Nutritional Metabolomics and Nutrigenomics their Role in Breast Cancer: An Overview

Jump To References Section

Authors

  • Department of Biochemistry, Biotechnology and Bioinformatics Avinashilingam Institute for Home Science and Higher Education for Women Coimbatore - 641 043 ,IN
  • Department of Biochemistry, Biotechnology and Bioinformatics Avinashilingam Institute for Home Science and Higher Education for Women Coimbatore - 641 043 ,IN

DOI:

https://doi.org/10.21048/IJND.2024.61.2.43710

Keywords:

Nutrition, breast cancer, nutrigenomics, gut microbiota, cancer, phytonutrients

Abstract

Breast cancer physiology is still being studied, and a patient's diet may have an impact on both their risk of developing the disease and how they respond to treatment once they are diagnosed. Nutritional treatments are essential public health initiatives since good nutrition plays a noteworthy role in illness prevention. In this postgenomic medicineera, the combination of nutritional, genomic, and proteomic disciplines has given rise to nutrigenomics and nutriproteomics. In particular, nutrigenomics and nutriproteomics concentrate on the interactions between nutrients and the human genome and proteome, respectively, and offer intuitions into the role that nutrition plays in the progression of cancer. A deeper understanding of nutrition and its underlying causes is anticipated to come from additional omics disciplines such as metabonomics, interactomics, and microbiomics. For the creation of individualised diets for women at risk of breast cancer, these domains offer a hitherto unheard-of potential. It will assist in determining a person's unique nutritional needs based on their genetic makeup, a personalised diet, and the relationship between diet and chronic diseases like cancer, opening up new perspectives on the complexity of breast cancer and paving the way for better management of the disease.

Downloads

Download data is not yet available.

Metrics

Metrics Loading ...

Published

2024-06-11

How to Cite

S., S., & Chalos , V. A. (2024). Nutritional Metabolomics and Nutrigenomics their Role in Breast Cancer: An Overview. The Indian Journal of Nutrition and Dietetics, 61(2), 216–231. https://doi.org/10.21048/IJND.2024.61.2.43710

Issue

Section

Review Articles

 

References

Vahid, F., Hajizadeghan, K. and Khodabakhshi, A. Nutritional metabolomics in diet-breast cancer relations: Current research, challenges, and future directions - A review. Biomed., 2023, 11, 1845. http://dx.doi.org/10.3390/biomedicines11071845

Thorne, J.L., Holen, I. and Corfe, B.M. The Second Nutrition and Cancer Networking Meeting Nutrition and Breast Cancer: Translating Evidence into Practice. Proc. Nutr. Soci., 2023, 82, 58-62.

Go, Y., Chung, M. and Park, Y. Dietary patterns for women with triple-negative breast cancer and dense breasts. Nutr. Cancer, 2016, 68, 1281-1288.

Asghar, W. and Khalid, N. Nutrigenetics and nutrigenomics, and precision nutrition. Nutr. Health, 2023, 29, 169-170.

Malcomson, F.C. and Mathers, J.C. Translation of nutrigenomic research for personalised and precision nutrition for cancer prevention and for cancer survivors. Redox Biol., 2023, 62, 102710.

McGee, E.E., Kiblawi, R., Playdon, M.C. and Eliassen, A.H. Nutritional metabolomics in cancer epidemiology: Current trends, challenges, and future directions, Curr. Nutr. Repo., 2019, 8, 202.

McGee, E.E., Kiblawi, R., Playdon, M.C. and Eliassen, A.H. Nutritional metabolomics in cancer epidemiology: Current trends, challenges, and future directions. Curr. Nutr. Repo., 2019, 8, 187-201.

Crowder, S.L., Playdon, M.C., Gudenkauf, L.M., Ose, J., Gigic, B., Greathouse, L. and Figueiredo, J.C. A molecular approach to understanding the role of diet in cancer-related fatigue: Challenges and future opportunities. Nutr., 2022, 14, 1496.

Scalbert, A., Brennan, L., Manach, C., Andres-Lacueva, C., Dragsted, L.O., Draper, J. and Wishart, D.S. The food metabolome: A window over dietary exposure. The Am. J. Clin. Nutr., 2014, 99, 1286-1308.

Schmidt, D.R., Patel, R., Kirsch, D.G., Lewis, C.A., Vander Heiden, M.G. and Locasale, J.W. Metabolomics in cancer research and emerging applications in clinical oncology. CA: A Cancer J. Clinic., 2021, 71, 333-358.

Ojo, O. Nutrition and chronic conditions. Nutr., 2019, 11, 459.

Cole, J.B., Florez, J.C. and Hirschhorn, J.N. Comprehensive genomic analysis of dietary habits in UK Biobank identifies hundreds of genetic associations. Nature Communications, 2020, 11, 1467.

Admoun, C. and Mayrovitz, H.N. The Etiology of Breast Cancer. Exon Publications, 2022, 21-30.

Marcum, J.A. Nutrigenetics/nutrigenomics, personalized nutrition, and precision healthcare. Curr. Nutr. Reports, 2020, 9, 338-345.

Bhattacharya, T., Dutta, S., Akter, R., Rahman, M.H., Karthika, C., Nagaswarupa, H.P. and Bungau, S. Role of phytonutrients in nutrigenetics and nutrigenomics perspective in curing breast cancer. Biomolec., 2021, 11, 1176.

Behl, T., Kumar, K., Brisc, C., Rus, M., Nistor-Cseppento, D.C., Bustea, C. and Bungau, S. Exploring the multifocal role of phytochemicals as immunomodulators. Biomed. Pharmacother., 2021, 133, 110959.

Younas, M., Hano, C., Giglioli-Guivarc’h, N. and Abbasi, B.H. Mechanistic evaluation of phytochemicals in breast cancer remedy: Current understanding and future perspectives. RSC Advances, 2018, 8, 29714-29744.

Barbieri, R., Coppo, E., Marchese, A., Daglia, M., Sobarzo-Sánchez, E., Nabavi, S.F. and Nabavi, S.M. Phytochemicals for human disease: An update on plant-derived compounds antibacterial activity. Microbiol. Res., 2017, 196, 44-68.

Rahman, M.A., Hannan, M.A., Dash, R., Rahman, M.H., Islam, R., Uddin, M.J. and Rhim, H. Phytochemicals as a complement to cancer chemotherapy: Pharmacological modulation of the autophagy-apoptosis pathway. Front. Pharmacol., 2021, 12, 639628.

Selvakumar, P., Badgeley, A., Murphy, P., Anwar, H., Sharma, U., Lawrence, K. and Lakshmikuttyamma, A. Flavonoids and other polyphenols act as epigenetic modifiers in breast cancer. Nutr., 2020, 12, 761.

Sirerol, J A., Rodríguez, M.L., Mena, S., Asensi, M.A., Estrela, J.M. and Ortega, A.L. Role of natural stilbenes in the prevention of cancer. Oxidative Medicine and Cellular Longevity, 2016, 2016, 3128951.

Siraj, M.A., Islam, M.A., Al Fahad, M.A., Kheya, H.R., Xiao, J. and Simal-Gandara, J. Cancer chemo-preventive role of dietary terpenoids by modulating Keap1-Nrf2-ARE signaling system- A comprehensive update. Appl. Sci., 2021, 11, 10806.

Kamran, S., Sinniah, A., Abdulghani, M.A. and Alshawsh, M. A. Therapeutic potential of certain terpenoids as anticancer agents: A scoping review. Cancers, 2022, 14, 1100.

Chatterjee, P., Gupta, S. and Banerjee, S. Understanding the role of the natural warriors: Phytochemicals in breast cancer chemoprevention. In Recent Front. Phytochem., 2023, 261-293. Elsevier.

Abdel-Daim, M.M., Abo-EL-Sooud, K., Aleya, L., Bungǎu, S.G., Najda, A. and Saluja, R. Alleviation of drugs and chemicals toxicity: Biomedical value of antioxidants. Oxidative Medicine and Cellular Longevity, 2018, 2018.

Page, M.J., McKenzie, J.E., Bossuyt, P.M., Boutron, I., Hoffmann, T.C., Mulrow, C.D. and Moher, D. Updating guidance for reporting systematic reviews: Development of the PRISMA 2020 statement. J. Clin. Epidemiol., 2021, 134, 103-112.

Lou, C., Yokoyama, S., Saiki, I. and Hayakawa, Y. Selective anticancer activity of hirsutine against HER2‑-positive breast cancer cells by inducing DNA damage. Oncol. Reports, 2015, 33, 2072-2076.

Du, J., Li, J., Song, D., Li, Q., Li, L., Li, B. and Li, L. Matrine exerts anti‑-breast cancer activity by mediating apoptosis and protective autophagy via the AKT/mTOR pathway in MCF‑-7 cells. Molec. Med. Reports, 2020, 22, 3659-3666.

Dordevic, D., Capikova, J., Dordevic, S., Tremlová, B., Gajdács, M. and Kushkevych, I. Sulfur content in foods and beverages and its role in human and animal metabolism: A scoping review of recent studies. Heliyon, 2023

Kowalska, K., Habrowska-Górczyńska, D.E., Domińska, K., Urbanek, K.A. and Piastowska-Ciesielska, A.W. Methylsulfonylmethane (organic sulfur) induces apoptosis and decreases invasiveness of prostate cancer cells. Environ. Toxicol. Pharmacol., 2018, 64, 101-111.

Ha, A.W., Hong, K.H., Kim, H.S. and Kim, W.K. Inorganic sulfur reduces cell proliferation by inhibiting of ErbB2 and ErbB3 protein and mRNA expression in MDA-MB-231 human breast cancer cells. Nutr. Res. Prac., 2013, 7, 89-95.

Murtaugh, M.A., Herrick, J., Sweeney, C., Guiliano, A., Baumgartner, K., Byers, T. and Slattery, M. Macronutrient composition influence on breast cancer risk in Hispanic and non-Hispanic white women: The 4-Corners Breast Cancer Study. Nutr. Cancer, 2011, 63, 185-195.

Passarelli, S., Free, C.M., Allen, L.H., Batis, C., Beal, T., Biltoft-Jensen, A.P. and Golden, C.D. Estimating national and subnational nutrient intake distributions of global diets. The Am. J. Clin. Nutr., 2022, 116, 551-560.

Arnold, M., Morgan, E., Rumgay, H., Mafra, A., Singh, D., Laversanne, M. and Soerjomataram, I. Current and future burden of breast cancer: Global statistics for 2020 and 2040. The Breast, 2022, 66, 15-23.

Alfaro, Y., Delgado, G., Cárabez, A., Anguiano, B. and Aceves, C. Iodine and doxorubicin, a good combination for mammary cancer treatment: Antineoplastic adjuvancy, chemoresistance inhibition, and cardioprotection. Molec. Cancer, 2013, 12, 1-11.

Dijck-Brouwer, D.J., Muskiet, F.A., Verheesen, R.H., Schaafsma, G., Schaafsma, A. and Geurts, J.M. Thyroidal and extrathyroidal requirements for iodine and selenium: A combined evolutionary and (Patho) physiological approach. Nutr., 2022, 14, 3886.

He, S., Wang, B., Lu, X., Miao, S., Yang, F., Zava, T. and Shi, Y. E. Iodine stimulates estrogen receptor singling and its systemic level is increased in surgical patients due to topical absorption. Oncotarget, 2018, 9, 375.

Wang, Z. and Su, C. Effects of folic acid deficiency on genetic damage in colorectal cancer cells. Am. J. Translat. Res., 2023, 15, 3162.

Essén, A., Santaolalla, A., Garmo, H., Hammar, N., Walldius, G., Jungner, I. and Van Hemelrijck, M. Baseline serum folate, vitamin B12 and the risk of prostate and breast cancer using data from the Swedish AMORIS cohort. Cancer Causes Control., 2019, 30, 603-615.

Ismail, S., Eljazzar, S. and Ganji, V. Intended and unintended benefits of folic acid fortificationA narrative review. Foods, 2023, 12, 1612.

