1
|
Lee AH, Rodriguez Jimenez DM, Meisel M. Limosilactobacillus reuteri - a probiotic gut commensal with contextual impact on immunity. Gut Microbes 2025; 17:2451088. [PMID: 39825615 DOI: 10.1080/19490976.2025.2451088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 12/10/2024] [Accepted: 01/02/2025] [Indexed: 01/20/2025] Open
Abstract
The gut microbiome plays a key role in human health, influencing various biological processes and disease outcomes. The historical roots of probiotics are traced back to Nobel Laureate Élie Metchnikoff, who linked the longevity of Bulgarian villagers to their consumption of sour milk fermented by Lactobacilli. His pioneering work led to the global recognition of probiotics as beneficial supplements, now a multibillion-dollar industry. Modern probiotics have been extensively studied for their immunomodulatory effects. Limosilactobacillus reuteri (L. reuteri), a widely used probiotic, has garnered significant attention for its systemic immune-regulatory properties, particularly in relation to autoimmunity and cancer. This review delves into the role of L. reuteri in modulating immune responses, with a focus on its impact on systemic diseases.
Collapse
Affiliation(s)
- Amanda H Lee
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Graduate Program of Microbiology and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | | | - Marlies Meisel
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| |
Collapse
|
2
|
Nama ASA, Sandeepa GM, Buddolla V, Mastan A. Advances in understanding therapeutic mechanisms of probiotics in cancer management, with special emphasis on breast cancer: A comprehensive review. Eur J Pharmacol 2025; 995:177410. [PMID: 39986595 DOI: 10.1016/j.ejphar.2025.177410] [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: 11/27/2024] [Revised: 02/01/2025] [Accepted: 02/19/2025] [Indexed: 02/24/2025]
Abstract
The increasing global prevalence of cancer, particularly breast cancer, necessitates the development of innovative therapeutic strategies. Probiotics, proficient in promoting gut health, have emerged as promising candidates for cancer treatment due to their immunomodulatory and potential anticancer properties. This review focuses on the therapeutic mechanisms of probiotics in breast cancer, examining their anticancer efficacy through metabolic, immune, and molecular mechanisms. Probiotics enhance cancer therapies, minimize side effects, and offer new adjuvant approaches in oncology. Recent advancements discussed in the review include the utilization of probiotics as oncolytic gene expression systems and drug delivery vectors, as well as personalized probiotic interventions aimed at optimizing cancer therapy. Clinical studies are critically evaluated, highlighting both the outcomes and limitations of probiotic use in cancer patients, particularly those suffering from breast cancer. Additionally, the review explores factors influencing anticancer effects of probiotics, focusing on their role in modulating the tumor microenvironment. Challenges in translating preclinical findings to clinical practice are discussed, along with future research directions, focusing on the relationship between probiotics, the microbiome, and cancer treatment. Ultimately, this review advocates for further investigation into the therapeutic potential of probiotics in breast cancer, aiming to harness their benefits in oncology.
Collapse
Affiliation(s)
- A S Angel Nama
- Department of Biotechnology, Vikrama Simhapuri University, Nellore, 524320, India
| | - G Mary Sandeepa
- Department of Biotechnology, Vikrama Simhapuri University, Nellore, 524320, India.
| | - Viswanath Buddolla
- Dr.Buddolla's Institute of Life Sciences (A unit of Dr. Buddolla's Research and Educational Society), Tirupati, 517506, India
| | - Anthati Mastan
- Dr.Buddolla's Institute of Life Sciences (A unit of Dr. Buddolla's Research and Educational Society), Tirupati, 517506, India.
| |
Collapse
|
3
|
Sharif-Askari EA, Atoui KM, Mteyrek AK, Fawaz LM. Probiotics and mediterranean diet for breast cancer management and prevention? Cell Stress 2025; 9:1-15. [PMID: 40417456 PMCID: PMC12096334 DOI: 10.15698/cst2025.05.303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2025] [Revised: 01/30/2025] [Accepted: 03/25/2025] [Indexed: 05/27/2025] Open
Abstract
The human gut microbiota, a diverse community of beneficial normal flora microorganisms, significantly influences physiological function and the immune response. Various microbiota strains have shown promise in supporting clinical treatment of chronic diseases, including cancer, by potentially providing antioxidative and anti-tumorigenic effects in both in vivo and in vitro studies. Breast cancer, which ranks amongst the top five cancer types common worldwide and particularly in Mediterranean countries, has been showing high incidence and prevalence. In breast cancer, microbiota composition, hormonal dynamics, and dietary choices are believed to play significant roles. Hence, the Mediterranean diet, known for its microbiota-friendly features, emerges as a potential protective factor against breast cancer development, highlighting the potential for personalized dietary strategies in cancer prevention. This comprehensive review highlights the emerging mechanisms by which probiotics support our immune system during different physiological activities. It also discusses their potential role, along with nutrition intervention, in improving essential clinical treatment outcomes in breast cancer patients and survivors, suggesting potential supportive strategies that go hand in hand with clinical strategies. Unfortunately, very little research addresses the possible clinical implications of probiotics and dietary habits on breast cancer, despite the promising results, calling for further studies and actions.
Collapse
Affiliation(s)
- Ehssan A. Sharif-Askari
- Biomedical Science Department, School of Arts & Sciences, Lebanese International University, Tyre, Lebanon
| | - Khadija M. Atoui
- Biomedical Science Department, School of Arts & Sciences, Lebanese International University, Tyre, Lebanon
| | - Ali K. Mteyrek
- Department of Biological and Chemical Sciences, School of Arts and Sciences, Lebanese International University, Tyre, Lebanon
| | - Lama M. Fawaz
- Neuroimmunology Unit, Montreal Neurological Institute, McGill University, Quebec, Canada
| |
Collapse
|
4
|
Liu Y, Ning H, Li Y, Li Y, Ma J. The microbiota in breast cancer: dysbiosis, microbial metabolites, and therapeutic implications. Am J Cancer Res 2025; 15:1384-1409. [PMID: 40371158 PMCID: PMC12070087 DOI: 10.62347/zjcf2843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 03/29/2025] [Indexed: 05/16/2025] Open
Abstract
The human microbiome plays a pivotal role in host health and disease, with emerging evidence underscoring its significant influence on the development and progression of breast cancer. Studies have revealed that dysbiosis in both the gut and breast tissue microbiota is strongly associated with an elevated risk of breast cancer. Distinct microbial profiles have been identified among healthy individuals, patients with benign breast conditions, and those with malignant tumors, with further variations observed across different ethnic groups and breast cancer subtypes. The complex interplay between breast cancer risk factors and microbial populations, coupled with the direct impact of microbial communities and their metabolites on inflammatory pathways and immune responses, underscores the importance of this field. Additionally, the interaction between gut microbiota and therapeutic modalities such as chemotherapy and radiotherapy is of particular interest, as these interactions can significantly influence treatment outcomes, either enhancing or diminishing efficacy. This review explores the effects of the Mediterranean diet, probiotics, prebiotics, and natural medicinal products on gut microbiota, emphasizing their potential as innovative therapeutic strategies. Notably, the use of engineered probiotics within the tumor microenvironment represents a promising frontier in breast cancer treatment. In conclusion, research on the human microbiome not only deepens our understanding of breast cancer pathogenesis but also lays the groundwork for the development of novel and targeted therapeutic interventions.
Collapse
Affiliation(s)
- Yan Liu
- Shanxi Province Cancer Hospital, Shanxi Medical UniversityTaiyuan 030006, Shanxi, China
| | - Haiyang Ning
- Shanxi Province Cancer Hospital, Shanxi Medical UniversityTaiyuan 030006, Shanxi, China
| | - Yifei Li
- Shanxi Province Cancer Hospital, Shanxi Medical UniversityTaiyuan 030006, Shanxi, China
| | - Yifan Li
- Shanxi Province Cancer Hospital, Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Cancer Hospital Affiliated to Shanxi Medical UniversityTaiyuan 030001, Shanxi, China
| | - Jinfeng Ma
- Shanxi Province Cancer Hospital, Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Cancer Hospital Affiliated to Shanxi Medical UniversityTaiyuan 030001, Shanxi, China
| |
Collapse
|
5
|
Wang Q, Zhao H, Ding H, Zhang H, Zhang J, Li L, Han B, Kai G. Cell-free supernatant of Clostridium leptum inhibits breast cancer cell proliferation. Lett Appl Microbiol 2025; 78:ovaf037. [PMID: 40074544 DOI: 10.1093/lambio/ovaf037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 02/26/2025] [Accepted: 03/11/2025] [Indexed: 03/14/2025]
Abstract
Breast cancer has emerged as the leading cause of global cancer incidence, surpassing lung cancer. Accumulating evidence suggests that probiotics exhibit inhibitory effect on breast cancer progression, highlighting the need to identify gut flora-derived probiotics with potential anti-breast cancer properties. Here, we investigated the effect of the cell-free supernatant of Clostridium leptum (ClCFS) on breast cancer cells by methyl thiazolyl tetrazolium (MTT) assay. Untargeted metabolomics analysis was employed to characterize metabolite alterations in ClCFS. Furthermore, the core targets were predicted by the protein-protein interaction network and the signaling pathways were enriched by the Kyoto Encyclopedia of Genes and Genomes analysis. Our findings demonstrated that ClCFS inhibited the proliferation of breast cancer cells and that the main metabolite of ClCFS might be acetylcarnitine. Utilizing network pharmacological analysis, we identified apoptosis-related signaling pathways as the principal mechanisms underlying ClCFS activity. Furthermore, five core targets of STAT3, IL-1β, BCL2, CASP3, and ESR1 were identified. This study elucidates the main bioactive constituent and the potential targets of ClCFS against breast cancer. It provides a new understanding of the pharmacological activity of ClCFS in breast cancer treatment.
Collapse
Affiliation(s)
- Qingling Wang
- Zhejiang Provincial International S&T Cooperation Base for Active Ingredients of Medicinal and Edible Plants and Health, Zhejiang Provincial Key TCM Laboratory for Chinese Resource Innovation and Transformation, School of Pharmaceutical Sciences, Jinhua Academy, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Huan Zhao
- Zhejiang Provincial International S&T Cooperation Base for Active Ingredients of Medicinal and Edible Plants and Health, Zhejiang Provincial Key TCM Laboratory for Chinese Resource Innovation and Transformation, School of Pharmaceutical Sciences, Jinhua Academy, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Huizhe Ding
- Zhejiang Provincial International S&T Cooperation Base for Active Ingredients of Medicinal and Edible Plants and Health, Zhejiang Provincial Key TCM Laboratory for Chinese Resource Innovation and Transformation, School of Pharmaceutical Sciences, Jinhua Academy, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Hao Zhang
- Zhejiang Provincial International S&T Cooperation Base for Active Ingredients of Medicinal and Edible Plants and Health, Zhejiang Provincial Key TCM Laboratory for Chinese Resource Innovation and Transformation, School of Pharmaceutical Sciences, Jinhua Academy, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Jizhou Zhang
- Oncology Department, Wenzhou TCM Hospital of Zhejiang Chinese Medical University, Wenzhou 325000, China
| | - Liqin Li
- Key Laboratory of Traditional Chinese Medicine for the Development and Clinical Transformation of Immunomodulatory Traditional Chinese Medicine in Zhejiang Province, Huzhou Central Hospital, The Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou 313002, China
| | - Bing Han
- Zhejiang Provincial International S&T Cooperation Base for Active Ingredients of Medicinal and Edible Plants and Health, Zhejiang Provincial Key TCM Laboratory for Chinese Resource Innovation and Transformation, School of Pharmaceutical Sciences, Jinhua Academy, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Guoyin Kai
- Zhejiang Provincial International S&T Cooperation Base for Active Ingredients of Medicinal and Edible Plants and Health, Zhejiang Provincial Key TCM Laboratory for Chinese Resource Innovation and Transformation, School of Pharmaceutical Sciences, Jinhua Academy, Zhejiang Chinese Medical University, Hangzhou 310053, China
| |
Collapse
|
6
|
Rezai S, Ghorbani E, Nazari SE, Rahmani F, Hassanian SM, Afshari A, Habibi Najafi MB, Avan A, Ryzhikov M, Soleimanpour S, Khazei M. Investigation of Lactobacillus Probiotics Derived from Traditional Dairy Products in Eliciting Anti-Tumor Responses in Mouse Colorectal Cancer Model. IRANIAN JOURNAL OF MEDICAL SCIENCES 2025; 50:247-259. [PMID: 40255226 PMCID: PMC12008654 DOI: 10.30476/ijms.2024.102396.3530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 08/07/2024] [Accepted: 09/06/2024] [Indexed: 04/22/2025]
Abstract
Background Colorectal cancer (CRC) is a serious health problem, and finding new treatments is important. There is growing evidence for the antitumor activity of probiotics. This study investigated the anti-cancer potential of a probiotic mix containing Lactobacillus plantarum, L. brevis, L. helveticus, and L. delbrueckii, alone or in combination with the chemotherapy drug 5-fluorouracil, against CRC. Methods The research was carried out in Mashhad in 2021. The cytotoxic effect of Lactobacillus isolates on CRC cells was investigated in two-dimensional and three-dimensional cell culture models. Histological staining and molecular approaches were used to investigate the regulatory mechanism of Lactobacillus isolates on cell migration, inflammation, fibrosis, cell cycle progression, apoptosis, and tumor necrosis in the CRC mouse model. Statistical analysis was performed using SPSS software version 20 with a significance level of P<0.05. The tests employed included the Kolmogorov-Smirnov, ANOVA, Dunnett's post hoc, and Kruskal-Wallis. Results Lactobacillus strains effectively suppressed tumor growth in CRC by promoting cell death and inhibiting fibrosis and inflammation. These bacteria regulated apoptosis-related genes such as Bcl-2-associated protein x (P=0.0033), and BCL-2 (P=0.0029), leading to increased tumor necrosis. Treatment with bacterial supernatants reduced tumor size and fibrosis by downregulating collagen type I, alpha 1 (Col1a1) (P=0.024), Col1a2 (P=0.0231), and actin alpha 2 (P=0.0466), and transforming growth factor-beta expression. Additionally, they suppressed inflammation by decreasing tumor necrosis factor-alpha (P=0.0001), interleukin 6, and IL-1β (P=0.0198) levels in tumor tissues. Furthermore, the treatment inhibited CRC cell migration by modulating epithelial cadherin (P=0.0198) and matrix metallopeptidase 2 (P=0.033) expression. Conclusion Findings indicated that co-administration of Lactobacillus isolates with 5-FU could improve the anti-tumor properties of the standard drug, 5-FU, supporting the therapeutic potential of these safe isolated lactic acid bacteria for CRC patients in vivo.
