Dersleri yüzünden oldukça stresli bir ruh haline sikiş hikayeleri bürünüp özel matematik dersinden önce rahatlayabilmek için amatör pornolar kendisini yatak odasına kapatan genç adam telefonundan porno resimleri açtığı porno filmini keyifle seyir ederek yatağını mobil porno okşar ruh dinlendirici olduğunu iddia ettikleri özel sex resim bir masaj salonunda çalışan genç masör hem sağlık hem de huzur sikiş için gelip masaj yaptıracak olan kadını gördüğünde porn nutku tutulur tüm gün boyu seksi lezbiyenleri sikiş dikizleyerek onları en savunmasız anlarında fotoğraflayan azılı erkek lavaboya geçerek fotoğraflara bakıp koca yarağını keyifle okşamaya başlar

GET THE APP

Journal of Clinical and Experimental Transplantation - Non-HLA Antibodies in Renal Transplantation, Where Do We Stand?
ISSN: 2475-7640

Journal of Clinical and Experimental Transplantation
Open Access

Like us on:

Our Group organises 3000+ Global Conferenceseries Events every year across USA, Europe & Asia with support from 1000 more scientific Societies and Publishes 700+ Open Access Journals which contains over 50000 eminent personalities, reputed scientists as editorial board members.

Open Access Journals gaining more Readers and Citations
700 Journals and 15,000,000 Readers Each Journal is getting 25,000+ Readers

This Readership is 10 times more when compared to other Subscription Journals (Source: Google Analytics)

Non-HLA Antibodies in Renal Transplantation, Where Do We Stand?

El Hennawy HM*
Transplant Surgery Section, Department of Surgery, Armed Forces Hospital-Southern Regions, Khamis Mushayt, KSA
*Corresponding Author: El Hennawy HM, MD, Transplant Surgery Section, Department of Surgery, Armed Forces Hospital-Southern Regions, Khamis Mushayt, 101, KSA, Tel: +966 503081770, Email: hennawyhany@hotmail.com

Received: 25-Nov-2018 / Accepted Date: 18-Dec-2018 / Published Date: 26-Dec-2018 DOI: 10.4172/2475-7640.1000125

Abstract

Objective: This review discusses current findings in the subject and addresses the clinical relevance of Non-HLA antibodies.
Methods: This traditional narrative review used PubMed and Medline searches for English language reports on Non HLA abs during last 20 years. The search included the key words: non-human leukocyte; antibodies; kidney; transplantation.
Results: 65 related articles and review were found.
Conclusion: Non-HLA immunity is associated with poor graft survival, rejection and chronic graft loss. Moreover, they could be used as biomarkers of ongoing immune response and as predictors of graft failure.

Keywords: Non-human leukocyte; Antibodies; Kidney; Transplantation

Abbreviations

Abs: Antibodies; AEPCA: Anti-Endothelial Precursor Cell Antibodies; AECA: Anti-Endothelial cell Antibodies; AMR: Antibody-Mediated Rejection; Anti-Col: Anti-Collagens; AT1R: Angiotensin II Type 1 Receptor; Anti-Ka1 tubulin: Anti-Tubulin ; AVA: Anti-Vimentin Antibody; C4D: Complement Component 4d; DSA: Donor-Specific Antibodies; EC: Endothelial Cell; ELISA: Enzyme- Linked Immunosorbent Assay; ETAR: Endothelin-1 Type A Receptor; FSGS: Focal Segmental Glomerulosclerosis; HLA: Human Leukocyte Antigen; MHC: Major Histocompatibility Complex; MICA: Major Histocompatibility Complex Class I Chain-Related Gene A; Non-HLA abs: Non-Human Leukocyte antibodies; XM: Crossmatch.

Introduction

Non-HLA antigens include antigens expressed on endothelial, epithelial cells, parenchymal cells and circulating immune cells [1-3]. Non HLA abs can be directed against auto- or allo-antigens and be either present pre-transplant or de novo formed post transplantation [3]. Furthermore, The most reported Non-HLA abs include those directed against Angiotensin II Type 1 Receptor (AT1R-Ab), Endothelin Type A Receptor (ETAR), MHC Class I Chain-Related Antigen A (MICA-Ab), Vimentin (AVA), Tubulin (anti-Ka1 tubulin), Collagens (anti-Col) Anti Endothelial Cell Antibodies (AECA), anti-heat shock protein, and antiphospholipid (Table 1) [4,5].

