Review Article | | Peer-Reviewed

Probiotics Application in the Treatment of Autoimmune Diseases and Enhancement of Efficacy Through Genetic Engineering

Received: 30 August 2024     Accepted: 7 November 2024     Published: 28 November 2024
Views:       Downloads:
Abstract

The development of multidrug resistance in most pathogenic microorganisms and the rapidly increasing prevalence of non-communicable diseases are becoming major health concerns worldwide. Among non-communicable diseases, autoimmune diseases are caused mainly by imbalances in the gut microbiota (dysbiosis). Gut microbiota colonization and immune system establishment started in the early years of life. A defect in the gut microbiota predominantly affects the proper functioning of immune cells. Hence, restoring gut dysbiosis has received considerable attention for the last few decades as a potential therapeutic option. In this regard, probiotics have been the focus of research during recent decades because of their safe history of use along with fermented foods and beverages. Currently, advanced research is being conducted on the use of probiotics as immunomodulatory mediators and for the amelioration of gut dysbiosis as therapeutic adjuncts in the treatment of autoimmune diseases. In addition, probiotics are genetically engineered to enhance treatment efficacy and to develop live biotherapeutics (LBP). In this review, research articles summarizing findings in autoimmune disease treatment via probiotic strains, emphasizing type 1 diabetes, rheumatoid arthritis, Graves' disease, systemic lupus erythematosus and inflammatory bowel disease in both clinical trials and animal models, were reviewed. Finally, promising results of genetic engineering of probiotics for use as biosensors, delivery of therapeutic proteins, and diagnosis of infections were reported.

Published in American Journal of Bioscience and Bioengineering (Volume 12, Issue 6)
DOI 10.11648/j.bio.20241206.12
Page(s) 97-115
Creative Commons

This is an Open Access article, distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution and reproduction in any medium or format, provided the original work is properly cited.

