Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 113
Filter
1.
Curr Opin Support Palliat Care ; 18(2): 73-77, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38652454

ABSTRACT

PURPOSE OF REVIEW: Disruption of the precious ecosystem of micro-organisms that reside in the gut - the gut microbiota - is rapidly emerging as a key driver of the adverse side effects/toxicities caused by numerous anti-cancer agents. Although the contribution of the gut microbiota to these toxicities is understood with ever increasing precision, the cause of microbial disruption (dysbiosis) remains poorly understood. Here, we discuss current evidence on the cause(s) of dysbiosis after cancer therapy, positioning breakdown of the intestinal mucosa (mucositis) as a central cause. RECENT FINDINGS: Dysbiosis in people with cancer has historically been attributed to extensive antibiotic use. However, evidence now suggests that certain antibiotics have minimal impacts on the microbiota. Indeed, recent evidence shows that the type of cancer therapy predicts microbiota composition independently of antibiotics. Given most anti-cancer drugs have modest effects on microbes directly, this suggests that their impact on the gut microenvironment, in particular the mucosa, which is highly vulnerable to cytotoxicity, is a likely cause of dysbiosis. Here, we outline evidence that support this hypothesis, and discuss the associated clinical implications/opportunities. SUMMARY: The concept that mucositis dictates microbiota compositions provides two important implications for clinical practice. Firstly, it reiterates the importance of prioritising the development of novel mucoprotectants that preserve mucosal integrity, and indirectly support microbial stability. Secondly, it provides an opportunity to identify dysbiotic events and associated consequences using readily accessible, minimally invasive biomarkers of mucositis such as plasma citrulline.


Subject(s)
Anti-Bacterial Agents , Antineoplastic Agents , Dysbiosis , Gastrointestinal Microbiome , Mucositis , Neoplasms , Humans , Gastrointestinal Microbiome/physiology , Gastrointestinal Microbiome/drug effects , Dysbiosis/microbiology , Dysbiosis/chemically induced , Mucositis/microbiology , Mucositis/chemically induced , Neoplasms/drug therapy , Neoplasms/microbiology , Antineoplastic Agents/adverse effects , Anti-Bacterial Agents/pharmacology , Intestinal Mucosa/microbiology
2.
Clin Oral Implants Res ; 34(9): 947-957, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37358250

ABSTRACT

OBJECTIVES: This study aimed to investigate the relationship between microbial communities and the severity of peri-implant mucosal bleeding in peri-implant mucositis. MATERIALS AND METHODS: Submucosal plaque samples were collected from 54 implants divided into the healthy implant (HI) group, peri-implant mucositis (PM) group, and peri-implantitis (PI) group. Sequencing of 16S rRNA was performed using the Illumina MiSeq platform. Alpha diversity (i.e., Shannon and Chao index) and beta diversity were used to measure microbial diversity within and between microbial communities, respectively. Differences in microbial taxa between groups were assessed via linear discriminate analysis effect size. Correlation between the modified sulcus bleeding index (mSBI) and microbial dysbiosis index (MDI) was examined using Spearman correlation analysis and linear models. RESULTS: The submucosal bacterial richness (Chao index) was positively correlated with the mean mSBI in the PM group. As the mean mSBI increased in the PM group, the beta diversity became closer to that of the PI group. In the PM group, the abundances of 47 genera were significantly correlated with the mean mSBI, and the MDI was positively associated with the mean mSBI. Fourteen of the forty-seven genera were discriminative taxa between the HI and PI groups, and the abundances of these biomarkers became closer to those in the PI group in the progression of peri-implant disease. CONCLUSIONS: A higher mSBI value corresponded to a higher risk of microbial dysbiosis in peri-implant mucositis. The biomarkers identified may be useful for monitoring the progression of peri-implant disease.


Subject(s)
Dental Implants , Mucositis , Peri-Implantitis , Periodontitis , Humans , Peri-Implantitis/microbiology , Dental Implants/adverse effects , Dental Implants/microbiology , Mucositis/microbiology , Dysbiosis , RNA, Ribosomal, 16S/genetics , Biomarkers
3.
Probiotics Antimicrob Proteins ; 15(5): 1079-1091, 2023 10.
Article in English | MEDLINE | ID: mdl-35639268

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.


Subject(s)
Antineoplastic Agents , Gastrointestinal Microbiome , Ligilactobacillus salivarius , Mucositis , Mice , Animals , Mucositis/chemically induced , Mucositis/metabolism , Mucositis/microbiology , Cytokines/metabolism , Temozolomide/adverse effects , Temozolomide/metabolism , Mice, Inbred ICR , Antineoplastic Agents/pharmacology , Intestinal Mucosa/microbiology
4.
Biofouling ; 38(8): 814-823, 2022 09.
Article in English | MEDLINE | ID: mdl-36250998

ABSTRACT

This systematic review evaluated the features of the progression of experimentally induced gingivitis and peri-implant mucositis in humans. Included were studies that evaluated clinical, immunological, or microbiological responses between experimentally induced gingivitis and peri-implant mucositis in periodontally healthy patients. A total of 887 articles were initially identified, but only 12 were included in the final analysis. Implants accumulate less biofilm and suffer the most heterogeneous alterations in the microbiota, in the abstinence of oral hygiene, compared with the tooth. Interestingly, although dental implants presented less biofilm accumulation, the peri-implant mucosa showed a more exacerbated clinical response than the gingival tissue. The risk of bias of the selected studies was moderate to low, with one study presenting serious risk. The progression events of peri-implant mucositis were similar to those of experimental gingivitis but led to a different host response. This review was registered in the PROSPERO database CRD420201 123360.


Subject(s)
Dental Implants , Gingivitis , Mucositis , Peri-Implantitis , Humans , Mucositis/microbiology , Biofilms , Peri-Implantitis/microbiology , Gingivitis/microbiology , Dental Implants/adverse effects
5.
Int J Mol Sci ; 23(3)2022 Feb 08.
Article in English | MEDLINE | ID: mdl-35163830

ABSTRACT

Although bacteria-free DNA in blood during systemic infection is mainly derived from bacterial death, translocation of the DNA from the gut into the blood circulation (gut translocation) is also possible. Hence, several mouse models with experiments on macrophages were conducted to explore the sources, influences, and impacts of bacteria-free DNA in sepsis. First, bacteria-free DNA and bacteriome in blood were demonstrated in cecal ligation and puncture (CLP) sepsis mice. Second, administration of bacterial lysate (a source of bacterial DNA) in dextran sulfate solution (DSS)-induced mucositis mice elevated blood bacteria-free DNA without bacteremia supported gut translocation of free DNA. The absence of blood bacteria-free DNA in DSS mice without bacterial lysate implies an impact of the abundance of bacterial DNA in intestinal contents on the translocation of free DNA. Third, higher serum cytokines in mice after injection of combined bacterial DNA with lipopolysaccharide (LPS), when compared to LPS injection alone, supported an influence of blood bacteria-free DNA on systemic inflammation. The synergistic effects of free DNA and LPS on macrophage pro-inflammatory responses, as indicated by supernatant cytokines (TNF-α, IL-6, and IL-10), pro-inflammatory genes (NFκB, iNOS, and IL-1ß), and profound energy alteration (enhanced glycolysis with reduced mitochondrial functions), which was neutralized by TLR-9 inhibition (chloroquine), were demonstrated. In conclusion, the presence of bacteria-free DNA in sepsis mice is partly due to gut translocation of bacteria-free DNA into the systemic circulation, which would enhance sepsis severity. Inhibition of the responses against bacterial DNA by TLR-9 inhibition could attenuate LPS-DNA synergy in macrophages and might help improve sepsis hyper-inflammation in some situations.


Subject(s)
Cytokines/blood , DNA, Bacterial/immunology , Dextran Sulfate/adverse effects , Lipopolysaccharides/immunology , Mucositis/immunology , Sepsis/immunology , Animals , Disease Models, Animal , Feces/microbiology , Interleukin-10/blood , Interleukin-6/blood , Lipopolysaccharides/adverse effects , Macrophages/drug effects , Macrophages/immunology , Mice , Mucositis/chemically induced , Mucositis/microbiology , Sepsis/chemically induced , Sepsis/microbiology , Tumor Necrosis Factor-alpha/blood
6.
Gut Microbes ; 14(1): 2018898, 2022.
Article in English | MEDLINE | ID: mdl-35012435

ABSTRACT

Acute intestinal mucositis is a common off-target effect of chemotherapy, leading to co-morbidities such as vomiting, diarrhea, sepsis, and death. We previously demonstrated that the presence of enteric bacteria modulates the extent of jejunal epithelial damage induced by doxorubicin (DXR) in mice. Despite conventional thinking of the crypt as a sterile environment, recent evidence suggests that bacterial signaling influences aISC function. In this study, we labeled aISCs using transgenic Lgr5-driven fluorescence or with immunostaining for OLFM4. We examined the effect of DXR in both germ free (GF) mice and mice depleted of microbiota using an established antimicrobial treatment protocol (AMBx). We found differences in DXR-induced loss of aISCs between GF mice and mice treated with AMBx. aISCs were decreased after DXR in GF mice, whereas AMBx mice retained aISC expression after DXR. Neither group of mice exhibited an inflammatory response to DXR, suggesting the difference in aISC retention was not due to differences in local tissue inflammation. Therefore, we suspected that there was a protective microbial signal present in the AMBx mice that was not present in the GF mice. 16S rRNA sequencing of jejunal luminal contents demonstrated that AMBx altered the fecal and jejunal microbiota. In the jejunal contents, AMBx mice had increased abundance of Ureaplasma and Burkholderia. These results suggest pro-survival signaling from microbiota in AMBx-treated mice to the aISCs, and that this signaling maintains aISCs in the face of chemotherapeutic injury. Manipulation of the enteric microbiota presents a therapeutic target for reducing the severity of chemotherapy-associated mucositis.


Subject(s)
Antineoplastic Agents/adverse effects , Doxorubicin/adverse effects , Jejunum/drug effects , Mucositis/prevention & control , Stem Cells/drug effects , Administration, Oral , Animals , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/pharmacology , Antineoplastic Agents/administration & dosage , Bacteria/classification , Bacteria/drug effects , Bacteria/genetics , Bacteria/isolation & purification , Cell Survival/drug effects , Doxorubicin/administration & dosage , Gastrointestinal Microbiome/drug effects , Germ-Free Life , Humans , Jejunum/cytology , Jejunum/microbiology , Mice , Mice, Inbred C57BL , Mucositis/microbiology , Stem Cells/cytology , Time Factors
7.
Dermatology ; 238(1): 53-59, 2022.
Article in English | MEDLINE | ID: mdl-33774629

ABSTRACT

BACKGROUND: Mycoplasma pneumoniae atypical pneumonia is frequently associated with erythema multiforme. Occasionally, a mycoplasma infection does not trigger any cutaneous but exclusively mucosal lesions. The term mucosal respiratory syndrome is employed to denote the latter condition. Available reviews do not address the possible association of mucosal respiratory syndrome with further atypical bacterial pathogens such as Chlamydophila pneumoniae, Chlamydophila psittaci, Coxiella burnetii, Francisella tularensis, or Legionella species. We therefore performed a systematic review of the literature addressing this issue in the National Library of Medicine, Excerpta Medica, and Web of Science databases. SUMMARY: We found 63 patients (≤18 years, n = 36; >18 years, n = 27; 54 males and 9 females) affected by a mucosal respiratory syndrome. Fifty-three cases were temporally associated with a M. pneumoniae and 5 with a C. pneumoniae infection. No cases temporally associated with C. psittaci, C. burnetii, F. tularensis, or Legionella species infection were found. Two cases were temporally associated with Epstein-Barr virus or influenzavirus B, respectively.


