Submitted:
31 December 2024
Posted:
03 January 2025
You are already at the latest version
Abstract
Keywords:
1. Introduction
2. Rejection: Definition and Types
2.1. Risk Factors for Rejection
2.2. Types of Kidney Transplantation with Increased Immunological Risk
2.2.1. DSAs - Incompatible Transplantation
2.2.1.1. HLA Epitopes Definition and Classification
2.2.1.2. Characteristics of Donor-Specific Antibodies Associated with Pathogenicity
2.2.2. ABO Incompatible Kidney Transplantation
2.2.2.1. Complications of ABO-Incompatible Kidney Transplantation
2.2.3. HLA Incompatible Kidney Transplantation
2.2.3.1. Alloantibody Detection Tests Include
3. Biomarkers of Graft Dysfunction
3.1. Biomarkers of Acute Rejection [5,8,29]
| Biomarker | Method of assessment | Pathogenic Characteristics/Function | Clinical Relevance |
|---|---|---|---|
| Donor-derived cell-free DNA (dd-cfDNA) | Quantitative PCR (qPCR), Digital Droplet PCR (ddPCR)*, Next-Generation Sequencing (NGS)*, Targeted NGS Panels, Fragmentation Analysis (Fragmentomics), Methylation-Specific Methods, Mass Spectrometry-Based Methods, Immunoassay-Based Methods, Microarray-Based Techniques | Fragments of DNA from apoptotic donor cells, originating from the transplanted kidney, released into the recipient's bloodstream and initiate immune reactions | Graft injury, AMR and TCMR |
|
DSAs anti-HLA class I anti-HLA class II |
Luminex Single Antigen Bead (SAB) Assay*, Flow cytometry crossmatch*, Complement-dependent cytotoxicity (CDC) Assay*, C1q Binding Assay, IgG Subclass Analysis, ELISA-Based Methods, Endothelial Cell Crossmatch |
Bind to donor HLA class I and II molecules on endothelial cells, activate classical complement pathway | Acute and Chronic AMR |
| Anti-MICA (MHC Class I-Related Chain A) Antibodies | Luminex-Based Assays*, ELISA*, Flow Cytometry, CDC Assay, Multiplex Immunoassays, Western Blot | Activate T lymphocytes and NK cells, leading to endothelial cell injury | Acute and Chronic AMR |
| Anti-AT1R (Angiotensin II Type 1 Receptor) Antibodies | Luminex-Based Assays, ELISA*, Cell-Based assays, Surface Plasmon Resonance (SPR), Multiplex Immunoassays | Immune and Inflammatory responses, Vasoconstriction, Vascular injury, Hypertension | Acute and Chronic AMR, Chronic allograft dysfunction, Fibrosis |
| Anti-VEGF (Vascular Endothelial Growth Factor) Antibodies | ELISA*, Surface Plasmon Resonance, Radioimmunoassay, Flow Cytometry, Functional Neutralization Assays, Multiplex Immunoassays, Western Blot | Retard endothelial repair and angiogenesis | Acute and Chronic AMR, Impaired vascular repair, Chronic allograft dysfunction, Fibrosis |
| Non-HLA Autoantibodies | ELISA*, Luminex-Based Assays*, Flow Cytometry, Western Blot, Immunoprecipitation, Functional Neutralization Assays, Surface Plasmon Resonance (SPR), Multiplex Immunohistochemistry or Immunofluorescence, Next-Generation Sequencing (NGS)-Based Approaches | chronic inflammation, classical Complement activation | Acute and Chronic AMR |
| Anti-C4d Antibodies | ELISA*, Flow Cytometry*, Solid-Phase Assays (SPA), Complement-Dependent Cytotoxicity (CDC) Crossmatch, Flow Cytometric Crossmatch | ongoing complement activation | Acute and Chronic AMR |
| Gene expression profiles (GEP) in peripheral blood | Commercially Available GEP TestsAlloMap (CareDx) and TruGraf (Transplant Genomics)*, Microarray analysis, Quantitative Real-Time PCR, Next-Generation Sequencing, Multiplex PCR Panels, Digital PCR | T-cell and B-cell activation | Early phases of acute rejection |
| iATP levels | ELISA*, Western Blotting, Flow Cytometry, RIA, Immunoprecipitation, Solid-Phase Assays (SPA) | Ischemic injury, mitochondrial dysfunction, inflammation, tubular injury | Increased levels suggest acute rejection, reduced levels suggest infection |
| Donor-specific IFN-gamma-producing lymphocytes | ELISPOT assay*, ELISA*, Lymphocyte Transformation Test (LTT)*, Flow Cytometry, Cytotoxicity Assays, Multiplex Bead-Assays | immune activation, immunologic memory, vascular damge, fibrosis | Increased levels predict acute rejection |
