Preprint
Review

This version is not peer-reviewed.

Exploring the Diverse Roles of Probiotic Strains in Aging, Cancer, Autoimmune Diseases, and Neurodegenerative Disorders

A peer-reviewed article of this preprint also exists.

Submitted:

08 September 2025

Posted:

10 September 2025

You are already at the latest version

Abstract
This review looks at how probiotics affect health issues like cancer, aging, autoimmune diseases, and neurodegenerative disorders, focusing on immune balance through different probiotic strain combinations. It discusses how various combinations activate PBMCs, NK cells, and CD8+ T cells in unique ways. Some combinations are helpful for autoimmune diseases, while others show potential in cancer treatment. Most formulations improve immune balance and aging when used as supplements. The research highlights varying levels of immune cell activation, with certain combinations promoting cell growth and activating NK and CD8+ T cells alongside feeder cells. In cancer-bearing humanized mice, oral administration of specific combinations with NK cell-based therapy enhanced tumor suppression, immune function, and bone health. Cancer patients taking probiotic supplements experienced increased cytokine production and improved PBMC and NK cell cytotoxicity. Probiotics' positive effects are also examined in aging, autoimmune diseases, and neurodegenerative disorders. Overall, the review emphasizes the important role of probiotics in maintaining immune balance and boosting immunotherapy results.
Keywords: 
;  ;  ;  ;  ;  ;  ;  ;  ;  ;  

1. Introduction and Background

In the early 20th century, Elie Metchnikoff discovered certain strains of gut bacteria essential for maintaining balance in the gut, which he named probiotics 1. These live, beneficial bacteria, often derived from fermented foods or the gut itself, offer numerous health benefits supported by studies and clinical trials 2,3. Research shows their potential to address gut issues, obesity, syndrome, allergies, heart disease, and even cancer 4−7. Probiotics help maintain the intestinal barrier and microbial balance by enhancing proteins and mucus that prevent pathogens and excessive immune responses 8,9. They interact with immune cells through direct contact and signaling molecules, boosting immunity while maintaining gut tolerance 10. Probiotics also strengthen defenses against infections by activating immune cells and increasing cytokines and antibodies 11. Additionally, they balance inflammation, helping with conditions like inflammatory bowel disease, allergies, and autoimmune disorders 12−15. By competing with pathogens and producing antimicrobial agents, probiotics contribute to immune stability 16. Components like exopolysaccharides, surface proteins, and secreted peptides interact with immune receptors, influencing signaling pathways 17−19. They also adhere to the gut lining and engage with immune cells via Toll-like receptors (TLRs) like TLR2, TLR4, and TLR9 12,20.
Probiotics play a crucial role in maintaining immune balance by activating Tregs, which promote tolerance to commensals and food antigens, reducing inflammation and allergic responses 21. They also impact the differentiation and function of T helper (Th) cell subsets and Tregs, driving immune responses like cell-mediated immunity or tolerance through cytokines like IL-10 and TGF-β 12,20. Probiotics aid in B-cell maturation into plasma cells that produce secretory IgA (sIgA), essential for defending mucosal surfaces against pathogens 22. The immune-modulating effects of probiotics depend heavily on the specific strains used, as different species and strains can uniquely affect cytokine responses and immune cell activity. For instance, Lactobacillus strains often enhance Th1-type immune responses, while Bifidobacterium strains typically promote anti-inflammatory effects 23. Most probiotics are lactic acid-producing bacteria, such as lactobacilli, streptococci, and bifidobacteria, emphasizing their potential for human health 24. Certain strains, like Lactococcus lactis subsp. Cremoris C60, have even been found to boost antigen presentation by dendritic cells, enhancing cytotoxic T cell responses critical for combating tumors and viruses 25,26.
Picking the right strain and dosage of probiotic bacteria is essential for achieving immune benefits. The effects of probiotics on immunity largely depend on the specific strains and the overall context, making careful selection key to meeting therapeutic goals. This review looks at how different probiotic formulations activate and boost immune cells, helping maintain immune balance in conditions like cancer, aging, autoimmune diseases, and neurodegenerative disorders.

2. Probiotics Formulation for Cancer Prevention and as Adjuvant Cancer Therapeutics

Cancer is the second leading cause of death globally, with no effective cure currently available, and cancer-related deaths are expected to rise 27. There is a pressing need for treatments that are highly effective and have minimal side effects 28−30. Current therapies often impact quality of life due to side effects, drug resistance, and affordability challenges 31. Interestingly, research suggests probiotics could help in cancer prevention and as an additional treatment. Probiotics contribute to cancer prevention and treatment by influencing immunoglobulin A production, stimulating macrophage activity, and reducing the toxicity of anti-cancer therapies 15,32−35. Probiotics boost immune responses and support tumor immunity by interacting with dendritic cells, macrophages, natural killer cells, and neutrophils, enhancing their function and cytokine production 12,36. This activation leads to the upregulation of co-stimulatory molecules on antigen-presenting cells and the release of cytokines to manage inflammation and immune cell recruitment, balancing pro- and anti-inflammatory pathways 26,37−39. Found in foods and supplements, probiotics enhance innate immunity by activating natural killer cells for antitumor effects and maintaining gut microbial balance 40−44. They also lower nitric oxide production and reduce pro-inflammatory cytokines 28,30,45−50. Even inactive probiotics and their byproducts provide similar health benefits, making them valuable in cancer prevention and treatment 51−56.
NK cells are crucial for innate immunity due to their lack of antigen specificity 57−59, while CD8+ T cells play a key role in adaptive cellular immunity, closely interacting with the innate immune system 60,61. Both cell types are essential in cancer therapy, forming the foundation of current treatments 62,63. Impaired function of these cells is associated with worse outcomes in cancer patients 64−70. NK cells also enhance CD8+ T cell responses against viral infections like cytomegalovirus 71,72. In cancer patients, NK cells show reduced cytotoxicity, IFN-γ secretion, survival, and expansion, linked to lower expression of CD16, NKG2D, and the zeta chain 67,73−79. CD8+ T cells in cancer patients exhibit decreased IFN-γ secretion and reduced expression of CD62L, CD28, CCR7, and CD127 80. Probiotics have shown potential by significantly enhancing NK cell cytotoxicity and IFN-γ secretion in both NK and CD8+ T cells 80. This approach also improves monocytes’ ability to synergistically stimulate IFN-γ secretion in NK cells in cancer patients and healthy individuals 81. Probiotics further impact DCs, regulatory T cells, and NK cells, strengthening intestinal defenses, although the effects vary depending on the probiotic strains used 1,42.
A study examined eight gram-positive bacterial strains, showing that formulations containing Streptococcus thermophilus, Bifidobacterium longum, Bifidobacterium breve, Bifidobacterium infantis, Lactobacillus acidophilus, Lactobacillus plantarum, Lactobacillus paracasei, and Lactobacillus bulgaricus stimulated both pro-inflammatory and anti-inflammatory cytokines and growth factors in NK cells 82. Notably, IFN-γ and IL-1Ra levels significantly increased 82. This NK cell activation by probiotics supports their anti-cancer effects 73,75,82. Probiotic-treated NK cells revealed that Lactobacillus species and Streptococcus thermophilus promoted a Th1-type cytokine profile, increasing IL-12 and IFN-γ while decreasing IL-10, whereas Bifidobacterium induced a Th2 profile with higher IL-10 and IL-6 compared to IL-12 and IFN-γ 82. Another study analyzed probiotic formulations and identified seven gram-positive strains, either as a mix of seven or four: Streptococcus thermophilus, Bifidobacterium longum, Bifidobacterium breve, Bifidobacterium infantis, Lactobacillus acidophilus, Lactobacillus plantarum, and Lactobacillus paracasei, or Streptococcus thermophilus, Lactobacillus acidophilus, Lactobacillus plantarum, and Lactobacillus paracasei, which demonstrated anti-cancer and anti-infection effects by increasing IFN-γ and reducing IL-10 83. Probiotic bacteria have been utilized to develop NK or CD8+T cell-based immunotherapies, which significantly boost the functional capabilities of these cells 73,75,84,85. A combination of eight strains (Streptococcus thermophilus, Bifidobacterium longum, Bifidobacterium breve, Bifidobacterium infantis, Lactobacillus acidophilus, Lactobacillus plantarum, Lactobacillus paracasei, and Lactobacillus bulgaricus) or seven strains (Streptococcus thermophilus, Bifidobacterium longum, Bifidobacterium breve, Bifidobacterium infantis, Lactobacillus acidophilus, Lactobacillus plantarum, and Lactobacillus paracasei) of gram-positive bacteria, along with feeder cells, significantly boosted the growth of NK and CD8+ T cells 86,87.
In individuals consuming Bifidobacterium, DCs showed increased expression of anti-tumor immunity-related genes, enhancing T cell activation 88,89. Lactobacilli play a key role in stimulating immune responses by influencing DCs, NK cells, and Th1 cells 28,90−92. Lactic acid bacteria boost host immunity by producing IL-2, which activates NK cells 93. Consuming Lactobacillus acidophilus is associated with higher serum levels of IFN-γ, IL-10, and increased CD4+ and CD8+ T cells 94−96. Oral administration of B. longum and B. breve to melanoma-bearing mice significantly reduced tumor volume, showing effects similar to PD-1 therapy alone. When combined with PD-1 therapy, these probiotics further reduced tumor volume 88,97. Probiotic-fed humanized mice exhibited improved NK-mediated cytotoxicity, increased IFN-γ secretion across tissues, and reduced tumor load, and also restored bone formation 74,98,99. Colorectal cancer patients experienced significant reductions in various cytokines when treated with probiotics 50. In mice with breast tumors, oral L. acidophilus administration reduced tumor burden and affected cytokine production 100,101. Lactobacillus plantarum and Lactobacillus salivarius show potential in preventing oral and colon cancers by modulating immune responses and suppressing COX-2 expression in tumors 102−104. Bifidobacterium longum, Lactobacillus acidophilus, and Lactobacillus plantarum consumption has shown promise in preventing and inhibiting cancers like colon, breast, liver, intestines, lungs, oral cavity, and pancreas 28,50,89,94−96,103−116. These findings highlight the potential of probiotics in cancer treatment, showcasing their ability to modulate immune responses for therapeutic benefits 111,114,115,117.
Probiotics also have shown efficacy as adjuvant therapeutics, offering protection against chemotherapy and radiation side effects like diarrhea, fever, and disruptions to intestinal microbiota in cancer patients. These advantages not only enhance patients’ quality of life but also help prevent interruptions or halts in treatment 118−125. These results highlight that probiotics not only provide therapeutic benefits against cancer but also improve outcomes when paired with other cancer treatments.

3. Probiotics Formulation to Restore Immune Function in Older Individuals

Aging brings about immunosenescence, characterized by a decline in both innate and adaptive immune functions, increased systemic inflammation, and alterations in the composition and diversity of gut microbiota 126,127. It significantly affects innate immunity, causing changes in the number, characteristics, and functionality of immune cells, which are closely linked to various diseases and infections 128−136. These changes increase susceptibility to infections, chronic diseases, and reduced vaccine effectiveness. In older adults, diminished immune function leads to higher risks of infections, cancer, autoimmune disorders, Alzheimer’s disease, atherosclerosis, vision problems like age-related macular degeneration, cardiovascular issues, coronary heart disease, liver fibrosis, neurodegenerative diseases, and exposure to pathogens 133−138. Additionally, cytokine levels, especially IFN-γ, are notably lower in the immune cells of older individuals 139,140.
The gut microbiota is essential for immune regulation, with aging-related dysbiosis playing a major role in immune dysfunction 141,142. Probiotics can restore gut microbial balance, enhance innate immunity, such as natural killer (NK) cells, and regulate inflammation 8,143. NK cells are thought to significantly influence longevity, as their proper function protects the elderly from infections, cancer, autoimmune disorders, and neurodegenerative diseases 133. Aging leads to decreased NK cell function, contributing to age-related health issues 133−136. NK cells are vital for fighting these illnesses and activating adaptive immune cells, ensuring a robust immune system in old age 129,140. Additionally, NK cell proliferation declines in the elderly. Research by Chuang Guo et al. found that the proinflammatory CD52+ NK cell subset increases in older adults, facilitating infection spread 144. Feeder cells, which support NK cell activation and growth, also lose effectiveness in older individuals due to reduced ligand expression and factor secretion 140. However, in vitro treatment with probiotics, alone or with feeder cells, has restored NK cell numbers and function in the elderly 140. Probiotic supplements have been found to moderately improve immune markers and boost NK cell function, an essential part of innate immunity, in healthy older adults 20. Studies show that probiotics significantly enhance NK cell cytotoxic activity in individuals aged 60 and above, helping with early infection defense and tumor surveillance 145. They also lower chronic low-grade inflammation, prevent infections, and promote healthy aging 145. Probiotics increase anti-inflammatory cytokines like IL-10 and TGF-β while reducing pro-inflammatory markers such as IL-6 and CRP, combating chronic inflammation 12,146. These treatments can activate T and B cells, improve immune monitoring, reduce the occurrence and severity of colds and gut infections, and strengthen vaccine responses 146.
The benefits vary depending on the probiotic strain used. Lactobacillus acidophilus helps balance gut bacteria and boost immunity during illness or antibiotic use 16. Lactobacillus fermentum strengthens the immune system and helps prevent gastrointestinal and respiratory infections 147,148. Lactobacillus casei/paracasei have anti-inflammatory properties, support the gut barrier, and aid in immune modulation 149. Lactobacillus plantarum promotes immune response and reduces gut inflammation 150. Lactobacillus rhamnosus enhances gut health, strengthens immune defenses, and eases digestive discomfort. Bifidobacterium longum reduces inflammation, protects against intestinal infections, and supports immune balance 151. Bifidobacterium bifidum improves digestive health and boosts the immune system. Bifidobacterium lactis helps prevent infections and supports vitamin production 152. For individuals over 60, typical probiotic doses range from 1 billion to 50 billion CFU (colony-forming units) daily, depending on the strain and health condition. Studies show supplementation duration usually varies between 3 to 12 weeks, with longer courses potentially needed for lasting effects 153. Probiotic formulations can be single-strain or multi-strain combinations, often including both Lactobacillus and Bifidobacterium species for synergistic benefits. Probiotics are generally safe for healthy elderly individuals 153.
Gerobiotics are an intriguing new kind of probiotics and related biotics designed to tackle aging-related issues like cellular senescence, mitochondrial dysfunction, and chronic inflammation 154−156. They go beyond gut health, offering potential anti-aging and immune-boosting benefits 154. Early studies suggest that strains like Lactobacillus plantarum, Lactobacillus rhamnosus, Bifidobacterium longum, and Lactobacillus helveticus may help improve aging processes and strengthen immunity 157.

4. Probiotics Formulation for Autoimmune Disease Therapeutics

Autoimmune diseases occur when the immune system becomes overactive and mistakenly attacks the body’s own tissues 158. Studies show that gut microbiota dysbiosis, an imbalance in the intestinal microbial community, is connected to the onset and progression of autoimmune conditions such as type 1 diabetes, rheumatoid arthritis, lupus, and multiple sclerosis 158. Probiotics offer a promising way to manage gut microbiota, restoring balance by promoting beneficial microbes and outcompeting harmful ones 159. They help strengthen the gut barrier by improving tight junctions, reducing gut permeability (“leaky gut”), and lowering exposure to inflammatory triggers 160,161. Probiotics also regulate immune responses by balancing T-helper cells (e.g., increasing anti-inflammatory Treg cells while reducing Th17/Th1-driven inflammation) and modulating cytokines (e.g., boosting IL-10 and TGF-β while decreasing TNF-α and IL-6) 23. Additionally, they interact with immune cell receptors (TLRs) to refine immune activation and produce helpful metabolites like short-chain fatty acids (SCFAs) that reduce inflammation 162.
Various probiotic strains hold promise for supporting patients with autoimmune diseases through their differentiation effects 163. Lactobacillus species such as L. casei, L. rhamnosus, L. acidophilus, and L. reuteri are known for their immunomodulatory and barrier-strengthening properties 157. Bifidobacterium species, such as B. bifidum and B. animalis, promote Treg induction and help reduce pro-inflammatory responses 164. Bacillus coagulans has been associated with easing arthritis symptoms and systemic inflammation 165. Next-generation probiotics (NGPs), developed through sequencing and bioinformatics, leverage synthetic biology and gene editing for targeted therapeutic applications 166. Probiotic formulations may involve single-strain or multispecies blends tailored to specific autoimmune conditions 167. Research in animals and humans indicates probiotics can delay or prevent autoimmune diabetes in NOD mice, lower inflammatory cytokines, and reduce joint damage in rheumatoid arthritis 153,168. They also improve gut microbiota composition in conditions like systemic lupus and multiple sclerosis. However, results vary based on strain, disease, and study design 169,170. While meta-analyses highlight their adjunctive benefits, probiotics are not yet standard primary treatments 171. The functional state of NK cells is crucial in infection control, disease progression, and prognosis. NK cell dysfunction has been linked to the development of autoimmune diseases, with changes in NK cell number or function impacting the entire immune system 133,172. One study showed a probiotic formulation with three gram-positive bacterial strains enhanced IL-10 levels in PBMCs and NK cells, providing benefits for autoimmune diseases compared to other combinations 83.
Creating probiotic formulations for autoimmune disease treatment involves selecting strains with known immunomodulatory benefits, ensuring they survive and thrive in the right locations, and tailoring them to the host and microbiome’s specific needs 173,174. These formulations often blend strains like Lactobacillus, Bifidobacterium, and Bacillus, sometimes enhanced with technologies like nano-encapsulation or genetic engineering to improve their impact. While promising, current research indicates that probiotics are most effective as supplementary therapies rather than standalone treatments until more clinical studies are completed 173.

5. Probiotics Formulation for Neurodegenerative Disorder Therapeutics

Neurodegenerative diseases such as Alzheimer’s, Parkinson’s, Amyotrophic lateral sclerosis (ALS), and Huntington’s are progressive conditions with complex origins, including protein buildup, oxidative stress, neuroinflammation, mitochondrial dysfunction, and immune system issues 175,176. The gut microbiome significantly influences these diseases via the microbiota–gut–brain axis, impacting neuroinflammation, gut barrier integrity, immune responses, and neurotransmitter production 177−179. Dysbiosis, or an imbalance in gut bacteria, is often observed in these disorders 173,180. It includes an increased presence of pro-inflammatory strains like Streptococcus, Alistipes, Ruminococcus, Enterococcus, and Desulfovibrio, alongside a reduction in beneficial butyrate-producing bacteria such as Faecalibacterium, Lachnospira, Roseburia, Blautia, and Prevotella 173,180.
Probiotics provide therapeutic benefits by replenishing beneficial bacteria, boosting neuroprotective metabolites like butyrate, and offering anti-inflammatory and antioxidant effects 143,177. They help maintain gut and blood-brain barrier integrity, influence neurotrophic factors and neurotransmitter pathways such as brain-derived neurotrophic factor (BDNF), serotonin, and gamma-aminobutyric acid (GABA), and support microglial balance 181−183. Probiotics reduce proinflammatory pathways, enhance vagus nerve signaling for improved brain communication and emotional regulation, and combat oxidative stress through antioxidant defenses, which is vital for neurodegenerative disorders 184,185. Moreover, they may prevent the buildup of harmful protein aggregates like amyloid-β and α-synuclein via immune modulation, improve gut barrier function, lower systemic proinflammatory endotoxins, and positively affect BDNF to promote neurogenesis and synaptic plasticity 8.
Combining multiple probiotic strains like Lactobacillus and Bifidobacterium species can synergistically improve motor and cognitive symptoms in Parkinson’s disease models 186,187. For effective results, a sufficient dose (usually ≥10^9 CFU per strain per dose) is crucial, and spore-forming bacteria like Bacillus subtilis add stability and improve gastrointestinal survival. Probiotic formulations targeting neurodegenerative diseases should aim to restore gut microbiome balance, enhance butyrate-producing bacteria, and reduce inflammation 188,189. Studies have suggested the potential implications of probiotics in depression treatment 190. Both clinical and preclinical studies highlight their exciting potential.

6. Conclusions

Probiotics are widely recognized for their positive impact on gut microbiota health, with recent studies highlighting their broader role in health and disease. They help restore immune function in aging and conditions like cancer, autoimmune, and neurodegenerative disorders. In cancer therapy, probiotics support immune balance and reduce chemotherapy and radiotherapy side effects. Combining probiotics with checkpoint inhibitors shows potential for improving patient outcomes. Research has shown probiotics can activate and expand anti-cancer immune cells such as NK and CD8+ T cells. They boost immune cell cytotoxic activity, IFN-γ secretion, CD8+ T cell numbers, and immune cell recruitment in tumors, enhancing the fight against cancer. Additionally, probiotics aid in restoring immune function in aging and other chronic diseases.

Conflicts of Interest

The authors declare that the work reviewed in this article was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

References

  1. Metchnikoff, E. Essais optimistes. (A. Maloine, 1907).
  2. 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] [PubMed]
  3. 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]
  4. Fotiadis, C.I.; Stoidis, C.N.; Spyropoulos, B.G.; Zografos, E.D. Role of probiotics, prebiotics and synbiotics in chemoprevention for colorectal cancer. World J. Gastroenterol. 2008, 14, 6453–6457. [Google Scholar] [CrossRef] [PubMed]
  5. Dongarrà, M.L.; Rizzello, V.; Muccio, L.; Fries, W.; Cascio, A.; Bonaccorsi, I.; Ferlazzo, G. Mucosal Immunology and Probiotics. Curr. Allergy Asthma Rep. 2012, 13, 19–26. [Google Scholar] [CrossRef]
  6. Özdemir, Ö. Various effects of different probiotic strains in allergic disorders: an update from laboratory and clinical data. Clin. Exp. Immunol. 2010, 160, 295–304. [Google Scholar] [CrossRef]
  7. Ouwehand, A. C. Antiallergic effects of probiotics. J Nutr 137, 794S-797S (2007).
  8. Chandrasekaran, P.; Weiskirchen, S.; Weiskirchen, R. Effects of Probiotics on Gut Microbiota: An Overview. Int. J. Mol. Sci. 2024, 25, 6022. [Google Scholar] [CrossRef]
  9. Gao, X.; Zhao, J.; Zhang, H.; Chen, W.; Zhai, Q. Modulation of gut health using probiotics: the role of probiotic effector molecules. J. Futur. Foods 2022, 2, 1–12. [Google Scholar] [CrossRef]
  10. 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]
  11. Dikiy, S.; Rudensky, A.Y. Principles of regulatory T cell function. Immunity 2023, 56, 240–255. [Google Scholar] [CrossRef]
  12. 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]
  13. Isolauri, E. , Sutas, Y., Kankaanpaa, P., Arvilommi, H. & Salminen, S. Probiotics: effects on immunity. Am J Clin Nutr 2001, 73, 444S–450S. [Google Scholar]
  14. Perdigon, G.; Demacias, M.; Alvarez, S.; Oliver, G.; Holgado, A. Systemic augmentation of the immune-response in mice by feeding fermented milks with lactobacillus-casei and lactobacillus-acidophilus. Immunology 1988, 63, 17–23. [Google Scholar]
  15. Park, J.-H.; Um, J.-I.; Lee, B.-J.; Goh, J.-S.; Park, S.-Y.; Kim, W.-S.; Kim, P.-H. Encapsulated Bifidobacterium bifidum potentiates intestinal IgA production. Cell. Immunol. 2002, 219, 22–27. [Google Scholar] [CrossRef]
  16. Wang, X.; Zhang, P.; Zhang, X. Probiotics Regulate Gut Microbiota: An Effective Method to Improve Immunity. Molecules 2021, 26, 6076. [Google Scholar] [CrossRef] [PubMed]
  17. 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. [Google Scholar] [CrossRef] [PubMed]
  18. Angelin, J.; Kavitha, M. Exopolysaccharides from probiotic bacteria and their health potential. Int. J. Biol. Macromol. 2020, 162, 853–865. [Google Scholar] [CrossRef]
  19. 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]
  20. 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]
  21. 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] [PubMed]
  22. 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. 2024, 155, 37–51. [Google Scholar] [CrossRef]
  23. 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, 1–13. [Google Scholar] [CrossRef]
  24. Abedi, E.; Hashemi, S.M.B. Lactic acid production – producing microorganisms and substrates sources-state of art. Heliyon 2020, 6. [Google Scholar] [CrossRef]
  25. 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]
  26. 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]
  27. Global Burden of Disease Cancer, Collaboration; Fitzmaurice, C.; Allen, C.; Barber, R.M.; Barregard, L.; Bhutta, Z.A.; Brenner, H.; Dicker, D.J.; Chimed-Orchir, O.; Dandona, R.; et al. Global, Regional, and National Cancer Incidence, Mortality, Years of Life Lost, Years Lived With Disability, and Disability-Adjusted Life-years for 32 Cancer Groups, 1990 to 2015: A systematic analysis for the global burden of disease study. JAMA Oncol. 2017, 3, 524–548, Erratum in JAMA Oncol. 2017, 3, 418. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  28. Maroof, H.; Hassan, Z.M.; Mobarez, A.M.; Mohamadabadi, M.A. Lactobacillus acidophilus Could Modulate the Immune Response Against Breast Cancer in Murine Model. J. Clin. Immunol. 2012, 32, 1353–1359. [Google Scholar] [CrossRef]
  29. Hassan, Z. Anti-cancer and Biotherapeutic Potentials of Probiotic Bacteria. J. Cancer Sci. Ther. 2019, 11. [Google Scholar] [CrossRef]
  30. 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]
  31. Nazir, Y.; Hussain, S.A.; Hamid, A.A.; Song, Y. Probiotics and Their Potential Preventive and Therapeutic Role for Cancer, High Serum Cholesterol, and Allergic and HIV Diseases. BioMed Res. Int. 2018, 2018, 1–17. [Google Scholar] [CrossRef]
  32. Fukushima, Y.; Kawata, Y.; Hara, H.; Terada, A.; Mitsuoka, T. Effect of a probiotic formula on intestinal immunoglobulin A production in healthy children. Int. J. Food Microbiol. 1998, 42, 39–44. [Google Scholar] [CrossRef]
  33. Rautava, S.; Arvilommi, H.; Isolauri, E. Specific Probiotics in Enhancing Maturation of IgA Responses in Formula-Fed Infants. Pediatr. Res. 2006, 60, 221–224. [Google Scholar] [CrossRef]
  34. Ustunol, Z. & Pestka, J. J. Probiotics in health: Their immunomodulatory potential against allergic disorders. J. Anim. Sci. 2004, 82, 274–274. [Google Scholar]
  35. Fuccio, L.; Guido, A.; Eusebi, L.H.; Laterza, L.; Grilli, D.; Cennamo, V.; Ceroni, L.; Barbieri, E.; Bazzoli, F. Effects of Probiotics for the Prevention and Treatment of Radiation-induced Diarrhea. J. Clin. Gastroenterol. 2009, 43, 506–513. [Google Scholar] [CrossRef]
  36. da Silva, R.B.; Münz, C. Natural killer cell activation by dendritic cells: balancing inhibitory and activating signals. Cell. Mol. Life Sci. 2011, 68, 3505–3518. [Google Scholar] [CrossRef]
  37. Zhang, Q.; Vignali, D.A. Co-stimulatory and Co-inhibitory Pathways in Autoimmunity. Immunity 2016, 44, 1034–1051. [Google Scholar] [CrossRef]
  38. 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. 2022, 135, 1–9. [Google Scholar] [CrossRef]
  39. Oerlemans, M.M.; Akkerman, R.; Ferrari, M.; Walvoort, M.T.; de Vos, P. Benefits of bacteria-derived exopolysaccharides on gastrointestinal microbiota, immunity and health. J. Funct. Foods 2021, 76. [Google Scholar] [CrossRef]
  40. Toi, M.; Hirota, S.; Tomotaki, A.; Sato, N.; Hozumi, Y.; Anan, K.; Nagashima, T.; Tokuda, Y.; Masuda, N.; Ohsumi, S.; et al. Probiotic Beverage with Soy Isoflavone Consumption for Breast Cancer Prevention: A Case-control Study. Curr. Nutr. Food Sci. 2013, 9, 194–200. [Google Scholar] [CrossRef]
  41. Fuller, R. Probiotics in man and animals. J. Appl. Bacteriol. 1989, 66, 365–78. [Google Scholar] [CrossRef]
  42. Matsuzaki, T.; Chin, J. Modulating immune responses with probiotic bacteria. Immunol. Cell Biol. 2000, 78, 67–73. [Google Scholar] [CrossRef]
  43. Gill, H.S.; Rutherfurd, K.J.; Cross, M.L.; Gopal, P.K. Enhancement of immunity in the elderly by dietary supplementation with the probiotic Bifidobacterium lactis HN019. Am. J. Clin. Nutr. 2001, 74, 833–839. [Google Scholar] [CrossRef]
  44. O’Toole, P.W.; Marchesi, J.R.; Hill, C. Next-generation probiotics: The spectrum from probiotics to live biotherapeutics. Nat. Microbiol. 2017, 2, 17057. [Google Scholar] [CrossRef]
  45. Thirabunyanon, M.; Boonprasom, P.; Niamsup, P. Probiotic potential of lactic acid bacteria isolated from fermented dairy milks on antiproliferation of colon cancer cells. Biotechnol. Lett. 2008, 31, 571–576. [Google Scholar] [CrossRef]
  46. Kumar, R.S.; Kanmani, P.; Yuvaraj, N.; Paari, K.A.; Pattukumar, V.; Thirunavukkarasu, C.; Arul, V. Lactobacillus plantarum AS1 Isolated from South Indian Fermented Food Kallappam Suppress 1,2-Dimethyl Hydrazine (DMH)-Induced Colorectal Cancer in Male Wistar Rats. Appl. Biochem. Biotechnol. 2011, 166, 620–631. [Google Scholar] [CrossRef]
  47. Tarrah, A.; De Castilhos, J.; Rossi, R.C.; da Silva Duarte, V.; 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]
  48. Jacouton, E.; Maravilla, E.T.; Boucard, A.-S.; Pouderous, N.; Vilela, A.P.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. 2019, 9, 3355. [Google Scholar] [CrossRef]
  49. Han, K.J.; Lee, N.-K.; Park, H.; Paik, H.-D. Anticancer and Anti-Inflammatory Activity of Probiotic Lactococcus lactis NK34. J. Microbiol. Biotechnol. 2015, 25, 1697–1701. [Google Scholar] [CrossRef]
  50. Zaharuddin, L.; Mokhtar, N.M.; Nawawi, K.N.M.; Ali, R.A.R. A randomized double-blind placebo-controlled trial of probiotics in post-surgical colorectal cancer. BMC Gastroenterol. 2019, 19, 131. [Google Scholar] [CrossRef]
  51. Nekouian, R.; Rasouli, B.S.; Ghadimi-Darsajini, A.; Iragian, G.-R. In vitro activity of probiotic Lactobacillus reuteri against gastric cancer progression by downregulation of urokinase plasminogen activator/urokinase plasminogen activator receptor gene expression. J. Cancer Res. Ther. 2017, 13, 246–251. [Google Scholar] [CrossRef]
  52. Adams, C.A. The probiotic paradox: live and dead cells are biological response modifiers. Nutr. Res. Rev. 2010, 23, 37–46. [Google Scholar] [CrossRef]
  53. Mattila-Sandholm, T.; Myllärinen, P.; Crittenden, R.; Mogensen, G.; Fondén, R.; Saarela, M. Technological challenges for future probiotic foods. Int. Dairy J. 2002, 12, 173–182. [Google Scholar] [CrossRef]
  54. Lee, H.A.; Kim, H.; Lee, K.-W.; Park, K.-Y. Dead Nano-SizedLactobacillus plantarumInhibits Azoxymethane/Dextran Sulfate Sodium-Induced Colon Cancer in Balb/c Mice. J. Med. Food 2015, 18, 1400–1405. [Google Scholar] [CrossRef]
  55. 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 Apc Min/+ mouse model. Appl. Microbiol. Biotechnol. 2016, 101, 1999–2019. [Google Scholar] [CrossRef]
  56. McIntosh, G.H. Probiotics and colon cancer prevention. Asia Pac J Clin Nutr. 1996, 5, 48–52. [Google Scholar]
  57. Vivier, E.; Raulet, D.H.; Moretta, A.; Caligiuri, M.A.; Zitvogel, L.; Lanier, L.L.; Yokoyama, W.M.; Ugolini, S. Innate or Adaptive Immunity? The Example of Natural Killer Cells. Science 2011, 331, 44–49. [Google Scholar] [CrossRef]
  58. A Cooper, M.; A Fehniger, T.; A Caligiuri, M. The biology of human natural killer-cell subsets. Trends Immunol. 2001, 22, 633–640. [Google Scholar] [CrossRef] [PubMed]
  59. Sun, J.C.; Lanier, L.L. Is There Natural Killer Cell Memory and Can It Be Harnessed by Vaccination? Cold Spring Harb. Perspect. Biol. 2017, 10, a029538. [Google Scholar] [CrossRef] [PubMed]
  60. Shaw, S.Y.; Tran, K.; Castoreno, A.B.; Peloquin, J.M.; Lassen, K.G.; Khor, B.; Aldrich, L.N.; Tan, P.H.; Graham, D.B.; Kuballa, P.; et al. Selective Modulation of Autophagy, Innate Immunity, and Adaptive Immunity by Small Molecules. ACS Chem. Biol. 2013, 8, 2724–2733. [Google Scholar] [CrossRef]
  61. Pant, H.; Hughes, A.; Miljkovic, D.; Schembri, M.; Wormald, P.; Macardle, P.; Grose, R.; Zola, H.; Krumbiegel, D. Accumulation of Effector Memory CD8+ T Cells in Nasal Polyps. Am. J. Rhinol. Allergy 2013, 27, e117–e126. [Google Scholar] [CrossRef]
  62. Tomala, J.; Chmelova, H.; Mrkvan, T.; Rihova, B.; Kovar, M. In Vivo Expansion of Activated Naive CD8+ T Cells and NK Cells Driven by Complexes of IL-2 and Anti-IL-2 Monoclonal Antibody As Novel Approach of Cancer Immunotherapy. J. Immunol. 2009, 183, 4904–4912. [Google Scholar] [CrossRef]
  63. Tanaka, J.; Toubai, T.; Miura, Y.; Tsutsumi, Y.; Kato, N.; Umehara, S.; Toyoshima, N.; Ohta, S.; Asaka, M.; Imamura, M. Differential expression of natural killer cell receptors (CD94/NKG2A) on T cells by the stimulation of G-CSF-mobilized peripheral blood mononuclear cells with anti-CD3 monoclonal antibody and cytokines: A study in stem cell donors. Transplant. Proc. 2004, 36, 2511–2512. [Google Scholar] [CrossRef]
  64. Burke, S.; Lakshmikanth, T.; Colucci, F.; Carbone, E. New views on natural killer cell-based immunotherapy for melanoma treatment. Trends Immunol. 2010, 31, 339–345. [Google Scholar] [CrossRef]
  65. Larsen, S.K.; Gao, Y.; Basse, P.H. NK cells in the tumor microenvironment. Crit. Rev. Oncog. 2014, 19, 91–105. [Google Scholar] [CrossRef]
  66. Nolibé, D.; Poupon, M.F. Enhancement of Pulmonary Metastases Induced by Decreased Lung Natural Killer Cell Activity. JNCI J. Natl. Cancer Inst. 1986, 77, 99–103. [Google Scholar] [CrossRef]
  67. Imai, K.; Matsuyama, S.; Miyake, S.; Suga, K.; Nakachi, K. Natural cytotoxic activity of peripheral-blood lymphocytes and cancer incidence: an 11-year follow-up study of a general population. Lancet 2000, 356, 1795–1799. [Google Scholar] [CrossRef]
  68. Harning, R.; Koo, G.; Szalay, J. Regulation of the metastasis of murine ocular melanoma by natural-killer cells. Investig. Ophthalmol. Vis. Sci. 1989, 30, 1909–1915. [Google Scholar]
  69. Coca, S.; et al. The prognostic significance of intratumoral natural killer cells in patients with colorectal carcinoma. Cancer 1997, 79, 2320–2328. [Google Scholar] [CrossRef]
  70. Zanetti, M. Tapping CD4 T Cells for Cancer Immunotherapy: The Choice of Personalized Genomics. J. Immunol. 2015, 194, 2049–2056. [Google Scholar] [CrossRef] [PubMed]
  71. Robbins, S.H.; Bessou, G.; Cornillon, A.; Zucchini, N.; Rupp, B.; Ruzsics, Z.; Sacher, T.; Tomasello, E.; Vivier, E.; Koszinowski, U.H.; et al. Natural Killer Cells Promote Early CD8 T Cell Responses against Cytomegalovirus. PLOS Pathog. 2007, 3, e123. [Google Scholar] [CrossRef]
  72. Wodarz, D.; Sierro, S.; Klenerman, P. Dynamics of killer T cell inflation in viral infections. J. R. Soc. Interface 2006, 4, 533–543. [Google Scholar] [CrossRef]
  73. 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]
  74. 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]
  75. 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 tumorsin vivoin humanized BLT mice; effect on tumor differentiation and response to chemotherapeutic drugs. OncoImmunology 2018, 7, e1426518. [Google Scholar] [CrossRef]
  76. Gogali, F.; Paterakis, G.; Rassidakis, G.Z.; Liakou, C.I.; Liapi, C. CD3CD16CD56brightImmunoregulatory NK Cells are Increased in the Tumor Microenvironment and Inversely Correlate with Advanced Stages in Patients with Papillary Thyroid Cancer. Thyroid 2013, 23, 1561–1568. [Google Scholar] [CrossRef]
  77. López-Cobo, S.; Pieper, N.; Campos-Silva, C.; García-Cuesta, E.M.; Reyburn, H.T.; Paschen, A.; Valés-Gómez, M. Impaired NK cell recognition of vemurafenib-treated melanoma cells is overcome by simultaneous application of histone deacetylase inhibitors. OncoImmunology 2017, 7, e1392426. [Google Scholar] [CrossRef]
  78. Ciszak, L. Impaired ζ chain expression and IFN-γ production in peripheral blood T and NK cells of patients with advanced lung cancer. Oncol. Rep. 1994, 21, 173–184. [Google Scholar] [CrossRef]
  79. 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] [PubMed]
  80. 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, 1–17. [Google Scholar] [CrossRef] [PubMed]
  81. Ko, M.-W.; Kaur, K.; Safaei, T.; Chen, W.; Sutanto, C.; Wong, P.; Jewett, A. Defective Patient NK Function Is Reversed by AJ2 Probiotic Bacteria or Addition of Allogeneic Healthy Monocytes. Cells 2022, 11, 697. [Google Scholar] [CrossRef] [PubMed]
  82. Bui, V.T.; Tseng, H.-C.; Kozlowska, A.; Maung, P.O.; Kaur, K.; Topchyan, P.; Jewett, A. Augmented IFN-γ and TNF-α 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]
  83. 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]
  84. Fujisaki, H.; Kakuda, H.; Shimasaki, N.; Imai, C.; Ma, J.; Lockey, T.; Eldridge, P.; Leung, W.H.; Campana, D. Expansion of Highly Cytotoxic Human Natural Killer Cells for Cancer Cell Therapy. Cancer Res. 2009, 69, 4010–4017. [Google Scholar] [CrossRef] [PubMed]
  85. Leivas, A.; Perez-Martínez, A.; Blanchard, M.J.; Martín-Clavero, E.; Fernández, 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] [PubMed]
  86. 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]
  87. 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]
  88. 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] [PubMed]
  89. Gianotti, L.; Morelli, L.; Galbiati, F.; Rocchetti, S.; Coppola, S.; Beneduce, A.; Gilardini, C.; Zonenschain, D.; Nespoli, A.; Braga, M. A randomized double-blind trial on perioperative administration of probiotics in colorectal cancer patients. World J. Gastroenterol. 2010, 16, 167–175. [Google Scholar] [CrossRef]
  90. 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]
  91. Aziz, N.; Bonavida, B. Activation of Natural Killer Cells by Probiotics. Forum Immunopathol. Dis. Ther. 2016, 7, 41–55. [Google Scholar] [CrossRef]
  92. Takagi, A.; Ikemura, H.; Matsuzaki, T.; Sato, M.; Nomoto, K.; Morotomi, M.; Yokokura, T. Relationship between the in vitro response of dendritic cells to Lactobacillus and prevention of tumorigenesis in the mouse. J. Gastroenterol. 2008, 43, 661–669. [Google Scholar] [CrossRef]
  93. Hachimura, S.; Totsuka, M.; Hosono, A. Immunomodulation by food: impact on gut immunity and immune cell function. Biosci. Biotechnol. Biochem. 2018, 82, 584–599. [Google Scholar] [CrossRef]
  94. 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 2018, 11, 857–864. [Google Scholar] [CrossRef]
  95. 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]
  96. 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. 2014, 44, 1–11. [Google Scholar] [CrossRef] [PubMed]
  97. 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] [PubMed]
  98. 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 tumorsin vivoin humanized BLT mice; effect on tumor differentiation and response to chemotherapeutic drugs. OncoImmunology 2018, 7, e1426518. [Google Scholar] [CrossRef] [PubMed]
  99. 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]
  100. Yazdi, M.H.; Dallal, M.M.S.; Hassan, Z.M.; Holakuyee, M.; Amiri, S.A.; Abolhassani, M.; Mahdavi, M. Oral administration of Lactobacillus acidophilusinduces IL-12 production in spleen cell culture of BALB/c mice bearing transplanted breast tumour. Br. J. Nutr. 2010, 104, 227–232. [Google Scholar] [CrossRef]
  101. Fooladi, A.A.I.; Yazdi, M.H.; Pourmand, M.R.; Mirshafiey, A.; Hassan, Z.M.; Azizi, T.; Mahdavi, M.; Dallal, M.M.S. Th1 Cytokine Production Induced by Lactobacillus acidophilus in BALB/c Mice Bearing Transplanted Breast Tumor. Jundishapur J. Microbiol. 2015, 8, e17354. [Google Scholar] [CrossRef]
  102. Zhang, M.; Wang, F.; Jiang, L.; Liu, R.; Zhang, L.; Lei, X.; Li, J.; Jiang, J.; Guo, H.; Fang, B.; et al. Lactobacillus Salivarius REN Inhibits Rat Oral Cancer Induced by 4-Nitroquioline 1-Oxide. Cancer Prev. Res. 2013, 6, 686–694. [Google Scholar] [CrossRef]
  103. Kamaluddin, W.N.F.W.M.; Rismayuddin, N.A.R.; Ismail, A.F.; Aidid, E.M.; 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]
  104. 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]
  105. Ohara, T.; Suzutani, T. Intake ofBifidobacterium longumand Fructooligosaccharides prevents Colorectal Carcinogenesis. Euroasian J. Hepato-Gastroenterology 2018, 8, 11–17. [Google Scholar] [CrossRef]
  106. Fahmy, C.A.; Gamal-Eldeen, A.M.; El-Hussieny, E.A.; Raafat, B.M.; Mehanna, N.S.; Talaat, R.M.; Shaaban, M.T. Bifidobacterium longumSuppresses Murine Colorectal Cancer through the Modulation of oncomiRs and Tumor Suppressor miRNAs. Nutr. Cancer 2019, 71, 688–700. [Google Scholar] [CrossRef] [PubMed]
  107. Reddy, B.S.; Rivenson, A. Inhibitory effect of Bifidobacterium longum on colon, mammary, and liver carcinogenesis induced by 2-amino-3-methylimidazo[4,5-f]quinoline, a food mutagen. Cancer Res. 1993, 53, 3914–3918. [Google Scholar]
  108. 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]
  109. 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 Apc Min/+ mouse model. Appl. Microbiol. Biotechnol. 2016, 101, 1999–2019. [Google Scholar] [CrossRef]
  110. 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]
  111. 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]
  112. Sasaki, T.; Fujimori, M.; Hamaji, Y.; Hama, Y.; Ito, K.; Amano, J.; Taniguchi, S. Genetically engineered Bifidobacterium longum for tumor-targeting enzyme-prodrug therapy of autochthonous mammary tumors in rats. Cancer Sci. 2006, 97, 649–657. [Google Scholar] [CrossRef] [PubMed]
  113. Chang, J.-H.; Shim, Y.Y.; Cha, S.-K.; Reaney, M.J.T.; Chee, K.M. Effect of Lactobacillus acidophilus KFRI342 on the development of chemically induced precancerous growths in the rat colon. J. Med Microbiol. 2012, 61, 361–368. [Google Scholar] [CrossRef] [PubMed]
  114. 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, 2719–28. [Google Scholar] [PubMed]
  115. Yazdi, M.H.; Mahdavi, M.; Kheradmand, E.; Shahverdi, A.R. The Preventive Oral Supplementation of a Selenium Nanoparticle-enriched Probiotic Increases the Immune Response and Lifespan of 4T1 Breast Cancer Bearing Mice. Arzneimittelforschung 2012, 62, 525–531. [Google Scholar] [CrossRef]
  116. 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]
  117. Shin, R.; Itoh, Y.; Kataoka, M.; Iino-Miura, S.; Miura, R.; Mizutani, T.; Fujisawa, T. Anti-tumor activity of heat-killedLactobacillus plantarumBF-LP284 on Meth-A tumor cells in BALB/c mice. Int. J. Food Sci. Nutr. 2016, 67, 641–649. [Google Scholar] [CrossRef]
  118. 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]
  119. Wada, M.; Nagata, S.; Saito, M.; Shimizu, T.; Yamashiro, Y.; Matsuki, T.; Asahara, T.; Nomoto, K. Effects of the enteral administration of Bifidobacterium breve on patients undergoing chemotherapy for pediatric malignancies. Support. Care Cancer 2009, 18, 751–759. [Google Scholar] [CrossRef]
  120. Urbancsek, H.; Kazar, T.; Mezes, I.; Neumann, K. Results of a double-blind, randomized study to evaluate the efficacy and safety of Antibiophilus® in patients with radiation-induced diarrhoea. Eur. J. Gastroenterol. Hepatol. 2001, 13, 391–396. [Google Scholar] [CrossRef]
  121. Giralt, J.; Regadera, J.P.; Verges, R.; Romero, J.; de la Fuente, I.; Biete, A.; Villoria, J.; Cobo, J.M.; Guarner, F. Effects of Probiotic Lactobacillus Casei DN-114 001 in Prevention of Radiation-Induced Diarrhea: Results From Multicenter, Randomized, Placebo-Controlled Nutritional Trial. Int. J. Radiat. Oncol. 2008, 71, 1213–1219. [Google Scholar] [CrossRef]
  122. Delia, P.; Sansotta, G.; Donato, V.; Messina, G.; Frosina, P.; Pergolizzi, S.; De Renzis, C.; Famularo, G. Prevention of radiation-induced diarrhea with the use of VSL#3, a new high-potency probiotic preparation. Am. J. Gastroenterol. 2002, 97, 2150–2152. [Google Scholar] [CrossRef] [PubMed]
  123. Delia, P. Use of probiotics for prevention of radiation-induced diarrhea. World J. Gastroenterol. 2007, 13, 912–5. [Google Scholar] [CrossRef]
  124. Chitapanarux, I.; Chitapanarux, T.; Traisathit, P.; Kudumpee, S.; Tharavichitkul, E.; Lorvidhaya, V. Randomized controlled trial of live lactobacillus acidophilus plus bifidobacterium bifidum in prophylaxis of diarrhea during radiotherapy in cervical cancer patients. Radiat. Oncol. 2010, 5, 31–31. [Google Scholar] [CrossRef]
  125. Mego, M.; Holec, V.; Drgona, L.; Hainova, K.; Ciernikova, S.; Zajac, V. Probiotic bacteria in cancer patients undergoing chemotherapy and radiation therapy. Complement. Ther. Med. 2013, 21, 712–723. [Google Scholar] [CrossRef]
  126. Goyani, P.; Christodoulou, R.; Vassiliou, E. Immunosenescence: Aging and Immune System Decline. Vaccines 2024, 12, 1314. [Google Scholar] [CrossRef]
  127. Nguyen, T.Q.T.; A Cho, K. Targeting immunosenescence and inflammaging: advancing longevity research. Exp. Mol. Med. 2025, 1–12. [Google Scholar] [CrossRef]
  128. Solana, R.; Pawelec, G.; Tarazona, R. Aging and Innate Immunity. Immunity 2006, 24, 491–494. [Google Scholar] [CrossRef] [PubMed]
  129. Gounder, S.S.; Abdullah, B.J.J.; Radzuanb, N.E.I.B.M.; Zain, F.D.B.M.; 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, 1–6. [Google Scholar] [CrossRef] [PubMed]
  130. Qi, C.; Liu, Q. Natural killer cells in aging and age-related diseases. Neurobiol. Dis. 2023, 183, 106156. [Google Scholar] [CrossRef] [PubMed]
  131. 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]
  132. Le Garff-Tavernier, M.; Béziat, V.; Decocq, J.; Siguret, V.; Gandjbakhch, F.; Pautas, E.; Debré, P.; Merle-Beral, H.; Vieillard, V. Human NK cells display major phenotypic and functional changes over the life span. Aging Cell 2010, 9, 527–535. [Google Scholar] [CrossRef]
  133. Camous, X.; Pera, A.; Solana, R.; Larbi, A. NK Cells in Healthy Aging and Age-Associated Diseases. J. Biomed. Biotechnol. 2012, 2012, 1–8. [Google Scholar] [CrossRef]
  134. Solana, R.; Campos, C.; Pera, A.; Tarazona, R. Shaping of NK cell subsets by aging. Curr. Opin. Immunol. 2014, 29, 56–61. [Google Scholar] [CrossRef]
  135. Hak, Ł.; Myśliwska, J.; Więckiewicz, J.; Szyndler, K.; Trzonkowski, P.; Siebert, J.; Myśliwski, A. NK cell compartment in patients with coronary heart disease. Immun. Ageing 2007, 4, 3–3. [Google Scholar] [CrossRef]
  136. Fasbender, F.; Widera, A.; Hengstler, J.G.; Watzl, C. Natural Killer Cells and Liver Fibrosis. Front. Immunol. 2016, 7, 19. [Google Scholar] [CrossRef]
  137. 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, 1–29. [Google Scholar] [CrossRef]
  138. 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]
  139. Hazeldine, J.; Lord, J.M. The impact of ageing on natural killer cell function and potential consequences for health in older adults. Ageing Res. Rev. 2013, 12, 1069–1078. [Google Scholar] [CrossRef] [PubMed]
  140. 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]
  141. 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]
  142. Bosco, N.; Noti, M. The aging gut microbiome and its impact on host immunity. Genes Immun. 2021, 22, 289–303. [Google Scholar] [CrossRef]
  143. 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. [Google Scholar] [CrossRef]
  144. 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, 1–20. [Google Scholar] [CrossRef]
  145. 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]
  146. Virk, M. S.; 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] [PubMed]
  147. Dempsey, E.; Corr, S.C. Lactobacillus spp. for Gastrointestinal Health: Current and Future Perspectives. Front. Immunol. 2022, 13, 840245. [Google Scholar] [CrossRef]
  148. 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]
  149. 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]
  150. 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]
  151. Yao, S.; Zhao, Z.; Wang, W.; Liu, X. Bifidobacterium Longum: Protection against Inflammatory Bowel Disease. J. Immunol. Res. 2021, 2021, 1–11. [Google Scholar] [CrossRef]
  152. Hidalgo-Cantabrana, C.; Delgado, S.; Ruiz, L.; Ruas-Madiedo, P.; Sánchez, B.; Margolles, A.; Britton, R.A.; Cani, P.D. Bifidobacteria and Their Health-Promoting Effects. Microbiol. Spectr. 2017, 5. [Google Scholar] [CrossRef]
  153. 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] [PubMed]
  154. Tsai, Y.-C.; Cheng, L.-H.; Liu, Y.-W.; Jeng, O.-J.; Lee, Y.-K. Gerobiotics: probiotics targeting fundamental aging processes. Biosci. Microbiota, Food Health 2021, 40, 1–11. [Google Scholar] [CrossRef]
  155. Mahamud, A.U.; Tanvir, I.A.; Kabir, E.; Samonty, I.; Chowdhury, A.H.; Rahman, A. Gerobiotics: Exploring the Potential and Limitations of Repurposing Probiotics in Addressing Aging Hallmarks and Chronic Diseases. Probiotics Antimicrob. Proteins 2025, 1–41. [Google Scholar] [CrossRef]
  156. Choudhary, P.; Kathuria, D.; Suri, S.; Bahndral, A.; Naveen, A.K. Probiotics- its functions and influence on the ageing process: A comprehensive review. Food Biosci. 2023, 52. [Google Scholar] [CrossRef]
  157. 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]
  158. 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]
  159. 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]
  160. Hollander, D.; Kaunitz, J.D. The “Leaky Gut”: Tight Junctions but Loose Associations? Dig. Dis. Sci. 2020, 65, 1277–1287. [Google Scholar] [CrossRef] [PubMed]
  161. 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]
  162. 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]
  163. Liu, Y.; Alookaran, J.J.; Rhoads, J.M. Probiotics in Autoimmune and Inflammatory Disorders. Nutrients 2018, 10, 1537. [Google Scholar] [CrossRef]
  164. 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]
  165. Mandel, D.R.; Eichas, K.; Holmes, J. Bacillus coagulans: a viable adjunct therapy for relieving symptoms of rheumatoid arthritis according to a randomized, controlled trial. BMC Complement. Altern. Med. 2010, 10, 1. [Google Scholar] [CrossRef]
  166. 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]
  167. Sr, D.K.; Krishnamurty, A.Y.; A, P.; Chander, A. Single Strain vs Multiple Strain Probiotics: The Clinician's Choice. Cureus 2025, 17, e86353. [Google Scholar] [CrossRef] [PubMed]
  168. Giri, P. S., Shah, F. & Dwivedi, M. K. in Probiotics in the Prevention and Management of Human Diseases (eds Mitesh Kumar Dwivedi, N. Amaresan, A. Sankaranarayanan, & E. Helen Kemp) 161-186 (Academic Press, 2022).
  169. Mirfeizi, Z.; Mahmoudi, M.; Faridzadeh, A. Probiotics as a complementary treatment in systemic lupus erythematosus: A systematic review. Heal. Sci. Rep. 2023, 6, e1640. [Google Scholar] [CrossRef]
  170. Mohamed, 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, 1–13. [Google Scholar] [CrossRef]
  171. Nikolova, V.L.; Cleare, A.J.; Young, A.H.; Stone, J.M. Updated Review and Meta-Analysis of Probiotics for the Treatment of Clinical Depression: Adjunctive vs. Stand-Alone Treatment. J. Clin. Med. 2021, 10, 647. [Google Scholar] [CrossRef] [PubMed]
  172. Yang, Y.; Day, J.; Guimaraes, F.S.; Wicks, I.P.; Louis, C. Natural killer cells in inflammatory autoimmune diseases. Clin. Transl. Immunol. 2021, 10, e1250. [Google Scholar] [CrossRef] [PubMed]
  173. de Salis, L. V. V. , Martins, L. S., Rodrigues, G. S. P. & de Oliveira, G. L. V. in Translational Autoimmunity Vol. 2 (ed Nima Rezaei) 269-294 (Academic Press, 2022).
  174. 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. (Lausanne) 2018, 5, 124. [Google Scholar] [CrossRef]
  175. Zhang, W.; Xiao, D.; Mao, Q.; Xia, H. Role of neuroinflammation in neurodegeneration development. Signal Transduct. Target. Ther. 2023, 8, 1–32. [Google Scholar] [CrossRef]
  176. 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]
  177. 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, 1–53. [Google Scholar] [CrossRef]
  178. 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] [PubMed]
  179. 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. [Google Scholar] [CrossRef] [PubMed]
  180. Origüela, V.; Lopez-Zaplana, A. Gut Microbiota: An Immersion in Dysbiosis, Associated Pathologies, and Probiotics. Microorganisms 2025, 13, 1084. [Google Scholar] [CrossRef]
  181. 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, 1–20. [Google Scholar] [CrossRef]
  182. 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] [PubMed]
  183. 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]
  184. 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] [PubMed]
  185. 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]
  186. 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]
  187. 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] [PubMed]
  188. Latif, A.; Shehzad, A.; Niazi, S.; Zahid, A.; Ashraf, W.; Iqbal, M.W.; Rehman, A.; Riaz, T.; Aadil, R.M.; Khan, I.M.; et al. Probiotics: mechanism of action, health benefits and their application in food industries. Front. Microbiol. 2023, 14, 1216674. [Google Scholar] [CrossRef]
  189. 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]
  190. 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, 1–13. [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.
Copyright: This open access article is published under a Creative Commons CC BY 4.0 license, which permit the free download, distribution, and reuse, provided that the author and preprint are cited in any reuse.
Prerpints.org logo

Preprints.org is a free preprint server supported by MDPI in Basel, Switzerland.

Subscribe

Disclaimer

Terms of Use

Privacy Policy

Privacy Settings

© 2025 MDPI (Basel, Switzerland) unless otherwise stated