Next Article in Journal
Expression Profiling of Coding and Noncoding RNAs in the Endometrium of Patients with Endometriosis
Previous Article in Journal
Prospective Variation of Cytokine Trends during COVID-19: A Progressive Approach from Disease Onset until Outcome
Previous Article in Special Issue
Integrating Genome-Scale Metabolic Models with Patient Plasma Metabolome to Study Endothelial Metabolism In Situ
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

Special Issue “Bioinformatics Study in Human Diseases: Integration of Omics Data for Personalized Medicine”

1
Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung 402, Taiwan
2
Research Assistant Center, Show Chwan Memorial Hospital, Changhua 500, Taiwan
3
Department of Pathology, Show Chwan Memorial Hospital, Changhua 500, Taiwan
4
National Institute of Cancer Research, National Health Research Institutes, Tainan 704, Taiwan
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(19), 10579; https://doi.org/10.3390/ijms251910579
Submission received: 27 September 2024 / Revised: 28 September 2024 / Accepted: 29 September 2024 / Published: 1 October 2024
The field of bioinformatics has made remarkable strides in recent years, revolutionizing our approach to understanding and treating human diseases. This Special Issue, “Bioinformatics Study in Human Diseases: Integration of Omics Data for Personalized Medicine”, showcases cutting-edge research that harnesses the power of bioinformatics and multi-omics integration to advance precision medicine.
The convergence of multiple scientific disciplines and technological advances has led to unprecedented growth in the field of multi-omics. This surge is evidenced by the more than doubling of multi-omics scientific publications within just two years (2022–2023) since its first referenced mention in 2002, as indexed by the National Library of Medicine [1]. Multi-omics approaches have demonstrated their capability to provide comprehensive insights into complex biological systems, representing a transformative force in health diagnostics and therapeutic strategies [2].
The 12 papers published in this issue demonstrate the breadth and depth of bioinformatics applications in various areas of medical research. From metabolic modeling to transcriptomics, these studies exemplify how computational approaches can unravel complex biological mechanisms and identify potential biomarkers and therapeutic targets.
The COVID-19 pandemic has highlighted the need for rapid and comprehensive bioinformatic analyses in understanding disease mechanisms. A study on COVID-19-induced pulmonary fibrosis demonstrates how bioinformatics can shed light on complex pathological processes. By analyzing publicly available data, the researchers show that COVID-19 infection drives endothelial lineage cells towards myofibroblasts, contributing to pulmonary fibrosis. This finding provides crucial insights into the long-term effects of COVID-19 and potential therapeutic targets [3].
One of the highlights is the innovative pipeline for integrating genome-scale metabolic models with patient plasma metabolome data, offering new insights into endothelial metabolism in trauma patients [4]. This approach aligns with the growing trend of integrating multiple omics layers to gain a more comprehensive understanding of biological systems [5,6].
In the realm of cancer research, several studies have made significant contributions. The computational analysis of FLT3 variations in acute myeloid leukemia provides valuable insights into the structural and functional impacts of mutations, potentially guiding future experimental and clinical studies [7]. Another study on cervical cancer elucidates the role of LAMB3 in promoting cancer cell migration, invasion, and survival [8]. These studies exemplify how multi-omics approaches can facilitate early disease detection and prevention, in addition to aiding in the discovery of crucial biomarkers for diagnosis, prognosis, and treatment monitoring [9,10].
Network-based approaches are applied in periodontitis research to identify master regulator transcription factors [11]. This aligns with the broader trend of using sophisticated computational utilities and stringent statistical methodologies to ensure accurate data interpretation in multi-omics research [12].
Several studies focus on the microbiome and its interactions with human health [13,14]. These studies contribute to the growing body of research recognizing the importance of the microbiome in multi-omics analyses and its impact on human health [15,16].
The exploration of epigenetic mechanisms in Alzheimer’s disease offers new perspectives on neurodegenerative disorders [17]. This research aligns with the broader goal of using multi-omics to provide a deep understanding of disease-associated molecular mechanisms [18,19].
In reproductive health, single-cell analysis reveals insights into testicular inflammation in idiopathic non-obstructive azoospermia [20]. This exemplifies the power of integrating various omics technologies to gain comprehensive insights into complex biological systems [21,22].
The application of supervised learning and multi-omics integration in kidney renal clear cell carcinoma research demonstrates the clinical significance of inner membrane mitochondrial protein (IMMT) in prognostic prediction and understanding the tumor immune microenvironment [23]. This study showcases how multi-omics can not only facilitate precision medicine by accounting for individual omics profiles [24,25] but also bridge the gaps between mitochondrial medicine and clinical oncology [26].
Other notable contributions include the identification of potential biomarkers for methamphetamine use disorder [27] and the elucidation of TGF-β signaling in saphenous vein graft failure [28], both of which have significant implications for personalized medicine and are in line with specific clinical scopes [29,30].
While multi-omics offers substantial promise, its research and large-scale application in healthcare present several challenges. These include cohesively integrating and normalizing data across varied omics platforms, managing the sheer volume and high dimensionality of multi-omics datasets, and addressing the ethical implications of managing sensitive health information [31,32].
For multi-omics to truly revolutionize healthcare, it demands rigorous validation, tangible real-world applications, and smooth integration into existing healthcare infrastructures. Key developments include targeted sampling methods, the use of artificial intelligence in formulating health indices, the integration of sophisticated n-of-1 statistical models such as digital twins, and the incorporation of blockchain technology for heightened data security [33,34].
As we move forward, the continued development and application of these computational approaches will undoubtedly play a crucial role in shaping the future of precision medicine. The field is poised to overcome the current challenges and realize its full potential in revolutionizing personalized healthcare [35,36].

Author Contributions

H.-Y.L.: Conceptualization, Methodology, Software, Validation, Investigation, Data curation, Writing—original draft, Visualization. P.-Y.C.: Conceptualization, Software, Validation, Resources, Writing—original draft, Writing—review & editing, Supervision, Funding acquisition. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the National Science and Technology Council, Taiwan (NSTC 112-2314-B-005-012 and 113-2314-B-442-002 (recipient H.-Y.L.); MOST 109-2314-B-442-001, NSTC 112-2314-B-442-001, and NSTC 113-2314-B-442-001-MY2 [recipient: P.-Y.C.]), the National Health Research Institutes (NHRI-109BCCO-MF-202015-01 [recipient: P.-Y.C.]), and Show Chwan Memorial Hospital, Taiwan (SRD-113016 [recipient: P.-Y.C.]).

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Mohr, A.E.; Ortega-Santos, C.P.; Whisner, C.M.; Klein-Seetharaman, J.; Jasbi, P. Navigating Challenges and Opportunities in Multi-Omics Integration for Personalized Healthcare. Biomedicines 2024, 12, 1496. [Google Scholar] [CrossRef] [PubMed]
  2. Hasin, Y.; Seldin, M.; Lusis, A. Multi-omics approaches to disease. Genome Biol. 2017, 18, 83. [Google Scholar] [CrossRef] [PubMed]
  3. Wu, X.; Zhang, D.; Bostrom, K.I.; Yao, Y. COVID-19 Infection May Drive EC-like Myofibroblasts towards Myofibroblasts to Contribute to Pulmonary Fibrosis. Int. J. Mol. Sci. 2023, 24, 11500. [Google Scholar] [CrossRef] [PubMed]
  4. Silva-Lance, F.; Montejano-Montelongo, I.; Bautista, E.; Nielsen, L.K.; Johansson, P.I.; Marin de Mas, I. Integrating Genome-Scale Metabolic Models with Patient Plasma Metabolome to Study Endothelial Metabolism In Situ. Int. J. Mol. Sci. 2024, 25, 5406. [Google Scholar] [CrossRef] [PubMed]
  5. Karczewski, K.J.; Snyder, M.P. Integrative omics for health and disease. Nat. Rev. Genet. 2018, 19, 299–310. [Google Scholar] [CrossRef]
  6. Subramanian, I.; Verma, S.; Kumar, S.; Jere, A.; Anamika, K. Multi-omics Data Integration, Interpretation, and Its Application. Bioinform. Biol. Insights 2020, 14, 1177932219899051. [Google Scholar] [CrossRef]
  7. Mirza, Z.; Al-Saedi, D.A.; Alganmi, N.; Karim, S. Landscape of FLT3 Variations Associated with Structural and Functional Impact on Acute Myeloid Leukemia: A Computational Study. Int. J. Mol. Sci. 2024, 25, 3419. [Google Scholar] [CrossRef]
  8. Wattanathavorn, W.; Seki, M.; Suzuki, Y.; Buranapraditkun, S.; Kitkumthorn, N.; Sasivimolrattana, T.; Bhattarakosol, P.; Chaiwongkot, A. Downregulation of LAMB3 Altered the Carcinogenic Properties of Human Papillomavirus 16-Positive Cervical Cancer Cells. Int. J. Mol. Sci. 2024, 25, 2535. [Google Scholar] [CrossRef]
  9. Chakraborty, S.; Sharma, G.; Karmakar, S.; Banerjee, S. Multi-OMICS approaches in cancer biology: New era in cancer therapy. Biochim. Biophys. Acta Mol. Basis Dis. 2024, 1870, 167120. [Google Scholar] [CrossRef]
  10. Menyhart, O.; Gyorffy, B. Multi-omics approaches in cancer research with applications in tumor subtyping, prognosis, and diagnosis. Comput. Struct. Biotechnol. J. 2021, 19, 949–960. [Google Scholar] [CrossRef]
  11. Vicencio, E.; Nunez-Belmar, J.; Cardenas, J.P.; Cortes, B.I.; Martin, A.J.M.; Maracaja-Coutinho, V.; Rojas, A.; Cafferata, E.A.; Gonzalez-Osuna, L.; Vernal, R.; et al. Transcriptional Signatures and Network-Based Approaches Identified Master Regulators Transcription Factors Involved in Experimental Periodontitis Pathogenesis. Int. J. Mol. Sci. 2023, 24, 14835. [Google Scholar] [CrossRef] [PubMed]
  12. Kaku, M.; Thant, L.; Dobashi, A.; Ono, Y.; Kitami, M.; Mizukoshi, M.; Arai, M.; Iwama, H.; Kitami, K.; Kakihara, Y.; et al. Multiomics analysis of cultured mouse periodontal ligament cell-derived extracellular matrix. Sci. Rep. 2024, 14, 354. [Google Scholar] [CrossRef] [PubMed]
  13. Mingaila, J.; Atzeni, A.; Burokas, A. A Comparison of Methods of Gut Microbiota Transplantation for Preclinical Studies. Int. J. Mol. Sci. 2023, 24, 12005. [Google Scholar] [CrossRef] [PubMed]
  14. Attia, H.; ElBanna, S.A.; Khattab, R.A.; Farag, M.A.; Yassin, A.S.; Aziz, R.K. Integrating Microbiome Analysis, Metabolomics, Bioinformatics, and Histopathology to Elucidate the Protective Effects of Pomegranate Juice against Benzo-alpha-pyrene-Induced Colon Pathologies. Int. J. Mol. Sci. 2023, 24, 10691. [Google Scholar] [CrossRef]
  15. Lloyd-Price, J.; Arze, C.; Ananthakrishnan, A.N.; Schirmer, M.; Avila-Pacheco, J.; Poon, T.W.; Andrews, E.; Ajami, N.J.; Bonham, K.S.; Brislawn, C.J.; et al. Multi-omics of the gut microbial ecosystem in inflammatory bowel diseases. Nature 2019, 569, 655–662. [Google Scholar] [CrossRef]
  16. Sun, B.; Wang, Y.; Bai, J.; Li, X.; Ma, L.; Man, S. Litchi Procyanidins Ameliorate DSS-Induced Colitis through Gut Microbiota-Dependent Regulation of Treg/Th17 Balance. J. Agric. Food Chem. 2024; ahead of print. [Google Scholar] [CrossRef]
  17. Yang, L.; Pang, X.; Guo, W.; Zhu, C.; Yu, L.; Song, X.; Wang, K.; Pang, C. An Exploration of the Coherent Effects between METTL3 and NDUFA10 on Alzheimer’s Disease. Int. J. Mol. Sci. 2023, 24, 10111. [Google Scholar] [CrossRef]
  18. Hampel, H.; Vergallo, A.; Perry, G.; Lista, S.; Alzheimer Precision Medicine, I. The Alzheimer Precision Medicine Initiative. J. Alzheimers Dis. 2019, 68, 1–24. [Google Scholar] [CrossRef]
  19. Zhou, C.; Guo, H.; Cao, S. Gene Network Analysis of Alzheimer’s Disease Based on Network and Statistical Methods. Entropy 2021, 23, 1365. [Google Scholar] [CrossRef]
  20. Xia, P.; Ouyang, S.; Shen, R.; Guo, Z.; Zhang, G.; Liu, X.; Yang, X.; Xie, K.; Wang, D. Macrophage-Related Testicular Inflammation in Individuals with Idiopathic Non-Obstructive Azoospermia: A Single-Cell Analysis. Int. J. Mol. Sci. 2023, 24, 8819. [Google Scholar] [CrossRef]
  21. Lahnemann, D.; Koster, J.; Szczurek, E.; McCarthy, D.J.; Hicks, S.C.; Robinson, M.D.; Vallejos, C.A.; Campbell, K.R.; Beerenwinkel, N.; Mahfouz, A.; et al. Eleven grand challenges in single-cell data science. Genome Biol. 2020, 21, 31. [Google Scholar] [CrossRef] [PubMed]
  22. Stuart, T.; Satija, R. Integrative single-cell analysis. Nat. Rev. Genet. 2019, 20, 257–272. [Google Scholar] [CrossRef] [PubMed]
  23. Chen, C.C.; Chu, P.Y.; Lin, H.Y. Supervised Learning and Multi-Omics Integration Reveals Clinical Significance of Inner Membrane Mitochondrial Protein (IMMT) in Prognostic Prediction, Tumor Immune Microenvironment and Precision Medicine for Kidney Renal Clear Cell Carcinoma. Int. J. Mol. Sci. 2023, 24, 8807. [Google Scholar] [CrossRef] [PubMed]
  24. Cheng, F.; Desai, R.J.; Handy, D.E.; Wang, R.; Schneeweiss, S.; Barabasi, A.L.; Loscalzo, J. Network-based approach to prediction and population-based validation of in silico drug repurposing. Nat. Commun. 2018, 9, 2691. [Google Scholar] [CrossRef]
  25. Ritchie, M.D.; Holzinger, E.R.; Li, R.; Pendergrass, S.A.; Kim, D. Methods of integrating data to uncover genotype-phenotype interactions. Nat. Rev. Genet. 2015, 16, 85–97. [Google Scholar] [CrossRef]
  26. Lin, H.Y.; Chu, P.Y. Mitochondrial calcium uniporter as biomarker and therapeutic target for breast cancer: Prognostication, immune microenvironment, epigenetic regulation and precision medicine. J. Adv. Res. 2024; ahead of print. [Google Scholar] [CrossRef]
  27. Jang, W.J.; Song, S.H.; Son, T.; Bae, J.W.; Lee, S.; Jeong, C.H. Identification of Potential Biomarkers for Diagnosis of Patients with Methamphetamine Use Disorder. Int. J. Mol. Sci. 2023, 24, 8672. [Google Scholar] [CrossRef]
  28. He, C.; Ye, P.; Zhang, X.; Esmaeili, E.; Li, Y.; Lu, P.; Cai, C. The Role of TGF-beta Signaling in Saphenous Vein Graft Failure after Peripheral Arterial Disease Bypass Surgery. Int. J. Mol. Sci. 2023, 24, 10381. [Google Scholar] [CrossRef]
  29. Chen, F.; Xu, Y.; Shi, K.; Zhang, Z.; Xie, Z.; Wu, H.; Ma, Y.; Zhou, Y.; Chen, C.; Yang, J.; et al. Multi-omics study reveals associations among neurotransmitter, extracellular vesicle-derived microRNA and psychiatric comorbidities during heroin and methamphetamine withdrawal. Biomed. Pharmacother. 2022, 155, 113685. [Google Scholar] [CrossRef]
  30. Michaud, M.E.; Mota, L.; Bakhtiari, M.; Thomas, B.E.; Tomeo, J.; Pilcher, W.; Contreras, M.; Ferran, C.; Bhasin, S.S.; Pradhan-Nabzdyk, L.; et al. Early Injury Landscape in Vein Harvest by Single-Cell and Spatial Transcriptomics. Circ. Res. 2024, 135, 110–134. [Google Scholar] [CrossRef]
  31. Alyass, A.; Turcotte, M.; Meyre, D. From big data analysis to personalized medicine for all: Challenges and opportunities. BMC Med. Genomics 2015, 8, 33. [Google Scholar] [CrossRef] [PubMed]
  32. Austin, C.P. Opportunities and challenges in translational science. Clin. Transl. Sci. 2021, 14, 1629–1647. [Google Scholar] [CrossRef] [PubMed]
  33. Bjornsson, B.; Borrebaeck, C.; Elander, N.; Gasslander, T.; Gawel, D.R.; Gustafsson, M.; Jornsten, R.; Lee, E.J.; Li, X.; Lilja, S.; et al. Digital twins to personalize medicine. Genome Med. 2019, 12, 4. [Google Scholar] [CrossRef] [PubMed]
  34. Kuo, T.T.; Kim, H.E.; Ohno-Machado, L. Blockchain distributed ledger technologies for biomedical and health care applications. J. Am. Med. Inform. Assoc. 2017, 24, 1211–1220. [Google Scholar] [CrossRef]
  35. Athieniti, E.; Spyrou, G.M. A guide to multi-omics data collection and integration for translational medicine. Comput. Struct. Biotechnol. J. 2023, 21, 134–149. [Google Scholar] [CrossRef]
  36. Pallocca, M.; Betti, M.; Baldinelli, S.; Palombo, R.; Bucci, G.; Mazzarella, L.; Tonon, G.; Ciliberto, G. Clinical bioinformatics desiderata for molecular tumor boards. Brief. Bioinform. 2024, 25, bbae447. [Google Scholar] [CrossRef]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Lin, H.-Y.; Chu, P.-Y. Special Issue “Bioinformatics Study in Human Diseases: Integration of Omics Data for Personalized Medicine”. Int. J. Mol. Sci. 2024, 25, 10579. https://doi.org/10.3390/ijms251910579

AMA Style

Lin H-Y, Chu P-Y. Special Issue “Bioinformatics Study in Human Diseases: Integration of Omics Data for Personalized Medicine”. International Journal of Molecular Sciences. 2024; 25(19):10579. https://doi.org/10.3390/ijms251910579

Chicago/Turabian Style

Lin, Hung-Yu, and Pei-Yi Chu. 2024. "Special Issue “Bioinformatics Study in Human Diseases: Integration of Omics Data for Personalized Medicine”" International Journal of Molecular Sciences 25, no. 19: 10579. https://doi.org/10.3390/ijms251910579

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop