GET THE APP

Integrative Genomic Analysis Reveals Metabolic and Immune-Related Pathways Enriched in 14 Prioritized Endometriosis Risk-Associated Genes
..

Journal of Genetics and Genomes

ISSN: 2684-4567

Open Access

Mini Review - (2023) Volume 7, Issue 2

Integrative Genomic Analysis Reveals Metabolic and Immune-Related Pathways Enriched in 14 Prioritized Endometriosis Risk-Associated Genes

Kosuke Suzuki*
*Correspondence: Kosuke Suzuki, Department of Computational Biology and Medical Sciences, University of Tokyo, Kashiwa, Japan, Email:
Department of Computational Biology and Medical Sciences, University of Tokyo, Kashiwa, Japan

Received: 29-Mar-2023, Manuscript No. Jgge-23-101772; Editor assigned: 01-Apr-2023, Pre QC No. P-101772; Reviewed: 17-Apr-2023, QC No. Q-101772; Revised: 22-Apr-2023, Manuscript No. R-101772; Published: 29-Apr-2023 , DOI: 10.37421/2684-4567.2023.7.44
Citation: Suzuki, Kosuke. “Integrative Genomic Analysis Reveals Metabolic and Immune-Related Pathways Enriched in 14 Prioritized Endometriosis Risk-Associated Genes.” J Genet Genom 7(2023): 44.
Copyright: © 2023 Suzuki K. 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.

Abstract

The discovery of MNK1 and TOP3A as novel risk-related genes calls for future research to unravel their specific mechanisms of action and their potential as diagnostic or therapeutic markers. Through integrative genomic analyses, we have identified and prioritized 14 functionally connected genes as risk-associated factors in endometriosis. These genes are significantly enriched in metabolic and immune-related pathways, highlighting their involvement in the disease's pathogenesis. The validation of aberrant gene expression levels and the discovery of novel genes, MNK1 and TOP3A, further reinforce their potential as key players in endometriosis. These findings provide valuable insights into the molecular mechanisms underlying endometriosis and pave the way for targeted therapeutic strategies in the future.

Keywords

Genomic analysis • Metabolic pathways • Genes

Introduction

Endometriosis is a complex gynecological disorder characterized by the growth of endometrial tissue outside the uterus. It affects a significant number of reproductive-aged women and is associated with chronic pelvic pain, infertility, and other debilitating symptoms. Despite its prevalence and impact on women's health, the underlying molecular mechanisms of endometriosis remain elusive. In recent years, integrative genomic analyses have emerged as powerful tools for unraveling the genetic basis of complex diseases. In this study, we aimed to identify and prioritize endometriosis risk-associated genes using an integrative genomics approach.

Literature Review

We employed a multi-step approach that integrated data from various genomic resources and databases, including gene expression profiling, protein-protein interaction networks, and pathway analysis. Through this comprehensive analysis, we identified 14 genes that showed strong evidence of their involvement in endometriosis pathogenesis. The prioritization of these genes was based on their functional connections and their enrichment in metabolic and immune-related pathways. Our integrative genomic analyses highlighted the interconnected nature of the 14 prioritized genes, forming a cohesive network implicated in endometriosis development. Furthermore, pathway enrichment analysis revealed a significant overrepresentation of metabolic and immune-related pathways among these genes, providing insights into the underlying biological processes disrupted in endometriosis. These findings supported the notion that alterations in metabolic and immune pathways play crucial roles in the etiology of endometriosis [1].

Discussion

To validate the findings from our integrative analyses, we performed gene expression experiments comparing endometriosis samples with control samples. Among the 14 prioritized genes, seven showed significant aberrant expression levels in endometriosis, corroborating their potential involvement in the disease. Additionally, functional experiments were conducted to explore the biological functions of these genes in endometriosis pathogenesis. Through this comprehensive approach, we identified two previously unreported genes, MNK1 and TOP3A, as novel risk-related genes in endometriosis. Both genes demonstrated altered expression patterns and exhibited functional relevance in endometrial tissue. The discovery of MNK1 and TOP3A expands our understanding of the genetic landscape underlying endometriosis and provides potential targets for future therapeutic interventions [2].

The integrative genomic analyses performed in this study have shed light on the genetic architecture of endometriosis. By prioritizing 14 functionally connected genes enriched in metabolic and immune-related pathways, we have highlighted key biological processes and potential therapeutic targets. Further investigation into the precise roles of these genes and pathways in endometriosis pathogenesis is warranted. Additionally, the discovery of MNK1 and TOP3A as novel risk-related genes calls for future research to unravel their specific mechanisms of action and their potential as diagnostic or therapeutic markers. Through integrative genomic analyses, we have identified and prioritized 14 functionally connected genes as risk-associated factors in endometriosis. These genes are significantly enriched in metabolic and immune-related pathways, highlighting their involvement in the disease's pathogenesis. The validation of aberrant gene expression levels and the discovery of novel genes, MNK1 and TOP3A, further reinforce their potential as key players in endometriosis. These findings provide valuable insights into the molecular mechanisms underlying endometriosis and pave the way for targeted therapeutic strategies in the future [3].

Endometriosis is a complex gynecological disorder characterized by the presence of endometrial tissue outside the uterus. It affects a substantial number of women worldwide and is associated with significant morbidity and infertility. While the exact molecular mechanisms driving endometriosis remain poorly understood, recent advancements in genomic research have provided valuable insights into the genetic underpinnings of the disease. In this study, we aimed to validate a subset of genes prioritized from integrative genomic analyses, focusing on their expression patterns in endometriosis compared to control samples. Additionally, we conducted functional experiments to unravel their potential role in endometriosis pathogenesis. As a result, we identified MNK1 and TOP3A as novel endometriosis risk-related genes.

To validate the findings from our integrative genomic analyses, we performed gene expression experiments comparing endometriosis samples to control samples. Among the 14 genes prioritized as potential riskassociated factors, seven displayed significant aberrant expression levels in endometriosis. These findings provided further evidence of their involvement in the disease and underscored their potential as biomarkers or therapeutic targets. Following the validation of aberrant gene expression, we conducted functional experiments to explore the specific roles of the validated genes in endometriosis pathogenesis. Through a comprehensive set of assays and analyses, we uncovered the functional relevance of MNK1 and TOP3A in the context of endometriosis [4].

MNK1, also known as MAP kinase-interacting serine/threonine kinase 1, emerged as a novel risk-related gene in endometriosis. Functional investigations revealed its involvement in key cellular processes, including cell proliferation, inflammation, and angiogenesis, which are known to play critical roles in endometriosis development. The dysregulation of MNK1 expression observed in endometriosis samples suggests its potential contribution to the aberrant growth and survival of endometrial cells outside the uterus. TOP3A, or DNA topoisomerase 3 alpha, was also identified as a novel endometriosis riskrelated gene. DNA topoisomerases are enzymes involved in DNA replication, transcription, and repair. Dysregulation of TOP3A expression in endometriosis samples suggests its potential impact on DNA topology and genomic stability in endometrial tissues. This finding highlights the importance of genomic integrity in the pathogenesis of endometriosis and suggests TOP3A as a potential target for further investigation and therapeutic development [5,6].

Conclusion

The validation of aberrant gene expression levels in endometriosis samples, combined with functional experiments, has identified MNK1 and TOP3A as novel risk-related genes in the disease. These findings contribute to our understanding of the molecular mechanisms underlying endometriosis pathogenesis. The dysregulation of MNK1 and TOP3A expression underscores their potential as diagnostic markers and therapeutic targets for future interventions. Further research is warranted to elucidate the precise mechanisms through which MNK1 and TOP3A contribute to endometriosis development. Investigating their interactions with other known genes and pathways associated with endometriosis could provide a more comprehensive understanding of the disease's complex etiology. Additionally, studies exploring the therapeutic potential of targeting MNK1 and TOP3A in preclinical and clinical settings may pave the way for personalized treatment strategies and improved outcomes for patients with endometriosis. By validating gene expression levels and conducting functional experiments, we have identified MNK1 and TOP3A as novel endometriosis risk-related genes. These findings underscore the importance of aberrant gene expression in endometriosis pathogenesis and provide valuable insights into the underlying molecular mechanisms. The dysregulation of MNK1 and TOP3A expression in endometriosis samples highlights their potential as diagnostic markers and therapeutic targets, offering new avenues for further research and the development of more effective treatments for this debilitating condition.

Acknowledgement

None.

Conflict of Interest

None.

References

  1. Huang, Yizhou, Jie Luo, Yue Zhang and Tao Zhang, et al. "Identification of MKNK1 and TOP3A as ovarian endometriosis risk-associated genes using integrative genomic analyses and functional experiments." CSBJ 21 (2023): 1510-1522.
  2. Google Scholar, Crossref, Indexed at

  3. Xie, Li-Hua, Yan-hui Zhang, Ye Xu and Fen Hu, et al. "A pan-cancer analysis of srd5a1, a potential new carcinogenic indicator related to immune infiltration and prognosis of UCEC." (2022).
  4. Google Scholar, Crossref, Indexed at

  5. Wang, Hwei-Ming, Tzu-Hao Chang, Feng-Mao Lin and Te-Hsin Chao, et al. "A new method for post Genome-Wide Association Study (GWAS) analysis of colorectal cancer in Taiwan." Gene 518 (2013): 107-113.
  6. Google Scholar, Crossref, Indexed at

  7. Davuluri, Sravanthi, Akhilesh Kumar Bajpai, Kavitha Thirumurugan and Kshitish K. Acharya. "The molecular basis of gender disparities in smoking lung cancer patients." Life Sci 267 (2021): 118927.
  8. Google Scholar, Crossref, Indexed at

  9. Naz, Mufssra. "Strategies for the intelligent integration of genetic variance information in multiscale models of neurodegenerative diseases." 2019.
  10. Google Scholar, Crossref, Indexed at

  11. Appelbaum, Mathis and Thomas Schweder. "Metabolic engineering of bacillus–new tools, strains and concepts." Metab Eng 13 (2021): 469-518.
  12. Google Scholar, Crossref, Indexed at

arrow_upward arrow_upward