Targets for Non-HLA Antibodies
 
  • Major histocompatibility complex class I chain-related gene: A (MICA), B (MICB)
  • Antibodies against G Protein-Coupled Receptors (GPCRs): AT1R (Angiotensin II type 1 receptor) and Endothelin-1 type A receptor (ETAR)
  • Antiendothelial cell antibodies (AECAs)
  • Anti endothelial precursor cell antibodies (AEPCA)
  • LG3 (Perlecan)
  • Intercellular adhesion molecule 4 (ICAM4)
  • Anti-GBM Glomerular basement membrane (GBM)
  • Antivimentin: Intermediate Filaments – Vimentin (AVA)
  • Duffy antibody (a chemokine receptor)
  • Agrin antibody
  • Fibronectin and
  • Anti collagen IV antibodies (anti-Col)
  • Myosin antibody
  • Anti-Ka1 tubulin antibody
  • Anti heat shock protein
  • Anti-phospholipid

Table 1: Targets for Non-HLA Antibodies.

Moreover, the triggers of activation or transition of these Non-HLA abs toward pathogenicity are likely acute rejection, hypoperfusion, ischemia reperfusion, calcineurin toxicity, infection, and recurrent diseases [6].

Non-HLA abs have a stronger role in graft dysfunction and rejection; Antibody-Mediated Rejection (AMR) or C4d deposition in the absence of circulating donor specific Non-HLA abs than previously thought [1,5-7]. The aim of this review is to shed light on Non-HLA abs development, mechanism of action, clinical relevance, and treatment.

Mechanism of NON-HLA antibodies production

Injury of graft endothelium by Non HLA abs can lead to exposure of neo-antigens which consecutively stimulate the production of antibodies against non-HLA antigens [1,4,5,7-9]. Furthermore, Cytokine storm during brain death and inflammation associated with an ischemia–reperfusion injury, vascular injury, and/or rejection may cause increased expression of cryptic autoantigens, and may stimulate Non-HLA abs production. Additionally, immune activation, tapered immunosuppression in transplant recipients may stimulate Non HLA abs production [10].

However, several studies reported other ways of Non-HLA abs development other than sensitization [1,11-13]. For example, an A5.1 mutation in the donor, which is related to the MICA*008 allele, is associated with a strongly increased MICA expression on donor endothelial cells compared to wild type donors and therefore these mutated MICA molecules are important targets for antibody formation [14]. Additionally, mismatching on certain amino acid residues leads to increased MICA antibody formation and it can be that based on the 3D-structure of MICA, these structures are more accessible for antibodies [1,13].

Conversely, inflammatory response induced by Non-HLA abs could sequentially upregulate HLA expression, increase the risk for a patient to develop HLA-specific antibodies and thus make the allograft more susceptible to an allo-immune response involving both humoral and cellular [1,2,4,5,8,15].

Numerous studies showed that patients with both HLA and non- HLA antibodies had lower graft survival rates compared to patients with either one of them [16]. It is assumed that HLA and non-HLA antibodies have a synergistic effect [4,15].

Non-HLA antibodies incidence and pathogenicity

Non-HLA abs may function as complement- and non-complementfixing antibodies and they may induce a large variety of allograft injuries, reflecting the complexity of their acute and chronic actions [17].

Complement-dependent and complement-independent mechanisms are not mutually exclusive [8,18]. For example, Anti-Vimentin Antibodies [AVA] seem to fix complement [19]. Similarly, MICA Abs have been shown to be more efficient at complement activation and have been associated with C4d AMR [2]. In contrast, 40%-50% of cases with severe vascular changes such as fibrinoid necrosis are C4dnegative, implicating involvement of either non-complement-fixing antibodies or other mediators, as noticed in of cases of AMR in the presence of AT1R-Ab or AECA that occurred without evidence of complement activation [2, 20,21].

Besides, antibodies can induce lysis of target-cells with membrane bound antibodies through activation of natural-killer cells, a process called antibody-dependent cell mediated cytotoxicity [1,22]. Furthermore, Non-HLA abs may also contribute to short and long-term structural changes in the arterial wall or duct epithelia that promote clotting or/and narrowing [8].

Additionally, The capability of Non-HLA abs to mediate allograft injury may depend on their specificity and affinity, density of the target antigen, and synergy with HLA antibodies [2].It is unlikely that Non- HLA abs can directly induce major graft damage since hyperacute rejection induced by these antibodies rarely occurs (Table 2) [23-37].

