1
|
Zhao Y, Dai Z, Lang Y, Li R, Zheng H, Mi J, He X, Liu J, Xiang R, Mei X, Liu Y, Wang Y, Guo H, Yang Q, Ren K, Yang T. Screening of Fecal Bacteroides Strains and Discovery of Bacteroides eggerthii S13-F8 with Protective Effects Against Chemotherapy-Induced Diarrhea. Probiotics Antimicrob Proteins 2025:10.1007/s12602-025-10595-2. [PMID: 40423879 DOI: 10.1007/s12602-025-10595-2] [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] [Accepted: 05/12/2025] [Indexed: 05/28/2025]
Abstract
Chemotherapy-induced diarrhea (CID) is a frequent gastrointestinal side effect in cancer patients, particularly associated with the use of 5-fluorouracil (5-FU). This study aimed to isolate multiple Bacteroides strains from the feces of healthy individuals and identify Bacteroides eggerthii (B. eggerthii) S13-F8 as the optimal candidate for alleviating CID. Whole-genome sequencing of B. eggerthii S13-F8 was conducted to uncover its functional characteristics and explore the potential mechanisms underlying its protective effects against CID. The anti-CID efficacy of B. eggerthii S13-F8 was assessed using multiple parameters, including diarrhea severity, food intake, and body weight changes. Comprehensive analyses, including blood tests, intestinal histopathology, colon transcriptomics, and fecal metagenomics, were performed to elucidate its underlying mechanisms. In a 5-FU-induced mouse model, B. eggerthii S13-F8 significantly alleviated weight loss and diarrhea. Histological examination revealed that B. eggerthii S13-F8 preserved the villus height-to-crypt depth (V/C) ratio and protected goblet cells in colonic tissues. Gene expression analysis showed that B. eggerthii S13-F8 upregulated protective markers, such as Aqp8, Slc26a3, and mucin-related genes (TFF3, FCGBP, and Muc2), while downregulating pro-inflammatory mediators, including IL-1α, IL-22, and Cxcl2. Furthermore, B. eggerthii S13-F8 modulated gut microbiota composition by suppressing pathogenic bacteria (Pseudomonas aeruginosa, Salmonella, γ-Proteobacteria, and Shigella) and enriching beneficial taxa, such as Lactobacillus and Akkermansia muciniphila. In conclusion, B. eggerthii S13-F8 demonstrates significant potential in mitigating severe diarrhea caused by 5-FU chemotherapy, providing a strong foundation for its development as a live biotherapeutic for CID treatment.
Collapse
Affiliation(s)
- Yi Zhao
- School of Pharmacy, Chengdu Medical College, Sichuan Province, Chengdu, China
| | - Zhengqun Dai
- School of Pharmacy, Chengdu Medical College, Sichuan Province, Chengdu, China
| | - Yanmei Lang
- School of Pharmacy, Chengdu Medical College, Sichuan Province, Chengdu, China
| | - Rui Li
- School of Pharmacy, Chengdu Medical College, Sichuan Province, Chengdu, China
| | - Hongyu Zheng
- School of Pharmacy, Chengdu Medical College, Sichuan Province, Chengdu, China
| | - Jiajia Mi
- School of Pharmacy, Chengdu Medical College, Sichuan Province, Chengdu, China
| | - Xiu He
- School of Pharmacy, Chengdu Medical College, Sichuan Province, Chengdu, China
| | - Jin Liu
- School of Pharmacy, Chengdu Medical College, Sichuan Province, Chengdu, China
| | - Rong Xiang
- Precision Medicine Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xueran Mei
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Yang Liu
- Scientific Research Center, Chengdu Medical College, Sichuan Province, Chengdu, China
| | - Yantang Wang
- Scientific Research Center, Chengdu Medical College, Sichuan Province, Chengdu, China
| | - Huijie Guo
- Scientific Research Center, Chengdu Medical College, Sichuan Province, Chengdu, China
| | - Qian Yang
- School of Pharmacy, Chengdu Medical College, Sichuan Province, Chengdu, China
| | - Ke Ren
- School of Laboratory Medicine, Chengdu Medical College, Sichuan Province, Chengdu, China.
| | - Tai Yang
- School of Pharmacy, Chengdu Medical College, Sichuan Province, Chengdu, China.
| |
Collapse
|
2
|
Liu Y, Ning H, Li Y, Li Y, Ma J. The microbiota in breast cancer: dysbiosis, microbial metabolites, and therapeutic implications. Am J Cancer Res 2025; 15:1384-1409. [PMID: 40371158 PMCID: PMC12070087 DOI: 10.62347/zjcf2843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 03/29/2025] [Indexed: 05/16/2025] Open
Abstract
The human microbiome plays a pivotal role in host health and disease, with emerging evidence underscoring its significant influence on the development and progression of breast cancer. Studies have revealed that dysbiosis in both the gut and breast tissue microbiota is strongly associated with an elevated risk of breast cancer. Distinct microbial profiles have been identified among healthy individuals, patients with benign breast conditions, and those with malignant tumors, with further variations observed across different ethnic groups and breast cancer subtypes. The complex interplay between breast cancer risk factors and microbial populations, coupled with the direct impact of microbial communities and their metabolites on inflammatory pathways and immune responses, underscores the importance of this field. Additionally, the interaction between gut microbiota and therapeutic modalities such as chemotherapy and radiotherapy is of particular interest, as these interactions can significantly influence treatment outcomes, either enhancing or diminishing efficacy. This review explores the effects of the Mediterranean diet, probiotics, prebiotics, and natural medicinal products on gut microbiota, emphasizing their potential as innovative therapeutic strategies. Notably, the use of engineered probiotics within the tumor microenvironment represents a promising frontier in breast cancer treatment. In conclusion, research on the human microbiome not only deepens our understanding of breast cancer pathogenesis but also lays the groundwork for the development of novel and targeted therapeutic interventions.
Collapse
Affiliation(s)
- Yan Liu
- Shanxi Province Cancer Hospital, Shanxi Medical UniversityTaiyuan 030006, Shanxi, China
| | - Haiyang Ning
- Shanxi Province Cancer Hospital, Shanxi Medical UniversityTaiyuan 030006, Shanxi, China
| | - Yifei Li
- Shanxi Province Cancer Hospital, Shanxi Medical UniversityTaiyuan 030006, Shanxi, China
| | - Yifan Li
- Shanxi Province Cancer Hospital, Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Cancer Hospital Affiliated to Shanxi Medical UniversityTaiyuan 030001, Shanxi, China
| | - Jinfeng Ma
- Shanxi Province Cancer Hospital, Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Cancer Hospital Affiliated to Shanxi Medical UniversityTaiyuan 030001, Shanxi, China
| |
Collapse
|
3
|
Sørum ME, Boulund U, De Pietri S, Weischendorff S, Enevold C, Rathe M, Als-Nielsen B, Hasle H, Pamp S, Stokholm J, Müller K. Changes in gut microbiota predict neutropenia after induction treatment in childhood acute lymphoblastic leukemia. Blood Adv 2025; 9:1508-1521. [PMID: 39561377 PMCID: PMC11985026 DOI: 10.1182/bloodadvances.2024013986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 09/19/2024] [Accepted: 10/11/2024] [Indexed: 11/21/2024] Open
Abstract
ABSTRACT Delayed neutrophil recovery during acute lymphoblastic leukemia (ALL) treatment increases the risk of infection and causes delay in chemotherapy. Emerging evidence implicates gut microbiota in neutrophil reconstitution after chemotherapy. We explored the interplay between the gut microbiota and neutrophil dynamics, including neutrophil chemoattractants, in 51 children with newly diagnosed ALL. Daily absolute neutrophil count (ANC), weekly plasma chemokines (CXCL1 and CXCL8), granulocyte colony-stimulating factor (G-CSF), and fecal samples were monitored until day 29 during ALL induction treatment. Fecal sequencing using 16S ribosomal RNA revealed an overall significant reduction in bacterial diversity and Enterococcus overgrowth throughout the induction treatment. Prolonged neutropenia (ANC <0.5 × 109 cells per L at day 36) and elevated chemokine levels were associated with a decreased abundance of genera from the Ruminococcaceae and Lachnospiraceae families, decreased Veillonella genus, and Enterococcus overgrowth from diagnosis and throughout induction treatment. G-CSF was upregulated in response to neutropenia but was unrelated to microbiota changes. Overall, this study revealed that a diminished abundance of specific intestinal commensals and Enterococcus overgrowth is associated with delayed neutrophil reconstitution and increased chemokine signaling, indicating that disruption of the microbiota may contribute to prolonged neutropenia. These findings lay the groundwork for future investigations into the mechanisms underlying these associations and their clinical implications for developing gut-sparring strategies to minimize the impact of gut dysbiosis on immune recovery.
Collapse
Affiliation(s)
- Maria Ebbesen Sørum
- Department of Pediatrics and Adolescent Medicine, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Ulrika Boulund
- Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Gentofte, Denmark
| | - Silvia De Pietri
- Department of Pediatrics and Adolescent Medicine, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Sarah Weischendorff
- Department of Pediatrics and Adolescent Medicine, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Christian Enevold
- Institute for Inflammation Research, Center for Rheumatology and Spine Diseases, University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Mathias Rathe
- Department of Pediatrics and Adolescent Medicine, The Hans Christian Andersen Children’s Hospital, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Bodil Als-Nielsen
- Department of Pediatrics and Adolescent Medicine, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Henrik Hasle
- Department of Pediatrics and Adolescent Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Sünje Pamp
- Research Group for Genomic Epidemiology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Jakob Stokholm
- Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Gentofte, Denmark
| | - Klaus Müller
- Department of Pediatrics and Adolescent Medicine, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
4
|
Yin Q, Li X, Xiong Y, Jiang Y, Ma S, Qian G. Bletilla oligosaccharides improved 5-fluorouracil-induced intestinal mucositis in mice by activating NF-κB signalling pathway and regulating intestinal microbiota. Front Pharmacol 2025; 16:1526274. [PMID: 40183076 PMCID: PMC11965902 DOI: 10.3389/fphar.2025.1526274] [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: 11/11/2024] [Accepted: 02/12/2025] [Indexed: 04/05/2025] Open
Abstract
Introduction The Bletilla oligosaccharides (BO) are active compounds extracted from Bletilla striata and have the strong protective effect on the gastrointestinal tract. Chemotherapeutic intestinal mucositis (CIM) is one of the toxic side effects of chemotherapeutic agents on the gastrointestinal tract. The aim of this study was to identify the structure of BO and evaluate the therapeutic effect of BO on 5-fluorouracil-induced intestinal mucosal inflammation. Methods BO were purified from DEAE52 cellulose. The structure of BO were characterised by HPGPC, GC-MS and NMR. In vivo, the mouse model of intestinal mucositis was established by intraperitoneal injection of 5-FU. The effect of BO on intestinal mucositis in mice was detected by assessing the levels of intestinal flora, ZO-1, occludin, and MUC-2, and inflammatory cytokines (IL-1β, IL-6, IL-10, and TNF-α). Results Structural characterisation showed that BO were the neutral polysaccharide composed mainly of glucose and mannose. The backbone of BO consisted of→4)-β-Manp-(1→, →4)-β-Glcp-(1→ and small →3,4)-α-Manp-(1→. The results of the in vivo experiment showed that the symptoms of diarrhoea, haematochezia and colonic mucosal lesions improved after administration of BO. Further experiments showed that BO not only reduced the levels of pro-inflammatory factors such as IL-1β, IL-6 and TNF-α, but also improved the expression of intestinal barrier protein and intestinal microbial community after BO treatment. Conclusion BO can relieve the progress of intestinal mucositis by relieving inflammation, protecting the intestinal epithelial barrier and regulating the intestinal microbiota. These data provide experimental evidence for the application of BO in chemotherapeutic intestinal mucositis.
Collapse
Affiliation(s)
- Qiuxiong Yin
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Xinran Li
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yanli Xiong
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yupeng Jiang
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Shengsuo Ma
- Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Guoqiang Qian
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
5
|
Yang S, Hao S, Ye H, Zhang X. Crosstalk between gut microbiota and cancer chemotherapy: current status and trends. Discov Oncol 2024; 15:833. [PMID: 39715958 DOI: 10.1007/s12672-024-01704-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 12/13/2024] [Indexed: 12/25/2024] Open
Abstract
BACKGROUND Chemotherapy is crucial in the management of tumors, but challenges such as chemoresistance and adverse reactions frequently lead to therapeutic delays or even premature cessation. A growing body of research underscores a profound connection between the gut microbiota (GM) and cancer chemotherapy (CC). This paper aims to pinpoint highly influential publications and monitor the current landscape and evolving trends within the realm of GM/CC research. METHODS On October 1st, 2024, a comprehensive search for GM/CC publications spanning the past 20 years from 2004 to 2023 was conducted utilizing the Web of Science Core Collection (WoSCC). The scope encompassed both articles and reviews, and the data was subsequently extracted. To gain insights into the evolution and dynamics of this research field, we employed bibliometric analysis tools such as the Bibliometrix R package, VOSviewer, and Microsoft Excel to visualize and analyze various dimensions, including prominent journals, leading authors, esteemed institutions, contributing countries/regions, highly cited papers, and frequently occurring keywords. RESULTS A total of 888 papers were obtained. The number of publications about GM/CC studies has increased gradually. China and the United States published the largest number of papers. The INSERM was in the leading position in publishers. The most productive authors were Zitvogel L from France. Cancers had the largest number of papers. Citation analysis explained the historical evolution and breakthroughs in GM/CC research. Highly cited papers and common keywords illustrated the status and trends of GM/CC research. Four clusters were identified, and the hot topics included the role of the GM in the efficacy and toxicity of CC, the targeting of the GM to improve the outcome of CC, the mechanism by which the GM affects CC, and the correlation of the GM with carcinogenesis and cancer therapy. Metabolism, GM-derived metabolites, tumor microenvironment, immunity, intestinal barrier, tumor microbiota and Fusobacterium nucleatum may become the new hotspots and trends of GM/CC research. CONCLUSION This study analyzed global publications and bibliometric characteristics of the links between GM and CC, identified highly cited papers in GM/CC, provided insight into the status, hotspots, and trends of global GM/CC research, and showed that the GM can be used to predict the efficacy and toxicity of CC and modifying the GM can improve the outcomes of chemotherapeutics, which may inform clinical researchers of future directions.
Collapse
Affiliation(s)
- Shanshan Yang
- Department of Traditional Chinese Medicine, Peking University First Hospital, Beijing, China
| | - Shaodong Hao
- Spleen-Stomach Department, Fangshan Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Hui Ye
- Department of Traditional Chinese Medicine, Peking University First Hospital, Beijing, China.
| | - Xuezhi Zhang
- Department of Traditional Chinese Medicine, Peking University First Hospital, Beijing, China.
| |
Collapse
|
6
|
Sun M, Tang D, Jia J, Wu Y, Yu C, Qiu R, Wang H, Tao S. The role of the gut microbiota in infectious complications during immunochemotherapy for diffuse large B-cell lymphoma. BMC Cancer 2024; 24:1570. [PMID: 39716091 DOI: 10.1186/s12885-024-13344-w] [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: 04/04/2024] [Accepted: 12/14/2024] [Indexed: 12/25/2024] Open
Abstract
BACKGROUND Infections are common complications and causes of death during immunochemotherapy in diffuse large B-cell lymphoma (DLBCL). The gut microbiota plays a significant role in bacterial infection, but its relationship and predictive capacity with infectious complications in DLBCL are unknown. METHODS We performed 16S rRNA gene sequencing of fecal samples collected from 41 patients with newly diagnosed DLBCL at baseline, after every two cycles of standard immunochemotherapy, during infection, and after infection recovery. Analysis of the diversity and species composition of these samples was used to evaluate the relationship between gut microbiota and bacterial infection. RESULTS Our findings demonstrate the dynamic changes of Enterobacteriaceae in patients with DLBCL during immunochemotherapy. The abundance of Enterobacteriaceae was markedly higher at baseline in patients who subsequently developed bacterial infection during immunochemotherapy than in those who did not (P < 0.0001), and showed a further increase during infection (P < 0.01), after recovery from the infection, the Enterobacteriaceae was significantly decreased (P < 0.001). While there was no significant change in patients who did not develop bacterial infection. The univariate and multivariate analysis showed that baseline abundance of Enterobacteriaceae > 4.5% was independently associated with post-immunochemotherapy bacterial infection. CONCLUSIONS Our findings suggest that the gut microbiota signatures differ between patients with DLBCL who do and do not develop bacterial infection. The baseline abundance of Enterobacteriaceae is associated with the post-immunochemotherapy bacterial infection, and it has certain predictive value. Detecting the changes of gut microbiota can help predict the risk of bacterial infection after immunochemotherapy.
Collapse
Affiliation(s)
- Man Sun
- Department of Oncology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1 Min-De Road, Nanchang, Jiangxi, 330006, China
| | - Duozhuang Tang
- Department of Hematology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1 Min-De Road, Nanchang, Jiangxi, 330006, China
- Jiangxi Provincial Key Laboratory of Hematological Diseases(2024SSY06052),Department of Hematology , The Second Affiliated Hospital of Nanchang University, 1 Min-De Road, Nanchang, Jiangxi, 330006, China
| | - Jie Jia
- Department of Oncology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1 Min-De Road, Nanchang, Jiangxi, 330006, China
| | - Yuanyuan Wu
- Department of Oncology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1 Min-De Road, Nanchang, Jiangxi, 330006, China
| | - Chenghui Yu
- Department of Hematology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1 Min-De Road, Nanchang, Jiangxi, 330006, China
- Jiangxi Provincial Key Laboratory of Hematological Diseases(2024SSY06052),Department of Hematology , The Second Affiliated Hospital of Nanchang University, 1 Min-De Road, Nanchang, Jiangxi, 330006, China
| | - Rongrong Qiu
- Department of Oncology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1 Min-De Road, Nanchang, Jiangxi, 330006, China
| | - Hua Wang
- Department of Oncology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1 Min-De Road, Nanchang, Jiangxi, 330006, China.
| | - Si Tao
- Department of Oncology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1 Min-De Road, Nanchang, Jiangxi, 330006, China.
| |
Collapse
|
7
|
Qin D, Fu W, Sun Y, Zhao L, Liu H, Fan D, Tan D, Ji X, Wang S. Protective Effects of Cereal-Based Fermented Beverages Against 5-Fluorouracil-Induced Intestinal Damage in Mice. Nutrients 2024; 16:4332. [PMID: 39770954 PMCID: PMC11679319 DOI: 10.3390/nu16244332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 11/27/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND 5-Fluorouracil (5-FU) is a common chemotherapeutic medication used to treat cancer. However, the intestinal tract may sustain oxidative damage as a result. OBJECTIVES The purpose of this study was to clarify the underlying molecular mechanisms and examine the preventive benefits of cereal-based fermented drinks (CFBs) against intestinal injury in mice caused by 5-FU. METHODS The mice were injected intraperitoneally with 5-FU to induce intestinal mucosal and treated with CFB. The factors for intestinal barrier integrity, oxidative stress and inflammation were measured. RESULTS The findings demonstrated that CFBs had high levels of polyphenol, flavonoids, and peptides and had in vitro high free radical scavenging capacity. Furthermore, CFBs effectively ameliorated 5-FU-induced intestinal epithelium damage, characterized by increasing intestinal tight junctions and reducing apoptosis in intestinal cells. These protective effects may attribute to the increased activity of antioxidant-related enzymes (SOD, CAT, and GSH) as well as decreased amounts of inflammatory and oxidative damage markers (IL-1β, TNF-α, and MDA) in the intestinal tract. CONCLUSIONS Overall, these results show that CFBs can mitigate intestinal damage caused by 5-FU by reducing oxidative stress, suggesting the potential utility of CFBs for therapeutic treatment against intestinal mucositis.
Collapse
Affiliation(s)
- Dongze Qin
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300071, China; (D.Q.); (W.F.); (Y.S.); (L.Z.); (H.L.); (D.F.); (X.J.)
| | - Wenhui Fu
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300071, China; (D.Q.); (W.F.); (Y.S.); (L.Z.); (H.L.); (D.F.); (X.J.)
| | - Yi Sun
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300071, China; (D.Q.); (W.F.); (Y.S.); (L.Z.); (H.L.); (D.F.); (X.J.)
| | - Lingda Zhao
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300071, China; (D.Q.); (W.F.); (Y.S.); (L.Z.); (H.L.); (D.F.); (X.J.)
| | - Haiwei Liu
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300071, China; (D.Q.); (W.F.); (Y.S.); (L.Z.); (H.L.); (D.F.); (X.J.)
| | - Dancai Fan
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300071, China; (D.Q.); (W.F.); (Y.S.); (L.Z.); (H.L.); (D.F.); (X.J.)
| | - Dongfei Tan
- Institute of Agro-Product Safety and Nutrition, Tianjin Academy of Agricultural Sciences (TAAS), Tianjin 300192, China;
| | - Xuemeng Ji
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300071, China; (D.Q.); (W.F.); (Y.S.); (L.Z.); (H.L.); (D.F.); (X.J.)
| | - Shuo Wang
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300071, China; (D.Q.); (W.F.); (Y.S.); (L.Z.); (H.L.); (D.F.); (X.J.)
| |
Collapse
|
8
|
Lima FRF, Monteiro CEDS, Damasceno SRB, Pantoja PDS, Franco ÁX, Silva RO, Sousa JAO, Mendes TS, Lima MA, Justino PFC, de Souza MHLP, Soares PMG. Bacillus clausii Attenuates 5-Fluorouracil-Induced Intestinal Mucositis in Mice. Pharmaceuticals (Basel) 2024; 17:1676. [PMID: 39770518 PMCID: PMC11678352 DOI: 10.3390/ph17121676] [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: 04/30/2024] [Revised: 08/06/2024] [Accepted: 08/12/2024] [Indexed: 01/11/2025] Open
Abstract
5-Fluorouracil (5-FU) is an antimetabolite widely prescribed in cancer treatments, but its use in highly proliferative tissues can cause significant problems such as mucositis. Bacillus clausii is a probiotic commonly used for protection against acute diarrhea, gastrointestinal dysbiosis and inflammatory bowel diseases. We investigated the effect of B. clausii on 5-FU intestinal mucositis in mice. B. clausii was administered concomitantly with 5-FU on the first day and alone for the other two days. After the third day of 5-FU (450 mg/kg, ip), the animals were euthanized. Ileum samples were removed for evaluation for histopathological and biochemical analyses (myeloperoxidase (MPO), glutathione (GSH), malondialdehyde (MDA), catalase (CAT), interleukin-1 beta (IL-1β) and tumor necrosis factor alpha (TNF-α). In addition, we investigated gastric emptying and intestinal transit, intestinal permeability, intestinal smooth muscle contractility, transepithelial electrical resistance and intestinal transport of water and electrolytes. B. clausii reduced histopathological scores and increased the villus/crypt ratio in all intestinal segments after mucositis. B. clausii attenuated 5-FU-induced weight loss. The probiotic treatment increased GSH levels, reduced MPO and CAT activity, and also reduced MDA, IL-1β and TNF-α levels. B. clausii attenuated the delay in gastric emptying, water and electrolyte secretion and intestinal hypercontractility, and increased 5-FU-induced intestinal permeability. Thus, our data suggest that B. clausii may be a potential candidate for the treatment of chemotherapy-induced intestinal mucositis.
Collapse
Affiliation(s)
- Francisco Ronaldo Farias Lima
- Laboratory of Gastrointestinal Physio-Pharmacology (LEFFAG), Federal University of Ceará, Coronel Nunes de Melo Street, 1315 Rodolfo Teófilo, Fortaleza 60416-030, CE, Brazil; (F.R.F.L.); (C.E.d.S.M.); (S.R.B.D.); (P.d.S.P.); (Á.X.F.); (J.A.O.S.); (T.S.M.); (M.A.L.); (P.F.C.J.); (M.H.L.P.d.S.)
| | - Carlos Eduardo da Silva Monteiro
- Laboratory of Gastrointestinal Physio-Pharmacology (LEFFAG), Federal University of Ceará, Coronel Nunes de Melo Street, 1315 Rodolfo Teófilo, Fortaleza 60416-030, CE, Brazil; (F.R.F.L.); (C.E.d.S.M.); (S.R.B.D.); (P.d.S.P.); (Á.X.F.); (J.A.O.S.); (T.S.M.); (M.A.L.); (P.F.C.J.); (M.H.L.P.d.S.)
| | - Samara Rodrigues Bonfim Damasceno
- Laboratory of Gastrointestinal Physio-Pharmacology (LEFFAG), Federal University of Ceará, Coronel Nunes de Melo Street, 1315 Rodolfo Teófilo, Fortaleza 60416-030, CE, Brazil; (F.R.F.L.); (C.E.d.S.M.); (S.R.B.D.); (P.d.S.P.); (Á.X.F.); (J.A.O.S.); (T.S.M.); (M.A.L.); (P.F.C.J.); (M.H.L.P.d.S.)
| | - Patricia da Silva Pantoja
- Laboratory of Gastrointestinal Physio-Pharmacology (LEFFAG), Federal University of Ceará, Coronel Nunes de Melo Street, 1315 Rodolfo Teófilo, Fortaleza 60416-030, CE, Brazil; (F.R.F.L.); (C.E.d.S.M.); (S.R.B.D.); (P.d.S.P.); (Á.X.F.); (J.A.O.S.); (T.S.M.); (M.A.L.); (P.F.C.J.); (M.H.L.P.d.S.)
| | - Álvaro Xavier Franco
- Laboratory of Gastrointestinal Physio-Pharmacology (LEFFAG), Federal University of Ceará, Coronel Nunes de Melo Street, 1315 Rodolfo Teófilo, Fortaleza 60416-030, CE, Brazil; (F.R.F.L.); (C.E.d.S.M.); (S.R.B.D.); (P.d.S.P.); (Á.X.F.); (J.A.O.S.); (T.S.M.); (M.A.L.); (P.F.C.J.); (M.H.L.P.d.S.)
| | - Renan Oliveira Silva
- Department of Physiology and Pharmacology, Federal University of Pernambuco, Av. da Engenharia-Cidade Universitária, Recife 50670-420, PE, Brazil;
| | - Johnatan Alisson Oliveira Sousa
- Laboratory of Gastrointestinal Physio-Pharmacology (LEFFAG), Federal University of Ceará, Coronel Nunes de Melo Street, 1315 Rodolfo Teófilo, Fortaleza 60416-030, CE, Brazil; (F.R.F.L.); (C.E.d.S.M.); (S.R.B.D.); (P.d.S.P.); (Á.X.F.); (J.A.O.S.); (T.S.M.); (M.A.L.); (P.F.C.J.); (M.H.L.P.d.S.)
| | - Tiago Santos Mendes
- Laboratory of Gastrointestinal Physio-Pharmacology (LEFFAG), Federal University of Ceará, Coronel Nunes de Melo Street, 1315 Rodolfo Teófilo, Fortaleza 60416-030, CE, Brazil; (F.R.F.L.); (C.E.d.S.M.); (S.R.B.D.); (P.d.S.P.); (Á.X.F.); (J.A.O.S.); (T.S.M.); (M.A.L.); (P.F.C.J.); (M.H.L.P.d.S.)
| | - Marcos Aurélio Lima
- Laboratory of Gastrointestinal Physio-Pharmacology (LEFFAG), Federal University of Ceará, Coronel Nunes de Melo Street, 1315 Rodolfo Teófilo, Fortaleza 60416-030, CE, Brazil; (F.R.F.L.); (C.E.d.S.M.); (S.R.B.D.); (P.d.S.P.); (Á.X.F.); (J.A.O.S.); (T.S.M.); (M.A.L.); (P.F.C.J.); (M.H.L.P.d.S.)
- Instituto Federal de Educação, Ciência e Tecnologia do Ceará-Campus Acopiara, Rodovia CE 060, Km 332, Vila Martins, Acopiara 63560-000, CE, Brazil
| | - Priscilla Fernanda Campos Justino
- Laboratory of Gastrointestinal Physio-Pharmacology (LEFFAG), Federal University of Ceará, Coronel Nunes de Melo Street, 1315 Rodolfo Teófilo, Fortaleza 60416-030, CE, Brazil; (F.R.F.L.); (C.E.d.S.M.); (S.R.B.D.); (P.d.S.P.); (Á.X.F.); (J.A.O.S.); (T.S.M.); (M.A.L.); (P.F.C.J.); (M.H.L.P.d.S.)
| | - Marcellus Henrique Loiola Ponte de Souza
- Laboratory of Gastrointestinal Physio-Pharmacology (LEFFAG), Federal University of Ceará, Coronel Nunes de Melo Street, 1315 Rodolfo Teófilo, Fortaleza 60416-030, CE, Brazil; (F.R.F.L.); (C.E.d.S.M.); (S.R.B.D.); (P.d.S.P.); (Á.X.F.); (J.A.O.S.); (T.S.M.); (M.A.L.); (P.F.C.J.); (M.H.L.P.d.S.)
| | - Pedro Marcos Gomes Soares
- Laboratory of Gastrointestinal Physio-Pharmacology (LEFFAG), Federal University of Ceará, Coronel Nunes de Melo Street, 1315 Rodolfo Teófilo, Fortaleza 60416-030, CE, Brazil; (F.R.F.L.); (C.E.d.S.M.); (S.R.B.D.); (P.d.S.P.); (Á.X.F.); (J.A.O.S.); (T.S.M.); (M.A.L.); (P.F.C.J.); (M.H.L.P.d.S.)
- Department of Morphology, Medical School, Federal University of Ceara, Rua Delmiro de Farias s/n, Rodolfo Teofilo, Fortaleza 60416-030, CE, Brazil
| |
Collapse
|
9
|
Otto-Dobos LD, Strehle LD, Loman BR, Seng MM, Sardesai SD, Williams NO, Gatti-Mays ME, Stover DG, Sudheendra PK, Wesolowski R, Andridge RR, Bailey MT, Pyter LM. Baseline gut microbiome alpha diversity predicts chemotherapy-induced gastrointestinal symptoms in patients with breast cancer. NPJ Breast Cancer 2024; 10:99. [PMID: 39548124 PMCID: PMC11568184 DOI: 10.1038/s41523-024-00707-6] [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: 04/16/2024] [Accepted: 10/23/2024] [Indexed: 11/17/2024] Open
Abstract
Chemotherapy frequently causes debilitating gastrointestinal symptoms, which are inadequately managed by current treatments. Recent research indicates the gut microbiome plays a role in the pathogenesis of these symptoms. The current study aimed to identify pre-chemotherapy microbiome markers that predict gastrointestinal symptom severity after breast cancer chemotherapy. Fecal samples, blood, and gastrointestinal symptom scores were collected from 59 breast cancer patients before, during, and after chemotherapy. Lower pre-chemotherapy microbiome alpha diversity and abundance of specific microbes (e.g., Faecalibacterium) predicted greater chemotherapy-induced gastrointestinal symptoms. Notably, tumor and diet characteristics were associated with lower pre-chemotherapy alpha diversity. Lower baseline alpha diversity also predicted higher chemotherapy-induced microbiome disruption, which was positively associated with diarrhea symptoms. The results indicate certain cancer patients have lower microbiome diversity before chemotherapy, which is predictive of greater chemotherapy-induced gastrointestinal symptoms and a less resilient microbiome. These patients may be strong candidates for pre-chemotherapy microbiome-directed preventative interventions (e.g., diet change).
Collapse
Affiliation(s)
- Lauren D Otto-Dobos
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA
| | - Lindsay D Strehle
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA
| | - Brett R Loman
- Center for Microbial Pathogenesis and Oral and Gastrointestinal Microbiology Research Affinity Group, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
- Department of Animal Sciences, University of Illinois, Urbana, IL, USA
| | - Melina M Seng
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA
| | - Sagar D Sardesai
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| | - Nicole O Williams
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| | - Margaret E Gatti-Mays
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| | - Daniel G Stover
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| | - Preeti K Sudheendra
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| | - Robert Wesolowski
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| | | | - Michael T Bailey
- Center for Microbial Pathogenesis and Oral and Gastrointestinal Microbiology Research Affinity Group, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Leah M Pyter
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA.
- Department of Psychiatry and Behavioral Health, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
10
|
Takasugi N, Uchida K, Shimoda K, Watanabe E, Kato S, Hiwatari M, Ikeda M, Kato M, Matsui H. Management of fulminant botulism after allogeneic hematopoietic cell transplantation. Pediatr Blood Cancer 2024; 71:e31309. [PMID: 39228072 DOI: 10.1002/pbc.31309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 08/20/2024] [Accepted: 08/21/2024] [Indexed: 09/05/2024]
Affiliation(s)
- Nao Takasugi
- Department of Pediatrics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kaname Uchida
- Department of Pediatrics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Konomi Shimoda
- Department of Pediatrics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Eiichiro Watanabe
- Department of Pediatric Surgery, The University of Tokyo, Tokyo, Japan
| | - Shota Kato
- Department of Pediatrics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Mitsuteru Hiwatari
- Department of Pediatrics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Pediatrics, Teikyo University School of Medicine, Tokyo, Japan
| | - Mahoko Ikeda
- Department of Infectious Diseases, The University of Tokyo, Tokyo, Japan
| | - Motohiro Kato
- Department of Pediatrics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hikoro Matsui
- Department of Pediatrics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
11
|
Marin J, Walewski V, Braun T, Dziri S, Magnan M, Denamur E, Carbonnelle E, Bridier-Nahmias A. Genomic evidence of Escherichia coli gut population diversity translocation in leukemia patients. mSphere 2024; 9:e0053024. [PMID: 39365076 PMCID: PMC11520291 DOI: 10.1128/msphere.00530-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 09/09/2024] [Indexed: 10/05/2024] Open
Abstract
Escherichia coli, a commensal species of the human gut, is an opportunistic pathogen that can reach extra-intestinal compartments, including the bloodstream and the bladder, among others. In non-immunosuppressed patients, purifying or neutral evolution of E. coli populations has been reported in the gut. Conversely, it has been suggested that when migrating to extra-intestinal compartments, E. coli genomes undergo diversifying selection as supported by strong evidence for adaptation. The level of genomic polymorphism and the size of the populations translocating from gut to extra-intestinal compartments is largely unknown. To gain insights into the pathophysiology of these translocations, we investigated the level of polymorphism and the evolutionary forces acting on the genomes of 77 E. coli isolated from various compartments in three immunosuppressed patients. Each patient had a unique strain, which was a mutator in one case. In all instances, we observed that translocation encompasses much of the genomic diversity present in the gut. The same signature of selection, whether purifying or diversifying, and as anticipated, neutral for mutator isolates, was observed in both the gut and bloodstream. Additionally, we found a limited number of non-specific mutations among compartments for non-mutator isolates. In all cases, urine isolates were dominated by neutral selection. These findings indicate that substantial proportions of populations are undergoing translocation and that they present a complex compartment-specific pattern of selection at the patient level.IMPORTANCEIt has been suggested that intra and extra-intestinal compartments differentially constrain the evolution of E. coli strains. Whether host particular conditions, such as immunosuppression, could affect the strain evolutionary trajectories remains understudied. We found that, in immunosuppressed patients, large fractions of E. coli gut populations are translocating with variable modifications of the signature of selection for commensal and pathogenic isolates according to the compartment and/or the patient. Such multiple site sampling should be performed in large cohorts of patients to gain a better understanding of E. coli extra-intestinal diseases.
Collapse
Affiliation(s)
- Julie Marin
- Université Sorbonne Paris Nord, INSERM, IAME, Bobigny, France
| | - Violaine Walewski
- APHP, HUPSSD, Hôpital Avicenne, Service de Microbiologie clinique, Bobigny, France
| | - Thorsten Braun
- Université Sorbonne Paris Nord, INSERM, IAME, Bobigny, France
- APHP, HUPSSD, Hôpital Avicenne, Service de Microbiologie clinique, Bobigny, France
| | - Samira Dziri
- APHP, HUPSSD, Hôpital Avicenne, Service de Microbiologie clinique, Bobigny, France
| | - Mélanie Magnan
- Université Paris Cité, INSERM, IAME, and APHP, Hôpital Bichat, Laboratoire de Génétique Moléculaire, Paris, France
| | - Erick Denamur
- Université Paris Cité, INSERM, IAME, and APHP, Hôpital Bichat, Laboratoire de Génétique Moléculaire, Paris, France
| | - Etienne Carbonnelle
- Université Sorbonne Paris Nord, INSERM, IAME, Bobigny, France
- APHP, HUPSSD, Hôpital Avicenne, Service de Microbiologie clinique, Bobigny, France
| | - Antoine Bridier-Nahmias
- Université Paris Cité, INSERM, IAME, and APHP, Hôpital Bichat, Laboratoire de Génétique Moléculaire, Paris, France
| |
Collapse
|
12
|
Khorashadizadeh S, Abbasifar S, Yousefi M, Fayedeh F, Moodi Ghalibaf A. The Role of Microbiome and Probiotics in Chemo-Radiotherapy-Induced Diarrhea: A Narrative Review of the Current Evidence. Cancer Rep (Hoboken) 2024; 7:e70029. [PMID: 39410854 PMCID: PMC11480522 DOI: 10.1002/cnr2.70029] [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: 11/11/2023] [Revised: 08/28/2024] [Accepted: 09/13/2024] [Indexed: 10/20/2024] Open
Abstract
BACKGROUND In this article, we review the most recent research on probiotics effects on diarrhea in both human and animal models of the condition along with the therapeutic potential of these compounds based on their findings. RECENT FINDINGS Nearly 50%-80% of cancer patients experience chemotherapy-induced diarrhea (CID), serious gastrointestinal toxicity of chemotherapeutic and radiation regimens that leads to prolonged hospitalizations, cardiovascular problems, electrolyte imbalances, disruptions in cancer treatment, poor cancer prognosis, and death. CID is typically categorized as osmotic diarrhea. The depletion of colonic crypts and villi by radiotherapy and chemotherapy agents interferes with the absorptive function of the intestine, thereby decreasing the absorption of chloride and releasing water into the intestinal lumen. Probiotic supplements have been found to be able to reverse the intestinal damage caused by chemo-radiation therapy by promoting the growth of crypt and villi and reducing inflammatory pathways. In addition, they support the modulation of immunological and angiogenesis responses in the gut as well as the metabolism of certain digestive enzymes by altering the gut microbiota. CONCLUSION Beyond the benefits of probiotics, additional clinical research is required to clarify the most effective strain combinations and dosages for preventing chemotherapy and radiotherapy diarrhea.
Collapse
Affiliation(s)
| | - Sara Abbasifar
- Student Research CommitteeBirjand University of Medical SciencesBirjandIran
| | - Mohammad Yousefi
- Student Research CommitteeBirjand University of Medical SciencesBirjandIran
| | - Farzad Fayedeh
- Student Research CommitteeBirjand University of Medical SciencesBirjandIran
| | | |
Collapse
|
13
|
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
|
14
|
Anderson CJ, Boeckaerts L, Chin P, Cardas JB, Xie W, Gonçalves A, Blancke G, Benson S, Rogatti S, Simpson MS, Davey A, Choi SM, Desmet S, Bushman SD, Goeminne G, Vandenabeele P, Desai MS, Vereecke L, Ravichandran KS. Metabolite-based inter-kingdom communication controls intestinal tissue recovery following chemotherapeutic injury. Cell Host Microbe 2024; 32:1469-1487.e9. [PMID: 39197455 DOI: 10.1016/j.chom.2024.07.026] [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: 01/04/2024] [Revised: 06/12/2024] [Accepted: 07/31/2024] [Indexed: 09/01/2024]
Abstract
Cytotoxic chemotherapies have devastating side effects, particularly within the gastrointestinal tract. Gastrointestinal toxicity includes the death and damage of the epithelium and an imbalance in the intestinal microbiota, otherwise known as dysbiosis. Whether dysbiosis is a direct contributor to tissue toxicity is a key area of focus. Here, from both mammalian and bacterial perspectives, we uncover an intestinal epithelial cell death-Enterobacteriaceae signaling axis that fuels dysbiosis. Specifically, our data demonstrate that chemotherapy-induced epithelial cell apoptosis and the purine-containing metabolites released from dying cells drive the inter-kingdom transcriptional re-wiring of the Enterobacteriaceae, including fundamental shifts in bacterial respiration and promotion of purine utilization-dependent expansion, which in turn delays the recovery of the intestinal tract. Inhibition of epithelial cell death or restriction of the Enterobacteriaceae to homeostatic levels reverses dysbiosis and improves intestinal recovery. These findings suggest that supportive therapies that maintain homeostatic levels of Enterobacteriaceae may be useful in resolving intestinal disease.
Collapse
Affiliation(s)
- Christopher J Anderson
- VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium; Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK.
| | - Laura Boeckaerts
- VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Priscilla Chin
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Javier Burgoa Cardas
- VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Wei Xie
- VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Amanda Gonçalves
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium; VIB BioImaging Core, Ghent, Belgium
| | - Gillian Blancke
- VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Sam Benson
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Sebastian Rogatti
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Mariska S Simpson
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Anna Davey
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Sze Men Choi
- VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | | | - Summer D Bushman
- Department of Infection and Immunity, Luxembourg Institute of Health, L-4354 Esch-sur-Alzette, Luxembourg
| | | | - Peter Vandenabeele
- VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Mahesh S Desai
- Department of Infection and Immunity, Luxembourg Institute of Health, L-4354 Esch-sur-Alzette, Luxembourg
| | - Lars Vereecke
- VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Kodi S Ravichandran
- VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium; Division of Immunobiology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
15
|
Zhang M, Guo H. Conversation between host and gut microbiota unveils a "silver bullet" therapeutic option for chemotherapy. Cell Host Microbe 2024; 32:1455-1457. [PMID: 39265529 DOI: 10.1016/j.chom.2024.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 08/12/2024] [Indexed: 09/14/2024]
Abstract
Chemotherapy is associated with the induction of intestinal microbiota dysbiosis and gastrointestinal injuries. In this Cell Host & Microbe issue, Anderson et al. demonstrate that chemotherapy-induced epithelial cell apoptosis drives microbiota imbalance and transcriptional rewiring, which in turn delays intestinal recovery.
Collapse
Affiliation(s)
- Mengdan Zhang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Hao Guo
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian 361102, China; Xiang'an Hospital of Xiamen University, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian 361102, China.
| |
Collapse
|
16
|
Teng Y, Li J, Guo J, Yan C, Wang A, Xia X. Alginate oligosaccharide improves 5-fluorouracil-induced intestinal mucositis by enhancing intestinal barrier and modulating intestinal levels of butyrate and isovalerate. Int J Biol Macromol 2024; 276:133699. [PMID: 38972652 DOI: 10.1016/j.ijbiomac.2024.133699] [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: 01/16/2024] [Revised: 07/03/2024] [Accepted: 07/04/2024] [Indexed: 07/09/2024]
Abstract
Chemotherapy-induced mucositis (CIM) is the typical side effect of chemotherapy. This study investigates the potential of alginate oligosaccharide (AOS) in ameliorating CIM induced by 5-fluorouracil (5-FU) in a murine model and its underlying mechanisms. AOS effectively mitigated body weight loss and histopathological damage, modulated inflammatory cytokines and attenuated the oxidative stress. AOS restored intestinal barrier integrity through enhancing expression of tight junction proteins via MLCK signaling pathway. AOS alleviated intestinal mucosal damage by inhibiting TLR4/MyD88/NF-κB signaling pathway, downregulating the pro-apoptotic protein Bax and upregulating the anti-apoptotic protein Bcl-2. Moreover, AOS significantly enriched intestinal Akkermansiaceae and increased the production of short-chain fatty acids (SCFAs), most notably butyrate and isovalerate. Pre-treatment with butyrate and isovalerate also alleviated 5-FU-induced CIM. In conclusion, AOS effectively mitigated CIM through strenghthening intestinal barrier, attenuating inflammation, and modulating gut microbiota and intestianl levels of butyrate and isovalerate. These finding indicate that AOS could be potentially utilized as a supplemental strategy for prevention or mitigation of CIM.
Collapse
Affiliation(s)
- Yue Teng
- Dalian Jinshiwan Laboratory, Dalian, Liaoning 116034, China; State Key Laboratory of Marine Food Processing and Safety Control, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning 116034, China
| | - Jiahui Li
- Dalian Jinshiwan Laboratory, Dalian, Liaoning 116034, China; State Key Laboratory of Marine Food Processing and Safety Control, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning 116034, China
| | - Jian Guo
- Dalian Jinshiwan Laboratory, Dalian, Liaoning 116034, China; State Key Laboratory of Marine Food Processing and Safety Control, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning 116034, China
| | - Chunhong Yan
- Dalian Jinshiwan Laboratory, Dalian, Liaoning 116034, China; State Key Laboratory of Marine Food Processing and Safety Control, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning 116034, China
| | - Ailing Wang
- Dalian Jinshiwan Laboratory, Dalian, Liaoning 116034, China; State Key Laboratory of Marine Food Processing and Safety Control, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning 116034, China
| | - Xiaodong Xia
- Dalian Jinshiwan Laboratory, Dalian, Liaoning 116034, China; State Key Laboratory of Marine Food Processing and Safety Control, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning 116034, China.
| |
Collapse
|
17
|
Gangaev A, van Sleen Y, Brandhorst N, Hoefakker K, Prajapati B, Singh A, Boerma A, van der Heiden M, Oosting SF, van der Veldt AAM, Hiltermann TJN, GeurtsvanKessel CH, Dingemans AMC, Smit EF, de Vries EGE, Haanen JBAG, Kvistborg P, van Baarle D. mRNA-1273 vaccination induces polyfunctional memory CD4 and CD8 T cell responses in patients with solid cancers undergoing immunotherapy or/and chemotherapy. Front Immunol 2024; 15:1447555. [PMID: 39257577 PMCID: PMC11385311 DOI: 10.3389/fimmu.2024.1447555] [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: 06/11/2024] [Accepted: 08/08/2024] [Indexed: 09/12/2024] Open
Abstract
Introduction Research has confirmed the safety and comparable seroconversion rates following SARS-CoV-2 vaccination in patients with solid cancers. However, the impact of cancer treatment on vaccine-induced T cell responses remains poorly understood. Methods In this study, we expand on previous findings within the VOICE trial by evaluating the functional and phenotypic composition of mRNA-1273-induced T cell responses in patients with solid tumors undergoing immunotherapy, chemotherapy, or both, compared to individuals without cancer. We conducted an ELISpot analysis on 386 participants to assess spike-specific T cell responses 28 days after full vaccination. Further in-depth characterization of using flow cytometry was performed on a subset of 63 participants to analyze the functional phenotype and differentiation state of spike-specific T cell responses. Results ELISpot analysis showed robust induction of spike-specific T cell responses across all treatment groups, with response rates ranging from 75% to 80%. Flow cytometry analysis revealed a distinctive cytokine production pattern across cohorts, with CD4 T cells producing IFNγ, TNF, and IL-2, and CD8 T cells producing IFNγ, TNF, and CCL4. Variations were observed in the proportion of monofunctional CD4 T cells producing TNF, particularly higher in individuals without cancer and patients treated with chemotherapy alone, while those treated with immunotherapy or chemoimmunotherapy predominantly produced IFNγ. Despite these differences, polyfunctional spike-specific memory CD4 and CD8 T cell responses were comparable across cohorts. Notably, immunotherapy-treated patients exhibited an expansion of spike-specific CD4 T cells with a terminally differentiated effector memory phenotype. Discussion These findings demonstrate that systemic treatment in patients with solid tumors does not compromise the quality of polyfunctional mRNA-1273-induced T cell responses. This underscores the importance of COVID-19 vaccination in patients with solid cancers undergoing systemic treatment.
Collapse
Affiliation(s)
- Anastasia Gangaev
- Division of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Yannick van Sleen
- Department of Medical Microbiology and Infection Prevention, University Medical Centre Groningen, Groningen, Netherlands
| | - Nicole Brandhorst
- Division of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Kelly Hoefakker
- Division of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Bimal Prajapati
- Department of Medical Microbiology and Infection Prevention, University Medical Centre Groningen, Groningen, Netherlands
| | - Amrita Singh
- Department of Medical Microbiology and Infection Prevention, University Medical Centre Groningen, Groningen, Netherlands
| | - Annemarie Boerma
- Department of Medical Microbiology and Infection Prevention, University Medical Centre Groningen, Groningen, Netherlands
| | - Marieke van der Heiden
- Department of Medical Microbiology and Infection Prevention, University Medical Centre Groningen, Groningen, Netherlands
| | - Sjoukje F. Oosting
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Astrid A. M. van der Veldt
- Department of Medical Oncology and Radiology & Nuclear Medicine, Erasmus Medical Center (MC)-Cancer Institute, Rotterdam, Netherlands
| | - T. Jeroen N. Hiltermann
- Department of Pulmonary Diseases, University Medical Centre Groningen, Groningen, Netherlands
| | - Corine H. GeurtsvanKessel
- Department of Viroscience, Erasmus Medical Center (MC) Cancer Institute, University Medical Centre, Rotterdam, Netherlands
| | | | - Egbert F. Smit
- Department of Thoracic Oncology, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Elisabeth G. E. de Vries
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - John B. A. G. Haanen
- Division of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Pia Kvistborg
- Division of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Debbie van Baarle
- Department of Medical Microbiology and Infection Prevention, University Medical Centre Groningen, Groningen, Netherlands
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Utrecht, Netherlands
| |
Collapse
|
18
|
Fernandes C, Miranda MCC, Roque CR, Paguada ALP, Mota CAR, Florêncio KGD, Pereira AF, Wong DVT, Oriá RB, Lima-Júnior RCP. Is There an Interplay between Environmental Factors, Microbiota Imbalance, and Cancer Chemotherapy-Associated Intestinal Mucositis? Pharmaceuticals (Basel) 2024; 17:1020. [PMID: 39204125 PMCID: PMC11357004 DOI: 10.3390/ph17081020] [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: 05/29/2024] [Revised: 07/24/2024] [Accepted: 08/01/2024] [Indexed: 09/03/2024] Open
Abstract
Interindividual variation in drug efficacy and toxicity is a significant problem, potentially leading to adverse clinical and economic public health outcomes. While pharmacogenetics and pharmacogenomics have long been considered the primary causes of such heterogeneous responses, pharmacomicrobiomics has recently gained attention. The microbiome, a community of microorganisms living in or on the human body, is a critical determinant of drug response and toxicity. Factors such as diet, lifestyle, exposure to xenobiotics, antibiotics use, illness, and genetics can influence the composition of the microbiota. Changes in the intestinal microbiota are particularly influential in drug responsiveness, especially in cancer chemotherapy. The microbiota can modulate an individual's response to a drug, affecting its bioavailability, clinical effect, and toxicity, affecting treatment outcomes and patient quality of life. For instance, the microbiota can convert drugs into active or toxic metabolites, influencing their efficacy and side effects. Alternatively, chemotherapy can also alter the microbiota, creating a bidirectional interplay. Probiotics have shown promise in modulating the microbiome and ameliorating chemotherapy side effects, highlighting the potential for microbiota-targeted interventions in improving cancer treatment outcomes. This opinion paper addresses how environmental factors and chemotherapy-induced dysbiosis impact cancer chemotherapy gastrointestinal toxicity.
Collapse
Affiliation(s)
- Camila Fernandes
- Department of Physiology and Pharmacology, and Drug Research and Development Center (NPDM), Faculty of Medicine, Federal University of Ceara, Rua Cel Nunes de Melo, 1000, Fortaleza 60430-270, Brazil; (C.F.); (A.L.P.P.); (C.A.R.M.); (K.G.D.F.); (A.F.P.); (D.V.T.W.)
| | | | - Cássia Rodrigues Roque
- Laboratory of Tissue Healing, Ontogeny, and Nutrition, Department of Morphology, and Institute of Biomedicine, Faculty of Medicine, Federal University of Ceara, Fortaleza 60430-170, Brazil; (C.R.R.); (R.B.O.)
| | - Ana Lizeth Padilla Paguada
- Department of Physiology and Pharmacology, and Drug Research and Development Center (NPDM), Faculty of Medicine, Federal University of Ceara, Rua Cel Nunes de Melo, 1000, Fortaleza 60430-270, Brazil; (C.F.); (A.L.P.P.); (C.A.R.M.); (K.G.D.F.); (A.F.P.); (D.V.T.W.)
| | - Carlos Adrian Rodrigues Mota
- Department of Physiology and Pharmacology, and Drug Research and Development Center (NPDM), Faculty of Medicine, Federal University of Ceara, Rua Cel Nunes de Melo, 1000, Fortaleza 60430-270, Brazil; (C.F.); (A.L.P.P.); (C.A.R.M.); (K.G.D.F.); (A.F.P.); (D.V.T.W.)
| | - Katharine Gurgel Dias Florêncio
- Department of Physiology and Pharmacology, and Drug Research and Development Center (NPDM), Faculty of Medicine, Federal University of Ceara, Rua Cel Nunes de Melo, 1000, Fortaleza 60430-270, Brazil; (C.F.); (A.L.P.P.); (C.A.R.M.); (K.G.D.F.); (A.F.P.); (D.V.T.W.)
| | - Anamaria Falcão Pereira
- Department of Physiology and Pharmacology, and Drug Research and Development Center (NPDM), Faculty of Medicine, Federal University of Ceara, Rua Cel Nunes de Melo, 1000, Fortaleza 60430-270, Brazil; (C.F.); (A.L.P.P.); (C.A.R.M.); (K.G.D.F.); (A.F.P.); (D.V.T.W.)
| | - Deysi Viviana Tenazoa Wong
- Department of Physiology and Pharmacology, and Drug Research and Development Center (NPDM), Faculty of Medicine, Federal University of Ceara, Rua Cel Nunes de Melo, 1000, Fortaleza 60430-270, Brazil; (C.F.); (A.L.P.P.); (C.A.R.M.); (K.G.D.F.); (A.F.P.); (D.V.T.W.)
| | - Reinaldo Barreto Oriá
- Laboratory of Tissue Healing, Ontogeny, and Nutrition, Department of Morphology, and Institute of Biomedicine, Faculty of Medicine, Federal University of Ceara, Fortaleza 60430-170, Brazil; (C.R.R.); (R.B.O.)
| | - Roberto César Pereira Lima-Júnior
- Department of Physiology and Pharmacology, and Drug Research and Development Center (NPDM), Faculty of Medicine, Federal University of Ceara, Rua Cel Nunes de Melo, 1000, Fortaleza 60430-270, Brazil; (C.F.); (A.L.P.P.); (C.A.R.M.); (K.G.D.F.); (A.F.P.); (D.V.T.W.)
| |
Collapse
|
19
|
Bum Lee J, Huang Y, Oya Y, Nutzinger J, LE Ang Y, Sooi K, Chul Cho B, Soo RA. Modulating the gut microbiome in non-small cell lung cancer: Challenges and opportunities. Lung Cancer 2024; 194:107862. [PMID: 38959670 DOI: 10.1016/j.lungcan.2024.107862] [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: 04/15/2024] [Revised: 06/19/2024] [Accepted: 06/22/2024] [Indexed: 07/05/2024]
Abstract
Despite the efficacy of immunotherapy in non-small cell lung cancer (NSCLC), the majority of the patients experience relapse with limited subsequent treatment options. Preclinical studies of various epithelial tumors, such as melanoma and NSCLC, have shown that harnessing the gut microbiome resulted in improvement of therapeutic responses to immunotherapy. Is this review, we summarize the role of microbiome, including lung and gut microbiome in the context of NSCLC, provide overview of the mechanisms of microbiome in efficacy and toxicity of chemotherapies and immunotherapies, and address current ongoing clinical trials for NSCLC including fecal microbiota transplantation (FMT) and live biotherapeutic products (LBPs).
Collapse
Affiliation(s)
- Jii Bum Lee
- Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Yiqing Huang
- Department of Haematology-Oncology, National University Cancer Institute, Singapore
| | - Yuko Oya
- Department of Respiratory Medicine, Fujita Health University, Toyoake, Japan
| | - Jorn Nutzinger
- Department of Haematology-Oncology, National University Cancer Institute, Singapore
| | - Yvonne LE Ang
- Department of Haematology-Oncology, National University Cancer Institute, Singapore
| | - Kenneth Sooi
- Department of Haematology-Oncology, National University Cancer Institute, Singapore
| | - Byoung Chul Cho
- Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Ross A Soo
- Department of Haematology-Oncology, National University Cancer Institute, Singapore.
| |
Collapse
|
20
|
Xu J, Han J, Jin S, Yu B, Li X, Ma X, Sun L, Li C, Zhao L, Ni X. Modulation of mercaptopurine intestinal toxicity and pharmacokinetics by gut microbiota. Biomed Pharmacother 2024; 177:116975. [PMID: 38925017 DOI: 10.1016/j.biopha.2024.116975] [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: 02/18/2024] [Revised: 06/04/2024] [Accepted: 06/15/2024] [Indexed: 06/28/2024] Open
Abstract
The interaction between the gut microbiota and mercaptopurine (6-MP), a crucial drug used in pediatric acute lymphoblastic leukemia (ALL) treatment, has not been extensively studied. Here we reveal the significant perturbation of gut microbiota after 2-week 6-MP treatment in beagles and mice followed by the functional prediction that showed impairment of SCFAs production and altered amino acid synthesis. And the targeted metabolomics in plasma also showed changes in amino acids. Additionally, targeted metabolomics analysis of feces showed changes in amino acids and SCFAs. Furthermore, ablating the intestinal microbiota by broad-spectrum antibiotics exacerbated the imbalance of amino acids, particularly leading to a significant decrease in the concentration of S-adenosylmethionine (SAM). Importantly, the depletion of gut microbiota worsened the damage of small intestine caused by 6-MP, resulting in increased intestinal permeability. Considering the relationship between toxicity and 6-MP metabolites, we conducted a pharmacokinetic study in pseudo germ-free rats to confirm that gut microbiota depletion altered the methylation metabolites of 6-MP. Specifically, the concentration of MeTINs, a secondary methylation metabolite, showed a negative correlation with SAM, the pivotal methyl donor. Additionally, we observed a strong correlation between Alistipes and SAM levels in both feces and plasma. In conclusion, our study demonstrates that 6-MP disrupts the gut microbiota, and depleting the gut microbiota exacerbates 6-MP-induced intestinal toxicity. Moreover, SAM derived from microbiota plays a crucial role in influencing plasma SAM and the methylation of 6-MP. These findings underscore the importance of comprehending the role of the gut microbiota in 6-MP metabolism and toxicity.
Collapse
Affiliation(s)
- Jiamin Xu
- Department of Pharmacy, Beijing Children's Hospital, Capital Medical University, Beijing 100045, China; Department of Pharmacy, Peking University Third Hospital, Beijing 100191, China
| | - Jiaqi Han
- Department of Pharmacy, Beijing Children's Hospital, Capital Medical University, Beijing 100045, China
| | - Siyao Jin
- Department of Pharmacy, Beijing Children's Hospital, Capital Medical University, Beijing 100045, China
| | - Boran Yu
- Department of Pharmacy, Beijing Children's Hospital, Capital Medical University, Beijing 100045, China
| | - Xiaona Li
- Department of Pharmacy, Peking University Third Hospital, Beijing 100191, China
| | - Xiangyu Ma
- Department of Pharmacy, Beijing Children's Hospital, Capital Medical University, Beijing 100045, China
| | | | | | - Libo Zhao
- Department of Pharmacy, Beijing Children's Hospital, Capital Medical University, Beijing 100045, China; Department of Pharmacy, Peking University Third Hospital, Beijing 100191, China.
| | - Xin Ni
- Department of Pharmacy, Beijing Children's Hospital, Capital Medical University, Beijing 100045, China.
| |
Collapse
|
21
|
Stringer AM, Hargreaves BM, Mendes RA, Blijlevens NMA, Bruno JS, Joyce P, Kamath S, Laheij AMGA, Ottaviani G, Secombe KR, Tonkaboni A, Zadik Y, Bossi P, Wardill HR. Updated perspectives on the contribution of the microbiome to the pathogenesis of mucositis using the MASCC/ISOO framework. Support Care Cancer 2024; 32:558. [PMID: 39080025 PMCID: PMC11289053 DOI: 10.1007/s00520-024-08752-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 07/19/2024] [Indexed: 08/02/2024]
Abstract
Advances in the treatment of cancer have significantly improved mortality rates; however, this has come at a cost, with many treatments still limited by their toxic side effects. Mucositis in both the mouth and gastrointestinal tract is common following many anti-cancer agents, manifesting as ulcerative lesions and associated symptoms throughout the alimentary tract. The pathogenesis of mucositis was first defined in 2004 by Sonis, and almost 20 years on, the model continues to be updated reflecting ongoing research initiatives and more sophisticated analytical techniques. The most recent update, published by the Multinational Association for Supportive Care in Cancer and the International Society for Oral Oncology (MASCC/ISOO), highlights the numerous co-occurring events that underpin mucositis development. Most notably, a role for the ecosystem of microorganisms that reside throughout the alimentary tract (the oral and gut microbiota) was explored, building on initial concepts proposed by Sonis. However, many questions remain regarding the true causal contribution of the microbiota and associated metabolome. This review aims to provide an overview of this rapidly evolving area, synthesizing current evidence on the microbiota's contribution to mucositis development and progression, highlighting (i) components of the 5-phase model where the microbiome may be involved, (ii) methodological challenges that have hindered advances in this area, and (iii) opportunities for intervention.
Collapse
Affiliation(s)
- Andrea M Stringer
- Clinical and Health Sciences, University of South Australia, Adelaide, 5000, Australia
| | - Benjamin M Hargreaves
- Clinical and Health Sciences, University of South Australia, Adelaide, 5000, Australia
| | - Rui Amaral Mendes
- Faculty of Medicine, University of Porto/CINTESIS@RISE, Porto, Portugal
- Department of Oral and Maxillofacial Medicine and Diagnostic Sciences, Case Western Reserve University, Cleveland, OH, 44106-7401, USA
| | - Nicole M A Blijlevens
- Department of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Julia S Bruno
- Molecular Oncology Center, Hospital Sírio-Libanês, São Paulo, Brazil
| | - Paul Joyce
- Centre for Pharmaceutical Innovation, Clinical and Health Sciences, University of South Australia, Adelaide, 5000, Australia
| | - Srinivas Kamath
- Centre for Pharmaceutical Innovation, Clinical and Health Sciences, University of South Australia, Adelaide, 5000, Australia
| | - Alexa M G A Laheij
- Department of Oral Medicine, Academic Centre for Dentistry Amsterdam, University of Amsterdam and VU University, Amsterdam, The Netherlands
- Department of Oral and Maxillofacial Surgery, UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Giulia Ottaviani
- Department of Surgical, Medical and Health Sciences, University of Trieste, Trieste, Italy
| | - Kate R Secombe
- The School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, 5005, Australia
| | - Arghavan Tonkaboni
- Department of Oral Medicine, School of Dentistry, Tehran University of Medical Sciences, Tehran, Iran
| | - Yehuda Zadik
- Department of Military Medicine and "Tzameret", Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
- Department of Oral Medicine, Sedation and Imaging, Faculty of Dental Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Paolo Bossi
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20072, Milan, Italy
- IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089, Milan, Italy
| | - Hannah R Wardill
- The School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, 5005, Australia.
- Supportive Oncology Research Group, Precision Cancer Medicine Theme, South Australian Health and Medical Research Institute, Level 5S, Adelaide, 5000, Australia.
| |
Collapse
|
22
|
He Y, Li F, Zhang Y, Zhu X, Lin Z, Li L, Nawaz S, Kulyar MFEA, Iqbal M, Li J. Pediococcus pentosaceus PP34 Ameliorates 5-Fluorouracil-Induced Intestinal Mucositis via Inhibiting Oxidative Stress and Restoring the Gut Microbiota. Probiotics Antimicrob Proteins 2024:10.1007/s12602-024-10324-1. [PMID: 39046671 DOI: 10.1007/s12602-024-10324-1] [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] [Accepted: 07/08/2024] [Indexed: 07/25/2024]
Abstract
Chemotherapy-induced intestinal mucositis based on 5-fluorouracil (5-FU) slows down the progress of cancer treatment and causes significant suffering to patients. Pediococcus pentosaceus (P. pentosaceus), as a type of LAB, has a range of probiotic properties, including antioxidant, immune benefits, and cholesterol-lowering effects, which are attracting increasing attention. However, studies on the protective effect of P. pentosaceus against chemotherapeutic-induced intestinal mucositis caused by 5-FU remain unclear. Therefore, this study aimed to investigate the potential relieving effects of P. pentosaceus PP34 on 5-FU-induced intestinal mucositis and its mechanism. In the present study, a P. pentosaceus PP34 solution (2 × 109 CFU/mL) was administered daily by gavage followed by intraperitoneal injection of 5-FU to model intestinal mucositis. The body weight, serum biochemical indices, jejunal pathological organization, and expression levels of inflammatory cytokines in the jejunum were examined. The results indicated that the mice induced with 5-FU developed typical intestinal mucositis symptoms and histopathological changes with intense inflammatory and oxidative responses. Moreover, the gut microbiota was disturbed, while PP34 effectively decreased the oxidative reactions and the expression levels of inflammatory mediators and regulated the gut microbiota in 5-FU-exposed mice. Taken together, the study indicated that P. pentosaceus PP34 ameliorates 5-Fluorouracil-induced intestinal mucositis via inhibiting oxidative stress and restoring the gut microbiota.
Collapse
Affiliation(s)
- Yuanyuan He
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Feiran Li
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Yu Zhang
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Xiaohui Zhu
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Zhengrong Lin
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Linxiao Li
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Shah Nawaz
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | | | - Mudassar Iqbal
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Jiakui Li
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China.
- College of Animals Husbandry and Veterinary Medicine, Tibet Agricultural and Animal Husbandry University, Linzhi, Tibet, 860000, People's Republic of China.
| |
Collapse
|
23
|
Lu L, Li F, Gao Y, Kang S, Li J, Guo J. Microbiome in radiotherapy: an emerging approach to enhance treatment efficacy and reduce tissue injury. Mol Med 2024; 30:105. [PMID: 39030525 PMCID: PMC11264922 DOI: 10.1186/s10020-024-00873-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 07/08/2024] [Indexed: 07/21/2024] Open
Abstract
Radiotherapy is a widely used cancer treatment that utilizes powerful radiation to destroy cancer cells and shrink tumors. While radiation can be beneficial, it can also harm the healthy tissues surrounding the tumor. Recent research indicates that the microbiota, the collection of microorganisms in our body, may play a role in influencing the effectiveness and side effects of radiation therapy. Studies have shown that specific species of bacteria living in the stomach can influence the immune system's response to radiation, potentially increasing the effectiveness of treatment. Additionally, the microbiota may contribute to adverse effects like radiation-induced diarrhea. A potential strategy to enhance radiotherapy outcomes and capitalize on the microbiome involves using probiotics. Probiotics are living microorganisms that offer health benefits when consumed in sufficient quantities. Several studies have indicated that probiotics have the potential to alter the composition of the gut microbiota, resulting in an enhanced immune response to radiation therapy and consequently improving the efficacy of the treatment. It is important to note that radiation can disrupt the natural balance of gut bacteria, resulting in increased intestinal permeability and inflammatory conditions. These disruptions can lead to adverse effects such as diarrhea and damage to the intestinal lining. The emerging field of radiotherapy microbiome research offers a promising avenue for optimizing cancer treatment outcomes. This paper aims to provide an overview of the human microbiome and its role in augmenting radiation effectiveness while minimizing damage.
Collapse
Affiliation(s)
- Lina Lu
- School of Chemical Engineering, Northwest Minzu University, No.1, Northwest New Village, Lanzhou, Gansu, 730030, China.
- Key Laboratory of Environment-Friendly Composite Materials of the State Ethnic Affairs Commission, Lanzhou, Gansu, China.
- Gansu Provincial Biomass Function Composites Engineering Research Center, Lanzhou, Gansu, China.
- Key Laboratory for Utility of Environment-Friendly Composite Materials and Biomass in, University of Gansu Province, Lanzhou, Gansu, China.
| | - Fengxiao Li
- Department of Pharmacy, the Affiliated Hospital of Qingdao University, Qingdao, China
| | | | - Shuhe Kang
- School of Chemical Engineering, Northwest Minzu University, No.1, Northwest New Village, Lanzhou, Gansu, 730030, China
- Key Laboratory of Environment-Friendly Composite Materials of the State Ethnic Affairs Commission, Lanzhou, Gansu, China
- Gansu Provincial Biomass Function Composites Engineering Research Center, Lanzhou, Gansu, China
- Key Laboratory for Utility of Environment-Friendly Composite Materials and Biomass in, University of Gansu Province, Lanzhou, Gansu, China
| | - Jia Li
- School of Chemical Engineering, Northwest Minzu University, No.1, Northwest New Village, Lanzhou, Gansu, 730030, China
- Key Laboratory of Environment-Friendly Composite Materials of the State Ethnic Affairs Commission, Lanzhou, Gansu, China
- Gansu Provincial Biomass Function Composites Engineering Research Center, Lanzhou, Gansu, China
- Key Laboratory for Utility of Environment-Friendly Composite Materials and Biomass in, University of Gansu Province, Lanzhou, Gansu, China
| | - Jinwang Guo
- School of Chemical Engineering, Northwest Minzu University, No.1, Northwest New Village, Lanzhou, Gansu, 730030, China
- Key Laboratory of Environment-Friendly Composite Materials of the State Ethnic Affairs Commission, Lanzhou, Gansu, China
- Gansu Provincial Biomass Function Composites Engineering Research Center, Lanzhou, Gansu, China
- Key Laboratory for Utility of Environment-Friendly Composite Materials and Biomass in, University of Gansu Province, Lanzhou, Gansu, China
| |
Collapse
|
24
|
Cazzaniga M, Cardinali M, Di Pierro F, Zonzini GB, Palazzi CM, Gregoretti A, Zerbinati N, Guasti L, Matera MR, Cavecchia I, Bertuccioli A. The Role of Short-Chain Fatty Acids, Particularly Butyrate, in Oncological Immunotherapy with Checkpoint Inhibitors: The Effectiveness of Complementary Treatment with Clostridium butyricum 588. Microorganisms 2024; 12:1235. [PMID: 38930617 PMCID: PMC11206605 DOI: 10.3390/microorganisms12061235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/06/2024] [Accepted: 06/16/2024] [Indexed: 06/28/2024] Open
Abstract
The discovery of immune checkpoints (CTLA-4, PD-1, and PD-L1) and their impact on the prognosis of oncological diseases have paved the way for the development of revolutionary oncological treatments. These treatments do not combat tumors with drugs "against" cancer cells but rather support and enhance the ability of the immune system to respond directly to tumor growth by attacking the cancer cells with lymphocytes. It has now been widely demonstrated that the presence of an adequate immune response, essentially represented by the number of TILs (tumor-infiltrating lymphocytes) present in the tumor mass decisively influences the response to treatments and the prognosis of the disease. Therefore, immunotherapy is based on and cannot be carried out without the ability to increase the presence of lymphocytic cells at the tumor site, thereby limiting and nullifying certain tumor evasion mechanisms, particularly those expressed by the activity (under positive physiological conditions) of checkpoints that restrain the response against transformed cells. Immunotherapy has been in the experimental phase for decades, and its excellent results have made it a cornerstone of treatments for many oncological pathologies, especially when combined with chemotherapy and radiotherapy. Despite these successes, a significant number of patients (approximately 50%) do not respond to treatment or develop resistance early on. The microbiota, its composition, and our ability to modulate it can have a positive impact on oncological treatments, reducing side effects and increasing sensitivity and effectiveness. Numerous studies published in high-ranking journals confirm that a certain microbial balance, particularly the presence of bacteria capable of producing short-chain fatty acids (SCFAs), especially butyrate, is essential not only for reducing the side effects of chemoradiotherapy treatments but also for a better response to immune treatments and, therefore, a better prognosis. This opens up the possibility that favorable modulation of the microbiota could become an essential complementary treatment to standard oncological therapies. This brief review aims to highlight the key aspects of using precision probiotics, such as Clostridium butyricum, that produce butyrate to improve the response to immune checkpoint treatments and, thus, the prognosis of oncological diseases.
Collapse
Affiliation(s)
- Massimiliano Cazzaniga
- Scientific & Research Department, Velleja Research, 20125 Milan, Italy; (M.C.); (F.D.P.)
- Microbiota International Clinical Society, 10123 Torino, Italy; (A.G.); (M.R.M.); (I.C.); (A.B.)
| | - Marco Cardinali
- Department of Internal Medicine, Infermi Hospital, AUSL Romagna, 47921 Rimini, Italy;
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61122 Urbino, Italy;
| | - Francesco Di Pierro
- Scientific & Research Department, Velleja Research, 20125 Milan, Italy; (M.C.); (F.D.P.)
- Microbiota International Clinical Society, 10123 Torino, Italy; (A.G.); (M.R.M.); (I.C.); (A.B.)
- Department of Medicine and Surgery, University of Insurbia, 21100 Varese, Italy; (N.Z.); (L.G.)
| | - Giordano Bruno Zonzini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61122 Urbino, Italy;
| | - Chiara Maria Palazzi
- Microbiota International Clinical Society, 10123 Torino, Italy; (A.G.); (M.R.M.); (I.C.); (A.B.)
| | - Aurora Gregoretti
- Microbiota International Clinical Society, 10123 Torino, Italy; (A.G.); (M.R.M.); (I.C.); (A.B.)
| | - Nicola Zerbinati
- Department of Medicine and Surgery, University of Insurbia, 21100 Varese, Italy; (N.Z.); (L.G.)
| | - Luigina Guasti
- Department of Medicine and Surgery, University of Insurbia, 21100 Varese, Italy; (N.Z.); (L.G.)
| | - Maria Rosaria Matera
- Microbiota International Clinical Society, 10123 Torino, Italy; (A.G.); (M.R.M.); (I.C.); (A.B.)
| | - Ilaria Cavecchia
- Microbiota International Clinical Society, 10123 Torino, Italy; (A.G.); (M.R.M.); (I.C.); (A.B.)
| | - Alexander Bertuccioli
- Microbiota International Clinical Society, 10123 Torino, Italy; (A.G.); (M.R.M.); (I.C.); (A.B.)
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61122 Urbino, Italy;
| |
Collapse
|
25
|
Guan Y, Wu D, Wang H, Liu N. Microbiome-driven anticancer therapy: A step forward from natural products. MLIFE 2024; 3:219-230. [PMID: 38948147 PMCID: PMC11211674 DOI: 10.1002/mlf2.12118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 12/25/2023] [Accepted: 01/25/2024] [Indexed: 07/02/2024]
Abstract
Human microbiomes, considered as a new emerging and enabling cancer hallmark, are increasingly recognized as critical effectors in cancer development and progression. Manipulation of microbiome revitalizing anticancer therapy from natural products shows promise toward improving cancer outcomes. Herein, we summarize our current understanding of the human microbiome-driven molecular mechanisms impacting cancer progression and anticancer therapy. We highlight the potential translational and clinical implications of natural products for cancer prevention and treatment by developing targeted therapeutic strategies as adjuvants for chemotherapy and immunotherapy against tumorigenesis. The challenges and opportunities for future investigations using modulation of the microbiome for cancer treatment are further discussed in this review.
Collapse
Affiliation(s)
- Yunxuan Guan
- State Key Laboratory of Systems Medicine for Cancer, Center for Single‐Cell Omics, School of Public HealthShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Di Wu
- State Key Laboratory of Systems Medicine for Cancer, Center for Single‐Cell Omics, School of Public HealthShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Hui Wang
- State Key Laboratory of Systems Medicine for Cancer, Center for Single‐Cell Omics, School of Public HealthShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Ning‐Ning Liu
- State Key Laboratory of Systems Medicine for Cancer, Center for Single‐Cell Omics, School of Public HealthShanghai Jiao Tong University School of MedicineShanghaiChina
| |
Collapse
|
26
|
Yan X, Bai L, Lv J, Qi P, Song X, Zhang L. Effects of Combined Live Bifidobacterium, Lactobacillus, Enterococcus, and Bacillus Cereus Tablets on the Structure and Function of the Intestinal Flora in Rabbits Undergoing Hepatic Artery Infusion Chemotherapy. BIOLOGY 2024; 13:327. [PMID: 38785809 PMCID: PMC11117994 DOI: 10.3390/biology13050327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 04/16/2024] [Accepted: 05/07/2024] [Indexed: 05/25/2024]
Abstract
Few studies have explored the biological mechanism by which probiotics alleviate adverse reactions to chemotherapy drugs after local hepatic chemotherapy perfusion by regulating the intestinal flora. This study investigates the effects of Combined Live Bifidobacterium, Lactobacillus, Enterococcus, and Bacillus Cereus Tablets on the intestinal microbial structure and intestinal barrier function, as well as the potential mechanism in rabbits after local hepatic chemotherapy infusion. Eighteen New Zealand White rabbits were randomly divided into a control group, a hepatic local chemotherapy perfusion group, and a hepatic local chemotherapy perfusion + Combined Live Bifidobacterium, Lactobacillus, Enterococcus, and Bacillus Cereus Tablets group to assess the effects of Combined Live Bifidobacterium, Lactobacillus, Enterococcus, and Bacillus Cereus Tablets on the adverse reactions. The administration of Combined Live Bifidobacterium, Lactobacillus, Enterococcus, and Bacillus Cereus Tablets alleviated the intestinal flora disorder caused by local hepatic perfusion chemotherapy, promoted the growth of beneficial bacteria, and inhibited the growth of harmful bacteria. The Combined Live Bifidobacterium, Lactobacillus, Enterococcus, and Bacillus Cereus Tablets also reduced the levels of serum pro-inflammatory cytokines and liver injury factors induced by local hepatic perfusion chemotherapy. Our findings indicate that Combined Live Bifidobacterium, Lactobacillus, Enterococcus, and Bacillus Cereus Tablets can ameliorate the toxicity and side effects of chemotherapy by regulating intestinal flora, blocking pro-inflammatory cytokines, reducing liver injury factors, and repairing the intestinal barrier. Probiotics may be used as a potential alternative therapeutic strategy to prevent the adverse reactions caused by chemotherapy with local hepatic perfusion.
Collapse
Affiliation(s)
- Xiangdong Yan
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, China; (X.Y.); (L.B.); (J.L.); (P.Q.); (X.S.)
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, China
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, The First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Liuhui Bai
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, China; (X.Y.); (L.B.); (J.L.); (P.Q.); (X.S.)
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, China
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, The First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Jin Lv
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, China; (X.Y.); (L.B.); (J.L.); (P.Q.); (X.S.)
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, China
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, The First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Ping Qi
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, China; (X.Y.); (L.B.); (J.L.); (P.Q.); (X.S.)
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, China
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, The First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Xiaojing Song
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, China; (X.Y.); (L.B.); (J.L.); (P.Q.); (X.S.)
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, China
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, The First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Lei Zhang
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, China; (X.Y.); (L.B.); (J.L.); (P.Q.); (X.S.)
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, China
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, The First Hospital of Lanzhou University, Lanzhou 730000, China
| |
Collapse
|
27
|
Kuo YR, Lin CH, Lin WS, Pan MH. L-Glutamine Substantially Improves 5-Fluorouracil-Induced Intestinal Mucositis by Modulating Gut Microbiota and Maintaining the Integrity of the Gut Barrier in Mice. Mol Nutr Food Res 2024; 68:e2300704. [PMID: 38656560 DOI: 10.1002/mnfr.202300704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 03/26/2024] [Indexed: 04/26/2024]
Abstract
SCOPE This study investigates the potential of glutamine to mitigate intestinal mucositis and dysbiosis caused by the chemotherapeutic agent 5-fluorouracil (5-FU). METHODS AND RESULTS Over twelve days, Institute of Cancer Research (ICR) mice are given low (0.5 mg kg-1) or high (2 mg kg-1) doses of L-Glutamine daily, with 5-FU (50 mg kg-1) administered between days six and nine. Mice receiving only 5-FU exhibited weight loss, diarrhea, abnormal cell growth, and colonic inflammation, correlated with decreased mucin proteins, increased endotoxins, reduced fecal short-chain fatty acids, and altered gut microbiota. Glutamine supplementation counteracted these effects by inhibiting the Toll-like receptor 4/nuclear factor kappa B (TLR4/NF-κB) pathway, modulating nuclear factor erythroid 2-related factor 2/heme oxygenase 1 (Nrf2/HO-1) oxidative stress proteins, and increasing mammalian target of rapamycin (mTOR) levels, thereby enhancing microbial diversity and protecting intestinal mucosa. CONCLUSIONS These findings underscore glutamine's potential in preventing 5-FU-induced mucositis by modulating gut microbiota and inflammation pathways.
Collapse
Affiliation(s)
- Ya-Ru Kuo
- Institute of Food Science and Technology, National Taiwan University, Taipei, 10617, Taiwan
| | - Cheng-Hung Lin
- Institute of Food Science and Technology, National Taiwan University, Taipei, 10617, Taiwan
| | - Wei-Sheng Lin
- Institute of Food Science and Technology, National Taiwan University, Taipei, 10617, Taiwan
- Department of Food Science, National Quemoy University, Quemoy County, 89250, Taiwan
| | - Min-Hsiung Pan
- Institute of Food Science and Technology, National Taiwan University, Taipei, 10617, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung City, 40402, Taiwan
- Department of Health and Nutrition Biotechnology, Asia University, Taichung City, 41354, Taiwan
| |
Collapse
|
28
|
Chen C, Xu JL, Gu ZC, Zhou SS, Wei GL, Gu JL, Ma HL, Feng YQ, Song ZW, Yan ZP, Deng S, Ding R, Li SL, Huo JG. Danggui Sini decoction alleviates oxaliplatin-induced peripheral neuropathy by regulating gut microbiota and potentially relieving neuroinflammation related metabolic disorder. Chin Med 2024; 19:58. [PMID: 38584284 PMCID: PMC10999090 DOI: 10.1186/s13020-024-00929-7] [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: 07/05/2023] [Accepted: 04/01/2024] [Indexed: 04/09/2024] Open
Abstract
BACKGROUND Danggui Sini decoction (DSD), a traditional Chinese medicine formula, has the function of nourishing blood, warming meridians, and unblocking collaterals. Our clinical and animal studies had shown that DSD can effectively protect against oxaliplatin (OXA)-induced peripheral neuropathy (OIPN), but the detailed mechanisms remain uncertain. Multiple studies have confirmed that gut microbiota plays a crucial role in the development of OIPN. In this study, the potential mechanism of protective effect of DSD against OIPN by regulating gut microbiota was investigated. METHODS The neuroprotective effects of DSD against OIPN were examined on a rat model of OIPN by determining mechanical allodynia, biological features of dorsal root ganglia (DRG) as well as proinflammatory indicators. Gut microbiota dysbiosis was characterized using 16S rDNA gene sequencing and metabolism disorders were evaluated using untargeted and targeted metabolomics. Moreover the gut microbiota mediated mechanisms were validated by antibiotic intervention and fecal microbiota transplantation. RESULTS DSD treatment significantly alleviated OIPN symptoms by relieving mechanical allodynia, preserving DRG integrity and reducing proinflammatory indicators lipopolysaccharide (LPS), IL-6 and TNF-α. Besides, DSD restored OXA induced intestinal barrier disruption, gut microbiota dysbiosis as well as systemic metabolic disorders. Correlation analysis revealed that DSD increased bacterial genera such as Faecalibaculum, Allobaculum, Dubosiella and Rhodospirillales_unclassified were closely associated with neuroinflammation related metabolites, including positively with short-chain fatty acids (SCFAs) and sphingomyelin (d18:1/16:0), and negatively with pi-methylimidazoleacetic acid, L-glutamine and homovanillic acid. Meanwhile, antibiotic intervention apparently relieved OIPN symptoms. Furthermore, fecal microbiota transplantation further confirmed the mediated effects of gut microbiota. CONCLUSION DSD alleviates OIPN by regulating gut microbiota and potentially relieving neuroinflammation related metabolic disorder.
Collapse
Affiliation(s)
- Chen Chen
- Department of Oncology, Yancheng TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Yancheng, 224001, Jiangsu, China
- Department of Oncology, Yancheng TCM Hospital, Yancheng, 224001, Jiangsu, China
- The Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Jian-Lin Xu
- Department of Oncology, Yancheng TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Yancheng, 224001, Jiangsu, China
- Department of Oncology, Yancheng TCM Hospital, Yancheng, 224001, Jiangsu, China
| | - Zhan-Cheng Gu
- Department of Oncology, Kunshan Hospital of Traditional Chinese Medicine, Suzhou, 215399, China
| | - Shan-Shan Zhou
- Department of Pharmaceutical Analysis, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, No. 100 Shizi Street Hongshan Road, Nanjing, 210028, Jiangsu, China
- Department of Metabolomics, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, Jiangsu, China
| | - Guo-Li Wei
- Department of Oncology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, No. 100 Shizi Street Hongshan Road, Nanjing, 210028, Jiangsu, China
- Department of Oncology, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, Jiangsu, China
- Department of Oncology, Nanjing Lishui District Hospital of Traditional Chinese Medicine, Nanjing, 211299, Jiangsu, China
| | - Jia-Lin Gu
- The Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Hai-Long Ma
- Department of Paediatrics, Yancheng TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Yancheng, 224001, Jiangsu, China
| | - Yan-Qi Feng
- The Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Zi-Wei Song
- The Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Zhan-Peng Yan
- Clinical Research Department of Chinese and Western Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, Jiangsu, China
| | - Shan Deng
- Department of Oncology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, No. 100 Shizi Street Hongshan Road, Nanjing, 210028, Jiangsu, China
- Department of Oncology, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, Jiangsu, China
| | - Rong Ding
- Department of Oncology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, No. 100 Shizi Street Hongshan Road, Nanjing, 210028, Jiangsu, China
- Department of Oncology, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, Jiangsu, China
| | - Song-Lin Li
- Department of Pharmaceutical Analysis, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, No. 100 Shizi Street Hongshan Road, Nanjing, 210028, Jiangsu, China.
- Department of Metabolomics, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, Jiangsu, China.
| | - Jie-Ge Huo
- Department of Oncology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, No. 100 Shizi Street Hongshan Road, Nanjing, 210028, Jiangsu, China.
- Department of Oncology, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, Jiangsu, China.
| |
Collapse
|
29
|
Reis SK, Socca EAR, de Souza BR, Genaro SC, Durán N, Fávaro WJ. Effects of probiotic supplementation on chronic inflammatory process modulation in colorectal carcinogenesis. Tissue Cell 2024; 87:102293. [PMID: 38244400 DOI: 10.1016/j.tice.2023.102293] [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: 09/17/2023] [Revised: 12/17/2023] [Accepted: 12/20/2023] [Indexed: 01/22/2024]
Abstract
The current study investigated the potential effects of probiotic supplementation on colorectal carcinogenesis chemically induced with 1,2-dimethylhydrazine (DMH) and treated with 5-fluorouracil (5FU)-based chemotherapy in mice. Animals were randomly allocated in five different groups: Control: which not receive any treatment throughout the experimental course; Colitis model group (DMH): treated with DMH; DMH+ 5FU: animals received I.P. (intraperitoneal) dose of chemotherapy on a weekly basis; DMH+PROB: animals received daily administrations (via gavage) of probiotics (Lactobacillus: acidophilus and paracasei, Bifidobacterium lactis and bifidum); and DMH+ PROB+ 5FU: animals received the same treatment as the previous groups. After ten-week treatment, mice's large intestine was collected and subjected to colon length, histopathological, periodic acid-schiff (PAS) staining and immunohistochemistry (TLR2, MyD88, NF-κB, IL-6, TLR4, TRIF, IRF-3, IFN-γ, Ki-67, KRAS, p53, IL-10, and TGF-β) analyzes. Variance (ANOVA) and Kruskal-Wallis tests were used for statistical analysis, at significance level p 0.05. Probiotics' supplementation has increased the production of Ki-67 cell-proliferation marker, reduced body weight, and colon shortening, as well as modulated the chronic inflammatory process in colorectal carcinogenesis by inhibiting NF-κB expression and mitigating mucin depletion. Thus, these findings lay a basis for guide future studies focused on probiotics' action mechanisms in tumor microenvironment which might have implications in clinical practice.
Collapse
Affiliation(s)
- Sabrina Karen Reis
- Faculty Medical Sciences, State University of Campinas (UNICAMP), Campinas, SP, Brazil; Laboratory of Urogenital Carcinogenesis and Immunotherapy, Department of Structural and Functional Biology, State University of Campinas (UNICAMP), Campinas, SP, Brazil.
| | - Eduardo Augusto Rabelo Socca
- Laboratory of Urogenital Carcinogenesis and Immunotherapy, Department of Structural and Functional Biology, State University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Bianca Ribeiro de Souza
- British Columbia's Gynecological Cancer Research (OVCARE) Program and Department of Obstetrics and Gynecology, University of British Columbia, Vancouver General Hospital, Vancouver, BC, Canada.
| | | | - Nelson Durán
- Laboratory of Urogenital Carcinogenesis and Immunotherapy, Department of Structural and Functional Biology, State University of Campinas (UNICAMP), Campinas, SP, Brazil; Nanomedicine Research Unit (Nanomed), Federal University of ABC (UFABC), Santo André, SP, Brazil
| | - Wagner José Fávaro
- Faculty Medical Sciences, State University of Campinas (UNICAMP), Campinas, SP, Brazil; Laboratory of Urogenital Carcinogenesis and Immunotherapy, Department of Structural and Functional Biology, State University of Campinas (UNICAMP), Campinas, SP, Brazil
| |
Collapse
|
30
|
Zeng T, Deng YH, Lin CH, Chen XX, Jia HX, Hu XW, Xia T, Ling Y, Zhang LH, Cao TF. A randomized trial of Bacteroides fragilis 839 on preventing chemotherapy-induced myelosuppression and gastrointestinal adverse effects in breast cancer patients. Asia Pac J Clin Nutr 2024; 33:23-32. [PMID: 38494684 PMCID: PMC11170005 DOI: 10.6133/apjcn.202403_33(1).0003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 01/04/2024] [Accepted: 11/10/2023] [Indexed: 03/19/2024]
Abstract
BACKGROUND AND OBJECTIVES To evaluate the potential benefits of Bacteroides fragilis 839 (BF839), a next-generation probiotics, in reducing myelosuppression and gastrointestinal toxicity associated with chemotherapy in breast cancer patient. METHODS AND STUDY DESIGN 40 women with early breast cancer were randomly assigned to the BF839 (n=20) or placebo (n=20) during the administration of adjuvant chemotherapy (4 cycles of epirubicin 100mg/m2 and cyclophosphamide 600mg/m2). Myelosuppression and gastrointestinal adverse effects were monitored in both groups. RESULTS Throughout the four treatment cycles, the percentage of patients experiencing myelosuppression was 42.5% in the BF839 group, significantly lower than the 66.3% observed in the control group (p=0.003). Two patients in the BF839 group and three patients in the placebo group received recombinant human granulocyte colony-stimulating factor (rhG-CSF) due to leuko-penia/neutropenia. When considering an ITT analysis, which included all patients regardless of rhG-CSF treatment, the BF839 group exhibited less reduction from baseline in white blood cells (-0.31±1.19 vs -1.15±0.77, p=0.012) and neutrophils (0.06±1.00 vs -0.84±0.85, p=0.004) compared to the placebo group. The difference became even more significant when excluding the patients who received rhG-CSF injections. Throughout the four treatment cycles, compared to the placebo group, the BF839 group had significantly lower rates of 3-4 grade nausea (35.0% vs 71.3%, p=0.001), vomiting (20.0% vs 45.0%, p=0.001), and diarrhea (15.0% vs 30.0%, p=0.023). CONCLUSIONS These findings suggest that BF839 has the potential to effectively mitigate myelosuppression and gastrointestinal toxicity associated with chemotherapy in breast cancer patients.
Collapse
Affiliation(s)
- Ting Zeng
- Clinical Nutrition, The Second Affiliated Hospital of Guangzhou Medical University
| | - Yu-Hong Deng
- Clinical Nutrition, The Second Affiliated Hospital of Guangzhou Medical University
| | - Chu-Hui Lin
- Clinical Nutrition, The Second Affiliated Hospital of Guangzhou Medical University
| | - Xin-Xin Chen
- Breast Surgery, The Second Affiliated Hospital of Guangzhou Medical University
| | - Hai-Xia Jia
- Breast Surgery, The Second Affiliated Hospital of Guangzhou Medical University
| | - Xiao-Wu Hu
- Breast Surgery, The Second Affiliated Hospital of Guangzhou Medical University
| | - Ting Xia
- Breast Surgery, The Second Affiliated Hospital of Guangzhou Medical University
| | - Yun Ling
- Breast Surgery, The Second Affiliated Hospital of Guangzhou Medical University
| | - Le-Hong Zhang
- Breast Surgery, The Second Affiliated Hospital of Guangzhou Medical University.
| | - Teng-Fei Cao
- Breast Surgery, The Second Affiliated Hospital of Guangzhou Medical University.
| |
Collapse
|
31
|
Cazzaniga M, Cardinali M, Di Pierro F, Zonzini GB, Palazzi CM, Gregoretti A, Zerbinati N, Guasti L, Bertuccioli A. The Potential Role of Probiotics, Especially Butyrate Producers, in the Management of Gastrointestinal Mucositis Induced by Oncologic Chemo-Radiotherapy. Int J Mol Sci 2024; 25:2306. [PMID: 38396981 PMCID: PMC10889689 DOI: 10.3390/ijms25042306] [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: 01/16/2024] [Revised: 02/08/2024] [Accepted: 02/13/2024] [Indexed: 02/25/2024] Open
Abstract
Many clinical studies have now highlighted how the composition of the intestinal microbiota can regulate the effects of many oncological therapies. In particular, the modulation of microbial composition has been shown to enhance their efficacy and reduce potential side effects. Numerous adverse events induced by chemotherapy and radiotherapy appear to be strongly associated with an alteration in the intestinal microbiota caused by these treatments. This supports the hypothesis that the modulation or correction of the microbiota may decrease the toxic impact of therapies, improving patient compliance and quality of life. Among the most debilitating disorders related to oncological treatments is certainly mucositis, and recent clinical data highlight how the deficiency of short-chain fatty acids, especially butyrate, and specifically the lack of certain bacterial groups responsible for its production (butyrate producers), is strongly associated with this disorder. It is hypothesized that restoring these elements may influence the onset and severity of adverse events. Therefore, the intake of probiotics, especially butyrate producers, and specifically Clostridium butyricum (CBM588), currently the only cultivable and usable strain with a history of data proving its safety, could be a valuable ally in oncological therapies, reducing the associated discomfort and improving compliance, efficacy, and quality of life for patients.
Collapse
Affiliation(s)
- Massimiliano Cazzaniga
- Scientific & Research Department, Velleja Research, 20125 Milano, Italy (F.D.P.)
- Microbiota International Clinical Society, 10123 Torino, Italy (A.B.)
| | - Marco Cardinali
- Department of Internal Medicine, Infermi Hospital, AUSL Romagna, 47921 Rimini, Italy
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61122 Urbino, Italy
| | - Francesco Di Pierro
- Scientific & Research Department, Velleja Research, 20125 Milano, Italy (F.D.P.)
- Microbiota International Clinical Society, 10123 Torino, Italy (A.B.)
- Department of Medicine and Surgery, University of Insurbia, 21100 Varese, Italy (L.G.)
| | | | | | - Aurora Gregoretti
- Microbiota International Clinical Society, 10123 Torino, Italy (A.B.)
| | - Nicola Zerbinati
- Department of Medicine and Surgery, University of Insurbia, 21100 Varese, Italy (L.G.)
| | - Luigina Guasti
- Department of Medicine and Surgery, University of Insurbia, 21100 Varese, Italy (L.G.)
| | - Alexander Bertuccioli
- Microbiota International Clinical Society, 10123 Torino, Italy (A.B.)
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61122 Urbino, Italy
| |
Collapse
|
32
|
Liu X, Li Y, Gu M, Xu T, Wang C, Chang P. Radiation enteropathy-related depression: A neglectable course of disease by gut bacterial dysbiosis. Cancer Med 2024; 13:e6865. [PMID: 38457257 PMCID: PMC10923036 DOI: 10.1002/cam4.6865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 12/08/2023] [Accepted: 12/12/2023] [Indexed: 03/10/2024] Open
Abstract
Radiation enteropathy (RE) is common in patients treated with radiotherapy for pelvic-abdominal cancers. Accumulating data indicate that gut commensal bacteria determine intestinal radiosensitivity. Radiotherapy can result in gut bacterial dysbiosis. Gut bacterial dysbiosis contributes to the pathogenesis of RE. Mild to moderate depressive symptoms can be observed in patients with RE in clinical settings; however, the rate of these symptoms has not been reported. Studies have demonstrated that gut bacterial dysbiosis induces depression. In the state of comorbidity, RE and depression may be understood as local and abscopal manifestations of gut bacterial disorders. The ability of comorbid depression to worsen inflammatory bowel disease (IBD) has long been demonstrated and is associated with dysfunction of cholinergic neural anti-inflammatory pathways. There is a lack of direct evidence for RE comorbid with depression. It is widely accepted that RE shares similar pathophysiologic mechanisms with IBD. Therefore, we may be able to draw on the findings of the relationship between IBD and depression. This review will explore the relationship between gut bacteria, RE, and depression in light of the available evidence and indicate a method for investigating the mechanisms of RE combined with depression. We will also describe new developments in the treatment of RE with probiotics, prebiotics, and fecal microbial transplantation.
Collapse
Affiliation(s)
- Xinliang Liu
- Department of Radiation Oncology and TherapyThe First Hospital of Jilin UniversityChangchunChina
| | - Ying Li
- Department of Radiation Oncology and TherapyThe First Hospital of Jilin UniversityChangchunChina
| | - Meichen Gu
- Department of Radiation Oncology and TherapyThe First Hospital of Jilin UniversityChangchunChina
| | - Tiankai Xu
- Department of Radiation Oncology and TherapyThe First Hospital of Jilin UniversityChangchunChina
| | - Chuanlei Wang
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery CenterThe First Hospital of Jilin UniversityChangchunChina
| | - Pengyu Chang
- Department of Radiation Oncology and TherapyThe First Hospital of Jilin UniversityChangchunChina
| |
Collapse
|
33
|
Spalinger MR, Scharl M. Microbiota Manipulation as an Emerging Concept in Cancer Therapy. Visc Med 2024; 40:2-11. [PMID: 38312366 PMCID: PMC10836949 DOI: 10.1159/000534810] [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: 08/31/2023] [Accepted: 10/24/2023] [Indexed: 02/06/2024] Open
Abstract
Background The human body is colonized by billions of bacteria that provide nutrients to the host, train our immune system, and importantly affect our heath. It has long been suggested that microbes might play a role in tumor pathogenesis; however, compelling evidence was only provided in the past decades when novel detection methods that do not depend on culturing techniques had been developed. Summary The microbiome impacts tumor development and anti-tumor therapies on various levels. Bacteria can promote or suppress tumor growth via direct interactions with cancer cells, production of metabolites that promote or inhibit tumor growth, and via stimulation or suppression of the local and systemic immune response. Cancer patients harbor a distinct microbiome when compared to healthy controls, which could potentially be employed to detect, identify, and treat cancer. Manipulation of the microbiome either via supplementation of single strains, bacterial consortia, fecal microbiota transfer or the use of pre- and probiotics has been suggested as therapeutic approach to directly target tumor growth or to enhance the efficacy of current state-of-the-art treatment options. Key Messages (1) Bacteria have a tremendous impact on anti-cancer immune responses. (2) Cancer patients harbor a distinct microbiome when compared to healthy controls. (3) The microbiome seems to be cancer-type specific. (4) Exploitation of bacteria to promote anti-tumor therapy is a novel, very promising venue in cancer treatment.
Collapse
Affiliation(s)
| | - Michael Scharl
- Department for Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
34
|
Kipchumba SK, Njuguna FM, Nyandiko WM. Bacterial Isolates and Characteristics of Children With Febrile Neutropenia on Treatment for Cancer at a Tertiary Hospital in Western Kenya. JCO Glob Oncol 2024; 10:e2300313. [PMID: 38301180 PMCID: PMC10846791 DOI: 10.1200/go.23.00313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/28/2023] [Accepted: 11/21/2023] [Indexed: 02/03/2024] Open
Abstract
PURPOSE This study aimed to identify the patient characteristics of children with febrile neutropenia, the associated bacterial organisms, and their sensitivity patterns. MATERIALS AND METHODS A descriptive cross-sectional study was conducted at the Moi Teaching and Referral Hospital (MTRH) pediatric oncology ward, from June 2021 to April 2022. A total of 110 children who developed fever and neutropenia during chemotherapy were enrolled. Blood samples for culture were collected aseptically. Patient characteristics were presented in frequency tables. Antimicrobial sensitivity patterns were plotted in tables against the bacterial isolates cultured. Chi-square/Fisher's exact test was used to determine any association between patient characteristics, bacterial growth, and antimicrobial sensitivity. RESULTS The majority (n = 66; 60%) were males. The median age was 6.3 years (standard deviation, 3.7). The majority of patients 71 (64.5%) had hematologic malignancies, the most common being AML. There was a significant association between severity of neutropenia and hematologic malignancies (P = .028). In total, 31/110 (28.2%) blood cultures were positive for bacterial growth. Gram-positive bacteria were more frequent (n = 20; 58.1%). The most common organism was Escherichia coli (n = 6; 18.2%), followed by Staphylococcus aureus (n = 5; 15.2%). All the isolates were sensitive to linezolid and vancomycin and also showed good sensitivity toward meropenem (n = 10/11; 90.9%). High resistance to cephalosporins was noted with ceftriaxone (n = 5/6; 83.3%), cefepime (n = 4/7; 57.1%), and ceftazidime (n = 3/4; 75%). CONCLUSION The most common malignancy associated with febrile neutropenia was AML. Gram-positive bacteria were the most common isolates. There was high resistance to cephalosporins.
Collapse
Affiliation(s)
- Samuel Kipkemoi Kipchumba
- Department of Child Health and Paediatrics, Moi University College of Health Sciences, Eldoret, Kenya
| | - Festus Muigai Njuguna
- Department of Child Health and Paediatrics, Moi University College of Health Sciences, Eldoret, Kenya
- Academic Model Providing Access to Healthcare, Eldoret, Kenya
| | - Winstone Mokaya Nyandiko
- Department of Child Health and Paediatrics, Moi University College of Health Sciences, Eldoret, Kenya
- Academic Model Providing Access to Healthcare, Eldoret, Kenya
| |
Collapse
|
35
|
Souza RO, Miranda VC, Quintanilha MF, Gallotti B, Oliveira SRM, Silva JL, Alvarez-Leite JI, Jesus LCL, Azevedo V, Vital KD, Fernandes SOA, Cardoso VN, Ferreira E, Nicoli JR, Martins FS. Evaluation of the Treatment with Akkermansia muciniphila BAA-835 of Chemotherapy-induced Mucositis in Mice. Probiotics Antimicrob Proteins 2024; 16:275-292. [PMID: 36652108 DOI: 10.1007/s12602-023-10040-2] [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: 01/05/2023] [Indexed: 01/19/2023]
Abstract
Mucositis is a high-incidence side effect in cancer patients undergoing chemotherapy. Next-generation probiotics are emerging as new therapeutic tools for managing various disorders. Studies have demonstrated the potential of Akkermansia muciniphila to increase the efficiency of anticancer treatment and to mitigate mucositis. Due to the beneficial effect of A. muciniphila on the host, we evaluated the dose-response, the microorganism viability, and the treatment protocol of A. muciniphila BAA-835 in a murine model of chemotherapy-induced mucositis. Female Balb/c mice were divided into groups that received either sterile 0.9% saline or A. muciniphila by gavage. Mucositis was induced using a single intraperitoneal injection of 5-fluorouracil. The animals were euthanized three days after the induction of mucositis, and tissue and blood were collected for analysis. Prevention of weight loss and small intestine shortening and reduction of neutrophil and eosinophil influx were observed when animals were pretreated with viable A. muciniphila at 1010 colony-forming units per mL (CFU/mL). The A. muciniphila improved mucosal damage by preserving tissue architecture and increasing villus height and goblet cell number. It also improved the integrity of the epithelial barrier, decreasing intestinal permeability and bacterial translocation. In addition, the treatment prevented the expansion of Enterobacteriaceae. The immunological parameters were also improved by decreasing the expression of pro-inflammatory cytokines (IL6, IL1β, and TNF) and increasing IL10. In conclusion, pretreatment with 1010 CFU/mL of viable A. muciniphila effectively controlled inflammation, protected the intestinal mucosa and the epithelial barrier, and prevented Enterobacteriaceae expansion in treated mice.
Collapse
Affiliation(s)
- Ramon O Souza
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Vivian C Miranda
- 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
| | - Bruno Gallotti
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Samantha R M Oliveira
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Janayne L Silva
- Departamento de Bioquímica E Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Jacqueline I Alvarez-Leite
- Departamento de Bioquímica E Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Luís C L Jesus
- Departamento de Genética, Ecologia E Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Vasco Azevedo
- Departamento de Genética, Ecologia E Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Kátia 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
| | - Enio Ferreira
- Departamento de Patologia Geral, Instituto de Ciências Biológicas, 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
| | - Flaviano S Martins
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil.
| |
Collapse
|
36
|
Bianchi MG, Chiu M, Taurino G, Bergamaschi E, Turroni F, Mancabelli L, Longhi G, Ventura M, Bussolati O. Amorphous silica nanoparticles and the human gut microbiota: a relationship with multiple implications. J Nanobiotechnology 2024; 22:45. [PMID: 38291460 PMCID: PMC10826219 DOI: 10.1186/s12951-024-02305-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 01/23/2024] [Indexed: 02/01/2024] Open
Abstract
Amorphous silica nanoparticles (ASNP) are among the nanomaterials that are produced in large quantities. ASNP have been present for a long time in several fast-moving consumer products, several of which imply exposure of the gastrointestinal tract, such as toothpastes, food additives, drug excipients, and carriers. Consolidated use and experimental evidence have consistently pointed to the very low acute toxicity and limited absorption of ASNP. However, slow absorption implies prolonged exposure of the intestinal epithelium to ASNP, with documented effects on intestinal permeability and immune gut homeostasis. These effects could explain the hepatic toxicity observed after oral administration of ASNP in animals. More recently, the role of microbiota in these and other ASNP effects has attracted increasing interest in parallel with the recognition of the role of microbiota in a variety of conditions. Although evidence for nanomaterial effects on microbiota is particularly abundant for materials endowed with bactericidal activities, a growing body of recent experimental data indicates that ASNPs also modify microbiota. The implications of these effects are recounted in this contribution, along with a discussion of the more important open issues and recommendations for future research.
Collapse
Affiliation(s)
- Massimiliano G Bianchi
- Lab. of General Pathology, Dept. of Medicine and Surgery, University of Parma, Parma, Italy.
- Interdepartmental Research Centre "Microbiome Research Hub", University of Parma, Parma, Italy.
| | - Martina Chiu
- Lab. of General Pathology, Dept. of Medicine and Surgery, University of Parma, Parma, Italy
| | - Giuseppe Taurino
- Lab. of General Pathology, Dept. of Medicine and Surgery, University of Parma, Parma, Italy
- Interdepartmental Research Centre "Microbiome Research Hub", University of Parma, Parma, Italy
| | - Enrico Bergamaschi
- Department of Public Health Sciences and Paediatrics, University of Turin, Turin, Italy
| | - Francesca Turroni
- Interdepartmental Research Centre "Microbiome Research Hub", University of Parma, Parma, Italy
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
| | - Leonardo Mancabelli
- Interdepartmental Research Centre "Microbiome Research Hub", University of Parma, Parma, Italy
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
| | - Giulia Longhi
- Interdepartmental Research Centre "Microbiome Research Hub", University of Parma, Parma, Italy
| | - Marco Ventura
- Interdepartmental Research Centre "Microbiome Research Hub", University of Parma, Parma, Italy
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
| | - Ovidio Bussolati
- Lab. of General Pathology, Dept. of Medicine and Surgery, University of Parma, Parma, Italy
- Interdepartmental Research Centre "Microbiome Research Hub", University of Parma, Parma, Italy
| |
Collapse
|
37
|
Okolo O, Honzel E, Britton WR, Yu VX, Flashner S, Martin C, Nakagawa H, Parikh AS. Experimental Modeling of Host-Bacterial Interactions in Head and Neck Squamous Cell Carcinoma. Cancers (Basel) 2023; 15:5810. [PMID: 38136355 PMCID: PMC10742111 DOI: 10.3390/cancers15245810] [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: 11/07/2023] [Accepted: 12/05/2023] [Indexed: 12/24/2023] Open
Abstract
The microscopic species colonizing the human body, collectively referred to as the microbiome, play a crucial role in the maintenance of tissue homeostasis, immunity, and the development of disease. There is evidence to suggest associations between alterations in the microbiome and the development of head and neck squamous cell carcinomas (HNSCC). The use of two-dimensional (2D) modeling systems has made significant strides in uncovering the role of microbes in carcinogenesis; however, direct mechanistic links remain in their infancy. Patient-derived three-dimensional (3D) HNSCC organoid and organotypic models have recently been described. Compared to 2D models, 3D organoid culture systems effectively capture the genetic and epigenetic features of parent tissue in a patient-specific manner and may offer a more nuanced understanding of the role of host-microbe responses in carcinogenesis. This review provides a topical literature review assessing the current state of the field investigating the role of the microbiome in HNSCC; including in vivo and in vitro modeling methods that may be used to characterize microbiome-epithelial interactions.
Collapse
Affiliation(s)
- Ogoegbunam Okolo
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY 10027, USA; (O.O.); (W.R.B.); (V.X.Y.); (S.F.); (C.M.); (H.N.)
- Columbia Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10027, USA;
| | - Emily Honzel
- Columbia Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10027, USA;
| | - William R. Britton
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY 10027, USA; (O.O.); (W.R.B.); (V.X.Y.); (S.F.); (C.M.); (H.N.)
- Columbia Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10027, USA;
| | - Victoria X. Yu
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY 10027, USA; (O.O.); (W.R.B.); (V.X.Y.); (S.F.); (C.M.); (H.N.)
- Department of Otolaryngology-Head and Neck Surgery, Columbia University, New York, NY 10027, USA
| | - Samuel Flashner
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY 10027, USA; (O.O.); (W.R.B.); (V.X.Y.); (S.F.); (C.M.); (H.N.)
| | - Cecilia Martin
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY 10027, USA; (O.O.); (W.R.B.); (V.X.Y.); (S.F.); (C.M.); (H.N.)
- Organoid and Cell Culture Core, Columbia University Digestive and Liver Diseases Research Center, Columbia University, New York, NY 10027, USA
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University, New York, NY 10027, USA
| | - Hiroshi Nakagawa
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY 10027, USA; (O.O.); (W.R.B.); (V.X.Y.); (S.F.); (C.M.); (H.N.)
- Organoid and Cell Culture Core, Columbia University Digestive and Liver Diseases Research Center, Columbia University, New York, NY 10027, USA
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University, New York, NY 10027, USA
| | - Anuraag S. Parikh
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY 10027, USA; (O.O.); (W.R.B.); (V.X.Y.); (S.F.); (C.M.); (H.N.)
- Department of Otolaryngology-Head and Neck Surgery, Columbia University, New York, NY 10027, USA
| |
Collapse
|
38
|
Wu WQ, Zhang YQ, Xu J, Tang ZX, Li SJ, Wei XY, Li L, Wu HQ, Ma X, Liu JS, Wu DP, Wu XJ. Risk Factors for Carbapenem-Resistant Enterobacteriaceae Colonization and the Effect on Clinical Outcomes and Prognosis in Allogeneic Hematopoietic Stem Cell Transplanted Patients. Infect Drug Resist 2023; 16:6821-6831. [PMID: 37904832 PMCID: PMC10613414 DOI: 10.2147/idr.s424048] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 09/14/2023] [Indexed: 11/01/2023] Open
Abstract
Purpose The current study assesses which are the main risk factors, clinical outcome and prognosis following the colonization of CRE in patients that underwent allo-HSCT. Patients and Methods A total of 343 patients subjected to allo-HSCT in the period comprised between June 2021 and June 2022 were enrolled in this retrospective study. The CRE colonization was diagnosed by clinical history and routine microbial culture of perirectal swab. In this regard, a clinical prediction model was designed based on independent risk factors underlying the pre-transplantation CRE colonization using a backward stepwise logistic regression, followed by the evaluation of its discrimination and calibration efficacies, along with clinical usefulness. Furthermore, univariate and multivariate Cox regression analyses were then conducted to assess the risk factors for post-transplantation clinical outcomes. Results Out of 343 patients enrolled in this study, 135 (39.3%) reported CRE colonization. The independent risk factor variables for CRE colonization were incorporated into the nomogram to build a prediction model, which showed an area under the curve of 0.767 (95% CI: 0.716-0.818), and well-fitted calibration curves (χ2 = 1.737, P = 0.9788). The patients with CRE colonization reported a significantly lower platelet engraftment rate with a higher risk of post-transplantation BSI when compared with the non-CRE colonization group (P = 0.02 and P < 0.001; respectively). The non-relapse mortality (NRM) value was higher in the CRE patients (P < 0.05), consistently with a survival probability that was thus significantly lower for the same timeframe (P < 0.05). Conclusion A reliable clinical prediction model for pre-transplantation CRE colonization was developed that demonstrated that the CRE colonization negatively affects platelet engraftment and survival outcomes following allo-HSCT.
Collapse
Affiliation(s)
- Wen-Qi Wu
- Department of Hematology, the First Affiliated Hospital of Soochow University, Suzhou, People’s Republic of China
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, Suzhou, People’s Republic of China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, People’s Republic of China
| | - Yu-Qi Zhang
- Department of Hematology, the First Affiliated Hospital of Soochow University, Suzhou, People’s Republic of China
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, Suzhou, People’s Republic of China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, People’s Republic of China
| | - Jie Xu
- Department of Hematology, the First Affiliated Hospital of Soochow University, Suzhou, People’s Republic of China
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, Suzhou, People’s Republic of China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, People’s Republic of China
| | - Zai-Xiang Tang
- Department of Epidemiology and Statistics, School of Public Health, Faculty of Medicine, Soochow University, Suzhou, People’s Republic of China
| | - Shi-Jia Li
- Department of Hematology, the First Affiliated Hospital of Soochow University, Suzhou, People’s Republic of China
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, Suzhou, People’s Republic of China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, People’s Republic of China
| | - Xi-Ya Wei
- Department of Hematology, the First Affiliated Hospital of Soochow University, Suzhou, People’s Republic of China
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, Suzhou, People’s Republic of China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, People’s Republic of China
| | - Ling Li
- Department of Hematology, the First Affiliated Hospital of Soochow University, Suzhou, People’s Republic of China
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, Suzhou, People’s Republic of China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, People’s Republic of China
| | - He-Qing Wu
- Department of Hematology, the First Affiliated Hospital of Soochow University, Suzhou, People’s Republic of China
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, Suzhou, People’s Republic of China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, People’s Republic of China
| | - Xiao Ma
- Department of Hematology, the First Affiliated Hospital of Soochow University, Suzhou, People’s Republic of China
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, Suzhou, People’s Republic of China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, People’s Republic of China
| | - Ji-Sheng Liu
- Department of Hematology, the First Affiliated Hospital of Soochow University, Suzhou, People’s Republic of China
- Department of Otolaryngology Head and Neck Surgery, the First Affiliated Hospital of Soochow University, Suzhou, People’s Republic of China
| | - De-Pei Wu
- Department of Hematology, the First Affiliated Hospital of Soochow University, Suzhou, People’s Republic of China
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, Suzhou, People’s Republic of China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, People’s Republic of China
| | - Xiao-Jin Wu
- Department of Hematology, the First Affiliated Hospital of Soochow University, Suzhou, People’s Republic of China
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, Suzhou, People’s Republic of China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, People’s Republic of China
| |
Collapse
|
39
|
Wang Y, Zhang X, Tang G, Deng P, Qin Y, Han J, Wang S, Sun X, Li D, Chen Z. The causal relationship between gut microbiota and bone mineral density: a Mendelian randomization study. Front Microbiol 2023; 14:1268935. [PMID: 37937216 PMCID: PMC10625988 DOI: 10.3389/fmicb.2023.1268935] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 10/09/2023] [Indexed: 11/09/2023] Open
Abstract
Background The gut microbiota has emerged as an intriguing and potentially influential factor in regulating bone health. However, the causal effect of the gut microbiota on bone mineral density (BMD) appears to differ throughout various life stages. Methods We conducted a Mendelian randomization (MR) analysis to investigate the potential causal relationship between gut microbiota and BMD in five distinct age groups: 0-15, 15-30, 30-45, 45-60, and 60 years and older. The analysis employed three different methods, namely MR-Egger, weighted median, and Inverse-variance weighting, to ensure the robustness of our findings, a series of sensitivity analyses were also conducted, such as horizontal pleiotropy tests, heterogeneity tests, and leave-one-out sensitivity tests. Results In the age group of 0-15 years, Eubacterium_fissicatena_group and Eubacterium_hallii_group were identified as risk factors for BMD. During the 15-30 age group, Phascolarctobacterium, Roseburia, and Ruminococcaceae_UCG_003 were found to be protective factors for BMD. In the 30-45 age group, Lachnospira genus demonstrated a protective effect on BMD, while Barnesiella and Lactococcus were identified as risk factors for BMD. Moving on to the 45-60 age group, Eubacterium_ventriosum_group, Lachnospiraceae_UCG_004, and Subdoligranulum were observed to be protective factors for BMD, while Eubacterium_coprostanoligenes_group, Fusicatenibacter, and Lactococcus were associated with an increased risk of BMD. In individuals aged 60 years and older, Fusicatenibacter and Ruminococcaceae_UCG_002 were also noted as risk factors for BMD. Conversely, Eubacterium_ruminantium_group, Ruminococcus_gauvreauii_group, Alistipes, and Coprococcus_3 were found to be protective factors for BMD, whereas Barnesiella and Sellimonas were identified as risk factors for BMD. Conclusion A robust causal relationship between gut microbiota and bone mineral density (BMD) exists throughout all stages of life, with Firmicutes phylum being the primary group associated with BMD across age groups. Gut microbiota linked with BMD primarily belong to the Firmicutes phylum across age groups. The diversity of gut microbiota phyla associated with BMD depicts relatively stable patterns during the ages of 0-45 years. However, for individuals aged 45 years and above, there is an observed increase in the number of gut microbiota species linked with BMD, and by the age of 60 years, a trend toward an increase in the Bacteroidetes phylum categories is proposed.
Collapse
Affiliation(s)
- Ying Wang
- Beijing University of Chinese Medicine, Beijing, China
- Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, China
| | - Xuejian Zhang
- Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, China
| | - Guangjun Tang
- Beijing University of Chinese Medicine, Beijing, China
- Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, China
| | - Pin Deng
- Institute of Basic Theory of Traditional Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuyan Qin
- Beijing University of Chinese Medicine, Beijing, China
- Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, China
| | - Jinglu Han
- Beijing University of Chinese Medicine, Beijing, China
- Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, China
| | - Shulong Wang
- Beijing University of Chinese Medicine, Beijing, China
- Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, China
| | - Xiaojie Sun
- Beijing University of Chinese Medicine, Beijing, China
- Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, China
| | - Dongxiao Li
- Beijing University of Chinese Medicine, Beijing, China
- Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, China
| | - Zhaojun Chen
- Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, China
| |
Collapse
|
40
|
Corley C, McElroy T, Sridharan B, Trujillo M, Simmons P, Kandel S, Sykes DJ, Robeson MS, Allen AR. Physiological and cognitive changes after treatments of cyclophosphamide, methotrexate, and fluorouracil: implications of the gut microbiome and depressive-like behavior. Front Neurosci 2023; 17:1212791. [PMID: 37869506 PMCID: PMC10587567 DOI: 10.3389/fnins.2023.1212791] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 09/08/2023] [Indexed: 10/24/2023] Open
Abstract
Introduction Chemotherapy-induced cognitive impairment colloquially referred to as chemobrain is a poorly understood phenomenon affecting a highly variable proportion of patients with breast cancer. Here we investigate the association between anxiety and despair-like behaviors in mice treated with cyclophosphamide, methotrexate, and fluorouracil (CMF) along with host histological, proteomic, gene expression, and gut microbial responses. Methods Forced swim and sociability tests were used to evaluate depression and despair-like behaviors. The tandem mass tag (TMT) proteomics approach was used to assess changes in the neural protein network of the amygdala and hippocampus. The composition of gut microbiota was assessed through 16S rRNA gene sequencing. Finally, quantitative reverse transcription polymerase chain reaction (qRT-PCR) was used to evaluate changes in intestinal gap junction markers. Results and discussion We observed that CMF induced social and despair-like behavior in mice 96 hours following treatment. Proteomic analysis identified changes in various proteins related to progressive neurological disease, working memory deficit, primary anxiety disorder, and gene expression revealing increases in NMDA and AMPA receptors in both the hippocampus and the amygdala because of CMF treatment. These changes finally, we observed immediate changes in the microbial population after chemotherapy treatment, with a notable abundance of Muribaculaceae and Romboutsia which may contribute to changes seen in the gut.
Collapse
Affiliation(s)
- Christa Corley
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, United States
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Taylor McElroy
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, United States
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Bhavana Sridharan
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, United States
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Madison Trujillo
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, United States
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Pilar Simmons
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, United States
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Sangam Kandel
- Department of Bioinformatics, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | | | - Michael S. Robeson
- Department of Bioinformatics, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Antiño R. Allen
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, United States
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| |
Collapse
|
41
|
Yamaguchi T, Gaowa A, Park EJ, Tawara I, Shimaoka M. Recombinant soluble thrombomodulin attenuates cisplatin-induced intestinal injury by inhibiting intestinal epithelial cell-derived cytokine secretion. Mol Biol Rep 2023; 50:8459-8467. [PMID: 37632632 DOI: 10.1007/s11033-023-08762-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 08/16/2023] [Indexed: 08/28/2023]
Abstract
BACKGROUND Intestinal injury is one of the main side-effects of cisplatin chemotherapy, impairing the quality of life in patients with cancer. In this study, we investigated the protective effects of recombinant soluble thrombomodulin (rsTM), which is a potent anti-inflammatory agent, on cisplatin-induced intestinal injury. METHODS We first evaluated the effects of rsTM on intestinal injury caused by cisplatin in mice in vivo. Disease progression was monitored by analyzing loss of body weight and histological changes in intestinal tissue. We then investigated the effects of rsTM on mouse intestinal organoid formation and growth in vitro. Gene expression levels were analyzed by quantitative real-time polymerase chain reaction and Western blotting. RESULTS rsTM treatment significantly attenuated the loss of body weight, histological damage and gene expression levels of pro-inflammatory cytokines such as interleukin-6, tumor necrosis factor-α and high-mobility group box-1 in a cisplatin-treated mouse model. Furthermore, rsTM alleviated the inflammatory response and apoptosis in a cisplatin-treated intestinal epithelial organoid model. CONCLUSION rsTM suppresses cisplatin-induced intestinal epithelial cell-derived cytokine production and alleviates intestinal mucositis.
Collapse
Affiliation(s)
- Takanori Yamaguchi
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie, 514-8507, Japan
- Department of Hematology and Oncology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Arong Gaowa
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie, 514-8507, Japan.
| | - Eun Jeong Park
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie, 514-8507, Japan
| | - Isao Tawara
- Department of Hematology and Oncology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Motomu Shimaoka
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie, 514-8507, Japan
| |
Collapse
|
42
|
Wu Y, Zhang Y, Zhang W, Huang Y, Lu X, Shang L, Zhou Z, Chen X, Li S, Cheng S, Song Y. The tremendous clinical potential of the microbiota in the treatment of breast cancer: the next frontier. J Cancer Res Clin Oncol 2023; 149:12513-12534. [PMID: 37382675 DOI: 10.1007/s00432-023-05014-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 06/19/2023] [Indexed: 06/30/2023]
Abstract
Although significant advances have been made in the diagnosis and treatment of breast cancer (BC) in recent years, BC remains the most common cancer in women and one of the main causes of death among women worldwide. Currently, more than half of BC patients have no known risk factors, emphasizing the significance of identifying more tumor-related factors. Therefore, we urgently need to find new therapeutic strategies to improve prognosis. Increasing evidence demonstrates that the microbiota is present in a wider range of cancers beyond colorectal cancer. BC and breast tissues also have different types of microbiotas that play a key role in carcinogenesis and in modulating the efficacy of anticancer treatment, for instance, chemotherapy, radiotherapy, and immunotherapy. In recent years, studies have confirmed that the microbiota can be an important factor directly and/or indirectly affecting the occurrence, metastasis and treatment of BC by regulating different biological processes, such as estrogen metabolism, DNA damage, and bacterial metabolite production. Here, we review the different microbiota-focused studies associated with BC and explore the mechanisms of action of the microbiota in BC initiation and metastasis and its application in various therapeutic strategies. We found that the microbiota has vital clinical value in the diagnosis and treatment of BC and could be used as a biomarker for prognosis prediction. Therefore, modulation of the gut microbiota and its metabolites might be a potential target for prevention or therapy in BC.
Collapse
Affiliation(s)
- Yang Wu
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, 150 Haping Road, Nangang District, Harbin, 150081, China
| | - Yue Zhang
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Wenwen Zhang
- Department of Gynecology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yuanxi Huang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, 150 Haping Road, Nangang District, Harbin, 150081, China
| | - Xiangshi Lu
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, 150 Haping Road, Nangang District, Harbin, 150081, China
| | - Lingmin Shang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, 150 Haping Road, Nangang District, Harbin, 150081, China
| | - Zhaoyue Zhou
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, 150 Haping Road, Nangang District, Harbin, 150081, China
| | - Xiaolu Chen
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, 150 Haping Road, Nangang District, Harbin, 150081, China
| | - Shuhui Li
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, 150 Haping Road, Nangang District, Harbin, 150081, China
| | - Shaoqiang Cheng
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, 150 Haping Road, Nangang District, Harbin, 150081, China.
| | - Yanni Song
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, 150 Haping Road, Nangang District, Harbin, 150081, China.
| |
Collapse
|
43
|
Wang Z, Zhou Y, Luo A, Heng X, Liu J, Wang H, Chu W. Lactobacillus salivarius CPU-01 Ameliorates Temozolomide-Induced Intestinal Mucositis by Modulating Gut Microbiota, Maintaining Intestinal Barrier, and Blocking Pro-inflammatory Cytokines. Probiotics Antimicrob Proteins 2023; 15:1079-1091. [PMID: 35639268 DOI: 10.1007/s12602-022-09955-z] [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: 05/18/2022] [Indexed: 10/18/2022]
Abstract
Chemotherapy-induced intestinal mucositis is one of the major toxic side effects in the treatment of cancer patients. The purpose of this study is to screen lactic acid bacteria which could alleviate intestinal inflammation and damage induced by chemotherapeutic agents and explore the possible underlying mechanisms. Lactobacillus salivarius CPU-01 was selected from traditional Chinese fermented foods due to its protective effects on the toxicity of temozolomide in Caenorhabditis elegans. Eighteen ICR mice were randomly divided into 3 groups including control group, temozolomide-induced intestinal mucositis group, and temozolomide + L. salivarius CPU-01 group, and were used to investigate the effect of L. salivarius CPU-01 on chemotherapy-induced intestinal mucositis. It has been demonstrated that the administration of L. salivarius CPU-01 can prevent colon shortening and alleviate colon tissue damage caused by temozolomide-induced intestinal mucositis in mice. L. salivarius CPU-01 relieved the intestinal microbiota disorders caused by temozolomide and contributed to the growth of beneficial bacteria, such as Lactobacillus, Clostridia UCG - 014_norank, and Akkermansia. In vivo experiments also indicated that L. salivarius CPU-01 can suppress the level of temozolomide-induced pro-inflammatory cytokines in serum and mRNA expression in the small intestine tissues. It was also found that L. salivarius CPU-01 significantly increased the expressions of intestinal tight junction (TJ) proteins, ZO-1, and Occludin proteins in mice treated with temozolomide. These findings suggest that L. salivarius CPU-01 can ameliorate temozolomide-induced intestinal mucositis by modulating gut microbiota, blocking pro-inflammatory cytokines, and repairing the intestinal barrier. These findings suggest probiotics may serve as a potential alternative therapeutic strategy for the prevention of chemotherapy-induced intestinal mucositis in the future.
Collapse
Affiliation(s)
- Zheng Wang
- Department of Pharmaceutical Microbiology, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, China
| | - Yuhong Zhou
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Aoxiang Luo
- Department of Pharmaceutical Microbiology, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, China
| | - Xing Heng
- Department of Pharmaceutical Microbiology, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, China
| | - Jinqiu Liu
- Department of Pharmaceutical Microbiology, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, China
| | - Huafu Wang
- Lishui People's Hospital, the Sixth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
| | - Weihua Chu
- Department of Pharmaceutical Microbiology, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
44
|
Wang Y, Han W, Wang N, Han M, Ban M, Dai J, Dong Y, Sun T, Xu J. The role of microbiota in the development and treatment of gastric cancer. Front Oncol 2023; 13:1224669. [PMID: 37841431 PMCID: PMC10572359 DOI: 10.3389/fonc.2023.1224669] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 08/28/2023] [Indexed: 10/17/2023] Open
Abstract
The stomach was once considered a sterile organ until the discovery of Helicobacter pylori (HP). With the application of high-throughput sequencing technology and macrogenomics, researchers have identified fungi and fivemajor bacterial phyla within the stomachs of healthy individuals. These microbial communities exert regulatory influence over various physiological functions, including energy metabolism and immune responses. HP is a well-recognized risk factor for gastric cancer, significantly altering the stomach's native microecology. Currently, numerous studies are centered on the mechanisms by which HP contributes to gastric cancer development, primarily involving the CagA oncoprotein. However, aside from exogenous infections such as HP and EBV, certain endogenous dysbiosis can also lead to gastric cancer through multiple mechanisms. Additionally, gut microbiota and its metabolites significantly impact the development of gastric cancer. The role of microbial therapies, including diet, phages, probiotics and fecal microbiota transplantation, in treating gastric cancer should not be underestimated. This review aims to study the mechanisms involved in the roles of exogenous pathogen infection and endogenous microbiota dysbiosis in the development of gastric cancer. Also, we describe the application of microbiota therapy in the treatment and prognosis of gastric cancer.
Collapse
Affiliation(s)
- Yiwen Wang
- Department of Breast Medicine 1, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang, China
- Department of Pharmacology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang, China
| | - Wenjie Han
- Department of Breast Medicine 1, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang, China
- Department of Pharmacology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang, China
| | - Na Wang
- Department of Breast Medicine 1, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang, China
- Department of Pharmacology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang, China
| | - Mengzhen Han
- Department of Breast Medicine 1, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang, China
- Department of Pharmacology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang, China
| | - Meng Ban
- Department of Bioinformatics, Kanghui Biotechnology Co., Ltd., Shenyang, China
| | - Jianying Dai
- School of Bioengineering, Dalian University of Technology, Dalian, Liaoning, China
| | - Yuesheng Dong
- School of Bioengineering, Dalian University of Technology, Dalian, Liaoning, China
| | - Tao Sun
- Department of Breast Medicine 1, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang, China
- Department of Oncology Medicine, Key Laboratory of Liaoning Breast Cancer Research, Shenyang, Liaoning, China
| | - Junnan Xu
- Department of Breast Medicine 1, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang, China
- Department of Pharmacology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang, China
- Department of Oncology Medicine, Key Laboratory of Liaoning Breast Cancer Research, Shenyang, Liaoning, China
| |
Collapse
|
45
|
Margolis EB, Maron G, Sun Y, Dallas RH, Allison KJ, Ferrolino J, Ross HS, Davis AE, Jia Q, Turner P, Mackay V, Morin CE, Triplett BM, Klein EJ, Englund JA, Tang L, Hayden RT. Microbiota Predict Infections and Acute Graft-Versus-Host Disease After Pediatric Allogeneic Hematopoietic Stem Cell Transplantation. J Infect Dis 2023; 228:627-636. [PMID: 37249910 PMCID: PMC10469318 DOI: 10.1093/infdis/jiad190] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 04/19/2023] [Accepted: 05/27/2023] [Indexed: 05/31/2023] Open
Abstract
BACKGROUND Despite preventive measures, infections continue to pose significant risks to pediatric allogeneic hematopoietic cell transplantation (allo-HCT) recipients. The gut microbiota has been linked to clinical outcomes following adult allo-HCT. This study evaluated whether similar disruptions or differing microbiota patterns were associated with infection risk in pediatric allo-HCT. METHODS In a prospective observational study, fecal samples were obtained from 74 children before conditioning and upon neutrophil recovery. Microbiome signatures identified through sequencing were examined for their associations with infections or acute graft-versus-host disease (aGVHD) in the first-year post-HCT using Cox proportional hazards analysis. RESULTS Microbiome disruption in adults, did not predict infection risk in pediatric allo-HCT. Unique microbiota signatures were associated with different infections or aGVHD. A ratio of strict and facultative anaerobes (eg, Lachnoclostridium, Parabacteroides) prior to conditioning predicted bacteremia risk (Cox hazard ratio [HR], 3.89). A distinct ratio of oral (eg, Rothia, Veillonella) to intestinal anaerobes (eg, Anaerobutyricum, Romboutsia) at neutrophil recovery predicted likelihood of bacterial infections (Cox HR, 1.81) and viral enterocolitis (Cox HR, 1.96). CONCLUSIONS Interactions between medical interventions, pediatric hosts, and microbial communities contribute to microbiota signatures that predict infections. Further multicenter study is necessary to validate the generalizability of these ratios as biomarkers.
Collapse
Affiliation(s)
- Elisa B Margolis
- Department of Infectious Diseases, St Jude Children’s Research Hospital
- Department of Pediatrics, University of Tennessee Health Sciences Center
| | - Gabriela Maron
- Department of Infectious Diseases, St Jude Children’s Research Hospital
- Department of Pediatrics, University of Tennessee Health Sciences Center
| | - Yilun Sun
- Department of Biostatistics, St Jude Children’s Research Hospital
| | - Ronald H Dallas
- Department of Infectious Diseases, St Jude Children’s Research Hospital
| | - Kim J Allison
- Department of Infectious Diseases, St Jude Children’s Research Hospital
| | - Jose Ferrolino
- Department of Infectious Diseases, St Jude Children’s Research Hospital
| | - Hailey S Ross
- Department of Infectious Diseases, St Jude Children’s Research Hospital
| | - Amy E Davis
- Department of Infectious Diseases, St Jude Children’s Research Hospital
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Sciences Center, Memphis
| | - Qidong Jia
- Department of Infectious Diseases, St Jude Children’s Research Hospital
| | - Paige Turner
- Department of Infectious Diseases, St Jude Children’s Research Hospital
| | - Victoria Mackay
- Department of Infectious Diseases, St Jude Children’s Research Hospital
| | - Cara E Morin
- Division of Radiology and Medical Imaging, Cincinnati Children's Hospital, Ohio
| | - Brandon M Triplett
- Department of Bone Marrow Transplantation and Cellular Therapy, St Jude Children’s Research Hospital, Memphis, Tennessee
| | | | | | - Li Tang
- Department of Biostatistics, St Jude Children’s Research Hospital
| | | |
Collapse
|
46
|
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: 0.5] [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
|
47
|
Cazzaniga M, Zonzini GB, Di Pierro F, Palazzi CM, Cardinali M, Bertuccioli A. Influence of the microbiota on the effectiveness and toxicity of oncological therapies, with a focus on chemotherapy. Pathol Oncol Res 2023; 29:1611300. [PMID: 37593337 PMCID: PMC10427764 DOI: 10.3389/pore.2023.1611300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Accepted: 07/18/2023] [Indexed: 08/19/2023]
Abstract
Recent studies have highlighted a possible correlation between microbiota composition and the pathogenesis of various oncological diseases. Also, many bacterial groups are now directly or indirectly associated with the capability of stimulating or inhibiting carcinogenic pathways. However, little is known about the importance and impact of microbiota patterns related to the efficacy and toxicity of cancer treatments. We have recently begun to understand how oncological therapies and the microbiota are closely interconnected and could influence each other. Chemotherapy effectiveness, for example, appears to be strongly influenced by the presence of some microorganisms capable of modulating the pharmacokinetics and pharmacodynamics of the compounds used, thus varying the real response and therefore the efficacy of the oncological treatment. Similarly, chemotherapeutic agents can modulate the microbiota with variations that could facilitate or avoid the onset of important side effects. This finding has or could have considerable relevance as it is possible that our ability to modulate and modify the microbial structure before, during, and after treatment could influence all the clinical parameters related to pharmacological treatments and, eventually, the prognosis of the disease.
Collapse
Affiliation(s)
| | | | - Francesco Di Pierro
- Scientific & Research Department, Velleja Research, Milano, Italy
- Department of Medicine and Surgery, University of Insurbia, Varese, Italy
| | | | - Marco Cardinali
- Department of Internal Medicine, Infermi Hospital, Azienda Unità Sanitaria Locale Romagna, Rimini, Italy
| | | |
Collapse
|
48
|
Tavares LM, de Jesus LCL, Batista VL, Barroso FAL, Dos Santos Freitas A, Campos GM, Américo MF, da Silva TF, Coelho-Rocha ND, Belo GA, Drumond MM, Mancha-Agresti P, Vital KD, Fernandes SOA, Cardoso VN, Birbrair A, Ferreira E, Martins FS, Laguna JG, Azevedo V. Synergistic synbiotic containing fructooligosaccharides and Lactobacillus delbrueckii CIDCA 133 alleviates chemotherapy-induced intestinal mucositis in mice. World J Microbiol Biotechnol 2023; 39:235. [PMID: 37365380 DOI: 10.1007/s11274-023-03679-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 06/15/2023] [Indexed: 06/28/2023]
Abstract
Intestinal mucositis is a commonly reported side effect in oncology patients undergoing chemotherapy and radiotherapy. Probiotics, prebiotics, and synbiotics have been investigated as alternative therapeutic approaches against intestinal mucositis due to their well-known anti-inflammatory properties and health benefits to the host. Previous studies showed that the potential probiotic Lactobacillus delbrueckii CIDCA 133 and the prebiotic Fructooligosaccharides (FOS) alleviated the 5-Fluorouracil (5-FU) chemotherapy-induced intestinal mucosa damage. Based on these previous beneficial effects, this work evaluated the anti-inflammatory property of the synbiotic formulation containing L. delbrueckii CIDCA 133 and FOS in mice intestinal mucosa inflammation induced by 5-FU. This work showed that the synbiotic formulation was able to modulate inflammatory parameters, including reduction of cellular inflammatory infiltration, gene expression downregulation of Tlr2, Nfkb1, and Tnf, and upregulation of the immunoregulatory Il10 cytokine, thus protecting the intestinal mucosa from epithelial damage caused by the 5-FU. The synbiotic also improved the epithelial barrier function by upregulating mRNA transcript levels of the short chain fatty acid (SCFA)-associated GPR43 receptor and the occludin tight junction protein, with the subsequent reduction of paracellular intestinal permeability. The data obtained showed that this synbiotic formulation could be a promising adjuvant treatment to be explored against inflammatory damage caused by 5-FU chemotherapy.
Collapse
Affiliation(s)
- Laísa Macedo Tavares
- Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Luís Cláudio Lima de Jesus
- Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Viviane Lima Batista
- Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | | | - Andria Dos Santos Freitas
- Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Gabriela Munis Campos
- Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Monique Ferrary Américo
- Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Tales Fernando da Silva
- Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Nina Dias Coelho-Rocha
- Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Giovanna Angeli Belo
- Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Mariana Martins Drumond
- Federal Center for Technological Education of Minas Gerais, Department of Biological Sciences, Belo Horizonte, Brazil
- Federal Center for Technological Education of Minas Gerais, Materials Engineering Post- Graduation Program, Belo Horizonte, Brazil
| | - Pamela Mancha-Agresti
- Federal Center for Technological Education of Minas Gerais, Department of Biological Sciences, Belo Horizonte, Brazil
- Federal Center for Technological Education of Minas Gerais, Materials Engineering Post- Graduation Program, Belo Horizonte, Brazil
| | - Kátia Duarte Vital
- Department of Clinical and Toxicological Analysis, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | | | - Valbert Nascimento Cardoso
- Department of Clinical and Toxicological Analysis, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Alexander Birbrair
- Department of General Pathology, Federal University of Minas Gerais, Belo Horizonte, Brazil
- Department of Dermatology, University of Wisconsin-Madison, Madison, WI, USA
- Department of Radiology, Columbia University Medical Center, New York, NY, USA
| | - Enio Ferreira
- Department of General Pathology, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | | | - Juliana Guimarães Laguna
- Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Vasco Azevedo
- Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil.
| |
Collapse
|
49
|
Reuss JM, Alonso-Gamo L, Garcia-Aranda M, Reuss D, Albi M, Albi B, Vilaboa D, Vilaboa B. Oral Mucosa in Cancer Patients-Putting the Pieces Together: A Narrative Review and New Perspectives. Cancers (Basel) 2023; 15:3295. [PMID: 37444405 DOI: 10.3390/cancers15133295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 06/15/2023] [Accepted: 06/18/2023] [Indexed: 07/15/2023] Open
Abstract
The oral mucosa is a key player in cancer patients and during cancer treatment. The increasing prevalence of cancer and cancer-therapy-associated side effects are behind the major role that oral mucosa plays in oncological patients. Oral mucositis is a debilitating severe complication caused by the early toxicity of chemo and/or radiotherapy that can restrict treatment outcome possibilities, even challenging a patient's survival. It has been referred to as the most feared cancer treatment complication. Predictive variables as to who will be affected, and to what extent, are still unclear. Additionally, oral mucositis is one of the sources of the increasing economic burden of cancer, not only for patients and their families but also for institutions and governments. All efforts should be implemented in the search for new approaches to minimize the apparently ineluctable outburst of oral mucositis during cancer treatment. New perspectives derived from different approaches to explaining the interrelation between oral mucositis and the oral microbiome or the similarities with genitourinary mucosa may help elucidate the biomolecular pathways and mechanisms behind oral mucosa cancer-therapy-related toxicity, and what is more important is its management in order to minimize treatment side effects and provide enhanced cancer support.
Collapse
Affiliation(s)
- Jose Manuel Reuss
- Department of Postgraduate Prosthodontics, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Laura Alonso-Gamo
- Department of Pediatrics, Hospital Infanta Cristina, 28981 Madrid, Spain
| | - Mariola Garcia-Aranda
- Centro Integral Oncológico Clara Campal, Department of Oncologic Radiotherapy, Hospital Universitario Sanchinarro, 28050 Madrid, Spain
| | - Debora Reuss
- Lecturer Dental School, Universidad San Pablo CEU, 28003 Madrid, Spain
| | - Manuel Albi
- Department of Gynecology and Obstetrics, Quironsalud Group Public Hospitals, 28223 Madrid, Spain
| | - Beatriz Albi
- Department of Gynecology and Obstetrics, Hospital Universitario Fundación Jiménez Díaz, 28040 Madrid, Spain
| | - Debora Vilaboa
- Aesthetic Dentistry Department, Universidad San Pablo CEU, 28003 Madrid, Spain
| | | |
Collapse
|
50
|
da Silva KS, Abboud KY, Schiebel CS, de Oliveira NMT, Bueno LR, de Mello Braga LLV, da Silveira BC, Santos IWFD, Gomes EDS, Gois MB, Cordeiro LMC, Maria Ferreira D. Polysaccharides from Passion Fruit Peels: From an Agroindustrial By-Product to a Viable Option for 5-FU-Induced Intestinal Damage. Pharmaceuticals (Basel) 2023; 16:912. [PMID: 37513823 PMCID: PMC10383750 DOI: 10.3390/ph16070912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 06/14/2023] [Accepted: 06/16/2023] [Indexed: 07/30/2023] Open
Abstract
Gastrointestinal mucositis is a serious and dose-limiting toxic side effect of oncologic treatment. Interruption of cancer treatment due to gastrointestinal mucositis leads to a significant decrease in cure rates and consequently to the deterioration of a patient's quality of life. Natural polysaccharides show a variety of beneficial effects, including a gastroprotective effect. Treatment with soluble dietary fiber (SDF) from yellow passion fruit (Passiflora edulis) biomass residues protected the gastric and intestinal mucosa in models of gastrointestinal injury. In this study, we investigated the protective therapeutic effect of SDF on 5-FU-induced mucositis in male and female mice. Oral treatment of the animals with SDF did not prevent weight loss but reduced the disease activity index and preserved normal intestinal function by alleviating diarrhea and altered gastrointestinal transit. SDF preserved the length of the colon and histological damage caused by 5-FU. SDF significantly restored the oxidative stress and inflammation in the intestine and the enlargement and swelling of the spleen induced by 5-FU. In conclusion, SDF may be a promising adjuvant strategy for the prevention and treatment of intestinal mucositis induced by 5-FU.
Collapse
Affiliation(s)
- Karien Sauruk da Silva
- Faculdades Pequeno Príncipe, Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Curitiba 80250-200, Brazil
- Instituto de Pesquisa Pelé Pequeno Príncipe, Av. Silva Jardim No 1532, Curitiba 80250-200, Brazil
| | - Kahlile Youssef Abboud
- Department of Biochemistry and Molecular Biology, Federal University of Paraná, Curitiba 81531-980, Brazil
| | - Carolina Silva Schiebel
- Faculdades Pequeno Príncipe, Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Curitiba 80250-200, Brazil
- Instituto de Pesquisa Pelé Pequeno Príncipe, Av. Silva Jardim No 1532, Curitiba 80250-200, Brazil
| | - Natalia Mulinari Turin de Oliveira
- Faculdades Pequeno Príncipe, Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Curitiba 80250-200, Brazil
- Instituto de Pesquisa Pelé Pequeno Príncipe, Av. Silva Jardim No 1532, Curitiba 80250-200, Brazil
| | - Laryssa Regis Bueno
- Faculdades Pequeno Príncipe, Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Curitiba 80250-200, Brazil
- Instituto de Pesquisa Pelé Pequeno Príncipe, Av. Silva Jardim No 1532, Curitiba 80250-200, Brazil
| | - Lara Luisa Valerio de Mello Braga
- Faculdades Pequeno Príncipe, Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Curitiba 80250-200, Brazil
- Instituto de Pesquisa Pelé Pequeno Príncipe, Av. Silva Jardim No 1532, Curitiba 80250-200, Brazil
| | - Bruna Carla da Silveira
- Faculdades Pequeno Príncipe, Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Curitiba 80250-200, Brazil
- Instituto de Pesquisa Pelé Pequeno Príncipe, Av. Silva Jardim No 1532, Curitiba 80250-200, Brazil
| | - Isabella Wzorek França Dos Santos
- Faculdades Pequeno Príncipe, Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Curitiba 80250-200, Brazil
- Instituto de Pesquisa Pelé Pequeno Príncipe, Av. Silva Jardim No 1532, Curitiba 80250-200, Brazil
| | - Everton Dos Santos Gomes
- Programa de Pós-Graduação em Imunologia, Universidade Federal da Bahia, Salvador 40231-300, Brazil
- Programa de Pós-Graduação em Biociências e Saúde, Universidade Federal de Rondonópolis, Rondonópolis 78736-900, Brazil
| | - Marcelo Biondaro Gois
- Programa de Pós-Graduação em Imunologia, Universidade Federal da Bahia, Salvador 40231-300, Brazil
- Programa de Pós-Graduação em Biociências e Saúde, Universidade Federal de Rondonópolis, Rondonópolis 78736-900, Brazil
| | | | - Daniele Maria Ferreira
- Faculdades Pequeno Príncipe, Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Curitiba 80250-200, Brazil
- Instituto de Pesquisa Pelé Pequeno Príncipe, Av. Silva Jardim No 1532, Curitiba 80250-200, Brazil
| |
Collapse
|