1
|
Sadri M, Shafaghat Z, Roozbehani M, Hoseinzadeh A, Mohammadi F, Arab FL, Minaeian S, Fard SR, Faraji F. Effects of Probiotics on Liver Diseases: Current In Vitro and In Vivo Studies. Probiotics Antimicrob Proteins 2024:10.1007/s12602-024-10431-z. [PMID: 39739162 DOI: 10.1007/s12602-024-10431-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/06/2024] [Indexed: 01/02/2025]
Abstract
Various types of liver or hepatic diseases cause the death of about 2 million people worldwide every year, of which 1 million die from the complications of cirrhosis and another million from hepatocellular carcinoma and viral hepatitis. Currently, the second most common solid organ transplant is the liver, and the current rate represents less than 10% of global transplant requests. Hence, finding new approaches to treat and prevent liver diseases is essential. In liver diseases, the interaction between the liver, gut, and immune system is crucial, and probiotics positively affect the human microbiota. Probiotics are a non-toxic and biosafe alternative to synthetic chemical compounds. Health promotion by lowering cholesterol levels, stimulating host immunity, the natural gut microbiota, and other functions are some of the activities of probiotics, and their metabolites, including bacteriocins, can exert antimicrobial effects against a broad range of pathogenic bacteria. The present review discusses the available data on the results of preclinical and clinical studies on the effects of probiotic administration on different types of liver diseases.
Collapse
Affiliation(s)
- Maryam Sadri
- Department of Immunology, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Shafaghat
- Department of Immunology, Iran University of Medical Sciences, Tehran, Iran
| | - Mona Roozbehani
- Vaccine Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Akram Hoseinzadeh
- Cancer Research Center, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Fatemeh Mohammadi
- Department of Immunology, School of Medicine, Mashhad University of Medicine Sciences, Mashhad, Iran
| | - Fahimeh Lavi Arab
- Department of Immunology, School of Medicine, Mashhad University of Medicine Sciences, Mashhad, Iran
| | - Sara Minaeian
- Antimicrobial Resistance Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medicine Sciences, Tehran, Iran
| | - Soheil Rahmani Fard
- Antimicrobial Resistance Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medicine Sciences, Tehran, Iran
| | - Fatemeh Faraji
- Antimicrobial Resistance Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medicine Sciences, Tehran, Iran.
| |
Collapse
|
2
|
Shamim MZ, Panda J, Mohanty G, Gogoi B, Patowary K, Mishra B, Mohanta YK. The Preventative and Curative Functions of Probiotics. APPLIED BIOTECHNOLOGY AND BIOINFORMATICS 2024:181-215. [DOI: 10.1002/9781119896869.ch8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
3
|
Mukherjee S, Kumar D, Guha D. Insights of probiotics as an alternative medicine for cancer therapy, mechanism, and applications. MEDICINE IN MICROECOLOGY 2024; 22:100111. [DOI: 10.1016/j.medmic.2024.100111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2025] Open
|
4
|
Yao B, Wei W, Zhang H. Efficacy of probiotics or synbiotics supplementation on chemotherapy-induced complications and gut microbiota dysbiosis in gastrointestinal cancer: a systematic review and meta-analysis. Eur J Clin Nutr 2024:10.1038/s41430-024-01542-5. [PMID: 39562823 DOI: 10.1038/s41430-024-01542-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 10/30/2024] [Accepted: 11/05/2024] [Indexed: 11/21/2024]
Abstract
This study aimed to systematically review the clinical efficacy of probiotics or synbiotics supplementation in the treatment of chemotherapy-induced complications and gut microbiota dysbiosis in patients with gastrointestinal cancer. A literature search was performed systematically using PubMed, Embase, Cochrane, Web of Science, Wanfang Data, and CNKI for randomized controlled trials of probiotics or synthetic supplementation on chemotherapy-induced complications and gut microbiota dysbiosis in gastrointestinal cancer up to December 2023. The outcome measures included chemotherapy-related complications and the the incidence of gut microbiotas. Fifteen studies were finally eligible for meta-analysis, involving 1356 patients. Meta-analysis results showed that the the incidence rates of chemotherapy-related complications such as nausea and vomiting [RR = 0.61, 95% CI (0.46,0.82), P = 0.0008] and diarrhea [RR = 0.47, 95% CI (0.32,0.68), P < 0.001] were significantly reduced after probiotic intervention. The number of intestinal flora changed significantly after intervention, such as bifidobacterium [SMD = 1.33, 95% CI (0.52,2.31), P = 0.001], Escherichia coli [SMD = -0.82, 95% CI (-1.26, -0.38), P = 0.0003], and the difference was statistically significant. Probiotics or synbiotics supplementation can reduce chemotherapy-induced complications in patients with gastrointestinal cancer and regulate the number of gut microbiotas to balance the intestinal microecology of the body.
Collapse
Affiliation(s)
- Bei Yao
- First school of clinical medicine, Shandong Traditional Chinese Medicine University, Jinan Shandong, 250014, China
| | - Wei Wei
- Acupuncture rehabilitation Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, GuangDong, 510006, China
| | - Huiping Zhang
- Oncology department, Jinan Hospital of Traditional Chinese Medicine, Jinan Shandong, 250012, China.
| |
Collapse
|
5
|
Thomas CJ, Delgado K, Sawant K, Roy J, Gupta U, Song CS, Poojary R, de Figueiredo P, Song J. Harnessing Bacterial Agents to Modulate the Tumor Microenvironment and Enhance Cancer Immunotherapy. Cancers (Basel) 2024; 16:3810. [PMID: 39594765 PMCID: PMC11593222 DOI: 10.3390/cancers16223810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 10/22/2024] [Accepted: 10/26/2024] [Indexed: 11/28/2024] Open
Abstract
Cancer immunotherapy has revolutionized cancer treatment by leveraging the immune system to attack tumors. However, its effectiveness is often hindered by the immunosuppressive tumor microenvironment (TME), where a complex interplay of tumor, stromal, and immune cells undermines antitumor responses and allows tumors to evade immune detection. This review explores innovative strategies to modify the TME and enhance immunotherapy outcomes, focusing on the therapeutic potential of engineered bacteria. These bacteria exploit the unique characteristics of the TME, such as abnormal vasculature and immune suppression, to selectively accumulate in tumors. Genetically modified bacteria can deliver therapeutic agents, including immune checkpoint inhibitors and cytokines, directly to tumor sites. This review highlights how bacterial therapeutics can target critical immune cells within the TME, such as myeloid-derived suppressor cells and tumor-associated macrophages, thereby promoting antitumor immunity. The combination of bacterial therapies with immune checkpoint inhibitors or adoptive cell transfer presents a promising strategy to counteract immune suppression. Continued research in this area could position bacterial agents as a powerful new modality to reshape the TME and enhance the efficacy of cancer immunotherapy, particularly for tumors resistant to conventional treatments.
Collapse
Affiliation(s)
- Christina James Thomas
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, MREB II, Room 3344, 8447 John Sharp Parkway, Bryan, TX 77807, USA; (C.J.T.); (K.D.)
| | - Kaylee Delgado
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, MREB II, Room 3344, 8447 John Sharp Parkway, Bryan, TX 77807, USA; (C.J.T.); (K.D.)
| | - Kamlesh Sawant
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, MREB II, Room 3344, 8447 John Sharp Parkway, Bryan, TX 77807, USA; (C.J.T.); (K.D.)
| | - Jacob Roy
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, MREB II, Room 3344, 8447 John Sharp Parkway, Bryan, TX 77807, USA; (C.J.T.); (K.D.)
| | - Udit Gupta
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, MREB II, Room 3344, 8447 John Sharp Parkway, Bryan, TX 77807, USA; (C.J.T.); (K.D.)
| | - Carly Shaw Song
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, MREB II, Room 3344, 8447 John Sharp Parkway, Bryan, TX 77807, USA; (C.J.T.); (K.D.)
| | - Rayansh Poojary
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, MREB II, Room 3344, 8447 John Sharp Parkway, Bryan, TX 77807, USA; (C.J.T.); (K.D.)
| | - Paul de Figueiredo
- Department of Molecular Microbiology and Immunology, The University of Missouri School of Medicine, Columbia, MO 65212, USA
| | - Jianxun Song
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, MREB II, Room 3344, 8447 John Sharp Parkway, Bryan, TX 77807, USA; (C.J.T.); (K.D.)
| |
Collapse
|
6
|
Liu J, He C, Tan W, Zheng JH. Path to bacteriotherapy: From bacterial engineering to therapeutic perspectives. Life Sci 2024; 352:122897. [PMID: 38971366 DOI: 10.1016/j.lfs.2024.122897] [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: 01/11/2024] [Revised: 06/30/2024] [Accepted: 07/03/2024] [Indexed: 07/08/2024]
Abstract
The major reason for the failure of conventional therapies is the heterogeneity and complexity of tumor microenvironments (TMEs). Many malignant tumors reprogram their surface antigens to evade the immune surveillance, leading to reduced antigen-presenting cells and hindered T-cell activation. Bacteria-mediated cancer immunotherapy has been extensively investigated in recent years. Scientists have ingeniously modified bacteria using synthetic biology and nanotechnology to enhance their biosafety with high tumor specificity, resulting in robust anticancer immune responses. To enhance the antitumor efficacy, therapeutic proteins, cytokines, nanoparticles, and chemotherapeutic drugs have been efficiently delivered using engineered bacteria. This review provides a comprehensive understanding of oncolytic bacterial therapies, covering bacterial design and the intricate interactions within TMEs. Additionally, it offers an in-depth comparison of the current techniques used for bacterial modification, both internally and externally, to maximize their therapeutic effectiveness. Finally, we outlined the challenges and opportunities ahead in the clinical application of oncolytic bacterial therapies.
Collapse
Affiliation(s)
- Jinling Liu
- The Affiliated Xiangtan Central Hospital of Hunan University, School of Biomedical Sciences, Hunan University, Changsha 410082, China; College of Biology, Hunan University, Changsha 410082, China
| | - Chongsheng He
- College of Biology, Hunan University, Changsha 410082, China
| | - Wenzhi Tan
- School of Food Science and Bioengineering, Changsha University of Science & Technology, Changsha, Hunan 410114, China.
| | - Jin Hai Zheng
- The Affiliated Xiangtan Central Hospital of Hunan University, School of Biomedical Sciences, Hunan University, Changsha 410082, China.
| |
Collapse
|
7
|
Zhang N, Ye S, Wang X, Wang K, Zhong F, Yao F, Liu J, Huang B, Xu F, Wang X. Hepatic Symbiotic Bacterium L. reuteri FLRE5K1 Inhibits the Development and Progression of Hepatocellular Carcinoma via Activating the IFN-γ/CXCL10/CXCR3 Pathway. Probiotics Antimicrob Proteins 2024; 16:1158-1171. [PMID: 37289406 DOI: 10.1007/s12602-023-10098-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/25/2023] [Indexed: 06/09/2023]
Abstract
Symbiotic bacteria participate in the formation of the structure and function of the tissues and organs in which they live, and play an essential role in maintaining the balance between health and disease. Lactobacillus reuteri FLRE5K1 was isolated from the liver of healthy mice and proved to be a probiotic with anti-melanoma activity in previous studies. The relationship between hepatic symbiotic probiotics and hepatocellular carcinoma (HCC) has not been reported yet. In the present study, L. reuteri FLRE5K1 was initially confirmed to successfully enter the liver after being administered by gavage, and the efficacy of probiotic feeding on HCC and its potential mechanism of inhibiting tumor progression were investigated by an orthotopic liver cancer model established. The results showed that L. reuteri FLRE5K1 significantly reduced the tumor formation rate and inhibited tumor growth in mice. From the perspective of mechanism, activation of the IFN-γ/CXCL10/CXCR3 pathway, as well as its positive feedback on the secretion of IFN-γ, induced the polarization of Th0 cell to Th1 cells and inhibited the differentiation of Tregs, which played a key role in the inhibitory effect of L. reuteri FLRE5K1 on the development and progression of HCC.
Collapse
Affiliation(s)
- Nan Zhang
- Jiangxi Province Key Laboratory of Laboratory Medicine, Department of Clinical Laboratory, the Second Affiliated Hospital of Nanchang University, Nanchang, 330047, Jiangxi, China
| | - Shuiwen Ye
- Department of Blood Transfusion, the Second Affiliated Hospital of Nanchang University, Nanchang, 330047, Jiangxi, China
| | - Xinlu Wang
- Jiangxi Province Key Laboratory of Laboratory Medicine, Department of Clinical Laboratory, the Second Affiliated Hospital of Nanchang University, Nanchang, 330047, Jiangxi, China
| | - Kang Wang
- Department of Traditional Chinese Medicine, the Second Affiliated Hospital of Nanchang University, Nanchang, 330047, Jiangxi, China
| | - Fangmin Zhong
- Jiangxi Province Key Laboratory of Laboratory Medicine, Department of Clinical Laboratory, the Second Affiliated Hospital of Nanchang University, Nanchang, 330047, Jiangxi, China
| | - Fangyi Yao
- Jiangxi Province Key Laboratory of Laboratory Medicine, Department of Clinical Laboratory, the Second Affiliated Hospital of Nanchang University, Nanchang, 330047, Jiangxi, China
| | - Jing Liu
- Jiangxi Province Key Laboratory of Laboratory Medicine, Department of Clinical Laboratory, the Second Affiliated Hospital of Nanchang University, Nanchang, 330047, Jiangxi, China
| | - Bo Huang
- Jiangxi Province Key Laboratory of Laboratory Medicine, Department of Clinical Laboratory, the Second Affiliated Hospital of Nanchang University, Nanchang, 330047, Jiangxi, China
| | - Feng Xu
- Jiangxi-Oai Joint Research Institute, Nanchang University, Nanchang, 330047, Jiangxi, China.
| | - Xiaozhong Wang
- Jiangxi Province Key Laboratory of Laboratory Medicine, Department of Clinical Laboratory, the Second Affiliated Hospital of Nanchang University, Nanchang, 330047, Jiangxi, China.
| |
Collapse
|
8
|
Vinothkanna A, Shi‐Liang X, Karthick Rajan D, Prathiviraj R, Sekar S, Zhang S, Wang B, Liu Z, Jia A. Feasible mechanisms and therapeutic potential of food probiotics to mitigate diabetes‐associated cancers: A comprehensive review and in silico validation. FOOD FRONTIERS 2024; 5:1476-1511. [DOI: 10.1002/fft2.406] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025] Open
Abstract
AbstractPeople with diabetes mellitus (DM) and hyperglycemia are linked with cancer risk. Diabetes and cancer have been corroborated by high morbidity and mortality rates. Studies revealed that elevated levels of insulin secretions trigger insulin‐like growth factor 1 (IGF‐1) production. Moreover, IGF‐1 is a key regulator involved in promoting cancer cell progression and is linked with DM. Cancer drug resistance and ototoxic effects can adversely affect the health and lifespan of an individual. However, naturally derived bioactive compounds are gaining attention for their nontoxic properties and specific behavior. Likewise, probiotics have also been regarded as safe and successful alternatives to treat DM‐linked cancers. The present review aims to highlight the therapeutic potential and feasible functions of probiotics to mitigate or inhibit DM‐associated cancers. Meanwhile, the intracellular signaling cascades involved in promoting DM‐linked cancer are enumerated for future prospective research. However, metabolomics interactions and protein–protein interactions are to be discussed for deeper insights into affirmative principles in diabetic‐linked cancers. Drug discovery and innovative preclinical evaluation need further adjuvant and immune‐enhancement therapies. Furthermore, the results of the in silico assessment could provide scientific excellence of IGF‐1 in diabetes and cancer. Overall, this review summarizes the mechanistic insights and therapeutic targets for diabetes‐associated cancer.
Collapse
Affiliation(s)
- Annadurai Vinothkanna
- Hainan Affiliated Hospital of Hainan Medical University Hainan General Hospital Haikou China
- School of Life and Health Sciences Hainan University Haikou China
| | - Xiang Shi‐Liang
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences Hainan University Haikou China
| | - Durairaj Karthick Rajan
- Department of Cell Biology, School of Life Sciences Central South University Changsha Hunan China
| | | | - Soundarapandian Sekar
- Department of Biotechnology Bharathidasan University Tiruchirappalli Tamil Nadu India
| | - Shubing Zhang
- Department of Cell Biology, School of Life Sciences Central South University Changsha Hunan China
| | - Bo Wang
- Hainan Affiliated Hospital of Hainan Medical University Hainan General Hospital Haikou China
| | - Zhu Liu
- School of Life and Health Sciences Hainan University Haikou China
| | - Ai‐Qun Jia
- Hainan Affiliated Hospital of Hainan Medical University Hainan General Hospital Haikou China
| |
Collapse
|
9
|
Wang W, Fan J, Zhang C, Huang Y, Chen Y, Fu S, Wu J. Targeted modulation of gut and intra-tumor microbiota to improve the quality of immune checkpoint inhibitor responses. Microbiol Res 2024; 282:127668. [PMID: 38430889 DOI: 10.1016/j.micres.2024.127668] [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: 08/08/2023] [Revised: 01/22/2024] [Accepted: 02/25/2024] [Indexed: 03/05/2024]
Abstract
Immune checkpoint inhibitor (ICI) therapies, such as those blocking the interaction of PD-1 with its ligands, can restore the immune-killing function of T cells. However, ICI therapy is clinically beneficial in only a small number of patients, and it is difficult to predict post-treatment outcomes, thereby limiting its widespread clinical use. Research suggests that gut microbiota can regulate the host immune system and affect cancer progression and treatment. Moreover, the effectiveness of immunotherapy is related to the composition of the patient's gut microbiota; different gut microbial strains can either activate or inhibit the immune response. However, the importance of the microbial composition within the tumor has not been explored until recently. This study describes recent advances in the crosstalk between microbes in tumors and gut microbiota, which can modulate the tumor microbiome by directly translocating into the tumor and altering the tumor microenvironment. This study focused on the potential manipulation of the tumor and gut microbiota using fecal microbiota transplantation (FMT), probiotics, antimicrobials, prebiotics, and postbiotics to enrich immune-boosting bacteria while decreasing unfavorable bacteria to proactively improve the efficacy of ICI treatments. In addition, the use of genetic technologies and nanomaterials to modify microorganisms can largely optimize tumor immunotherapy and advance personalized and precise cancer treatment.
Collapse
Affiliation(s)
- WeiZhou Wang
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - JunYing Fan
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Chi Zhang
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Yuan Huang
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Yue Chen
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, Sichuan 646000, China; Department of Nuclear Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - ShaoZhi Fu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China; Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, Sichuan 646000, China.
| | - JingBo Wu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China; Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, Sichuan 646000, China; Academician (Expert) Workstation of Sichuan Province, Luzhou, Sichuan 646000, China.
| |
Collapse
|
10
|
Mirzadeh MA, Eslami M, Ghanbari A, Zarbakhsh S, Yosefi S, Pakdel A. Coadministration of doxorubicin with vitamin D3, Lactobacillus acidophilus, and Lactobacillus casei in the 4T1 mouse model of breast cancer: anticancer and enteroprotective effects. Med Oncol 2024; 41:111. [PMID: 38592504 DOI: 10.1007/s12032-024-02346-0] [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: 01/21/2024] [Accepted: 02/27/2024] [Indexed: 04/10/2024]
Abstract
The use of doxorubicin (Dox) in the treatment of breast cancer negatively affects the intestines and other tissues. Many studies have proven that probiotics and vitamin D3 have antitumor and intestinal tissue-protecting properties. To achieve effectiveness and minimize side effects, the current study aims to administer Dox together with probiotics (Lactobacillus acidophilus and Lactobacillus casei) and vitamin D3. Forty-two female BALB/c inbred mice were divided into six groups: Group 1 (Control), Group 2 (Dox), Group 3 (Dox and probiotics), Group 4 (Dox and vitamin D3), Group 5 (Dox, probiotics, and vitamin D3), and Group 6 (probiotics and vitamin D3). The 4T1 mouse carcinoma cell line was injected into the mammary fat pad of each mouse. Gene expression was examined using quantitative real-time PCR. The treated groups (except group 6) showed significantly reduced tumor volume and weight compared to the control group (P < 0.05, P < 0.01). Probiotics/vitamin D3 with Dox reduced chemotherapy toxicity and a combination of supplements had a significant protective effect against Dox (P < 0.05, 0.01, 0.001). The treated groups (except 6) had significantly higher expression of Bax/Caspase 3 genes and lower expression of Bcl-2 genes than the control group (P < 0.05, 0.01). Coadministration of Dox with probiotics and vitamin D3 showed promising results in reducing tumor size, protecting intestinal tissue and influencing gene expression, suggesting a strategy to enhance the effectiveness of breast cancer treatment while reducing side effects.
Collapse
Affiliation(s)
- Mohammad Ali Mirzadeh
- Department of Biochemistry , Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Majid Eslami
- Department of Bacteriology and Virology, Semnan University of Medical Sciences, Semnan, Iran
| | - Ali Ghanbari
- Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran
| | - Sam Zarbakhsh
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Sedighe Yosefi
- Department of Biochemistry , Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Abbas Pakdel
- Department of Biochemistry , Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran.
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran.
| |
Collapse
|
11
|
de Castilhos J, Tillmanns K, Blessing J, Laraño A, Borisov V, Stein-Thoeringer CK. Microbiome and pancreatic cancer: time to think about chemotherapy. Gut Microbes 2024; 16:2374596. [PMID: 39024520 PMCID: PMC11259062 DOI: 10.1080/19490976.2024.2374596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 06/26/2024] [Indexed: 07/20/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive cancer characterized by late diagnosis, rapid progression, and a high mortality rate. Its complex biology, characterized by a dense, stromal tumor environment with an immunosuppressive milieu, contributes to resistance against standard treatments like chemotherapy and radiation. This comprehensive review explores the dynamic role of the microbiome in modulating chemotherapy efficacy and outcomes in PDAC. It delves into the microbiome's impact on drug metabolism and resistance, and the interaction between microbial elements, drugs, and human biology. We also highlight the significance of specific bacterial species and microbial enzymes in influencing drug action and the immune response in the tumor microenvironment. Cutting-edge methodologies, including artificial intelligence, low-biomass microbiome analysis and patient-derived organoid models, are discussed, offering insights into the nuanced interactions between microbes and cancer cells. The potential of microbiome-based interventions as adjuncts to conventional PDAC treatments are discussed, paving the way for personalized therapy approaches. This review synthesizes recent research to provide an in-depth understanding of how the microbiome affects chemotherapy efficacy. It focuses on elucidating key mechanisms and identifying existing knowledge gaps. Addressing these gaps is crucial for enhancing personalized medicine and refining cancer treatment strategies, ultimately improving patient outcomes.
