1
|
Futamura K, Tsujita M, Kosugi T, Ryuge A, Okada M, Hiramitsu T, Narumi S, Takeda A, Watarai Y, Morozumi K, Maruyama S. Urinary Basigin/CD147 is a useful marker of acute T cell-mediated rejection in kidney transplant recipients. Ren Fail 2025; 47:2479574. [PMID: 40125925 PMCID: PMC11934155 DOI: 10.1080/0886022x.2025.2479574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 02/23/2025] [Accepted: 03/06/2025] [Indexed: 03/25/2025] Open
Abstract
BACKGROUND Acute T cell-mediated rejection (ATCMR) is a severe negative outcome of kidney transplantation; however, it currently has no reliable marker in Japan. METHODS This cross-sectional study was conducted at the Japanese Red Cross Aichi Medical Center Nagoya Daini Hospital from 2016 to 2018 to determine whether plasma or urinary Basigin/CD147 is an effective marker of ATCMR. Plasma and urinary samples were obtained when episode graft biopsies were performed. RESULTS Forty-six kidney transplant recipients received graft biopsies. Three of them missed plasma and urinary samples and three in ATCMR were on postrejection treatment. Graft biopsy results revealed ATCMR in 12 of them, calcineurin inhibitor nephrotoxicity (CIN) in nine, chronic active antibody-mediated rejection (CAAMR) in nine, BK nephropathy, recurrence IgA nephropathy, necrotic glomerulonephritis, and infection-related glomerulonephritis in one each, and other complications in six. The urinary Basigin/CD147 levels of patients in the ATCMR group [759.4 (490.0, 843.0)] pg/gCre were significantly higher than the levels of patients in the CAAMR [247.0 (157.1, 288.8)] and CIN groups [379.1 (264.7, 456.7)] pg/gCre (p < 0.001). No statistical difference in plasma Basigin/CD147 levels was observed between those groups. At a urinary Basigin/CD147 of 631.5 µg/gCre, 75% sensitivity and 84% specificity with an area under the curve of 0.80 were attained for the diagnosis of graft rejection. CONCLUSION Urinary Basigin/CD147 may be a potential marker for ATCMR in kidney transplant recipients. Further studies will be needed to clarify the effectiveness of Basigin/CD147.
Collapse
Affiliation(s)
- Kenta Futamura
- Department of Kidney Disease Center, Transplant Nephrology and Surgery, Japanese Red Cross Aichi Medical Center Nagoya Daini Hospital, Nagoya, Japan
| | - Makoto Tsujita
- Department of Kidney Disease Center, Transplant Nephrology and Surgery, Japanese Red Cross Aichi Medical Center Nagoya Daini Hospital, Nagoya, Japan
- Department of Nephrology, Masuko Memorial Hospital, Nagoya, Japan
| | - Tomoki Kosugi
- Department of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Akihiro Ryuge
- Internal Medicine, Ryuge Internal Medicine Kamiotai Clinic, Nagoya, Japan
| | - Manabu Okada
- Department of Kidney Disease Center, Transplant Nephrology and Surgery, Japanese Red Cross Aichi Medical Center Nagoya Daini Hospital, Nagoya, Japan
| | - Takahisa Hiramitsu
- Department of Kidney Disease Center, Transplant Nephrology and Surgery, Japanese Red Cross Aichi Medical Center Nagoya Daini Hospital, Nagoya, Japan
| | - Shunji Narumi
- Department of Kidney Disease Center, Transplant Nephrology and Surgery, Japanese Red Cross Aichi Medical Center Nagoya Daini Hospital, Nagoya, Japan
| | - Asami Takeda
- Department of Kidney Disease Center, Transplant Nephrology and Surgery, Japanese Red Cross Aichi Medical Center Nagoya Daini Hospital, Nagoya, Japan
- Department of Nephrology, Masuko Memorial Hospital, Nagoya, Japan
| | | | - Kunio Morozumi
- Department of Nephrology, Masuko Memorial Hospital, Nagoya, Japan
| | - Shoichi Maruyama
- Department of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
2
|
Azim S, Rousselle T, Zubair H, Shetty AC, Archer KJ, Marshall JN, Rajabi A, Lara CM, Mustofa S, Drachenberg C, Bromberg J, Menon M, Maluf DG, Akalin E, Mas VR. Epithelial-Immune-Stromal Interactions Define Divergent Repair and Fibrosis Pathways After Acute Kidney Injury in Human Renal Transplants. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.30.651080. [PMID: 40364910 PMCID: PMC12073942 DOI: 10.1101/2025.04.30.651080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2025]
Abstract
Acute kidney injury (AKI) is a major cause of early graft dysfunction after kidney transplantation, particularly in recipients of high-risk donor kidneys prone to ischemia-reperfusion injury. However, the cellular mechanisms dictating whether injury resolves or progresses to fibrosis remain unclear. This study combines single-nucleus RNA sequencing and imaging mass cytometry (IMC) analysis of human kidney allograft biopsies collected within eight weeks posttransplant, stratified by long-term functional outcomes. Grafts that recovered function were enriched in regenerative proximal tubular (PT) cells co-expressing PROM1, CD24, and injury markers, consistent with scattered tubular cells (STCs). In contrast, non-recovering grafts contained a unique subpopulation of transitional proximal tubule cells (tPT4) characterized by dedifferentiation, loss of epithelial identity, and acquisition of fibroblast-like features. Fibroblast trajectory analysis revealed a profibrotic lineage, progressing from stromal progenitors to myofibroblasts, exclusive to nonrecovery grafts. Immune profiling showed divergent macrophage (MΦ) polarization, with reparative MΦ2 cells and regulatory dendritic cell (DC)-like signatures in recovering grafts, versus inflammatory MΦ1 and pro-fibrotic DCs in non-recovery. IMC confirmed spatial colocalization of injured tubules, activated fibroblasts, and immune cells in fibrotic regions, validated in an independent cohort. Functional assays demonstrated that ischemic epithelial injury activated monocyte-derived MΦs with mixed inflammatory/reparative profiles and induced fibroblast-related gene expression, while PAX8 knockdown impaired epithelial proliferation and promoted pro-inflammatory signaling. These findings reveal epithelial cell plasticity as a central driver of divergent repair outcomes following renal transplant AKI and highlight epithelial-immune-stromal crosstalk as a therapeutic target to promote recovery and prevent chronic graft injury. One Sentence Summary Single-cell and spatial mapping of human kidney transplants reveal regenerative and fibrotic cell programs across tubular, immune, and stromal compartments that determine whether acute injury resolves or progresses to chronic allograft injury.
Collapse
|
3
|
Liu J, Zeng D, Wang Y, Deng F, Wu S, Deng Z. Identification of druggable targets in acute kidney injury by proteome- and transcriptome-wide Mendelian randomization and bioinformatics analysis. Biol Direct 2025; 20:38. [PMID: 40148878 PMCID: PMC11951703 DOI: 10.1186/s13062-025-00631-0] [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: 01/28/2025] [Accepted: 03/11/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND Acute kidney injury (AKI) remains a critical condition with limited therapeutic options, predominantly managed by renal replacement therapy. The challenge of developing targeted treatments persists. METHODS We integrated genetic data related to druggable proteins and gene expression with AKI genome-wide association study (GWAS) findings. Based on multi-omics Mendelian randomization (MR), we identified the potential causal influence of 5,883 unique proteins and genes on AKI. We also performed using reverse MR and external cohort-based analysis to verify the robustness of this causal relationship. Expression patterns of these targets were examined using bulk transcriptome and single-cell transcriptome data. In addition, drug repurposing analyses were conducted to explore the potential of existing medications. We also constructed a molecular interaction network to explore the interplay between identified targets and known drugs. RESULTS Genetically predicted levels of seven proteins and twelve genes were associated with an increased risk of AKI. Of these, six targets (NCF1, TNFRSF1B, APEH, ACADSB, ADD1, and FAM3B) were prioritized based on robust evidence and validated in independent cohorts. Reverse MR showed a one-way causal relationship of targets. These targets are predominantly expressed in proximal tubular cells, endothelial cells, collecting duct-principal cells, and immune cells within both AKI-affected and normal tissues. Several promising drug repurposing opportunities were identified, such as telmisartan-NCF1, calcitriol-ACADSB, and ethinyl estradiol-ACADSB. The molecular interaction mapping and pathway integration analysis provided further insights, suggesting potential strategies for combinatorial therapies. CONCLUSIONS This extensive investigation identified several promising therapeutic targets for AKI and highlighted opportunities for drug repurposing. These findings offer valuable insights that could shape future research and the development of targeted treatments.
Collapse
Affiliation(s)
- Jiachen Liu
- Department of Urology, The Second Xiangya Hospital at Central South University, Changsha, 410011, Hunan, China
- Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Dianjie Zeng
- Department of Urology, The Second Xiangya Hospital at Central South University, Changsha, 410011, Hunan, China
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yinhuai Wang
- Department of Urology, The Second Xiangya Hospital at Central South University, Changsha, 410011, Hunan, China
| | - Fei Deng
- Department of Urology, The Second Xiangya Hospital at Central South University, Changsha, 410011, Hunan, China
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Shuiqing Wu
- Department of Urology, The Second Xiangya Hospital at Central South University, Changsha, 410011, Hunan, China.
| | - Zebin Deng
- Department of Urology, The Second Xiangya Hospital at Central South University, Changsha, 410011, Hunan, China.
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
4
|
Peng J, Wang S, Pan X, Wu M, Zhan X, Wang D, Zhu G, Wang W, Tang H, An N, Pei J. Identification of ALDH2 as a novel target for the treatment of acute kidney injury in kidney transplantation based on WGCNA and machine learning algorithms and exploration of its potential mechanism of action using animal experiments. Front Immunol 2025; 16:1536800. [PMID: 40103812 PMCID: PMC11913804 DOI: 10.3389/fimmu.2025.1536800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 02/13/2025] [Indexed: 03/20/2025] Open
Abstract
Background Acute kidney injury (AKI) after kidney transplantation is one of the main causes of graft loss and poor patient prognosis, and it is important to explore new targets for treating AKI in kidney transplantation. Methods Based on the kidney transplantation AKI-related dataset GSE30718, the most relevant modular genes for AKI among them were firstly screened using WGCNA and intersected with the DEGs, and the intersected genes were used as candidate genes for kidney transplantation AKI. Second, machine learning algorithms were utilized to identify the key genes among them, and the HPA database was used to explore the expression landscape. Next, we constructed a rat renal IRI model and explored the role of key genes in renal IRI. Finally, we combined ssGSEA enrichment analysis with animal experiments to further validate the potential mechanism of action of key genes. Results In total, we identified 98 of the most relevant modular genes for AKI and 417 DEGs, which intersected to yield a total of 24 AKI candidate genes. Next, we intersected the key genes identified by three types of machine learning, namely, Random Forest, LASSO regression analysis and SVM, and obtained a total of 1 intersected gene as ALDH2, which we used as a key gene in kidney transplantation AKI. Using the HPA database, we found that ALDH2 has a high expression level in renal tissues and is mainly located in renal tubular epithelial cells. Next, we found in a rat renal IRI model that increasing the expression of ALDH2 alleviated the impairment of renal function and decreased the expression of NGAL, a marker of tubular injury, and BAX, an apoptotic protein, as well as reducing the expression of the inflammatory factors IL1β and IL6. Finally, using ssGSEA enrichment analysis and animal experiments, we further found that ALDH2 was able to inhibit the activation of the MAPK signaling pathway. Conclusion ALDH2 may serve as a novel target for the treatment of kidney transplantation AKI, and increasing the expression level of ALDH2 has a protective effect on renal IRI, and this protective effect may be achieved by inhibiting the MAPK signaling pathway.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Nini An
- Department of Pediatric Surgrey, Guizhou Provincial People’s Hospital, Guiyang, China
| | - Jun Pei
- Department of Pediatric Surgrey, Guizhou Provincial People’s Hospital, Guiyang, China
| |
Collapse
|
5
|
Broecker V, Toulza F, Brännström M, Ernst A, Roufosse C, Carbonnel M, Alkattan Z, Mölne J. Transcript analysis of uterus transplant cervical biopsies using the Banff Human Organ Transplant panel. Am J Transplant 2025; 25:329-342. [PMID: 39216690 DOI: 10.1016/j.ajt.2024.08.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 08/10/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
Uterus transplantation is being more widely implemented in clinical practice. Monitoring of rejection is routinely done for cervical biopsies and is dependent on histopathological assessment, as rejections are clinically silent and nonhistological biomarkers are missing. Until this gap is filled, it is important to corroborate the histopathological diagnosis of rejection through independent methods such as gene expression analysis. In this study, we compared our previously published scoring system for grading rejection in uterus transplant cervical biopsies to the gene expression profile in the same biopsy. For this, we used the Banff Human Organ Transplant gene panel to analyze the expression of 788 genes in 75 paraffin-embedded transplant cervical biopsies with a spectrum of histologic findings, as well as in 24 cervical biopsies from healthy controls. We found that gene expression in borderline changes did not differ from normal transplants, whereas the genes with increased expression in mild rejections overlapped with previously published rejection-associated transcripts. Moderate/severe rejection samples showed a gene expression pattern characterized by a mixture of rejection-associated and tissue injury-associated genes and a decrease in epithelial transcripts. In summary, our findings support our proposed scoring system for rejection but argue against the treatment of borderline changes.
Collapse
Affiliation(s)
- Verena Broecker
- Department of Clinical Pathology, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden; Department of Obstetrics and Gynecology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Sweden.
| | - Frederic Toulza
- Department of Immunology and Inflammation, Imperial College, Centre for Inflammatory Disease, London, United Kingdom
| | - Mats Brännström
- Department of Obstetrics and Gynecology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Angela Ernst
- Institute of Medical Statistics and Computational Biology, University Hospital of Cologne, Germany; Institute for AI and Informatics in Medicine, Technical University of Munich, Germany
| | - Candice Roufosse
- Department of Immunology and Inflammation, Imperial College, Centre for Inflammatory Disease, London, United Kingdom
| | - Marie Carbonnel
- Department of Obstetrics and Gynecology, Foch Hospital, University of Versailles-Saint-Quentin-en-Yvelines Paris Saclay, Suresnes, France
| | - Zeinab Alkattan
- Department of Obstetrics and Gynecology, Halland Hospital, Varberg, Region Halland, Sweden
| | - Johan Mölne
- Department of Clinical Pathology, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden; Institute of Biomedicine, Department of Laboratory Medicine, Sahlgrenska Academy, University of Gothenburg, Sweden
| |
Collapse
|
6
|
Halloran PF, Madill-Thomsen KS, Böhmig G, Bromberg J, Budde K, Barner M, Mackova M, Chang J, Einecke G, Eskandary F, Gupta G, Myślak M, Viklicky O, Akalin E, Alhamad T, Anand S, Arnol M, Baliga R, Banasik M, Bingaman A, Blosser CD, Brennan D, Chamienia A, Chow K, Ciszek M, de Freitas D, Dęborska-Materkowska D, Debska-Ślizień A, Djamali A, Domański L, Durlik M, Fatica R, Francis I, Fryc J, Gill J, Gill J, Glyda M, Gourishankar S, Grenda R, Gryczman M, Hruba P, Hughes P, Jittirat A, Jurekovic Z, Kamal L, Kamel M, Kant S, Kasiske B, Kojc N, Konopa J, Lan J, Mannon R, Matas A, Mazurkiewicz J, Miglinas M, Müller T, Narins S, Naumnik B, Patel A, Perkowska-Ptasińska A, Picton M, Piecha G, Poggio E, Bloudíčkova SR, Samaniego-Picota M, Schachtner T, Shin S, Shojai S, Sikosana MLN, Slatinská J, Smykal-Jankowiak K, Solanki A, Veceric Haler Ž, Vucur K, Weir MR, Wiecek A, Włodarczyk Z, Yang H, Zaky Z. Subthreshold rejection activity in many kidney transplants currently classified as having no rejection. Am J Transplant 2025; 25:72-87. [PMID: 39117038 DOI: 10.1016/j.ajt.2024.07.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 06/19/2024] [Accepted: 07/31/2024] [Indexed: 08/10/2024]
Abstract
Most kidney transplant patients who undergo biopsies are classified as having no rejection based on consensus thresholds. However, we hypothesized that because these patients have normal adaptive immune systems, T cell-mediated rejection (TCMR) and antibody-mediated rejection (ABMR) may exist as subthreshold activity in some transplants currently classified as no rejection. To examine this question, we studied genome-wide microarray results from 5086 kidney transplant biopsies (from 4170 patients). An updated molecular archetypal analysis designated 56% of biopsies as no rejection. Subthreshold molecular TCMR and/or ABMR activity molecular activity was detectable as elevated classifier scores in many biopsies classified as no rejection, with ABMR activity in many TCMR biopsies and TCMR activity in many ABMR biopsies. In biopsies classified as no rejection histologically and molecularly, molecular TCMR classifier scores correlated with increases in histologic TCMR features and molecular injury, lower estimated glomerular filtration rate, and higher risk of graft loss, and molecular ABMR activity correlated with increased glomerulitis and donor-specific antibody. No rejection biopsies with high subthreshold TCMR or ABMR activity had a higher probability of having TCMR or ABMR, respectively, diagnosed in a future biopsy. We conclude that many kidney transplant recipients have unrecognized subthreshold TCMR or ABMR activity, with significant implications for future problems.
Collapse
Affiliation(s)
- Philip F Halloran
- Department of Medicine, Division of Nephrology & Transplantation Immunology, University of Alberta, Canada
| | | | - Georg Böhmig
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Austria
| | | | - Klemens Budde
- Department of Nephrology, Charite-Medical University of Berlin, Germany
| | | | | | | | - Gunilla Einecke
- Department of Nephrology, Medical University of Hannover, Germany
| | - Farsad Eskandary
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Austria
| | - Gaurav Gupta
- Department of Internal Medicine, Division of Nephrology, Virginia Commonwealth University, USA
| | - Marek Myślak
- Department of Clinical Interventions, Department of Nephrology and Kidney Transplantation SPWSZ Hospital, Pomeranian Medical University, Poland
| | - Ondrej Viklicky
- Department of Nephrology and Transplant Center, Institute for Experimental and Clinical Medicine, Czech Republic
| | - Enver Akalin
- Albert Einstein College of Medicine, Montefiore Medical Center, USA
| | - Tarek Alhamad
- Division of Nephrology, Washington University at St. Louis, USA
| | | | - Miha Arnol
- Department of Nephrology, University of Ljubljana, Slovenia
| | | | - Mirosław Banasik
- Department of Nephrology and Transplantation Medicine, Medical University of Wrocław, Poland
| | - Adam Bingaman
- Department of Surgery, Methodist Transplant and Specialty Hospital, USA
| | | | - Daniel Brennan
- Department of Medicine, Johns Hopkins University School of Medicine, USA
| | - Andrzej Chamienia
- Department of Nephrology, Transplantology and Internal Diseases, Medical University of Gdańsk, Poland
| | - Kevin Chow
- Department of Nephrology, The Royal Melbourne Hospital, Australia
| | - Michał Ciszek
- Department of Immunology, Transplantology and Internal Diseases, Warsaw Medical University, Poland
| | - Declan de Freitas
- Department of Renal Research, Manchester Royal Infirmary, United Kingdom
| | | | - Alicja Debska-Ślizień
- Department of Nephrology, Transplantology and Internal Medicine, Medical University of Gdańsk, Poland
| | | | - Leszek Domański
- Department of Nephrology, Transplantology and Internal Medicine, Pomeranian Medical University, Poland
| | - Magdalena Durlik
- Department of Transplantology, Immunology, Nephrology and Internal Diseases, Warsaw Medical University, Poland
| | - Richard Fatica
- Department of Kidney Medicine, Cleveland Clinic Foundation, USA
| | | | - Justyna Fryc
- 1st Department of Nephrology and Transplantation With Dialysis Unit, Medical University in Bialystok, Poland
| | | | | | | | - Sita Gourishankar
- Department of Medicine, Division of Nephrology & Transplantation Immunology, University of Alberta, Canada
| | - Ryszard Grenda
- Department of Nephrology, Kidney Transplantation and Hypertension, The Children's Memorial Health Institute, Poland
| | - Marta Gryczman
- Department of Nephrology and Kidney Transplantation, Pomeranian Medical University, Poland
| | - Petra Hruba
- Department of Nephrology, Institute for Experimental and Clinical Medicine, Czech Republic
| | - Peter Hughes
- Department of Nephrology, The Royal Melbourne Hospital, Australia
| | | | - Zeljka Jurekovic
- Renal Replacement Therapy, Department of Nephrology, University Hospital Merkur, Croatia
| | - Layla Kamal
- Division of Nephrology, Department of Medicine, Virginia Commonwealth University, USA
| | | | - Sam Kant
- Division of Nephrology & Comprehensive Transplant Center, Department of Medicine, Johns Hopkins University School of Medicine, USA
| | | | - Nika Kojc
- Department of Pathology, University of Ljubljana, Slovenia
| | - Joanna Konopa
- Department of Nephrology, Transplantology and Internal Diseases, Medical University of Gdańsk, Poland
| | | | - Roslyn Mannon
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, USA
| | - Arthur Matas
- Department of Surgery, Division of Transplantation, University on Minnesota, USA
| | | | - Marius Miglinas
- Nephrology and Kidney Transplantation Unit, Nephrology Center, Vilnius University Hospital Santaros Klinikos, Lithuania
| | - Thomas Müller
- Nephrology Department, University Hospital Zurich, Switzerland
| | | | - Beata Naumnik
- 1st Department of Nephrology and Transplantation With Dialysis Unit, Medical University in Bialystok, Poland
| | | | | | - Michael Picton
- Department of Renal Medicine, Manchester Royal Infirmary, United Kingdom
| | - Grzegorz Piecha
- Department of Nephrology, Transplantation and Internal Medicine, Silesian Medical University, Poland
| | - Emilio Poggio
- Department of Kidney Medicine, Glickman Urological and Kidney Institute, Cleveland Clinic Foundation, USA
| | | | | | - Thomas Schachtner
- Department of Surgery and Transplantation, University Hospital Zurich, Switzerland
| | - Sung Shin
- Department of Laboratory Medicine, University of Ulsan College of Medicine/Assan Medical Center, South Korea
| | - Soroush Shojai
- Division of Nephrology, Department of Medicine, University of Alberta, USA
| | - Majid L N Sikosana
- Department of Medicine, Division of Nephrology & Transplantation Immunology, University of Alberta, Canada
| | - Janka Slatinská
- Department of Nephrology, Institute for Experimental and Clinical Medicine, Czech Republic
| | | | | | | | - Ksenija Vucur
- Department of Nephrology, University Hospital Merkur, Croatia
| | - Matthew R Weir
- Department of Medicine, Division of Nephrology, University of Maryland, USA
| | - Andrzej Wiecek
- Department of Nephrology, Transplantation and Internal Medicine, Silesian Medical University, Poland
| | | | - Harold Yang
- Department of Surgery, PinnacleHealth Transplant Associates, USA
| | | |
Collapse
|
7
|
Kumar D, Raju N, Tanriover B, Azzouz L, Moinuddin I, Philogene M, Kamal L, McDougan F, Massey HD, Muthusamy S, Lee I, Halloran P, Gupta G. Tissue-based Gene Expression Diagnosis of Mild and Moderate T-cell-mediated Rejection to Guide Therapy in Kidney Transplants. Transplantation 2024:00007890-990000000-00962. [PMID: 39710875 DOI: 10.1097/tp.0000000000005296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
BACKGROUND Mild histologic lesions of tubulo-interstitial inflammation could represent a "response-to-wounding" rather than allorecognition. Tissue gene expression may complement histopathology for T-cell-mediated rejection (TCMR) diagnostics. METHODS We report on the incorporation of tissue gene expression testing using a Molecular Microscope Diagnostic System into the management of kidney transplant biopsies with suspected TCMR. Patients (N = 209) were divided into 3 groups based upon diagnosis and TCMR therapy (with high-dose steroids and/or anti-thymocyte globulin): Group 1: Untreated histologic TCMR with molecular quiescence (H+M-); Group 2: Treated histologic and molecular TCMR (H+M+); and Group 3: Controls, with no histologic or molecular (H-M-) rejection. RESULTS At biopsy, estimated glomerular filtration rate was worse (P = 0.006) in H+M+ (N = 35; 33 ± 22 mL/min/1.73 m2) and H+M- (N = 30; 40 ± 18 mL/min/1.73 m2) groups; compared with H-M- (N = 144; 47 ± 24 mL/min/1.73 m2) group. In H+M- biopsies, mean molecular acute kidney injury scores (0.33 versus 0.10; P = 0.03) were higher than in H-M-; while molecular TCMR was lower compared with H+M+ (0.04 versus 0.54; P < 0.001). At 12 m postbiopsy estimated glomerular filtration rate remained low (P < 0.001) in H+M+ (38 ± 22 mL/min/1.73 m2) but improved in untreated H+M- (44 ± 22 mL/min/1.73 m2) and H-M- (50 ± 23 mL/min/1.73 m2) groups. At a mean follow-up of 2.1 ± 1.2 y post-index biopsy, death-censored graft survival was lower in H+M+ (74%) than in H+M- (90%) and H-M- (92%; P = 0.001). H+M+ cases had numerically higher rejection on follow-up biopsy (20%) than H+M- (7%) (P = 0.12) and de novo donor-specific antibody formation (H+M+ 24%; H+M- 10%; P = 0.13). CONCLUSIONS In this large single-center study, biopsies with untreated histological TCMR and molecular quiescence had comparable clinical outcomes to cases with no rejection, whereas those with histologic and tissue gene expression confirmed TCMR had inferior outcomes.
Collapse
Affiliation(s)
- Dhiren Kumar
- Division of Nephrology, Virginia Commonwealth University, Richmond, VA
| | - Nihar Raju
- Division of Nephrology, Virginia Commonwealth University, Richmond, VA
| | | | - Louiza Azzouz
- Division of Nephrology, Virginia Commonwealth University, Richmond, VA
| | - Irfan Moinuddin
- Division of Nephrology, Virginia Commonwealth University, Richmond, VA
| | - Mary Philogene
- Division of Nephrology, Virginia Commonwealth University, Richmond, VA
| | - Layla Kamal
- Division of Nephrology, Virginia Commonwealth University, Richmond, VA
| | - Felecia McDougan
- Division of Nephrology, Virginia Commonwealth University, Richmond, VA
| | - Hugh Davis Massey
- Division of Nephrology, Virginia Commonwealth University, Richmond, VA
| | | | - Inkoo Lee
- Division of Nephrology, Virginia Commonwealth University, Richmond, VA
| | | | - Gaurav Gupta
- Division of Nephrology, Virginia Commonwealth University, Richmond, VA
| |
Collapse
|
8
|
Su X, Bai M, Shang Y, Du Y, Xu S, Lin X, Xiao Y, Zhang Y, Chen H, Zhang A. Slc25a21 in cisplatin-induced acute kidney injury: a new target for renal tubular epithelial protection by regulating mitochondrial metabolic homeostasis. Cell Death Dis 2024; 15:891. [PMID: 39695098 DOI: 10.1038/s41419-024-07231-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 10/31/2024] [Accepted: 11/06/2024] [Indexed: 12/20/2024]
Abstract
Acute kidney injury (AKI) is a significant global health issue, which is often caused by cisplatin therapy and characterized by mitochondrial dysfunction. Restoring mitochondrial homeostasis in tubular cells could exert therapeutic effects. Here, we investigated Slc25a21, a mitochondrial carrier, as a potential target for AKI intervention. Renal Slc25a21 expression is negatively associated with kidney function in both AKI patients and cisplatin-induced murine models. Sustaining renal expression of Slc25a21 slowed down AKI progression by reducing cellular apoptosis, necroptosis, and the inflammatory response, likely through its regulation of 2-oxoadipate conversion. Slc25a21 is highly expressed in proximal tubular epithelial cells, and its down-regulation contributes to compromised mitochondrial biogenesis and integrity, as well as impaired oxidative phosphorylation. Mechanistically, reduced Slc25a21 in AKI disrupts mitochondrial 2-oxoadipate transport, affecting related metabolites influx and the tricarboxylic acid cycle. These findings demonstrate a previously unappreciated metabolic function of Slc25a21 in tubular cells, and suggest that targeting mitochondrial metabolic homeostasis by sustaining Slc25a21 expression could be a potential novel therapeutic strategy for AKI.
Collapse
Affiliation(s)
- Xin Su
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Guangzhou Road 72, Nanjing, 210008, China.
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China.
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, 210029, China.
| | - Mi Bai
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Guangzhou Road 72, Nanjing, 210008, China
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, 210029, China
| | - Yaqiong Shang
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Guangzhou Road 72, Nanjing, 210008, China
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, 210029, China
| | - Yang Du
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Guangzhou Road 72, Nanjing, 210008, China
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, 210029, China
| | - Shuang Xu
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Guangzhou Road 72, Nanjing, 210008, China
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, 210029, China
| | - Xiuli Lin
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Guangzhou Road 72, Nanjing, 210008, China
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, 210029, China
| | - Yunzhi Xiao
- Centre for Computational Biology and Programme in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, 8 College Road, 169857, Singapore, Singapore
| | - Yue Zhang
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Guangzhou Road 72, Nanjing, 210008, China
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, 210029, China
| | - Huimei Chen
- Centre for Computational Biology and Programme in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, 8 College Road, 169857, Singapore, Singapore.
| | - Aihua Zhang
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Guangzhou Road 72, Nanjing, 210008, China.
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China.
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, 210029, China.
| |
Collapse
|
9
|
Liu X, Li X, Li H, Guan B, Jiang Y, Zheng C, Kong D. Annexin A1: a key regulator of T cell function and bone marrow adiposity in aplastic anaemia. J Physiol 2024; 602:6125-6152. [PMID: 39373986 DOI: 10.1113/jp286148] [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: 12/15/2023] [Accepted: 07/31/2024] [Indexed: 10/08/2024] Open
Abstract
This study investigates the role of Annexin A1 (ANXA1) in regulating T cell function and its implications in bone marrow adiposity in aplastic anaemia (AA). Utilizing single-cell sequencing analysis, we compared bone marrow tissues from AA patients and healthy individuals, focusing on T cell subgroups and their impact on bone marrow pathology. Our findings reveal a significant activation of CD8+ T cells in AA, driven by reduced ANXA1 expression. This heightened T cell activity promotes adipogenesis in bone marrow-derived mesenchymal stem cells via IFN-γ secretion. Overexpression of ANXA1 was found to suppress this process, suggesting its therapeutic potential in AA treatment. The study highlights ANXA1 as a crucial regulator in the AA-associated immune microenvironment and bone marrow adiposity. KEY POINTS: This study found that ANXA1 is significantly downregulated in AA and provides detailed insights into its critical role in the disease. The study demonstrates the excessive activation of CD8+ T cells in the progression of AA. The research shows that the overexpression of ANXA1 can effectively inhibit the activation of CD8+ T cells. The study confirms that overexpression of ANXA1 reduces the secretion of the cytokine IFN-γ, decreases adipogenesis in bone marrow-derived mesenchymal stem cells and may improve AA symptoms. This research provides new molecular targets for the treatment of AA.
Collapse
Affiliation(s)
- Xia Liu
- Department of Respiratory Intervention, Children's Hospital Affiliated to Shandong University, Jinan, China
| | - Xiaomei Li
- Tumor Research and Therapy Center, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- JiNan Key Laboratory of Basic and Clinical Translational Research in Radiobiology, Jinan, China
| | - Hui Li
- Department of Hematology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Bingxin Guan
- Department of Pathology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yang Jiang
- Institute of Biotherapy for Hematological Malignancies, Shandong University, Jinan, China
- Shandong University-Karolinska Institute Collaborative Laboratory for Stem Cell Research, Shandong University, Jinan, China
| | - Chengyun Zheng
- Department of Hematology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Institute of Biotherapy for Hematological Malignancies, Shandong University, Jinan, China
- Shandong University-Karolinska Institute Collaborative Laboratory for Stem Cell Research, Shandong University, Jinan, China
| | - Dexiao Kong
- Department of Hematology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Institute of Biotherapy for Hematological Malignancies, Shandong University, Jinan, China
- Shandong University-Karolinska Institute Collaborative Laboratory for Stem Cell Research, Shandong University, Jinan, China
| |
Collapse
|
10
|
Deng Z, Dong Z, Wang Y, Dai Y, Liu J, Deng F. Identification of TACSTD2 as novel therapeutic targets for cisplatin-induced acute kidney injury by multi-omics data integration. Hum Genet 2024; 143:1061-1080. [PMID: 38369676 DOI: 10.1007/s00439-024-02641-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Accepted: 01/11/2024] [Indexed: 02/20/2024]
Abstract
Cisplatin-induced acute kidney injury (CP-AKI) is a common complication in cancer patients. Although ferroptosis is believed to contribute to the progression of CP-AKI, its mechanisms remain incompletely understood. In this study, after initially processed individual omics datasets, we integrated multi-omics data to construct a ferroptosis network in the kidney, resulting in the identification of the key driver TACSTD2. In vitro and in vivo results showed that TACSTD2 was notably upregulated in cisplatin-treated kidneys and BUMPT cells. Overexpression of TACSTD2 accelerated ferroptosis, while its gene disruption decelerated ferroptosis, likely mediated by its potential downstream targets HMGB1, IRF6, and LCN2. Drug prediction and molecular docking were further used to propose that drugs targeting TACSTD2 may have therapeutic potential in CP-AKI, such as parthenolide, progesterone, premarin, estradiol and rosiglitazone. Our findings suggest a significant association between ferroptosis and the development of CP-AKI, with TACSTD2 playing a crucial role in modulating ferroptosis, which provides novel perspectives on the pathogenesis and treatment of CP-AKI.
Collapse
Affiliation(s)
- Zebin Deng
- Department of Urology, The Second Xiangya Hospital at Central South University, 139 Middle Renmin Road, Changsha, 410011, Hunan, China
| | - Zheng Dong
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, GA, USA
- Department of Nephrology, The Second Xiangya Hospital at Central South University, Changsha, Hunan, China
| | - Yinhuai Wang
- Department of Urology, The Second Xiangya Hospital at Central South University, 139 Middle Renmin Road, Changsha, 410011, Hunan, China
| | - Yingbo Dai
- Department of Urology, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, Guangdong, China
| | - Jiachen Liu
- Xiangya Hospital, Central South University, Changsha, Hunan, China.
- The Center of Systems Biology and Data Science, Xiangya School of Medicine, Central South University, Changsha, Hunan, People's Republic of China.
| | - Fei Deng
- Department of Urology, The Second Xiangya Hospital at Central South University, 139 Middle Renmin Road, Changsha, 410011, Hunan, China.
- Department of Nephrology, The Second Xiangya Hospital at Central South University, Changsha, Hunan, China.
| |
Collapse
|
11
|
Pei J, Zhang J, Yu C, Luo J, Wen S, Hua Y, Wei G. Transcriptomics-based exploration of shared M1-type macrophage-related biomarker in acute kidney injury after kidney transplantation and acute rejection after kidney transplantation. Transpl Immunol 2024; 85:102066. [PMID: 38815767 DOI: 10.1016/j.trim.2024.102066] [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: 12/23/2023] [Revised: 05/12/2024] [Accepted: 05/27/2024] [Indexed: 06/01/2024]
Abstract
BACKGROUND Macrophage type 1 (M1) cells are associated with both acute kidney injury (AKI) during kidney transplantation and acute rejection (AR) after kidney transplantation. Our study explored M1-related biomarkers involved in both AKI and AR and their potential biological functions. METHODS Based on the Gene Expression Omnibus (GEO) database, the immune cell infiltration levels and differentially expressed genes were examined in AKI and AR in the kidney transplantation; M1-related genes shared in AKI and AR were identified using weighted gene co-expression analysis (WGCNA) system. Subsequently, protein-protein interaction (PPI) networks and machine learning methods to identify Hub genes and construct diagnostic models. Both AKI model and AR rat models were built to validate the expressions of Hub genes and test the injury phenotype, oxidative stress markers, and inflammatory factors. Finally, the transcription factor (TF)-Hub gene and micro-RNA (miRNA)-Hub gene regulatory networks were constructed based on identified Hub genes. RESULTS Out of 2167 differential expression genes (DEGs) in AKI and 2100 DEGs in AR, four M1-related Hub genes were obtained by PPI networks and machine learning methods, namely GBP2, TYROBP, CCR5, and TLR8. The calibration curves in the nomogram diagnostic model for these four Hub genes suggested the same predictive probability as an ideal model for AKI and AR after kidney transplantation (AUC values of the area under the ROC curve were all >0.7). The same observations were confirmed in ischemia reperfusion injury (IRI) and AR rat models by identifying common four Hub genes (GBP2, TYROBP, TLR8, and CCR5). Western blots showed that these four Hub genes were significantly different in rat models of IRI and AR (all p<0.05). Compared with the control group, IRI and AR groups showed aggravated histopathological damage and increased secretion of oxidative stress markers and inflammatory factors in rat kidneys (all p<0.05). Finally, TF-Hub and miRNA-Hub gene regulatory networks were constructed to provide a theoretical basis for the regulation of Hub genes. CONCLUSION We identified four macrophage M1-related Hub genes shared among AKI and AR after kidney transplantation. These genes may be considered for diagnosis of AKI and AR after kidney transplantation.
Collapse
Affiliation(s)
- Jun Pei
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, China; Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, China
| | - Jie Zhang
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, China; Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, China
| | - Chengjun Yu
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, China; Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, China
| | - Jin Luo
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, China; Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, China
| | - Sheng Wen
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, China; Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, China
| | - Yi Hua
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, China; Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, China.
| | - Guanghui Wei
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, China; Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing, China.
| |
Collapse
|
12
|
Reichardt C, Brandt S, Bernhardt A, Krause A, Lindquist JA, Weinert S, Geffers R, Franz T, Kahlfuss S, Dudeck A, Mathew A, Rana R, Isermann B, Mertens PR. DNA-binding protein-A promotes kidney ischemia/reperfusion injury and participates in mitochondrial function. Kidney Int 2024; 106:241-257. [PMID: 38821446 DOI: 10.1016/j.kint.2024.05.009] [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: 12/08/2023] [Revised: 04/23/2024] [Accepted: 05/08/2024] [Indexed: 06/02/2024]
Abstract
DNA-binding protein-A (DbpA; gene: Ybx3) belongs to the cold shock protein family with known functions in cell cycling, transcription, translation, and tight junction communication. In chronic nephritis, DbpA is upregulated. However, its activities in acute injury models, such as kidney ischemia/reperfusion injury (IRI), are unclear. To study this, mice harboring Ybx3+/+, Ybx3+/- or the Ybx3-/- genotype were characterized over 24 months and following experimental kidney IRI. Mitochondrial function, number and integrity were analyzed by mitochondrial stress tests, MitoTracker staining and electron microscopy. Western Blot, immunohistochemistry and flow cytometry were performed to quantify tubular cell damage and immune cell infiltration. DbpA was found to be dispensable for kidney development and tissue homeostasis under healthy conditions. Furthermore, endogenous DbpA protein localizes within mitochondria in primary tubular epithelial cells. Genetic deletion of Ybx3 elevates the mitochondrial membrane potential, lipid uptake and metabolism, oxygen consumption rates and glycolytic activities of tubular epithelial cells. Ybx3-/- mice demonstrated protection from IRI with less immune cell infiltration, endoplasmic reticulum stress and tubular cell damage. A presumed protective mechanism was identified via upregulated antioxidant activities and reduced ferroptosis, when Ybx3 was deleted. Thus, our studies reveal DbpA acts as a mitochondrial protein with profound adverse effects on cell metabolism and highlights a protective effect against IRI when Ybx3 is genetically deleted. Hence, preemptive DbpA targeting in situations with expected IRI, such as kidney transplantation or cardiac surgery, may preserve post-procedure kidney function.
Collapse
Affiliation(s)
- Charlotte Reichardt
- Clinic of Nephrology and Hypertension, Diabetes and Endocrinology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Health Campus Immunology, Infectiology and Inflammation (GCI3), Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Sabine Brandt
- Clinic of Nephrology and Hypertension, Diabetes and Endocrinology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Health Campus Immunology, Infectiology and Inflammation (GCI3), Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Anja Bernhardt
- Clinic of Nephrology and Hypertension, Diabetes and Endocrinology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Health Campus Immunology, Infectiology and Inflammation (GCI3), Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Anna Krause
- Clinic of Nephrology and Hypertension, Diabetes and Endocrinology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Health Campus Immunology, Infectiology and Inflammation (GCI3), Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Jonathan A Lindquist
- Clinic of Nephrology and Hypertension, Diabetes and Endocrinology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Health Campus Immunology, Infectiology and Inflammation (GCI3), Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Sönke Weinert
- Health Campus Immunology, Infectiology and Inflammation (GCI3), Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Clinic of Cardiology and Angiology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Robert Geffers
- Genome Analytics Research Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Tobias Franz
- Health Campus Immunology, Infectiology and Inflammation (GCI3), Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Institute of Molecular and Clinical Immunology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Sascha Kahlfuss
- Health Campus Immunology, Infectiology and Inflammation (GCI3), Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Institute of Molecular and Clinical Immunology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Institute of Medical Microbiology and Hospital Hygiene, Medical Faculty, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Anne Dudeck
- Health Campus Immunology, Infectiology and Inflammation (GCI3), Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Institute of Molecular and Clinical Immunology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Akash Mathew
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Leipzig, Leipzig, Germany
| | - Rajiv Rana
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Leipzig, Leipzig, Germany
| | - Berend Isermann
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Leipzig, Leipzig, Germany
| | - Peter R Mertens
- Clinic of Nephrology and Hypertension, Diabetes and Endocrinology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Health Campus Immunology, Infectiology and Inflammation (GCI3), Otto-von-Guericke University Magdeburg, Magdeburg, Germany.
| |
Collapse
|
13
|
Mengel M, Mannon RB. No Time for Cancel Culture: The Importance of Banff Pathology Criteria and Clinical Outcomes. J Am Soc Nephrol 2024; 35:829-832. [PMID: 38857893 PMCID: PMC11230712 DOI: 10.1681/asn.0000000000000411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2024] Open
Affiliation(s)
- Michael Mengel
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta, Canada
| | - Roslyn B. Mannon
- Division of Nephrology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska
- Medical Service, Nebraska Western Iowa VA Healthcare System, Omaha, Nebraska
| |
Collapse
|
14
|
Hinze C, Lovric S, Halloran PF, Barasch J, Schmidt-Ott KM. Epithelial cell states associated with kidney and allograft injury. Nat Rev Nephrol 2024; 20:447-459. [PMID: 38632381 PMCID: PMC11660082 DOI: 10.1038/s41581-024-00834-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/27/2024] [Indexed: 04/19/2024]
Abstract
The kidney epithelium, with its intricate arrangement of highly specialized cell types, constitutes the functional core of the organ. Loss of kidney epithelium is linked to the loss of functional nephrons and a subsequent decline in kidney function. In kidney transplantation, epithelial injury signatures observed during post-transplantation surveillance are strong predictors of adverse kidney allograft outcomes. However, epithelial injury is currently neither monitored clinically nor addressed therapeutically after kidney transplantation. Several factors can contribute to allograft epithelial injury, including allograft rejection, drug toxicity, recurrent infections and postrenal obstruction. The injury mechanisms that underlie allograft injury overlap partially with those associated with acute kidney injury (AKI) and chronic kidney disease (CKD) in the native kidney. Studies using advanced transcriptomic analyses of single cells from kidney or urine have identified a role for kidney injury-induced epithelial cell states in exacerbating and sustaining damage in AKI and CKD. These epithelial cell states and their associated expression signatures are also observed in transplanted kidney allografts, suggesting that the identification and characterization of transcriptomic epithelial cell states in kidney allografts may have potential clinical implications for diagnosis and therapy.
Collapse
Affiliation(s)
- Christian Hinze
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - Svjetlana Lovric
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - Philip F Halloran
- Alberta Transplant Applied Genomics Centre, Edmonton, Alberta, Canada
- Department of Medicine, Division of Nephrology and Transplant Immunology, University of Alberta, Edmonton, Alberta, Canada
| | - Jonathan Barasch
- Division of Nephrology, Columbia University, New York City, NY, USA
| | - Kai M Schmidt-Ott
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
15
|
Madill-Thomsen K, Halloran P. Precision diagnostics in transplanted organs using microarray-assessed gene expression: concepts and technical methods of the Molecular Microscope® Diagnostic System (MMDx). Clin Sci (Lond) 2024; 138:663-685. [PMID: 38819301 PMCID: PMC11147747 DOI: 10.1042/cs20220530] [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: 02/22/2024] [Revised: 04/26/2024] [Accepted: 05/02/2024] [Indexed: 06/01/2024]
Abstract
There is a major unmet need for improved accuracy and precision in the assessment of transplant rejection and tissue injury. Diagnoses relying on histologic and visual assessments demonstrate significant variation between expert observers (as represented by low kappa values) and have limited ability to assess many biological processes that produce little histologic changes, for example, acute injury. Consensus rules and guidelines for histologic diagnosis are useful but may have errors. Risks of over- or under-treatment can be serious: many therapies for transplant rejection or primary diseases are expensive and carry risk for significant adverse effects. Improved diagnostic methods could alleviate healthcare costs by reducing treatment errors, increase treatment efficacy, and serve as useful endpoints for clinical trials of new agents that can improve outcomes. Molecular diagnostic assessments using microarrays combined with machine learning algorithms for interpretation have shown promise for increasing diagnostic precision via probabilistic assessments, recalibrating standard of care diagnostic methods, clarifying ambiguous cases, and identifying potentially missed cases of rejection. This review describes the development and application of the Molecular Microscope® Diagnostic System (MMDx), and discusses the history and reasoning behind many common methods, statistical practices, and computational decisions employed to ensure that MMDx scores are as accurate and precise as possible. MMDx provides insights on disease processes and highly reproducible results from a comparatively small amount of tissue and constitutes a general approach that is useful in many areas of medicine, including kidney, heart, lung, and liver transplants, with the possibility of extrapolating lessons for understanding native organ disease states.
Collapse
Affiliation(s)
- Katelynn S. Madill-Thomsen
- Department of Medicine, University of Alberta, Edmonton, AB, Canada
- Alberta Transplant Applied Genomics Center, University of Alberta, Edmonton, AB, Canada
| | - Philip F. Halloran
- Department of Medicine, University of Alberta, Edmonton, AB, Canada
- Alberta Transplant Applied Genomics Center, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
16
|
Yaffe HC, von Ahrens D, Urioste A, Mas VR, Akalin E. Impact of Deceased-donor Acute Kidney Injury on Kidney Transplantation. Transplantation 2024; 108:1283-1295. [PMID: 37990359 DOI: 10.1097/tp.0000000000004848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2023]
Abstract
Even as record numbers of deceased donors are undergoing organ recovery, the global transplant community continues to struggle with a shortage of donor organs and a high organ discard rate. Acute kidney injury (AKI) occurs in many hospitalized patients, including up to 25% of patients in critical condition. Registry studies have shown a significant increase in nonrecovery or organ discard rates in AKI donors, despite most studies reporting similar clinical outcomes compared with non-AKI donors. This review aims to capture the salient information learned from these studies and to summarize the efforts that have been made to gain a more granular understanding of how kidneys from donors with AKI behave posttransplant. In particular, we reviewed the studies that analyzed the clinical outcomes in different stages of AKI and AKI in marginal donors, such as kidney donor profile index of >85%, older donors, and donation after circulatory death donors. We summarized studies investigating molecular biomarkers, transcriptomics, and possible future therapeutic targets for postdonation AKI.
Collapse
Affiliation(s)
- Hillary C Yaffe
- Montefiore Einstein Center for Transplantation, Department of Surgery, Montefiore Medical Center, Bronx, NY
- Albert Einstein College of Medicine, Bronx, NY
| | - Dagny von Ahrens
- Montefiore Einstein Center for Transplantation, Department of Surgery, Montefiore Medical Center, Bronx, NY
- Albert Einstein College of Medicine, Bronx, NY
| | - Alejandra Urioste
- Surgical Sciences Division, University of Maryland School of Medicine, Baltimore, MD
| | - Valeria R Mas
- Surgical Sciences Division, University of Maryland School of Medicine, Baltimore, MD
| | - Enver Akalin
- Albert Einstein College of Medicine, Bronx, NY
- Montefiore Einstein Center for Transplantation, Department of Medicine, Montefiore Medical Center, Bronx, NY
| |
Collapse
|
17
|
Urie RR, Morris A, Farris D, Hughes E, Xiao C, Chen J, Lombard E, Feng J, Li JZ, Goldstein DR, Shea LD. Biomarkers from subcutaneous engineered tissues predict acute rejection of organ allografts. SCIENCE ADVANCES 2024; 10:eadk6178. [PMID: 38748794 PMCID: PMC11095459 DOI: 10.1126/sciadv.adk6178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 04/10/2024] [Indexed: 05/19/2024]
Abstract
Invasive graft biopsies assess the efficacy of immunosuppression through lagging indicators of transplant rejection. We report on a microporous scaffold implant as a minimally invasive immunological niche to assay rejection before graft injury. Adoptive transfer of T cells into Rag2-/- mice with mismatched allografts induced acute cellular allograft rejection (ACAR), with subsequent validation in wild-type animals. Following murine heart or skin transplantation, scaffold implants accumulate predominantly innate immune cells. The scaffold enables frequent biopsy, and gene expression analyses identified biomarkers of ACAR before clinical signs of graft injury. This gene signature distinguishes ACAR and immunodeficient respiratory infection before injury onset, indicating the specificity of the biomarkers to differentiate ACAR from other inflammatory insult. Overall, this implantable scaffold enables remote evaluation of the early risk of rejection, which could potentially be used to reduce the frequency of routine graft biopsy, reduce toxicities by personalizing immunosuppression, and prolong transplant life.
Collapse
Affiliation(s)
- Russell R. Urie
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Aaron Morris
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Diana Farris
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Elizabeth Hughes
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Chengchuan Xiao
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Judy Chen
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
- Program in Immunology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Elizabeth Lombard
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jiane Feng
- Animal Phenotyping Core, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jun Z. Li
- Department of Human Genetics, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Daniel R. Goldstein
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
- Program in Immunology, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Lonnie D. Shea
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
18
|
Bradford STJ, Wu H, Kirita Y, Chen C, Malvin NP, Yoshimura Y, Muto Y, Humphreys BD. TNIK depletion induces inflammation and apoptosis in injured renal proximal tubule epithelial cells. Am J Physiol Renal Physiol 2024; 326:F827-F838. [PMID: 38482555 PMCID: PMC11386974 DOI: 10.1152/ajprenal.00262.2023] [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: 08/28/2023] [Revised: 03/05/2024] [Accepted: 03/05/2024] [Indexed: 04/12/2024] Open
Abstract
In the aftermath of acute kidney injury (AKI), surviving proximal tubule epithelia repopulate injured tubules to promote repair. However, a portion of cells fail to repair [termed failed-repair proximal tubule cells (FR-PTCs)] and exert ongoing proinflammatory and profibrotic effects. To better understand the molecular drivers of the FR-PTC state, we reanalyzed a mouse ischemia-reperfusion injury single-nucleus RNA-sequencing (snRNA-seq) atlas to identify Traf2 and Nck interacting kinase (Tnik) to be exclusively expressed in FR-PTCs but not in healthy or acutely injured proximal tubules after AKI (2 and 6 wk) in mice. We confirmed expression of Tnik protein in injured mouse and human tissues by immunofluorescence. Then, to determine the functional role of Tnik in FR-PTCs, we depleted TNIK with siRNA in two human renal proximal tubule epithelial cell lines (primary and immortalized hRPTECs) and analyzed each by bulk RNA-sequencing. Pathway analysis revealed significant upregulation of inflammatory signaling pathways, whereas pathways associated with differentiated proximal tubules such as organic acid transport were significantly downregulated. TNIK gene knockdown drove reduced cell viability and increased apoptosis, including differentially expressed poly(ADP-ribose) polymerase (PARP) family members, cleaved PARP-1 fragments, and increased annexin V binding to phosphatidylserine. Together, these results indicate that Tnik upregulation in FR-PTCs acts in a compensatory fashion to suppress inflammation and promote proximal tubule epithelial cell survival after injury. Modulating TNIK activity may represent a prorepair therapeutic strategy after AKI.NEW & NOTEWORTHY The molecular drivers of successful and failed repair in the proximal tubule after acute kidney injury (AKI) are incompletely understood. We identified Traf2 and Nck interacting kinase (Tnik) to be exclusively expressed in failed-repair proximal tubule cells after AKI. We tested the effect of siTNIK depletion in two proximal tubule cell lines followed by bulk RNA-sequencing analysis. Our results indicate that TNIK acts to suppress inflammatory signaling and apoptosis in injured renal proximal tubule epithelial cells to promote cell survival.
Collapse
Affiliation(s)
- Shayna T J Bradford
- Division of Nephrology, Department of Medicine, Washington University in St. Louis School of Medicine, St. Louis, Missouri, United States
| | - Haojia Wu
- Division of Nephrology, Department of Medicine, Washington University in St. Louis School of Medicine, St. Louis, Missouri, United States
| | - Yuhei Kirita
- Department of Nephrology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Changfeng Chen
- Division of Nephrology, Department of Medicine, Washington University in St. Louis School of Medicine, St. Louis, Missouri, United States
| | - Nicole P Malvin
- Division of Nephrology, Department of Medicine, Washington University in St. Louis School of Medicine, St. Louis, Missouri, United States
| | - Yasuhiro Yoshimura
- Division of Nephrology, Department of Medicine, Washington University in St. Louis School of Medicine, St. Louis, Missouri, United States
| | - Yoshiharu Muto
- Division of Nephrology, Department of Medicine, Washington University in St. Louis School of Medicine, St. Louis, Missouri, United States
| | - Benjamin D Humphreys
- Division of Nephrology, Department of Medicine, Washington University in St. Louis School of Medicine, St. Louis, Missouri, United States
- Department of Developmental Biology, Washington University in St. Louis School of Medicine, St. Louis, Missouri, United States
| |
Collapse
|
19
|
Halloran PF, Madill-Thomsen K, Mackova M, Aliabadi-Zuckermann AZ, Cadeiras M, Crespo-Leiro MG, Depasquale EC, Deng M, Gökler J, Hall SA, Kim DH, Kobashigawa J, Macdonald P, Potena L, Shah K, Stehlik J, Zuckermann A, Reeve J. Molecular states associated with dysfunction and graft loss in heart transplants. J Heart Lung Transplant 2024; 43:508-518. [PMID: 38042442 DOI: 10.1016/j.healun.2023.11.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 10/23/2023] [Accepted: 11/21/2023] [Indexed: 12/04/2023] Open
Abstract
BACKGROUND We explored the changes in gene expression correlating with dysfunction and graft failure in endomyocardial biopsies. METHODS Genome-wide microarrays (19,462 genes) were used to define mRNA changes correlating with dysfunction (left ventricular ejection fraction [LVEF] ≤ 55) and risk of graft loss within 3 years postbiopsy. LVEF data was available for 1,013 biopsies and survival data for 779 patients (74 losses). Molecular classifiers were built for predicting dysfunction (LVEF ≤ 55) and postbiopsy 3-year survival. RESULTS Dysfunction is correlated with dedifferentiation-decreased expression of normal heart transcripts, for example, solute carriers, along with increased expression of inflammation genes. Many genes with reduced expression in dysfunction were matrix genes such as fibulin 1 and decorin. Gene ontology (GO) categories suggested matrix remodeling and inflammation, not rejection. Genes associated with the risk of failure postbiopsy overlapped dysfunction genes but also included genes affecting microcirculation, for example, arginase 2, which reduces NO production, and endothelin 1. GO terms also reflected increased glycolysis and response to hypoxia, but decreased VEGF and angiogenesis pathways. T cell-mediated rejection was associated with reduced survival and antibody-mediated rejection with relatively good survival, but the main determinants of survival were features of parenchymal injury. Both dysfunction and graft loss were correlated with increased biopsy expression of BNP (gene NPPB). Survival probability classifiers divided hearts into risk quintiles, with actuarial 3-year postbiopsy survival >95% for the highest versus 50% for the lowest. CONCLUSIONS Dysfunction in transplanted hearts reflects dedifferentiation, decreased matrix genes, injury, and inflammation. The risk of short-term loss includes these changes but is also associated with microcirculation abnormalities, glycolysis, and response to hypoxia.
Collapse
Affiliation(s)
- Philip F Halloran
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada.
| | | | - Martina Mackova
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | | | | | | | | | - Mario Deng
- Ronald Reagan UCLA Medical Center, Los Angeles, California
| | - Johannes Gökler
- Department of Cardiac Surgery, Medical University of Vienna, Vienna, Austria
| | | | - Daniel H Kim
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | | | - Peter Macdonald
- The Victor Chang Cardiac Research Institute, Sydney, Australia
| | - Luciano Potena
- Heart Failure and Transplant Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Keyur Shah
- Department of Cardiology, Virginia Commonwealth University, Richmond, Virginia
| | - Josef Stehlik
- Department of Medicine, University of Utah, Salt Lake City, Utah
| | - Andreas Zuckermann
- Department of Cardiac Surgery, Medical University of Vienna, Vienna, Austria
| | - Jeff Reeve
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
20
|
Qi P, Huang MJ, Wu W, Ren XW, Zhai YZ, Qiu C, Zhu HY. Exploration of potential biomarkers and therapeutic targets for trauma-related acute kidney injury. Chin J Traumatol 2024; 27:97-106. [PMID: 38296680 DOI: 10.1016/j.cjtee.2024.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 12/02/2023] [Accepted: 01/02/2024] [Indexed: 02/02/2024] Open
Abstract
PURPOSE Acute kidney injury (AKI) is one of the most common functional injuries observed in trauma patients. However, certain trauma medications may exacerbate renal injury. Therefore, the early detection of trauma-related AKI holds paramount importance in improving trauma prognosis. METHODS Qualified datasets were selected from public databases, and common differentially expressed genes related to trauma-induced AKI and hub genes were identified through enrichment analysis and the establishment of protein-protein interaction (PPI) networks. Additionally, the specificity of these hub genes was investigated using the sepsis dataset and conducted a comprehensive literature review to assess their plausibility. The raw data from both datasets were downloaded using R software (version 4.2.1) and processed with the "affy" package19 for correction and normalization. RESULTS Our analysis revealed 585 upregulated and 629 downregulated differentially expressed genes in the AKI dataset, along with 586 upregulated and 948 downregulated differentially expressed genes in the trauma dataset. Concurrently, the establishment of the PPI network and subsequent topological analysis highlighted key hub genes, including CD44, CD163, TIMP metallopeptidase inhibitor 1, cytochrome b-245 beta chain, versican, membrane spanning 4-domains A4A, mitogen-activated protein kinase 14, and early growth response 1. Notably, their receiver operating characteristic curves displayed areas exceeding 75%, indicating good diagnostic performance. Moreover, our findings postulated a unique molecular mechanism underlying trauma-related AKI. CONCLUSION This study presents an alternative strategy for the early diagnosis and treatment of trauma-related AKI, based on the identification of potential biomarkers and therapeutic targets. Additionally, this study provides theoretical references for elucidating the mechanisms of trauma-related AKI.
Collapse
Affiliation(s)
- Peng Qi
- Department of Emergency, First Medical Center of Chinese PLA General Hospital, Beijing, 100853, China
| | - Meng-Jie Huang
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Beijing, 100853, China
| | - Wei Wu
- Department of Anesthesiology, First Medical Center of Chinese PLA General Hospital, Beijing, 100853, China
| | - Xue-Wen Ren
- Department of Emergency, First Medical Center of Chinese PLA General Hospital, Beijing, 100853, China
| | - Yong-Zhi Zhai
- Department of Emergency, First Medical Center of Chinese PLA General Hospital, Beijing, 100853, China
| | - Chen Qiu
- Department of Orthopedics, Fourth Medical Center of Chinese PLA General Hospital, Beijing, 100853, China.
| | - Hai-Yan Zhu
- Department of Emergency, First Medical Center of Chinese PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
21
|
Herz CT, Diebold M, Kainz A, Mayer KA, Doberer K, Kozakowski N, Halloran PF, Böhmig GA. Morphologic and Molecular Features of Antibody-Mediated Transplant Rejection: Pivotal Role of Molecular Injury as an Independent Predictor of Renal Allograft Functional Decline. Transpl Int 2023; 36:12135. [PMID: 38169771 PMCID: PMC10758445 DOI: 10.3389/ti.2023.12135] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 12/04/2023] [Indexed: 01/05/2024]
Abstract
Current knowledge about the factors correlating with functional decline and subsequent failure of kidney allografts in antibody-mediated rejection (ABMR) is limited. We conducted a cohort study involving 75 renal allograft recipients diagnosed with late ABMR occurring at least 6 months after transplantation. The study aimed to examine the correlation of molecular and histologic features with estimated glomerular filtration rate (eGFR) trajectories and death-censored graft survival. We focused on sum scores reflecting histologic ABMR activity versus chronicity and molecular scores of ABMR probability (ABMRProb), injury-repair response (IRRAT) and fibrosis (ciprob). In multivariable Cox analysis, a Banff lesion-based chronicity index (ci+ct+cg[x2]; hazard ratio per interquartile range [IQR]: 1.97 [95% confidence interval: 0.97 to 3.99]) and IRRAT (1.93 [0.96 to 3.89]) showed the strongest associations with graft failure. Among biopsy variables, IRRAT exhibited the highest relative variable importance and emerged as the sole independent predictor of eGFR slope (change per IQR: -4.2 [-7.8 to -0.6] mL/min/1.73 m2/year). In contrast, morphologic chronicity associated with baseline eGFR only. We conclude that the extent of molecular injury is a robust predictor of renal function decline. Transcriptome analysis has the potential to improve outcome prediction and possibly identify modifiable injury, guiding targeted therapeutic interventions.
Collapse
Affiliation(s)
- Carsten T. Herz
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Matthias Diebold
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
- Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Alexander Kainz
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Katharina A. Mayer
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Konstantin Doberer
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | | | - Philip F. Halloran
- Alberta Transplant Applied Genomics Centre, ATAGC, University of Alberta, Edmonton, AB, Canada
| | - Georg A. Böhmig
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
22
|
Chatterjee E, Rodosthenous RS, Kujala V, Gokulnath P, Spanos M, Lehmann HI, de Oliveira GP, Shi M, Miller-Fleming TW, Li G, Ghiran IC, Karalis K, Lindenfeld J, Mosley JD, Lau ES, Ho JE, Sheng Q, Shah R, Das S. Circulating extracellular vesicles in human cardiorenal syndrome promote renal injury in a kidney-on-chip system. JCI Insight 2023; 8:e165172. [PMID: 37707956 PMCID: PMC10721327 DOI: 10.1172/jci.insight.165172] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 09/08/2023] [Indexed: 09/16/2023] Open
Abstract
BACKGROUNDCardiorenal syndrome (CRS) - renal injury during heart failure (HF) - is linked to high morbidity. Whether circulating extracellular vesicles (EVs) and their RNA cargo directly impact its pathogenesis remains unclear.METHODSWe investigated the role of circulating EVs from patients with CRS on renal epithelial/endothelial cells using a microfluidic kidney-on-chip (KOC) model. The small RNA cargo of circulating EVs was regressed against serum creatinine to prioritize subsets of functionally relevant EV-miRNAs and their mRNA targets investigated using in silico pathway analysis, human genetics, and interrogation of expression in the KOC model and in renal tissue. The functional effects of EV-RNAs on kidney epithelial cells were experimentally validated.RESULTSRenal epithelial and endothelial cells in the KOC model exhibited uptake of EVs from patients with HF. HF-CRS EVs led to higher expression of renal injury markers (IL18, LCN2, HAVCR1) relative to non-CRS EVs. A total of 15 EV-miRNAs were associated with creatinine, targeting 1,143 gene targets specifying pathways relevant to renal injury, including TGF-β and AMPK signaling. We observed directionally consistent changes in the expression of TGF-β pathway members (BMP6, FST, TIMP3) in the KOC model exposed to CRS EVs, which were validated in epithelial cells treated with corresponding inhibitors and mimics of miRNAs. A similar trend was observed in renal tissue with kidney injury. Mendelian randomization suggested a role for FST in renal function.CONCLUSIONPlasma EVs in patients with CRS elicit adverse transcriptional and phenotypic responses in a KOC model by regulating biologically relevant pathways, suggesting a role for EVs in CRS.TRIAL REGISTRATIONClinicalTrials.gov NCT03345446.FUNDINGAmerican Heart Association (AHA) (SFRN16SFRN31280008); National Heart, Lung, and Blood Institute (1R35HL150807-01); National Center for Advancing Translational Sciences (UH3 TR002878); and AHA (23CDA1045944).
Collapse
Affiliation(s)
- Emeli Chatterjee
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Rodosthenis S. Rodosthenous
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, Massachusetts, USA
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | | | - Priyanka Gokulnath
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Michail Spanos
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Helge Immo Lehmann
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | | | | | | | - Guoping Li
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Ionita Calin Ghiran
- Department of Anesthesia, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Katia Karalis
- Emulate, Inc., Boston, Massachusetts, USA
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, USA
| | - JoAnn Lindenfeld
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Jonathan D. Mosley
- Department of Biomedical Informatics and
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Emily S. Lau
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Jennifer E. Ho
- Cardiovascular Institute, Division of Cardiovascular Medicine, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | | | - Ravi Shah
- Vanderbilt Translational and Clinical Research Center, Cardiology Division, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Saumya Das
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
23
|
Xie M, Xie R, Huang P, Yap DYH, Wu P. GADD45A and GADD45B as Novel Biomarkers Associated with Chromatin Regulators in Renal Ischemia-Reperfusion Injury. Int J Mol Sci 2023; 24:11304. [PMID: 37511062 PMCID: PMC10379085 DOI: 10.3390/ijms241411304] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/21/2023] [Accepted: 07/03/2023] [Indexed: 07/30/2023] Open
Abstract
Chromatin regulators (CRs) are essential upstream regulatory factors of epigenetic modification. The role of CRs in the pathogenesis of renal ischemia-reperfusion injury (IRI) remains unclear. We analyzed a bioinformatic analysis on the differentially expressed chromatin regulator genes in renal IRI patients using data from public domains. The hub CRs identified were used to develop a risk prediction model for renal IRI, and their expressions were also validated using Western blot, qRT-PCR, and immunohistochemistry in a murine renal IRI model. We also examined the relationships between hub CRs and infiltrating immune cells in renal IRI and used network analysis to explore drugs that target hub CRs and their relevant downstream microRNAs. The results of machine learning methods showed that five genes (DUSP1, GADD45A, GADD45B, GADD45G, HSPA1A) were upregulated in renal IRI, with key roles in the cell cycle, p38 MAPK signaling pathway, p53 signaling pathway, FoxO signaling pathway, and NF-κB signaling pathway. Two genes from the network, GADD45A and GADD45B (growth arrest and DNA damage-inducible protein 45 alpha and beta), were chosen for the renal IRI risk prediction model. They all showed good performance in the testing and validation cohorts. Mice with renal IRI showed significantly upregulated GADD45A and GADD45B expression within kidneys compared to sham-operated mice. GADD45A and GADD45B showed correlations with plasmacytoid dendritic cells (pDCs) in infiltrating immune cell analysis and enrichment in the MAPK pathway based on the weighted gene co-expression network analysis (WGCNA) method. Candidate drugs that target GADD45A and GADD45B include beta-escin, sertraline, primaquine, pimozide, and azacyclonol. The dysregulation of GADD45A and GADD45B is related to renal IRI and the infiltration of pDCs, and drugs that target GADD45A and GADD45B may have therapeutic potential for renal IRI.
Collapse
Affiliation(s)
- Ming Xie
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Ruiyan Xie
- Division of Nephrology, Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong 999077, China
| | - Pengcheng Huang
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Desmond Y H Yap
- Division of Nephrology, Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong 999077, China
| | - Peng Wu
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
24
|
Ray N, Park SJ, Jung H, Kim J, Korcsmaros T, Moon Y. Stress-responsive Gdf15 counteracts renointestinal toxicity via autophagic and microbiota reprogramming. Commun Biol 2023; 6:602. [PMID: 37270567 DOI: 10.1038/s42003-023-04965-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 05/22/2023] [Indexed: 06/05/2023] Open
Abstract
The integrated stress response (ISR) plays a pivotal role in the cellular stress response, primarily through global translational arrest and the upregulation of cellular adaptation-linked molecules. Growth differentiation factor 15 (Gdf15) is a potent stress-responsive biomarker of clinical inflammatory and metabolic distress in various types of diseases. Herein, we assess whether ISR-driven cellular stress contributes to pathophysiological outcomes by modulating Gdf15. Clinical transcriptome analysis demonstrates that PKR is positively associated with Gdf15 expression in patients with renal injury. Gdf15 expression is dependent on protein kinase R (PKR)-linked ISR during acute renointestinal distress in mice and genetic ablation of Gdf15 aggravates chemical-induced lesions in renal tissues and the gut barrier. An in-depth evaluation of the gut microbiota indicates that Gdf15 is associated with the abundance of mucin metabolism-linked bacteria and their enzymes. Moreover, stress-responsive Gdf15 facilitates mucin production and cellular survival via the reorganization of the autophagy regulatory network. Collectively, ISR-activated Gdf15 counteracts pathological processes via the protective reprogramming of the autophagic network and microbial community, thereby providing robust predictive biomarkers and interventions against renointestinal distress.
Collapse
Affiliation(s)
- Navin Ray
- Laboratory of Mucosal Exposome and Biomodulation, Department of Integrative Biomedical Sciences, Pusan National University, Yangsan, Korea
| | - Seung Jun Park
- Laboratory of Mucosal Exposome and Biomodulation, Department of Integrative Biomedical Sciences, Pusan National University, Yangsan, Korea
| | - Hoyung Jung
- Laboratory of Mucosal Exposome and Biomodulation, Department of Integrative Biomedical Sciences, Pusan National University, Yangsan, Korea
| | - Juil Kim
- Laboratory of Mucosal Exposome and Biomodulation, Department of Integrative Biomedical Sciences, Pusan National University, Yangsan, Korea
| | - Tamas Korcsmaros
- Division of Digestive Diseases, Faculty of Medicine, Imperial College London, London, UK
- Earlham Institute, Norwich Research Park, Norwich, UK
- Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
| | - Yuseok Moon
- Laboratory of Mucosal Exposome and Biomodulation, Department of Integrative Biomedical Sciences, Pusan National University, Yangsan, Korea.
- Earlham Institute, Norwich Research Park, Norwich, UK.
- Graduate Program of Genomic Data Sciences, Pusan National University, Yangsan, Korea.
| |
Collapse
|
25
|
Li J, Li T, Li Z, Song Z, Gong X. Nephroprotective mechanisms of Rhizoma Chuanxiong and Radix et Rhizoma Rhei against acute renal injury and renal fibrosis based on network pharmacology and experimental validation. Front Pharmacol 2023; 14:1154743. [PMID: 37229255 PMCID: PMC10203597 DOI: 10.3389/fphar.2023.1154743] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 04/25/2023] [Indexed: 05/27/2023] Open
Abstract
The molecular mechanisms of Rhizoma Chuanxiong (Chuanxiong, CX) and Rhei Radix et Rhizoma (Dahuang, DH) in treating acute kidney injury (AKI) and subsequent renal fibrosis (RF) were investigated in this study by applying network pharmacology and experimental validation. The results showed that aloe-emodin, (-)-catechin, beta-sitosterol, and folic acid were the core active ingredients, and TP53, AKT1, CSF1R, and TGFBR1 were the core target genes. Enrichment analyses showed that the key signaling pathways were the MAPK and IL-17 signaling pathways. In vivo experiments confirmed that Chuanxiong and Dahuang pretreatments significantly inhibited the levels of SCr, BUN, UNAG, and UGGT in contrast media-induced acute kidney injury (CIAKI) rats (p < 0.001). The results of Western blotting showed that compared with the control group, the protein levels of p-p38/p38 MAPK, p53, and Bax in the contrast media-induced acute kidney injury group were significantly increased, and the levels of Bcl-2 were significantly reduced (p < 0.001). Chuanxiong and Dahuang interventions significantly reversed the expression levels of these proteins (p < 0.01). The localization and quantification of p-p53 expression in immunohistochemistry technology also support the aforementioned results. In conclusion, our data also suggest that Chuanxiong and Dahuang may inhibit tubular epithelial cell apoptosis and improve acute kidney injury and renal fibrosis by inhibiting p38 MAPK/p53 signaling.
Collapse
|
26
|
Bayır H, Dixon SJ, Tyurina YY, Kellum JA, Kagan VE. Ferroptotic mechanisms and therapeutic targeting of iron metabolism and lipid peroxidation in the kidney. Nat Rev Nephrol 2023; 19:315-336. [PMID: 36922653 DOI: 10.1038/s41581-023-00689-x] [Citation(s) in RCA: 124] [Impact Index Per Article: 62.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/07/2023] [Indexed: 03/17/2023]
Abstract
Ferroptosis is a mechanism of regulated necrotic cell death characterized by iron-dependent, lipid peroxidation-driven membrane destruction that can be inhibited by glutathione peroxidase 4. Morphologically, it is characterized by cellular, organelle and cytoplasmic swelling and the loss of plasma membrane integrity, with the release of intracellular components. Ferroptosis is triggered in cells with dysregulated iron and thiol redox metabolism, whereby the initial robust but selective accumulation of hydroperoxy polyunsaturated fatty acid-containing phospholipids is further propagated through enzymatic and non-enzymatic secondary mechanisms, leading to formation of oxidatively truncated electrophilic species and their adducts with proteins. Thus, ferroptosis is dependent on the convergence of iron, thiol and lipid metabolic pathways. The kidney is particularly susceptible to redox imbalance. A growing body of evidence has linked ferroptosis to acute kidney injury in the context of diverse stimuli, such as ischaemia-reperfusion, sepsis or toxins, and to chronic kidney disease, suggesting that ferroptosis may represent a novel therapeutic target for kidney disease. However, further work is needed to address gaps in our understanding of the triggers, execution and spreading mechanisms of ferroptosis.
Collapse
Affiliation(s)
- Hülya Bayır
- Department of Critical Care Medicine, Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, USA.
- Center for Free Radical and Antioxidant Health, Departments of Environmental Health, Pharmacology and Chemical Biology, Chemistry, Radiation Oncology, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Pediatrics, Division of Critical Care and Hospital Medicine, Redox Health Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA.
| | - Scott J Dixon
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Yulia Y Tyurina
- Center for Free Radical and Antioxidant Health, Departments of Environmental Health, Pharmacology and Chemical Biology, Chemistry, Radiation Oncology, University of Pittsburgh, Pittsburgh, PA, USA
| | - John A Kellum
- Department of Critical Care Medicine, Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Valerian E Kagan
- Center for Free Radical and Antioxidant Health, Departments of Environmental Health, Pharmacology and Chemical Biology, Chemistry, Radiation Oncology, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
27
|
Smith RN, Rosales IA, Tomaszewski KT, Mahowald GT, Araujo-Medina M, Acheampong E, Bruce A, Rios A, Otsuka T, Tsuji T, Hotta K, Colvin R. Utility of Banff Human Organ Transplant Gene Panel in Human Kidney Transplant Biopsies. Transplantation 2023; 107:1188-1199. [PMID: 36525551 PMCID: PMC10132999 DOI: 10.1097/tp.0000000000004389] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
BACKGROUND Microarray transcript analysis of human renal transplantation biopsies has successfully identified the many patterns of graft rejection. To evaluate an alternative, this report tests whether gene expression from the Banff Human Organ Transplant (B-HOT) probe set panel, derived from validated microarrays, can identify the relevant allograft diagnoses directly from archival human renal transplant formalin-fixed paraffin-embedded biopsies. To test this hypothesis, principal components (PCs) of gene expressions were used to identify allograft diagnoses, to classify diagnoses, and to determine whether the PC data were rich enough to identify diagnostic subtypes by clustering, which are all needed if the B-HOT panel can substitute for microarrays. METHODS RNA was isolated from routine, archival formalin-fixed paraffin-embedded tissue renal biopsy cores with both rejection and nonrejection diagnoses. The B-HOT panel expression of 770 genes was analyzed by PCs, which were then tested to determine their ability to identify diagnoses. RESULTS PCs of microarray gene sets identified the Banff categories of renal allograft diagnoses, modeled well the aggregate diagnoses, showing a similar correspondence with the pathologic diagnoses as microarrays. Clustering of the PCs identified diagnostic subtypes including non-chronic antibody-mediated rejection with high endothelial expression. PCs of cell types and pathways identified new mechanistic patterns including differential expression of B and plasma cells. CONCLUSIONS Using PCs of gene expression from the B-Hot panel confirms the utility of the B-HOT panel to identify allograft diagnoses and is similar to microarrays. The B-HOT panel will accelerate and expand transcript analysis and will be useful for longitudinal and outcome studies.
Collapse
Affiliation(s)
- Rex N Smith
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- Center for Transplantation Sciences, Massachusetts General Hospital, Boston, MA
| | - Ivy A Rosales
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- Center for Transplantation Sciences, Massachusetts General Hospital, Boston, MA
| | - Kristen T Tomaszewski
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- Center for Transplantation Sciences, Massachusetts General Hospital, Boston, MA
| | - Grace T Mahowald
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Milagros Araujo-Medina
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Ellen Acheampong
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Amy Bruce
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Andrea Rios
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Takuya Otsuka
- Department of Surgical Pathology, Hokkaido University Hospital, Sapporo, Japan
| | - Takahiro Tsuji
- Department of Pathology, Sapporo City General Hospital, Sapporo, Japan
| | - Kiyohiko Hotta
- Department of Urology, Hokkaido University Hospital, Sapporo, Japan
| | - Robert Colvin
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- Center for Transplantation Sciences, Massachusetts General Hospital, Boston, MA
| |
Collapse
|
28
|
Halawi A, El Kurdi AB, Vernon KA, Solhjou Z, Choi JY, Saad AJ, Younis NK, Elfekih R, Mohammed MT, Deban CA, Weins A, Abdi R, Riella LV, De Serres SA, Cravedi P, Greka A, Khoueiry P, Azzi JR. Uncovering a novel role of focal adhesion and interferon-gamma in cellular rejection of kidney allografts at single cell resolution. Front Immunol 2023; 14:1139358. [PMID: 37063857 PMCID: PMC10102512 DOI: 10.3389/fimmu.2023.1139358] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 02/23/2023] [Indexed: 04/03/2023] Open
Abstract
BackgroundKidney transplant recipients are currently treated with nonspecific immunosuppressants that cause severe systemic side effects. Current immunosuppressants were developed based on their effect on T-cell activation rather than the underlying mechanisms driving alloimmune responses. Thus, understanding the role of the intragraft microenvironment will help us identify more directed therapies with lower side effects.MethodsTo understand the role of the alloimmune response and the intragraft microenvironment in cellular rejection progression, we conducted a Single nucleus RNA sequencing (snRNA-seq) on one human non-rejecting kidney allograft sample, one borderline sample, and T-cell mediated rejection (TCMR) sample (Banff IIa). We studied the differential gene expression and enriched pathways in different conditions, in addition to ligand-receptor (L-R) interactions.ResultsPathway analysis of T-cells in borderline sample showed enrichment for allograft rejection pathway, suggesting that the borderline sample reflects an early rejection. Hence, this allows for studying the early stages of cellular rejection. Moreover, we showed that focal adhesion (FA), IFNg pathways, and endomucin (EMCN) were significantly upregulated in endothelial cell clusters (ECs) of borderline compared to ECs TCMR. Furthermore, we found that pericytes in TCMR seem to favor endothelial permeability compared to borderline. Similarly, T-cells interaction with ECs in borderline differs from TCMR by involving DAMPS-TLRs interactions.ConclusionOur data revealed novel roles of T-cells, ECs, and pericytes in cellular rejection progression, providing new clues on the pathophysiology of allograft rejection.
Collapse
Affiliation(s)
- Ahmad Halawi
- Transplantation Research Center, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Abdullah B. El Kurdi
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | | | - Zhabiz Solhjou
- Transplantation Research Center, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
- Scripps Clinic Medical Group, San Diego, CA, United States
| | - John Y. Choi
- Transplantation Research Center, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Anis J. Saad
- Transplantation Research Center, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Nour K. Younis
- Transplantation Research Center, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Rania Elfekih
- Transplantation Research Center, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Mostafa Tawfeek Mohammed
- Transplantation Research Center, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
- Clinical Pathology Department, Faculty of Medicine, Minia University, Minia, Egypt
| | - Christa A. Deban
- Transplantation Research Center, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Astrid Weins
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
| | - Reza Abdi
- Transplantation Research Center, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Leonardo V. Riella
- Department of Medicine, Division of Nephrology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, MA, United States
| | - Sasha A. De Serres
- Transplantation Unit, Renal Division, Department of Medicine, University Health Center of Quebec, Faculty of Medicine, Laval University, Québec, QC, Canada
| | - Paolo Cravedi
- Translational Transplant Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Anna Greka
- The Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, United States
- Department of Medicine, Brigham and Women’s Hospital, Boston, MA, United States
| | - Pierre Khoueiry
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Jamil R. Azzi
- Transplantation Research Center, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
- *Correspondence: Jamil R. Azzi,
| |
Collapse
|
29
|
The Molecular Diagnosis Might Be Clinically Useful in Discrepant Kidney Allograft Biopsy Findings: An Analysis of Clinical Outcomes. Transplantation 2023; 107:485-494. [PMID: 36117252 PMCID: PMC9875837 DOI: 10.1097/tp.0000000000004284] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
BACKGROUND The Molecular Microscope Diagnostic System (MMDx) may overcome histology shortcomings. Previous studies have simply examined discrepant findings but have not attempted to determine clinical endpoints. To measure performance, clinical outcomes are strongly required. METHODS This single-center cohort study described discrepancies between MMDx and histology from 51 kidney transplant recipients (KTRs) and analyzed 72 indication biopsies, including 21 follow-up biopsies. Clinical performance was assessed by a combined endpoint of graft failure, rejection on follow-up biopsy, de novo donor-specific antibody, and improvement of kidney allograft function upon antirejection treatment. RESULTS MMDx agreed in 33 (65%) and differed in 18 (35%) of 51 KTRs. Most discrepancies occurred in biopsies called no rejection by MMDx and rejection by histology (15/24, 63%). In contrast, in biopsies called rejection by MMDx, 3 were classified as no rejection by histology (3/27, 11%). Discrepant findings between MMDx and histology occurred following delayed graft function and MMDx from biopsies with a low percentage of cortex. Among 15 biopsies classified as no rejection by MMDx but rejection by histology, the clinical course suggested no rejection in 9 cases. Six KTRs reached the endpoint, showing predominant t ≥ 2 lesions. CONCLUSIONS The most often occurring discrepancy is rejection by histology but no rejection by MMDx. As more KTRs do not meet the combined endpoint for rejection, MMDx might be clinically useful in these discrepant cases. Although strong histological findings have priority in indicating the treatment, clinical implementation of MMDx could strengthen treatment strategies.
Collapse
|
30
|
The Molecular Microscope Diagnostic System: Assessment of Rejection and Injury in Heart Transplant Biopsies. Transplantation 2023; 107:27-44. [PMID: 36508644 DOI: 10.1097/tp.0000000000004323] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
This review describes the development of the Molecular Microscope Diagnostic System (MMDx) for heart transplant endomyocardial biopsies (EMBs). MMDx-Heart uses microarrays to measure biopsy-based gene expression and ensembles of machine learning algorithms to interpret the results and compare each new biopsy to a large reference set of earlier biopsies. MMDx assesses T cell-mediated rejection (TCMR), antibody-mediated rejection (AMR), recent parenchymal injury, and atrophy-fibrosis, continually "learning" from new biopsies. Rejection-associated transcripts mapped in kidney transplants and experimental systems were used to identify TCMR, AMR, and recent injury-induced inflammation. Rejection and injury emerged as gradients of intensity, rather than binary classes. AMR was one-third donor-specific antibody (DSA)-negative, and many EMBs first considered to have no rejection displayed minor AMR-like changes, with increased probability of DSA positivity and subtle inflammation. Rejection-associated transcript-based algorithms now classify EMBs as "Normal," "Minor AMR changes," "AMR," "possible AMR," "TCMR," "possible TCMR," and "recent injury." Additionally, MMDx uses injury-associated transcript sets to assess the degree of parenchymal injury and atrophy-fibrosis in every biopsy and study the effect of rejection on the parenchyma. TCMR directly injures the parenchyma whereas AMR usually induces microcirculation stress but relatively little initial parenchymal damage, although slowly inducing parenchymal atrophy-fibrosis. Function (left ventricular ejection fraction) and short-term risk of failure are strongly determined by parenchymal injury. These discoveries can guide molecular diagnostic applications, either as a central MMDx system or adapted to other platforms. MMDx can also help calibrate noninvasive blood-based biomarkers to avoid unnecessary biopsies and monitor response to therapy.
Collapse
|
31
|
Li H, Dixon EE, Wu H, Humphreys BD. Comprehensive single-cell transcriptional profiling defines shared and unique epithelial injury responses during kidney fibrosis. Cell Metab 2022; 34:1977-1998.e9. [PMID: 36265491 PMCID: PMC9742301 DOI: 10.1016/j.cmet.2022.09.026] [Citation(s) in RCA: 106] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 07/19/2022] [Accepted: 09/28/2022] [Indexed: 01/12/2023]
Abstract
The underlying cellular events driving kidney fibrogenesis and metabolic dysfunction are incompletely understood. Here, we employed single-cell combinatorial indexing RNA sequencing to analyze 24 mouse kidneys from two fibrosis models. We profiled 309,666 cells in one experiment, representing 50 cell types/states encompassing epithelial, endothelial, immune, and stromal populations. Single-cell analysis identified diverse injury states of the proximal tubule, including two distinct early-phase populations with dysregulated lipid and amino acid metabolism, respectively. Lipid metabolism was defective in the chronic phase but was transiently activated in the very early stages of ischemia-induced injury, where we discovered increased lipid deposition and increased fatty acid β-oxidation. Perilipin 2 was identified as a surface marker of intracellular lipid droplets, and its knockdown in vitro disrupted cell energy state maintenance during lipid accumulation. Surveying epithelial cells across nephron segments identified shared and unique injury responses. Stromal cells exhibited high heterogeneity and contributed to fibrogenesis by epithelial-stromal crosstalk.
Collapse
Affiliation(s)
- Haikuo Li
- Division of Nephrology, Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Eryn E Dixon
- Division of Nephrology, Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Haojia Wu
- Division of Nephrology, Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Benjamin D Humphreys
- Division of Nephrology, Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA; Department of Developmental Biology, Washington University in St. Louis, St. Louis, MO, USA.
| |
Collapse
|
32
|
Peng L, Liu D, Liu H, Xia M, Wan L, Li M, Zhao J, Tang C, Chen G, Qu X, Dong Z, Liu H. Bombesin receptor-activated protein exacerbates cisplatin-induced AKI by regulating the degradation of SIRT2. Nephrol Dial Transplant 2022; 37:2366-2385. [PMID: 35488871 DOI: 10.1093/ndt/gfac164] [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: 10/17/2021] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Acute kidney injury (AKI) is a public health problem with no specific therapies in the clinic and the underlying pathogenesis of AKI remains obscure. Bombesin receptor-activated protein (BRAP, C6ORF89 protein) was initially discovered as a ligand for a previously orphan G-protein-coupled receptor bombesin-like receptor-3. At present, accepted biological effects of BRAP include cell cycle progression, wound repair and the activation of histone deacetylases. However, its role in kidney disease is unknown. In this study we have investigated the role of BRAP and underlying mechanisms involved in cisplatin (CP)-induced AKI. METHODS Here we used Bc004004 (homologous of C6ORF89 in mice) knockout mice and HK2 cells to investigate the effect of BRAP on AKI in vitro and in vivo. We analyzed ChIP-Seq and RNA-Seq data to search for the upstream regulators of BRAP and downstream mediators of BRAP action in AKI. Immunostaining, real-time polymerase chain reaction (PCR), co-immunoprecipitation, a dual-luciferase reporter assay and ChIP-PCR assay were applied to reveal the upstream and downstream regulation mechanism of BRAP during cisplatin-induced AKI. RESULTS BRAP was downregulated in mice and human kidneys with AKI. Global Bc004004 deletion alleviated tubular cell apoptosis and necroptosis in CP-induced AKI mice, whereas local overexpression of BRAP in kidneys aggravated them. Pan-caspase inhibitor Z-VAD pretreatment attenuated CP-induced blood creatinine increase and kidney injury in wild-type mice but not in BRAP -/- mice. The activation of mixed lineage kinase like-domain was magnified by Z-VAD in CP-treated mice, especially in BRAP -/- mice. The cytoprotective effect of Z-VAD was more substantial than necrostatin-1 (Nec-1, an inhibitor of necroptosis) in CP-treated human kidney proximal tubular epithelial (HK2) cells. Furthermore, Nec-1 pretreatment reduced the CP-induced cell death in BRAP overexpression HK2 cells but did not work in cells with normal BRAP levels. We determined that CP treatment activated the nuclear factor-κB subunit P65 and inhibition of P65 increased the messenger RNA (mRNA) levels of BRAP in HK2 cells. The chromatin immunoprecipitation assay and dual-luciferase reporter gene assay verified P65 binding to the C6ORF89 promoter and reduced its mRNA expression upon CP treatment. Next we found that sirtuin 2 (SIRT2) was downregulated in CP-induced AKI and BRAP levels directly impacted the protein levels of SIRT2. Our findings further confirmed that BRAP regulates the SIRT2 protein levels by affecting SIRT2's interactions with E3 ubiquitin ligase HRD1 and subsequent proteasomal degradation. CONCLUSIONS Our results demonstrated that BRAP played an important role in tubular cell apoptosis and necroptosis during CP-induced AKI. Safe and efficient BRAP inhibitors might be effective therapeutic options for AKI.
Collapse
Affiliation(s)
- Liang Peng
- Department of Nephrology, Second Xiangya Hospital of Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Di Liu
- Department of Nephrology, Second Xiangya Hospital of Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Haiyang Liu
- Department of Nephrology, Second Xiangya Hospital of Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Ming Xia
- Department of Nephrology, Second Xiangya Hospital of Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Lili Wan
- Department of Nephrology, Second Xiangya Hospital of Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Mei Li
- Department of Nephrology, Second Xiangya Hospital of Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Junyong Zhao
- Department of Nephrology, Second Xiangya Hospital of Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Chengyuan Tang
- Department of Nephrology, Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Guochun Chen
- Department of Nephrology, Second Xiangya Hospital of Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Xiangpin Qu
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Zheng Dong
- Department of Nephrology, Second Xiangya Hospital of Central South University, Changsha, Hunan, China.,Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University and Charlie Norwood VA Medical Center, Augusta, GA, USA
| | - Hong Liu
- Department of Nephrology, Second Xiangya Hospital of Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| |
Collapse
|
33
|
Assessing the Relationship Between Molecular Rejection and Parenchymal Injury in Heart Transplant Biopsies. Transplantation 2022; 106:2205-2216. [PMID: 35968995 DOI: 10.1097/tp.0000000000004231] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
BACKGROUND The INTERHEART study (ClinicalTrials.gov #NCT02670408) used genome-wide microarrays to detect rejection in endomyocardial biopsies; however, many heart transplants with no rejection have late dysfunction and impaired survival. We used the microarray measurements to develop a molecular classification of parenchymal injury. METHODS In 1320 endomyocardial biopsies from 645 patients previously studied for rejection-associated transcripts, we measured the expression of 10 injury-induced transcript sets: 5 induced by recent injury; 2 reflecting macrophage infiltration; 2 normal heart transcript sets; and immunoglobulin transcripts, which correlate with time. We used archetypal clustering to assign injury groups. RESULTS Injury transcript sets correlated with impaired function. Archetypal clustering based on the expression of injury transcript sets assigned each biopsy to 1 of 5 injury groups: 87 Severe-injury, 221 Late-injury, and 3 with lesser degrees of injury, 376 No-injury, 526 Mild-injury, and 110 Moderate-injury. Severe-injury had extensive loss of normal transcripts (dedifferentiation) and increase in macrophage and injury-induced transcripts. Late-injury was characterized by high immunoglobulin transcript expression. In Severe- and Late-injury, function was depressed, and short-term graft failure was increased, even in hearts with no rejection. T cell-mediated rejection almost always had parenchymal injury, and 85% had Severe- or Late-injury. In contrast, early antibody-mediated rejection (ABMR) had little injury, but late ABMR often had the Late-injury state. CONCLUSION Characterizing heart transplants for their injury state provides new understanding of dysfunction and outcomes and demonstrates the differential impact of T cell-mediated rejection versus ABMR on the parenchyma. Slow deterioration from ABMR emerges as a major contributor to late dysfunction.
Collapse
|
34
|
Zhang Y, Cai J, Lu W, Xu S, Qu M, Zhao S, Ding X. Comprehensive Network-Based Analyses Reveal Novel Renal Function-Related Targets in Acute Kidney Injury. Front Genet 2022; 13:907145. [PMID: 35860471 PMCID: PMC9289212 DOI: 10.3389/fgene.2022.907145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 05/23/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Acute kidney injury (AKI) is a common clinical syndrome with limited methods of treatment and diagnosis. Although several molecules associated with AKI have been discovered, molecular mechanisms underlying AKI still remain unclear. Weighted gene co-expression network analysis (WGCNA) is a novel method to uncover the relationship between co-expression genes and clinical traits at the system level. Methods: First, by employing WGCNA in transcriptional data on 30 patients with well/poor functioning kidney graft, we identified two co-expression modules that were significantly related to serum creatinine (SCr). Second, based on the modules, potential small molecular compound candidates for developing targeted therapeutics were obtained by connectivity map analysis. Furthermore, multiple validations of expression in space/time were carried out with two classical AKI models in vivo and other five databases of over 152 samples. Results: Two of the 14 modules were found to be closely correlated with SCr. Function enrichment analysis illustrated that one module was enriched in the immune system, while the other was in the metabolic process. Six key renal function-related genes (RFRGs) were finally obtained. Such genes performed well in cisplatin-induced or cecal ligation and puncture-induced AKI mouse models. Conclusion: The analysis suggests that WGCNA is a proper method to connect clinical traits with genome data to find novel targets in AKI. The kidney tissue with worse renal function tended to develop a “high immune but low metabolic activity” expression pattern. Also, ACSM2A, GLYAT, CORO1A, DPEP1, ALDH7A1, and EPHX2 are potential targets of molecular diagnosis and treatment in AKI.
Collapse
Affiliation(s)
- Yang Zhang
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jieru Cai
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Medical Center of Kidney Disease, Shanghai, China
- Kidney and Dialysis Institute of Shanghai, Shanghai, China
- Kidney and Blood Purification Key Laboratory of Shanghai, Shanghai, China
| | - Wei Lu
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Sujuan Xu
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Mengdi Qu
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shuan Zhao
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Medical Center of Kidney Disease, Shanghai, China
- Kidney and Dialysis Institute of Shanghai, Shanghai, China
- Kidney and Blood Purification Key Laboratory of Shanghai, Shanghai, China
- *Correspondence: Xiaoqiang Ding, ; Shuan Zhao,
| | - Xiaoqiang Ding
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Medical Center of Kidney Disease, Shanghai, China
- Kidney and Dialysis Institute of Shanghai, Shanghai, China
- Kidney and Blood Purification Key Laboratory of Shanghai, Shanghai, China
- *Correspondence: Xiaoqiang Ding, ; Shuan Zhao,
| |
Collapse
|
35
|
Halloran PF, Böhmig GA, Bromberg J, Einecke G, Eskandary FA, Gupta G, Myslak M, Viklicky O, Perkowska-Ptasinska A, Madill-Thomsen KS. Archetypal Analysis of Injury in Kidney Transplant Biopsies Identifies Two Classes of Early AKI. Front Med (Lausanne) 2022; 9:817324. [PMID: 35463013 PMCID: PMC9021747 DOI: 10.3389/fmed.2022.817324] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 03/07/2022] [Indexed: 01/07/2023] Open
Abstract
All transplanted kidneys are subjected to some degree of injury as a result of the donation-implantation process and various post-transplant stresses such as rejection. Because transplants are frequently biopsied, they present an opportunity to explore the full spectrum of kidney response-to-wounding from all causes. Defining parenchymal damage in transplanted organs is important for clinical management because it determines function and survival. In this study, we classified the scenarios associated with parenchymal injury in genome-wide microarray results from 1,526 kidney transplant indication biopsies collected during the INTERCOMEX study. We defined injury groups by using archetypal analysis (AA) of scores for gene sets and classifiers previously identified in various injury states. Six groups and their characteristics were defined in this population: No injury, minor injury, two classes of acute kidney injury ("AKI," AKI1, and AKI2), chronic kidney disease (CKD), and CKD combined with AKI. We compared the two classes of AKI, namely, AKI1 and AKI2. AKI1 had a poor function and increased parenchymal dedifferentiation but minimal response-to-injury and inflammation, instead having increased expression of PARD3, a gene previously characterized as being related to epithelial polarity and adherens junctions. In contrast, AKI2 had a poor function and increased response-to-injury, significant inflammation, and increased macrophage activity. In random forest analysis, the most important predictors of function (estimated glomerular filtration rate) and graft loss were injury-based molecular scores, not rejection scores. AKI1 and AKI2 differed in 3-year graft survival, with better survival in the AKI2 group. Thus, injury archetype analysis of injury-induced gene expression shows new heterogeneity in kidney response-to-wounding, revealing AKI1, a class of early transplants with a poor function but minimal inflammation or response to injury, a deviant response characterized as PC3, and an increased risk of failure. Given the relationship between parenchymal injury and kidney survival, further characterization of the injury phenotypes in kidney transplants will be important for an improved understanding that could have implications for understanding native kidney diseases (ClinicalTrials.gov #NCT01299168).
Collapse
Affiliation(s)
- Philip F Halloran
- Alberta Transplant Applied Genomics Centre, Edmonton, AB, Canada.,Division of Nephrology and Transplant Immunology, Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Georg A Böhmig
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Jonathan Bromberg
- Department of Surgery, University of Maryland, Baltimore, MD, United States
| | - Gunilla Einecke
- Department of Nephrology, Hannover Medical School, Hannover, Germany
| | - Farsad A Eskandary
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Gaurav Gupta
- Division of Nephrology, Virginia Commonwealth University, Richmond, VA, United States
| | - Marek Myslak
- Department of Clinical Interventions, Department of Nephrology and Kidney Transplantation Samodzielny Publiczny Wojewódzki Szpital Zespolony (SPWSZ) Hospital, Pomeranian Medical University, Szczecin, Poland
| | - Ondrej Viklicky
- Department of Nephrology and Transplant Center, Institute for Clinical and Experimental Medicine, Prague, Czechia
| | - Agnieszka Perkowska-Ptasinska
- Department of Transplantation Medicine, Nephrology and Internal Diseases, Medical University of Warsaw, Warsaw, Poland
| | | | | |
Collapse
|
36
|
Ray N, Jeong H, Kwon D, Kim J, Moon Y. Antibiotic Exposure Aggravates Bacteroides-Linked Uremic Toxicity in the Gut-Kidney Axis. Front Immunol 2022; 13:737536. [PMID: 35401522 PMCID: PMC8988921 DOI: 10.3389/fimmu.2022.737536] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 03/04/2022] [Indexed: 12/25/2022] Open
Abstract
Epidemiological and experimental evidence has implicated a potent link between antibiotic exposure and susceptibility to various diseases. Clinically, antibiotic treatment during platinum chemotherapy is associated with poor prognosis in patients with malignancy. In the present study, mucosal antibiotic exposure was assessed for its impact on renal distress as a sequela of platinum-based chemotherapy. Clinical transcriptome dataset-based evaluations demonstrated that levels of dysbiosis-responsive genes were elevated during renal distress, indicating pathological communications between gut and kidney. Experimentally, mucosal exposure to streptomycin aggravated platinum-induced renal tubular lesions in a mouse model. Moreover, antibiotic-induced dysbiosis increased susceptibility to gut mucosal inflammation, epithelial disruption, and bacterial exposure in response to cisplatin treatment. Further investigation of the luminal microbes indicated that antibiotic-induced dysbiosis promoted the dominance of Bacteroides species. Moreover, the functional assessment of dysbiotic microbiota predicted tryptophan metabolic pathways. In particular, dysbiosis-responsive Bacteroides acidifaciens was associated with the production of the uremic toxin indoxyl sulfate and renal injuries. The results of this study including bacterial community-based evaluations provide new predictive insights into the interorgan communications and interventions against dysbiosis-associated disorders.
Collapse
Affiliation(s)
- Navin Ray
- Laboratory of Mucosal Exposome and Biomodulation, Department of Integrative Biomedical Sciences, Pusan National University, Yangsan, South Korea
| | - Hoyoung Jeong
- Laboratory of Mucosal Exposome and Biomodulation, Department of Integrative Biomedical Sciences, Pusan National University, Yangsan, South Korea
- Graduate Program of Genomic Data Sciences, Pusan National University, Yangsan, South Korea
| | - Dasom Kwon
- Laboratory of Mucosal Exposome and Biomodulation, Department of Integrative Biomedical Sciences, Pusan National University, Yangsan, South Korea
| | - Juil Kim
- Laboratory of Mucosal Exposome and Biomodulation, Department of Integrative Biomedical Sciences, Pusan National University, Yangsan, South Korea
| | - Yuseok Moon
- Laboratory of Mucosal Exposome and Biomodulation, Department of Integrative Biomedical Sciences, Pusan National University, Yangsan, South Korea
- Graduate Program of Genomic Data Sciences, Pusan National University, Yangsan, South Korea
- *Correspondence: Yuseok Moon,
| |
Collapse
|
37
|
Madill-Thomsen KS, Abouljoud M, Bhati C, Ciszek M, Durlik M, Feng S, Foroncewicz B, Francis I, Grąt M, Jurczyk K, Klintmalm G, Krasnodębski M, McCaughan G, Miquel R, Montano-Loza A, Moonka D, Mucha K, Myślak M, Pączek L, Perkowska-Ptasińska A, Piecha G, Reichman T, Sanchez-Fueyo A, Tronina O, Wawrzynowicz-Syczewska M, Więcek A, Zieniewicz K, Halloran PF. The molecular phenotypes of injury, steatohepatitis, and fibrosis in liver transplant biopsies in the INTERLIVER study. Am J Transplant 2022; 22:909-926. [PMID: 34780106 DOI: 10.1111/ajt.16890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 10/19/2021] [Accepted: 10/21/2021] [Indexed: 01/25/2023]
Abstract
To extend previous molecular analyses of rejection in liver transplant biopsies in the INTERLIVER study (ClinicalTrials.gov #NCT03193151), the present study aimed to define the gene expression selective for parenchymal injury, fibrosis, and steatohepatitis. We analyzed genome-wide microarray measurements from 337 liver transplant biopsies from 13 centers. We examined expression of genes previously annotated as increased in injury and fibrosis using principal component analysis (PCA). PC1 reflected parenchymal injury and related inflammation in the early posttransplant period, slowly regressing over many months. PC2 separated early injury from late fibrosis. Positive PC3 identified a distinct mildly inflamed state correlating with histologic steatohepatitis. Injury PCs correlated with liver function and histologic abnormalities. A classifier trained on histologic steatohepatitis predicted histologic steatohepatitis with cross-validated AUC = 0.83, and was associated with pathways reflecting metabolic abnormalities distinct from fibrosis. PC2 predicted histologic fibrosis (AUC = 0.80), as did a molecular fibrosis classifier (AUC = 0.74). The fibrosis classifier correlated with matrix remodeling pathways with minimal overlap with those selective for steatohepatitis, although some biopsies had both. Genome-wide assessment of liver transplant biopsies can not only detect molecular changes induced by rejection but also those correlating with parenchymal injury, steatohepatitis, and fibrosis, offering potential insights into disease mechanisms for primary diseases.
Collapse
Affiliation(s)
| | | | - Chandra Bhati
- Virginia Commonwealth University, Richmond, Virginia, USA
| | - Michał Ciszek
- Department of Immunology, Transplantology and Internal Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Magdalena Durlik
- Department of Transplant Medicine, Nephrology and Internal Diseases, Medical University of Warsaw, Warsaw, Poland
| | - Sandy Feng
- University of California San Francisco, San Francisco, California, USA
| | - Bartosz Foroncewicz
- Department of Immunology, Transplantology and Internal Medicine, Medical University of Warsaw, Warsaw, Poland
| | | | - Michał Grąt
- Department of General, Transplant and Liver Surgery, Medical University of Warsaw, Warsaw, Poland
| | - Krzysztof Jurczyk
- Department of Infectious Diseases, Hepatology and Liver Transplantation, Pomeranian Medical University, Szczecin, Poland
| | | | - Maciej Krasnodębski
- Department of General, Transplant and Liver Surgery, Medical University of Warsaw, Warsaw, Poland
| | - Geoff McCaughan
- Centenary Research Institute, Australian National Liver Transplant Unit, Royal Prince Alfred Hospital, The University of Sydney, Sydney, New South Wales, Australia
| | | | | | | | - Krzysztof Mucha
- Department of Immunology, Transplantology and Internal Medicine, Medical University of Warsaw, Warsaw, Poland
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Marek Myślak
- Department of Clinical Interventions, Department of Nephrology and Kidney Transplantation SPWSZ Hospital, Pomeranian Medical University, Szczecin, Poland
| | - Leszek Pączek
- Department of Immunology, Transplantology and Internal Medicine, Medical University of Warsaw, Warsaw, Poland
| | | | - Grzegorz Piecha
- Department of Nephrology, Transplantation and Internal Medicine, Medical University of Silesia, Katowice, Poland
| | | | | | - Olga Tronina
- Department of Transplant Medicine, Nephrology and Internal Diseases, Medical University of Warsaw, Warsaw, Poland
| | - Marta Wawrzynowicz-Syczewska
- Department of Infectious Diseases, Hepatology and Liver Transplantation, Pomeranian Medical University, Szczecin, Poland
| | - Andrzej Więcek
- Department of Nephrology, Transplantation and Internal Medicine, Medical University of Silesia, Katowice, Poland
| | - Krzysztof Zieniewicz
- Department of General, Transplant and Liver Surgery, Medical University of Warsaw, Warsaw, Poland
| | | |
Collapse
|
38
|
Halloran PF, Reeve J, Madill-Thomsen KS, Demko Z, Prewett A, Billings P. The Trifecta Study: Comparing Plasma Levels of Donor-derived Cell-Free DNA with the Molecular Phenotype of Kidney Transplant Biopsies. J Am Soc Nephrol 2022; 33:387-400. [PMID: 35058354 PMCID: PMC8819982 DOI: 10.1681/asn.2021091191] [Citation(s) in RCA: 79] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 11/15/2021] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND The relationship between the donor-derived cell-free DNA fraction (dd-cfDNA[%]) in plasma in kidney transplant recipients at time of indication biopsy and gene expression in the biopsied allograft has not been defined. METHODS In the prospective, multicenter Trifecta study, we collected tissue from 300 biopsies from 289 kidney transplant recipients to compare genome-wide gene expression in biopsies with dd-cfDNA(%) in corresponding plasma samples drawn just before biopsy. Rejection was assessed with the microarray-based Molecular Microscope Diagnostic System using automatically assigned rejection archetypes and molecular report sign-outs, and histology assessments that followed Banff guidelines. RESULTS The median time of biopsy post-transplantation was 455 days (5 days to 32 years), with a case mix similar to that of previous studies: 180 (60%) no rejection, 89 (30%) antibody-mediated rejection (ABMR), and 31 (10%) T cell-mediated rejection (TCMR) and mixed. In genome-wide mRNA measurements, all 20 top probe sets correlating with dd-cfDNA(%) were previously annotated for association with ABMR and all types of rejection, either natural killer (NK) cell-expressed (e.g., GNLY, CCL4, TRDC, and S1PR5) or IFN-γ-inducible (e.g., PLA1A, IDO1, CXCL11, and WARS). Among gene set and classifier scores, dd-cfDNA(%) correlated very strongly with ABMR and all types of rejection, reasonably strongly with active TCMR, and weakly with inactive TCMR, kidney injury, and atrophy fibrosis. Active ABMR, mixed, and active TCMR had the highest dd-cfDNA(%), whereas dd-cfDNA(%) was lower in late-stage ABMR and less-active TCMR. By multivariate random forests and logistic regression, molecular rejection variables predicted dd-cfDNA(%) better than histologic variables. CONCLUSIONS The dd-cfDNA(%) at time of indication biopsy strongly correlates with active molecular rejection and has the potential to reduce unnecessary biopsies. CLINICAL TRIAL REGISTRATION NUMBER NCT04239703.
Collapse
Affiliation(s)
- Philip F. Halloran
- Alberta Transplant Applied Genomics Center, Edmonton, Canada,Department of Medicine, University of Alberta, Edmonton, Canada,Transcriptome Sciences Inc., Edmonton, Canada
| | - Jeff Reeve
- Alberta Transplant Applied Genomics Center, Edmonton, Canada
| | - Katelynn S. Madill-Thomsen
- Alberta Transplant Applied Genomics Center, Edmonton, Canada,Transcriptome Sciences Inc., Edmonton, Canada
| | | | | | | | | |
Collapse
|
39
|
Zhang Y, Zhao H, Su Q, Wang C, Chen H, Shen L, Ma L, Zhu T, Chen W, Jiang H, Chen J. Novel Plasma Biomarker-Based Model for Predicting Acute Kidney Injury After Cardiac Surgery: A Case Control Study. Front Med (Lausanne) 2022; 8:799516. [PMID: 35096889 PMCID: PMC8795513 DOI: 10.3389/fmed.2021.799516] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 12/10/2021] [Indexed: 12/16/2022] Open
Abstract
Introduction: Acute kidney injury (AKI) after cardiac surgery is independently associated with a prolonged hospital stay, increased cost of care, and increased post-operative mortality. Delayed elevation of serum creatinine (SCr) levels requires novel biomarkers to provide a prediction of AKI after cardiac surgery. Our objective was to find a novel blood biomarkers combination to construct a model for predicting AKI after cardiac surgery and risk stratification. Methods: This was a case-control study. Weighted Gene Co-expression Network Analysis (WGCNA) was applied to Gene Expression Omnibus (GEO) dataset GSE30718 to seek potential biomarkers associated with AKI. We measured biomarker levels in venous blood samples of 67 patients with AKI after cardiac surgery and 59 control patients in two cohorts. Clinical data were collected. We developed a multi-biomarker model for predicting cardiac-surgery-associated AKI and compared it with a traditional clinical-factor-based model. Results: From bioinformatics analysis and previous articles, we found 6 potential plasma biomarkers for the prediction of AKI. Among them, 3 biomarkers, such as growth differentiation factor 15 (GDF15), soluble suppression of tumorigenicity 2 (ST2, IL1RL1), and soluble urokinase plasminogen activator receptor (uPAR) were found to have prediction ability for AKI (area under the curve [AUC] > 0.6) in patients undergoing cardiac surgery. They were then incorporated into a multi-biomarker model for predicting AKI (C-statistic: 0.84, Brier 0.15) which outperformed the traditional clinical-factor-based model (C-statistic: 0.73, Brier 0.16). Conclusion: Our research validated a promising plasma multi-biomarker model for predicting AKI after cardiac surgery.
Collapse
Affiliation(s)
- Yichi Zhang
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.,Key Laboratory of Nephropathy, Hangzhou, China.,Institute of Nephropathy, Zhejiang University, Hangzhou, China.,Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, China
| | - Haige Zhao
- Department of Cardiothoracic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qun Su
- Department of Intensive Care Unit, The First Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Cuili Wang
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.,Key Laboratory of Nephropathy, Hangzhou, China.,Institute of Nephropathy, Zhejiang University, Hangzhou, China.,Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, China
| | - Hongjun Chen
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.,Key Laboratory of Nephropathy, Hangzhou, China.,Institute of Nephropathy, Zhejiang University, Hangzhou, China.,Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, China
| | - Lingling Shen
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.,Key Laboratory of Nephropathy, Hangzhou, China.,Institute of Nephropathy, Zhejiang University, Hangzhou, China.,Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, China
| | - Liang Ma
- Department of Cardiothoracic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Tingting Zhu
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.,Key Laboratory of Nephropathy, Hangzhou, China.,Institute of Nephropathy, Zhejiang University, Hangzhou, China.,Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, China
| | - Wenqing Chen
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.,Key Laboratory of Nephropathy, Hangzhou, China.,Institute of Nephropathy, Zhejiang University, Hangzhou, China.,Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, China
| | - Hong Jiang
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.,Key Laboratory of Nephropathy, Hangzhou, China.,Institute of Nephropathy, Zhejiang University, Hangzhou, China.,Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, China
| | - Jianghua Chen
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.,Key Laboratory of Nephropathy, Hangzhou, China.,Institute of Nephropathy, Zhejiang University, Hangzhou, China.,Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, China
| |
Collapse
|
40
|
Salem FE, Perin L, Sedrakyan S, Angeletti A, Ghiggeri G, Coccia MC, Ross M, Fribourg M, Cravedi P. The spatially resolved transcriptional profile of acute T cell-mediated rejection in a kidney allograft. Kidney Int 2022; 101:131-136. [PMID: 34555393 PMCID: PMC9387544 DOI: 10.1016/j.kint.2021.09.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 08/31/2021] [Accepted: 09/03/2021] [Indexed: 01/03/2023]
Abstract
Analysis of the transcriptional profile of graft biopsies represents a promising strategy to study T cell-mediated-rejection (TCMR), also known as acute cellular rejection. However, bulk RNA sequencing of graft biopsies may not capture the focal nature of acute rejection. Herein, we used the whole exome GeoMX Digital Space Profiling platform to study five tubular and three glomerular regions of interest in the kidney graft biopsy from a patient with a chronic-active TCMR episode and in analogous areas from two different normal kidney control biopsies. All kidney sections were from paraffin blocks. Overall, inflammatory genes were significantly upregulated in the tubular areas of the TCMR biopsy and showed an enrichment for gene-ontology terms associated with T-cell activation, differentiation, and proliferation. Enrichment analysis of the 100 genes with the highest coefficient of variation across the TCMR tubular regions of interest revealed that these highly variable genes are involved in kidney development and injury and interestingly do not associate with the 2019 Banff classification pathology scores within the individual regions of interest. Spatial transcriptomics allowed us to unravel a previously unappreciated variability across different areas of the TCMR biopsy related to the graft response to the alloimmune attack, rather than to the immune cells. Thus, our approach has the potential to decipher clinically relevant, new pathogenic mechanisms, and therapeutic targets in acute cellular rejection and other kidney diseases with a focal nature.
Collapse
Affiliation(s)
- Fadi E. Salem
- Department of Pathology & Molecular and Cell Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Laura Perin
- GOFARR Laboratory for Organ Regenerative Research and Cell Therapeutics in Urology, Saban Research Institute, Division of Urology, Children’s Hospital Los Angeles, Los Angeles, CA, USA
| | - Sargis Sedrakyan
- GOFARR Laboratory for Organ Regenerative Research and Cell Therapeutics in Urology, Saban Research Institute, Division of Urology, Children’s Hospital Los Angeles, Los Angeles, CA, USA
| | - Andrea Angeletti
- Nephrology, Dialysis and Transplantation Unit, Giannina Gaslini Scientific Institute for Research, Hospitalization and Healthcare, Genoa, Italy
| | - GianMarco Ghiggeri
- Nephrology, Dialysis and Transplantation Unit, Giannina Gaslini Scientific Institute for Research, Hospitalization and Healthcare, Genoa, Italy
| | - Maria Cristina Coccia
- Pathological Anatomy Unit, Giannina Gaslini Scientific Institute for Research, Hospitalization and Healthcare, Genoa, Italy
| | - Marty Ross
- NanoString Technologies Inc., Seattle, WA, USA
| | - Miguel Fribourg
- Department of Medicine, Translational Transplant Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Paolo Cravedi
- Department of Medicine, Translational Transplant Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
41
|
Fusfeld L, Menon S, Gupta G, Lawrence C, Masud SF, Goss TF. US payer budget impact of a microarray assay with machine learning to evaluate kidney transplant rejection in for-cause biopsies. J Med Econ 2022; 25:515-523. [PMID: 35345966 DOI: 10.1080/13696998.2022.2059221] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
AIM This study evaluates the economic impact to US commercial payers of MMDx-Kidney used in conjunction with histologic evaluation of for-cause kidney transplant biopsies. MATERIALS AND METHODS An Excel-based model was developed to assess the cost impact of histology plus MMDx-Kidney versus histology alone for the evaluation of potential rejection in kidney transplant patients who receive a for-cause biopsy. Different model time periods were assessed, ranging from 1 to 5 years post-biopsy. A targeted literature review was used to identify parameter estimates, validated by two external clinicians with expertise in managing kidney transplant rejection. A sensitivity analysis was conducted to evaluate the relative impact of key clinical and cost parameters. In particular, the model identified the magnitude of MMDx-Kidney's impact on graft failure from rejection that would be required for MMDx-Kidney to be cost-neutral. RESULTS By more accurately characterizing rejection, MMDx-Kidney is estimated to increase antirejection treatment costs by $1,126 per test. Nevertheless, a break-even analysis shows that the costs of MMDx-Kidney and anti-rejection medication, as well as the costs associated with an increase in the number of patients with functioning transplants, may be offset by reductions in costs associated with graft failure (i.e. costs of hospitalizations, dialysis, and repeat transplants) over 5 years, assuming MMDx-Kidney reduces annual graft failure from rejection by at least 5%. For the base case, with a 25% relative reduction in annual rate of graft failures from rejection, MMDx-Kidney increases overall costs incurred in the first year of the model but starts generating savings by the second year of the model. CONCLUSIONS Compared with histologic evaluation of for-cause kidney transplant biopsies alone, the use of MMDx-Kidney in conjunction with histologic evaluation improves the diagnoses of graft dysfunction and may have the potential to generate overall savings from reductions in rejection-related graft failure.
Collapse
Affiliation(s)
- Lauren Fusfeld
- Boston Healthcare Associates, Inc. (now a Veranex company), Boston, MA, USA
| | - Sreeranjani Menon
- Boston Healthcare Associates, Inc. (now a Veranex company), Boston, MA, USA
| | - Gaurav Gupta
- Division of Nephrology, Department of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | | | - Salwa F Masud
- Boston Healthcare Associates, Inc. (now a Veranex company), Boston, MA, USA
| | - Thomas F Goss
- Boston Healthcare Associates, Inc. (now a Veranex company), Boston, MA, USA
| |
Collapse
|
42
|
Pajenda S, Zawedde F, Kapps S, Wagner L, Schmidt A, Winnicki W, O’Connell D, Gerges D. Urinary C3 levels associated with sepsis and acute kidney injury-A pilot study. PLoS One 2021; 16:e0259777. [PMID: 34767613 PMCID: PMC8589214 DOI: 10.1371/journal.pone.0259777] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 10/18/2021] [Indexed: 12/01/2022] Open
Abstract
Acute kidney injury (AKI) is an abrupt deterioration of renal function often caused by severe clinical disease such as sepsis, and patients require intensive care. Acute-phase parameters for systemic inflammation are well established and used in routine clinical diagnosis, but no such parameters are known for AKI and inflammation at the local site of tissue damage, namely the nephron. Therefore, we sought to investigate complement factors C3a/C3 in urine and urinary sediment cells. After the development of a C3a/C3-specific mouse monoclonal antibody (3F7E2), urine excretion from ICU sepsis patients was examined by dot blot and immunoblotting. This C3a/C3 ELISA and a C3a ELISA were used to obtain quantitative data over 24 hours for 6 consecutive days. Urine sediment cells were analyzed for topology of expression. Patients with severe infections (n = 85) showed peak levels of C3a/C3 on the second day of ICU treatment. The majority (n = 59) showed C3a/C3 levels above 20 μg/ml at least once in the first 6 days after admission. C3a was detectable on all 6 days. Peak C3a/C3 levels correlated negatively with peak C-reactive protein (CRP) levels. No relationship was found between peak C3a/C3 with peak leukocyte count, age, or AKI stage. Analysis of urine sediment cells identified C3a/C3-producing epithelial cells with reticular staining patterns and cells with large-granular staining. Opsonized bacteria were detected in patients with urinary tract infections. In critically ill sepsis patients with AKI, urinary C3a/C3 inversely correlated with serum CRP. Whether urinary C3a/C3 has a protective function through autophagy, as previously shown for cisplatin exposure, or is a by-product of sepsis caused by pathogenic stimuli to the kidney must remain open in this study. However, our data suggest that C3a/C3 may function as an inverse acute-phase parameter that originates in the kidney and is detectable in urine.
Collapse
Affiliation(s)
- Sahra Pajenda
- Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Florence Zawedde
- Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Sebastian Kapps
- Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Ludwig Wagner
- Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Alice Schmidt
- Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Wolfgang Winnicki
- Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - David O’Connell
- School of Biomolecular and Biomedical Science, University College Dublin, Belfield, Dublin, Ireland
| | - Daniela Gerges
- Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
- * E-mail:
| |
Collapse
|
43
|
Halloran PF, Madill-Thomsen KS, Böhmig GA, Myslak M, Gupta G, Kumar D, Viklicky O, Perkowska-Ptasinska A, Famulski KS. A 2-fold Approach to Polyoma Virus (BK) Nephropathy in Kidney Transplants: Distinguishing Direct Virus Effects From Cognate T Cell-mediated Inflammation. Transplantation 2021; 105:2374-2384. [PMID: 34310102 DOI: 10.1097/tp.0000000000003884] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND BK nephropathy (BKN) in kidney transplants diagnosed by histology is challenging because it involves damage from both virus activity and cognate T cell-mediated inflammation, directed against alloantigens (rejection) or viral antigens. The present study of indication biopsies from the Integrated Diagnostic System in the International Collaborative Microarray Study Extension study measured major capsid viral protein 2 (VP2) mRNA to assess virus activity and a T cell-mediated rejection (TCMR) classifier to assess cognate T cell-mediated inflammation. METHODS Biopsies were assessed by local standard-of-care histology and by genome-wide microarrays and Molecular Microscope Diagnostic System (MMDx) algorithms to detect rejection and injury. In a subset of 102 biopsies (50 BKN and 52 BKN-negative biopsies with various abnormalities), we measured VP2 transcripts by real-time polymerase chain reaction. RESULTS BKN was diagnosed in 55 of 1679 biopsies; 30 had cognate T cell-mediated activity assessed by by MMDx and TCMR lesions, but only 3 of 30 were histologically diagnosed as TCMR. We developed a BKN probability classifier that predicted histologic BKN (area under the curve = 0.82). Virus activity (VP2 expression) was highly selective for BKN (area under the curve = 0.94) and correlated with acute injury, atrophy-fibrosis, macrophage activation, and the BKN classifier, but not with the TCMR classifier. BKN with molecular TCMR had more tubulitis and inflammation than BKN without molecular TCMR. In 5 BKN cases with second biopsies, VP2 mRNA decreased in second biopsies, whereas in 4 of 5 TCMR classifiers, scores increased. Genes and pathways associated with BKN and VP2 mRNA were similar, reflecting injury, inflammation, and macrophage activation but none was selective for BKN. CONCLUSIONS Risk-benefit decisions in BKN may be assisted by quantitative assessment of the 2 major pathologic processes, virus activity and cognate T cell-mediated inflammation.
Collapse
Affiliation(s)
- Philip F Halloran
- Alberta Transplant Applied Genomics Centre, Edmonton, AB, Canada
- Department of Medicine, Division of Nephrology and Transplant Immunology, University of Alberta, Edmonton, AB, Canada
| | | | - Georg A Böhmig
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Marek Myslak
- Department of Nephrology and Kidney Transplantation, SPWSZ Hospital in Szczecin, Pomeranian Medical University, Szczecin, Poland
| | - Gaurav Gupta
- Division of Nephrology, Virginia Commonwealth University, Richmond, VA
| | - Dhiren Kumar
- Division of Nephrology, Virginia Commonwealth University, Richmond, VA
| | - Ondrej Viklicky
- Department of Nephrology and Transplant Center, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | | | | |
Collapse
|
44
|
Guo X, Qi X, Fan P, Gilbert M, La AD, Liu Z, Bertz R, Kellum JA, Chen Y, Wang L. Effect of ondansetron on reducing ICU mortality in patients with acute kidney injury. Sci Rep 2021; 11:19409. [PMID: 34593872 PMCID: PMC8484575 DOI: 10.1038/s41598-021-98734-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 09/01/2021] [Indexed: 12/02/2022] Open
Abstract
The purpose of this study is to identify medications with potentially beneficial effects on decreasing mortality in patients with acute kidney injury (AKI) while in the intensive care unit (ICU). We used logistic regression to investigate associations between medications received and ICU mortality in patients with AKI in the MIMIC III database. Drugs associated with reduced mortality were then validated using the eICU database. Propensity score matching (PSM) was used for matching the patients’ baseline severity of illness followed by a chi-square test to calculate the significance of drug use and mortality. Finally, we examined gene expression signatures to explore the drug’s molecular mechanism on AKI. While several drugs demonstrated potential beneficial effects on reducing mortality, most were used for potentially fatal illnesses (e.g. antibiotics, cardiac medications). One exception was found, ondansetron, a drug without previously identified life-saving effects, has correlation with lower mortality among AKI patients. This association was confirmed in a subsequent analysis using the eICU database. Based on the comparison of gene expression signatures, the presumed therapeutic effect of ondansetron may be elicited through the NF-KB pathway and JAK-STAT pathway. Our findings provide real-world evidence to support clinical trials of ondansetron for treatment of AKI.
Collapse
Affiliation(s)
- Xiaojiang Guo
- Department of Pharmaceutical Sciences, Computational Chemical Genomics Screening Center, School of Pharmacy, University of Pittsburgh, 5607 Baum Boulevard, Pittsburgh, PA, 15206, USA
| | - Xiguang Qi
- Department of Pharmaceutical Sciences, Computational Chemical Genomics Screening Center, School of Pharmacy, University of Pittsburgh, 5607 Baum Boulevard, Pittsburgh, PA, 15206, USA
| | - Peihao Fan
- Department of Pharmaceutical Sciences, Computational Chemical Genomics Screening Center, School of Pharmacy, University of Pittsburgh, 5607 Baum Boulevard, Pittsburgh, PA, 15206, USA
| | - Michael Gilbert
- Department of Pharmacy and Therapeutics, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, 15206, USA
| | - Andrew D La
- Department of Pharmacy and Therapeutics, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, 15206, USA
| | - Zeyu Liu
- The Dietrich School of Arts & Sciences, University of Pittsburgh, Pittsburgh, PA, 15206, USA
| | - Richard Bertz
- Department of Pharmaceutical Sciences, Computational Chemical Genomics Screening Center, School of Pharmacy, University of Pittsburgh, 5607 Baum Boulevard, Pittsburgh, PA, 15206, USA
| | - John A Kellum
- The Center for Critical Care Nephrology Department of Critical Care Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15206, USA.
| | - Yu Chen
- Eli Lilly and Company, Lilly Corporate Center, Indiana, IN, 46225, USA.
| | - Lirong Wang
- Department of Pharmaceutical Sciences, Computational Chemical Genomics Screening Center, School of Pharmacy, University of Pittsburgh, 5607 Baum Boulevard, Pittsburgh, PA, 15206, USA.
| |
Collapse
|
45
|
Urbanellis P, McEvoy CM, Škrtić M, Kaths JM, Kollmann D, Linares I, Ganesh S, Oquendo F, Sharma M, Mazilescu L, Goto T, Noguchi Y, John R, Mucsi I, Ghanekar A, Bagli D, Konvalinka A, Selzner M, Robinson LA. Transcriptome Analysis of Kidney Grafts Subjected to Normothermic Ex Vivo Perfusion Demonstrates an Enrichment of Mitochondrial Metabolism Genes. Transplant Direct 2021; 7:e719. [PMID: 34258386 PMCID: PMC8270593 DOI: 10.1097/txd.0000000000001157] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 03/18/2021] [Accepted: 03/20/2021] [Indexed: 12/28/2022] Open
Abstract
Normothermic ex vivo kidney perfusion (NEVKP) has demonstrated superior outcomes for donation-after-cardiovascular death grafts compared with static cold storage (SCS). To determine the mechanisms responsible for this, we performed an unbiased genome-wide microarray analysis. METHODS Kidneys from 30-kg Yorkshire pigs were subjected to 30 min of warm ischemia followed by 8 h of NEVKP or SCS, or no storage, before autotransplantation. mRNA expression was analyzed on renal biopsies on postoperative day 3. Gene set enrichment analysis was performed using hallmark gene sets, Gene Ontology, and pathway analysis. RESULTS The gene expression profile of NEVKP-stored grafts closely resembled no storage kidneys. Gene set enrichment analysis demonstrated enrichment of fatty acid metabolism and oxidative phosphorylation following NEVKP, whereas SCS-enriched gene sets were related to mitosis, cell cycle checkpoint, and reactive oxygen species (q < 0.05). Pathway analysis demonstrated enrichment of lipid oxidation/metabolism, the Krebs cycle, and pyruvate metabolism in NEVKP compared with SCS (q < 0.05). Comparison of our findings with external data sets of renal ischemia-reperfusion injury revealed that SCS-stored grafts demonstrated similar gene expression profiles to ischemia-reperfusion injury, whereas the profile of NEVKP-stored grafts resembled recovered kidneys. CONCLUSIONS Increased transcripts of key mitochondrial metabolic pathways following NEVKP storage may account for improved donation-after-cardiovascular death graft function, compared with SCS, which promoted expression of genes typically perturbed during IRI.
Collapse
Affiliation(s)
- Peter Urbanellis
- Soham and Shaila Ajmera Family Transplant Centre, Toronto General Hospital, University Health Network, Toronto, ON, Canada
- Canadian Donation and Transplantation Research Program, Edmonton, AB, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Caitriona M. McEvoy
- Soham and Shaila Ajmera Family Transplant Centre, Toronto General Hospital, University Health Network, Toronto, ON, Canada
- Canadian Donation and Transplantation Research Program, Edmonton, AB, Canada
- Division of Nephrology, Department of Medicine, University Health Network, Toronto, ON, Canada
| | - Marko Škrtić
- Division of Nephrology, Department of Medicine, University of Toronto, Toronto, ON, Canada
- Program in Cell Biology, The Hospital for Sick Children Research Institute, Toronto, ON, Canada
| | - J. Moritz Kaths
- Soham and Shaila Ajmera Family Transplant Centre, Toronto General Hospital, University Health Network, Toronto, ON, Canada
- Canadian Donation and Transplantation Research Program, Edmonton, AB, Canada
| | - Dagmar Kollmann
- Soham and Shaila Ajmera Family Transplant Centre, Toronto General Hospital, University Health Network, Toronto, ON, Canada
| | - Ivan Linares
- Soham and Shaila Ajmera Family Transplant Centre, Toronto General Hospital, University Health Network, Toronto, ON, Canada
- Canadian Donation and Transplantation Research Program, Edmonton, AB, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Sujani Ganesh
- Soham and Shaila Ajmera Family Transplant Centre, Toronto General Hospital, University Health Network, Toronto, ON, Canada
| | - Fabiola Oquendo
- Soham and Shaila Ajmera Family Transplant Centre, Toronto General Hospital, University Health Network, Toronto, ON, Canada
| | - Manraj Sharma
- Soham and Shaila Ajmera Family Transplant Centre, Toronto General Hospital, University Health Network, Toronto, ON, Canada
| | - Laura Mazilescu
- Soham and Shaila Ajmera Family Transplant Centre, Toronto General Hospital, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Program in Cell Biology, The Hospital for Sick Children Research Institute, Toronto, ON, Canada
| | - Toru Goto
- Soham and Shaila Ajmera Family Transplant Centre, Toronto General Hospital, University Health Network, Toronto, ON, Canada
| | - Yuki Noguchi
- Soham and Shaila Ajmera Family Transplant Centre, Toronto General Hospital, University Health Network, Toronto, ON, Canada
| | - Rohan John
- Laboratory Medicine and Pathobiology, Toronto General Hospital, University of Toronto, Toronto, ON, Canada
| | - Istvan Mucsi
- Canadian Donation and Transplantation Research Program, Edmonton, AB, Canada
- Division of Nephrology, Department of Medicine, University Health Network, Toronto, ON, Canada
| | - Anand Ghanekar
- Soham and Shaila Ajmera Family Transplant Centre, Toronto General Hospital, University Health Network, Toronto, ON, Canada
| | - Darius Bagli
- Departments of Surgery (Urology) and Physiology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Ana Konvalinka
- Soham and Shaila Ajmera Family Transplant Centre, Toronto General Hospital, University Health Network, Toronto, ON, Canada
- Canadian Donation and Transplantation Research Program, Edmonton, AB, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Division of Nephrology, Department of Medicine, University Health Network, Toronto, ON, Canada
- Laboratory Medicine and Pathobiology, Toronto General Hospital, University of Toronto, Toronto, ON, Canada
| | - Markus Selzner
- Soham and Shaila Ajmera Family Transplant Centre, Toronto General Hospital, University Health Network, Toronto, ON, Canada
- Canadian Donation and Transplantation Research Program, Edmonton, AB, Canada
| | - Lisa A. Robinson
- Program in Cell Biology, The Hospital for Sick Children Research Institute, Toronto, ON, Canada
- Division of Nephrology, The Hospital for Sick Children, Toronto, ON, Canada
| |
Collapse
|
46
|
Impact of Belatacept Conversion on Renal Function, Histology, and Gene Expression in Kidney Transplant Patients With Chronic Active Antibody-mediated Rejection. Transplantation 2021; 105:660-667. [PMID: 32510913 DOI: 10.1097/tp.0000000000003278] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
BACKGROUND Here, we present our initial experience with a prospective protocol of belatacept conversion in patients with chronic active antibody-mediated rejection (caAMR) and a high degree of chronicity at the time of diagnosis. METHODS We converted 19 patients (mean age, 45 ± 12 y) with biopsy-proven caAMR from tacrolimus to belatacept at a median of 44 months post-kidney transplant. RESULTS At a median of 29 months (interquartile range, 16-46 mo) postconversion, death-censored graft and patient survivals were 89% and 95%, respectively. When compared to a 1:2 propensity-matched control cohort from the INSERM U970 registry maintained on calcineurin inhibitor, the belatacept group had progressive improvement (P = 0.02) in estimated glomerular filtration rate from a mean of 33.9 ± 10 at baseline to 37.8 ± 13 at 6 months and 38.5 ± 12 mL/min/1.73 m2 at 12 months postconversion, as compared to a steady decline noted in the controls (36.2 [baseline] → 33.1 [6 mo] → 32.7 mL/min/1.73 m2 [12 mo] of follow-up). A paired histologic comparison of preconversion and postconversion (performed at median 9.5 mo postconversion) biopsies showed no worsening in microvascular inflammation or chronicity. The paired tissue gene expression analysis showed improved mean total rejection score (0.68 ± 0.26-0.56 ± 0.33; P = 0.02) and a trend toward improved antibody-mediated rejection score (0.64 ± 0.34-0.56 ± 0.39; P = 0.06). CONCLUSIONS Here, we report that in patients diagnosed with caAMR who were not subjected to intensive salvage immunosuppressive therapies, isolated belatacept conversion alone was associated with stabilization in renal function. These results are bolstered by molecular evidence of improved inflammation.
Collapse
|
47
|
Ide S, Kobayashi Y, Ide K, Strausser SA, Abe K, Herbek S, O'Brien LL, Crowley SD, Barisoni L, Tata A, Tata PR, Souma T. Ferroptotic stress promotes the accumulation of pro-inflammatory proximal tubular cells in maladaptive renal repair. eLife 2021; 10:68603. [PMID: 34279220 PMCID: PMC8318592 DOI: 10.7554/elife.68603] [Citation(s) in RCA: 105] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Accepted: 07/17/2021] [Indexed: 12/14/2022] Open
Abstract
Overwhelming lipid peroxidation induces ferroptotic stress and ferroptosis, a non-apoptotic form of regulated cell death that has been implicated in maladaptive renal repair in mice and humans. Using single-cell transcriptomic and mouse genetic approaches, we show that proximal tubular (PT) cells develop a molecularly distinct, pro-inflammatory state following injury. While these inflammatory PT cells transiently appear after mild injury and return to their original state without inducing fibrosis, after severe injury they accumulate and contribute to persistent inflammation. This transient inflammatory PT state significantly downregulates glutathione metabolism genes, making the cells vulnerable to ferroptotic stress. Genetic induction of high ferroptotic stress in these cells after mild injury leads to the accumulation of the inflammatory PT cells, enhancing inflammation and fibrosis. Our study broadens the roles of ferroptotic stress from being a trigger of regulated cell death to include the promotion and accumulation of proinflammatory cells that underlie maladaptive repair.
Collapse
Affiliation(s)
- Shintaro Ide
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, United States
| | - Yoshihiko Kobayashi
- Department of Cell Biology, Duke University School of Medicine, Durham, United States
| | - Kana Ide
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, United States
| | - Sarah A Strausser
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, United States
| | - Koki Abe
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, United States
| | - Savannah Herbek
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, United States
| | - Lori L O'Brien
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, United States
| | - Steven D Crowley
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, United States
| | - Laura Barisoni
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, United States.,Department of Pathology, Duke University School of Medicine, Durham, United States
| | - Aleksandra Tata
- Department of Cell Biology, Duke University School of Medicine, Durham, United States
| | - Purushothama Rao Tata
- Department of Cell Biology, Duke University School of Medicine, Durham, United States.,Regeneration Next, Duke University, Durham, United States.,Duke Cancer Institute, Duke University School of Medicine, Durham, United States
| | - Tomokazu Souma
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, United States.,Regeneration Next, Duke University, Durham, United States
| |
Collapse
|
48
|
Correlation of Donor-Derived Cell-free DNA with Histology and Molecular Diagnoses of Kidney Transplant Biopsies. Transplantation 2021; 106:1061-1070. [PMID: 34075006 DOI: 10.1097/tp.0000000000003838] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Circulating donor-derived cell free DNA (cfDNA), a minimally invasive diagnostic tool for kidney transplant rejection, was validated using traditional histology. The Molecular Microscope (MMDx) tissue gene expression platform may provide increased precision to traditional histology. METHODS In this single-center prospective study of 208 biopsies (median=5.8 months) post-transplant, we report on the calibration of cfDNA with simultaneous biopsy assessments using MMDx and histology by Area under the curve (AUC) analyses for optimal criterion, as well as for, previously published cfDNA cut-offs ≤0.21% to 'rule-out' rejection and ≥1% to 'rule-in' rejection. RESULTS There were significant discrepancies between histology and MMDx, with MMDx identifying more antibody-mediated rejection (65; 31%) than histology (43; 21%); the opposite was true for T-cell mediated rejection [TCMR; histology: 27 (13%) vs MMDx: 13 (6%)]. Most of the TCMR discrepancies were seen for histologic borderline/1A TCMR. AUC Curves for cfDNA and prediction of rejection were slightly better with MMDx (AUC=0.80; 95%CI: 0.74-0.86) vs. histology (AUC=0.75; 95%CI: 0.69-0.81). A cfDNA≤0.21% had similar sensitivity (~91%) to 'rule-out' rejection by histology and MMDx. Specificity was slightly higher with MMDx (92%) compared with histology (85%) to 'rule-in' rejection using cfDNA criterion≥1%. Strong positive quantitative correlations were observed between cfDNA scores and molecular acute kidney injury (AKI) for both 'rejection' and 'nonrejection' biopsies. CONCLUSIONS Molecular diagnostics using tissue gene expression and blood-based donor-derived cell-free DNA may add precision to some cases of traditional histology. The positive correlation of cfDNA with molecular AKI suggests a dose-dependent association with tissue injury irrespective of rejection characteristics.
Collapse
|
49
|
Halloran PF, Böhmig GA, Bromberg JS, Budde K, Gupta G, Einecke G, Eskandary F, Madill-Thomsen K, Reeve J. Discovering novel injury features in kidney transplant biopsies associated with TCMR and donor aging. Am J Transplant 2021; 21:1725-1739. [PMID: 33107191 DOI: 10.1111/ajt.16374] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 09/27/2020] [Accepted: 10/19/2020] [Indexed: 01/25/2023]
Abstract
We previously characterized the molecular changes in acute kidney injury (AKI) and chronic kidney disease (CKD) in kidney transplant biopsies, but parenchymal changes selective for specific types of injury could be missed by such analyses. The present study searched for injury changes beyond AKI and CKD related to specific scenarios, including correlations with donor age. We defined injury using previously defined gene sets and classifiers and used principal component analysis to discover new injury dimensions. As expected, Dimension 1 distinguished normal vs. injury, and Dimension 2 separated early AKI from late CKD, correlating with time posttransplant. However, Dimension 3 was novel, distinguishing a set of genes related to epithelial polarity (e.g., PARD3) that were increased in early AKI and decreased in T cell-mediated rejection (TCMR) but not in antibody-mediated rejection. Dimension 3 was increased in kidneys from older donors and was particularly important in survival of early kidneys. Thus high Dimension 3 scores emerge as a previously unknown element in the kidney response-to-injury that affects epithelial polarity genes and is increased in AKI but depressed in TCMR, indicating that in addition to general injury elements, certain injury elements are selective for specific pathologic mechanisms. (ClinicalTrials.gov NCT01299168).
Collapse
Affiliation(s)
- Philip F Halloran
- Alberta Transplant Applied Genomics Centre, Edmonton, Alberta, Canada.,Department of Medicine, Division of Nephrology and Transplant Immunology, University of Alberta, Edmonton, Alberta, Canada
| | - Georg A Böhmig
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | | | - Klemens Budde
- Charite-Medical University of Berlin, Berlin, Germany
| | - Gaurav Gupta
- Division of Nephrology, Virginia Commonwealth University, Richmond, Virginia
| | | | - Farsad Eskandary
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | | | - Jeff Reeve
- Alberta Transplant Applied Genomics Centre, Edmonton, Alberta, Canada
| | | |
Collapse
|
50
|
Tang Y, Yang X, Shu H, Yu Y, Pan S, Xu J, Shang Y. Bioinformatic analysis identifies potential biomarkers and therapeutic targets of septic-shock-associated acute kidney injury. Hereditas 2021; 158:13. [PMID: 33863396 PMCID: PMC8052759 DOI: 10.1186/s41065-021-00176-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 03/30/2021] [Indexed: 01/22/2023] Open
Abstract
Background Sepsis and septic shock are life-threatening diseases with high mortality rate in intensive care unit (ICU). Acute kidney injury (AKI) is a common complication of sepsis, and its occurrence is a poor prognostic sign to septic patients. We analyzed co-differentially expressed genes (co-DEGs) to explore relationships between septic shock and AKI and reveal potential biomarkers and therapeutic targets of septic-shock-associated AKI (SSAKI). Methods Two gene expression datasets (GSE30718 and GSE57065) were downloaded from the Gene Expression Omnibus (GEO). The GSE57065 dataset included 28 septic shock patients and 25 healthy volunteers and blood samples were collected within 0.5, 24 and 48 h after shock. Specimens of GSE30718 were collected from 26 patients with AKI and 11 control patents. AKI-DEGs and septic-shock-DEGs were identified using the two datasets. Subsequently, Gene Ontology (GO) functional analysis, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis, and protein-protein interaction (PPI) network analysis were performed to elucidate molecular mechanisms of DEGs. We also evaluated co-DEGs and corresponding predicted miRNAs involved in septic shock and AKI. Results We identified 62 DEGs in AKI specimens and 888, 870, and 717 DEGs in septic shock blood samples within 0.5, 24 and 48 h, respectively. The hub genes of EGF and OLFM4 may be involved in AKI and QPCT, CKAP4, PRKCQ, PLAC8, PRC1, BCL9L, ATP11B, KLHL2, LDLRAP1, NDUFAF1, IFIT2, CSF1R, HGF, NRN1, GZMB, and STAT4 may be associated with septic shock. Besides, co-DEGs of VMP1, SLPI, PTX3, TIMP1, OLFM4, LCN2, and S100A9 coupled with corresponding predicted miRNAs, especially miR-29b-3p, miR-152-3p, and miR-223-3p may be regarded as promising targets for the diagnosis and treatment of SSAKI in the future. Conclusions Septic shock and AKI are related and VMP1, SLPI, PTX3, TIMP1, OLFM4, LCN2, and S100A9 genes are significantly associated with novel biomarkers involved in the occurrence and development of SSAKI. Supplementary Information The online version contains supplementary material available at 10.1186/s41065-021-00176-y.
Collapse
Affiliation(s)
- Yun Tang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1277, Jiefang Avenue, Wuhan, 430022, China
| | - Xiaobo Yang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1277, Jiefang Avenue, Wuhan, 430022, China
| | - Huaqing Shu
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1277, Jiefang Avenue, Wuhan, 430022, China
| | - Yuan Yu
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1277, Jiefang Avenue, Wuhan, 430022, China
| | - Shangwen Pan
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1277, Jiefang Avenue, Wuhan, 430022, China
| | - Jiqian Xu
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1277, Jiefang Avenue, Wuhan, 430022, China
| | - You Shang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1277, Jiefang Avenue, Wuhan, 430022, China.
| |
Collapse
|