-dependent binding affinity to the neonatal Fc receptor (FcRn) in the endosomal compartments. This reclamation pathway rescues albumin from urinary losses and cellular catabolism, extending its serum half-life. Albumin that has been altered by oxidation, glycation, or carbamylation or because of other bound ligands that do not bind to FcRn traffics to the lysosome. This molecular sorting mechanism reclaims physiological albumin and eliminates potentially toxic albumin. The clinical importance of PT albumin metabolism has also increased as albumin is now being used to bind therapeutic agents to extend their half-life and minimize filtration and kidney injury. The purpose of this review is to update and integrate evolving information regarding the reabsorption and processing of albumin by proximal tubule cells including discussion of genetic disorders and therapeutic considerations.
Collapse
Affiliation(s)
- Bruce A. Molitoris
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, United States
- Dept.of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana, United States
| | - Ruben M. Sandoval
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, United States
| | - Shiv Pratap S. Yadav
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, United States
| | - Mark C. Wagner
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, United States
| |
Collapse
|
22
|
Molitoris BA, Sandoval RM, Wagner MC. Intravital Multiphoton Microscopy as a Tool for Studying Renal Physiology, Pathophysiology and Therapeutics. Front Physiol 2022; 13:827280. [PMID: 35399274 PMCID: PMC8988037 DOI: 10.3389/fphys.2022.827280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 02/09/2022] [Indexed: 11/13/2022] Open
Abstract
Intravital multiphoton microscopy has empowered investigators to study dynamic cell and subcellular processes in vivo within normal and disease organs. Advances in hardware, software, optics, transgenics and fluorescent probe design and development have enabled new quantitative approaches to create a disruptive technology pioneering advances in understanding of normal biology, disease pathophysiology and therapies. Offering superior spatial and temporal resolution with high sensitivity, investigators can follow multiple processes simultaneously and observe complex interactions between different cell types, intracellular organelles, proteins and track molecules for cellular uptake, intracellular trafficking, and metabolism in a cell specific fashion. The technique has been utilized in the kidney to quantify multiple dynamic processes including capillary flow, permeability, glomerular function, proximal tubule processes and determine the effects of diseases and therapeutic mechanisms. Limitations include the depth of tissue penetration with loss of sensitivity and resolution due to scattered emitted light. Tissue clearing technology has virtually eliminated penetration issues for fixed tissue studies. Use of multiphoton microscopy in preclinical animal models offers distinct advantages resulting in new insights into physiologic processes and the pathophysiology and treatment of diseases.
Collapse
|
23
|
Kitching AR, Hickey MJ. Immune cell behaviour and dynamics in the kidney - insights from in vivo imaging. Nat Rev Nephrol 2022; 18:22-37. [PMID: 34556836 DOI: 10.1038/s41581-021-00481-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/11/2021] [Indexed: 02/08/2023]
Abstract
The actions of immune cells within the kidney are of fundamental importance in kidney homeostasis and disease. In disease settings such as acute kidney injury, anti-neutrophil cytoplasmic antibody-associated vasculitis, lupus nephritis and renal transplant rejection, immune cells resident within the kidney and those recruited from the circulation propagate inflammatory responses with deleterious effects on the kidney. As in most forms of inflammation, intravital imaging - particularly two-photon microscopy - has been critical to our understanding of immune cell responses in the renal microvasculature and interstitium, enabling visualization of immune cell dynamics over time rather than statically. These studies have demonstrated differences in the recruitment and function of these cells from those in more conventional vascular beds, and provided a wealth of information on the actions of blood-borne immune cells such as neutrophils, monocytes and T cells, as well as kidney-resident mononuclear phagocytes, in a range of diseases affecting different kidney compartments. In particular, in vivo imaging has furthered our understanding of leukocyte function within the glomerulus in acute glomerulonephritis, and in the tubulointerstitium and interstitial microvasculature during acute kidney injury and following transplantation, revealing mechanisms of immune surveillance, antigen presentation and inflammation in the kidney.
Collapse
Affiliation(s)
- A Richard Kitching
- Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Clayton, Victoria, Australia. .,Departments of Nephrology and Paediatric Nephrology, Monash Medical Centre, Clayton, Victoria, Australia.
| | - Michael J Hickey
- Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Clayton, Victoria, Australia
| |
Collapse
|
24
|
Isaksson GL, Nielsen MB, Hinrichs GR, Krogstrup NV, Zachar R, Stubmark H, Svenningsen P, Madsen K, Bistrup C, Jespersen B, Birn H, Palarasah Y, Jensen BL. Proteinuria is accompanied by intratubular complement activation and apical membrane deposition of C3dg and C5b-9 in kidney transplant recipients. Am J Physiol Renal Physiol 2021; 322:F150-F163. [PMID: 34927448 PMCID: PMC8791842 DOI: 10.1152/ajprenal.00300.2021] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Proteinuria predicts accelerated decline in kidney function in kidney transplant recipients (KTRs). We hypothesized that aberrant filtration of complement factors causes intraluminal activation, apical membrane attack on tubular cells, and progressive injury. Biobanked samples from two previous studies in albuminuric KTRs were used. The complement-activation split products C3c, C3dg, and soluble C5b-9-associated C9 neoantigen were analyzed by ELISA in urine and plasma using neoepitope-specific antibodies. Urinary extracellular vesicles (uEVs) were enriched by lectin and immunoaffinity isolation and analyzed by immunoblot analysis. Urine complement excretion increased significantly in KTRs with an albumin-to-creatinine ratio of ≥300 mg/g compared with <30 mg/g. Urine C3dg and C9 neoantigen excretion correlated significantly to changes in albumin excretion from 3 to 12 mo after transplantation. Fractional excretion of C9 neoantigen was significantly higher than for albumin, indicating postfiltration generation. C9 neoantigen was detected in uEVs in six of the nine albuminuric KTRs but was absent in non-albuminuric controls (n = 8). In C9 neoantigen-positive KTRs, lectin affinity enrichment of uEVs from the proximal tubules yielded signal for iC3b, C3dg, C9 neoantigen, and Na+-glucose transporter 2 but only weakly for aquaporin 2. Coisolation of podocyte markers and Tamm–Horsfall protein was minimal. Our findings show that albuminuria is associated with aberrant filtration and intratubular activation of complement with deposition of C3 activation split products and C5b-9-associated C9 neoantigen on uEVs from the proximal tubular apical membrane. Intratubular complement activation may contribute to progressive kidney injury in proteinuric kidney grafts. NEW & NOTEWORTHY The present study proposes a mechanistic coupling between proteinuria and aberrant filtration of complement precursors, intratubular complement activation, and apical membrane attack in kidney transplant recipients. C3dg and C5b-9-associated C9 neoantigen associate with proximal tubular apical membranes as demonstrated in urine extracellular vesicles. The discovery suggests intratubular complement as a mediator between proteinuria and progressive kidney damage. Inhibitors of soluble and/or luminal complement activation with access to the tubular lumen may be beneficial.