Collapse
Affiliation(s)
- Shaghayegh Rezai
- Department of Medical Microbiology and Virology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Elnaz Ghorbani
- Department of Medical Microbiology and Virology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyedeh Elnaz Nazari
- Department of Medical Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Farzad Rahmani
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mahdi Hassanian
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Asma Afshari
- Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Amir Avan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Human Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mikhail Ryzhikov
- Saint Louis University, School of Medicine, Saint Louis, MO, USA
| | - Saman Soleimanpour
- Department of Medical Microbiology and Virology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Khazei
- Department of Medical Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
7
|
Argyris DG, Markaki MP, Afaloniati H, Karagiannis GS, Poutahidis T, Angelopoulou K. Suppression of chemically induced mammary cancer by early-life oral administration of cholera toxin in mice is associated with aberrant regulation of Bmp and Notch signaling pathways. Mol Biol Rep 2025; 52:150. [PMID: 39841292 DOI: 10.1007/s11033-025-10271-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 01/16/2025] [Indexed: 01/23/2025]
Abstract
BACKGROUND Lately, significant attention has been drawn towards the potential efficacy of cholera toxin (CT)-an exotoxin produced by the small intestine pathogenic bacterium Vibrio cholera-in modulating cancer-promoting events. In a recent study, we demonstrated that early-life oral administration of non-pathogenic doses of CT in mice suppressed chemically-induced carcinogenesis in tissues distantly located from the gut. In the mammary gland, CT pretreatment was shown to reduce tumor multiplicity, increase apoptosis and alter the expression of several cancer-related molecules. In the present work we investigated the protumorigenic mammary microenvironment for possible associations between early life CT administration and the expression of key components of the Bmp and Notch signaling pathways. METHODS AND RESULTS Total RNA from mammary tissue samples were retrieved from a recent experiment where FVB/N female mice were preconditioned with CT and later treated with the carcinogen 7,12-dimethylbenzanthracene (DMBA). Real-time PCR was used for relative quantification of gene expression. Our results revealed that CT anti-tumor effects significantly correlated with deregulation of crucial BMP pathway elements, with downregulation of Bmp7 ligand and upregulation of inhibitory Smad6 being the most prominent alterations observed. Concerning Notch signaling pathway, significantly elevated gene expression levels in the CT-treated DMBA mice, as compared to their non-treated counterparts, were also identified at the ligand-receptor level. CONCLUSIONS These findings suggest that CT tumor protective effects in the mammary gland are associated with discerning deregulation of components of both Bmp and Notch signaling pathways and provide insights into the mechanisms underlying CT's anti-cancer outcome.
Collapse
MESH Headings
- Animals
- Female
- Signal Transduction/drug effects
- Mice
- Receptors, Notch/metabolism
- Receptors, Notch/genetics
- Cholera Toxin/administration & dosage
- Cholera Toxin/pharmacology
- Administration, Oral
- Bone Morphogenetic Proteins/metabolism
- Bone Morphogenetic Proteins/genetics
- Mammary Neoplasms, Experimental/chemically induced
- Mammary Neoplasms, Experimental/drug therapy
- Mammary Neoplasms, Experimental/metabolism
- Mammary Neoplasms, Experimental/genetics
- 9,10-Dimethyl-1,2-benzanthracene
- Mammary Glands, Animal/drug effects
- Mammary Glands, Animal/metabolism
- Gene Expression Regulation, Neoplastic/drug effects
Collapse
Affiliation(s)
- Dimitrios G Argyris
- Laboratory of Biochemistry and Toxicology, School of Veterinary Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
- Tumor Microenvironment of Metastasis Program, Montefiore-Einstein Cancer Center, Bronx, NY, USA
| | - Maria P Markaki
- Laboratory of Biochemistry and Toxicology, School of Veterinary Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Hara Afaloniati
- Laboratory of Biochemistry and Toxicology, School of Veterinary Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - George S Karagiannis
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
- Tumor Microenvironment of Metastasis Program, Montefiore-Einstein Cancer Center, Bronx, NY, USA
| | - Theofilos Poutahidis
- Laboratory of Pathology, School of Veterinary Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Katerina Angelopoulou
- Laboratory of Biochemistry and Toxicology, School of Veterinary Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece.
| |
Collapse
|
8
|
Arnone AA, Tsai YT, Cline JM, Wilson AS, Westwood B, Seger ME, Chiba A, Howard-McNatt M, Levine EA, Thomas A, Soto-Pantoja DR, Cook KL. Endocrine-targeting therapies shift the breast microbiome to reduce estrogen receptor-α breast cancer risk. Cell Rep Med 2025; 6:101880. [PMID: 39742868 PMCID: PMC11866439 DOI: 10.1016/j.xcrm.2024.101880] [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: 08/07/2023] [Revised: 05/14/2024] [Accepted: 11/26/2024] [Indexed: 01/04/2025]
Abstract
Studies indicate that breast tissue has a distinct modifiable microbiome population. We demonstrate that endocrine-targeting therapies, such as tamoxifen, reshape the non-cancerous breast microbiome to influence tissue metabolism and reduce tumorigenesis. Using 16S sequencing, we found that tamoxifen alters β-diversity and increases Firmicutes abundance, including Lactobacillus spp., in mammary glands (MGs) of mice and non-human primates. Immunohistochemistry showed that lipoteichoic acid (LTA)-positive bacteria were elevated in tamoxifen-treated breast tissue. In B6.MMTV-PyMT mice, intra-nipple probiotic bacteria injections reduced tumorigenesis, altered metabolic gene expression, and decreased tumor proliferation. Probiotic-conditioned media selectively reduced viability in estrogen receptor-positive (ER+) breast cancer cells and altered mitochondrial metabolism in non-cancerous epithelial cells. Human tumor samples revealed that LTA-positive bacteria negatively correlated with Ki67, suggesting that endocrine therapies influence tumor-associated microbiota to regulate proliferation. Our data indicate that endocrine-targeting therapies modify the breast microbiome, corresponding with a shift in tissue metabolism to potentially reduce ER+ breast cancer risk.
Collapse
Affiliation(s)
- Alana A Arnone
- Department of Surgery, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA; Department of Physiology and Pharmacology, Wake Forest University Health Sciences, Winston-Salem, NC 27157, USA
| | - Yu-Ting Tsai
- Department of Surgery, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA; Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - J Mark Cline
- Department of Pathology, Section on Comparative Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA; Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Adam S Wilson
- Department of Surgery, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Brian Westwood
- Department of Surgery, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Meghan E Seger
- Department of Surgery, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Akiko Chiba
- Department of Surgery, Duke University School of Medicine, Durham, NC 27710, USA; Durham VA Medical Center, Department of Surgery, Durham, NC 27705, USA; Duke Cancer Institute, Duke University School of Medicine, Durham, NC 27710, USA
| | - Marissa Howard-McNatt
- Department of General Surgery, Section of Surgical Oncology, Wake Forest University School of Medicine, Winston-Salem NC, 27157, USA
| | - Edward A Levine
- Department of General Surgery, Section of Surgical Oncology, Wake Forest University School of Medicine, Winston-Salem NC, 27157, USA
| | - Alexandra Thomas
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC 27710, USA
| | - David R Soto-Pantoja
- Department of Surgery, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA; Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA; Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Katherine L Cook
- Department of Surgery, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA; Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA; Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA.
| |
Collapse
|
9
|
Kowalczyk M, Radziwill-Bienkowska JM, Marć MA, Jastrząb R, Mytych J, Siedlecki P, Szczepankowska AK. Screening for probiotic properties and potential immunogenic effects of lactobacilli strains isolated from various food products. Front Microbiol 2024; 15:1430582. [PMID: 39534506 PMCID: PMC11554495 DOI: 10.3389/fmicb.2024.1430582] [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: 05/10/2024] [Accepted: 10/09/2024] [Indexed: 11/16/2024] Open
Abstract
Introduction Deceleration of disease progression and re-establishment of microbial balance in the gut is often achieved by application of lactobacilli strains. Their beneficial effects are associated with probiotic properties, which may be accompanied by immunomodulatory action at mucosal surfaces. Methods To single out such strains, we screened almost three hundred lactobacilli isolates from eight genera and various food sources for acid and bile salt tolerance, adhesion to mucin as well as hemolytic activity and antibiotic susceptibility. Moreover, the immunomodulatory effects of cell-free supernatant (CFS) fractions of five lactobacilli strains were assessed using an in vitro cell line model. Results and discussion By our rationalized selection approach, we identified thirty-five strains with probiotic potential and biosafety features. Additionally, we showed that CFS from the Lactiplantibacillus L_4 strain downregulates proinflammatory cytokines IL-8 and IL-1β. In contrast, IL-8 expression was found to increase after treatment with CFSs of Lactiplantibacillus L_2 and L_5 and IL-1β was upregulated by CFSs of Lactiplantibacillus L_1 and Lactiplantibacillus L_3. Overall, our result delineate a rational approach of selecting lactobacilli strains for probiotic development to support the gut microbiota equilibrium and reinforce the host immune system.
Collapse
Affiliation(s)
- Magdalena Kowalczyk
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | | | | | - Rafał Jastrząb
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
- Research and Development Center, Olimp Laboratories, Dębica, Poland
| | - Jennifer Mytych
- Research and Development Center, Olimp Laboratories, Dębica, Poland
| | - Paweł Siedlecki
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | | |
Collapse
|
10
|
Sanwlani R, Bramich K, Mathivanan S. Role of probiotic extracellular vesicles in inter-kingdom communication and current technical limitations in advancing their therapeutic utility. EXTRACELLULAR VESICLES AND CIRCULATING NUCLEIC ACIDS 2024; 5:509-526. [PMID: 39697628 PMCID: PMC11648425 DOI: 10.20517/evcna.2024.39] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 08/24/2024] [Accepted: 09/05/2024] [Indexed: 12/20/2024]
Abstract
Diverse functions of probiotic extracellular vesicles (EVs) have been extensively studied over the past decade, proposing their role in inter-kingdom communication. Studies have explored their therapeutic role in pathophysiological processes ranging from cancer, immunoregulation, and ulcerative colitis to stress-induced depression. These studies have highlighted the significant and novel potential of probiotic EVs for therapeutic applications, offering immense promise in addressing several unmet clinical needs. Additionally, probiotic EVs are being explored as vehicles for targeted delivery approaches. However, the realization of clinical utility of probiotic EVs is hindered by several knowledge gaps, pitfalls, limitations, and challenges, which impede their wider acceptance by the scientific community. Among these, limited knowledge of EV biogenesis, markers and regulators in bacteria, variations in cargo due to culture conditions or EV isolation method, and lack of proper understanding of gut uptake and demonstration of in vivo effect are some important issues. This review aims to summarize the diverse roles of probiotic EVs in health and disease conditions. More importantly, it discusses the significant knowledge gaps and limitations that stand in the way of the therapeutic utility of probiotic EVs. Furthermore, the importance of addressing these gaps and limitations with technical advances such as rigorous omics has been discussed.
Collapse
Affiliation(s)
| | | | - Suresh Mathivanan
- Correspondence to: Prof. Suresh Mathivanan, Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, Science Drive, Melbourne 3086, Victoria, Australia. E-mail:
| |
Collapse
|
11
|
Xie B, Zhou X, Luo C, Fang Y, Wang Y, Wei J, Cai L, Chen T. Reversal of Platinum-based Chemotherapy Resistance in Ovarian Cancer by Naringin Through Modulation of The Gut Microbiota in a Humanized Nude Mouse Model. J Cancer 2024; 15:4430-4447. [PMID: 38947385 PMCID: PMC11212103 DOI: 10.7150/jca.96448] [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: 03/20/2024] [Accepted: 05/08/2024] [Indexed: 07/02/2024] Open
Abstract
As a chemotherapy agent, cisplatin (DDP) is often associated with drug resistance and gastrointestinal toxicity, factors that severely limit therapeutic efficacy in patients with ovarian cancer (OC). Naringin has been shown to increase sensitivity to cisplatin, but whether the intestinal microbiota is associated with this effect has not been reported so far. In this study, we applied a humanized mouse model for the first time to evaluate the reversal of cisplatin resistance by naringin, as well as naringin combined with the microbiota in ovarian cancer. The results showed that naringin combined with Bifidobacterium animalis subsp. lactis NCU-01 had an inhibitory effect on the tumor, significantly reducing tumor size (p<0.05), as well as the concentrations of serum tumor markers CA125 and HE4, increased the relative abundance of Bifidobacterium and Bacteroides, inhibit Toll-like receptor 4 (TLR4)/nuclear factor κB (NF-κB)-induced intestinal inflammation and increase the expression of intestinal permeability-associated proteins ZO-1 (p<0.001) and occludin (p<0.01). In conclusion, the above data demonstrate how naringin combined with Bifidobacterium animalis subsp. lactis NCU-01 reverses cisplatin resistance in ovarian cancer by modulating the intestinal microbiota, inhibiting the TLR4/NF-κB signaling pathway and modulating the p38MAPK signaling pathway.
Collapse
Affiliation(s)
- Bingqing Xie
- Department of Obstetrics & Gynecology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi, China
| | - Xiaoni Zhou
- Department of Obstetrics & Gynecology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi, China
| | - Chuanlin Luo
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang 330031, Jiangxi, China
| | - Yilin Fang
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang 330031, Jiangxi, China
| | - Yufei Wang
- Queen Mary School, Nanchang University, Nanchang 330031, China
| | - Jing Wei
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang 330031, Jiangxi, China
| | - Liping Cai
- Department of Obstetrics & Gynecology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi, China
| | - Tingtao Chen
- Department of Obstetrics & Gynecology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi, China
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang 330031, Jiangxi, China
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang,330031, Jiangxi, China
| |
Collapse
|
12
|
Valsecchi AA, Ferrari G, Paratore C, Dionisio R, Vignani F, Sperone P, Vellani G, Novello S, Di Maio M. Gut and local microbiota in patients with cancer: increasing evidence and potential clinical applications. Crit Rev Oncol Hematol 2024; 197:104328. [PMID: 38490281 DOI: 10.1016/j.critrevonc.2024.104328] [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: 07/13/2023] [Revised: 12/19/2023] [Accepted: 03/11/2024] [Indexed: 03/17/2024] Open
Abstract
In recent years, cancer research has highlighted the role of disrupted microbiota in carcinogenesis and cancer recurrence. However, microbiota may also interfere with drug metabolism, influencing the efficacy of cancer drugs, especially immunotherapy, and modulating the onset of adverse events. Intestinal micro-organisms can be altered by external factors, such as use of antibiotics, proton pump inhibitors treatment, lifestyle and the use of prebiotics or probiotics. The aim of our review is to provide a picture of the current evidence about preclinical and clinical data of the role of gut and local microbiota in malignancies and its potential clinical role in cancer treatments. Standardization of microbiota sequencing approaches and its modulating strategies within prospective clinical trials could be intriguing for two aims: first, to provide novel potential biomarkers both for early cancer detection and for therapeutic effectiveness; second, to propose personalized and "microbiota-tailored" treatment strategies.
Collapse
Affiliation(s)
- Anna Amela Valsecchi
- Department of Oncology, University of Turin, Città della Salute e della Scienza di Torino, Turin, Italy
| | - Giorgia Ferrari
- Department of Oncology, University of Turin, San Luigi Gonzaga Hospital, Orbassano, Italy
| | - Chiara Paratore
- Department of Oncology, ASL TO4, Ivrea Community Hospital, Ivrea, Italy.
| | - Rossana Dionisio
- Department of Oncology, University of Turin, Mauriziano Hospital, Turin, Italy
| | - Francesca Vignani
- Department of Oncology, University of Turin, Mauriziano Hospital, Turin, Italy
| | - Paola Sperone
- Department of Oncology, University of Turin, San Luigi Gonzaga Hospital, Orbassano, Italy
| | - Giorgio Vellani
- Department of Oncology, ASL TO4, Ivrea Community Hospital, Ivrea, Italy
| | - Silvia Novello
- Department of Oncology, University of Turin, San Luigi Gonzaga Hospital, Orbassano, Italy
| | - Massimo Di Maio
- Department of Oncology, University of Turin, Città della Salute e della Scienza di Torino, Turin, Italy
| |
Collapse
|
13
|
Feng K, Ren F, Shang Q, Wang X, Wang X. Association between oral microbiome and breast cancer in the east Asian population: A Mendelian randomization and case-control study. Thorac Cancer 2024; 15:974-986. [PMID: 38485288 PMCID: PMC11045337 DOI: 10.1111/1759-7714.15280] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 02/26/2024] [Accepted: 02/29/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND The causal relationship between breast cancer (BC) and the oral microbiome remains unclear. In this case-control study, using two-sample Mendelian randomization (MR), we thoroughly explored the relationship between the oral microbiome and BC in the East Asian population. METHODS Genetic summary data related to oral microbiota and BC were collected from genome-wide association studies involving participants of East Asian descent. MR estimates were generated by conducting various analyses. Sequencing data from a case-control study were used to verify the validity of these findings. RESULTS MR analysis revealed that 30 tongue and 37 salivary bacterial species were significantly associated with BC. Interestingly, in both tongue and salivary microbiomes, we observed the causal effect of six genera, namely, Aggregatibacter, Streptococcus, Prevotella, Haemophilus, Lachnospiraceae, Oribacterium, and Solobacterium, on BC. Our case-control study findings suggest differences in specific bacteria between patients with BC and healthy controls. Moreover, sequencing data confirmed the MR analysis results, demonstrating that compared with the healthy control group, the BC group had a higher relative abundance of Pasteurellaceae and Streptococcaceae but a lower relative abundance of Bacteroidaceae. CONCLUSIONS Our MR analysis suggests that the oral microbiome exerts a causative effect on BC risk, supported by the sequencing data of a case-control study. In the future, studies should be undertaken to comprehensively understand the complex interaction mechanisms between the oral microbiota and BC.
Collapse
Affiliation(s)
- Kexin Feng
- Department of Breast Surgical OncologyNational Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Fei Ren
- Department of Breast Surgical OncologyNational Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Qingyao Shang
- Department of Breast Surgical OncologyNational Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Xin Wang
- Department of Breast Surgical OncologyNational Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Xiang Wang
- Department of Breast Surgical OncologyNational Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
14
|
Afaloniati H, Aindelis G, Spyridopoulou K, Lagou MK, Tsingotjidou A, Chlichlia K, Erdman SE, Poutahidis T, Angelopoulou K. Peri-weaning cholera toxin consumption suppresses chemically-induced carcinogenesis in mice. Int J Cancer 2024; 154:1097-1110. [PMID: 38095490 DOI: 10.1002/ijc.34816] [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: 08/29/2023] [Revised: 10/27/2023] [Accepted: 11/17/2023] [Indexed: 01/23/2024]
Abstract
Gastrointestinal bacteria are known to have an impact on local and systemic immunity, and consequently either promote or suppress cancer development. Following the notion that perinatal bacterial exposure might confer immune system competency for life, we investigated whether early-life administration of cholera-toxin (CT), a protein exotoxin of the small intestine pathogenic bacterium Vibrio cholerae, may shape local and systemic immunity to impart a protective effect against tumor development in epithelia distantly located from the gut. For that, newborn mice were orally treated with low non-pathogenic doses of CT and later challenged with the carcinogen 7,12-dimethylbenzanthracene (DMBA), known to cause mainly mammary, but also skin, lung and stomach cancer. Our results revealed that CT suppressed the overall incidence and multiplicity of tumors, with varying efficiencies among cancer types, and promoted survival. Harvesting mouse tissues at an earlier time-point (105 instead of 294 days), showed that CT does not prevent preneoplastic lesions per se but it rather hinders their evolution into tumors. CT pretreatment universally increased apoptosis in the cancer-prone mammary, lung and nonglandular stomach, and altered the expression of several cancer-related molecules. Moreover, CT had a long-term effect on immune system cells and factors, the most prominent being the systemic neutrophil decrease. Finally, CT treatment significantly affected gut bacterial flora composition, leading among others to a major shift from Clostridia to Bacilli class abundance. Overall, these results support the notion that early-life CT consumption is able to affect host's immune, microbiome and gene expression profiles toward the prevention of cancer.
Collapse
Affiliation(s)
- Hara Afaloniati
- Laboratory of Biochemistry and Toxicology, School of Veterinary Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Georgios Aindelis
- Department of Molecular Biology and Genetics, Democritus University of Thrace, University Campus Dragana, Alexandroupolis, Greece
| | - Katerina Spyridopoulou
- Department of Molecular Biology and Genetics, Democritus University of Thrace, University Campus Dragana, Alexandroupolis, Greece
| | - Maria K Lagou
- Laboratory of Pathology, School of Veterinary Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Anastasia Tsingotjidou
- Laboratory of Anatomy, Histology and Embryology, School of Veterinary Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Katerina Chlichlia
- Department of Molecular Biology and Genetics, Democritus University of Thrace, University Campus Dragana, Alexandroupolis, Greece
| | - Suzan E Erdman
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Theofilos Poutahidis
- Laboratory of Pathology, School of Veterinary Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Katerina Angelopoulou
- Laboratory of Biochemistry and Toxicology, School of Veterinary Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
15
|
Summer M, Sajjad A, Ali S, Hussain T. Exploring the underlying correlation between microbiota, immune system, hormones, and inflammation with breast cancer and the role of probiotics, prebiotics and postbiotics. Arch Microbiol 2024; 206:145. [PMID: 38461447 DOI: 10.1007/s00203-024-03868-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 01/18/2024] [Accepted: 01/24/2024] [Indexed: 03/12/2024]
Abstract
According to recent research, bacterial imbalance in the gut microbiota and breast tissue may be linked to breast cancer. It has been discovered that alterations in the makeup and function of different types of bacteria found in the breast and gut may contribute to growth and advancement of breast cancer in several ways. The main role of gut microbiota is to control the metabolism of steroid hormones, such as estrogen, which are important in raising the risk of breast cancer, especially in women going through menopause. On the other hand, because the microbiota can influence mucosal and systemic immune responses, they are linked to the mutual interactions between cancer cells and their local environment in the breast and the gut. In this regard, the current review thoroughly explains the mode of action of probiotics and microbiota to eradicate the malignancy. Furthermore, immunomodulation by microbiota and probiotics is described with pathways of their activity.
Collapse
Affiliation(s)
- Muhammad Summer
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, Government College University, Lahore, 54000, Pakistan
| | - Ayesha Sajjad
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, Government College University, Lahore, 54000, Pakistan
| | - Shaukat Ali
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, Government College University, Lahore, 54000, Pakistan.
| | - Tauqeer Hussain
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, Government College University, Lahore, 54000, Pakistan
| |
Collapse
|
16
|
Feng P, Xue X, Bukhari I, Qiu C, Li Y, Zheng P, Mi Y. Gut microbiota and its therapeutic implications in tumor microenvironment interactions. Front Microbiol 2024; 15:1287077. [PMID: 38322318 PMCID: PMC10844568 DOI: 10.3389/fmicb.2024.1287077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 01/08/2024] [Indexed: 02/08/2024] Open
Abstract
The development of cancer is not just the growth and proliferation of a single transformed cell, but its tumor microenvironment (TME) also coevolves with it, which is primarily involved in tumor initiation, development, metastasis, and therapeutic responses. Recent years, TME has been emerged as a potential target for cancer diagnosis and treatment. However, the clinical efficacy of treatments targeting the TME, especially its specific components, remains insufficient. In parallel, the gut microbiome is an essential TME component that is crucial in cancer immunotherapy. Thus, assessing and constructing frameworks between the gut microbiota and the TME can significantly enhance the exploration of effective treatment strategies for various tumors. In this review the role of the gut microbiota in human cancers, including its function and relationship with various tumors was summarized. In addition, the interaction between the gut microbiota and the TME as well as its potential applications in cancer therapeutics was described. Furthermore, it was summarized that fecal microbiota transplantation, dietary adjustments, and synthetic biology to introduce gut microbiota-based medical technologies for cancer treatment. This review provides a comprehensive summary for uncovering the mechanism underlying the effects of the gut microbiota on the TME and lays a foundation for the development of personalized medicine in further studies.
Collapse
Affiliation(s)
- Pengya Feng
- Key Laboratory of Helicobacter Pylori, Microbiota and Gastrointestinal Cancer of Henan Province, Marshall Medical Research Center, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Children Rehabilitation Medicine, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Gastroenterology, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xia Xue
- Key Laboratory of Helicobacter Pylori, Microbiota and Gastrointestinal Cancer of Henan Province, Marshall Medical Research Center, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Gastroenterology, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ihtisham Bukhari
- Key Laboratory of Helicobacter Pylori, Microbiota and Gastrointestinal Cancer of Henan Province, Marshall Medical Research Center, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Gastroenterology, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chunjing Qiu
- Key Laboratory of Helicobacter Pylori, Microbiota and Gastrointestinal Cancer of Henan Province, Marshall Medical Research Center, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Gastroenterology, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yingying Li
- Key Laboratory of Helicobacter Pylori, Microbiota and Gastrointestinal Cancer of Henan Province, Marshall Medical Research Center, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Gastroenterology, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Pengyuan Zheng
- Key Laboratory of Helicobacter Pylori, Microbiota and Gastrointestinal Cancer of Henan Province, Marshall Medical Research Center, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Gastroenterology, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yang Mi
- Key Laboratory of Helicobacter Pylori, Microbiota and Gastrointestinal Cancer of Henan Province, Marshall Medical Research Center, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Gastroenterology, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
17
|
Zhou Y, Liu X, Gao W, Luo X, Lv J, Wang Y, Liu D. The role of intestinal flora on tumor immunotherapy: recent progress and treatment implications. Heliyon 2024; 10:e23919. [PMID: 38223735 PMCID: PMC10784319 DOI: 10.1016/j.heliyon.2023.e23919] [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: 08/28/2023] [Revised: 12/08/2023] [Accepted: 12/15/2023] [Indexed: 01/16/2024] Open
Abstract
Immunotherapy, specifically immune checkpoint inhibitors, has emerged as a promising approach for treating malignant tumors. The gut, housing approximately 70 % of the body's immune cells, is abundantly populated with gut bacteria that actively interact with the host's immune system. Different bacterial species within the intestinal flora are in a delicate equilibrium and mutually regulate each other. However, when this balance is disrupted, pathogenic microorganisms can dominate, adversely affecting the host's metabolism and immunity, ultimately promoting the development of disease. Emerging researches highlight the potential of interventions such as fecal microflora transplantation (FMT) to improve antitumor immune response and reduce the toxicity of immunotherapy. These remarkable findings suggest the major role of intestinal flora in the development of cancer immunotherapy and led us to the hypothesis that intestinal flora transplantation may be a new breakthrough in modifying immunotherapy side effects.