Copyright

Copyright © The Author(s), 2024. Published by Science Publishing Group

Keywords

Autoimmune, Dysbiosis, Genetic Engineering, Gut Microbiota, Probiotics

References
[1] Aladeboyeje O, Şanli NÖ. Fermented Traditional Probiotic Beverages of Turkish Origin: A Concise Review. International Journal of Life Sciences and Biotechnology 2021; 4: 546–64.
[2] Hill C, Guarner F, Reid G, Gibson GR, Merenstein DJ, Pot B, et al. The International Scientific Association for Probiotics and Prebiotics consensus statement on the scope and appropriate use of the term probiotic. Nat Rev Gastroenterol Hepatol 2014; 11: 506–14.
[3] Binda S, Hill C, Johansen E, Obis D, Pot B, Sanders ME, et al. Criteria to Qualify Microorganisms as “Probiotic” in Foods and Dietary Supplements. Front Microbiol 2020; 11: 1662.
[4] Pandey KavitaR, Naik SureshR, Vakil BabuV. Probiotics, prebiotics and synbiotics- a review. J Food Sci Technol 2015; 52: 7577–87.
[5] Sener, Bulut, Güneş Bayir. Probiotics and Relationship Between Probiotics and Cancer Types. Bezmialem Science 2021; 9: 490–7.
[6] Zhao, Liao, Wei, Ma, Wang F, Chen, et al. Potential Ability of Probiotics in the Prevention and Treatment of Colorectal Cancer. Clin Med Insights Oncol 2023; 17: 11795549231188225.
[7] Mugwanda K, Hamese S, Van Zyl WF, Prinsloo E, Du Plessis M, Dicks LMT, et al. Recent advances in genetic tools for engineering probiotic lactic acid bacteria. Bioscience Reports 2023; 43: BSR20211299.
[8] Al-Salami H, Caccetta R, Golocorbin-Kon S, Mikov M. Probiotics Applications in Autoimmune Diseases. In: Rigobelo E, editor. Probiotics, InTech; 2012.
[9] Lerner A, Jeremias P, Matthias T. The World Incidence and Prevalence of Autoimmune Diseases is Increasing. IJCD 2016; 3: 151–5.
[10] Liu Y, Alookaran JJ, Rhoads JM. Probiotics in Autoimmune and Inflammatory Disorders. Nutrients 2018; 10: 1537.
[11] Hara N, Alkanani AK, Ir D, Robertson CE, Wagner BD, Frank DN, et al. The role of the intestinal microbiota in type 1 diabetes. Clinical Immunology 2013; 146: 112–9.
[12] Mishra, Wang S, Nagpal R, Miller B, Singh R, Taraphder S, et al. Probiotics and Prebiotics for the Amelioration of Type 1 Diabetes: Present and Future Perspectives. Microorganisms 2019; 7: 67.
[13] Drexhage HA, Dik WA, Leenen PJM, Versnel MA. The Immune Pathogenesis of Type 1 Diabetes: Not Only Thinking Outside the Cell but Also Outside the Islet and Out of the Box. Diabetes 2016; 65: 2130–3.
[14] Paschou SA, Papadopoulou-Marketou N, Chrousos GP, Kanaka-Gantenbein C. On type 1 diabetes mellitus pathogenesis. Endocrine Connections 2018; 7: R38–46.
[15] Dovi KS, Bajinka O, Conteh I. Evidence and possible mechanisms of probiotics in the management of type 1 diabetes mellitus. J Diabetes Metab Disord 2022; 21: 1081–94.
[16] Zhang X, Zhang Y, Luo L, Le Y, Li Y, Yuan F, et al. The Beneficial Effects of a Multispecies Probiotic Supplement on Glycaemic Control and Metabolic Profile in Adults with Type 1 Diabetes: A Randomised, Double-Blinded, Placebo-Controlled Pilot-Study. DMSO 2023; Volume 16: 829–40.
[17] Kamada N, Seo S-U, Chen GY, Núñez G. Role of the gut microbiota in immunity and inflammatory disease. Nat Rev Immunol 2013; 13: 321–35.
[18] Harbison JE, Roth‐Schulze AJ, Giles LC, Tran CD, Ngui KM, Penno MA, et al. Gut microbiome dysbiosis and increased intestinal permeability in children with islet autoimmunity and type 1 diabetes: A prospective cohort study. Pediatr Diabetes 2019: pedi.12865.
[19] Valladares R, Sankar D, Li N, Williams E, Lai K-K, Abdelgeliel AS, et al. Lactobacillus johnsonii N6.2 Mitigates the Development of Type 1 Diabetes in BB-DP Rats. PLoS ONE 2010; 5: e10507.
[20] Dolpady J, Sorini C, Di Pietro C, Cosorich I, Ferrarese R, Saita D, et al. Oral Probiotic VSL#3 Prevents Autoimmune Diabetes by Modulating Microbiota and Promoting Indoleamine 2,3-Dioxygenase-Enriched Tolerogenic Intestinal Environment. Journal of Diabetes Research 2016; 2016: 1–12.
[21] Paun A, Yau C, Danska JS. The Influence of the Microbiome on Type 1 Diabetes. The Journal of Immunology 2017; 198: 590–5.
[22] Vatanen T, Franzosa EA, Schwager R, Tripathi S, Arthur TD, Vehik K, et al. The human gut microbiome in early-onset type 1 diabetes from the TEDDY study. Nature 2018; 562: 589–94.
[23] Zhang Y-Z. Inflammatory bowel disease: Pathogenesis. WJG 2014; 20: 91.
[24] Marcial GE, Ford AL, Haller MJ, Gezan SA, Harrison NA, Cai D, et al. Lactobacillus johnsonii N6.2 Modulates the Host Immune Responses: A Double-Blind, Randomized Trial in Healthy Adults. Front Immunol 2017; 8: 655.
[25] Ferro M, Charneca S, Dourado E, Guerreiro CS, Fonseca JE. Probiotic Supplementation for Rheumatoid Arthritis: A Promising Adjuvant Therapy in the Gut Microbiome Era. Front Pharmacol 2021; 12: 711788.
[26] De Oliveira GLV, Leite AZ, Higuchi BS, Gonzaga MI, Mariano VS. Intestinal dysbiosis and probiotic applications in autoimmune diseases. Immunology 2017; 152: 1–12.
[27] Paul AK, Paul A, Jahan R, Jannat K, Bondhon TA, Hasan A, et al. Probiotics and Amelioration of Rheumatoid Arthritis: Significant Roles of Lactobacillus casei and Lactobacillus acidophilus. Microorganisms 2021; 9: 1070.
[28] Bungau SG, Behl T, Singh A, Sehgal A, Singh S, Chigurupati S, et al. Targeting Probiotics in Rheumatoid Arthritis. Nutrients 2021; 13: 3376.
[29] Asoudeh F, Djafarian K, Akhalghi M, Mahmoudi M, Jamshidi AR, Farhadi E, et al. The effect of probiotic cheese consumption on inflammatory and anti-inflammatory markers, disease severity, and symptoms in patients with rheumatoid arthritis: study protocol for a randomized, double-blind, placebo-controlled trial. Trials 2022; 23: 180.
[30] Hatakka K, Martio J, Korpela M, Herranen M, Poussa T, Laasanen T, et al. Effects of probiotic therapy on the activity and activation of mild rheumatoid arthritis – a pilot study. Scandinavian Journal of Rheumatology 2003; 32: 211–5.
[31] Abhari K, Shekarforoush SS, Hosseinzadeh S, Nazifi S, Sajedianfard J, Eskandari MH. The effects of orally administered Bacillus coagulans and inulin on prevention and progression of rheumatoid arthritis in rats. Food & Nutrition Research 2016; 60: 30876.
[32] Vaghef-Mehrabany E, Alipour B, Homayouni-Rad A, Sharif S-K, Asghari-Jafarabadi M, Zavvari S. Probiotic supplementation improves inflammatory status in patients with rheumatoid arthritis. Nutrition 2014; 30: 430–5.
[33] Mohammed, Khattab, Ahmed, Turk, Sakr, M. Khalil A, et al. The therapeutic effect of probiotics on rheumatoid arthritis: a systematic review and meta-analysis of randomized control trials. Clin Rheumatol 2017; 36: 2697–707.
[34] Achi SC, Talahalli RR, Halami PM. Prophylactic effects of probiotic Bifidobacterium spp. in the resolution of inflammation in arthritic rats. Appl Microbiol Biotechnol 2019; 103: 6287–96.
[35] Amdekar, Singh, Kumar, Sharma, Singh. Lactobacillus casei and Lactobacillus acidophilus Regulate Inflammatory Pathway and Improve Antioxidant Status in Collagen-Induced Arthritic Rats. Journal of Interferon & Cytokine Research 2013; 33: 1–8.
[36] Fan, Yang, Ross, Stanton, Shi, Zhao J, et al. Protective effects of Bifidobacterium adolescentis on collagen-induced arthritis in rats depend on timing of administration. Food Funct 2020; 11: 4499–511.
[37] Alipour B, Homayouni‐Rad A, Vaghef‐Mehrabany E, Sharif SK, Vaghef‐Mehrabany L, Asghari‐Jafarabadi M, et al. Effects of L actobacillus casei supplementation on disease activity and inflammatory cytokines in rheumatoid arthritis patients: a randomized double–blind clinical trial. Int J of Rheum Dis 2014; 17: 519–27.
[38] Aqaeinezhad Rudbane SM, Rahmdel S, Abdollahzadeh SM, Zare M, Bazrafshan A, Mazloomi SM. The efficacy of probiotic supplementation in rheumatoid arthritis: a meta-analysis of randomized, controlled trials. Inflammopharmacol 2018; 26: 67–76.
[39] Cannarella, Mari, Alcântara, Iryioda, Costa, Oliveira SR, et al. Mixture of probiotics reduces inflammatory biomarkers and improves the oxidative/nitrosative profile in people with rheumatoid arthritis. Nutrition 2021; 89: 111282.
[40] Bodkhe R, Balakrishnan B, Taneja V. The role of microbiome in rheumatoid arthritis treatment. Therapeutic Advances in Musculoskeletal 2019; 11: 1759720X1984463.
[41] Pružinská K, Slovák L, Dráfi F, Poništ S, Juránek I, Chrastina M, et al. Enhanced Anti-Inflammatory Effect of the Combination of Lactiplantibacillus plantarum LS/07 with Methotrexate Compared to Their Monotherapies Studied in Experimental Arthritis. Molecules 2022; 28: 297.
[42] Smith TJ, Hegedüs L. Graves’ Disease. N Engl J Med 2016; 375: 1552–65.
[43] Khan M, S. Lone S, Faiz S, Farooq I, Majid S. Graves’ Disease: Pathophysiology, Genetics and Management. In: Gensure R, editor. Graves’ Disease, IntechOpen; 2021.
[44] Deng Y, Wang J, Xie G, Zou G, Li S, Zhang J, et al. Correlation between gut microbiota and the development of Graves’ disease: A prospective study. iScience 2023; 26: 107188.
[45] Lane LC, Wood CL, Cheetham T. Graves’ disease: moving forwards. Arch Dis Child 2023; 108: 276–81.
[46] Jacobson EM, Tomer Y. The CD40, CTLA-4, thyroglobulin, TSH receptor, and PTPN22 gene quintet and its contribution to thyroid autoimmunity: Back to the future. Journal of Autoimmunity 2007; 28: 85–98.
[47] Knezevic J, Starchl C, Tmava Berisha A, Amrein K. Thyroid-Gut-Axis: How Does the Microbiota Influence Thyroid Function? Nutrients 2020; 12: 1769.
[48] Frohlich E, Wahl R. Microbiota and Thyroid Interaction in Health and Disease. Trends in Endocrinology & Metabolism 2019; 30: 479–90.
[49] Shu Q, Kang C, Li J, Hou Z, Xiong M, Wang X, et al. Effect of probiotics or prebiotics on thyroid function: A meta-analysis of eight randomized controlled trials. PLoS ONE 2024; 19: e0296733.
[50] Moshkelgosha S, Verhasselt HL, Masetti G, Covelli D, Biscarini F, Horstmann M, et al. Modulating gut microbiota in a mouse model of Graves’ orbitopathy and its impact on induced disease. Microbiome 2021; 9: 45.
[51] Lin B, Zhao F, Liu Y, Wu X, Feng J, Jin X, et al. Randomized Clinical Trial: Probiotics Alleviated Oral-Gut Microbiota Dysbiosis and Thyroid Hormone Withdrawal-Related Complications in Thyroid Cancer Patients Before Radioiodine Therapy Following Thyroidectomy. Front Endocrinol 2022; 13: 834674.
[52] Huo D, Cen C, Chang H, Ou Q, Jiang S, Pan Y, et al. Probiotic Bifidobacterium longum supplied with methimazole improved the thyroid function of Graves’ disease patients through the gut-thyroid axis. Commun Biol 2021; 4: 1046.
[53] Spaggiari G, Brigante G, De Vincentis S, Cattini U, Roli L, De Santis MC, et al. Probiotics Ingestion Does Not Directly Affect Thyroid Hormonal Parameters in Hypothyroid Patients on Levothyroxine Treatment. Front Endocrinol 2017; 8: 316.
[54] Brusca SB, Abramson SB, Scher JU. Microbiome and mucosal inflammation as extra-articular triggers for rheumatoid arthritis and autoimmunity. Current Opinion in Rheumatology 2014; 26: 101–7.
[55] Mok CC, Lau CS. Pathogenesis of systemic lupus erythematosus. J Clin Pathol 2003; 56: 481–90.
[56] Wang Y, Zhu D, Ortiz-Velez LC, Perry JL, Pennington MW, Hyser JM, et al. A bioengineered probiotic for the oral delivery of a peptide Kv1.3 channel blocker to treat rheumatoid arthritis. Proc Natl Acad Sci USA 2023; 120: e2211977120.
[57] Liu Z, Davidson A. Taming lupus—a new understanding of pathogenesis is leading to clinical advances. Nat Med 2012; 18: 871–82.
[58] Lopez P, Sánchez B, Margolles A, Suárez A. Intestinal dysbiosis in systemic lupus erythematosus: cause or consequence? Current Opinion in Rheumatology 2016; 28: 515–22.
[59] Kim J-W, Kwok S-K, Choe J-Y, Park S-H. Recent Advances in Our Understanding of the Link between the Intestinal Microbiota and Systemic Lupus Erythematosus. IJMS 2019; 20: 4871.
[60] Mirfeizi, Mahmoudi, Faridzadeh. Probiotics as a complementary treatment in systemic lupus erythematosus: A systematic review. Health Science Reports 2023; 6: e1640.
[61] Guo X, Yang X, Li Q, Shen X, Zhong H, Yang Y. The Microbiota in Systemic Lupus Erythematosus: An Update on the Potential Function of Probiotics. Front Pharmacol 2021; 12: 759095.
[62] Ugarte-Gil MF, González LA, Alarcón GS. Lupus: the new epidemic. Lupus 2019; 28: 1031–50.
[63] Bagavant H, Dunkleberger ML, Wolska N, Sroka M, Rasmussen A, Adrianto I, et al. Antibodies to Periodontogenic Bacteria are Associated with Higher Disease Activity in Lupus Patients 2019. ClinExpRheumatol. 2019; 37(1): 106–111.
[64] Khorasani S, Mahmoudi M, Kalantari MR, Lavi Arab F, Esmaeili S, Mardani F, et al. Amelioration of regulatory T cells by Lactobacillus delbrueckii and Lactobacillus rhamnosus in pristane‐induced lupus mice model. Journal Cellular Physiology 2019; 234: 9778–86.
[65] Mu Q, Zhang H, Liao X, Lin K, Liu H, Edwards MR, et al. Control of lupus nephritis by changes of gut microbiota. Microbiome 2017; 5: 73.
[66] Cabana-Puig X, Mu Q, Lu R, Swartwout B, Abdelhamid L, Zhu J, et al. Lactobacillus spp. act in synergy to attenuate splenomegaly and lymphadenopathy in lupus-prone MRL/lpr mice. Front Immunol 2022; 13: 923754.
[67] Kim DS, Park Y, Choi J-W, Park S-H, Cho M-L, Kwok S-K. Lactobacillus acidophilus Supplementation Exerts a Synergistic Effect on Tacrolimus Efficacy by Modulating Th17/Treg Balance in Lupus-Prone Mice via the SIGNR3 Pathway. Front Immunol 2021; 12: 696074.
[68] Hevia A, Milani C, López P, Cuervo A, Arboleya S, Duranti S, et al. Intestinal Dysbiosis Associated with Systemic Lupus Erythematosus. mBio 2014; 5: e01548-14.
[69] Chen, Lin, Xiao, Zhang, Zhao, Wang, et al. Gut microbiota in systemic lupus erythematosus: A fuse and a solution. Journal of Autoimmunity 2022; 132: 102867.
[70] Li, Meng, Chen, Zhao, Zhang. Gut Microbiota in Lupus: a Butterfly Effect? Curr Rheumatol Rep 2021; 23: 27.
[71] Widhani A, Djauzi S, Suyatna FD, Dewi BE. Changes in Gut Microbiota and Systemic Inflammation after Synbiotic Supplementation in Patients with Systemic Lupus Erythematosus: A Randomized, Double-Blind, Placebo-Controlled Trial. Cells 2022; 11: 3419.
[72] Zhou J, Li M, Chen Q, Li X, Chen L, Dong Z, et al. Programmable probiotics modulate inflammation and gut microbiota for inflammatory bowel disease treatment after effective oral delivery. Nat Commun 2022; 13: 3432.
[73] Roy S, Dhaneshwar S. Role of prebiotics, probiotics, and synbiotics in management of inflammatory bowel disease: Current perspectives. World J Gastroenterol 2023; 29: 2078–100.
[74] Guan Q. A Comprehensive Review and Update on the Pathogenesis of Inflammatory Bowel Disease. Journal of Immunology Research 2019; 2019: 1–16.
[75] Choy MC, Visvanathan K, De Cruz P. An Overview of the Innate and Adaptive Immune System in Inflammatory Bowel Disease: Inflammatory Bowel Diseases 2017; 23: 2–13.
[76] Nagahori M, Nemoto Y, Watanabe M. Pathogenesis of Inflammatory Bowel Diseases. Intest Res 2010; 8: 9.
[77] Nell, Suerbaum, Josenhans. The impact of the microbiota on the pathogenesis of IBD: lessons from mouse infection models. Nat Rev Microbiol 2010; 8: 564–77.
[78] Saleh M, Elson CO. Experimental Inflammatory Bowel Disease: Insights into the Host-Microbiota Dialog. Immunity 2011; 34: 293–302.
[79] Nishida A, Inoue R, Inatomi O, Bamba S, Naito Y, Andoh A. Gut microbiota in the pathogenesis of inflammatory bowel disease. Clin J Gastroenterol 2018; 11: 1–10.
[80] Bai A-P, Ouyang Q. Probiotics and inflammatory bowel diseases. Postgraduate Medical Journal 2006; 82: 376–82.
[81] Chen Y, Jin Y, Stanton C, Ross RP, Wang Z, Zhao J, et al. Dose-response efficacy and mechanisms of orally administered CLA-producing Bifidobacterium breve CCFM683 on DSS-induced colitis in mice. Journal of Functional Foods 2020; 75: 104245.
[82] Komaki S, Haque A, Miyazaki H, Matsumoto T, Nakamura S. Unexpected effect of probiotics by Lactococcus lactis subsp. lactis against colitis induced by dextran sulfate sodium in mice. Journal of Infection and Chemotherapy 2020; 26: 549–53.
[83] Sandes, Figueiredo, Pedroso, Sant’Anna, Acurcio L, Abatemarco Junior, et al. Weissella paramesenteroides WpK4 plays an immunobiotic role in gut-brain axis, reducing gut permeability, anxiety-like and depressive-like behaviors in murine models of colitis and chronic stress. Food Research International 2020; 137: 109741.
[84] Silveira DSC, Veronez LC, Lopes-Júnior LC, Anatriello E, Brunaldi MO, Pereira-da-Silva G. Lactobacillus bulgaricus inhibits colitis-associated cancer via a negative regulation of intestinal inflammation in azoxymethane/dextran sodium sulfate model. WJG 2020; 26: 6782–94.
[85] Xia, Chen, Wang, Yang, Song X, Xiong, et al. Lactobacillus plantarum AR113 alleviates DSS-induced colitis by regulating the TLR4/MyD88/NF-κB pathway and gut microbiota composition. Journal of Functional Foods 2020; 67: 103854.
[86] Li, 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.
[87] Chen Y, Zhang L, Hong G, Huang C, Qian W, Bai T, et al. Probiotic mixtures with aerobic constituent promoted the recovery of multi-barriers in DSS-induced chronic colitis. Life Sciences 2020; 240: 117089.
[88] Liu X, Yu R, Zou K. Probiotic Mixture VSL#3 Alleviates Dextran Sulfate Sodium-induced Colitis in Mice by Downregulating T Follicular Helper Cells. CURR MED SCI 2019; 39: 371–8.
[89] Zhang, Zhao, Zhu, Ma, Ma, Zhang. Probiotic Mixture Protects Dextran Sulfate Sodium-Induced Colitis by Altering Tight Junction Protein Expressions and Increasing Tregs. Mediators of Inflammation 2018; 2018: 1–11.
[90] Pradhan D, Singh R, Tyagi A, H.M. R, Batish VK, Grover S. Assessing the Safety and Efficacy of Lactobacillus plantarum MTCC 5690 and Lactobacillus fermentum MTCC 5689 in Colitis Mouse Model. Probiotics & Antimicro Prot 2019; 11: 910–20.
[91] Selvamani S, Mehta V, Ali El Enshasy H, Thevarajoo S, El Adawi H, Zeini I, et al. Efficacy of Probiotics-Based Interventions as Therapy for Inflammatory Bowel Disease: A Recent Update. Saudi Journal of Biological Sciences 2022; 29: 3546–67.
[92] Fedorak R, Demeria D. Probiotic Bacteria in the Prevention and the Treatment of Inflammatory Bowel Disease. Gastroenterology Clinics of North America 2012; 41: 821–42.
[93] Tamaki H, Nakase H, Inoue S, Kawanami C, Itani T, Ohana M, et al. Efficacy of probiotic treatment with Bifidobacterium longum 536 for induction of remission in active ulcerative colitis: A randomized, double-blinded, placebo-controlled multicenter trial. Digestive Endoscopy 2016; 28: 67–74.
[94] Sun Y-Y, Li M, Li Y-Y, Li L-X, Zhai W-Z, Wang P, et al. The effect of Clostridium butyricum on symptoms and fecal microbiota in diarrhea-dominant irritable bowel syndrome: a randomized, double-blind, placebo-controlled trial. Sci Rep 2018; 8: 2964.
[95] Fedorak, Feagan, Hotte, Leddin, Dieleman, Petrunia DM, et al. The Probiotic VSL#3 Has Anti-inflammatory Effects and Could Reduce Endoscopic Recurrence After Surgery for Crohn’s Disease. Clinical Gastroenterology and Hepatology 2015; 13: 928-935. e2.
[96] Kamarli Altun H, Akal Yildiz E, Department of Nutrition and Dietetics, Eastern Mediterranean University School of Health Sciences, Famagusta, North Cyprus, Akin M, Department of Gastroenterology, Akdeniz University School of Medicine, Antalya, Turkey. Effects of synbiotic therapy in mild-to-moderately active ulcerative colitis: A randomized placebo-controlled study. Turk J Gastroenterol 2019; 30: 313–20.
[97] Pilarczyk Zurek M, Zwolinska Wcisło M, Mach T, Okon K, Adamski P, Heczko PB, et al. Influence of Lactobacillus and Bifidobacterium Combination on the Gut Microbiota, Clinical Course, and Local Gut Inflammation in Patients with Ulcerative Colitis: A Preliminary, Single-center, Open-label Study. J Prob Health 2017; 05.
[98] Palumbo VD, Romeo M, Gammazza AM, Carini F, Damiani P, Damiano G, et al. The long-term effects of probiotics in the therapy of ulcerative colitis: A clinical study. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2016; 160: 372–7.
[99] Barra M, Danino T, Garrido D. Engineered Probiotics for Detection and Treatment of Inflammatory Intestinal Diseases. Front Bioeng Biotechnol 2020; 8: 265.
[100] Veiga, Suez, Derrien, Elinav. Moving from probiotics to precision probiotics. Nat Microbiol 2020; 5: 878–80.
[101] Takiishi T, Korf H, Van Belle TL, Robert S, Grieco FA, Caluwaerts S, et al. Reversal of autoimmune diabetes by restoration of antigen-specific tolerance using genetically modified Lactococcus lactis in mice. J Clin Invest 2012; 122: 1717–25.
[102] Robert S, Gysemans C, Takiishi T, Korf H, Spagnuolo I, Sebastiani G, et al. Oral Delivery of Glutamic Acid Decarboxylase (GAD)-65 and IL10 by Lactococcus lactis Reverses Diabetes in Recent-Onset NOD Mice. Diabetes 2014; 63: 2876–87.
[103] Duan, Liu, March. Engineered Commensal Bacteria Reprogram Intestinal Cells Into Glucose-Responsive Insulin-Secreting Cells for the Treatment of Diabetes. Diabetes 2015; 64: 1794–803.
[104] Riglar DT, Giessen TW, Baym M, Kerns SJ, Niederhuber MJ, Bronson RT, et al. Engineered bacteria can function in the mammalian gut long-term as live diagnostics of inflammation. Nat Biotechnol 2017; 35: 653–8.
[105] McKay R, Hauk P, Quan D, Bentley WE. Development of Cell-Based Sentinels for Nitric Oxide: Ensuring Marker Expression and Unimodality. ACS Synth Biol 2018; 7: 1694–701.
[106] Hanson ML, Hixon JA, Li W, Felber BK, Anver MR, Stewart CA, et al. Oral Delivery of IL-27 Recombinant Bacteria Attenuates Immune Colitis in Mice. Gastroenterology 2014; 146: 210-221. e13.
[107] Liu, Li, Zhang, Xu, Wang, Sun. Oral engineered Bifidobacterium longum expressing rhMnSOD to suppress experimental colitis. International Immunopharmacology 2018; 57: 25–32.
[108] Ciocan D, Elinav E. Engineering bacteria to modulate host metabolism. Acta Physiologica 2023; 238: e14001.
[109] Zmora, Zilberman-Schapira, Suez, Mor, Dori-Bachash, Bashiardes S, et al. Personalized Gut Mucosal Colonization Resistance to Empiric Probiotics Is Associated with Unique Host and Microbiome Features. Cell 2018; 174: 1388-1405. e21.
[110] Cordaillat-Simmons M, Rouanet A, Pot B. Live biotherapeutic products: the importance of a defined regulatory framework. Exp Mol Med 2020; 52: 1397–406.
Cite This Article
  • APA Style

    Kebede, B., Tilahun, A. (2024). Probiotics Application in the Treatment of Autoimmune Diseases and Enhancement of Efficacy Through Genetic Engineering. American Journal of Bioscience and Bioengineering, 12(6), 97-115. https://doi.org/10.11648/j.bio.20241206.12

    Copy | Download

    ACS Style

    Kebede, B.; Tilahun, A. Probiotics Application in the Treatment of Autoimmune Diseases and Enhancement of Efficacy Through Genetic Engineering. Am. J. BioSci. Bioeng. 2024, 12(6), 97-115. doi: 10.11648/j.bio.20241206.12

    Copy | Download

    AMA Style

    Kebede B, Tilahun A. Probiotics Application in the Treatment of Autoimmune Diseases and Enhancement of Efficacy Through Genetic Engineering. Am J BioSci Bioeng. 2024;12(6):97-115. doi: 10.11648/j.bio.20241206.12

    Copy | Download

  • @article{10.11648/j.bio.20241206.12,
      author = {Betemariam Kebede and Adeba Tilahun},
      title = {Probiotics Application in the Treatment of Autoimmune Diseases and Enhancement of Efficacy Through Genetic Engineering
    },
      journal = {American Journal of Bioscience and Bioengineering},
      volume = {12},
      number = {6},
      pages = {97-115},
      doi = {10.11648/j.bio.20241206.12},
      url = {https://doi.org/10.11648/j.bio.20241206.12},
      eprint = {https://article.sciencepublishinggroup.com/pdf/10.11648.j.bio.20241206.12},
      abstract = {The development of multidrug resistance in most pathogenic microorganisms and the rapidly increasing prevalence of non-communicable diseases are becoming major health concerns worldwide. Among non-communicable diseases, autoimmune diseases are caused mainly by imbalances in the gut microbiota (dysbiosis). Gut microbiota colonization and immune system establishment started in the early years of life. A defect in the gut microbiota predominantly affects the proper functioning of immune cells. Hence, restoring gut dysbiosis has received considerable attention for the last few decades as a potential therapeutic option. In this regard, probiotics have been the focus of research during recent decades because of their safe history of use along with fermented foods and beverages. Currently, advanced research is being conducted on the use of probiotics as immunomodulatory mediators and for the amelioration of gut dysbiosis as therapeutic adjuncts in the treatment of autoimmune diseases. In addition, probiotics are genetically engineered to enhance treatment efficacy and to develop live biotherapeutics (LBP). In this review, research articles summarizing findings in autoimmune disease treatment via probiotic strains, emphasizing type 1 diabetes, rheumatoid arthritis, Graves' disease, systemic lupus erythematosus and inflammatory bowel disease in both clinical trials and animal models, were reviewed. Finally, promising results of genetic engineering of probiotics for use as biosensors, delivery of therapeutic proteins, and diagnosis of infections were reported.
    },
     year = {2024}
    }
    

    Copy | Download

  • TY  - JOUR
    T1  - Probiotics Application in the Treatment of Autoimmune Diseases and Enhancement of Efficacy Through Genetic Engineering
    
    AU  - Betemariam Kebede
    AU  - Adeba Tilahun
    Y1  - 2024/11/28
    PY  - 2024
    N1  - https://doi.org/10.11648/j.bio.20241206.12
    DO  - 10.11648/j.bio.20241206.12
    T2  - American Journal of Bioscience and Bioengineering
    JF  - American Journal of Bioscience and Bioengineering
    JO  - American Journal of Bioscience and Bioengineering
    SP  - 97
    EP  - 115
    PB  - Science Publishing Group
    SN  - 2328-5893
    UR  - https://doi.org/10.11648/j.bio.20241206.12
    AB  - The development of multidrug resistance in most pathogenic microorganisms and the rapidly increasing prevalence of non-communicable diseases are becoming major health concerns worldwide. Among non-communicable diseases, autoimmune diseases are caused mainly by imbalances in the gut microbiota (dysbiosis). Gut microbiota colonization and immune system establishment started in the early years of life. A defect in the gut microbiota predominantly affects the proper functioning of immune cells. Hence, restoring gut dysbiosis has received considerable attention for the last few decades as a potential therapeutic option. In this regard, probiotics have been the focus of research during recent decades because of their safe history of use along with fermented foods and beverages. Currently, advanced research is being conducted on the use of probiotics as immunomodulatory mediators and for the amelioration of gut dysbiosis as therapeutic adjuncts in the treatment of autoimmune diseases. In addition, probiotics are genetically engineered to enhance treatment efficacy and to develop live biotherapeutics (LBP). In this review, research articles summarizing findings in autoimmune disease treatment via probiotic strains, emphasizing type 1 diabetes, rheumatoid arthritis, Graves' disease, systemic lupus erythematosus and inflammatory bowel disease in both clinical trials and animal models, were reviewed. Finally, promising results of genetic engineering of probiotics for use as biosensors, delivery of therapeutic proteins, and diagnosis of infections were reported.
    
    VL  - 12
    IS  - 6
    ER  - 

    Copy | Download

Author Information
  • Ethiopian Institute of Agricultural Research, National Agricultural Biotechnology Research Center, National Microbial Biotechnology Research Program, Addis Ababa, Ethiopia

  • Ethiopian Institute of Agricultural Research, National Agricultural Biotechnology Research Center, National Microbial Biotechnology Research Program, Addis Ababa, Ethiopia

  • Sections