1
|
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: 0] [Impact Index Per Article: 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
|
2
|
Li X, Hu B, Zheng J, Pan Z, Cai Y, Zhao M, Jin X, Li ZQ. Probiotics Alleviate Chemotherapy-Associated Intestinal Mucosal Injury via the TLR4-NFκB Signaling Pathway. Drug Des Devel Ther 2023; 17:2183-2192. [PMID: 37521036 PMCID: PMC10386857 DOI: 10.2147/dddt.s403087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 07/17/2023] [Indexed: 08/01/2023] Open
Abstract
Introduction Temozolomide (TMZ) induces intestinal mucosa injury that cannot be fully counteracted by supportive treatment. Probiotics regulate gut microbial composition and the host immune system and may alleviate this side effect. We aimed to investigate the potential and mechanism of Lactobacillus rhamnosus GG (LGG) in relieving intestinal mucosal injury induced by TMZ. Methods Glioblastoma mice were divided into four groups: CON (control), LGG (109 CFU/mL, treated for 7 days), TMZ (50 mg/kg·d, treated for 5 days), LGG+TMZ (LGG for 7 days and TMZ subsequently for 5 days). Body weight, food intake, and fecal pH were recorded. Intestinal tissue samples were collected 1 day after the end of TMZ treatment. Degree of damage to intestine, expression of IL1β, IL6, TNFα, and IL10 in jejunum were determined. Levels of tight-junction proteins (ZO1, occludin), TLR4, IKKβ, IκBα, and P65 with their phosphorylation in jejunum were measured. Results Decreases in body weight, food intake, spleen index in the TMZ group were mitigated in the LGG+TMZ group, and the degree of intestinal shortening and damage to jejunum villus were also alleviated. The expression of tight-junction proteins in the LGG+TMZ group was significantly greater than that in the TMZ group. IκBα in intestinal tissue significantly decreased in the TMZ group, phos-IKKβ and phos-P65 increased compared to the CON group, and LGG reversed such changes in IκBα and phos-P65 in the LGG+TMZ group. Intestinal inflammatory cytokines were significantly increased in the TMZ group, but lower in the LGG+TMZ group. Moreover, expression of TLR4 in LGG group was significantly lower than that in the CON group. LGG inhibited the rise of TLR4 after TMZ in the LGG+TMZ group compared to the TMZ group. Conclusion LGG inhibits the activation of the TLR4-NFκB pathway and alleviates intestinal mucosal inflammation induced by TMZ, thereby protect the jejunum villi and mucosal physical barrier.
Collapse
Affiliation(s)
- Xiaochong Li
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, People’s Republic of China
| | - Bowen Hu
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, People’s Republic of China
| | - Jiachen Zheng
- Emergency Center, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, People’s Republic of China
- The Second Clinical School, Wuhan University, Wuhan, Hubei, 430071, People’s Republic of China
| | - Zhiyong Pan
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, People’s Republic of China
| | - Yuxiang Cai
- Department of Pathology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, People’s Republic of China
| | - Mingjuan Zhao
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, People’s Republic of China
| | - Xiaoqing Jin
- Emergency Center, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, People’s Republic of China
| | - Zhi-Qiang Li
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, People’s Republic of China
| |
Collapse
|
3
|
Lu DX, Liu F, Wu H, Liu HX, Chen BY, Yan J, Lu Y, Sun ZG. Wumei pills attenuates 5-fluorouracil-induced intestinal mucositis through Toll-like receptor 4/myeloid differentiation factor 88/nuclear factor-κB pathway and microbiota regulation. World J Gastroenterol 2022; 28:4574-4599. [PMID: 36157934 PMCID: PMC9476879 DOI: 10.3748/wjg.v28.i32.4574] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 01/05/2022] [Accepted: 07/18/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Radiotherapy and chemotherapy can kill tumor cells and improve the survival rate of cancer patients. However, they can also damage normal cells and cause serious intestinal toxicity, leading to gastrointestinal mucositis[1]. Traditional Chinese medicine is effective in improving the side effects of chemotherapy. Wumei pills (WMP) was originally documented in the Treatise on Exogenous Febrile Diseases. It has a significant effect on chronic diarrhea and other gastrointestinal diseases, but it is not clear whether it affects chemotherapy-induced intestinal mucositis (CIM).
AIM To explore the potential mechanism of WMP in the treatment of CIM through experimental research.
METHODS We used an intraperitoneal injection of 5-fluorouracil (5-Fu) to establish a CIM mouse model and an oral gavage of WMP decoction (11325 and 22650 mg/kg) to evaluate the efficacy of WMP in CIM. We evaluated the effect of WMP on CIM by observing the general conditions of the mice (body weight, food intake, spleen weight, diarrhea score, and hematoxylin and eosin stained tissues). The expression of tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), IL-1β, and myeloperoxidase (MPO), as well as the Toll-like receptor 4/myeloid differentiation factor 88/nuclear factor-κB (TLR4/MyD88/NF-κB) signaling pathway proteins and tight junction proteins (zonula occludens-1, claudin-1, E-cadherin, and mucin-2) was determined. Furthermore, intestinal permeability, intestinal flora, and the levels of short-chain fatty acids (SCFA) were also assessed.
RESULTS WMP effectively improved the body weight, spleen weight, food intake, diarrhea score, and inflammatory status of the mice with intestinal mucositis, which preliminarily confirmed the efficacy of WMP in CIM. Further experiments showed that in addition to reducing the levels of TNF-α, IL-1β, IL-6, and MPO and inhibiting the expression of the TLR4/MyD88/NF-κB pathway proteins, WMP also repaired the integrity of the mucosal barrier of mice, regulated the intestinal flora, and increased the levels of SCFA (such as butyric acid).
CONCLUSION WMP can play a therapeutic role in CIM by alleviating inflammation, restoring the mucosal barrier, and regulating gut microbiota.
Collapse
Affiliation(s)
- Dong-Xue Lu
- Department of Nutrition, Acupuncture and Moxibustion and Massage College & Health Preservation and Rehabilitation College, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China
| | - Feng Liu
- Department of Orthopaedics, Nanjing Pukou District Chinese Medicine Hospital, Nanjing 210000, Jiangsu Province, China
| | - Hua Wu
- Department of Nutrition, Acupuncture and Moxibustion and Massage College & Health Preservation and Rehabilitation College, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China
| | - Hai-Xia Liu
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210000, Jiangsu Province, China
| | - Bing-Yu Chen
- Department of Geriatrics, The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou 451150, Henan Province, China
| | - Jing Yan
- Key Laboratory for Metabolic Diseases in Chinese Medicine, The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China
| | - Yin Lu
- Key Pharmacology Laboratory of Jiangsu Province, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China
| | - Zhi-Guang Sun
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China
| |
Collapse
|
4
|
Pagani IS, Poudel G, Wardill HR. A Gut Instinct on Leukaemia: A New Mechanistic Hypothesis for Microbiota-Immune Crosstalk in Disease Progression and Relapse. Microorganisms 2022; 10:microorganisms10040713. [PMID: 35456764 PMCID: PMC9029211 DOI: 10.3390/microorganisms10040713] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/15/2022] [Accepted: 03/23/2022] [Indexed: 02/05/2023] Open
Abstract
Despite significant advances in the treatment of Chronic Myeloid and Acute Lymphoblastic Leukaemia (CML and ALL, respectively), disease progression and relapse remain a major problem. Growing evidence indicates the loss of immune surveillance of residual leukaemic cells as one of the main contributors to disease recurrence and relapse. More recently, there was an appreciation for how the host’s gut microbiota predisposes to relapse given its potent immunomodulatory capacity. This is especially compelling in haematological malignancies where changes in the gut microbiota have been identified after treatment, persisting in some patients for years after the completion of treatment. In this hypothesis-generating review, we discuss the interaction between the gut microbiota and treatment responses, and its capacity to influence the risk of relapse in both CML and ALL We hypothesize that the gut microbiota contributes to the creation of an immunosuppressive microenvironment, which promotes tumour progression and relapse.