Collapse
Affiliation(s)
- Juliana de Castilhos
- Translational Microbiome Research, Internal Medicine I and M3 Research Center, University Hospital Tuebingen, Tübingen, Germany
- Cluster of Excellence “Controlling Microbes to Fight Infections” (CMFI), University of Tuebingen, Tübingen, Germany
| | - Katharina Tillmanns
- Translational Microbiome Research, Internal Medicine I and M3 Research Center, University Hospital Tuebingen, Tübingen, Germany
- Cluster of Excellence “Controlling Microbes to Fight Infections” (CMFI), University of Tuebingen, Tübingen, Germany
| | - Jana Blessing
- Translational Microbiome Research, Internal Medicine I and M3 Research Center, University Hospital Tuebingen, Tübingen, Germany
- Cluster of Excellence “Controlling Microbes to Fight Infections” (CMFI), University of Tuebingen, Tübingen, Germany
| | - Arnelyn Laraño
- Translational Microbiome Research, Internal Medicine I and M3 Research Center, University Hospital Tuebingen, Tübingen, Germany
- Cluster of Excellence “Controlling Microbes to Fight Infections” (CMFI), University of Tuebingen, Tübingen, Germany
| | - Vadim Borisov
- Translational Microbiome Research, Internal Medicine I and M3 Research Center, University Hospital Tuebingen, Tübingen, Germany
- Cluster of Excellence “Controlling Microbes to Fight Infections” (CMFI), University of Tuebingen, Tübingen, Germany
| | - Christoph K. Stein-Thoeringer
- Translational Microbiome Research, Internal Medicine I and M3 Research Center, University Hospital Tuebingen, Tübingen, Germany
- Cluster of Excellence “Controlling Microbes to Fight Infections” (CMFI), University of Tuebingen, Tübingen, Germany
| |
Collapse
|
12
|
Zhao X, Zhao J, Li D, Yang H, Chen C, Qin M, Wen Z, He Z, Xu L. Akkermansia muciniphila: A potential target and pending issues for oncotherapy. Pharmacol Res 2023; 196:106916. [PMID: 37690533 DOI: 10.1016/j.phrs.2023.106916] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 09/02/2023] [Accepted: 09/07/2023] [Indexed: 09/12/2023]
Abstract
In the wake of the development of metagenomic, metabolomic, and metatranscriptomic approaches, the intricate interactions between the host and various microbes are now being progressively understood. Numerous studies have demonstrated evident changes in gut microbiota during the process of a variety of diseases, such as diabetes, obesity, aging, and cancers. Notably, gut microbiota is viewed as a potential source of novel therapeutics. Currently, Next-generation probiotics (NGPs) are gaining popularity as therapeutic agents that alter the gut microbiota and affect cancer development. Akkermansia muciniphila (A. muciniphila), a representative commensal bacterium, has received substantial attention over the past decade as a promising NGP. The components and metabolites of A. muciniphila can directly or indirectly affect tumorigenesis, in particular through its effects on antitumor immunosurveillance, including the stimulation of pattern recognition receptors (PRRs), which also leads to better outcomes in a variety of situations, including the prevention and curation of cancers. In this article, we systematically summarize the role of A. muciniphila in tumorigenesis (involving gastrointestinal and non-gastrointestinal cancers) and in tumor therapy. In particular, we carefully discuss some critical scientific issues that need to be solved for the future using A. muciniphila as a representative beneficial bacterium in tumor treatment, which might provide bright clues and assistance for the application of drugs targeting A. muciniphila in clinical oncotherapy.
Collapse
Affiliation(s)
- Xu Zhao
- Guizhou University Medical College, Guiyang 550025, Guizhou Province, China; Special Key Laboratory of Gene Detection &Therapy of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou 563000, China; Department of Immunology, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Juanjuan Zhao
- Special Key Laboratory of Gene Detection &Therapy of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou 563000, China; Department of Immunology, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Dongmei Li
- Special Key Laboratory of Gene Detection &Therapy of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou 563000, China; Department of Immunology, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Han Yang
- Special Key Laboratory of Gene Detection &Therapy of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou 563000, China; Department of Immunology, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Chao Chen
- Guizhou University Medical College, Guiyang 550025, Guizhou Province, China; Special Key Laboratory of Gene Detection &Therapy of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Ming Qin
- Special Key Laboratory of Gene Detection &Therapy of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Zhenke Wen
- Institutes of Biology and Medical Sciences, Soochow Univeristy, Jiangsu 215000, China
| | - Zhixu He
- Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical University, Zunyi, Guizhou 563000, China.
| | - Lin Xu
- Guizhou University Medical College, Guiyang 550025, Guizhou Province, China; Special Key Laboratory of Gene Detection &Therapy of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou 563000, China; Department of Immunology, Zunyi Medical University, Zunyi, Guizhou 563000, China; Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical University, Zunyi, Guizhou 563000, China.
| |
Collapse
|
13
|
Choi E, Murray B, Choi S. Biofilm and Cancer: Interactions and Future Directions for Cancer Therapy. Int J Mol Sci 2023; 24:12836. [PMID: 37629016 PMCID: PMC10454087 DOI: 10.3390/ijms241612836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/13/2023] [Accepted: 08/14/2023] [Indexed: 08/27/2023] Open
Abstract
There is a growing body of evidence supporting the significant role of bacterial biofilms in the pathogenesis of various human diseases, including cancer. Biofilms are polymicrobial communities enclosed within an extracellular matrix composed of polysaccharides, proteins, extracellular DNA, and lipids. This complex matrix provides protection against antibiotics and host immune responses, enabling the microorganisms to establish persistent infections. Moreover, biofilms induce anti-inflammatory responses and metabolic changes in the host, further facilitating their survival. Many of these changes are comparable to those observed in cancer cells. This review will cover recent research on the role of bacterial biofilms in carcinogenesis, especially in colorectal (CRC) and gastric cancers, emphasizing the shared physical and chemical characteristics of biofilms and cancer. This review will also discuss the interactions between bacteria and the tumor microenvironment, which can facilitate oncogene expression and cancer progression. This information will provide insight into developing new therapies to identify and treat biofilm-associated cancers, such as utilizing bacteria as delivery vectors, using bacteria to upregulate immune function, or more selectively targeting biofilms and cancer for their shared traits.
Collapse
Affiliation(s)
- Euna Choi
- Department of Biology, Union University, Jackson, TN 38305, USA; (E.C.); (B.M.)
| | - Ben Murray
- Department of Biology, Union University, Jackson, TN 38305, USA; (E.C.); (B.M.)
| | - Sunga Choi
- Department of Bioinformatics and Biosystems, Seongnam Campus of Korea Polytechnics, Seongnam-si 13122, Republic of Korea
| |
Collapse
|
14
|
Zhu Z, Peng L, Zhou H, Gu H, Tang Y, Zhou Z, Xiang L, Wang Y. Low-fat dairy consumption and the risk of lung cancer: A large prospective cohort study. Cancer Med 2023; 12:16558-16569. [PMID: 37329182 PMCID: PMC10469841 DOI: 10.1002/cam4.6249] [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: 03/04/2023] [Revised: 05/22/2023] [Accepted: 06/02/2023] [Indexed: 06/18/2023] Open
Abstract
BACKGROUND Despite the possible contribution of dairy products to the development or prevention of cancers, there is a lack of epidemiological evidence linking low-fat dairy consumption to the risk of developing lung cancer. This research was conducted to fill this knowledge gap. METHODS The data for this research were collected from the Prostate, Lung, Colorectal, and Ovarian (PLCO) Cancer Screening Trial. The Cox proportional risk model was employed to evaluate the link between low-fat dairy consumption and the risk of developing lung cancer. Hazard ratios (HRs) and 95% confidence intervals (CIs) were measured in both unadjusted and adjusted models. A series of predefined subgroup analyses were performed to identify potential effect modifiers, and several sensitivity analyses were conducted to assess the stability of the findings. RESULTS The study included data from 98,459 individuals. During a total of 869,807.9 follow-up person-years, 1642 cases of lung cancer were observed, with an incidence of 0.189 cases for every 100 person-years. In the fully adjusted model, participants in the highest quartile of low-fat dairy consumption had a significantly decreased risk of lung cancer compared to the ones in the lowest quartile (HRquartile 4 vs. 1 : 0.769, 95% CI: 0.664, 0.891, ptrend = 0.005). The restricted cubic spline plot revealed an inverse nonlinear dose-response relationship between low-fat dairy consumption and lung cancer risk (pnonlinearity = 0.008). Subgroup analyses demonstrated that the inverse association was stronger among participants with higher daily caloric intake (pinteraction = 0.031). Various sensitivity analyses produced consistent results. CONCLUSION Consuming more low-fat dairy products is significantly linked to a reduced risk of developing lung cancer, indicating that an appropriate increase in the use of low-fat dairy products may help prevent lung cancer.
Collapse
Affiliation(s)
- Zhiyong Zhu
- Department of Gastrointestinal SurgeryThe Second Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Linglong Peng
- Department of Gastrointestinal SurgeryThe Second Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - He Zhou
- Laboratory of Cancer Biology Department of OncologyUniversity of OxfordOxfordUK
- The Second Department of Gastrointestinal SurgeryAffiliated Hospital of North Sichuan Medical CollegeNanchongChina
| | - Haitao Gu
- Department of Gastrointestinal SurgeryThe Second Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Yunhao Tang
- Department of Gastrointestinal SurgeryThe Second Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Zhihang Zhou
- Department of GastroenterologyThe Second Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Ling Xiang
- Department of Clinical NutritionThe Second Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Yaxu Wang
- Department of Gastrointestinal SurgeryThe Second Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| |
Collapse
|
15
|
Henderson EA, Lukomski S, Boone BA. Emerging applications of cancer bacteriotherapy towards treatment of pancreatic cancer. Front Oncol 2023; 13:1217095. [PMID: 37588093 PMCID: PMC10425600 DOI: 10.3389/fonc.2023.1217095] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 06/26/2023] [Indexed: 08/18/2023] Open
Abstract
Pancreatic cancer is a highly aggressive form of cancer with a five-year survival rate of only ten percent. Pancreatic ductal adenocarcinoma (PDAC) accounts for ninety percent of those cases. PDAC is associated with a dense stroma that confers resistance to current treatment modalities. Increasing resistance to cancer treatments poses a challenge and a need for alternative therapies. Bacterial mediated cancer therapies were proposed in the late 1800s by Dr. William Coley when he injected osteosarcoma patients with live streptococci or a fabrication of heat-killed Streptococcus pyogenes and Serratia marcescens known as Coley's toxin. Since then, several bacteria have gained recognition for possible roles in potentiating treatment response, enhancing anti-tumor immunity, and alleviating adverse effects to standard treatment options. This review highlights key bacterial mechanisms and structures that promote anti-tumor immunity, challenges and risks associated with bacterial mediated cancer therapies, and applications and opportunities for use in PDAC management.
