1
|
Alhamdan F, Coppolino A, Sheikh A, Miele A, Lee S, Gasiewski A, Brescia P, Wood I, Venkat A, Thaniyavarn T, Jacob S, Keshk M, Meadowcroft S, Banday MM, Khan MM, Hayes D, Chandrekar A, Goldberg H, Guleria I, Sharma NS. Distinct Non-Human Leukocyte Antigen Antibody Signatures Correlate with Endothelial Crossmatch Status in Lung and Renal Transplant Recipients. Int J Mol Sci 2024; 25:10562. [PMID: 39408890 PMCID: PMC11476851 DOI: 10.3390/ijms251910562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/12/2024] [Accepted: 09/19/2024] [Indexed: 10/20/2024] Open
Abstract
Non-HLA antibodies against heterogeneous targets on endothelial cells have been associated with allograft injuries. The endothelial cell crossmatch (ECXM) is used in the detection of non-HLA antibodies but remains non-discriminatory for specific antibody identification. The primary objective of this study was to delineate the specific non-HLA antibody signatures associated with ECXM positivity and to determine the correlation of ECXM status and non-HLA antibody signatures on allograft health. Serum specimens from 25 lung transplant recipients (LTRs) and 13 renal transplant recipients (RTRs) were collected as part of clinical evaluation, and testing for angiotensin II receptor type 1 (AT1R) and donor-specific MHC class I chain-related gene A (MICA) antibodies and ECXM was performed. Remnant sera were tested for non-HLA antibodies using the LABScreen™ Autoantibody (LSAUT) Group 1, 2, and 3 kits (One Lambda, Inc., Los Angeles, CA, USA). In both cohorts, the concordance of AT1R and MICA together or individually with ECXM+ status was poor (<0.7), suggesting the presence of other unaccounted antibodies. Autoantibody profiling revealed three distinct clusters targeting fibrotic products, cytoskeletal proteins, and cell signaling molecules. A comparative analysis of ECXM+ and ECXM- specimens identified nine and five differentially expressed antibodies in the LTR and RTR cohorts, respectively. Employing machine learning techniques (variable importance, feature selection, ROC-AUC), we derived a five-antibody panel (TNFα, collagen V, CXCL11, GDNF, GAPDH) and a two-antibody panel (TNFα, CXCL9) that effectively discriminated between ECXM+ and ECXM- status in the LTR and RTR cohorts, respectively. Distinct antibody signatures were identified in LTR and RTR cohorts that correlated with ECXM+ status and were associated with allograft dysfunction.
Collapse
Affiliation(s)
- Fahd Alhamdan
- Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (F.A.); (A.C.); (A.S.); (A.M.); (S.L.); (A.G.); (P.B.); (I.W.); (A.V.); (T.T.); (S.J.); (M.K.); (S.M.); (M.M.B.); (M.M.K.); (A.C.); (H.G.); (I.G.)
| | - Antonio Coppolino
- Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (F.A.); (A.C.); (A.S.); (A.M.); (S.L.); (A.G.); (P.B.); (I.W.); (A.V.); (T.T.); (S.J.); (M.K.); (S.M.); (M.M.B.); (M.M.K.); (A.C.); (H.G.); (I.G.)
| | - Adil Sheikh
- Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (F.A.); (A.C.); (A.S.); (A.M.); (S.L.); (A.G.); (P.B.); (I.W.); (A.V.); (T.T.); (S.J.); (M.K.); (S.M.); (M.M.B.); (M.M.K.); (A.C.); (H.G.); (I.G.)
| | - Anna Miele
- Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (F.A.); (A.C.); (A.S.); (A.M.); (S.L.); (A.G.); (P.B.); (I.W.); (A.V.); (T.T.); (S.J.); (M.K.); (S.M.); (M.M.B.); (M.M.K.); (A.C.); (H.G.); (I.G.)
| | - Stefi Lee
- Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (F.A.); (A.C.); (A.S.); (A.M.); (S.L.); (A.G.); (P.B.); (I.W.); (A.V.); (T.T.); (S.J.); (M.K.); (S.M.); (M.M.B.); (M.M.K.); (A.C.); (H.G.); (I.G.)
- VA Boston Medical Center, Boston, MA 02130, USA
| | - Allison Gasiewski
- Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (F.A.); (A.C.); (A.S.); (A.M.); (S.L.); (A.G.); (P.B.); (I.W.); (A.V.); (T.T.); (S.J.); (M.K.); (S.M.); (M.M.B.); (M.M.K.); (A.C.); (H.G.); (I.G.)
| | - Peter Brescia
- Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (F.A.); (A.C.); (A.S.); (A.M.); (S.L.); (A.G.); (P.B.); (I.W.); (A.V.); (T.T.); (S.J.); (M.K.); (S.M.); (M.M.B.); (M.M.K.); (A.C.); (H.G.); (I.G.)
| | - Isabelle Wood
- Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (F.A.); (A.C.); (A.S.); (A.M.); (S.L.); (A.G.); (P.B.); (I.W.); (A.V.); (T.T.); (S.J.); (M.K.); (S.M.); (M.M.B.); (M.M.K.); (A.C.); (H.G.); (I.G.)
| | - Arvin Venkat
- Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (F.A.); (A.C.); (A.S.); (A.M.); (S.L.); (A.G.); (P.B.); (I.W.); (A.V.); (T.T.); (S.J.); (M.K.); (S.M.); (M.M.B.); (M.M.K.); (A.C.); (H.G.); (I.G.)
| | - Tany Thaniyavarn
- Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (F.A.); (A.C.); (A.S.); (A.M.); (S.L.); (A.G.); (P.B.); (I.W.); (A.V.); (T.T.); (S.J.); (M.K.); (S.M.); (M.M.B.); (M.M.K.); (A.C.); (H.G.); (I.G.)