| sCD30 | ELISA*, Western Blotting, Flow Cytometry, Immunoprecipitation AssayCytotoxicity Assays, Multiplex Bead-Assays | a glycoprotein expressed on human CD4 + and CD8 + T cells that secrete Th2-type cytokines | reflects recipients who may generate an alloimmune response against a grafted kidney |
| Donor-specific IFN-gamma-producing lymphocytes | ELISA*, LTT*, IFN-gamma enzyme-linked immunospot (ELISPOT) assay*, Flow Cytometry, Cytotoxicity Assays, Multiplex Bead assay, Solid phase assays | Direct cytotoxicity, endothelial injury, activation of macrophages and dendritic cells, expression of MHC class I and II | reflect immunologic memory and correlate with the risk of post-transplant rejection episodes |
| Torque Tenovirus (TTV) | ELISA*, Western Blotting, Immunofluoresence assay, PCR for TTV DNA, Imunoprecipitation, Multiplex Bead assay | TTV viral load in peripheral blood might reflect the intensity of host immunosuppression | Active immune responses |
| Plasma endothelial microparticles | ELISA*, Flow cytometry*, Luminex-Based Assays, Western blotting, immunoelectron microscopy, ELISPOT assays | endothelial dysfunction | Early phases of AMR |
| Titin, lipopolysaccharide-binding protein, peptidase inhibitor 16, complement factor D, mannose-binding lectin, protein Z-dependent protease, 2 -microglobulin | ELISA*, Luminex-Based Assays*, Western Blot, Flow Cytometry | complement activation, dysregulation of coangulation, chronic inflamation leading to fibrosis | Proteins Increased in AR |
| Kininogen-1, afamin, serine protease inhibitor, phosphatidylcholine-sterol acyltransferase, and sex hormone-binding globulin | ELISA*, Luminex-Based Assays*, Western Blot, Flow Cytometry | reduce oxidative stress, promote anti-inflammatory and vasoprotective effects, reduce inflammation | Proteins Decreased in AR |
| Mitochondrial DNA (mtDNA) | ELISA*, Immunofluorescence Assay (IFA)*, Luminex-Based Assays, Western Blot, , Immunoprecipitation assay, RIA | direct cellular injury, cytokine production, vascular injury, fibrosis | AR, vascular injury, and chronic graft dysfunction |
| Anti-LG3 (Perlecan Fragment) Antibodies | ELISA*, Luminex-Based Assays*, Western Blot, Immunofluorescence Assay (IFA), Flow Cytometry, Functional Assays | amplify complement activation | AR, thrombotic microangiopathy, chronic graft dysfunction, microvascular inflamation, |
| Anti-Endothelial Cell Antibodies (AECA) | Flow Cytometry*, ELISA*, Immunoprecipitation assay, Western Blotting, Luminex-Based assays | endothelial activation | AR, chronic graft dysfunction, microvascular inflamation, |
| MicroRNAs (e.g., miR-21, miR-155) | ELISA*, RNA immunoprecipitation*, Western Blotting,RT-qPCR, Multiplex assay | Non-coding RNAs regulating gene expression in immune and inflammatory pathways. | Altered expression patterns correlate with acute rejection. |
| TIM-3 | ELISA*, Flow Cytometry*, Multiplex-Bead assays*, Western Blotting | Immune checkpoint protein regulating T-cell activation. Elevated levels indicate failed immune regulation. | Increased levels predict acute rejection and immune activation. |
| Perforin | ELISA*, Flow Cytometry*, Multiplex-Bead assays*, Western Blotting | Protein secreted by T-cells and NK cells; cytotoxic activity | Increased levels indicate acute rejection. |
| Granzyme B | ELISA*, Flow Cytometry*, Multiplex-Bead assays*, Western Blotting | Protein secreted by T-cells and NK cells; cytotoxic activity and apoptosis | Increased levels are associated with T-cell-mediated acute rejection. |
| CXCL9 and CXCL10 (Monokine induced by gamma-interferon and Interferon-inducible protein-10) | ELISA*, Multiplex-Bead assays*, Flow Cytometry, Western Blotting, RIA | Stimulate T-cell recruitment to the kidney graft during rejection. | Increased levels in urine predict acute rejection |
| Pro-inflammatory Cytokines (e.g., IL-6 IL-2, IL-17, TNF-a) | ELISA*, Flow Cytometry*, Multiplex-Bead assays*, Western Blotting | Pro-inflammatory Cytokines (e.g., IL-6 IL-2, IL-17, TNF-a) | Active immune responses, acute rejection |
3.1.1. Biomarkers of Acute and Chronic Rejection Detected in Peripheral Blood Samples
3.1.2. Urinary Biomarkers: They Can Provide Real-Time Information About Kidney Function. However, They Can Degrade Rapidly After Collection and Standardization Is Necessary [2,8,28,42]