Collapse
Affiliation(s)
- Emily A. Henderson
- Department of Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, WV, United States
| | - Slawomir Lukomski
- Department of Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, WV, United States
- West Virginia Cancer Institute, West Virginia University, Morgantown, WV, United States
| | - Brian A. Boone
- Department of Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, WV, United States
- West Virginia Cancer Institute, West Virginia University, Morgantown, WV, United States
- Department of Surgery, West Virginia University, Morgantown, WV, United States
| |
Collapse
|
16
|
Pourmollaei S, Farshbaf-Khalili A, Barzegari A, Bastani S, Babaie S, Fattahi A, Shahnazi M. Anticancer Effect of Enterococcus faecium, Isolated from Vaginal Fluid, on Ovarian Cancer Cells. IRANIAN BIOMEDICAL JOURNAL 2023; 27:205-13. [PMID: 37525437 PMCID: PMC10507285 DOI: 10.61186/ibj.3846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 05/31/2023] [Indexed: 12/17/2023]
Abstract
Background Given the association between cervicovaginal microbiota and OVC, we investigated the effect of Enterococcus faecium conditioned medium (CM) on OVC (Caov-4) cells. Methods CM was obtained from the bacterium E. faecium isolated from the vagina of healthy women. The Caov-4 cells were treated with varying concentrations of CM that comprised co-cultured bacteria with 0.2, 0.5, 1, 1.5, and 2 OD for 12, 24, and 48 h. The apoptosis and growth of cancer cells were evaluated by 4′,6-diamidino-2-phenylindole (DAPI) staining, flow cytometry, and DNA laddering assay. Moreover, the expression of PTEN, BAX, BCL2, and AKT1 genes were analyzed using real-time PCR. Results The CM at a concentration of 0.5 OD from the cultured bacteria and incubation time of 48 h showed the highest negative effect on the viability of cancer cells. The CM treatment increased DNA fragmentation and also induced apoptosis in Caov-4 cells. Interestingly, CM could decrease the expression of proapoptotic genes were less, while antiapoptotic genes were more than fluorouracil in the presence of CM. Conclusion CM of human-derived E. faecium could have an anticancer effect on OVC cells in a concentration- and time-dependent manner. This study demonstrated that E. faecium secretes anticancer substances into the CM, which could directly affect the viability and apoptosis of cancer cells.
Collapse
Affiliation(s)
- Soraya Pourmollaei
- Department of Midwifery, Faculty of Nursing and Midwifery, Tabriz University of Medical Sciences, Tabriz, Iran
- Students' Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Azizeh Farshbaf-Khalili
- Aging Research Institute, Physical Medicine, and Rehabilitation Research Centre, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Abolfazl Barzegari
- Research Center of Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sepideh Bastani
- Research Center of Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Soraya Babaie
- Aging Research Institute, Physical Medicine, and Rehabilitation Research Centre, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Fattahi
- Womenʼs Reproductive Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahnaz Shahnazi
- Department of Midwifery, Faculty of Nursing and Midwifery, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
17
|
Zhao T, Wang H, Liu Z, Liu Y, Li B, Huang X. Recent Perspective of Lactobacillus in Reducing Oxidative Stress to Prevent Disease. Antioxidants (Basel) 2023; 12:antiox12030769. [PMID: 36979017 PMCID: PMC10044891 DOI: 10.3390/antiox12030769] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/19/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
During oxidative stress, an important factor in the development of many diseases, cellular oxidative and antioxidant activities are imbalanced due to various internal and external factors such as inflammation or diet. The administration of probiotic Lactobacillus strains has been shown to confer a range of antibacterial, anti-inflammatory, antioxidant, and immunomodulatory effects in the host. This review focuses on the potential role of oxidative stress in inflammatory bowel diseases (IBD), cancer, and liver-related diseases in the context of preventive and therapeutic effects associated with Lactobacillus. This article reviews studies in cell lines and animal models as well as some clinical population reports that suggest that Lactobacillus could alleviate basic symptoms and related abnormal indicators of IBD, cancers, and liver damage, and covers evidence supporting a role for the Nrf2, NF-κB, and MAPK signaling pathways in the effects of Lactobacillus in alleviating inflammation, oxidative stress, aberrant cell proliferation, and apoptosis. This review also discusses the unmet needs and future directions in probiotic Lactobacillus research including more extensive mechanistic analyses and more clinical trials for Lactobacillus-based treatments.
Collapse
Affiliation(s)
- Tingting Zhao
- School of Public Health, Lanzhou University, Lanzhou 730033, China
- Institute of Animal Husbandry and Veterinary, Tibet Academy of Agricultural and Animal Husbandry Sciences, Lhasa 850000, China
| | - Haoran Wang
- School of Public Health, Lanzhou University, Lanzhou 730033, China
- Institute of Animal Husbandry and Veterinary, Tibet Academy of Agricultural and Animal Husbandry Sciences, Lhasa 850000, China
| | - Zhenjiang Liu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Yang Liu
- Institute of Animal Husbandry and Veterinary, Tibet Academy of Agricultural and Animal Husbandry Sciences, Lhasa 850000, China
| | - Bin Li
- Institute of Animal Husbandry and Veterinary, Tibet Academy of Agricultural and Animal Husbandry Sciences, Lhasa 850000, China
| | - Xiaodan Huang
- School of Public Health, Lanzhou University, Lanzhou 730033, China
- Institute of Animal Husbandry and Veterinary, Tibet Academy of Agricultural and Animal Husbandry Sciences, Lhasa 850000, China
| |
Collapse
|
18
|
Noor S, Ali S, Riaz S, Sardar I, Farooq MA, Sajjad A. Chemopreventive role of probiotics against cancer: a comprehensive mechanistic review. Mol Biol Rep 2023; 50:799-814. [PMID: 36324027 DOI: 10.1007/s11033-022-08023-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 10/10/2022] [Indexed: 11/06/2022]
Abstract
Probiotics use different mechanisms such as intestinal barrier improvement, bacterial translocation and maintaining gut microbiota homeostasis to treat cancer. Probiotics' ability to induce apoptosis against tumor cells makes them more effective to treat cancer. Moreover, probiotics stimulate immune function through an immunomodulation mechanism that induces an anti-tumor effect. There are different strains of probiotics, but the most important ones are lactic acid bacteria (LAB) having antagonistic and anti-mutagenic activities. Live and dead probiotics have anti-inflammatory, anti-proliferative, anti-oxidant and anti-metastatic properties which are useful to fight against different diseases, especially cancer. The main focus of this article is to review the anti-cancerous properties of probiotics and their role in the reduction of different types of cancer. However, further investigations are in progress to improve the efficiency of probiotics in cancer treatment.
Collapse
Affiliation(s)
- Shehzeen Noor
- Applied Entomology and Medical Toxicology and Laboratory, Department of Zoology, Government College University, Lahore, Pakistan
| | - Shaukat Ali
- Applied Entomology and Medical Toxicology and Laboratory, Department of Zoology, Government College University, Lahore, Pakistan.
| | - Shumaila Riaz
- Applied Entomology and Medical Toxicology and Laboratory, Department of Zoology, Government College University, Lahore, Pakistan
| | - Iqra Sardar
- Applied Entomology and Medical Toxicology and Laboratory, Department of Zoology, Government College University, Lahore, Pakistan
| | - Muhammad Adeel Farooq
- Applied Entomology and Medical Toxicology and Laboratory, Department of Zoology, Government College University, Lahore, Pakistan
| | - Ayesha Sajjad
- Applied Entomology and Medical Toxicology and Laboratory, Department of Zoology, Government College University, Lahore, Pakistan
| |
Collapse
|
19
|
Schemczssen-Graeff Z, Pileggi M. Probiotics and live biotherapeutic products aiming at cancer mitigation and patient recover. Front Genet 2022; 13:921972. [PMID: 36017495 PMCID: PMC9395637 DOI: 10.3389/fgene.2022.921972] [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: 04/17/2022] [Accepted: 07/13/2022] [Indexed: 11/13/2022] Open
Abstract
Molecular biology techniques allowed access to non-culturable microorganisms, while studies using analytical chemistry, as Liquid Chromatography and Tandem Mass Spectrometry, showed the existence of a complex communication system among bacteria, signaled by quorum sensing molecules. These approaches also allowed the understanding of dysbiosis, in which imbalances in the microbiome diversity, caused by antibiotics, environmental toxins and processed foods, lead to the constitution of different diseases, as cancer. Colorectal cancer, for example, can originate by a dysbiosis configuration, which leads to biofilm formation, production of toxic metabolites, DNA damage in intestinal epithelial cells through the secretion of genotoxins, and epigenetic regulation of oncogenes. However, probiotic strains can also act in epigenetic processes, and so be use for recovering important intestinal functions and controlling dysbiosis and cancer mitigation through the metabolism of drugs used in chemotherapy, controlling the proliferation of cancer cells, improving the immune response of the host, regulation of cell differentiation and apoptosis, among others. There are still gaps in studies on the effectiveness of the use of probiotics, therefore omics and analytical chemistry are important approaches to understand the role of bacterial communication, formation of biofilms, and the effects of probiotics and microbiome on chemotherapy. The use of probiotics, prebiotics, synbiotics, and metabiotics should be considered as a complement to other more invasive and hazard therapies, such chemotherapy, surgery, and radiotherapy. The study of potential bacteria for cancer treatment, as the next-generation probiotics and Live Biotherapeutic Products, can have a controlling action in epigenetic processes, enabling the use of these bacteria for the mitigation of specific diseases through changes in the regulation of genes of microbiome and host. Thus, it is possible that a path of medicine in the times to come will be more patient-specific treatments, depending on the environmental, genetic, epigenetic and microbiome characteristics of the host.
Collapse
Affiliation(s)
- Zelinda Schemczssen-Graeff
- Comparative Immunology Laboratory, Department of Microbiology, Parasitology and Pathology, Federal University of Paraná, Curitiba, Brazil
| | - Marcos Pileggi
- Environmental Microbiology Laboratory, Structural and Molecular Biology and Genetics Department, Life Sciences and Health Institute, Ponta Grossa State University, Ponta Grossa, Brazil
- *Correspondence: Marcos Pileggi,
| |
Collapse
|
20
|
Wan L, Wu C, Wu Q, Luo S, Liu J, Xie X. Impact of probiotics use on clinical outcomes of immune checkpoint inhibitors therapy in cancer patients. Cancer Med 2022; 12:1841-1849. [PMID: 35770869 PMCID: PMC9883533 DOI: 10.1002/cam4.4994] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 06/09/2022] [Accepted: 06/19/2022] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND Dysbiosis of the gut microbiota can lead to impaired therapeutic effect of immune checkpoint inhibitors (ICIs). This study aimed to investigate the use of probiotics on the clinical outcomes of cancer patients receiving ICIs therapy. METHOD PubMed, EMBASE, and the Cochrane Library database were searched to retrieve relevant studies that exploring the relationship between probiotics and the efficacy of ICIs. The primary endpoints included overall survival (OS) and progression-free survival (PFS), evaluated by the hazard rations (HRs) with 95% confidence intervals (CI), and the secondary endpoint was objective response rate (ORR), evaluated by the odd ratio (OR) with a 95% CI. RESULTS A total of five studies including 1031 patients were eligible for analysis. Our results indicated that the use of probiotics was associated with a superior OS (HR = 0.50, 95% CI: 0.30-0.85, p = 0.01) and PFS (HR = 0.51, 95% CI: 0.42-0.61, p < 0.01), but had no relationship with ORR (OR = 2.11, 95%CI: 0.51-8.65, p = 0.30) in non-small cell lung cancer (NSCLC) patients. CONCLUSIONS Probiotics were positively correlated with OS and PFS in NSCLC patients administrated with ICIs, but had no relationship with ORR.
