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 - An Overview on Transplantation Tolerance
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)
  • Editorial   
  • J Clin Exp Transplant 8: 161, Vol 8(2)

An Overview on Transplantation Tolerance

Helen Cribb*
Department of Veterinary and Agricultural Sciences, University of Melbourne, Australia
*Corresponding Author: Helen Cribb, Department of Veterinary and Agricultural Sciences, University of Melbourne, Australia, Email: cribbhel@gmail.com

Received: 01-Mar-2023 / Manuscript No. jcet-23-92547 / Editor assigned: 04-Mar-2023 / PreQC No. jcet-23-92547 / Reviewed: 18-Mar-2023 / QC No. jcet-23-92547 / Revised: 24-Mar-2023 / Manuscript No. jcet-23-92547 / Published Date: 30-Mar-2023

Abstract

Organ transplantation has become a common practice in modern medicine. With advancements in surgical techniques and immunosuppressive therapies, the success rates of organ transplantation have significantly increased, providing patients with a new lease of life. However, transplantation is not without its challenges. The immune system of recipients recognizes transplanted organs as foreign, triggering an immune response that can lead to graft rejection. This process is mediated by various immune cells and molecules, including T cells, B cells, antibodies, cytokines, and complement proteins. To prevent graft rejection, patients receive immunosuppressive drugs that suppress their immune system, but these drugs have side effects and can increase the risk of infections and cancer. Therefore, there is a need for alternative strategies to induce transplantation tolerance, which refers to the state of immune unresponsiveness towards the transplanted organ. Transplantation tolerance can eliminate or reduce the need for immunosuppressive drugs and improve the long-term outcomes of transplantation. In this article, we will review the current understanding of transplantation tolerance and the strategies that are being developed to achieve it.

Keywords

Organ transplantation; Immunosuppressive; Destruction

Introduction

Mechanisms of transplant rejection

Before discussing transplantation tolerance, it is important to understand the mechanisms of transplant rejection. Transplant rejection can occur through two main pathways: direct and indirect allorecognition. Direct allorecognition occurs when recipient T cells recognize donor MHC molecules presented on the surface of donor cells or donor APCs (antigen-presenting cells) in the context of self-MHC molecules [1]. This recognition leads to the activation of recipient T cells, which can then migrate to the transplanted organ and cause tissue damage through various effector mechanisms, such as cytotoxicity, cytokine release, and recruitment of inflammatory cells. Indirect all recognition, on the other hand, occurs when recipient APCs take up donor antigens from the transplanted organ and present them to recipient T cells in the context of self-MHC molecules. This recognition also leads to the activation of recipient T cells and the subsequent destruction of the transplanted organ [2].

Strategies for transplantation tolerance

To achieve transplantation tolerance, several strategies are being developed that aim to modify the immune system of recipients or the transplantation process itself. These strategies can be broadly classified into two categories: central and peripheral tolerance induction [3].

Central tolerance induction

Central tolerance induction aims to eliminate or suppress the donor-reactive T cells in the thymus, where T cells develop and undergo selection. This can be achieved by several methods, including:

• Thymic transplantation: Thymic transplantation involves the transplantation of the thymus gland from the donor into the recipient before or at the time of solid organ transplantation. This allows the recipient to develop a new population of T cells that are tolerant to the donor antigens. However, thymic transplantation is technically challenging and has limited clinical application [4].

• Hematopoietic stem cell transplantation (HSCT): HSCT involves the transplantation of hematopoietic stem cells from the donor into the recipient, which can differentiate into all blood cells, including T cells. If the donor cells engraft in the recipient’s bone marrow and generate a new immune system, the recipient may become tolerant to the donor antigens [5]. HSCT is currently used to treat certain blood disorders and cancers, but its application in solid organ transplantation is limited due to the risk of graft-versus-host disease (GVHD), a condition in which the donor immune cells attack the recipient’s tissues [6].

• Chimerism induction: Chimerism induction involves the administration of non-myeloablative (low-dose) conditioning regimens, followed by the infusion of donor bone marrow or peripheral blood stem cells. This results in the presence of both recipient and donor immune cells in the recipient’s body, leading to the development of mixed chimerism, in which the recipient’s immune system becomes tolerant to the donor antigens. Chimerism induction has been successful in inducing transplantation tolerance in animal models and some clinical trials [7].

Peripheral tolerance induction

Peripheral tolerance induction aims to suppress or redirect the donor-reactive T cells in the periphery, where they encounter the transplanted organ. This can be achieved by several methods, including:

• Costimulation blockade: Costimulation blockade involves the administration of antibodies that block the interaction between Costimulatory molecules on T cells and APCs, thereby inhibiting T cell activation. This approach has been successful in preventing rejection in animal models and some clinical trials, but its efficacy in inducing transplantation tolerance remains to be established [8].

• Treg cell therapy: Treg cells are a subset of CD4+ T cells that have immune-suppressive properties and play a role in maintaining peripheral tolerance. Treg cell therapy involves the isolation and expansion of Treg cells from the recipient or donor, followed by their infusion into the recipient before or after solid organ transplantation. This approach has shown promise in inducing transplantation tolerance in preclinical studies and some clinical trials [9].

• Gene editing: Gene editing technologies, such as CRISPR/ Cas9 [10]

Conclusion

This is one of the first retrospective comparison studies that evaluated the outcomes of heart transplantation using organs preserved and transported using the system compared with SCS. Although the group showed longer total allograft ischemic time, patients using required fewer units of blood product for perioperative transfusion and had similar early-term survival compared with SCS. Although a multicenter trial is warranted to further validate findings described in this study, accepting organs from a more remote location may be a safe and practical strategy to expand the donor pool by using advanced technologies, such as the system.

References

  1. Modell B, Darlison M (2008) Global epidemiology of haemoglobin disorders and derived service indicators. Bull World Health Organ 86:480–487.
  2. Indexed at, Google Scholar, Crossref

  3. Meng X, Gao X, Chen X, Yu J (2021) Umbilical cord-derived mesenchymal stem cells exert anti-fibrotic action on hypertrophic scar-derived fibroblasts in co-culture by inhibiting the activation of the TGF β1/Smad3 pathway. Exp Ther Med 21:210.
  4. Indexed at, Google Scholar, Crossref

  5. Yang LL, Lambert SR, Lynn MJ, Stulting RD (1999) Long-term results of corneal graft survival in infants and children with peters anomaly. Ophthalmology 106:833-848.
  6. Indexed at, Google Scholar, Crossref

  7. Binenbaum G, Zackai EH, Walker BM, Coleman K (2008) Sclerocornea associated with the chromosome 22q11.2 deletion syndrome. Am J Med Genet A 146: 904-909.
  8. Indexed at, Google Scholar, Crossref

  9. Pagano L, Shah H, Al Ibrahim O, Gadhvi KA, Coco G, et al. (2022) Update on Suture Techniques in Corneal Transplantation: A Systematic Review. J Clin Med 11:10-78.
  10. Indexed at, Google Scholar, Crossref

  11. Meng X, Gao X, Chen X, Yu J (2021) Umbilical cord-derived mesenchymal stem cells exert anti-fibrotic action on hypertrophic scar-derived fibroblasts in co-culture by inhibiting the activation of the TGF β1/Smad3 pathway. Exp Ther Med 21:210.
  12. Indexed at, Google Scholar, Crossref

  13. Matsuzaki T, Yoshizato K (1998) Role of hair papilla cells on induction and regeneration processes of hair follicles. Wound Repair Regen 6:524-530.
  14. Indexed at, Google Scholar, Crossref

  15. Musto P, Simeon V, Todoerti K, Neri A (2016) Primary Plasma Cell Leukemia: Identity Card 2016. Curr Treat Options Oncol 17(4):19-25.
  16. Indexed at, Google Scholar, Crossref

  17. Giordano G, Blanchini F, Bruno R, Colaneri P, di Filippo A, et al. (2020) Modelling the COVID-19 epidemic and implementation of population-wide interventions in Italy. Nature Medicine 26: 855–860.
  18. Indexed at, Google Scholar, Crossref

  19. Winskill P, Walker PGT, Griffin JT, Ghani AC (2017) Modelling the cost-effectiveness of introducing the RTS, S malaria vaccine relative to scaling up other malaria interventions in sub-Saharan Africa. BMJ Global Health 2: 1–10.
  20. Google Scholar, Crossref

Citation: Cribb H (2023) An Overview on Transplantation Tolerance. J Clin Exp Transplant 8: 161.

Copyright: © 2023 Cribb H. 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