- VA Boston Medical Center, Boston, MA 02130, USA
| | - Selvin Jacob
- Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (F.A.); (A.C.); (A.S.); (A.M.); (S.L.); (A.G.); (P.B.); (I.W.); (A.V.); (T.T.); (S.J.); (M.K.); (S.M.); (M.M.B.); (M.M.K.); (A.C.); (H.G.); (I.G.)
| | - Mohamed Keshk
- Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (F.A.); (A.C.); (A.S.); (A.M.); (S.L.); (A.G.); (P.B.); (I.W.); (A.V.); (T.T.); (S.J.); (M.K.); (S.M.); (M.M.B.); (M.M.K.); (A.C.); (H.G.); (I.G.)
| | - Stacia Meadowcroft
- Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (F.A.); (A.C.); (A.S.); (A.M.); (S.L.); (A.G.); (P.B.); (I.W.); (A.V.); (T.T.); (S.J.); (M.K.); (S.M.); (M.M.B.); (M.M.K.); (A.C.); (H.G.); (I.G.)
| | - Mudassir M. Banday
- Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (F.A.); (A.C.); (A.S.); (A.M.); (S.L.); (A.G.); (P.B.); (I.W.); (A.V.); (T.T.); (S.J.); (M.K.); (S.M.); (M.M.B.); (M.M.K.); (A.C.); (H.G.); (I.G.)
| | - Mohd Moin Khan
- Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (F.A.); (A.C.); (A.S.); (A.M.); (S.L.); (A.G.); (P.B.); (I.W.); (A.V.); (T.T.); (S.J.); (M.K.); (S.M.); (M.M.B.); (M.M.K.); (A.C.); (H.G.); (I.G.)
| | - Don Hayes
- Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA;
| | - Anil Chandrekar
- Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (F.A.); (A.C.); (A.S.); (A.M.); (S.L.); (A.G.); (P.B.); (I.W.); (A.V.); (T.T.); (S.J.); (M.K.); (S.M.); (M.M.B.); (M.M.K.); (A.C.); (H.G.); (I.G.)
| | - Hilary Goldberg
- Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (F.A.); (A.C.); (A.S.); (A.M.); (S.L.); (A.G.); (P.B.); (I.W.); (A.V.); (T.T.); (S.J.); (M.K.); (S.M.); (M.M.B.); (M.M.K.); (A.C.); (H.G.); (I.G.)
| | - Indira Guleria
- Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (F.A.); (A.C.); (A.S.); (A.M.); (S.L.); (A.G.); (P.B.); (I.W.); (A.V.); (T.T.); (S.J.); (M.K.); (S.M.); (M.M.B.); (M.M.K.); (A.C.); (H.G.); (I.G.)
| | - Nirmal S. Sharma
- Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (F.A.); (A.C.); (A.S.); (A.M.); (S.L.); (A.G.); (P.B.); (I.W.); (A.V.); (T.T.); (S.J.); (M.K.); (S.M.); (M.M.B.); (M.M.K.); (A.C.); (H.G.); (I.G.)
- VA Boston Medical Center, Boston, MA 02130, USA
| |
Collapse
|
2
|
Ming Y, Peng B, Guo X, Luo W, Shao M, Cheng K, Luo Q, Zou Y. Posttransplant-Alloantibodies Against MICA Antigens Associated With Decreased Long-Term Allograft Survival of Kidney Transplant Recipients. Transplant Proc 2022; 54:1801-1808. [DOI: 10.1016/j.transproceed.2022.03.064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 02/17/2022] [Accepted: 03/26/2022] [Indexed: 10/14/2022]
|
3
|
Lammerts RGM, Altulea D, Hepkema BG, Sanders JS, van den Born J, Berger SP. Antigen and Cell-Based Assays for the Detection of Non-HLA Antibodies. Front Immunol 2022; 13:864671. [PMID: 35603145 PMCID: PMC9122123 DOI: 10.3389/fimmu.2022.864671] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 03/16/2022] [Indexed: 01/20/2023] Open
Abstract
To date, human leukocyte antigens (HLA) have been the major focus in the approach to acute and chronic antibody-mediated rejection (AMBR) in solid-organ transplantation. However, evidence from the clinic and published studies has shown that non-HLA antibodies, particularly anti-endothelial cell antibodies (AECAs), are found either in the context of AMBR or synergistically in the presence of donor-specific anti-HLA antibodies (DSA). Numerous studies have explored the influence of AECAs on clinical outcomes, yet the determination of the exact clinical relevance of non-HLA antibodies in organ transplantation is not fully established. This is due to highly heterogeneous study designs including differences in testing methods and outcome measures. Efforts to develop reliable and sensitive diagnostic non-HLA antibody tests are continuously made. This is essential considering the technical difficulties of non-HLA antibody assays and the large variation in reported incidences of antibodies. In addition, it is important to take donor specificity into account in order to draw clinically relevant conclusions from non-HLA antibody assays. Here, we provide an overview of non-HLA solid-phase and cell-based crossmatch assays for use in solid-organ transplantation that are currently available, either in a research setting or commercially.