Collapse
Affiliation(s)
- Yimin Zhou
- School of Basic Medical Sciences, Shandong University, Jinan 250011, China
| | - Xiangdong Liu
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250021, China
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China
| | - Wei Gao
- School of Basic Medical Sciences, Shandong University, Jinan 250011, China
| | - Xin Luo
- School of Basic Medical Sciences, Shandong University, Jinan 250011, China
| | - Junying Lv
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250021, China
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China
| | - Yunshan Wang
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250021, China
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China
| | - Duanrui Liu
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250021, China
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China
| |
Collapse
|
18
|
He J, Li H, Jia J, Liu Y, Zhang N, Wang R, Qu W, Liu Y, Jia L. Mechanisms by which the intestinal microbiota affects gastrointestinal tumours and therapeutic effects. MOLECULAR BIOMEDICINE 2023; 4:45. [PMID: 38032415 PMCID: PMC10689341 DOI: 10.1186/s43556-023-00157-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 11/08/2023] [Indexed: 12/01/2023] Open
Abstract
The intestinal microbiota is considered to be a forgotten organ in human health and disease. It maintains intestinal homeostasis through various complex mechanisms. A significant body of research has demonstrated notable differences in the gut microbiota of patients with gastrointestinal tumours compared to healthy individuals. Furthermore, the dysregulation of gut microbiota, metabolites produced by gut bacteria, and related signal pathways can partially explain the mechanisms underlying the occurrence and development of gastrointestinal tumours. Therefore, this article summarizes the latest research progress on the gut microbiota and gastrointestinal tumours. Firstly, we provide an overview of the composition and function of the intestinal microbiota and discuss the mechanisms by which the intestinal flora directly or indirectly affects the occurrence and development of gastrointestinal tumours by regulating the immune system, producing bacterial toxins, secreting metabolites. Secondly, we present a detailed analysis of the differences of intestinal microbiota and its pathogenic mechanisms in colorectal cancer, gastric cancer, hepatocellular carcinoma, etc. Lastly, in terms of treatment strategies, we discuss the effects of the intestinal microbiota on the efficacy and toxic side effects of chemotherapy and immunotherapy and address the role of probiotics, prebiotics, FMT and antibiotic in the treatment of gastrointestinal tumours. In summary, this article provides a comprehensive review of the pathogenic mechanisms of and treatment strategies pertaining to the intestinal microbiota in patients with gastrointestinal tumours. And provide a more comprehensive and precise scientific basis for the development of microbiota-based treatments for gastrointestinal tumours and the prevention of such tumours.
Collapse
Affiliation(s)
- Jikai He
- Central Laboratory, Bayannur Hospital, Bayannur, 015000, Inner Mongolia, China
| | - Haijun Li
- Department of Gastrointestinal Surgery, Inner Mongolia Autonomous Region People's Hospital, Hohhot, 010017, Inner Mongolia, China
| | - Jiaqi Jia
- Graduate School of Youjiang Medical University for Nationalities, No. 98 Chengcheng Road, Youjiang District, Baise City, 533000, China
| | - Yang Liu
- Central Laboratory, Bayannur Hospital, Bayannur, 015000, Inner Mongolia, China
| | - Ning Zhang
- Central Laboratory, Bayannur Hospital, Bayannur, 015000, Inner Mongolia, China
| | - Rumeng Wang
- Central Laboratory, Bayannur Hospital, Bayannur, 015000, Inner Mongolia, China
| | - Wenhao Qu
- Graduate School of Youjiang Medical University for Nationalities, No. 98 Chengcheng Road, Youjiang District, Baise City, 533000, China
| | - Yanqi Liu
- Department of Gastroenterology, Affiliated Hospital of Inner Mongolia Medical University, Hohhot City, 010050, Inner Mongolia, China.
| | - Lizhou Jia
- Central Laboratory, Bayannur Hospital, Bayannur, 015000, Inner Mongolia, China.
| |
Collapse
|
19
|
Avtanski D, Reddy V, Stojchevski R, Hadzi-Petrushev N, Mladenov M. The Microbiome in the Obesity-Breast Cancer Axis: Diagnostic and Therapeutic Potential. Pathogens 2023; 12:1402. [PMID: 38133287 PMCID: PMC10747404 DOI: 10.3390/pathogens12121402] [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: 11/05/2023] [Revised: 11/25/2023] [Accepted: 11/27/2023] [Indexed: 12/23/2023] Open
Abstract
A growing body of evidence has demonstrated a relationship between the microbiome, adiposity, and cancer development. The microbiome is emerging as an important factor in metabolic disease and cancer pathogenesis. This review aimed to highlight the role of the microbiome in obesity and its association with cancer, with a particular focus on breast cancer. This review discusses how microbiota dysbiosis may contribute to obesity and obesity-related diseases, which are linked to breast cancer. It also explores the potential of the gut microbiome to influence systemic immunity, leading to carcinogenesis via the modulation of immune function. This review underscores the potential use of the microbiome profile as a diagnostic tool and treatment target, with strategies including probiotics, fecal microbiota transplantation, and dietary interventions. However, this emphasizes the need for more research to fully understand the complex relationship between the microbiome, metabolic disorders, and breast cancer. Future studies should focus on elucidating the mechanisms underlying the impact of the microbiome on breast cancer and exploring the potential of the microbiota profile as a biomarker and treatment target.
Collapse
Affiliation(s)
- Dimiter Avtanski
- Friedman Diabetes Institute, Lenox Hill Hospital, Northwell Health, New York, NY 10022, USA;
- Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| | - Varun Reddy
- New York Institute of Technology, College of Osteopathic Medicine, Old Westbury, NY 11545, USA;
| | - Radoslav Stojchevski
- Friedman Diabetes Institute, Lenox Hill Hospital, Northwell Health, New York, NY 10022, USA;
- Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| | - Nikola Hadzi-Petrushev
- Faculty of Natural Sciences and Mathematics, Institute of Biology, Ss. Cyril and Methodius University, 1000 Skopje, North Macedonia; (N.H.-P.); (M.M.)
| | - Mitko Mladenov
- Faculty of Natural Sciences and Mathematics, Institute of Biology, Ss. Cyril and Methodius University, 1000 Skopje, North Macedonia; (N.H.-P.); (M.M.)
| |
Collapse
|
20
|
Shi Q, Wang J, Zhou M, Zheng R, Zhang X, Liu B. Gut Lactobacillus contribute to the progression of breast cancer by affecting the anti-tumor activities of immune cells in the TME of tumor-bearing mice. Int Immunopharmacol 2023; 124:111039. [PMID: 37862739 DOI: 10.1016/j.intimp.2023.111039] [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: 07/19/2023] [Revised: 10/07/2023] [Accepted: 10/08/2023] [Indexed: 10/22/2023]
Abstract
Studies have proven that gut microbiota dysbiosis may influence the carcinogenesis and outcomes of multiple cancers. However, it is still unclear whether gut microbiota dysbiosis affect the progression of breast cancer, especially triple-negative breast cancer. In the present study, by using gut microbiota dysbiosis murine model established by treatment of mice with streptomycin, we found Lactobacillus and the metabolite-lactic acid are the pivotal factors for 4T1 tumor progression. In fact, streptomycin-treated mice exhibited slower tumor growth, in parallel with less abundance of Lactobacillus in the gut. Supplementation with Lactobacillus resulted in a rapid tumor growth, following a decrease in the expression of mRNAs for anti-tumor-related factors but an increase in the M2 polarization. The elevated percentages of IFN-γ-producing CD4+T cells and CD8+T cells in the tumor microenvironment of streptomycin-treated tumor-bearing mice may be vanished by supplementation of Lactobacillus. It seems likely that lactobacillus-mediated pro-tumor effect is related to the production of lactic acid. A decrease in the levels of lactic acid in the cecal feces and tumor tissues were observed in streptomycin-treated tumor bearing mice. However, supplementation of Lactobacillus can restore streptomycin-reduced concentration of lactic acid in the tumor tissues, suggesting that gut Lactobacillus are the source of lactic acid. Bioinformatics analysis result suggests high concentration of lactic acid in tumor sites may be related to the diminished anti-tumor immunity in the TME. This study reveals a correlation between gut Lactobacillus and tumor progression in a murine 4T1 tumor model, providing experimental evidence for clinical treatment of breast cancer.
Collapse
Affiliation(s)
- Qi Shi
- Department of Pathogenic Microbiology, School of Basic Medical Science, China Medical University, Shenyang 110122, China
| | - Jia Wang
- Department of Pathogenic Microbiology, School of Basic Medical Science, China Medical University, Shenyang 110122, China
| | - Mengnan Zhou
- Department of Pathogenic Microbiology, School of Basic Medical Science, China Medical University, Shenyang 110122, China
| | - Rui Zheng
- Department of Pathogenic Microbiology, School of Basic Medical Science, China Medical University, Shenyang 110122, China
| | - Xiaoli Zhang
- Department of Pathogenic Microbiology, School of Basic Medical Science, China Medical University, Shenyang 110122, China
| | - Beixing Liu
- Department of Pathogenic Microbiology, School of Basic Medical Science, China Medical University, Shenyang 110122, China.
| |
Collapse
|
21
|
Abdel Tawab FI, Abd Elkadr MH, Sultan AM, Hamed EO, El-Zayat AS, Ahmed MN. Probiotic potentials of lactic acid bacteria isolated from Egyptian fermented food. Sci Rep 2023; 13:16601. [PMID: 37789063 PMCID: PMC10547719 DOI: 10.1038/s41598-023-43752-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 09/27/2023] [Indexed: 10/05/2023] Open
Abstract
Lactic acid bacteria (LAB) are of major concern due to their health benefits. Fermented food products comprise variable LAB demonstrating probiotic properties. Discovering and evaluating new probiotics in fermented food products poses a global economic and health importance. Therefore, the present work aimed to investigate and evaluate the probiotic potentials of LAB strains isolated from Egyptian fermented food. In this study, we isolated and functionally characterized 100 bacterial strains isolated from different Egyptian fermented food sources as probiotics. Only four LAB strains amongst the isolated LAB showed probiotic attributes and are considered to be safe for their implementation as feed or dietary supplements. Additionally, they were shown to exert antimicrobial activities against pathogenic bacteria and anticancer effects against the colon cancer cell line Caco-2. The Enterococcus massiliensis IS06 strain was exclusively reported in this study as a probiotic strain with high antimicrobial, antioxidant, and anti-colon cancer activity. Hitherto, few studies have focused on elucidating the impact of probiotic supplementation in vivo. Therefore, in the current study, the safety of the four strains was tested in vivo through the supplementation of rats with potential probiotic strains for 21 days. The results revealed that probiotic bacterial supplementation in rats did not adversely affect the general health of rats. The Lactiplantibacillus plantarum IS07 strain significantly increased the growth performance of rats. Furthermore, the four strains exhibited increased levels of antioxidants such as superoxide dismutase and glutathione in vivo. Consistently, all strains also showed high antioxidant activity of the superoxide dismutase enzyme in vitro. Overall, these findings demonstrated that these isolated potential probiotics harbor desirable characteristics and can be applied widely as feed additives for animals or as dietary supplements for humans to exert their health benefits and combat serious diseases.
Collapse
Affiliation(s)
- Fatma I Abdel Tawab
- Oil Crops Biotechnology Lab, Agricultural Genetic Engineering Institute, Agricultural Research Center, Giza, Egypt
| | - Menna H Abd Elkadr
- Microbiology Lab, Research Park, Faculty of Agriculture, Cairo University, Giza, Egypt
| | - Amany M Sultan
- Department of Biochemistry, Toxicology Unit, Animal Health Research Institute, Agricultural Research Center, Giza, Egypt
| | - Ehdaa O Hamed
- Department of Biochemistry, Toxicology Unit, Animal Health Research Institute, Agricultural Research Center, Giza, Egypt
| | - Ayatollah S El-Zayat
- Department of Microbiology, Faculty of Agriculture, Cairo University, El-Gamaa Street, Giza, 12613, Egypt
| | - Marwa N Ahmed
- Department of Microbiology, Faculty of Agriculture, Cairo University, El-Gamaa Street, Giza, 12613, Egypt.
| |
Collapse
|
22
|
Hoskinson C, Jiang RY, Stiemsma LT. Elucidating the roles of the mammary and gut microbiomes in breast cancer development. Front Oncol 2023; 13:1198259. [PMID: 37664075 PMCID: PMC10470065 DOI: 10.3389/fonc.2023.1198259] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 07/26/2023] [Indexed: 09/05/2023] Open
Abstract
The mammary microbiome is a newly characterized bacterial niche that might offer biological insight into the development of breast cancer. Together with in-depth analysis of the gut microbiome in breast cancer, current evidence using next-generation sequencing and metabolic profiling suggests compositional and functional shifts in microbial consortia are associated with breast cancer. In this review, we discuss the fundamental studies that have progressed this important area of research, focusing on the roles of both the mammary tissue microbiome and the gut microbiome. From the literature, we identified the following major conclusions, (I) There are unique breast and gut microbial signatures (both compositional and functional) that are associated with breast cancer, (II) breast and gut microbiome compositional and breast functional dysbiosis represent potential early events of breast tumor development, (III) specific breast and gut microbes confer host immune responses that can combat breast tumor development and progression, and (IV) chemotherapies alter the microbiome and thus maintenance of a eubiotic microbiome may be key in breast cancer treatment. As the field expectantly advances, it is necessary for the role of the microbiome to continue to be elucidated using multi-omic approaches and translational animal models in order to improve predictive, preventive, and therapeutic strategies for breast cancer.
Collapse
Affiliation(s)
- Courtney Hoskinson
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
| | | | - Leah T. Stiemsma
- Natural Science Division, Pepperdine University, Malibu, CA, United States
| |
Collapse
|
23
|
Jing Y, Feng B, Gao J, Li J, Zhou G, Sun Z, Wang Y. BLAB2CancerKD: a knowledge graph database focusing on the association between lactic acid bacteria and cancer, but beyond. Database (Oxford) 2023; 2023:7176387. [PMID: 37221044 DOI: 10.1093/database/baad036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Revised: 04/23/2023] [Accepted: 04/28/2023] [Indexed: 05/25/2023]
Abstract
In a broad sense, lactic acid bacteria (LAB) is a general term for Gram-positive bacteria that can produce lactic acid by utilizing fermentable carbohydrates. It is widely used in essential fields such as industry, agriculture, animal husbandry and medicine. At the same time, LAB are closely related to human health. They can regulate human intestinal flora and improve gastrointestinal function and body immunity. Cancer, a disease in which some cells grow out of control and spread to other body parts, is one of the leading causes of human death worldwide. In recent years, the potential of LAB in cancer treatment has attracted attention. Mining knowledge from the scientific literature significantly accelerates its application in cancer treatment. Using 7794 literature studies of LAB cancer as source data, we have processed 16 543 biomedical concepts and 23 091 associations by using automatic text mining tools combined with manual curation of domain experts. An ontology containing 31 434 pieces of structured data is constructed. Finally, based on ontology, a knowledge graph (KG) database, which is called Beyond 'Lactic acid bacteria to Cancer Knowledge graph Database' (BLAB2CancerKD), is constructed by using KG and web technology. BLAB2CancerKD presents all the relevant knowledge intuitively and clearly in various data presentation forms, and the interactive system function also makes it more efficient. BLAB2CancerKD will be continuously updated to advance the research and application of LAB in cancer therapy. Researchers can visit BLAB2CancerKD at. Database URL http://110.40.139.2:18095/.
