1
|
Ignatiou A, Pitsouli C. Host-diet-microbiota interplay in intestinal nutrition and health. FEBS Lett 2024; 598:2482-2517. [PMID: 38946050 DOI: 10.1002/1873-3468.14966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Accepted: 06/11/2024] [Indexed: 07/02/2024]
Abstract
The intestine is populated by a complex and dynamic assortment of microbes, collectively called gut microbiota, that interact with the host and contribute to its metabolism and physiology. Diet is considered a key regulator of intestinal microbiota, as ingested nutrients interact with and shape the resident microbiota composition. Furthermore, recent studies underscore the interplay of dietary and microbiota-derived nutrients, which directly impinge on intestinal stem cells regulating their turnover to ensure a healthy gut barrier. Although advanced sequencing methodologies have allowed the characterization of the human gut microbiome, mechanistic studies assessing diet-microbiota-host interactions depend on the use of genetically tractable models, such as Drosophila melanogaster. In this review, we first discuss the similarities between the human and fly intestines and then we focus on the effects of diet and microbiota on nutrient-sensing signaling cascades controlling intestinal stem cell self-renewal and differentiation, as well as disease. Finally, we underline the use of the Drosophila model in assessing the role of microbiota in gut-related pathologies and in understanding the mechanisms that mediate different whole-body manifestations of gut dysfunction.
Collapse
Affiliation(s)
- Anastasia Ignatiou
- Department of Biological Sciences, University of Cyprus, Nicosia, Cyprus
| | - Chrysoula Pitsouli
- Department of Biological Sciences, University of Cyprus, Nicosia, Cyprus
| |
Collapse
|
2
|
Al-Asfour A, Bhardwaj RG, Karched M. Growth Suppression of Oral Squamous Cell Carcinoma Cells by Lactobacillus Acidophilus. Int Dent J 2024; 74:1151-1160. [PMID: 38679518 PMCID: PMC11561490 DOI: 10.1016/j.identj.2024.03.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 03/18/2024] [Accepted: 03/28/2024] [Indexed: 05/01/2024] Open
Abstract
OBJECTIVES Oral squamous cell carcinoma (OSCC) is a highly aggressive form of oral cancer. Probiotic lactobacilli have demonstrated anticancer effects, whilst their interaction with Streptococcus mutans in this context remains unexplored. The objective of this study was to investigate the antiproliferative effect of Lactobacillus acidophilus on OSCC and to understand the effect of S mutans on OSCCs and whether it affects the antiproliferative potential of L acidophilus when co-exposed to OSCC. METHODS The human head and neck squamous cell carcinoma cells of the oral cavity (HNO97 cell line) were exposed to cultures of L acidophilus and S mutans separately and in combination. Further, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay was performed to assess the viability of HNO97 cells. Bacterial adhesion to HNO97 cells was examined by confocal microscopy and apoptosis by Nexin staining. To understand the underlying mechanism of apoptosis, expression of the tumour necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) gene and protein were determined by real-time polymerase chain reaction and quantitative enzyme-linked immunosorbent assay, respectively. RESULTS A significant decrease (53%-56%) in the viability of HNO97 cells on exposure to L acidophilus, S mutans, and the 2 species together demonstrated the antiproliferative activity of L acidophilus and S mutans. Both bacteria showed adhesion to HNO97 cells. The expression of the TRAIL gene increased 5-fold in HNO97 cells on treatment with L acidophilus and S mutans, which further increased to ∼17-fold with both species present. Expression levels of the TRAIL protein were significantly (P < .05) increased in bacteria-treated cell lysates. Further, bacteria-treated HNO97 cells exhibited lower live and intact cell percentages with higher proportions of cells in early and late apoptotic stages. CONCLUSIONS L acidophilus exhibits the antiproliferative activity against OSCC cells possibly partially via a TRAIL-induced mechanism of apoptosis, which is not affected by the presence of S mutans. These findings may encourage further investigation into the possible therapeutic application of probiotic L acidophilus in OSCC.
Collapse
Affiliation(s)
- Adel Al-Asfour
- Department of Surgical Sciences, College of Dentistry, Kuwait University, Kuwait City, Kuwait
| | - Radhika G Bhardwaj
- Oral Microbiology Research Laboratory, Department of Bioclinical Sciences, College of Dentistry, Kuwait University, Kuwait City, Kuwait; Department of Biotechnology, School of Arts and Science, American International University, Kuwait
| | - Maribasappa Karched
- Oral Microbiology Research Laboratory, Department of Bioclinical Sciences, College of Dentistry, Kuwait University, Kuwait City, Kuwait.
| |
Collapse
|
3
|
Mobasherpour P, Yavarmanesh M, Edalatian Dovom MR. Antitumor properties of traditional lactic acid bacteria: Short-chain fatty acid production and interleukin 12 induction. Heliyon 2024; 10:e36183. [PMID: 39253228 PMCID: PMC11382311 DOI: 10.1016/j.heliyon.2024.e36183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 08/11/2024] [Accepted: 08/12/2024] [Indexed: 09/11/2024] Open
Abstract
This paper presents an in vitro evaluation of antitumor properties through producing short-chain fatty acids and inducing interleukin 12. In addition, it offers the most important and functional probiotic properties of 24 Lactobacillus gasseri, Lactiplantibacillus plantarum, Lactobacillus acidophilus, and Limosilactobacillus fermentum strains isolated from humans, foods, and fermented foods. To this end, survival in an acidic environment (pH = 2.5), tolerance in bile salt, viability in the presence of pepsin-pancreatin, adhesion percentage, antibiotic resistance, auto-aggregation, and potential percentage of co-aggregation are studied in contact with three human intestinal pathogens. These pathogens are Escherichia coli O157: H7 NCTC 12900, Salmonella enterica subsp. enterica ATCC 13076, and Listeria monocytogenes ATTC 7644. Also, in vitro induction amount of IL-12 in mouse splenocytes is investigated to evaluate antitumor properties by 19 strains of L. gasseri and L. plantarum along with the development of short-chain fatty acids (SCFA) by 5 strains of L. fermentum and L. acidophilus. Gas Chromatography Flame Ionization Detector (GC-FID) and enzyme-linked immunosorbent assay (ELISA) were used to measure short-chain fatty acids and IL-12, respectively. All strains had high viability under acidic conditions. The highest levels of pancreatin and pepsin resistance were found in strains LF56, LF57, LF55, OF, and F and strains LF56, LF57, and A7, respectively. All strains except LF56 had high resistance to bile salts. L. gasseri 54C had the highest average adhesion score (hydrophobicity) of 62.9 % among 19 strains. Despite the susceptibility of different strains of L. plantarum to the tested antibiotics, M8 and M11, S2G, A7, LF55, LF57, and 5G were resistant to kanamycin and chloramphenicol, respectively. Also, 21G was resistant to ampicillin, LF56 to tetracycline and M8, and M11, LF56, and 21G to Erythromycin. In addition, L. gasseri showed moderate resistance to ampicillin, erythromycin, and tetracycline, while L. fermentum ATCC 9338 showed good resistance to ampicillin, erythromycin, and chloramphenicol. In this respect, L. plantarum LF56 and gasseri 54C had the highest average auto-aggregation and co-aggregation against three pathogenic bacteria, respectively. The highest and lowest levels of acetic acid as short-chain fatty acids were produced by L. fermentum 19SH isolated from Horre 41.62 and L. fermentum 21SH from fermented seeds 27.047, respectively. Moreover, L. fermentum, with the OF code of traditional-fermented food origin, produced the most isobutyric acid, butyric acid, and valeric acid, with values of 0.6828, 0.74165, and 0.49915 mmol, respectively. L. fermentum isolated from the human origin with code F produced the most isovaleric acid of 1.1874 mmol. All the tested strains produced good propionic acid except L. fermentum 21SH from fermented seeds. Among strains, L. plantarum M11 isolated from milk and L. gasseri 52B from humans had the highest in vitro induction of IL-12, which is probably related to their cell wall compositions and structure.
Collapse
Affiliation(s)
- Parinaz Mobasherpour
- Department of Food Science and Technology, Faculty of Agriculture, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Masoud Yavarmanesh
- Department of Food Science and Technology, Faculty of Agriculture, Ferdowsi University of Mashhad, Mashhad, Iran
| | | |
Collapse
|
4
|
Saha B, A T R, Adhikary S, Banerjee A, Radhakrishnan AK, Duttaroy AK, Pathak S. Exploring the Relationship Between Diet, Lifestyle and Gut Microbiome in Colorectal Cancer Development: A Recent Update. Nutr Cancer 2024; 76:789-814. [PMID: 39207359 DOI: 10.1080/01635581.2024.2367266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 05/18/2024] [Accepted: 06/05/2024] [Indexed: 09/04/2024]
Abstract
Colorectal cancer (CRC) is one of the major causes of cancer-related mortality worldwide. Despite advances in treatment modalities, its prevalence continues to rise, notably among younger populations. Unhealthy dietary habits, sedentary routines, and obesity have been identified as one of the key contributors to the development of colorectal cancer, apart from genetic and epigenetic modifications. Recognizing the profound impact of diet and lifestyle on the intricate gut microbiota ecosystem offers a promising avenue for understanding CRC development and its treatment. Gut dysbiosis, characterized by imbalances favoring harmful microbes over beneficial ones, has emerged as a defining feature of CRC. Changes in diet and lifestyle can profoundly alter the composition of gut microbes and the metabolites they produce, potentially contributing to CRC onset. Focusing on recent evidence, this review discussed various dietary factors, such as high consumption of red and processed meats and low fiber intake, and lifestyle factors, including obesity, lack of physical activity, smoking, and excessive alcohol consumption, that influence the gut microbiome composition and elevate CRC risk.
Collapse
Affiliation(s)
- Biki Saha
- Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Chennai, India
| | - Rithi A T
- Department of Pharmacology, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Chennai, India
| | - Subhamay Adhikary
- Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Chennai, India
| | - Antara Banerjee
- Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Chennai, India
| | - Arun Kumar Radhakrishnan
- Department of Pharmacology, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Chennai, India
| | - Asim K Duttaroy
- Department of Nutrition, Institute of Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Surajit Pathak
- Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Chennai, India
| |
Collapse
|
5
|
Saad HM, Oda SS, Alexiou A, Papadakis M, Mahmoud MH, Batiha GES, Khalifa E. Hepatoprotective activity of Lactéol® forte and quercetin dihydrate against thioacetamide-induced hepatic cirrhosis in male albino rats. J Cell Mol Med 2024; 28:e18196. [PMID: 38534093 DOI: 10.1111/jcmm.18196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 11/20/2023] [Accepted: 02/04/2024] [Indexed: 03/28/2024] Open
Abstract
Liver cirrhosis is a silent disease in humans and is experimentally induced by many drugs and toxins as thioacetamide (TAA) in particular, which is the typical model for experimental induction of hepatic fibrosis. Thus, the objective of the present study was to elucidate the possible protective effects of lactéol® forte (LF) and quercetin dihydrate (QD) against TAA-induced hepatic damage in male albino rats. Induction of hepatotoxicity was performed by TAA injection (200 mg/kg I/P, twice/ week) in rats. LF (1 × 109 CFU/rat 5 times/week) and QD (50 mg/kg 5 times/week) treated groups were administered concurrently with TAA injection (200 mg/kg I/P, twice/ week). The experimental treatments were conducted for 12 weeks. Hepatotoxicity was evaluated biochemically by measuring alanine aminotransferase (ALT), aspartate aminotransferase (AST) and gamma-glutamyl transferase (GGT) in the serum and histopathologically with the scoring of histopathological changes besides histochemical assessment of collagen by Masson's trichrome and immunohistochemical analysis for α-smooth muscle actin (α-SMA), Ki67 and caspase-3 expression in liver sections. Our results indicated that LF and QD attenuated some biochemical changes and histochemical markers in TAA-mediated hepatotoxicity in rats by amelioration of biochemical markers and collagen, α-SMA, Ki67 and caspase3 Immunoexpression. Additionally, LF and QD supplementation downregulated the proliferative, necrotic, fibroblastic changes, eosinophilic intranuclear inclusions, hyaline globules and Mallory-like bodies that were detected histopathologically in the TAA group. In conclusion, LF showed better hepatic protection than QD against TAA-induced hepatotoxicity in rats by inhibiting inflammatory reactions with the improvement of some serum hepatic transaminases, histopathological picture and immunohistochemical markers.
Collapse
Affiliation(s)
- Hebatallah M Saad
- Department of Pathology, Faculty of Veterinary Medicine, Matrouh University, Matrouh, Egypt
| | - Samah S Oda
- Department of Pathology, Faculty of Veterinary Medicine, Alexandria University, Abees, Alexandria Province, Egypt
| | - Athanasios Alexiou
- University Centre for Research & Development, Chandigarh University, Mohali, Punjab, India
- Department of Research & Development, Funogen, Athens, Greece
- Department of Research & Development, AFNP Med, Wien, Austria
- Department of Science and Engineering, Novel Global Community Educational Foundation, Hebersham, New South Wales, Germany
| | - Marios Papadakis
- Department of Surgery II, University Hospital Witten-Herdecke, Heusnerstrasse 40, University of Witten-Herdecke, Wuppertal, Germany
| | - Mohamed H Mahmoud
- Department of Biochemistry, College of Science, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, AlBeheira, Egypt
| | - Eman Khalifa
- Department of Microbiology, Faculty of Veterinary Medicine, Matrouh University, Matrouh, Egypt
| |
Collapse
|
6
|
Reis SK, Socca EAR, de Souza BR, Genaro SC, Durán N, Fávaro WJ. Effects of probiotic supplementation on chronic inflammatory process modulation in colorectal carcinogenesis. Tissue Cell 2024; 87:102293. [PMID: 38244400 DOI: 10.1016/j.tice.2023.102293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 12/17/2023] [Accepted: 12/20/2023] [Indexed: 01/22/2024]
Abstract
The current study investigated the potential effects of probiotic supplementation on colorectal carcinogenesis chemically induced with 1,2-dimethylhydrazine (DMH) and treated with 5-fluorouracil (5FU)-based chemotherapy in mice. Animals were randomly allocated in five different groups: Control: which not receive any treatment throughout the experimental course; Colitis model group (DMH): treated with DMH; DMH+ 5FU: animals received I.P. (intraperitoneal) dose of chemotherapy on a weekly basis; DMH+PROB: animals received daily administrations (via gavage) of probiotics (Lactobacillus: acidophilus and paracasei, Bifidobacterium lactis and bifidum); and DMH+ PROB+ 5FU: animals received the same treatment as the previous groups. After ten-week treatment, mice's large intestine was collected and subjected to colon length, histopathological, periodic acid-schiff (PAS) staining and immunohistochemistry (TLR2, MyD88, NF-κB, IL-6, TLR4, TRIF, IRF-3, IFN-γ, Ki-67, KRAS, p53, IL-10, and TGF-β) analyzes. Variance (ANOVA) and Kruskal-Wallis tests were used for statistical analysis, at significance level p 0.05. Probiotics' supplementation has increased the production of Ki-67 cell-proliferation marker, reduced body weight, and colon shortening, as well as modulated the chronic inflammatory process in colorectal carcinogenesis by inhibiting NF-κB expression and mitigating mucin depletion. Thus, these findings lay a basis for guide future studies focused on probiotics' action mechanisms in tumor microenvironment which might have implications in clinical practice.
Collapse
Affiliation(s)
- Sabrina Karen Reis
- Faculty Medical Sciences, State University of Campinas (UNICAMP), Campinas, SP, Brazil; Laboratory of Urogenital Carcinogenesis and Immunotherapy, Department of Structural and Functional Biology, State University of Campinas (UNICAMP), Campinas, SP, Brazil.
| | - Eduardo Augusto Rabelo Socca
- Laboratory of Urogenital Carcinogenesis and Immunotherapy, Department of Structural and Functional Biology, State University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Bianca Ribeiro de Souza
- British Columbia's Gynecological Cancer Research (OVCARE) Program and Department of Obstetrics and Gynecology, University of British Columbia, Vancouver General Hospital, Vancouver, BC, Canada.
| | | | - Nelson Durán
- Laboratory of Urogenital Carcinogenesis and Immunotherapy, Department of Structural and Functional Biology, State University of Campinas (UNICAMP), Campinas, SP, Brazil; Nanomedicine Research Unit (Nanomed), Federal University of ABC (UFABC), Santo André, SP, Brazil
| | - Wagner José Fávaro
- Faculty Medical Sciences, State University of Campinas (UNICAMP), Campinas, SP, Brazil; Laboratory of Urogenital Carcinogenesis and Immunotherapy, Department of Structural and Functional Biology, State University of Campinas (UNICAMP), Campinas, SP, Brazil
| |
Collapse
|
7
|
Roy R, Singh SK. The Microbiome Modulates the Immune System to Influence Cancer Therapy. Cancers (Basel) 2024; 16:779. [PMID: 38398170 PMCID: PMC10886470 DOI: 10.3390/cancers16040779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 02/08/2024] [Accepted: 02/08/2024] [Indexed: 02/25/2024] Open
Abstract
The gut microbiota composition can affect the tumor microenvironment and its interaction with the immune system, thereby having implications for treatment predictions. This article reviews the studies available to better understand how the gut microbiome helps the immune system fight cancer. To describe this fact, different mechanisms and approaches utilizing probiotics to improve advancements in cancer treatment will be discussed. Moreover, not only calorie intake but also the variety and quality of diet can influence cancer patients' immunotherapy treatment because dietary patterns can impair immunological activities either by stimulating or suppressing innate and adaptive immunity. Therefore, it is interesting and critical to understand gut microbiome composition as a biomarker to predict cancer immunotherapy outcomes and responses. Here, more emphasis will be given to the recent development in immunotherapies utilizing microbiota to improve cancer therapies, which is beneficial for cancer patients.
Collapse
Affiliation(s)
- Ruchi Roy
- UICentre for Drug Discovery, The University of Illinois, Chicago, IL 60612, USA
| | - Sunil Kumar Singh
- Department of Surgery, Division of Surgical Oncology, The University of Illinois at Chicago, Chicago, IL 60612, USA
| |
Collapse
|
8
|
Gou H, Su H, Liu D, Wong CC, Shang H, Fang Y, Zeng X, Chen H, Li Y, Huang Z, Fan M, Wei C, Wang X, Zhang X, Li X, Yu J. Traditional Medicine Pien Tze Huang Suppresses Colorectal Tumorigenesis Through Restoring Gut Microbiota and Metabolites. Gastroenterology 2023; 165:1404-1419. [PMID: 37704113 DOI: 10.1053/j.gastro.2023.08.052] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 08/02/2023] [Accepted: 08/26/2023] [Indexed: 09/15/2023]
Abstract
BACKGROUND & AIMS Pien Tze Huang (PZH) is a well-established traditional medicine with beneficial effects against inflammation and cancer. We aimed to explore the chemopreventive effect of PZH in colorectal cancer (CRC) through modulating gut microbiota. METHODS CRC mouse models were established by azoxymethane plus dextran sulfate sodium treatment or in Apcmin/+ mice treated with or without PZH (270 mg/kg and 540 mg/kg). Gut barrier function was determined by means of intestinal permeability assays and transmission electron microscopy. Fecal microbiota and metabolites were analyzed by means of metagenomic sequencing and liquid chromatography mass spectrometry, respectively. Germ-free mice or antibiotic-treated mice were used as models of microbiota depletion. RESULTS PZH inhibited colorectal tumorigenesis in azoxymethane plus dextran sulfate sodium-treated mice and in Apcmin/+ mice in a dose-dependent manner. PZH treatment altered the gut microbiota profile, with an increased abundance of probiotics Pseudobutyrivibrio xylanivorans and Eubacterium limosum, while pathogenic bacteria Aeromonas veronii, Campylobacter jejuni, Collinsella aerofaciens, and Peptoniphilus harei were depleted. In addition, PZH increased beneficial metabolites taurine and hypotaurine, bile acids, and unsaturated fatty acids, and significantly restored gut barrier function. Transcriptomic profiling revealed that PZH inhibited PI3K-Akt, interleukin-17, tumor necrosis factor, and cytokine-chemokine signaling. Notably, the chemopreventive effect of PZH involved both microbiota-dependent and -independent mechanisms. Fecal microbiota transplantation from PZH-treated mice to germ-free mice partly recapitulated the chemopreventive effects of PZH. PZH components ginsenoside-F2 and ginsenoside-Re demonstrated inhibitory effects on CRC cells and primary organoids, and PZH also inhibited tumorigenesis in azoxymethane plus dextran sulfate sodium-treated germ-free mice. CONCLUSIONS PZH manipulated gut microbiota and metabolites toward a more favorable profile, improved gut barrier function, and suppressed oncogenic and pro-inflammatory pathways, thereby suppressing colorectal carcinogenesis.
Collapse
Affiliation(s)
- Hongyan Gou
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shenzhen Research Institute, Hong Kong Special Administrative Region, China
| | - Hao Su
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shenzhen Research Institute, Hong Kong Special Administrative Region, China
| | - Dehua Liu
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shenzhen Research Institute, Hong Kong Special Administrative Region, China
| | - Chi Chun Wong
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shenzhen Research Institute, Hong Kong Special Administrative Region, China
| | - Haiyun Shang
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shenzhen Research Institute, Hong Kong Special Administrative Region, China
| | - Yi Fang
- Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xianyi Zeng
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shenzhen Research Institute, Hong Kong Special Administrative Region, China
| | - Huarong Chen
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shenzhen Research Institute, Hong Kong Special Administrative Region, China; Department of Anesthesia and Intensive Care and Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Yan Li
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shenzhen Research Institute, Hong Kong Special Administrative Region, China
| | - Ziheng Huang
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shenzhen Research Institute, Hong Kong Special Administrative Region, China
| | - Miao Fan
- Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Chunxian Wei
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shenzhen Research Institute, Hong Kong Special Administrative Region, China
| | - Xin Wang
- Department of Pathology, the First Hospital of Hebei Medical University, Hebei, China
| | - Xiang Zhang
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shenzhen Research Institute, Hong Kong Special Administrative Region, China
| | - Xiaoxing Li
- Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jun Yu
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shenzhen Research Institute, Hong Kong Special Administrative Region, China.
| |
Collapse
|
9
|
Luo S, Ru J, Mirzaei MK, Xue J, Peng X, Ralser A, Mejías-Luque R, Gerhard M, Deng L. Gut virome profiling identifies an association between temperate phages and colorectal cancer promoted by Helicobacter pylori infection. Gut Microbes 2023; 15:2257291. [PMID: 37747149 PMCID: PMC10578192 DOI: 10.1080/19490976.2023.2257291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 09/06/2023] [Indexed: 09/26/2023] Open
Abstract
Colorectal cancer (CRC) is one of the most commonly diagnosed cancers worldwide. While a close correlation between chronic Helicobacter pylori infection and CRC has been reported, the role of the virome has been overlooked. Here, we infected Apc-mutant mouse models and C57BL/6 mice with H. pylori and conducted a comprehensive metagenomics analysis of H. pylori-induced changes in lower gastrointestinal tract bacterial and viral communities. We observed an expansion of temperate phages in H. pylori infected Apc+/1638N mice at the early stage of carcinogenesis. Some of the temperate phages were predicted to infect bacteria associated with CRC, including Enterococcus faecalis. We also observed a high prevalence of virulent genes, such as flgJ, cwlJ, and sleB, encoded by temperate phages. In addition, we identified phages associated with pre-onset and onset of H. pylori-promoted carcinogenesis. Through co-occurrence network analysis, we found strong associations between the viral and bacterial communities in infected mice before the onset of carcinogenesis. These findings suggest that the expansion of temperate phages, possibly caused by prophage induction triggered by H. pylori infection, may have contributed to the development of CRC in mice by interacting with the bacterial community.
