1
|
Bae SH, Lee EJ, Hwang J, Hong SS, Chang YW, Nam B. Novel non-invasive and quantitative assessment of the renal function of transplanted kidneys using Doppler ultrasonography with the vascular index of superb microvascular imaging. Ultrasonography 2025; 44:160-169. [PMID: 40068674 PMCID: PMC11938797 DOI: 10.14366/usg.24176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 01/21/2025] [Accepted: 02/06/2025] [Indexed: 03/28/2025] Open
Abstract
PURPOSE This study assessed the reproducibility and clinical value of the vascular index (VI), derived from superb microvascular imaging (SMI) using Doppler ultrasonography, for evaluating renal function in transplanted kidneys. METHODS This retrospective study included 63 renal transplant patients who underwent grayscale and Doppler ultrasonography with SMI from January 2022 to February 2023. The VI of the transplanted kidneys was measured using three methods (VIbox, VIF1, VIF2). The VI was compared across chronic kidney disease (CKD) groups categorized by estimated glomerular filtration rate (eGFR) and Kidney Disease: Improving Global Outcomes (KDIGO) CKD risk groups based on eGFR and albuminuria. The correlation between VI and renal function was evaluated. Univariate and multivariate linear regression analyses were used to identify predictors of eGFR. RESULTS Significant differences in VI were observed among CKD groups based on eGFR (VIbox, P=0.001; VIF1, P<0.001; VIF2, P<0.001) and KDIGO CKD groups based on eGFR and albuminuria (VIbox, P=0.039; VIF1, P=0.001; VIF2, P<0.001). VIF1 and VIF2 demonstrated moderate/high correlations with eGFR (r=0.627, P<0.001 and r=0.657, P<0.001, respectively) and serum creatinine (r=-0.626, P<0.001 and r=-0.649, P<0.001, respectively). VIbox moderately correlated with eGFR (r=0.445, P<0.001). Multivariate regression identified the urine albumincreatinine ratio (ACR) (adjusted odds ratio [aOR], 1.122; 95% confidence interval [CI], -0.007 to, 0.000; P=0.030) and VIF2 (aOR, 1.114; 95% CI, 0.466 to 1.235; P<0.001) were independently associated with eGFR. CONCLUSION The VI measured by drawing a region of interest along the border of the transplanted kidney in SMI (VIF2) is highly reproducible and correlates well with eGFR. Both VIF2 and ACR independently predict eGFR.
Collapse
Affiliation(s)
- Sung Hwan Bae
- Department of Radiology, Soonchunhyang University Seoul Hospital, Soonchunhyang University College of Medicine, Seoul, Korea
| | - Eun Ji Lee
- Department of Radiology, Soonchunhyang University Seoul Hospital, Soonchunhyang University College of Medicine, Seoul, Korea
| | - Jiyoung Hwang
- Department of Radiology, Soonchunhyang University Seoul Hospital, Soonchunhyang University College of Medicine, Seoul, Korea
| | - Seong Sook Hong
- Department of Radiology, Soonchunhyang University Seoul Hospital, Soonchunhyang University College of Medicine, Seoul, Korea
| | - Yun-Woo Chang
- Department of Radiology, Soonchunhyang University Seoul Hospital, Soonchunhyang University College of Medicine, Seoul, Korea
| | - Boda Nam
- Department of Radiology, Soonchunhyang University Seoul Hospital, Soonchunhyang University College of Medicine, Seoul, Korea
| |
Collapse
|
2
|
Nast CC. Shape, Size, and Spatial Relationships: Peritubular Capillary Features in Kidney Fibrosis and Disease Progression. Clin J Am Soc Nephrol 2025; 20:01277230-990000000-00530. [PMID: 39792456 PMCID: PMC11835152 DOI: 10.2215/cjn.0000000647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Affiliation(s)
- Cynthia C Nast
- Department of Pathology, Cedars-Sinai Medical Center, Los Angeles, California
| |
Collapse
|
3
|
Hu Y, Lei Y, Yu M, Zhang Y, Huang X, Zhang G, Deng Q. Ultrasound super-resolution imaging for the assessment of renal allograft dysfunction: A pilot study. Heliyon 2024; 10:e36515. [PMID: 39247269 PMCID: PMC11380004 DOI: 10.1016/j.heliyon.2024.e36515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 08/16/2024] [Accepted: 08/16/2024] [Indexed: 09/10/2024] Open
Abstract
Background The purpose of this study was to examine the feasibility and practical application of ultrasound (US) super-resolution imaging (SRI) in evaluating microvasculature and measuring renal allograft function. Methods Sixteen consecutive patients who received kidney transplants were prospectively enrolled. The patients were assigned as: normal allograft function (n = 6), and allograft malfunction (n = 10). Localizing each potential contrast signal resulted in super-resolution images (SRI). SRI was utilized to assess micro-vessel density (MVD) and microvascular flow rate, whereas contrast-enhanced (CE) US images were statistically processed to get the time to peak (TTP) and peak intensity. Logistic regression was utilized to evaluate their relationship. Results US SRI may be utilized effectively on allografts to show microvasculature with significantly higher resolution than typical color Doppler flow and CEUS pictures. In the multivariate analysis, MVD and TTP were significant US markers of renal allograft failure (p = 0.031 and p = 0.045). The combination of MVD and TTP produced an AUC of 0.783 (p < 0.05) for allograft dysfunction. Conclusions SRI can accurately portray the microvasculature of renal allografts, while MVD and TTP are appropriate US markers for assessing renal allograft failure.
Collapse
Affiliation(s)
- Yugang Hu
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, 430061, China
| | - Yumeng Lei
- Department of Medical Ultrasound, China Resources & Wisco General Hospital, Wuhan University of Science and Technology, Wuhan, 430080, China
| | - Meihui Yu
- Department of Medical Ultrasound, China Resources & Wisco General Hospital, Wuhan University of Science and Technology, Wuhan, 430080, China
| | - Yao Zhang
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, 430061, China
| | - Xingyue Huang
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, 430061, China
| | - Ge Zhang
- Department of Medical Ultrasound, China Resources & Wisco General Hospital, Wuhan University of Science and Technology, Wuhan, 430080, China
| | - Qing Deng
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, 430061, China
| |
Collapse
|
4
|
Ogurlu B, Hamelink TL, Van Tricht IM, Leuvenink HGD, De Borst MH, Moers C, Pool MBF. Utilizing pathophysiological concepts of ischemia-reperfusion injury to design renoprotective strategies and therapeutic interventions for normothermic ex vivo kidney perfusion. Am J Transplant 2024; 24:1110-1126. [PMID: 38184242 DOI: 10.1016/j.ajt.2024.01.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 12/13/2023] [Accepted: 01/01/2024] [Indexed: 01/08/2024]
Abstract
Normothermic machine perfusion (NMP) has emerged as a promising tool for the preservation, viability assessment, and repair of deceased-donor kidneys prior to transplantation. These kidneys inevitably experience a period of ischemia during donation, which leads to ischemia-reperfusion injury when NMP is subsequently commenced. Ischemia-reperfusion injury has a major impact on the renal vasculature, metabolism, oxygenation, electrolyte balance, and acid-base homeostasis. With an increased understanding of the underlying pathophysiological mechanisms, renoprotective strategies and therapeutic interventions can be devised to minimize additional injury during normothermic reperfusion, ensure the safe implementation of NMP, and improve kidney quality. This review discusses the pathophysiological alterations in the vasculature, metabolism, oxygenation, electrolyte balance, and acid-base homeostasis of deceased-donor kidneys and delineates renoprotective strategies and therapeutic interventions to mitigate renal injury and improve kidney quality during NMP.
Collapse
Affiliation(s)
- Baran Ogurlu
- Department of Surgery - Organ Donation and Transplantation, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.
| | - Tim L Hamelink
- Department of Surgery - Organ Donation and Transplantation, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Isa M Van Tricht
- Department of Surgery - Organ Donation and Transplantation, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Henri G D Leuvenink
- Department of Surgery - Organ Donation and Transplantation, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Martin H De Borst
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Cyril Moers
- Department of Surgery - Organ Donation and Transplantation, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Merel B F Pool
- Department of Surgery - Organ Donation and Transplantation, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| |
Collapse
|
5
|
Niu X, Xu C, Cheuk YC, Xu X, Liang L, Zhang P, Rong R. Characterizing hub biomarkers for post-transplant renal fibrosis and unveiling their immunological functions through RNA sequencing and advanced machine learning techniques. J Transl Med 2024; 22:186. [PMID: 38378674 PMCID: PMC10880303 DOI: 10.1186/s12967-024-04971-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 02/09/2024] [Indexed: 02/22/2024] Open
Abstract
BACKGROUND Kidney transplantation stands out as the most effective renal replacement therapy for patients grappling with end-stage renal disease. However, post-transplant renal fibrosis is a prevalent and irreversible consequence, imposing a substantial clinical burden. Unfortunately, the clinical landscape remains devoid of reliable biological markers for diagnosing post-transplant renal interstitial fibrosis. METHODS We obtained transcriptome and single-cell sequencing datasets of patients with renal fibrosis from NCBI Gene Expression Omnibus (GEO). Subsequently, we employed Weighted Gene Co-Expression Network Analysis (WGCNA) to identify potential genes by integrating core modules and differential genes. Functional enrichment analysis was conducted to unveil the involvement of potential pathways. To identify key biomarkers for renal fibrosis, we utilized logistic analysis, a LASSO-based tenfold cross-validation approach, and gene topological analysis within Cytoscape. Furthermore, histological staining, Western blotting (WB), and quantitative PCR (qPCR) experiments were performed in a murine model of renal fibrosis to verify the identified hub genes. Moreover, molecular docking and molecular dynamics simulations were conducted to explore possible effective drugs. RESULTS Through WGCNA, the intersection of core modules and differential genes yielded a compendium of 92 potential genes. Logistic analysis, LASSO-based tenfold cross-validation, and gene topological analysis within Cytoscape identified four core genes (CD3G, CORO1A, FCGR2A, and GZMH) associated with renal fibrosis. The expression of these core genes was confirmed through single-cell data analysis and validated using various machine learning methods. Wet experiments also verified the upregulation of these core genes in the murine model of renal fibrosis. A positive correlation was observed between the core genes and immune cells, suggesting their potential role in bolstering immune system activity. Moreover, four potentially effective small molecules (ZINC000003830276-Tessalon, ZINC000003944422-Norvir, ZINC000008214629-Nonoxynol-9, and ZINC000085537014-Cobicistat) were identified through molecular docking and molecular dynamics simulations. CONCLUSION Four potential hub biomarkers most associated with post-transplant renal fibrosis, as well as four potentially effective small molecules, were identified, providing valuable insights for studying the molecular mechanisms underlying post-transplant renal fibrosis and exploring new targets.
Collapse
Affiliation(s)
- Xinhao Niu
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Key Laboratory of Organ Transplantation, Shanghai, 200032, China
| | - Cuidi Xu
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Key Laboratory of Organ Transplantation, Shanghai, 200032, China
| | - Yin Celeste Cheuk
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Key Laboratory of Organ Transplantation, Shanghai, 200032, China
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Xiaoqing Xu
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Key Laboratory of Organ Transplantation, Shanghai, 200032, China
| | - Lifei Liang
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Key Laboratory of Organ Transplantation, Shanghai, 200032, China
| | - Pingbao Zhang
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Key Laboratory of Organ Transplantation, Shanghai, 200032, China
| | - Ruiming Rong
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
- Shanghai Key Laboratory of Organ Transplantation, Shanghai, 200032, China.
| |
Collapse
|
6
|
Steegh FMEG, Keijbeck AA, de Hoogt PA, Rademakers T, Houben AJHM, Reesink KD, Stehouwer CDA, Daemen MJAP, Peutz-Kootstra CJ. Capillary rarefaction: a missing link in renal and cardiovascular disease? Angiogenesis 2024; 27:23-35. [PMID: 37326760 DOI: 10.1007/s10456-023-09883-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 05/28/2023] [Indexed: 06/17/2023]
Abstract
Patients with chronic kidney disease (CKD) have an increased risk for cardiovascular morbidity and mortality. Capillary rarefaction may be both one of the causes as well as a consequence of CKD and cardiovascular disease. We reviewed the published literature on human biopsy studies and conclude that renal capillary rarefaction occurs independently of the cause of renal function decline. Moreover, glomerular hypertrophy may be an early sign of generalized endothelial dysfunction, while peritubular capillary loss occurs in advanced renal disease. Recent studies with non-invasive measurements show that capillary rarefaction is detected systemically (e.g., in the skin) in individuals with albuminuria, as sign of early CKD and/or generalized endothelial dysfunction. Decreased capillary density is found in omental fat, muscle and heart biopsies of patients with advanced CKD as well as in skin, fat, muscle, brain and heart biopsies of individuals with cardiovascular risk factors. No biopsy studies have yet been performed on capillary rarefaction in individuals with early CKD. At present it is unknown whether individuals with CKD and cardiovascular disease merely share the same risk factors for capillary rarefaction, or whether there is a causal relationship between rarefaction in renal and systemic capillaries. Further studies on renal and systemic capillary rarefaction, including their temporal relationship and underlying mechanisms are needed. This review stresses the importance of preserving and maintaining capillary integrity and homeostasis in the prevention and management of renal and cardiovascular disease.
Collapse
Affiliation(s)
- Floor M E G Steegh
- Department of Pathology, Maastricht University Medical Centre+, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Anke A Keijbeck
- Department of Pathology, Maastricht University Medical Centre+, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Patrick A de Hoogt
- Surgery, Maastricht University Medical Centre+, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Timo Rademakers
- Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, The Netherlands
| | - Alfons J H M Houben
- Internal Medicine, Maastricht University Medical Centre+, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Koen D Reesink
- Biomedical Engineering, Maastricht University Medical Centre+, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Coen D A Stehouwer
- Internal Medicine, Maastricht University Medical Centre+, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Mat J A P Daemen
- Department of Pathology, UMC Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | - Carine J Peutz-Kootstra
- Department of Pathology, Maastricht University Medical Centre+, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands.
- Department of Pathology, Gelre Ziekenhuizen, Apeldoorn, The Netherlands.