Non HLA Antibodies Incidence and Mechanism of Action
Antibody Incidence Mechanism of Action
Major histocompatibility complex class I chain-related gene A
(MICA)
*13.9% and 5.4% pre and post-transplant, respectively [25]. *Complement-activating antibodies (fix C1q) [2,23].
*Activate NK cell via MICA/NKG2D interactions with subsequent cytotoxic proteins and IFN-γ release [26].
Angiotensin II type 1 receptor (AT1R) AT1R *22% [27], 23% [28], 47%[29] and 59%
[30] using a cutoff ≥ 9 units/ml. *Higher rate of AT1R-Ab positivity in patients with previous transplants [31].
*Activate complement independent pathways. In addition, increased tissue factor expression and thrombotic occlusion [18,23].
*Induce Erk1/2 signal transduction cascade that directly affect endothelial and vascular smooth muscle cells [23].
*Increase DNA binding activity of NF-B transcription factor, and increase expression of NF-B proinflammatory target genes such are chemokines 1 and RANTES [23].
Endothelin-1 type A receptor (ETAR)    *Damaging endothelial cells and increasing downstream effectors of GPCR signaling.
*Cause obliterative vasculopathy and progressive tissue fibrosis [32].
Antiendothelial cell antibodies (AECAs)   *23 % [1,7]
*In 50% of renal patients who had DSA to HLA [1,7].
*Higher rate of AECA positivity was found in patients with failed renal transplants
[1,7].
*Activate endothelial cell and produce of inflammatory cytokines [2,3].
* Increase HLA expression on endothelial cells, which may explain the severity of antibody-mediated injury in recipients when both AECAs and HLA-DSA were detected [2].
*Lead to AMR by activating complement [34].
  LG3 (Perlecan)   *Cause vascular injury and neointimal formation.
*Elicit humoral immune responses that accelerate immune-mediated vascular injury [35].
Intercellular adhesion molecule 4 (ICAM4)   *Activate Erk-mitogen-activated protein kinase pathway.
* Activate endothelial cell via induction of downstream proinflammatory signaling pathways [36].
Anti-GBM   *Targeting perlecan via proteolysis and degradation of perlecan induce profound changes in its biological activity [37].

Table 2: Non HLA antibodies incidence and mechanism of action.

Non-HLA Antibodies as Biomarkers of Injury

On the other hand, other studies claimed that Non-HLA abs may represent a marker for injury or humoral activation rather than having independent pathogenic potential [2,11]. Therefore, in the near future, Non-HLA abs may be used as biomarkers of ongoing immune response and herald the need for more suitable immunosuppression [8].

Compartment specificity

Non-HLA immune responses, including anti-MICA antibodies, were detected against kidney compartment-specific antigens, with highest post-transplant recognition for renal pelvis and cortex specific antigens (78%). The compartment specificity of selected antibodies was confirmed by IHC [7].

Clinical relevance of Non-HLA antibodies in renal transplantation

Non-HLA immunity has a much stronger role in clinical transplantation than previously thought. 10% of cases with C4d positivity fail to show circulating anti-HLA antibody is suggestive that Non-HLA abs also are to be considered [38]. In contrast to immunity against HLA mediated by antibodies present before transplantation, which leads to early acute graft rejection, non-HLA immunity is associated with chronic graft loss [39]. Moreover, the influence of non- HLA directed immunity was of similar magnitude to that of antibodies against HLA on long term follow up (Table 3) [39].