Collapse
Affiliation(s)
- Shiqi Luo
- Institute of Virology, Helmholtz Centre Munich — German Research Centre for Environmental Health, Neuherberg, Germany
- Chair for Preventions of Microbial Diseases, School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Jinlong Ru
- Institute of Virology, Helmholtz Centre Munich — German Research Centre for Environmental Health, Neuherberg, Germany
- Chair for Preventions of Microbial Diseases, School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Mohammadali Khan Mirzaei
- Institute of Virology, Helmholtz Centre Munich — German Research Centre for Environmental Health, Neuherberg, Germany
- Chair for Preventions of Microbial Diseases, School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Jinling Xue
- Institute of Virology, Helmholtz Centre Munich — German Research Centre for Environmental Health, Neuherberg, Germany
- Chair for Preventions of Microbial Diseases, School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Xue Peng
- Institute of Virology, Helmholtz Centre Munich — German Research Centre for Environmental Health, Neuherberg, Germany
- Faculty of Biology, Biocenter, Ludwig Maximilian University of Munich, Munich, Germany
| | - Anna Ralser
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich, Munich, Germany
| | - Raquel Mejías-Luque
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich, Munich, Germany
- German Center for Infection Research (DZIF), Munich Partner Site, Munich, Germany
| | - Markus Gerhard
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich, Munich, Germany
- German Center for Infection Research (DZIF), Munich Partner Site, Munich, Germany
| | - Li Deng
- Institute of Virology, Helmholtz Centre Munich — German Research Centre for Environmental Health, Neuherberg, Germany
- Chair for Preventions of Microbial Diseases, School of Life Sciences, Technical University of Munich, Freising, Germany
| |
Collapse
|
10
|
Tosti E, Srivastava N, Edelmann W. Vaccination and Microbiota Manipulation Approaches for Colon Cancer Prevention in Rodent Models. Cancer Prev Res (Phila) 2023; 16:429-438. [PMID: 37012205 DOI: 10.1158/1940-6207.capr-23-0015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 03/16/2023] [Accepted: 03/31/2023] [Indexed: 04/05/2023]
Abstract
Colorectal cancer represents the third most common cancer type worldwide and is a leading cause of cancer-related mortality in the United States and Western countries. Rodent models have been invaluable to study the etiology of colorectal cancer and to test novel chemoprevention avenues. In the past, the laboratory mouse has become one of the best preclinical models for these studies due to the availability of genetic information for commonly used mouse strains with well-established and precise gene targeting and transgenic techniques. Well-established chemical mutagenesis technologies are also being used to develop mouse and rat models of colorectal cancer for prevention and treatment studies. In addition, xenotransplantation of cancer cell lines and patient-derived xenografts has been useful for preclinical prevention studies and drug development. This review focuses on the recent use of rodent models to evaluate the utility of novel strategies in the prevention of colon cancers including immune prevention approaches and the manipulation of the intestinal microbiota.
Collapse
Affiliation(s)
- Elena Tosti
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York
| | - Nityanand Srivastava
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York
| | - Winfried Edelmann
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York
| |
Collapse
|
11
|
Gholami A, Montazeri-Najafabady N, Ashoori Y, Kazemi K, Heidari R, Omidifar N, Karimzadeh I, Ommati MM, Abootalebi SN, Golkar N. The ameliorating effect of limosilactobacillus fermentum and its supernatant postbiotic on cisplatin-induced chronic kidney disease in an animal model. BMC Complement Med Ther 2023; 23:243. [PMID: 37461012 PMCID: PMC10351115 DOI: 10.1186/s12906-023-04068-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 07/04/2023] [Indexed: 07/20/2023] Open
Abstract
BACKGROUND Chronic kidney disease (CKD) is a worldwide public health problem affecting millions of people. Probiotics and postbiotics are associated with valuable compounds with antibacterial, anti-inflammatory, and immunomodulatory effects, preserving renal function in CKD patients. The current study is aimed to evaluate the efficacy of Limosilactobacillus fermentum (L. fermentum) and its postbiotic in an animal model of cisplatin-induced CKD. METHODS The animals were divided into four experimental groups (normal mice, CKD mice with no treatment, CKD mice with probiotic treatment, and CKD mice with postbiotic treatment). CKD mice were induced by a single dose of cisplatin 10 mg/kg, intraperitoneally. For 28 days, the cultured probiotic bacteria and its supernatant (postbiotic) were delivered freshly to the related groups through their daily water. Then, blood urea nitrogen (BUN) and creatinine (Cr) of plasma samples as well as glutathione (GSH), lipid peroxidation, reactive oxygen species, and total antioxidant capacity of kidneys were assessed in the experimental mice groups. In addition, histopathological studies were performed on the kidneys. RESULTS Application of L. fermentum probiotic, and especially postbiotics, significantly decreased BUN and Cr (P < 0.0001) as well as ROS formation and lipid peroxidation levels (P < 0.0001) along with increased total antioxidant capacity and GSH levels (P < 0.001). The histopathologic images also confirmed their renal protection effect. Interestingly, the postbiotic displayed more effectiveness than the probiotic in some assays. The improvement effect on renal function in the current model is mainly mediated by oxidative stress markers in the renal tissue. CONCLUSIONS In conclusion, it was found that the administration of L. fermentum probiotic, and particularly its postbiotic in cisplatin-induced CKD mice, showed promising effects and could successfully improve renal function in the animal model of CKD. Therefore, probiotics and postbiotics are considered as probably promising alternative supplements to be used for CKD.
Collapse
Affiliation(s)
- Ahmad Gholami
- Biotechnology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Nima Montazeri-Najafabady
- Biotechnology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Endocrinology and Metabolism Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Yousef Ashoori
- Biotechnology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Clinical Pharmacy, School of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Kimia Kazemi
- Biotechnology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Reza Heidari
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, P.O. Box 71348-14336, Shiraz, Iran.
| | - Navid Omidifar
- Biotechnology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Pathology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Iman Karimzadeh
- Department of Clinical Pharmacy, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Mehdi Ommati
- Henan Key Laboratory of Environmental and Animal Product Safety, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan, 471000, China
| | - Seyedeh Narjes Abootalebi
- Biotechnology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Division of Intensive Care Unit, Department of Pediatrics, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Nasim Golkar
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, P.O. Box 71348-14336, Shiraz, Iran.
- Department of Pharmaceutics, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
12
|
Singh S, Sharma P, Sarma DK, Kumawat M, Tiwari R, Verma V, Nagpal R, Kumar M. Implication of Obesity and Gut Microbiome Dysbiosis in the Etiology of Colorectal Cancer. Cancers (Basel) 2023; 15:1913. [PMID: 36980799 PMCID: PMC10047102 DOI: 10.3390/cancers15061913] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/12/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
The complexity and variety of gut microbiomes within and among individuals have been extensively studied in recent years in connection to human health and diseases. Our growing understanding of the bidirectional communication between metabolic diseases and the gut microbiome has also highlighted the significance of gut microbiome dysbiosis in the genesis and development of obesity-related cancers. Therefore, it is crucial to comprehend the possible role of the gut microbiota in the crosstalk between obesity and colorectal cancer (CRC). Through the induction of gut microbial dysbiosis, gut epithelial barrier impairment, metabolomic dysregulation, chronic inflammation, or dysregulation in energy harvesting, obesity may promote the development of colorectal tumors. It is well known that strategies for cancer prevention and treatment are most effective when combined with a healthy diet, physical activity, and active lifestyle choices. Recent studies also suggest that an improved understanding of the complex linkages between the gut microbiome and various cancers as well as metabolic diseases can potentially improve cancer treatments and overall outcomes. In this context, we herein review and summarize the clinical and experimental evidence supporting the functional role of the gut microbiome in the pathogenesis and progression of CRC concerning obesity and its metabolic correlates, which may pave the way for the development of novel prognostic tools for CRC prevention. Therapeutic approaches for restoring the microbiome homeostasis in conjunction with cancer treatments are also discussed herein.
Collapse
Affiliation(s)
- Samradhi Singh
- Indian Council of Medical Research-National Institute for Research in Environmental Health, Bhopal 462030, India
| | - Poonam Sharma
- Indian Council of Medical Research-National Institute for Research in Environmental Health, Bhopal 462030, India
| | - Devojit Kumar Sarma
- Indian Council of Medical Research-National Institute for Research in Environmental Health, Bhopal 462030, India
| | - Manoj Kumawat
- Indian Council of Medical Research-National Institute for Research in Environmental Health, Bhopal 462030, India
| | - Rajnarayan Tiwari
- Indian Council of Medical Research-National Institute for Research in Environmental Health, Bhopal 462030, India
| | - Vinod Verma
- Stem Cell Research Centre, Sanjay Gandhi Post-Graduate Institute of Medical Sciences, Lucknow 226014, India
| | - Ravinder Nagpal
- Department of Nutrition and Integrative Physiology, Florida State University, Tallahassee, FL 32302, USA
| | - Manoj Kumar
- Indian Council of Medical Research-National Institute for Research in Environmental Health, Bhopal 462030, India
| |
Collapse
|
13
|
Fuochi V, Spampinato M, Distefano A, Palmigiano A, Garozzo D, Zagni C, Rescifina A, Li Volti G, Furneri PM. Soluble peptidoglycan fragments produced by Limosilactobacillus fermentum with antiproliferative activity are suitable for potential therapeutic development: A preliminary report. Front Mol Biosci 2023; 10:1082526. [PMID: 36876040 PMCID: PMC9975264 DOI: 10.3389/fmolb.2023.1082526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 01/31/2023] [Indexed: 02/17/2023] Open
Abstract
Currently, the use of probiotic strains and their products represents a promising innovative approach as an antagonist treatment against many human diseases. Previous studies showed that a strain of Limosilactobacillus fermentum (LAC92), previously defined as Lactobacillus fermentum, exhibited a suitable amensalistic property. The present study aimed to purify the active components from LAC92 to evaluate the biological properties of soluble peptidoglycan fragments (SPFs). The cell-free supernatant (CFS) and bacterial cells were separated after 48 h of growth in MRS medium broth and treated for isolation of SPFs. Antimicrobial activity and proliferation analysis on the human cell line HTC116 were performed using technologies such as xCELLigence, count and viability, and clonogenic analysis. MALDI-MS investigation and docking analysis were performed to determine the molecular structure and hypothetical mode of action, respectively. Our results showed that the antimicrobial activity was mainly due to SPFs. Moreover, the results obtained when investigating the SPF effect on the cell line HCT116 showed substantial preliminary evidence, suggesting their significant cytostatic and quite antiproliferative properties. Although MALDI was unable to identify the molecular structure, it was subsequently revealed by analysis of the bacterial genome. The amino acid structure is called peptide 92. Furthermore, we confirmed by molecular docking studies the interaction of peptide 92 with MDM2 protein, the negative regulator of p53. This study showed that SPFs from the LAC92 strain exerted anticancer effects on the human colon cancer HCT116 cell line via antiproliferation and inducing apoptosis. These findings indicated that this probiotic strain might be a potential candidate for applications in functional products in the future. Further examination is needed to understand the specific advantages of this probiotic strain and improve its functional features to confirm these data. Moreover, deeper research on peptide 92 could increase our knowledge and help us understand if it will be possible to apply to specific diseases such as CRC.