Collapse
Affiliation(s)
- Ilaria S. Pagani
- Cancer Program, Precision Medicine Theme, South Australian Health & Medical Research Institute, Adelaide 5000, Australia; (G.P.); (H.R.W.)
- Faculty of Health and Medical Sciences, School of Medicine, University of Adelaide, Adelaide 5000, Australia
- Correspondence:
| | - Govinda Poudel
- Cancer Program, Precision Medicine Theme, South Australian Health & Medical Research Institute, Adelaide 5000, Australia; (G.P.); (H.R.W.)
- Faculty of Health and Medical Sciences, School of Medicine, University of Adelaide, Adelaide 5000, Australia
| | - Hannah R. Wardill
- Cancer Program, Precision Medicine Theme, South Australian Health & Medical Research Institute, Adelaide 5000, Australia; (G.P.); (H.R.W.)
- Faculty of Health and Medical Sciences, School of Biomedicine, University of Adelaide, Adelaide 5000, Australia
| |
Collapse
|
5
|
Jugan MC, Wouda RM, Higginbotham ML. Preliminary evaluation of probiotic effects on gastrointestinal signs in dogs with multicentric lymphoma undergoing multi-agent chemotherapy: A randomised, placebo-controlled study. Vet Rec Open 2021; 8:e2. [PMID: 33981436 PMCID: PMC8109851 DOI: 10.1002/vro2.2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 12/11/2020] [Accepted: 01/13/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Gastrointestinal (GI) toxicity is a major dose-limiting factor in dogs undergoing chemotherapy. A proposed mechanism of GI toxicity includes chemotherapy-driven GI dysbiosis. This study was designed to determine the effects of probiotic administration on GI side-effects in dogs receiving multi-agent chemotherapy. METHODS Ten client-owned dogs with multicentric lymphoma were enrolled in a prospective, randomised, placebo-controlled single-blinded study. On the first day of the cyclophosphamide doxorubicin vincristine prednisone (CHOP)-based chemotherapy protocol, dogs were randomised to receive either daily oral probiotic at a dose of 200 × 109 cfu/10 kg (n = 5) or daily oral placebo (n = 5). Complete blood count, faecal score (FS), faecal microbiome analysis (qPCR) and adverse events scores were performed at baseline and on the day of each subsequent chemotherapy dose, as well as 3 days after doxorubicin (days 0, 7, 14, 21, 24 and 28). RESULTS Overall, 40% of dogs had an abnormal GI microbiome at baseline, specifically decreased faecal C. hiranonis and Fusobacterium abundances. Dogs receiving probiotics had increased faecal Streptococcus (p = 0.02) and E. coli. (p = 0.01). No dogs receiving probiotics experienced diarrhoea (FS ≥ 3.5) compared to four of five receiving placebo. (F 2.895; p = 0.13). CONCLUSION GI microbiome dysbiosis was common in this group of dogs with multicentric lymphoma. Probiotics were well-tolerated, with no negative side effects. Further studies are needed to explore broader microbiome and metabolome changes, as well as clinical benefit.
Collapse
Affiliation(s)
- Maria C. Jugan
- Department of Clinical SciencesCollege of Veterinary MedicineKansas State UniversityManhattanKansasUSA
| | - Raelene M. Wouda
- Department of Clinical SciencesCollege of Veterinary MedicineKansas State UniversityManhattanKansasUSA
| | - Mary Lynn Higginbotham
- Department of Clinical SciencesCollege of Veterinary MedicineKansas State UniversityManhattanKansasUSA
| |
Collapse
|
6
|
A Perspective on the Role of Microbiome for Colorectal Cancer Treatment. Cancers (Basel) 2021; 13:cancers13184623. [PMID: 34572850 PMCID: PMC8468110 DOI: 10.3390/cancers13184623] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 09/13/2021] [Accepted: 09/13/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Colorectal cancer is the third most diagnosed cancer worldwide and contributes significantly to global mortality and morbidity. The gut microbiome, composed of the trillions of microbes endemic to the human gastrointestinal tract, has been shown to be implicated in colorectal cancer oncogenesis; however, the roles of microbiota and dysbiosis in CRC treatment remain poorly understood. This review sought to characterize this relationship and in doing so, identify how these interactions may inform future treatments in the form of synbiotics designed to alter the host microbiota to achieve optimized treatment outcomes. Abstract In healthy hosts, trillions of microbes colonise the gut and oral cavity in a well-balanced state, maintaining a mutually beneficial relationship. Loss of this balance, termed dysbiosis, is strongly implicated in the pathogenesis of colorectal cancer (CRC). However, the roles of microbiota and dysbiosis in CRC treatment remain poorly understood. Recent studies suggest that the gut microbiota has the ability to affect the host response to chemotherapeutic agents by enhancing drug efficacy, promoting chemoresistance and mediating chemotherapy-induced toxicity and side effects via a variety of mechanisms. Several other studies have also proposed manipulation of the microbiota to optimise CRC treatment. In this review, we summarise the current advancement of knowledge on how microbiota and CRC treatments interact with each other and how this interaction may shed some light on the development of personalised microbiota manipulations that improve CRC treatment outcomes.
Collapse
|
7
|
Gunasegaran B, Neilsen PM, Smid SD. P53 activation suppresses irinotecan metabolite SN-38-induced cell damage in non-malignant but not malignant epithelial colonic cells. Toxicol In Vitro 2020; 67:104908. [PMID: 32502622 DOI: 10.1016/j.tiv.2020.104908] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 05/25/2020] [Accepted: 05/30/2020] [Indexed: 12/12/2022]
Abstract
Nutlin-3a is a p53 activator and potential cyclotherapy approach that may also mitigate side effects of chemotherapeutic drugs in the treatment of colorectal cancer. We investigated cell proliferation in a panel of colorectal cancer (CRC) cell lines with wild-type or mutant p53, as well as a non-tumorigenic fetal intestinal cell line following Nutlin-3a treatment (10 μM). We then assessed apoptosis at 24 and 48 h following administration of the active irinotecan metabolite, SN-38 (0.001 μM - 1 μM), alone or following pre-treatment with Nutlin-3a (10 μM). Nutlin-3a treatment (10 μM) significantly reduced proliferation in wild-type p53 expressing cell lines (FHS 74 and HCT116+/+) at 72 and 96 h, but was without effect in cell lines with mutated or deleted p53 (Caco-2, SW480, and HCT 116-/-). SN-38 treatment induced significant apoptosis in all cell lines after 48 h. Nutlin-3a unexpectedly increased cell death in the p53 wild-type CRC cell line, HCT116+/+, while Nutlin-3a pre-treatment provided protection from SN-38 in the p53 wild-type normal cell line, FHs 74. These results demonstrate Nutlin-3a's selective growth-arresting efficacy in p53 wild-type non-malignant intestinal cell lines, enabling the selective targeting of malignant cells with chemotherapy drugs. These studies highlight the potential of Nutlin-3a to minimise intestinal mucosal damage following chemotherapy.