Collapse
Affiliation(s)
- Rosa G. M. Lammerts
- Transplantation Immunology, Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
- *Correspondence: Rosa G. M. Lammerts,
| | - Dania Altulea
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Bouke G. Hepkema
- Transplantation Immunology, Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Jan-Stephan Sanders
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Jacob van den Born
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Stefan P. Berger
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|
4
|
Baranwal AK, Agarwal SK, Mehra N. MHC Class I Related Chain A (MICA) Antibodies - A Potential Cause of Renal Allograft Rejection. Indian J Nephrol 2021; 31:583-586. [PMID: 35068770 PMCID: PMC8722556 DOI: 10.4103/ijn.ijn_407_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 10/25/2020] [Indexed: 11/09/2022] Open
Abstract
MHC class I related chain A (MICA) antibodies, especially those directed against the donor in absence of donor-specific anti-HLA antibodies have been reported to be possibly associated with renal allograft rejection in sensitized recipients. We are the first ones to present a case series of five patients who underwent primary live related donor renal transplantation in non-sensitized recipients either in the presence of donor-specific MICA antibodies (MICA-DSA) or developed de novo. Four of them presented characteristics of either accelerated, acute or chronic antibody-mediated rejection (AMR) attributable to the presence of MICA DSA. This case series emphasizes that AMR due to MICA-DSA is amenable to treatment with conventional regimens for treatment of AMR and there is a need for screening of MICA antibodies especially those directed against the donor on case to case basis.
Collapse
Affiliation(s)
- Ajay Kumar Baranwal
- Department of Transplant Immunology and Immunogenetics, All India Institute of Medical Sciences, Pune, Maharashtra, India.,Department of Pathology, Command Hospital, Pune, Maharashtra, India
| | - Sanjay Kumar Agarwal
- Department of Nephrology, All India Institute of Medical Sciences, New Delhi, India
| | - Narinder Mehra
- Department of Transplant Immunology and Immunogenetics, All India Institute of Medical Sciences, Pune, Maharashtra, India.,National Chair, ICMR, New Delhi, India
| |
Collapse
|
5
|
Fuertes MB, Domaica CI, Zwirner NW. Leveraging NKG2D Ligands in Immuno-Oncology. Front Immunol 2021; 12:713158. [PMID: 34394116 PMCID: PMC8358801 DOI: 10.3389/fimmu.2021.713158] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 07/02/2021] [Indexed: 12/14/2022] Open
Abstract
Immune checkpoint inhibitors (ICI) revolutionized the field of immuno-oncology and opened new avenues towards the development of novel assets to achieve durable immune control of cancer. Yet, the presence of tumor immune evasion mechanisms represents a challenge for the development of efficient treatment options. Therefore, combination therapies are taking the center of the stage in immuno-oncology. Such combination therapies should boost anti-tumor immune responses and/or target tumor immune escape mechanisms, especially those created by major players in the tumor microenvironment (TME) such as tumor-associated macrophages (TAM). Natural killer (NK) cells were recently positioned at the forefront of many immunotherapy strategies, and several new approaches are being designed to fully exploit NK cell antitumor potential. One of the most relevant NK cell-activating receptors is NKG2D, a receptor that recognizes 8 different NKG2D ligands (NKG2DL), including MICA and MICB. MICA and MICB are poorly expressed on normal cells but become upregulated on the surface of damaged, transformed or infected cells as a result of post-transcriptional or post-translational mechanisms and intracellular pathways. Their engagement of NKG2D triggers NK cell effector functions. Also, MICA/B are polymorphic and such polymorphism affects functional responses through regulation of their cell-surface expression, intracellular trafficking, shedding of soluble immunosuppressive isoforms, or the affinity of NKG2D interaction. Although immunotherapeutic approaches that target the NKG2D-NKG2DL axis are under investigation, several tumor immune escape mechanisms account for reduced cell surface expression of NKG2DL and contribute to tumor immune escape. Also, NKG2DL polymorphism determines functional NKG2D-dependent responses, thus representing an additional challenge for leveraging NKG2DL in immuno-oncology. In this review, we discuss strategies to boost MICA/B expression and/or inhibit their shedding and propose that combination strategies that target MICA/B with antibodies and strategies aimed at promoting their upregulation on tumor cells or at reprograming TAM into pro-inflammatory macrophages and remodeling of the TME, emerge as frontrunners in immuno-oncology because they may unleash the antitumor effector functions of NK cells and cytotoxic CD8 T cells (CTL). Pursuing several of these pipelines might lead to innovative modalities of immunotherapy for the treatment of a wide range of cancer patients.