Collapse
Affiliation(s)
- Virginia Fuochi
- Dipartimento di Scienze Biomediche e Biotecnologiche (BIOMETEC), Università di Catania, Catania, Italy.,Center of Excellence for the Acceleration of Harm Reduction (CoEHAR), University of Catania, Catania, Italy
| | - Mariarita Spampinato
- Dipartimento di Scienze Biomediche e Biotecnologiche (BIOMETEC), Università di Catania, Catania, Italy
| | - Alfio Distefano
- Dipartimento di Scienze Biomediche e Biotecnologiche (BIOMETEC), Università di Catania, Catania, Italy
| | - Angelo Palmigiano
- CNR, Institute for Polymers, Composites and Biomaterials (IPCB), Catania, Italy
| | - Domenico Garozzo
- CNR, Institute for Polymers, Composites and Biomaterials (IPCB), Catania, Italy
| | - Chiara Zagni
- Dipartimento di Scienze del Farmaco e della Salute, Università di Catania, Catania, Italy
| | - Antonio Rescifina
- Dipartimento di Scienze del Farmaco e della Salute, Università di Catania, Catania, Italy
| | - Giovanni Li Volti
- Dipartimento di Scienze Biomediche e Biotecnologiche (BIOMETEC), Università di Catania, Catania, Italy.,Center of Excellence for the Acceleration of Harm Reduction (CoEHAR), University of Catania, Catania, Italy
| | - Pio Maria Furneri
- Dipartimento di Scienze Biomediche e Biotecnologiche (BIOMETEC), Università di Catania, Catania, Italy.,Center of Excellence for the Acceleration of Harm Reduction (CoEHAR), University of Catania, Catania, Italy
| |
Collapse
|
14
|
Hadinia N, Edalatian Dovom MR, Yavarmanesh M. The effect of fermentation conditions (temperature, salt concentration, and pH) with lactobacillus strains for producing Short Chain Fatty Acids. Lebensm Wiss Technol 2022. [DOI: 10.1016/j.lwt.2022.113709] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
15
|
Role of a mixed probiotic product, VSL#3, in the prevention and treatment of colorectal cancer. Eur J Pharmacol 2022; 930:175152. [PMID: 35835181 DOI: 10.1016/j.ejphar.2022.175152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 07/03/2022] [Accepted: 07/07/2022] [Indexed: 12/09/2022]
Abstract
Colorectal cancer (CRC) is a multifactorial disease. The incidence of this type of cancer in younger patients has increased in recent years, and more strategies are needed to prevent and delay the progression of CRC. Probiotics play an adjunctive role in the prevention and treatment of CRC and can not only prevent the onset and delay the progression of disease but also reduce the side effects after the application of anti-cancer drugs. The anti-cancer effect of individual probiotics has been extensively studied, and the exact curative effect of various probiotics has been found, but the anti-cancer effect of mixed probiotics is still not well summarized. In this review, we discuss the positive effects of mixed probiotics on CRC and the related mechanisms of action, especially VSL#3 (VSL Pharmaceuticals, Inc., Gaithersburg, MD, USA), thus providing new ideas for the treatment of CRC. Moreover, we suggest the need to search for more therapeutic possibilities, especially via the research and application of synbiotics and postbiotics.
Collapse
|
16
|
Biodetoxification and Protective Properties of Probiotics. Microorganisms 2022; 10:microorganisms10071278. [PMID: 35888997 PMCID: PMC9319832 DOI: 10.3390/microorganisms10071278] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/17/2022] [Accepted: 06/22/2022] [Indexed: 11/17/2022] Open
Abstract
Probiotic consumption is recognized as being generally safe and correlates with multiple and valuable health benefits. However, the mechanism by which it helps detoxify the body and its anti-carcinogenic and antimutagenic potential is less discussed. A widely known fact is that globalization and mass food production/cultivation make it impossible to keep all possible risks under control. Scientists associate the multitude of diseases in the days when we live with these risks that threaten the population’s safety in terms of food. This review aims to explore whether the use of probiotics may be a safe, economically viable, and versatile tool in biodetoxification despite the numerous risks associated with food and the limited possibility to evaluate the contaminants. Based on scientific data, this paper focuses on the aspects mentioned above and demonstrates the probiotics’ possible risks, as well as their anti-carcinogenic and antimutagenic potential. After reviewing the probiotic capacity to react with pathogens, fungi infection, mycotoxins, acrylamide toxicity, benzopyrene, and heavy metals, we can conclude that the specific probiotic strain and probiotic combinations bring significant health outcomes. Furthermore, the biodetoxification maximization process can be performed using probiotic-bioactive compound association.
Collapse
|
17
|
Cellular Carcinogenesis: Role of Polarized Macrophages in Cancer Initiation. Cancers (Basel) 2022; 14:cancers14112811. [PMID: 35681791 PMCID: PMC9179569 DOI: 10.3390/cancers14112811] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 05/26/2022] [Accepted: 06/02/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Inflammation is a hallmark of many cancers. Macrophages are key participants in innate immunity and important drivers of inflammation. When chronically polarized beyond normal homeostatic responses to infection, injury, or aging, macrophages can express several pro-carcinogenic phenotypes. In this review, evidence supporting polarized macrophages as endogenous sources of carcinogenesis is discussed. In addition, the depletion or modulation of macrophages by small molecule inhibitors and probiotics are reviewed as emerging strategies in cancer prevention. Abstract Inflammation is an essential hallmark of cancer. Macrophages are key innate immune effector cells in chronic inflammation, parainflammation, and inflammaging. Parainflammation is a form of subclinical inflammation associated with a persistent DNA damage response. Inflammaging represents low-grade inflammation due to the dysregulation of innate and adaptive immune responses that occur with aging. Whether induced by infection, injury, or aging, immune dysregulation and chronic macrophage polarization contributes to cancer initiation through the production of proinflammatory chemokines/cytokines and genotoxins and by modulating immune surveillance. This review presents pre-clinical and clinical evidence for polarized macrophages as endogenous cellular carcinogens in the context of chronic inflammation, parainflammation, and inflammaging. Emerging strategies for cancer prevention, including small molecule inhibitors and probiotic approaches, that target macrophage function and phenotype are also discussed.
Collapse
|
18
|
Nagao-Kitamoto H, Kitamoto S, Kamada N. Inflammatory bowel disease and carcinogenesis. Cancer Metastasis Rev 2022; 41:301-316. [PMID: 35416564 DOI: 10.1007/s10555-022-10028-4] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 03/27/2022] [Indexed: 11/24/2022]
Abstract
Colorectal cancer (CRC) is the third most common cancer and the fourth most common cause of cancer mortality worldwide. Colitis-associated colorectal cancer (CAC) is a subtype of CRC associated with inflammatory bowel disease (IBD). It is well known that individuals with IBD have a 2-3 times higher risk of developing CRC than those who do not, rendering CAC a major cause of death in this group. Although the etiology and pathogenesis of CAC are incompletely understood, animal models of chronic inflammation and human cohort data indicate that changes in the intestinal environment, including host response dysregulation and gut microbiota perturbations, may contribute to the development of CAC. Genomic alterations are a hallmark of CAC, with patterns that are distinct from those in sporadic CRC. The discovery of the biological changes that underlie the development of CAC is ongoing; however, current data suggest that chronic inflammation in IBD increases the risk of developing CAC. Therefore, a deeper understanding of the precise mechanisms by which inflammation triggers genetic alterations and disrupts intestinal homeostasis may provide insight into novel therapeutic strategies for the prevention of CAC.
Collapse
Affiliation(s)
- Hiroko Nagao-Kitamoto
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan, 1150 W. Medical Center Drive, Ann Arbor, MI, 48109, USA.
- WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan.
| | - Sho Kitamoto
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan, 1150 W. Medical Center Drive, Ann Arbor, MI, 48109, USA
- WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
| | - Nobuhiko Kamada
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan, 1150 W. Medical Center Drive, Ann Arbor, MI, 48109, USA.
- WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan.
| |
Collapse
|
19
|
Abd Ellatif SA, Bouqellah NA, Abu-Serie MM, Razik ESA, Al-Surhanee AA, Askary AE, Daigham GE, Mahfouz AY. Assessment of probiotic efficacy and anticancer activities of Lactiplantibacillus plantarum ESSG1 (MZ683194.1) and Lactiplantibacillus pentosus ESSG2 (MZ683195.1) isolated from dairy products. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:39684-39701. [PMID: 35112259 DOI: 10.1007/s11356-022-18537-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 01/03/2022] [Indexed: 06/14/2023]
Abstract
Resistance to antibiotics is on the rise, and its indiscriminate usage has resulted in human and animal management constraints. In the research for an innovative treatment to diminish antimicrobial resistance, lactic acid bacteria (LAB) throw light on diminishing this problem in public health. As a result, this paper looked at the efficacy of LAB isolates and their active metabolites to combat pathogens, reduce antibiotic use in clinical settings, and explore the anticancer potential of 8 strains of LAB isolated from dairy products. Antifungal and antibacterial potential of LAB isolates against selected crop pathogenic fungi and food pathogenic bacteria had been estimated. Results revealed that all isolates exert antioxidant efficacy relating to DPPH, NO scavenging ability, reducing power, superoxide anion, hydroxyl radical, and anti-lipid peroxidation potential. Additionally, 12B isolate exert the highest anticancer upshot with IC50 values of 43.98 ± 0.4; 36.7 ± 0.6, 43.1 ± 0.8, and 35.1 ± 0.3 μg/ml, versus Caco-2, MCF-7, HepG-2, and PC3 cell lines respectively, whereas 13B isolate significantly had the highest selectivity index between peripheral blood mononuclear cells (PBMCs) and the tested human cancer cell lines compared to 5-fluorouracil. 13B was the most apoptosis-dependent death inducer for all human cancer cell lines besides exerting the lowest percentage of apoptosis against PBMCs suggesting its safety against PBMCs. The most promising strains 12B and 13B were identified by 16S rRNA sequencing as Lactiplantibacillus plantarum ESSG1 (MZ683194.1) and Lactiplantibacillus pentosus ESSG2 (MZ683195.1). LAB and their extracts are superb substitutive, safe, and efficient antimicrobial, antioxidant, and antitumor curative agents.
Collapse
Affiliation(s)
- Sawsan A Abd Ellatif
- Bioprocess development Department, Genetic Engineering and Biotechnology Research Institute (GEBRI), City for Scientific Research and Technology Applications, New Borg El-Arab, 21934, Alexandria, Egypt
| | - Nahla Alsayed Bouqellah
- Science College, Biology Department, Taibah University, 42317- 8599, Al-Madinah Al-Munawara, Kingdom of Saudi Arabia
| | - Marwa M Abu-Serie
- Medical Biotechnology Department, Genetic Engineering and Biotechnology Research Institute (GEBRI), the City of Scientific Research and Technology Applications, New Borg El-Arab, 21934, Alexandria, Egypt
| | - Elsayed S Abdel Razik
- Plant Protection and Biomolecular Diagnosis Department, Arid Lands Cultivation Research Institute, City for Scientific Research and Technology Applications, New Borg El-Arab, 21934, Alexandria, Egypt
| | - Ameena A Al-Surhanee
- Biology Department, College of Science, Jouf University, Sakaka, 2014, Kingdom of Saudi Arabia
| | - Ahmad El Askary
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, P.O. Box 11099, Taif, 21944, Saudi Arabia
| | - Ghadir E Daigham
- Botany and Microbiology Department, Faculty of Science, Al-Azhar University (Girls Branch), Cairo, Egypt
| | - Amira Y Mahfouz
- Botany and Microbiology Department, Faculty of Science, Al-Azhar University (Girls Branch), Cairo, Egypt.
| |
Collapse
|
20
|
Hou K, Wu ZX, Chen XY, Wang JQ, Zhang D, Xiao C, Zhu D, Koya JB, Wei L, Li J, Chen ZS. Microbiota in health and diseases. Signal Transduct Target Ther 2022; 7:135. [PMID: 35461318 PMCID: PMC9034083 DOI: 10.1038/s41392-022-00974-4] [Citation(s) in RCA: 916] [Impact Index Per Article: 305.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 03/11/2022] [Accepted: 03/15/2022] [Indexed: 02/07/2023] Open
Abstract
The role of microbiota in health and diseases is being highlighted by numerous studies since its discovery. Depending on the localized regions, microbiota can be classified into gut, oral, respiratory, and skin microbiota. The microbial communities are in symbiosis with the host, contributing to homeostasis and regulating immune function. However, microbiota dysbiosis can lead to dysregulation of bodily functions and diseases including cardiovascular diseases (CVDs), cancers, respiratory diseases, etc. In this review, we discuss the current knowledge of how microbiota links to host health or pathogenesis. We first summarize the research of microbiota in healthy conditions, including the gut-brain axis, colonization resistance and immune modulation. Then, we highlight the pathogenesis of microbiota dysbiosis in disease development and progression, primarily associated with dysregulation of community composition, modulation of host immune response, and induction of chronic inflammation. Finally, we introduce the clinical approaches that utilize microbiota for disease treatment, such as microbiota modulation and fecal microbial transplantation.