Clinical Relevance of Non-HLA antibodies
Antibody Clinical Relevance
Antivimentin (AVA) *Expression increases during rejection [2].
*Post-transplant development of IgG AVA was a risk factor associated with chronic injury such as interstitial fibrosis and tubular atrophy [27, 40-41].
Major histocompatibility complex class I chain-related gene: A
(MICA)
*Correlated with rejection (acute and chronic) and poor allograft survival (only significant in low immunological risk transplantations: well matched for the HLA) [1,7,42,44]
* Contrary to expectations, patients with positive pretransplant MICA antibodies had superior death-censored renal allograft survival when compared with MICA-negative patients [1].
Anti-endothelial precursor cell antibodies
(AEPCA)
*Strongly associated with acute rejections and increased serum creatinine levels at 3 and 6 months post-Tx [45].
Angiotensin II type 1 receptor (AT1R) *Associated with a higher incidence of graft loss [1,28,49], severe rejection [chronic and acute rejection (AMR and cellular mediated) and malignant hypertension [7,27,29,46].
*Patients with bothAT1R-Ab and HLA-DSA had greater incidence of allograft damage and graft loss [29,46-47].
*Patients with anti-AT1R Abs level >9 U/ml run a higher risk of graft failure independently of classical immunological risk factors [28].
*Patients with both anti-AT1R and DSA had lower graft survival than those with DSA alone [48].
Endothelin-1 type A receptor (ETAR) *Associated with a higher incidence of graft loss and rejection during the first post-transplant year [1,5,49].
*Vasculopathy or arteritis were observed in patients with anti-ETAR = 2.5 U/mL (p=0.0275) [5].
Duffy antibody (a chemokine receptor) *Associated with chronic renal allograft histological injury [7].
Agrin antibody *Associated with transplant glomerulopathy [7].
fibronectin and collagen IV antibodies *A significant risk factor for development of transplant glomerulopathy, a chronic lesion characterized by duplication of glomerular basement [27,50].
Antiendothelial cell antibodies (AECA) *AECAs are a risk marker for acute rejection [51]
*associate with both severe rejection (cellular mediated rejection and (AMR)) in kidney transplant recipients [2,52].
*high prevalence of C4d negative microcirculation injury [53].

Table 3: Clinical relevance of non-HLA antibodies.

Non-HLA antibodies monitoring and graft failure prediction

Many of the late graft failures attributable to non-HLA effects might be preventable [39]. The possibility of identifying recipients at increased risk of late graft loss before transplantation could be used to fashion specific immunosuppressive strategies for these patients [39-54]. For instance, the detection of anti-AT1R Abs seems to be a complementary risk factor for the identification of patients with higher immunological risk. Moreover, Banasik et al. proved that the occurrence of pre-transplant anti-AT1R Abs >9 U/ml is an independent risk factor for graft failure [28,29]. Therefore, monitoring for Non HLA abs should mirror that performed for HLA-DSA to identify those high risk patients [2].

Other possible uses of Non-HLA antibodies

Pre-transplant auto-antibody titers could have implications in terms of organ allocation. For instance, avoid use of organs with expected long cold ischemic time or coming from a donor after cardiocirculatory arrest for patients with elevated pre-transplant autoantibody titers [10].

Furthermore, Pre-transplant autoantibody levels could be added to the current clinical and laboratory variables used to assess the risk of rejection or delayed graft function, which in turn, could help transplant physicians select the most appropriate induction therapy [10]. For example, Pre-transplantation screening of recipients for AT1R-Abs may help to improve individual risk assessment and offer patients with AT1R-Abs preemptive specific treatment. Unfortunately, early AMR due to non-HLA antibodies is rare and seems difficult to predict by currently available assays including the AT1R-Ab-ELISA [53].

Who should be tested for Non-HLA antibodies?

Philogene et al. suggested performing pre-transplant Non HLA abs testing and post-transplant monitoring for high risk group of patients [2]. The risk factors include re-transplanted, male gender, young age, and those with FSGS at time of transplantation were positive for AT1RAbs and AECAs prior to transplantation [2]. Furthermore, testing for non-HLA antibodies is often performed when histological evidence suggests an antibody mediated process in the absence of HLA-DSA [2].

Non-HLA abs and Pediatric age group

Chaudhuri et al. reported that 24% of children with renal transplant have de novo antibodies, mostly directed against HLA. 6% of de novo antibodies were DSA Ab and 6% anti MHC class 1 related chain A (MICA), and were equally found either on steroid-free or steroidbased regimens. The presence of anti HLA and anti-MICA Ab was significantly associated with acute and chronic rejection with faster graft loss [54].

Interestingly, Matthew et al. reported a case of hyperacute rejection in 17 month old boy due to non-HLA antibodies. Pre-transplant Single antigen testing confirmed the absence of Donor Specific HLA Abs (DSA). Moreover, initial, final flow cross matches and 2 days post-Txp HLA-DSA were negative. Pre-Txp (pre-14 days) and post-Txp (post- 24 days) samples were sent out for AT1R Abs screening and donor specific endothelial cell crossmatch (XM-One). The XM-One assay using endothelial precursors isolated from the donor as targets was strongly positive using a pre-Txp serum but negative using post-Txp serum. Approximately two month’s post-Txp, the patient developed HLA Abs, on top of the AT1R antibodies [55].

