Journal of the Pancreas Open Access

  • ISSN: 1590-8577
  • Journal h-index: 80
  • Journal CiteScore: 29.12
  • Journal Impact Factor: 19.45*
  • Average acceptance to publication time (5-7 days)
  • Average article processing time (30-45 days) Less than 5 volumes 30 days
    8 - 9 volumes 40 days
    10 and more volumes 45 days
Reach us +44 7460731551

Mini Review - (2024) Volume 25, Issue 1

Exploring Genetic Factors in Pancreatic Disorders: A Review of Etiological Insights
Michael Brown*#
 
1Italy
#Equally contribution
 
*Correspondence: Michael Brown, Italy, Email:

, Manuscript No. ipp-24-19029 ; Published: 19-Feb-2024, DOI: 10.35841/1590-8577-25.1.843

Abstract

          

Introduction

Pancreatic disorders represent a significant health burden globally, with conditions such as pancreatic cancer, chronic pancreatitis, and pancreatic cysts posing considerable challenges in diagnosis and treatment. While environmental factors such as diet and lifestyle play crucial roles, genetic predispositions also contribute significantly to the development of these disorders. Understanding the genetic underpinnings of pancreatic disorders is vital for elucidating disease mechanisms, identifying at-risk individuals, and developing targeted therapies. This review provides a comprehensive examination of genetic factors associated with pancreatic disorders, highlighting recent advances and promising avenues for further research [1].

Pancreatic cancer is one of the deadliest malignancies, characterized by its aggressive nature and poor prognosis. While somatic mutations in genes such as KRAS, TP53, CDKN2A, and SMAD4 are well-established drivers of pancreatic cancer development, inherited genetic variants also play a significant role. Studies have identified several susceptibility loci associated with familial pancreatic cancer, including mutations in BRCA1, BRCA2, PALB2, ATM, and STK11. These genes are involved in DNA repair pathways, highlighting the importance of genomic stability in pancreatic tumorigenesis [2].

Furthermore, germline mutations in genes associated with hereditary cancer syndromes, such as Lynchsyndrome and familial adenomatous polyposis (FAP), increase the risk of pancreatic cancer development. Advances in next-generation sequencing technologies have facilitated the identification of rare susceptibility variants with moderate to high penetrance, providing valuable insights into the genetic architecture of pancreatic cancer [3].

Chronic pancreatitis is characterized by persistent inflammation of the pancreas, leading to irreversible damage and fibrosis. While alcohol abuse and smoking are well-established risk factors, genetic factors also contribute to disease susceptibility. Mutations in genes encoding digestive enzymes such as PRSS1, SPINK1, and CFTR disrupt pancreatic function and predispose individuals to chronic pancreatitis. Additionally, variants in genes involved in immune regulation and inflammatory pathways, including CTRC and CPA1, have been implicated in disease pathogenesis [4].

Family-based studies have revealed clustering of chronic pancreatitis cases, supporting the role of genetic predisposition in disease etiology. Furthermore, gene-environment interactions may modulate disease penetrance, with certain genetic variants conferring susceptibility only in the presence of specific environmental triggers. Elucidating the interplay between genetic and environmental factors is crucial for understanding disease heterogeneity and guiding personalized treatment strategies for chronic pancreatitis patients [5].

Pancreatic cysts are fluid-filled lesions that range from benign to premalignant or malignant neoplasms. While most pancreatic cysts are incidental findings and remain asymptomatic, a subset can progress to invasive pancreatic cancer. Genetic alterations drive the progression of pancreatic cysts through various molecular pathways, including activation of KRAS signaling, inactivation of tumor suppressor genes, and dysregulation of DNA repair mechanisms [6].

Genetic factors significantly contribute to pancreatic disorders, including pancreatic cancer, chronic pancreatitis, and pancreatic cysts. While environmental influences play a role, understanding the genetic underpinnings is crucial for disease management. In pancreatic cancer, mutations in genes like KRAS and TP53 are well-known, but inherited variants in BRCA1, BRCA2, and others also increase risk. Chronic pancreatitis often involves mutations in digestive enzyme genes like PRSS1 and SPINK1. Pancreatic cysts, potentially malignant, exhibit mutations in genes like GNAS and RNF43. Integrating genetic insights with environmental factors offers promise for personalized treatment and improved patient outcomes in these challenging conditions [7].

Recent genomic profiling studies have identified recurrent mutations in genes such as GNAS, RNF43, and CTNNB1 in different subtypes of pancreatic cysts, providing insights into their molecular classification and pathogenesis. Furthermore, germline mutations in genes associated with hereditary pancreatic cancer syndromes, such as PRSS1 and SPINK1, predispose individuals to the development of specific cystic lesions. Understanding the genetic basis of pancreatic cysts is essential for risk stratification, early detection, and surveillance of high-risk individuals [8].