Collapse
Affiliation(s)
- Kaijian Hou
- Department of Endocrine and Metabolic Diseases, Longhu Hospital, The First Affiliated Hospital of Medical College of Shantou University, Shantou, Guangdong, 515000, China
| | - Zhuo-Xun Wu
- Department of Pharmaceutical Sciences, Institute for Biotechnology, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
| | - Xuan-Yu Chen
- Department of Pharmaceutical Sciences, Institute for Biotechnology, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
| | - Jing-Quan Wang
- Department of Pharmaceutical Sciences, Institute for Biotechnology, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
| | - Dongya Zhang
- Microbiome Research Center, Moon (Guangzhou) Biotech Ltd, Guangzhou, 510535, China
| | - Chuanxing Xiao
- Department of Endocrine and Metabolic Diseases, Longhu Hospital, The First Affiliated Hospital of Medical College of Shantou University, Shantou, Guangdong, 515000, China
| | - Dan Zhu
- Department of Endocrine and Metabolic Diseases, Longhu Hospital, The First Affiliated Hospital of Medical College of Shantou University, Shantou, Guangdong, 515000, China
| | - Jagadish B Koya
- Department of Pharmaceutical Sciences, Institute for Biotechnology, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
| | - Liuya Wei
- School of Pharmacy, Weifang Medical University, Weifang, Shandong, 261053, China
| | - Jilin Li
- Department of Cardiovascular, The Second Affiliated Hospital of Medical College of Shantou University, Shantou, Guangdong, 515000, China
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, Institute for Biotechnology, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
| |
Collapse
|
21
|
Gastrointestinal Microbiota Dysbiosis Associated with SARS-CoV-2 Infection in Colorectal Cancer: The Implication of Probiotics. GASTROENTEROLOGY INSIGHTS 2022. [DOI: 10.3390/gastroent13010006] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The complexity of coronavirus disease 2019 (COVID-19)’s pathophysiology is such that microbial dysbiosis in the lung and gastrointestinal (GI) microbiota may be involved in its pathogenic process. GI microbiota dysbiosis has been associated with respiratory disorders, including COVID-19, as well as sporadic colorectal cancer (CRC) through imbalanced microbiota and compromised immune response. It is pertinent to understand the possible role of probiotics in stabilizing the microbial environment and maintaining the integrity of the respiratory and GI tracts in SARS-CoV-2 induced dysbiosis and colorectal carcinogenesis. The long-term implication of SARS-CoV-2 in GI dysbiosis via microbiota-gut-lung cross-talk could increase the risk of new CRC diagnosis or worsen the condition of previously diagnosed individuals. Recent knowledge shows that the immune-modulatory response to probiotics is shifting the beneficial use of probiotics towards the treatment of various diseases. In this review, we highlight the potential impact of probiotics on SARS-CoV-2 infection associated with CRC through microbiota imbalance in COVID-19 patients.
Collapse
|
22
|
Davoodvandi A, Fallahi F, Tamtaji OR, Tajiknia V, Banikazemi Z, Fathizadeh H, Abbasi-Kolli M, Aschner M, Ghandali M, Sahebkar A, Taghizadeh M, Mirzaei H. An Update on the Effects of Probiotics on Gastrointestinal Cancers. Front Pharmacol 2021; 12:680400. [PMID: 34992527 PMCID: PMC8724544 DOI: 10.3389/fphar.2021.680400] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 11/26/2021] [Indexed: 12/28/2022] Open
Abstract
Because of their increasing prevalence, gastrointestinal (GI) cancers are regarded as an important global health challenge. Microorganisms residing in the human GI tract, termed gut microbiota, encompass a large number of living organisms. The role of the gut in the regulation of the gut-mediated immune responses, metabolism, absorption of micro- and macro-nutrients and essential vitamins, and short-chain fatty acid production, and resistance to pathogens has been extensively investigated. In the past few decades, it has been shown that microbiota imbalance is associated with the susceptibility to various chronic disorders, such as obesity, irritable bowel syndrome, inflammatory bowel disease, asthma, rheumatoid arthritis, psychiatric disorders, and various types of cancer. Emerging evidence has shown that oral administration of various strains of probiotics can protect against cancer development. Furthermore, clinical investigations suggest that probiotic administration in cancer patients decreases the incidence of postoperative inflammation. The present review addresses the efficacy and underlying mechanisms of action of probiotics against GI cancers. The safety of the most commercial probiotic strains has been confirmed, and therefore these strains can be used as adjuvant or neo-adjuvant treatments for cancer prevention and improving the efficacy of therapeutic strategies. Nevertheless, well-designed clinical studies are still needed for a better understanding of the properties and mechanisms of action of probiotic strains in mitigating GI cancer development.
Collapse
Affiliation(s)
- Amirhossein Davoodvandi
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Farzaneh Fallahi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Omid Reza Tamtaji
- Students’ Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Vida Tajiknia
- Department of Surgery, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zarrin Banikazemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Hadis Fathizadeh
- Department of Laboratory Sciences, Sirjan Faculty of Medicine Sciences, Sirjan, Iran
| | - Mohammad Abbasi-Kolli
- Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Maryam Ghandali
- School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohsen Taghizadeh
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
23
|
Lactobacillus fermentum and Lactobacillus crispatus Do Not Have Cytotoxic Effects on HN5 Oral Squamous Cell Carcinoma Cell Line. Int J Dent 2021; 2021:3034068. [PMID: 34621315 PMCID: PMC8492272 DOI: 10.1155/2021/3034068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 08/28/2021] [Accepted: 09/09/2021] [Indexed: 11/26/2022] Open
Abstract
Background The oral environment has a very complex normal flora and a wide variety of bacteria including lactobacilli. Studies have shown oral microbial flora has important influence in the development of oral cancer. Squamous cell carcinomas account for more than 90% of cancers in oral cavity. Lactobacilli are known as one of the newest methods for the prevention and treatment of cancers. Previous studies on the effects of probiotics on oral cancer cells are very limited, and only two species of Lactobacillus which are not present in the normal oral microflora have been studied. Due to the unknown effects of lactobacilli on oral cancer, this study aimed to investigate the effect of two species of lactobacilli of oral cavity on oral cancer cells. Methods and Materials The effects of the supernatant of two lactobacilli, namely, fermentum and crispatus were studied on HN5-cancer cells. The MTT method was used to study the effects of lactobacilli on inhibition of cancer cell growth. Results The results showed that these lactobacilli do not prevent the progression of oral cancer cells. Moreover, the results showed that the acidic medium had the most effect on reducing the growth of oral cancer cells. Conclusion Due to the different effects of lactobacilli on various cancer types, the effects of two Lactobacillus crispatus and Lactobacillus fermentum on other oral cancer cell lines may be different from what has been reported in this study.
Collapse
|
24
|
Bazireh H, Shariati P, Azimzadeh Jamalkandi S, Ahmadi A, Boroumand MA. Isolation of Novel Probiotic Lactobacillus and Enterococcus Strains From Human Salivary and Fecal Sources. Front Microbiol 2020; 11:597946. [PMID: 33343539 PMCID: PMC7746552 DOI: 10.3389/fmicb.2020.597946] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 11/11/2020] [Indexed: 12/29/2022] Open
Abstract
Probiotics are non-pathogenic microorganisms that can interact with the gastrointestinal microbiota. They have numerous beneficial health effects that include enhancement of the host immune response, antiallergic, antimicrobial, anti-cancer, and anti-inflammatory properties. Probiotics are capable of restoring the impaired microbiome of a dysbiotic gut. They can be isolated from different environments. However, it is frequently suggested that probiotics for human use should come from human sources. The objective of this study was to isolate and characterize novel probiotic strains from the saliva and feces of healthy human individuals. To meet the criteria for probiotic attributes, the isolates were subjected to numerous standard morphological and biochemical tests. These tests included Gram staining, catalase tests, antibiotic susceptibility testing, hemolytic and antagonistic evaluation, tolerance tests involving temperature, NaCl levels, pH and bile salts, adherence ability assays, and genotypic characterization involving 16S rRNA gene sequencing. From 26 saliva and 11 stool samples, 185 microbial strains were isolated. Based on morphological and biochemical characteristics, 14 potential probiotic candidates were selected and identified genotypically. The new strains belonged to Lactobacillus fermentum, Enterococcus faecium, and Enterococcus hire. The selected strains were non-hemolytic, showed high tolerance to low pH and bile salts, and strong adherence abilities. Furthermore, the strains displayed a wide range of antimicrobial activities, particularly against antibiotic-resistant pathogens such as methicillin resistant Staphylococcus aureus (MRSA). Moreover, five of the selected isolates demonstrated antiproliferative features against human colon cancer cell line (Caco-2). The results of this investigation confirm the diversity of microbial populations in the human gut and saliva, and since these strains are of human origin, they will highly likely display maximal activities in food and drugs set for human use. Hence, the new strains of this study require additional in vivo experiments to assess their health-promoting effects.
Collapse
Affiliation(s)
- Homa Bazireh
- Department of Bioprocess Engineering, Institute of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Parvin Shariati
- Department of Bioprocess Engineering, Institute of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Sadegh Azimzadeh Jamalkandi
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Ali Ahmadi
- Molecular Biology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mohammad Ali Boroumand
- Department of Pathology and Laboratory Medicine, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
25
|
Silveira DSC, Veronez LC, Lopes-Júnior LC, Anatriello E, Brunaldi MO, Pereira-da-Silva G. Lactobacillus bulgaricus inhibits colitis-associated cancer via a negative regulation of intestinal inflammation in azoxymethane/dextran sodium sulfate model. World J Gastroenterol 2020; 26:6782-6794. [PMID: 33268961 PMCID: PMC7684459 DOI: 10.3748/wjg.v26.i43.6782] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 05/28/2020] [Accepted: 10/01/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Colitis-associated cancer (CAC) accounts for 2%-3% of colorectal cancer (CRC) cases preceded by inflammatory bowel diseases (IBD) such as Crohn's disease and ulcerative colitis. Intestinal microbiota has been reported to play a central role in the pathogenesis of IBD and CAC. Recently, numerous prebiotics and probiotics have being investigated as antitumor agents due to their capacity to modulate inflammatory responses. Previous studies have indicated that lactic acid bacteria could be successfully used in managing sporadic CRC, however little is known about their role in CAC.
AIM To investigate the effect of the probiotic Lactobacillus bulgaricus (L. bulgaricus) during the development of an experimental model of colitis associated colon cancer (CAC).
METHODS C57BL/6 mice received an intraperitoneal injection of azoxymethane (10 mg/kg), followed by three cycles of sodium dextran sulphate diluted in water (5% w/v). Probiotic group received daily L. bulgaricus. Intestinal inflammation was determined by scoring clinical signs. Cytokines levels were determined from colon and/or tumor samples by ELISA BD OptEIATM kits. The level of significance was set at P < 0.05. Graphs were generated and statistical analysis performed using the software GraphPad Prism 6.0.
RESULTS L. bulgaricus treatment inhibited of total tumor volume and mean size of tumors. In addition, the probiotic also attenuated the clinical signs of intestinal inflammation inducing a decrease in intestinal and tumor levels of IL-6, TNF-α, IL-17, IL-23 and IL-1β.
CONCLUSION Our results suggest a potential chemopreventive effect of probiotic on CAC. L. bulgaricus regulates the inflammatory response and preventing CAC.
Collapse
Affiliation(s)
- Denise Sayuri Calheiros Silveira
- Department of Biochemistry and Immunology, University of São Paulo at Ribeirão Preto Medical School, Ribeirão Preto 14049-900, SP, Brazil
| | - Luciana Chain Veronez
- Department of Biochemistry and Immunology, University of São Paulo at Ribeirão Preto Medical School, Ribeirão Preto 14049-900, SP, Brazil
| | - Luís Carlos Lopes-Júnior
- Health Sciences Center, Federal University of Espírito Santo - UFES, Vitória 29043-900, ES, Brazil
| | - Elen Anatriello
- Institute of Science and Technology, Federal University of São Paulo, UNIFESP, São José dos Campos 12231-280, SP, Brazil
| | | | - Gabriela Pereira-da-Silva
- Department of Maternal-Infant Nursing and Public Health, University of São Paulo at Ribeirão Preto College of Nursing, Ribeirão Preto 14040-902, SP, Brazil
| |
Collapse
|
26
|
Cruz BCS, Sarandy MM, Messias AC, Gonçalves RV, Ferreira CLLF, Peluzio MCG. Preclinical and clinical relevance of probiotics and synbiotics in colorectal carcinogenesis: a systematic review. Nutr Rev 2020; 78:667-687. [PMID: 31917829 DOI: 10.1093/nutrit/nuz087] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
CONTEXT Recent evidence suggests that modulation of the gut microbiota may help prevent colorectal cancer. OBJECTIVE The aim of this systematic review was to investigate the role of probiotics and synbiotics in the prevention of colorectal cancer and to clarify potential mechanisms involved. DATA SOURCES The PubMed, ScienceDirect, and LILACS databases were searched for studies conducted in humans or animal models and published up to August 15, 2018. STUDY SELECTION Clinical trials and placebo-controlled experimental studies that evaluated the effects of probiotics and synbiotics in colorectal cancer and cancer associated with inflammatory bowel disease were included. Of 247 articles identified, 31 remained after exclusion criteria were applied. A search of reference lists identified 5 additional studies, for a total of 36 included studies. DATA EXTRACTION Two authors independently assessed risk of bias of included studies and extracted data. Data were pooled by type of study, ie, preclinical or clinical. RESULTS The results showed positive effects of probiotics and synbiotics in preventing colorectal cancer. The main mechanisms identified were alterations in the composition and metabolic activity of the intestinal microbiota; reduction of inflammation; induction of apoptosis and inhibition of tumor growth; modulation of immune responses and cell proliferation; enhanced function of the intestinal barrier; production of compounds with anticarcinogenic activity; and modulation of oxidative stress. CONCLUSIONS Probiotics or synbiotics may help prevent colorectal cancer, but additional studies in humans are required to better inform clinical practice.
Collapse
Affiliation(s)
- Bruna C S Cruz
- Department of Nutrition and Health, Nutritional Biochemistry Laboratory, Universidade Federal de Viçosa, Viçosa, Minas Gerais, Brazil
| | - Mariáurea M Sarandy
- Department of Animal Biology, Experimental Pathology Laboratory, Universidade Federal de Viçosa, Viçosa, Minas Gerais, Brazil
| | - Anny C Messias
- Department of Nutrition and Health, Nutritional Biochemistry Laboratory, Universidade Federal de Viçosa, Viçosa, Minas Gerais, Brazil
| | - Reggiani V Gonçalves
- Department of Animal Biology, Experimental Pathology Laboratory, Universidade Federal de Viçosa, Viçosa, Minas Gerais, Brazil
| | - Célia L L F Ferreira
- Institute of Biotechnology Applied to Agriculture (BIOAGRO), Laboratory of Dairy Cultures, Universidade Federal de Viçosa, Viçosa, Minas Gerais, Brazil
| | - Maria C G Peluzio
- Department of Nutrition and Health, Nutritional Biochemistry Laboratory, Universidade Federal de Viçosa, Viçosa, Minas Gerais, Brazil
| |
Collapse
|
27
|
Brasiel PGDA, Dutra Luquetti SCP, Peluzio MDCG, Novaes RD, Gonçalves RV. Preclinical Evidence of Probiotics in Colorectal Carcinogenesis: A Systematic Review. Dig Dis Sci 2020; 65:3197-3210. [PMID: 31960202 DOI: 10.1007/s10620-020-06062-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 01/09/2020] [Indexed: 12/24/2022]
Abstract
BACKGROUND Colorectal cancer, the second major cause of cancer deaths, imposes a major health burden worldwide. There is growing evidence that supports that the use of probiotics is effective against various diseases, especially in gastrointestinal diseases, including the colorectal cancer, but the differences between the strains, dose, and frequency used are not yet clear. AIMS To perform a systematic review to compile the results of studies carried out in animal models and investigated the effect of probiotics on colorectal carcinogenesis. METHODS Studies were selected in PubMed/MEDLINE and Scopus according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines. Search filters were developed using three parameters: probiotics, colorectal cancer, and animal model. RESULTS From a structured search, we discovered 34 original articles and submitted them to a risk of bias analysis using SYRCLE's tool. The studies show a great diversity of models, most were conducted in rats (55.8%) and used 1,2 dimethylhydrazine as the drug to induce colorectal carcinogenesis (61.7%). The vast majority of trials investigated Lactobacillus (64%) and Bifidobacterium (29.4%) strains. Twenty-six (86.6%) studies found significant reduction in lesions or tumors in the animals that received probiotics. The main methodological limitation was the insufficient amount of information for the adequate reproducibility of the trials, which indicated a high risk of bias due to incomplete characterization of the experimental design. CONCLUSIONS The different probiotics' strains showed anti-carcinogenic effect, reduced the development of lesions and intestinal tumors, antioxidant and immunomodulatory activity, and reduced fecal bacterial enzymes.
Collapse
Affiliation(s)
| | | | | | - Rômulo Dias Novaes
- Department of Structural Biology, Federal University of Alfenas, Alfenas, MG, Brazil
| | | |
Collapse
|
28
|
Aponte M, Murru N, Shoukat M. Therapeutic, Prophylactic, and Functional Use of Probiotics: A Current Perspective. Front Microbiol 2020; 11:562048. [PMID: 33042069 PMCID: PMC7516994 DOI: 10.3389/fmicb.2020.562048] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 08/12/2020] [Indexed: 12/12/2022] Open
Abstract
Probiotics are considered as the twenty-first century panpharmacon due to their competent remedial power to cure from gastrointestinal dysbiosis, systematic metabolic diseases, and genetic impairments up to complicated neurodegenerative disorders. They paved the way for an innovative managing of various severe diseases through palatable food products. The probiotics' role as a "bio-therapy" increased their significance in food and medicine due to many competitive advantages over traditional treatment therapies. Their prophylactic and therapeutic potential has been assessed through hundreds of preclinical and clinical studies. In addition, the food industry employs probiotics as functional and nutraceutical ingredients to enhance the added value of food product in terms of increased health benefits. However, regardless of promising health-boosting effects, the probiotics' efficacy still needs an in-depth understanding of systematic mechanisms and factors supporting the healthy actions.
Collapse
Affiliation(s)
- Maria Aponte
- Department of Agricultural Sciences, University of Naples Federico II, Portici, Italy
| | - Nicoletta Murru
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, Naples, Italy
| | - Mahtab Shoukat
- Department of Agricultural Sciences, University of Naples Federico II, Portici, Italy
| |
Collapse
|
29
|
A critical review of antibiotic resistance in probiotic bacteria. Food Res Int 2020; 136:109571. [PMID: 32846610 DOI: 10.1016/j.foodres.2020.109571] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 07/16/2020] [Accepted: 07/17/2020] [Indexed: 12/12/2022]
Abstract
Probiotics are defined as live microorganisms that, when administered in adequate amounts, confer a health benefit upon the host. At present, probiotics are gaining popularity worldwide and are widely used in food and medicine. Consumption of probiotics is increasing with further in-depth research on the relationship between intestinal flora and host health. Most people pay more attention to the function of probiotics but ignore their potential risks, such as infection and antibiotic resistance transfer to pathogenic microbes. Physiological functions, effects and mechanisms of action of probiotics were covered in this review, as well as the antibiotic resistance phenotypes, mechanisms and genes found in probiotics. Typical cases of antibiotic resistance of probiotics were also highlighted, as well as the potential risks (including pathogenicity, infectivity and excessive immune response) and corresponding strategies (dosage, formulation, and administration route). This timely study provides an avenue for further research, development and application of probiotics.
Collapse
|
30
|
Jakubczyk D, Leszczyńska K, Górska S. The Effectiveness of Probiotics in the Treatment of Inflammatory Bowel Disease (IBD)-A Critical Review. Nutrients 2020; 12:nu12071973. [PMID: 32630805 PMCID: PMC7400428 DOI: 10.3390/nu12071973] [Citation(s) in RCA: 173] [Impact Index Per Article: 34.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 06/19/2020] [Accepted: 06/30/2020] [Indexed: 12/17/2022] Open
Abstract
Inflammatory bowel disease (IBD), which affects millions of people worldwide, includes two separate diseases: Crohn's disease (CD) and ulcerative colitis (UC). Although the background (chronic inflammatory state) and some of the symptoms of CD and UC are similar, both diseases differ from each other. It is becoming clear that a combination of many factors, in particular genetic background, host immune response and microbial reduced diversity status are associated with IBD. One potential strategy to prevent/treat IBD is gut modulation by probiotics. Over the last twenty years, many publications have focused on the role of probiotics in the course of IBD. The review discusses the utility of different strains of probiotics, especially Bifidobacterium spp., in all factors potentially involved in the etiology of IBD. The probiotic modulatory properties among different study models (cell lines, animal models of colitis, clinical study) are discussed and probiotic usefulness is assessed in relation to the treatment, prevention, and remission of diseases.
Collapse
|
31
|
Aindelis G, Chlichlia K. Modulation of Anti-Tumour Immune Responses by Probiotic Bacteria. Vaccines (Basel) 2020; 8:vaccines8020329. [PMID: 32575876 PMCID: PMC7350223 DOI: 10.3390/vaccines8020329] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 05/29/2020] [Accepted: 06/16/2020] [Indexed: 12/13/2022] Open
Abstract
There is a growing amount of evidence to support the beneficial role of a balanced intestinal microbiota, or distinct members thereof, in the manifestation and progression of malignant tumours, not only in the gastrointestinal tract but also in distant tissues as well. Intriguingly, bacterial species have been demonstrated to be indispensable modulatory agents of widely-used immunotherapeutic or chemotherapeutic regiments. However, the exact contribution of commensal bacteria to immunity, as well as to neoplasia formation and response to treatment, has not been fully elucidated, and most of the current knowledge acquired from animal models has yet to be translated to human subjects. Here, recent advances in understanding the interaction of gut microbes with the immune system and the modulation of protective immune responses to cancer, either naturally or in the context of widely-used treatments, are reviewed, along with the implications of these observations for future therapeutic approaches. In this regard, bacterial species capable of facilitating optimal immune responses against cancer have been surveyed. According to the findings summarized here, we suggest that strategies incorporating probiotic bacteria and/or modulation of the intestinal microbiota can be used as immune adjuvants, aiming to optimize the efficacy of cancer immunotherapies and conventional anti-tumour treatments.