Detection of Non-HLA Antibody

Considering the technical difficulties of current Non-HLA abs assays and the large variation in reported incidences of antibodies even with the same assays, continuous efforts to develop reliable and sensitive diagnostic tests are essential. Besides, measuring a panel of antibodies instead of one antibody at a time will provide valuable information regarding the role of Non-HLA abs in rejection and could eventually help identifying different risk profiles for rejection and impaired graft survival [1].

Currently, Non-HLA abs can be reliably detected by solid-phase assays (antibodies targeting G protein-coupled receptors (angiotensin type 1 receptor), MICA, collagen-V, vimentin), immunofluorescence (antibodies against antigens expressed on umbilical vein endothelial cells), ELISA or flow-crossmatch techniques (antibodies against donor endothelial progenitors) (Table 4) [3].

Non HLA Detection
Antibody Test
AT1R Single plex ELISA [100% specificity and 88% sensitivity] [9]
ETAR Single plex ELISA [9]
MICA Single antigen Luminex assays [9]
AECA * Indirect immunofluorescence HUVECs test.
* Flow cytometry endothelial crossmatch test (ECXM) [62]
Anti-endothelial precursor cell antibodies
(AEPCA)
Endothelial precursor cell crossmatch [45]
Multiple non-HLA
Antibodies
*Luminex based assay [4]
*Multiplex solid-phase assays [9]

Table 4: Non HLA detection.

At present, use of both ELISA and cytotoxicity assays in parallel for pre-transplant testing seems judicious to allow a separation of anti- HLA from anti-non-HLA activities [14].

Current Treatment Modalities for Pathogenic Non-HLA Antibodies

The presence of Non-HLA abs are not an absolute contraindication to transplantation, but rather may suggest previous or ongoing tissue injury, and may be useful in identifying patients who should be treated either prior to transplantation or post-transplantation to avoid graft injury [2].

Furthermore, immunologic risk stratification before transplantation, by comprehensive diagnostic assessment strategies focusing on both HLA-DSA and Non HLA abs responses, could help to better define subphenotypes of antibody-mediated rejection, or delayed graft function, and lead to timely initiation of targeted therapies [10]. Accordingly, early treatment of patients with increased immunologic risk factors and with circulating Non HLA abs is required [2].

Treatment to reduce levels of Non HLA abs is similar to what is commonly used for HLA antibodies (intravenous immunoglobulin, plasmapheresis, rituximab, and bortezomib) [56]. However, Combination therapies with Plasmapheresis (pre- and/or posttransplant), intravenous immunoglobulin (100mg/kg) and rituximab may lead to more durable antibody elimination [2,9,57].

Angiotensin receptor blockers such as losartan have also been used to block the activity of angiotensin receptor in patients with AT1R-Abmediated rejection [54,57]. However, a more recent study shows that chronic use of losartan can upregulate AT1R expression resulting in worse outcomes [58].

Recently, bortezomib was used to block the production of anti- LG3 auto-antibodies triggered by exosome-like vesicles may prove useful to help define therapeutic options for preventing auto-antibody production before transplantation [10,59].

Conclusions

The role of Non-HLA abs in renal transplantation is progressively being recognized. Non-HLA immunity is associated with poor graft survival, rejection and chronic graft loss. Moreover, they could be used as biomarkers of ongoing immune response and as predictors of graft failure. Therefore, they may herald the need for more suitable immunosuppression. Strong efforts to investigate Non HLA abs and their effect on graft outcome are still ongoing.

Disclosure

The author declared no competing interests.

References

  1. Michielsen LA, van Zuilen AD, Krebber MM, Verhaar MC, Otten HG (2016) Clinical value of non- HLA antibodies in kidney transplantation: Still an enigma? Transplant Rev (Orlando) 30: 195-202.
  2. Philogene MC, Jackson AM (2016) Non-HLA antibodies in transplantation: when do they matter? Curr Opin Organ Transplant 21: 427-432.
  3. Jackson AM, Sigdel TK, Delville M, Hsieh SC, Dai H, et al. (2015) Endothelial cell antibodies associated with novel targets and increased rejection. J Am Soc Nephrol 26:1161-1171.
  4. Zhang X, Reinsmoen NL (2017) Impact of Non-Human Leukocyte Antigen-Specific Antibodies in Kidney and Heart Transplantation. Front Immunol 8: 434.
  5. Banasik M, Boratyńska M, Kościelska-Kasprzak K, Krajewska M, Mazanowska O, et al. (2014) The impact of non-HLA antibodies directed against endothelin-1 type A receptors (ETAR) on early renal transplant outcomes.Transpl Immunol 30:24-29.
  6. Sigdel TK, Li L, Tran TQ, Khatri P, Naesens M, et al. (2012) Non-HLA antibodies to immunogenic epitopes predict the evolution of chronic renal allograft injury. J Am Soc Nephrol 23: 750-763.
  7. Li L, Wadia P, Chen R, Kambham N, Naesens M, et al. (2009) Identifying compartment-specific non-HLA targets  after renal transplantation by integratingtranscriptome and "antibodyome" measures. Proc Natl Acad Sci USA 106: 4148-4153.
  8. Remuzzi G, Chiaramonte S, Perico N, Ronco C (2009) Humoral immunity in kidney transplantation: what clinicians need to know. Contrib Nephrol 162:vii-ix.
  9. Jordan P, Kübler D (1996) Autoimmune diseases: nuclear autoantigens can be found at the cell-surface. Mol Biol Rep 22: 63-66.
  10. Cardinal H, Dieudé M, Hébert MJ (2017) The Emerging Importance of Non-HLA Autoantibodies in Kidney Transplant Complications. J Am Soc Nephrol. 28: 400-406.
  11. Valenzuela NM, Reed EF (2013) Antibodies in Transplantation: The Effects of HLA and Non-HLA Antibody Binding and Mechanisms of Injury. Methods Mol Biol 1034: 41-70.
  12. Sánchez-Zapardiel E, Castro-Panete MJ, Castillo-Rama M, Morales P, Lora-Pablos D, et al. (2013) Harmful effect of preformed anti-MICA antibodies on renal allograft evolution in early post-transplantation period. Transplantation 96: 70-78.
  13. Cox ST, Stephens HA, Fernando R, Karasu A, Harber M, et al. (2011) Major histocompatibility complex class I-related chain A allele mismatching, antibodies, and rejection in renal transplantation. Hum Immunol 72: 827-834.
  14. Tonnerre P, Gérard N, Chatelais M, Poli C, Allard S, et al. (2013) MICA variant promotes allosensitization after kidney transplantation. J Am Soc Nephrol 24: 954-966.
  15. Reinsmoen NL (2013) Role of angiotensin II type 1 receptor-activating antibodies in solid organ transplantation. Hum Immunol 74:1474-1477.
  16. Cardinal H, Dieudé M, Brassard N, Qi S, Patey N, et al. (2013) Antiperlecan antibodies are novel accelerators of immune-mediated vascular injury. Am J Transplant 13: 861-874.
  17. Dragun D, Hegner B (2009) Non-HLA antibodies post-transplantation: clinical relevance and treatment in solid organ transplantation. Contrib Nephrol 162: 129-139.
  18. Wehner J, Morrell CN, Reynolds T, Rodriguez ER, Baldwin WM (2007) Antibody and complement in transplant vasculopathy. Circ Res 100: 191-203.
  19. Mahesh B, Leong HS, McCormack A, Sarathchandra P, Holder A, et al. (2007) Autoantibodies to vimentin cause accelerated rejection of cardiac allografts. Am J Pathol 170: 1415-1427.
  20. Nickeleit V, Mihatsch MJ (2003) Kidney transplants, antibodies and rejection: is C4d a magic marker? Nephrol Dial Transplant 18: 2232-2239.
  21. Ming Y, Hu J, Luo Q, Ding X, Luo W, et al. (2015) Acute antibody-mediated rejection in presence of MICA-DSA and successful renal re-transplant with negative-mica virtual crossmatch. PLoS One 10: e0127861.
  22. Suviolahti E, Ge S, Nast CC, Mirocha J, Karasyov A (2015) Genes associated with antibody-dependent cell activation are overexpressed in renal biopsies from patients with antibody-mediated rejection. Transpl Immunol 32: 9-17.
  23. Dragun D, Müller DN, Bräsen JH, Fritsche L, Nieminen-Kelhä M, et al. (2005) Angiotensin II type 1-receptor activating antibodies in renal-allograft rejection. N Engl J Med 352: 558-569.
  24. Sumitran-Karuppan S, Tyden G, Reinholt F, Berg U, Moller E (1997) Hyperacute rejections of two consecutive renal allografts and early loss of the third transplant caused by non-HLA antibodies specific for endothelial cells. Transpl Immunol 5: 321-327.
  25. Lemy A, Andrien M, Wissing KM, Ryhahi K, Vandersarren A, et al. (2010) Major histocompatibility complex class 1 chain-related antigen a antibodies: sensitizing events and impact on renal graft outcomes. Transplantation 90: 168-174.
  26. Carapito R, Bahram S (2015) Genetics, genomics, and evolutionary biology of NKG2D ligands. Immunol Rev 267: 88-116.
  27. Philogene MC, Zhou S, Lonze BE, Bagnasco S, Alasfar S (2018) Pre-transplant Screening for Non-HLA Antibodies: Who should be Tested? Hum Immunol 79: 195-202.
  28. Banasik M, Boratyńska M, Kościelska-Kasprzak K, Kamińska D, Bartoszek D (2014) The influence of non-HLA antibodies directed against angiotensin II type 1 receptor (AT1R) on early renal transplant outcomes. Transpl Int 27: 1029-1038.
  29. Giral M, Foucher Y, Dufay A, Van Huyen JP, Renaudin K, et al. (2013) Pretransplant sensitization against angiotensin II type 1 receptor is a risk factor for acuterejection and graft loss. Am J Transplant 13: 2567-2576.
  30. Lee J, Huh KH, Park Y,Park BG, Yang J, et al. (2017) The clinicopathological relevance of pre-transplant anti-angiotensin II type 1 receptor antibodies in renal transplantation. Nephrol Dial Transplant 32: 1244-1250.
  31. Taniguchi M, Rebellato LM, Cai J, Hopfield J, Briley KP, et al. (2013) Higher risk of kidney graft failure in the presence of anti-angiotensin II type-1 receptor antibodies. Am J Transplant 13: 2577-2589.
  32. Dragun D, Catar R, Philippe A (2016) Non-HLA antibodies against endothelial targets bridging allo- and autoimmunity. Kidney Int 90: 280-288.
  33. Han F, Lv R, Jin J, Wu J, Chen Y, et al. (2009) Pre-transplant serum concentrations of anti-endothelial cell antibody in panel reactive antibody negative renal recipients and its impact on acute rejection. Clin Chem Lab Med 47: 1265-1269
  34. Niikura T, Yamamoto I, Nakada Y, Kamejima S, Katsumata H, et al. (2015) Probable C4d-negative accelerated acute antibody-mediated rejection due to non-HLA antibodies. Nephrology (Carlton) 20: 75-78.
  35. Soulez M, Pilon EA, Dieudé M, Cardinal H, Brassard N, et al. (2012) The perlecan fragment LG3 is a novel regulator of obliterative remodeling associated with allograft vascular rejection. Circ Res 110: 94-104.
  36. Lawson C, Holder AL, Stanford RE, Smith J, Rose ML (2005) Anti-intracellular adhesion molecule-1antibodies in sera of heart transplant recipients: a role in endothelial activation. Transplant 80: 264-271.
  37. Cailhier JF, Laplante P, Hebert MJ (2006) Endothelial apoptosis and chronic transplant vasculopathy: recent results, novel mechanisms. Am J Transplant 6: 247-253.
  38. Tinckam KJ, Chandraker A (2006) Mechanisms and role of HLA and non-HLA alloantibodies. Clin J Am Soc Nephrol 1: 404-414.
  39. Opelz G (2005) Non-HLA transplantation immunity revealed by lymphocytotoxic antibodies. Lancet 365: 1570-1576.
  40. Besarani D, Cerundolo L, Smith JD, Procter J, Barnardo MC, et al. (2014) Role of anti-vimentin antibodies in renal transplantation. Transplantation 98: 72-78.
  41. Jurcevic S, Ainsworth ME, Pomerance A, Smith JD, Robinson DR, et al. (2001) Antivimentin antibodies are an independent predictor of transplant-associated coronary artery disease after cardiac transplantation. Transplantation 71: 886-892.
  42. Sumitran-Holgersson S, Wilczek HE, Holgersson J, Söderström K (2002) Identification of the nonclassical HLA molecules, mica, as targets for humoral immunityassociated with irreversible rejection of kidney allografts.Transplantation 74: 268-277.
  43. Mizutani K, Terasaki P, Rosen A, Esquenazi V, Miller J, et al. (2005) Serial ten-year follow-up of HLA and MICA antibody production prior to kidney graft failure. Am J Transplant 5: 2265-2272.
  44. Angaswamy N, Tiriveedhi V, Sarma NJ, Subramanian V, Klein C, et al. (2013) Interplay between immune responses to HLA and non-HLA self-antigens in allograft rejection. Hum Immunol 74: 1478-1485.
  45. Almahri A (2015) The clinical importance of non-HLA specific antibodies in kidney transplantation, Laboratory for Transplantation and Regenerative Medicine, Department of Clinical Chemistry and Transfusion Medicine Institute of Biomedicine, the Sahlgrenska Academy, University of Gothenburg.
  46. Zhang J, Wang M, Liang J, Zhang M, LiuXH, et al. (2017) The presence of antiangiotensin II type-1 receptor antibodies adversely affect kidney graft outcomes. Int J Environ Res Public Health 2017;14: 500.
  47. Sumitran-Holgersson S (2008) Relevance of MICA and other non-HLA antibodies in clinical transplantation. Curr Opin Immunol 20: 607-613.
  48. Banasik M, Boratyńska M, Kościelska-Kasprzak K, Kamińska D, Zmonarski S, et al. (2014) Non-HLA antibodies: angiotensin II type 1 receptor (anti-AT1R) and endothelin-1 type A receptor (anti-ETAR) are associated with renal allograft injury and graft loss. Transplant Proc 46: 2618-2621.
  49. Angaswamy N, Klein C, Tiriveedhi V, Gaut J, Anwar S, et al. (2014) Immune responses to collagen-IV and fibronectin in renal transplant recipients with Transplant glomerulopathy. Am J Transplant 14: 685-693.
  50. Ronda C, Borba SC, Ferreira SC, Glotz D, Ianhez LE, et al. (2011) Non-human leukocyte antigen antibodies reactive with endothelial cells could be involved in early loss of renal allografts. Transplant Proc 43: 1345-1348.
  51. Sun Q, Cheng Z, Cheng D, Chen J, Ji S (2011) De novo development of circulating anti-endothelial cell antibodies rather than pre-existing antibodies is associated with post-transplant allograft rejection. Kidney Int 79: 655-662.
  52. Breimer ME, Rydberg L, Jackson AM, Lucas DP, Zachary AA (2009) Multicenter evaluation of a novel endothelial cell crossmatch test in kidney transplantation. Transplantation 87: 549-556.
  53. Amico P, Hِnger G, Bielmann D, Lutz D, Garzoni D, et al. (2008) Incidence and prediction of early antibody-mediated rejection due to non-human leukocyte antigen-antibodies. Transplantation 85: 1557-1563.
  54. Chaudhuri A, Ozawa M, Everly MJ, Ettenger R, Dharnidharka V, et al. (2013) The clinical impact of humoral immunity in pediatric renal transplantation. J Am Soc Nephrol 24: 655-664.
  55. Cusick M, Bobrowski A, Haarberg KM, Tambur AAR (2015) Potential hyperacute renal rejection due to non-hla antibody in a pediatric patient. Human Immunology 76: 157.
  56. Djamali A, Kaufman DB, Ellis TM, Zhong W, Matas A, et al. (2014) Diagnosis and management of antibody-mediated rejection: current status and novel approaches. Am J Transplant 14: 255-271.
  57. Lukitsch I, Kehr J, Chaykovska L, Wallukat G, Nieminen-Kelhä M, et al. (2012) Renal ischemia and transplantation predispose to vascular constriction mediated by angiotensin II type 1 receptor-activating antibodies. Transplantation 94: 8-13.
  58. Song MA, Dasgupta C, Zhang L (2015) Chronic Losartan Treatment Up-Regulates AT1R and Increases the Heart Vulnerability to Acute Onset of Ischemia and Reperfusion Injury in Male Rats. PLoS One 10: e0132712.
  59. Dieudé M, Bell C, Turgeon J, Beillevaire D, Pomerleau L (2015) The 20S proteasome core, active within apoptotic exosome-like vesicles, induces autoantibody production and accelerates rejection. Sci Transl Med 7: 318ra200.

Citation: El Hennawy HM (2018) Non-HLA Antibodies in Renal Transplantation, Where Do We Stand?. J Clin Exp Transplant 3: 125. DOI: 10.4172/2475-7640.1000125

Copyright: © 2018 El Hennawy HM. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Top