1
|
Peñalver Bernabé B, Oliveira ML, Wolf PG, McLeod A, Gabel K, Cares K, Robinson N, DiPiazza B, Varady K, Tussing-Humphreys L. Intermittent Fasting: Implications for Obesity-Related Colorectal Tumorigenesis. Endocrinol Metab Clin North Am 2025; 54:61-83. [PMID: 39919878 DOI: 10.1016/j.ecl.2024.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 02/09/2025]
Abstract
Obesity is associated with metabolic and immune perturbations (ie, insulin resistance, increased inflammation, and oxidative stress), circadian rhythm dysregulation, and gut microbial changes that can promote colorectal tumorigenesis. Colorectal cancer (CRC) is the third most incident cancer in the United States. This narrative review examines the effects of intermittend fasting on factors influencing colon tumorigenesis, such as body weight, metabolic and immune markers, circadian rythm, and the gut microbiota in humans. Findings suggest that intermittent fasting regimens can lead to weight loss and shifts in metabolic markers, which could be preventive for CRC but effects on the gut microbiota composition and functions still remains elusive.
Collapse
Affiliation(s)
- Beatriz Peñalver Bernabé
- Department of Biomedical Engineering, University of Illinois Chicago, 851 South Morgan Street, Chicago, IL, USA; Center for Bioinformatics and Quantitative Biology, University of Illinois Chicago, Chicago, IL, USA
| | - Manoela Lima Oliveira
- Department of Kinesiology and Nutrition, University of Illinois Chicago, 1919 West Taylor Street, Chicago, IL, USA; University of Illinois Cancer Center, Chicago, IL, USA
| | - Patricia G Wolf
- Department of Nutrition Science, Purdue University, 700 Mitch Daniels Boulevard, West Lafayette, IN, USA; Purdue Institute for Cancer Research, West Lafayette, IN, USA
| | - Andrew McLeod
- Department of Kinesiology and Nutrition, University of Illinois Chicago, 1919 West Taylor Street, Chicago, IL, USA; University of Illinois Cancer Center, Chicago, IL, USA
| | - Kelsey Gabel
- Department of Kinesiology and Nutrition, University of Illinois Chicago, 1919 West Taylor Street, Chicago, IL, USA; Department of Nutrition Science, Purdue University, 700 Mitch Daniels Boulevard, West Lafayette, IN, USA
| | - Kate Cares
- Department of Kinesiology and Nutrition, University of Illinois Chicago, 1919 West Taylor Street, Chicago, IL, USA
| | - Nadia Robinson
- College of Nursing, University of Illinois Chicago, 845 South Damen Avenue, MC 802, Chicago, IL, USA
| | - Brittany DiPiazza
- Department of Kinesiology and Nutrition, University of Illinois Chicago, 1919 West Taylor Street, Chicago, IL, USA
| | - Krista Varady
- Department of Kinesiology and Nutrition, University of Illinois Chicago, 1919 West Taylor Street, Chicago, IL, USA
| | - Lisa Tussing-Humphreys
- Department of Kinesiology and Nutrition, University of Illinois Chicago, 1919 West Taylor Street, Chicago, IL, USA; University of Illinois Cancer Center, Chicago, IL, USA.
| |
Collapse
|
2
|
Fei L, Propato AP, Lotti G, Nardini P, Guasti D, Polvani S, Bani D, Galli A, Casini D, Cantini G, Chiaramonti D, Luconi M. Tailor-made Biochar enhances the anti-tumour effects of butyrate-glycerides in colorectal cancer. Biomed Pharmacother 2025; 184:117900. [PMID: 39921946 DOI: 10.1016/j.biopha.2025.117900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/08/2024] [Revised: 01/28/2025] [Accepted: 02/03/2025] [Indexed: 02/10/2025] Open
Abstract
Colorectal cancer (CRC) is the third most common cancer and the second cause of cancer death in the world. Emerging evidence suggests that the short-chain-fatty-acid butyrate diet-assumed or produced by gut microbiota may interfere with CRC. Novel, more focused and effective anti-cancer natural molecules selectively acting on tumour cells are required to improve patients' compliance compared to more aggressive drug-based schemes. This study explored the in vitro anti-cancer effects of a novel green compound consisting of butyrate-glycerides (BMDG) alone or absorbed on tailor-made Biochar (BMDG-Biochar) or on activated-carbon Norit-B (BMDG-Norit), by using two CRC cell lines, HCT116 and HT29. Tailor-made Biochar characterised by a larger share of meso and macroporosity compared to commercially available activated-carbon Norit-B, with micro-pored ultrastructure, displayed superior performances as a BMDG carrier, with higher absorption/release properties. BMDG, in particular when absorbed on Biochar, interfered significantly with CRC cell proliferation compared to BMDG-Norit that showed no effect. Analysis of cell metabolism revealed a superior sensitivity of HCT116 to the inhibitory effect of BMDG-Biochar. This compound specifically induced a shift from a glycolytic metabolism in particular in HCT116 cells where glycolysis supports the aggressive phenotype, towards the mitochondrial respiration that characterises the more differentiated and less aggressive HT29 cells. Biochar's ability to deliver the butyrate-glyceride bioactive mixture and to exert in vitro combined anti-cancer activity in colorectal cancer, interfering with the Warburg effect that characterises the aggressive CRC forms, opens future translational opportunities to develop new orally assumed green molecules as promising anti-cancer strategies for CRC.
Collapse
Affiliation(s)
- Laura Fei
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy; Join-Laboratory Biodelivery, University of Florence, Florence, Italy
| | - Arianna Pia Propato
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy; Join-Laboratory Biodelivery, University of Florence, Florence, Italy
| | - Giulia Lotti
- Join-Laboratory Biodelivery, University of Florence, Florence, Italy; RE-CORD, Viale Kennedy 182, 50038, Scarperia e San Piero, Florence, Italy
| | - Patrizia Nardini
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Daniele Guasti
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Simone Polvani
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy; Join-Laboratory Biodelivery, University of Florence, Florence, Italy
| | - Daniele Bani
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Andrea Galli
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy; Join-Laboratory Biodelivery, University of Florence, Florence, Italy
| | - David Casini
- Join-Laboratory Biodelivery, University of Florence, Florence, Italy; RE-CORD, Viale Kennedy 182, 50038, Scarperia e San Piero, Florence, Italy
| | - Giulia Cantini
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy; Join-Laboratory Biodelivery, University of Florence, Florence, Italy
| | - David Chiaramonti
- Join-Laboratory Biodelivery, University of Florence, Florence, Italy; RE-CORD, Viale Kennedy 182, 50038, Scarperia e San Piero, Florence, Italy; DENERG-Politecnico di Torino and RE-CORD, Corso Duca degli Abruzzi 24, Torino 10129, Italy
| | - Michaela Luconi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy; Join-Laboratory Biodelivery, University of Florence, Florence, Italy.
| |
Collapse
|
3
|
Lai X, Liu B, Wan Y, Zhou P, Li W, Hu W, Gong W. Metformin alleviates colitis-associated colorectal cancer via inhibition of the TLR4/MyD88/NFκB/MAPK pathway and macrophage M2 polarization. Int Immunopharmacol 2025; 144:113683. [PMID: 39602956 DOI: 10.1016/j.intimp.2024.113683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/09/2024] [Revised: 11/13/2024] [Accepted: 11/18/2024] [Indexed: 11/29/2024]
Abstract
BACKGROUND Colon inflammation plays an essential role in the development and progression of colorectal cancer. Emerging evidence from clinical and animal studies indicates that metformin may reduce the risk of colorectal cancer through its anti-inflammatory effects. AIMS To investigate the efficacy of metformin in reducing the risk of colorectal cancer and the possible pathways and mechanisms. METHODS The Enterotoxigenic Bacteroides Fragilis (ETBF)/azoxymethane (AOM)/dextran sulfate sodium (DSS) mouse model was established and low-dose metformin (125 mg/kg) or high-dose metformin (250 mg/kg) was administered daily by gavage. Colon tumors were counted, and colon tissue was stained with hematoxylin and eosin (HE) and Periodic Acid-Schiff's and Alcian Blue (PAS-AB). Colon Ki67, ZO-1 Muc2, Claudin-1, Occludin, MPO, reactive oxygen species (ROS), E-cadherin, CD206 and Arg-1 expression were detected by immunohistochemistry or immunofluorescence staining. NF-κB pathway-related protein expression was assessed by Western blot. Fecal short-chain fatty acid (SCFA) levels were also examined. RESULTS Our results showed that low- or high-dose metformin ameliorates colonic mucosal damage, reduces colonic inflammation, and eventually inhibits colorectal tumorigenesis in the ETBF/AOM/DSS mouse model. Our further research found that metformin suppresses the expression of TLR4/MyD88/NFκB/MAPK pathway-related proteins, modulates macrophage M2 polarization and increases SCFA levels in colon contents, which may be the mechanisms by which metformin exerts a protective effect against colon carcinogenesis. CONCLUSION Metformin inhibited colorectal tumorigenesis by suppressing the TLR4/MyD88/NFκB/MAPK pathway, modulating macrophage M2 polarization and increasing SCFA levels. It holds promise as a potentially effective treatment for colorectal cancer.
Collapse
Affiliation(s)
- Xueying Lai
- Department of Gastroenterology, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong, 510086, China; Department of Gastroenterology, The Affiliated Panyu Central Hospital of Guangzhou Medical University, Guangzhou 511400, China
| | - Bin Liu
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yu Wan
- Department of Gastroenterology, The Affiliated Panyu Central Hospital of Guangzhou Medical University, Guangzhou 511400, China
| | - Ping Zhou
- Department of Gastroenterology, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong, 510086, China
| | - Wanjun Li
- Department of Gastroenterology, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong, 510086, China
| | - Wei Hu
- Department of Gastroenterology, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong, 510086, China
| | - Wei Gong
- Department of Gastroenterology, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong, 510086, China.
| |
Collapse
|
4
|
Yang S, Wang Y, Sheng L, Cui W, Ma C. The effect of fecal bile acids on the incidence and risk-stratification of colorectal cancer: an updated systematic review and meta-analysis. Sci Rep 2025; 15:740. [PMID: 39753873 PMCID: PMC11698987 DOI: 10.1038/s41598-024-84801-6] [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] [Academic Contribution Register] [Received: 07/03/2024] [Accepted: 12/27/2024] [Indexed: 01/06/2025] Open
Abstract
Recent studies suggest the role of gut microbes in bile acid metabolism in the development and progression of colorectal cancer. However, the surveys of the association between fecal bile acid concentrations and colorectal cancer (CRC) have been inconsistent. We searched online to identify relevant cross-sectional and case-control studies published online in the major English language databases (Medline, Embase, Web of Science, AMED, and CINAHL) up to January 1, 2024. We selected studies according to inclusion and exclusion criteria and extracted data from them. RevMan 5.3 was used to perform the meta-analyses. In CRC risk meta-analysis, the effect size of CA (cholic acid), CDCA (chenodeoxycholic acid), DCA (deoxycholic acid), and UDCA (ursodeoxycholic acid) were significantly higher (CA: standardized mean difference [SMD] = 0.41, 95% confidence interval [CI]: 0.5-0.76, P = 0.02; CDCA: SMD = 0.35, 95% CI: 0.09-0.62, P = 0.009; DCA: SMD = 0.33,95% CI: 0.03-0.64, P = 0.03; UDCA: SMD = 0.46, 95% CI: 0.14-0.78, P = 0.005), and the combined effect size was significantly higher in the high-risk than the low-risk CRC group (SMD = 0.36, 95% CI: 0.21-0.51, P < 0.00001). In the CRC incidence meta-analysis, the effect sizes of CA and CDCA were significantly higher (CA: SMD = 0.42, 95% CI: 0.04-0.80, P = 0.03; CDCA: SMD = 0.61, 95% CI: 0.26-0.96, P = 0.00079), and their combined effect size was also significantly higher in the high-risk compared to low-risk CRC group (SMD = 0.39, 95% CI: 0.09-0.68, P = 0.01). Only one cross-sectional study suggested a higher concentration of CDCA, DCA, and UDCA in the stool of the CRC high-risk group than the low-risk group. These findings indicate that higher fecal concentrations of bile acid may be associated with a higher risk/incidence of CRC.
Collapse
Affiliation(s)
- Shaohui Yang
- Department of Colorectal Surgery, Ningbo Medical Center Lihuili Hospital, Ningbo, 315000, China
| | - Yu Wang
- Department of Colorectal Surgery, Ningbo Medical Center Lihuili Hospital, Ningbo, 315000, China
| | - Lijuan Sheng
- Gulou Street Community Health Service Center, Ningbo, 315000, China
| | - Wei Cui
- Department of Colorectal Surgery, Ningbo Medical Center Lihuili Hospital, Ningbo, 315000, China
| | - Chenyang Ma
- Department of Colorectal Surgery, Ningbo Medical Center Lihuili Hospital, Ningbo, 315000, China.
| |
Collapse
|
5
|
Xia D, Jin L, Wang B, Jin Y, Zheng Q, Xu J, Chen S. Alpha-glucosidase inhibitor decreases the risk of colorectal adenoma in the aged with Type 2 diabetes. Sci Rep 2025; 15:583. [PMID: 39748054 PMCID: PMC11696837 DOI: 10.1038/s41598-024-84294-3] [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] [Academic Contribution Register] [Received: 07/27/2024] [Accepted: 12/23/2024] [Indexed: 01/04/2025] Open
Abstract
The rapidly aging population is fueling a surge in diabetes, especially Type 2, which heightens colorectal cancer (CRC) risk. Colorectal adenoma, a precursor, compounds this trend. Although alpha-glucosidase inhibitors are effective hypoglycemic drugs working in the GI tract, the link between them and colorectal adenoma formation remains unexplored. A retrospective cross-sectional study was conducted on type 2 diabetes patients aged 60 and above using data from Wenzhou Central Hospital from January 2021 to May 2024. We used multivariable logistic regression and propensity score matching analysis (PSM) to calculate adjusted ORs for colorectal adenoma, controlling for potential confounders. A total of 311 subjects were enrolled in the study, with a mean age of 67.55 years. 138 (44.4%) were diagnosed with colorectal adenoma. Multivariate logistic regression analysis revealed that the AGI (Alpha-glucosidase inhibitor) Group had an adjusted OR of 0.399 (95% CI = 0.22-0.723, p = 0.002) compared to those with AGI free people. A similar trend was also observed in the PSM analysis (OR = 0.362, 95% CI = 0.176-0.744, p = 0.004). Subgroup analysis reveals hypertension as a potential modulator of the inverse relationship between AGI and colorectal adenoma occurrence post-PSM (p = 0.049). And AGI reduces serum iron levels, both before (p = 0.01) and after PSM (p = 0.028). In summary, our findings indicate that AGI significantly mitigates the risk of colorectal adenoma among individuals aged 60 and above, particularly among those afflicted with hypertension. Additionally, it substantially decreases serum iron levels.
Collapse
Affiliation(s)
- Dingchao Xia
- Department of Infectious Diseases, Wenzhou Central Hospital, Wenzhou, 325000, Zhejiang, China
- Department of Infectious Diseases, Wenzhou Sixth People's Hospital, Wenzhou, 325000, Zhejiang, China
| | - Lanling Jin
- Department of Neurology, Pujiang County People's Hospital, Wenzhou, Jinhua, 322200, Zhejiang, China
| | - Binfeng Wang
- Department of Gastroenterology, Affiliated Yueqing Hospital,Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Yi Jin
- Department of Rheumatology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Qun Zheng
- Department of Rheumatology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Jie Xu
- Department of Rheumatology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
| | - Senzhong Chen
- Department of Gerontology, Wenzhou Central Hospital, Wenzhou, 325000, Zhejiang, China.
| |
Collapse
|
6
|
Brusnic O, Onisor D, Boicean A, Hasegan A, Ichim C, Guzun A, Chicea R, Todor SB, Vintila BI, Anderco P, Porr C, Dura H, Fleaca SR, Cristian AN. Fecal Microbiota Transplantation: Insights into Colon Carcinogenesis and Immune Regulation. J Clin Med 2024; 13:6578. [PMID: 39518717 PMCID: PMC11547077 DOI: 10.3390/jcm13216578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/03/2024] [Revised: 10/24/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
Colorectal cancer (CRC) constitutes a significant global health challenge, with recent studies underscoring the pivotal role of the gut microbiome in its pathogenesis and progression. Fecal microbiota transplantation (FMT) has emerged as a compelling therapeutic approach, offering the potential to modulate microbial composition and optimize treatment outcomes. Research suggests that specific bacterial strains are closely linked to CRC, influencing both its clinical management and therapeutic interventions. Moreover, the gut microbiome's impact on immunotherapy responsiveness heralds new avenues for personalized medicine. Despite the promise of FMT, safety concerns, particularly in immunocompromised individuals, remain a critical issue. Clinical outcomes vary widely, influenced by genetic predispositions and the specific transplantation methodologies employed. Additionally, rigorous donor selection and screening protocols are paramount to minimize risks and maximize therapeutic efficacy. The current body of literature advocates for the establishment of standardized protocols and further clinical trials to substantiate FMT's role in CRC management. As our understanding of the microbiome deepens, FMT is poised to become a cornerstone in CRC treatment, underscoring the imperative for continued research and clinical validation.
Collapse
Affiliation(s)
- Olga Brusnic
- Department of Gastroenterology, University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540142 Târgu Mures, Romania; (O.B.); (D.O.)
| | - Danusia Onisor
- Department of Gastroenterology, University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540142 Târgu Mures, Romania; (O.B.); (D.O.)
| | - Adrian Boicean
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania; (A.B.); (A.H.); (A.G.); (R.C.); (B.I.V.); (P.A.); (C.P.); (H.D.); (S.R.F.); (A.N.C.)
| | - Adrian Hasegan
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania; (A.B.); (A.H.); (A.G.); (R.C.); (B.I.V.); (P.A.); (C.P.); (H.D.); (S.R.F.); (A.N.C.)
| | - Cristian Ichim
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania; (A.B.); (A.H.); (A.G.); (R.C.); (B.I.V.); (P.A.); (C.P.); (H.D.); (S.R.F.); (A.N.C.)
| | - Andreea Guzun
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania; (A.B.); (A.H.); (A.G.); (R.C.); (B.I.V.); (P.A.); (C.P.); (H.D.); (S.R.F.); (A.N.C.)
| | - Radu Chicea
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania; (A.B.); (A.H.); (A.G.); (R.C.); (B.I.V.); (P.A.); (C.P.); (H.D.); (S.R.F.); (A.N.C.)
| | - Samuel Bogdan Todor
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania; (A.B.); (A.H.); (A.G.); (R.C.); (B.I.V.); (P.A.); (C.P.); (H.D.); (S.R.F.); (A.N.C.)
| | - Bogdan Ioan Vintila
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania; (A.B.); (A.H.); (A.G.); (R.C.); (B.I.V.); (P.A.); (C.P.); (H.D.); (S.R.F.); (A.N.C.)
| | - Paula Anderco
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania; (A.B.); (A.H.); (A.G.); (R.C.); (B.I.V.); (P.A.); (C.P.); (H.D.); (S.R.F.); (A.N.C.)
| | - Corina Porr
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania; (A.B.); (A.H.); (A.G.); (R.C.); (B.I.V.); (P.A.); (C.P.); (H.D.); (S.R.F.); (A.N.C.)
| | - Horatiu Dura
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania; (A.B.); (A.H.); (A.G.); (R.C.); (B.I.V.); (P.A.); (C.P.); (H.D.); (S.R.F.); (A.N.C.)
| | - Sorin Radu Fleaca
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania; (A.B.); (A.H.); (A.G.); (R.C.); (B.I.V.); (P.A.); (C.P.); (H.D.); (S.R.F.); (A.N.C.)
| | - Adrian Nicolae Cristian
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania; (A.B.); (A.H.); (A.G.); (R.C.); (B.I.V.); (P.A.); (C.P.); (H.D.); (S.R.F.); (A.N.C.)
| |
Collapse
|
7
|
Sharma DK, Ramadass B, Callary SA, Meade A, Dash R, Clothier R, Atkins GJ, Solomon LB, Ramasamy B. The effect of prebiotic fibre on the gut microbiome and surgical outcomes in patients with prosthetic joint infection (PENGUIN) - study protocol for a randomised, double-blind, placebo-controlled trial (ACTRN12623001273673). Nutr J 2024; 23:132. [PMID: 39455990 PMCID: PMC11515416 DOI: 10.1186/s12937-024-01034-z] [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] [Academic Contribution Register] [Received: 02/13/2024] [Accepted: 10/14/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Prosthetic Joint Infection (PJI) is the most devastating complication of arthroplasty surgery and affects 1-5% of patients. Despite strict adherence to aseptic protocols and preventive measures, infection is the most common reason for revision arthroplasty, and the incidence is increasing. Treatment of PJI is challenging and often requires repeated major surgeries with sequentially poor results. The continued occurrence of PJI, and persistence after treatment, brings into question the current treatment paradigm. Preclinical evidence suggests a link between altered gut health and the risk of PJI in arthroplasty patients. Resistant starches helps to restore gut physiology by enhancing the beneficial microbiome and producing short-chain fatty acids, which have several health-conferring properties. The primary aim of this study is to investigate the effect of a commercially available prebiotic fibre formulation on the gut microbiome in PJI patients planned for a two-stage revision surgery. METHODS A double-blind placebo-controlled trial will assess the effect of 8-week supplementation of a commercially available prebiotic supplement in patients presenting with first-time PJI undergoing two-stage revision surgery. The supplementation phase will start after the first stage revision, and 80 patients will be randomised to receive either a test product (34 g of resistant starch) or a placebo (custard powder) daily for eight weeks. Stool and blood specimens will be collected at baseline, four weeks and eight weeks after the first-stage surgery and once at second-stage surgery. Gut microbiome profile, inflammatory cytokines and gut permeability biomarkers will be measured. Tissue specimens will be collected intra-operatively during first and second-stage surgeries. Baseline dietary patterns and gut symptoms will be recorded using validated questionnaires. Treatment outcomes will be reported for both cohorts using the Delphi criterion at one and two years after second-stage surgery. DISCUSSION This will be the first study to investigate the relationship between gut health optimisation and preventing PJI recurrence in arthroplasty patients. If supplementation with resistant starch improves gut health and reduces systemic inflammation, optimising the gut microbiome will be a recommended preoperative management strategy for arthroplasty patients. TRIAL REGISTRATION NO ACTRN12623001273673.
Collapse
Affiliation(s)
- Deepti K Sharma
- Department of Orthopaedics and Trauma, Royal Adelaide Hospital, Adelaide, 5000, Australia
- Centre for Orthopaedic and Trauma Research, University of Adelaide, Adelaide, 5000, Australia
| | - Balamurugan Ramadass
- Centre for Orthopaedic and Trauma Research, University of Adelaide, Adelaide, 5000, Australia
- Centre of Excellence for Clinical Microbiome Research (CCMR), All India Institute of Medical Sciences, Bhubaneswar, Odisha, India
| | - Stuart A Callary
- Department of Orthopaedics and Trauma, Royal Adelaide Hospital, Adelaide, 5000, Australia
- Centre for Orthopaedic and Trauma Research, University of Adelaide, Adelaide, 5000, Australia
| | - Anthony Meade
- Central Northern Adelaide Renal and Transplantation Service, Royal Adelaide Hospital, Adelaide, Australia
| | - Rishikesh Dash
- Centre of Excellence for Clinical Microbiome Research (CCMR), All India Institute of Medical Sciences, Bhubaneswar, Odisha, India
| | - Robyn Clothier
- Department of Orthopaedics and Trauma, Royal Adelaide Hospital, Adelaide, 5000, Australia
- Centre for Orthopaedic and Trauma Research, University of Adelaide, Adelaide, 5000, Australia
| | - Gerald J Atkins
- Centre for Orthopaedic and Trauma Research, University of Adelaide, Adelaide, 5000, Australia
| | - L Bogdan Solomon
- Department of Orthopaedics and Trauma, Royal Adelaide Hospital, Adelaide, 5000, Australia
- Centre for Orthopaedic and Trauma Research, University of Adelaide, Adelaide, 5000, Australia
| | - Boopalan Ramasamy
- Department of Orthopaedics and Trauma, Royal Adelaide Hospital, Adelaide, 5000, Australia.
- Centre for Orthopaedic and Trauma Research, University of Adelaide, Adelaide, 5000, Australia.
| |
Collapse
|
8
|
Daniel N, Farinella R, Chatziioannou AC, Jenab M, Mayén AL, Rizzato C, Belluomini F, Canzian F, Tavanti A, Keski-Rahkonen P, Hughes DJ, Campa D. Genetically predicted gut bacteria, circulating bacteria-associated metabolites and pancreatic ductal adenocarcinoma: a Mendelian randomisation study. Sci Rep 2024; 14:25144. [PMID: 39448785 PMCID: PMC11502931 DOI: 10.1038/s41598-024-77431-5] [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] [Academic Contribution Register] [Received: 08/22/2024] [Accepted: 10/22/2024] [Indexed: 10/26/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has high mortality and rising incidence rates. Recent data indicate that the gut microbiome and associated metabolites may play a role in the development of PDAC. To complement and inform observational studies, we investigated associations of genetically predicted abundances of individual gut bacteria and genetically predicted circulating concentrations of microbiome-associated metabolites with PDAC using Mendelian randomisation (MR). Gut microbiome-associated metabolites were identified through a comprehensive search of Pubmed, Exposome Explorer and Human Metabolome Database. Single Nucleotide Polymorphisms (SNPs) associated by Genome-Wide Association Studies (GWAS) with circulating levels of 109 of these metabolites were collated from Pubmed and the GWAS catalogue. SNPs for 119 taxonomically defined gut genera were selected from a meta-analysis performed by the MiBioGen consortium. Two-sample MR was conducted using GWAS summary statistics from the Pancreatic Cancer Cohort Consortium (PanScan) and the Pancreatic Cancer Case-Control Consortium (PanC4), including a total of 8,769 cases and 7,055 controls. Inverse variance-weighted MR analyses were performed along with sensitivity analyses to assess potential violations of MR assumptions. Nominally significant associations were noted for genetically predicted circulating concentrations of mannitol (odds ratio per standard deviation [ORSD] = 0.97; 95% confidence interval [CI]: 0.95-0.99, p = 0.006), methionine (ORSD= 0.97; 95%CI: 0.94-1.00, p = 0.031), stearic acid (ORSD= 0.93; 95%CI: 0.87-0.99, p = 0.027), carnitine = (ORSD=1.01; 95% CI: 1.00-1.03, p = 0.027), hippuric acid (ORSD= 1.02; 95%CI: 1.00-1.04, p = 0.038) and 3-methylhistidine (ORSD= 1.05; 95%CI: 1.01-1.10, p = 0.02). Two gut microbiome genera were associated with reduced PDAC risk; Clostridium sensu stricto 1 (OR: 0.88; 95%CI: 0.78-0.99, p = 0.027) and Romboutsia (OR: 0.87; 95%CI: 0.80-0.96, p = 0.004). These results, though based only on genetically predicted gut microbiome characteristics and circulating bacteria-related metabolite concentrations, provide evidence for causal associations with pancreatic carcinogenesis.
Collapse
Affiliation(s)
- Neil Daniel
- Molecular Epidemiology of Cancer Group, UCD Conway Institute, School of Biomedical and Biomolecular Sciences, University College Dublin, Dublin, Ireland
| | | | | | - Mazda Jenab
- Nutrition and Metabolism Branch, International Agency for Research on Cancer (IARC), Lyon, France
| | - Ana-Lucia Mayén
- Nutrition and Metabolism Branch, International Agency for Research on Cancer (IARC), Lyon, France
| | | | | | - Federico Canzian
- Genomic Epidemiology Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | - Pekka Keski-Rahkonen
- Nutrition and Metabolism Branch, International Agency for Research on Cancer (IARC), Lyon, France
| | - David J Hughes
- Molecular Epidemiology of Cancer Group, UCD Conway Institute, School of Biomedical and Biomolecular Sciences, University College Dublin, Dublin, Ireland.
| | - Daniele Campa
- Department of Biology, University of Pisa, Pisa, Italy
| |
Collapse
|
9
|
Liu M, Lu Y, Xue G, Han L, Jia H, Wang Z, Zhang J, Liu P, Yang C, Zhou Y. Role of short-chain fatty acids in host physiology. Animal Model Exp Med 2024; 7:641-652. [PMID: 38940192 PMCID: PMC11528394 DOI: 10.1002/ame2.12464] [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] [Academic Contribution Register] [Received: 04/10/2024] [Accepted: 06/08/2024] [Indexed: 06/29/2024] Open
Abstract
Short-chain fatty acids (SCFAs) are major metabolites produced by the gut microbiota through the fermentation of dietary fiber, and they have garnered significant attention due to their close association with host health. As important mediators between the gut microbiota and the host, SCFAs serve as energy substrates for intestinal epithelial cells and maintain homeostasis in host immune and energy metabolism by influencing host epigenetics, activating G protein-coupled receptors, and inhibiting pathogenic microbial infections. This review provides a comprehensive summary of SCFAs synthesis and metabolism and offering an overview of the latest research progress on their roles in protecting gut health, enhancing energy metabolism, mitigating diseases such as cancer, obesity, and diabetes, modulating the gut-brain axis and gut-lung axis, and promoting bone health.
Collapse
Affiliation(s)
- Mingyue Liu
- Stem Cell Storage Center, Hebei Reproductive Health Hospital, Hebei Women and Children's Health HospitalHebei Research Institute For Reproductive HealthShijiazhuangChina
| | - Yubo Lu
- School of Electronic Information and Electrical EngineeringShanghai Jiao Tong UniversityShanghaiChina
| | - Guoyu Xue
- Stem Cell Storage Center, Hebei Reproductive Health Hospital, Hebei Women and Children's Health HospitalHebei Research Institute For Reproductive HealthShijiazhuangChina
| | - Le Han
- Prevention Health Section, Hebei Reproductive Health Hospital, Hebei Women and Children's Health HospitalHebei Research Institute For Reproductive HealthShijiazhuangChina
| | - Hanbing Jia
- Department of Medical Imaging, Hebei Reproductive Health Hospital, Hebei Women and Children's Health HospitalHebei Research Institute For Reproductive HealthShijiazhuangChina
| | - Zi Wang
- Department of Medical Imaging, Hebei Reproductive Health Hospital, Hebei Women and Children's Health HospitalHebei Research Institute For Reproductive HealthShijiazhuangChina
| | - Jia Zhang
- Department of Obstetrical, Hebei Reproductive Health Hospital, Hebei Women and Children's Health HospitalHebei Research Institute For Reproductive HealthShijiazhuangChina
| | - Peng Liu
- Department of Clinical Laboratory, Hebei Reproductive Health Hospital, Hebei Women and Children's Health HospitalHebei Research Institute For Reproductive HealthShijiazhuangChina
| | - Chaojuan Yang
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering MedicineBeihang UniversityBeijingChina
| | - Yingjie Zhou
- Department of Obstetrics and Gynecology, Hebei Reproductive Health Hospital, Hebei Women and Children's Health HospitalHebei Research Institute For Reproductive HealthShijiazhuangChina
| |
Collapse
|
10
|
Lu S, Wang C, Ma J, Wang Y. Metabolic mediators: microbial-derived metabolites as key regulators of anti-tumor immunity, immunotherapy, and chemotherapy. Front Immunol 2024; 15:1456030. [PMID: 39351241 PMCID: PMC11439727 DOI: 10.3389/fimmu.2024.1456030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/27/2024] [Accepted: 08/27/2024] [Indexed: 10/04/2024] Open
Abstract
The human microbiome has recently emerged as a focal point in cancer research, specifically in anti-tumor immunity, immunotherapy, and chemotherapy. This review explores microbial-derived metabolites, emphasizing their crucial roles in shaping fundamental aspects of cancer treatment. Metabolites such as short-chain fatty acids (SCFAs), Trimethylamine N-Oxide (TMAO), and Tryptophan Metabolites take the spotlight, underscoring their diverse origins and functions and their profound impact on the host immune system. The focus is on SCFAs' remarkable ability to modulate immune responses, reduce inflammation, and enhance anti-tumor immunity within the intricate tumor microenvironment (TME). The review critically evaluates TMAO, intricately tied to dietary choices and gut microbiota composition, assessing its implications for cancer susceptibility, progression, and immunosuppression. Additionally, the involvement of tryptophan and other amino acid metabolites in shaping immune responses is discussed, highlighting their influence on immune checkpoints, immunosuppression, and immunotherapy effectiveness. The examination extends to their dynamic interaction with chemotherapy, emphasizing the potential of microbial-derived metabolites to alter treatment protocols and optimize outcomes for cancer patients. A comprehensive understanding of their role in cancer therapy is attained by exploring their impacts on drug metabolism, therapeutic responses, and resistance development. In conclusion, this review underscores the pivotal contributions of microbial-derived metabolites in regulating anti-tumor immunity, immunotherapy responses, and chemotherapy outcomes. By illuminating the intricate interactions between these metabolites and cancer therapy, the article enhances our understanding of cancer biology, paving the way for the development of more effective treatment options in the ongoing battle against cancer.
Collapse
Affiliation(s)
- Shan Lu
- Department of General Practice, The Second Hospital of Jilin University, Changchun, China
| | - Chunling Wang
- Medical Affairs Department, The Second Hospital of Jilin University, Changchun, China
| | - Jingru Ma
- Department of Clinical Laboratory, the Second Hospital of Jilin University, Changchun, China
| | - Yichao Wang
- Department of Obstetrics and Gynecology, the Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
11
|
Shieh C, Thompson HJ, McLaughlin E, Chiang CW, Hussan H. Advancements in Understanding and Preventing Obesity-Related Colon Cancer. Cancer J 2024; 30:357-369. [PMID: 39312456 DOI: 10.1097/ppo.0000000000000744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 09/25/2024]
Abstract
ABSTRACT Obesity and colorectal cancer are global public health issues, with the prevalence of both conditions increasing over the last 4 decades. In the United States alone, the prevalence of obesity is greater than 40%, and this percentage is projected to increase past 50% by 2030. This review focuses on understanding the association between obesity and the risk of colorectal cancer while also highlighting hypotheses about molecular mechanisms underlying the link between these disease processes. We also consider whether those linkages can be disrupted via weight loss therapies, including lifestyle modifications, pharmacotherapy, bariatric surgery, and endobariatrics.
Collapse
Affiliation(s)
- Christine Shieh
- From the Department of Gastroenterology, University of California, Davis, Sacramento, CA
| | - Henry J Thompson
- Cancer Prevention Laboratory, Colorado State University, Fort Collins, CO
| | | | - Chien-Wei Chiang
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, OH
| | | |
Collapse
|
12
|
Maukonen M, Koponen KK, Havulinna AS, Kaartinen NE, Niiranen T, Méric G, Pajari AM, Knight R, Salomaa V, Männistö S. Associations of plant-based foods, red and processed meat, and dairy with gut microbiome in Finnish adults. Eur J Nutr 2024; 63:2247-2260. [PMID: 38753173 PMCID: PMC11377619 DOI: 10.1007/s00394-024-03406-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/28/2023] [Accepted: 04/16/2024] [Indexed: 09/06/2024]
Abstract
PURPOSE Population-based studies on the associations of plant-based foods, red meat or dairy with gut microbiome are scarce. We examined whether the consumption of plant-based foods (vegetables, potatoes, fruits, cereals), red and processed meat (RPM) or dairy (fermented milk, cheese, other dairy products) are related to gut microbiome in Finnish adults. METHODS We utilized data from the National FINRISK/FINDIET 2002 Study (n = 1273, aged 25-64 years, 55% women). Diet was assessed with 48-hour dietary recalls. Gut microbiome was analyzed using shallow shotgun sequencing. We applied multivariate analyses with linear models and permutational ANOVAs adjusted for relevant confounders. RESULTS Fruit consumption was positively (beta = 0.03, SE = 0.01, P = 0.04), while a dairy subgroup including milk, cream and ice-creams was inversely associated (beta=-0.03, SE 0.01, P = 0.02) with intra-individual gut microbiome diversity (alpha-diversity). Plant-based foods (R2 = 0.001, P = 0.03) and dairy (R2 = 0.002, P = 0.01) but not RPM (R2 = 0.001, P = 0.38) contributed to the compositional differences in gut microbiome (beta-diversity). Plant-based foods were associated with several butyrate producers/cellulolytic species including Roseburia hominis. RPM associations included an inverse association with R. hominis. Dairy was positively associated with several lactic producing/probiotic species including Lactobacillus delbrueckii and potentially opportunistic pathogens including Citrobacter freundii. Dairy, fermented milk, vegetables, and cereals were associated with specific microbial functions. CONCLUSION Our results suggest a potential association between plant-based foods and dairy or their subgroups with microbial diversity measures. Furthermore, our findings indicated that all the food groups were associated with distinct overall microbial community compositions. Plant-based food consumption particularly was associated with a larger number of putative beneficial species.
Collapse
Affiliation(s)
- Mirkka Maukonen
- Finnish Institute for Health and Welfare (THL), Helsinki, Finland.
| | - Kari K Koponen
- Finnish Institute for Health and Welfare (THL), Helsinki, Finland
| | - Aki S Havulinna
- Finnish Institute for Health and Welfare (THL), Helsinki, Finland
- Institute for Molecular Medicine Finland, FIMM-HiLIFE, Helsinki, Finland
| | | | - Teemu Niiranen
- Finnish Institute for Health and Welfare (THL), Helsinki, Finland
| | - Guillaume Méric
- Baker Heart and Diabetes Institute, Melbourne, Australia
- University of Melbourne, Melbourne, Australia
- Monash University, Melbourne, Australia
- La Trobe University, Melbourne, Australia
| | | | - Rob Knight
- University of California San Diego, La Jolla, CA, USA
| | - Veikko Salomaa
- Finnish Institute for Health and Welfare (THL), Helsinki, Finland
| | - Satu Männistö
- Finnish Institute for Health and Welfare (THL), Helsinki, Finland
| |
Collapse
|
13
|
Hamamah S, Lobiuc A, Covasa M. Antioxidant Role of Probiotics in Inflammation-Induced Colorectal Cancer. Int J Mol Sci 2024; 25:9026. [PMID: 39201713 PMCID: PMC11354872 DOI: 10.3390/ijms25169026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/19/2024] [Revised: 08/15/2024] [Accepted: 08/16/2024] [Indexed: 09/03/2024] Open
Abstract
Colorectal cancer (CRC) continues to be a significant contributor to global morbidity and mortality. Emerging evidence indicates that disturbances in gut microbial composition, the formation of reactive oxygen species (ROS), and the resulting inflammation can lead to DNA damage, driving the pathogenesis and progression of CRC. Notably, bacterial metabolites can either protect against or contribute to oxidative stress by modulating the activity of antioxidant enzymes and influencing signaling pathways that govern ROS-induced inflammation. Additionally, microbiota byproducts, when supplemented through probiotics, can affect tumor microenvironments to enhance treatment efficacy and selectively mediate the ROS-induced destruction of CRC cells. This review aims to discuss the mechanisms by which taxonomical shifts in gut microbiota and related metabolites such as short-chain fatty acids, secondary bile acids, and trimethylamine-N-oxide influence ROS concentrations to safeguard or promote the onset of inflammation-mediated CRC. Additionally, we focus on the role of probiotic species in modulating ROS-mediated signaling pathways that influence both oxidative status and inflammation, such as Nrf2-Keap1, NF-κB, and NLRP3 to mitigate carcinogenesis. Overall, a deeper understanding of the role of gut microbiota on oxidative stress may aid in delaying or preventing the onset of CRC and offer new avenues for adjunct, CRC-specific therapeutic interventions such as cancer immunotherapy.
Collapse
Affiliation(s)
- Sevag Hamamah
- Department of Basic Medical Sciences, College of Osteopathic Medicine, Western University of Health Sciences, Pomona, CA 91766, USA;
- Department of Internal Medicine, Scripps Mercy Hospital, San Diego, CA 92103, USA
| | - Andrei Lobiuc
- Department of Medicine and Biomedical Sciences, College of Medicine and Biological Science, University of Suceava, 7200229 Suceava, Romania;
| | - Mihai Covasa
- Department of Basic Medical Sciences, College of Osteopathic Medicine, Western University of Health Sciences, Pomona, CA 91766, USA;
- Department of Medicine and Biomedical Sciences, College of Medicine and Biological Science, University of Suceava, 7200229 Suceava, Romania;
| |
Collapse
|
14
|
Balendra V, Rosenfeld R, Amoroso C, Castagnone C, Rossino MG, Garrone O, Ghidini M. Postbiotics as Adjuvant Therapy in Cancer Care. Nutrients 2024; 16:2400. [PMID: 39125280 PMCID: PMC11314502 DOI: 10.3390/nu16152400] [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] [Academic Contribution Register] [Received: 06/04/2024] [Revised: 07/14/2024] [Accepted: 07/18/2024] [Indexed: 08/12/2024] Open
Abstract
Postbiotics are defined as a preparation of inanimate microorganisms and/or their components that confers a health benefit to the host. They range from cell wall fragments to metabolites, bacterial lysates, extracellular vesicles, and short-chain fatty acids (SCFAs). Postbiotics may influence carcinogenesis via a variety of mechanisms. They can promote homeostatic immune responses, reduce inflammation, induce selective cytotoxicity against tumor cells, as well as the enabling the control of tumor cell proliferation and enhancing intestinal epithelial barrier function. Therefore, probiotics can serve as an adjunct strategy in anticancer treatment together with chemotherapy and immunotherapy. Up to now, the only relevant postbiotics used as interventions in oncological patients remain vitamin K molecules, with few phase-II and III trials available. In fact, postbiotics' levels are strictly dependent on the gut microbiota's composition, which may vary between individuals and can be altered under different physiological and pathological conditions. Therefore, the lack of consistent clinical evidence supporting postbiotics' efficacy is due to their poor bioavailability, short half-life, and fluctuating levels. Synbiotics, a mixture of prebiotics and probiotics, are expected to have a more homogeneous bioavailability with respect to postbiotics and may have greater potential for future development. In this review, we focus on the role of postbiotics as an adjuvant therapy in cancer treatment.
Collapse
Affiliation(s)
| | - Roberto Rosenfeld
- Oncology Unit, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (R.R.); (M.G.R.); (O.G.)
| | - Chiara Amoroso
- Gastroenterology and Endoscopy Unit, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy;
| | | | - Maria Grazia Rossino
- Oncology Unit, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (R.R.); (M.G.R.); (O.G.)
| | - Ornella Garrone
- Oncology Unit, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (R.R.); (M.G.R.); (O.G.)
| | - Michele Ghidini
- Oncology Unit, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (R.R.); (M.G.R.); (O.G.)
| |
Collapse
|
15
|
Duan J, Du P, Jiang T, Ma X, Sun J, Liang J, Wang J, Liu C. Exploring the mechanism of enterotoxicity mediated by the microbiome-butyrate-PPAR axis in podophyllotoxin through the toxicological evidence chain (TEC) concept. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 280:116548. [PMID: 38850705 DOI: 10.1016/j.ecoenv.2024.116548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 02/21/2024] [Revised: 05/25/2024] [Accepted: 06/02/2024] [Indexed: 06/10/2024]
Abstract
Podophyllotoxin (PPT) is a lignan derived from the roots and stems of the Podophyllum plant. However, its enterotoxicity restricts its clinical application. The underlying mechanisms by which PPT exerts its action remain largely elusive. This study aimed to evaluate the molecular mechanisms underlying PPT-induced enterotoxicity utilizing the concept of toxicological evidence chain. Changes in body weight, behavior, and histopathological and biochemical markers in rats were observed. Additionally, microbiome, metabolome, and transcriptome analyses were integrated to identify potential microorganisms, metabolic markers, and major pathways using a co-occurrence network. Our findings suggested that PPT induced pathological changes in rats, including weight loss, diarrhea, and inflammation accompanied by increased levels of IFN-γ, IL-5, IL-6, GRO/KC, and IL-12p70. The decrease in butyrate levels in the PPT group may be related to the enrichment of Firmicutes. The reduction of butyrate levels may impair the expression of PPARγ, subsequently promoting Escherichia-Shigella proliferation. Additionally, the suppression of PPARs pathway may result in the increased production of inflammatory factors, contributing to enterotoxicity. This study offers a novel understanding of the molecular mechanisms underlying PPT-induced enterotoxicity, making a significant contribution to developing strategies to mitigate PPT toxicity and prevent associated diseases.
Collapse
Affiliation(s)
- Jiajia Duan
- Department of Clinical Laboratory, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang 471003, China.
| | - Peipei Du
- Department of Clinical Laboratory, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang 471003, China.
| | - Tao Jiang
- Department of Clinical Laboratory, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang 471003, China.
| | - Xiao Ma
- Department of Clinical Laboratory, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang 471003, China.
| | - Jiaxing Sun
- Department of Clinical Laboratory, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang 471003, China.
| | - Jin Liang
- Department of Clinical Laboratory, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang 471003, China.
| | - Jingjing Wang
- Department of Clinical Laboratory, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang 471003, China.
| | - Chuanxin Liu
- Luoyang Key Laboratory of Clinical Multiomics and Translational Medicine, Henan Key Laboratory of Rare Diseases, Endocrinology and Metabolism Center, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang 471003, China.
| |
Collapse
|
16
|
Oliero M, Cuisiniere T, Ajayi AS, Gerkins C, Hajjar R, Fragoso G, Calvé A, Vennin Rendos H, Mathieu-Denoncourt A, Dagbert F, De Broux É, Loungnarath R, Schwenter F, Sebajang H, Ratelle R, Wassef R, Richard C, Duperthuy M, Gravel AE, Vincent AT, Santos MM. Putrescine Supplementation Limits the Expansion of pks+ Escherichia coli and Tumor Development in the Colon. CANCER RESEARCH COMMUNICATIONS 2024; 4:1777-1792. [PMID: 38934090 PMCID: PMC11261243 DOI: 10.1158/2767-9764.crc-23-0355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 10/05/2023] [Revised: 04/11/2024] [Accepted: 06/24/2024] [Indexed: 06/28/2024]
Abstract
Escherichia coli that harbor the polyketide synthase (pks) genomic island produce colibactin and are associated with sporadic colorectal cancer development. Given the considerable prevalence of pks+ bacteria in healthy individuals, we sought to identify strategies to limit the growth and expansion of pks+ E. coli. We found that culture supernatants of the probiotic strain E. coli Nissle 1917 were able to inhibit the growth of the murine pathogenic strain pks+ E. coli NC101 (EcNC101). We performed a nontargeted analysis of the metabolome in supernatants from several E. coli strains and identified putrescine as a potential postbiotic capable of suppressing EcNC101 growth in vitro. The effect of putrescine supplementation was then evaluated in the azoxymethane/dextran sulfate sodium mouse model of colorectal cancer in mice colonized with EcNC101. Putrescine supplementation inhibited the growth of pks+ E. coli, reduced the number and size of colonic tumors, and downmodulated the release of inflammatory cytokines in the colonic lumen. Additionally, putrescine supplementation led to shifts in the composition and function of gut microbiota, characterized by an increase in the Firmicutes/Bacteroidetes ratio and enhanced acetate production. The effect of putrescine was further confirmed in vitro using a pks+ E. coli strain isolated from a patient with colorectal cancer. These results suggest that probiotic-derived metabolites can be used as an alternative to live bacteria in individuals at risk of developing colorectal cancer due to the presence of pks+ bacteria in their colon. SIGNIFICANCE Putrescine supplementation inhibits the growth of cancer-promoting bacteria in the gut, lowers inflammation, and reduces colon cancer development. The consumption of healthy foods rich in putrescine may be a potential prophylactic approach for individuals at risk of developing colorectal cancer due to the presence of pks+ bacteria in their colon.
Collapse
Affiliation(s)
- Manon Oliero
- Nutrition and Microbiome Laboratory, Institut du cancer de Montréal, Centre de recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, Canada.
| | - Thibault Cuisiniere
- Nutrition and Microbiome Laboratory, Institut du cancer de Montréal, Centre de recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, Canada.
| | - Ayodeji S. Ajayi
- Nutrition and Microbiome Laboratory, Institut du cancer de Montréal, Centre de recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, Canada.
| | - Claire Gerkins
- Nutrition and Microbiome Laboratory, Institut du cancer de Montréal, Centre de recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, Canada.
| | - Roy Hajjar
- Nutrition and Microbiome Laboratory, Institut du cancer de Montréal, Centre de recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, Canada.
- Department of Surgery, Faculty of Medicine, Université de Montréal, Montréal, Canada.
| | - Gabriela Fragoso
- Nutrition and Microbiome Laboratory, Institut du cancer de Montréal, Centre de recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, Canada.
| | - Annie Calvé
- Nutrition and Microbiome Laboratory, Institut du cancer de Montréal, Centre de recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, Canada.
| | - Hervé Vennin Rendos
- Nutrition and Microbiome Laboratory, Institut du cancer de Montréal, Centre de recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, Canada.
| | - Annabelle Mathieu-Denoncourt
- Department of Microbiology, Infectiology and Immunology, Faculty of Medicine, Université de Montréal, Montréal, Canada.
| | - François Dagbert
- Digestive Surgery Service, Department of Surgery, Centre hospitalier de l’Université de Montréal (CHUM), Montréal, Canada.
| | - Éric De Broux
- Digestive Surgery Service, Department of Surgery, Centre hospitalier de l’Université de Montréal (CHUM), Montréal, Canada.
| | - Rasmy Loungnarath
- Digestive Surgery Service, Department of Surgery, Centre hospitalier de l’Université de Montréal (CHUM), Montréal, Canada.
| | - Frank Schwenter
- Digestive Surgery Service, Department of Surgery, Centre hospitalier de l’Université de Montréal (CHUM), Montréal, Canada.
| | - Herawaty Sebajang
- Digestive Surgery Service, Department of Surgery, Centre hospitalier de l’Université de Montréal (CHUM), Montréal, Canada.
| | - Richard Ratelle
- Digestive Surgery Service, Department of Surgery, Centre hospitalier de l’Université de Montréal (CHUM), Montréal, Canada.
| | - Ramses Wassef
- Digestive Surgery Service, Department of Surgery, Centre hospitalier de l’Université de Montréal (CHUM), Montréal, Canada.
| | - Carole Richard
- Department of Surgery, Faculty of Medicine, Université de Montréal, Montréal, Canada.
- Digestive Surgery Service, Department of Surgery, Centre hospitalier de l’Université de Montréal (CHUM), Montréal, Canada.
| | - Marylise Duperthuy
- Department of Microbiology, Infectiology and Immunology, Faculty of Medicine, Université de Montréal, Montréal, Canada.
| | - Andrée E. Gravel
- Drug Discovery Platform, Research Institute McGill University Health Centre, Montreal, Canada.
| | - Antony T. Vincent
- Département des sciences animales, Faculté des sciences de l’agriculture et de l’alimentation, Université Laval, Quebec City, Canada.
- Institut de biologie intégrative et des systèmes, Université Laval, Quebec City, Canada.
| | - Manuela M. Santos
- Nutrition and Microbiome Laboratory, Institut du cancer de Montréal, Centre de recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, Canada.
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montréal, Canada.
| |
Collapse
|
17
|
Leung HKM, Lo EKK, Zhang F, Felicianna, Ismaiah MJ, Chen C, El-Nezami H. Modulation of Gut Microbial Biomarkers and Metabolites in Cancer Management by Tea Compounds. Int J Mol Sci 2024; 25:6348. [PMID: 38928054 PMCID: PMC11203446 DOI: 10.3390/ijms25126348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/29/2024] [Revised: 05/24/2024] [Accepted: 05/27/2024] [Indexed: 06/28/2024] Open
Abstract
Cancers are causing millions of deaths and leaving a huge clinical and economic burden. High costs of cancer drugs are limiting their access to the growing number of cancer cases. The development of more affordable alternative therapy could reach more patients. As gut microbiota plays a significant role in the development and treatment of cancer, microbiome-targeted therapy has gained more attention in recent years. Dietary and natural compounds can modulate gut microbiota composition while providing broader and more accessible access to medicine. Tea compounds have been shown to have anti-cancer properties as well as modulate the gut microbiota and their related metabolites. However, there is no comprehensive review that focuses on the gut modulatory effects of tea compounds and their impact on reshaping the metabolic profiles, particularly in cancer models. In this review, the effects of different tea compounds on gut microbiota in cancer settings are discussed. Furthermore, the relationship between these modulated bacteria and their related metabolites, along with the mechanisms of how these changes led to cancer intervention are summarized.
Collapse
Affiliation(s)
- Hoi Kit Matthew Leung
- School of Biological Sciences, University of Hong Kong, Pokfulam, Hong Kong SAR 999077, China; (H.K.M.L.); (E.K.K.L.); (F.Z.); (F.); (M.J.I.); (C.C.)
| | - Emily Kwun Kwan Lo
- School of Biological Sciences, University of Hong Kong, Pokfulam, Hong Kong SAR 999077, China; (H.K.M.L.); (E.K.K.L.); (F.Z.); (F.); (M.J.I.); (C.C.)
| | - Fangfei Zhang
- School of Biological Sciences, University of Hong Kong, Pokfulam, Hong Kong SAR 999077, China; (H.K.M.L.); (E.K.K.L.); (F.Z.); (F.); (M.J.I.); (C.C.)
| | - Felicianna
- School of Biological Sciences, University of Hong Kong, Pokfulam, Hong Kong SAR 999077, China; (H.K.M.L.); (E.K.K.L.); (F.Z.); (F.); (M.J.I.); (C.C.)
| | - Marsena Jasiel Ismaiah
- School of Biological Sciences, University of Hong Kong, Pokfulam, Hong Kong SAR 999077, China; (H.K.M.L.); (E.K.K.L.); (F.Z.); (F.); (M.J.I.); (C.C.)
| | - Congjia Chen
- School of Biological Sciences, University of Hong Kong, Pokfulam, Hong Kong SAR 999077, China; (H.K.M.L.); (E.K.K.L.); (F.Z.); (F.); (M.J.I.); (C.C.)
| | - Hani El-Nezami
- School of Biological Sciences, University of Hong Kong, Pokfulam, Hong Kong SAR 999077, China; (H.K.M.L.); (E.K.K.L.); (F.Z.); (F.); (M.J.I.); (C.C.)
- Institute of Public Health and Clinical Nutrition, School of Medicine, University of Eastern Finland, FI-70211 Kuopio, Finland
| |
Collapse
|
18
|
Kuru-Yaşar R, Üstün-Aytekin Ö. The Crucial Roles of Diet, Microbiota, and Postbiotics in Colorectal Cancer. Curr Nutr Rep 2024; 13:126-151. [PMID: 38483752 PMCID: PMC11133122 DOI: 10.1007/s13668-024-00525-z] [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] [Academic Contribution Register] [Accepted: 02/25/2024] [Indexed: 05/30/2024]
Abstract
PURPOSE OF REVIEW Colorectal cancer is the second deadliest cancer in the world, and its prevalence has been increasing alarmingly in recent years. After researchers discovered the existence of dysbiosis in colorectal cancer, they considered the use of probiotics in the treatment of colorectal cancer. However, for various reasons, including the low safety profile of probiotics in susceptible and immunocompromised patient5s, and the risk of developing antibiotic resistance, researchers have shifted their focus to non-living cells, their components, and metabolites. This review aims to comprehensively evaluate the literature on the effects of diet, microbiota, and postbiotics on colorectal cancer and the future of postbiotics. RECENT FINDINGS The link between diet, gut microbiota, and colorectal cancer has been established primarily as a relationship rather than a cause-effect relationship. The gut microbiota can convert gastrointestinal tract and dietary factors into either onco-metabolites or tumor suppressor metabolites. There is serious dysbiosis in the microbiota in colorectal cancer. Postbiotics appear to be promising agents in the prevention and treatment of colorectal cancer. It has been shown that various postbiotics can selectively induce apoptosis in CRC, inhibit cell proliferation, growth, invasion, and migration, modulate the immune system, suppress carcinogenic signaling pathways, maintain intestinal epithelial integrity, and have a synergistic effect with chemotherapy drugs. However, it is also reported that some postbiotics are ineffective and may be risky in terms of safety profile in some patients. Many issues need to be researched about postbiotics. Large-scale, randomized, double-blind clinical studies are needed.
Collapse
Affiliation(s)
- Rüya Kuru-Yaşar
- Department of Nutrition and Dietetics, Hamidiye Faculty of Health Sciences, University of Health Sciences, 34668, Istanbul, Türkiye
| | - Özlem Üstün-Aytekin
- Department of Nutrition and Dietetics, Hamidiye Faculty of Health Sciences, University of Health Sciences, 34668, Istanbul, Türkiye.
| |
Collapse
|
19
|
Papadimitriou N, Kim A, Kawaguchi ES, Morrison J, Diez-Obrero V, Albanes D, Berndt SI, Bézieau S, Bien SA, Bishop DT, Bouras E, Brenner H, Buchanan DD, Campbell PT, Carreras-Torres R, Chan AT, Chang-Claude J, Conti DV, Devall MA, Dimou N, Drew DA, Gruber SB, Harrison TA, Hoffmeister M, Huyghe JR, Joshi AD, Keku TO, Kundaje A, Küry S, Le Marchand L, Lewinger JP, Li L, Lynch BM, Moreno V, Newton CC, Obón-Santacana M, Ose J, Pellatt AJ, Peoples AR, Platz EA, Qu C, Rennert G, Ruiz-Narvaez E, Shcherbina A, Stern MC, Su YR, Thomas DC, Thomas CE, Tian Y, Tsilidis KK, Ulrich CM, Um CY, Visvanathan K, Wang J, White E, Woods MO, Schmit SL, Macrae F, Potter JD, Hopper JL, Peters U, Murphy N, Hsu L, Gunter MJ, Gauderman WJ. Genome-wide interaction study of dietary intake of fibre, fruits, and vegetables with risk of colorectal cancer. EBioMedicine 2024; 104:105146. [PMID: 38749303 PMCID: PMC11112268 DOI: 10.1016/j.ebiom.2024.105146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/11/2023] [Revised: 04/15/2024] [Accepted: 04/19/2024] [Indexed: 05/26/2024] Open
Abstract
BACKGROUND Consumption of fibre, fruits and vegetables have been linked with lower colorectal cancer (CRC) risk. A genome-wide gene-environment (G × E) analysis was performed to test whether genetic variants modify these associations. METHODS A pooled sample of 45 studies including up to 69,734 participants (cases: 29,896; controls: 39,838) of European ancestry were included. To identify G × E interactions, we used the traditional 1--degree-of-freedom (DF) G × E test and to improve power a 2-step procedure and a 3DF joint test that investigates the association between a genetic variant and dietary exposure, CRC risk and G × E interaction simultaneously. FINDINGS The 3-DF joint test revealed two significant loci with p-value <5 × 10-8. Rs4730274 close to the SLC26A3 gene showed an association with fibre (p-value: 2.4 × 10-3) and G × fibre interaction with CRC (OR per quartile of fibre increase = 0.87, 0.80, and 0.75 for CC, TC, and TT genotype, respectively; G × E p-value: 1.8 × 10-7). Rs1620977 in the NEGR1 gene showed an association with fruit intake (p-value: 1.0 × 10-8) and G × fruit interaction with CRC (OR per quartile of fruit increase = 0.75, 0.65, and 0.56 for AA, AG, and GG genotype, respectively; G × E -p-value: 0.029). INTERPRETATION We identified 2 loci associated with fibre and fruit intake that also modify the association of these dietary factors with CRC risk. Potential mechanisms include chronic inflammatory intestinal disorders, and gut function. However, further studies are needed for mechanistic validation and replication of findings. FUNDING National Institutes of Health, National Cancer Institute. Full funding details for the individual consortia are provided in acknowledgments.
Collapse
Affiliation(s)
- Nikos Papadimitriou
- Nutrition and Metabolism Branch, International Agency for Research on Cancer, Lyon, France
| | - Andre Kim
- Division of Biostatistics, Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Eric S Kawaguchi
- Division of Biostatistics, Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - John Morrison
- Division of Biostatistics, Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Virginia Diez-Obrero
- Unit of Biomarkers and Susceptibility, Oncology Data Analytics Program, Catalan Institute of Oncology, Barcelona, 08908, Spain; Colorectal Cancer Group, ONCOBELL Program, Bellvitge Biomedical Research Institute, Barcelona, 08908, Spain; Consortium for Biomedical Research in Epidemiology and Public Health, Barcelona, 08908, Spain; Department of Clinical Sciences, Faculty of Medicine and Health Sciences and Universitat de Barcelona Institute of Complex Systems (UBICS), University of Barcelona (UB), L'Hospitalet de Llobregat, 08908, Barcelona, Spain
| | - Demetrius Albanes
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Sonja I Berndt
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Stéphane Bézieau
- Service de Génétique Médicale, Centre Hospitalier Universitaire (CHU) Nantes, Nantes, France
| | - Stephanie A Bien
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - D Timothy Bishop
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, UK
| | - Emmanouil Bouras
- Department of Hygiene and Epidemiology, University of Ioannina School of Medicine, Ioannina, Greece
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany; Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumour Diseases (NCT), Heidelberg, Germany; German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Daniel D Buchanan
- Colorectal Oncogenomics Group, Department of Clinical Pathology, Melbourne Medical School, The University of Melbourne, Parkville, Australia; University of Melbourne Centre for Cancer Research, The University of Melbourne, Parkville, Australia; Genomic Medicine and Family Cancer Clinic, The Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Peter T Campbell
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Robert Carreras-Torres
- Colorectal Cancer Group, ONCOBELL Program, Bellvitge Biomedical Research Institute, Barcelona, 08908, Spain; Digestive Diseases and Microbiota Group, Girona Biomedical Research Institute (IDIBGI), 17190 Salt, Girona, Spain
| | - Andrew T Chan
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA; Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Broad Institute of Harvard and MIT, Cambridge, MA, USA; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA, USA; Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
| | - Jenny Chang-Claude
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany; University Medical Centre Hamburg-Eppendorf, University Cancer Centre Hamburg (UCCH), Hamburg, Germany
| | - David V Conti
- Division of Biostatistics, Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Matthew A Devall
- Center for Public Health Genomics, Department of Public Health Sciences, University of Virginia, Charlottesville, VA, USA; Department of Public Health Sciences, Center for Public Health Genomics, Charlottesville, VA, USA
| | - Niki Dimou
- Nutrition and Metabolism Branch, International Agency for Research on Cancer, Lyon, France
| | - David A Drew
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Stephen B Gruber
- Department of Medical Oncology & Therapeutics Research and Center for Precision Medicine, City of Hope National Medical Center, Duarte, CA, USA
| | - Tabitha A Harrison
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Michael Hoffmeister
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jeroen R Huyghe
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Amit D Joshi
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
| | - Temitope O Keku
- Center for Gastrointestinal Biology and Disease, University of North Carolina, Chapel Hill, NC, USA
| | - Anshul Kundaje
- Department of Genetics, Stanford University, Stanford, CA, USA; Department of Computer Science, Stanford University, Stanford, CA, USA
| | - Sébastien Küry
- Service de Génétique Médicale, Centre Hospitalier Universitaire (CHU) Nantes, Nantes, France
| | | | - Juan Pablo Lewinger
- Division of Biostatistics, Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Li Li
- Department of Family Medicine, University of Virginia, Charlottesville, VA, USA
| | - Brigid M Lynch
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Victoria, Australia; Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, Victoria, Australia
| | - Victor Moreno
- Unit of Biomarkers and Susceptibility, Oncology Data Analytics Program, Catalan Institute of Oncology, Barcelona, 08908, Spain; Colorectal Cancer Group, ONCOBELL Program, Bellvitge Biomedical Research Institute, Barcelona, 08908, Spain; Consortium for Biomedical Research in Epidemiology and Public Health, Barcelona, 08908, Spain; Department of Clinical Sciences, Faculty of Medicine and Health Sciences and Universitat de Barcelona Institute of Complex Systems (UBICS), University of Barcelona (UB), L'Hospitalet de Llobregat, 08908, Barcelona, Spain
| | - Christina C Newton
- Department of Population Science, American Cancer Society, Atlanta, GA, USA
| | - Mireia Obón-Santacana
- Unit of Biomarkers and Susceptibility, Oncology Data Analytics Program, Catalan Institute of Oncology, Barcelona, 08908, Spain; Colorectal Cancer Group, ONCOBELL Program, Bellvitge Biomedical Research Institute, Barcelona, 08908, Spain; Consortium for Biomedical Research in Epidemiology and Public Health, Barcelona, 08908, Spain
| | - Jennifer Ose
- Huntsman Cancer Institute, Salt Lake City, UT, USA; Department of Population Health Sciences, University of Utah, Salt Lake City, UT, USA
| | - Andrew J Pellatt
- Department of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Anita R Peoples
- Huntsman Cancer Institute, Salt Lake City, UT, USA; Department of Population Health Sciences, University of Utah, Salt Lake City, UT, USA
| | - Elizabeth A Platz
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Conghui Qu
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Gad Rennert
- Department of Community Medicine and Epidemiology, Lady Davis Carmel Medical Center, Haifa, Israel; Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel; Clalit National Cancer Control Center, Haifa, Israel
| | - Edward Ruiz-Narvaez
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Anna Shcherbina
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Mariana C Stern
- Division of Biostatistics, Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Yu-Ru Su
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Duncan C Thomas
- Division of Biostatistics, Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Claire E Thomas
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Yu Tian
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany; School of Public Health, Capital Medical University, Beijing, China
| | - Konstantinos K Tsilidis
- Department of Hygiene and Epidemiology, University of Ioannina School of Medicine, Ioannina, Greece; Department of Epidemiology and Biostatistics, Imperial College London, School of Public Health, London, UK
| | - Cornelia M Ulrich
- Huntsman Cancer Institute, Salt Lake City, UT, USA; Department of Population Health Sciences, University of Utah, Salt Lake City, UT, USA
| | - Caroline Y Um
- Department of Population Science, American Cancer Society, Atlanta, GA, USA
| | - Kala Visvanathan
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Jun Wang
- Division of Biostatistics, Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA; Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Emily White
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA; Department of Epidemiology, University of Washington School of Public Health, Seattle, WA, USA
| | - Michael O Woods
- Memorial University of Newfoundland, Discipline of Genetics, St. John's, Canada
| | - Stephanie L Schmit
- Genomic Medicine Institute, Cleveland Clinic, Cleveland, OH, USA; Population and Cancer Prevention Program, Case Comprehensive Cancer Center, Cleveland, OH, USA
| | - Finlay Macrae
- The Royal Melbourne Hospital, Melbourne, Victoria, Australia
| | - John D Potter
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - John L Hopper
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Victoria, Australia; Department of Epidemiology, School of Public Health and Institute of Health and Environment, Seoul National University, Seoul, South Korea
| | - Ulrike Peters
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA; Department of Epidemiology, University of Washington School of Public Health, Seattle, WA, USA.
| | - Neil Murphy
- Nutrition and Metabolism Branch, International Agency for Research on Cancer, Lyon, France
| | - Li Hsu
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA; Department of Biostatistics, University of Washington, Seattle, WA, USA.
| | - Marc J Gunter
- Nutrition and Metabolism Branch, International Agency for Research on Cancer, Lyon, France; Department of Epidemiology and Biostatistics, Imperial College London, School of Public Health, London, UK.
| | - W James Gauderman
- Division of Biostatistics, Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
20
|
Soldado-Gordillo A, Álvarez-Mercado AI. Epigenetics, Microbiota, and Breast Cancer: A Systematic Review. Life (Basel) 2024; 14:705. [PMID: 38929688 PMCID: PMC11204553 DOI: 10.3390/life14060705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/05/2024] [Revised: 05/14/2024] [Accepted: 05/28/2024] [Indexed: 06/28/2024] Open
Abstract
Breast cancer is the most frequently diagnosed cancer in women worldwide. According to recent studies, alterations in the microbiota and epigenetic modulations are risk factors for this disease. This systematic review aims to determine the possible associations between the intestinal and mammary microbial populations, epigenetic modifications, and breast cancer. To achieve this objective, we conducted a literature search in the PubMed, Web of Science, and Science Direct databases following the PRISMA guidelines. Although no results are yet available in humans, studies in mice suggest a protective effect of maternal dietary interventions with bioactive compounds on the development of breast tumors in offspring. These dietary interventions also modified the gut microbiota, increasing the relative abundance of short-chain fatty acid-producing taxa and preventing mammary carcinogenesis. In addition, short-chain fatty acids produced by the microbiota act as epigenetic modulators. Furthermore, some authors indicate that stress alters the gut microbiota, promoting breast tumor growth through epigenetic and gene expression changes in the breast tumor microenvironment. Taken together, these findings show the ability of epigenetic modifications and alterations of the microbiota associated with environmental factors to modulate the development, aggressiveness, and progression of breast cancer.
Collapse
Affiliation(s)
- Alba Soldado-Gordillo
- Department of Biochemistry and Molecular Biology 2, School of Pharmacy, Campus de Cartuja s/n, 18071 Granada, Spain;
| | - Ana Isabel Álvarez-Mercado
- Department Pharmacology, School of Pharmacy, Campus de Cartuja s/n, 18071 Granada, Spain
- Institute of Nutrition and Food Technology “José Mataix”, Biomedical Research Center, Parque Tecnológico Ciencias de la Salud, Avda. del Conocimiento s/n, Armilla, 18016 Granada, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, Complejo Hospitalario Universitario de Granada, 18071 Granada, Spain
| |
Collapse
|
21
|
Zhou H, Deng N, Li Y, Hu X, Yu X, Jia S, Zheng C, Gao S, Wu H, Li K. Distinctive tumorigenic significance and innovative oncology targets of SUMOylation. Theranostics 2024; 14:3127-3149. [PMID: 38855173 PMCID: PMC11155398 DOI: 10.7150/thno.97162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/10/2024] [Accepted: 05/13/2024] [Indexed: 06/11/2024] Open
Abstract
Protein SUMOylation, a post-translational modification, intricately regulates diverse biological processes including gene expression, cell cycle progression, signaling pathway transduction, DNA damage response, and RNA metabolism. This modification contributes to the acquisition of tumorigenicity and the maintenance of cancer hallmarks. In malignancies, protein SUMOylation is triggered by various cellular stresses, promoting tumor initiation and progression. This augmentation is orchestrated through its specific regulatory mechanisms and characteristic biological functions. This review focuses on elucidating the fundamental regulatory mechanisms and pathological functions of the SUMO pathway in tumor pathogenesis and malignant evolution, with particular emphasis on the tumorigenic potential of SUMOylation. Furthermore, we underscore the potential therapeutic benefits of targeting the SUMO pathway, paving the way for innovative anti-tumor strategies by perturbing this dynamic and reversible modifying process.
Collapse
Affiliation(s)
- Heng Zhou
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China; Key Laboratory of Molecular Pathology and Epidemiology of Gastric Cancer in the Universities of Liaoning Province, Shenyang, Liaoning 110001, China
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Na Deng
- Department of Hematology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Yanshu Li
- Department of Cell Biology, Key Laboratory of Cell Biology, National Health Commission of the PRC and Key Laboratory of Medical Cell Biology, Ministry of Education of the PRC, China Medical University, Shenyang, Liaoning 110122, China
| | - Xiaoyun Hu
- Scientific Experimental Center, School of Pharmacy, China Medical University, Shenyang, Liaoning 110122, China
| | - Xue Yu
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China; Key Laboratory of Molecular Pathology and Epidemiology of Gastric Cancer in the Universities of Liaoning Province, Shenyang, Liaoning 110001, China
| | - Shiheng Jia
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China; Key Laboratory of Molecular Pathology and Epidemiology of Gastric Cancer in the Universities of Liaoning Province, Shenyang, Liaoning 110001, China
| | - Chen Zheng
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China; Key Laboratory of Molecular Pathology and Epidemiology of Gastric Cancer in the Universities of Liaoning Province, Shenyang, Liaoning 110001, China
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Shan Gao
- Department of Gynecology and Obstetrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Huizhe Wu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning 110122, China
- Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation; Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education; China Medical University, Shenyang, Liaoning 110122, China
- Shenyang Kangwei Medical Laboratory Analysis Co. LTD, Liaoning Province, China
| | - Kai Li
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China; Key Laboratory of Molecular Pathology and Epidemiology of Gastric Cancer in the Universities of Liaoning Province, Shenyang, Liaoning 110001, China
| |
Collapse
|
22
|
Profir M, Roşu OA, Creţoiu SM, Gaspar BS. Friend or Foe: Exploring the Relationship between the Gut Microbiota and the Pathogenesis and Treatment of Digestive Cancers. Microorganisms 2024; 12:955. [PMID: 38792785 PMCID: PMC11124004 DOI: 10.3390/microorganisms12050955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/11/2024] [Revised: 04/25/2024] [Accepted: 05/06/2024] [Indexed: 05/26/2024] Open
Abstract
Digestive cancers are among the leading causes of cancer death in the world. However, the mechanisms of cancer development and progression are not fully understood. Accumulating evidence in recent years pointing to the bidirectional interactions between gut dysbiosis and the development of a specific type of gastrointestinal cancer is shedding light on the importance of this "unseen organ"-the microbiota. This review focuses on the local role of the gut microbiota imbalance in different digestive tract organs and annexes related to the carcinogenic mechanisms. Microbiota modulation, either by probiotic administration or by dietary changes, plays an important role in the future therapies of various digestive cancers.
Collapse
Affiliation(s)
- Monica Profir
- Department of Oncology, Elias University Emergency Hospital, 011461 Bucharest, Romania; (M.P.); (O.A.R.)
- Department of Morphological Sciences, Cell and Molecular Biology and Histology, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Oana Alexandra Roşu
- Department of Oncology, Elias University Emergency Hospital, 011461 Bucharest, Romania; (M.P.); (O.A.R.)
| | - Sanda Maria Creţoiu
- Department of Morphological Sciences, Cell and Molecular Biology and Histology, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Bogdan Severus Gaspar
- Surgery Clinic, Emergency Clinical Hospital of Bucharest, 014461 Bucharest, Romania;
- Department of Surgery, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
| |
Collapse
|
23
|
Facchin S, Bertin L, Bonazzi E, Lorenzon G, De Barba C, Barberio B, Zingone F, Maniero D, Scarpa M, Ruffolo C, Angriman I, Savarino EV. Short-Chain Fatty Acids and Human Health: From Metabolic Pathways to Current Therapeutic Implications. Life (Basel) 2024; 14:559. [PMID: 38792581 PMCID: PMC11122327 DOI: 10.3390/life14050559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/25/2024] [Revised: 04/24/2024] [Accepted: 04/25/2024] [Indexed: 05/26/2024] Open
Abstract
The gastrointestinal tract is home to trillions of diverse microorganisms collectively known as the gut microbiota, which play a pivotal role in breaking down undigested foods, such as dietary fibers. Through the fermentation of these food components, short-chain fatty acids (SCFAs) such as acetate, propionate, and butyrate are produced, offering numerous health benefits to the host. The production and absorption of these SCFAs occur through various mechanisms within the human intestine, contingent upon the types of dietary fibers reaching the gut and the specific microorganisms engaged in fermentation. Medical literature extensively documents the supplementation of SCFAs, particularly butyrate, in the treatment of gastrointestinal, metabolic, cardiovascular, and gut-brain-related disorders. This review seeks to provide an overview of the dynamics involved in the production and absorption of acetate, propionate, and butyrate within the human gut. Additionally, it will focus on the pivotal roles these SCFAs play in promoting gastrointestinal and metabolic health, as well as their current therapeutic implications.
Collapse
Affiliation(s)
- Sonia Facchin
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University Hospital of Padua, 35128 Padua, Italy (L.B.); (B.B.)
| | - Luisa Bertin
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University Hospital of Padua, 35128 Padua, Italy (L.B.); (B.B.)
| | - Erica Bonazzi
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University Hospital of Padua, 35128 Padua, Italy (L.B.); (B.B.)
| | - Greta Lorenzon
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University Hospital of Padua, 35128 Padua, Italy (L.B.); (B.B.)
| | - Caterina De Barba
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University Hospital of Padua, 35128 Padua, Italy (L.B.); (B.B.)
| | - Brigida Barberio
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University Hospital of Padua, 35128 Padua, Italy (L.B.); (B.B.)
| | - Fabiana Zingone
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University Hospital of Padua, 35128 Padua, Italy (L.B.); (B.B.)
| | - Daria Maniero
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University Hospital of Padua, 35128 Padua, Italy (L.B.); (B.B.)
| | - Marco Scarpa
- General Surgery Unit, Department of Surgery, Oncology and Gastroenterology, University of Padova, 35138 Padua, Italy (C.R.); (I.A.)
| | - Cesare Ruffolo
- General Surgery Unit, Department of Surgery, Oncology and Gastroenterology, University of Padova, 35138 Padua, Italy (C.R.); (I.A.)
| | - Imerio Angriman
- General Surgery Unit, Department of Surgery, Oncology and Gastroenterology, University of Padova, 35138 Padua, Italy (C.R.); (I.A.)
| | - Edoardo Vincenzo Savarino
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University Hospital of Padua, 35128 Padua, Italy (L.B.); (B.B.)
| |
Collapse
|
24
|
Alves S, Santos-Pereira C, Oliveira CSF, Preto A, Chaves SR, Côrte-Real M. Enhancement of Acetate-Induced Apoptosis of Colorectal Cancer Cells by Cathepsin D Inhibition Depends on Oligomycin A-Sensitive Respiration. Biomolecules 2024; 14:473. [PMID: 38672489 PMCID: PMC11048611 DOI: 10.3390/biom14040473] [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] [Academic Contribution Register] [Received: 03/18/2024] [Revised: 04/08/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024] Open
Abstract
Colorectal cancer (CRC) is a leading cause of death worldwide. Conventional therapies are available with varying effectiveness. Acetate, a short-chain fatty acid produced by human intestinal bacteria, triggers mitochondria-mediated apoptosis preferentially in CRC but not in normal colonocytes, which has spurred an interest in its use for CRC prevention/therapy. We previously uncovered that acetate-induced mitochondrial-mediated apoptosis in CRC cells is significantly enhanced by the inhibition of the lysosomal protease cathepsin D (CatD), which indicates both mitochondria and the lysosome are involved in the regulation of acetate-induced apoptosis. Herein, we sought to determine whether mitochondrial function affects CatD apoptotic function. We found that enhancement of acetate-induced apoptosis by CatD inhibition depends on oligomycin A-sensitive respiration. Mechanistically, the potentiating effect is associated with an increase in cellular and mitochondrial superoxide anion accumulation and mitochondrial mass. Our results provide novel clues into the regulation of CatD function and the effect of tumor heterogeneity in the outcome of combined treatment using acetate and CatD inhibitors.
Collapse
Affiliation(s)
| | | | | | | | - Susana R. Chaves
- CBMA—Centre of Molecular and Environmental Biology, Department of Biology, University of Minho, 4710-057 Braga, Portugal; (S.A.); (C.S.-P.); (C.S.F.O.); (A.P.)
| | - Manuela Côrte-Real
- CBMA—Centre of Molecular and Environmental Biology, Department of Biology, University of Minho, 4710-057 Braga, Portugal; (S.A.); (C.S.-P.); (C.S.F.O.); (A.P.)
| |
Collapse
|
25
|
Garvey M. Intestinal Dysbiosis: Microbial Imbalance Impacts on Colorectal Cancer Initiation, Progression and Disease Mitigation. Biomedicines 2024; 12:740. [PMID: 38672096 PMCID: PMC11048178 DOI: 10.3390/biomedicines12040740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/08/2024] [Revised: 03/22/2024] [Accepted: 03/25/2024] [Indexed: 04/28/2024] Open
Abstract
The human gastrointestinal tract houses a diverse range of microbial species that play an integral part in many biological functions. Several preclinical studies using germ-free mice models have demonstrated that the gut microbiome profoundly influences carcinogenesis and progression. Colorectal cancer appears to be associated with microbial dysbiosis involving certain bacterial species, including F. nucleatum, pks+ E. coli, and B. fragilis, with virome commensals also disrupted in patients. A dysbiosis toward these pro-carcinogenic species increases significantly in CRC patients, with reduced numbers of the preventative species Clostridium butyicum, Roseburia, and Bifidobacterium evident. There is also a correlation between Clostridium infection and CRC. F. nucleatum, in particular, is strongly associated with CRC where it is associated with therapeutic resistance and poor outcomes in patients. The carcinogenic mode of action of pathogenic bacteria in CRC is a result of genotoxicity, epigenetic alterations, ROS generation, and pro-inflammatory activity. The aim of this review is to discuss the microbial species and their impact on colorectal cancer in terms of disease initiation, progression, and metastasis. The potential of anticancer peptides as anticancer agents or adjuvants is also discussed, as novel treatment options are required to combat the high levels of resistance to current pharmaceutical options.
Collapse
Affiliation(s)
- Mary Garvey
- Department of Life Science, Atlantic Technological University, F91 YW50 Sligo, Ireland;
- Centre for Precision Engineering, Materials and Manufacturing Research (PEM), Atlantic Technological University, F91 YW50 Sligo, Ireland
| |
Collapse
|
26
|
Cao Q, Tian Y, Deng Z, Yang F, Chen E. Epigenetic Alteration in Colorectal Cancer: Potential Diagnostic and Prognostic Implications. Int J Mol Sci 2024; 25:3358. [PMID: 38542332 PMCID: PMC10969857 DOI: 10.3390/ijms25063358] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/03/2024] [Revised: 03/01/2024] [Accepted: 03/12/2024] [Indexed: 01/03/2025] Open
Abstract
Colorectal cancer (CRC), a prevalent malignant tumor of the digestive system, ranks as the third and second in global incidence and mortality, respectively, in 2020, with 1.93 million new cases (≈10% of all cancers). There are 940,000 deaths (≈9.4% of all cancers), and the incidence of CRC in younger patients (under 50 years of age) has become a new trend. The pathogenesis of CRC is primarily attributed to a series of genetic and epigenetic abnormalities within normal colonic epithelial cells, coupled with the reshaping of the tumor microenvironment in the surrounding stroma. This process leads to the transformation of colorectal adenomas into invasive adenocarcinomas. Although genetic changes are known to be the primary driving force in the occurrence and progression of CRC, recent research indicates that epigenetic regulation serves as a crucial molecular marker in cancer, playing a significant role in the pathological and physiological control of interactions between genetics and the environment. This review discusses the current global epidemiology of CRC, its risk factors, and preventive treatment strategies. The current study explores the latest advancements in the epigenetic regulation of CRC, including DNA methylation, histone modifications, and non-coding RNAs (ncRNAs). These developments hold potential as screening tools, prognostic biomarkers, and therapeutic targets for CRC.
Collapse
Affiliation(s)
- Qing Cao
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Northwest University, Xi’an 710069, China; (Q.C.); (Y.T.); (Z.D.); (F.Y.)
- Provincial Key Laboratory of Biotechnology of Shaanxi Province, Northwest University, Xi’an 710069, China
| | - Ye Tian
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Northwest University, Xi’an 710069, China; (Q.C.); (Y.T.); (Z.D.); (F.Y.)
- Provincial Key Laboratory of Biotechnology of Shaanxi Province, Northwest University, Xi’an 710069, China
| | - Zhiyi Deng
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Northwest University, Xi’an 710069, China; (Q.C.); (Y.T.); (Z.D.); (F.Y.)
- Provincial Key Laboratory of Biotechnology of Shaanxi Province, Northwest University, Xi’an 710069, China
| | - Fangfang Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Northwest University, Xi’an 710069, China; (Q.C.); (Y.T.); (Z.D.); (F.Y.)
- Provincial Key Laboratory of Biotechnology of Shaanxi Province, Northwest University, Xi’an 710069, China
| | - Erfei Chen
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Northwest University, Xi’an 710069, China; (Q.C.); (Y.T.); (Z.D.); (F.Y.)
- Provincial Key Laboratory of Biotechnology of Shaanxi Province, Northwest University, Xi’an 710069, China
- School of Medicine, Northwest University, Xi’an 710069, China
| |
Collapse
|
27
|
Zhang Y, Dong L, Dai X, Huang Y, Gao Y, Wang F. Modulation of intestinal metabolites by calorie restriction and its association with gut microbiota in a xenograft model of colorectal cancer. Discov Oncol 2024; 15:46. [PMID: 38386206 PMCID: PMC10884396 DOI: 10.1007/s12672-024-00897-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 09/22/2023] [Accepted: 02/18/2024] [Indexed: 02/23/2024] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is a common malignant tumor, and its occurrence and development are closely related to dysbiosis of gut microbes. Previously, we found calorie restriction altered the composition of the microbial community in a colorectal cancer mouse model and inhibited in vivo growth of CRC cells. Here, we aim to further investigate alteration in the intestinal metabolites and explore the interplay between gut microbiota and intestinal metabolites upon calorie restriction. METHODS Human colorectal cancer HCT116 cells were used to establish a colorectal cancer xenograft mouse model. The changes of intestinal metabolites in the ad libitum group and calorie restriction group were investigated through untargeted metabolomics analysis. The integrative analysis of gut microbiota and metabolites to elucidate the associations between gut microbiota and intestinal metabolites. RESULTS Compared with the mice in the ad libitum group, mice upon calorie restriction exhibited downregulation of Isoleucyl-Valine, and upregulation of D-Proline, 1-Palmitoylphosphatidylcholine, and 4-Trimethylammoniobutanoic acid. Additionally, an integrative analysis of gut microbiota and metabolites revealed that Lactobacillus, Parabacteroides and rC4-4 genus were upregulated in the calorie restriction group and positively correlated with D-Proline, 4-Trimethylammoniobutanoic acid or 1-Palmitoylphosphatidylcholine, while negatively correlated with Isoleucyl-Valine. In contrast, the Nitrospirae and Deferribacteres phylum exhibited opposite trends. CONCLUSION Calorie restriction affects the abundance of gut microbes such as Nitrospirae phylum and Lactobacillus genus in mouse model of colorectal cancer, leading to changes in the metabolites such as D-Proline、Isoleucyl-Valine, which contributes to the suppression of in vivo growth of CRC by calorie restriction.
Collapse
Affiliation(s)
- Yuhuan Zhang
- Department of Gastroenterology, General Hospital, Ningxia Medical University, 804 Shengli South Street, Yinchuan, 750004, Ningxia, People's Republic of China
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, China
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750004, Ningxia, People's Republic of China
| | - Lintao Dong
- Department of Gastroenterology, General Hospital, Ningxia Medical University, 804 Shengli South Street, Yinchuan, 750004, Ningxia, People's Republic of China
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, China
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750004, Ningxia, People's Republic of China
| | - Xingchen Dai
- Department of Gastroenterology, General Hospital, Ningxia Medical University, 804 Shengli South Street, Yinchuan, 750004, Ningxia, People's Republic of China
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, China
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750004, Ningxia, People's Republic of China
| | - Yongli Huang
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750004, Ningxia, People's Republic of China
| | - Yujing Gao
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750004, Ningxia, People's Republic of China.
- National Health Commission Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan, 750004, Ningxia, People's Republic of China.
| | - Fang Wang
- Department of Gastroenterology, General Hospital, Ningxia Medical University, 804 Shengli South Street, Yinchuan, 750004, Ningxia, People's Republic of China.
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Ningxia Medical University, 1160 Shengli Street, Yinchuan, 750004, Ningxia, People's Republic of China.
| |
Collapse
|
28
|
Li X, Wu D, Li Q, Gu J, Gao W, Zhu X, Yin W, Zhu R, Zhu L, Jiao N. Host-microbiota interactions contributing to the heterogeneous tumor microenvironment in colorectal cancer. Physiol Genomics 2024; 56:221-234. [PMID: 38073489 DOI: 10.1152/physiolgenomics.00103.2023] [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] [Academic Contribution Register] [Received: 09/01/2023] [Revised: 11/15/2023] [Accepted: 12/01/2023] [Indexed: 01/23/2024] Open
Abstract
Colorectal cancer (CRC) exhibits pronounced heterogeneity and is categorized into four widely accepted consensus molecular subtypes (CMSs) with unique tumor microenvironments (TMEs). However, the intricate landscape of the microbiota and host-microbiota interactions within these TMEs remains elusive. Using RNA-sequencing data from The Cancer Genome Atlas, we analyzed the host transcriptomes and intratumoral microbiome profiles of CRC samples. Distinct host genes and microbial genera were identified among the CMSs. Immune microenvironments were evaluated using CIBERSORTx and ESTIMATE, and microbial coabundance patterns were assessed with FastSpar. Through LASSO penalized regression, we explored host-microbiota associations for each CMS. Our analysis revealed distinct host gene signatures within the CMSs, which encompassed ferroptosis-related genes and specific immune microenvironments. Moreover, we identified 293, 153, 66, and 109 intratumoral microbial genera with differential abundance, and host-microbiota associations contributed to distinct TMEs, characterized by 829, 1,270, 634, and 1,882 robust gene-microbe associations for each CMS in CMS1-CMS4, respectively. CMS1 featured inflammation-related HSF1 activation and gene interactions within the endothelin pathway and Flammeovirga. Integrin-related genes displayed positive correlations with Sutterella in CMS2, whereas CMS3 spotlighted microbial associations with biosynthetic and metabolic pathways. In CMS4, genes involved in collagen biosynthesis showed positive associations with Sutterella, contributing to disruptions in homeostasis. Notably, immune-rich subtypes exhibited pronounced ferroptosis dysregulation, potentially linked to tissue microbial colonization. This comprehensive investigation delineates the diverse landscapes of the TME within each CMS, incorporating host genes, intratumoral microbiota, and their complex interactions. These findings shed light on previously uncharted mechanisms underpinning CRC heterogeneity and suggest potential therapeutic targets.NEW & NOTEWORTHY This study determined the following: 1) providing a comprehensive landscape of consensus molecular subtype (CMS)-specific tumor microenvironments (TMEs); 2) constructing CMS-specific networks, including host genes, intratumoral microbiota, and enriched pathways, analyzing their associations to uncover unique patterns that demonstrate the intricate interplay within the TME; and 3) revealing a connection between immune-rich subtypes and ferroptosis activation, suggesting a potential regulatory role of the microbiota in ferroptosis dysregulation of the colorectal cancer TME.
Collapse
Affiliation(s)
- Xiaoyi Li
- Department of Nephrology, Children's Hospital, National Clinical Research Center for Child Health, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Dingfeng Wu
- Department of Nephrology, Children's Hospital, National Clinical Research Center for Child Health, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Qiuyu Li
- Department of Nephrology, Children's Hospital, National Clinical Research Center for Child Health, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Jinglan Gu
- Department of Nephrology, Children's Hospital, National Clinical Research Center for Child Health, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Wenxing Gao
- The Shanghai Tenth People's Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, People's Republic of China
| | - Xinyue Zhu
- The Shanghai Tenth People's Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, People's Republic of China
| | - Wenjing Yin
- The Shanghai Tenth People's Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, People's Republic of China
| | - Ruixin Zhu
- The Shanghai Tenth People's Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, People's Republic of China
| | - Lixin Zhu
- Department of Colorectal Surgery, Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Na Jiao
- Department of Nephrology, Children's Hospital, National Clinical Research Center for Child Health, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| |
Collapse
|
29
|
Pham NHT, Joglekar MV, Wong WKM, Nassif NT, Simpson AM, Hardikar AA. Short-chain fatty acids and insulin sensitivity: a systematic review and meta-analysis. Nutr Rev 2024; 82:193-209. [PMID: 37290429 PMCID: PMC10777678 DOI: 10.1093/nutrit/nuad042] [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] [Academic Contribution Register] [Indexed: 06/10/2023] Open
Abstract
CONTEXT There is substantial evidence that reduced short-chain fatty acids (SCFAs) in the gut are associated with obesity and type 2 diabetes, although findings from clinical interventions that can increase SCFAs are inconsistent. OBJECTIVE This systematic review and meta-analysis aimed to assess the effect of SCFA interventions on fasting glucose, fasting insulin, and homeostatic model assessment of insulin resistance (HOMA-IR). DATA SOURCES Relevant articles published up to July 28, 2022, were extracted from PubMed and Embase using the MeSH (Medical Subject Headings) terms of the defined keywords [(short-chain fatty acids) AND (obesity OR diabetes OR insulin sensitivity)] and their synonyms. Data analyses were performed independently by two researchers who used the Cochrane meta-analysis checklist and the PRISMA guidelines. DATA EXTRACTION Clinical studies and trials that measured SCFAs and reported glucose homeostasis parameters were included in the analysis. Standardized mean differences (SMDs) with 95%CIs were calculated using a random-effects model in the data extraction tool Review Manager version 5.4 (RevMan 5.4). The risk-of-bias assessment was performed following the Cochrane checklist for randomized and crossover studies. DATA ANALYSIS In total, 6040 nonduplicate studies were identified, 23 of which met the defined criteria, reported fasting insulin, fasting glucose, or HOMA-IR values, and reported change in SCFA concentrations post intervention. Meta-analyses of these studies indicated that fasting insulin concentrations were significantly reduced (overall effect: SMD = -0.15; 95%CI = -0.29 to -0.01, P = 0.04) in treatment groups, relative to placebo groups, at the end of the intervention. Studies with a confirmed increase in SCFAs at the end of intervention also had a significant effect on lowering fasting insulin (P = 0.008). Elevated levels of SCFAs, compared with baseline levels, were associated with beneficial effects on HOMA-IR (P < 0.00001). There was no significant change in fasting glucose concentrations. CONCLUSION Increased postintervention levels of SCFAs are associated with lower fasting insulin concentrations, offering a beneficial effect on insulin sensitivity. SYSTEMATIC REVIEW REGISTRATION PROSPERO registration number CRD42021257248.
Collapse
Affiliation(s)
- Nhan H T Pham
- are with the Diabetes and Islet Biology Group, School of Medicine, Western Sydney University, Campbelltown, New South Wales, Australia
- are with the School of Life Sciences, University of Technology Sydney, Ultimo, New South Wales, Australia
| | - Mugdha V Joglekar
- are with the Diabetes and Islet Biology Group, School of Medicine, Western Sydney University, Campbelltown, New South Wales, Australia
| | - Wilson K M Wong
- are with the Diabetes and Islet Biology Group, School of Medicine, Western Sydney University, Campbelltown, New South Wales, Australia
| | - Najah T Nassif
- are with the School of Life Sciences, University of Technology Sydney, Ultimo, New South Wales, Australia
| | - Ann M Simpson
- are with the School of Life Sciences, University of Technology Sydney, Ultimo, New South Wales, Australia
| | - Anandwardhan A Hardikar
- are with the Diabetes and Islet Biology Group, School of Medicine, Western Sydney University, Campbelltown, New South Wales, Australia
- is with the Department of Science and Environment, Roskilde University, Roskilde, Denmark
| |
Collapse
|
30
|
Wei J, Zheng Z, Hou X, Jia F, Yuan Y, Yuan F, He F, Hu L, Zhao L. Echinacoside inhibits colorectal cancer metastasis via modulating the gut microbiota and suppressing the PI3K/AKT signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2024; 318:116866. [PMID: 37429503 DOI: 10.1016/j.jep.2023.116866] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 04/25/2023] [Revised: 06/10/2023] [Accepted: 06/27/2023] [Indexed: 07/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Echinacoside (ECH) is the dominant phenylethanoid glycoside-structured compound identified from our developed herbal formula Huangci granule, which has been previously reported to inhibit the invasion and metastasis of CRC and prolong patients' disease-free survival duration. Though ECH has inhibitory activity against aggressive colorectal cancer (CRC) cells, its anti-metastasis effect in vivo and the action mechanism is undetermined. Given that ECH has an extremely low bioavailability and gut microbiota drives the CRC progression, we hypothesized that ECH could inhibit metastatic CRC by targeting the gut microbiome. AIM OF THE STUDY The purpose of this study was to investigate the impact of ECH on colorectal cancer liver metastasis in vivo and its potential mechanisms. MATERIALS AND METHODS An intrasplenic injection-induced liver metastatic model was established to examine the efficiency of ECH on tumor metastasis in vivo. Fecal microbiota from the model group and the ECH group were separately transplanted into pseudo-sterile CRLM mice in order to verify the role of gut flora in the ECH anti-metastatic effect. The 16S rRNA gene sequence was applied to analyze the structure and composition of the gut microbiota after ECH intervention, and the effect of ECH on short-chain fatty acid (SCFAs)-producing bacteria growth was proven by anaerobic culturing in vitro. GC-MS was applied to quantitatively analyze the serum SCFAs levels in mice. RNA-seq was performed to detect the gene changes involving tumor-promoting signaling pathway. RESULTS ECH inhibited CRC metastasis in a dose-dependent manner in the metastatic colorectal cancer (mCRC) mouse model. Manipulation of gut bacteria in the mCRC mouse model further proved that SCFA-generating gut bacteria played an indispensable role in mediating the antimetastatic action of ECH. Under an anaerobic condition, ECH benefited SCFA-producing microbiota without affecting the total bacterial load, presenting a dose-dependent promotion on the growth of a butyrate producer, Faecalibacterium prausnitzii (F.p). Furthermore, ECH-reshaped or F.p-colonized microbiota with a high butyrate-producing capability inhibited liver metastasis by suppressing PI3K/AKT signaling and reversing the epithelial-mesenchymal transition (EMT) process, whereas this anti-metastatic ability was abrogated by the butyrate synthase inhibitor heptanoyl-CoA. CONCLUSION This study demonstrated that ECH exhibits oral anti-metastatic efficacy by facilitating butyrate-producing gut bacteria, which downregulates PI3K/AKT signaling and EMT. It hints at a novel role for ECH in CRC therapy.
Collapse
Affiliation(s)
- Jiao Wei
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zongmei Zheng
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xinxin Hou
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Fengjing Jia
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuan Yuan
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Fuwen Yuan
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Feng He
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Liang Hu
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Ling Zhao
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
31
|
Kwao-Zigah G, Bediako-Bowan A, Boateng PA, Aryee GK, Abbang SM, Atampugbire G, Quaye O, Tagoe EA. Microbiome Dysbiosis, Dietary Intake and Lifestyle-Associated Factors Involve in Epigenetic Modulations in Colorectal Cancer: A Narrative Review. Cancer Control 2024; 31:10732748241263650. [PMID: 38889965 PMCID: PMC11186396 DOI: 10.1177/10732748241263650] [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] [Academic Contribution Register] [Received: 11/02/2023] [Revised: 05/18/2024] [Accepted: 06/04/2024] [Indexed: 06/20/2024] Open
Abstract
Background: Colorectal cancer is the second cause of cancer mortality and the third most commonly diagnosed cancer worldwide. Current data available implicate epigenetic modulations in colorectal cancer development. The health of the large bowel is impacted by gut microbiome dysbiosis, which may lead to colon and rectum cancers. The release of microbial metabolites and toxins by these microbiotas has been shown to activate epigenetic processes leading to colorectal cancer development. Increased consumption of a 'Westernized diet' and certain lifestyle factors such as excessive consumption of alcohol have been associated with colorectal cancer.Purpose: In this review, we seek to examine current knowledge on the involvement of gut microbiota, dietary factors, and alcohol consumption in colorectal cancer development through epigenetic modulations.Methods: A review of several published articles focusing on the mechanism of how changes in the gut microbiome, diet, and excessive alcohol consumption contribute to colorectal cancer development and the potential of using these factors as biomarkers for colorectal cancer diagnosis.Conclusions: This review presents scientific findings that provide a hopeful future for manipulating gut microbiome, diet, and alcohol consumption in colorectal cancer patients' management and care.
Collapse
Affiliation(s)
- Genevieve Kwao-Zigah
- Department of Biochemistry, Cell and Molecular Biology/West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), University of Ghana, Accra, Ghana
| | - Antionette Bediako-Bowan
- Department of Surgery, University of Ghana Medical School, Accra, Ghana
- Department of Surgery, Korle Bu Teaching Hospital, Accra, Ghana
| | - Pius Agyenim Boateng
- Department of Biochemistry, Cell and Molecular Biology/West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), University of Ghana, Accra, Ghana
| | - Gloria Kezia Aryee
- Department of Medical Laboratory Sciences, University of Ghana, Accra, Ghana
| | - Stacy Magdalene Abbang
- Department of Biochemistry, Cell and Molecular Biology/West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), University of Ghana, Accra, Ghana
| | - Gabriel Atampugbire
- Department of Biochemistry, Cell and Molecular Biology/West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), University of Ghana, Accra, Ghana
| | - Osbourne Quaye
- Department of Biochemistry, Cell and Molecular Biology/West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), University of Ghana, Accra, Ghana
| | - Emmanuel A. Tagoe
- Department of Medical Laboratory Sciences, University of Ghana, Accra, Ghana
| |
Collapse
|
32
|
Hussan H, Clinton SK, Grainger EM, Webb M, Wang C, Webb A, Needleman B, Noria S, Zhu J, Choueiry F, Pietrzak M, Bailey MT. Distinctive patterns of sulfide- and butyrate-metabolizing bacteria after bariatric surgery: potential implications for colorectal cancer risk. Gut Microbes 2023; 15:2255345. [PMID: 37702461 PMCID: PMC10501170 DOI: 10.1080/19490976.2023.2255345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 04/18/2023] [Revised: 08/09/2023] [Accepted: 08/31/2023] [Indexed: 09/14/2023] Open
Abstract
Despite improved cardiometabolic outcomes following bariatric surgery, its long-term impact on colorectal cancer (CRC) risk remains uncertain. In parallel, the influence of bariatric surgery on the host microbiome and relationships with disease outcomes is beginning to be appreciated. Therefore, we investigated the impact of Roux-en-Y gastric bypass (RYGB) and vertical sleeve gastrectomy (VSG) on the patterns of sulfide-reducing and butyrate-producing bacteria, which are hypothesized to modulate CRC risk after bariatric surgery. In this single-center, cross-sectional study, we included 15 pre-surgery subjects with severe obesity and patients who are at a median (range) of 25.6 (9.9-46.5) months after RYGB (n = 16) or VSG (n = 10). The DNA abundance of fecal bacteria and enzymes involved in butyrate and sulfide metabolism were identified using metagenomic sequencing. Differences between pre-surgery and post-RYGB or post-VSG cohorts were quantified using the linear discriminant analysis (LDA) effect size (LEfSe) method. Our sample was predominantly female (87%) with a median (range) age of 46 (23-71) years. Post-RYGB and post-VSG patients had a higher DNA abundance of fecal sulfide-reducing bacteria than pre-surgery controls (LDA = 1.3-4.4, p < .05). The most significant enrichments were for fecal E. coli, Acidaminococcus and A. finegoldii after RYGB, and for A. finegoldii, S. vestibularis, V. parvula after VSG. As for butyrate-producing bacteria, R. faecis was more abundant, whereas B. dentium and A. hardus were lower post-RYGB vs. pre-surgery. B. dentium was also lower in post-VSG vs. pre-surgery. Consistent with these findings, our analysis showed a greater enrichment of sulfide-reducing enzymes after bariatric surgery, especially RYGB, vs. pre-surgery. The DNA abundance of butyrate-producing enzymes was lower post-RYGB. In conclusion, the two most used bariatric surgeries, RYGB and VSG, are associated with microbiome patterns that are potentially implicated in CRC risk. Future studies are needed to validate and understand the impact of these microbiome changes on CRC risk after bariatric surgery.
Collapse
Affiliation(s)
- Hisham Hussan
- Division of Gastroenterology, Department of Internal Medicine, University of California, Davis; Sacramento, CA, USA
- The UC Davis Comprehensive Cancer Center, Sacramento, CA, USA
| | - Steven K. Clinton
- Division of Medical Oncology; Department of Internal Medicine, The Ohio StateUniversity, Columbus, OH, USA
- The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Elizabeth M. Grainger
- Division of Medical Oncology; Department of Internal Medicine, The Ohio StateUniversity, Columbus, OH, USA
- The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Maxine Webb
- Division of Medical Oncology; Department of Internal Medicine, The Ohio StateUniversity, Columbus, OH, USA
- The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Cankun Wang
- Division of Biomedical Informatics, Department of Biomedical Informatics, The Ohio State University, Columbus, OH, USA
| | - Amy Webb
- Division of Biomedical Informatics, Department of Biomedical Informatics, The Ohio State University, Columbus, OH, USA
| | - Bradley Needleman
- Center for Minimally Invasive Surgery; Department of General Surgery, The Ohio State University, Columbus, OH, USA
| | - Sabrena Noria
- Center for Minimally Invasive Surgery; Department of General Surgery, The Ohio State University, Columbus, OH, USA
| | - Jiangjiang Zhu
- The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
- The Department of Human Sciences, The Ohio State University, Columbus, OH, USA
| | - Fouad Choueiry
- The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
- The Department of Human Sciences, The Ohio State University, Columbus, OH, USA
| | - Maciej Pietrzak
- Division of Biomedical Informatics, Department of Biomedical Informatics, The Ohio State University, Columbus, OH, USA
| | - Michael T. Bailey
- The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
- Center for Microbial Pathogenesis, Abigail Wexner Research Institute at Nationwide Children’s Hospital and Department of Pediatrics, Columbus, OH, USA
- The Oral and Gastrointestinal Microbiology Research Affinity Group, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, USA
| |
Collapse
|
33
|
Iwasaka C, Ninomiya Y, Nakagata T, Nanri H, Watanabe D, Ohno H, Tanisawa K, Konishi K, Murakami H, Tsunematsu Y, Sato M, Watanabe K, Miyachi M. Association between physical activity and the prevalence of tumorigenic bacteria in the gut microbiota of Japanese adults: a cross-sectional study. Sci Rep 2023; 13:20841. [PMID: 38012174 PMCID: PMC10682492 DOI: 10.1038/s41598-023-47442-9] [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] [Academic Contribution Register] [Received: 12/01/2022] [Accepted: 11/14/2023] [Indexed: 11/29/2023] Open
Abstract
Escherichia coli harboring polyketide synthase (pks+ E. coli) has been suggested to contribute to colorectal cancer development. Physical activity is strongly associated with lower colorectal cancer risks, but its effects on pks+ E. coli remain unclear. The aim of this study was to investigate the association between pks+ E. coli prevalence and physical activity. A cross-sectional study was conducted on 222 Japanese adults (27-79-years-old, 73.9% female). Triaxial accelerometers were used to measure light-intensity physical activity, moderate-to-vigorous intensity physical activity, the physical activity level, step-count, and time spent inactive. Fecal samples collected from participants were used to determine the prevalence of pks+ E. coli. Multivariate logistic regression analysis and restricted cubic spline curves were used to examine the association between pks+ E. coli prevalence and physical activity. The prevalence of pks+ E. coli was 26.6% (59/222 participants). The adjusted odds ratios (ORs) and 95% confidence intervals (CIs) for the highest tertile with reference to the lowest tertile of physical activity variables were as follows: light-intensity physical activity (OR 0.63; 95% CI 0.26-1.5), moderate-to-vigorous intensity physical activity (OR 0.85; 95% CI 0.39-1.87), physical activity level (OR 0.69; 95% CI 0.32-1.51), step-count (OR 0.92; 95% CI 0.42-2.00) and time spent inactive (OR 1.30; 95% CI 0.58-2.93). No significant dose-response relationship was found between all physical activity variables and pks+ E. coli prevalence. Our findings did not suggest that physical activity has beneficial effects on the prevalence of pks+ E. coli. Longitudinal studies targeting a large population are needed to clarify this association.
Collapse
Affiliation(s)
- Chiharu Iwasaka
- Department of Physical Activity Research, National Institutes of Biomedical Innovation, Health and Nutrition, Tokyo, Japan
| | - Yuka Ninomiya
- Department of Physical Activity Research, National Institutes of Biomedical Innovation, Health and Nutrition, Tokyo, Japan
| | - Takashi Nakagata
- Department of Physical Activity Research, National Institutes of Biomedical Innovation, Health and Nutrition, Tokyo, Japan
- Laboratory of Gut Microbiome for Health, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | - Hinako Nanri
- Department of Physical Activity Research, National Institutes of Biomedical Innovation, Health and Nutrition, Tokyo, Japan.
- Laboratory of Gut Microbiome for Health, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan.
| | - Daiki Watanabe
- Department of Physical Activity Research, National Institutes of Biomedical Innovation, Health and Nutrition, Tokyo, Japan
- Faculty of Sport Sciences, Waseda University, Saitama, 359-1192, Japan
| | - Harumi Ohno
- Department of Physical Activity Research, National Institutes of Biomedical Innovation, Health and Nutrition, Tokyo, Japan
- Department of Nutrition, Kiryu University, Gunma, Japan
| | - Kumpei Tanisawa
- Department of Physical Activity Research, National Institutes of Biomedical Innovation, Health and Nutrition, Tokyo, Japan
- Faculty of Sport Sciences, Waseda University, Saitama, 359-1192, Japan
| | - Kana Konishi
- Department of Physical Activity Research, National Institutes of Biomedical Innovation, Health and Nutrition, Tokyo, Japan
- Faculty of Food and Nutritional Sciences, Toyo University, Gunma, Japan
| | - Haruka Murakami
- Department of Physical Activity Research, National Institutes of Biomedical Innovation, Health and Nutrition, Tokyo, Japan
- Faculty of Sport and Health Science, Ritsumeikan University, Shiga, Japan
| | - Yuta Tsunematsu
- Department of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Michio Sato
- Department of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Kenji Watanabe
- Department of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Motohiko Miyachi
- Department of Physical Activity Research, National Institutes of Biomedical Innovation, Health and Nutrition, Tokyo, Japan.
- Faculty of Sport Sciences, Waseda University, Saitama, 359-1192, Japan.
| |
Collapse
|
34
|
Almanza-Aguilera E, Cano A, Gil-Lespinard M, Burguera N, Zamora-Ros R, Agudo A, Farràs M. Mediterranean diet and olive oil, microbiota, and obesity-related cancers. From mechanisms to prevention. Semin Cancer Biol 2023; 95:103-119. [PMID: 37543179 DOI: 10.1016/j.semcancer.2023.08.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/27/2022] [Revised: 07/02/2023] [Accepted: 08/01/2023] [Indexed: 08/07/2023]
Abstract
Olive oil (OO) is the main source of added fat in the Mediterranean diet (MD). It is a mix of bioactive compounds, including monounsaturated fatty acids, phytosterols, simple phenols, secoiridoids, flavonoids, and terpenoids. There is a growing body of evidence that MD and OO improve obesity-related factors. In addition, obesity has been associated with an increased risk for several cancers: endometrial, oesophageal adenocarcinoma, renal, pancreatic, hepatocellular, gastric cardia, meningioma, multiple myeloma, colorectal, postmenopausal breast, ovarian, gallbladder, and thyroid cancer. However, the epidemiological evidence linking MD and OO with these obesity-related cancers, and their potential mechanisms of action, especially those involving the gut microbiota, are not clearly described or understood. The goals of this review are 1) to update the current epidemiological knowledge on the associations between MD and OO consumption and obesity-related cancers, 2) to identify the gut microbiota mechanisms involved in obesity-related cancers, and 3) to report the effects of MD and OO on these mechanisms.
Collapse
Affiliation(s)
- Enrique Almanza-Aguilera
- Unit of Nutrition and Cancer, Epidemiology Research Program, Catalan Institute of Oncology (ICO), Bellvitge Biomedical Research Institute (IDIBELL), 08908 L'Hospitalet de Llobregat, Spain
| | - Ainara Cano
- Food Research, AZTI, Basque Research and Technology Alliance (BRTA), Parque Tecnológico de Bizkaia, Astondo Bidea, Edificio 609, 48160, Derio, Spain
| | - Mercedes Gil-Lespinard
- Unit of Nutrition and Cancer, Epidemiology Research Program, Catalan Institute of Oncology (ICO), Bellvitge Biomedical Research Institute (IDIBELL), 08908 L'Hospitalet de Llobregat, Spain
| | - Nerea Burguera
- Food Research, AZTI, Basque Research and Technology Alliance (BRTA), Parque Tecnológico de Bizkaia, Astondo Bidea, Edificio 609, 48160, Derio, Spain
| | - Raul Zamora-Ros
- Unit of Nutrition and Cancer, Epidemiology Research Program, Catalan Institute of Oncology (ICO), Bellvitge Biomedical Research Institute (IDIBELL), 08908 L'Hospitalet de Llobregat, Spain; Department of Nutrition, Food Sciences, and Gastronomy, Food Innovation Network (XIA), Institute for Research on Nutrition and Food Safety (INSA), Faculty of Pharmacy and Food Sciences University of Barcelona, Barcelona, Spain.
| | - Antonio Agudo
- Unit of Nutrition and Cancer, Epidemiology Research Program, Catalan Institute of Oncology (ICO), Bellvitge Biomedical Research Institute (IDIBELL), 08908 L'Hospitalet de Llobregat, Spain
| | - Marta Farràs
- Unit of Nutrition and Cancer, Epidemiology Research Program, Catalan Institute of Oncology (ICO), Bellvitge Biomedical Research Institute (IDIBELL), 08908 L'Hospitalet de Llobregat, Spain.
| |
Collapse
|
35
|
Celiberto F, Aloisio A, Girardi B, Pricci M, Iannone A, Russo F, Riezzo G, D’Attoma B, Ierardi E, Losurdo G, Di Leo A. Fibres and Colorectal Cancer: Clinical and Molecular Evidence. Int J Mol Sci 2023; 24:13501. [PMID: 37686308 PMCID: PMC10488173 DOI: 10.3390/ijms241713501] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/29/2023] [Revised: 08/30/2023] [Accepted: 08/30/2023] [Indexed: 09/10/2023] Open
Abstract
Colorectal cancer (CRC) is one of the leading causes of mortality for cancer in industrialized countries. The link between diet and CRC is well-known, and presumably CRC is the type of cancer which is most influenced by dietary habits. In Western countries, an inadequate dietary intake of fibers is endemic, and this could be a driving factor in the increase of CRC incidence. Indeed, several epidemiologic studies have elucidated an inverse relationship between daily fiber intake and risk of CRC. Long-term prognosis in CRC survivors is also dependent on dietary fibers. Several pathogenetic mechanisms may be hypothesized. Fibers may interfere with the metabolism of bile acids, which may promote colon carcinogenesis. Further, fibers are often contained in vegetables which, in turn, contain large amounts of antioxidant agents like resveratrol, polyphenols, or phytoestrogens. Moreover, fibers can be digested by commensal flora, thus producing compounds such as butyrate, which exerts an antiproliferative effect. Finally, fibers may modulate gut microbiota, whose composition has shown to be associated with CRC onset. In this regard, dietary interventions based on high-fiber-containing diets are ongoing to prevent CRC development, especially in patients with high potential for this type of tumor. Despite the fact that outcomes are preliminary, encouraging results have been observed.
Collapse
Affiliation(s)
- Francesca Celiberto
- Section of Gastroenterology, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari, 70124 Bari, Italy
- Ph.D. Course in Organs and Tissues Transplantation and Cellular Therapies, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari, 70124 Bari, Italy
| | - Adriana Aloisio
- Section of Gastroenterology, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari, 70124 Bari, Italy
| | | | | | - Andrea Iannone
- Section of Gastroenterology, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari, 70124 Bari, Italy
| | - Francesco Russo
- Functional Gastrointestinal Disorders Research Group, National Institute of Gastroenterology IRCCS “Saverio de Bellis”, 70013 Castellana Grotte, Italy; (F.R.); (G.R.); (B.D.)
| | - Giuseppe Riezzo
- Functional Gastrointestinal Disorders Research Group, National Institute of Gastroenterology IRCCS “Saverio de Bellis”, 70013 Castellana Grotte, Italy; (F.R.); (G.R.); (B.D.)
| | - Benedetta D’Attoma
- Functional Gastrointestinal Disorders Research Group, National Institute of Gastroenterology IRCCS “Saverio de Bellis”, 70013 Castellana Grotte, Italy; (F.R.); (G.R.); (B.D.)
| | - Enzo Ierardi
- Section of Gastroenterology, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari, 70124 Bari, Italy
| | - Giuseppe Losurdo
- Section of Gastroenterology, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari, 70124 Bari, Italy
| | - Alfredo Di Leo
- Section of Gastroenterology, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari, 70124 Bari, Italy
| |
Collapse
|
36
|
Thoda C, Touraki M. Probiotic-Derived Bioactive Compounds in Colorectal Cancer Treatment. Microorganisms 2023; 11:1898. [PMID: 37630458 PMCID: PMC10456921 DOI: 10.3390/microorganisms11081898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/30/2023] [Revised: 07/14/2023] [Accepted: 07/24/2023] [Indexed: 08/27/2023] Open
Abstract
Colorectal cancer (CRC) is a multifactorial disease with increased morbidity and mortality rates globally. Despite advanced chemotherapeutic approaches for the treatment of CRC, low survival rates due to the regular occurrence of drug resistance and deleterious side effects render the need for alternative anticancer agents imperative. Accumulating evidence supports that gut microbiota imbalance precedes the establishment of carcinogenesis, subsequently contributing to cancer progression and response to anticancer therapy. Manipulation of the gut microbiota composition via the administration of probiotic-derived bioactive compounds has gradually attained the interest of scientific communities as a novel therapeutic strategy for CRC. These compounds encompass miscellaneous metabolic secreted products of probiotics, including bacteriocins, short-chain fatty acids (SCFAs), lactate, exopolysaccharides (EPSs), biosurfactants, and bacterial peptides, with profound anti-inflammatory and antiproliferative properties. This review provides a classification of postbiotic types and a comprehensive summary of the current state of research on their biological role against CRC. It also describes how their intricate interaction with the gut microbiota regulates the proper function of the intestinal barrier, thus eliminating gut dysbiosis and CRC development. Finally, it discusses the future perspectives in precision-medicine approaches as well as the challenges of their synthesis and optimization of administration in clinical studies.
Collapse
Affiliation(s)
| | - Maria Touraki
- Laboratory of General Biology, Department of Genetics, Development and Molecular Biology, School of Biology, Faculty of Sciences, Aristotle University of Thessaloniki, 54 124 Thessaloniki, Greece;
| |
Collapse
|
37
|
Chalova P, Tazky A, Skultety L, Minichova L, Chovanec M, Ciernikova S, Mikus P, Piestansky J. Determination of short-chain fatty acids as putative biomarkers of cancer diseases by modern analytical strategies and tools: a review. Front Oncol 2023; 13:1110235. [PMID: 37441422 PMCID: PMC10334191 DOI: 10.3389/fonc.2023.1110235] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/28/2022] [Accepted: 06/13/2023] [Indexed: 07/15/2023] Open
Abstract
Short-chain fatty acids (SCFAs) are the main metabolites produced by bacterial fermentation of non-digestible carbohydrates in the gastrointestinal tract. They can be seen as the major flow of carbon from the diet, through the microbiome to the host. SCFAs have been reported as important molecules responsible for the regulation of intestinal homeostasis. Moreover, these molecules have a significant impact on the immune system and are able to affect inflammation, cardiovascular diseases, diabetes type II, or oncological diseases. For this purpose, SCFAs could be used as putative biomarkers of various diseases, including cancer. A potential diagnostic value may be offered by analyzing SCFAs with the use of advanced analytical approaches such as gas chromatography (GC), liquid chromatography (LC), or capillary electrophoresis (CE) coupled with mass spectrometry (MS). The presented review summarizes the importance of analyzing SCFAs from clinical and analytical perspective. Current advances in the analysis of SCFAs focused on sample pretreatment, separation strategy, and detection methods are highlighted. Additionally, it also shows potential areas for the development of future diagnostic tools in oncology and other varieties of diseases based on targeted metabolite profiling.
Collapse
Affiliation(s)
- Petra Chalova
- Department of Pharmaceutical Analysis and Nuclear Pharmacy, Faculty of Pharmacy, Comenius University, Bratislava, Slovakia
- Biomedical Research Center of the Slovak Academy of Sciences, Institute of Virology, