IJMZ_2024v14n2

International Journal of Molecular Zoology 2024, Vol.14, No.2, 72-83 http://animalscipublisher.com/index.php/ijmz 80 8 Future Directions and Perspectives 8.1 Emerging trends and future research areas in immunotolerance enhancement The field of xenotransplantation has seen significant advancements, particularly with the use of genetically modified pigs to overcome immunological barriers. Emerging trends in this area include the development of multi-modified pigs through techniques such as "combineering" and "gene stacking", which allow for the integration of multiple xenoprotective transgenes at a single locus. This approach has shown promise in inhibiting short to mid-term xenograft rejection (Fischer et al., 2016). Additionally, the use of CRISPR/Cas9 technology to disrupt genes responsible for major xenoantigens has been effective in reducing antigenicity and improving pig-to-human compatibility (Li et al., 2021). Future research should focus on refining these genetic modifications and exploring new gene-editing techniques to further enhance immunotolerance. 8.2 Potential breakthroughs and technological innovations Potential breakthroughs in xenotransplantation are likely to come from advancements in both genetic engineering and immunosuppressive therapies. The successful use of genetically modified pig kidneys in brain-dead human recipients, which showed no signs of hyperacute or antibody-mediated rejection, is a significant milestone (Montgomery et al., 2022). Innovations in immunosuppressive regimens, such as the use of anti-CD40 monoclonal antibodies, have also demonstrated improved graft survival in preclinical models (Mohiuddin et al., 2014; Yamamoto et al., 2019). Furthermore, the development of in vitro methods to evaluate human-to-pig xeno-immune responses can accelerate the identification of ideal genetic modifications, thereby expediting the clinical application of xenotransplantation (Li et al., 2021). Continued research in these areas is essential for achieving long-term graft survival and reducing the need for lifelong immunosuppression. 8.3 Collaboration opportunities and interdisciplinary approaches The complexity of xenotransplantation necessitates a multidisciplinary approach, involving collaboration between geneticists, immunologists, transplant surgeons, and bioengineers. Interdisciplinary research can facilitate the development of more effective genetic modifications and immunosuppressive strategies. For instance, combining expertise in gene editing with advancements in immunology has already led to the creation of genetically engineered pigs that express human complement regulatory genes, significantly improving graft survival (Yang and Sykes, 2007; Fischer et al., 2016). Collaborative efforts should also focus on addressing the remaining physiological incompatibilities and zoonotic risks associated with xenotransplantation (Lu et al., 2020; Lei et al., 2022). By fostering partnerships across various scientific disciplines, the field can move closer to the goal of clinical application and ultimately solve the organ shortage crisis. In summary, the future of xenotransplantation lies in the continued refinement of genetic modifications, the development of innovative immunosuppressive therapies, and the fostering of interdisciplinary collaborations. These efforts will pave the way for successful clinical trials and the eventual widespread use of xenotransplantation to address the critical shortage of human organs. 9 Concluding Remarks The research on enhancing immunotolerance in genetically modified pigs for xenotransplantation has made significant strides. Key genetic modifications, such as the deletion of carbohydrate xenoantigens and the expression of human complement and coagulation-regulatory proteins, have been shown to improve graft survival and function. For instance, pigs with nine genetic modifications, including the deletion of all three known carbohydrate xenoantigens and the expression of human CD46, CD55, thrombomodulin, endothelial cell protein C receptor, human hemeoxygenase-1, and human CD47, have demonstrated promising results in preclinical studies. These modifications help mitigate hyperacute rejection, acute humoral xenograft rejection, and immune cell-mediated rejection, which are major barriers to successful xenotransplantation. The findings suggest that genetically modified pigs could potentially serve as viable organ donors for humans, addressing the critical shortage of human organs for transplantation. Future research should focus on optimizing the combination of genetic modifications to further enhance graft survival and function. Additionally, clinical trials are necessary to validate the efficacy and safety of these genetically modified pig organs in human recipients.

RkJQdWJsaXNoZXIy MjQ4ODYzNA==