Collapse
|
32
|
Legesse Bedada T, Feto TK, Awoke KS, Garedew AD, Yifat FT, Birri DJ. Probiotics for cancer alternative prevention and treatment. Biomed Pharmacother 2020; 129:110409. [PMID: 32563987 DOI: 10.1016/j.biopha.2020.110409] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 06/03/2020] [Accepted: 06/13/2020] [Indexed: 02/06/2023] Open
Abstract
Cancer is a fatal malignancy with high clinical significance and remains one of the major causes of illness and death. It has no suitable cure existing till now. The safety and stability of the standard chemotherapeutics drugs and synthetic agents used to manage cancer are doubtful. These agents are affecting the quality of life or contributing for development of drug resistance and are not affordable to the majority of the patients. Therefore, scientists are looking into clinical management of the cancer with high efficiency. This review focuses on the role of probiotics as alternative prevention and treatment of cancer. In this regard, we discuss the alternative cancer biotherapeutic drugs including live or dead probiotics and their metabolites, such as short chain fatty acids, inhibitory compounds of protein, polysaccharide, nucleic acid and ferrichrome in in vitro, in vivo and clinical studies. We also discuss the effectiveness of these biotherapeutics in prevention and treatment of various types of cancer linked with probiotic bacterial or fungal strains, probiotic dose, and time of exposure. More in vivo mainly clinical trials are necessary to further reveal and approve the significant role of live and dead probiotics as well as their metabolic products in cancer prevention and treatment. Finally, the majority of the positive results provided by probiotic treatments are limited to experimental settings. To minimize side effects associated with probiotics, short and long term effect studies in the direction of methodology standardization are required.
Collapse
Affiliation(s)
- Tesfaye Legesse Bedada
- Public Health Microbiology Research Team, Ethiopian Public Health Institute, P. O. Box: 1242, Addis Ababa, Ethiopia.
| | - Tatek Kasim Feto
- Public Health Microbiology Research Team, Ethiopian Public Health Institute, P. O. Box: 1242, Addis Ababa, Ethiopia.
| | - Kaleab Sebsibe Awoke
- Public Health Microbiology Research Team, Ethiopian Public Health Institute, P. O. Box: 1242, Addis Ababa, Ethiopia.
| | - Asnake Desalegn Garedew
- Division of Microbiology and Infectious Diseases, Department of Microbial, Cellular and Molecular Biology, Addis Ababa University, Addis Ababa, P. O. Box: 1176, Ethiopia.
| | - Fitsum Tigu Yifat
- Division of Microbiology and Infectious Diseases, Department of Microbial, Cellular and Molecular Biology, Addis Ababa University, Addis Ababa, P. O. Box: 1176, Ethiopia.
| | - Dagim Jirata Birri
- Division of Microbiology and Infectious Diseases, Department of Microbial, Cellular and Molecular Biology, Addis Ababa University, Addis Ababa, P. O. Box: 1176, Ethiopia.
| |
Collapse
|
33
|
Si H, Yang Q, Hu H, Ding C, Wang H, Lin X. Colorectal cancer occurrence and treatment based on changes in intestinal flora. Semin Cancer Biol 2020; 70:3-10. [PMID: 32404293 DOI: 10.1016/j.semcancer.2020.05.004] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 04/29/2020] [Accepted: 05/05/2020] [Indexed: 02/07/2023]
Abstract
Colorectal cancer (CRC) is the most common cancer in the world, and its incidence has been increasing in recent years. The occurrence of CRC is believed to be related to a variety of factors. Epidemiological data indicate that CRC is mainly affected by environmental factors, eating habits, physical activity and genetic factors. As a newly recognized functional component, the intestinal microbiota plays important roles in preventing CRC formation and maintaining intestinal immunity. In this review, we summarize the mechanisms by which the gut microbiota causes CRC through alterations to immune function, focusing on the mechanisms by which intestinal microbial dysfunction promotes CRC. Furthermore, we describe the changes in the intestinal flora observed in CRC and their potential for CRC treatment with the goal of facilitating future research on the roles of the intestinal flora.
Collapse
Affiliation(s)
- Huifang Si
- Department of Gastroenterology, Huaihe Hospital of Henan University, 115 Ximen Street, Kaifeng 475000, Henan, China
| | - Qing Yang
- Department of Anesthesiology, Huaihe Hospital of Henan University, 115 Ximen Street, Kaifeng 475000, Henan, China
| | - Hong Hu
- Department of Gastroenterology, Huaihe Hospital of Henan University, 115 Ximen Street, Kaifeng 475000, Henan, China
| | - Chunsheng Ding
- Department of Gastroenterology, Huaihe Hospital of Henan University, 115 Ximen Street, Kaifeng 475000, Henan, China
| | - Huichao Wang
- Department of Nephrology, First Affiliated Hospital of Henan University, 357 Ximen Street, Kaifeng 475000, Henan, China
| | - Xuhong Lin
- Department of Clinical Laboratory, Translational Medicine Center, Huaihe Hospital of Henan University, 115 Ximen Street, Kaifeng 475000, Henan, China.
| |
Collapse
|
34
|
Khadivi R, Razavilar V, Anvar A, Akbari Adreghani B. Aflatoxin M1-Binding Ability of Selected Lactic Acid Bacteria Strains and Saccharomyces boulardii in the Experimentally Contaminated Milk Treated with Some Biophysical Factors. ARCHIVES OF RAZI INSTITUTE 2020; 75:63-73. [PMID: 32292004 DOI: 10.22092/ari.2019.123985.1265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 03/04/2019] [Indexed: 09/30/2022]
Abstract
There is a growing concern regarding the recurrent observation of aflatoxins (AFs) in the milk of lactating animals. Regarding this, the present study was conducted to assess the aflatoxin M1 (AFM1)-binding ability of three species, namely Lactobacillus rhamnosus, L. plantarum, and Saccharomyces boulardii, inAFM1-contaminatedmilk. The mentioned species were administeredatthe concentrations of107 and 109 CFU/mLto skimmed milk contaminated with 0.5 and 0.75 ng/mL AFM1 within the incubation times of 30 and 90 min at 4&deg;C and 37&deg;C. Lactobacillus rhamnosus was found to have the best binding ability at the concentrations of 107 and 109 (CFU/ml), rendering 82% and 90% removal in the milk samples with 0.5 and 0.75 ng/ml AFM1, respectively. Accordingly, this value at 107 and 109 CFU/ml of L. plantarum was obtained 89% and 82% with 0.75 ng/ml of AFM1, respectively. For S. boulardii at 107 and 109 CFU/ml, the rates were respectively estimated at 75% and 90% with 0.75 ng/ml of AFM1. The best AFM1-binding levels for L. rhamnosus, L. plantarum, and S. boulardii were 91.82&plusmn;10.9%, 89.33&plusmn;0.58%, and 93.20&plusmn;10.9, respectively, at the concentrations of 1&times;109, 1&times;107, and 1&times;107 CFU/ml at 37, 4, and 37&deg;C, respectively. In this study, the maximum AFM1 binding (100.0&plusmn;0.58) occurred while a combination of the aforementioned probiotics was employed at a concentration of 1&times;107 CFU/ml at 37&deg;C with 0.5 ng/ml AFM1, followed by the combination of L. rhamnosus and L. plantarum (95.86&plusmn;10.9) at a concentration of 1&times;109 CFU/ml at the same temperature with 0.75 ng/ml AFM1. It was concluded that the use of S. boulardii in combination with Lactobacillus rhamnosus and L. plantarum, which bind AFM1 in milk, can decrease the risk of AFM1 in dairy products.
Collapse
Affiliation(s)
- R Khadivi
- Department of Food Hygiene, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - V Razavilar
- Department of Food Hygiene, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - A Anvar
- Department of Food Hygiene, Science and Research Branch, Islamic Azad University, Tehran, Iran.,Department of Food Hygiene, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - B Akbari Adreghani
- Food and Drug Laboratory Research Center, Food and Drug Organization, Ministry of Health and Medical Education, Tehran, Iran
| |
Collapse
|
35
|
The Role of Probiotics in Colorectal Cancer Management. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:3535982. [PMID: 32148539 PMCID: PMC7048916 DOI: 10.1155/2020/3535982] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 01/16/2020] [Accepted: 01/23/2020] [Indexed: 12/13/2022]
Abstract
Colorectal cancer (CRC) is one of the most common cancerous diseases worldwide and causes leading cancer-associated deaths. Several factors are related to the incidence of CRC such as unhealthy diet and lifestyle, heredity, metabolic disorders, and genetic factors. Even though several advanced medical procedures are available for CRC treatment, the survival rates are poor with many adverse treatments associated side effects, which affects the quality of life. Probiotics are a well-known bioactive candidate for the treatment of several diseases and ill-health conditions. The recent scientific evidence suggested that probiotic supplementation protects the CRC patients from treatment-associated adverse effects. The manuscript summarizes the influence of probiotic supplementation on the health status of CRC patients and discusses the possible mechanism behind the protective effect of probiotics against CRC. The literature survey revealed that beneficial impact of probiotic supplementation depends on several factors such as strain, dosage, duration of the intervention, host physiology, and other food supplements. The probiotic intervention improves the microbiota, releases antimicrobials and anticarcinogenic agents, helps to remove carcinogens, and improves the intestinal permeability, tight junction function, and enzyme activity in CRC patients. Besides, not all probiotic strains exhibit anti-CRC activities; it is necessary to screen the potent strain for the development of a probiotic-based therapeutic agent to control or prevent the incidence of CRC.
Collapse
|
36
|
Settanni CR, Quaranta G, Bibbò S, Gasbarrini A, Cammarota G, Ianiro G. Oral supplementation with lactobacilli to prevent colorectal cancer in preclinical models. MINERVA GASTROENTERO 2019; 66:48-69. [PMID: 31760735 DOI: 10.23736/s1121-421x.19.02631-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Colorectal cancer (CRC) is still a major threat for public health, as it is the third most common cancer in men and the second in women and it ranks second among tumors in terms of mortality. Evidence from the last decades emphasizes the complex role of gut microbial composition in CRC development. Historically, it is believed that dairy products, a source of lactobacilli and other lactic acid bacteria, are beneficial for human health and help in preventing CRC. We searched online literature for trials evaluating the preventive role of lactobacilli in CRC animal models. Most of selected studied assessed a relevant role of lactobacilli in preventing CRC and precursor lesions. Mechanisms through which this effect was achieved are supposed to regard immunomodulation, regulation of apoptosis, gut microbial modulation, genes expression, reduction of oxidative stress and others. Lactobacilli oral supplementation is reported to be effective in preventing CRC in animal models, even if the underlying mechanisms of action are still not fully understood.
