GET THE APP

Vasoactive Intestinal Polypeptide and its Impact on Immune Modulation
..

Human Genetics & Embryology

ISSN: 2161-0436

Open Access

Perspective - (2023) Volume 14, Issue 4

Vasoactive Intestinal Polypeptide and its Impact on Immune Modulation

Srushti Dovgan*
*Correspondence: Srushti Dovgan, Department of Anatomy and Embryology, University of Sadat City, Sadat, Egypt, Email:
Department of Anatomy and Embryology, University of Sadat City, Sadat, Egypt

Received: 24-Oct-2023, Manuscript No. hgec-23-122612; Editor assigned: 26-Oct-2023, Pre QC No. P-122612; Reviewed: 09-Nov-2023, QC No. Q-122612; Revised: 14-Nov-2023, Manuscript No. R-122612; Published: 21-Nov-2023 , DOI: 10.37421/2161-0436.2023.14.226
Citation: Dovgan, Srushti. “Vasoactive Intestinal Polypeptide and its Impact on Immune Modulation.” Human Genet Embryol 14 (2023): 226.
Copyright: © 2023 Dovgan S. 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.

Introduction

Vasoactive Intestinal Polypeptide (VIP) is a neuropeptide that plays a crucial role in the regulation of various physiological processes, including immune modulation. Originally identified for its vasoactive properties in the intestine, VIP has since been recognized as a potent immunomodulation, influencing both innate and adaptive immune responses. This article explores the multifaceted roles of VIP in immune modulation, highlighting its impact on different components of the immune system and its therapeutic potential. VIP is a 28-amino acid neuropeptide belonging to the glucagon/secretin superfamily. It is synthesized and released by various cell types, including neurons, endocrine cells, and immune cells. VIP exerts its effects by binding to specific G protein-coupled receptors, namely VPAC1 and VPAC2 receptors, which are widely distributed in immune cells, as well as other tissues [1].

Description

VIP has been shown to dampen inflammatory responses by inhibiting the release of pro-inflammatory cytokines such as Interleukin-6 (IL-6), Tumour Necrosis Factor-alpha (TNF-α), and interleukin-1 beta (IL-1β). This antiinflammatory action contributes to the resolution of acute inflammation and may have implications for the treatment of inflammatory disorders. VIP can influence phagocytic activity in macrophages and neutrophils, enhancing the clearance of pathogens. Additionally, VIP promotes tolerogenic dendritic cell phenotypes, leading to altered antigen presentation and subsequent modulation of T cell responses. Understanding these mechanisms is essential for elucidating VIP's role in immune homeostasis. The relationship between the nervous and immune systems is dynamic, with bidirectional communication influencing physiological responses. VIP, being a neuropeptide, exemplifies this interaction by modulating both neuronal and immune cell activities. Understanding the crosstalk between the nervous and immune systems, particularly in the context of VIP signalling, provides insights into the broader regulatory networks governing immune function [2,3].

VIP exerts a profound influence on T cell differentiation and function. It has been reported to promote regulatory T cell development, contributing to immune tolerance and suppression of excessive immune responses. Furthermore, VIP can inhibit the proliferation and cytokine production of effector T cells, providing a regulatory checkpoint in adaptive immunity. While the impact of VIP on B cells is less understood compared to T cells, emerging evidence suggests that VIP may modulate B cell differentiation and antibody production. Further research is needed to elucidate the specific mechanisms underlying VIP's interactions with B cells and its implications for humoral immunity [4].

Given its immunosuppressive properties, VIP has been investigated as a potential therapeutic agent for autoimmune disorders such as rheumatoid arthritis, multiple sclerosis, and inflammatory bowel diseases. Clinical trials exploring the use of VIP or VIP receptor agonists in these conditions are underway, with promising preliminary results. VIP has been implicated in the regulation of allergic responses, particularly in the context of asthma and allergic rhinitis. Understanding the interplay between VIP and allergic inflammation may unveil novel therapeutic avenues for managing allergic disorders. Despite the promising therapeutic potential of VIP, several challenges exist in translating preclinical findings into clinical applications. Issues such as stability, delivery methods, and potential side effects need to be addressed. Additionally, further research is warranted to fully comprehend the intricate signalling pathways and cellular interactions involved in VIP-mediated immune modulation [5].

Conclusion

Vasoactive Intestinal Polypeptide stands out as a key player in immune modulation, exerting intricate control over both innate and adaptive immune responses. Its diverse effects on different immune cell types make it a potential therapeutic target for various immune-related disorders. Continued research into the mechanisms of VIP-mediated immune regulation and the development of targeted therapies hold promise for advancing our understanding of immune homeostasis and improving clinical outcomes in immune-mediated diseases. Ongoing research continues to unveil new aspects of VIP biology, including its roles in tissue-specific immunity, interactions with the micro biome, and involvement in chronic inflammatory conditions. Exploring these emerging research areas will deepen our understanding of VIP's contributions to immune regulation and may uncover novel therapeutic opportunities. Despite significant progress, many questions remain unanswered. Elucidating the precise molecular mechanisms underlying VIP's immunomodulatory effects, identifying specific downstream signalling pathways, and unravelling the complex interplay between VIP and other immune modulators are essential for maximizing the therapeutic potential of VIP-based interventions.

Acknowledgement

None.

Conflict of Interest

There are no conflicts of interest by author.

References

  1. De Herder, Wouter W., Jens F. Rehfeld, Mark Kidd and Irvin M. Modlin. "A short history of neuroendocrine tumours and their peptide hormones." Best Pract Res Clin Endocrinol Metab 30 (2016): 3-17.
  2. Google Scholar, Crossref, Indexed at

  3. Reindl, T., P. Degenhardt, W. Luck and T. Riebel, et al. "Der VIP-produzierende Tumor in der Padiatrie als differenzialdiagnose der protrahierten Diarrho." Klin Padiatr 217 (2004): 264-269.
  4. Google Scholar, Crossref, Indexed at

  5. Gonzalez, Carolina Bonilla, Juliana Rusinque, Camila Uribe and Ailim Carias, et al. "Pancreatic VIPoma as a differential diagnosis in chronic pediatric diarrhea: A case report and review of the literature." J Med Case Rep 12 (2021): 195.
  6. Google Scholar, Crossref, Indexed at

  7. Hussein, Mohammed Salah, RIdha A. Shaqaqiq and Khaled Mubarak Aldhuaina, et al. "Etiology, Assessment and Management of WDHA (Watery Diarrhea, Hypokalemia, and Achlorhydria) and VIPoma." J Pharm Res Int 33 (2021): 69-75.
  8. Google Scholar

  9. LeLeiko, Neal S., Michael S. Gee and Peter M. Sadow. "Case 13-2010: An 18.5-month-old girl with watery diarrhea and poor weight gain." N Engl J Med 362 (2010): 1619-1626.
  10. Google Scholar, Crossref, Indexed at

Google Scholar citation report
Citations: 309

Human Genetics & Embryology received 309 citations as per Google Scholar report

Human Genetics & Embryology peer review process verified at publons

Indexed In

 
arrow_upward arrow_upward