3.2. Biomarkers in Renal Transplantation Ischemia Reperfusion Injury and Delayed Graft Function
3.3. Other Types of Biomarkers [58]
3.3.1. Genes Overexpressed in the Common Rejection Module.
3.3.2. Transcriptomic Biomarkers
3.3.3. Epigenetic Biomarkers
3.3.4. Proteomic Biomarkers
3.3.5. Metabolomic Biomarkers
3.3.6. Cellular Biomarkers
4. Conclusions
Author Contributions
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- J. D. Schold, L. D. Buccini, D. A. Goldfarb, S. M. Flechner, E. D. Poggio, and A. R. Sehgal, ‘Association between Kidney Transplant Center Performance and the Survival Benefit of Transplantation Versus Dialysis’, Clin. J. Am. Soc. Nephrol., vol. 9, no. 10, pp. 1773–1780, Oct. 2014. [CrossRef]
- M. Salvadori and A. Tsalouchos, ‘Biomarkers in renal transplantation: An updated review’, World J. Transplant., vol. 7, no. 3, p. 161, 2017. [CrossRef]
- C. G. Rabbat, K. E. Thorpe, J. D. Russell, and D. N. Churchill, ‘Comparison of Mortality Risk for Dialysis Patients and Cadaveric First Renal Transplant Recipients in Ontario, Canada’, J. Am. Soc. Nephrol., vol. 11, no. 5, pp. 917–922, May 2000. [CrossRef]
- G. G. Garcia, P. Harden, and J. Chapman, ‘The global role of kidney transplantation’:, Curr. Opin. Organ Transplant., vol. 17, no. 4, pp. 362–367, Aug. 2012. [CrossRef]
- M. Naesens, J. Friedewald, V. Mas, B. Kaplan, and M. M. Abecassis, ‘A Practical Guide to the Clinical Implementation of Biomarkers for Subclinical Rejection Following Kidney Transplantation’, Transplantation, vol. 104, no. 4, pp. 700–707, Apr. 2020. [CrossRef]
- ‘Biomarkers and surrogate endpoints: Preferred definitions and conceptual framework’, Clin. Pharmacol. Ther., vol. 69, no. 3, pp. 89–95, Mar. 2001. [CrossRef]
- R. M. Califf, ‘Biomarker definitions and their applications’, Exp. Biol. Med., vol. 243, no. 3, pp. 213–221, Feb. 2018. [CrossRef]
- A. Kataria, A. Athreya, and G. Gupta, ‘Biomarkers in Kidney Transplantation’, Adv. Kidney Dis. Health, vol. 31, no. 5, pp. 427–435, Sep. 2024. [CrossRef]
- J. E. Cooper, ‘Evaluation and Treatment of Acute Rejection in Kidney Allografts’, Clin. J. Am. Soc. Nephrol., vol. 15, no. 3, pp. 430–438, Mar. 2020. [CrossRef]
- N. Singh, H. Samant, A. Hawxby, and M. D. Samaniego, ‘Biomarkers of rejection in kidney transplantation’, Curr. Opin. Organ Transplant., vol. 24, no. 1, pp. 103–110, Feb. 2019. [CrossRef]
- M. González-Molina et al., ‘Immune response and histology of humoral rejection in kidney transplantation’, Nefrología, vol. 36, no. 4, pp. 354–367, Jul. 2016. [CrossRef]
- M. A. Rafiq et al., ‘Differential outcomes in 3 types of acute antibody-mediated rejection’, Clin. Transplant., vol. 23, no. 6, pp. 951–957, Nov. 2009. [CrossRef]
- A. Djamali, D. B. Kaufman, T. M. Ellis, W. Zhong, A. Matas, and M. Samaniego, ‘Diagnosis and Management of Antibody-Mediated Rejection: Current Status and Novel Approaches’, Am. J. Transplant., vol. 14, no. 2, pp. 255–271, Feb. 2014. [CrossRef]
- A. Loupy et al., ‘The Banff 2019 Kidney Meeting Report (I): Updates on and clarification of criteria for T cell– and antibody-mediated rejection’, Am. J. Transplant., vol. 20, no. 9, pp. 2318–2331, Sep. 2020. [CrossRef]
- G. Agarwal, C. D. Diskin, T. A. Williams, and V. Kumar, ‘Late Antibody-Mediated Rejection in Kidney Transplant Recipients: Outcomes after Intravenous Immunoglobulin Therapy’, Clin. Transpl., vol. 32, pp. 111–118, 2016.
- J. L. Platt, G. M. Vercellotti, B. J. Lindman, T. R. Oegema, F. H. Bach, and A. P. Dalmasso, ‘Release of heparan sulfate from endothelial cells. Implications for pathogenesis of hyperacute rejection.’, J. Exp. Med., vol. 171, no. 4, pp. 1363–1368, Apr. 1990. [CrossRef]
- B. Sis et al., ‘Banff ’09 Meeting Report: Antibody Mediated Graft Deterioration and Implementation of Banff Working Groups’, Am. J. Transplant., vol. 10, no. 3, pp. 464–471, Mar. 2010. [CrossRef]
- T. Shimizu, T. Tanabe, H. Shirakawa, K. Omoto, H. Ishida, and K. Tanabe, ‘Acute vascular rejection after renal transplantation and isolated v-lesion’, Clin. Transplant., vol. 26, no. s24, pp. 2–8, Jul. 2012. [CrossRef]
- S. C. Jordan and A. A. Vo, ‘Donor-specific antibodies in allograft recipients: etiology, impact and therapeutic approaches’, Curr. Opin. Organ Transplant., vol. 19, no. 6, pp. 591–597, Dec. 2014. [CrossRef]
- J. Callemeyn, B. Lamarthée, A. Koenig, P. Koshy, O. Thaunat, and M. Naesens, ‘Allorecognition and the spectrum of kidney transplant rejection’, Kidney Int., vol. 101, no. 4, pp. 692–710, Apr. 2022. [CrossRef]
- S. Lionaki, K. Panagiotellis, A. Iniotaki, and J. N. Boletis, ‘Incidence and Clinical Significance of De Novo Donor Specific Antibodies after Kidney Transplantation’, Clin. Dev. Immunol., vol. 2013, pp. 1–9, 2013. [CrossRef]
- M. Mohamed et al., ‘ABO incompatibility in renal transplantation’, World J. Transplant., vol. 11, no. 9, pp. 388–399, Sep. 2021. [CrossRef]
- M. E. Breimer et al., ‘Blood group ABO-incompatible kidney transplantation biochemical and immunochemical studies of blood group A glycolipid antigens in human kidney and characterization of the antibody response (antigen specificity and antibody class) in O recipients receiving A2 grafts’, Transplant. Proc., vol. 19, no. 1 Pt 1, pp. 226–230, Feb. 1987.