Collapse
Affiliation(s)
- Mercedes Beatriz Fuertes
- Laboratorio de Fisiopatología de la Inmunidad Innata, Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina
| | - Carolina Inés Domaica
- Laboratorio de Fisiopatología de la Inmunidad Innata, Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina
| | - Norberto Walter Zwirner
- Laboratorio de Fisiopatología de la Inmunidad Innata, Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina.,Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
6
|
Baranwal AK, Bhat DK, Goswami S, Agarwal SK, Kaur G, Mehra N. Clinical relevance of major histocompatibility complex class I chain-related molecule A (MICA) antibodies in live donor renal transplantation - Indian Experience. Scand J Immunol 2020; 92:e12923. [PMID: 32593197 DOI: 10.1111/sji.12923] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 05/29/2020] [Accepted: 06/22/2020] [Indexed: 11/28/2022]
Abstract
Antibody-mediated rejections (AMR) in the absence of circulating anti-HLA-DSA have highlighted the role of non-HLA antibodies, particularly those directed against endothelial cells. Of these, MICA (major histocompatibility complex class I chain-related molecule A) antibodies are the most notable and important because of their potential in promoting graft rejections. Limited studies have focused on the impact of MICA donor-specific antibodies (DSA) on graft outcome as compared to those that are not donor-specific (NDSA). We evaluated pre- and post-transplant sera at POD 7, 30, 90, 180 and the time of biopsy from 206 consecutive primary live donor renal transplant recipients for anti-MICA and anti-HLA antibodies using single antigen bead assay on a Luminex platform. Recipients who developed MICA antibodies and their donors were phenotyped for MICA alleles. For the purpose of antibody analysis, patients were categorized into three major groups: biopsy-proven AMR, acute cellular rejection (ACR) and those with no rejection episodes (NRE). During the mean follow-up period of 17.37 ± 6.88 months, 16 of the 206 recipients developed AMR, while ACR was observed in only 13 cases. A quarter (25%) of the AMR cases had anti-MICA antibodies as compared to 7.7% of those experiencing ACR and 6.2% of the NRE group. Allelic typing revealed that all MICA Ab +ve AMR cases were due to the presence of donor-specific antibodies. MICA-DSA even in the absence of HLA-DSA was significantly associated with AMR but not with ACR when compared with the NRE group (P = <.01).
Collapse
Affiliation(s)
- Ajay Kumar Baranwal
- Department of Transplant Immunology and Immunogenetics, All India Institute of Medical Sciences, New Delhi, India.,Department of Pathology, Command Hospital, Pune, India
| | - Deepali K Bhat
- Department of Transplant Immunology and Immunogenetics, All India Institute of Medical Sciences, New Delhi, India.,Cellular & Molecular Therapeutic Branch, NHLBI, National Institute of Health (NIH), Bethesda, MD, USA
| | - Sanjeev Goswami
- Department of Transplant Immunology and Immunogenetics, All India Institute of Medical Sciences, New Delhi, India
| | - Sanjay Kumar Agarwal
- Department of Nephrology, All India Institute of Medical Sciences, New Delhi, India
| | - Gurvinder Kaur
- Department of Transplant Immunology and Immunogenetics, All India Institute of Medical Sciences, New Delhi, India.,Laboratory Oncology, Dr BR Ambedkar Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India
| | - Narinder Mehra
- Department of Transplant Immunology and Immunogenetics, All India Institute of Medical Sciences, New Delhi, India.,National Chair, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
7
|
Luo Q, Luo W, Zhu Q, Huang H, Peng H, Liu R, Xie M, Li S, Li M, Hu X, Zou Y. Tumor-Derived Soluble MICA Obstructs the NKG2D Pathway to Restrain NK Cytotoxicity. Aging Dis 2020; 11:118-128. [PMID: 32010486 PMCID: PMC6961768 DOI: 10.14336/ad.2019.1017] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 06/28/2019] [Indexed: 12/31/2022] Open
Abstract
The natural killer group 2D (NKG2D) receptor and its ligands play important roles in immune surveillance. In this study, we observed that the average serum soluble MICA (sMICA) concentration of 174 hepatocellular carcinoma (HCC) patients was significantly higher than that in 80 healthy subjects (602.17 ± 338.15 vs. 72.26 ± 87.88 pg/ml, t = 3.107, P=0.002). The levels of serum sMICA in 44 HCC patients with initial levels above 400 pg/ml declined significantly after surgical removal of the liver cancer tissue (P<0.001). Moreover, the mean survival time of HCC patients who had sMICA above 400 pg/ml was significantly shorter than that HCC patients with lower sMICA levels (P<0.001). Using the reporter cell line (NKG2D-2B4) in which activation of the NKG2D receptor pathway results in GFP expression based on the stimulation of immobilized rMICA, we showed that the number of GFP-expressing cells decreased sharply in presence of sMICA. Upon adding sMICA, the release of cytokines IFN-γ, TNF-α, and IL-8 by NK cell line (NKL) under stimulation of immobilized rMICA was blocked. Using MICA-expressing cells as the target cells, we observed that about 80% of target cells were killed by NKL at E:T of 10:1, but in presence of sMICAhigh serum of HCC patients, the dead target cells were reduced to 30.8%. Compared in presence of sMICAlow serum from HCC patients, there were 63.7% of target cells dead (p=0.043). Thus, our data suggested that sMICA obstructs the activation of NKG2D pathway to protect tumor cells from NK cell-mediated cytotoxicity.