- , Porthoslaan 39, 6213 CN, Maastricht, The Netherlands.
| |
Collapse
|
7
|
van der Weijden J, De Hoogt PA, Leufkens MME, Keijbeck AA, van Goor H, van den Heuvel MC, Cleutjens JPM, Moers C, Snoeijs MG, Navis GJ, van Londen M, Nolte IM, Berger SP, De Borst MH, Peutz-Kootstra CJ. The relationship of peritubular capillary density with glomerular volume and kidney function in living kidney donors. J Nephrol 2023; 36:2111-2124. [PMID: 37768545 PMCID: PMC10543576 DOI: 10.1007/s40620-023-01734-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 07/03/2023] [Indexed: 09/29/2023]
Abstract
BACKGROUND Peritubular capillary rarefaction plays an important role in the progression of chronic kidney disease. Little is known about the relation between peritubular capillary density, glomerular volume and filtration rate in the healthy kidney. METHODS In this single-center study, we included 69 living kidney donors who donated between 2005 and 2008 and had representative renal biopsies available. In all donors, glomerular filtration rate was measured using 125I-Iothalamate before donation and at five years after donation. Before donation, the increase in glomerular filtration rate after dopamine stimulation was measured. Glomerular volume and peritubular capillary density were determined in biopsies taken at the time of transplantation. Pearson's correlation coefficient and linear regression were used to assess relations between parameters. RESULTS Mean donor age was 52 ± 11 years and mean measured glomerular filtration rate was 119 ± 22 mL/min before donation and 82 ± 15 mL/min at five years after donation. While peritubular capillary density (measured by either number of peritubular capillaries/50,000 μm2 or number of peritubular capillaries/tubule) was not associated with measured glomerular filtration rate before or after donation, number of peritubular capillaries/tubule was associated with the increase in measured glomerular filtration rate after dopamine stimulation (St.β = 0.33, p = 0.004), and correlated positively with glomerular volume (R = 0.24, p = 0.047). Glomerular volume was associated with unstimulated measured glomerular filtration rate before donation (St.β = 0.31, p = 0.01) and at five years (St.β = 0.30, p = 0.01) after donation, independent of age. CONCLUSIONS In summary, peritubular capillary density was not related to unstimulated kidney function before or after kidney donation, in contrast to glomerular volume. However, number of peritubular capillaries/tubule correlated with the increase in glomerular filtration rate after dopamine stimulation in healthy kidneys, and with glomerular volume. These findings suggest that peritubular capillary density and glomerular volume differentially affect kidney function in healthy living kidney donors.
Collapse
Affiliation(s)
- J van der Weijden
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Hanzeplein 1, P.O. Box AA53, 9713 GZ, Groningen, The Netherlands.
| | - P A De Hoogt
- Department of Vascular Surgery, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - M M E Leufkens
- Department of Pathology, Maastricht University Medical Center+, Cardiovascular Research Institute Maastricht (CARIM), Maastricht, The Netherlands
| | - A A Keijbeck
- Department of Pathology, Maastricht University Medical Center+, Cardiovascular Research Institute Maastricht (CARIM), Maastricht, The Netherlands
| | - H van Goor
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - M C van den Heuvel
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - J P M Cleutjens
- Department of Pathology, Maastricht University Medical Center+, Cardiovascular Research Institute Maastricht (CARIM), Maastricht, The Netherlands
| | - C Moers
- Department of Surgery, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - M G Snoeijs
- Department of Vascular Surgery, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - G J Navis
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Hanzeplein 1, P.O. Box AA53, 9713 GZ, Groningen, The Netherlands
| | - M van Londen
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Hanzeplein 1, P.O. Box AA53, 9713 GZ, Groningen, The Netherlands
| | - I M Nolte
- Department of Epidemiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - S P Berger
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Hanzeplein 1, P.O. Box AA53, 9713 GZ, Groningen, The Netherlands
| | - M H De Borst
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Hanzeplein 1, P.O. Box AA53, 9713 GZ, Groningen, The Netherlands
| | - C J Peutz-Kootstra
- Department of Pathology, Maastricht University Medical Center+, Cardiovascular Research Institute Maastricht (CARIM), Maastricht, The Netherlands
- Department of Pathology, Gelre Ziekenhuizen, Apeldoorn, The Netherlands
| |
Collapse
|
8
|
Rao JS, Pruett TL. Immunology of the transplanted cryopreserved kidney. Cryobiology 2023; 110:1-7. [PMID: 36640932 DOI: 10.1016/j.cryobiol.2023.01.003] [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: 11/10/2022] [Revised: 12/28/2022] [Accepted: 01/10/2023] [Indexed: 01/13/2023]
Abstract
Transplantation has substituted dysfunctional organs with healthy organs from donors to significantly lower morbidity and mortality associated with end-stage organ disease. Since the advent of transplantation, the promise of functional replacement has attracted an exponential mismatch between organ supply and demand. Theoretical proposals to counter the increasing needs have either been to create a source through genetic engineering of porcine donors for xenotransplantation (with more potent immunosuppression protocols) or recreate one's organ in a pig using interspecies blastocyst complementation for exogenic organ transplantation (without immunosuppression). Another promising avenue has been organ banking through cryopreservation for transplantation. Although ice free preservation and acceptable early function following rewarming is critical for success in transplantation, the immunological response that predominantly defines short- and long-term graft survival has failed to captivate attention to date. It is well sorted that thermal and metabolic stress incurred at 4 °C during recovery and reperfusion of organs for clinical transplantation has varying impact on graft survival. Considering the magnitude of cellular imbalance and injury at sub-zero/ultralow temperatures in addition to the chemical toxicity of cryoprotective agents (CPA), it is essential to assess and address the immunological response associated following transplantation to maximize the success of cryopreservation.
Collapse
Affiliation(s)
- Joseph Sushil Rao
- Division of Solid Organ Transplantation, Department of Surgery, University of Minnesota, Minneapolis, MN, USA; Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, MN, USA.
| | - Timothy L Pruett
- Division of Solid Organ Transplantation, Department of Surgery, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
9
|
Ultrasound localization microscopy of the human kidney allograft on a clinical ultrasound scanner. Kidney Int 2023; 103:930-935. [PMID: 36841476 DOI: 10.1016/j.kint.2023.01.027] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 01/18/2023] [Accepted: 01/27/2023] [Indexed: 02/27/2023]
Abstract
Chronic kidney disease is a major medical problem, causing more than a million deaths each year worldwide. Peripheral kidney microvascular damage characterizes most chronic kidney diseases, yet noninvasive and quantitative diagnostic tools to measure this are lacking. Ultrasound Localization Microscopy (ULM) can assess tissue microvasculature with unprecedented resolution. Here, we optimized methods on 35 kidney transplants and studied the feasibility of ULM in seven human kidney allografts with a standard low frame rate ultrasound scanner to access microvascular damage. Interlobar, arcuate, cortical radial vessels, and part of the medullary organization were visible on ULM density maps. The medullary vasa recta can be seen but are not as clear as the cortical vessels. Acquisition parameters were derived from Contrast-Enhanced Ultrasound examinations by increasing the duration of the recorded clip at the same plane. ULM images were compared with Color Doppler, Advanced Dynamic Flow, and Superb Microvascular Imaging with a contrast agent. Despite some additional limitations due to movement and saturation artifacts, ULM identified vessels two to four times thinner compared with Doppler modes. The mean ULM smallest analyzable vessel cross section was 0.3 ± 0.2 mm in the seven patients. Additionally, ULM was able to provide quantitative information on blood velocities in the cortical area. Thus, this proof-of-concept study has shown ULM to be a promising imaging technique for qualitative and quantitative microvascular assessment. Imaging native kidneys in patients with kidney diseases will be needed to identify their ULM biomarkers.
Collapse
|
10
|
Akalay S, Hosgood SA. How to Best Protect Kidneys for Transplantation-Mechanistic Target. J Clin Med 2023; 12:jcm12051787. [PMID: 36902572 PMCID: PMC10003664 DOI: 10.3390/jcm12051787] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 02/15/2023] [Accepted: 02/17/2023] [Indexed: 02/25/2023] Open
Abstract
The increasing number of patients on the kidney transplant waiting list underlines the need to expand the donor pool and improve kidney graft utilization. By protecting kidney grafts adequately from the initial ischemic and subsequent reperfusion injury occurring during transplantation, both the number and quality of kidney grafts could be improved. The last few years have seen the emergence of many new technologies to abrogate ischemia-reperfusion (I/R) injury, including dynamic organ preservation through machine perfusion and organ reconditioning therapies. Although machine perfusion is gradually making the transition to clinical practice, reconditioning therapies have not yet progressed from the experimental setting, pointing towards a translational gap. In this review, we discuss the current knowledge on the biological processes implicated in I/R injury and explore the strategies and interventions that are being proposed to either prevent I/R injury, treat its deleterious consequences, or support the reparative response of the kidney. Prospects to improve the clinical translation of these therapies are discussed with a particular focus on the need to address multiple aspects of I/R injury to achieve robust and long-lasting protective effects on the kidney graft.
Collapse
Affiliation(s)
- Sara Akalay
- Department of Development and Regeneration, Laboratory of Pediatric Nephrology, KU Leuven, 3000 Leuven, Belgium
| | - Sarah A. Hosgood
- Department of Surgery, University of Cambridge, Cambridge CB2 0QQ, UK
- Correspondence:
| |
Collapse
|
11
|
Kwiatkowska E, Kwiatkowski S, Dziedziejko V, Tomasiewicz I, Domański L. Renal Microcirculation Injury as the Main Cause of Ischemic Acute Kidney Injury Development. BIOLOGY 2023; 12:biology12020327. [PMID: 36829602 PMCID: PMC9953191 DOI: 10.3390/biology12020327] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 02/04/2023] [Accepted: 02/09/2023] [Indexed: 02/22/2023]
Abstract
Acute kidney injury (AKI) can result from multiple factors. The main cause is reduced renal perfusion. Kidneys are susceptible to ischemia due to the anatomy of microcirculation that wraps around the renal tubules-peritubular capillary (PTC) network. Cortical and medullary superficial tubules have a large share in transport and require the supply of oxygen for ATP production, while it is the cortex that receives almost 100% of the blood flowing through the kidneys and the medulla only accounts for 5-10% of it. This difference makes the tubules present in the superficial layer of the medulla very susceptible to ischemia. Impaired blood flow causes damage to the endothelium, with an increase in its prothrombotic and pro-adhesive properties. This causes congestion in the microcirculation of the renal medulla. The next stage is the migration of pericytes with the disintegration of these vessels. The phenomenon of destruction of small vessels is called peritubular rarefaction, attributed as the main cause of further irreversible changes in the damaged kidney leading to the development of chronic kidney disease. In this article, we will present the characteristic structure of renal microcirculation, its regulation, and the mechanism of damage in acute ischemia, and we will try to find methods of prevention with particular emphasis on the inhibition of the renin-angiotensin-aldosterone system.
Collapse
Affiliation(s)
- Ewa Kwiatkowska
- Department of Nephrology, Transplantology and Internal Medicine, Pomeranian Medical University in Szczecin, Powstańców Wlkp, 72, 70-204 Szczecin, Poland
- Correspondence:
| | - Sebastian Kwiatkowski
- Department of Obstetrician and Gynecology, Pomeranian Medical University in Szczecin, Powstańców Wlkp, 72, 70-204 Szczecin, Poland
| | - Violetta Dziedziejko
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp, 72, 70-204 Szczecin, Poland
| | - Izabela Tomasiewicz
- Department of Nephrology, Transplantology and Internal Medicine, Pomeranian Medical University in Szczecin, Powstańców Wlkp, 72, 70-204 Szczecin, Poland
| | - Leszek Domański
- Department of Nephrology, Transplantology and Internal Medicine, Pomeranian Medical University in Szczecin, Powstańców Wlkp, 72, 70-204 Szczecin, Poland
| |
Collapse
|
12
|
Mawad H, Pinard L, Medani S, Chagnon M, Boucquemont J, Turgeon J, Dieudé M, Hamelin K, Rimbaud AK, Belayachi A, Yang B, Collette S, Sénécal L, Foster BJ, Hébert MJ, Cardinal H. Hypothermic Perfusion Modifies the Association Between Anti-LG3 Antibodies and Delayed Graft Function in Kidney Recipients. Transpl Int 2023; 36:10749. [PMID: 36891519 PMCID: PMC9986256 DOI: 10.3389/ti.2023.10749] [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: 07/06/2022] [Accepted: 01/26/2023] [Indexed: 02/22/2023]
Abstract
We previously reported associations between autoantibodies to the LG3 fragment of perlecan, anti-LG3, and a higher risk of delayed graft function (DGF) in kidney transplant recipients. Here, we aimed to determine whether some factors that modulate ischemia-reperfusion injury (IRI) can modify this association. We performed a retrospective cohort study in kidney transplant recipients in 2 university-affiliated centers. In 687 patients, we show that high pre-transplant anti-LG3 are associated with DGF when the kidney is transported on ice (odds ratio (OR): 1.75, 95% confidence interval 1.02-3.00), but not when placed on hypothermic perfusion pump (OR: 0.78, 95% CI 0.43-1.37). In patients with DGF, high pre-transplant anti-LG3 are associated with a higher risk of graft failure (subdistribution hazard ratio (SHR): 4.07, 95% CI: 1.80, 9.22), while this was not the case in patients with immediate graft function (SHR: 0.50, 95% CI 0.19, 1.29). High anti-LG3 levels are associated with a higher risk of DGF in kidneys exposed to cold storage, but not when hypothermic pump perfusion is used. High anti-LG3 are also associated with a higher risk of graft failure in patients who experience DGF, a clinical manifestation of severe IRI.
Collapse
Affiliation(s)
- Habib Mawad
- Research centre, Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
| | - Louis Pinard
- Research centre, Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
| | - Samar Medani
- Research centre, Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
| | - Miguel Chagnon
- Department of Mathematics and Statistics, Université de Montréal, Montreal, QC, Canada
| | - Julie Boucquemont
- Montreal Children's Hospital, McGill University, Montreal, QC, Canada
| | - Julie Turgeon
- Research centre, Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada.,Canadian Donation and Transplantation Research Program, Edmonton, AB, Canada
| | - Mélanie Dieudé
- Research centre, Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada.,Canadian Donation and Transplantation Research Program, Edmonton, AB, Canada.,Héma-Québec, Québec, QC, Canada
| | - Katia Hamelin
- Research centre, Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
| | | | - Ali Belayachi
- Research centre, Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
| | - Bing Yang
- Research centre, Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
| | - Suzon Collette
- Canadian Donation and Transplantation Research Program, Edmonton, AB, Canada.,Department of Medicine, Université de Montréal, Montréal, QC, Canada.,Hôpital Maisonneuve-Rosemont, Montreal, QC, Canada
| | - Lynne Sénécal
- Canadian Donation and Transplantation Research Program, Edmonton, AB, Canada.,Department of Medicine, Université de Montréal, Montréal, QC, Canada.,Hôpital Maisonneuve-Rosemont, Montreal, QC, Canada
| | - Bethany J Foster
- Montreal Children's Hospital, McGill University, Montreal, QC, Canada.,Canadian Donation and Transplantation Research Program, Edmonton, AB, Canada
| | - Marie-Josée Hébert
- Research centre, Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada.,Canadian Donation and Transplantation Research Program, Edmonton, AB, Canada.,Héma-Québec, Québec, QC, Canada.,Department of Medicine, Université de Montréal, Montréal, QC, Canada
| | - Héloïse Cardinal
- Research centre, Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada.,Canadian Donation and Transplantation Research Program, Edmonton, AB, Canada.,Héma-Québec, Québec, QC, Canada.,Department of Medicine, Université de Montréal, Montréal, QC, Canada
| |
Collapse
|
13
|
Ghajar-Rahimi G, Agarwal A. Endothelial KLF11 as a Nephroprotectant in AKI. KIDNEY360 2022; 3:1302-1305. [PMID: 36176668 PMCID: PMC9416841 DOI: 10.34067/kid.0003422022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 06/14/2022] [Indexed: 06/16/2023]
Affiliation(s)
- Gelare Ghajar-Rahimi
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama
- Division of Cardiothoracic Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama
| | - Anupam Agarwal
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Birmingham Veterans Administration Health Care Services, Birmingham, Alabama
| |
Collapse
|
14
|
Lan S, Yang B, Migneault F, Turgeon J, Bourgault M, Dieudé M, Cardinal H, Hickey MJ, Patey N, Hébert MJ. Caspase-3-dependent peritubular capillary dysfunction is pivotal for the transition from acute to chronic kidney disease after acute ischemia-reperfusion injury. Am J Physiol Renal Physiol 2021; 321:F335-F351. [PMID: 34338031 DOI: 10.1152/ajprenal.00690.2020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 07/21/2021] [Indexed: 11/22/2022] Open
Abstract
Ischemia-reperfusion injury (IRI) is a major risk factor for chronic renal failure. Caspase-3, an effector responsible for apoptosis execution, is activated within the peritubular capillary (PTC) in the early stage of IRI-induced acute kidney injury (AKI). Recently, we showed that caspase-3-dependent microvascular rarefaction plays a key role in fibrosis development after mild renal IRI. Here, we further characterized the role of caspase-3 in microvascular dysfunction and progressive renal failure in both mild and severe AKI, by performing unilateral renal artery clamping for 30/60 min with contralateral nephrectomy in wild-type (C57BL/6) or caspase-3-/- mice. In both forms of AKI, caspase-3-/- mice showed better long-term outcomes despite worse initial tubular injury. After 3 wk, they showed reduced PTC injury, decreased PTC collagen deposition and α-smooth muscle actin expression, and lower tubular injury scores compared with wild-type animals. Caspase-3-/- mice with severe IRI also showed better preservation of long-term renal function. Intravital imaging and microcomputed tomography revealed preserved PTC permeability and better terminal capillary density in caspase-3-/- mice. Collectively, these results demonstrate the pivotal importance of caspase-3 in regulating long-term renal function after IRI and establish the predominant role of PTC dysfunction as a major contributor to progressive renal dysfunction.NEW & NOTEWORTHY Our findings demonstrate the pivotal importance of caspase-3 in regulating renal microvascular dysfunction, fibrogenesis, and long-term renal impairment after acute kidney injury induced by ischemia-reperfusion injury. Furthermore, this study establishes the predominant role of peritubular capillary integrity as a major contributor to progressive renal dysfunction after ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Shanshan Lan
- Research Centre, Centre hospitalier de l'Université de Montréal, Montreal, Quebec, Canada
- Canadian Donation Transplant Research Program, Edmonton, Alberta, Canada
- Université de Montréal, Montreal, Quebec, Canada
| | - Bing Yang
- Research Centre, Centre hospitalier de l'Université de Montréal, Montreal, Quebec, Canada
- Canadian Donation Transplant Research Program, Edmonton, Alberta, Canada
- Université de Montréal, Montreal, Quebec, Canada
| | - Francis Migneault
- Research Centre, Centre hospitalier de l'Université de Montréal, Montreal, Quebec, Canada
- Canadian Donation Transplant Research Program, Edmonton, Alberta, Canada
| | - Julie Turgeon
- Research Centre, Centre hospitalier de l'Université de Montréal, Montreal, Quebec, Canada
- Canadian Donation Transplant Research Program, Edmonton, Alberta, Canada
| | - Maude Bourgault
- Research Centre, Centre hospitalier de l'Université de Montréal, Montreal, Quebec, Canada
| | - Mélanie Dieudé
- Research Centre, Centre hospitalier de l'Université de Montréal, Montreal, Quebec, Canada
- Canadian Donation Transplant Research Program, Edmonton, Alberta, Canada
- Université de Montréal, Montreal, Quebec, Canada
| | - Héloïse Cardinal
- Research Centre, Centre hospitalier de l'Université de Montréal, Montreal, Quebec, Canada
- Canadian Donation Transplant Research Program, Edmonton, Alberta, Canada
- Université de Montréal, Montreal, Quebec, Canada
| | - Michael J Hickey
- Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Clayton, Victoria, Australia
| | - Natacha Patey
- Research Centre, Centre hospitalier de l'Université de Montréal, Montreal, Quebec, Canada
- Université de Montréal, Montreal, Quebec, Canada
- Department of Pathology, Centre Hospitalier Universitaire Sainte-Justine, Université de Montréal, Montreal, Quebec, Canada
| | - Marie-Josée Hébert
- Research Centre, Centre hospitalier de l'Université de Montréal, Montreal, Quebec, Canada
- Canadian Donation Transplant Research Program, Edmonton, Alberta, Canada
- Université de Montréal, Montreal, Quebec, Canada
| |
Collapse
|
15
|
Wewers TM, Schulz A, Nolte I, Pavenstädt H, Brand M, Di Marco GS. Circulating Soluble Fms-like Tyrosine Kinase in Renal Diseases Other than Preeclampsia. J Am Soc Nephrol 2021; 32:1853-1863. [PMID: 34155060 PMCID: PMC8455271 DOI: 10.1681/asn.2020111579] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 04/20/2021] [Indexed: 02/04/2023] Open
Abstract
Soluble Fms-like tyrosine kinase (sFlt-1/sVEGFR1) is a naturally occurring antagonist of vascular endothelial growth factor (VEGF). Despite being a secreted, soluble protein lacking cytoplasmic and transmembrane domains, sFlt-1 can act locally and be protective against excessive microenvironmental VEGF concentration or exert autocrine functions independently of VEGF. Circulating sFlt-1 may indiscriminately affect endothelial function and the microvasculature of distant target organs. The clinical significance of excess sFlt-1 in kidney disease was first shown in preeclampsia, a major renal complication of pregnancy. However, circulating sFlt-1 levels appear to be increased in various diseases with varying degrees of renal impairment. Relevant clinical associations between circulating sFlt-1 and severe outcomes (e.g., endothelial dysfunction, renal impairment, cardiovascular disease, and all-cause mortality) have been observed in patients with CKD and after kidney transplantation. However, sFlt-1 appears to be protective against renal dysfunction-associated aggravation of atherosclerosis and diabetic nephropathy. Therefore, in this study, we provide an update on sFlt-1 in several kidney diseases other than preeclampsia, discuss clinical findings and experimental studies, and briefly consider its use in clinical practice.
Collapse
Affiliation(s)
- Theresa M. Wewers
- Department of Internal Medicine D, University Hospital Muenster, Muenster, Germany,Small Animal Hospital, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Annika Schulz
- Department of Internal Medicine D, University Hospital Muenster, Muenster, Germany
| | - Ingo Nolte
- Small Animal Hospital, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Hermann Pavenstädt
- Department of Internal Medicine D, University Hospital Muenster, Muenster, Germany
| | - Marcus Brand
- Department of Internal Medicine D, University Hospital Muenster, Muenster, Germany
| | - Giovana S. Di Marco
- Department of Internal Medicine D, University Hospital Muenster, Muenster, Germany,Correspondence: Giovana S. Di Marco, Albert-Schweitzer-Campus 1, Building A14, 48149 Münster, Germany.
| |
Collapse
|
16
|
Doreille A, Azzi F, Larivière-Beaudoin S, Karakeussian-Rimbaud A, Trudel D, Hébert MJ, Dieudé M, Patey N, Cardinal H. Acute Kidney Injury, Microvascular Rarefaction, and Estimated Glomerular Filtration Rate in Kidney Transplant Recipients. Clin J Am Soc Nephrol 2021; 16:415-426. [PMID: 33648972 PMCID: PMC8011007 DOI: 10.2215/cjn.07270520] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 01/14/2021] [Indexed: 02/04/2023]
Abstract
BACKGROUND AND OBJECTIVES Animal studies suggest that microvascular rarefaction is a key factor in the acute kidney disease to CKD transition. Hence, delayed graft function appears as a unique human model of AKI to further explore the role of microvascular rarefaction in kidney transplant recipients. Here, we assessed whether delayed graft function is associated with peritubular capillary loss and evaluated the association between this loss and long-term kidney graft function. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS This observational, retrospective cohort study included 61 participants who experienced delayed graft function and 130 who had immediate graft function. We used linear regression models to evaluate associations between delayed graft function and peritubular capillary density expressed as the percentage of efficient cortical area occupied by peritubular capillaries in pre- and post-transplant graft biopsies. eGFRs 1 and 3 years post-transplant were secondary outcomes. RESULTS Post-transplant biopsies were performed at a median of 113 days (interquartile range, 101-128) after transplantation. Peritubular capillary density went from 15.4% to 11.5% in patients with delayed graft function (median change, -3.7%; interquartile range, -6.6% to -0.8%) and from 19.7% to 15.1% in those with immediate graft function (median change, -4.5%; interquartile range, -8.0% to -0.8%). Although the unadjusted change in peritubular capillary density was similar between patients with and without delayed graft function, delayed graft function was associated with more peritubular capillary loss in the multivariable analysis (adjusted difference in change, -2.9%; 95% confidence interval, -4.0 to -1.8). Pretransplant peritubular capillary density and change in peritubular capillary density were associated with eGFR 1 and 3 years post-transplantation. CONCLUSIONS Perioperative AKI is associated with lower density in peritubular capillaries before transplantation and with loss of peritubular capillaries following transplantation. Lower peritubular capillary density is linked to lower long-term eGFR.
Collapse
Affiliation(s)
- Alice Doreille
- Centre de recherche du Centre hospitalier de l’Université de Montréal, Immunopathology axis, Montreal, Quebec, Canada,Faculté de Médecine, Université Paris-Sud, Paris, France
| | - Féryel Azzi
- Institut du cancer de Montréal, Montreal, Quebec, Canada
| | - Stéphanie Larivière-Beaudoin
- Centre de recherche du Centre hospitalier de l’Université de Montréal, Immunopathology axis, Montreal, Quebec, Canada,Canadian Donation and Transplantation Research Program, Edmonton, Alberta, Canada
| | - Annie Karakeussian-Rimbaud
- Centre de recherche du Centre hospitalier de l’Université de Montréal, Immunopathology axis, Montreal, Quebec, Canada,Canadian Donation and Transplantation Research Program, Edmonton, Alberta, Canada
| | - Dominique Trudel
- Institut du cancer de Montréal, Montreal, Quebec, Canada,Pathology Department, Centre hospitalier de l’Université de Montréal, Montreal, Quebec, Canada
| | - Marie-Josée Hébert
- Centre de recherche du Centre hospitalier de l’Université de Montréal, Immunopathology axis, Montreal, Quebec, Canada,Canadian Donation and Transplantation Research Program, Edmonton, Alberta, Canada,Nephrology Department, Centre hospitalier de l’Université de Montréal, Montreal, Quebec, Canada
| | - Mélanie Dieudé
- Centre de recherche du Centre hospitalier de l’Université de Montréal, Immunopathology axis, Montreal, Quebec, Canada,Canadian Donation and Transplantation Research Program, Edmonton, Alberta, Canada
| | - Natacha Patey
- Centre de recherche du Centre hospitalier de l’Université de Montréal, Immunopathology axis, Montreal, Quebec, Canada,Pathology Department, Sainte-Justine Hospital, Montreal, Quebec, Canada
| | - Héloïse Cardinal
- Centre de recherche du Centre hospitalier de l’Université de Montréal, Immunopathology axis, Montreal, Quebec, Canada,Canadian Donation and Transplantation Research Program, Edmonton, Alberta, Canada,Nephrology Department, Centre hospitalier de l’Université de Montréal, Montreal, Quebec, Canada
| |
Collapse
|
17
|
Abstract
Interstitial fibrosis with tubule atrophy (IF/TA) is the response to virtually any sustained kidney injury and correlates inversely with kidney function and allograft survival. IF/TA is driven by various pathways that include hypoxia, renin-angiotensin-aldosterone system, transforming growth factor (TGF)-β signaling, cellular rejection, inflammation and others. In this review we will focus on key pathways in the progress of renal fibrosis, diagnosis and therapy of allograft fibrosis. This review discusses the role and origin of myofibroblasts as matrix producing cells and therapeutic targets in renal fibrosis with a particular focus on renal allografts. We summarize current trends to use multi-omic approaches to identify new biomarkers for IF/TA detection and to predict allograft survival. Furthermore, we review current imaging strategies that might help to identify and follow-up IF/TA complementary or as alternative to invasive biopsies. We further discuss current clinical trials and therapeutic strategies to treat kidney fibrosis.Supplemental Visual Abstract; http://links.lww.com/TP/C141.
Collapse
|
18
|
Peritubular Capillary Rarefaction: An Underappreciated Regulator of CKD Progression. Int J Mol Sci 2020; 21:ijms21218255. [PMID: 33158122 PMCID: PMC7662781 DOI: 10.3390/ijms21218255] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Accepted: 10/29/2020] [Indexed: 12/15/2022] Open
Abstract
Peritubular capillary (PTC) rarefaction is commonly detected in chronic kidney disease (CKD) such as hypertensive nephrosclerosis and diabetic nephropathy. Moreover, PTC rarefaction prominently correlates with impaired kidney function and predicts the future development of end-stage renal disease in patients with CKD. However, it is still underappreciated that PTC rarefaction is a pivotal regulator of CKD progression, primarily because the molecular mechanisms of PTC rarefaction have not been well-elucidated. In addition to the established mechanisms (reduced proangiogenic factors and increased anti-angiogenic factors), recent studies discovered significant contribution of the following elements to PTC loss: (1) prompt susceptibility of PTC to injury, (2) impaired proliferation of PTC, (3) apoptosis/senescence of PTC, and (4) pericyte detachment from PTC. Mainly based on the recent and novel findings in basic research and clinical study, this review describes the roles of the above-mentioned elements in PTC loss and focuses on the major factors regulating PTC angiogenesis, the assessment of PTC rarefaction and its surrogate markers, and an overview of the possible therapeutic agents to mitigate PTC rarefaction during CKD progression. PTC rarefaction is not only a prominent histological characteristic of CKD but also a central driving force of CKD progression.
Collapse
|
19
|
de Boer A, Pieters TT, Harteveld AA, Blankestijn PJ, Bos C, Froeling M, Goldschmeding R, Hoogduin HJM, Joles JA, Petri BJ, Verhaar MC, Leiner T, Nguyen TQ, van Zuilen AD. Validation of multiparametric MRI by histopathology after nephrectomy: a case study. MAGNETIC RESONANCE MATERIALS IN PHYSICS BIOLOGY AND MEDICINE 2020; 34:377-387. [PMID: 32954447 PMCID: PMC8154819 DOI: 10.1007/s10334-020-00887-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 07/23/2020] [Accepted: 09/01/2020] [Indexed: 11/28/2022]
Abstract
OBJECTIVES Renal multiparametric MRI (mpMRI) is a promising tool to monitor renal allograft health to enable timely treatment of chronic allograft nephropathy. This study aims to validate mpMRI by whole-kidney histology following transplantectomy. MATERIALS AND METHODS A patient with kidney transplant failure underwent mpMRI prior to transplantectomy. The mpMRI included blood oxygenation level-dependent (BOLD) MRI, T1 and T2 mapping, diffusion-weighted imaging (DWI), 2D phase contrast (2DPC) and arterial spin labeling (ASL). Parenchymal mpMRI measures were compared to normative values obtained in 19 healthy controls. Differences were expressed in standard deviations (SD) of normative values. The mpMRI measures were compared qualitatively to histology. RESULTS The mpMRI showed a heterogeneous parenchyma consistent with extensive interstitial hemorrhage on histology. A global increase in T1 (+ 3.0 SD) and restricted diffusivity (- 3.6 SD) were consistent with inflammation and fibrosis. Decreased T2 (- 1.8 SD) indicated fibrosis or hemorrhage. ASL showed diminished cortical perfusion (- 2.9 SD) with patent proximal arteries. 2DPC revealed a 69% decrease in renal perfusion. Histological evaluation showed a dense inflammatory infiltrate and fibrotic changes, consistent with mpMRI results. Most interlobular arteries were obliterated while proximal arteries were patent, consistent with ASL findings. DISCUSSION mpMRI findings correlated well with histology both globally as well as locally.
Collapse
Affiliation(s)
- Anneloes de Boer
- Department of Radiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.
| | - Tobias T Pieters
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Anita A Harteveld
- Department of Radiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Peter J Blankestijn
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Clemens Bos
- Department of Radiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Martijn Froeling
- Department of Radiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Roel Goldschmeding
- Department of Pathology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Hans J M Hoogduin
- Department of Radiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Jaap A Joles
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Bart-Jeroen Petri
- Department of Vascular Surgery, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Marianne C Verhaar
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Tim Leiner
- Department of Radiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Tri Q Nguyen
- Department of Pathology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Arjan D van Zuilen
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
20
|
Xu-Dubois YC, Ahmadpoor P, Brocheriou I, Louis K, Arzouk Snanoudj N, Rouvier P, Taupin JL, Corchia A, Galichon P, Barrou B, Giraud S, Hauet T, Jouanneau C, Rodenas A, Placier S, Niasse A, Ouchelouche S, Naimi BY, Akil E, Hertig A, Buob D, Rondeau E. Microvasculature partial endothelial mesenchymal transition in early posttransplant biopsy with acute tubular necrosis identifies poor recovery renal allografts. Am J Transplant 2020; 20:2400-2412. [PMID: 32167213 DOI: 10.1111/ajt.15847] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 03/05/2020] [Accepted: 03/08/2020] [Indexed: 01/25/2023]
Abstract
Acute tubular necrosis (ATN), a frequent histopathological feature in the early post-renal transplant biopsy, affects long-term graft function. Appropriate markers to identify patients at risk of no or incomplete recovery after delayed graft function are lacking. In this study, we first included 41 renal transplant patients whose biopsy for cause during the first month after transplantation showed ATN lesions. Using partial microvasculature endothelial (fascin, vimentin) and tubular epithelial (vimentin) to mesenchymal transition markers, detected by immunohistochemistry, we found a significant association between partial endothelial to mesenchymal transition and poor graft function recovery (Spearman's rho = -0.55, P = .0005). Transforming growth factor-β1 was strongly expressed in these phenotypic changed endothelial cells. Extent of ATN was also correlated with short- and long-term graft dysfunction. However, the association of extensive ATN with long-term graft dysfunction (24 months posttransplant) was observed only in patients with partial endothelial to mesenchymal transition marker expression in their grafts (Spearman's rho = -0.64, P = .003), but not in those without. The association of partial endothelial to mesenchymal transition with worse renal graft outcome was confirmed on 34 other early biopsies with ATN from a second transplant center. Our results suggest that endothelial cell activation at the early phase of renal transplantation plays a detrimental role.
Collapse
Affiliation(s)
- Yi-Chun Xu-Dubois
- Sorbonne Université, Institut national de la santé et de la recherche médicale (INSERM), Unité mixte de recherche (UMR), Paris, France.,Public Health, Assistance Publique-Hôpitaux de Paris (AP-HP), Tenon Hospital, Paris, France.,Kidney Transplantation, AP-HP, Tenon Hospital, Paris, France
| | | | | | - Kevin Louis
- Sorbonne Université, Institut national de la santé et de la recherche médicale (INSERM), Unité mixte de recherche (UMR), Paris, France
| | - Nadia Arzouk Snanoudj
- Department of Kidney Transplantation, AP-HP, Pitié Salpêtrière Hospital, Paris, France
| | - Philippe Rouvier
- Department of Pathology, AP-HP, Pitié Salpêtrière Hospital, Paris, France
| | - Jean-Luc Taupin
- Laboratoire d'Immunologie et Histocompatibilité Hôpital Saint-Louis, Paris, France.,Université Paris Diderot, INSERM UMR, Institut Universitaire d'Hématologie, Paris, France
| | - Anthony Corchia
- Sorbonne Université, Institut national de la santé et de la recherche médicale (INSERM), Unité mixte de recherche (UMR), Paris, France
| | - Pierre Galichon
- Sorbonne Université, Institut national de la santé et de la recherche médicale (INSERM), Unité mixte de recherche (UMR), Paris, France.,Kidney Transplantation, AP-HP, Tenon Hospital, Paris, France
| | - Benoit Barrou
- Department of Kidney Transplantation, AP-HP, Pitié Salpêtrière Hospital, Paris, France
| | - Sébastien Giraud
- Service de Biochimie, CHU de Poitiers, Pôle Biospharm, Poitiers, France.,INSERM UMR 1082 IRTOMIT, Poitiers, France
| | - Thierry Hauet
- Service de Biochimie, CHU de Poitiers, Pôle Biospharm, Poitiers, France.,INSERM UMR 1082 IRTOMIT, Poitiers, France.,Faculté de Médecine et de Pharmacie, Université de Poitiers, Poitiers, France
| | - Chantal Jouanneau
- Sorbonne Université, Institut national de la santé et de la recherche médicale (INSERM), Unité mixte de recherche (UMR), Paris, France
| | - Anita Rodenas
- Department of Pathology, AP-HP, Tenon Hospital, Paris, France
| | - Sandrine Placier
- Sorbonne Université, Institut national de la santé et de la recherche médicale (INSERM), Unité mixte de recherche (UMR), Paris, France
| | - Aissata Niasse
- Sorbonne Université, Institut national de la santé et de la recherche médicale (INSERM), Unité mixte de recherche (UMR), Paris, France
| | - Souhila Ouchelouche
- Sorbonne Université, Institut national de la santé et de la recherche médicale (INSERM), Unité mixte de recherche (UMR), Paris, France
| | | | | | - Alexandre Hertig
- Sorbonne Université, Institut national de la santé et de la recherche médicale (INSERM), Unité mixte de recherche (UMR), Paris, France.,Kidney Transplantation, AP-HP, Tenon Hospital, Paris, France.,Department of Kidney Transplantation, AP-HP, Pitié Salpêtrière Hospital, Paris, France
| | - David Buob
- Sorbonne Université, Institut national de la santé et de la recherche médicale (INSERM), Unité mixte de recherche (UMR), Paris, France.,Department of Pathology, AP-HP, Tenon Hospital, Paris, France
| | - Eric Rondeau
- Sorbonne Université, Institut national de la santé et de la recherche médicale (INSERM), Unité mixte de recherche (UMR), Paris, France.,Kidney Transplantation, AP-HP, Tenon Hospital, Paris, France
| |
Collapse
|
21
|
Ullah MM, Basile DP. Role of Renal Hypoxia in the Progression From Acute Kidney Injury to Chronic Kidney Disease. Semin Nephrol 2020; 39:567-580. [PMID: 31836039 DOI: 10.1016/j.semnephrol.2019.10.006] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Over the past 20 years, there has been an increased appreciation of the long-term sequelae of acute kidney injury (AKI) and the potential development of chronic kidney disease (CKD). Several pathophysiologic features have been proposed to mediate AKI to CKD progression including maladaptive alterations in tubular, interstitial, inflammatory, and vascular cells. These alterations likely interact to culminate in the progression to CKD. In this article we focus primarily on evidence of vascular rarefaction secondary to AKI, and the potential mechanisms by which rarefaction occurs in relation to other alterations in tubular and interstitial compartments. We further focus on the potential that rarefaction contributes to renal hypoxia. Consideration of the role of hypoxia in AKI to CKD transition focuses on experimental evidence of persistent renal hypoxia after AKI and experimental maneuvers to evaluate the influence of hypoxia, per se, in progressive disease. Finally, consideration of methods to evaluate hypoxia in patients is provided with the suggestion that noninvasive measurement of renal hypoxia may provide insight into progression in post-AKI patients.
Collapse
Affiliation(s)
- Md Mahbub Ullah
- Department of Anatomy, Cell Biology and Physiology, Indiana University, Indianapolis, IN
| | - David P Basile
- Department of Medicine, Division of Nephrology, Indiana University, Indianapolis, IN.
| |
Collapse
|
22
|
Hysi E, He X, Fadhel MN, Zhang T, Krizova A, Ordon M, Farcas M, Pace KT, Mintsopoulos V, Lee WL, Kolios MC, Yuen DA. Photoacoustic imaging of kidney fibrosis for assessing pretransplant organ quality. JCI Insight 2020; 5:136995. [PMID: 32298239 DOI: 10.1172/jci.insight.136995] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 04/09/2020] [Indexed: 12/12/2022] Open
Abstract
Roughly 10% of the world's population has chronic kidney disease (CKD). In its advanced stages, CKD greatly increases the risk of hospitalization and death. Although kidney transplantation has revolutionized the care of advanced CKD, clinicians have limited ways of assessing donor kidney quality. Thus, optimal donor kidney-recipient matching cannot be performed, meaning that some patients receive damaged kidneys that function poorly. Fibrosis is a form of chronic damage often present in donor kidneys, and it is an important predictor of future renal function. Currently, no safe, easy-to-perform technique exists that accurately quantifies renal fibrosis. We describe a potentially novel photoacoustic (PA) imaging technique that directly images collagen, the principal component of fibrotic tissue. PA imaging noninvasively quantifies whole kidney fibrotic burden in mice, and cortical fibrosis in pig and human kidneys, with outstanding accuracy and speed. Remarkably, 3-dimensional PA imaging exhibited sufficiently high resolution to capture intrarenal variations in collagen content. We further show that PA imaging can be performed in a setting that mimics human kidney transplantation, suggesting the potential for rapid clinical translation. Taken together, our data suggest that PA collagen imaging is a major advance in fibrosis quantification that could have widespread preclinical and clinical impact.
Collapse
Affiliation(s)
- Eno Hysi
- Department of Physics, Ryerson University, Toronto, Canada.,Institute for Biomedical Engineering, Science and Technology (iBEST), a partnership between Ryerson University and St. Michael's Hospital, Toronto, Canada
| | - Xiaolin He
- Institute for Biomedical Engineering, Science and Technology (iBEST), a partnership between Ryerson University and St. Michael's Hospital, Toronto, Canada.,Division of Nephrology, Department of Medicine, St. Michael's Hospital, Unity Health Toronto and University of Toronto, Toronto, Canada.,Keenan Research Centre for Biomedical Science and
| | - Muhannad N Fadhel
- Department of Physics, Ryerson University, Toronto, Canada.,Institute for Biomedical Engineering, Science and Technology (iBEST), a partnership between Ryerson University and St. Michael's Hospital, Toronto, Canada
| | - Tianzhou Zhang
- Division of Nephrology, Department of Medicine, St. Michael's Hospital, Unity Health Toronto and University of Toronto, Toronto, Canada.,Keenan Research Centre for Biomedical Science and
| | - Adriana Krizova
- Keenan Research Centre for Biomedical Science and.,Department of Laboratory Medicine, St. Michael's Hospital, Unity Health Toronto, Toronto, Canada
| | - Michael Ordon
- Keenan Research Centre for Biomedical Science and.,Department of Laboratory Medicine, St. Michael's Hospital, Unity Health Toronto, Toronto, Canada.,Division of Urology, Department of Surgery, St. Michael's Hospital, Unity Health Toronto and University of Toronto, Toronto, Ontario, Canada
| | - Monica Farcas
- Keenan Research Centre for Biomedical Science and.,Department of Laboratory Medicine, St. Michael's Hospital, Unity Health Toronto, Toronto, Canada.,Division of Urology, Department of Surgery, St. Michael's Hospital, Unity Health Toronto and University of Toronto, Toronto, Ontario, Canada
| | - Kenneth T Pace
- Keenan Research Centre for Biomedical Science and.,Department of Laboratory Medicine, St. Michael's Hospital, Unity Health Toronto, Toronto, Canada.,Division of Urology, Department of Surgery, St. Michael's Hospital, Unity Health Toronto and University of Toronto, Toronto, Ontario, Canada
| | - Victoria Mintsopoulos
- Keenan Research Centre for Biomedical Science and.,Interdepartmental Division of Critical Care Medicine, St. Michael's Hospital, University of Toronto, Toronto, Canada
| | - Warren L Lee
- Keenan Research Centre for Biomedical Science and.,Interdepartmental Division of Critical Care Medicine, St. Michael's Hospital, University of Toronto, Toronto, Canada
| | - Michael C Kolios
- Department of Physics, Ryerson University, Toronto, Canada.,Institute for Biomedical Engineering, Science and Technology (iBEST), a partnership between Ryerson University and St. Michael's Hospital, Toronto, Canada
| | - Darren A Yuen
- Institute for Biomedical Engineering, Science and Technology (iBEST), a partnership between Ryerson University and St. Michael's Hospital, Toronto, Canada.,Division of Nephrology, Department of Medicine, St. Michael's Hospital, Unity Health Toronto and University of Toronto, Toronto, Canada.,Keenan Research Centre for Biomedical Science and
| |
Collapse
|
23
|
Chen Q, Yu J, Rush BM, Stocker SD, Tan RJ, Kim K. Ultrasound super-resolution imaging provides a noninvasive assessment of renal microvasculature changes during mouse acute kidney injury. Kidney Int 2020; 98:355-365. [PMID: 32600826 DOI: 10.1016/j.kint.2020.02.011] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 01/22/2020] [Accepted: 02/06/2020] [Indexed: 12/26/2022]
Abstract
Acute kidney injury (AKI) is a risk factor for the development of chronic kidney disease (CKD). One mechanism for this phenomenon is renal microvascular rarefaction and subsequent chronic impairment in perfusion. However, diagnostic tools to monitor the renal microvasculature in a noninvasive and quantitative manner are still lacking. Ultrasound super-resolution imaging is an emerging technology that can identify microvessels with unprecedented resolution. Here, we applied this imaging technique to identify microvessels in the unilateral ischemia-reperfusion injury mouse model of AKI-to-CKD progression in vivo. Kidneys from 21 and 42 day post- ischemia-reperfusion injury, the contralateral uninjured kidneys, and kidneys from sham-operated mice were examined by ultrasound super-resolution and histology. Renal microvessels were successfully identified by this imaging modality with a resolution down to 32 μm. Renal fibrosis was observed in all kidneys with ischemia-reperfusion injury and was associated with a significant reduction in kidney size, cortical thickness, relative blood volume, and microvascular density as assessed by this imaging. Tortuosity of the cortical microvasculature was also significantly increased at 42 days compared to sham. These vessel density measurements correlated significantly with CD31 immunohistochemistry (R2=0.77). Thus, ultrasound super-resolution imaging provides unprecedented resolution and is capable of noninvasive quantification of renal vasculature changes associated with AKI-to-CKD progression in mice. Hence, this technique could be a promising diagnostic tool for monitoring progressive kidney disease.
Collapse
Affiliation(s)
- Qiyang Chen
- Department of Bioengineering, University of Pittsburgh School of Engineering, Pittsburgh, Pennsylvania, USA; Center for Ultrasound Molecular Imaging and Therapeutics, Department of Medicine, Heart and Vascular Institute, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Jaesok Yu
- Department of Bioengineering, University of Pittsburgh School of Engineering, Pittsburgh, Pennsylvania, USA; Center for Ultrasound Molecular Imaging and Therapeutics, Department of Medicine, Heart and Vascular Institute, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Brittney M Rush
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Sean D Stocker
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Roderick J Tan
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA.
| | - Kang Kim
- Department of Bioengineering, University of Pittsburgh School of Engineering, Pittsburgh, Pennsylvania, USA; Center for Ultrasound Molecular Imaging and Therapeutics, Department of Medicine, Heart and Vascular Institute, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA; Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA; McGowan Institute of Regenerative Medicine, University of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA; Department of Mechanical Engineering and Materials Science, University of Pittsburgh School of Engineering, Pittsburgh, Pennsylvania, USA.
| |
Collapse
|
24
|
Woda CB, Bruneau S, Mak AL, Haskova Z, Liu K, Ghosh CC, Briscoe DM. Calcineurin inhibitors augment endothelial-to-mesenchymal transition by enhancing proliferation in association with cytokine-mediated activation. Biochem Biophys Res Commun 2019; 519:667-673. [PMID: 31542230 PMCID: PMC7119266 DOI: 10.1016/j.bbrc.2019.09.043] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Accepted: 09/11/2019] [Indexed: 12/21/2022]
Abstract
Calcineurin Inhibitors (CNIs) are routinely used for immunosuppression following solid organ transplantation. However, the prolonged use of these agents lead to organ fibrosis which limits their efficacy. CNIs induce TGFβ expression, which is reported to augment endothelial-to-mesenchymal transition (EndMT), but their role in this process is not known. In these studies, we find that the CNIs FK506 and cyclosporine (CsA) are potent to increase endothelial cell (EC) proliferation using established in vitro assays (P < 0.05). Furthermore, using phosphokinase arrays, we find that each CNI activates the MAPK and Akt/mTOR signaling pathways, and that pharmacological inhibition of each pathway targets CNI-induced proliferative responses (P < 0.001). EndMT was evaluated by FACS for N-cadherin and CD31 expression and by qPCR for the expression of α-smooth muscle actin, N-cadherin and Snail. We find that CNIs do not directly induce dedifferentiation, while TGFβ and hypoxia induce EndMT in small numbers of EC. In contrast, the treatment of EC with the inflammatory cytokine TNFα was potent to elicit an EndMT response, and its effects were most notably in EC following proliferation/doubling. Taken together, these observations suggest that CNIs elicit proliferative responses, which enhance EndMT in association with local inflammation. The clinical implications of these findings are that anti-proliferative therapeutics have high potential to target the initiation of this EndMT response.
Collapse
Affiliation(s)
- Craig B Woda
- The Transplant Research Program and the Division of Nephrology, Boston Children's Hospital, Boston, MA, 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
| | - Sarah Bruneau
- The Transplant Research Program and the Division of Nephrology, Boston Children's Hospital, Boston, MA, 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
| | - Anne Linde Mak
- The Transplant Research Program and the Division of Nephrology, Boston Children's Hospital, Boston, MA, 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
| | - Zdenka Haskova
- The Transplant Research Program and the Division of Nephrology, Boston Children's Hospital, Boston, MA, 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
| | - Kaifeng Liu
- The Transplant Research Program and the Division of Nephrology, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Chandra C Ghosh
- The Transplant Research Program and the Division of Nephrology, Boston Children's Hospital, Boston, MA, 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
| | - David M Briscoe
- The Transplant Research Program and the Division of Nephrology, Boston Children's Hospital, Boston, MA, 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
25
|
Menshikh A, Scarfe L, Delgado R, Finney C, Zhu Y, Yang H, de Caestecker MP. Capillary rarefaction is more closely associated with CKD progression after cisplatin, rhabdomyolysis, and ischemia-reperfusion-induced AKI than renal fibrosis. Am J Physiol Renal Physiol 2019; 317:F1383-F1397. [PMID: 31509009 PMCID: PMC6879932 DOI: 10.1152/ajprenal.00366.2019] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Revised: 08/26/2019] [Accepted: 09/09/2019] [Indexed: 02/07/2023] Open
Abstract
Acute kidney injury (AKI) is a strong independent predictor of mortality and often results in incomplete recovery of renal function, leading to progressive chronic kidney disease (CKD). Many clinical trials have been conducted on the basis of promising preclinical data, but no therapeutic interventions have been shown to improve long-term outcomes after AKI. This is partly due to the failure of preclinical studies to accurately model clinically relevant injury and long-term outcomes on CKD progression. Here, we evaluated the long-term effects of AKI on CKD progression in three animal models reflecting diverse etiologies of AKI: repeat-dose cisplatin, rhabdomyolysis, and ischemia-reperfusion injury. Using transdermal measurement of glomerular filtration rate as a clinically relevant measure of kidney function and quantification of peritubular capillary density to measure capillary rarefaction, we showed that repeat-dose cisplatin caused capillary rarefaction and decreased renal function in mice without a significant increase in interstitial fibrosis, whereas rhabdomyolysis-induced AKI led to severe interstitial fibrosis, but renal function and peritubular capillary density were preserved. Furthermore, long-term experiments in mice with unilateral ischemia-reperfusion injury showed that restoration of renal function 12 wk after a contralateral nephrectomy was associated with increasing fibrosis, but a reversal of capillary rarefaction was seen at 4 wk. These data demonstrate that clear dissociation between kidney function and fibrosis in these models of AKI to CKD progression and suggest that peritubular capillary rarefaction is more strongly associated with CKD progression than renal fibrosis.
Collapse
Affiliation(s)
- Anna Menshikh
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Lauren Scarfe
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Rachel Delgado
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Charlene Finney
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Yuantee Zhu
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Haichun Yang
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Mark P de Caestecker
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
26
|
Situmorang GR, Sheerin NS. Ischaemia reperfusion injury: mechanisms of progression to chronic graft dysfunction. Pediatr Nephrol 2019; 34:951-963. [PMID: 29603016 PMCID: PMC6477994 DOI: 10.1007/s00467-018-3940-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 02/18/2018] [Accepted: 03/02/2018] [Indexed: 12/18/2022]
Abstract
The increasing use of extended criteria organs to meet the demand for kidney transplantation raises an important question of how the severity of early ischaemic injury influences long-term outcomes. Significant acute ischaemic kidney injury is associated with delayed graft function, increased immune-associated events and, ultimately, earlier deterioration of graft function. A comprehensive understanding of immediate molecular events that ensue post-ischaemia and their potential long-term consequences are key to the discovery of novel therapeutic targets. Acute ischaemic injury primarily affects tubular structure and function. Depending on the severity and persistence of the insult, this may resolve completely, leading to restoration of normal function, or be sustained, resulting in persistent renal impairment and progressive functional loss. Long-term effects of acute renal ischaemia are mediated by several mechanisms including hypoxia, HIF-1 activation, endothelial dysfunction leading to vascular rarefaction, sustained pro-inflammatory stimuli involving innate and adaptive immune responses, failure of tubular cells to recover and epigenetic changes. This review describes the biological relevance and interaction of these mechanisms based on currently available evidence.
Collapse
Affiliation(s)
- Gerhard R Situmorang
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
- Urology Department, Faculty of Medicine Universitas Indonesia - Cipto Mangunkusumo Hospital, Jakarta, 10430, Indonesia
| | - Neil S Sheerin
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK.
| |
Collapse
|
27
|
Sun IO, Santelli A, Abumoawad A, Eirin A, Ferguson CM, Woollard JR, Lerman A, Textor SC, Puranik AS, Lerman LO. Loss of Renal Peritubular Capillaries in Hypertensive Patients Is Detectable by Urinary Endothelial Microparticle Levels. Hypertension 2019; 72:1180-1188. [PMID: 30354805 DOI: 10.1161/hypertensionaha.118.11766] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Hypertension, an important cause of chronic kidney disease, is characterized by peritubular capillary (PTC) loss. Circulating levels of endothelial microparticles (EMPs) reflect systemic endothelial injury. We hypothesized that systemic and urinary PTC-EMPs levels would reflect renal microvascular injury in hypertensive patients. We prospectively measured by flow cytometry renal vein, inferior vena cava, and urinary levels of EMPs in essential (n=14) and renovascular (RVH; n=24) hypertensive patients and compared them with peripheral blood and urinary levels in healthy volunteers (n=14). PTC-EMPs were identified as urinary exosomes positive for the PTC marker plasmalemmal-vesicle-associated protein. In 7 RVH patients, PTC and fibrosis were also quantified in renal biopsy, and in 18 RVH patients, PTC-EMPs were measured again 3 months after continued medical therapy with or without stenting (n=9 each). Renal vein and systemic PTC-EMPs levels were not different among the groups, whereas their urinary levels were elevated in both RVH and essential hypertension versus healthy volunteers (56.8%±12.7% and 62.8%±10.7% versus 34.0%±17.8%; both P≤0.001). Urinary PTC-EMPs levels correlated directly with blood pressure and inversely with estimated glomerular filtration rate. Furthermore, in RVH, urinary PTC-EMPs levels correlated directly with stenotic kidney hypoxia, histological PTC count, and fibrosis and inversely with cortical perfusion. Three months after treatment, the change in urinary PTC-EMPs levels correlated inversely with a change in renal function ( r=-0.582; P=0.011). Therefore, urinary PTC-EMPs levels are increased in hypertensive patients and may reflect renal microcirculation injury, whereas systemic PTC-EMPs levels are unchanged. Urinary PTC-EMPs may be useful as novel biomarkers of intrarenal capillary loss.
Collapse
Affiliation(s)
- In O Sun
- From the Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN (I.O.S., A.S., A.A., A.E., C.M.F., J.R.W., S.C.T., A.S.P., L.O.L.).,Division of Nephrology, Department of Internal Medicine, Presbyterian Medical Center, Jeonju, Korea (I.O.S.)
| | - Adrian Santelli
- From the Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN (I.O.S., A.S., A.A., A.E., C.M.F., J.R.W., S.C.T., A.S.P., L.O.L.)
| | - Abdelrhman Abumoawad
- From the Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN (I.O.S., A.S., A.A., A.E., C.M.F., J.R.W., S.C.T., A.S.P., L.O.L.)
| | - Alfonso Eirin
- From the Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN (I.O.S., A.S., A.A., A.E., C.M.F., J.R.W., S.C.T., A.S.P., L.O.L.)
| | - Christopher M Ferguson
- From the Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN (I.O.S., A.S., A.A., A.E., C.M.F., J.R.W., S.C.T., A.S.P., L.O.L.)
| | - John R Woollard
- From the Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN (I.O.S., A.S., A.A., A.E., C.M.F., J.R.W., S.C.T., A.S.P., L.O.L.)
| | - Amir Lerman
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN (A.L.)
| | - Stephen C Textor
- From the Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN (I.O.S., A.S., A.A., A.E., C.M.F., J.R.W., S.C.T., A.S.P., L.O.L.)
| | - Amrutesh S Puranik
- From the Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN (I.O.S., A.S., A.A., A.E., C.M.F., J.R.W., S.C.T., A.S.P., L.O.L.)
| | - Lilach O Lerman
- From the Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN (I.O.S., A.S., A.A., A.E., C.M.F., J.R.W., S.C.T., A.S.P., L.O.L.)
| |
Collapse
|
28
|
Wewers TM, Mayer AB, Pfleiderer A, Beul K, Schmidt R, Heitplatz B, Van Marck V, Nolte I, Pavenstädt H, Reuter S, Brand M, Di Marco GS. Increased soluble fms-like tyrosine kinase 1 after ischemia reperfusion contributes to adverse clinical outcomes following kidney transplantation. Kidney Int 2019; 95:1091-1102. [PMID: 30824181 DOI: 10.1016/j.kint.2018.11.023] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 10/05/2018] [Accepted: 11/02/2018] [Indexed: 12/20/2022]
Abstract
Renal ischemia reperfusion injury (IRI) adversely affects clinical outcomes following kidney transplantation. Understanding the cellular mechanisms and the changes in gene/protein expression following IRI may help to improve these outcomes. Serum soluble fms-like tyrosine kinase 1 (sFlt-1), a circulating antiangiogenic protein, is increased in the first week following kidney transplantation. We evaluated the casual relationship of elevated sFlt-1 levels with renal microvascular dysfunction following IRI in a longitudinal study of 93 kidney transplant recipients and in several animal models. Transplant recipients with higher sFlt-1 levels had higher odds of delayed graft function, graft rejection, impaired graft function, and death. In a subgroup of 25 participants who underwent kidney biopsy within 4 months of kidney transplantation, peritubular capillary area was lower in those with elevated serum sFtl-1 levels. The administration of recombinant sFlt-1 into rodents resulted in significant structural and functional changes of the renal microvasculature, including reduced peritubular capillary density and intracapillary blood volume, and lead to increased expression of inflammatory genes and increased fibrosis. In a murine model of IRI, the kidney was a site of sFlt-1 production, and systemic neutralization of sFlt-1 preserved peritubular capillary density and alleviated renal fibrosis. Our data indicate that high sFlt-1 levels after IRI play an important role in the pathogenesis of microvascular dysfunction, thereby contributing to adverse clinical outcomes following kidney transplantation.
Collapse
Affiliation(s)
- Theresa M Wewers
- Department of Internal Medicine D, University Hospital Münster, Münster, Germany; Small Animal Hospital, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Anna B Mayer
- Department of Internal Medicine D, University Hospital Münster, Münster, Germany
| | - Alexander Pfleiderer
- Department of Internal Medicine D, University Hospital Münster, Münster, Germany
| | - Katrin Beul
- Department of Internal Medicine D, University Hospital Münster, Münster, Germany
| | - Rene Schmidt
- Institute for Biostatistics and Clinical Research, University Hospital Münster, Münster, Germany
| | - Barbara Heitplatz
- Department of Pathology, University Hospital Münster, Münster, Germany
| | - Veerle Van Marck
- Department of Pathology, University Hospital Münster, Münster, Germany
| | - Ingo Nolte
- Small Animal Hospital, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Hermann Pavenstädt
- Department of Internal Medicine D, University Hospital Münster, Münster, Germany
| | - Stefan Reuter
- Department of Internal Medicine D, University Hospital Münster, Münster, Germany
| | - Marcus Brand
- Department of Internal Medicine D, University Hospital Münster, Münster, Germany
| | - Giovana S Di Marco
- Department of Internal Medicine D, University Hospital Münster, Münster, Germany.
| |
Collapse
|
29
|
Doreille A, Dieudé M, Cardinal H. The determinants, biomarkers, and consequences of microvascular injury in kidney transplant recipients. Am J Physiol Renal Physiol 2018; 316:F9-F19. [PMID: 30379097 DOI: 10.1152/ajprenal.00163.2018] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Independent of the initial cause of kidney disease, microvascular injury to the peritubular capillary network appears to play a central role in the development of interstitial fibrosis in both native and transplanted kidney disease. This association is explained by mechanisms such as the upregulation of profibrotic genes and epigenetic changes induced by hypoxia, capillary leakage, endothelial and pericyte transition to interstitial fibroblasts, as well as modifications in the secretome of endothelial cells. Alloimmune injury due to antibody-mediated rejection and ischemia-reperfusion injury are the two main etiologies of microvascular damage in kidney transplant recipients. The presence of circulating donor-specific anti-human leukocyte antigen (HLA) antibodies, histological findings, such as diffuse C4d staining in peritubular capillaries, and the extent and severity of peritubular capillaritis, are commonly used clinically to provide both diagnostic and prognostic information. Complement-dependent assays, circulating non-HLA antibodies, or evaluation of the microvasculature with novel imaging techniques are the subject of ongoing studies.
Collapse
Affiliation(s)
- Alice Doreille
- Research Centre, Centre Hospitalier de l'Université de Montréal , Montreal, Quebec , Canada.,Université Paris-Sud , Paris , France
| | - Mélanie Dieudé
- Research Centre, Centre Hospitalier de l'Université de Montréal , Montreal, Quebec , Canada.,Canadian Donation and Transplantation Research Program, Montreal, Quebec, Canada
| | - Heloise Cardinal
- Research Centre, Centre Hospitalier de l'Université de Montréal , Montreal, Quebec , Canada.,Canadian Donation and Transplantation Research Program, Montreal, Quebec, Canada
| |
Collapse
|
30
|
Yang B, Lan S, Dieudé M, Sabo-Vatasescu JP, Karakeussian-Rimbaud A, Turgeon J, Qi S, Gunaratnam L, Patey N, Hébert MJ. Caspase-3 Is a Pivotal Regulator of Microvascular Rarefaction and Renal Fibrosis after Ischemia-Reperfusion Injury. J Am Soc Nephrol 2018; 29:1900-1916. [PMID: 29925521 DOI: 10.1681/asn.2017050581] [Citation(s) in RCA: 101] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 05/11/2018] [Indexed: 12/31/2022] Open
Abstract
Background Ischemia-reperfusion injury (IRI) is a major risk factor for chronic renal failure. Here, we characterize the different modes of programmed cell death in the tubular and microvascular compartments during the various stages of IRI-induced AKI, and their relative importance to renal fibrogenesis.Methods We performed unilateral renal artery clamping for 30 minutes and contralateral nephrectomy in wild-type mice (C57BL/6) or caspase-3-/- mice.Results Compared with their wild-type counterparts, caspase-3-/- mice in the early stage of AKI had high urine cystatin C levels, tubular injury scores, and serum creatinine levels. Electron microscopy revealed evidence of tubular epithelial cell necrosis in caspase-3-/- mice, and immunohistochemistry showed upregulation of the necroptosis marker receptor-interacting serine/threonine-protein kinase 3 (RIPK3) in renal cortical sections. Western blot analysis further demonstrated enhanced levels of phosphorylated RIPK3 in the kidneys of caspase-3-/- mice. In contrast, caspase-3-/- mice had less microvascular congestion and activation in the early and extension phases of AKI. In the long term (3 weeks after IRI), caspase-3-/- mice had reduced microvascular rarefaction and renal fibrosis, as well as decreased expression of α-smooth muscle actin and reduced collagen deposition within peritubular capillaries. Moreover, caspase-3-/- mice exhibited signs of reduced tubular ischemia, including lower tubular expression of hypoxia-inducible factor-1α and improved tubular injury scores.Conclusions These results establish the pivotal importance of caspase-3 in regulating microvascular endothelial cell apoptosis and renal fibrosis after IRI. These findings also demonstrate the predominant role of microvascular over tubular injury as a driver of progressive renal damage and fibrosis after IRI.
Collapse
Affiliation(s)
- Bing Yang
- Research Centre, Centre hospitalier de l'Université de Montréal, Montreal, Quebec, Canada.,Canadian National Transplant Research Program, Edmonton, Alberta, Canada.,Université de Montréal, Montreal, Quebec, Canada
| | - Shanshan Lan
- Research Centre, Centre hospitalier de l'Université de Montréal, Montreal, Quebec, Canada.,Canadian National Transplant Research Program, Edmonton, Alberta, Canada.,Université de Montréal, Montreal, Quebec, Canada
| | - Mélanie Dieudé
- Research Centre, Centre hospitalier de l'Université de Montréal, Montreal, Quebec, Canada.,Canadian National Transplant Research Program, Edmonton, Alberta, Canada.,Université de Montréal, Montreal, Quebec, Canada
| | | | - Annie Karakeussian-Rimbaud
- Research Centre, Centre hospitalier de l'Université de Montréal, Montreal, Quebec, Canada.,Canadian National Transplant Research Program, Edmonton, Alberta, Canada
| | - Julie Turgeon
- Research Centre, Centre hospitalier de l'Université de Montréal, Montreal, Quebec, Canada.,Canadian National Transplant Research Program, Edmonton, Alberta, Canada
| | - Shijie Qi
- Research Centre, Centre hospitalier de l'Université de Montréal, Montreal, Quebec, Canada.,Canadian National Transplant Research Program, Edmonton, Alberta, Canada
| | - Lakshman Gunaratnam
- Canadian National Transplant Research Program, Edmonton, Alberta, Canada.,London Health Sciences Centre, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada; and
| | - Natalie Patey
- Research Centre, Centre hospitalier de l'Université de Montréal, Montreal, Quebec, Canada; .,Canadian National Transplant Research Program, Edmonton, Alberta, Canada.,Université de Montréal, Montreal, Quebec, Canada.,Department of Pathology, Centre hospitalier universitaire Sainte-Justine, Université de Montréal, Montreal, Quebec, Canada
| | - Marie-Josée Hébert
- Research Centre, Centre hospitalier de l'Université de Montréal, Montreal, Quebec, Canada; .,Canadian National Transplant Research Program, Edmonton, Alberta, Canada.,Université de Montréal, Montreal, Quebec, Canada
| |
Collapse
|
31
|
Cardinal H, Dieudé M, Hébert MJ. Endothelial Dysfunction in Kidney Transplantation. Front Immunol 2018; 9:1130. [PMID: 29875776 PMCID: PMC5974048 DOI: 10.3389/fimmu.2018.01130] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 05/04/2018] [Indexed: 12/17/2022] Open
Abstract
Kidney transplantation entails a high likelihood of endothelial injury. The endothelium is a target of choice for injury by ischemia-reperfusion, alloantibodies, and autoantibodies. A certain degree of ischemia-reperfusion injury inevitably occurs in the immediate posttransplant setting and can manifest as delayed graft function. Acute rejection episodes, whether T-cell or antibody-mediated, can involve the graft micro- and macrovasculature, leading to endothelial injury and adverse long-term consequences on graft function and survival. In turn, caspase-3 activation in injured and dying endothelial cells favors the release of extracellular vesicles (apoptotic bodies and apoptotic exosome-like vesicles) that further enhance autoantibody production, complement deposition, and microvascular rarefaction. In this review, we present the evidence for endothelial injury, its causes and long-term consequences on graft outcomes in the field of kidney transplantation.
Collapse
Affiliation(s)
- Héloïse Cardinal
- Research Centre, Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada.,Canadian National Transplant Research Program, Montreal, QC, Canada.,University of Montreal, Montreal, QC, Canada
| | - Mélanie Dieudé
- Research Centre, Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada.,Canadian National Transplant Research Program, Montreal, QC, Canada
| | - Marie-Josée Hébert
- Research Centre, Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada.,Canadian National Transplant Research Program, Montreal, QC, Canada.,University of Montreal, Montreal, QC, Canada
| |
Collapse
|
32
|
Castellano G, Franzin R, Stasi A, Divella C, Sallustio F, Pontrelli P, Lucarelli G, Battaglia M, Staffieri F, Crovace A, Stallone G, Seelen M, Daha MR, Grandaliano G, Gesualdo L. Complement Activation During Ischemia/Reperfusion Injury Induces Pericyte-to-Myofibroblast Transdifferentiation Regulating Peritubular Capillary Lumen Reduction Through pERK Signaling. Front Immunol 2018; 9:1002. [PMID: 29875766 PMCID: PMC5974049 DOI: 10.3389/fimmu.2018.01002] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 04/23/2018] [Indexed: 12/21/2022] Open
Abstract
Pericytes are one of the principal sources of scar-forming myofibroblasts in chronic kidneys disease. However, the modulation of pericyte-to-myofibroblast transdifferentiation (PMT) in the early phases of acute kidney injury is poorly understood. Here, we investigated the role of complement in inducing PMT after transplantation. Using a swine model of renal ischemia/reperfusion (I/R) injury, we found the occurrence of PMT after 24 h of I/R injury as demonstrated by reduction of PDGFRβ+/NG2+ cells with increase in myofibroblasts marker αSMA. In addition, PMT was associated with significant reduction in peritubular capillary luminal diameter. Treatment by C1-inhibitor (C1-INH) significantly preserved the phenotype of pericytes maintaining microvascular density and capillary lumen area at tubulointerstitial level. In vitro, C5a transdifferentiated human pericytes in myofibroblasts, with increased αSMA expression in stress fibers, collagen I production, and decreased antifibrotic protein Id2. The C5a-induced PMT was driven by extracellular signal-regulated kinases phosphorylation leading to increase in collagen I release that required both non-canonical and canonical TGFβ pathways. These results showed that pericytes are a pivotal target of complement activation leading to a profibrotic maladaptive cellular response. Our studies suggest that C1-INH may be a potential therapeutic strategy to counteract the development of PMT and capillary lumen reduction in I/R injury.
Collapse
Affiliation(s)
- Giuseppe Castellano
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
| | - Rossana Franzin
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
| | - Alessandra Stasi
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
| | - Chiara Divella
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
| | - Fabio Sallustio
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy.,Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari Aldo Moro, Bari, Italy
| | - Paola Pontrelli
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
| | - Giuseppe Lucarelli
- Urology, Andrology and Renal Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
| | - Michele Battaglia
- Urology, Andrology and Renal Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
| | - Francesco Staffieri
- Veterinary Surgery Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
| | - Antonio Crovace
- Veterinary Surgery Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
| | - Giovanni Stallone
- Nephrology, Dialysis and Transplantation Unit, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Marc Seelen
- Division of Nephrology, Department of Internal Medicine, University of Groningen, University Medical Centre Groningen, Groningen, Netherlands
| | - Mohamed R Daha
- Division of Nephrology, Department of Internal Medicine, University of Groningen, University Medical Centre Groningen, Groningen, Netherlands.,Department of Nephrology, Leiden University Medical Centre, Leiden, Netherlands
| | - Giuseppe Grandaliano
- Nephrology, Dialysis and Transplantation Unit, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Loreto Gesualdo
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
| |
Collapse
|
33
|
Changes in Renal Peritubular Capillaries in Canine and Feline Chronic Kidney Disease. J Comp Pathol 2018; 160:79-83. [DOI: 10.1016/j.jcpa.2018.03.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 03/19/2018] [Accepted: 03/24/2018] [Indexed: 02/01/2023]
|
34
|
Zhao H, Alam A, Soo AP, George AJT, Ma D. Ischemia-Reperfusion Injury Reduces Long Term Renal Graft Survival: Mechanism and Beyond. EBioMedicine 2018; 28:31-42. [PMID: 29398595 PMCID: PMC5835570 DOI: 10.1016/j.ebiom.2018.01.025] [Citation(s) in RCA: 214] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 01/18/2018] [Accepted: 01/20/2018] [Indexed: 01/10/2023] Open
Abstract
Ischemia-reperfusion injury (IRI) during renal transplantation often initiates non-specific inflammatory responses that can result in the loss of kidney graft viability. However, the long-term consequence of IRI on renal grafts survival is uncertain. Here we review clinical evidence and laboratory studies, and elucidate the association between early IRI and later graft loss. Our critical analysis of previous publications indicates that early IRI does contribute to later graft loss through reduction of renal functional mass, graft vascular injury, and chronic hypoxia, as well as subsequent fibrosis. IRI is also known to induce kidney allograft dysfunction and acute rejection, reducing graft survival. Therefore, attempts have been made to substitute traditional preserving solutions with novel agents, yielding promising results. Ischaemia reperfusion injury (IRI) potentiates delayed renal graft function and causes reduction in renal graft survival IRI causes innate immune system activation, hypoxic injury, inflammation and graft vascular disease Reducing prolonged cold ischaemic time improves graft survival Novel protective strategies include mesenchymal stem cells, machine perfusion, and ex vivo preservation solution saturated with gas. Further studies are needed to investigate the long-term effects of novel ex vivo preservation agents
Collapse
Affiliation(s)
- Hailin Zhao
- Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea & Westminster Hospital, London, UK
| | - Azeem Alam
- Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea & Westminster Hospital, London, UK
| | - Aurelie Pac Soo
- Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea & Westminster Hospital, London, UK
| | | | - Daqing Ma
- Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea & Westminster Hospital, London, UK.
| |
Collapse
|
35
|
Kumar S. Cellular and molecular pathways of renal repair after acute kidney injury. Kidney Int 2018; 93:27-40. [PMID: 29291820 DOI: 10.1016/j.kint.2017.07.030] [Citation(s) in RCA: 176] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2017] [Revised: 07/23/2017] [Accepted: 07/31/2017] [Indexed: 12/31/2022]
Abstract
The acutely injured mammalian kidney mounts a cellular and molecular response to repair itself. However, in patchy regions such intrinsic processes are impaired and dysregulated leading to chronic kidney disease. Currently, no therapy exists to treat established acute kidney injury per se. Strategies to augment human endogenous repair processes and retard associated profibrotic responses are urgently required. Recent studies have identified injury-induced activation of the intrinsic molecular driver of epithelial regeneration and induction of partial epithelial to the mesenchymal state, respectively. Activation of key developmental transcription factors drive such processes; however, whether these recruit comparable gene regulatory networks with target genes similar to those in nephrogenesis is unclear. Extensive complex molecular cross-talk between the nephron epithelia and immune, interstitial, and endothelial cells regulate renal recovery. In vitro-based M1/M2 macrophage subtypes have been increasingly linked to renal repair; however, the precise contribution of in vivo macrophage plasticity to repair responses is poorly understood. Endothelial cell-pericyte intimacy, balance of the angiocrine/antiangiocrine system, and endothelial cell-regulated inflammatory processes have an impact on renal recovery and fibrosis. Close scrutiny of cellular and molecular pathways in repairing human kidneys is imperative for the identification of promising therapeutic targets and biomarker of human renal repair processes.
Collapse
Affiliation(s)
- Sanjeev Kumar
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA.
| |
Collapse
|
36
|
Gosset C, Viglietti D, Rabant M, Vérine J, Aubert O, Glotz D, Legendre C, Taupin JL, Duong Van-Huyen JP, Loupy A, Lefaucheur C. Circulating donor-specific anti-HLA antibodies are a major factor in premature and accelerated allograft fibrosis. Kidney Int 2017; 92:729-742. [DOI: 10.1016/j.kint.2017.03.033] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Revised: 02/21/2017] [Accepted: 03/16/2017] [Indexed: 11/24/2022]
|
37
|
The intragraft microenvironment as a central determinant of chronic rejection or local immunoregulation/tolerance. Curr Opin Organ Transplant 2016; 22:55-63. [PMID: 27898465 DOI: 10.1097/mot.0000000000000373] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
PURPOSE OF REVIEW Chronic rejection is associated with persistent mononuclear cell recruitment, endothelial activation and proliferation, local tissue hypoxia and related biology that enhance effector immune responses. In contrast, the tumor microenvironment elicits signals/factors that inhibit effector T cell responses and rather promote immunoregulation locally within the tissue itself. The identification of immunoregulatory check points and/or secreted factors that are deficient within allografts is of great importance in the understanding and prevention of chronic rejection. RECENT FINDINGS The relative deficiency of immunomodulatory molecules (cell surface and secreted) on microvascular endothelial cells within the intragraft microenvironment, is of functional importance in shaping the phenotype of rejection. These regulatory molecules include coinhibitory and/or intracellular regulatory signals/factors that enhance local activation of T regulatory cells. For example, semaphorins may interact with endothelial cells and CD4 T cells to promote local tolerance. Additionally, metabolites and electrolytes within the allograft microenvironment may regulate local effector and regulatory cell responses. SUMMARY Multiple factors within allografts shape the microenvironment either towards local immunoregulation or proinflammation. Promoting the expression of intragraft cell surface or secreted molecules that support immunoregulation will be critical for long-term graft survival and/or alloimmune tolerance.
Collapse
|
38
|
Oberhuber R, Riede G, Cardini B, Bernhard D, Messner B, Watschinger K, Steger C, Brandacher G, Pratschke J, Golderer G, Werner ER, Maglione M. Impaired Endothelial Nitric Oxide Synthase Homodimer Formation Triggers Development of Transplant Vasculopathy - Insights from a Murine Aortic Transplantation Model. Sci Rep 2016; 6:37917. [PMID: 27883078 PMCID: PMC5121662 DOI: 10.1038/srep37917] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 11/04/2016] [Indexed: 12/17/2022] Open
Abstract
Transplant vasculopathy (TV) represents a major obstacle to long-term graft survival and correlates with severity of ischemia reperfusion injury (IRI). Donor administration of the nitric oxide synthases (NOS) co-factor tetrahydrobiopterin has been shown to prevent IRI. Herein, we analysed whether tetrahydrobiopterin is also involved in TV development. Using a fully allogeneic mismatched (BALB/c to C57BL/6) murine aortic transplantation model grafts subjected to long cold ischemia time developed severe TV with intimal hyperplasia (α-smooth muscle actin positive cells in the neointima) and endothelial activation (increased P-selectin expression). Donor pretreatment with tetrahydrobiopterin significantly minimised these changes resulting in only marginal TV development. Severe TV observed in the non-treated group was associated with increased protein oxidation and increased occurrence of endothelial NOS monomers in the aortic grafts already during graft procurement. Tetrahydrobiopterin supplementation of the donor prevented all these early oxidative changes in the graft. Non-treated allogeneic grafts without cold ischemia time and syngeneic grafts did not develop any TV. We identified early protein oxidation and impaired endothelial NOS homodimer formation as plausible mechanistic explanation for the crucial role of IRI in triggering TV in transplanted aortic grafts. Therefore, targeting endothelial NOS in the donor represents a promising strategy to minimise TV.
Collapse
Affiliation(s)
- Rupert Oberhuber
- Centre of Operative Medicine, Department of Visceral, Transplant and Thoracic Surgery, Medical University Innsbruck, Innsbruck, Austria
| | - Gregor Riede
- Centre of Operative Medicine, Department of Visceral, Transplant and Thoracic Surgery, Medical University Innsbruck, Innsbruck, Austria
| | - Benno Cardini
- Centre of Operative Medicine, Department of Visceral, Transplant and Thoracic Surgery, Medical University Innsbruck, Innsbruck, Austria
| | - David Bernhard
- Cardiac Surgery Research Laboratory, University Clinic for Cardiac Surgery, Medical University Innsbruck, Innsbruck, Austria
| | - Barbara Messner
- Cardiac Surgery Research Laboratory, Department of Surgery, Vienna Medical University, Austria
| | - Katrin Watschinger
- Division of Biological Chemistry, Biocenter, Medical University Innsbruck, Innsbruck, Austria
| | - Christina Steger
- Institute of Pathology, Academic Teaching Hospital Feldkirch, Feldkirch, Austria
| | - Gerald Brandacher
- Centre of Operative Medicine, Department of Visceral, Transplant and Thoracic Surgery, Medical University Innsbruck, Innsbruck, Austria
- Department of Plastic and Reconstructive Surgery, Vascularized Composite Allotransplantation (VCA) Laboratory, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Johann Pratschke
- Centre of Operative Medicine, Department of Visceral, Transplant and Thoracic Surgery, Medical University Innsbruck, Innsbruck, Austria
- Department of General-, Visceral- and Transplantation Surgery, Charité, Campus Virchow Klinikum, Berlin, Germany
| | - Georg Golderer
- Division of Biological Chemistry, Biocenter, Medical University Innsbruck, Innsbruck, Austria
| | - Ernst R. Werner
- Division of Biological Chemistry, Biocenter, Medical University Innsbruck, Innsbruck, Austria
| | - Manuel Maglione
- Centre of Operative Medicine, Department of Visceral, Transplant and Thoracic Surgery, Medical University Innsbruck, Innsbruck, Austria
| |
Collapse
|
39
|
Bábíčková J, Klinkhammer BM, Buhl EM, Djudjaj S, Hoss M, Heymann F, Tacke F, Floege J, Becker JU, Boor P. Regardless of etiology, progressive renal disease causes ultrastructural and functional alterations of peritubular capillaries. Kidney Int 2016; 91:70-85. [PMID: 27678159 DOI: 10.1016/j.kint.2016.07.038] [Citation(s) in RCA: 124] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 07/15/2016] [Accepted: 07/28/2016] [Indexed: 12/28/2022]
Abstract
Progressive renal diseases are associated with rarefaction of peritubular capillaries, but the ultrastructural and functional alterations of the microvasculature are not well described. To study this, we analyzed different time points during progressive kidney damage and fibrosis in 3 murine models of different disease etiologies. These models were unilateral ureteral obstruction, unilateral ischemia-reperfusion injury, and Col4a3-deficient mice, we analyzed ultrastructural alterations in patient biopsy specimens. Compared with kidneys of healthy mice, we found a significant and progressive reduction of peritubular capillaries in all models analyzed. Ultrastructurally, compared with the kidneys of control mice, focal widening of the subendothelial space and higher numbers of endothelial vacuoles and caveolae were found in fibrotic kidneys. Quantitative analysis showed that peritubular capillary endothelial cells in fibrotic kidneys had significantly and progressively reduced numbers of fenestrations and increased thickness of the cell soma and lamina densa of the capillary basement membrane. Similar ultrastructural changes were also observed in patient's kidney biopsy specimens. Compared with healthy murine kidneys, fibrotic kidneys had significantly increased extravasation of Evans blue dye in all 3 models. The extravasation could be visualized using 2-photon microscopy in real time in living animals and was mainly localized to capillary branching points. Finally, fibrotic kidneys in all models exhibited a significantly greater degree of interstitial deposition of fibrinogen. Thus, peritubular capillaries undergo significant ultrastructural and functional alterations during experimental progressive renal diseases, independent of the underlying injury. Analyses of these alterations could provide read-outs for the evaluation of therapeutic approaches targeting the renal microvasculature.
Collapse
Affiliation(s)
- Janka Bábíčková
- Institute of Pathology, RWTH University of Aachen, Aachen, Germany; Division of Nephrology, RWTH University of Aachen, Aachen, Germany; Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University, Bratislava, Slovakia; Institute for Clinical and Translational Research, Biomedical Research Center SAS, Bratislava, Slovakia
| | | | - Eva M Buhl
- Institute of Pathology, RWTH University of Aachen, Aachen, Germany; Division of Nephrology, RWTH University of Aachen, Aachen, Germany
| | - Sonja Djudjaj
- Institute of Pathology, RWTH University of Aachen, Aachen, Germany
| | - Mareike Hoss
- Institute of Pathology, RWTH University of Aachen, Aachen, Germany; Electron Microscopy Facility, RWTH University of Aachen, Aachen, Germany
| | - Felix Heymann
- Division of Gastroenterology, RWTH University of Aachen, Aachen, Germany
| | - Frank Tacke
- Division of Gastroenterology, RWTH University of Aachen, Aachen, Germany
| | - Jürgen Floege
- Division of Nephrology, RWTH University of Aachen, Aachen, Germany
| | - Jan U Becker
- Institute of Pathology, University Hospital Cologne, Cologne, Germany
| | - Peter Boor
- Institute of Pathology, RWTH University of Aachen, Aachen, Germany; Division of Nephrology, RWTH University of Aachen, Aachen, Germany.
| |
Collapse
|
40
|
Peritubular Capillary Basement Membrane Multilayering in Renal Allograft Biopsies of Patients With De Novo Donor-Specific Antibodies. Transplantation 2016; 100:889-97. [PMID: 26413993 DOI: 10.1097/tp.0000000000000908] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Severe peritubular capillary basement membrane multilayering (PTCBML) is part of the Banff definition of chronic antibody-mediated rejection. We retrospectively investigated whether assessment of the mean number of layers of basement membrane (BM) around peritubular capillaries (PTC) can be used in a cohort of patients with de novo donor-specific antibodies (dnDSA) as an early marker to predict long-term antibody-mediated injury. METHODS This is a retrospective cohort study with 151 electron microscopy samples from 54 patients with dnDSA, assessed at around 1 year after transplantation, for a mean number of BM layers around PTC and in serial biopsies. Graft survival and time to transplant glomerulopathy (TG) development were estimated in survival analyses. RESULTS We found that a mean PTCBML count greater than 2.5 layers assessed in a sample of 25 PTCs around 1 year after transplantation is indicative of the development of TG in patients with dnDSA (P = 0.001). In addition, in patients with serial biopsies available for electron microscopy analysis, we could distinguish 2 groups: patients with a mean PTCBML count of 2.5 or less on all biopsies, and patients who developed greater than 2.5 layers at any time after transplantation. The latter group reflected dnDSA patients at risk for TG development (P < 0.001). In patients with dnDSA, PTCBML score added significantly to the sensitivity and specificity of prediction of TG compared with microcirculation injury score alone. CONCLUSIONS Our results highlight the potential value of assessing the mean number of BM in PTC for early prediction of progression to chronic antibody-mediated injury.
Collapse
|
41
|
Xie J, Li X, Meng D, Liang Q, Wang X, Wang L, Wang R, Xiang M, Chen S. Transduction of interleukin-10 through renal artery attenuates vascular neointimal proliferation and infiltration of immune cells in rat renal allograft. Immunol Lett 2016; 176:105-13. [PMID: 27317647 DOI: 10.1016/j.imlet.2016.06.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Revised: 05/24/2016] [Accepted: 06/13/2016] [Indexed: 12/14/2022]
Abstract
Renal transplantation is the treatment of choice for end-stage renal failure. Although acute rejection is not a major issue anymore, chronic rejection, especially vascular rejection, is still a major factor that might lead to allograft dysfunction on the long term. The role of the local immune-regulating cytokine interleukin-10 (IL-10) in chronic renal allograft is unclear. Many clinical observations showed that local IL-10 level was negatively related to kidney allograft function. It is unknown this negative relationship was the result of immunostimulatory property or insufficient immunosuppression property of local IL-10. We performed ex vivo transduction before transplantation through artery of the renal allograft using adeno-associated viral vectors carrying IL-10 gene. Twelve weeks after transplantation, we found intrarenal IL-10 gene transduction significantly inhibited arterial neointimal proliferation, the number of occluded intrarenal artery, interstitial fibrosis, peritubular capillary congestion and glomerular inflammation in renal allografts compared to control allografts receiving PBS or vectors carrying YFP. IL-10 transduction increased serum IL-10 level at 4 weeks but not at 8 and 12 weeks. Renal IL-10 level increased while serum creatinine decreased significantly in IL-10 group at 12 weeks compared to PBS or YFP controls. Immunohistochemical staining showed unchanged total T cells (CD3) and B cells (CD45R/B220), decreased cytotoxic T cells (CD8), macrophages (CD68) and increased CD4+ and FoxP3+ cells in IL-10 group. In summary, intrarenal IL-10 inhibited the allograft rejection while modulated immune response.
Collapse
Affiliation(s)
- Jingxin Xie
- Department of Physiology and Pathophysiology, Fudan University Shanghai Medical College, Shanghai, China
| | - Xueyi Li
- Department of Physiology and Pathophysiology, Fudan University Shanghai Medical College, Shanghai, China
| | - Dan Meng
- Department of Physiology and Pathophysiology, Fudan University Shanghai Medical College, Shanghai, China
| | - Qiujuan Liang
- Department of Physiology and Pathophysiology, Fudan University Shanghai Medical College, Shanghai, China
| | - Xinhong Wang
- Department of Physiology and Pathophysiology, Fudan University Shanghai Medical College, Shanghai, China
| | - Li Wang
- Department of Physiology and Pathophysiology, Fudan University Shanghai Medical College, Shanghai, China
| | - Rui Wang
- Department of Physiology and Pathophysiology, Fudan University Shanghai Medical College, Shanghai, China
| | - Meng Xiang
- Department of Physiology and Pathophysiology, Fudan University Shanghai Medical College, Shanghai, China
| | - Sifeng Chen
- Department of Physiology and Pathophysiology, Fudan University Shanghai Medical College, Shanghai, China.
| |
Collapse
|
42
|
Deng Y, Guo Y, Liu P, Zeng R, Ning Y, Pei G, Li Y, Chen M, Guo S, Li X, Han M, Xu G. Blocking protein phosphatase 2A signaling prevents endothelial-to-mesenchymal transition and renal fibrosis: a peptide-based drug therapy. Sci Rep 2016; 6:19821. [PMID: 26805394 PMCID: PMC4726189 DOI: 10.1038/srep19821] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 12/18/2015] [Indexed: 02/04/2023] Open
Abstract
Endothelial-to-mesenchymal transition (EndMT) contributes to the emergence of fibroblasts and plays a significant role in renal interstitial fibrosis. Protein phosphatase 2A (PP2A) is a major serine/threonine protein phosphatase in eukaryotic cells and regulates many signaling pathways. However, the significance of PP2A in EndMT is poorly understood. In present study, the role of PP2A in EndMT was evaluated. We demonstrated that PP2A activated in endothelial cells (EC) during their EndMT phenotype acquisition and in the mouse model of obstructive nephropathy (i.e., UUO). Inhibition of PP2A activity by its specific inhibitor prevented EC undergoing EndMT. Importantly, PP2A activation was dependent on tyrosine nitration at 127 in the catalytic subunit of PP2A (PP2Ac). Our renal-protective strategy was to block tyrosine127 nitration to inhibit PP2A activation by using a mimic peptide derived from PP2Ac conjugating a cell penetrating peptide (CPP: TAT), termed TAT-Y127WT. Pretreatment withTAT-Y127WT was able to prevent TGF-β1-induced EndMT. Administration of the peptide to UUO mice significantly ameliorated renal EndMT level, with preserved density of peritubular capillaries and reduction in extracellular matrix deposition. Taken together, these results suggest that inhibiting PP2Ac nitration using a mimic peptide is a potential preventive strategy for EndMT in renal fibrosis.
Collapse
Affiliation(s)
- Yuanjun Deng
- Division of Nephrology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Yanyan Guo
- Division of Nephrology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Ping Liu
- Division of Nephrology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Rui Zeng
- Division of Nephrology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Yong Ning
- Division of Nephrology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Guangchang Pei
- Division of Nephrology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Yueqiang Li
- Division of Nephrology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Meixue Chen
- Division of Nephrology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Shuiming Guo
- Division of Nephrology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Xiaoqing Li
- Division of Nephrology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Min Han
- Division of Nephrology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Gang Xu
- Division of Nephrology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| |
Collapse
|
43
|
Bruneau S, Wedel J, Fakhouri F, Nakayama H, Boneschansker L, Irimia D, Daly KP, Briscoe DM. Translational implications of endothelial cell dysfunction in association with chronic allograft rejection. Pediatr Nephrol 2016; 31:41-51. [PMID: 25903640 PMCID: PMC4619184 DOI: 10.1007/s00467-015-3094-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 03/03/2015] [Accepted: 03/12/2015] [Indexed: 12/20/2022]
Abstract
Advances in therapeutics have dramatically improved short-term graft survival, but the incidence of chronic rejection has not changed in the past 20 years. New insights into mechanism are sorely needed at this time and it is hoped that the development of predictive biomarkers will pave the way for the emergence of preventative therapeutics. In this review, we discuss a paradigm suggesting that sequential changes within graft endothelial cells (EC) lead to an intragraft microenvironment that favors the development of chronic rejection. Key initial events include EC injury, activation and uncontrolled leukocyte-induced angiogenesis. We propose that all of these early changes in the microvasculature lead to abnormal blood flow patterns, local tissue hypoxia, and an associated overexpression of HIF-1α-inducible genes, including vascular endothelial growth factor. We also discuss how cell intrinsic regulators of mTOR-mediated signaling within EC are of critical importance in microvascular stability and may thus have a role in the inhibition of chronic rejection. Finally, we discuss recent findings indicating that miRNAs may regulate EC stability, and we review their potential as novel non-invasive biomarkers of allograft rejection. Overall, this review provides insights into molecular events, genes, and signals that promote chronic rejection and their potential as biomarkers that serve to support the future development of interruption therapeutics.
Collapse
Affiliation(s)
- Sarah Bruneau
- Transplant Research Program, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
- Division of Nephrology, Department of Medicine, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- INSERM UMR S-1064, Institut de Transplantation Urologie-Nephrologie (ITUN), Centre Hospitalier Universitaire (CHU) de Nantes, University of Nantes, Nantes, France
| | - Johannes Wedel
- Transplant Research Program, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
- Division of Nephrology, Department of Medicine, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Fadi Fakhouri
- INSERM UMR S-1064, Institut de Transplantation Urologie-Nephrologie (ITUN), Centre Hospitalier Universitaire (CHU) de Nantes, University of Nantes, Nantes, France
| | - Hironao Nakayama
- Transplant Research Program, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
- Vascular Biology Program, Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Leo Boneschansker
- Transplant Research Program, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
- Division of Nephrology, Department of Medicine, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Department of Surgery, BioMEMS Resource Center, Massachusetts General Hospital, Harvard Medical School, Shriners Hospitals for Children, Boston, MA, USA
| | - Daniel Irimia
- Department of Surgery, BioMEMS Resource Center, Massachusetts General Hospital, Harvard Medical School, Shriners Hospitals for Children, Boston, MA, USA
| | - Kevin P Daly
- Transplant Research Program, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
- Division of Nephrology, Department of Medicine, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - David M Briscoe
- Transplant Research Program, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA.
- Division of Nephrology, Department of Medicine, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA.
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
44
|
Lankhorst S, Baelde HJ, Clahsen-van Groningen MC, Smedts FMM, Danser AHJ, van den Meiracker AH. Effect of high salt diet on blood pressure and renal damage during vascular endothelial growth factor inhibition with sunitinib. Nephrol Dial Transplant 2015; 31:914-21. [PMID: 26681729 DOI: 10.1093/ndt/gfv410] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 11/10/2015] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND Antiangiogenic treatment with the multitargeted vascular endothelial growth factor (VEGF) receptor inhibitor sunitinib associates with a blood pressure (BP) rise and glomerular renal injury. Recent evidence indicates that VEGF derived from tubular cells is required for maintenance of the peritubular vasculature. In the present study, we focussed on tubular and glomerular pathology induced by sunitinib and explored whether a high salt (HS) diet augments the BP rise and renal abnormalities. METHODS Normotensive Wistar Kyoto (WKY) rats were exposed to a normal salt (NS) or HS diet for 2 weeks and subsequently for 8 days to sunitinib or vehicle administration after which the rats were euthanized and kidneys excised. Mean arterial pressure (MAP) was telemetrically measured. Urine was sampled for proteinuria and endothelinuria, and blood for measurement of endothelin-1, creatinine and cystatin C. RESULTS Compared with the NS diet, MAP rapidly rose by 27 ± 3 mmHg with the HS diet. On sunitinib, MAP rose further by 15 ± 1 with the NS and by 23 ± 4 mmHg with the HS diet (P < 0.05). The HS diet itself had no effect on proteinuria, endothelinuria or the plasma levels of endothelin-1, creatinine and cystatin C. Only with the HS diet, sunitinib administration massively increased proteinuria and endothelinuria and these two parameters were related (r = 0.50, P < 0.01). Likewise, renal glomerular pathology was enhanced during sunitinib with the HS diet, whereas tubulointerstitial injury or reduced peritubular capillary density did not occur. CONCLUSIONS An HS diet induces a marked BP rise in WKY rats and exacerbates both the magnitude of the BP rise and glomerular injury induced by sunitinib.
Collapse
Affiliation(s)
- Stephanie Lankhorst
- Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Hans J Baelde
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Frank M M Smedts
- Department of Pathology, Reinier de Graaf Gasthuis, Delft, The Netherlands
| | - A H Jan Danser
- Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Anton H van den Meiracker
- Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
45
|
Use of [18F]FDG Positron Emission Tomography to Monitor the Development of Cardiac Allograft Rejection. Transplantation 2015; 99:e132-9. [PMID: 25675207 DOI: 10.1097/tp.0000000000000618] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Positron emission tomography (PET) has the potential to be a specific, sensitive and quantitative diagnostic test for transplant rejection. To test this hypothesis, we evaluated F-labeled fluorodeoxyglucose ([F]FDG) and N-labeled ammonia ([N]NH3) small animal PET imaging in a well-established murine cardiac rejection model. METHODS Heterotopic transplants were performed using minor major histocompatibility complex-mismatched B6.C-H2 donor hearts in C57BL/6(H-2) recipients. C57BL/6 donor hearts into C57BL/6 recipients served as isograft controls. [F]FDG PET imaging was performed weekly between posttransplant days 7 and 42, and the percent injected dose was computed for each graft. [N]NH3 imaging was performed to evaluate myocardial perfusion. RESULTS There was a significant increase in [F]FDG uptake in allografts from day 14 to day 21 (1.6% to 5.2%; P < 0.001) and uptake in allografts was significantly increased on posttransplant days 21 (5.2% vs 0.9%; P = 0.005) and 28 (4.8% vs 0.9%; P = 0.006) compared to isograft controls. Furthermore, [F]FDG uptake correlated with an increase in rejection grade within allografts between days 14 and 28 after transplantation. Finally, the uptake of [N]NH3 was significantly lower relative to the native heart in allografts with chronic vasculopathy compared to isograft controls on day 28 (P = 0.01). CONCLUSIONS PET imaging with [F]FDG can be used after transplantation to monitor the evolution of rejection. Decreased uptake of [N]NH3 in rejecting allografts may be reflective of decreased myocardial blood flow. These data suggest that combined [F]FDG and [N]NH3 PET imaging could be used as a noninvasive, quantitative technique for serial monitoring of allograft rejection and has potential application in human transplant recipients.
Collapse
|
46
|
Boor P, Bábíčková J, Steegh F, Hautvast P, Martin IV, Djudjaj S, Nakagawa T, Ehling J, Gremse F, Bücher E, Eriksson U, van Roeyen CR, Eitner F, Lammers T, Floege J, Peutz-Kootstra CJ, Ostendorf T. Role of Platelet-Derived Growth Factor-CC in Capillary Rarefaction in Renal Fibrosis. THE AMERICAN JOURNAL OF PATHOLOGY 2015. [DOI: 10.1016/j.ajpath.2015.04.022] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
47
|
Boor P, Floege J. Renal allograft fibrosis: biology and therapeutic targets. Am J Transplant 2015; 15:863-86. [PMID: 25691290 DOI: 10.1111/ajt.13180] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Revised: 11/30/2014] [Accepted: 12/19/2014] [Indexed: 01/25/2023]
Abstract
Renal tubulointerstitial fibrosis is the final common pathway of progressive renal diseases. In allografts, it is assessed with tubular atrophy as interstitial fibrosis/tubular atrophy (IF/TA). IF/TA occurs in about 40% of kidney allografts at 3-6 months after transplantation, increasing to 65% at 2 years. The origin of renal fibrosis in the allograft is complex and includes donor-related factors, in particular in case of expanded criteria donors, ischemia-reperfusion injury, immune-mediated damage, recurrence of underlying diseases, hypertensive damage, nephrotoxicity of immunosuppressants, recurrent graft infections, postrenal obstruction, etc. Based largely on studies in the non-transplant setting, there is a large body of literature on the role of different cell types, be it intrinsic to the kidney or bone marrow derived, in mediating renal fibrosis, and the number of mediator systems contributing to fibrotic changes is growing steadily. Here we review the most important cellular processes and mediators involved in the progress of renal fibrosis, with a focus on the allograft situation, and discuss some of the challenges in translating experimental insights into clinical trials, in particular fibrosis biomarkers or imaging modalities.
Collapse
Affiliation(s)
- P Boor
- Division of Nephrology and Clinical Immunology, RWTH University of Aachen, Aachen, Germany; Department of Pathology, RWTH University of Aachen, Aachen, Germany; Institute of Molecular Biomedicine, Bratislava, Slovakia
| | | |
Collapse
|
48
|
Basile DP, Yoder MC. Renal endothelial dysfunction in acute kidney ischemia reperfusion injury. Cardiovasc Hematol Disord Drug Targets 2015; 14:3-14. [PMID: 25088124 DOI: 10.2174/1871529x1401140724093505] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Revised: 05/15/2014] [Accepted: 05/30/2014] [Indexed: 01/11/2023]
Abstract
Acute kidney injury is associated with alterations in vascular tone that contribute to an overall reduction in GFR. Studies in animal models indicate that ischemia triggers alterations in endothelial function that contribute significantly to the overall degree and severity of a kidney injury. Putative mediators of vasoconstriction that may contribute to the initial loss of renal blood flow and GFR are highlighted. In addition, there is discussion of how intrinsic damage to the endothelium impairs homeostatic responses in vascular tone as well as promotes leukocyte adhesion and exacerbating the reduction in renal blood flow. The timing of potential therapies in animal models as they relate to the evolution of AKI, as well as the limitations of such approaches in the clinical setting are discussed. Finally, we discuss how acute kidney injury induces permanent alterations in renal vascular structure. We posit that the cause of the sustained impairment in kidney capillary density results from impaired endothelial growth responses and suggest that this limitation is a primary contributing feature underlying progression of chronic kidney disease.
Collapse
Affiliation(s)
| | - Mervin C Yoder
- Department of Cellular & Integrative Physiology, Indiana University School of Medicine, 635 Barnhill Drive, Med Sci 334, Indianapolis, IN 46202, USA.
| |
Collapse
|
49
|
Abstract
Interstitial fibrosis is a hallmark structural correlate of progressive and chronic kidney disease. There remain many uncertainties about how to best measure interstitial fibrosis both in research settings and in evaluations of renal biopsies performed for management of individual patients. Areas of uncertainty include determination of the composition of the matrix in a fibrotic parenchyma, the definition of how the interstitium is involved by fibrosing injuries, the choice of histologic stains for evaluation of renal fibrosis, and the reproducibility and robustness of measures currently employed by pathologists, both with and without the assistance of computerized imaging and assessments. In this review, we address some of these issues while citing the key studies that illustrate these difficulties. We point to future approaches that may allow a more accurate and meaningful assessment of renal interstitial fibrosis.
Collapse
|
50
|
The Interplay between inflammation and fibrosis in kidney transplantation. BIOMED RESEARCH INTERNATIONAL 2014; 2014:750602. [PMID: 24991565 PMCID: PMC4065724 DOI: 10.1155/2014/750602] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Accepted: 05/11/2014] [Indexed: 12/29/2022]
Abstract
Serial surveillance renal allograft biopsies have shown that early subclinical inflammation constitutes a risk factor for the development of interstitial fibrosis. More recently, it has been observed that persistent inflammation is also associated with fibrosis progression and chronic humoral rejection, two histological conditions associated with poor allograft survival. Treatment of subclinical inflammation with steroid boluses prevents progression of fibrosis and preserves renal function in patients treated with a cyclosporine-based regimen. Subclinical inflammation has been reduced after the introduction of tacrolimus based regimens, and it has been shown that immunosuppressive schedules that are effective in preventing acute rejection and subclinical inflammation may prevent the progression of fibrosis and chronic humoral rejection. On the other hand, minimization protocols are associated with progression of fibrosis, and noncompliance with the immunosuppressive regime constitutes a major risk factor for chronic humoral rejection. Thus, adequate immunosuppressive treatment, avoiding minimization strategies and reinforcing educational actions to prevent noncompliance, is at present an effective approach to combat the progression of fibrosis.
Collapse
|