Subject(s)
Mucositis/complications , Mucositis/microbiology , Respiratory Tract Infections/complications , Respiratory Tract Infections/microbiology , Chlamydophila pneumoniae , Humans , Mucositis/diagnosis , Mycoplasma pneumoniae , Respiratory Tract Infections/diagnosis , Syndrome
8.
Life Sci ; 289: 120243, 2022 Jan 15.
Article in English | MEDLINE | ID: mdl-34922941

ABSTRACT

Intestinal mucositis (IM) is a critical side-effect associated with antineoplastic therapy. Treatment available is only palliative and often not effective. However, alternative therapeutic strategies, such as probiotics, have attracted significant attention due to their immune-modulatory action in several diseases. Thus, the present study aims to elucidate the therapeutic potential of the probiotic strain Bifidobacterium longum 51A in a murine model of mucositis induced by irinotecan. Due to the scarcity of studies on dose-response and viability (probiotic vs paraprobiotic), we first evaluated which dose and cell viability would be most effective in treating mucositis. In this study, the oral pretreatment with viable B. longum 51A at a concentration of 1 × 109 CFU/mL reduced the daily disease activity index (p < 0.01), protected the intestinal architecture, preserved the length of the intestine (p < 0.05), and reduced intestinal permeability (p < 0.01), inflammation, and oxidative damage (p < 0.01) induced by irinotecan. Also, treatment with B. longum 51A increased the production of secretory immunoglobulin A (p < 0.05) in the intestinal fluid of mice with mucositis. Furthermore, B. longum 51A reversed the mucositis-induced increase in Enterobacteriaceae bacterial group in the gut (p < 0.01). In conclusion, these results showed that oral administration of B. longum 51A protects mice against intestinal damage caused by irinotecan, suggesting its use as a potential probiotic in therapy during mucositis.


Subject(s)
Bifidobacterium longum , Gastrointestinal Microbiome/drug effects , Intestinal Diseases , Irinotecan/adverse effects , Mucositis , Probiotics/pharmacology , Animals , Female , Intestinal Diseases/chemically induced , Intestinal Diseases/microbiology , Intestinal Diseases/therapy , Irinotecan/pharmacology , Mice , Mice, Inbred BALB C , Mucositis/chemically induced , Mucositis/microbiology , Mucositis/therapy
9.
Front Immunol ; 12: 677870, 2021.
Article in English | MEDLINE | ID: mdl-34220823

ABSTRACT

Background: 16S sequencing results are often used for Machine Learning (ML) tasks. 16S gene sequences are represented as feature counts, which are associated with taxonomic representation. Raw feature counts may not be the optimal representation for ML. Methods: We checked multiple preprocessing steps and tested the optimal combination for 16S sequencing-based classification tasks. We computed the contribution of each step to the accuracy as measured by the Area Under Curve (AUC) of the classification. Results: We show that the log of the feature counts is much more informative than the relative counts. We further show that merging features associated with the same taxonomy at a given level, through a dimension reduction step for each group of bacteria improves the AUC. Finally, we show that z-scoring has a very limited effect on the results. Conclusions: The prepossessing of microbiome 16S data is crucial for optimal microbiome based Machine Learning. These preprocessing steps are integrated into the MIPMLP - Microbiome Preprocessing Machine Learning Pipeline, which is available as a stand-alone version at: https://github.com/louzounlab/microbiome/tree/master/Preprocess or as a service at http://mip-mlp.math.biu.ac.il/Home Both contain the code, and standard test sets.


Subject(s)
Bacteria/classification , Bacteria/genetics , Colitis/microbiology , Gastrointestinal Microbiome/genetics , Machine Learning , Mucositis/microbiology , Adult , Animals , Colitis/metabolism , Female , Hematopoietic Stem Cell Transplantation , Humans , Interleukin-1alpha/metabolism , Mice , Mucositis/metabolism , Neural Networks, Computer , Phylogeny , Pregnancy , Progesterone/metabolism , RNA, Ribosomal, 16S/genetics
11.
Bull Cancer ; 108(7-8): 761-770, 2021.
Article in French | MEDLINE | ID: mdl-33933290

ABSTRACT

Mucositis is defined as inflammatory and/or ulcerative lesions of the oral and/or gastrointestinal tract. It occurs in approximately 40% to 50% of adults patients receiving conventional chemotherapy and up to 75% of patients receiving high dose chemotherapy as conditioning for hematopoietic stem cell transplantation. It is a toxic side effect, which strongly impairs quality of life and leads to refractory pain, increasing risk of infection and even therapeutic modifications. Despite improvements made, the management of mucositis remains a challenge and is still not consensual. A multicentric survey of practices concerning the preventive and curative management of chemo-induced mucositis in pediatric oncology department in France was carried out using a standardized questionnaire. Results confirm heterogeneous practices and the small progress made during the last decade. This national survey and an analysis of the recent literature leads to propose guidelines for the prevention and treatment of oral mucositis in children receiving treatment for cancer.


Subject(s)
Antineoplastic Agents/adverse effects , Mucositis/chemically induced , Practice Patterns, Physicians'/standards , Cancer Care Facilities/statistics & numerical data , Child , France , Health Care Surveys/statistics & numerical data , Hematopoietic Stem Cell Transplantation , Humans , Laser Therapy/methods , Mucositis/complications , Mucositis/microbiology , Mucositis/prevention & control , Oral Hygiene , Pain Management , Quality of Life , Transplantation Conditioning/adverse effects
12.
Anaerobe ; 68: 102361, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33781900

ABSTRACT

Colorectal cancer (CRC) has become a serious threat to human life and health. Most patients are diagnosed at the late stage of advanced CRC, resulting in losing their best opportunity for surgical treatment. Chemotherapy plays a crucial role in the control and treatment of advanced CRC. However, the cytotoxicity of chemotherapeutic drugs can easily cause the imbalance of gut flora, damage the barrier of the gastrointestinal mucosa, and mediate mucosal inflammation of the digestive tract, which is called "gastrointestinal mucositis." This mucositis can affect the quality of life of the host and even threaten their lives. Several studies reported the association between chemotherapy-mediated gastrointestinal mucositis in CRC and gut dysbiosis. However, the underlying mechanisms of this association are still unclear. The alternative or complementary treatments to reshape gut microbiota and slow down the side effects of chemotherapy have shown the improvement of gastrointestinal mucositis following chemotherapy in the CRC condition. This review will summarize and discuss the evidence of the association between chemotherapy-mediated gastrointestinal mucositis in CRC and altered gut microbiota from in vivo and clinical studies. The possible mechanisms of gastrointestinal mucositis, including the destruction of the gastrointestinal mucosal barrier, the induction of gut dysbiosis, and histopathological changes in the gut of CRC with chemotherapy will be illustrated. In addition, the nonpharmacological interventions and phytochemical extracts by using the manipulation of the microbial population for therapeutic purposes for relieving side effects of chemotherapy as well as a cancer treatment would be summarized and discussed in this review.


Subject(s)
Antineoplastic Agents/therapeutic use , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/microbiology , Gastrointestinal Microbiome/drug effects , Animals , Antineoplastic Agents/adverse effects , Colorectal Neoplasms/pathology , Gastrointestinal Tract/microbiology , Gastrointestinal Tract/pathology , Humans , Mucositis/etiology , Mucositis/microbiology , Mucositis/pathology
13.
Dermatology ; 237(2): 230-235, 2021.
Article in English | MEDLINE | ID: mdl-32222707

ABSTRACT

BACKGROUND: Mycoplasma pneumoniae pneumonia is sometimes associated with skin or mucous membrane eruptions. Available reviews do not address the association of Chlamydophila pneumoniae pneumonia with skin eruptions. We therefore conducted a systematic review of the literature addressing this issue. The National Library of Medicine, Excerpta Medica, and Web of Science databases were employed. SUMMARY: In two reports, skin lesions and especially urticaria were more common (p < 0.05) in atypical pneumonia caused by C. pneumoniae as compared with M. pneumoniae. We found 47 patients (<18 years, n = 16; ≥18 years, n = 31) affected by a C. pneumoniae atypical pneumonia, which was associated with erythema nodosum, erythema multiforme minus, erythema multiforme majus, isolated mucositis, or cutaneous vasculitis. We also found the case of a boy with C. pneumoniae pneumonia and acute generalized exanthematous pustulosis. We did not find any case of C. pneumoniae respiratory infection associated with either Gianotti-Crosti syndrome, pityriasis lichenoides et varioliformis acuta Mucha-Habermann, or varicella-like skin eruptions.


Subject(s)
Chlamydophila Infections/complications , Chlamydophila pneumoniae , Pneumonia/complications , Pneumonia/microbiology , Skin Diseases/microbiology , Erythema Multiforme/microbiology , Erythema Nodosum/microbiology , Humans , Mucositis/microbiology , Skin Diseases, Vascular/microbiology , Urticaria/microbiology
14.
Clin Exp Dermatol ; 46(3): 420-429, 2021 Apr.
Article in English | MEDLINE | ID: mdl-32918499

ABSTRACT

Mycoplasma pneumoniae-induced rash and mucositis is the most accurate diagnosis for patients with blistering mucocutaneous disease provoked by an infection. Recent literature suggests expansion of the name is required, as other infections have caused a clinically similar presentation. This review provides a concise update on current understanding of M. pneumoniae-induced rash and mucositis and other reactive infectious mucocutaneous eruptions.


Subject(s)
Exanthema/microbiology , Mucositis/microbiology , Mycoplasma Infections/diagnosis , Mycoplasma pneumoniae , Anti-Bacterial Agents/therapeutic use , Diagnosis, Differential , Exanthema/diagnosis , Humans , Incidence , Mucositis/diagnosis , Mycoplasma Infections/classification , Mycoplasma Infections/drug therapy , Mycoplasma Infections/epidemiology , Stevens-Johnson Syndrome/diagnosis , Stevens-Johnson Syndrome/microbiology , Terminology as Topic
16.
J Appl Microbiol ; 130(6): 2063-2074, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33128836

ABSTRACT

AIMS: To evaluate a mixture of selected lactic acid bacteria (LAB) (a riboflavin-producer, a folate-producer and an immunomodulatory strain) as co-adjuvant for 5-fluorouracil (5-FU) chemotherapy in cell culture and using a 4T1 cell animal model of breast cancer. METHODS AND RESULTS: The viability of Caco-2 cells exposed to 5-FU and/or LAB was analysed. Mice bearing breast tumour were treated with 5-FU and/or LAB. Tumour growth was measured. Intestinal mucositis (IM) was evaluated in small intestine; haematological parameters and plasma cytokines were determined. The bacterial mixture did not negatively affect the cytotoxic activity of 5-FU on Caco-2 cells. The LAB mixture attenuated the IM and prevented blood cell decreases associated with 5-FU treatment. Mice that received 5-FU and LAB mixture decreased tumour growth and showed modulation of systemic cytokines modified by both tumour growth and 5-FU treatment. The LAB mixture by itself delayed tumour growth. CONCLUSIONS: The mixture of selected LAB was able to reduce the side-effects associated with chemotherapy without affecting its primary anti-tumour activity. SIGNIFICANCE AND IMPACT OF THE STUDY: This bacterial mixture could prevent the interruption of conventional oncologic therapies by reducing undesirable side-effects. In addition, this blend would provide essential nutrients (vitamins) to oncology patients.


Subject(s)
Adjuvants, Immunologic , Breast Neoplasms/therapy , Fluorouracil/therapeutic use , Lactobacillales/immunology , Lactobacillales/metabolism , Animals , Antineoplastic Agents/therapeutic use , Caco-2 Cells , Cell Line , Cell Survival , Cytokines/blood , Disease Models, Animal , Female , Folic Acid/metabolism , Humans , Immunomodulation , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Mice , Mice, Inbred BALB C , Mucositis/microbiology , Mucositis/pathology , Riboflavin/metabolism , Vitamins
17.
Biomed Pharmacother ; 133: 111012, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33254017

ABSTRACT

The beneficial effects of prebiotic, such as fructo-oligosaccharides (FOS), in intestinal inflammation have been demonstrated in several studies. Herein, we evaluate whether joint treatment with FOS, both before and during mucositis, had additional beneficial effects and investigated the mechanisms underlying in the action of FOS on the intestinal barrier. BALB/c mice were randomly divided into five groups: CTR (without mucositis + saline solution), FOS (without mucositis + 6 % FOS), MUC (mucositis + saline solution), PT (mucositis + 6 % FOS supplementation before disease induction), and TT (mucositis + 6 % FOS supplementation before and during disease induction). Mucositis was induced by intraperitoneal injection (300 mg/kg) of 5-fluorouracil (5-FU). After 72 h, the animals were euthanized and intestinal permeability (IP), tight junction, bacterial translocation (BT), histology and morphometry, and immunoglobulin A secretory (sIgA), inflammatory infiltrate, and production of short-chain fatty acids (acetate, butyrate and propionate) were evaluated. The MUC group showed an increase in the IP, BT, and inflammatory infiltrate but a decrease in the tight junction expression and butyrate and propionate levels (P < 0.05). In the PT and TT groups, FOS supplementation maintained the IP, tight junction expression, and propionate concentration within physiologic levels, increased butyrate levels, and reduced BT and inflammatory infiltrate (P < 0.05). Total treatment with FOS (TT group) was more effective in maintaining histological score, morphometric parameters, and sIgA production. Thus, total treatment (prophylactic and therapeutic supplementation) with FOS was more effective than pretreatment alone, in reducing 5-FU-induced damage to the intestinal barrier.


Subject(s)
Bacteria/drug effects , Fatty Acids, Volatile/metabolism , Gastrointestinal Microbiome/drug effects , Ileum/drug effects , Intestinal Mucosa/drug effects , Mucositis/chemically induced , Oligosaccharides/pharmacology , Prebiotics , Tight Junctions/drug effects , Acetates/metabolism , Animals , Bacteria/metabolism , Bacterial Translocation/drug effects , Butyrates/metabolism , Disease Models, Animal , Fluorouracil , Ileum/metabolism , Ileum/microbiology , Ileum/pathology , Immunoglobulin A, Secretory/metabolism , Inflammation Mediators/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Male , Mice, Inbred BALB C , Mucositis/metabolism , Mucositis/microbiology , Mucositis/pathology , Permeability , Propionates/metabolism , Tight Junctions/metabolism , Tight Junctions/microbiology , Tight Junctions/pathology
18.
Biomed Pharmacother ; 134: 111152, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33373916