Collapse
Affiliation(s)
- Yi Jing
- Faculty of Science, The University of New South Wales, High Street, Sydney, New South Wales 2052, Australia
- Inner Mongolia Autonomous Region Key Laboratory of Big Data Research and Application for Agriculture and Animal Husbandry, Zhaowuda Road No. 306, Hohhot 010018, China
| | - Baiyang Feng
- Inner Mongolia Autonomous Region Key Laboratory of Big Data Research and Application for Agriculture and Animal Husbandry, Zhaowuda Road No. 306, Hohhot 010018, China
- College of Computer and Information Engineering, Inner Mongolia Agricultural University, Erdos East Street No. 29, Hohhot 010011, China
| | - Jing Gao
- Inner Mongolia Autonomous Region Key Laboratory of Big Data Research and Application for Agriculture and Animal Husbandry, Zhaowuda Road No. 306, Hohhot 010018, China
- College of Computer and Information Engineering, Inner Mongolia Agricultural University, Erdos East Street No. 29, Hohhot 010011, China
- Inner Mongolia Autonomous Region Big Data Center, Chilechuan Street No. 1, Hohhot 010091, China
| | - Jin Li
- Inner Mongolia Autonomous Region Key Laboratory of Big Data Research and Application for Agriculture and Animal Husbandry, Zhaowuda Road No. 306, Hohhot 010018, China
- College of Computer and Information Engineering, Inner Mongolia Agricultural University, Erdos East Street No. 29, Hohhot 010011, China
| | - Ganghui Zhou
- Inner Mongolia Autonomous Region Key Laboratory of Big Data Research and Application for Agriculture and Animal Husbandry, Zhaowuda Road No. 306, Hohhot 010018, China
- College of Computer and Information Engineering, Inner Mongolia Agricultural University, Erdos East Street No. 29, Hohhot 010011, China
| | - Zhihong Sun
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Zhaowuda Road No. 306, Hohhot 010018, China
| | - Yufei Wang
- The Affiliated Hospital of Inner Mongolia Medical University, Tongdao North road No.1, Hohhot 010050, China
| |
Collapse
|
24
|
Thakker DP, Narayanan R. Arginine deiminase produced by lactic acid bacteria as a potent anti-cancer drug. Med Oncol 2023; 40:175. [PMID: 37171497 DOI: 10.1007/s12032-023-02043-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Accepted: 04/30/2023] [Indexed: 05/13/2023]
Abstract
Bacterial-based cancer immunotherapy has recently gained widespread attention due to its exceptional mechanism of rich pathogen-associated molecular patterns in anti-cancer immune responses. Contrary to conventional cancer therapies such as surgery, chemotherapy, radiation and phototherapy, bacteria-based cancer immunotherapy has the unique ability to suppress cancer by selectively accumulating and growing in tumours. In the view of this, several bacterial strains are being used for the treatment of cancer. Of which, lactic acid bacteria are a powerful, albeit still inadequately understood bacteria that possess a wide source of bioactive chemicals. Lactic acid bacteria metabolites, such as bacteriocins, short-chain fatty acids, exopolysaccharides show antitumour property. Amino acid pathways, which have lately been focussed as a new strategy to cancer therapy, are key element of the adaptability and dysregulation of metabolic pathways identified in proliferation of tumour cells. Arginine metabolism, in particular, has been shown to be critical for cancer therapy. As a result, better understanding of arginine metabolism in LAB and cancer cells could lead to new cancer therapeutic targets. This review will outline current advances in the interaction of arginine metabolism with cancer therapy and propose an arginine deiminase expression system to combat cancer more effectively.
Collapse
Affiliation(s)
- Darshali P Thakker
- Department of Genetic Engineering, College of Engineering & Technology, SRM Institute of Science and Technology, SRM Nagar, Kattankulathur, 603203, Tamil Nadu, India
| | - Rajnish Narayanan
- Department of Genetic Engineering, College of Engineering & Technology, SRM Institute of Science and Technology, SRM Nagar, Kattankulathur, 603203, Tamil Nadu, India.
| |
Collapse
|
25
|
Abdel-Hamid HA, Marey H, Ibrahim MFG. Hemin protects against cell stress induced by estrogen and progesterone in rat mammary glands via modulation of Nrf2/HO-1 and NF-κB pathways. Cell Stress Chaperones 2023; 28:289-301. [PMID: 36930344 PMCID: PMC10167073 DOI: 10.1007/s12192-023-01337-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 02/19/2023] [Accepted: 03/08/2023] [Indexed: 03/18/2023] Open
Abstract
Mammary gland hyperplasia is one of the risk factors for breast cancer. Till date, there is no study that has addressed the effect of hemin in this condition. Thus, this study was designed to evaluate the effect of the heme oxygenase 1 (HO-1) inducer (hemin) and its inhibitor (zinc protoporphyrin-IX) (ZnPP-IX) on mammary gland hyperplasia (MGH) induced by estrogen and progesterone in adult albino rats. Forty adult female albino rats were divided into the control group, MGH group, MGH + Hemin group, and MGH + Hemin + ZnPP-IX group. Serum levels of estradiol and progesterone were measured. Breast tissues were taken for estimation of oxidative, inflammatory, and apoptotic markers. Mammary gland histology was performed, and expression of Ki-67, Beclin, and P53 in breast tissue was also measured. Estrogen and progesterone administration induced hyperplasia of cells lining the ducts of the breast tissues associated with increased diameter and height of the nipples as well as increased oxidative stress markers, inflammatory markers, antiapoptotic markers, and cell autophagy. Hemin administration during induction of MGH can reverse all the affected parameters. Then, these effects were abolished by ZnPP-IX administration. We concluded that hemin administration can antagonize the cell stress induced by estrogen and progesterone and protect against the development of mammary gland hyperplasia via modulation of Nrf2/HO-1 and NF-κB pathways.
Collapse
Affiliation(s)
- Heba A. Abdel-Hamid
- Department of Medical Physiology, Faculty of Medicine, Minia University, Minia, 61111 Egypt
- Department of Medical Physiology, Faculty of Medicine, Al-Baha University, Al Baha, Saudi Arabia
| | - Heba Marey
- Department of Medical Biochemistry, Faculty of Medicine, Minia University, Minia, 61111 Egypt
| | | |
Collapse
|
26
|
Kunika, Frey N, Rangrez AY. Exploring the Involvement of Gut Microbiota in Cancer Therapy-Induced Cardiotoxicity. Int J Mol Sci 2023; 24:7261. [PMID: 37108423 PMCID: PMC10138392 DOI: 10.3390/ijms24087261] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 04/04/2023] [Accepted: 04/12/2023] [Indexed: 04/29/2023] Open
Abstract
Trillions of microbes in the human intestinal tract, including bacteria, viruses, fungi, and protozoa, are collectively referred to as the gut microbiome. Recent technological developments have led to a significant increase in our understanding of the human microbiome. It has been discovered that the microbiome affects both health and the progression of diseases, including cancer and heart disease. Several studies have indicated that the gut microbiota may serve as a potential target in cancer therapy modulation, by enhancing the effectiveness of chemotherapy and/or immunotherapy. Moreover, altered microbiome composition has been linked to the long-term effects of cancer therapy; for example, the deleterious effects of chemotherapy on microbial diversity can, in turn, lead to acute dysbiosis and serious gastrointestinal toxicity. Specifically, the relationship between the microbiome and cardiac diseases in cancer patients following therapy is poorly understood. In this article, we provide a summary of the role of the microbiome in cancer treatment, while also speculating on a potential connection between treatment-related microbial changes and cardiotoxicity. Through a brief review of the literature, we further explore which bacterial families or genera were differentially affected in cancer treatment and cardiac disease. A deeper understanding of the link between the gut microbiome and cardiotoxicity caused by cancer treatment may help lower the risk of this critical and potentially fatal side effect.
Collapse
Affiliation(s)
- Kunika
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, 69120 Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, 69120 Heidelberg, Germany
| | - Norbert Frey
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, 69120 Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, 69120 Heidelberg, Germany
| | - Ashraf Y. Rangrez
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, 69120 Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, 69120 Heidelberg, Germany
| |
Collapse
|
27
|
Tanitame M, Sugawara Y, Lu Y, Matsuyama S, Kanemura S, Fukao A, Tsuji I. Dairy consumption and incident risk of thyroid cancer in Japan: a pooled analysis of the Miyagi Cohort Study and the Ohsaki Cohort Study. Eur J Nutr 2023; 62:251-259. [PMID: 35951088 DOI: 10.1007/s00394-022-02979-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 07/29/2022] [Indexed: 02/07/2023]
Abstract
PURPOSE The impact of dairy consumption on thyroid cancer is unclear. The purpose of this study was to elucidate the association between dairy consumption and the risk of thyroid cancer in Japanese people. METHODS The association between dairy consumption and the risk of thyroid cancer in Japanese people was examined by conducting a pooled analysis of two prospective studies of residents in Miyagi Prefecture, Japan. Data from 64,340 men and women aged 40-79 years registered in the Miyagi Cohort Study in 1990 and in the Ohsaki Cohort Study in 1994 were analyzed. Dairy consumption was assessed at baseline using a self-administered food frequency questionnaire and was divided into quartiles based on the weight (in grams) of total dairy consumption per day. RESULTS During 1,075,018 person-years of follow-up, there were 190 incident cases of thyroid cancer (29 men and 161 women). The hazard ratios (HRs) and 95% confidence intervals (CIs) for thyroid cancer incidence in the highest quartile of dairy consumption compared with the lowest quartile were 0.83 (95% CIs 0.28-2.43, P-trend = 0.823) for men and 0.67 (95% CIs 0.42-1.06, P-trend = 0.056) for women. After stratification for BMI, a decreased risk was observed in women with BMI ≥ 25 kg/m2 (HRs: 0.37, 95% CIs 0.18-0.79, P-trend = 0.010). CONCLUSION Dairy consumption is inversely associated with the risk of thyroid cancer in women with BMI ≥ 25 kg/m2.
Collapse
Affiliation(s)
- Marina Tanitame
- Division of Epidemiology, Department of Health Informatics and Public Health, Graduate School of Medicine, Tohoku University School of Public Health, 2-1, Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| | - Yumi Sugawara
- Division of Epidemiology, Department of Health Informatics and Public Health, Graduate School of Medicine, Tohoku University School of Public Health, 2-1, Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan.
| | - Yukai Lu
- Division of Epidemiology, Department of Health Informatics and Public Health, Graduate School of Medicine, Tohoku University School of Public Health, 2-1, Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| | - Sanae Matsuyama
- Division of Epidemiology, Department of Health Informatics and Public Health, Graduate School of Medicine, Tohoku University School of Public Health, 2-1, Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| | - Seiki Kanemura
- Division of Epidemiology, Miyagi Prefectural Cancer Research Center, Natori, Miyagi, Japan
| | - Akira Fukao
- Miyagi Cancer Society, Sendai, Miyagi, Japan
| | - Ichiro Tsuji
- Division of Epidemiology, Department of Health Informatics and Public Health, Graduate School of Medicine, Tohoku University School of Public Health, 2-1, Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| |
Collapse
|
28
|
Thekkekkara D, Manjula SN, Mishra N, Bhatt S, Shilpi S. Synbiotics in the Management of Breast Cancer. SYNBIOTICS FOR THE MANAGEMENT OF CANCER 2023:289-304. [DOI: 10.1007/978-981-19-7550-9_14] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
29
|
Nandi D, Parida S, Sharma D. The gut microbiota in breast cancer development and treatment: The good, the bad, and the useful! Gut Microbes 2023; 15:2221452. [PMID: 37305949 PMCID: PMC10262790 DOI: 10.1080/19490976.2023.2221452] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 05/26/2023] [Indexed: 06/13/2023] Open
Abstract
Regardless of the global progress in early diagnosis and novel therapeutic regimens, breast carcinoma poses a devastating threat, and the advances are somewhat marred by high mortality rates. Breast cancer risk prediction models based on the known risk factors are extremely useful, but a large number of breast cancers develop in women with no/low known risk. The gut microbiome exerts a profound impact on the host health and physiology and has emerged as a pivotal frontier in breast cancer pathogenesis. Progress in metagenomic analysis has enabled the identification of specific changes in the host microbial signature. In this review, we discuss the microbial and metabolomic changes associated with breast cancer initiation and metastatic progression. We summarize the bidirectional impact of various breast cancer-related therapies on gut microbiota and vice-versa. Finally, we discuss the strategies to modulate the gut microbiota toward a more favorable state that confers anticancer effects.
Collapse
Affiliation(s)
- Deeptashree Nandi
- Dept. of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA
| | - Sheetal Parida
- Dept. of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA
| | - Dipali Sharma
- Dept. of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA
| |
Collapse
|
30
|
Lacticaseibacillus rhamnosus Probio-M9-Driven Mouse Mammary Tumor-Inhibitory Effect Is Accompanied by Modulation of Host Gut Microbiota, Immunity, and Serum Metabolome. Nutrients 2022; 15:nu15010005. [PMID: 36615662 PMCID: PMC9824041 DOI: 10.3390/nu15010005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 12/10/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022] Open
Abstract
Gut microbiome may influence tumor growth and cancer treatment efficacy, so it is a potential target for tumor prevention/treatment. This pilot study investigated the preventive and therapeutic effects of a probiotic strain, Lacticaseibacillus rhamnosus Probio-M9 (Probio-M9), against murine mammary cancer. Thirty-six female mice were randomly divided into three groups (n = 12 per group): control (without tumor transplantation), model (tumor transplantation; no probiotic administration), and probiotic (30-day oral gavage of probiotic, started seven days before tumor transplantation). Changes in tumor size were recorded, and blood, tumor tissue, and stool samples were collected at the end of the trial for analyses. Comparing with the model group, the probiotic group had a significantly smaller tumor volume (p < 0.05), a higher fecal microbiota Shannon diversity index, with significant modifications in the gut microbiota structure (p < 0.05), characterized by more Alistipes sp._2, Porphyromonadaceae bacterium_7, and Bacteroidales bacterium 55_9 (p < 0.05). Additionally, Probio-M9 administration elevated the serum IFN-γ, IL-9, IL-13, and IL-27 levels and several metabolites (e.g., pyridoxal, nicotinic acid, 3-hydroxybutyric acid, glutamine; p < 0.05), while reducing IL-5 (p < 0.05). These changes might be associated with the protective effect of Probio-M9 against mammary tumor growth. Thus, probiotic administration could harness host gut microbiome in anti-cancer responses.
