Application and Challenges of Using Probiotic Lactobacillus and Bifidobacterium to Enhance Overall Health and Manage Diseases
Abstract
Simple Summary
Abstract
1. Introduction and Background
2. Mechanism of Action, Application, and Challenges of Lactobacillus and Bifidobacterium in Cancer Prevention and Their Role as Adjuvant Cancer Therapies
3. Mechanism of Action, Application, and Challenges of Utilizing Lactobacillus and Bifidobacterium to Restore Immune Function in Older Individuals
4. Mechanism of Action, Application, and Challenges of Utilizing Lactobacillus and Bifidobacterium in Autoimmune Disease Therapy
5. Mechanism of Action, Application, and Challenges of Utilizing Lactobacillus and Bifidobacterium in Neurodegenerative Disorders
6. Clinical Outcomes That Align with the Findings from the Preclinical Studies
7. Research Gaps Contributing to Inconsistencies Between Clinical Outcomes and the Findings from the Preclinical Studies
8. Conclusions
Funding
Conflicts of Interest
Abbreviations
| TLR | Toll-like receptor |
| Tregs | Regulatory T cells |
| Th | T helper |
| IL | Interleukin |
| TGF-β | Transforming growth factor-beta |
| IgA | Immunoglobulin A |
| DCs | Dendritic cells |
| SCFA | Short-chain fatty acids |
| MyD88 | Myeloid differentiation primary response 88 |
| DNA | Deoxyribonucleic acid |
| NK | Natural killer |
| CD8 | Cluster of differentiation 8 |
| IFN-γ | Interferon-gamma |
| iNOS | Inducible nitric oxide synthase |
| NO | Nitric oxide |
| NKG2D | Natural killer group 2, member D |
| CCR7 | C-C chemokine receptor type 7 |
| PD-1 | Programmed death protein-1 |
| CRP | C-reactive protein |
| GABA | Gamma-aminobutyric acid |
| IBS | Irritable bowel syndrome |
| IBD | Inflammatory bowel disease |
| RA | Rheumatoid arthritis |
| ALS | Amyotrophic lateral sclerosis |
| GBA | Gut–brain axis |
| CNS | Central nervous system |
| BDNF | Brain-derived neurotrophic factor |
References
- Metchnikoff, E. Essais Optimistes; A. Maloine: Paris, France, 1907; p. iii. 438p. [Google Scholar]
- Eslami, M.; Yousefi, B.; Kokhaei, P.; Hemati, M.; Nejad, Z.R.; Arabkari, V.; Namdar, A. Importance of probiotics in the prevention and treatment of colorectal cancer. J. Cell Physiol. 2019, 234, 17127–17143. [Google Scholar] [CrossRef]
- Sanders, M.E.; Merenstein, D.J.; Reid, G.; Gibson, G.R.; Rastall, R.A. Probiotics and prebiotics in intestinal health and disease: From biology to the clinic. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 605–616. [Google Scholar] [CrossRef]
- Chandrasekaran, P.; Weiskirchen, S.; Weiskirchen, R. Effects of Probiotics on Gut Microbiota: An Overview. Int. J. Mol. Sci. 2024, 25, 6022. [Google Scholar] [CrossRef]
- Gao, X.; Zhao, J.; Zhang, H.; Chen, W.; Zhai, Q. Modulation of gut health using probiotics: The role of probiotic effector molecules. J. Future Foods 2022, 2, 1–12. [Google Scholar] [CrossRef]
- Nogueira, D.S.; de Oliveira, L.M.; Amorim, C.C.O.; Gazzinelli-Guimarães, A.C.; Barbosa, F.S.; Oliveira, F.M.S.; Kraemer, L.; Mattos, M.; Cardoso, M.S.; Resende, N.M.; et al. Eosinophils mediate SIgA production triggered by TLR2 and TLR4 to control Ascaris suum infection in mice. PLoS Pathog. 2021, 17, e1010067. [Google Scholar] [CrossRef]
- Dikiy, S.; Rudensky, A.Y. Principles of regulatory T cell function. Immunity 2023, 56, 240–255. [Google Scholar] [CrossRef] [PubMed]
- Cristofori, F.; Dargenio, V.N.; Dargenio, C.; Miniello, V.L.; Barone, M.; Francavilla, R. Anti-Inflammatory and Immunomodulatory Effects of Probiotics in Gut Inflammation: A Door to the Body. Front. Immunol. 2021, 12, 578386. [Google Scholar] [CrossRef] [PubMed]
- Wang, X.; Zhang, P.; Zhang, X. Probiotics Regulate Gut Microbiota: An Effective Method to Improve Immunity. Molecules 2021, 26, 6076. [Google Scholar] [CrossRef]
- Lee, C.-G.; Cha, K.H.; Kim, G.-C.; Im, S.-H.; Kwon, H.-K. Exploring probiotic effector molecules and their mode of action in gut–immune interactions. FEMS Microbiol. Rev. 2023, 47, fuad046. [Google Scholar] [CrossRef] [PubMed]
- Angelin, J.; Kavitha, M. Exopolysaccharides from probiotic bacteria and their health potential. Int. J. Biol. Macromol. 2020, 162, 853–865. [Google Scholar] [CrossRef]
- Li, J.; Feng, S.; Yu, L.; Zhao, J.; Tian, F.; Chen, W.; Zhai, Q. Capsular polysaccarides of probiotics and their immunomodulatory roles. Food Sci. Hum. Wellness 2022, 11, 1111–1120. [Google Scholar] [CrossRef]
- Mazziotta, C.; Tognon, M.; Martini, F.; Torreggiani, E.; Rotondo, J.C. Probiotics Mechanism of Action on Immune Cells and Beneficial Effects on Human Health. Cells 2023, 12, 184. [Google Scholar] [CrossRef]
- Okeke, E.B.; Uzonna, J.E. The Pivotal Role of Regulatory T Cells in the Regulation of Innate Immune Cells. Front. Immunol. 2019, 10, 680. [Google Scholar] [CrossRef]
- Wang, R.; Yu, Y.F.; Yu, W.R.; Sun, S.Y.; Lei, Y.M.; Li, Y.X.; Lu, C.X.; Zhai, J.N.; Bai, F.R.; Ren, F.; et al. Roles of Probiotics, Prebiotics, and Postbiotics in B-Cell-Mediated Immune Regulation. J. Nutr. 2025, 155, 37–51. [Google Scholar] [CrossRef]
- Abbaszadeh, S.H.; Hosseini, S.R.A.; Mahmoodpoor, A.; Yousefi, M.; Lotfi-Dizaji, L.; Mameghani, M.E. Investigating the Role of Probiotics in Modulating T Cells and the Immune Response: A Systematic Review. Indian J. Microbiol. 2024. [Google Scholar] [CrossRef]
- Abedi, E.; Hashemi, S.M.B. Lactic acid production—Producing microorganisms and substrates sources-state of art. Heliyon 2020, 6, e04974. [Google Scholar] [CrossRef]
- Saito, S.; Cao, D.-Y.; Maekawa, T.; Tsuji, N.M.; Okuno, A. Lactococcus lactis subsp. cremoris C60 Upregulates Macrophage Function by Modifying Metabolic Preference in Enhanced Anti-Tumor Immunity. Cancers 2024, 16, 1928. [Google Scholar] [CrossRef] [PubMed]
- Saito, S.; Okuno, A.; Peng, Z.; Cao, D.Y.; Tsuji, N.M. Probiotic lactic acid bacteria promote anti-tumor immunity through enhanced major histocompatibility complex class I-restricted antigen presentation machinery in dendritic cells. Front. Immunol. 2024, 15, 1335975. [Google Scholar] [CrossRef] [PubMed]
- Wu, Z.; Xia, F.; Lin, R. Global burden of cancer and associated risk factors in 204 countries and territories, 1980–2021: A systematic analysis for the GBD 2021. J. Hematol. Oncol. 2024, 17, 119. [Google Scholar] [CrossRef] [PubMed]
- Luo, Q.; Smith, D.P. Global cancer burden: Progress, projections, and challenges. Lancet 2025, 406, 1536–1537. [Google Scholar] [CrossRef]
- Bowie, K. Cancer deaths expected to reach 18 million by 2050, major study forecasts. BMJ 2025, 390, r2022. [Google Scholar] [CrossRef]
- Force, L.M.; Kocarnik, J.M.; May, M.L.; Bhangdia, K.; Crist, A.; Penberthy, L.; Pritchett, N.; Acheson, A.; Deitesfeld, L.; A, B.; et al. The global, regional, and national burden of cancer, 1990–2023, with forecasts to 2050: A systematic analysis for the Global Burden of Disease Study 2023. Lancet 2025, 406, 1565–1586. [Google Scholar] [CrossRef]
- Calvert, M.; Wilson, R.; Peipert, J.D. Symptoms, side effects, quality of life and financial toxicity matter to patients with cancer, we need to do a better job of capturing and reporting these concepts in trials and clinical care. BMJ Oncol. 2025, 4, e000879. [Google Scholar] [CrossRef]
- Yang, Y.; Pan, M.; Xia, X.; Liang, J.; Yin, X.; Ju, Q.; Hao, J. Effect of dietary probiotics intake on cancer mortality: A cohort study of NHANES 1999–2018. Sci. Rep. 2025, 15, 959. [Google Scholar] [CrossRef] [PubMed]
- Pyo, Y.; Kwon, K.H.; Jung, Y.J. Probiotic Functions in Fermented Foods: Anti-Viral, Immunomodulatory, and Anti-Cancer Benefits. Foods 2024, 13, 2386. [Google Scholar] [CrossRef]
- Shah, A.B.; Baiseitova, A.; Zahoor, M.; Ahmad, I.; Ikram, M.; Bakhsh, A.; Shah, M.A.; Ali, I.; Idress, M.; Ullah, R.; et al. Probiotic significance of Lactobacillus strains: A comprehensive review on health impacts, research gaps, and future prospects. Gut Microbes 2024, 16, 2431643. [Google Scholar] [CrossRef] [PubMed]
- Faghfoori, Z.; Faghfoori, M.H.; Saber, A.; Izadi, A.; Yari Khosroushahi, A. Anticancer effects of bifidobacteria on colon cancer cell lines. Cancer Cell Int. 2021, 21, 258. [Google Scholar] [CrossRef]
- Aburjaile, F.F.; de Jesus, L.C.L.; da Silva, T.F.; Drumond, M.M.; de Oliveira Carvalho, R.D.; Azevedo, V.; Mancha-Agresti, P.D.C. Chapter 12—Lactic acid bacteria in gut microbiota, probiotics and disease prevention. In Lactic Acid Bacteria in Food Biotechnology; Ray, R.C., Paramithiotis, S., de Carvalho Azevedo, V.A., Montet, D., Eds.; Elsevier: Amsterdam, The Netherlands, 2022; pp. 207–219. [Google Scholar]
- Dempsey, E.; Corr, S.C. Lactobacillus spp. for Gastrointestinal Health: Current and Future Perspectives. Front. Immunol. 2022, 13, 840245. [Google Scholar] [CrossRef]
- Pei, B.; Peng, S.; Huang, C.; Zhou, F. Bifidobacterium modulation of tumor immunotherapy and its mechanism. Cancer Immunol. Immunother. 2024, 73, 94. [Google Scholar] [CrossRef] [PubMed]
- Sharma, D.; Gajjar, D.; Seshadri, S. Understanding the role of gut microfloral bifidobacterium in cancer and its potential therapeutic applications. Microbiome Res. Rep. 2024, 3, 3. [Google Scholar] [CrossRef]
- Amonkar, R.; Uy, A.A.; Ramirez, P.; Patel, H.; Jeong, J.J.; Shoyele, N.O.; Vaghela, V.; Lakshmikuttyamma, A. Engineered Bifidobacterium Strains Colonization at Tumor Sites: A Novel Approach to the Delivery of Cancer Treatments. Cancers 2025, 17, 2487. [Google Scholar] [CrossRef]
- Śliżewska, K.; Markowiak-Kopeć, P.; Śliżewska, W. The Role of Probiotics in Cancer Prevention. Cancers 2020, 13, 20. [Google Scholar] [CrossRef]
- Kahouli, I.; Malhotra, M.; Westfall, S.; Alaoui-Jamali, M.A.; Prakash, S. Design and validation of an orally administrated active L. fermentum-L. acidophilus probiotic formulation using colorectal cancer ApcMin/+mouse model. Appl. Microbiol. Biotechnol. 2017, 101, 1999–2019. [Google Scholar] [CrossRef]
- Hou, M.; Yu, Q.Q.; Yang, L.; Zhao, H.; Jiang, P.; Qin, L.; Zhang, Q. The role of short-chain fatty acid metabolism in the pathogenesis, diagnosis and treatment of cancer. Front. Oncol. 2024, 14, 1451045. [Google Scholar] [CrossRef]
- Jiang, Y.; Yang, Z. A functional and genetic overview of exopolysaccharides produced by Lactobacillus plantarum. J. Funct. Foods 2018, 47, 229–240. [Google Scholar] [CrossRef]
- Yang, X.; Yang, Z.; Wang, Y.; Zeng, H.; Wang, B. Proteomics and metabolomics elucidate the biosynthetic pathway of acid stress-induced exopolysaccharides and its impact on growth phenotypes in Lactiplantibacillus plantarum HMX2. Food Chem. 2025, 476, 143431. [Google Scholar] [CrossRef] [PubMed]
- Wang, Y.; Wang, Y.; Sun, T.; Xu, J. Bacteriocins in Cancer Treatment: Mechanisms and Clinical Potentials. Biomolecules 2024, 14, 831. [Google Scholar] [CrossRef] [PubMed]
- Thoda, C.; Touraki, M. Molecular Mechanisms of Probiotic Action Against Gastrointestinal Cancers. Int. J. Mol. Sci. 2025, 26, 7857. [Google Scholar] [CrossRef]
- Sankarapandian, V.; Venmathi Maran, B.A.; Rajendran, R.L.; Jogalekar, M.P.; Gurunagarajan, S.; Krishnamoorthy, R.; Gangadaran, P.; Ahn, B.C. An Update on the Effectiveness of Probiotics in the Prevention and Treatment of Cancer. Life 2022, 12, 59. [Google Scholar] [CrossRef] [PubMed]
- Liu, X.; Zhang, J.; Yi, T.; Li, H.; Tang, X.; Liu, D.; Wu, D.; Li, Y. Decoding tumor angiogenesis: Pathways, mechanisms, and future directions in anti-cancer strategies. Biomark. Res. 2025, 13, 62. [Google Scholar] [CrossRef]
- Liu, Z.-L.; Chen, H.-H.; Zheng, L.-L.; Sun, L.-P.; Shi, L. Angiogenic signaling pathways and anti-angiogenic therapy for cancer. Signal Transduct. Target. Ther. 2023, 8, 198. [Google Scholar] [CrossRef]
- Poggi, A.; Benelli, R.; Venè, R.; Costa, D.; Ferrari, N.; Tosetti, F.; Zocchi, M.R. Human Gut-Associated Natural Killer Cells in Health and Disease. Front. Immunol. 2019, 10, 961. [Google Scholar] [CrossRef]
- Aziz, N.; Bonavida, B. Activation of Natural Killer Cells by Probiotics. Immunopathol. Dis. Ther. 2016, 7, 41–55. [Google Scholar] [CrossRef]
- Saleena, L.A.K.; Teo, M.Y.M.; How, Y.H.; In, L.L.A.; Pui, L.P. Immunomodulatory action of Lactococcus lactis. J. Biosci. Bioeng. 2023, 135, 1–9. [Google Scholar] [CrossRef] [PubMed]
- Oerlemans, M.M.P.; Akkerman, R.; Ferrari, M.; Walvoort, M.T.C.; de Vos, P. Benefits of bacteria-derived exopolysaccharides on gastrointestinal microbiota, immunity and health. J. Funct. Foods 2021, 76, 104289. [Google Scholar] [CrossRef]
- Górska, A.; Przystupski, D.; Niemczura, M.J.; Kulbacka, J. Probiotic Bacteria: A Promising Tool in Cancer Prevention and Therapy. Curr. Microbiol. 2019, 76, 939–949. [Google Scholar] [CrossRef]
- Tarrah, A.; de Castilhos, J.; Rossi, R.C.; Duarte, V.d.S.; Ziegler, D.R.; Corich, V.; Giacomini, A. In vitro Probiotic Potential and Anti-cancer Activity of Newly Isolated Folate-Producing Streptococcus thermophilus Strains. Front. Microbiol. 2018, 9, 2214. [Google Scholar] [CrossRef]
- Jacouton, E.; Torres Maravilla, E.; Boucard, A.S.; Pouderous, N.; Pessoa Vilela, A.P.; Naas, I.; Chain, F.; Azevedo, V.; Langella, P.; Bermúdez-Humarán, L.G. Anti-tumoral Effects of Recombinant Lactococcus lactis Strain Secreting IL-17A Cytokine. Front. Microbiol. 2018, 9, 3355. [Google Scholar] [CrossRef]
- Zaharuddin, L.; Mokhtar, N.M.; Muhammad Nawawi, K.N.; Raja Ali, R.A. A randomized double-blind placebo-controlled trial of probiotics in post-surgical colorectal cancer. BMC Gastroenterol. 2019, 19, 131. [Google Scholar] [CrossRef] [PubMed]
- Vallejos, O.P.; Bueno, S.M.; Kalergis, A.M. Probiotics in inflammatory bowel disease: Microbial modulation and therapeutic prospects. Trends Mol. Med. 2025, 31, 731–742. [Google Scholar] [CrossRef]
- Sivan, A.; Corrales, L.; Hubert, N.; Williams, J.B.; Aquino-Michaels, K.; Earley, Z.M.; Benyamin, F.W.; Lei, Y.M.; Jabri, B.; Alegre, M.-L.; et al. Commensal Bifidobacterium promotes antitumor immunity and facilitates anti-PD-L1 efficacy. Science 2015, 350, 1084–1089. [Google Scholar] [CrossRef]
- Bui, V.T.; Tseng, H.C.; Kozlowska, A.; Maung, P.O.; Kaur, K.; Topchyan, P.; Jewett, A. Augmented IFN-gamma and TNF-alpha Induced by Probiotic Bacteria in NK Cells Mediate Differentiation of Stem-Like Tumors Leading to Inhibition of Tumor Growth and Reduction in Inflammatory Cytokine Release; Regulation by IL-10. Front. Immunol. 2015, 6, 576. [Google Scholar] [CrossRef]
- Rosenberg, J.; Huang, J. CD8(+) T Cells and NK Cells: Parallel and Complementary Soldiers of Immunotherapy. Curr. Opin. Chem. Eng. 2018, 19, 9–20. [Google Scholar] [CrossRef] [PubMed]
- Dunai, C.; Collins, C.P.; Barao, I.; Murphy, W.J. Chapter 1—NK cells and CD8 T cells in cancer immunotherapy: Similar functions by different mechanisms. In Successes and Challenges of NK Immunotherapy; Bonavida, B., Jewett, A., Eds.; Academic Press: Cambridge, MA, USA, 2021; pp. 3–31. [Google Scholar]
- Viel, S.; Vivier, E.; Walzer, T.; Marçais, A. Targeting metabolic dysfunction of CD8 T cells and natural killer cells in cancer. Nat. Rev. Drug Discov. 2025, 24, 190–208. [Google Scholar] [CrossRef] [PubMed]
- Kaur, K.; Cook, J.; Park, S.H.; Topchyan, P.; Kozlowska, A.; Ohanian, N.; Fang, C.; Nishimura, I.; Jewett, A. Novel Strategy to Expand Super-Charged NK Cells with Significant Potential to Lyse and Differentiate Cancer Stem Cells: Differences in NK Expansion and Function between Healthy and Cancer Patients. Front. Immunol. 2017, 8, 297. [Google Scholar] [CrossRef]
- Kaur, K.; Kozlowska, A.K.; Topchyan, P.; Ko, M.W.; Ohanian, N.; Chiang, J.; Cook, J.; Maung, P.O.; Park, S.H.; Cacalano, N.; et al. Probiotic-Treated Super-Charged NK Cells Efficiently Clear Poorly Differentiated Pancreatic Tumors in Hu-BLT Mice. Cancers 2019, 12, 63. [Google Scholar] [CrossRef]
- Kaur, K.; Topchyan, P.; Kozlowska, A.K.; Ohanian, N.; Chiang, J.; Maung, P.O.; Park, S.H.; Ko, M.W.; Fang, C.; Nishimura, I.; et al. Super-charged NK cells inhibit growth and progression of stem-like/poorly differentiated oral tumors in vivo in humanized BLT mice; effect on tumor differentiation and response to chemotherapeutic drugs. Oncoimmunology 2018, 7, e1426518. [Google Scholar] [CrossRef] [PubMed]
- Lopez-Cobo, S.; Pieper, N.; Campos-Silva, C.; Garcia-Cuesta, E.M.; Reyburn, H.T.; Paschen, A.; Vales-Gomez, M. Impaired NK cell recognition of vemurafenib-treated melanoma cells is overcome by simultaneous application of histone deacetylase inhibitors. Oncoimmunology 2018, 7, e1392426. [Google Scholar] [CrossRef]
- Kaur, K.; Ko, M.-W.; Ohanian, N.; Cook, J.; Jewett, A. Osteoclast-expanded super-charged NK-cells preferentially select and expand CD8+ T cells. Sci. Rep. 2020, 10, 20363. [Google Scholar] [CrossRef]
- Leivas, A.; Perez-Martinez, A.; Blanchard, M.J.; Martin-Clavero, E.; Fernandez, L.; Lahuerta, J.J.; Martinez-Lopez, J. Novel treatment strategy with autologous activated and expanded natural killer cells plus anti-myeloma drugs for multiple myeloma. Oncoimmunology 2016, 5, e1250051. [Google Scholar] [CrossRef]
- Kaur, K.; Topchyan, P.; Jewett, A. Supercharged Natural Killer (sNK) Cells Inhibit Melanoma Tumor Progression and Restore Endogenous NK Cell Function in Humanized BLT Mice. Cancers 2025, 17, 2430. [Google Scholar] [CrossRef]
- Kaur, K.; Reese, P.; Chiang, J.; Jewett, A. Natural Killer Cell Therapy Combined with Probiotic Bacteria Supplementation Restores Bone Integrity in Cancer by Promoting IFN-γ Production. Cells 2025, 14, 1347. [Google Scholar] [CrossRef]
- Agah, S.; Alizadeh, A.M.; Mosavi, M.; Ranji, P.; Khavari-Daneshvar, H.; Ghasemian, F.; Bahmani, S.; Tavassoli, A. More Protection of Lactobacillus acidophilus Than Bifidobacterium bifidum Probiotics on Azoxymethane-Induced Mouse Colon Cancer. Probiotics Antimicrob. Proteins 2019, 11, 857–864. [Google Scholar] [CrossRef]
- Gui, Q.F.; Lu, H.F.; Zhang, C.X.; Xu, Z.R.; Yang, Y.H. Well-balanced commensal microbiota contributes to anti-cancer response in a lung cancer mouse model. Genet. Mol. Res. 2015, 14, 5642–5651. [Google Scholar] [CrossRef]
- Urbanska, A.M.; Bhathena, J.; Cherif, S.; Prakash, S. Orally delivered microencapsulated probiotic formulation favorably impacts polyp formation in APC (Min/+) model of intestinal carcinogenesis. Artif. Cells Nanomed. Biotechnol. 2016, 44, 1–11. [Google Scholar] [CrossRef] [PubMed]
- Imani Fooladi, A.A.; Yazdi, M.H.; Pourmand, M.R.; Mirshafiey, A.; Hassan, Z.M.; Azizi, T.; Mahdavi, M.; Soltan Dallal, M.M. Th1 Cytokine Production Induced by Lactobacillus acidophilus in BALB/c Mice Bearing Transplanted Breast Tumor. Jundishapur J. Microbiol. 2015, 8, e17354. [Google Scholar] [CrossRef] [PubMed]
- Ohara, T.; Suzutani, T. Intake of Bifidobacterium longum and Fructo-oligosaccharides prevents Colorectal Carcinogenesis. Euroasian J. Hepatogastroenterol. 2018, 8, 11–17. [Google Scholar] [CrossRef] [PubMed]
- Fahmy, C.A.; Gamal-Eldeen, A.M.; El-Hussieny, E.A.; Raafat, B.M.; Mehanna, N.S.; Talaat, R.M.; Shaaban, M.T. Bifidobacterium longum Suppresses Murine Colorectal Cancer through the Modulation of oncomiRs and Tumor Suppressor miRNAs. Nutr. Cancer 2019, 71, 688–700. [Google Scholar] [CrossRef]
- Kuugbee, E.D.; Shang, X.; Gamallat, Y.; Bamba, D.; Awadasseid, A.; Suliman, M.A.; Zang, S.; Ma, Y.; Chiwala, G.; Xin, Y.; et al. Structural Change in Microbiota by a Probiotic Cocktail Enhances the Gut Barrier and Reduces Cancer via TLR2 Signaling in a Rat Model of Colon Cancer. Dig. Dis. Sci. 2016, 61, 2908–2920. [Google Scholar] [CrossRef]
- Walia, S.; Kamal, R.; Kanwar, S.S.; Dhawan, D.K. Cyclooxygenase as a target in chemoprevention by probiotics during 1,2-dimethylhydrazine induced colon carcinogenesis in rats. Nutr. Cancer 2015, 67, 603–611. [Google Scholar] [CrossRef]
- Mendes, M.C.S.; Paulino, D.S.; Brambilla, S.R.; Camargo, J.A.; Persinoti, G.F.; Carvalheira, J.B.C. Microbiota modification by probiotic supplementation reduces colitis associated colon cancer in mice. World J. Gastroenterol. 2018, 24, 1995–2008. [Google Scholar] [CrossRef] [PubMed]
- Hu, J.; Wang, C.; Ye, L.; Yang, W.; Huang, H.; Meng, F.; Shi, S.; Ding, Z. Anti-tumour immune effect of oral administration of Lactobacillus plantarum to CT26 tumour-bearing mice. J. Biosci. 2015, 40, 269–279. [Google Scholar] [CrossRef] [PubMed]
- Kassayová, M.; Bobrov, N.; Strojný, L.; Orendáš, P.; Demečková, V.; Jendželovský, R.; Kubatka, P.; Kisková, T.; Kružliak, P.; Adamkov, M.; et al. Anticancer and Immunomodulatory Effects of Lactobacillus plantarum LS/07, Inulin and Melatonin in NMU-induced Rat Model of Breast Cancer. Anticancer. Res. 2016, 36, 2719–2728. [Google Scholar]
- Wan Mohd Kamaluddin, W.N.F.; Rismayuddin, N.A.R.; Ismail, A.F.; Mohamad Aidid, E.; Othman, N.; Mohamad, N.A.H.; Arzmi, M.H. Probiotic inhibits oral carcinogenesis: A systematic review and meta-analysis. Arch. Oral Biol. 2020, 118, 104855. [Google Scholar] [CrossRef]
- Kita, A.; Fujiya, M.; Konishi, H.; Tanaka, H.; Kashima, S.; Iwama, T.; Ijiri, M.; Murakami, Y.; Takauji, S.; Goto, T.; et al. Probiotic-derived ferrichrome inhibits the growth of refractory pancreatic cancer cells. Int. J. Oncol. 2020, 57, 721–732. [Google Scholar] [CrossRef]
- Shin, R.; Itoh, Y.; Kataoka, M.; Iino-Miura, S.; Miura, R.; Mizutani, T.; Fujisawa, T. Anti-tumor activity of heat-killed Lactobacillus plantarum BF-LP284 on Meth-A tumor cells in BALB/c mice. Int. J. Food Sci. Nutr. 2016, 67, 641–649. [Google Scholar] [CrossRef]
- Wang, Y.H.; Yao, N.; Wei, K.K.; Jiang, L.; Hanif, S.; Wang, Z.X.; Pei, C.X. The efficacy and safety of probiotics for prevention of chemoradiotherapy-induced diarrhea in people with abdominal and pelvic cancer: A systematic review and meta-analysis. Eur. J. Clin. Nutr. 2016, 70, 1246–1253. [Google Scholar] [CrossRef]
- Schlienger de Alba, B.N.; Espinosa Andrews, H. Benefits and Challenges of Encapsulating Bifidobacterium Probiotic Strains with Bifidogenic Prebiotics. Probiotics Antimicrob. Proteins 2024, 16, 1790–1800. [Google Scholar] [CrossRef]
- Mejía-Caballero, A.; Salas-Villagrán, V.A.; Jiménez-Serna, A.; Farrés, A. Challenges in the production and use of probiotics as therapeuticals in cancer treatment or prevention. J. Ind. Microbiol. Biotechnol. 2021, 48, kuab052. [Google Scholar] [CrossRef] [PubMed]
- Hidalgo-Cantabrana, C.; Delgado, S.; Ruiz, L.; Ruas-Madiedo, P.; Sánchez, B.; Margolles, A. Bifidobacteria and Their Health-Promoting Effects. Microbiol. Spectr. 2017, 5, 73–98. [Google Scholar] [CrossRef]
- Xie, A.; Gao, M.; Du, H.; Pan, X. Next-generation probiotics delivery: Innovations and applications of single-cell encapsulation. Curr. Opin. Food Sci. 2025, 61, 101234. [Google Scholar] [CrossRef]
- Goyani, P.; Christodoulou, R.; Vassiliou, E. Immunosenescence: Aging and Immune System Decline. Vaccines 2024, 12, 1314. [Google Scholar] [CrossRef] [PubMed]
- Nguyen, T.Q.T.; Cho, K.A. Targeting immunosenescence and inflammaging: Advancing longevity research. Exp. Mol. Med. 2025, 57, 1881–1892. [Google Scholar] [CrossRef]
- Gounder, S.S.; Abdullah, B.J.J.; Radzuanb, N.; Zain, F.; Sait, N.B.M.; Chua, C.; Subramani, B. Effect of Aging on NK Cell Population and Their Proliferation at Ex Vivo Culture Condition. Anal. Cell. Pathol. 2018, 2018, 7871814. [Google Scholar] [CrossRef]
- Qi, C.; Liu, Q. Natural killer cells in aging and age-related diseases. Neurobiol. Dis. 2023, 183, 106156. [Google Scholar] [CrossRef]
- Brauning, A.; Rae, M.; Zhu, G.; Fulton, E.; Admasu, T.D.; Stolzing, A.; Sharma, A. Aging of the Immune System: Focus on Natural Killer Cells Phenotype and Functions. Cells 2022, 11, 1017. [Google Scholar] [CrossRef]
- Fasbender, F.; Widera, A.; Hengstler, J.G.; Watzl, C. Natural Killer Cells and Liver Fibrosis. Front. Immunol. 2016, 7, 19. [Google Scholar] [CrossRef]
- Kaur, K.; Jewett, A. Decreased surface receptors, function, and suboptimal osteoclasts-induced cell expansion in natural killer (NK) cells of elderly subjects. Aging 2025, 17, 798–821. [Google Scholar] [CrossRef] [PubMed]
- Li, X.; Li, C.; Zhang, W.; Wang, Y.; Qian, P.; Huang, H. Inflammation and aging: Signaling pathways and intervention therapies. Signal Transduct. Target. Ther. 2023, 8, 239. [Google Scholar] [CrossRef]
- Li, Z.; Zhang, Z.; Ren, Y.; Wang, Y.; Fang, J.; Yue, H.; Ma, S.; Guan, F. Aging and age-related diseases: From mechanisms to therapeutic strategies. Biogerontology 2021, 22, 165–187. [Google Scholar] [CrossRef] [PubMed]
- Yoo, J.Y.; Groer, M.; Dutra, S.V.O.; Sarkar, A.; McSkimming, D.I. Gut Microbiota and Immune System Interactions. Microorganisms 2020, 8, 1587. [Google Scholar] [CrossRef] [PubMed]
- Bosco, N.; Noti, M. The aging gut microbiome and its impact on host immunity. Genes Immun. 2021, 22, 289–303. [Google Scholar] [CrossRef] [PubMed]
- Guo, C.; Wu, M.; Huang, B.; Zhao, R.; Jin, L.; Fu, B.; Wang, P.; Wang, D.; Zheng, M.; Fang, J.; et al. Single-cell transcriptomics reveal a unique memory-like NK cell subset that accumulates with ageing and correlates with disease severity in COVID-19. Genome Med. 2022, 14, 46. [Google Scholar] [CrossRef]
- Mafe, A.N.; Edo, G.I.; Majeed, O.S.; Gaaz, T.S.; Akpoghelie, P.O.; Isoje, E.F.; Igbuku, U.A.; Owheruo, J.O.; Opiti, R.A.; Garba, Y.; et al. A review on probiotics and dietary bioactives: Insights on metabolic well-being, gut microbiota, and inflammatory responses. Food Chem. Adv. 2025, 6, 100919. [Google Scholar] [CrossRef]
- Boichuk, S.; Galembikova, A.; Vollmer, D. Enhancement of NK Cell Cytotoxic Activity and Immunoregulatory Effects of a Natural Product Supplement Across a Wide Age Span: A 30-Day In Vivo Human Study. Int. J. Mol. Sci. 2025, 26, 2897. [Google Scholar] [CrossRef]
- Virk, M.S.; Virk, M.A.; He, Y.; Tufail, T.; Gul, M.; Qayum, A.; Rehman, A.; Rashid, A.; Ekumah, J.-N.; Han, X.; et al. The Anti-Inflammatory and Curative Exponent of Probiotics: A Comprehensive and Authentic Ingredient for the Sustained Functioning of Major Human Organs. Nutrients 2024, 16, 546. [Google Scholar] [CrossRef]
- Zhao, Y.; Hong, K.; Zhao, J.; Zhang, H.; Zhai, Q.; Chen, W. Lactobacillus fermentum and its potential immunomodulatory properties. J. Funct. Foods 2019, 56, 21–32. [Google Scholar] [CrossRef]
- Qin, D.; Ma, Y.; Wang, Y.; Hou, X.; Yu, L. Contribution of Lactobacilli on Intestinal Mucosal Barrier and Diseases: Perspectives and Challenges of Lactobacillus casei. Life 2022, 12, 1910. [Google Scholar] [CrossRef]
- Zeng, Z.; Huang, Z.; Yue, W.; Nawaz, S.; Chen, X.; Liu, J. Lactobacillus plantarum modulate gut microbiota and intestinal immunity in cyclophosphamide-treated mice model. Biomed. Pharmacother. 2023, 169, 115812. [Google Scholar] [CrossRef] [PubMed]
- Yao, S.; Zhao, Z.; Wang, W.; Liu, X. Bifidobacterium Longum: Protection against Inflammatory Bowel Disease. J. Immunol. Res. 2021, 2021, 8030297. [Google Scholar] [CrossRef]
- Maftei, N.M.; Raileanu, C.R.; Balta, A.A.; Ambrose, L.; Boev, M.; Marin, D.B.; Lisa, E.L. The Potential Impact of Probiotics on Human Health: An Update on Their Health-Promoting Properties. Microorganisms 2024, 12, 234. [Google Scholar] [CrossRef]
- Gao, Y.; Liang, Q.; Sun, J.; Wu, X.; Song, Y.; Xu, Y.; Nie, H.; Huang, J.; Mu, G. Probiotic potential of lactic acid bacteria with antioxidant properties in modulating health: Mechanisms, applications, and future directions. Food Biosci. 2025, 66, 106181. [Google Scholar] [CrossRef]
- Feng, T.; Wang, J. Oxidative stress tolerance and antioxidant capacity of lactic acid bacteria as probiotic: A systematic review. Gut Microbes 2020, 12, 1801944. [Google Scholar] [CrossRef]
- Dell’Anno, M.; Giromini, C.; Reggi, S.; Cavalleri, M.; Moscatelli, A.; Onelli, E.; Rebucci, R.; Sundaram, T.S.; Coranelli, S.; Spalletta, A.; et al. Evaluation of Adhesive Characteristics of L. plantarum and L. reuteri Isolated from Weaned Piglets. Microorganisms 2021, 9, 1587. [Google Scholar] [CrossRef]
- Kumar, A.; Sivamaruthi, B.S.; Dey, S.; Kumar, Y.; Malviya, R.; Prajapati, B.G.; Chaiyasut, C. Probiotics as modulators of gut-brain axis for cognitive development. Front. Pharmacol. 2024, 15, 1348297. [Google Scholar] [CrossRef]
- O’Riordan, K.J.; Moloney, G.M.; Keane, L.; Clarke, G.; Cryan, J.F. The gut microbiota-immune-brain axis: Therapeutic implications. Cell Rep. Med. 2025, 6, 101982. [Google Scholar] [CrossRef]
- Ansari, F.; Neshat, M.; Pourjafar, H.; Jafari, S.M.; Samakkhah, S.A.; Mirzakhani, E. The role of probiotics and prebiotics in modulating of the gut-brain axis. Front. Nutr. 2023, 10, 1173660. [Google Scholar] [CrossRef]
- Dicks, L.M.T. Gut Bacteria and Neurotransmitters. Microorganisms 2022, 10, 1838. [Google Scholar] [CrossRef]
- Chen, Y.; Xu, J.; Chen, Y. Regulation of Neurotransmitters by the Gut Microbiota and Effects on Cognition in Neurological Disorders. Nutrients 2021, 13, 2099. [Google Scholar] [CrossRef]
- Wachamo, S.; Gaultier, A. The emerging role of microbiota derived SCFAs in neurodegenerative disorders. Brain Behav. Immun.—Health 2025, 46, 101012. [Google Scholar] [CrossRef]
- Guo, B.; Zhang, J.; Zhang, W.; Chen, F.; Liu, B. Gut microbiota-derived short chain fatty acids act as mediators of the gut-brain axis targeting age-related neurodegenerative disorders: A narrative review. Crit. Rev. Food Sci. Nutr. 2025, 65, 265–286. [Google Scholar] [CrossRef]
- Caetano-Silva, M.E.; Rund, L.; Hutchinson, N.T.; Woods, J.A.; Steelman, A.J.; Johnson, R.W. Inhibition of inflammatory microglia by dietary fiber and short-chain fatty acids. Sci. Rep. 2023, 13, 2819. [Google Scholar] [CrossRef]
- Li, S.C.; Hsu, W.F.; Chang, J.S.; Shih, C.K. Combination of Lactobacillus acidophilus and Bifidobacterium animalis subsp. lactis Shows a Stronger Anti-Inflammatory Effect than Individual Strains in HT-29 Cells. Nutrients 2019, 11, 969. [Google Scholar] [CrossRef]
- Aghamohammad, S.; Sepehr, A.; Miri, S.T.; Najafi, S.; Pourshafie, M.R.; Rohani, M. The Role of Combining Probiotics in Preventing and Controlling Inflammation: A Focus on the Anti-Inflammatory and Immunomodulatory Effects of Probiotics in an In Vitro Model of IBD. Can. J. Gastroenterol. Hepatol. 2022, 2022, 2045572. [Google Scholar] [CrossRef]
- Ku, S.; Haque, M.A.; Jang, M.J.; Ahn, J.; Choe, D.; Jeon, J.I.; Park, M.S. The role of Bifidobacterium in longevity and the future of probiotics. Food Sci. Biotechnol. 2024, 33, 2097–2110. [Google Scholar] [CrossRef]
- Setbo, E.; Campbell, K.; O’Cuiv, P.; Hubbard, R. Utility of Probiotics for Maintenance or Improvement of Health Status in Older People—A Scoping Review. J. Nutr. Health Aging 2019, 23, 364–372. [Google Scholar] [CrossRef]
- Hutchinson, A.N.; Bergh, C.; Kruger, K.; Sűsserová, M.; Allen, J.; Améen, S.; Tingö, L. The Effect of Probiotics on Health Outcomes in the Elderly: A Systematic Review of Randomized, Placebo-Controlled Studies. Microorganisms 2021, 9, 1344. [Google Scholar] [CrossRef]
- Huang, X.; Huang, L.; Lu, J.; Cheng, L.; Wu, D.; Li, L.; Zhang, S.; Lai, X.; Xu, L. The relationship between telomere length and aging-related diseases. Clin. Exp. Med. 2025, 25, 72. [Google Scholar] [CrossRef]
- Li, S.; Liu, Z.; Zhang, J.; Li, L. Links between telomere dysfunction and hallmarks of aging. Mutat. Res.—Genet. Toxicol. Environ. Mutagen. 2023, 888, 503617. [Google Scholar] [CrossRef]
- Shi, F.; Peng, J.; Li, H.; Liu, D.; Han, L.; Wang, Y.; Liu, Q.; Liu, Q. Probiotics as a targeted intervention in anti-ageing: A review. Biomarkers 2024, 29, 577–585. [Google Scholar] [CrossRef]
- Choudhary, P.; Kathuria, D.; Suri, S.; Bahndral, A.; Kanthi Naveen, A. Probiotics- its functions and influence on the ageing process: A comprehensive review. Food Biosci. 2023, 52, 102389. [Google Scholar] [CrossRef]
- Han, S.; Lu, Y.; Xie, J.; Fei, Y.; Zheng, G.; Wang, Z.; Liu, J.; Lv, L.; Ling, Z.; Berglund, B.; et al. Probiotic Gastrointestinal Transit and Colonization After Oral Administration: A Long Journey. Front. Cell Infect. Microbiol. 2021, 11, 609722. [Google Scholar] [CrossRef]
- Xiao, Y.; Huang, L.; Zhao, J.; Chen, W.; Lu, W. The gut core microbial species Bifidobacterium longum: Colonization, mechanisms, and health benefits. Microbiol. Res. 2025, 290, 127966. [Google Scholar] [CrossRef]
- Kanimozhi, N.V.; Sukumar, M. Aging through the lens of the gut microbiome: Challenges and therapeutic opportunities. Arch. Gerontol. Geriatr. Plus 2025, 2, 100142. [Google Scholar] [CrossRef]
- Cabello-Olmo, M.; Oneca, M.; Torre, P.; Díaz, J.V.; Encio, I.J.; Barajas, M.; Araña, M. Influence of Storage Temperature and Packaging on Bacteria and Yeast Viability in a Plant-Based Fermented Food. Foods 2020, 9, 302. [Google Scholar] [CrossRef]
- Castro-Herrera, V.M.; Fisk, H.L.; Wootton, M.; Lown, M.; Owen-Jones, E.; Lau, M.; Lowe, R.; Hood, K.; Gillespie, D.; Hobbs, F.D.R.; et al. Combination of the Probiotics Lacticaseibacillus rhamnosus GG and Bifidobacterium animalis subsp. lactis, BB-12 Has Limited Effect on Biomarkers of Immunity and Inflammation in Older People Resident in Care Homes: Results From the Probiotics to Reduce Infections iN CarE home reSidentS Randomized, Controlled Trial. Front. Immunol. 2021, 12, 643321. [Google Scholar] [CrossRef]
- Andrade, J.C.; Almeida, D.; Domingos, M.; Seabra, C.L.; Machado, D.; Freitas, A.C.; Gomes, A.M. Commensal Obligate Anaerobic Bacteria and Health: Production, Storage, and Delivery Strategies. Front. Bioeng. Biotechnol. 2020, 8, 550. [Google Scholar] [CrossRef]
- Jain, P.; Joshi, N.; Sahu, V.; Dominic, A.; Aggarwal, S. Autoimmune diseases and microbiome targeted therapies. Int. Rev. Cell Mol. Biol. 2025, 395, 133–156. [Google Scholar] [CrossRef]
- Smolinska, S.; Popescu, F.-D.; Zemelka-Wiacek, M. A Review of the Influence of Prebiotics, Probiotics, Synbiotics, and Postbiotics on the Human Gut Microbiome and Intestinal Integrity. J. Clin. Med. 2025, 14, 3673. [Google Scholar] [CrossRef]
- Giri, P.S.; Shah, F.; Dwivedi, M.K. Chapter 12—Probiotics and prebiotics in the suppression of autoimmune diseases. In Probiotics in the Prevention and Management of Human Diseases; Dwivedi, M.K., Amaresan, N., Sankaranarayanan, A., Kemp, E.H., Eds.; Academic Press: Cambridge, MA, USA, 2022; pp. 161–186. [Google Scholar]