Collapse
Affiliation(s)
- Qizhi Luo
- 1Department of Immunology, Basic Medical School of Central South University, Changsha, Hunan, China
| | - Weiguang Luo
- 1Department of Immunology, Basic Medical School of Central South University, Changsha, Hunan, China.,2Department of Physiology, University of Texas Southwestern Medical Center at Dallas, TX, USA
| | - Quan Zhu
- 1Department of Immunology, Basic Medical School of Central South University, Changsha, Hunan, China
| | - Hongjun Huang
- 3Cancer Hospital of Hunan, Xiangya Medical School, Central South University, Changsha, Hunan, China
| | - Huiyun Peng
- 1Department of Immunology, Basic Medical School of Central South University, Changsha, Hunan, China
| | - Rongjiao Liu
- 1Department of Immunology, Basic Medical School of Central South University, Changsha, Hunan, China
| | - Min Xie
- 1Department of Immunology, Basic Medical School of Central South University, Changsha, Hunan, China
| | - Shili Li
- 1Department of Immunology, Basic Medical School of Central South University, Changsha, Hunan, China
| | - Ming Li
- 1Department of Immunology, Basic Medical School of Central South University, Changsha, Hunan, China
| | - Xiaocui Hu
- 3Cancer Hospital of Hunan, Xiangya Medical School, Central South University, Changsha, Hunan, China
| | - Yizhou Zou
- 1Department of Immunology, Basic Medical School of Central South University, Changsha, Hunan, China
| |
Collapse
|
8
|
Zou Y, Luo W, Guo J, Luo Q, Deng M, Lu Z, Fang Y, Zhang CC. NK cell-mediated anti-leukemia cytotoxicity is enhanced using a NKG2D ligand MICA and anti-CD20 scfv chimeric protein. Eur J Immunol 2018; 48:1750-1763. [PMID: 30063799 DOI: 10.1002/eji.201847550] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2018] [Revised: 07/06/2018] [Accepted: 07/30/2018] [Indexed: 01/08/2023]
Abstract
NK cells are important innate cytotoxic lymphocytes that have potential in treatment of leukemia. Engagement of NKG2D receptor on NK cells enhances the target cytotoxicity. Here, we produced a fusion protein consisting of the extracellular domain of the NKG2D ligand MICA and the anti-CD20 single-chain variable fragment (scfv). This recombinant protein is capable of binding both NK cells and CD20+ tumor cells. Using a human NKG2D reporter cell system we developed, we showed that this fusion protein could decorate CD20+ tumor cells with MICA extracellular domain and activate NK through NKG2D. We further demonstrated that this protein could specifically induce the ability of a NK cell line (NKL) and primary NK cells to lyse CD20+ leukemia cells. Moreover, we found that downregulation of surface HLA class I expression in the target cells improved NKL-mediated killing. Our results demonstrated that this recombinant protein specifically lyses leukemia cells by NK cells, which may lead to development of a novel strategy for treating leukemia and other tumors.
Collapse
Affiliation(s)
- Yizhou Zou
- Department of Immunology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Weiguang Luo
- Department of Immunology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- Department of Physiology, UT Southwestern Medical Center at Dallas, TX, USA
| | - Jing Guo
- Department of Immunology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Qizhi Luo
- Department of Immunology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Mi Deng
- Department of Physiology, UT Southwestern Medical Center at Dallas, TX, USA
| | - Zhigang Lu
- Department of Physiology, UT Southwestern Medical Center at Dallas, TX, USA
| | - Yi Fang
- Department of Physiology, UT Southwestern Medical Center at Dallas, TX, USA
| | - Cheng Cheng Zhang
- Department of Physiology, UT Southwestern Medical Center at Dallas, TX, USA
| |
Collapse
|
9
|
Chowdhry M, Makroo RN, Kakkar B, Thakur Y, Kumar M, Singh M. A positive complement dependent cytotoxicity immunoglobulin G crossmatch due to auto-antibodies with a negative luminex bead assays in a renal transplant recipient: A Diagnostic dilemma. Asian J Transfus Sci 2018; 12:160-164. [PMID: 30692803 PMCID: PMC6327756 DOI: 10.4103/ajts.ajts_95_17] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 11/10/2017] [Indexed: 11/26/2022] Open
Abstract
Transplant recipients are always at a risk of developing anti-human leukocyte antigen (HLA) antibodies due to prior sensitizing events such as blood transfusions, multiple pregnancies, or transplantation. Unexpected positive outcomes can be seen in complement dependent cytotoxicity (CDC) based assays due to underlying autoimmune disorders or pharmacological treatment (rituximab/intravenous immunoglobulin/anti-thymocyte globulin administration), therefore, limiting its value. CDC based assay results strongly depend on the vitality of the donor lymphocytes, highlighting another major limitation of this assay. Thus, as an alternative approach, solid phase based crossmatch assays were introduced which function independently of the cell quality and have higher sensitivity and specificity in detecting anti-HLA antibodies. We describe a case where the patient awaiting renal transplantation from living related donor was evaluated by pretransplant histocompatibility testing for the detection of anti-HLA antibodies. The histocompatibility testing revealed positive CDC anti-human globulin and flow crossmatch along with negative Luminex based assays (HLA antibody screen, luminex crossmatch, and luminex single bead assay). Detailed histocompatibility workup revealed immunoglobulin G autoantibodies which were complement activating and lympocytoxic in nature.