Collapse
Affiliation(s)
- Bavani Gunasegaran
- Discipline of Pharmacology, Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, South Australia, Australia
| | - Paul M Neilsen
- School of Health, Medical and Applied Sciences, Central Queensland University, Queensland, Australia; Centre for Personalized Cancer Medicine, Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, South Australia, Australia
| | - Scott D Smid
- Discipline of Pharmacology, Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, South Australia, Australia.
| |
Collapse
|
8
|
Wu J, Gan Y, Li M, Chen L, Liang J, Zhuo J, Luo H, Xu N, Wu X, Wu Q, Lin Z, Su Z, Liu Y. Patchouli alcohol attenuates 5-fluorouracil-induced intestinal mucositis via TLR2/MyD88/NF-kB pathway and regulation of microbiota. Biomed Pharmacother 2020; 124:109883. [PMID: 32004938 DOI: 10.1016/j.biopha.2020.109883] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 01/07/2020] [Accepted: 01/10/2020] [Indexed: 02/09/2023] Open
Abstract
Intestinal mucositis causes great suffering to cancer patients who undergo chemotherapy and radiotherapy. Owing to the uncertain side effects of anticancer drugs to attenuate patients' intestinal mucositis, many studies focused on traditional Chinese medicine (TCM). Patchouli alcohol (PA) is an active compound extracted from Pogostemon cablin, and has potent gastrointestinal protective effect. However, whether PA has an effect on intestinal mucositis is still unknown. Therefore, we established a rat model of intestinal mucositis via intraperitoneal injection of 5-fluorouracil, and intragastrically administrated PA (10, 20, and 40 mg/kg) to evaluate the effect of PA on intestinal mucositis. The routine observation (body weight, food intake, and diarrhea) in rats was used to detect whether PA had an effect on intestinal mucositis. Levels of inflammatory cytokines (TNF-α, IL-1β, IL-6, IL-10, and MPO), mucosal barrier proteins (zonula occludens -1 (ZO-1), claudin-1, occludin, myosin light chain (MLC), and mucin-2) and intestinal microbiota were determined to elucidate the underlying mechanism of PA action on intestinal mucositis in rats. The results showed that PA could effectively improve body weight, food intake, and diarrhea in intestinal mucositis rats, preliminary confirming PA efficacy. Further experiments revealed that PA not only decreased the levels of TNF-α, IL-1β, IL-6, and MPO but also increased the level of IL-10 significantly. In addition, the expression of mucosal barrier proteins and microbiota community were also improved after PA treatment in diseased rats. Hence, PA may prevent the development and progression of intestinal mucositis by improving inflammation, protecting mucosal barrier, and regulating intestinal microbiota.
Collapse
Affiliation(s)
- Jiazhen Wu
- Guangdong Provincial Key Laboratory of New Drug Development and Research of Chinese Medicine, Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Yuxuan Gan
- Guangdong Provincial Key Laboratory of New Drug Development and Research of Chinese Medicine, Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Muxia Li
- Guangdong Provincial Key Laboratory of New Drug Development and Research of Chinese Medicine, Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Liping Chen
- Faculty of Health Sciences, University of Macau, Macao, China
| | - Jiali Liang
- Faculty of Science and Engineering, Macquarie University, Balaclava Road, Macquarie Park, NSW, Sydney, 2109, Australia
| | - Jianyi Zhuo
- Guangdong Provincial Key Laboratory of New Drug Development and Research of Chinese Medicine, Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Huijuan Luo
- Guangdong Provincial Key Laboratory of New Drug Development and Research of Chinese Medicine, Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Nan Xu
- Guangdong Provincial Key Laboratory of New Drug Development and Research of Chinese Medicine, Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Xue Wu
- Guangdong Provincial Key Laboratory of New Drug Development and Research of Chinese Medicine, Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Qiduan Wu
- The First Affiliated Hospital of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Zhixiu Lin
- Guangdong Provincial Key Laboratory of New Drug Development and Research of Chinese Medicine, Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Ziren Su
- Guangdong Provincial Key Laboratory of New Drug Development and Research of Chinese Medicine, Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China; Dongguan & Guangzhou University of Chinese Medicine Cooperative Academy of Mathematical Engineering for Chinese Medicine, Dongguan, 523808, China.
| | - Yuhong Liu
- Guangdong Provincial Key Laboratory of New Drug Development and Research of Chinese Medicine, Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| |
Collapse
|
9
|
Chang CW, Lee HC, Li LH, Chiang Chiau JS, Wang TE, Chuang WH, Chen MJ, Wang HY, Shih SC, Liu CY, Tsai TH, Chen YJ. Fecal Microbiota Transplantation Prevents Intestinal Injury, Upregulation of Toll-Like Receptors, and 5-Fluorouracil/Oxaliplatin-Induced Toxicity in Colorectal Cancer. Int J Mol Sci 2020; 21:ijms21020386. [PMID: 31936237 PMCID: PMC7013718 DOI: 10.3390/ijms21020386] [Citation(s) in RCA: 122] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Revised: 12/18/2019] [Accepted: 01/06/2020] [Indexed: 02/07/2023] Open
Abstract
FOLFOX (5-fluorouracil, leucovorin, and oxaliplatin), a 5-fluorouracil (5-FU)-based chemotherapy regimen, is one of most common therapeutic regimens for colorectal cancer. However, intestinal mucositis is a common adverse effect for which no effective preventive strategies exist. Moreover, the efficacy and the safety of fecal microbiota transplants (FMT) in cancer patients treated with anti-neoplastic agents are still scant. We investigated the effect of FMT on FOLFOX-induced mucosal injury. BALB/c mice implanted with syngeneic CT26 colorectal adenocarcinoma cells were orally administered FMT daily during and two days after five-day injection of FOLFOX regimen for seven days. Administration of FOLFOX significantly induced marked levels of diarrhea and intestinal injury. FMT reduced the severity of diarrhea and intestinal mucositis. Additionally, the number of goblet cells and zonula occludens-1 decreased, while apoptotic and NF-κB-positive cells increased following FOLFOX treatment. The expression of toll-like receptors (TLRs), MyD88, and serum IL-6 were upregulated following FOLFOX treatment. These responses were attenuated following FMT. The disrupted fecal gut microbiota composition was also restored by FMT after FOLFOX treatment. Importantly, FMT did not cause bacteremia and safely alleviated FOLFOX-induced intestinal mucositis in colorectal cancer-bearing mice. The putative mechanism may involve the gut microbiota TLR-MyD88-NF-κB signaling pathway in mice with implanted colorectal carcinoma cells.
Collapse
Affiliation(s)
- Ching-Wei Chang
- Institute of Traditional Medicine, National Yang-Ming University, Taipei 11221, Taiwan;
- Division of Gastroenterology, Department of Internal Medicine, MacKay Memorial Hospital, Taipei 10449, Taiwan; (T.-E.W.); (M.-J.C.); (H.-Y.W.); (S.-C.S.)
- Department of Medical Research, MacKay Memorial Hospital, New Taipei City 25173, Taiwan; (H.-C.L.); (L.-H.L.); (J.-S.C.C.)
- Department of Medicine, MacKay Medical College, New Taipei City 25245, Taiwan
- MacKay Junior College of Medicine, Nursing, and Management, New Taipei City 11260, Taiwan
| | - Hung-Chang Lee
- Department of Medical Research, MacKay Memorial Hospital, New Taipei City 25173, Taiwan; (H.-C.L.); (L.-H.L.); (J.-S.C.C.)
- Department of Medicine, MacKay Medical College, New Taipei City 25245, Taiwan
- MacKay Junior College of Medicine, Nursing, and Management, New Taipei City 11260, Taiwan
- MacKay Children’s Hospital, Taipei 10449, Taiwan
| | - Li-Hui Li
- Department of Medical Research, MacKay Memorial Hospital, New Taipei City 25173, Taiwan; (H.-C.L.); (L.-H.L.); (J.-S.C.C.)
| | - Jen-Shiu Chiang Chiau
- Department of Medical Research, MacKay Memorial Hospital, New Taipei City 25173, Taiwan; (H.-C.L.); (L.-H.L.); (J.-S.C.C.)
| | - Tsang-En Wang
- Division of Gastroenterology, Department of Internal Medicine, MacKay Memorial Hospital, Taipei 10449, Taiwan; (T.-E.W.); (M.-J.C.); (H.-Y.W.); (S.-C.S.)
- Department of Medicine, MacKay Medical College, New Taipei City 25245, Taiwan
- MacKay Junior College of Medicine, Nursing, and Management, New Taipei City 11260, Taiwan
| | - Wei-Hung Chuang
- Institute of Biomedical Informatics, Center for Systems and Synthetic Biology, National Yang-Ming University, Taipei 11221, Taiwan;
| | - Ming-Jen Chen
- Division of Gastroenterology, Department of Internal Medicine, MacKay Memorial Hospital, Taipei 10449, Taiwan; (T.-E.W.); (M.-J.C.); (H.-Y.W.); (S.-C.S.)
- Department of Medicine, MacKay Medical College, New Taipei City 25245, Taiwan
- MacKay Junior College of Medicine, Nursing, and Management, New Taipei City 11260, Taiwan
| | - Horng-Yuan Wang
- Division of Gastroenterology, Department of Internal Medicine, MacKay Memorial Hospital, Taipei 10449, Taiwan; (T.-E.W.); (M.-J.C.); (H.-Y.W.); (S.-C.S.)
- Department of Medicine, MacKay Medical College, New Taipei City 25245, Taiwan
- MacKay Junior College of Medicine, Nursing, and Management, New Taipei City 11260, Taiwan
| | - Shou-Chuan Shih
- Division of Gastroenterology, Department of Internal Medicine, MacKay Memorial Hospital, Taipei 10449, Taiwan; (T.-E.W.); (M.-J.C.); (H.-Y.W.); (S.-C.S.)
- Department of Medicine, MacKay Medical College, New Taipei City 25245, Taiwan
- MacKay Junior College of Medicine, Nursing, and Management, New Taipei City 11260, Taiwan
| | - Chia-Yuan Liu
- Institute of Traditional Medicine, National Yang-Ming University, Taipei 11221, Taiwan;
- Division of Gastroenterology, Department of Internal Medicine, MacKay Memorial Hospital, Taipei 10449, Taiwan; (T.-E.W.); (M.-J.C.); (H.-Y.W.); (S.-C.S.)
- Department of Medical Research, MacKay Memorial Hospital, New Taipei City 25173, Taiwan; (H.-C.L.); (L.-H.L.); (J.-S.C.C.)
- Department of Medicine, MacKay Medical College, New Taipei City 25245, Taiwan
- MacKay Junior College of Medicine, Nursing, and Management, New Taipei City 11260, Taiwan
- Correspondence: (C.-Y.L.); (T.-H.T.); (Y.-J.C.); Tel.: +886-2-2543-3535 (ext. 3993) (C.-Y.L.); +886-2-2826-7115 (T.-H.T.); +886-2-2809-4661 (ext. 2301) (Y.-J.C.)
| | - Tung-Hu Tsai
- Institute of Traditional Medicine, National Yang-Ming University, Taipei 11221, Taiwan;
- Department of Chemical Engineering, National United University, Miaoli 36063, Taiwan
- Correspondence: (C.-Y.L.); (T.-H.T.); (Y.-J.C.); Tel.: +886-2-2543-3535 (ext. 3993) (C.-Y.L.); +886-2-2826-7115 (T.-H.T.); +886-2-2809-4661 (ext. 2301) (Y.-J.C.)
| | - Yu-Jen Chen
- Institute of Traditional Medicine, National Yang-Ming University, Taipei 11221, Taiwan;
- Department of Medical Research, MacKay Memorial Hospital, New Taipei City 25173, Taiwan; (H.-C.L.); (L.-H.L.); (J.-S.C.C.)
- Department of Medicine, MacKay Medical College, New Taipei City 25245, Taiwan
- MacKay Junior College of Medicine, Nursing, and Management, New Taipei City 11260, Taiwan
- Department of Radiation Oncology, MacKay Memorial Hospital, Taipei 10449, Taiwan
- Correspondence: (C.-Y.L.); (T.-H.T.); (Y.-J.C.); Tel.: +886-2-2543-3535 (ext. 3993) (C.-Y.L.); +886-2-2826-7115 (T.-H.T.); +886-2-2809-4661 (ext. 2301) (Y.-J.C.)
| |
Collapse
|
10
|
Gao Y, Sun Q, Yang X, Lu W, Zhao Y, Ge W, Yang Y, Xu X, Zhang J. Orally administered salecan ameliorates methotrexate-induced intestinal mucositis in mice. Cancer Chemother Pharmacol 2019; 84:105-116. [DOI: 10.1007/s00280-019-03854-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Accepted: 05/02/2019] [Indexed: 01/21/2023]
|
11
|
Khan S, Wardill HR, Bowen JM. Role of toll-like receptor 4 (TLR4)-mediated interleukin-6 (IL-6) production in chemotherapy-induced mucositis. Cancer Chemother Pharmacol 2018; 82:31-37. [PMID: 29845394 DOI: 10.1007/s00280-018-3605-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 05/22/2018] [Indexed: 12/12/2022]
Abstract
Despite significant advances in our ability to treat cancer, cytotoxic chemotherapy continues to be the mainstay treatment for many solid tumours. Chemotherapy is commonly associated with a raft of largely manageable adverse events; however, gastrointestinal (GI) toxicity (also termed mucositis) remains a significant challenge with little in the way of preventative and therapeutic options. The inability to manage GI complications likely reflects our incomplete understanding of its aetiology and the idiosyncrasies of each chemotherapeutic agent. This review highlights aims to provide a narrative for the involvement of Toll-like receptor (TLR4) in the development of chemotherapy-induced GI mucositis, an already emerging theme within this field. Particular focus will be placed upon the signalling interaction between TLR4 and interleukin (IL)-6. This parallels recent preclinical findings showing that TLR4 knockout mice, which are protected from developing severe GI mucositis, completely lack an IL-6 response. As such, we suggest that this signalling pathway presents as a novel mechanism with potential for therapeutic intervention.
Collapse
Affiliation(s)
- S Khan
- Adelaide Medical School, The University of Adelaide, Adelaide, Australia
| | - Hannah R Wardill
- Adelaide Medical School, The University of Adelaide, Adelaide, Australia.
- Centre for Nutrition and Gastrointestinal Disease, South Australian Health and Medical Research Institute, North Terrace, Adelaide, SA, 5000, Australia.
| | - J M Bowen
- Adelaide Medical School, The University of Adelaide, Adelaide, Australia
| |
Collapse
|
12
|
Konstantinov SR. Diet, microbiome, and colorectal cancer. Best Pract Res Clin Gastroenterol 2017; 31:675-681. [PMID: 29566911 DOI: 10.1016/j.bpg.2017.09.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 09/03/2017] [Indexed: 01/31/2023]
Abstract
The scientific interests in the colorectal cancer (CRC) associated microbiome have increased significantly in the past decade. Mechanistically, several members of the human microbiome and products thereof have been implicated as inductors of the pathogenic inflammation related to CRC. Conversely, the activities of the human intestinal microbial community influenced by specific diet might confer a protective effect against the CRC risks and progression. As the microbiome is both a key contributor and one of the tools to prevent CRC, the current review gives a summary of the CRC-associated microbiome and the dietary strategies relevant to CRC. As more evidences become available, new microbiome-based treatments and specific diets may emerge to reduce the CRC risk and improve CRC patients' quality of life.
Collapse
Affiliation(s)
- Sergey R Konstantinov
- Department of Gasteroenterology and Hepatology, Erasmus MC - University Medical Center Rotterdam, 's Gravendijkwal 230, NL-3015, CE Rotterdam, The Netherlands.
| |
Collapse
|
13
|
Carvalho RD, Breyner N, Menezes-Garcia Z, Rodrigues NM, Lemos L, Maioli TU, da Gloria Souza D, Carmona D, de Faria AMC, Langella P, Chatel JM, Bermúdez-Humarán LG, Figueiredo HCP, Azevedo V, de Azevedo MS. Secretion of biologically active pancreatitis-associated protein I (PAP) by genetically modified dairy Lactococcus lactis NZ9000 in the prevention of intestinal mucositis. Microb Cell Fact 2017; 16:27. [PMID: 28193209 PMCID: PMC5307810 DOI: 10.1186/s12934-017-0624-x] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 01/03/2017] [Indexed: 12/22/2022] Open
Abstract
Background Mucositis is one of the most relevant gastrointestinal inflammatory conditions in humans, generated by the use of chemotherapy drugs, such as 5-fluoracil (5-FU). 5-FU-induced mucositis affects 80% of patients undergoing oncological treatment causing mucosal gut dysfunctions and great discomfort. As current therapy drugs presents limitations in alleviating mucositis symptoms, alternative strategies are being pursued. Recent studies have shown that the antimicrobial pancreatitis-associated protein (PAP) has a protective role in intestinal inflammatory processes. Indeed, it was demonstrated that a recombinant strain of Lactococcus lactis expressing human PAP (LL-PAP) could prevent and improve murine DNBS-induced colitis, an inflammatory bowel disease (IBD) that causes severe inflammation of the colon. Hence, in this study we sought to evaluate the protective effects of LL-PAP on 5-FU-induced experimental mucositis in BALB/c mice as a novel approach to treat the disease. Results Our results show that non-recombinant L. lactis NZ9000 have antagonistic activity, in vitro, against the enteroinvasive gastrointestinal pathogen L. monocytogenes and confirmed PAP inhibitory effect against Opportunistic E. faecalis. Moreover, L. lactis was able to prevent histological damage, reduce neutrophil and eosinophil infiltration and secretory Immunoglobulin-A in mice injected with 5-FU. Recombinant lactococci carrying antimicrobial PAP did not improve those markers of inflammation, although its expression was associated with villous architecture preservation and increased secretory granules density inside Paneth cells in response to 5-FU inflammation. Conclusions We have demonstrated for the first time that L. lactis NZ9000 by itself, is able to prevent 5-FU-induced intestinal inflammation in BALB/c mice. Moreover, PAP delivered by recombinant L. lactis strain showed additional protective effects in mice epithelium, revealing to be a promising strategy to treat intestinal mucositis.
Collapse
Affiliation(s)
- Rodrigo D Carvalho
- Federal University of Minas Gerais (UFMG-ICB), Av. Antônio Carlos, 6627, CP 486, Belo Horizonte, MG, 31270-901, Brazil
| | - Natalia Breyner
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350, Jouy-En-Josas, France
| | - Zelia Menezes-Garcia
- Federal University of Minas Gerais (UFMG-ICB), Av. Antônio Carlos, 6627, CP 486, Belo Horizonte, MG, 31270-901, Brazil
| | - Nubia M Rodrigues
- Federal University of Minas Gerais (UFMG-ICB), Av. Antônio Carlos, 6627, CP 486, Belo Horizonte, MG, 31270-901, Brazil
| | - Luisa Lemos
- Federal University of Minas Gerais (UFMG-ICB), Av. Antônio Carlos, 6627, CP 486, Belo Horizonte, MG, 31270-901, Brazil
| | - Tatiane U Maioli
- Federal University of Minas Gerais (UFMG-ICB), Av. Antônio Carlos, 6627, CP 486, Belo Horizonte, MG, 31270-901, Brazil
| | - Danielle da Gloria Souza
- Federal University of Minas Gerais (UFMG-ICB), Av. Antônio Carlos, 6627, CP 486, Belo Horizonte, MG, 31270-901, Brazil.,Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350, Jouy-En-Josas, France
| | - Denise Carmona
- Federal University of Minas Gerais (UFMG-ICB), Av. Antônio Carlos, 6627, CP 486, Belo Horizonte, MG, 31270-901, Brazil
| | - Ana M C de Faria
- Federal University of Minas Gerais (UFMG-ICB), Av. Antônio Carlos, 6627, CP 486, Belo Horizonte, MG, 31270-901, Brazil
| | - Philippe Langella
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350, Jouy-En-Josas, France
| | - Jean-Marc Chatel
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350, Jouy-En-Josas, France
| | - Luis G Bermúdez-Humarán
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350, Jouy-En-Josas, France
| | - Henrique C P Figueiredo
- Federal University of Minas Gerais (UFMG-ICB), Av. Antônio Carlos, 6627, CP 486, Belo Horizonte, MG, 31270-901, Brazil
| | - Vasco Azevedo
- Federal University of Minas Gerais (UFMG-ICB), Av. Antônio Carlos, 6627, CP 486, Belo Horizonte, MG, 31270-901, Brazil
| | - Marcela S de Azevedo
- Federal University of Minas Gerais (UFMG-ICB), Av. Antônio Carlos, 6627, CP 486, Belo Horizonte, MG, 31270-901, Brazil.
| |
Collapse
|
14
|
Cabrera-Perez J, Babcock JC, Dileepan T, Murphy KA, Kucaba TA, Badovinac VP, Griffith TS. Gut Microbial Membership Modulates CD4 T Cell Reconstitution and Function after Sepsis. THE JOURNAL OF IMMUNOLOGY 2016; 197:1692-8. [PMID: 27448587 DOI: 10.4049/jimmunol.1600940] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 06/29/2016] [Indexed: 12/29/2022]
Abstract
Transient lymphopenia is one hallmark of sepsis, and emergent data indicate the CD4 T cell compartment in sepsis survivors is numerically and functionally altered (when examined at the Ag-specific level) compared with nonseptic control subjects. Previous data from our laboratory demonstrated Ag-independent, lymphopenia-induced homeostatic proliferation to be a contributing mechanism by which CD4 T cells numerically recover in sepsis survivors. However, we reasoned it is also formally possible that some CD4 T cells respond directly to Ag expressed by gut-resident microbes released during polymicrobial sepsis. The effect of gut microbiome leakage on CD4 T cells is currently unknown. In this study, we explored the number and function of endogenous CD4 T cells specific for segmented filamentous bacterium (SFB) after cecal ligation and puncture (CLP)-induced sepsis using mice that either contained or lacked SFB as a normal gut-resident microbe. Interestingly, SFB-specific CD4 T cells underwent Ag-driven proliferation in CLP-treated SFB(+), but not in SFB(-), mice. Moreover, CLP-treated SFB(+) mice showed resistance to secondary lethal infection with recombinant SFB Ag-expressing virulent Listeria (but not wild-type virulent Listeria), suggesting the CLP-induced polymicrobial sepsis primed for a protective response by the SFB-specific CD4 T cells. Thus, our data demonstrate that the numerical recovery and functional responsiveness of Ag-specific CD4 T cells in sepsis survivors is, in part, modulated by the intestinal barrier's health discreetly defined by individual bacterial populations of the host's microbiome.
Collapse
Affiliation(s)
- Javier Cabrera-Perez
- Microbiology, Immunology, and Cancer Biology Graduate Program, University of Minnesota, Minneapolis, MN 55455; Medical Scientist Training Program, University of Minnesota, Minneapolis, MN 55455
| | - Jeffrey C Babcock
- Medical Student Summer Research Program in Infection and Immunity, University of Minnesota, Minneapolis, MN 55455
| | | | | | - Tamara A Kucaba
- Department of Urology, University of Minnesota, Minneapolis, MN 55455
| | - Vladimir P Badovinac
- Department of Pathology, University of Iowa, Iowa City, IA 52242; Interdisciplinary Program in Immunology, University of Iowa, Iowa City, IA 52242
| | - Thomas S Griffith
- Microbiology, Immunology, and Cancer Biology Graduate Program, University of Minnesota, Minneapolis, MN 55455; Center for Immunology, University of Minnesota, Minneapolis, MN 55455; Department of Urology, University of Minnesota, Minneapolis, MN 55455; Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455; and Minneapolis VA Health Care System, Minneapolis, MN 55417
| |
Collapse
|
15
|
Krezalek MA, Skowron KB, Guyton KL, Shakhsheer B, Hyoju S, Alverdy JC. The intestinal microbiome and surgical disease. Curr Probl Surg 2016; 53:257-93. [PMID: 27497246 DOI: 10.1067/j.cpsurg.2016.06.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 06/07/2016] [Indexed: 12/12/2022]
Affiliation(s)
- Monika A Krezalek
- Department of Surgery, Center for Surgical Infection Research and Therapeutics, Pritzker School of Medicine, University of Chicago, Chicago, IL
| | - Kinga B Skowron
- Department of Surgery, Center for Surgical Infection Research and Therapeutics, Pritzker School of Medicine, University of Chicago, Chicago, IL
| | - Kristina L Guyton
- Department of Surgery, Center for Surgical Infection Research and Therapeutics, Pritzker School of Medicine, University of Chicago, Chicago, IL
| | - Baddr Shakhsheer
- Department of Surgery, Center for Surgical Infection Research and Therapeutics, Pritzker School of Medicine, University of Chicago, Chicago, IL
| | - Sanjiv Hyoju
- Department of Surgery, Center for Surgical Infection Research and Therapeutics, Pritzker School of Medicine, University of Chicago, Chicago, IL
| | - John C Alverdy
- Department of Surgery, Center for Surgical Infection Research and Therapeutics, Pritzker School of Medicine, University of Chicago, Chicago, IL.
| |
Collapse
|
16
|
Abstract
PURPOSE OF REVIEW Microbiota secrete a multitude of factors that either confer virulence or promote colonization because they are continuously challenged by host immune responses. The dynamic interplay between the host's immune response and microbiota eventually determines the outcome for the host: health or disease. Toll-like receptors (TLRs) play a key role in this interplay as they can recognize both microbial and host-derived ligands on the basis of the context in which recognition occurs. RECENT FINDINGS Evidence is accumulating that conventional cancer therapies alter interactions and cross talks between the host and microbiota. This has been shown for intestinal mucositis, a common side-effect of various cancer therapies. Advances have been made in the development of new and less toxic cancer strategies. One promising field is immunotherapy on the basis of TLR activation through recognition of microbial-associated molecular patterns. SUMMARY Evidence is emerging, indicating that existing cancer therapies have implications on the composition and functionality of the host-microbiota environment. This may favor the colonization of pathogens and build up the overall toxicity of the drug. Exploitation of the host-microbiota cross talks mediated by TLRs is an emerging and promising field in the search for new, less toxic anticancer strategies.
Collapse
|
17
|
Bateman E, Weaver E, Klein G, Wignall A, Wozniak B, Plews E, Mayo B, White I, Keefe D. Serum-derived bovine immunoglobulin/protein isolate in the alleviation of chemotherapy-induced mucositis. Support Care Cancer 2015; 24:377-385. [PMID: 26081596 PMCID: PMC4669373 DOI: 10.1007/s00520-015-2806-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 06/08/2015] [Indexed: 12/14/2022]
Abstract
BACKGROUND Gastrointestinal (GI) mucositis caused by chemotherapy is associated with diarrhoea and intestinal barrier disruption caused by apoptosis, immune dysfunction and microbiome alterations. Serum-derived bovine immunoglobulin/protein isolate (SBI) has been shown to manage HIV-associated enteropathy and irritable bowel syndrome with diarrhoea (IBS-D). We investigated in a rat model whether SBI was effective in alleviating symptoms of irinotecan-induced GI mucositis. METHODS Animals were gavaged with 250 or 500 mg/kg of SBI twice daily for 4 days, before intraperitoneal administration of 200 mg/kg irinotecan. Twice daily gavaging of SBI continued for 6 days post-irinotecan. Animals were monitored for bodyweight changes and incidence of diarrhoea and clinical symptoms of stress. Tissues and blood samples were collected at necropsy 6 h, and 2, 4 and 6 days post-irinotecan. H&E-stained colon and jejunum were analysed for histological damage. RESULTS The overall incidence, severity and duration of diarrhoea, and clinical symptoms of mucositis were decreased in irinotecan-treated animals that had received SBI. Animals receiving 500 mg/kg SBI also tended to lose less bodyweight than animals treated only with irinotecan (P > 0.10). SBI-gavaged animals had less pronounced irinotecan-induced changes in neutrophil (P = 0.04959) and lymphocyte (P = 0.0035) levels, and lower tissue damage scores than those receiving irinotecan alone (P < 0.0001). CONCLUSIONS Twice daily oral gavage of SBI was well-tolerated and reduced the incidence, severity and duration of irinotecan-induced mucositis. SBI was associated with less pronounced changes in inflammatory cell levels and tissue damage to colon and jejunum. Ongoing experiments aim to investigate the mechanisms of SBI-associated gastrointestinal protection.
Collapse
Affiliation(s)
- Emma Bateman
- Mucositis Research Group, School of Medicine, Faculty of Health Sciences, University of Adelaide, Level 4 Hanson Institute Building, Frome Road, Adelaide, SA, 5000, Australia.
| | | | | | - Anthony Wignall
- Mucositis Research Group, School of Medicine, Faculty of Health Sciences, University of Adelaide, Level 4 Hanson Institute Building, Frome Road, Adelaide, SA, 5000, Australia
| | - Belinda Wozniak
- Mucositis Research Group, School of Medicine, Faculty of Health Sciences, University of Adelaide, Level 4 Hanson Institute Building, Frome Road, Adelaide, SA, 5000, Australia
| | - Erin Plews
- Mucositis Research Group, School of Medicine, Faculty of Health Sciences, University of Adelaide, Level 4 Hanson Institute Building, Frome Road, Adelaide, SA, 5000, Australia
| | - Bronwen Mayo
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia
| | - Imogen White
- School of Medical Sciences, Faculty of Health Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Dorothy Keefe
- Mucositis Research Group, School of Medicine, Faculty of Health Sciences, University of Adelaide, Level 4 Hanson Institute Building, Frome Road, Adelaide, SA, 5000, Australia.,SA Cancer Service, Royal Adelaide Hospital, Adelaide, SA, Australia
| |
Collapse
|
18
|
Generoso SDV, Rodrigues NM, Trindade LM, Paiva NC, Cardoso VN, Carneiro CM, Ferreira AVDM, Faria AMC, Maioli TU. Dietary supplementation with omega-3 fatty acid attenuates 5-fluorouracil induced mucositis in mice. Lipids Health Dis 2015; 14:54. [PMID: 26063053 PMCID: PMC4473827 DOI: 10.1186/s12944-015-0052-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 05/28/2015] [Indexed: 12/14/2022] Open
Abstract
Background Studies showed the positive effects of omega-3 fatty acid (n-3 FA) for the treatment of inflammatory bowel disease as it alleviated the symptoms and promoted better mucosal integrity. The objective of this study was to determine whether a diet with the addition of n-3 FA helps control the inflammation observed in 5-fluorouracil (5-FU) induced mucositis. Methods BALB/c mice were randomly divided into four groups as follows: 1: control (CTL), fed a standard chow diet; 2: CTL + n-3 FA – n-3 FA, fed a diet with n-3; 3: mucositis (MUC), fed a standard chow diet and subjected to mucositis; and 4: MUC+ n-3 FA, fed a diet with n-3 FA and subjected to mucositis. On the 8th day, the animals of the MUC and MUC + n-3 FA groups received an intraperitoneal injection of 300 mg/kg 5-FU for mucositis induction. After 24 h or 72 h, all mice were euthanized and evaluated for intestinal permeability, bacterial translocation, intestinal histology and apoptosis. Results Mice that received the diet with n-3 FA and a 5-FU injection showed less weight loss compared to the animals of the MUC group (p < 0.005). Decreased intestinal permeability and bacterial translocation were also observed in animals fed n-3 FA, and these mice underwent mucositis compared to the MUC group (p < 0.005). These data were associated with mucosal integrity and a reduced number of apoptotic cells in the ileum mucosa compared to the mice that received the control diet and 5-FU injection. Conclusion Together, these results show that omega-3 fatty acid decreases the mucosal damage caused by 5-FU-induced mucositis.
Collapse
Affiliation(s)
| | - Núbia Morais Rodrigues
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
| | - Luísa Martins Trindade
- Departamento de Nutrição, Escola de Enfermagem, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
| | - Nivea Carolina Paiva
- Núcleo de Pesquisa em Ciências Biológicas, Instituto de Ciências Exatas e Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Brazil.
| | - Valbert Nascimento Cardoso
- Departamento de Análises Clínicas e Toxicológicas, Escola de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
| | - Cláudia Martins Carneiro
- Núcleo de Pesquisa em Ciências Biológicas, Instituto de Ciências Exatas e Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Brazil.
| | | | - Ana Maria Caetano Faria
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
| | - Tatiani Uceli Maioli
- Departamento de Nutrição, Escola de Enfermagem, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
| |
Collapse
|
19
|
Frank M, Hennenberg EM, Eyking A, Rünzi M, Gerken G, Scott P, Parkhill J, Walker AW, Cario E. TLR signaling modulates side effects of anticancer therapy in the small intestine. THE JOURNAL OF IMMUNOLOGY 2015; 194:1983-95. [PMID: 25589072 DOI: 10.4049/jimmunol.1402481] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Intestinal mucositis represents the most common complication of intensive chemotherapy, which has a severe adverse impact on quality of life of cancer patients. However, the precise pathophysiology remains to be clarified, and there is so far no successful therapeutic intervention. In this study, we investigated the role of innate immunity through TLR signaling in modulating genotoxic chemotherapy-induced small intestinal injury in vitro and in vivo. Genetic deletion of TLR2, but not MD-2, in mice resulted in severe chemotherapy-induced intestinal mucositis in the proximal jejunum with villous atrophy, accumulation of damaged DNA, CD11b(+)-myeloid cell infiltration, and significant gene alterations in xenobiotic metabolism, including a decrease in ABCB1/multidrug resistance (MDR)1 p-glycoprotein (p-gp) expression. Functionally, stimulation of TLR2 induced synthesis and drug efflux activity of ABCB1/MDR1 p-gp in murine and human CD11b(+)-myeloid cells, thus inhibiting chemotherapy-mediated cytotoxicity. Conversely, TLR2 activation failed to protect small intestinal tissues genetically deficient in MDR1A against DNA-damaging drug-induced apoptosis. Gut microbiota depletion by antibiotics led to increased susceptibility to chemotherapy-induced mucosal injury in wild-type mice, which was suppressed by administration of a TLR2 ligand, preserving ABCB1/MDR1 p-gp expression. Findings were confirmed in a preclinical model of human chemotherapy-induced intestinal mucositis using duodenal biopsies by demonstrating that TLR2 activation limited the toxic-inflammatory reaction and maintained assembly of the drug transporter p-gp. In conclusion, this study identifies a novel molecular link between innate immunity and xenobiotic metabolism. TLR2 acts as a central regulator of xenobiotic defense via the multidrug transporter ABCB1/MDR1 p-gp. Targeting TLR2 may represent a novel therapeutic approach in chemotherapy-induced intestinal mucositis.
Collapse
Affiliation(s)
- Magdalena Frank
- Division of Gastroenterology and Hepatology, University Hospital of Essen, D-45147 Essen, Germany; Medical School, University of Duisburg-Essen, D-45122 Essen, Germany
| | - Eva Maria Hennenberg
- Division of Gastroenterology and Hepatology, University Hospital of Essen, D-45147 Essen, Germany; Medical School, University of Duisburg-Essen, D-45122 Essen, Germany
| | - Annette Eyking
- Division of Gastroenterology and Hepatology, University Hospital of Essen, D-45147 Essen, Germany; Medical School, University of Duisburg-Essen, D-45122 Essen, Germany
| | - Michael Rünzi
- Medical School, University of Duisburg-Essen, D-45122 Essen, Germany; Division of Gastroenterology and Metabolic Diseases, Kliniken Essen Süd, D-45239 Essen, Germany
| | - Guido Gerken
- Division of Gastroenterology and Hepatology, University Hospital of Essen, D-45147 Essen, Germany; Medical School, University of Duisburg-Essen, D-45122 Essen, Germany
| | - Paul Scott
- Pathogen Genomics Group, Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, United Kingdom; and
| | - Julian Parkhill
- Pathogen Genomics Group, Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, United Kingdom; and
| | - Alan W Walker
- Pathogen Genomics Group, Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, United Kingdom; and Microbiology Group, Rowett Institute of Nutrition and Health, University of Aberdeen, Aberdeen AB21 9SB, United Kingdom
| | - Elke Cario
- Division of Gastroenterology and Hepatology, University Hospital of Essen, D-45147 Essen, Germany; Medical School, University of Duisburg-Essen, D-45122 Essen, Germany;
| |
Collapse
|
20
|
Li TT, Ogino S, Qian ZR. Toll-like receptor signaling in colorectal cancer: Carcinogenesis to cancer therapy. World J Gastroenterol 2014; 20:17699-17708. [PMID: 25548469 PMCID: PMC4273121 DOI: 10.3748/wjg.v20.i47.17699] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Revised: 08/27/2014] [Accepted: 11/19/2014] [Indexed: 02/06/2023] Open
Abstract
Toll-like receptors (TLRs) are germ line encoded innate immune sensors that recognize conserved microbial structures and host alarmins, and signal expression of major histocompatibility complex proteins, costimulatory molecules, and inflammatory mediators by macrophages, neutrophils, dendritic cells, and other cell types. These protein receptors are characterized by their ability to respond to invading pathogens promptly by recognizing particular TLR ligands, including flagellin and lipopolysaccharide of bacteria, nucleic acids derived from viruses, and zymosan of fungi. There are 2 major TLR pathways; one is mediated by myeloid differentiation factor 88 (MYD88) adaptor proteins, and the other is independent of MYD88. The MYD88-dependent pathway involves early-phase activation of nuclear factor of kappa light polypeptide gene enhancer in B-cells 1 (NF-κB1) and all the TLRs, except TLR3, have been shown to activate this pathway. TLR3 and TLR4 act via MYD88-independent pathways with delayed activation of NF-κB signaling. TLRs play a vital role in activating immune responses. TLRs have been shown to mediate inflammatory responses and maintain epithelial barrier homeostasis, and are highly likely to be involved in the activation of a number of pathways following cancer therapy. Colorectal cancer (CRC) is one of the most common cancers, and accounts for almost half a million deaths annually worldwide. Inflammation is considered a risk factor for many common malignancies including cancers of the colorectum. The key molecules involved in inflammation-driven carcinogenesis include TLRs. As sensors of cell death and tissue remodeling, TLRs may have a universal role in cancer; stimulation of TLRs to activate the innate immune system has been a legitimate therapeutic strategy for some years. TLRs 3/4/7/8/9 are all validated targets for cancer therapy, and a number of companies are developing agonists and vaccine adjuvants. On the other hand, antagonists may favor inhibition of signaling responsible for autoimmune responses. In this paper, we review TLR signaling in CRC from carcinogenesis to cancer therapy.
Collapse
|
21
|
Kuiken NSS, Rings EHHM, Tissing WJE. Risk analysis, diagnosis and management of gastrointestinal mucositis in pediatric cancer patients. Crit Rev Oncol Hematol 2014; 94:87-97. [PMID: 25560731 DOI: 10.1016/j.critrevonc.2014.12.009] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Revised: 11/17/2014] [Accepted: 12/11/2014] [Indexed: 12/12/2022] Open
Abstract
Mucositis is a complex inflammatory reaction of the mucous membranes of the alimentary tract upon chemotherapy and radiotherapy treatment in oncology patients. Mucositis can be subdivided in oral and gastrointestinal mucositis (GI mucositis). The damage to the gastrointestinal tract compromises the intestinal function and thereby the nutritional status and the quality of life, and eventually affects survival. The literature on GI mucositis focuses mainly on adults. This review focuses on data available on GI mucositis in pediatric cancer patients. An evaluation of the clinical presentation and consequences of GI mucositis in children is outlined. The review summarizes key issues for clinicians with respect to risk analysis for developing mucositis and the diagnosis of this condition in children. Information on these issues is obtained from clinical trials in children and adults, and from animal models. Diagnostic tools and assessment of severity of GI mucositis in children is elaborated on. Furthermore, the clinical management of the symptoms and consequences of GI mucositis in children, with specific focus on nutritional support, are discussed.
Collapse
Affiliation(s)
- Nicoline S S Kuiken
- Department of Pediatric Oncology, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, The Netherlands; Department of Pediatric Gastroenterology and Hepatology, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, The Netherlands
| | - Edmond H H M Rings
- Department of Pediatrics, Leiden University Medical Center, The Netherlands; Department of Pediatrics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Wim J E Tissing
- Department of Pediatric Oncology, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, The Netherlands.
| |
Collapse
|
22
|
Martin J, Howard SC, Pillai A, Vogel P, Naren AP, Davis S, Ringwald- Smith K, Buddington K, Buddington RK. The weaned pig as a model for Doxorubicin-induced mucositis. Chemotherapy 2014; 60:24-36. [PMID: 25341697 PMCID: PMC7762628 DOI: 10.1159/000365725] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Accepted: 07/03/2014] [Indexed: 01/23/2023]
Abstract
BACKGROUND Chemotherapy-induced mucositis (CIM) complicates cancer therapy and limits maximum tolerated doses and efficacy. Rodent models do not reproducibly mimic clinical CIM, so alternative models are needed. METHODS CIM severity was assessed after weaned pigs were treated with doxorubicin (5 and 3.75 mg/kg) using clinical observations, laboratory parameters and gastrointestinal structure and functions. Bovine colostrum was provided as an experimental intervention to the pigs treated receiving the 3.75 mg/kg dose. RESULTS Doxorubin at 3.75 mg/kg decreased food intake and weight gain (p < 0.05) and caused diarrhea and vomiting that coincided with damage to the small intestine mucosa based on histological scoring (p < 0.05). It resulted in higher serum TNF-α concentrations, increased chloride secretion and reduced brush border membrane disaccharidase activities and carrier-mediated glucose uptake (all p < 0.05). The gastrointestinal damage and dysfunction resemble the clinical and laboratory features of CIM in humans; these can be partially prevented by providing cow colostrum. CONCLUSION The weaned pig is a relevant large animal for studying CIM and evaluating existing and experimental interventions for mucositis.
Collapse
Affiliation(s)
- Jamee Martin
- Department of Health and Sport Sciences, University of Memphis, Memphis, Tenn., USA
| | - Scott C. Howard
- Department of Oncology and International Outreach Program, Memphis, Tenn., USA
| | - Asha Pillai
- Department of Bone Marrow Transplantation and Cellular Therapy, Memphis, Tenn., USA
| | - Peter Vogel
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, Tenn., USA
| | - Anjaparavanda P. Naren
- Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Steven Davis
- Department of Abbott Nutrition, Columbus, Ohio, USA
| | - Karen Ringwald- Smith
- Department of Clinical Nutrition, St. Jude Children’s Research Hospital, Memphis, Tenn., USA
| | - Karyl Buddington
- Department of Animal Care Facilities, University of Memphis, Memphis, Tenn., USA
| | - Randal K. Buddington
- Department of Health and Sport Sciences, University of Memphis, Memphis, Tenn., USA
| |
Collapse
|
23
|
Zhan YS, Tan SW, Mao W, Jiang J, Liu HL, Wu B. Chemotherapy mediates intestinal injury via p53/p53 upregulated modulator of apoptosis (PUMA) signaling pathway. J Dig Dis 2014; 15:425-34. [PMID: 24814616 DOI: 10.1111/1751-2980.12157] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVE The aim of this study was to investigate the potential mechanism and signaling pathway involved in chemotherapy-induced intestinal mucosal injury (CIMI), which is a common physiopathological problem in patients with cancer. METHODS For the in vivo experiment, mice received intraperitoneal injection of 5-fluorouracil (5-FU) at a dose of 75 mg/kg/day for 1, 3 or 5 days. Villus height and crypt depth of the small intestine, cell apoptosis and proliferation were then examined to determine the extent of CIMI. The expressions of Akt, p53, PUMA and p21 were evaluated both in vivo in mice models and in vitro in the IEC-6 and HCT116 cell lines. RESULTS After 5-FU therapy both the intestinal villus height (275.93 μm vs 164.52 μm, P < 0.001) and crypt depth (64.13 μm vs 42.48 μm, P < 0.001) were decreased. The apoptotic index was greatly increased from 0.32% to 15.84% (P < 0.001) and proliferation was suppressed (63.58% vs 39.15%, P < 0.001). Additionally, p53 expression was significantly increased in the intestinal crypt along with the expressions of PUMA and p21. Western blot showed that the administration of 5-FU induced p53/PUMA-mediated apoptosis and upregulated p21 expression to suppress cell proliferation. CONCLUSION Chemotherapy might mediate intestinal injury via p53/PUMA-mediated apoptotic signaling and the suppression of proliferation in response to p21.
Collapse
Affiliation(s)
- Ya Shi Zhan
- Department of Gastroenterology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | | | | | | | | | | |
Collapse
|
24
|
Vanhoecke B, De Ryck T, Stringer A, Van de Wiele T, Keefe D. Microbiota and their role in the pathogenesis of oral mucositis. Oral Dis 2014; 21:17-30. [PMID: 24456144 DOI: 10.1111/odi.12224] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Revised: 12/17/2013] [Accepted: 01/11/2014] [Indexed: 02/06/2023]
Abstract
Oral mucositis in patients undergoing cancer therapy is a significant problem. Its prevalence ranges between 20 and 100%, depending on treatment type and protocols and patient-based variables. Mucositis is self-limiting when uncomplicated by infection. Unfortunately, the incidence of developing a local or systemic infection during the course of the treatment is very high. At this stage, it is unclear which role oral microbiota play in the onset, duration, and severity of oral mucositis. Nevertheless, there is growing interest in this underexplored topic, and new studies are being undertaken to unravel their impact on the pathogenesis of mucositis.
Collapse
Affiliation(s)
- B Vanhoecke
- Faculty of Bioscience Engineering, Laboratory of Microbial Ecology and Technology, Ghent University, Ghent, Belgium; Department of Medicine, Mucositis Research Group, The University of Adelaide, Adelaide, SA, Australia
| | | | | | | | | |
Collapse
|
25
|
Live and heat-killed Lactobacillus rhamnosus GG upregulate gene expression of pro-inflammatory cytokines in 5-fluorouracil-pretreated Caco-2 cells. Support Care Cancer 2014; 22:1647-54. [PMID: 24500789 DOI: 10.1007/s00520-014-2137-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2013] [Accepted: 01/13/2014] [Indexed: 01/16/2023]
Abstract
PURPOSE This study investigates whether post-chemotherapeutic use of live and heat-killed Lactobacillus rhamnosus GG can modulate the expression of three pro-inflammatory cytokines in 5-fluorouracil (5-FU)-induced intestinal mucositis in vitro. METHODS Live L. rhamnosus GG and heat-killed L. rhamnosus GG were observed using scanning electron microscopy. To establish the duration required for optimal expression of tumor necrosis factor-α (TNF-α), monocyte chemotactic protein-1 (MCP-1), and interleukin-12 (IL-12), 5 μM of 5-FU was selected to treat 10-day-old Caco-2 cells for 4, 6, 8, and 24 h. Caco-2 cells were treated with 5-FU (5 μM) for 4 h, followed by the administration of live L. rhamnosus GG (multiplicity of infection = 25), and heat-killed L. rhamnosus GG for 2 and 4 h. Finally, total cellular RNA was isolated to quantify mRNA expression of TNF-α, MCP-1, and IL-12 using real-time PCR. RESULTS The results demonstrated that heat-killed L. rhamnosus GG remained structurally intact with elongation. A biphasic upregulated expression of TNF-α, MCP-1, and IL-12 was observed in 5-FU-treated Caco-2 cells at 4 and 24 h. Compared to non-L. rhamnosus GG controls in 5-FU-pretreated Caco-2 cells, a 2-h treatment of heat-killed L. rhamnosus GG significantly upregulated the MCP-1 expression (p < 0.05), and both live and heat-killed L. rhamnosus GG treatments lasting 4 h upregulated the TNF-α and MCP-1 expression (p < 0.05). Only live L. rhamnosus GG upregulated the IL-12 expression (p < 0.05). CONCLUSIONS Post-chemotherapeutic use of live or heat-killed L. rhamnosus GG can upregulate the gene expression of 5-FU-induced pro-inflammatory cytokines in Caco-2 cells. Human intestinal epithelium may be vulnerable to the post-chemotherapeutic use of L. rhamnosus GG in 5-FU-induced mucositis that requires further in vivo studies for clarification.
Collapse
|