- C. Morath, M. Zeier, B. Döhler, G. Opelz, and C. Süsal, ‘ABO-Incompatible Kidney Transplantation’, Front. Immunol., vol. 8, Mar. 2017. [CrossRef]
- S. Sethi, J. Choi, M. Toyoda, A. Vo, A. Peng, and S. C. Jordan, ‘Desensitization: Overcoming the Immunologic Barriers to Transplantation’, J. Immunol. Res., vol. 2017, pp. 1–11, 2017. [CrossRef]
- Y. Park, E. J. Ko, B. H. Chung, and C. W. Yang, ‘Kidney transplantation in highly sensitized recipients’, Kidney Res. Clin. Pract., vol. 40, no. 3, pp. 355–370, Sep. 2021. [CrossRef]
- R. A. Montgomery, D. S. Warren, D. L. Segev, and A. A. Zachary, ‘HLA incompatible renal transplantation’:, Curr. Opin. Organ Transplant., vol. 17, no. 4, pp. 386–392, Aug. 2012. [CrossRef]
- M. Quaglia, G. Merlotti, G. Guglielmetti, G. Castellano, and V. Cantaluppi, ‘Recent Advances on Biomarkers of Early and Late Kidney Graft Dysfunction’, Int. J. Mol. Sci., vol. 21, no. 15, p. 5404, Jul. 2020. [CrossRef]
- J. C. Cicciarelli et al., ‘Renal Transplant Patients Biopsied for Cause and Tested for C4d, DSA, and IgG Subclasses and C1q: Which Humoral Markers Improve Diagnosis and Outcomes?’, J. Immunol. Res., vol. 2017, pp. 1–14, 2017. [CrossRef]
- R. D. Bloom et al., ‘Cell-Free DNA and Active Rejection in Kidney Allografts’, J. Am. Soc. Nephrol., vol. 28, no. 7, pp. 2221–2232, Jul. 2017. [CrossRef]
- B. D. Tait et al., ‘Consensus Guidelines on the Testing and Clinical Management Issues Associated With HLA and Non-HLA Antibodies in Transplantation’, Transplantation, vol. 95, no. 1, pp. 19–47, Jan. 2013. [CrossRef]
- C. Lefaucheur et al., ‘Clinical recommendations for posttransplant assessment of anti–HLA (Human Leukocyte Antigen) donor-specific antibodies: A Sensitization in Transplantation: Assessment of Risk consensus document’, Am. J. Transplant., vol. 23, no. 1, pp. 115–132, Jan. 2023. [CrossRef]
- M. Risti and M. D. G. Bicalho, ‘MICA and NKG2D: Is There an Impact on Kidney Transplant Outcome?’, Front. Immunol., vol. 8, Feb. 2017. [CrossRef]
- R. Carapito et al., ‘The MHC class I MICA gene is a histocompatibility antigen in kidney transplantation’, Nat. Med., vol. 28, no. 5, pp. 989–998, May 2022. [CrossRef]
- Z.-Y. Kang, C. Liu, W. Liu, and D.-H. Li, ‘Effect of anti-angiotensin II type 1 receptor antibodies on the outcomes of kidney transplantation: a systematic review and meta-analysis’, Nephrol. Dial. Transplant., vol. 37, no. 6, pp. 1171–1180, May 2022. [CrossRef]
- W. Cheungpasitporn et al., ‘Intravitreal Antivascular Endothelial Growth Factor Therapy May Induce Proteinuria and Antibody Mediated Injury in Renal Allografts’, Transplantation, vol. 99, no. 11, pp. 2382–2386, Nov. 2015. [CrossRef]
- Q. Zhang and E. F. Reed, ‘The importance of non-HLA antibodies in transplantation’, Nat. Rev. Nephrol., vol. 12, no. 8, pp. 484–495, Aug. 2016. [CrossRef]
- V. Nickeleit, ‘Kidney transplants, antibodies and rejection: is C4d a magic marker?’, Nephrol. Dial. Transplant., vol. 18, no. 11, pp. 2232–2239, Nov. 2003. [CrossRef]
- C. M. Tower et al., ‘Plasma C4d+ Endothelial Microvesicles Increase in Acute Antibody-Mediated Rejection’, Transplantation, vol. 101, no. 9, pp. 2235–2243, Sep. 2017. [CrossRef]
- S. R. Martins et al., ‘Cell-derived microparticles and von Willebrand factor in Brazilian renal transplant recipients’, Nephrology, vol. 24, no. 12, pp. 1304–1312, Dec. 2019. [CrossRef]
- G. V. C. Freue et al., ‘Proteomic Signatures in Plasma during Early Acute Renal Allograft Rejection’, Mol. Cell. Proteomics, vol. 9, no. 9, pp. 1954–1967, Sep. 2010. [CrossRef]
- H.-Y. Jung et al., ‘Potential urinary extracellular vesicle protein biomarkers of chronic active antibody-mediated rejection in kidney transplant recipients’, J. Chromatogr. B, vol. 1138, p. 121958, Feb. 2020. [CrossRef]
- D. Chen et al., ‘Noninvasive detection of acute renal allograft rejection by measurement of soluble Tim-3 in urine’, Mol. Med. Rep., vol. 16, no. 1, pp. 915–921, Jan. 2017. [CrossRef]
- G. Joelsons, T. Domenico, L. F. Gonçalves, and R. C. Manfro, ‘Non-invasive messenger RNA transcriptional evaluation in human kidney allograft dysfunction’, Braz. J. Med. Biol. Res., vol. 51, no. 7, p. e6904, 2018. [CrossRef]
- A.-M. Pașatu-Cornea, E. Ciciu, and L.-A. Tuță, ‘Perforin: An intriguing protein in allograft rejection immunology (Review)’, Exp. Ther. Med., vol. 24, no. 2, p. 519, Jun. 2022. [CrossRef]
- T. K. Sigdel et al., ‘The Identification of Novel Potential Injury Mechanisms and Candidate Biomarkers in Renal Allograft Rejection by Quantitative Proteomics’, Mol. Cell. Proteomics, vol. 13, no. 2, pp. 621–631, Feb. 2014. [CrossRef]
- F. L. Nauta et al., ‘Albuminuria, Proteinuria, and Novel Urine Biomarkers as Predictors of Long-term Allograft Outcomes in Kidney Transplant Recipients’, Am. J. Kidney Dis., vol. 57, no. 5, pp. 733–743, May 2011. [CrossRef]
- M. G. J. Snoeijs et al., ‘Tubular Epithelial Injury and Inflammation After Ischemia and Reperfusion in Human Kidney Transplantation’, Ann. Surg., vol. 253, no. 3, pp. 598–604, Mar. 2011. [CrossRef]
- T. C. Van Smaalen et al., ‘Extracellular histone release by renal cells after warm and cold ischemic kidney injury: Studies in an ex-vivo porcine kidney perfusion model’, PLOS ONE, vol. 18, no. 1, p. e0279944, Jan. 2023. [CrossRef]
- W. W. Hancock, W. Gao, K. L. Faia, and V. Csizmadia, ‘Chemokines and their receptors in allograft rejection’, Curr. Opin. Immunol., vol. 12, no. 5, pp. 511–516, Oct. 2000. [CrossRef]
- M. Fischereder, ‘The role of chemokines in acute renal allograft rejection and chronic allograft injury’, Front. Biosci., vol. Volume, no. 14, p. 1807, 2009. [CrossRef]
- B. Yadav, N. Prasad, V. Agarwal, V. Agarwal, and M. Jain, ‘Hidden Granzyme B-Mediated Injury in Chronic Active Antibody-Mediated Rejection’, Exp. Clin. Transplant., vol. 18, no. 7, pp. 778–784, Dec. 2020. [CrossRef]
- J.-H. Lim et al., ‘Omics-based biomarkers for diagnosis and prediction of kidney allograft rejection’, Korean J. Intern. Med., vol. 37, no. 3, pp. 520–533, May 2022. [CrossRef]
- M. E. Ziegler et al., ‘Apolipoprotein A1 and C-Terminal Fragment of α-1 Antichymotrypsin Are Candidate Plasma Biomarkers Associated With Acute Renal Allograft Rejection’, Transplantation, vol. 92, no. 4, pp. 388–395, Aug. 2011. [CrossRef]
- H.-Y. Jung et al., ‘Potential urinary extracellular vesicle protein biomarkers of chronic active antibody-mediated rejection in kidney transplant recipients’, J. Chromatogr. B, vol. 1138, p. 121958, Feb. 2020. [CrossRef]
- J.-H. Lim et al., ‘Novel urinary exosomal biomarkers of acute T cell-mediated rejection in kidney transplant recipients: A cross-sectional study’, PLOS ONE, vol. 13, no. 9, p. e0204204, Sep. 2018. [CrossRef]
- I. Mühlberger, P. Perco, R. Fechete, B. Mayer, and R. Oberbauer, ‘Biomarkers in Renal Transplantation Ischemia Reperfusion Injury’, Transplantation, vol. 88, no. 3S, pp. S14–S19, Aug. 2009. [CrossRef]
- X. Lai, X. Zheng, J. M. Mathew, L. Gallon, J. R. Leventhal, and Z. J. Zhang, ‘Tackling Chronic Kidney Transplant Rejection: Challenges and Promises’, Front. Immunol., vol. 12, p. 661643, May 2021. [CrossRef]
- H. Wu et al., ‘Single-Cell Transcriptomics of a Human Kidney Allograft Biopsy Specimen Defines a Diverse Inflammatory Response’, J. Am. Soc. Nephrol., vol. 29, no. 8, pp. 2069–2080, Aug. 2018. [CrossRef]
- X. Xiang, J. Zhu, G. Dong, and Z. Dong, ‘Epigenetic Regulation in Kidney Transplantation’, Front. Immunol., vol. 13, p. 861498, Apr. 2022. [CrossRef]
- S. Herath, J. Erlich, A. Y. M. Au, and Z. H. Endre, ‘Advances in Detection of Kidney Transplant Injury’, Mol. Diagn. Ther., vol. 23, no. 3, pp. 333–351, Jun. 2019. [CrossRef]
- U. Christians, J. Klawitter, and J. Klawitter, ‘Biomarkers in Transplantation—Proteomics and Metabolomics’, Ther. Drug Monit., vol. 38, no. Supplement 1, pp. S70–S74, Apr. 2016. [CrossRef]
| Genes Overexpressed in the Common Rejection Module | |||
|---|---|---|---|
| Biomarker | Method of assessment | Pathogenic Characteristics/Function | Clinical Relevance |
| BASP1 | Microarray analysis, Next-Generation Sequencing (NGS), Quantitative Real-Time PCR (qPCR), RNA Sequencing (RNA-seq) and computational gene expression scoring | Activate signaling pathways, cytotoxicity, inflamation, immune reactions, T cell recruitment | Acute rejection, Chronic allograft dysfunction |
| CD6 | |||
| CXCL10 | |||
| CXCL9 | |||
| INPP5D | |||
| ISG20 | |||
| LCK | |||
| NKG7 | |||
| PSMB9 | |||
| RUNX3 | |||
| TAP1 | |||
| Transcriptomic Biomarkers | |||
| 4-gene model (vimentin, NKCC2, E-cadherin, and 18S rRNA) | Gene expression profiling using microarray and next-generation sequencing technologies | Increased metabolic activity, Cellular Stress, tissue damage, impaired repair mechanisms | Ischemia-Reperfusion Injury, Acute rejection, Fibrosis |
| 11 genes | Gene expression profiling using microarray and next-generation sequencing technologies | ||
| Epigenetic Biomarkers | |||
| Foxp3 DNA demethylation | Quantitative Real-Time PCR (qRT-PCR), Chromatin Immunoprecipitation (ChIP), Advanced Single-Cell Techniques, Flow Cytometry | Non-coding RNAs regulating gene expression in immune and inflammatory pathways. They reflect changes in gene expression without altering the DNA sequence | Correlate with intragraft regulatory T cells, indicating better graft outcomes |
| PD1 DNA methylation in memory CD8+ T cells | DNA Methylation Analysis Techniques, Genome-Wide Methylation Analysis, Single-Cell Methylation Analysis, Flow cytometry | Acute and chronic rejection | |
| microRNAs (miRs): miR-21, miR-200b, miR-150, miR-155, miR-192, miR-200b, miR-423-3p, miR-145-5p, miR-148a, miR-142-3p, miR-204, miR-211 | Quantitative Real-Time PCR (qRT-PCR), Northern Blotting, Microarray Analysis, Next-Generation Sequencing (NGS), Fluorescence In Situ Hybridization (FISH), Lateral Flow Assay (miRNA Detection Kit) | IFTA,Chronic allograft dysfunction), Acute rejection | |
| Proteomic Biomarkers | |||
| S100A8, S100A9, IL-6, IL-8, MCP-1, Cystatic-C | High-throughput proteomic techniques (LC-MS, iTRAQ, etc.) | Inflamation, immune response, and tissue injury | Acute and chronic rejection |
| Metabolomic Biomarkers | |||
| NAD, 1-MN, cholesterol sulfate, GABA, nicotinic acid, NADPH, proline, spermidine | Liquid Chromatography-Tandem Mass Spectrometry (LC-MS/MS), High-Performance Liquid Chromatography (HPLC), Gas Chromatography-Mass Spectrometry (GC-MS) | Inflammaton, oxidative stress, immune responses, inpaired remodeling | TCMR or overall chronic allograft nephropathy |
| Cellular Biomarkers | |||
| alloreactive CD8+ T cells, particularly effector memory T cells (TEMRA and EM | Flow cytometry | Inflammation, cytotoxicity | Rejection, Chronic allograft dysfunction |
| CD154+ T-cytotoxic memory cells | |||
| ratio of T follicular helper cells and T follicular regulatory cells (Tfh/Tfr) | |||
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2025 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