Collapse
Affiliation(s)
- Carlo R Settanni
- Digestive Disease Center, Agostino Gemelli University Polyclinic, IRCCS and Foundation, Sacred Heart Catholic University, Rome, Italy
| | - Gianluca Quaranta
- Institute of Microbiology, Agostino Gemelli University Polyclinic, IRCCS and Foundation, Sacred Heart Catholic University, Rome, Italy
| | - Stefano Bibbò
- Digestive Disease Center, Agostino Gemelli University Polyclinic, IRCCS and Foundation, Sacred Heart Catholic University, Rome, Italy
| | - Antonio Gasbarrini
- Digestive Disease Center, Agostino Gemelli University Polyclinic, IRCCS and Foundation, Sacred Heart Catholic University, Rome, Italy
| | - Giovanni Cammarota
- Digestive Disease Center, Agostino Gemelli University Polyclinic, IRCCS and Foundation, Sacred Heart Catholic University, Rome, Italy
| | - Gianluca Ianiro
- Digestive Disease Center, Agostino Gemelli University Polyclinic, IRCCS and Foundation, Sacred Heart Catholic University, Rome, Italy -
| |
Collapse
|
37
|
Naghmouchi K, Belguesmia Y, Bendali F, Spano G, Seal BS, Drider D. Lactobacillus fermentum: a bacterial species with potential for food preservation and biomedical applications. Crit Rev Food Sci Nutr 2019; 60:3387-3399. [PMID: 31729242 DOI: 10.1080/10408398.2019.1688250] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Lactic acid-producing bacteria are the most commonly used probiotics that play an important role in protecting the host against harmful microorganisms, strengthening the host immune system, improving feed digestibility, and reducing metabolic disorders. Lactobacillus fermentum (Lb. fermentum) is a Gram-positive bacterium belonging to Lactobacillus genus, and many reportedly to enhance the immunologic response as well as prevent community-acquired gastrointestinal and upper respiratory infections. Additionally, Lb. fermentum strains produce diverse and potent antimicrobial peptides, which can be applied as food preservative agents or as alternatives to antibiotics. Further functions attributed to probiotic Lb. fermentum strains are their abilities to decrease the level of blood stream cholesterol (as cholesterol-lowering agents) and to potentially help prevent alcoholic liver disease and colorectal cancer among humans. Finally, Lb. fermentum is a key microorganism in sourdough technology, contributing to flavor, texture, or health-promoting dough ingredients, and has recently been used to develop new foods stuffs such as fortified and functional foods with beneficial attributes for human health. Development of such new foodstuffs are currently taking important proportions of the food industry market. Furthermore, an increasing awareness of the consumers prompts the food-makers to implement alternative environmental friendly solutions in the production processes and/or suitable biological alternative to limit the use of antibiotics in feed and food. Here, we give an account on the application of Lb. fermentum strains in the biomedical and food preservation fields, with a focus on probiotic features such as bacteriocin production. We also summarize the use of Lb. fermentum as cell factories with the aim to improve the efficacy and health value of functional food.
Collapse
Affiliation(s)
- Karim Naghmouchi
- Department of Pharmaceutical Chemistry, College of Clinical Pharmacy, Al Baha University, Saudi Arabia.,Faculté des Sciences de Tunis, Université de Tunis El Manar, LR01ES05 Biochimie et Biotechnologie, Tunis, Tunisie
| | - Yanath Belguesmia
- Université Lille, INRA, ISA, Université d'Artois, Université Littoral Côte d'Opale, EA 7394-ICV Institut Charles Viollette, Lille, France
| | - Farida Bendali
- Laboratoire de Microbiologie Appliquée, Faculté des Sciences de la Nature et de la Vie, Université de Bejaia, Bejaia, Algeria
| | - Giuseppe Spano
- Dipartimento di Scienze Agrarie, degli Alimenti e dell'Ambiente, Università di Foggia, Foggia, Italy
| | - Bruce S Seal
- Biology Program, Oregon State University Cascades, Bend, Oregon, USA
| | - Djamel Drider
- Université Lille, INRA, ISA, Université d'Artois, Université Littoral Côte d'Opale, EA 7394-ICV Institut Charles Viollette, Lille, France
| |
Collapse
|
38
|
Lee JE, Lee J, Kim JH, Cho N, Lee SH, Park SB, Koh B, Kang D, Kim S, Yoo HM. Characterization of the Anti-Cancer Activity of the Probiotic Bacterium Lactobacillus fermentum Using 2D vs. 3D Culture in Colorectal Cancer Cells. Biomolecules 2019; 9:E557. [PMID: 31581581 PMCID: PMC6843223 DOI: 10.3390/biom9100557] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 09/25/2019] [Accepted: 09/30/2019] [Indexed: 02/07/2023] Open
Abstract
The aim of this study was to investigate the potential anti-cancer effects of probiotic cell-free supernatant (CFS) treatment using Lactobacillusfermentum for colorectal cancer (CRC) in 3D culture systems. Cell viability was assessed using MTS (3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium, inner salt) assays, whereas apoptosis was monitored through RT-qPCR analysis of Bax, Bak, Noxa, and Bid mRNA expressions in addition to flow cytometry analysis of Lactobacillus cell-free supernatant (LCFS) treatment. Our results showed that the anti-cancer effect of LCFS on cell viability was pronouncedly enhanced in 3D-cultured HCT-116 cells, which was linked to the increased level of cleaved caspase 3. Additionally, upregulation of apoptotic marker gene mRNA transcription was dramatically increased in 3D cultured cells compared to 2D systems. In conclusion, this study suggests that LCFS enhances the activation of intrinsic apoptosis in HCT-116 cells and the potential anti-cancer effects of Lactobacilli mixtures in 3D culture systems. All in all, our study highlights the benefits of 3D culture models over 2D culture modeling in studying the anti-cancer effects of probiotics.
Collapse
Affiliation(s)
- Joo-Eun Lee
- Stem Cell Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea.
| | - Jina Lee
- Center for Bioanalysis, Korea Research Institute of Standards and Science (KRISS), Daejeon 34113, Korea.
| | - Ji Hyun Kim
- Center for Bioanalysis, Korea Research Institute of Standards and Science (KRISS), Daejeon 34113, Korea.
| | - Namki Cho
- College of Pharmacy and Research Institute of Drug Development, Chonnam National University, Gwangju 61186, Korea.
| | - Sung Hoon Lee
- College of Pharmacy, Chung-Ang University, Seoul 06974, Korea.
| | - Sung Bum Park
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34114, Korea.
| | - Byumseok Koh
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34114, Korea.
| | - Dukjin Kang
- Center for Bioanalysis, Korea Research Institute of Standards and Science (KRISS), Daejeon 34113, Korea.
| | - Seil Kim
- Center for Bioanalysis, Korea Research Institute of Standards and Science (KRISS), Daejeon 34113, Korea.
- Convergent Research Center for Emerging Virus Infection, Korea Research Institute of Chemical Technology, Daejeon 34114, Korea.
- Department of Bio-Analysis Science, University of Science & Technology (UST), Daejeon 34113, Korea.
| | - Hee Min Yoo
- Center for Bioanalysis, Korea Research Institute of Standards and Science (KRISS), Daejeon 34113, Korea.
| |
Collapse
|
39
|
Zaharuddin L, Mokhtar NM, Muhammad Nawawi KN, Raja Ali RA. A randomized double-blind placebo-controlled trial of probiotics in post-surgical colorectal cancer. BMC Gastroenterol 2019; 19:131. [PMID: 31340751 PMCID: PMC6657028 DOI: 10.1186/s12876-019-1047-4] [Citation(s) in RCA: 151] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Accepted: 07/17/2019] [Indexed: 12/31/2022] Open
Abstract
Background Our study aimed to determine the effect of probiotic consumption containing six viable microorganisms of 30 × 1010 cfu Lactobacillus and Bifidobacteria strains for six months on clinical outcomes and inflammatory cytokines (TNF-α, IFN-γ, IL-6, IL-10, IL-12, IL-17A, IL-17C and IL-22) in patients with colorectal cancer. Methods Fifty-two patients with colorectal cancer were randomized at four weeks after surgery to receive either a placebo (n = 25) or 30 billion colony-forming unit (CFU) of a mixture of six viable strains including 107 mg of Lactobacillus acidophilus BCMC® 12,130, Lactobacillus lactis BCMC® 12,451, Lactobacillus casei subsp BCMC® 12,313, Bifidobacterium longum BCMC® 02120, Bifidobacterium bifidum BCMC® 02290 and Bifidobacterium infantis BCMC® 02129 (n = 27). Patients were instructed to take the product orally twice daily for six months. Infection status, diarrhea or hospital admission were recorded throughout the study. Blood was taken pre- and post-intervention to measure TNF-α, IFN-γ, IL-6, IL-10, IL-12, IL-17A, IL-17C and IL-22 using ELISA multiplex kit. Results The majority of cases (~ 70%) were in Duke’s C colorectal cancer for both groups. No surgical infection occurred and no antibiotics were required. Chemotherapy induced diarrhea was observed in both groups. Significant reduction in the level of pro-inflammatory cytokine, TNF-α, IL-6, IL-10, IL-12, IL-17A, IL-17C and IL-22 were observed in CRC patients who received probiotics as compared to pre-treatment level (P < 0.05). However, there was no significant difference in the IFN-γ in both groups. Conclusions We have shown that probiotics containing six viable microorganisms of Lactobacillus and Bifidobacteria strains are safe to be consumed at four weeks after surgery in colorectal cancer patients and have reduced pro-inflammatory cytokines (except for IFN-gamma). Probiotic may modify intestinal microenvironment resulting in a decline in pro-inflammatory cytokines. Trial registration NCT03782428; retrospectively registered on 20th December 2018.
Collapse
Affiliation(s)
- Liyana Zaharuddin
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, 56000 Cheras, Kuala Lumpur, Malaysia
| | - Norfilza Mohd Mokhtar
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, 56000 Cheras, Kuala Lumpur, Malaysia
| | - Khairul Najmi Muhammad Nawawi
- GUT Research group, Department of Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latiff, Bandar Tun Razak, 56000 Cheras, Kuala Lumpur, Malaysia
| | - Raja Affendi Raja Ali
- GUT Research group, Department of Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latiff, Bandar Tun Razak, 56000 Cheras, Kuala Lumpur, Malaysia.
| |
Collapse
|
40
|
Eslami M, Yousefi B, Kokhaei P, Hemati M, Nejad ZR, Arabkari V, Namdar A. Importance of probiotics in the prevention and treatment of colorectal cancer. J Cell Physiol 2019; 234:17127-17143. [PMID: 30912128 DOI: 10.1002/jcp.28473] [Citation(s) in RCA: 98] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 02/10/2019] [Accepted: 02/19/2019] [Indexed: 12/14/2022]
Affiliation(s)
- Majid Eslami
- Cancer Research Center, Semnan University of Medical Sciences Semnan Iran
| | - Bahman Yousefi
- Department of Immunology Semnan University of Medical Sciences Semnan Iran
| | - Parviz Kokhaei
- Cancer Research Center, Semnan University of Medical Sciences Semnan Iran
- Immune and Gene Therapy Lab, Cancer Centre Karolinska, Karolinska University Hospital Stockholm Sweden
| | - Maral Hemati
- Department of Immunology Semnan University of Medical Sciences Semnan Iran
| | | | - Vahid Arabkari
- Discipline of Pathology, Lambe Institute for Translational Research, Clinical Science Institute, School of Medicine, National University of Ireland Galway Ireland
| | - Afshin Namdar
- Department of Dentistry Faculty of Medicine and Dentistry, University of Alberta Edmonton Alberta Canada
| |
Collapse
|
41
|
Namvar Rad M, Razavilar V, Anvar SAA, Akbari-Adergani B. Selected bio-physical factors affecting the efficiency ofBifidobacterium animalis lactisandLactobacillus delbrueckii bulgaricusto degrade aflatoxin M1in artificially contaminated milk. J Food Saf 2018. [DOI: 10.1111/jfs.12463] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- Maryam Namvar Rad
- Student of Food Hygiene, Science and Research Branch, Islamic Azad University; Tehran Iran
| | - Vadood Razavilar
- Department of Food Hygiene, Science and Research Branch, Islamic Azad University; Tehran Iran
| | - Seyed Amir Ali Anvar
- Department of Food Hygiene, Science and Research Branch, Islamic Azad University; Tehran Iran
| | - Behrouz Akbari-Adergani
- Food and Drug Laboratory Research Center, Food and Drug Organization, Ministry of Health and Medical Education; Tehran Iran
| |
Collapse
|