DePhillipo, N.N., Aman, Z.S., Kennedy, M.I., Begley, J.P., Moatshe, G. and LaPrade, R.F. Efficacy of vitamin C supplementation on collagen synthesis and oxidative stress after musculoskeletal injuries: A systematic review. Orthopaedic J. Sports Med., 2018, 6, 2325967118804544.

Carr, A.C. and Maggini, S. Vitamin C and immune function. Nutr., 2017, 9, 1211.

Rock, C.L., Thomson, C., Gansler, T., Gapstur, S.M., McCullough, M.L., Patel, A.V. and Doyle, C. American Cancer Society guideline for diet and physical activity for cancer prevention. CA: A Cancer J. Clinic., 2020, 70, 245-271.

Chungchunlam, S.M. and Moughan, P.J. Comparative bioavailability of vitamins in human foods sourced from animals and plants. Cri. Rev. Food Sci. Nutr., 2023, 1-36.

Didier, A.J., Stiene, J., Fang, L., Watkins, D., Dworkin, L.D. and Creeden, J.F. Antioxidant and anti-tumor effects of dietary vitamins A, C, and E. Antioxidants, 2023, 12, 632.

Dao, D.Q., Ngo, T.C., Thong, N.M. and Nam, P.C. Is vitamin A an antioxidant or a pro-oxidant? The J. Phys. Chem., 2017, 121, 9348-9357.

Pantavos, A., Ruiter, R., Feskens, E.F., de Keyser, C.E., Hofman, A., Stricker, B.H. and Kiefte-de Jong, J.C. Total dietary antioxidant capacity, individual antioxidant intake and breast cancer risk: the R otterdam study. Int. J. Cancer, 2015, 136, 2178-2186.

Zitvogel, L., Daillère, R., Roberti, M.P., Routy, B. and Kroemer, G. Anticancer effects of the microbiome and its products. Nature Rev. Microbiol., 2017, 15, 465-478.

Shreiner, A.B., Kao, J.Y. and Young, V.B. The gut microbiome in health and in disease. Curr. Opin. Gastroenterol., 2015, 31, 69.

Bhatt, A.P., Redinbo, M.R. and Bultman, S.J. The role of the microbiome in cancer development and therapy. CA: A Cancer J. Clinic., 2017, 67, 326-344.

Su, Q. and Liu, Q. Factors affecting gut microbiome in daily diet. Front. Nutr., 2021, 218.

Wu, N., Yang, X., Zhang, R., Li, J., Xiao, X., Hu, Y. and Zhu, B. Dysbiosis signature of fecal microbiota in colorectal cancer patients. Microb. Ecol., 2013, 66, 462-470.

Xuan, C., Shamonki, J.M., Chung, A., DiNome, M.L., Chung, M., Sieling, P.A. and Lee, D.J. Microbial dysbiosis is associated with human breast cancer. PloS one, 2014, 9, e83744.

Hill, E. B., Grainger, E.M., Young, G.S., Clinton, S.K. and Spees, C.K. Application of the updated WCRF/AICR cancer prevention score as an outcome for cancer survivors participating in a tailored and intensive dietary and physical activity intervention. Nutr., 2022, 14, 4751.

Murtaza, B., Hichami, A., Khan, A.S., Ghiringhelli, F. and Khan, N.A. Alteration in taste perception in cancer: Causes and strategies of treatment. Front. Physiol., 2017, 8, 134.

Szenderák, J., Fróna, D. and Rákos, M. Consumer acceptance of plant-based meat substitutes: A narrative review. Foods, 2022, 11, 1274.

Tang, H., Zhang, Y., Cao, B., Liang, Y., Na, R., Yang, Z. and Shang, L. Knowledge, attitudes and behaviors toward healthy eating among Chinese cancer patients treated with chemotherapy: A systematic review. Asia-Pacific J. Oncol. Nursing, 2022, 100163.

Lutes, C., Zelig, R. and Radler, D.R. Safety and feasibility of intermittent fasting during chemotherapy for breast cancer: A review of the literature. Topics in Clin. Nutr., 2020, 35, 168-177.

Tosti, V., Bertozzi, B. and Fontana, L. Health benefits of the Mediterranean diet: Metabolic and molecular mechanisms. The J. Gerontol. Series A., 2018, 73, 318-326.