Collapse
Affiliation(s)
- Gustaf Lissel Isaksson
- Dept. of Molecular Medicine - Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark.,Dept. of Nephrology, Odense University Hospital, Odense, Denmark
| | - Marie Bodilsen Nielsen
- Dept. of Renal Medicine, Aarhus University Hospital, Aarhus, Denmark.,Dept of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Gitte Rye Hinrichs
- Dept. of Molecular Medicine - Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark.,Dept. of Nephrology, Odense University Hospital, Odense, Denmark
| | | | - Rikke Zachar
- Dept. of Molecular Medicine - Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark
| | - Heidi Stubmark
- Dept. of Molecular Medicine - Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark
| | - Per Svenningsen
- Dept. of Molecular Medicine - Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark
| | - Kirsten Madsen
- Dept. of Molecular Medicine - Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark.,Dept. of Pathology, Odense University Hospital, Odense, Denmark
| | - Claus Bistrup
- Dept. of Nephrology, Odense University Hospital, Odense, Denmark.,Dept. of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Bente Jespersen
- Dept. of Renal Medicine, Aarhus University Hospital, Aarhus, Denmark.,Dept. of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | | | - Henrik Birn
- Dept. of Renal Medicine, Aarhus University Hospital, Aarhus, Denmark.,Dept of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Yaseelan Palarasah
- Dept. of Molecular Medicine - Cancer and Inflammation Research, University of Southern Denmark, Odense, Denmark
| | - Boye L Jensen
- Dept. of Molecular Medicine - Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
25
|
Ballermann BJ, Nyström J, Haraldsson B. The Glomerular Endothelium Restricts Albumin Filtration. Front Med (Lausanne) 2021; 8:766689. [PMID: 34912827 PMCID: PMC8667033 DOI: 10.3389/fmed.2021.766689] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Accepted: 11/05/2021] [Indexed: 12/29/2022] Open
Abstract
Inflammatory activation and/or dysfunction of the glomerular endothelium triggers proteinuria in many systemic and localized vascular disorders. Among them are the thrombotic microangiopathies, many forms of glomerulonephritis, and acute inflammatory episodes like sepsis and COVID-19 illness. Another example is the chronic endothelial dysfunction that develops in cardiovascular disease and in metabolic disorders like diabetes. While the glomerular endothelium is a porous sieve that filters prodigious amounts of water and small solutes, it also bars the bulk of albumin and large plasma proteins from passing into the glomerular filtrate. This endothelial barrier function is ascribed predominantly to the endothelial glycocalyx with its endothelial surface layer, that together form a relatively thick, mucinous coat composed of glycosaminoglycans, proteoglycans, glycolipids, sialomucins and other glycoproteins, as well as secreted and circulating proteins. The glycocalyx/endothelial surface layer not only covers the glomerular endothelium; it extends into the endothelial fenestrae. Some glycocalyx components span or are attached to the apical endothelial cell plasma membrane and form the formal glycocalyx. Other components, including small proteoglycans and circulating proteins like albumin and orosomucoid, form the endothelial surface layer and are bound to the glycocalyx due to weak intermolecular interactions. Indeed, bound plasma albumin is a major constituent of the endothelial surface layer and contributes to its barrier function. A role for glomerular endothelial cells in the barrier of the glomerular capillary wall to protein filtration has been demonstrated by many elegant studies. However, it can only be fully understood in the context of other components, including the glomerular basement membrane, the podocytes and reabsorption of proteins by tubule epithelial cells. Discovery of the precise mechanisms that lead to glycocalyx/endothelial surface layer disruption within glomerular capillaries will hopefully lead to pharmacological interventions that specifically target this important structure.
Collapse
Affiliation(s)
| | - Jenny Nyström
- Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| | - Börje Haraldsson
- Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
26
|
Gong E, Perin L, Da Sacco S, Sedrakyan S. Emerging Technologies to Study the Glomerular Filtration Barrier. Front Med (Lausanne) 2021; 8:772883. [PMID: 34901088 PMCID: PMC8655839 DOI: 10.3389/fmed.2021.772883] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 11/04/2021] [Indexed: 11/16/2022] Open
Abstract
Kidney disease is characterized by loss of glomerular function with clinical manifestation of proteinuria. Identifying the cellular and molecular changes that lead to loss of protein in the urine is challenging due to the complexity of the filtration barrier, constituted by podocytes, glomerular endothelial cells, and glomerular basement membrane. In this review, we will discuss how technologies like single cell RNA sequencing and bioinformatics-based spatial transcriptomics, as well as in vitro systems like kidney organoids and the glomerulus-on-a-chip, have contributed to our understanding of glomerular pathophysiology. Knowledge gained from these studies will contribute toward the development of personalized therapeutic approaches for patients affected by proteinuric diseases.
Collapse
Affiliation(s)
- Emma Gong
- Division of Urology, GOFARR Laboratory for Organ Regenerative Research and Cell Therapeutics, Children's Hospital Los Angeles, Saban Research Institute, Los Angeles, CA, United States
| | - Laura Perin
- Division of Urology, GOFARR Laboratory for Organ Regenerative Research and Cell Therapeutics, Children's Hospital Los Angeles, Saban Research Institute, Los Angeles, CA, United States.,Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Stefano Da Sacco
- Division of Urology, GOFARR Laboratory for Organ Regenerative Research and Cell Therapeutics, Children's Hospital Los Angeles, Saban Research Institute, Los Angeles, CA, United States.,Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Sargis Sedrakyan
- Division of Urology, GOFARR Laboratory for Organ Regenerative Research and Cell Therapeutics, Children's Hospital Los Angeles, Saban Research Institute, Los Angeles, CA, United States.,Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
27
|
Yang Y, Kowalkowski K, Ciurlionis R, Buck WR, Glaser KB, Albert DH, Blomme EAG. Identification of VEGF Signaling Inhibition-Induced Glomerular Injury in Rats through Site-Specific Urinary Biomarkers. Int J Mol Sci 2021; 22:ijms222312629. [PMID: 34884436 PMCID: PMC8657489 DOI: 10.3390/ijms222312629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 11/15/2021] [Accepted: 11/17/2021] [Indexed: 12/05/2022] Open
Abstract
Cancer therapies targeting the vascular endothelial growth factor (VEGF) signaling pathway can lead to renal damage by disrupting the glomerular ultrafiltration apparatus. The objective of the current study was to identify sensitive biomarkers for VEGF inhibition-induced glomerular changes in rats. Male Sprague-Dawley rats were administered an experimental VEGF receptor (VEGFR) inhibitor, ABT-123, for seven days to investigate the correlation of several biomarkers with microscopic and ultrastructural changes. Glomeruli obtained by laser capture microdissection were also subjected to gene expression analysis to investigate the underlying molecular events of VEGFR inhibition in glomerulus. ABT-123 induced characteristic glomerular ultrastructural changes in rats, including fusion of podocyte foot processes, the presence of subendothelial electron-dense deposits, and swelling and loss of fenestrations in glomerular endothelium. The subtle morphological changes cannot be detected with light microscopy or by changes in standard clinical chemistry and urinalysis. However, urinary albumin increased 44-fold as early as Day three. Urinary β2-microglobulin levels were also increased. Other urinary biomarkers that are typically associated with tubular injury were not significantly impacted. Such patterns in urinary biomarkers can provide valuable diagnostic insight to VEGF inhibition therapy-induced glomeruli injuries.
Collapse
Affiliation(s)
- Yi Yang
- Correspondence: ; Tel.: +1-847-937-8893
| | | | | | | | | | | | | |
Collapse
|
28
|
Wang H, Zhang S, Guo J. Lipotoxic Proximal Tubular Injury: A Primary Event in Diabetic Kidney Disease. Front Med (Lausanne) 2021; 8:751529. [PMID: 34760900 PMCID: PMC8573085 DOI: 10.3389/fmed.2021.751529] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Accepted: 09/27/2021] [Indexed: 01/23/2023] Open
Abstract
The pathogenesis of diabetic nephropathy is a complex process that has a great relationship with lipotoxicity. Since the concept of “nephrotoxicity” was proposed, many studies have confirmed that lipotoxicity plays a significant role in the progression of diabetic nephropathy and causes various renal dysfunction. This review will make a brief summary of renal injury caused by lipotoxicity that occurs primarily and predominantly in renal tubules during diabetic progression, further leading to glomerular dysfunction. The latest research suggests that lipotoxicity-mediated tubular injury may be a major event in diabetic nephropathy.
Collapse
Affiliation(s)
- Hua Wang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shu Zhang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jia Guo
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Department of Nephrology, Nephropathy Research Institutes of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
29
|
Uchida Y, Torisu K, Ueki K, Tsuruya K, Nakano T, Kitazono T. Autophagy gene ATG7 regulates albumin transcytosis in renal tubule epithelial cells. Am J Physiol Renal Physiol 2021; 321:F572-F586. [PMID: 34541900 DOI: 10.1152/ajprenal.00172.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 09/13/2021] [Indexed: 01/03/2023] Open
Abstract
Receptor-mediated albumin transport in proximal tubule epithelial cells (PTECs) is important to control proteinuria. Autophagy is an evolutionarily conserved degradation pathway, and its role in intracellular trafficking through interactions with the endocytic pathway has recently been highlighted. Here, we determined whether autophagy regulates albumin transcytosis in PTECs and suppresses albumin-induced cytotoxicity using human proximal tubule (HK-2) cells. The neonatal Fc receptor (FcRn), a receptor for albumin transcytosis, is partially colocalized with autophagosomes. Recycling of FcRn was attenuated, and FcRn accumulated in autophagy-related 7 (ATG7) knockdown HK-2 cells. Colocalization of FcRn with RAB7-positive late endosomes and RAB11-positive recycling endosomes was reduced in ATG7 knockdown cells, which decreased recycling of FcRn to the plasma membrane. In ATG7 or autophagy-related 5 (ATG5) knockdown cells and Atg5 or Atg7 knockout mouse embryonic fibroblasts, albumin transcytosis was significantly reduced and intracellular albumin accumulation was increased. Finally, the release of kidney injury molecule-1, a marker of tubule injury, from ATG7 or ATG5 knockdown cells was increased in response to excess albumin. In conclusion, suppression of autophagy in tubules impairs FcRn transport, thereby inhibiting albumin transcytosis. The resulting accumulation of albumin induces cytotoxicity in tubules.NEW & NOTEWORTHY Albumin transport in proximal tubule epithelial cells (PTECs) is important to control proteinuria. The neonatal Fc receptor (FcRn), a receptor for albumin transcytosis, is partially colocalized with autophagosomes. Recycling of FcRn to the plasma membrane was decreased in autophagy-related 7 (ATG7) knockdown cells. In addition, albumin transcytosis was decreased in ATG7 or autophagy-related 5 (ATG5) knockdown PTECs. Finally, release of kidney injury molecule-1 from ATG7 or ATG5 knockdown cells was increased in response to excess albumin.