Collapse
|
31
|
Devoy C, Flores Bueso Y, Tangney M. Understanding and harnessing triple-negative breast cancer-related microbiota in oncology. Front Oncol 2022; 12:1020121. [PMID: 36505861 PMCID: PMC9730816 DOI: 10.3389/fonc.2022.1020121] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 10/31/2022] [Indexed: 11/27/2022] Open
Abstract
Bacterial inhabitants of the body have the potential to play a role in various stages of cancer initiation, progression, and treatment. These bacteria may be distal to the primary tumour, such as gut microbiota, or local to the tissue, before or after tumour growth. Breast cancer is well studied in this context. Amongst breast cancer types, Triple Negative Breast Cancer (TNBC) is more aggressive, has fewer treatment options than receptor-positive breast cancers, has an overall worse prognosis and higher rates of reoccurrence. Thus, an in-depth understanding of the bacterial influence on TNBC progression and treatment is of high value. In this regard, the Gut Microbiota (GM) can be involved in various stages of tumour progression. It may suppress or promote carcinogenesis through the release of carcinogenic metabolites, sustenance of proinflammatory environments and/or the promotion of epigenetic changes in our genome. It can also mediate metastasis and reoccurrence through interactions with the immune system and has been recently shown to influence chemo-, radio-, and immune-therapies. Furthermore, bacteria have also been found to reside in normal and malignant breast tissue. Several studies have now described the breast and breast tumour microbiome, with the tumour microbiota of TNBC having the least taxonomic diversity among all breast cancer types. Here, specific conditions of the tumour microenvironment (TME) - low O2, leaky vasculature and immune suppression - are supportive of tumour selective bacterial growth. This innate bacterial ability could enable their use as delivery agents for various therapeutics or as diagnostics. This review aims to examine the current knowledge on bacterial relevance to TNBC and potential uses while examining some of the remaining unanswered questions regarding mechanisms underpinning observed effects.
Collapse
Affiliation(s)
- Ciaran Devoy
- Cancer Research@UCC, College of Medicine and Health, University College Cork, Cork, Ireland,SynBio Center, University College Cork, Cork, Ireland,APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Yensi Flores Bueso
- Cancer Research@UCC, College of Medicine and Health, University College Cork, Cork, Ireland,SynBio Center, University College Cork, Cork, Ireland,APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Mark Tangney
- Cancer Research@UCC, College of Medicine and Health, University College Cork, Cork, Ireland,SynBio Center, University College Cork, Cork, Ireland,APC Microbiome Ireland, University College Cork, Cork, Ireland,School of Pharmacy, College of Medicine and Health, University College Cork, Cork, Ireland,*Correspondence: Mark Tangney,
| |
Collapse
|
32
|
Wang Z, Li L, Wang S, Wei J, Qu L, Pan L, Xu K. The role of the gut microbiota and probiotics associated with microbial metabolisms in cancer prevention and therapy. Front Pharmacol 2022; 13:1025860. [PMID: 36452234 PMCID: PMC9702994 DOI: 10.3389/fphar.2022.1025860] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 11/01/2022] [Indexed: 11/29/2023] Open
Abstract
Cancer is the second leading cause of elevated mortality worldwide. Thus, the development of drugs and treatments is needed to enhance the survival rate of the cancer-affected population. Recently, gut microbiota research in the healthy development of the human body has garnered widespread attention. Many reports indicate that changes in the gut microbiota are strongly associated with chronic inflammation-related diseases, including colitis, liver disease, and cancer within the intestine and the extraintestinal tract. Different gut bacteria are vital in the occurrence and development of tumors within the gut and extraintestinal tract. The human gut microbiome has significant implications for human physiology, including metabolism, nutrient absorption, and immune function. Moreover, diet and lifestyle habits are involved in the evolution of the human microbiome throughout the lifetime of the host and are involved in drug metabolism. Probiotics are a functional food with a protective role in cancer development in animal models. Probiotics alter the gut microbiota in the host; thus, beneficial bacterial activity is stimulated, and detrimental activity is inhibited. Clinical applications have revealed that some probiotic strains could reduce the occurrence of postoperative inflammation among cancer patients. An association network was constructed by analyzing the previous literature to explore the role of probiotics from the anti-tumor perspective. Therefore, it provides direction and insights for research on tumor treatment.
Collapse
Affiliation(s)
- Zijun Wang
- Hubei Engineering Technology Research Center of Chinese Materia Medica Processing, College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
| | - Lanqing Li
- Hubei Engineering Technology Research Center of Chinese Materia Medica Processing, College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
| | - Shunshun Wang
- Hubei Engineering Technology Research Center of Chinese Materia Medica Processing, College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
| | - Jing Wei
- Hubei Engineering Technology Research Center of Chinese Materia Medica Processing, College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
| | - Linghang Qu
- Hubei Engineering Technology Research Center of Chinese Materia Medica Processing, College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
| | - Lianhong Pan
- Chongqing Key Laboratory of Development and Utilization of Genuine Medicinal Materials in Three Gorges Reservoir Area, Chongqing Engineering Research Center of Antitumor Natural Drugs, Chongqing Three Gorges Medical College, Chongqing, China
| | - Kang Xu
- Hubei Engineering Technology Research Center of Chinese Materia Medica Processing, College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
| |
Collapse
|
33
|
Son YM, Kim J. The Microbiome-Immune Axis Therapeutic Effects in Cancer Treatments. J Microbiol Biotechnol 2022; 32:1086-1097. [PMID: 36116940 PMCID: PMC9628962 DOI: 10.4014/jmb.2208.08002] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 08/15/2022] [Accepted: 08/16/2022] [Indexed: 12/15/2022]
Abstract
During the last decades, research and therapeutic methods in cancer treatment have been evolving. As the results, nowadays, cancer patients are receiving several types of treatments, ranging from chemotherapy and radiation therapy to surgery and immunotherapy. In fact, most cancer patients take a combination of current anti-cancer therapies to improve the efficacy of treatment. However, current strategies still cause some side effects to patients, such as pain and depression. Therefore, there is the need to discover better ways to eradicate cancer whilst minimizing side effects. Recently, immunotherapy, particularly immune checkpoint blockade, is rising as an effective anti-cancer treatment. Unlike chemotherapy or radiation therapy, immunotherapy has few side effects and a higher tumor cell removal efficacy depend on cellular immunological mechanisms. Moreover, recent studies suggest that tissue immune responses are regulated by their microbiome composition. Each tissue has their specific microenvironment, which makes their microbiome composition different, particularly in the context of different types of cancer, such as breast, colorectal, kidney, lung, and skin. Herein, we review the current understanding of the relationship of immune responses and tissue microbiome in cancer in both animal and human studies. Moreover, we discuss the cancermicrobiome-immune axis in the context of cancer development and treatment. Finally, we speculate on strategies to control tissue microbiome alterations that may synergistically affect the immune system and impact cancer treatment outcomes.
Collapse
Affiliation(s)
- Young Min Son
- Department of Systems Biotechnology, Chung-Ang University, Anseong 17546, Republic of Korea,Corresponding author Phone: +82-31-670-4792 E-mail:
| | - Jihwan Kim
- Department of Systems Biotechnology, Chung-Ang University, Anseong 17546, Republic of Korea
| |
Collapse
|
34
|
Interaction between gut microbiota and tumour chemotherapy. Clin Transl Oncol 2022; 24:2330-2341. [DOI: 10.1007/s12094-022-02919-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 08/01/2022] [Indexed: 11/26/2022]
|
35
|
Wu H, Ganguly S, Tollefsbol TO. Modulating Microbiota as a New Strategy for Breast Cancer Prevention and Treatment. Microorganisms 2022; 10:microorganisms10091727. [PMID: 36144329 PMCID: PMC9503838 DOI: 10.3390/microorganisms10091727] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 08/23/2022] [Accepted: 08/25/2022] [Indexed: 11/18/2022] Open
Abstract
Breast cancer (BC) is the most common cancer in women in the United States. There has been an increasing incidence and decreasing mortality rate of BC cases over the past several decades. Many risk factors are associated with BC, such as diet, aging, personal and family history, obesity, and some environmental factors. Recent studies have shown that healthy individuals and BC patients have different microbiota composition, indicating that microbiome is a new risk factor for BC. Gut and breast microbiota alterations are associated with BC prognosis. This review will evaluate altered microbiota populations in gut, breast tissue, and milk of BC patients, as well as mechanisms of interactions between microbiota modulation and BC. Probiotics and prebiotics are commercially available dietary supplements to alleviate side-effects of cancer therapies. They also shape the population of human gut microbiome. This review evaluates novel means of modulating microbiota by nutritional treatment with probiotics and prebiotics as emerging and promising strategies for prevention and treatment of BC. The mechanistic role of probiotic and prebiotics partially depend on alterations in estrogen metabolism, systematic immune regulation, and epigenetics regulation.
Collapse
Affiliation(s)
- Huixin Wu
- Department of Biology, University of Alabama at Birmingham, 1300 University Boulevard, Birmingham, AL 35294, USA
| | - Sebanti Ganguly
- Department of Biology, University of Alabama at Birmingham, 1300 University Boulevard, Birmingham, AL 35294, USA
| | - Trygve O. Tollefsbol
- Department of Biology, University of Alabama at Birmingham, 1300 University Boulevard, Birmingham, AL 35294, USA
- Integrative Center for Aging Research, University of Alabama Birmingham, 1530 3rd Avenue South, Birmingham, AL 35294, USA
- O’Neal Comprehensive Cancer Center, University of Alabama Birmingham, 1802 6th Avenue South, Birmingham, AL 35294, USA
- Nutrition Obesity Research Center, University of Alabama Birmingham, 1675 University Boulevard, Birmingham, AL 35294, USA
- Comprehensive Diabetes Center, University of Alabama Birmingham, 1825 University Boulevard, Birmingham, AL 35294, USA
- University Wide Microbiome Center, University of Alabama Birmingham, 845 19th Street South, Birmingham, AL 35294, USA
- Correspondence: ; Tel.: +1-205-934-4573; Fax: +1-205-975-6097
| |
Collapse
|
36
|
Dicks LMT, Vermeulen W. Do Bacteria Provide an Alternative to Cancer Treatment and What Role Does Lactic Acid Bacteria Play? Microorganisms 2022; 10:microorganisms10091733. [PMID: 36144335 PMCID: PMC9501580 DOI: 10.3390/microorganisms10091733] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 08/17/2022] [Accepted: 08/26/2022] [Indexed: 11/16/2022] Open
Abstract
Cancer is one of the leading causes of mortality and morbidity worldwide. According to 2022 statistics from the World Health Organization (WHO), close to 10 million deaths have been reported in 2020 and it is estimated that the number of cancer cases world-wide could increase to 21.6 million by 2030. Breast, lung, thyroid, pancreatic, liver, prostate, bladder, kidney, pelvis, colon, and rectum cancers are the most prevalent. Each year, approximately 400,000 children develop cancer. Treatment between countries vary, but usually includes either surgery, radiotherapy, or chemotherapy. Modern treatments such as hormone-, immuno- and antibody-based therapies are becoming increasingly popular. Several recent reports have been published on toxins, antibiotics, bacteriocins, non-ribosomal peptides, polyketides, phenylpropanoids, phenylflavonoids, purine nucleosides, short chain fatty acids (SCFAs) and enzymes with anticancer properties. Most of these molecules target cancer cells in a selective manner, either directly or indirectly through specific pathways. This review discusses the role of bacteria, including lactic acid bacteria, and their metabolites in the treatment of cancer.
Collapse
|
37
|
Ali A, Ara A, Kashyap MK. Gut microbiota: Role and Association with Tumorigenesis in Different Malignancies. Mol Biol Rep 2022; 49:8087-8107. [PMID: 35543828 DOI: 10.1007/s11033-022-07357-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 03/01/2022] [Accepted: 03/10/2022] [Indexed: 02/07/2023]
Abstract
The microbiota has been associated with different cancer and may vary from patient to patient. A specific microbial strain can alter the progression of cancer and therapeutic outcome in response to anti-cancer therapy. The variations in microbiota contributed due to the individual microbiome of the microorganism are responsible for diverse clinical outcomes. The expansion of microbiota subpopulation during dysbiosis can lead to toxin production, inducing inflammation and cancer. The microbiota can be a dual-edged sword because it can be tumor-suppressive or oncogenic in the case of the gut. The transition of cancer cells from early to late-stage also impacts the composition of the microbiota, and this alteration could change the behavior of cancer. Multi-omics platforms derived data from an individual's multi-dimensional data (DNA, mRNA, microRNA, protein, metabolite, microbiota, and microbiome), i.e., individualome, to exploit it for personalized tailored treatment for different cancers in a precise manner. A number of studies suggest the importance of microbiota and its add-in suitability to existing treatment options for different malignancies. Furthermore, in vitro, and in vivo studies and cancer clinical trials suggest that probiotics have driven modulation of gut microbiota and other sites discourage the aggressive behavior and progression of different cancers.
Collapse
Affiliation(s)
- Altamas Ali
- Department of Biosciences, Jamia Millia Islamia (A central University), Jamia Nagar, 110025, New Delhi, India
| | - Anam Ara
- Department of Biosciences, Jamia Millia Islamia (A central University), Jamia Nagar, 110025, New Delhi, India
| | - Manoj Kumar Kashyap
- Amity Stem Cell Institute/Amity Medical School, Amity University Haryana, Amity Education Valley, Panchgaon (Manesar), Gurugram, HR, 122413, India.
| |
Collapse
|
38
|
The crosstalk of the human microbiome in breast and colon cancer: A metabolomics analysis. Crit Rev Oncol Hematol 2022; 176:103757. [PMID: 35809795 DOI: 10.1016/j.critrevonc.2022.103757] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 06/28/2022] [Accepted: 07/04/2022] [Indexed: 11/20/2022] Open
Abstract
The human microbiome's role in colon and breast cancer is described in this review. Understanding how the human microbiome and metabolomics interact with breast and colon cancer is the chief area of this study. First, the role of the gut and distal microbiome in breast and colon cancer is investigated, and the direct relationship between microbial dysbiosis and breast and colon cancer is highlighted. This work also focuses on the many metabolomic techniques used to locate prospective biomarkers, make an accurate diagnosis, and research new therapeutic targets for cancer treatment. This review clarifies the influence of anti-tumor medications on the microbiota and the proactive measures that can be taken to treat cancer using a variety of therapies, including radiotherapy, chemotherapy, next-generation biotherapeutics, gene-based therapy, integrated omics technology, and machine learning.
Collapse
|
39
|
The Effect of the Gut Microbiota on Systemic and Anti-Tumor Immunity and Response to Systemic Therapy against Cancer. Cancers (Basel) 2022; 14:cancers14153563. [PMID: 35892821 PMCID: PMC9330582 DOI: 10.3390/cancers14153563] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/08/2022] [Accepted: 07/19/2022] [Indexed: 12/12/2022] Open
Abstract
Gut microbiota can have opposing functions from pro-tumorigenic to anti-tumorigenic effects. Increasing preclinical and clinical evidence suggests that the intestinal microbiota affects cancer patients’ response to immune checkpoint inhibitors (ICIs) immunotherapy, such as anti-programmed cell death protein 1 (PD-1) and its ligand (PD-L1) and anti-cytotoxic T lymphocyte-associated protein 4 (CTLA-4). Microbiota-induced inflammation possibly contributes to tumor growth and cancer development. Microbiota-derived metabolites can also be converted to carcinogenic agents related to genetic mutations and DNA damage in organs such as the colon. However, other attributes of microbiota, such as greater diversity and specific bacterial species and their metabolites, are linked to better clinical outcomes and potentially improved anti-tumor immunity. In addition, the intratumoral microbial composition strongly affects T-cell-mediated cytotoxicity and anti-tumor immune surveillance, adding more complexity to the cancer-microbiome-immune axis. Despite the emerging clinical evidence for the activity of the gut microbiota in immuno-oncology, the fundamental mechanisms of such activity are not well understood. This review provides an overview of underlying mechanisms by which the gut microbiota and its metabolites enhance or suppress anti-tumor immune responses. Understanding such mechanisms allows for better design of microbiome-specific treatment strategies to improve the clinical outcome in cancer patients undergoing systemic therapy.
Collapse
|
40
|
Di Modica M, Arlotta V, Sfondrini L, Tagliabue E, Triulzi T. The Link Between the Microbiota and HER2+ Breast Cancer: The New Challenge of Precision Medicine. Front Oncol 2022; 12:947188. [PMID: 35912227 PMCID: PMC9326166 DOI: 10.3389/fonc.2022.947188] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 06/22/2022] [Indexed: 12/12/2022] Open
Abstract
The microbiota is emerging as a key player in cancer due to its involvement in several host physiological functions, including digestion, development of the immune system, and modulation of endocrine function. Moreover, its participation in the efficacy of anticancer treatments has been well described. For instance, the involvement of the breast microbiota in breast cancer (BC) development and progression has gained ground in the past several years. In this review, we report and discuss new findings on the impact of the gut and breast microbiota on BC, focusing on the HER2+ BC subtype, and the possibility of defining microbial signatures that are associated with disease aggressiveness, treatment response, and therapy toxicity. We also discuss novel insights into the mechanisms through which microorganism-host interactions occur and the possibility of microbiota editing in the prevention and treatment optimization of BC.
Collapse
Affiliation(s)
- Martina Di Modica
- Molecular Targeting Unit, Department of Research, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Nazionale dei Tumori, Milan, Italy
| | - Valeria Arlotta
- Molecular Targeting Unit, Department of Research, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Nazionale dei Tumori, Milan, Italy
| | - Lucia Sfondrini
- Molecular Targeting Unit, Department of Research, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Nazionale dei Tumori, Milan, Italy
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, Milan, Italy
| | - Elda Tagliabue
- Molecular Targeting Unit, Department of Research, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Nazionale dei Tumori, Milan, Italy
- *Correspondence: Elda Tagliabue,
| | - Tiziana Triulzi
- Molecular Targeting Unit, Department of Research, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Nazionale dei Tumori, Milan, Italy
| |
Collapse
|
41
|
Song X, Wei C, Li X. The Relationship Between Microbial Community and Breast Cancer. Front Cell Infect Microbiol 2022; 12:849022. [PMID: 35782150 PMCID: PMC9245449 DOI: 10.3389/fcimb.2022.849022] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 05/24/2022] [Indexed: 11/18/2022] Open
Abstract
Breast cancer (BC) is the most common cancer in women and the leading cause of cancer-related deaths in women worldwide. Recent research studies have shown that the intestinal flora is related to the occurrence and progression of BC. Notably, some evidence identifies a unique microbial community in breast tissue, a site previously thought to be sterile. In addition, breast tumors have their own specific microbial community, distinct from normal mammary gland tissue, and all of them may result from intestinal flora. Some microbial community in breast tissue may lead to the occurrence and development of BC. This review focuses on the relationship between the microbial community and breast cancer, which will lay a solid theoretical foundation for further understanding the local microenvironment of BC and developing effective targeted therapeutic drugs.
Collapse
Affiliation(s)
- Xuelian Song
- Department of The Graduate Student, Shandong First Medical University, Tai’an, China
| | - Changran Wei
- Department of The First Clinical Medical School, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiangqi Li
- Department of Breast Surgery, The Second Affiliated Hospital of Shandong First Medical University, Tai’an, China
| |
Collapse
|
42
|
Ma Z, Qu M, Wang X. Analysis of Gut Microbiota in Patients with Breast Cancer and Benign Breast Lesions. Pol J Microbiol 2022; 71:217-226. [PMID: 35675827 PMCID: PMC9252143 DOI: 10.33073/pjm-2022-019] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 03/21/2022] [Indexed: 11/05/2022] Open
Abstract
Breast cancer (BC) and benign breast lesions (BBLs) are common diseases in women worldwide. The gut microbiota plays a vital role in regulating breast diseases' formation, progression, and therapy response. Hence, we explored the structure and function of gut microflora in patients with BC and BBLs. A cohort of 66 subjects was enrolled in the study. Twenty-six subjects had BC, 20 subjects had BBLs, and 20 matched healthy controls. High throughput 16S ribosomal RNA (16S rRNA) gene sequencing technology was used to determine the microbial community structure. Compared with healthy individuals, BC patients had significantly lower alpha diversity indices (Sobs index, p = 0.019; Chao1 index, p = 0.033). Sobs and Chao1 indices were also lower in patients with BBLs than healthy individuals, without statistical significance (p = 0.279, p = 0.314, respectively). Both unweighted and weighted UniFrac analysis showed that beta diversity differed significantly among the three groups (p = 3.376e-14, p < 0.001, respectively). Compared with healthy individuals, the levels of Porphyromonas and Peptoniphilus were higher in BC patients (p = 0.004, p = 0.007, respectively), whereas Escherichia and Lactobacillus were more enriched in the benign breast lesion group (p < 0.001, p = 0.011, respectively). Our study indicates that patients with BC and BBLs may undergo significant changes in intestinal microbiota. These findings can help elucidate the role of intestinal flora in BC and BBLs patients.
Collapse
Affiliation(s)
- Zhijun Ma
- Department of Surgical Oncology, The Affiliated Hospital of Qinghai University, Xining, China
| | - Manli Qu
- Graduate School of Qinghai University, Xining, China
| | - Xiaowu Wang
- Department of Surgical Oncology, The Affiliated Hospital of Qinghai University, Xining, China
| |
Collapse
|
43
|
Wenhui Y, Zhongyu X, Kai C, Zhaopeng C, Jinteng L, Mengjun M, Zepeng S, Yunshu C, Peng W, Yanfeng W, Huiyong S. Variations in the Gut Microbiota in Breast Cancer Occurrence and Bone Metastasis. Front Microbiol 2022; 13:894283. [PMID: 35722347 PMCID: PMC9204246 DOI: 10.3389/fmicb.2022.894283] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 04/29/2022] [Indexed: 01/18/2023] Open
Abstract
Breast cancer is the most common cancer in women and the second most common cancer overall. Although advancements in the early diagnosis and therapy of breast cancer have occurred in recent years, the prognosis of breast cancer bone metastasis remains poor and this type of cancer is rarely cured. The gut microbiota is indispensable for internal homeostasis and regulates various biological processes. Understanding the gut microbiota profiles in normal controls (NCs), breast cancer patients with no metastasis (BNs), and breast cancer patients with bone metastasis (BMs) may shed light on the development of diagnostic and therapeutic targets for breast cancer and bone metastasis. We comprehensively analyzed the gut microbiota from NCs, BNs, and BMs and found that the community diversity decreased in the order of NCs, BNs, and BMs. Streptococcus, Campylobacter and Moraxellaceae showed higher abundances in BNs and BMs than in NCs. The lack of Megamonas and Akkermansia in the BM compared with those in the NC and BN groups was considered related to bone metastasis. Additionally, based on the distinct gut microbiota profiles, we predicted that lipid transportation and metabolism, as well as folate biosynthesis, participate in breast cancer occurrence and that steroid hormone biosynthesis influences bone metastasis. Our study demonstrated that variations in gut microbiota are associated with breast cancer occurrence and bone metastasis, providing attractive targets to develop therapeutic and diagnostic methods.
Collapse
Affiliation(s)
- Yu Wenhui
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Xie Zhongyu
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Chen Kai
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Cai Zhaopeng
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Li Jinteng
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Ma Mengjun
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Su Zepeng
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Che Yunshu
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Wang Peng
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
- *Correspondence: Wang Peng,
| | - Wu Yanfeng
- Center for Biotherapy, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
- Wu Yanfeng,
| | - Shen Huiyong
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
- Shen Huiyong,
| |
Collapse
|
44
|
Meng Z, Ye Z, Zhu P, Zhu J, Fang S, Qiu T, Li Y, Meng L. New Developments and Opportunities of Microbiota in Treating Breast Cancers. Front Microbiol 2022; 13:818793. [PMID: 35633703 PMCID: PMC9134200 DOI: 10.3389/fmicb.2022.818793] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Accepted: 04/04/2022] [Indexed: 01/01/2023] Open
Abstract
Despite the prevalence of breast cancer (BC), over half of BC cases are unrelated to known risk factors, which highlights the importance of uncovering more cancer-related factors. Currently, the microbiota has been proven to be a potent modulator of the tumor environment in BC, which regulates the immune balance in tumor-related networks. Through a large amount of data accumulation, the microbiota has shown many possibilities to reveal more insights into the development or control of BC. To expand the potential benefits of patients with BC, this study discusses the distribution profile and the effect mechanism of BC-related microbiota on tumors and further discusses its impact on different tumor therapies. Finally, we summarize the possibility of targeting microbiological therapies to improve BC treatment or in combination with other therapies.
Collapse
Affiliation(s)
- Zihui Meng
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, China
| | - Zixuan Ye
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, China
| | - Pengrong Zhu
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, China
| | | | | | - Tianzhu Qiu
- Department of Oncology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- *Correspondence: Tianzhu Qiu,
| | - Yanan Li
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, China
- Yanan Li,
| | - Lijuan Meng
- Department of Geriatric Oncology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Lijuan Meng,
| |
Collapse
|
45
|
Wang Y, Dong J, Wang J, Chi W, Zhou W, Tian Q, Hong Y, Zhou X, Ye H, Tian X, Hu R, Wong A. Assessing the drug resistance profiles of oral probiotic lozenges. J Oral Microbiol 2022; 14:2019992. [PMID: 35024089 PMCID: PMC8745366 DOI: 10.1080/20002297.2021.2019992] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Background Probiotic lozenges have been developed to harvest the benefits of probiotics for oral health, but their long-term consumption may encourage the transfer of resistance genes from probiotics to commensals, and eventually to disease-causing bacteria. Aim To screen commercial probiotic lozenges for resistance to antibiotics, characterize the resistance determinants, and examine their transferability in vitro. Results Probiotics of all lozenges were resistant to glycopeptide, sulfonamide, and penicillin antibiotics, while some were resistant to aminoglycosides and cephalosporins. High minimum inhibitory concentrations (MICs) were detected for streptomycin (>128 µg/mL) and chloramphenicol (> 512 µg/mL) for all probiotics but only one was resistant to piperacillin (MIC = 32 µg/mL). PCR analysis detected erythromycin (erm(T), ermB or mefA) and fluoroquinolone (parC or gyr(A)) resistance genes in some lozenges although there were no resistant phenotypes. The dfrD, cat-TC, vatE, aadE, vanX, and aph(3")-III or ant(2")-I genes conferring resistance to trimethoprim, chloramphenicol, quinupristin/dalfopristin, vancomycin, and streptomycin, respectively, were detected in resistant probiotics. The rifampicin resistance gene rpoB was also present. We found no conjugal transfer of streptomycin resistance genes in our co-incubation experiments. Conclusion Our study represents the first antibiotic resistance profiling of probiotics from oral lozenges, thus highlighting the health risk especially in the prevailing threat of drug resistance globally.