Despite significant advances in understanding the genetic basis of pancreatic disorders, several challenges remain. The complex interplay between genetic, environmental, and lifestyle factors complicates risk assessment and disease prediction. Moreover, the heterogeneous nature of pancreatic disorders necessitates large-scale collaborative efforts to identify rare genetic variants with clinical significance. Integrating multi-omics data, including genomics, transcriptomics, and epigenomics, holds promise for elucidating disease mechanisms and identifying novel therapeutic targets [9].

Additionally, leveraging artificial intelligence and machine learning algorithms can facilitate the integration of diverse datasets and enhance predictive modeling for personalized medicine approaches. Finally, translating genetic discoveries into clinical practice requires interdisciplinary collaboration among researchers, clinicians, and policymakers to ensure equitable access to genetic testing and targeted therapies for individuals at risk of pancreatic disorders [10].

Conclusion

Genetic factors play a significant role in the etiology of pancreatic disorders, including pancreatic cancer, chronic pancreatitis, and pancreatic cysts. Advances in genomic technologies have uncovered a complex landscape of genetic susceptibility variants, providing valuable insights into disease mechanisms and risk stratification. Integrating genetic information with clinical and environmental data holds promise for personalized risk assessment, early detection, and targeted therapies for individuals at risk of pancreatic disorders. Continued research efforts are needed to unravel the intricate genetic networks underlying pancreatic pathologies and translate these findings into clinical practice to improve patient outcomes

References

Indexed at, Google Scholar, Cross Ref

[PMID: 35883588]

Indexed at, Google Scholar, Cross Ref

Indexed at, Google Scholar, Cross Ref

Indexed at, Google Scholar, Cross Ref

Indexed at, Google Scholar, Cross Ref

Indexed at, Google Scholar, Cross Ref

Indexed at, Google Scholar, Cross Ref

[ PMID: 33374418]
Indexed at, Google Scholar, Cross Ref

[PMID: 31181644]

Indexed at, Google Scholar, Cross Ref

  1. Gentiluomo M, Canzian F, Nicolini A, Gemignani F, Landi S, Campa D. Germline genetic variability in pancreatic cancer risk and prognosis. Semin Cancer Biol. 2022;79:105-131. [PMID: 32818625]
  2. Mahoney AL, Nassif NT, O’Brien BA, Simpson AM. Pancreatic Transdifferentiation Using β-Cell Transcription Factors for Type 1 Diabetes Treatment. Cells. 2022;11(14):2145.
  3. Balakrishnan S, Dhavamani S, Prahalathan C. β-Cell specific transcription factors in the context of diabetes mellitus and β-cell regeneration. Mech Dev. 2020;163:103634. [PMID: 32711047]
  4. Sarwar A, Zhu M, Su Q, Zhu Z, Yang T, Chen Y, et al. Targeting mitochondrial dysfunctions in pancreatic cancer evokes new therapeutic opportunities. Crit Rev Oncol Hematol. 2022:103858. [PMID: 36257540]
  5. Sikdar N, Saha G, Dutta A, Ghosh S, Shrikhande SV, Banerjee S. Genetic alterations of periampullary and pancreatic ductal adenocarcinoma: an overview. Curr Genomics. 2018;19(6):444-63. [PMID: 30258276]
  6. Mocci E, Milne RL, Hopper JL, John EM, Andrulis IL, Chung WK, et al. Risk of pancreatic cancer in breast cancer families from the breast cancer family registry. Cancer Epidemiol Biomarkers Prev. 2013;22(5):803-11. [PMID: 23456555]
  7. Gambella A, Kalantari S, Cadamuro M, Quaglia M, Delvecchio M, Fabris L, et al. The Landscape of HNF1B Deficiency: A Syndrome Not Yet Fully Explored. Cells. 2023;12(2):307. [PMID: 36672242]
  8. Elhag DA, Kumar M, Al Khodor S. Exploring the triple interaction between the host genome, the epigenome, and the gut microbiome in type 1 diabetes. Int J Mol Sci. 2020;22(1):125.
  9. Pontious C, Kaul S, Hong M, Hart PA, Krishna SG, Lara LF, et al. Cathepsin E expression and activity: Role in the detection and treatment of pancreatic cancer. Pancreatology. 2019;19(7):951-6. [PMID: 31582345]
                                                                                                                                                                                                            Indexed at, Google Scholar, Cross Ref
  10. Silva-Vaz P, Abrantes AM, Castelo-Branco M, Gouveia A, Botelho MF, Tralhão JG. Murine models of acute pancreatitis: a critical appraisal of clinical relevance. Int J Mol Sci. 2019;20(11):2794.

Copyright: 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 work is properly cited.