- Mirfeizi, Z.; Mahmoudi, M.; Faridzadeh, A. Probiotics as a complementary treatment in systemic lupus erythematosus: A systematic review. Health Sci. Rep. 2023, 6, e1640. [Google Scholar] [CrossRef]
- Mohamed, A.a.H.; Shafie, A.; Al-Samawi, R.I.; Jamali, M.C.; Ashour, A.A.; Felemban, M.F.; Alqarni, A.; Ahmad, I.; Mansuri, N.; Ahmad, F.; et al. The role of probiotics in promoting systemic immune tolerance in systemic lupus erythematosus. Gut Pathog. 2025, 17, 45. [Google Scholar] [CrossRef] [PubMed]
- Liu, Y.; Alookaran, J.J.; Rhoads, J.M. Probiotics in Autoimmune and Inflammatory Disorders. Nutrients 2018, 10, 1537. [Google Scholar] [CrossRef]
- Kesavelu Sr, D.; Krishnamurty, A.Y.; Chander, A. Single Strain vs Multiple Strain Probiotics: The Clinician’s Choice. Cureus 2025, 17, e86353. [Google Scholar] [CrossRef]
- de Salis, L.V.V.; Martins, L.S.; Rodrigues, G.S.P.; de Oliveira, G.L.V. Chapter 13—Dysbiosis and probiotic applications in autoimmune diseases. In Translational Autoimmunity; Rezaei, N., Ed.; Academic Press: Cambridge, MA, USA, 2022; Volume 2, pp. 269–294. [Google Scholar]
- McFarland, L.V.; Evans, C.T.; Goldstein, E.J.C. Strain-Specificity and Disease-Specificity of Probiotic Efficacy: A Systematic Review and Meta-Analysis. Front. Med. 2018, 5, 124. [Google Scholar] [CrossRef]
- Abouelela, M.E.; Helmy, Y.A. Next-Generation Probiotics as Novel Therapeutics for Improving Human Health: Current Trends and Future Perspectives. Microorganisms 2024, 12, 430. [Google Scholar] [CrossRef]
- Hollander, D.; Kaunitz, J.D. The “Leaky Gut”: Tight Junctions but Loose Associations? Dig. Dis. Sci. 2020, 65, 1277–1287. [Google Scholar] [CrossRef] [PubMed]
- Horowitz, A.; Chanez-Paredes, S.D.; Haest, X.; Turner, J.R. Paracellular permeability and tight junction regulation in gut health and disease. Nat. Rev. Gastroenterol. Hepatol. 2023, 20, 417–432. [Google Scholar] [CrossRef]
- Dwivedi, M.; Kumar, P.; Laddha, N.C.; Kemp, E.H. Induction of regulatory T cells: A role for probiotics and prebiotics to suppress autoimmunity. Autoimmun. Rev. 2016, 15, 379–392. [Google Scholar] [CrossRef]
- Gavzy, S.J.; Kensiski, A.; Lee, Z.L.; Mongodin, E.F.; Ma, B.; Bromberg, J.S. Bifidobacterium mechanisms of immune modulation and tolerance. Gut Microbes 2023, 15, 2291164. [Google Scholar] [CrossRef]
- Pyclik, M.; Srutkova, D.; Schwarzer, M.; Górska, S. Bifidobacteria cell wall-derived exo-polysaccharides, lipoteichoic acids, peptidoglycans, polar lipids and proteins—Their chemical structure and biological attributes. Int. J. Biol. Macromol. 2020, 147, 333–349. [Google Scholar] [CrossRef] [PubMed]
- Ji, H.-F.; Li, M.; Han, X.; Fan, Y.-T.; Yang, J.-J.; Long, Y.; Yu, J.; Ji, H.-Y. Lactobacilli-Mediated Regulation of the Microbial–Immune Axis: A Review of Key Mechanisms, Influencing Factors, and Application Prospects. Foods 2025, 14, 1763. [Google Scholar] [CrossRef]
- Yang, Y.; Day, J.; Souza-Fonseca Guimaraes, F.; Wicks, I.P.; Louis, C. Natural killer cells in inflammatory autoimmune diseases. Clin. Transl. Immunol. 2021, 10, e1250. [Google Scholar] [CrossRef]
- Camous, X.; Pera, A.; Solana, R.; Larbi, A. NK cells in healthy aging and age-associated diseases. J. Biomed. Biotechnol. 2012, 2012, 195956. [Google Scholar] [CrossRef]
- Chen, P.-C.; Kaur, K.; Ko, M.-W.; Huerta-Yepez, S.; Jain, Y.; Jewett, A. Regulation of Cytotoxic Immune Effector Function by AJ3 Probiotic Bacteria in Amyotrophic Lateral Sclerosis (ALS). Crit. Rev. Immunol. 2023, 43, 13–26. [Google Scholar] [CrossRef]
- Zheng, Y.; Zhang, Z.; Tang, P.; Wu, Y.; Zhang, A.; Li, D.; Wang, C.-Z.; Wan, J.-Y.; Yao, H.; Yuan, C.-S. Probiotics fortify intestinal barrier function: A systematic review and meta-analysis of randomized trials. Front. Immunol. 2023, 14, 1143548. [Google Scholar] [CrossRef] [PubMed]
- Kwoji, I.D.; Aiyegoro, O.A.; Okpeku, M.; Adeleke, M.A. Multi-Strain Probiotics: Synergy among Isolates Enhances Biological Activities. Biology 2021, 10, 322. [Google Scholar] [CrossRef] [PubMed]
- Heczko, P.B.; Giemza, M.; Ponikiewska, W.; Strus, M. Importance of Lactobacilli for Human Health. Microorganisms 2024, 12, 2382. [Google Scholar] [CrossRef]
- Shahali, A.; Soltani, R.; Akbari, V. Probiotic Lactobacillus and the potential risk of spreading antibiotic resistance: A systematic review. Res. Pharm. Sci. 2023, 18, 468–477. [Google Scholar] [CrossRef] [PubMed]
- Li, T.; Teng, D.; Mao, R.; Hao, Y.; Wang, X.; Wang, J. A critical review of antibiotic resistance in probiotic bacteria. Food Res. Int. 2020, 136, 109571. [Google Scholar] [CrossRef]
- Uusitalo, U.; Mramba, L.K.; Aronsson, C.A.; Vehik, K.; Yang, J.; Hummel, S.; Lernmark, Å.; Rewers, M.; Hagopian, W.; McIndoe, R.; et al. HLA Genotype and Probiotics Modify the Association Between Timing of Solid Food Introduction and Islet Autoimmunity in the TEDDY Study. Diabetes Care 2023, 46, 1839–1847. [Google Scholar] [CrossRef]
- Karahan, F. Environmental Factors Affecting the Gut Microbiota and Their Consequences. Nat. Cell Sci. 2024, 2, 133–140. [Google Scholar] [CrossRef]
- Paray, B.A.; Albeshr, M.F.; Jan, A.T.; Rather, I.A. Leaky Gut and Autoimmunity: An Intricate Balance in Individuals Health and the Diseased State. Int. J. Mol. Sci. 2020, 21, 9770. [Google Scholar] [CrossRef]
- Rossi, F.; Amadoro, C.; Gasperi, M.; Colavita, G. Lactobacilli Infection Case Reports in the Last Three Years and Safety Implications. Nutrients 2022, 14, 1178. [Google Scholar] [CrossRef]
- Radcliffe, C.V.; Anahtar, M.N.; Hohmann, E.L.; Turbett, S.E.; Dugdale, C.M.; Paras, M.L. Lactobacillus Bacteremia and Endovascular Infections: A Retrospective Study of 100 Patients. Open Forum Infect. Dis. 2025, 12, ofaf466. [Google Scholar] [CrossRef]
- Abdelhamid, A.G.; El-Masry, S.S.; El-Dougdoug, N.K. Probiotic Lactobacillus and Bifidobacterium strains possess safety characteristics, antiviral activities and host adherence factors revealed by genome mining. EPMA J. 2019, 10, 337–350. [Google Scholar] [CrossRef] [PubMed]
- Doncheva, N.T.; Palasca, O.; Yarani, R.; Litman, T.; Anthon, C.; Groenen, M.A.M.; Stadler, P.F.; Pociot, F.; Jensen, L.J.; Gorodkin, J. Human pathways in animal models: Possibilities and limitations. Nucleic Acids Res. 2021, 49, 1859–1871. [Google Scholar] [CrossRef] [PubMed]
- Ma, T.; Shen, X.; Shi, X.; Sakandar, H.A.; Quan, K.; Li, Y.; Jin, H.; Kwok, L.-Y.; Zhang, H.; Sun, Z. Targeting gut microbiota and metabolism as the major probiotic mechanism—An evidence-based review. Trends Food Sci. Technol. 2023, 138, 178–198. [Google Scholar] [CrossRef]
- Díaz-Peña, R. Personalized Medicine in Autoimmune Diseases. J. Pers. Med. 2021, 11, 1181. [Google Scholar] [CrossRef]
- Zhang, W.; Xiao, D.; Mao, Q.; Xia, H. Role of neuroinflammation in neurodegeneration development. Signal Transduct. Target. Ther. 2023, 8, 267. [Google Scholar] [CrossRef] [PubMed]
- Garofalo, M.; Pandini, C.; Bordoni, M.; Pansarasa, O.; Rey, F.; Costa, A.; Minafra, B.; Diamanti, L.; Zucca, S.; Carelli, S.; et al. Alzheimer’s, Parkinson’s Disease and Amyotrophic Lateral Sclerosis Gene Expression Patterns Divergence Reveals Different Grade of RNA Metabolism Involvement. Int. J. Mol. Sci. 2020, 21, 9500. [Google Scholar] [CrossRef]
- Loh, J.S.; Mak, W.Q.; Tan, L.K.S.; Ng, C.X.; Chan, H.H.; Yeow, S.H.; Foo, J.B.; Ong, Y.S.; How, C.W.; Khaw, K.Y. Microbiota–gut–brain axis and its therapeutic applications in neurodegenerative diseases. Signal Transduct. Target. Ther. 2024, 9, 37. [Google Scholar] [CrossRef] [PubMed]
- Liu, L.; Huh, J.R.; Shah, K. Microbiota and the gut-brain-axis: Implications for new therapeutic design in the CNS. eBioMedicine 2022, 77, 103908. [Google Scholar] [CrossRef]
- Ashique, S.; Mohanto, S.; Ahmed, M.G.; Mishra, N.; Garg, A.; Chellappan, D.K.; Omara, T.; Iqbal, S.; Kahwa, I. Gut-brain axis: A cutting-edge approach to target neurological disorders and potential synbiotic application. Heliyon 2024, 10, e34092. [Google Scholar] [CrossRef]
- Origüela, V.; Lopez-Zaplana, A. Gut Microbiota: An Immersion in Dysbiosis, Associated Pathologies, and Probiotics. Microorganisms 2025, 13, 1084. [Google Scholar] [CrossRef]
- Beikmohammadi, M.; Halimi, S.; Fard, N.A.; Wen, W. Therapeutic Potential of Probiotics: A Review of Their Role in Modulating Inflammation. Probiotics Antimicrob. Proteins 2025. [Google Scholar] [CrossRef]
- Meher, A.K.; Acharya, B.; Sahu, P.K. Probiotics: Bridging the interplay of a healthy gut and psychoneurological well-being. Food Bioeng. 2024, 3, 126–147. [Google Scholar] [CrossRef]
- Ong, J.S.; Lew, L.C.; Hor, Y.Y.; Liong, M.T. Probiotics: The Next Dietary Strategy against Brain Aging. Prev. Nutr. Food Sci. 2022, 27, 1–13. [Google Scholar] [CrossRef]
- Sharma, R.; Gupta, D.; Mehrotra, R.; Mago, P. Psychobiotics: The Next-Generation Probiotics for the Brain. Curr. Microbiol. 2021, 78, 449–463. [Google Scholar] [CrossRef] [PubMed]
- Ma, J.; Piao, X.; Mahfuz, S.; Long, S.; Wang, J. The interaction among gut microbes, the intestinal barrier and short chain fatty acids. Anim. Nutr. 2022, 9, 159–174. [Google Scholar] [CrossRef]
- di Vito, R.; Conte, C.; Traina, G. A Multi-Strain Probiotic Formulation Improves Intestinal Barrier Function by the Modulation of Tight and Adherent Junction Proteins. Cells 2022, 11, 2617. [Google Scholar] [CrossRef] [PubMed]
- Fusco, W.; Lorenzo, M.B.; Cintoni, M.; Porcari, S.; Rinninella, E.; Kaitsas, F.; Lener, E.; Mele, M.C.; Gasbarrini, A.; Collado, M.C.; et al. Short-Chain Fatty-Acid-Producing Bacteria: Key Components of the Human Gut Microbiota. Nutrients 2023, 15, 2211. [Google Scholar] [CrossRef]
- Yamazaki, S. Diverse roles of dendritic cell and regulatory T cell crosstalk in controlling health and disease. Int. Immunol. 2024, 37, 5–14. [Google Scholar] [CrossRef]
- Pittet, M.J.; Di Pilato, M.; Garris, C.; Mempel, T.R. Dendritic cells as shepherds of T cell immunity in cancer. Immunity 2023, 56, 2218–2230. [Google Scholar] [CrossRef]
- Gu, Q.; Yin, Y.; Yan, X.; Liu, X.; Liu, F.; McClements, D.J. Encapsulation of multiple probiotics, synbiotics, or nutrabiotics for improved health effects: A review. Adv. Colloid. Interface Sci. 2022, 309, 102781. [Google Scholar] [CrossRef]
- Hasaniani, N.; Ghasemi-Kasman, M.; Halaji, M.; Rostami-Mansoor, S. Bifidobacterium breve Probiotic Compared to Lactobacillus casei Causes a Better Reduction in Demyelination and Oxidative Stress in Cuprizone-Induced Demyelination Model of Rat. Mol. Neurobiol. 2024, 61, 498–509. [Google Scholar] [CrossRef]
- Liu, L.; Wang, H.; Guo, S.; Liu, S.; Du, Y.; Wang, L.; He, D.; Mo, X.; Zhang, H.; Cheng, Q.; et al. The emerging role of the gut microbiome in depression: Implications for precision medicine. Mol. Psychiatry 2025. [Google Scholar] [CrossRef]
- Abd Mutalib, N.; Syed Mohamad, S.A.; Jusril, N.A.; Hasbullah, N.I.; Mohd Amin, M.C.I.; Ismail, N.H. Lactic Acid Bacteria (LAB) and Neuroprotection, What Is New? An Up-To-Date Systematic Review. Pharmaceuticals 2023, 16, 712. [Google Scholar] [CrossRef]
- Lim, S.-M.; Zaki Ramli, M.; Ahmad Alwi, N.A.; Mani, V.; Majeed, A.B.A.; Ramasamy, K. Chapter 79—Probiotics and Neuroprotection. In Diet and Nutrition in Dementia and Cognitive Decline; Martin, C.R., Preedy, V.R., Eds.; Academic Press: San Diego, CA, USA, 2015; pp. 859–868. [Google Scholar]
- Blake, S.J.; Wolf, Y.; Boursi, B.; Lynn, D.J. Role of the microbiota in response to and recovery from cancer therapy. Nat. Rev. Immunol. 2024, 24, 308–325. [Google Scholar] [CrossRef]
- Mahooti, M.; Safaei, F.; Firuzpour, F.; Abdolalipour, E.; Zare, D.; Sanami, S.; Safavi, M.; Mirdamadi, S. Exploring the role of inflammatory regulatory effects of probiotics as adjuvants in cancer development management with considering possible challenges: A comprehensive review. Inflammopharmacology 2025, 33, 3823–3841. [Google Scholar] [CrossRef]
- Zhuang, K.; Luo, H.; Zeng, M.; Chan, S.C.L.; Gong, M.; Wang, Y. Effects of probiotics, prebiotics, and synbiotics on gut microbiota in older adults: A systematic review and meta-analysis of randomized controlled trials. Nutr. J. 2025, 24, 147. [Google Scholar] [CrossRef]
- Ale, E.C.; Binetti, A.G. Role of Probiotics, Prebiotics, and Synbiotics in the Elderly: Insights Into Their Applications. Front. Microbiol. 2021, 12, 631254. [Google Scholar] [CrossRef]
- Jouriani, F.H.; Torkamaneh, M.; Torfeh, M.; Ashrafian, F.; Aghamohammad, S.; Rohani, M. Native Lactobacillus and Bifidobacterium probiotics modulate autophagy genes and exert anti-inflammatory effect. Sci. Rep. 2025, 15, 25006. [Google Scholar] [CrossRef]
- Ringel-Kulka, T.; Palsson, O.S.; Maier, D.; Carroll, I.; Galanko, J.A.; Leyer, G.; Ringel, Y. Probiotic bacteria Lactobacillus acidophilus NCFM and Bifidobacterium lactis Bi-07 versus placebo for the symptoms of bloating in patients with functional bowel disorders: A double-blind study. J. Clin. Gastroenterol. 2011, 45, 518–525. [Google Scholar] [CrossRef]
- Sarita, B.; Samadhan, D.; Hassan, M.Z.; Kovaleva, E.G. A comprehensive review of probiotics and human health-current prospective and applications. Front. Microbiol. 2024, 15, 1487641. [Google Scholar] [CrossRef]
- Jena, R.; Singh, N.A.; Ahmed, N.; Choudhury, P.K. Bifidobacteria in antibiotic-associated dysbiosis: Restoring balance in the gut microbiome. World J. Microbiol. Biotechnol. 2025, 41, 297. [Google Scholar] [CrossRef]
- Butel, M.J. Probiotics, gut microbiota and health. Med. Mal. Infect. 2014, 44, 1–8. [Google Scholar] [CrossRef]
- Kamareddine, L.; Najjar, H.; Sohail, M.U.; Abdulkader, H.; Al-Asmakh, M. The Microbiota and Gut-Related Disorders: Insights from Animal Models. Cells 2020, 9, 2401. [Google Scholar] [CrossRef]
- Whelan, K.; Alexander, M.; Gaiani, C.; Lunken, G.; Holmes, A.; Staudacher, H.M.; Theis, S.; Marco, M.L. Design and reporting of prebiotic and probiotic clinical trials in the context of diet and the gut microbiome. Nat. Microbiol. 2024, 9, 2785–2794. [Google Scholar] [CrossRef]
- Liu, X.F.; Shao, J.H.; Liao, Y.T.; Wang, L.N.; Jia, Y.; Dong, P.J.; Liu, Z.Z.; He, D.D.; Li, C.; Zhang, X. Regulation of short-chain fatty acids in the immune system. Front. Immunol. 2023, 14, 1186892. [Google Scholar] [CrossRef]
- Gilbert, J.A.; Azad, M.B.; Bäckhed, F.; Blaser, M.J.; Byndloss, M.; Chiu, C.Y.; Chu, H.; Dugas, L.R.; Elinav, E.; Gibbons, S.M.; et al. Clinical translation of microbiome research. Nat. Med. 2025, 31, 1099–1113. [Google Scholar] [CrossRef]
- Wickens, K.; Black, P.; Stanley, T.V.; Mitchell, E.; Barthow, C.; Fitzharris, P.; Purdie, G.; Crane, J. A protective effect of Lactobacillus rhamnosus HN001 against eczema in the first 2 years of life persists to age 4 years. Clin. Exp. Allergy 2012, 42, 1071–1079. [Google Scholar] [CrossRef]
- Liu, Y.; Zhou, X.; Ye, W.; Liu, Y.; Luo, J.; Tang, X.; Wang, J.; Liu, C.; Zhou, H. Effect of Lactobacillus rhamnosus LZ260E on allergic symptoms and intestinal microbiota in β-lactoglobulin–sensitized mice. J. Funct. Foods 2024, 113, 106045. [Google Scholar] [CrossRef]
- Yamasaki, M.; Minesaki, M.; Iwakiri, A.; Miyamoto, Y.; Ogawa, K.; Nishiyama, K.; Tsend-Ayush, C.; Oyunsuren, T.; Li, Y.; Nakano, T.; et al. Lactobacillus plantarum 06CC2 reduces hepatic cholesterol levels and modulates bile acid deconjugation in Balb/c mice fed a high-cholesterol diet. Food Sci. Nutr. 2020, 8, 6164–6173. [Google Scholar] [CrossRef]
- Al-Sadi, R.; Nighot, P.; Nighot, M.; Haque, M.; Rawat, M.; Ma, T.Y. Lactobacillus acidophilus Induces a Strain-specific and Toll-Like Receptor 2-Dependent Enhancement of Intestinal Epithelial Tight Junction Barrier and Protection Against Intestinal Inflammation. Am. J. Pathol. 2021, 191, 872–884. [Google Scholar] [CrossRef]
- Wang, G.; Xu, Q.; Jin, X.; Hang, F.; Liu, Z.; Zhao, J.; Zhang, H.; Chen, W. Effects of lactobacilli with different regulatory behaviours on tight junctions in mice with dextran sodium sulphate-induced colitis. J. Funct. Foods 2018, 47, 107–115. [Google Scholar] [CrossRef]
- Liao, Z.; Quintana, Y. Challenges to Global Standardization of Outcome Measures. AMIA Jt. Summits Transl. Sci. Proc. 2021, 2021, 404–409. [Google Scholar]
- Chung, K.C.; Malay, S.; Shauver, M.J. The Complexity of Conducting a Multicenter Clinical Trial: Taking It to the Next Level Stipulated by the Federal Agencies. Plast. Reconstr. Surg. 2019, 144, 1095e–1103e. [Google Scholar] [CrossRef]
- Matera, M. Bifidobacteria, Lactobacilli... when, how and why to use them. Glob. Pediatr. 2024, 8, 100139. [Google Scholar] [CrossRef]
- Livingstone, K.M.; Ramos-Lopez, O.; Pérusse, L.; Kato, H.; Ordovas, J.M.; Martínez, J.A. Precision nutrition: A review of current approaches and future endeavors. Trends Food Sci. Technol. 2022, 128, 253–264. [Google Scholar] [CrossRef]
- Lyu, R.; Qu, Y.; Divaris, K.; Wu, D. Methodological Considerations in Longitudinal Analyses of Microbiome Data: A Comprehensive Review. Genes 2023, 15, 51. [Google Scholar] [CrossRef]
- 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]





| Factors | Preclinical Studies | Clinical Studies |
|---|---|---|
| Gut Microbiota/ physiology/immune system | It can be regulated. Probiotics might temporarily colonize mice and significantly influence immune markers or gut barrier function because of their simplified microbiota. | Humans have highly diverse microbiomes and immune adaptations that influence the survival, activity, and metabolic effects of Lactobacillus. Probiotic colonization tends to be temporary, with metabolic or immunomodulatory impacts often being weaker and highly personalized. |
| Diet/lifestyle | Diet and lifestyle can be adjusted or personalized. | Variable dietary factors, medications, and previous microbiome exposures contribute to confounding factors. |
| Genetic and environmental factors | Genetically uniform strains are common, and environmental conditions can be managed. | Substantial genetic heterogeneity makes it challenging to manage due to population variability. |
| Duration | Short-term interventions and acute effects | Long-term chronic effects and safety assessments are not as frequently studied. |
| Sample size | Can reach 100 or more, and it is easy to scale up. | Often ranging from 10 to 1000, it is not always easy to scale up. |
| Mechanism | SCFAs increased acetate and propionate levels, providing barrier protection. | Outcomes depend on diet and transient colonization. |
| Boosted Treg cells while reducing pro-inflammatory cytokines. | Small reductions in inflammation markers with subject variability. | |
| In colitis models, SCFAs enhanced junction integrity by increasing ZO-1, occludin, and claudin-1. | Some improvements in mucosal barrier proteins have been noted, though functional outcomes are modest. | |
| mitigated stress and anxiety through the vagus-OXT axis. | Evidence is limited, but perinatal mood benefits have been observed in pilot RCTs. | |
| Consistent anti-inflammatory responses, enhanced mucosal IgA production, or regulated cytokine activity. | Variable effects, occasionally showing statistical significance in specific subpopulations, but often not reproducible across different studies. | |
| Endpoint measurements | Invasive sampling, such as from tissue or the intestinal lumen, provides detailed mechanistic insights | Non-invasive methods, such as stool samples and blood biomarkers, offer insights but limit mechanistic resolution. |
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. |
© 2025 by the author. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Kaur, K. Application and Challenges of Using Probiotic Lactobacillus and Bifidobacterium to Enhance Overall Health and Manage Diseases. Diseases 2025, 13, 345. https://doi.org/10.3390/diseases13100345
Kaur K. Application and Challenges of Using Probiotic Lactobacillus and Bifidobacterium to Enhance Overall Health and Manage Diseases. Diseases. 2025; 13(10):345. https://doi.org/10.3390/diseases13100345
Chicago/Turabian StyleKaur, Kawaljit. 2025. "Application and Challenges of Using Probiotic Lactobacillus and Bifidobacterium to Enhance Overall Health and Manage Diseases" Diseases 13, no. 10: 345. https://doi.org/10.3390/diseases13100345
APA StyleKaur, K. (2025). Application and Challenges of Using Probiotic Lactobacillus and Bifidobacterium to Enhance Overall Health and Manage Diseases. Diseases, 13(10), 345. https://doi.org/10.3390/diseases13100345