Collapse
Affiliation(s)
- Luying Wan
- Department of OncologyThe First Affiliated Hospital of Fujian Medical UniversityFuzhouFujianChina
| | - Chunlan Wu
- Department of OncologyThe First Affiliated Hospital of Fujian Medical UniversityFuzhouFujianChina
| | - Qin Wu
- Department of OncologyThe First Affiliated Hospital of Fujian Medical UniversityFuzhouFujianChina
| | - Shuimei Luo
- Department of OncologyThe First Affiliated Hospital of Fujian Medical UniversityFuzhouFujianChina
| | - Junjin Liu
- Department of Infectious DiseasesThe First Affiliated Hospital of Fujian Medical UniversityFuzhouFujianChina
| | - Xianhe Xie
- Department of OncologyThe First Affiliated Hospital of Fujian Medical UniversityFuzhouFujianChina,Fujian Key Laboratory of Precision Medicine for CancerThe First Affiliated Hospital of Fujian Medical UniversityFuzhouFujianChina
| |
Collapse
|
21
|
Tegegne BA, Kebede B. Probiotics, their prophylactic and therapeutic applications in human health development: A review of the literature. Heliyon 2022; 8:e09725. [PMID: 35785237 PMCID: PMC9240980 DOI: 10.1016/j.heliyon.2022.e09725] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 04/03/2022] [Accepted: 06/09/2022] [Indexed: 02/07/2023] Open
Abstract
Antibiotics do not differentiate between good and bad germs, disrupting normal microflora and causing vitamin deficiency in the human body. They also kill healthy bacteria in the gut and genital tract on a large scale, weakening the host's defense mechanism. Probiotics are a colony of bacteria that live in our intestines and are regarded as a metabolic 'organ' due to their beneficial effects on human health, including metabolism and immunological function. They are used in clinical settings to prevent and treat conditions such as diarrhoea, colon cancer, hypertension, diabetes, acute pancreatitis, Helicobacter pylori infection, ventilator-associated pneumonia, migraine and autism. Probiotics may modify immunological activity by increasing innate and adaptive immune responses, altering microbial habitat in the intestine, improving gut barrier function, competitive adherence to the mucosa and epithelium, and producing antimicrobial compounds. The aim of this study is to index that further in depth researches to be conducted on probiotics pivotal role in the prophylaxis and therapeutic usage for a variety of disease that may or may not have treatment alternatives. Key words such as probiotics, microbiota, prophylactics, and therapeutic applications were searched extensively in research databases such as PubMed, PubMed Central (PMC), Scopus, Web of Science, Research Gate, Google Scholar, and Cochrane Library. This concise narrative review article summarized primarily the history, selection, mechanism/mode of action, recent advances in prophylactic and therapeutic applications, and future directions in the use of probiotics for prophylactic and therapeutic applications.
Collapse
|
22
|
Yue Y, Wang S, Shi J, Xie Q, Li N, Guan J, Evivie SE, Liu F, Li B, Huo G. Effects of Lactobacillus acidophilus KLDS1.0901 on Proliferation and Apoptosis of Colon Cancer Cells. Front Microbiol 2022; 12:788040. [PMID: 35250903 PMCID: PMC8895954 DOI: 10.3389/fmicb.2021.788040] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 12/22/2021] [Indexed: 12/20/2022] Open
Abstract
Colon cancer is the most common type of malignant tumor. The cytotoxicity effect of lactic acid bacteria may be active by inhibiting cancer cell proliferation, producing anticancer compounds, and inducing apoptosis in cancer cells, but the mechanism is unclear. Our previous study revealed that Lactobacillus acidophilus KLDS1.0901 has good probiotic properties. In this study, We screened out the highest inhibition rate of L. acidophilus KLDS1.0901 and assessed the effects on the proliferation of HT-29, Caco-2, and IEC-6 cells. Then, the apoptosis mechanism of HT-29 cells was studied when treated with L. acidophilus KLDS1.0901. Results showed that L. acidophilus KLDS1.0901 inhibited the proliferation of HT-29 and Caco-2 cells in a dose-dependent manner and reached the maximum under the condition of multiplicity of infection (MOI) = 100 (rate of Lactobacillus to cells) at 48 h. With the increase in time and MOI, reactive oxygen species in HT-29 cells, the apoptosis rates of HT-29 cells were increased, and the amount of blue fluorescence of the cells was also increased after Hoechst 33258 staining. Furthermore, L. acidophilus KLDS1.0901 reduced the mitochondrial membrane potential of HT-29 cells. Notably, 1,133 differentially expressed genes were screened by transcriptomics research, including 531 up-regulated genes and 602 down-regulated genes. These genes were involved in the nuclear factor κB and PI3K-AKT signaling pathways related to the apoptosis of HT-29 cells. These findings suggested that L. acidophilus KLDS1.0901 has the potential to be used in the development of a new type of functional foods for adjuvant treatment of colon cancer.
Collapse
Affiliation(s)
- Yingxue Yue
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin, China
- Food College, Northeast Agricultural University, Harbin, China
| | - Song Wang
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin, China
- Food College, Northeast Agricultural University, Harbin, China
| | - Jialu Shi
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin, China
- Food College, Northeast Agricultural University, Harbin, China
| | - Qinggang Xie
- Heilongjiang Feihe Dairy Co., Ltd., Qiqihaer, China
| | - Na Li
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin, China
- Food College, Northeast Agricultural University, Harbin, China
| | - Jiaqi Guan
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin, China
- Food College, Northeast Agricultural University, Harbin, China
| | - Smith Etareri Evivie
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin, China
- Department of Animal Science, Faculty of Agriculture, University of Benin, Benin City, Nigeria
- Department of Food Science and Human Nutrition, Faculty of Agriculture, University of Benin, Benin City, Nigeria
| | - Fei Liu
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin, China
- Food College, Northeast Agricultural University, Harbin, China
| | - Bailiang Li
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin, China
- Food College, Northeast Agricultural University, Harbin, China
| | - Guicheng Huo
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin, China
- Food College, Northeast Agricultural University, Harbin, China
| |
Collapse
|
23
|
Role of probiotics in the management of cervical cancer: An update. Clin Nutr ESPEN 2022; 48:5-16. [DOI: 10.1016/j.clnesp.2022.02.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 12/27/2021] [Accepted: 02/16/2022] [Indexed: 12/14/2022]
|
24
|
Pham F, Moinard-Butot F, Coutzac C, Chaput N. Cancer and immunotherapy: a role for microbiota composition. Eur J Cancer 2021; 155:145-154. [PMID: 34375896 DOI: 10.1016/j.ejca.2021.06.051] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 06/25/2021] [Accepted: 06/28/2021] [Indexed: 02/07/2023]
Abstract
Human microbiota plays a key role in preserving homeostasis; therefore, alteration in its composition is associated with susceptibility to various diseases. Recent findings suggest that gut microbiota may influence response to cancer treatment, especially immune checkpoint blockers (ICBs). The development of ICBs has changed outcomes of patients with cancer and has allowed sustained recovery. Unfortunately, some patients do not respond to ICBs, and microbiota may be a promising new biomarker to identify patients who will have benefit from ICBs. This review presents relationship between microbiome composition or microbiota-derived metabolites and response to ICBs or immune-related adverse events. Furthermore, we will present different strategies to modulate microbiota composition in patients to enhance ICB efficacy or dampen their toxicities which could lead to the emergence of interesting complementary treatments.
Collapse
Affiliation(s)
- Fiona Pham
- Department of Pharmacy, Hôpital Henri Mondor, AP-HP Créteil, F-94000, France; Department of Pharmacy, Centre Intercommunal de Créteil Créteil, F-94000, France
| | - Fabien Moinard-Butot
- Department of Medical Oncology, Strasbourg Europe Institute of Oncology Strasbourg, F-67033, France
| | - Clélia Coutzac
- Centre Léon Bérard, Cancer Research Center of Lyon (CRCL), Lyon, F-69008, France.
| | - Nathalie Chaput
- Laboratory of Immunomonitoring in Oncology, Gustave Roussy Cancer Center, Villejuif, F-94805, France; University Paris-Saclay, School of Pharmacy, Chatenay-Malabry, F-92296, France.
| |
Collapse
|
25
|
Casas-Solís J, Huizar-López MDR, Irecta-Nájera CA, Pita-López ML, Santerre A. Immunomodulatory Effect of Lactobacillus casei in a Murine Model of Colon Carcinogenesis. Probiotics Antimicrob Proteins 2021; 12:1012-1024. [PMID: 31797281 DOI: 10.1007/s12602-019-09611-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
We previously reported beneficial effects of the probiotic strain Lactobacillus casei 393 in hindering colon carcinogenesis in a 1,2-dimethylhydrazine (DMH)-induced BALB/c mouse model of colon cancer. In the present study, we investigated the effect of preventive administration of L. casei 393 on the levels of selected pro- and anti-inflammatory circulating cytokines, as well as subpopulations of splenic T cells. The resulting experimental data on IFNγ, TNFα, IL-10, and colon histological features demonstrated that administration of L. casei 2 weeks before DMH treatment impaired the pro-inflammatory effect of DMH, while maintaining the levels of the three cytokines as well as colon histology; it also modulated splenic CD4+, CD8+, and NK T cell subpopulations. The preventive administration of L. casei to DMH-treated mice increased IL-17A synthesis and Treg percentages, further indicating a tumor-protecting role. Together, the results suggest that the colon-cancer-protective properties of L. casei 393 involve the dampening of inflammation through cytokine homeostasis and the maintenance of a healthy T cell subpopulation dynamic. For these reasons, probiotics such as L. casei may contribute to the health of the host as they promote optimal control of the immune response. Further, they may be used as prophylactic agents in combination with standard therapies against colon cancer.
Collapse
Affiliation(s)
- Josefina Casas-Solís
- Departamento de Biología Celular y Molecular, Centro Universitario de Ciencias Biológicas y Agropecuarias, Universidad de Guadalajara, Carretera Guadalajara-Nogales Km 15.5, Las Agujas, CP, 45110, Zapopan, Jalisco, México
| | - María Del Rosario Huizar-López
- Departamento de Biología Celular y Molecular, Centro Universitario de Ciencias Biológicas y Agropecuarias, Universidad de Guadalajara, Carretera Guadalajara-Nogales Km 15.5, Las Agujas, CP, 45110, Zapopan, Jalisco, México
| | - Cesar Antonio Irecta-Nájera
- Departamento de Salud, El Colegio de La Frontera Sur, Carretera a Reforma Km15.5 s/n, Ra ElGuieno 2ª Sección, 86280, Villahermosa, Tabasco, México
| | - María Luisa Pita-López
- Departamento de Ciencias Básicas para la Salud, CIBIMEC, Centro Universitario del Sur, Universidad de Guadalajara, Av. Enrique Arreola Silva 883, CP4900, Cd. Guzmán, Guadalajara, Jalisco, México
| | - Anne Santerre
- Departamento de Biología Celular y Molecular, Centro Universitario de Ciencias Biológicas y Agropecuarias, Universidad de Guadalajara, Carretera Guadalajara-Nogales Km 15.5, Las Agujas, CP, 45110, Zapopan, Jalisco, México.
| |
Collapse
|
26
|
Huang X, Pan J, Xu F, Shao B, Wang Y, Guo X, Zhou S. Bacteria-Based Cancer Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2003572. [PMID: 33854892 PMCID: PMC8025040 DOI: 10.1002/advs.202003572] [Citation(s) in RCA: 131] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 11/03/2020] [Indexed: 05/24/2023]
Abstract
In the past decade, bacteria-based cancer immunotherapy has attracted much attention in the academic circle due to its unique mechanism and abundant applications in triggering the host anti-tumor immunity. One advantage of bacteria lies in their capability in targeting tumors and preferentially colonizing the core area of the tumor. Because bacteria are abundant in pathogen-associated molecular patterns that can effectively activate the immune cells even in the tumor immunosuppressive microenvironment, they are capable of enhancing the specific immune recognition and elimination of tumor cells. More attractively, during the rapid development of synthetic biology, using gene technology to enable bacteria to be an efficient producer of immunotherapeutic agents has led to many creative immunotherapy paradigms. The combination of bacteria and nanomaterials also displays infinite imagination in the multifunctional endowment for cancer immunotherapy. The current progress report summarizes the recent advances in bacteria-based cancer immunotherapy with specific foci on the applications of naive bacteria-, engineered bacteria-, and bacterial components-based cancer immunotherapy, and at the same time discusses future directions in this field of research based on the present developments.
Collapse
Affiliation(s)
- Xuehui Huang
- Key Laboratory of Advanced Technologies of MaterialsMinistry of EducationSchool of Materials Science and EngineeringSouthwest Jiaotong UniversityChengdu610031China
| | - Jingmei Pan
- Key Laboratory of Advanced Technologies of MaterialsMinistry of EducationSchool of Materials Science and EngineeringSouthwest Jiaotong UniversityChengdu610031China
| | - Funeng Xu
- Key Laboratory of Advanced Technologies of MaterialsMinistry of EducationSchool of Materials Science and EngineeringSouthwest Jiaotong UniversityChengdu610031China
| | - Binfen Shao
- School of Life Science and EngineeringSouthwest Jiaotong UniversityChengdu610031China
| | - Yi Wang
- School of Life Science and EngineeringSouthwest Jiaotong UniversityChengdu610031China
| | - Xing Guo
- Key Laboratory of Advanced Technologies of MaterialsMinistry of EducationSchool of Materials Science and EngineeringSouthwest Jiaotong UniversityChengdu610031China
| | - Shaobing Zhou
- Key Laboratory of Advanced Technologies of MaterialsMinistry of EducationSchool of Materials Science and EngineeringSouthwest Jiaotong UniversityChengdu610031China
| |
Collapse
|
27
|
Jafari-Nasab T, Khaleghi M, Farsinejad A, Khorrami S. Probiotic potential and anticancer properties of Pediococcus sp. isolated from traditional dairy products. BIOTECHNOLOGY REPORTS (AMSTERDAM, NETHERLANDS) 2021; 29:e00593. [PMID: 33598413 PMCID: PMC7868823 DOI: 10.1016/j.btre.2021.e00593] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 11/23/2020] [Accepted: 01/18/2021] [Indexed: 02/06/2023]
Abstract
Herein, 18 lactic acid bacteria isolated from 30 samples of traditional dairy products were identified, and their probiotic potential was evaluated. According to the results, almost all strains showed the probiotic properties sufficiently, though M1 had better characterise. 16S rRNA gene sequencing revealed that this strain belongs to the Pediococcus sp. (<95 % similarity). This strain had substantial antipathogenic activity and did not show any worrying antibiotic resistance. Also, the strain was resistant to high concentrations of bile salt (1 %), NaCl (6.5 %), and low pH (2). Furthermore, it was revealed that cell-free supernatant (CFS), heat-killed cells and live cells derived from M1 significantly decreased the viability of MCF-7 cells so that the CFS resulted in 85 % cell death. Flow cytometry and western blot analysis determined that this compound induced apoptosis in the cancerous cells through increasing the BAX protein expression and decreasing the Bcl-2 protein expression.
Collapse
Affiliation(s)
- Tayebeh Jafari-Nasab
- Department of Biology, Faculty of Sciences, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Moj Khaleghi
- Department of Biology, Faculty of Sciences, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Alireza Farsinejad
- Department of Hematology and Medical Laboratory Sciences, Faculty of Allied Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Sadegh Khorrami
- Department of Biology, Faculty of Sciences, Shahid Bahonar University of Kerman, Kerman, Iran
| |
Collapse
|
28
|
Oliva M, Mulet-Margalef N, Ochoa-De-Olza M, Napoli S, Mas J, Laquente B, Alemany L, Duell EJ, Nuciforo P, Moreno V. Tumor-Associated Microbiome: Where Do We Stand? Int J Mol Sci 2021; 22:1446. [PMID: 33535583 PMCID: PMC7867144 DOI: 10.3390/ijms22031446] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 01/26/2021] [Accepted: 01/28/2021] [Indexed: 12/14/2022] Open
Abstract
The study of the human microbiome in oncology is a growing and rapidly evolving field. In the past few years, there has been an exponential increase in the number of studies investigating associations of microbiome and cancer, from oncogenesis and cancer progression to resistance or sensitivity to specific anticancer therapies. The gut microbiome is now known to play a significant role in antitumor immune responses and in predicting the efficacy of immune-checkpoint inhibitors in cancer patients. Beyond the gut, the tumor-associated microbiome-microbe communities located either in the tumor or within its body compartment-seems to interact with the local microenvironment and the tumor immune contexture, ultimately impacting cancer progression and treatment outcome. However, pre-clinical research focusing on causality and mechanistic pathways as well as proof-of-concept studies are still needed to fully understand the potential clinical utility of microbiome in cancer patients. Moreover, there is a need for the standardization of methodology and the implementation of quality control across microbiome studies to allow for a better interpretation and greater comparability of the results reported between them. This review summarizes the accumulating evidence in the field and discusses the current and upcoming challenges of microbiome studies.
Collapse
Affiliation(s)
- Marc Oliva
- Medical Oncology Department, Catalan Institute of Oncology L’Hospitalet de Llobregat, 08908 Catalonia, Spain; (N.M.-M.); (B.L.)
- ONCOBELL Program, Bellvitge Biomedical Research Institute (IDIBELL), L’Hospitalet de Llobregat, 08908 Catalonia, Spain; (J.M.); (E.J.D.); (V.M.)
| | - Nuria Mulet-Margalef
- Medical Oncology Department, Catalan Institute of Oncology L’Hospitalet de Llobregat, 08908 Catalonia, Spain; (N.M.-M.); (B.L.)
- ONCOBELL Program, Bellvitge Biomedical Research Institute (IDIBELL), L’Hospitalet de Llobregat, 08908 Catalonia, Spain; (J.M.); (E.J.D.); (V.M.)
| | - Maria Ochoa-De-Olza
- Service of Immuno-Oncology, Department of Oncology, Lausanne University Hospital, 1011 Lausanne, Switzerland;
- Ludwig Institute for Cancer Research, University of Lausanne, 1066 Lausanne, Switzerland
| | - Stefania Napoli
- Molecular Oncology Group, Vall d’Hebron Institute of Oncology, 08035 Barcelona, Spain; (S.N.); (P.N.)
| | - Joan Mas
- ONCOBELL Program, Bellvitge Biomedical Research Institute (IDIBELL), L’Hospitalet de Llobregat, 08908 Catalonia, Spain; (J.M.); (E.J.D.); (V.M.)
- Oncology Data Analytics Program, Catalan Institute of Oncology (ICO), L’Hospitalet de Llobregat, 08908 Catalonia, Spain
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), 28029 Madrid, Spain;
| | - Berta Laquente
- Medical Oncology Department, Catalan Institute of Oncology L’Hospitalet de Llobregat, 08908 Catalonia, Spain; (N.M.-M.); (B.L.)
- ONCOBELL Program, Bellvitge Biomedical Research Institute (IDIBELL), L’Hospitalet de Llobregat, 08908 Catalonia, Spain; (J.M.); (E.J.D.); (V.M.)
| | - Laia Alemany
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), 28029 Madrid, Spain;
- Cancer Epidemiology Research Program, Catalan Institute of Oncology, L’Hospitalet de Llobregat, 08908 Catalonia, Spain
- EPIBELL Program, Bellvitge Biomedical Research Institute (IDIBELL), L’Hospitalet de Llobregat, 08908 Catalonia, Spain
| | - Eric J. Duell
- ONCOBELL Program, Bellvitge Biomedical Research Institute (IDIBELL), L’Hospitalet de Llobregat, 08908 Catalonia, Spain; (J.M.); (E.J.D.); (V.M.)
- Oncology Data Analytics Program, Catalan Institute of Oncology (ICO), L’Hospitalet de Llobregat, 08908 Catalonia, Spain
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), 28029 Madrid, Spain;
| | - Paolo Nuciforo
- Molecular Oncology Group, Vall d’Hebron Institute of Oncology, 08035 Barcelona, Spain; (S.N.); (P.N.)
| | - Victor Moreno
- ONCOBELL Program, Bellvitge Biomedical Research Institute (IDIBELL), L’Hospitalet de Llobregat, 08908 Catalonia, Spain; (J.M.); (E.J.D.); (V.M.)
- Oncology Data Analytics Program, Catalan Institute of Oncology (ICO), L’Hospitalet de Llobregat, 08908 Catalonia, Spain
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), 28029 Madrid, Spain;
| |
Collapse
|
29
|
Cancer Related Anemia: An Integrated Multitarget Approach and Lifestyle Interventions. Nutrients 2021; 13:nu13020482. [PMID: 33535496 PMCID: PMC7912724 DOI: 10.3390/nu13020482] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 01/26/2021] [Accepted: 01/28/2021] [Indexed: 12/16/2022] Open
Abstract
Cancer is often accompanied by worsening of the patient's iron profile, and the resulting anemia could be a factor that negatively impacts antineoplastic treatment efficacy and patient survival. The first line of therapy is usually based on oral or intravenous iron supplementation; however, many patients remain anemic and do not respond. The key might lie in the pathogenesis of the anemia itself. Cancer-related anemia (CRA) is characterized by a decreased circulating serum iron concentration and transferrin saturation despite ample iron stores, pointing to a more complex problem related to iron homeostatic regulation and additional factors such as chronic inflammatory status. This review explores our current understanding of iron homeostasis in cancer, shedding light on the modulatory role of hepcidin in intestinal iron absorption, iron recycling, mobilization from liver deposits, and inducible regulators by infections and inflammation. The underlying relationship between CRA and systemic low-grade inflammation will be discussed, and an integrated multitarget approach based on nutrition and exercise to improve iron utilization by reducing low-grade inflammation, modulating the immune response, and supporting antioxidant mechanisms will also be proposed. Indeed, a Mediterranean-based diet, nutritional supplements and exercise are suggested as potential individualized strategies and as a complementary approach to conventional CRA therapy.
Collapse
|
30
|
Pimentel TC, Costa WKAD, Barão CE, Rosset M, Magnani M. Vegan probiotic products: A modern tendency or the newest challenge in functional foods. Food Res Int 2021; 140:110033. [DOI: 10.1016/j.foodres.2020.110033] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 12/02/2020] [Accepted: 12/13/2020] [Indexed: 02/06/2023]
|
31
|
The role of Lactobacillus plantarum 299v in supporting treatment of selected diseases. Cent Eur J Immunol 2021; 45:488-493. [PMID: 33613097 PMCID: PMC7882405 DOI: 10.5114/ceji.2020.101515] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 01/29/2019] [Indexed: 12/13/2022] Open
Abstract
Alterations in composition of human gut microbiome can lead to its dysbiosis. It is associated with gastrointestinal side effects during anti-cancer treatment, antibiotics administration, or infectious agents. There are studies confirming positive effect of consuming Lactobacillus plantarum 299v on intestinal microflora. This review summarizes the current knowledge about the role of L. plantarum 299v in supporting treatment of selected diseases, such as cancer, irritable bowel syndrome (IBS), and Clostridium difficile infection. The immunomodulating properties of L. plantarum 299v include an increase in the level of anti-inflammatory cytokines, which reduce the risk of cancer and improve the efficacy of regimens. The intake of L. plantarum 299v provides benefits for IBS patients, mainly due to normalization of stool and relief of abdominal pain, which significantly improves the quality of life of IBS patients. In addition, the intake of L. plantarum 299v prevents C. difficile-associated diarrhea among patients receiving antibiotic treatment. Due to the limited possibilities of treating these diseases and numerous complications of cancer treatment, there is a need for new therapeutic strategies. The administration of L. plantarum 299v seems to be useful in these cases.
Collapse
|
32
|
Mohammadi M, Shadnoush M, Sohrabvandi S, Yousefi M, Khorshidian N, Mortazavian AM. Probiotics as potential detoxification tools for mitigation of pesticides: a mini review. Int J Food Sci Technol 2020. [DOI: 10.1111/ijfs.14880] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Mehrdad Mohammadi
- Department of Food Technology Research National Nutrition and Food Technology Research Institute, Faculty of Nutrition Sciences and Food Technology Shahid Beheshti University of Medical Sciences Tehran Iran
| | - Mahdi Shadnoush
- Department of Clinical Nutrition Faculty of Nutrition Sciences and Food Technology National Nutrition and Food Technology Research Institute Shahid Beheshti University of Medical Sciences Tehran Iran
| | - Sara Sohrabvandi
- Department of Food Technology Research National Nutrition and Food Technology Research Institute, Faculty of Nutrition Sciences and Food Technology Shahid Beheshti University of Medical Sciences Tehran Iran
| | - Mojtaba Yousefi
- Food Safety Research Center (Salt) Semnan University of Medical Sciences Semnan Iran
| | - Nasim Khorshidian
- Food Safety Research Center (Salt) Semnan University of Medical Sciences Semnan Iran
| | - Amir M. Mortazavian
- Food Safety Research Center Shahid Beheshti University of Medical Sciences Tehran Iran
| |
Collapse
|
33
|
Saberian M, Shahidi Delshad E, Habibi M. The Effect of Bifidobacterium Bifidum Supernatant and Cell Mass on the Proliferation Potential of Rat Bone Marrow-Derived Stromal Cells. IRANIAN JOURNAL OF MEDICAL SCIENCES 2020; 45:269-276. [PMID: 32801416 PMCID: PMC7395953 DOI: 10.30476/ijms.2019.45772] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Background Mesenchymal stem cells (MSCs) are widely used to treat various diseases, however, their proliferative potential reduces after a number of passages. It has been shown that some probiotics such as Bifidobacterium Bifidum (B. bifidum) affect the proliferation of various cell lineages. The present study aimed to investigate the effect of B. bifidum on the proliferation of rat bone marrow stromal cells (rBMSCs) and to develop a method for compensating their proliferation reduction after some passages. Methods The present experimental study was conducted at Tehran University of Medical Sciences, Tehran, Iran, in 2017. The stromal cells were isolated from rBMSCs and their mesenchymal properties were confirmed by osteogenic and adipogenic differentiation media and staining. B. bifidum was cultured and the B. bifidum supernatant (BS) and bacterial cell mass (BCM) were extracted. The rBMSCs were treated with different concentrations of BS and BCM. The MTT assay was performed to measure the number of viable cells in the culture. Cell proliferation was analyzed using the paired-sample t test. Results Cell proliferation increased as the concentration of bacteria was increased logarithmically (0, 0.1, 0.3, 0.9, 3, 9, 30 μL/mL). In comparison with BS, cells treated with BCM showed increased cell proliferation at lower concentrations. This effect was caused by removing the "de Man, Rogosa, and Sharpe" (MRS) broth medium from the BCM culture. The optimal concentration of bacteria with the most significant effect on rBMSCs proliferation was determined. Conclusion A significant increase in the proliferation of stromal cells was observed; confirming the stimulatory potential of probiotics (B. bifidum) on various cells. The use of products containing probiotic bacteria can increase the proliferation potential of BMSCs.
Collapse
Affiliation(s)
- Mostafa Saberian
- Department of Cellular and Molecular Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences. Tehran, Iran
| | - Elham Shahidi Delshad
- Shahid Rajaei Cardiovascular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Masoud Habibi
- Department of Genetics, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| |
Collapse
|
34
|
Pourmollaei S, Barzegari A, Farshbaf-Khalili A, Nouri M, Fattahi A, Shahnazi M, Dittrich R. Anticancer effect of bacteria on cervical cancer: Molecular aspects and therapeutic implications. Life Sci 2020; 246:117413. [PMID: 32035929 DOI: 10.1016/j.lfs.2020.117413] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 01/27/2020] [Accepted: 02/04/2020] [Indexed: 12/21/2022]
Abstract
Cervical cancer is the second common cancer and the third leading cause of cancer deaths among women in less developed countries. It has been indicated that changes in vaginal microbiome play an important role in the occurrence and development of cervical cancer. However, studies have shown that probiotics play an effective role in fighting cancer by affecting pathogenic bacteria, inducing cancer cells apoptosis, and other anticancer activities. Therefore, the purpose of the present study is reviewing the anticancer effect of cervicovaginal bacteria and their potential for cervical cancer treatment.
Collapse
Affiliation(s)
- Soraya Pourmollaei
- Department of Midwifery, Faculty of Nursing and Midwifery, Tabriz University of Medical Sciences, Tabriz, Iran; Students' Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Abolfazl Barzegari
- Research Center of Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Azizeh Farshbaf-Khalili
- Physical Medicine and Rehabilitation Research Centre, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Nouri
- Department of Reproductive Biology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran; Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Fattahi
- Department of Reproductive Biology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran; Women's Reproductive Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Obstetrics and Gynecology, Erlangen University Hospital, Friedrich-Alexander-Universität of Erlangen-Nürnberg, Germany.
| | - Mahnaz Shahnazi
- Department of Midwifery, Faculty of Nursing and Midwifery, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Ralf Dittrich
- Department of Obstetrics and Gynecology, Erlangen University Hospital, Friedrich-Alexander-Universität of Erlangen-Nürnberg, Germany
| |
Collapse
|
35
|
Caruana JC, Walper SA. Bacterial Membrane Vesicles as Mediators of Microbe - Microbe and Microbe - Host Community Interactions. Front Microbiol 2020; 11:432. [PMID: 32265873 PMCID: PMC7105600 DOI: 10.3389/fmicb.2020.00432] [Citation(s) in RCA: 147] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 03/02/2020] [Indexed: 01/18/2023] Open
Abstract
Bacterial membrane vesicles are proteoliposomal nanoparticles produced by both Gram-negative and Gram-positive bacteria. As they originate from the outer surface of the bacteria, their composition and content is generally similar to the parent bacterium’s membrane and cytoplasm. However, there is ample evidence that preferential packaging of proteins, metabolites, and toxins into vesicles does occur. Incorporation into vesicles imparts a number of benefits to the cargo, including protection from degradation by other bacteria, the host organism, or environmental factors, maintenance of a favorable microenvironment for enzymatic activity, and increased potential for long-distance movement. This enables vesicles to serve specialized functions tailored to changing or challenging environments, particularly in regard to microbial community interactions including quorum sensing, biofilm formation, antibiotic resistance, antimicrobial peptide expression and deployment, and nutrient acquisition. Additionally, based on their contents, vesicles play crucial roles in host-microbe interactions as carriers of virulence factors and other modulators of host cell function. Here, we discuss recent advances in our understanding of how vesicles function as signals both within microbial communities and between pathogenic or commensal microbes and their mammalian hosts. We also highlight a few areas that are currently ripe for additional research, including the mechanisms of selective cargo packaging into membrane vesicles and of cargo processing once it enters mammalian host cells, the function of vesicles in transfer of nucleic acids among bacteria, and the possibility of engineering commensal bacteria to deliver cargo of interest to mammalian hosts in a controlled manner.
Collapse
Affiliation(s)
- Julie C Caruana
- American Society for Engineering Education, Washington, DC, United States
| | - Scott A Walper
- US Naval Research Laboratory, Center for Biomolecular Science and Engineering, Washington, DC, United States
| |
Collapse
|
36
|
Zhang D, Li S, Wang N, Tan HY, Zhang Z, Feng Y. The Cross-Talk Between Gut Microbiota and Lungs in Common Lung Diseases. Front Microbiol 2020; 11:301. [PMID: 32158441 PMCID: PMC7052046 DOI: 10.3389/fmicb.2020.00301] [Citation(s) in RCA: 245] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 02/10/2020] [Indexed: 12/12/2022] Open
Abstract
Emerging findings indicate there is a vital cross-talk between gut microbiota and the lungs, which is known as gut-lung axis. The gut disturbances in lung diseases including allergy, asthma, chronic obstructive pulmonary disease, cystic fibrosis and lung cancer were observed by extensive studies. Investigating how gut microbiota impact other distant organs is of great interest in recent years. Although it has not been fully understood whether the disturbance is the cause or effect of lung diseases, alterations in the gut microbial species and metabolites have been linked to changes in immune responses and inflammation as well as the disease development in the lungs. In this article, we systemically review the role and mechanisms underlying the changes in the constituent of gut microbiota and metabolites in lung diseases. In particular, the roles of gut-lung axis in mediating immune responses and reshaping inflammation are highlighted. Furthermore, we discuss the potential of strategies to manipulate the gut microbiota and metabolites as the therapeutic approach for lung diseases.
Collapse
Affiliation(s)
- Dapeng Zhang
- First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Sha Li
- School of Chinese Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Ning Wang
- School of Chinese Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Hor-Yue Tan
- School of Chinese Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Zhimin Zhang
- First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yibin Feng
- School of Chinese Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
37
|
Yang JJ, Yu D, Xiang YB, Blot W, White E, Robien K, Sinha R, Park Y, Takata Y, Lazovich D, Gao YT, Zhang X, Lan Q, Bueno-de-Mesquita B, Johansson I, Tumino R, Riboli E, Tjønneland A, Skeie G, Quirós JR, Johansson M, Smith-Warner SA, Zheng W, Shu XO. Association of Dietary Fiber and Yogurt Consumption With Lung Cancer Risk: A Pooled Analysis. JAMA Oncol 2020; 6:e194107. [PMID: 31647500 PMCID: PMC6813596 DOI: 10.1001/jamaoncol.2019.4107] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 07/13/2019] [Indexed: 12/14/2022]
Abstract
Importance Dietary fiber (the main source of prebiotics) and yogurt (a probiotic food) confer various health benefits via modulating the gut microbiota and metabolic pathways. However, their associations with lung cancer risk have not been well investigated. Objective To evaluate the individual and joint associations of dietary fiber and yogurt consumption with lung cancer risk and to assess the potential effect modification of the associations by lifestyle and other dietary factors. Design, Setting, and Participants This pooled analysis included 10 prospective cohorts involving 1 445 850 adults from studies that were conducted in the United States, Europe, and Asia. Data analyses were performed between November 2017 and February 2019. Using harmonized individual participant data, hazard ratios and 95% confidence intervals for lung cancer risk associated with dietary fiber and yogurt intakes were estimated for each cohort by Cox regression and pooled using random-effects meta-analysis. Participants who had a history of cancer at enrollment or developed any cancer, died, or were lost to follow-up within 2 years after enrollment were excluded. Exposures Dietary fiber intake and yogurt consumption measured by validated instruments. Main Outcomes and Measures Incident lung cancer, subclassified by histologic type (eg, adenocarcinoma, squamous cell carcinoma, and small cell carcinoma). Results The analytic sample included 627 988 men, with a mean (SD) age of 57.9 (9.0) years, and 817 862 women, with a mean (SD) age of 54.8 (9.7) years. During a median follow-up of 8.6 years, 18 822 incident lung cancer cases were documented. Both fiber and yogurt intakes were inversely associated with lung cancer risk after adjustment for status and pack-years of smoking and other lung cancer risk factors: hazard ratio, 0.83 (95% CI, 0.76-0.91) for the highest vs lowest quintile of fiber intake; and hazard ratio, 0.81 (95% CI, 0.76-0.87) for high vs no yogurt consumption. The fiber or yogurt associations with lung cancer were significant in never smokers and were consistently observed across sex, race/ethnicity, and tumor histologic type. When considered jointly, high yogurt consumption with the highest quintile of fiber intake showed more than 30% reduced risk of lung cancer than nonyogurt consumption with the lowest quintile of fiber intake (hazard ratio, 0.67 [95% CI, 0.61-0.73] in total study populations; hazard ratio, 0.69 [95% CI, 0.54-0.89] in never smokers), suggesting potential synergism. Conclusions and Relevance Dietary fiber and yogurt consumption was associated with reduced risk of lung cancer after adjusting for known risk factors and among never smokers. Our findings suggest a potential protective role of prebiotics and probiotics against lung carcinogenesis.