Collapse
Affiliation(s)
- Yushi Uchida
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kumiko Torisu
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- Department of Integrated Therapy for Chronic Kidney Disease, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kenji Ueki
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | | | - Toshiaki Nakano
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takanari Kitazono
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
30
|
Moriyama T, Hasegawa F, Miyabe Y, Akiyama K, Karasawa K, Uchida K, Nitta K. Intracellular trafficking pathway of albumin in glomerular epithelial cells. Biochem Biophys Res Commun 2021; 574:97-103. [PMID: 34450430 DOI: 10.1016/j.bbrc.2021.08.043] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 08/18/2021] [Indexed: 10/20/2022]
Abstract
The intracellular trafficking pathway of albumin in podocytes remains controversial. We therefore analysed albumin endocytosis through caveolae, subsequent transcytosis, and exocytosis. In Western blot and immunofluorescence analysis in vitro, methyl-beta-cyclodextrin (MBCD) treatment significantly decreased the expression of caveolin-1 and albumin in cultured human podocytes after incubation with albumin; additionally, MBCD interfered with albumin endocytosis through caveolae in the experiment using Transwell plates. In the immunofluorescence analysis, albumin was incubated with cultured human podocytes, and colocalisation analysis with organelles and cytoskeletons in the podocytes showed that albumin particles colocalised with caveolin-1 and Fc-receptor but not clathrin in endocytosis, colocalised with actin cytoskeleton but not microtubules in transcytosis, and colocalised with early endosomes and lysosomes but not proteasome, endoplasmic reticulum, or Golgi apparatus. In the electron microscopic analysis of podocytes in nephrotic syndrome model mice, gold-labelled albumin was shown as endocytosis, transcytosis, and exocytosis with caveolae. These results indicate the intracellular trafficking of albumin through podocytes. Albumin enters through caveolae with the Fc-receptor, moves along actin, and reaches the early endosome, where some of them are sorted for lysosomal degradation, and others are directly transported outside the cells through exocytosis. This intracellular pathway may be a new aetiological hypothesis for albuminuria.
Collapse
Affiliation(s)
- Takahito Moriyama
- Department of Nephrology, Tokyo Women's Medical University, Tokyo, Japan.
| | - Fumio Hasegawa
- Department of Nephrology, Tokyo Women's Medical University, Tokyo, Japan.
| | - Yoei Miyabe
- Department of Nephrology, Tokyo Women's Medical University, Tokyo, Japan.
| | - Kenichi Akiyama
- Department of Nephrology, Tokyo Women's Medical University, Tokyo, Japan.
| | - Kazunori Karasawa
- Department of Nephrology, Tokyo Women's Medical University, Tokyo, Japan.
| | - Keiko Uchida
- Department of Nephrology, Tokyo Women's Medical University, Tokyo, Japan.
| | - Kosaku Nitta
- Department of Nephrology, Tokyo Women's Medical University, Tokyo, Japan.
| |
Collapse
|
31
|
Ormonde C, Laranjinha I, Gil C, Gonçalves M, A Gaspar A. Glycosuria in primary glomerulopathies: prevalence and prognostic significance. J Bras Nefrol 2021; 44:26-31. [PMID: 34424258 PMCID: PMC8943878 DOI: 10.1590/2175-8239-jbn-2021-0115] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 06/16/2021] [Indexed: 11/21/2022] Open
Abstract
Introduction: Tubular damage is common in glomerular diseases (GD). Glycosuria is a marker of tubular dysfunction and may be used to detect tubular lesion and CKD progression. The aim of this study was to evaluate the prevalence and prognostic value of glycosuria at the time of diagnosis in primary glomerulopathies (PG). Methods: We conducted a 24-month retrospective study in patients diagnosed with PG in our center between 2009 and 2020. We excluded diabetic patients, use of SGLT2 inhibitors, transplant patients, and secondary GD. Patients were divided in two groups according to their glycosuria status at diagnosis. Results: We studied 115 patients. Global prevalence of glycosuria was 10% (n=11) and membranous nephropathy (MN) had the highest prevalence (n=5, 17.9%). We found that patients with glycosuria had higher serum creatinine (2.4 vs. 1.2 mg/dL, p=0.030), higher albuminuria (4.8 vs. 1.9 g/g, p=0.004), and lower serum albumin (2.3 vs. 3.2 g/dL, p=0.021). We did not find association with histological prognostic factors. At the end of follow-up, patients with glycosuria had higher prevalence of the composite outcome of stage 5D CKD or 50% increase in basal SCr (45.5% vs. 17.3%, p=0.037). In patients with MN, results were similar but we were able to find an association of glycosuria with more severe interstitial fibrosis and tubular atrophy (25.0 vs. 0.0 %, p=0.032). Conclusion: Ten percent of our patients with PG have glycosuria. Glycosuria at the time of diagnosis was associated with more severe clinical presentation and worst renal outcome. The association with higher albuminuria suggests that tubular function has an impact on the severity and outcomes of PG.
Collapse
Affiliation(s)
- Carolina Ormonde
- Hospital do Divino Espírito Santo, Avenida D.Manuel I, 9500-370, Ponta Delgada, Portugal
| | - Ivo Laranjinha
- Hospital de Santa Cruz, Centro Hospitalar Lisboa Ocidental, Avenida Prof. Dr. Reinaldo dos Santos, 2790-134, Carnaxide, Portugal
| | - Célia Gil
- Hospital de Santa Cruz, Centro Hospitalar Lisboa Ocidental, Avenida Prof. Dr. Reinaldo dos Santos, 2790-134, Carnaxide, Portugal
| | - Margarida Gonçalves
- Hospital de Santa Cruz, Centro Hospitalar Lisboa Ocidental, Avenida Prof. Dr. Reinaldo dos Santos, 2790-134, Carnaxide, Portugal
| | - August A Gaspar
- Hospital de Santa Cruz, Centro Hospitalar Lisboa Ocidental, Avenida Prof. Dr. Reinaldo dos Santos, 2790-134, Carnaxide, Portugal
| |
Collapse
|
32
|
Wang D, Sant S, Ferrell N. A Biomimetic In Vitro Model of the Kidney Filtration Barrier Using Tissue-Derived Glomerular Basement Membrane. Adv Healthc Mater 2021; 10:e2002275. [PMID: 34218528 DOI: 10.1002/adhm.202002275] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 05/24/2021] [Indexed: 01/28/2023]
Abstract
The glomerular filtration barrier (GFB) filters the blood to remove toxins while retaining high molecular weight proteins in the circulation. The glomerular basement membrane (GBM) and podocytes, highly specialized epithelial cells, are critical components of the filtration barrier. The GBM serves as a physical barrier to passage of molecules into the filtrate. Podocytes adhere to the filtrate side of the GBM and further restrict passage of high molecular weight molecules into the filtrate. Here, a 3D cell culture model of the glomerular filtration barrier to evaluate the role of the GBM and podocytes in mediating molecular diffusion is developed. GBM is isolated from mammalian kidneys to recapitulate the composition and mechanics of the in vivo basement membrane. The GFB model exhibits molecular selectivity that is comparable to the in vivo filtration barrier. The GBM alone provides a stringent barrier to passage of albumin and Ficoll. Podocytes further restrict molecular diffusion. Damage to the GBM that is typical of diabetic kidney disease is simulated using hypochlorous acid and results in increased molecular diffusion. This system can serve as a platform to evaluate the effects of GBM damage, podocyte injury, and reciprocal effects of altered podocyte-GBM interactions on kidney microvascular permeability.
Collapse
Affiliation(s)
- Dan Wang
- Department of Medicine, Division of Nephrology, Vanderbilt University Medical Center, 1161 21st Ave. South, Nashville, TN, 37232, USA
| | - Snehal Sant
- Department of Medicine, Division of Nephrology, Vanderbilt University Medical Center, 1161 21st Ave. South, Nashville, TN, 37232, USA
| | - Nicholas Ferrell
- Department of Medicine, Division of Nephrology, Vanderbilt University Medical Center, 1161 21st Ave. South, Nashville, TN, 37232, USA
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
- Vanderbilt Center for Kidney Disease, S3223 Medical Center North, Nashville, TN, 37232, USA
| |
Collapse
|
33
|
Gburek J, Konopska B, Gołąb K. Renal Handling of Albumin-From Early Findings to Current Concepts. Int J Mol Sci 2021; 22:ijms22115809. [PMID: 34071680 PMCID: PMC8199105 DOI: 10.3390/ijms22115809] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 05/20/2021] [Accepted: 05/25/2021] [Indexed: 12/29/2022] Open
Abstract
Albumin is the main protein of blood plasma, lymph, cerebrospinal and interstitial fluid. The protein participates in a variety of important biological functions, such as maintenance of proper colloidal osmotic pressure, transport of important metabolites and antioxidant action. Synthesis of albumin takes place mainly in the liver, and its catabolism occurs mostly in vascular endothelium of muscle, skin and liver, as well as in the kidney tubular epithelium. Long-lasting investigation in this area has delineated the principal route of its catabolism involving glomerular filtration, tubular endocytic uptake via the multiligand scavenger receptor tandem—megalin and cubilin-amnionless complex, as well as lysosomal degradation to amino acids. However, the research of the last few decades indicates that also additional mechanisms may operate in this process to some extent. Direct uptake of albumin in glomerular podocytes via receptor for crystallizable region of immunoglobulins (neonatal FC receptor) was demonstrated. Additionally, luminal recycling of short peptides into the bloodstream and/or back into tubular lumen or transcytosis of whole molecules was suggested. The article discusses the molecular aspects of these processes and presents the major findings and controversies arising in the light of the research concerning the last decade. Their better characterization is essential for further research into pathophysiology of proteinuric renal failure and development of effective therapeutic strategies.
Collapse
|
34
|
Martins JR, Haenni D, Bugarski M, Polesel M, Schuh C, Hall AM. Intravital kidney microscopy: entering a new era. Kidney Int 2021; 100:527-535. [PMID: 34015315 DOI: 10.1016/j.kint.2021.02.042] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 02/01/2021] [Accepted: 02/16/2021] [Indexed: 02/07/2023]
Abstract
The development of intravital imaging with multiphoton microscopy has had a major impact on kidney research. It provides the unique opportunity to visualize dynamic behavior of cells and organelles in their native environment and to relate this to the complex 3-dimensional structure of the organ. Moreover, changes in cell/organelle function can be followed in real time in response to physiological interventions or disease-causing insults. However, realizing the enormous potential of this exciting approach has necessitated overcoming several substantial practical hurdles. In this article, we outline the nature of these challenges and how a variety of technical advances have provided effective solutions. In particular, improvements in laser/microscope technology, fluorescent probes, transgenic animals, and abdominal windows are collectively making previously opaque processes visible. Meanwhile, the rise of machine learning-based image analysis is facilitating the rapid generation of large amounts of quantitative data, amenable to deeper statistical interrogation. Taken together, the increased capabilities of multiphoton imaging are opening up huge new possibilities to study structure-function relationships in the kidney in unprecedented detail. In addition, they are yielding important new insights into cellular mechanisms of tissue damage, repair, and adaptive remodeling during disease states. Thus, intravital microscopy is truly entering an exciting new era in translational kidney research.
Collapse
Affiliation(s)
- Joana R Martins
- Center for Microscopy and Image Analysis, University of Zurich, Zurich, Switzerland
| | - Dominik Haenni
- Center for Microscopy and Image Analysis, University of Zurich, Zurich, Switzerland; Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | - Milica Bugarski
- Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | | | - Claus Schuh
- Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | - Andrew M Hall
- Institute of Anatomy, University of Zurich, Zurich, Switzerland; Department of Nephrology, University Hospital Zurich, Zurich, Switzerland.
| |
Collapse
|
35
|
Affiliation(s)
- Thomas Benzing
- From Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine, and University Hospital Cologne, and the Excellence Cluster CECAD, University of Cologne, Cologne, Germany (T.B.); and the Section of Nephrology, Department of Medicine, Boston Medical Center and Boston University School of Medicine, Boston (D.S.)
| | - David Salant
- From Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine, and University Hospital Cologne, and the Excellence Cluster CECAD, University of Cologne, Cologne, Germany (T.B.); and the Section of Nephrology, Department of Medicine, Boston Medical Center and Boston University School of Medicine, Boston (D.S.)
| |
Collapse
|
36
|
Dunn KW, Molitoris BA, Dagher PC. The Indiana O'Brien Center for Advanced Renal Microscopic Analysis. Am J Physiol Renal Physiol 2021; 320:F671-F682. [PMID: 33682441 DOI: 10.1152/ajprenal.00007.2021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The Indiana O'Brien Center for Advanced Microscopic Analysis is a National Institutes of Health (NIH) P30-funded research center dedicated to the development and dissemination of advanced methods of optical microscopy to support renal researchers throughout the world. The Indiana O'Brien Center was founded in 2002 as an NIH P-50 project with the original goal of helping researchers realize the potential of intravital multiphoton microscopy as a tool for understanding renal physiology and pathophysiology. The center has since expanded into the development and implementation of large-scale, high-content tissue cytometry. The advanced imaging capabilities of the center are made available to renal researchers worldwide via collaborations and a unique fellowship program. Center outreach is accomplished through an enrichment core that oversees a seminar series, an informational website, and a biennial workshop featuring hands-on training from members of the Indiana O'Brien Center and imaging experts from around the world.
Collapse
Affiliation(s)
- Kenneth W Dunn
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Bruce A Molitoris
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Pierre C Dagher
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| |
Collapse
|
37
|
Abstract
Upper respiratory and pulmonary diseases are the primary manifestations of coronavirus disease 2019 (COVID-19). However, kidney involvement has also been recognized and extensively described. A large percentage of affected patients present with acute kidney injury (AKI). However, specific phenotypic aspects of AKI or other renal manifestations of COVID-19 remain sparsely characterized. Many reports indicate that proteinuria can be detected in AKI associated with COVID-19 (CoV-AKI) despite CoV-AKI being largely described as a form of acute tubular injury. On the other hand, individuals of African ancestry with the high-risk APOL1 genotype are uniquely at risk of developing collapsing glomerulopathy when they are infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the entity now known as COVID-19-associated nephropathy (COVAN). Patients with COVAN typically present with nephrotic-range proteinuria. The exact incidence of proteinuria in COVID-19 is unclear due to heterogeneity in the frequency with which proteinuria has been assessed in cases of COVID-19, as well as methodological differences in the way proteinuria is measured and/or reported. In this review we discuss the current evidence of proteinuria as a manifestation of COVID-19 and elaborate on potential pathophysiological mechanisms associated with it.
Collapse
Affiliation(s)
- Muner M B Mohamed
- Department of Nephrology, Ochsner Health System, New Orleans, LA, USA
| | - Juan Carlos Q Velez
- Department of Nephrology, Ochsner Health System, New Orleans, LA, USA
- Ochsner Clinical School, University of Queensland (Australia), New Orleans, LA, USA
| |
Collapse
|
38
|
Vaghela R, Arkudas A, Horch RE, Hessenauer M. Actually Seeing What Is Going on - Intravital Microscopy in Tissue Engineering. Front Bioeng Biotechnol 2021; 9:627462. [PMID: 33681162 PMCID: PMC7925911 DOI: 10.3389/fbioe.2021.627462] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 01/26/2021] [Indexed: 12/21/2022] Open
Abstract
Intravital microscopy (IVM) study approach offers several advantages over in vitro, ex vivo, and 3D models. IVM provides real-time imaging of cellular events, which provides us a comprehensive picture of dynamic processes. Rapid improvement in microscopy techniques has permitted deep tissue imaging at a higher resolution. Advances in fluorescence tagging methods enable tracking of specific cell types. Moreover, IVM can serve as an important tool to study different stages of tissue regeneration processes. Furthermore, the compatibility of different tissue engineered constructs can be analyzed. IVM is also a promising approach to investigate host reactions on implanted biomaterials. IVM can provide instant feedback for improvising tissue engineering strategies. In this review, we aim to provide an overview of the requirements and applications of different IVM approaches. First, we will discuss the history of IVM development, and then we will provide an overview of available optical modalities including the pros and cons. Later, we will summarize different fluorescence labeling methods. In the final section, we will discuss well-established chronic and acute IVM models for different organs.
Collapse
Affiliation(s)
- Ravikumar Vaghela
- Department of Plastic and Hand Surgery, University Hospital of Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Andreas Arkudas
- Department of Plastic and Hand Surgery, University Hospital of Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Raymund E Horch
- Department of Plastic and Hand Surgery, University Hospital of Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Maximilian Hessenauer
- Department of Plastic and Hand Surgery, University Hospital of Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| |
Collapse
|
39
|
Prutskova NP, Seliverstova EV. The Effect of Protein Overload on
Reabsorption of Different Proteins in Frog Renal Tubules. J EVOL BIOCHEM PHYS+ 2021. [DOI: 10.1134/s0022093021010105] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
40
|
Pafundi PC, Garofalo C, Galiero R, Borrelli S, Caturano A, Rinaldi L, Provenzano M, Salvatore T, De Nicola L, Minutolo R, Sasso FC. Role of Albuminuria in Detecting Cardio-Renal Risk and Outcome in Diabetic Subjects. Diagnostics (Basel) 2021; 11:290. [PMID: 33673215 PMCID: PMC7918197 DOI: 10.3390/diagnostics11020290] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 02/09/2021] [Accepted: 02/10/2021] [Indexed: 12/29/2022] Open
Abstract
The clinical significance of albuminuria in diabetic subjects and the impact of its reduction on the main cardiorenal outcomes by different drug classes are among the most interesting research focuses of recent years. Although nephrologists and cardiologists have been paying attention to the study of proteinuria for years, currently among diabetics, increased urine albumin excretion ascertains the highest cardio-renal risk. In fact, diabetes is a condition by itself associated with a high-risk of both micro/macrovascular complications. Moreover, proteinuria reduction in diabetic subjects by several treatments lowers both renal and cardiovascular disease progression. The 2019 joint ESC-EASD guidelines on diabetes, prediabetes and cardiovascular (CV) disease assign to proteinuria a crucial role in defining CV risk level in the diabetic patient. In fact, proteinuria by itself allows the diabetic patient to be staged at very high CV risk, thus affecting the choice of anti-hyperglycemic drug class. The purpose of this review is to present a clear update on the role of albuminuria as a cardio-renal risk marker, starting from pathophysiological mechanisms in support of this role. Besides this, we will show the prognostic value in observational studies, as well as randomized clinical trials (RCTs) demonstrating the potential improvement of cardio-renal outcomes in diabetic patients by reducing proteinuria.
Collapse
Affiliation(s)
- Pia Clara Pafundi
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Piazza Luigi Miraglia 2, 80138 Naples, Italy; (P.C.P.); (C.G.); (R.G.); (S.B.); (A.C.); (L.R.); (L.D.N.)
| | - Carlo Garofalo
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Piazza Luigi Miraglia 2, 80138 Naples, Italy; (P.C.P.); (C.G.); (R.G.); (S.B.); (A.C.); (L.R.); (L.D.N.)
| | - Raffaele Galiero
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Piazza Luigi Miraglia 2, 80138 Naples, Italy; (P.C.P.); (C.G.); (R.G.); (S.B.); (A.C.); (L.R.); (L.D.N.)
| | - Silvio Borrelli
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Piazza Luigi Miraglia 2, 80138 Naples, Italy; (P.C.P.); (C.G.); (R.G.); (S.B.); (A.C.); (L.R.); (L.D.N.)
| | - Alfredo Caturano
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Piazza Luigi Miraglia 2, 80138 Naples, Italy; (P.C.P.); (C.G.); (R.G.); (S.B.); (A.C.); (L.R.); (L.D.N.)
| | - Luca Rinaldi
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Piazza Luigi Miraglia 2, 80138 Naples, Italy; (P.C.P.); (C.G.); (R.G.); (S.B.); (A.C.); (L.R.); (L.D.N.)
| | - Michele Provenzano
- Renal Unit, Department of Health Sciences, “Magna Graecia” University, Viale Europa, 88100 Catanzaro, Italy;
| | - Teresa Salvatore
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Via De Crecchio 7, 80138 Naples, Italy;
| | - Luca De Nicola
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Piazza Luigi Miraglia 2, 80138 Naples, Italy; (P.C.P.); (C.G.); (R.G.); (S.B.); (A.C.); (L.R.); (L.D.N.)
| | - Roberto Minutolo
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Piazza Luigi Miraglia 2, 80138 Naples, Italy; (P.C.P.); (C.G.); (R.G.); (S.B.); (A.C.); (L.R.); (L.D.N.)
| | - Ferdinando Carlo Sasso
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Piazza Luigi Miraglia 2, 80138 Naples, Italy; (P.C.P.); (C.G.); (R.G.); (S.B.); (A.C.); (L.R.); (L.D.N.)
| |
Collapse
|
41
|
Gong Q, Yin J, Wang M, He L, Lei F, Luo Y, Yang S, Feng Y, Li J, Du L. Comprehensive study of dexamethasone on albumin biogenesis during normal and pathological renal conditions. PHARMACEUTICAL BIOLOGY 2020; 58:1252-1262. [PMID: 33332210 PMCID: PMC7751422 DOI: 10.1080/13880209.2020.1855214] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Accepted: 11/19/2020] [Indexed: 06/12/2023]
Abstract
CONTEXT Dexamethasone (DXM) has an anti-immunoinflammatory effect, and is often used in acute kidney injury (AKI). However, the effects of DXM on albumin (ALB) have not been fully studied. OBJECTIVE To investigate the effects of DXM on ALB production and renal function. MATERIALS AND METHODS Male Wistar rats were divided into normal and DXM groups (0.25, 0.5, 1 mg/kg for 5 days) (n = 15) for a dose-dependent study. Rats were divided into normal group and DXM groups (0.5 mg/kg for 3, 5, 7 days) (n = 9) for a time-dependent study. In AKI experiment, rats were divided into normal (saline), cisplatin (CP, 5 mg/kg, i.v.), CP + DXM groups (0.25, 0.5 and 1 mg/kg, i.m.) (n = 16). The blood and the organs were isolated for analysis. RESULTS In normal, serum ALB (sALB) and serum total protein (sTP) increased in DXM group with sALB increased 19.8-32.2% (from small to large dosages); and 30.2-32.5.6% (from 3 to 7 days of DXM); sTP 15.7-22.6% and 14.2-24.3%; urine ALB (uALB) 31.5-392.3%, and 1047.2-1390.8%; urine TP (uTP) 0.68-173.1% and 98.0-504.9%, compared with normal groups. DXM increased the mRNA expression of Cebp and Hnf, suppressing podocin. In AKI, DXM decreased serum BUN (53.7%), serum Cre (73.4%), sALB (30.0%), sTP (18.7%), uALB (74.5%), uTP (449.3%), rescuing the suppressed podocin in kidney. CONCLUSIONS DXM acts on Cebp and Hnf and promotes ALB production. This finding helps to evaluate the rationale of DXM for kidney injury.
Collapse
Affiliation(s)
- Qin Gong
- School of Pharmacy, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
- Pharmacology Laboratory, State Key Laboratory of Innovative Drugs and Efficient Energy-Saving Pharmaceutical Equipment, Nanchang, China
| | - Jilei Yin
- Department of Traditional Chinese Medicine, Jiangsu Union Technical Institute Lianyungang Branch Institute of Traditional Chinese Medicine, Lianyungang, China
| | - Mulan Wang
- Pharmacology Laboratory, State Key Laboratory of Innovative Drugs and Efficient Energy-Saving Pharmaceutical Equipment, Nanchang, China
| | - Luling He
- School of Pharmacy, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
- Pharmacology Laboratory, State Key Laboratory of Innovative Drugs and Efficient Energy-Saving Pharmaceutical Equipment, Nanchang, China
| | - Fan Lei
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Yingying Luo
- School of Pharmacy, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
- Pharmacology Laboratory, State Key Laboratory of Innovative Drugs and Efficient Energy-Saving Pharmaceutical Equipment, Nanchang, China
| | - Shilin Yang
- School of Pharmacy, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
- Pharmacology Laboratory, State Key Laboratory of Innovative Drugs and Efficient Energy-Saving Pharmaceutical Equipment, Nanchang, China
| | - Yulin Feng
- School of Pharmacy, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
- Pharmacology Laboratory, State Key Laboratory of Innovative Drugs and Efficient Energy-Saving Pharmaceutical Equipment, Nanchang, China
| | - Jun Li
- School of Pharmacy, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
- Pharmacology Laboratory, State Key Laboratory of Innovative Drugs and Efficient Energy-Saving Pharmaceutical Equipment, Nanchang, China
| | - Lijun Du
- School of Pharmacy, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
- Pharmacology Laboratory, State Key Laboratory of Innovative Drugs and Efficient Energy-Saving Pharmaceutical Equipment, Nanchang, China
- School of Life Sciences, Tsinghua University, Beijing, China
| |
Collapse
|
42
|
Cetin N, Kiraz ZK, Sav NM. Urine hepcidin, netrin-1, neutrophil gelatinase-associated lipocalin and C-C motif chemokine ligand 2 levels in multicystic dysplastic kidney. J Bras Nefrol 2020; 42:280-289. [PMID: 32818222 PMCID: PMC7657047 DOI: 10.1590/2175-8239-jbn-2019-0022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 04/22/2020] [Indexed: 11/23/2022] Open
Abstract
Introduction: Glomerular hyperfiltration may lead to proteinuria and chronic kidney disease
in unilateral multicystic dysplastic kidney (MCDK). We aimed to investigate
the urine neutrophil-gelatinase-associated lipocalin (NGAL), netrin-1,
hepcidin, and C-C motif chemokine ligand-2 (MCP-1/CCL-2) levels in patients
with MCDK. Methods: Thirty-two patients and 25 controls were included. The urine hepcidin,
netrin-1, NGAL, and MCP-1/CCL-2 levels were determined by ELISA. Results: The
patients had higher serum creatinine
(Cr) levels, urine albumin, and netrin-1/
Cr ratio with lower GFR. There were
positive correlations between urine
protein/Cr, MCP-1/CCL-2/Cr, and
netrin-1 with NGAL (r = 0.397, p =
0.031; r = 0.437, p = 0.041, r = 0.323, p
= 0.042, respectively). Urine netrin-1/Cr
was positively correlated with MCP-1/
CCL-2/Cr (r = 0.356, p = 0.045). There
were positive associations between the
presence of proteinuria and netrin-1/
Cr, MCP-1/CCL-2/Cr, and NGAL/Cr
[Odds ratio (OR): 1.423, p = 0.037,
OR: 1.553, p = 0.033, OR: 2.112, p
= 0.027, respectively)]. ROC curve
analysis showed that netrin-1/Cr,
MCP-1/CCL-2/Cr, and NGAL/Cr had
high predictive values for determining
proteinuria p = 0.027, p = 0.041,
p = 0.035, respectively). Urine hepcidin/
Cr was negatively correlated with
tubular phosphorus reabsorption and
was positively correlated with urine
NGAL/Cr (r = -0.418, p = 0.019; r
= 0.682, p = 0.000; respectively). Conclusions: MCP-1/CCL-2 may play a role in the development of proteinuria in MCDK.
Netrin-1 may be a protective factor against proteinuria-induced renal
injury. Urine hepcidin/Cr may reflect proximal tubule damage in MCDK. Urine
NGAL/Cr may be a predictor of tubule damage by proteinuria.
Collapse
|
43
|
Miyai N, Shiozaki M, Terada K, Takeshita T, Utsumi M, Miyashita K, Arita M. Exaggerated blood pressure response to exercise is associated with subclinical vascular impairment in healthy normotensive individuals. Clin Exp Hypertens 2020; 43:56-62. [PMID: 32799691 DOI: 10.1080/10641963.2020.1806292] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
BACKGROUND This study was designed to evaluate the possible association between an exaggerated blood pressure (BP) response to exercise and subclinical vascular impairment in normotensive individuals. METHODS The study participants consisted of 92 untreated normotensive men (aged 42 ± 9 years) without a history of cardiovascular disease or stroke. A graded exercise test was conducted using a bicycle ergometer, and the mean arterial pressure (MAP) during submaximal exercise was evaluated. The brachial-ankle pulse wave velocity (baPWV) was measured as an index of arterial stiffness. The second peak of radial systolic BP (SBP2) was used as an estimate of central BP. The albumin-to-creatinine ratio (ACR) values were determined as the mean of two nonconsecutive spot urine specimens. RESULTS Compared with individuals with a normal response (MAP z-score < +1.0, n = 60), those with an exaggerated BP response to exercise (MAP z-score ≥ +1.0, n = 32) exhibited significantly higher baPWV (1412 ± 158 vs. 1250 ± 140 cm/s), radial SBP2 (122 ± 11 vs. 106 ± 13 mmHg), and greater log-ACR (0.93 ± 0.30 vs. 0.59 ± 0.23 mg/gCre). Multiple regression analysis revealed that an exaggerated BP response to exercise was significantly associated with baPWV (β = 0.198, P= .043), radial SBP2 (β = 0.156, P = .049), and log-ACR (β = 0.276, P = .006) independent of potential confounding factors. CONCLUSIONS These results suggest that subclinical vascular impairment is associated with an exaggerated increase in BP during exercise even in the absence of clinical hypertension.
Collapse
Affiliation(s)
- Nobuyuki Miyai
- School of Health and Nursing Science, Wakayama Medical University , Wakayama, Japan
| | - Maki Shiozaki
- Department of Hygiene, School of Medicine, Wakayama Medical University , Wakayama, Japan
| | - Kazufumi Terada
- Faculty of Budo and Sport Studies, Tenri University , Nara, Japan
| | - Tatsuya Takeshita
- Department of Public Health, School of Medicine, Wakayama Medical University , Wakayama, Japan
| | - Miyoko Utsumi
- School of Health and Nursing Science, Wakayama Medical University , Wakayama, Japan
| | - Kazuhisa Miyashita
- Department of Hygiene, School of Medicine, Wakayama Medical University , Wakayama, Japan
| | - Mikio Arita
- School of Health and Nursing Science, Wakayama Medical University , Wakayama, Japan.,Department of Cardiology, Sumiya Rehabilitation Hospital , Wakayama, Japan
| |
Collapse
|
44
|
Hosohata K, Matsuoka H, Iwanaga K, Kumagai E. Urinary vanin-1 associated with chronic kidney disease in hypertensive patients: A pilot study. J Clin Hypertens (Greenwich) 2020; 22:1458-1465. [PMID: 32869948 PMCID: PMC8029830 DOI: 10.1111/jch.13959] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 05/05/2020] [Accepted: 05/14/2020] [Indexed: 12/12/2022]
Abstract
Hypertension and chronic kidney disease (CKD) are serious interrelated public health problems. Despite the monitoring and control of high blood pressure, symptoms of CKD are not usually apparent in its early stages. Previously, we reported the utility of urinary vanin-1 as an early biomarker of kidney injury in spontaneously hypertensive rats, but it remains unknown whether urinary vanin-1 is associated with CKD in humans. In this study, we estimated associations between urinary vanin-1 and parameters of kidney function in a cross-sectional study of hypertensive patients. We measured concentrations of vanin-1 using spot urine from 147 adult hypertensive patients (mean age, 72.8 years; 39.5% women). Patients were divided into 2 groups based on the median of the estimated glomerular filtration rate (eGFR). The group with eGFR < 60 mL/min per 1.73 m2 showed significantly higher levels of urinary vanin-1 than those with eGFR ≥ 60 mL/min per 1.73 m2. On univariate analysis, urinary vanin-1 as well as neutrophil gelatinase-associated lipocalin (NGAL) showed significant negative correlations with eGFR; however, multivariate analysis revealed that urinary vanin-1, but not NGAL, significantly correlated with eGFR. In addition, urinary vanin-1 had a significant positive correlation with the urinary protein-to-creatinine ratio (UPCR) (r = 0.21; P = .021) and albumin-to-creatinine ratio (UACR) (r = 0.61; P < .01). In conclusion, urinary vanin-1 is associated with lower eGFR and higher UPCR and UACR, and might be a potential marker of decreased kidney function in hypertensive patients. Further studies are needed to confirm these findings.
Collapse
Affiliation(s)
- Keiko Hosohata
- Education and Research Center of Clinical PharmacyOsaka University of Pharmaceutical SciencesOsakaJapan
| | | | - Kazunori Iwanaga
- Education and Research Center of Clinical PharmacyOsaka University of Pharmaceutical SciencesOsakaJapan
| | | |
Collapse
|
45
|
Butt L, Unnersjö-Jess D, Höhne M, Edwards A, Binz-Lotter J, Reilly D, Hahnfeldt R, Ziegler V, Fremter K, Rinschen MM, Helmstädter M, Ebert LK, Castrop H, Hackl MJ, Walz G, Brinkkoetter PT, Liebau MC, Tory K, Hoyer PF, Beck BB, Brismar H, Blom H, Schermer B, Benzing T. A molecular mechanism explaining albuminuria in kidney disease. Nat Metab 2020; 2:461-474. [PMID: 32694662 DOI: 10.1038/s42255-020-0204-y] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Accepted: 04/07/2020] [Indexed: 01/18/2023]
Abstract
Mammalian kidneys constantly filter large amounts of liquid, with almost complete retention of albumin and other macromolecules in the plasma. Breakdown of the three-layered renal filtration barrier results in loss of albumin into urine (albuminuria) across the wall of small renal capillaries, and is a leading cause of chronic kidney disease. However, exactly how the renal filter works and why its permeability is altered in kidney diseases is poorly understood. Here we show that the permeability of the renal filter is modulated through compression of the capillary wall. We collect morphometric data prior to and after onset of albuminuria in a mouse model equivalent to a human genetic disease affecting the renal filtration barrier. Combining quantitative analyses with mathematical modelling, we demonstrate that morphological alterations of the glomerular filtration barrier lead to reduced compressive forces that counteract filtration pressure, thereby resulting in capillary dilatation, and ultimately albuminuria. Our results reveal distinct functions of the different layers of the filtration barrier and expand the molecular understanding of defective renal filtration in chronic kidney disease.
Collapse
Affiliation(s)
- Linus Butt
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - David Unnersjö-Jess
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
- Royal Institute of Technology, Stockholm, Sweden
| | - Martin Höhne
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Aurelie Edwards
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Julia Binz-Lotter
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Dervla Reilly
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Robert Hahnfeldt
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Vera Ziegler
- Institute of Physiology, University of Regensburg, Regensburg, Germany
| | - Katharina Fremter
- Institute of Physiology, University of Regensburg, Regensburg, Germany
| | - Markus M Rinschen
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
- Center for Mass Spectrometry and Metabolomics, The Scripps Research Institute, La Jolla, CA, USA
| | - Martin Helmstädter
- Department of Medicine IV, Medical Center - University of Freiburg, Faculty of Medicine, and Signalling Research Centres BIOSS and CIBSS, University of Freiburg,, Freiburg, Germany
| | - Lena K Ebert
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Hayo Castrop
- Institute of Physiology, University of Regensburg, Regensburg, Germany
| | - Matthias J Hackl
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Gerd Walz
- Department of Medicine IV, Medical Center - University of Freiburg, Faculty of Medicine, and Signalling Research Centres BIOSS and CIBSS, University of Freiburg,, Freiburg, Germany
| | - Paul T Brinkkoetter
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Max C Liebau
- Department of Pediatrics and Center for Molecular Medicine, University of Cologne, Faculty of Medicine and University Hospital of Cologne, Cologne, Germany
| | - Kálmán Tory
- MTA-SE Lendület Nephrogenetic Laboratory, 1st Department of Pediatrics, Semmelweis University, Budapest, Hungary
| | - Peter F Hoyer
- University Children's Hospital, Clinic for Pediatrics II, University of Duisburg-Essen, Essen, Germany
| | - Bodo B Beck
- Institute of Human Genetics and Center for Molecular Medicine, University of Cologne, Faculty of Medicine and University Hospital of Cologne, Cologne, Germany
| | | | - Hans Blom
- Royal Institute of Technology, Stockholm, Sweden
| | - Bernhard Schermer
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
- CECAD, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Thomas Benzing
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany.
- CECAD, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany.
| |
Collapse
|
46
|
Reconsidering Garth Robinson: fluid flow and the glomerular filtration barrier. Curr Opin Nephrol Hypertens 2020; 29:273-279. [PMID: 32235269 DOI: 10.1097/mnh.0000000000000606] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW The goal of this review is to present recent models of the filtration barrier that may suggest mechanism-based treatments for proteinuric renal disease. The vast majority of renal failure occurs in diseases of glomerular proteinuria. The physiology of the filtration barrier remains incompletely understood, preventing invention of mechanism-based therapies. Research is currently dominated by molecular biology approaches to the kidney instead of engineering-based filtration and transport models. RECENT FINDINGS Reexamination of two older paradigms (basement membrane and slit diaphragm) and critical analysis of newer models may provide mechanistic insight to guide further research. We expand on our theory of podocyte-basement membrane mechanical interactions and speculate on mechanisms of action of the leading treatment for proteinuria, angiotensin blockade. SUMMARY Treatment of proteinuria remains largely empiric and based on inhibition of the renin-angiotensin-aldosterone system, with additional benefit from statins and vitamin D. Improved definition of transport phenomena in the capillary wall may suggest rational design of new interventions.
Collapse
|
47
|
Zheng Y, Guan H, Zhou X, Xu Y, Fu C, Xiao J, Ye Z. The association of renal tubular inflammatory and injury markers with uric acid excretion in chronic kidney disease patients. Int Urol Nephrol 2020; 52:923-932. [PMID: 32232720 DOI: 10.1007/s11255-020-02447-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Accepted: 03/16/2020] [Indexed: 01/05/2023]
Abstract
AIM To investigate the correlation of renal tubular inflammatory and injury markers with renal uric acid excretion in chronic kidney disease (CKD) patients. METHODS Seventy-three patients with CKD were enrolled. Fasting blood and morning urine sample were collected for routine laboratory measurements. At the same time, 24 h of urine was collected for urine biochemistry analyses, and 10 ml was extracted from the 24-h urine sample to further detect renal tubular inflammatory and injury markers, including interleukin-18 (IL-18), interleukin 1β (IL-1β), neutrophil gelatinase-associated lipocalin (NGAL) and kidney injury molecule-1 (KIM-1). The patients were divided into three tertile groups according to their 24-h urinary uric acid (24-h UUA) levels (UUA1: 24-h UUA ≤ 393.12 mg; UUA2: 393.12 < 24-h UUA ≤ 515.76 mg; UUA3: 24-h UUA > 515.76 mg). The general clinical and biochemical indexes were compared. Multivariable linear regression models were used to test the association of IL-18/Urinary creatinine concentration (IL-18/CR), IL-1β/CR, NGAL/CR and KIM-1/CR with renal uric acid excretion indicators. RESULTS All of tested renal tubular inflammation- and injury-related urinary markers were negatively associated with 24-h UUA and UEUA, and the negative correlation still persisted after adjusting for multiple influencing factors including urinary protein and eGFR. Further group analyses showed that these makers were significantly higher in the UUA1 than in the UUA3 group. CONCLUSIONS Our findings suggest that markers of urinary interstitial inflammation and injury in CKD patients are significantly correlated with 24-h UUA and Urinary excretion of uric acid (UEUA), and those with high 24-h UUA have lower levels of these markers. Renal uric acid excretion may also reflect the inflammation and injury of renal tubules under certain conditions.
Collapse
Affiliation(s)
- Yuqi Zheng
- Department of Nephrology, Huadong Hospital Affiliated With Fudan University, No. 221 West Yan'an Road, Shanghai, 200040, P.R. China.,Shanghai Key Laboratory of Clinical Geriatric Medicine, No. 221 West Yan'an Road, Shanghai, 200040, P.R. China
| | - Haochen Guan
- Department of Nephrology, Huadong Hospital Affiliated With Fudan University, No. 221 West Yan'an Road, Shanghai, 200040, P.R. China.,Shanghai Key Laboratory of Clinical Geriatric Medicine, No. 221 West Yan'an Road, Shanghai, 200040, P.R. China
| | - Xun Zhou
- Department of Nephrology, Huadong Hospital Affiliated With Fudan University, No. 221 West Yan'an Road, Shanghai, 200040, P.R. China.,Shanghai Key Laboratory of Clinical Geriatric Medicine, No. 221 West Yan'an Road, Shanghai, 200040, P.R. China
| | - Ying Xu
- Department of Nephrology, Huadong Hospital Affiliated With Fudan University, No. 221 West Yan'an Road, Shanghai, 200040, P.R. China.,Shanghai Key Laboratory of Clinical Geriatric Medicine, No. 221 West Yan'an Road, Shanghai, 200040, P.R. China
| | - Chensheng Fu
- Department of Nephrology, Huadong Hospital Affiliated With Fudan University, No. 221 West Yan'an Road, Shanghai, 200040, P.R. China.,Shanghai Key Laboratory of Clinical Geriatric Medicine, No. 221 West Yan'an Road, Shanghai, 200040, P.R. China
| | - Jing Xiao
- Department of Nephrology, Huadong Hospital Affiliated With Fudan University, No. 221 West Yan'an Road, Shanghai, 200040, P.R. China. .,Shanghai Key Laboratory of Clinical Geriatric Medicine, No. 221 West Yan'an Road, Shanghai, 200040, P.R. China.
| | - Zhibin Ye
- Department of Nephrology, Huadong Hospital Affiliated With Fudan University, No. 221 West Yan'an Road, Shanghai, 200040, P.R. China. .,Shanghai Key Laboratory of Clinical Geriatric Medicine, No. 221 West Yan'an Road, Shanghai, 200040, P.R. China.
| |
Collapse
|
48
|
Abstract
PURPOSE OF REVIEW Megalin is well known for its role in the reabsorption of proteins from the ultrafiltrate. Recent studies suggest that megalin also reabsorbs renin and angiotensinogen. Indeed, without megalin urinary renin and angiotensinogen levels massively increase, and even prorenin becomes detectable in urine. RECENT FINDINGS Intriguingly, megalin might also contribute to renal angiotensin production, as evidenced from studies in megalin knockout mice. This review discusses these topics critically, concluding that urinary renin-angiotensin system components reflect diminished reabsorption rather than release from renal tissue sites and that alterations in renal renin levels or megalin-dependent signaling need to be ruled out before concluding that angiotensin production at renal tissue sites is truly megalin dependent. Future studies should evaluate megalin-mediated renin/angiotensinogen transcytosis (allowing interstitial angiotensin generation), and determine whether megalin prefers prorenin over renin, thus explaining why urine normally contains no prorenin.
Collapse
Affiliation(s)
- Yuan Sun
- Department of Internal Medicine, Division of Pharmacology and Vascular Medicine, Erasmus MC, room EE1418b, Wytemaweg 80, 3015 CN, Rotterdam, The Netherlands
- Department of Physiology, Shenzhen University Health Science Center, Shenzhen University, Shenzhen, China
- Translational Medicine Collaborative Innovation Center, The Second Clinical Medical College (Shenzhen People's Hospital) of Jinan University, Shenzhen, China
| | - Xifeng Lu
- Department of Physiology, Shenzhen University Health Science Center, Shenzhen University, Shenzhen, China
| | - A H Jan Danser
- Department of Internal Medicine, Division of Pharmacology and Vascular Medicine, Erasmus MC, room EE1418b, Wytemaweg 80, 3015 CN, Rotterdam, The Netherlands.
| |
Collapse
|
49
|
Immunotoxin SS1P is rapidly removed by proximal tubule cells of kidney, whose damage contributes to albumin loss in urine. Proc Natl Acad Sci U S A 2020; 117:6086-6091. [PMID: 32123080 DOI: 10.1073/pnas.1919038117] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Recombinant immunotoxins (RITs) are chimeric proteins composed of an Fv and a protein toxin being developed for cancer treatment. The Fv brings the toxin to the cancer cell, but most of the RITs do not reach the tumor and are removed by other organs. To identify cells responsible for RIT removal, and the pathway by which RITs reach these cells, we studied SS1P, a 63-kDa RIT that targets mesothelin-expressing tumors and has a short serum half-life. The major organs that remove RIT were identified by live mouse imaging of RIT labeled with FNIR-Z-759. Cells responsible for SS1P removal were identified by immunohistochemistry and intravital two-photon microscopy of kidneys of rats. The primary organ of SS1P removal is kidney followed by liver. In the kidney, SS1P passes through the glomerulus, is taken up by proximal tubular cells, and transferred to lysosomes. In the liver, macrophages are involved in removal. The short half-life of SS1P is due to its very rapid filtration by the kidney followed by degradation in proximal tubular cells of the kidney. In mice treated with SS1P, proximal tubular cells are damaged and albumin in the urine is increased. SS1P uptake by kidney is reduced by coadministration of l-lysine. Our data suggests that l-lysine administration to humans might prevent SS1P-mediated kidney damage, reduce albumin loss in urine, and alleviate capillary leak syndrome.
Collapse
|
50
|
Hsu CY, Hsu RK, Liu KD, Yang J, Anderson A, Chen J, Chinchilli VM, Feldman HI, Garg AX, Hamm L, Himmelfarb J, Kaufman JS, Kusek JW, Parikh CR, Ricardo AC, Rosas SE, Saab G, Sha D, Siew ED, Sondheimer J, Taliercio JJ, Yang W, Go AS. Impact of AKI on Urinary Protein Excretion: Analysis of Two Prospective Cohorts. J Am Soc Nephrol 2019; 30:1271-1281. [PMID: 31235617 PMCID: PMC6622423 DOI: 10.1681/asn.2018101036] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 03/30/2019] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Prior studies of adverse renal consequences of AKI have almost exclusively focused on eGFR changes. Less is known about potential effects of AKI on proteinuria, although proteinuria is perhaps the strongest risk factor for future loss of renal function. METHODS We studied enrollees from the Assessment, Serial Evaluation, and Subsequent Sequelae of AKI (ASSESS-AKI) study and the subset of the Chronic Renal Insufficiency Cohort (CRIC) study enrollees recruited from Kaiser Permanente Northern California. Both prospective cohort studies included annual ascertainment of urine protein-to-creatinine ratio, eGFR, BP, and medication use. For hospitalized participants, we used inpatient serum creatinine measurements obtained as part of clinical care to define an episode of AKI (i.e., peak/nadir inpatient serum creatinine ≥1.5). We performed mixed effects regression to examine change in log-transformed urine protein-to-creatinine ratio after AKI, controlling for time-updated covariates. RESULTS At cohort entry, median eGFR was 62.9 ml/min per 1.73 m2 (interquartile range [IQR], 46.9-84.6) among 2048 eligible participants, and median urine protein-to-creatinine ratio was 0.12 g/g (IQR, 0.07-0.25). After enrollment, 324 participants experienced at least one episode of hospitalized AKI during 9271 person-years of follow-up; 50.3% of first AKI episodes were Kidney Disease Improving Global Outcomes stage 1 in severity, 23.8% were stage 2, and 25.9% were stage 3. In multivariable analysis, an episode of hospitalized AKI was independently associated with a 9% increase in the urine protein-to-creatinine ratio. CONCLUSIONS Our analysis of data from two prospective cohort studies found that hospitalization for an AKI episode was independently associated with subsequent worsening of proteinuria.
Collapse
Affiliation(s)
- Chi-yuan Hsu
- Department of Medicine, University of California, San Francisco, San Francisco, California;,Division of Research, Kaiser Permanente Northern California, Oakland, California
| | - Raymond K. Hsu
- Department of Medicine, University of California, San Francisco, San Francisco, California
| | - Kathleen D. Liu
- Department of Medicine, University of California, San Francisco, San Francisco, California
| | - Jingrong Yang
- Division of Research, Kaiser Permanente Northern California, Oakland, California
| | | | - Jing Chen
- Medicine, Tulane University, New Orleans, Louisiana
| | - Vernon M. Chinchilli
- Department of Public Health Sciences, Penn State College of Medicine, Hershey, Pennsylvania
| | | | - Amit X. Garg
- Department of Medicine, Western University, London, Ontario, Canada
| | - Lee Hamm
- Medicine, Tulane University, New Orleans, Louisiana
| | | | - James S. Kaufman
- Veterans Affairs New York Harbor Healthcare System, New York, New York;,Department of Medicine, New York University School of Medicine, New York, New York
| | | | - Chirag R. Parikh
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Ana C. Ricardo
- Department of Medicine, University of Illinois, Chicago, Illinois
| | - Sylvia E. Rosas
- Kidney and Hypertension Unit, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts
| | - Georges Saab
- Case Western Reserve University and Metrohealth Medical Center, Cleveland, Ohio
| | - Daohang Sha
- Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Edward D. Siew
- Vanderbilt University Medical Center and Nashville Veterans Affairs Hospital, Nashville, Tennessee
| | - James Sondheimer
- Department of Medicine, Wayne State University School of Medicine, Detroit, Michigan; and
| | | | - Wei Yang
- Department of Biostatistics and Epidemiology, and
| | - Alan S. Go
- Department of Medicine, University of California, San Francisco, San Francisco, California;,Division of Research, Kaiser Permanente Northern California, Oakland, California
| | | |
Collapse
|