1
|
da Silva TF, Glória RDA, Americo MF, Freitas ADS, de Jesus LCL, Barroso FAL, Laguna JG, Coelho-Rocha ND, Tavares LM, le Loir Y, Jan G, Guédon É, Azevedo VADC. Unlocking the Potential of Probiotics: A Comprehensive Review on Research, Production, and Regulation of Probiotics. Probiotics Antimicrob Proteins 2024; 16:1687-1723. [PMID: 38539008 DOI: 10.1007/s12602-024-10247-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/12/2024] [Indexed: 10/02/2024]
Abstract
This review provides a comprehensive overview of the current state of probiotic research, covering a wide range of topics, including strain identification, functional characterization, preclinical and clinical evaluations, mechanisms of action, therapeutic applications, manufacturing considerations, and future directions. The screening process for potential probiotics involves phenotypic and genomic analysis to identify strains with health-promoting properties while excluding those with any factor that could be harmful to the host. In vitro assays for evaluating probiotic traits such as acid tolerance, bile metabolism, adhesion properties, and antimicrobial effects are described. The review highlights promising findings from in vivo studies on probiotic mitigation of inflammatory bowel diseases, chemotherapy-induced mucositis, dysbiosis, obesity, diabetes, and bone health, primarily through immunomodulation and modulation of the local microbiota in human and animal models. Clinical studies demonstrating beneficial modulation of metabolic diseases and human central nervous system function are also presented. Manufacturing processes significantly impact the growth, viability, and properties of probiotics, and the composition of the product matrix and supplementation with prebiotics or other strains can modify their effects. The lack of regulatory oversight raises concerns about the quality, safety, and labeling accuracy of commercial probiotics, particularly for vulnerable populations. Advancements in multi-omics approaches, especially probiogenomics, will provide a deeper understanding of the mechanisms behind probiotic functionality, allowing for personalized and targeted probiotic therapies. However, it is crucial to simultaneously focus on improving manufacturing practices, implementing quality control standards, and establishing regulatory oversight to ensure the safety and efficacy of probiotic products in the face of increasing therapeutic applications.
Collapse
Affiliation(s)
- Tales Fernando da Silva
- Institute of Biological Sciences, Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
- UMR1253, INRAE, L'Institut Agro Rennes Angers, STLO, Rennes, France
| | - Rafael de Assis Glória
- Institute of Biological Sciences, Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Monique Ferrary Americo
- Institute of Biological Sciences, Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Andria Dos Santos Freitas
- Institute of Biological Sciences, Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Luis Claudio Lima de Jesus
- Institute of Biological Sciences, Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Fernanda Alvarenga Lima Barroso
- Institute of Biological Sciences, Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Juliana Guimarães Laguna
- Institute of Biological Sciences, Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Nina Dias Coelho-Rocha
- Institute of Biological Sciences, Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Laisa Macedo Tavares
- Institute of Biological Sciences, Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Yves le Loir
- UMR1253, INRAE, L'Institut Agro Rennes Angers, STLO, Rennes, France
| | - Gwénaël Jan
- UMR1253, INRAE, L'Institut Agro Rennes Angers, STLO, Rennes, France
| | - Éric Guédon
- UMR1253, INRAE, L'Institut Agro Rennes Angers, STLO, Rennes, France
| | - Vasco Ariston de Carvalho Azevedo
- Institute of Biological Sciences, Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil.
| |
Collapse
|
2
|
Xu Y, Du H, Chen Y, Ma C, Zhang Q, Li H, Xie Z, Hong Y. Targeting the gut microbiota to alleviate chemotherapy-induced toxicity in cancer. Crit Rev Microbiol 2024; 50:564-580. [PMID: 37439132 DOI: 10.1080/1040841x.2023.2233605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 05/22/2023] [Accepted: 06/30/2023] [Indexed: 07/14/2023]
Abstract
Despite ongoing breakthroughs in novel anticancer therapies, chemotherapy remains a mainstream therapeutic modality in different types of cancer. Unfortunately, chemotherapy-related toxicity (CRT) often leads to dose limitation, and even results in treatment termination. Over the past few years, accumulating evidence has indicated that the gut microbiota is extensively engaged in various toxicities initiated by chemotherapeutic drugs, either directly or indirectly. The gut microbiota can now be targeted to reduce the toxicity of chemotherapy. In the current review, we summarized the clinical relationship between the gut microbiota and CRT, as well as the critical role of the gut microbiota in the occurrence and development of CRT. We then summarized the key mechanisms by which the gut microbiota modulates CRT. Furthermore, currently available strategies to mitigate CRT by targeting the gut microbiota were summarized and discussed. This review offers a novel perspective for the mitigation of diverse chemotherapy-associated toxic reactions in cancer patients and the future development of innovative drugs or functional supplements to alleviate CRT via targeting the gut microbiota.
Collapse
Affiliation(s)
- Yaning Xu
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, China
| | - Haiyan Du
- Department of Hepatology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Yuchun Chen
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, China
| | - Chong Ma
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, China
| | - Qian Zhang
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, China
| | - Hao Li
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, China
| | - Zhiyong Xie
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, China
| | - Yanjun Hong
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, China
| |
Collapse
|
3
|
Hong Y, Song G, Feng X, Niu J, Wang L, Yang C, Luo X, Zhou S, Ma W. The Probiotic Kluyveromyces lactis JSA 18 Alleviates Obesity and Hyperlipidemia in High-Fat Diet C57BL/6J Mice. Foods 2024; 13:1124. [PMID: 38611428 PMCID: PMC11011337 DOI: 10.3390/foods13071124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 03/30/2024] [Accepted: 04/03/2024] [Indexed: 04/14/2024] Open
Abstract
Obesity poses a significant threat to various health conditions such as heart diseases, diabetes, high blood pressure, and heart attack, with the gut microbiota playing a crucial role in maintaining the body's energy balance. We identified a novel probiotic fungal strain, Kluyveromyces lactis JSA 18 (K. lactis), which was isolated from yak milk and was found to possess anti-obesity properties. Additionally, Lactobacillus plantarum CGMCC 8198 (LP8198) from our previous study was also included to evaluate its anti-obesity properties. The findings indicated that K. lactis caused a notable reduction in weight gain, liver and fat indexes, and hyperlipidemia in mice fed a high-fat diet (HFD). Administering K. lactis and LP8198 to mice on a high-fat diet resulted in a reduction of serum triglyceride levels. Furthermore, the supplements reduced ALT and AST activity, and inhibited the production of inflammatory cytokines such as TNF-α and IL-1β. In addition, lipid metabolism was enhanced by the downregulation of ACC1, PPAR-γ, SREBP-1, and Fasn. Moreover, this study found that K. lactis and LP8198 have little effect on gut bacteria. Additionally, K. lactis partially influenced intestinal fungi, while LP8198 had a minor influence on gut mycobiota. The main goal of this research was to show how effective K. lactis can be as a probiotic in combating obesity.
Collapse
Affiliation(s)
- Yingxiang Hong
- Key Laboratory of Industrial Fermentation Microbiology of the Ministry of Education, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China; (Y.H.); (G.S.); (X.F.); (J.N.); (L.W.); (C.Y.); (X.L.); (W.M.)
| | - Guodong Song
- Key Laboratory of Industrial Fermentation Microbiology of the Ministry of Education, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China; (Y.H.); (G.S.); (X.F.); (J.N.); (L.W.); (C.Y.); (X.L.); (W.M.)
| | - Xiaoqian Feng
- Key Laboratory of Industrial Fermentation Microbiology of the Ministry of Education, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China; (Y.H.); (G.S.); (X.F.); (J.N.); (L.W.); (C.Y.); (X.L.); (W.M.)
| | - Jialei Niu
- Key Laboratory of Industrial Fermentation Microbiology of the Ministry of Education, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China; (Y.H.); (G.S.); (X.F.); (J.N.); (L.W.); (C.Y.); (X.L.); (W.M.)
| | - Lu Wang
- Key Laboratory of Industrial Fermentation Microbiology of the Ministry of Education, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China; (Y.H.); (G.S.); (X.F.); (J.N.); (L.W.); (C.Y.); (X.L.); (W.M.)
| | - Caini Yang
- Key Laboratory of Industrial Fermentation Microbiology of the Ministry of Education, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China; (Y.H.); (G.S.); (X.F.); (J.N.); (L.W.); (C.Y.); (X.L.); (W.M.)
| | - Xuegang Luo
- Key Laboratory of Industrial Fermentation Microbiology of the Ministry of Education, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China; (Y.H.); (G.S.); (X.F.); (J.N.); (L.W.); (C.Y.); (X.L.); (W.M.)
| | - Sa Zhou
- Key Laboratory of Industrial Fermentation Microbiology of the Ministry of Education, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China; (Y.H.); (G.S.); (X.F.); (J.N.); (L.W.); (C.Y.); (X.L.); (W.M.)
| | - Wenjian Ma
- Key Laboratory of Industrial Fermentation Microbiology of the Ministry of Education, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China; (Y.H.); (G.S.); (X.F.); (J.N.); (L.W.); (C.Y.); (X.L.); (W.M.)
- Qilu Institute of Technology, Jinan 250200, China
| |
Collapse
|
4
|
Calazans APCT, Milani TMS, Prata AS, Clerici MTPS, Nicoli JR, Martins FS, Borges MC. A Functional Bread Fermented with Saccharomyces cerevisiae UFMG A-905 Prevents Allergic Asthma in Mice. Curr Dev Nutr 2024; 8:102142. [PMID: 38655128 PMCID: PMC11035053 DOI: 10.1016/j.cdnut.2024.102142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 03/13/2024] [Accepted: 03/18/2024] [Indexed: 04/26/2024] Open
Abstract
Background The administration of probiotics has been shown to be beneficial in asthma. The administration of Saccharomyces cerevisiae UFMG A-905 prevented asthma development. Traditionally, probiotics are administered using dairy-based matrices, but other vehicles (e.g., fruit juices, biscuits, candies, and breads) can be used. Objectives This study aimed to assess the effect of bread fermented with S. cerevisiae UFMG A-905 in asthma prevention. Methods Three breads were produced: fermented with commercial yeast, fermented with S. cerevisiae UFMG A-905, and fermented with S. cerevisiae UFMG A-905 with the addition of alginate microcapsules containing live S. cerevisiae UFMG A-905. Characterization of the microbial composition of the breads was performed. Male Balb/c mice were sensitized and challenged with ovalbumin. Breads were administered 10 d before the first sensitization and during sensitization and challenge protocol. Yeast fecal count, in vivo airway hyperresponsiveness, and airway and lung inflammation were assessed. Results In UFMG A-905 bread, there was an increase in yeast number and a decrease in total and lactic acid bacteria. Animals that received S. cerevisiae UFMG A-905 fermented bread with microcapsules had a significant increase in yeast recovery from feces. S. cerevisiae UFMG A-905-fermented breads partially reduced airway inflammation, decreasing eosinophils and IL5 and IL13 concentrations. When adding microcapsules, the bread also diminished airway hyperresponsiveness and increased IL17A concentrations. Conclusions S. cerevisiae UFMG A-905 was able to generate long-fermentation breads. Microcapsules were a safe and viable way to inoculate the live yeast into food. The administration of breads fermented with S. cerevisiae UFMG A-905 prevented asthma-like characteristics, being more pronounced when the breads contained microcapsules with live yeast.
Collapse
Affiliation(s)
| | | | - Ana Silvia Prata
- Department of Food Engineering and Technology, School of Food Engineering, University of Campinas, Campinas, Brazil
| | | | - Jacques Robert Nicoli
- Department of Microbiology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Flaviano Santos Martins
- Department of Microbiology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Marcos Carvalho Borges
- Department of Internal Medicine, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| |
Collapse
|
5
|
Milani TMS, Sandy CM, Calazans APCT, Silva RQ, Fonseca VMB, Martins FS, Borges MC. Dose-Response Effect of Saccharomyces cerevisiae UFMG A-905 on the Prevention of Asthma in an Animal Model. Probiotics Antimicrob Proteins 2024; 16:53-61. [PMID: 36445686 DOI: 10.1007/s12602-022-10014-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/18/2022] [Indexed: 11/30/2022]
Abstract
Probiotics should be administered in adequate amounts to confer health benefits. Probiotic dose-response studies are still missing. Saccharomyces cerevisiae UFMG A-905 prevented asthma development; however, the ideal dose has not been investigated. We evaluated the optimal dose and administration regimen of S. cerevisiae UFMG A-905 in the prevention of asthma. Male Balb/c mice were sensitized intraperitoneally with ovalbumin (OVA) and challenged with OVA intranasally. Mice received, via gavage, daily or alternate-day S. cerevisiae UFMG A-905. In daily regimen, different concentrations (107, 108, or 109 CFU/mL) were given 10 days before OVA sensitization and during challenges. In alternate-day regimen, a concentration of 109 CFU/mL was administered three times per week for 5 weeks, starting 2 weeks prior to the first sensitization. After the last challenge, in vivo bronchial hyperresponsiveness and airway and lung inflammation were assessed. OVA-challenged mice, when compared to saline-challenged mice, presented a significant increase in bronchial hyperresponsiveness and airway and lung inflammation. Daily and alternate-day administration of 109 CFU/mL of S. cerevisiae UFMG A-905 significantly reduced bronchial hyperresponsiveness; lower concentrations of S. cerevisiae UFMG A-905 did not significantly reduce bronchial hyperresponsiveness. Daily regimen with the highest concentration significantly reduced total cell number, eosinophil count in the BAL, and the levels of IL-4, IL-5, and IL-13. Daily administration of S. cerevisiae UFMG A-905 at 107 and 108 CFU/mL and alternate-day regimen did not significantly decrease airway and lung inflammation. S. cerevisiae UFMG A-905 led to a significant attenuation of bronchial hyperresponsiveness and lung inflammation in a dose-dependent manner.
Collapse
Affiliation(s)
- Thamires M S Milani
- Department of Internal Medicine, Ribeirao Preto Medical School, University of São Paulo, São Paulo, SP, Brazil
| | - Camila M Sandy
- Department of Internal Medicine, Ribeirao Preto Medical School, University of São Paulo, São Paulo, SP, Brazil
| | | | - Rosana Q Silva
- Department of Internal Medicine, Ribeirao Preto Medical School, University of São Paulo, São Paulo, SP, Brazil
| | - Vanessa M B Fonseca
- Department of Internal Medicine, Ribeirao Preto Medical School, University of São Paulo, São Paulo, SP, Brazil
| | - Flaviano S Martins
- Department of Microbiology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, São Paulo, MG, Brazil
| | - Marcos C Borges
- Department of Internal Medicine, Ribeirao Preto Medical School, University of São Paulo, São Paulo, SP, Brazil.
| |
Collapse
|
6
|
López-Gómez L, Alcorta A, Abalo R. Probiotics and Probiotic-like Agents against Chemotherapy-Induced Intestinal Mucositis: A Narrative Review. J Pers Med 2023; 13:1487. [PMID: 37888098 PMCID: PMC10607965 DOI: 10.3390/jpm13101487] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 10/02/2023] [Accepted: 10/09/2023] [Indexed: 10/28/2023] Open
Abstract
Cancer chemotherapy has allowed many patients to survive, but not without risks derived from its adverse effects. Drugs, such as 5-fluorouracil, irinotecan, oxaliplatin, methotrexate, and others, as well as different drug combinations trigger intestinal mucositis that may cause or contribute to anorexia, pain, diarrhea, weight loss, systemic infections, and even death. Dysbiosis is a hallmark of chemotherapy-induced intestinal mucositis and diarrhea, and, therefore, strategies aimed at modulating intestinal microbiota may be useful to counteract and prevent those dreadful effects. This narrative review offers an overview of the studies performed to test the efficacy of probiotics and probiotic-like agents against chemotherapy-induced intestinal mucositis and its consequences. Microbiota modulation through the oral administration of different probiotics (mainly strains of Lactobacillus and Bifidobacterium), probiotic mixtures, synbiotics, postbiotics, and paraprobiotics has been tested in different animal models and in some clinical trials. Regulation of dysbiosis, modulation of epithelial barrier permeability, anti-inflammatory effects, modulation of host immune response, reduction of oxidative stress, or prevention of apoptosis are the main mechanisms involved in their beneficial effects. However, the findings are limited by the great heterogeneity of the preclinical studies and the relative lack of studies in immunocompromised animals, as well as the scarce availability of results from clinical trials. Despite this, the results accumulated so far are promising. Hopefully, with the aid of these agents, intestinal mucositis will be less impactful to the cancer patient in the near future.
Collapse
Affiliation(s)
- Laura López-Gómez
- Department of Basic Health Sciences, Faculty of Health Sciences, University Rey Juan Carlos (URJC), 28922 Alcorcón, Spain; (L.L.-G.); (A.A.)
- High Performance Research Group in Physiopathology and Pharmacology of the Digestive System (NeuGut-URJC), University Rey Juan Carlos (URJC), 28922 Alcorcón, Spain
| | - Alexandra Alcorta
- Department of Basic Health Sciences, Faculty of Health Sciences, University Rey Juan Carlos (URJC), 28922 Alcorcón, Spain; (L.L.-G.); (A.A.)
| | - Raquel Abalo
- Department of Basic Health Sciences, Faculty of Health Sciences, University Rey Juan Carlos (URJC), 28922 Alcorcón, Spain; (L.L.-G.); (A.A.)
- High Performance Research Group in Physiopathology and Pharmacology of the Digestive System (NeuGut-URJC), University Rey Juan Carlos (URJC), 28922 Alcorcón, Spain
- Associated I+D+i Unit to the Institute of Medicinal Chemistry (IQM), Scientific Research Superior Council (CSIC), 28006 Madrid, Spain
- Working Group of Basic Sciences on Pain and Analgesia of the Spanish Pain Society, 28046 Madrid, Spain
- Working Group of Basic Sciences on Cannabinoids of the Spanish Pain Society, 28046 Madrid, Spain
| |
Collapse
|
7
|
Oliveira SRM, Campos LL, Amaral MNS, Galotti B, Ricci MF, Vital KD, Souza RO, Uetanabaro APT, Junqueira MS, Silva AM, Fernandes SOA, Cardoso VN, Nicoli JR, Martins FS. Evaluation of a Functional Craft Wheat Beer Fermented with Saccharomyces cerevisiae UFMG A-905 to treat Salmonella Typhimurium infection in mice. Probiotics Antimicrob Proteins 2023; 15:1180-1192. [PMID: 35907169 DOI: 10.1007/s12602-022-09973-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/25/2022] [Indexed: 11/26/2022]
Abstract
Functional foods containing probiotics are generally administered as dairy products. Non-dairy beverages are another possibility, but probiotic functionality must be confirmed in such vehicles. In the present study, a craft wheat beer brewed with the probiotic yeast Saccharomyces cerevisiae UFMG A-905 (905) was evaluated in a murine model of Salmonella Typhimurium infection. Unfiltered or filtered beer brewed with 905, a commercial wheat beer used as a negative control, or saline were administered orally to mice before and during oral S. Typhimurium challenge. High fecal levels of yeast were only counted in mice treated with the unfiltered 905 beer, which also had reduced mortality and body weight loss due to S. Typhimurium infection. Increased levels of intestinal IgA, translocation to liver and spleen, liver and intestinal lesions, pro-inflammatory cytokines in liver and ileum, and hepatic and intestinal myeloperoxidase and eosinophilic peroxidase activities were observed in animals infected with S. Typhimurium. All these parameters were reduced by the treatment with unfiltered 905 beer. In conclusion, the results show that a craft wheat beer brewed with S. cerevisiae UFMG A-905 maintained the probiotic properties of this yeast when administered orally to mice challenged with S. Typhimurium.
Collapse
Affiliation(s)
- Samantha R M Oliveira
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Avenida Antônio Carlos, Belo Horizonte, MG, 6627, 31270-901, Brazil
- Departamento de Ciências Biológicas, Universidade Estadual de Santa Cruz, Ilhéus, BA, Brazil
| | - Lara L Campos
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Avenida Antônio Carlos, Belo Horizonte, MG, 6627, 31270-901, Brazil
| | - Maisa N S Amaral
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Avenida Antônio Carlos, Belo Horizonte, MG, 6627, 31270-901, Brazil
| | - Bruno Galotti
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Avenida Antônio Carlos, Belo Horizonte, MG, 6627, 31270-901, Brazil
| | - Mayra F Ricci
- Departamento de Patologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Katia D Vital
- Departamento de Análises Clínicas E Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Ramon O Souza
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Avenida Antônio Carlos, Belo Horizonte, MG, 6627, 31270-901, Brazil
| | - Ana Paula T Uetanabaro
- Departamento de Ciências Biológicas, Universidade Estadual de Santa Cruz, Ilhéus, BA, Brazil
| | - Mateus S Junqueira
- Departamento de Engenharia de Alimentos, Universidade Federal de São João del Rei, Sete Lagoas, MG, Brazil
| | - Andreia M Silva
- Departamento de Engenharia de Alimentos, Universidade Federal de São João del Rei, Sete Lagoas, MG, Brazil
| | - Simone O A Fernandes
- Departamento de Análises Clínicas E Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Valbert N Cardoso
- Departamento de Análises Clínicas E Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Jacques R Nicoli
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Avenida Antônio Carlos, Belo Horizonte, MG, 6627, 31270-901, Brazil.
| | - Flaviano S Martins
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Avenida Antônio Carlos, Belo Horizonte, MG, 6627, 31270-901, Brazil
| |
Collapse
|
8
|
Obermüller B, Singer G, Kienesberger B, Mittl B, Stadlbauer V, Horvath A, Miekisch W, Fuchs P, Schweiger M, Pajed L, Till H, Castellani C. Probiotic OMNi-BiOTiC ® 10 AAD Reduces Cyclophosphamide-Induced Inflammation and Adipose Tissue Wasting in Mice. Nutrients 2023; 15:3655. [PMID: 37630845 PMCID: PMC10458463 DOI: 10.3390/nu15163655] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 08/15/2023] [Accepted: 08/16/2023] [Indexed: 08/27/2023] Open
Abstract
Cancer therapy is often associated with severe side effects such as drug induced weight loss, also known as chemotherapy-induced cachexia. The aim of this study was to investigate the effects of a multispecies probiotic (OMNi-BiOTiC® 10 AAD) in a chemotherapy mouse model. A total of 24 male BALB/c mice were gavage-fed with the probiotic formulation or water, once a day for 3 weeks. In the third week, the mice received intraperitoneal cyclophosphamide. At euthanasia, the organs were dissected, and serum was sampled for cytokine analysis. Tight junction components, myosin light chain kinase, mucins, and apoptosis markers were detected in the ileum and colon using histological analyses and qRT-PCR. Lipolysis was analyzed by enzymatic activity assay, Western blotting analyses, and qRT-PCR in WAT. The fecal microbiome was measured with 16S-rRNA gene sequencing from stool samples, and fecal volatile organic compounds analysis was performed using gas chromatography/mass spectrometry. The probiotic-fed mice exhibited significantly less body weight loss and adipose tissue wasting associated with a reduced CGI58 mediated lipolysis. They showed significantly fewer pro-inflammatory cytokines and lower gut permeability compared to animals fed without the probiotic. The colons of the probiotic-fed animals showed lower inflammation scores and less goblet cell loss. qRT-PCR revealed no differences in regards to tight junction components, mucins, or apoptosis markers. No differences in microbiome alpha diversity, but differences in beta diversity, were observed between the treatment groups. Taxonomic analysis showed that the probiotic group had a lower relative abundance of Odoribacter and Ruminococcus-UCG014 and a higher abundance of Desulfovibrio. VOC analysis yielded no significant differences. The results of this study indicate that oral administration of the multispecies probiotic OMNi-BiOTiC® 10 AAD could mitigate cyclophosphamide-induced chemotherapy side effects.
Collapse
Affiliation(s)
- Beate Obermüller
- Department of Paediatric and Adolescent Surgery, Medical University of Graz, 8036 Graz, Austria; (B.O.); (B.K.); (B.M.); (H.T.); (C.C.)
| | - Georg Singer
- Department of Paediatric and Adolescent Surgery, Medical University of Graz, 8036 Graz, Austria; (B.O.); (B.K.); (B.M.); (H.T.); (C.C.)
| | - Bernhard Kienesberger
- Department of Paediatric and Adolescent Surgery, Medical University of Graz, 8036 Graz, Austria; (B.O.); (B.K.); (B.M.); (H.T.); (C.C.)
- Department of Paediatric Surgery, Clinical Center of Klagenfurt, 9020 Klagenfurt, Austria
| | - Barbara Mittl
- Department of Paediatric and Adolescent Surgery, Medical University of Graz, 8036 Graz, Austria; (B.O.); (B.K.); (B.M.); (H.T.); (C.C.)
| | - Vanessa Stadlbauer
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria;
- Center of Biomarker Research (CBmed), 8010 Graz, Austria;
| | - Angela Horvath
- Center of Biomarker Research (CBmed), 8010 Graz, Austria;
| | - Wolfram Miekisch
- Department of Anesthesiology and Intensive Care & Pain Therapy, Rostock University Medical Center, 18057 Rostock, Germany; (W.M.); (P.F.)
| | - Patricia Fuchs
- Department of Anesthesiology and Intensive Care & Pain Therapy, Rostock University Medical Center, 18057 Rostock, Germany; (W.M.); (P.F.)
| | - Martina Schweiger
- Institute of Molecular Biosciences, BioTechMed-Graz, BioHealth-Graz, University of Graz, 8010 Graz, Austria; (M.S.); (L.P.)
| | - Laura Pajed
- Institute of Molecular Biosciences, BioTechMed-Graz, BioHealth-Graz, University of Graz, 8010 Graz, Austria; (M.S.); (L.P.)
| | - Holger Till
- Department of Paediatric and Adolescent Surgery, Medical University of Graz, 8036 Graz, Austria; (B.O.); (B.K.); (B.M.); (H.T.); (C.C.)
| | - Christoph Castellani
- Department of Paediatric and Adolescent Surgery, Medical University of Graz, 8036 Graz, Austria; (B.O.); (B.K.); (B.M.); (H.T.); (C.C.)
- Department of Anesthesiology and Intensive Care Medicine, Weiz District Hospital, 8160 Weiz, Austria
| |
Collapse
|
9
|
Mego M, Danis R, Chovanec J, Jurisova S, Bystricky B, Porsok S, Konkolovsky P, Vaclav V, Wagnerova M, Streško M, Brezinova B, Rečková M, Sutekova D, Pazderova N, Novisedlakova M, Zomborska E, Ciernikova S, Svetlovska D, Drgona L. Randomized double-blind, placebo-controlled multicenter phase III study of prevention of irinotecan-induced diarrhea by a probiotic mixture containing Bifidobacterium BB-12 ®Lactobacillus rhamnosus LGG ® in colorectal cancer patients. Front Oncol 2023; 13:1168654. [PMID: 37601667 PMCID: PMC10438450 DOI: 10.3389/fonc.2023.1168654] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 07/10/2023] [Indexed: 08/22/2023] Open
Abstract
Background The incidence of irinotecan-induced diarrhea varies between 60-90%, by which the incidence of severe diarrhea is 20-40%. The objective of this phase III trial was to determine the effectiveness of the probiotic mixture containing Bifidobacterium, BB-12® and Lactobacillus rhamnosus, LGG® in the prophylaxis of irinotecan-induced diarrhea in metastatic colorectal cancer patients due to a reduction in the activity of intestinal beta-D-glucuronidase. Methods From March 2016 to May 2022, a total of 242 patients with colorectal cancer starting a new line of irinotecan-based therapy were registered to the study in 11 cancer centers in Slovakia. Patients were randomized in a ratio 1:1 to probiotic formula vs. placebo that was administered for 6 weeks. Each capsule of Probio-Tec® BG-Vcap-6.5 contained 2.7x109 colony-forming units (CFU) of 2 lyophilized probiotic strains Bifidobacterium, BB-12® (50%) and Lactobacillus rhamnosus GG, LGG® (50%). Results Administration of probiotics compared to placebo was not associated with a significant reduction of grade 3/4 diarrhea (placebo arm 11.8% vs. probiotic arm 7.9%, p=0.38). Neither the overall incidence of diarrhea (46.2% vs. 41.2%, p=0.51) nor the incidence of enterocolitis (3.4% vs. 0.9%, p=0.37) was different in the placebo vs. probiotic arm. Subgroup analysis revealed that patients with colostomy had higher incidence of any diarrhea and grade 3/4 diarrhea in the placebo arm compared to the probiotic arm (48.5% vs. 22.2%, p=0.06 and 15.2% vs. 0%, p=0.06, respectively). Moreover, patients on probiotic arm had significantly better diarrhea-free survival (HR = 0.41, 95%CI 0.18 - 0.95, p=0.05) and needed less loperamide (p=0.01) compared to patients on placebo arm. We did not observe any infection caused by probiotic strains used in this study. Conclusion This study failed to achieve its primary endpoint, and results suggest a lack of benefit of administered probiotic formula for the prevention of irinotecan-induced diarrhea. However, subgroup analysis suggests a possible benefit in patients with colostomy.
Collapse
Affiliation(s)
- Michal Mego
- Faculty of Medicine, Comenius University and National Cancer Institute, Bratislava, Slovakia
| | - Radoslav Danis
- Faculty of Medicine, Comenius University, Bratislava, Slovakia
| | - Jozef Chovanec
- Department of Oncology, St. Jacob Hospital, Bardejov, Slovakia
| | - Silvia Jurisova
- Faculty of Medicine, Comenius University and National Cancer Institute, Bratislava, Slovakia
| | | | - Stefan Porsok
- Faculty of Medicine, Comenius University and National Cancer Institute, Bratislava, Slovakia
| | | | - Vladimir Vaclav
- Department of Oncology, University Hospital Milosrdni Bratia, Bratislava, Slovakia
| | - Maria Wagnerova
- Department of Oncology, East Slovakia Comprehensive Cancer Center, Kosice, Slovakia
| | - Marian Streško
- Department of Oncology, Faculty Hospital, Trnava, Trebisov, Slovakia
| | | | - Mária Rečková
- Department of Oncology, Regional Cancer Center, Poprad, Slovakia
| | - Dagmar Sutekova
- Department of Oncology, University Hospital Martin, Martin, Slovakia
| | - Natalia Pazderova
- Faculty of Medicine, Comenius University and National Cancer Institute, Bratislava, Slovakia
| | - Mária Novisedlakova
- Faculty of Medicine, Comenius University and National Cancer Institute, Bratislava, Slovakia
- Department of Oncology, University Hospital Milosrdni Bratia, Bratislava, Slovakia
| | - Eva Zomborska
- Faculty of Medicine, Comenius University and National Cancer Institute, Bratislava, Slovakia
| | - Sona Ciernikova
- Biomedical Center, Cancer Research Institute, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Daniela Svetlovska
- Faculty of Medicine, Comenius University and National Cancer Institute, Bratislava, Slovakia
| | - Lubos Drgona
- Faculty of Medicine, Comenius University and National Cancer Institute, Bratislava, Slovakia
| |
Collapse
|
10
|
Muller G, de Godoy VR, Dário MG, Duval EH, Alves-Jr SL, Bücker A, Rosa CA, Dunn B, Sherlock G, Stambuk BU. Improved Sugarcane-Based Fermentation Processes by an Industrial Fuel-Ethanol Yeast Strain. J Fungi (Basel) 2023; 9:803. [PMID: 37623574 PMCID: PMC10456111 DOI: 10.3390/jof9080803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/26/2023] [Accepted: 07/27/2023] [Indexed: 08/26/2023] Open
Abstract
In Brazil, sucrose-rich broths (cane juice and/or molasses) are used to produce billions of liters of both fuel ethanol and cachaça per year using selected Saccharomyces cerevisiae industrial strains. Considering the important role of feedstock (sugar) prices in the overall process economics, to improve sucrose fermentation the genetic characteristics of a group of eight fuel-ethanol and five cachaça industrial yeasts that tend to dominate the fermentors during the production season were determined by array comparative genomic hybridization. The widespread presence of genes encoding invertase at multiple telomeres has been shown to be a common feature of both baker's and distillers' yeast strains, and is postulated to be an adaptation to sucrose-rich broths. Our results show that only two strains (one fuel-ethanol and one cachaça yeast) have amplification of genes encoding invertase, with high specific activity. The other industrial yeast strains had a single locus (SUC2) in their genome, with different patterns of invertase activity. These results indicate that invertase activity probably does not limit sucrose fermentation during fuel-ethanol and cachaça production by these industrial strains. Using this knowledge, we changed the mode of sucrose metabolism of an industrial strain by avoiding extracellular invertase activity, overexpressing the intracellular invertase, and increasing its transport through the AGT1 permease. This approach allowed the direct consumption of the disaccharide by the cells, without releasing glucose or fructose into the medium, and a 11% higher ethanol production from sucrose by the modified industrial yeast, when compared to its parental strain.
Collapse
Affiliation(s)
- Gabriela Muller
- Department of Biochemistry, Federal University of Santa Catarina, Florianópolis, Santa Catarina 88040-900, Brazil; (G.M.); (V.R.d.G.); (M.G.D.); (E.H.D.); (S.L.A.-J.); (A.B.)
| | - Victor R. de Godoy
- Department of Biochemistry, Federal University of Santa Catarina, Florianópolis, Santa Catarina 88040-900, Brazil; (G.M.); (V.R.d.G.); (M.G.D.); (E.H.D.); (S.L.A.-J.); (A.B.)
| | - Marcelo G. Dário
- Department of Biochemistry, Federal University of Santa Catarina, Florianópolis, Santa Catarina 88040-900, Brazil; (G.M.); (V.R.d.G.); (M.G.D.); (E.H.D.); (S.L.A.-J.); (A.B.)
| | - Eduarda H. Duval
- Department of Biochemistry, Federal University of Santa Catarina, Florianópolis, Santa Catarina 88040-900, Brazil; (G.M.); (V.R.d.G.); (M.G.D.); (E.H.D.); (S.L.A.-J.); (A.B.)
| | - Sergio L. Alves-Jr
- Department of Biochemistry, Federal University of Santa Catarina, Florianópolis, Santa Catarina 88040-900, Brazil; (G.M.); (V.R.d.G.); (M.G.D.); (E.H.D.); (S.L.A.-J.); (A.B.)
| | - Augusto Bücker
- Department of Biochemistry, Federal University of Santa Catarina, Florianópolis, Santa Catarina 88040-900, Brazil; (G.M.); (V.R.d.G.); (M.G.D.); (E.H.D.); (S.L.A.-J.); (A.B.)
| | - Carlos A. Rosa
- Departamento de Microbiologia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil;
| | - Barbara Dunn
- Department of Genetics, Stanford University, Stanford, CA 94305, USA; (B.D.); (G.S.)
| | - Gavin Sherlock
- Department of Genetics, Stanford University, Stanford, CA 94305, USA; (B.D.); (G.S.)
| | - Boris U. Stambuk
- Department of Biochemistry, Federal University of Santa Catarina, Florianópolis, Santa Catarina 88040-900, Brazil; (G.M.); (V.R.d.G.); (M.G.D.); (E.H.D.); (S.L.A.-J.); (A.B.)
| |
Collapse
|
11
|
Trindade LM, Torres L, Matos ID, Miranda VC, de Jesus LCL, Cavalcante G, de Souza Oliveira JJ, Cassali GD, Mancha-Agresti P, de Carvalho Azevedo VA, Maioli TU, Cardoso VN, Martins FDS, de Vasconcelos Generoso S. Paraprobiotic Lacticaseibacillus rhamnosus Protects Intestinal Damage in an Experimental Murine Model of Mucositis. Probiotics Antimicrob Proteins 2023; 15:338-350. [PMID: 34524605 DOI: 10.1007/s12602-021-09842-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/27/2021] [Indexed: 12/17/2022]
Abstract
Intestinal mucositis (IM) is a common side effect resulting from cancer treatment. However, the management so far has not been very effective. In the last years, the role of the gut microbiota in the development and severity of mucositis has been studied. Therefore, the use of probiotics and paraprobiotics could have a potential therapeutic effect on IM. The aim of our study was to investigate the impact of the administration of Lacticaseibacillus rhamnosus (L. rhamnosus) CGMCC1.3724 and the paraprobiotic on IM in mice. For 13 days, male Balb/c mice were divided into six groups: control (CTL) and mucositis (MUC)/0.1 mL of saline; CTL LrV and MUC LrV/0.1 mL of 108 CFU of viable Lr; CTL LrI and MUC LrI/0.1 mL of 108 CFU of inactivated Lr. On the 10th day, mice from the MUC, MUC LrV, and MUC LrI groups received an intraperitoneal injection (300 mg/kg) of 5-fluorouracil to induce mucositis. The results showed that the administration of the chemotherapeutic agent increased the weight loss and intestinal permeability of the animals in the MUC and MUC LrV groups. However, administration of paraprobiotic reduced weight loss and maintained PI at physiological levels. The paraprobiotic also preserved the villi and intestinal crypts, reduced the inflammatory infiltrate, and increased the mucus secretion, Muc2 gene expression, and Treg cells frequency.
Collapse
Affiliation(s)
- Luísa Martins Trindade
- Programa de Pós-Graduação Em Ciência de Alimentos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Lícia Torres
- Programa de Pós-Graduação Em Bioquímica E Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Isabel David Matos
- Programa de Pós-Graduação Em Nutrição E Saúde, Departamento de Nutrição, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Vivian Correia Miranda
- Programa de Pós-Graduação Em Microbiologia, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Luís Cláudio Lima de Jesus
- Programa de Pós-Graduação Em Genética, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Gregório Cavalcante
- Programa de Pós-Graduação Em Bioquímica E Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Geovanni Dantas Cassali
- Departamento de Patologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Pamela Mancha-Agresti
- Programa de Pós-Graduação Em Genética, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Vasco Ariston de Carvalho Azevedo
- Programa de Pós-Graduação Em Genética, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Tatiani Uceli Maioli
- Programa de Pós-Graduação Em Nutrição E Saúde, Departamento de Nutrição, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Valbert Nascimento Cardoso
- Departamento de Análises Clínicas E Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Flaviano Dos Santos Martins
- Programa de Pós-Graduação Em Microbiologia, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Simone de Vasconcelos Generoso
- Programa de Pós-Graduação Em Nutrição E Saúde, Departamento de Nutrição, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
| |
Collapse
|
12
|
Lactobacillus gasseri JM1 Isolated from Infant Feces Alleviates Colitis in Mice via Protecting the Intestinal Barrier. Nutrients 2022; 15:nu15010139. [PMID: 36615796 PMCID: PMC9823819 DOI: 10.3390/nu15010139] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/22/2022] [Accepted: 12/24/2022] [Indexed: 12/29/2022] Open
Abstract
Ulcerative colitis (UC) is a chronic and recurrent inflammatory bowel disease, and the intestinal barrier is an important line of defense against intestinal disease. Herein, we investigated the effect of Lactobacillus gasseri JM1 at different doses (1 × 106, 1 × 107, 1 × 108 CFU/day) on colitis mice and explored the possible mechanism. The results showed that L. gasseri JM1 alleviated DSS-induced colitis in mice, with reductions in disease activity index (DAI), histological scores and myeloperoxidase activity as well as alleviation of colonic shortening. Furthermore, L. gasseri JM1 regulated the levels of inflammatory cytokines TNF-α, IL-6, IL-1β, and IL-10; restored the expression of Claudin-3, Occludin, ZO-1, and MUC2; and increased the number of goblet cells and acidic mucin. The 16S rDNA sequencing results indicated that intervention with L. gasseri JM1 balanced the gut microbiota structure by elevating the abundance of beneficial bacteria (Oscillospira, Clostridium and Ruminococcus) and decreasing that of harmful bacteria (Shigella and Turicibacter). Meanwhile, the contents of short-chain fatty acids (SCFAs) increased. In conclusion, L. gasseri JM1 could alleviate intestinal barrier damage in colitis mice by modulating the tight junction structures, intestinal mucus layer, inflammatory cytokines, gut microbiota, and SCFAs. It can be considered a potential preventive strategy to alleviate colitis injury.
Collapse
|
13
|
Huang J, Hwang AYM, Jia Y, Kim B, Iskandar M, Mohammed AI, Cirillo N. Experimental Chemotherapy-Induced Mucositis: A Scoping Review Guiding the Design of Suitable Preclinical Models. Int J Mol Sci 2022; 23:15434. [PMID: 36499758 PMCID: PMC9737148 DOI: 10.3390/ijms232315434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 12/01/2022] [Accepted: 12/04/2022] [Indexed: 12/12/2022] Open
Abstract
Mucositis is a common and most debilitating complication associated with the cytotoxicity of chemotherapy. The condition affects the entire alimentary canal from the mouth to the anus and has a significant clinical and economic impact. Although oral and intestinal mucositis can occur concurrently in the same individual, these conditions are often studied independently using organ-specific models that do not mimic human disease. Hence, the purpose of this scoping review was to provide a comprehensive yet systematic overview of the animal models that are utilised in the study of chemotherapy-induced mucositis. A search of PubMed/MEDLINE and Scopus databases was conducted to identify all relevant studies. Multiple phases of filtering were conducted, including deduplication, title/abstract screening, full-text screening, and data extraction. Studies were reported according to the updated Preferred Reporting Items for Systematic reviews and Meta-Analyses Extension for Scoping Reviews (PRISMA-ScR) guidelines. An inter-rater reliability test was conducted using Cohen's Kappa score. After title, abstract, and full-text screening, 251 articles met the inclusion criteria. Seven articles investigated both chemotherapy-induced intestinal and oral mucositis, 198 articles investigated chemotherapy-induced intestinal mucositis, and 46 studies investigated chemotherapy-induced oral mucositis. Among a total of 205 articles on chemotherapy-induced intestinal mucositis, 103 utilised 5-fluorouracil, 34 irinotecan, 16 platinum-based drugs, 33 methotrexate, and 32 other chemotherapeutic agents. Thirteen articles reported the use of a combination of 5-fluorouracil, irinotecan, platinum-based drugs, or methotrexate to induce intestinal mucositis. Among a total of 53 articles on chemotherapy-induced oral mucositis, 50 utilised 5-fluorouracil, 2 irinotecan, 2 methotrexate, 1 topotecan and 1 with other chemotherapeutic drugs. Three articles used a combination of these drugs to induce oral mucositis. Various animal models such as mice, rats, hamsters, piglets, rabbits, and zebrafish were used. The chemotherapeutic agents were introduced at various dosages via three routes of administration. Animals were mainly mice and rats. Unlike intestinal mucositis, most oral mucositis models combined mechanical or chemical irritation with chemotherapy. In conclusion, this extensive assessment of the literature revealed that there was a large variation among studies that reproduce oral and intestinal mucositis in animals. To assist with the design of a suitable preclinical model of chemotherapy-induced alimentary tract mucositis, animal types, routes of administration, dosages, and types of drugs were reported in this study. Further research is required to define an optimal protocol that improves the translatability of findings to humans.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Nicola Cirillo
- Melbourne Dental School, The University of Melbourne, Carlton, VIC 3053, Australia
| |
Collapse
|
14
|
The Intestinal Redox System and Its Significance in Chemotherapy-Induced Intestinal Mucositis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:7255497. [PMID: 35585883 PMCID: PMC9110227 DOI: 10.1155/2022/7255497] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/04/2022] [Accepted: 04/09/2022] [Indexed: 12/12/2022]
Abstract
Chemotherapy-induced intestinal mucositis (CIM) is a significant dose-limiting adverse reaction brought on by the cancer treatment. Multiple studies reported that reactive oxygen species (ROS) is rapidly produced during the initial stages of chemotherapy, when the drugs elicit direct damage to intestinal mucosal cells, which, in turn, results in necrosis, mitochondrial dysfunction, and ROS production. However, the mechanism behind the intestinal redox system-based induction of intestinal mucosal injury and necrosis of CIM is still undetermined. In this article, we summarized relevant information regarding the intestinal redox system, including the composition and regulation of redox enzymes, ROS generation, and its regulation in the intestine. We innovatively proposed the intestinal redox “Tai Chi” theory and revealed its significance in the pathogenesis of CIM. We also conducted an extensive review of the English language-based literatures involving oxidative stress (OS) and its involvement in the pathological mechanisms of CIM. From the date of inception till July 31, 2021, 51 related articles were selected. Based on our analysis of these articles, only five chemotherapeutic drugs, namely, MTX, 5-FU, cisplatin, CPT-11, and oxaliplatin were shown to trigger the ROS-based pathological mechanisms of CIM. We also discussed the redox system-mediated modulation of CIM pathogenesis via elaboration of the relationship between chemotherapeutic drugs and the redox system. It is our belief that this overview of the intestinal redox system and its role in CIM pathogenesis will greatly enhance research direction and improve CIM management in the future.
Collapse
|
15
|
Savassi B, Cordeiro BF, Silva SH, Oliveira ER, Belo G, Figueiroa AG, Alves Queiroz MI, Faria AMC, Alves J, da Silva TF, Campos GM, Esmerino EA, Rocha RS, Freitas MQ, Silva MC, Cruz AG, Vital KD, Fernandes SO, Cardoso VN, Acurcio LB, Jan G, Le Loir Y, Gala-Garcia A, do Carmo FLR, Azevedo V. Lyophilized Symbiotic Mitigates Mucositis Induced by 5-Fluorouracil. Front Pharmacol 2021; 12:755871. [PMID: 34955828 PMCID: PMC8703075 DOI: 10.3389/fphar.2021.755871] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 11/01/2021] [Indexed: 11/22/2022] Open
Abstract
Mucositis is an adverse effect of cancer chemotherapies using 5-Fluorouracil (5-FU). It is characterized by mucosal inflammation, pain, diarrhea, and weight loss. Some studies reported promising healing effects of probiotic strains, when associated with prebiotics, as adjuvant treatment of mucositis. We developed a lyophilized symbiotic product, containing skimmed milk, supplemented with whey protein isolate (WPI) and with fructooligosaccharides (FOS), and fermented by Lactobacillus casei BL23, Lactiplantibacillus plantarum B7, and Lacticaseibacillus rhamnosus B1. In a mice 5-FU mucositis model, this symbiotic lyophilized formulation was able to reduce weight loss and intestinal permeability. This last was determined in vivo by quantifying blood radioactivity after oral administration of 99mTc-DTPA. Finally, histological damages caused by 5-FU-induced mucositis were monitored. Consumption of the symbiotic formulation caused a reduced score of inflammation in the duodenum, ileum, and colon. In addition, it decreased levels of pro-inflammatory cytokines IL-1β, IL-6, IL-17, and TNF-α in the mice ileum. The symbiotic product developed in this work thus represents a promising adjuvant treatment of mucositis.
Collapse
Affiliation(s)
- Bruna Savassi
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Bárbara F. Cordeiro
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Sara H. Silva
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Emiliano R. Oliveira
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Giovanna Belo
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | | | | | - Ana Maria Caetano Faria
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Juliana Alves
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Tales Fernando da Silva
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Gabriela Munis Campos
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Erick A. Esmerino
- Faculdade de Veterinária, Universidade Federal Fluminense (UFF), Niterói, Brazil
| | - Ramon S. Rocha
- Faculdade de Veterinária, Universidade Federal Fluminense (UFF), Niterói, Brazil
- Departamento de Alimentos, Ciência e Tecnologia Do Rio de Janeiro (IFRJ), Instituto Federal de Educação, Rio de Janeiro, Brazil
| | - Monica Q. Freitas
- Faculdade de Veterinária, Universidade Federal Fluminense (UFF), Niterói, Brazil
| | - Marcia C. Silva
- Departamento de Alimentos, Ciência e Tecnologia Do Rio de Janeiro (IFRJ), Instituto Federal de Educação, Rio de Janeiro, Brazil
| | - Adriano G. Cruz
- Departamento de Alimentos, Ciência e Tecnologia Do Rio de Janeiro (IFRJ), Instituto Federal de Educação, Rio de Janeiro, Brazil
| | - Kátia Duarte Vital
- Departamento de Análises Clínicas e Toxicológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Simone O.A. Fernandes
- Departamento de Análises Clínicas e Toxicológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Valbert N. Cardoso
- Departamento de Análises Clínicas e Toxicológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Leonardo Borges Acurcio
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Gwénaël Jan
- INRAE, STLO, Institut Agro, Agrocampus Ouest, Rennes, France
| | - Yves Le Loir
- INRAE, STLO, Institut Agro, Agrocampus Ouest, Rennes, France
| | - Alfonso Gala-Garcia
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
- Faculdade de Odontologia, Universidade Federal da Bahia (UFBA), Salvador, Brazil
| | - Fillipe Luiz R. do Carmo
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
- INRAE, STLO, Institut Agro, Agrocampus Ouest, Rennes, France
| | - Vasco Azevedo
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| |
Collapse
|
16
|
Yue B, Gao R, Wang Z, Dou W. Microbiota-Host-Irinotecan Axis: A New Insight Toward Irinotecan Chemotherapy. Front Cell Infect Microbiol 2021; 11:710945. [PMID: 34722328 PMCID: PMC8553258 DOI: 10.3389/fcimb.2021.710945] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Accepted: 09/23/2021] [Indexed: 12/19/2022] Open
Abstract
Irinotecan (CPT11) and its active metabolite ethyl-10-hydroxy-camptothecin (SN38) are broad-spectrum cytotoxic anticancer agents. Both cause cell death in rapidly dividing cells (e.g., cancer cells, epithelial cells, hematopoietic cells) and commensal bacteria. Therefore, CPT11 can induce a series of toxic side-effects, of which the most conspicuous is gastrointestinal toxicity (nausea, vomiting, diarrhea). Studies have shown that the gut microbiota modulates the host response to chemotherapeutic drugs. Targeting the gut microbiota influences the efficacy and toxicity of CPT11 chemotherapy through three key mechanisms: microbial ecocline, catalysis of microbial enzymes, and immunoregulation. This review summarizes and explores how the gut microbiota participates in CPT11 metabolism and mediates host immune dynamics to affect the toxicity and efficacy of CPT11 chemotherapy, thus introducing a new concept that is called "microbiota-host-irinotecan axis". Also, we emphasize the utilization of bacterial β-glucuronidase-specific inhibitor, dietary interventions, probiotics and strain-engineered interventions as emergent microbiota-targeting strategies for the purpose of improving CPT11 chemotherapy efficiency and alleviating toxicity.
Collapse
Affiliation(s)
- Bei Yue
- The MOE Key Laboratory of Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines, and the SATCM Key Laboratory of New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine (SHUTCM), Shanghai, China
| | - Ruiyang Gao
- The MOE Key Laboratory of Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines, and the SATCM Key Laboratory of New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine (SHUTCM), Shanghai, China
| | - Zhengtao Wang
- The MOE Key Laboratory of Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines, and the SATCM Key Laboratory of New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine (SHUTCM), Shanghai, China
| | - Wei Dou
- The MOE Key Laboratory of Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines, and the SATCM Key Laboratory of New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine (SHUTCM), Shanghai, China
| |
Collapse
|
17
|
Miknevicius P, Zulpaite R, Leber B, Strupas K, Stiegler P, Schemmer P. The Impact of Probiotics on Intestinal Mucositis during Chemotherapy for Colorectal Cancer: A Comprehensive Review of Animal Studies. Int J Mol Sci 2021; 22:9347. [PMID: 34502251 PMCID: PMC8430988 DOI: 10.3390/ijms22179347] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 07/26/2021] [Accepted: 08/21/2021] [Indexed: 02/07/2023] Open
Abstract
Colorectal cancer (CRC) is the second most commonly diagnosed cancer in females (incidence 16.4/10,000) and the third in males (incidence 23.4/10,000) worldwide. Surgery, chemotherapy (CTx), radiation therapy (RTx), or a combined treatment of those are the current treatment modalities for primary CRC. Chemotherapeutic drug-induced gastrointestinal (GIT) toxicity mainly presents as mucositis and diarrhea. Preclinical studies revealed that probiotic supplementation helps prevent CTx-induced side effects by reducing oxidative stress and proinflammatory cytokine production and promoting crypt cell proliferation. Moreover, probiotics showed significant results in preventing the loss of body weight (BW) and reducing diarrhea. However, further clinical studies are needed to elucidate the exact doses and most promising combination of strains to reduce or prevent chemotherapy-induced side effects. The aim of this review is to overview currently available literature on the impact of probiotics on CTx-induced side effects in animal studies concerning CRC treatment and discuss the potential mechanisms based on experimental studies' outcomes.
Collapse
Affiliation(s)
- Povilas Miknevicius
- General, Visceral and Transplant Surgery, Department of Surgery, Medical University of Graz, Auenbruggerpl. 2, 8036 Graz, Austria; (P.M.); (R.Z.); (B.L.); (P.S.)
- Faculty of Medicine, Vilnius University, 03101 Vilnius, Lithuania;
| | - Ruta Zulpaite
- General, Visceral and Transplant Surgery, Department of Surgery, Medical University of Graz, Auenbruggerpl. 2, 8036 Graz, Austria; (P.M.); (R.Z.); (B.L.); (P.S.)
- Faculty of Medicine, Vilnius University, 03101 Vilnius, Lithuania;
| | - Bettina Leber
- General, Visceral and Transplant Surgery, Department of Surgery, Medical University of Graz, Auenbruggerpl. 2, 8036 Graz, Austria; (P.M.); (R.Z.); (B.L.); (P.S.)
| | - Kestutis Strupas
- Faculty of Medicine, Vilnius University, 03101 Vilnius, Lithuania;
| | - Philipp Stiegler
- General, Visceral and Transplant Surgery, Department of Surgery, Medical University of Graz, Auenbruggerpl. 2, 8036 Graz, Austria; (P.M.); (R.Z.); (B.L.); (P.S.)
| | - Peter Schemmer
- General, Visceral and Transplant Surgery, Department of Surgery, Medical University of Graz, Auenbruggerpl. 2, 8036 Graz, Austria; (P.M.); (R.Z.); (B.L.); (P.S.)
| |
Collapse
|
18
|
Coutinho JOPA, Quintanilha MF, Campos MRA, Ferreira E, de Menezes GCA, Rosa LH, Rosa CA, Vital KD, Fernandes SOA, Cardoso VN, Nicoli JR, Tiago FCP, Martins FS. Antarctic Strain of Rhodotorula mucilaginosa UFMGCB 18,377 Attenuates Mucositis Induced by 5-Fluorouracil in Mice. Probiotics Antimicrob Proteins 2021; 14:486-500. [PMID: 34255281 DOI: 10.1007/s12602-021-09817-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/01/2021] [Indexed: 12/17/2022]
Abstract
Mucositis is one of the most strenuous side effects caused by chemotherapy drugs, such as 5-fluorouracil (5-FU), during the treatment of several types of cancers. The disease is so prevalent and aggressive that many patients cannot resist such symptoms. However, despite its frequency and clinical significance, there is no effective treatment to prevent or treat mucositis. Thus, the use of probiotics as an adjuvant for the treatment has gained prominence. In the present study, we evaluated the effectiveness of oral administration of the Antarctic strain of Rhodotorula mucilaginosa UFMGCB 18,377 as an alternative to minimize side effects of 5-FU-induced mucositis in mice. Body weight, food consumption, stool consistency, and presence of blood in the feces were assessed daily in mice orally treated or not with the yeast and submitted or not to experimental mucositis. Blood, bones, and intestinal tissues and fluid were used to determine intestinal permeability and immunological, microbiological, and histopathological parameters. Treatment with R. mucilaginosa UFMGCB 18,377 was able to decrease clinical signs of the disease, such as reduction of food intake and body weight loss, and also decreased the number of intestinal enterobacteria and intestinal length shortening. Additionally, treatment was able to decrease the levels of MPO and EPO activities and inflammatory infiltrates, as well as the histopathological lesions characteristic of mucositis in the jejunum and ileum. Results of the present study showed that the oral administration of R. mucilaginosa UFMGCB 18,377 protected mice against mucositis induced by 5-FU.
Collapse
Affiliation(s)
- Joana O P A Coutinho
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Mônica F Quintanilha
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Marina R A Campos
- Departamento de Patologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Enio Ferreira
- Departamento de Patologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Graciéle C A de Menezes
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Luiz H Rosa
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Carlos A Rosa
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Katia D Vital
- Departamento de Análises Clínicas E Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Simone O A Fernandes
- Departamento de Análises Clínicas E Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Valbert N Cardoso
- Departamento de Análises Clínicas E Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Jacques R Nicoli
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Fabiana C P Tiago
- Centro Federal de Educação Tecnológica de Minas Gerais (CEFET-MG), Belo Horizonte, MG, Brazil
| | - Flaviano S Martins
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil. .,Laboratório de Agentes Bioterapêuticos, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos 6627, Belo Horizonte, MG, 30270-901, Brazil.
| |
Collapse
|
19
|
Adibi S, Seferovic D, Tribble GD, Alcorn JL, Fakhouri WD. Surfactant Protein A and Microbiome Composition in Patients With Atraumatic Intraoral Lesions. FRONTIERS IN ORAL HEALTH 2021; 2:663483. [PMID: 35048007 PMCID: PMC8757703 DOI: 10.3389/froh.2021.663483] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 03/22/2021] [Indexed: 12/03/2022] Open
Abstract
Oral ulcers are lesions that occur due to disruption of epithelial integrity of the mucosa of the oral cavity. Intraoral ulcers are often associated with pain, redness, symptoms of discomfort, and blood hemorrhage. The etiology for many oral ulcers is local trauma, systemic health conditions, or medication; for other ulcers the cause is less clear. This pilot study aims to evaluate the salivary components and microbiome in patients with atraumatic pre-ulcerous and ulcerous oral lesions compared to control individuals, while considering three common risk factors for atraumatic ulcers, smoking, stress, and gender. This study uses matched age, sex, and ethnicity samples from healthy otherwise and oral lesion patients to investigate the changes in salivary surfactant protein A (SP-A) and examines the prevalence and diversity of the salivary oral microflora. The goal is to determine if there are factors in saliva that have the potential to be used as biomarkers for risk of developing atraumatic oral ulcers. Our data show that the average level of SP-A is significantly reduced in female smokers compared to non-smoker healthy females. The average level of SP-A in female oral lesion patients is reduced compared to controls. The microbiome composition is significantly affected by smoking and the level of SP-A. Comparing the control participants and oral lesion patients, there are 16 species of bacteria that are significantly different, and all of these bacteria are significantly affected by smoking and SP-A. LEfSe analysis identified five bacteria that may represent potential biomarkers. This preliminary study demonstrates the potential of the oral microbiome to act as a biomarker for oral ulcer risk and infers potential mechanistic links between risk factors and alterations in innate immune mechanisms such as SP-A levels.
Collapse
Affiliation(s)
- Shawn Adibi
- Department of General Practice and Dental Public Health, University of Texas Health Science Center at Houston School of Dentistry, Houston, TX, United States
| | - Davor Seferovic
- Department of General Practice and Dental Public Health, University of Texas Health Science Center at Houston School of Dentistry, Houston, TX, United States
| | - Gena D. Tribble
- Department of Periodontics and Dental Hygiene, University of Texas School of Dentistry at Houston, Houston, TX, United States
| | - Joseph L. Alcorn
- Department of Pediatrics, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Walid D. Fakhouri
- Department of Diagnostic and Biomedical Sciences, University of Texas Health Science Center at Houston School of Dentistry, Houston, TX, United States
- Genetics and Epigenetics Program, MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, United States
- *Correspondence: Walid D. Fakhouri
| |
Collapse
|
20
|
Luo Y, Zhou T. Connecting the dots: Targeting the microbiome in drug toxicity. Med Res Rev 2021; 42:83-111. [PMID: 33856076 DOI: 10.1002/med.21805] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/22/2021] [Accepted: 03/31/2021] [Indexed: 12/13/2022]
Abstract
The gut microbiota has a vast influence on human health and its role in initiating, aggravating, or ameliorating diseases is beginning to emerge. Recently, its contribution to heterogeneous toxicological responses is also gaining attention, especially in drug-induced toxicity. Whether they are orally administered or not, drugs may interact with the gut microbiota directly or indirectly, which leads to altered toxicity. Present studies focus more on the unidirectional influence of how xenobiotics disturb intestinal microbial composition and functions, and thus induce altered homeostasis. However, interactions between the gut microbiota and xenobiotics are bidirectional and the impact of the gut microbiota on xenobiotics, especially on drugs, should not be neglected. Thus, in this review, we focus on how the gut microbiota modulates drug toxicity by highlighting the microbiome, microbial enzyme, and microbial metabolites. We connect the dots between drugs, the microbiome, microbial enzymes or metabolites, drug metabolites, and host toxicological responses to facilitate the discovery of microbial targets and mechanisms associated with drug toxicity. Besides this, current mainstream strategies to manipulate drug toxicity by targeting the microbiome are summarized and discussed. The review provides technical reference for the evaluation of medicinal properties in the research and development of innovative drugs, and for the future exploitation of strategies to reduce drug toxicity by targeting the microbiome.
Collapse
Affiliation(s)
- Yusha Luo
- Department of Pharmaceutical Analysis, School of Pharmacy, Second Military Medical University, Shanghai, China.,Shanghai Key Laboratory for Pharmaceutical Metabolite Research, School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Tingting Zhou
- Department of Pharmaceutical Analysis, School of Pharmacy, Second Military Medical University, Shanghai, China.,Shanghai Key Laboratory for Pharmaceutical Metabolite Research, School of Pharmacy, Second Military Medical University, Shanghai, China
| |
Collapse
|
21
|
In Vitro and In Vivo Evaluation of the Probiotic Potential of Antarctic Yeasts. Probiotics Antimicrob Proteins 2021; 13:1338-1354. [PMID: 33759043 DOI: 10.1007/s12602-021-09758-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/19/2021] [Indexed: 02/07/2023]
Abstract
Antarctica is one of the most pristine and inhospitable regions of the planet, mostly inhabited by microorganisms that survive due to unusual metabolic pathways to adapt to its extreme conditions, which could be interesting for the selection of new probiotics. The aim of the present study was to screen in vitro and in vivo putative probiotics among 254 yeasts isolated from different habitats of Antarctica. In vitro selection evaluated functional (growth at 37 °C, resistance to simulated gastric environment, and to bile salts), safety (degradation of mucin, production of β-haemolysis and resistance to antifungal drugs), and beneficial (production of antagonistic substances and adhesion to pathogens) properties. Twelve yeasts were able to grow at 37 °C, one of which was eliminated to present β-haemolytic ability. The remained yeasts resisted to gastric simulation and bile salts, but none presented antagonism against the pathogens tested. Because of the high co-aggregation with Salmonella enterica Typhimurium and growth yield, Rhodotorula mucilaginosa UFMGCB 18377 and Saccharomyces cerevisiae UFMGCB 11120 were selected for in vivo steps using mice challenged with S. Typhimurium. Both yeasts reached high faecal population levels when daily administered, but only R. mucilaginosa UFMGCB 18377 protected mice against Salmonella infection presenting a higher survival and reduced weight loss, bacterial translocation to the liver, sIgA intestinal levels, and intestinal and hepatic MPO and EPO activities. Our in vitro and in vivo results suggest that R. mucilaginosa UFMGCB 18377 presents probiotic potential and deserve further studies as candidate of probiotic by-products. In addition, this is the first screening study of yeasts isolated from Antarctic environments and of Rhodotorula genus for probiotic use.
Collapse
|
22
|
Batista VL, da Silva TF, de Jesus LCL, Coelho-Rocha ND, Barroso FAL, Tavares LM, Azevedo V, Mancha-Agresti P, Drumond MM. Probiotics, Prebiotics, Synbiotics, and Paraprobiotics as a Therapeutic Alternative for Intestinal Mucositis. Front Microbiol 2020; 11:544490. [PMID: 33042054 PMCID: PMC7527409 DOI: 10.3389/fmicb.2020.544490] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Accepted: 08/24/2020] [Indexed: 12/12/2022] Open
Abstract
Intestinal mucositis, a cytotoxic side effect of the antineoplastic drug 5-fluorouracil (5-FU), is characterized by ulceration, inflammation, diarrhea, and intense abdominal pain, making it an important issue for clinical medicine. Given the seriousness of the problem, therapeutic alternatives have been sought as a means to ameliorate, prevent, and treat this condition. Among the alternatives available to address this side effect of treatment with 5-FU, the most promising has been the use of probiotics, prebiotics, synbiotics, and paraprobiotics. This review addresses the administration of these "biotics" as a therapeutic alternative for intestinal mucositis caused by 5-FU. It describes the effects and benefits related to their use as well as their potential for patient care.
Collapse
Affiliation(s)
- Viviane Lima Batista
- Laboratório de Genética Celular e Molecular (LGCM), Departamento de Biologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Tales Fernando da Silva
- Laboratório de Genética Celular e Molecular (LGCM), Departamento de Biologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Luís Cláudio Lima de Jesus
- Laboratório de Genética Celular e Molecular (LGCM), Departamento de Biologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Nina Dias Coelho-Rocha
- Laboratório de Genética Celular e Molecular (LGCM), Departamento de Biologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Fernanda Alvarenga Lima Barroso
- Laboratório de Genética Celular e Molecular (LGCM), Departamento de Biologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Laisa Macedo Tavares
- Laboratório de Genética Celular e Molecular (LGCM), Departamento de Biologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Vasco Azevedo
- Laboratório de Genética Celular e Molecular (LGCM), Departamento de Biologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Pamela Mancha-Agresti
- Laboratório de Genética Celular e Molecular (LGCM), Departamento de Biologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
- Faculdade de Minas, FAMINAS-BH, Belo Horizonte, Brazil
| | - Mariana Martins Drumond
- Laboratório de Genética Celular e Molecular (LGCM), Departamento de Biologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
- Centro Federal de Educação Tecnológica de Minas Gerais (CEFET/MG), Departamento de Ciências Biológicas, Belo Horizonte, Brazil
| |
Collapse
|
23
|
A novel kefir product (PFT) inhibits Ehrlich ascites carcinoma in mice via induction of apoptosis and immunomodulation. BMC Complement Med Ther 2020; 20:127. [PMID: 32345289 PMCID: PMC7189677 DOI: 10.1186/s12906-020-02901-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Accepted: 03/23/2020] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND The popularity of fermented foods such as kefir, kuniss, and tofu has been greatly increasing over the past several decades, and the ability of probiotic bacteria to exert anticancer effects has recently become the focus of research. While we have recently demonstrated the ability of the novel kefir product PFT (Probiotics Fermentation Technology) to exert anticancer effects in vitro, here we demonstrate its ability to inhibit Ehrlich ascites carcinoma (EAC) in mice. METHODS Mice were inoculated intramuscularly with EAC cells to develop solid tumors. PFT was administered orally (2 g/kg/day) to mice 6 days/week, either 2 days before tumor cell inoculation or 9 days after inoculation to mice bearing solid tumors. Tumor growth, blood lymphocyte levels, cell cycle progression, apoptosis, apoptotic regulator expression, TNF-α expression, changes in mitochondrial membrane potential (MMP), PCNA, and CD4+ and CD8+ T cells in tumor cells were quantitatively evaluated by flow cytometry or RT-PCR. Further studies in vitro were carried out where EAC cells along with several other human cancer cell lines were cultured in the presence of PFT (0-5 mg/mL). Percent cell viability and IC50 was estimated by MTT assay. RESULTS Our data shows that PFT exerts the following: 1) inhibition of tumor incidence and tumor growth; 2) inhibition of cellular proliferation via a marked decrease in the expression of tumor marker PCNA; 3) arrest of the tumor cell cycle in the sub-G0/G1 phase, signifying apoptosis; 4) induction of apoptosis in cancer cells via a mitochondrial-dependent pathway as indicated by the up-regulation of p53 expression, increased Bax/Bcl-2 ratio, decrease in the polarization of MMP, and caspase-3 activation; and 5) immunomodulation with an increase in the number of infiltrating CD4+ and CD8+ T cells and an enhancement of TNF-α expression within the tumor. CONCLUSIONS PFT reduces tumor incidence and tumor growth in mice with EAC by inducing apoptosis in EAC cells via the mitochondrial-dependent pathway, suppressing cancer cell proliferation, and stimulating the immune system. PFT may be a useful agent for cancer prevention.
Collapse
|
24
|
Animal models of mucositis: critical tools for advancing pathobiological understanding and identifying therapeutic targets. Curr Opin Support Palliat Care 2020; 13:119-133. [PMID: 30925531 DOI: 10.1097/spc.0000000000000421] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE OF REVIEW Mucositis remains a prevalent, yet poorly managed side effect of anticancer therapies. Mucositis affecting both the oral cavity and gastrointestinal tract predispose to infection and require extensive supportive management, contributing to the growing economic burden associated with cancer care. Animal models remain a critical aspect of mucositis research, providing novel insights into its pathogenesis and revealing therapeutic targets. The current review aims to provide a comprehensive overview of the current animal models used in mucositis research. RECENT FINDINGS A wide variety of animal models of mucositis exist highlighting the highly heterogenous landscape of supportive oncology and the unique cytotoxic mechanisms of different anticancer agents. Golden Syrian hamsters remain the gold-standard species for investigation of oral mucositis induced by single dose and fractionated radiation as well as chemoradiation. There is no universally accepted gold-standard model for the study of gastrointestinal mucositis, with rats, mice, pigs and dogs all offering unique perspectives on its pathobiology. SUMMARY Animal models are a critical aspect of mucositis research, providing unprecedent insight into the pathobiology of mucositis. Introduction of tumour-bearing models, cyclic dosing scheduled, concomitant agents and genetically modified animals have been integral in refining our understanding of mucositis.
Collapse
|
25
|
Hu M, Wu X, Luo M, Wei H, Xu D, Xu F. Lactobacillus rhamnosus FLRH93 protects against intestinal damage in mice induced by 5-fluorouracil. J Dairy Sci 2020; 103:5003-5018. [PMID: 32229117 DOI: 10.3168/jds.2019-17836] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Accepted: 01/07/2020] [Indexed: 12/24/2022]
Abstract
5-Fluorouracil (5-FU) is widely used as a chemotherapeutic drug for the treatment of cancer but it has toxic side effects. It can induce severe intestinal damage and even lead to death. The purpose of this study was to investigate whether milk fermented with Lactobacillus rhamnosus FLRH93 could alleviate intestinal damage induced by 5-FU. The results of injury intervention in a mouse model showed that milk fermented with Lb. rhamnosus FLRH93 significantly ameliorated intestinal injury caused by 5-FU. The results of hematoxylin and eosin staining showed that mice fed Lb. rhamnosus FLRH93 preserved the villus/crypt ratio and reduced the loss of goblet cells in ileum sections of 5-FU-treated animal. Further, administration of fermented milk upregulated expression of Bcl-2 in the intestinal tract and downregulated the expression of NLRP3, thus reducing the production of inflammatory factors interleukin 1-β and tumor necrosis factor-α. The survival rate of mice treated with fermented milk was twice that of mice not fed fermented milk after continuous oral administration of 5-FU. In conclusion, Lb. rhamnosus FLRH93 has positive effects on body injury and could be used to prevent intestinal damage caused by cancer chemotherapy.
Collapse
Affiliation(s)
- Miaomiao Hu
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, 330047, China
| | - Xiaoli Wu
- College of Basic Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, 330004, China
| | - Meng Luo
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, 330047, China
| | - Hua Wei
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, 330047, China; Jiangxi-OAI Joint Research Institute, Nanchang University, 235 Nanjing East Road, Nanchang, 330047, China
| | - Di Xu
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, 330047, China.
| | - Feng Xu
- Jiangxi-OAI Joint Research Institute, Nanchang University, 235 Nanjing East Road, Nanchang, 330047, China.
| |
Collapse
|
26
|
Abstract
Twenty-five years ago, the cytotoxic drug irinotecan (IRT) was first approved in Japan for the treatment of cancer. For more than two decades, the IRT prodrug has largely contributed to the treatment of solid tumors worldwide. Nowadays, this camptothecin derivative targeting topoisomerase 1 remains largely used in combination regimen, like FOLFIRI and FOLFIRINOX, to treat metastatic or advanced solid tumors, such as colon, gastric and pancreatic cancers and others. This review highlights recent discoveries in the field of IRT and its derivatives, including analogues of the active metabolite SN38 (such as FL118), the recently approved liposomal form Nal-IRI and SN38-based immuno-conjugates currently in development (such as sacituzumab govitecan). New information about the IRT mechanism of action are presented, including the discovery of a new protein target, the single-stranded DNA-binding protein FUBP1. Significant progress has been made also to better understand and manage the main limiting toxicities of IRT, chiefly neutropenia and diarrhea. The role of drug-induced inflammation and dysbiosis is underlined and strategies to limit the intestinal toxicity of IRT are discussed (use of β-glucuronidase inhibitors, plant extracts, probiotics). The detailed knowledge of the metabolism of IRT has enabled the identification of potential biomarkers to guide patient selection and to limit drug-induced toxicities, but no robust IRT-specific therapeutic biomarker has been approved yet. IRT is a versatile chemotherapeutic agent which combines well with a variety of anticancer drugs. It offers a large range of drug combinations with cytotoxic agents, targeted products and immuno-active biotherapeutics, to treat a variety of advanced solid carcinoma, sarcoma and cancers with progressive central nervous system diseases. A quarter of century after its first launch, IRT remains an essential anticancer drug, largely prescribed, useful to many patients and scientifically inspiring.
Collapse
|
27
|
Fonseca JF, Alvim LB, Nunes ÁC, Oliveira FMS, Amaral RS, Caliari MV, Nicoli JR, Neumann E, Gomes MA. Probiotic effect of Bifidobacterium longum 5 1A and Weissella paramesenteroides WpK4 on gerbils infected with Giardia lamblia. J Appl Microbiol 2019; 127:1184-1191. [PMID: 31155822 DOI: 10.1111/jam.14338] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 03/13/2019] [Accepted: 05/23/2019] [Indexed: 12/25/2022]
Abstract
AIMS The objective of this study was to assess the probiotic potential of genuine strains of Bifidobacterium longum 51A and Weissella paramesenteroides WpK4, in experimental giardiasis. METHODS AND RESULTS The bacteria were administered orally to gerbils (Meriones unguiculatus) 10 days before oral infection with trophozoites of Giardia lamblia. After 7 days of infection, the animals were euthanized and portions of the duodenum were processed for histopathologic, histochemical and morphometric assessment. The height of the intestinal crypts and crypt/villi ratio were higher in infected groups (P < 0·05) than in noninfected groups. The area of mucus production was higher (P < 0·05) in infected animals pretreated with B. longum 51A than in other groups. The parasitic load of the animals that received both bacteria decreased significantly (P < 0·05) compared to the ones of the control group. CONCLUSIONS Our results suggest a probiotic function of B. longum 51A and W. paramesenteroides WpK4 and may result in their use as a prophylactic and therapeutic alternative for promoting human and animal health. SIGNIFICANCE AND IMPACT OF THE STUDY Bifidobacterium longum 51A and W. paramesenteroides WpK4 may constitute prophylactic alternatives, reversing the emergence of side effects and resistance observed in the conventional treatment of giardiasis.
Collapse
Affiliation(s)
- J F Fonseca
- Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - L B Alvim
- Department of General Biology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Á C Nunes
- Department of General Biology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - F M S Oliveira
- Department of General Pathology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - R S Amaral
- Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - M V Caliari
- Department of General Pathology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - J R Nicoli
- Department of Microbiology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - E Neumann
- Department of Microbiology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - M A Gomes
- Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
28
|
The pathogenesis of mucositis: updated perspectives and emerging targets. Support Care Cancer 2019; 27:4023-4033. [PMID: 31286231 DOI: 10.1007/s00520-019-04893-z] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 05/22/2019] [Indexed: 12/13/2022]
Abstract
Mucositis research and treatment are a rapidly evolving field providing constant new avenues of research and potential therapies. The MASCC/ISOO Mucositis Study Group regularly assesses available literature relating to pathogenesis, mechanisms, and novel therapeutic approaches and distils this to summary perspectives and recommendations. Reviewers assessed 164 articles published between January 2011 and June 2016 to identify progress made since the last review and highlight new targets for further investigation. Findings were organized into sections including established and emerging mediators of toxicity, potential insights from technological advances in mucositis research, and perspective. Research momentum is accelerating for mucositis pathogenesis, and with this has come utilization of new models and interventions that target specific mechanisms of injury. Technological advances have the potential to revolutionize the field of mucositis research, although focused effort is needed to move rationally targeted interventions to the clinical setting.
Collapse
|
29
|
Wang Y, Sun L, Chen S, Guo S, Yue T, Hou Q, Feng M, Xu H, Liu Y, Wang P, Pan Y. The administration of Escherichia coli Nissle 1917 ameliorates irinotecan-induced intestinal barrier dysfunction and gut microbial dysbiosis in mice. Life Sci 2019; 231:116529. [PMID: 31173781 DOI: 10.1016/j.lfs.2019.06.004] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 05/28/2019] [Accepted: 06/02/2019] [Indexed: 02/08/2023]
Abstract
AIMS The present study investigated the effect of Escherichia coli Nissle 1917 (EcN) on irinotecan-induced intestinal barrier dysfunction and gut microbial dysbiosis in a mouse model and in the human colonic cells lines Caco-2. MATERIALS AND METHODS Male BALB/c mice received irinotecan intraperitoneal injection with or without EcN administration intragastrically. Body weight, diarrhea severity, intestinal permeability and histopathological analysis of ileum epithelia of mice from different groups were assessed. The expression and localization of tight junction proteins were examined using western blot and immunofluorescence. Gut microbiota structure and diversity were measured with 16 S rRNA sequencing. Caco-2 monolayers were incubated with EcN culture supernatant (EcNsup) or SN-38 and the monolayer barrier function was assessed by transepithelial electrical resistance (TER) and FITC-dextran 4000 Da (FD-4) flux. KEY FINDINGS Pretreatment with EcN significantly attenuated irinotecan-induced weight loss and diarrhea in mice. In addition, EcN inhibited the increased intestinal permeability and decreased Claudin-1 expression in irinotecan-treated mice. Furthermore, irinotecan treatment decreased the diversity of gut microbiota and increased the relative abundance of Proteobacteria compared to control group. EcN administration ameliorated the gut microbiota dysbiosis. In Caco-2 monolayers, EcNsup ameliorated the decreased TER and increased FD-4 flux elicited by SN-38. Moreover, EcNsup attenuated SN-38-induced altered localization and distribution of Claudin-1 in Caco-2 monolayers. SIGNIFICANCE Our results indicated that the administration of EcN protected against irinotecan-induced intestinal injury by regulating intestinal barrier function and gut microbiota.
Collapse
Affiliation(s)
- Yurong Wang
- Division of General Surgery, Peking University First Hospital, Peking University, 8 Xi ShiKu Street, Beijing 100034, People's Republic of China
| | - Lie Sun
- Division of General Surgery, Peking University First Hospital, Peking University, 8 Xi ShiKu Street, Beijing 100034, People's Republic of China
| | - Shanwen Chen
- Division of General Surgery, Peking University First Hospital, Peking University, 8 Xi ShiKu Street, Beijing 100034, People's Republic of China
| | - Shihao Guo
- Division of General Surgery, Peking University First Hospital, Peking University, 8 Xi ShiKu Street, Beijing 100034, People's Republic of China
| | - Taohua Yue
- Division of General Surgery, Peking University First Hospital, Peking University, 8 Xi ShiKu Street, Beijing 100034, People's Republic of China
| | - Qisheng Hou
- Division of General Surgery, Peking University First Hospital, Peking University, 8 Xi ShiKu Street, Beijing 100034, People's Republic of China
| | - Mei Feng
- Division of General Surgery, Peking University First Hospital, Peking University, 8 Xi ShiKu Street, Beijing 100034, People's Republic of China
| | - Hao Xu
- Division of General Surgery, Peking University First Hospital, Peking University, 8 Xi ShiKu Street, Beijing 100034, People's Republic of China
| | - Yucun Liu
- Division of General Surgery, Peking University First Hospital, Peking University, 8 Xi ShiKu Street, Beijing 100034, People's Republic of China
| | - Pengyuan Wang
- Division of General Surgery, Peking University First Hospital, Peking University, 8 Xi ShiKu Street, Beijing 100034, People's Republic of China
| | - Yisheng Pan
- Division of General Surgery, Peking University First Hospital, Peking University, 8 Xi ShiKu Street, Beijing 100034, People's Republic of China.
| |
Collapse
|
30
|
Treatment with selenium-enriched Saccharomyces cerevisiae UFMG A-905 partially ameliorates mucositis induced by 5-fluorouracil in mice. Cancer Chemother Pharmacol 2019; 84:117-126. [PMID: 31079219 DOI: 10.1007/s00280-019-03865-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 05/04/2019] [Indexed: 02/07/2023]
Abstract
PURPOSE Gastrointestinal mucositis is a major problem associated with cancer therapy. To minimize these deleterious effects, simultaneous administration of antioxidant components, such as selenium, can be considered. There is a growing interest in the use of yeasts because they are able to convert inorganic selenium into selenomethionine. In the present study, oral administration of Saccharomyces cerevisiae UFMG A-905 enriched with selenium was evaluated as an alternative in minimizing the side effects of 5FU-induced mucositis in mice. METHODS Mice body weight, food consumption, faeces consistency and the presence of blood in faeces were assessed daily during experimental mucositis induced by 5-fluorouracil (5FU). Blood was used for intestinal permeability determination, and small intestine for oxidative stress, immunological and histopathological examination. RESULTS The increased intestinal permeability observed with mucositis induction was partially reverted by S. cerevisiae and selenium-enriched yeast. Both treatments were able to reduce myeloperoxidase activity, but only selenium-enriched yeast reduced eosinophil peroxidase activity. CXCL1/KC levels, histopathological tissue damage and oxidative stress (lipid peroxidation and nitrite production) in the small intestine were reduced by both treatments; however, this reduction was always higher when treatment with selenium-enriched yeast was evaluated. CONCLUSIONS Results of the present study showed that the oral administration of S. cerevisiae UFMG A-905 protected mice against mucositis induced by 5-FU, and that this effect was potentiated when the yeast was enriched with selenium.
Collapse
|
31
|
De Jesus LCL, Drumond MM, de Carvalho A, Santos SS, Martins FS, Ferreira Ê, Fernandes RS, de Barros ALB, do Carmo FL, Perez PF, Azevedo V, Mancha-Agresti P. Protective effect of Lactobacillus delbrueckii subsp. Lactis CIDCA 133 in a model of 5 Fluorouracil-Induced intestinal mucositis. J Funct Foods 2019. [DOI: 10.1016/j.jff.2018.12.027] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
|
32
|
Thomsen M, Clarke S, Vitetta L. The role of adjuvant probiotics to attenuate intestinal inflammatory responses due to cancer treatments. Benef Microbes 2018; 9:899-916. [PMID: 30232908 DOI: 10.3920/bm2017.0172] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Chemotherapy and radiotherapy treatment regimens for gastrointestinal, peritoneal and pelvic tumours can disrupt the intestinal microbiome and intestinal epithelia. Such disturbances can provoke symptoms such as diarrhoea, nausea and vomiting. Chemotherapy and radiotherapy induced gastrointestinal toxicity aggravating intestinal microbiome dysbiosis is postulated to adversely alter the intestinal microbiome, with a consequent induced pro-inflammatory effect that disrupts the intestinal microbiome-epithelia-mucosal immunity axis. Although not widely recognised, the intestinal mucosa is the largest and most densely and dynamically populated immune-environment. Cancer treatment adverse effects that affect intestinal and mucosal cells inadvertently target and disrupt resident intestinal macrophages, the cells that marshal immune activity in the intestinal mucosa by shaping pro-inflammatory and anti-inflammatory activities to control and eradicate infectious insults and maintain local homeostasis. Pathobionts (bacteria capable of pathogenic pro-inflammatory activity) and noxious environmental and bacterial antigens use the intestinal epithelia and gap junctions as a point of entry into the systemic circulation. This translocation movement promotes toxic sequelae that obstruct intestinal macrophage functions resulting in uncontrolled local and systemic pro-inflammatory activity, loss of phagocytic function and loss of expression of tight junction proteins. Probiotic bacteria as an adjunctive treatment shows efficacy in ameliorating enteropathies such as mucositis/diarrhoea resulting from chemotherapy or radiotherapy regimens. As such we posit that an important benefit that warrants a further focused research effort is the administration of adjuvant probiotics to help reduce the incidence of febrile neutropenia.
Collapse
Affiliation(s)
- M Thomsen
- 1 The University of Sydney, School of Medicine, Faculty of Medicine and Health, NSW 2006, Australia
| | - S Clarke
- 1 The University of Sydney, School of Medicine, Faculty of Medicine and Health, NSW 2006, Australia.,2 Northern Clinical School, Kolling Institute of Medical Research, Pacific Hwy, St Leonards NSW 2065, Australia
| | - L Vitetta
- 1 The University of Sydney, School of Medicine, Faculty of Medicine and Health, NSW 2006, Australia.,3 Medlab Clinical Ltd., 66 McCauley St., Sydney, 2006 NSW, Australia
| |
Collapse
|
33
|
Cordeiro BF, Oliveira ER, da Silva SH, Savassi BM, Acurcio LB, Lemos L, Alves JDL, Carvalho Assis H, Vieira AT, Faria AMC, Ferreira E, Le Loir Y, Jan G, Goulart LR, Azevedo V, Carvalho RDDO, do Carmo FLR. Whey Protein Isolate-Supplemented Beverage, Fermented by Lactobacillus casei BL23 and Propionibacterium freudenreichii 138, in the Prevention of Mucositis in Mice. Front Microbiol 2018; 9:2035. [PMID: 30258413 PMCID: PMC6143704 DOI: 10.3389/fmicb.2018.02035] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 08/13/2018] [Indexed: 12/12/2022] Open
Abstract
Mucositis is a clinically important gastrointestinal inflammatory infirmity, generated by antineoplastic drugs cytotoxic effects. The inflammatory process caused by this disease frequently leads to derangements in the alimentary tract and great malaise for the patient. Novel strategies are necessary for its prevention or treatment, as currently available treatments of mucositis have several limitations in relieving its symptoms. In this context, several research groups have investigated the use of probiotic bacteria, and in particular dairy bacterial strains. Compelling evidences reveal that milk fermented by certain probiotic bacteria has the capacity to ameliorate intestinal inflammatory disorders. In addition, innovative probiotic delivery strategies, based on probiotics incorporation into protective matrices, such as whey proteins, were able to increase the therapeutic effect of probiotic strains by providing extra protection for bacteria against environmental stresses. Therefore, in this study, we evaluated the role of the whey protein isolate (WPI), when added to skim milk fermented by Lactobacillus casei BL23 (L. casei BL23) or by Propionibacterium freudenreichii CIRM-BIA138 (P. freudenreichii 138), as a protective matrix against in vitro stress challenges. In addition, we investigated the therapeutic effect of these fermented beverages in a murine model of mucositis induced by 5-Fluorouracil (5-FU). Our results demonstrated that milk supplementation with 30% (w/v) of WPI increases the survival rate of both strains when challenged with acid, bile salts, high temperature and cold storage stresses, compared to fermented skim milk without the addition of WPI. Moreover, treatment with the probiotic beverages prevented weight loss and intestinal damages in mice receiving 5-FU. We conclude that the presence of WPI maximizes the anti-inflammatory effects of L. casei BL23, but not for P. freudenreichii 138, suggesting that whey protein enhancement of probiotic activity might be strain-dependent.
Collapse
Affiliation(s)
- Bárbara F. Cordeiro
- Institute of Biological Sciences, Federal University of Minas Gerais (ICB/UFMG), Belo Horizonte, Brazil
| | - Emiliano R. Oliveira
- Institute of Biological Sciences, Federal University of Minas Gerais (ICB/UFMG), Belo Horizonte, Brazil
| | - Sara H. da Silva
- Institute of Biological Sciences, Federal University of Minas Gerais (ICB/UFMG), Belo Horizonte, Brazil
| | - Bruna M. Savassi
- Institute of Biological Sciences, Federal University of Minas Gerais (ICB/UFMG), Belo Horizonte, Brazil
| | - Leonardo B. Acurcio
- Institute of Biological Sciences, Federal University of Minas Gerais (ICB/UFMG), Belo Horizonte, Brazil
| | - Luisa Lemos
- Institute of Biological Sciences, Federal University of Minas Gerais (ICB/UFMG), Belo Horizonte, Brazil
| | - Juliana de L. Alves
- Institute of Biological Sciences, Federal University of Minas Gerais (ICB/UFMG), Belo Horizonte, Brazil
| | - Helder Carvalho Assis
- Institute of Biological Sciences, Federal University of Minas Gerais (ICB/UFMG), Belo Horizonte, Brazil
| | - Angélica T. Vieira
- Institute of Biological Sciences, Federal University of Minas Gerais (ICB/UFMG), Belo Horizonte, Brazil
| | - Ana M. C. Faria
- Institute of Biological Sciences, Federal University of Minas Gerais (ICB/UFMG), Belo Horizonte, Brazil
| | - Enio Ferreira
- Institute of Biological Sciences, Federal University of Minas Gerais (ICB/UFMG), Belo Horizonte, Brazil
| | | | - Gwénaël Jan
- STLO, INRA, Agrocampus Ouest, Rennes, France
| | - Luiz R. Goulart
- Institute of Biotechnology, Federal University of Uberlândia, Uberlândia, Brazil
| | - Vasco Azevedo
- Institute of Biological Sciences, Federal University of Minas Gerais (ICB/UFMG), Belo Horizonte, Brazil
| | - Rodrigo D. de O. Carvalho
- Institute of Biological Sciences, Federal University of Minas Gerais (ICB/UFMG), Belo Horizonte, Brazil
| | - Fillipe L. R. do Carmo
- Institute of Biological Sciences, Federal University of Minas Gerais (ICB/UFMG), Belo Horizonte, Brazil
- STLO, INRA, Agrocampus Ouest, Rennes, France
| |
Collapse
|
34
|
Abstract
PURPOSE OF REVIEW To provide an updated perspective on the use of probiotics as adjuvant treatment strategy for patients suffering from or at risk of developing mucositis. RECENT FINDINGS Studies suggest that oral and intestinal microbiota could be relevant to mucositis development and treatment, but no clear high-risk pattern has been identified and no single probiotic formulation has emerged from human clinical trials for strong recommendation. Promising results from available clinical trials suggest their use in patients with peri-implant mucositis or at risk of anticancer treatment-related oral or intestinal mucositis. In general, a positive effects of Lactobacillus species is becoming consistent, particularly Lactobacillus reuteri, in the treatment of peri-implant mucositis and Lactobacillus brevi CD2 in the prevention of chemoradiotherapy-related oral mucositis. However, several limitations still need to be addressed by future research. Nonetheless, their use appears to be safe. Therefore, decision to consider the use of probiotics ultimately depends on the preference of the clinicians. SUMMARY In the ongoing era of 'precision medicine', efforts should be directed toward the identification of high-risk patient populations which could benefit most from targeted interventions with probiotics enabling an improvement of clinical outcomes and quality of life in a cost-effective manner.
Collapse
Affiliation(s)
- Emanuele Cereda
- Clinical Nutrition and Dietetics Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | | | | |
Collapse
|
35
|
Lemme-Dumit JM, Polti MA, Perdigón G, Galdeano CM. Probiotic bacteria cell walls stimulate the activity of the intestinal epithelial cells and macrophage functionality. Benef Microbes 2017; 9:153-164. [PMID: 29124968 DOI: 10.3920/bm2016.0220] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The effect of oral administration of probiotic bacteria cell walls (PBCWs) in the stimulation of the immune system in healthy BALB/c mice was evaluated. We focused our investigation mainly on intestinal epithelial cells (IECs) which are essential for coordinating an adequate mucosal immune response and on the functionality of macrophages. The probiotic bacteria and their cell walls were able to stimulate the IECs exhibiting an important activation and cytokine releases. Supplementation with PBCWs promoted macrophage activation from peritoneum and spleen, indicating that the PBCWs oral administration was able to improve the functionality of the macrophages. In addition, the PBCWs increased immunoglobulin A (IgA)-producing cells in the gut lamina propria in a similar way to probiotic bacteria, but this supplementation did not have an effect on the population of goblet cells in the small intestine epithelium. These results indicate that the probiotic bacteria and their cell walls have an important immunoregulatory effect on the IECs without altering the homeostatic environment but with an increase in IgA+ producing cells and in the innate immune cells, mainly those distant from the gut such as spleen and peritoneum. These findings about the capacity of the cell walls from probiotic bacteria to stimulate key cells, such as IECs and macrophages, and to improve the functioning of the immune system, suggest that those structures could be applied as a new oral adjuvant.
Collapse
Affiliation(s)
- J M Lemme-Dumit
- 1 Laboratorio de Inmunología, Centro de Referencia para Lactobacilos (CERELA-CONICET). Chacabuco 145, San Miguel de Tucumán 4000, Tucumán, Argentina.,2 Facultad de Bioquímica, Química y Farmacia, Universidad Nacional de Tucumán, Ayacucho 471, San Miguel de Tucumán 4000, Tucumán, Argentina
| | - M A Polti
- 3 Planta Piloto de Procesos Industriales Microbiológicos (PROIMI-CONICET), Av. Belgrano y Pasaje Caseros, San Miguel de Tucumán 4000, Tucumán, Argentina.,4 Facultad de Ciencias Naturales e Instituto Miguel Lillo, Universidad Nacional de Tucumán. Miguel Lillo 205, San Miguel de Tucumán 4000, Tucumán, Argentina
| | - G Perdigón
- 1 Laboratorio de Inmunología, Centro de Referencia para Lactobacilos (CERELA-CONICET). Chacabuco 145, San Miguel de Tucumán 4000, Tucumán, Argentina.,2 Facultad de Bioquímica, Química y Farmacia, Universidad Nacional de Tucumán, Ayacucho 471, San Miguel de Tucumán 4000, Tucumán, Argentina
| | - C Maldonado Galdeano
- 1 Laboratorio de Inmunología, Centro de Referencia para Lactobacilos (CERELA-CONICET). Chacabuco 145, San Miguel de Tucumán 4000, Tucumán, Argentina.,2 Facultad de Bioquímica, Química y Farmacia, Universidad Nacional de Tucumán, Ayacucho 471, San Miguel de Tucumán 4000, Tucumán, Argentina
| |
Collapse
|
36
|
In vivo probiotic and antimicrobial photodynamic therapy as alternative therapies against cryptococcosis are ineffective. Vet Microbiol 2017; 211:169-173. [PMID: 29102114 DOI: 10.1016/j.vetmic.2017.08.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 08/18/2017] [Accepted: 08/23/2017] [Indexed: 12/27/2022]
Abstract
Cryptococcosis, an invasive fungal infection distributed worldwide that affects both domestic and wild animals, has incredible rates regarding treatment failure, leading to the necessity of the development of new therapies. In this way, we aimed to evaluate the probiotic (Saccharomyces boulardii, Lactobacillus paracasei ST-11, and Lactobacillus rhamnosus GG) and antimicrobial photodynamic alternative therapies against Cryptococcus gattii in a murine model. Although previous studies suggest that these therapies can be promising against cryptococcosis, our experimental conditions for both probiotic and antimicrobial photodynamic therapies (aPDT) were not able to improve the survival of mice with cryptococcosis, even with the treatment combined with fluconazole. Our results may help other researchers to find the best protocol to test alternative therapies against Cryptococcus gattii.
Collapse
|
37
|
Kato S, Hamouda N, Kano Y, Oikawa Y, Tanaka Y, Matsumoto K, Amagase K, Shimakawa M. Probiotic Bifidobacterium bifidum
G9-1 attenuates 5-fluorouracil-induced intestinal mucositis in mice via suppression of dysbiosis-related secondary inflammatory responses. Clin Exp Pharmacol Physiol 2017; 44:1017-1025. [DOI: 10.1111/1440-1681.12792] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 05/30/2017] [Accepted: 06/01/2017] [Indexed: 12/31/2022]
Affiliation(s)
- Shinichi Kato
- Division of Pathological Sciences; Department of Pharmacology and Experimental Therapeutics; Kyoto Pharmaceutical University; Kyoto Japan
| | - Nahla Hamouda
- Division of Pathological Sciences; Department of Pharmacology and Experimental Therapeutics; Kyoto Pharmaceutical University; Kyoto Japan
| | - Yoshitaro Kano
- Division of Pathological Sciences; Department of Pharmacology and Experimental Therapeutics; Kyoto Pharmaceutical University; Kyoto Japan
| | - Yousuke Oikawa
- R&D Center; Biofermin Pharmaceutical Co., Ltd.; Kobe Japan
| | - Yoshiki Tanaka
- R&D Center; Biofermin Pharmaceutical Co., Ltd.; Kobe Japan
| | - Kenjiro Matsumoto
- Division of Pathological Sciences; Department of Pharmacology and Experimental Therapeutics; Kyoto Pharmaceutical University; Kyoto Japan
| | - Kikuko Amagase
- Division of Pathological Sciences; Department of Pharmacology and Experimental Therapeutics; Kyoto Pharmaceutical University; Kyoto Japan
| | | |
Collapse
|