Collapse
Affiliation(s)
- Mohit Chowdhry
- Department of Transfusion Medicine and Immunology, Indraprastha Apollo Hospitals, New Delhi, India
| | - Raj Nath Makroo
- Department of Transfusion Medicine and Immunology, Indraprastha Apollo Hospitals, New Delhi, India
| | - Brinda Kakkar
- Department of Transfusion Medicine, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Yogita Thakur
- Department of Transfusion Medicine and Immunology, Indraprastha Apollo Hospitals, New Delhi, India
| | - Manoj Kumar
- Department of Transfusion Medicine and Immunology, Indraprastha Apollo Hospitals, New Delhi, India
| | - Mandhata Singh
- Department of Transfusion Medicine and Immunology, Indraprastha Apollo Hospitals, New Delhi, India
| |
Collapse
|
10
|
Kang ES, Choi SI, Park YH, Park GB, Jang HR. Results of Questionnaire Survey of Current Immune Monitoring Practice of Transplant Clinicians and Clinical Pathologists in Korea: Basis for Establishment of Harmonized Immune Monitoring Guidelines. KOREAN JOURNAL OF TRANSPLANTATION 2018. [DOI: 10.4285/jkstn.2018.32.2.13] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Affiliation(s)
- Eun-Suk Kang
- Department of Laboratory Medicine and Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Soo In Choi
- Department of Laboratory Medicine and Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Youn Hee Park
- Department of Laboratory Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Geum Borae Park
- Department of Laboratory Medicine, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Korea
| | - Hye Ryon Jang
- Department of Internal Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
11
|
Abstract
PURPOSE OF REVIEW In recent years, there has been increasing awareness and appreciation for the role of humoral immune responses in lung allograft rejection. This review summarizes our current understanding of this role and the associated challenges. RECENT FINDINGS Recent studies have described a syndrome of acute antibody-mediated rejection with a generally poor response to therapy and a high mortality. In addition, there is significant evidence implicating donor-specific human leukocyte antigen antibodies in the development of chronic lung allograft dysfunction. However, the optimal intervention to mitigate the risk of chronic lung allograft dysfunction after donor-specific human leukocyte antigen antibodies development remains unclear. SUMMARY There is mounting evidence that humoral immune responses play an important role in lung allograft rejection. However, therapeutic implications of this increased awareness have been limited. Indeed, there is insufficient evidence to adequately guide therapy, and the optimal treatment is unknown.
Collapse
|
12
|
Daniel V, Süsal C, Scherer S, Tran H, Gombos P, Trojan K, Sadeghi M, Morath C, Opelz G. Endothelial precursor cell cross-match using Tie-2-enriched spleen cells. Clin Transplant 2017; 31. [PMID: 28925558 DOI: 10.1111/ctr.13118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/28/2017] [Indexed: 11/28/2022]
Abstract
BACKGROUND Non-HLA antibodies against human endothelial progenitor cells (EPC) in pre-transplant recipient serum can have a deleterious influence on the graft. EPC enriched from peripheral blood have been commonly used for EPC cross-matching. In the present study, we describe cross-matches using EPC enriched from fresh or frozen-thawed spleen cell preparations, thereby widening the sample source for deceased-donor cross-matching and retrospective studies. METHODS EPC cross-matches were performed retrospectively using spleen cells and the flow cytometric XM-ONE cross-match test kit. RESULTS Healthy controls (n = 28) showed no IgG antibodies against EPC. When sera of 11 random dialysis patients were studied, 2 patients (18%) exhibited IgG EPC antibodies. When pre-transplant sera of 20 kidney graft recipients with good long-term graft outcome (serum creatinine 1.0 ± 0.2 mg/dL measured 2463 ± 324 days post-transplant) were investigated using frozen-thawed and then separated Tie-2-enriched spleen cells of the original transplant donor, 3 patients (15%) had pre-transplant IgG EPC antibodies. When pre-transplant sera of 5 patients with intra-operative graft loss were studied employing the original donor spleen cells, 4 (80%) patients showed IgG EPC antibodies. CONCLUSIONS Cross-matches with spleen cell-derived EPC using the XM-ONE assay are technically possible. Our very preliminary experience suggests clinical relevance.
Collapse
Affiliation(s)
- Volker Daniel
- Transplantation-Immunology, Institute of Immunology, University-Hospital Heidelberg, Heidelberg, Germany
| | - Caner Süsal
- Transplantation-Immunology, Institute of Immunology, University-Hospital Heidelberg, Heidelberg, Germany
| | - Sabine Scherer
- Transplantation-Immunology, Institute of Immunology, University-Hospital Heidelberg, Heidelberg, Germany
| | - Hien Tran
- Transplantation-Immunology, Institute of Immunology, University-Hospital Heidelberg, Heidelberg, Germany
| | - Petra Gombos
- Transplantation-Immunology, Institute of Immunology, University-Hospital Heidelberg, Heidelberg, Germany.,Department of Surgery, University-Hospital Heidelberg, Heidelberg, Germany
| | - Karina Trojan
- Transplantation-Immunology, Institute of Immunology, University-Hospital Heidelberg, Heidelberg, Germany
| | - Mahmoud Sadeghi
- Transplantation-Immunology, Institute of Immunology, University-Hospital Heidelberg, Heidelberg, Germany.,Department of Surgery, University-Hospital Heidelberg, Heidelberg, Germany
| | - Christian Morath
- Department of Nephrology, Center for Internal Medicine, University-Hospital Heidelberg, Heidelberg, Germany
| | - Gerhard Opelz
- Transplantation-Immunology, Institute of Immunology, University-Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
13
|
Tan K, Zheng K, Li D, Lu H, Wang S, Sun X. Impact of adipose tissue or umbilical cord derived mesenchymal stem cells on the immunogenicity of human cord blood derived endothelial progenitor cells. PLoS One 2017; 12:e0178624. [PMID: 28562647 PMCID: PMC5451078 DOI: 10.1371/journal.pone.0178624] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 05/16/2017] [Indexed: 12/30/2022] Open
Abstract
The application of autologous endothelial progenitor cell (EPC) transplantation is a promising approach in therapeutic cardiovascular diseases and ischemic diseases. In this study, we compared the immunogenicity of EPCs, adipose tissue (AD)-derived mesenchymal stem cells (MSCs) and umbilical cord (UC)-derived MSCs by flow cytometry and the mixed lymphocyte reaction. The impact of AD-MSCs and UC-MSCs on the immunogenicity of EPCs was analyzed by the mixed lymphocyte reaction and cytokine secretion in vitro and was further tested by allogenic peripheral blood mononuclear cell (PBMC) induced immuno-rejection on a cell/matrigel graft in an SCID mouse model. EPCs and AD-MSCs express higher levels of MHC class I than UC-MSCs. All three kinds of cells are negative for MHC class II. UC-MSCs also express lower levels of IFN-γ receptor mRNA when compared with EPCs and AD-MSCs. EPCs can stimulate higher rates of proliferation of lymphocytes than AD-MSCs and UC-MSCs. Furthermore, AD-MSCs and UC-MSCs can modulate immune response and inhibit lymphocyte proliferation induced by EPCs, mainly through inhibition of the proliferation of CD8+ T cells. Compared with UC-MSCs, AD-MSCs can significantly improve vessel formation and maintain the integrity of neovascular structure in an EPC+MSC/matrigel graft in SCID mice, especially under allo-PBMC induced immuno-rejection. In conclusion, our study shows that AD-MSC is a powerful candidate to minimize immunological rejection and improve vessel formation in EPC transplantation treatment.
Collapse
Affiliation(s)
- Kefang Tan
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Sciences, Central South University, Changsha, Hunan, China
- National Engineering and Research Center of Human Stem Cell, Changsha, Hunan, China
| | - Ke Zheng
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Sciences, Central South University, Changsha, Hunan, China
- National Engineering and Research Center of Human Stem Cell, Changsha, Hunan, China
| | - Daiye Li
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Sciences, Central South University, Changsha, Hunan, China
- National Engineering and Research Center of Human Stem Cell, Changsha, Hunan, China
| | - Haiyuan Lu
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Sciences, Central South University, Changsha, Hunan, China
- Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Siqi Wang
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Sciences, Central South University, Changsha, Hunan, China
- National Engineering and Research Center of Human Stem Cell, Changsha, Hunan, China
| | - Xuan Sun
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Sciences, Central South University, Changsha, Hunan, China
- National Engineering and Research Center of Human Stem Cell, Changsha, Hunan, China
- * E-mail:
| |
Collapse
|
14
|
Baranwal AK, Mehra NK. Major Histocompatibility Complex Class I Chain-Related A (MICA) Molecules: Relevance in Solid Organ Transplantation. Front Immunol 2017; 8:182. [PMID: 28293239 PMCID: PMC5329007 DOI: 10.3389/fimmu.2017.00182] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2016] [Accepted: 02/08/2017] [Indexed: 11/27/2022] Open
Abstract
An ever growing number of reports on graft rejection and/or failure even with good HLA matches have highlighted an important role of non-HLA antigens in influencing allograft immunity. The list of non-HLA antigens that have been implicated in graft rejection in different types of organ transplantation has already grown long. Of these, the Major Histocompatibility Complex class I chain-related molecule A (MICA) is one of the most polymorphic and extensively studied non-HLA antigenic targets especially in the kidney transplantation. Humoral response to MICA antigens has repeatedly been associated with lower graft survival and an increased risk of acute and chronic rejection following kidney and liver transplantation with few studies showing conflicting results. Although there are clear indications of MICA antibodies being associated with adverse graft outcome, a definitive consensus on this relationship has not been arrived yet. Furthermore, only a few studies have dealt with the impact of MICA donor-specific antibodies as compared to those that are not donor specific on graft outcome. In addition to the membrane bound form, a soluble isoform of MICA (sMICA), which has the potential to engage the natural killer cell-activating receptor NKG2D resulting in endocytosis and degradation of receptor–ligand interaction complex leading to suppression of NKG2D-mediated host innate immunity, has been a subject of intense discussion. Most studies on sMICA have been directed toward understanding their influence on tumor growth, with limited literature focusing its role in transplant biology. Furthermore, a unique dimorphism (methionine to valine) at position 129 in the α2 domain categorizes MICA alleles into strong (MICA-129 met) and weak (MICA-129 val) binders of NKG2D receptor depending on whether they have methionine or valine at this position. Although the implications of MICA 129 dimorphism have been highlighted in hematopoietic stem cell transplantation, its role in solid organ transplantation is yet to be explored. This review summarizes the currently available information on MICA antibodies, soluble MICA, and MICA-129 dimorphism in a setting of solid organ transplantation.
Collapse
Affiliation(s)
| | - Narinder K Mehra
- All India Institute of Medical Sciences , New Delhi, Delhi , India
| |
Collapse
|
15
|
Risti M, Bicalho MDG. MICA and NKG2D: Is There an Impact on Kidney Transplant Outcome? Front Immunol 2017; 8:179. [PMID: 28289413 PMCID: PMC5326783 DOI: 10.3389/fimmu.2017.00179] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 02/07/2017] [Indexed: 01/06/2023] Open
Abstract
This paper aims to present an overview of MICA and natural killer group 2 member D (NKG2D) genetic and functional interactions and their impact on kidney transplant outcome. Organ transplantation has gone from what can accurately be called a “clinical experiment” to a routine and reliable practice, which has proven to be clinically relevant, life-saving and cost-effective when compared with non-transplantation management strategies of both chronic and acute end-stage organ failures. The kidney is the most frequently transplanted organ in the world (transplant-observatory1). The two treatment options for end-stage renal disease (ESRD) are dialysis and/or transplantation. Compared with dialysis, transplantation is associated with significant improvements in quality of life and overall longevity. A strong relationship exists between allograft loss and human leukocyte antigens (HLA) antibodies (Abs). HLA Abs are not the only factor involved in graft loss, as multiple studies have shown that non-HLA antigens are also involved, even when a patient has a good HLA matche and receives standard immunosuppressive therapy. A deeper understanding of other biomarkers is therefore important, as it is likely to lead to better monitoring (and consequent success) of organ transplants. The objective is to fill the void left by extensive reviews that do not often dive this deep into the importance of MICA and NKG2D in allograft acceptance and their partnership in the immune response. There are few papers that explore the relationship between these two protagonists when it comes to kidney transplantation. This is especially true for the role of NKG2D in kidney transplantation. These reasons give a special importance to this review, which aims to be a helpful tool in the hands of researchers in this field.
Collapse
Affiliation(s)
- Matilde Risti
- LIGH - Immunogenetics and Histocompatibility Laboratory, Federal University of Paraná , Curitiba , Brazil
| | - Maria da Graça Bicalho
- LIGH - Immunogenetics and Histocompatibility Laboratory, Federal University of Paraná , Curitiba , Brazil
| |
Collapse
|
16
|
Analysis of Sera of Recipients with Allograft Rejection Indicates That Keratin 1 Is the Target of Anti-Endothelial Antibodies. J Immunol Res 2017; 2017:8679841. [PMID: 28265584 PMCID: PMC5318619 DOI: 10.1155/2017/8679841] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 01/04/2017] [Indexed: 11/24/2022] Open
Abstract
Anti-endothelial cell antibodies (AECAs) are usually directed against the surface antigens on the vascular endothelial cells. Clinical studies suggest a pathogenic role for nonhuman leukocyte antigen in antibody-mediated rejection; however, the antigens on the donor vascular endothelium that serve as the first-line targets for an immune response during allograft rejection have not been fully identified. Here, we used immunoprecipitation and mass spectrometry to identify antigens from the sera of kidney transplant recipients who were experiencing antibody-mediated rejection. Keratin 1 (KRT1) was identified as a novel antigenic target expressed on endothelial cells. To validate our finding, we produced recombinant proteins representing the three most common alleles of KRT1. The serum used for immunoprecipitation showed a strong reaction to KRT1 recombinants in western blot and ELISA. In the kidney transplant cohort, more AECA-positive recipients than AECA-negative recipients had KRT1 antibodies (32.2% versus 11.9%, p = 0.002). Sera from 255 renal recipients were tested by ELISA. Of the 77 recipients with deteriorating graft function (serum creatinine > 120 μmol/L), 23 had anti-KRT1 antibodies. KRT1-IgG positivity was, therefore, associated with a higher risk of kidney transplant rejection (29.9% (23/77) versus 16.9% (30/178), p = 0.0187). A better understanding of this antigenic target will improve long-term allograft survival.
Collapse
|
17
|
|
18
|
Xu Y, Dou D, Ran X, Liu C, Chen J. Integrative analysis of proteomics and metabolomics of anaphylactoid reaction induced by Xuesaitong injection. J Chromatogr A 2015; 1416:103-11. [DOI: 10.1016/j.chroma.2015.09.019] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2015] [Revised: 08/07/2015] [Accepted: 09/07/2015] [Indexed: 11/30/2022]
|