ABSTRACT

BACKGROUND: 5-Fluorouracil (5-FU)-based chemotherapy is first-line chemotherapy for colorectal cancer. However, 5-FU-induced intestinal mucositis (FUIIM) is a common adverse effect that severely impairs drug tolerance and results in poor patient health. METHODS: Male C57BL/6 mice were given 5-FU (50 mg/kg/day, i.p.) and treated with MPH-966 (5 and 7.5 mg/kg/day, p.o.) for five days. The body weight loss and the amount of food intake, and histopathological findings were recorded and analyzed. In addition, the neutrophil infiltration, levels of neutrophil serine proteases and pro-inflammatory cytokines, and tight junction proteins expression in intestinal tissues were determined. The ecology of gut microbiota was performed through next-generation sequencing technologies. RESULTS: Neutrophil elastase (NE) overexpression is a key feature in FUIIM. This study showed that treatment with the specific NE inhibitor MPH-966 (7.5 mg/kg/day, p.o.) significantly reversed 5-FU-induced loss in body weight and food intake; reversed villous atrophy; significantly suppressed myeloperoxidase, NE, and proteinase 3 activity; and reduced pro-inflammatory cytokine expression in an FUIIM mouse model. In addition, MPH-966 prevented 5-FU-induced intestinal barrier dysfunction, as indicated by the modulated expression of the tight junction proteins zonula occludin-1 and occludin. MPH-966 also reversed 5-FU-induced changes in gut microbiota diversity and abundances, specifically the Firmicutes-to-Bacteroidetes ratio; Muribaculaceae, Ruminococcaceae, and Eggerthellaceae abundances at the family level; and Candidatus Arthromitus abundance at the genus level. CONCLUSION: These data indicate that NE inhibitor is a key treatment candidate to alleviate FUIIM by regulating abnormal inflammatory responses, intestinal barrier dysfunction, and gut microbiota imbalance.


Subject(s)
Gastrointestinal Microbiome/drug effects , Intestinal Mucosa/drug effects , Leukocyte Elastase/antagonists & inhibitors , Mucositis/prevention & control , Neutrophils/drug effects , Serine Proteinase Inhibitors/pharmacology , Animals , Cell Line , Cytokines/metabolism , Disease Models, Animal , Dysbiosis , Fluorouracil , Inflammation Mediators/metabolism , Intestinal Mucosa/enzymology , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Leukocyte Elastase/metabolism , Male , Mice, Inbred C57BL , Mucositis/enzymology , Mucositis/microbiology , Mucositis/pathology , Neutrophil Infiltration/drug effects , Neutrophils/enzymology , Occludin/metabolism , Permeability , Rats , Zonula Occludens-1 Protein/metabolism
19.
Microbiome ; 8(1): 112, 2020 07 25.
Article in English | MEDLINE | ID: mdl-32711581

ABSTRACT

BACKGROUND: The increasing incidence of cancer and intestinal mucositis induced by chemotherapeutics are causing worldwide concern. Many approaches such as fecal microbiota transplantation (FMT) have been used to minimize mucositis. However, it is still unknown whether FMT from a donor with beneficial gut microbiota results in more effective intestinal function in the recipient. Recently, we found that alginate oligosaccharides (AOS) benefit murine gut microbiota through increasing "beneficial" microbes to rescue busulfan induced mucositis. RESULTS: In the current investigation, FMT from AOS-dosed mice improved small intestine function over FMT from control mice through the recovery of gene expression and an increase in the levels of cell junction proteins. FMT from AOS-dosed mice showed superior benefits over FMT from control mice on recipient gut microbiotas through an increase in "beneficial" microbes such as Leuconostocaceae and recovery in blood metabolome. Furthermore, the correlation of gut microbiota and blood metabolites suggested that the "beneficial" microbe Lactobacillales helped with the recovery of blood metabolites, while the "harmful" microbe Mycoplasmatales did not. CONCLUSION: The data confirm our hypothesis that FMT from a donor with superior microbes leads to a more profound recovery of small intestinal function. We propose that gut microbiota from naturally produced AOS-treated donor may be used to prevent small intestinal mucositis induced by chemotherapeutics or other factors in recipients. Video Abstract.


Subject(s)
Alginates/pharmacology , Fecal Microbiota Transplantation , Intestine, Small/microbiology , Intestine, Small/pathology , Microbiota/drug effects , Mucositis/microbiology , Mucositis/therapy , Oligosaccharides/pharmacology , Animals , Busulfan/adverse effects , Intestine, Small/drug effects , Male , Mice , Mice, Inbred ICR , Mucositis/chemically induced
SELECTION OF CITATIONS
SEARCH DETAIL
...