Collapse
Affiliation(s)
- Yi Wang
- Department of Orthodontics, School and Hospital of Stomatology, Wenzhou Medical University, University Town, Wenzhou, Zhejiang Province, China
| | - Jingya Dong
- Department of Orthodontics, School and Hospital of Stomatology, Wenzhou Medical University, University Town, Wenzhou, Zhejiang Province, China
| | - Junyi Wang
- Department of Biology, College of Science and Technology, Wenzhou-Kean University, Wenzhou, Zhejiang Province, China
| | - Wei Chi
- Department of Biology, College of Science and Technology, Wenzhou-Kean University, Wenzhou, Zhejiang Province, China
| | - Wei Zhou
- Department of Biology, College of Science and Technology, Wenzhou-Kean University, Wenzhou, Zhejiang Province, China
| | - Qiwen Tian
- Department of Biology, College of Science and Technology, Wenzhou-Kean University, Wenzhou, Zhejiang Province, China
| | - Yue Hong
- Department of Orthodontics, School and Hospital of Stomatology, Wenzhou Medical University, University Town, Wenzhou, Zhejiang Province, China
| | - Xuan Zhou
- Department of Biology, College of Science and Technology, Wenzhou-Kean University, Wenzhou, Zhejiang Province, China
| | - Hailv Ye
- Department of Biology, College of Science and Technology, Wenzhou-Kean University, Wenzhou, Zhejiang Province, China
| | - Xuechen Tian
- Zhejiang Bioinformatics International Science and Technology Cooperation Center, Wenzhou, Zhejiang Province, China.,Wenzhou Municipal Key Lab for Applied Biomedical and Biopharmaceutical Informatics, Wenzhou, Zhejiang Province, China
| | - Rongdang Hu
- Department of Orthodontics, School and Hospital of Stomatology, Wenzhou Medical University, University Town, Wenzhou, Zhejiang Province, China
| | - Aloysius Wong
- Department of Biology, College of Science and Technology, Wenzhou-Kean University, Wenzhou, Zhejiang Province, China.,Zhejiang Bioinformatics International Science and Technology Cooperation Center, Wenzhou, Zhejiang Province, China.,Wenzhou Municipal Key Lab for Applied Biomedical and Biopharmaceutical Informatics, Wenzhou, Zhejiang Province, China
| |
Collapse
|
46
|
Baruah R, Ray M, Halami PM. Preventive and Therapeutic aspects of Fermented Foods. J Appl Microbiol 2022; 132:3476-3489. [PMID: 35000256 DOI: 10.1111/jam.15444] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 01/05/2022] [Accepted: 01/05/2022] [Indexed: 11/30/2022]
Abstract
In recent times, the status of some fermented foods which are considered as functional foods that confer health benefits in certain disease conditions has grown rapidly. The health benefits of fermented foods are due to the presence of probiotic microbes and the bioactive compound formed during fermentation. Microbes involved and metabolites produced by them are highly species-specific and contribute to the authenticity of the fermented foods. Several studies pertaining to the effect of fermented foods on various disease conditions have been conducted in recent years using both animal models and clinical trials on humans. This review focuses on the impact of fermented foods on conditions like diabetes, cardiovascular disease (CVD), obesity, gastrointestinal disorder, cancer and neurodegenerative disorders.
Collapse
Affiliation(s)
- Rwivoo Baruah
- Microbiology & Fermentation Technology Department, CSIR-Central Food Technological Research Institute, Mysuru, 570020, India
| | - Mousumi Ray
- Microbiology & Fermentation Technology Department, CSIR-Central Food Technological Research Institute, Mysuru, 570020, India
| | - Prakash M Halami
- Microbiology & Fermentation Technology Department, CSIR-Central Food Technological Research Institute, Mysuru, 570020, India
| |
Collapse
|
47
|
Yang X, Guo Y, Chen C, Shao B, Zhao L, Zhou Q, Liu J, Wang G, Yuan W, Sun Z. Interaction between intestinal microbiota and tumour immunity in the tumour microenvironment. Immunology 2021; 164:476-493. [PMID: 34322877 PMCID: PMC8517597 DOI: 10.1111/imm.13397] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 07/18/2021] [Accepted: 07/20/2021] [Indexed: 11/27/2022] Open
Abstract
In recent years, an increasing number of studies have reported that intestinal microbiota have an important effect on tumour immunity by affecting the tumour microenvironment (TME). The intestinal microbiota are closely associated with various immune cells, such as T lymphocytes, natural killer cells (NK cells) and macrophages. Some bacteria, such as Akkermansia muciniphila (A. muciniphila) and Lactobacillus reuteri (L. reuteri), have been shown to improve the effect of tumour immunity. Furthermore, microbial imbalance, such as the increased abundance of Fusobacterium nucleatum (F. nucleatum) and Helicobacter hepaticus (H. hepaticus), generally causes tumour formation and progression. In addition, some microbiota also play important roles in tumour immunotherapy, especially PD-L1-related therapies. Therefore, what is the relationship between these processes and how do they affect each other? In this review, we summarize the interactions and corresponding mechanisms among the intestinal microbiota, immune system and TME to facilitate the research and development of new targeted drugs and provide new approaches to tumour therapy.
Collapse
Affiliation(s)
- Xiuxiu Yang
- Department of Colorectal SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- School of MedicineZhengzhou UniversityZhengzhouChina
- Academy of Medical SciencesZhengzhou UniversityZhengzhouChina
| | - Yaxin Guo
- Academy of Medical SciencesZhengzhou UniversityZhengzhouChina
- Department of Basic MedicalAcademy of Medical Sciences of Zhengzhou UniversityZhengzhouChina
- Henan Academy of Medical and Pharmaceutical SciencesZhengzhou UniversityZhengzhouChina
| | - Chen Chen
- Academy of Medical SciencesZhengzhou UniversityZhengzhouChina
- School of Life SciencesZhengzhou UniversityZhengzhouChina
| | - Bo Shao
- Department of Colorectal SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Academy of Medical SciencesZhengzhou UniversityZhengzhouChina
| | - Luyang Zhao
- Academy of Medical SciencesZhengzhou UniversityZhengzhouChina
- Department of Basic MedicalAcademy of Medical Sciences of Zhengzhou UniversityZhengzhouChina
- Henan Academy of Medical and Pharmaceutical SciencesZhengzhou UniversityZhengzhouChina
| | - Quanbo Zhou
- Department of Colorectal SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Jinbo Liu
- Department of Colorectal SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Guixian Wang
- Department of Colorectal SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Weitang Yuan
- Department of Colorectal SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Zhenqiang Sun
- Department of Colorectal SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Academy of Medical SciencesZhengzhou UniversityZhengzhouChina
| |
Collapse
|
48
|
Liu C, Zheng J, Ou X, Han Y. Anti-cancer Substances and Safety of Lactic Acid Bacteria in Clinical Treatment. Front Microbiol 2021; 12:722052. [PMID: 34721321 PMCID: PMC8548880 DOI: 10.3389/fmicb.2021.722052] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 09/08/2021] [Indexed: 01/06/2023] Open
Abstract
Lactic acid bacteria (LAB) are a kind of Gram-positive bacteria which can colonize in the biological gastrointestinal tract and play a variety of probiotic roles. LAB have a wide range of applications in industry, animal husbandry, planting, food safety, and medical science fields. Previous studies on LAB have typically concentrated on their effects on improving the digestion and absorption of the gastrointestinal tract, regulating the balance of the microflora, and inhibiting the production and accumulation of toxic substances. The resistance of LAB to cancer is a topic of growing interest and relevance. This paper provided a summary of bio-active substances of LAB when they act against cancer, as well as the safety of LAB in clinical cancer treatment. Moreover, this paper further discussed several possible directions for future research and the potential application of LAB as anti-cancer therapy.
Collapse
Affiliation(s)
- Chaoran Liu
- College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Jiaqi Zheng
- College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Xuan Ou
- College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Yuzhu Han
- College of Animal Science and Technology, Southwest University, Chongqing, China
- Immunology Research Center, Medical Research Institute, Southwest University, Chongqing, China
| |
Collapse
|
49
|
McKee AM, Kirkup BM, Madgwick M, Fowler WJ, Price CA, Dreger SA, Ansorge R, Makin KA, Caim S, Le Gall G, Paveley J, Leclaire C, Dalby M, Alcon-Giner C, Andrusaite A, Feng TY, Di Modica M, Triulzi T, Tagliabue E, Milling SW, Weilbaecher KN, Rutkowski MR, Korcsmáros T, Hall LJ, Robinson SD. Antibiotic-induced disturbances of the gut microbiota result in accelerated breast tumor growth. iScience 2021; 24:103012. [PMID: 34522855 PMCID: PMC8426205 DOI: 10.1016/j.isci.2021.103012] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 04/29/2021] [Accepted: 08/17/2021] [Indexed: 02/08/2023] Open
Abstract
The gut microbiota's function in regulating health has seen it linked to disease progression in several cancers. However, there is limited research detailing its influence in breast cancer (BrCa). This study found that antibiotic-induced perturbation of the gut microbiota significantly increases tumor progression in multiple BrCa mouse models. Metagenomics highlights the common loss of several bacterial species following antibiotic administration. One such bacteria, Faecalibaculum rodentium, rescued this increased tumor growth. Single-cell transcriptomics identified an increased number of cells with a stromal signature in tumors, and subsequent histology revealed an increased abundance of mast cells in the tumor stromal regions. We show that administration of a mast cell stabilizer, cromolyn, rescues increased tumor growth in antibiotic treated animals but has no influence on tumors from control cohorts. These findings highlight that BrCa-microbiota interactions are different from other cancers studied to date and suggest new research avenues for therapy development.
Collapse
Affiliation(s)
- Alastair M. McKee
- Gut Microbes and Health Programme, Quadram Institute Bioscience, Norwich Research Park, Norwich, NR4 7AU, UK
| | - Benjamin M. Kirkup
- Gut Microbes and Health Programme, Quadram Institute Bioscience, Norwich Research Park, Norwich, NR4 7AU, UK
| | - Matthew Madgwick
- Gut Microbes and Health Programme, Quadram Institute Bioscience, Norwich Research Park, Norwich, NR4 7AU, UK
- Earlham Institute, Norwich Research Park, Norwich, NR4 7UZ, UK
| | - Wesley J. Fowler
- Gut Microbes and Health Programme, Quadram Institute Bioscience, Norwich Research Park, Norwich, NR4 7AU, UK
| | - Christopher A. Price
- Gut Microbes and Health Programme, Quadram Institute Bioscience, Norwich Research Park, Norwich, NR4 7AU, UK
| | - Sally A. Dreger
- Gut Microbes and Health Programme, Quadram Institute Bioscience, Norwich Research Park, Norwich, NR4 7AU, UK
| | - Rebecca Ansorge
- Gut Microbes and Health Programme, Quadram Institute Bioscience, Norwich Research Park, Norwich, NR4 7AU, UK
| | - Kate A. Makin
- Faculty of Medicine and Health Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| | - Shabhonam Caim
- Gut Microbes and Health Programme, Quadram Institute Bioscience, Norwich Research Park, Norwich, NR4 7AU, UK
| | - Gwenaelle Le Gall
- Faculty of Medicine and Health Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| | - Jack Paveley
- Gut Microbes and Health Programme, Quadram Institute Bioscience, Norwich Research Park, Norwich, NR4 7AU, UK
| | - Charlotte Leclaire
- Gut Microbes and Health Programme, Quadram Institute Bioscience, Norwich Research Park, Norwich, NR4 7AU, UK
| | - Matthew Dalby
- Gut Microbes and Health Programme, Quadram Institute Bioscience, Norwich Research Park, Norwich, NR4 7AU, UK
| | - Cristina Alcon-Giner
- Gut Microbes and Health Programme, Quadram Institute Bioscience, Norwich Research Park, Norwich, NR4 7AU, UK
| | - Anna Andrusaite
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medicine, Veterinary Medicine and Life Sciences, University of Glasgow, Glasgow G12 8TA, UK
| | - Tzu-Yu Feng
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA
| | - Martina Di Modica
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Instituto Nazionale di Tumori, Milan, 20133, Italy
| | - Tiziana Triulzi
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Instituto Nazionale di Tumori, Milan, 20133, Italy
| | - Elda Tagliabue
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Instituto Nazionale di Tumori, Milan, 20133, Italy
| | - Simon W.F. Milling
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medicine, Veterinary Medicine and Life Sciences, University of Glasgow, Glasgow G12 8TA, UK
| | - Katherine N. Weilbaecher
- Department of Internal Medicine, Division of Molecular Oncology, Washington University in St Louis, St. Louis, MO, 63110, USA
| | - Melanie R. Rutkowski
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA
| | - Tamás Korcsmáros
- Gut Microbes and Health Programme, Quadram Institute Bioscience, Norwich Research Park, Norwich, NR4 7AU, UK
- Earlham Institute, Norwich Research Park, Norwich, NR4 7UZ, UK
| | - Lindsay J. Hall
- Gut Microbes and Health Programme, Quadram Institute Bioscience, Norwich Research Park, Norwich, NR4 7AU, UK
- Faculty of Medicine and Health Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
- Chair of Intestinal Microbiome, School of Life Sciences, ZIEL – Institute for Food & Health, Technical University of Munich, 85354 Freising, Germany
| | - Stephen D. Robinson
- Gut Microbes and Health Programme, Quadram Institute Bioscience, Norwich Research Park, Norwich, NR4 7AU, UK
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| |
Collapse
|
50
|
Administration of Lactobacillus reuteri Combined with Clostridium butyricum Attenuates Cisplatin-Induced Renal Damage by Gut Microbiota Reconstitution, Increasing Butyric Acid Production, and Suppressing Renal Inflammation. Nutrients 2021; 13:nu13082792. [PMID: 34444952 PMCID: PMC8402234 DOI: 10.3390/nu13082792] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 07/28/2021] [Accepted: 08/09/2021] [Indexed: 01/02/2023] Open
Abstract
Cisplatin-induced nephrotoxicity is associated with gut microbiota disturbance. The present study aimed to investigate whether supplementation of Lactobacillus reuteri and Clostridium butyricum (LCs) had a protective effect on cisplatin-induced nephrotoxicity through reconstruction of gut microbiota. Wistar rats were given different treatments: control, cisplatin (Cis), cisplatin + C. butyricum and L. reuteri (Cis+LCs), and C. butyricum and L. reuteri (LCs). We observed that cisplatin-treated rats supplemented with LCs exhibited significantly decreased renal inflammation (KIM-1, F4/80, and MPO), oxidative stress, fibrosis (collagen IV, fibronectin, and a-SMA), apoptosis, concentration of blood endotoxin and indoxyl sulfate, and increased fecal butyric acid production compared with those without supplementation. In addition, LCs improved the cisplatin-induced microbiome dysbiosis by maintaining a healthy gut microbiota structure and diversity; depleting Escherichia-Shigella and the Enterobacteriaceae family; and enriching probiotic Bifidobacterium, Ruminococcaceae, Ruminiclostridium_9, and Oscillibacter. Moreover, the LCs intervention alleviated the cisplatin-induced intestinal epithelial barrier impairment. This study indicated LCs probiotic serves as a mediator of the gut–kidney axis in cisplatin-induced nephrotoxicity to restore the intestinal microbiota composition, thereby suppressing uremic toxin production and enhancing butyrate production. Furthermore, the renoprotective effect of LCs is partially mediated by increasing the anti-inflammatory effects and maintaining the integrity of the intestinal barrier.
Collapse
|