Collapse
Affiliation(s)
- Jae Jeong Yang
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Danxia Yu
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Yong-Bing Xiang
- State Key Laboratory of Oncogene and Related Genes & Department of Epidemiology, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - William Blot
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt University Medical Center, Nashville, Tennessee
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Emily White
- Cancer Prevention Program, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Kim Robien
- Department of Exercise and Nutrition Sciences, Milken Institute School of Public Health, George Washington University, Washington, District of Columbia
| | - Rashmi Sinha
- Division of Epidemiology & Genetics, National Cancer Institute, Bethesda, Maryland
| | - Yikyung Park
- Division of Public Health Sciences, Department of Surgery, Washington University School of Medicine, St Louis, Missouri
| | - Yumie Takata
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt University Medical Center, Nashville, Tennessee
- College of Public Health and Human Sciences, Oregon State University, Corvallis
| | - DeAnn Lazovich
- Division of Epidemiology & Community Health, School of Public Health, University of Minnesota, Minneapolis
- Masonic Cancer Center, University of Minnesota, Minneapolis
| | - Yu-Tang Gao
- State Key Laboratory of Oncogene and Related Genes & Department of Epidemiology, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xuehong Zhang
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| | - Qing Lan
- Division of Epidemiology & Genetics, National Cancer Institute, Bethesda, Maryland
| | - Bas Bueno-de-Mesquita
- Department for Determinants of Chronic Diseases, National Institute for Public Health and the Environment, Bilthoven, the Netherlands
- Department of Gastroenterology and Hepatology, University Medical Centre, Utrecht, the Netherlands
| | | | - Rosario Tumino
- Cancer Registry and Histopathology Department, Civic-M.P. Arezzo Hospital, American Samoa, Ragusa, Italy
| | - Elio Riboli
- Faculty of Medicine, School of Public Health, Imperial College, London, United Kingdom
| | - Anne Tjønneland
- Diet, Genes and Environment, Danish Cancer Society Research Center, Copenhagen, Denmark
- Denmark Department of Public Health, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Guri Skeie
- Department of Community Medicine, UIT, The Arctic University of Norway, Tromsø, Norway
| | | | - Mattias Johansson
- Genetic Epidemiology Group, International Agency for Research on Cancer, Lyons, France
| | - Stephanie A. Smith-Warner
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Wei Zheng
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt University Medical Center, Nashville, Tennessee
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Xiao-Ou Shu
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt University Medical Center, Nashville, Tennessee
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
38
|
Heydari Z, Rahaie M, Alizadeh AM. Different anti-inflammatory effects of Lactobacillus acidophilus and Bifidobactrum bifidioum in hepatocellular carcinoma cancer mouse through impact on microRNAs and their target genes. JOURNAL OF NUTRITION & INTERMEDIARY METABOLISM 2019. [DOI: 10.1016/j.jnim.2019.100096] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
39
|
Gaucher F, Bonnassie S, Rabah H, Marchand P, Blanc P, Jeantet R, Jan G. Review: Adaptation of Beneficial Propionibacteria, Lactobacilli, and Bifidobacteria Improves Tolerance Toward Technological and Digestive Stresses. Front Microbiol 2019; 10:841. [PMID: 31068918 PMCID: PMC6491719 DOI: 10.3389/fmicb.2019.00841] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 04/02/2019] [Indexed: 01/15/2023] Open
Abstract
This review deals with beneficial bacteria, with a focus on lactobacilli, propionibacteria, and bifidobacteria. As being recognized as beneficial bacteria, they are consumed as probiotics in various food products. Some may also be used as starters in food fermentation. In either case, these bacteria may be exposed to various environmental stresses during industrial production steps, including drying and storage, and during the digestion process. In accordance with their adaptation to harsh environmental conditions, they possess adaptation mechanisms, which can be induced by pretreatments. Adaptive mechanisms include accumulation of compatible solutes and of energy storage compounds, which can be largely modulated by the culture conditions. They also include the regulation of energy production pathways, as well as the modulation of the cell envelop, i.e., membrane, cell wall, surface layers, and exopolysaccharides. They finally lead to the overexpression of molecular chaperones and of stress-responsive proteases. Triggering these adaptive mechanisms can improve the resistance of beneficial bacteria toward technological and digestive stresses. This opens new perspectives for the improvement of industrial processes efficiency with regard to the survival of beneficial bacteria. However, this bibliographical survey evidenced that adaptive responses are strain-dependent, so that growth and adaptation should be optimized case-by-case.
Collapse
Affiliation(s)
- Floriane Gaucher
- STLO, Agrocampus Ouest, Institut National de la Recherche Agronomique, Paris, France
- Bioprox, Levallois-Perret, France
| | - Sylvie Bonnassie
- STLO, Agrocampus Ouest, Institut National de la Recherche Agronomique, Paris, France
- Science de la Vie et de la Terre, Université de Rennes 1, Rennes, France
| | - Houem Rabah
- STLO, Agrocampus Ouest, Institut National de la Recherche Agronomique, Paris, France
- Pôle Agronomique Ouest, Bba, Rennes, France
| | | | | | - Romain Jeantet
- STLO, Agrocampus Ouest, Institut National de la Recherche Agronomique, Paris, France
| | - Gwénaël Jan
- STLO, Agrocampus Ouest, Institut National de la Recherche Agronomique, Paris, France
| |
Collapse
|
40
|
Calabuig-Jiménez L, Betoret E, Betoret N, Patrignani F, Barrera C, Seguí L, Lanciotti R, Dalla Rosa M. High pressures homogenization (HPH) to microencapsulate L. salivarius spp. salivarius in mandarin juice. Probiotic survival and in vitro digestion. J FOOD ENG 2019. [DOI: 10.1016/j.jfoodeng.2018.07.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
41
|
Önal Darilmaz D, Sönmez Ş, Beyatli Y. The effects of inulin as a prebiotic supplement and the synbiotic interactions of probiotics to improve oxalate degrading activity. Int J Food Sci Technol 2018. [DOI: 10.1111/ijfs.13912] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Derya Önal Darilmaz
- Department of Biotechnology and Molecular Biology; Faculty of Science and Letters; Aksaray University; 68100 Aksaray Turkey
| | - Şule Sönmez
- Department of Biology; Faculty of Science; Gazi University; 06500 Ankara Turkey
| | - Yavuz Beyatli
- Department of Biology; Faculty of Science; Gazi University; 06500 Ankara Turkey
| |
Collapse
|
42
|
Dasari S, Ali SM, Zheng G, Chen A, Dontaraju VS, Bosland MC, Kajdacsy-Balla A, Munirathinam G. Vitamin K and its analogs: Potential avenues for prostate cancer management. Oncotarget 2017; 8:57782-57799. [PMID: 28915711 PMCID: PMC5593683 DOI: 10.18632/oncotarget.17997] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 04/15/2017] [Indexed: 01/27/2023] Open
Abstract
Epidemiological studies have demonstrated a relationship between cancer incidence and dietary habits. Especially intake of certain essential nutrients like vitamins has been shown to be beneficial in experimental studies and some clinical trials. Vitamin K (VK) is an essential nutrient involved in the blood clotting cascade, and there are considerable experimental data demonstrating its potential anticancer activity in several cancer types including prostate cancer. Previous in vitro and in vivo studies have focused mainly on anti-oxidative effects as the underlying anticancer mechanism of VK. However, recent studies reveal that VK inhibits the growth of cancer cells through other mechanisms, including apoptosis, cell cycle arrest, autophagy, and modulation of various transcription factors such as Myc and Fos. In the present review, we focus on the anticancer effect of dietary VK and its analogs on prostate cancer, with an emphasis on the signaling pathways that are activated following exposure to these compounds. This review also highlights the potential of VK and its derivatives as an adjuvant treatment in combination with other vitamins or with chemotherapeutic drugs. Based on our recent results and a review of the existing literature, we present evidence that VK and its derivatives can potentially be explored as cancer therapy, especially for prostate cancer.
Collapse
Affiliation(s)
- Subramanyam Dasari
- Department of Biomedical Sciences, College of Medicine, University of Illinois, Rockford, IL, USA
| | - Syed M Ali
- Department of Biomedical Sciences, College of Medicine, University of Illinois, Rockford, IL, USA
| | - Guoxing Zheng
- Department of Biomedical Sciences, College of Medicine, University of Illinois, Rockford, IL, USA
| | - Aoshuang Chen
- Department of Biomedical Sciences, College of Medicine, University of Illinois, Rockford, IL, USA
| | | | - Maarten C Bosland
- Department of Pathology, University of Illinois at Chicago, Chicago, IL, USA
| | | | - Gnanasekar Munirathinam
- Department of Biomedical Sciences, College of Medicine, University of Illinois, Rockford, IL, USA
| |
Collapse
|
43
|
Kathera C, Zhang J, Janardhan A, Sun H, Ali W, Zhou X, He L, Guo Z. Interacting partners of FEN1 and its role in the development of anticancer therapeutics. Oncotarget 2017; 8:27593-27602. [PMID: 28187440 PMCID: PMC5432360 DOI: 10.18632/oncotarget.15176] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 01/24/2017] [Indexed: 11/25/2022] Open
Abstract
Protein-protein interaction (PPI) plays a key role in cellular communication, Protein-protein interaction connected with each other with hubs and nods involved in signaling pathways. These interactions used to develop network based biomarkers for early diagnosis of cancer. FEN1(Flap endonuclease 1) is a central component in cellular metabolism, over expression and decrease of FEN1 levels may cause cancer, these regulation changes of Flap endonuclease 1reported in many cancer cells, to consider this data may needs to develop a network based biomarker. The current review focused on types of PPI, based on nature, detection methods and its role in cancer. Interacting partners of Flap endonuclease 1 role in DNA replication repair and development of anticancer therapeutics based on Protein-protein interaction data.
Collapse
Affiliation(s)
- Chandrasekhar Kathera
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Jing Zhang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Avilala Janardhan
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Hongfang Sun
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Wajid Ali
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Xiaolong Zhou
- The Laboratory of Animal Genetics, Breeding, and Reproduction, College of Animal Science and Technology, Zhejiang Agriculture and Forestry University, Hangzhou, China
| | - Lingfeng He
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Zhigang Guo
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| |
Collapse
|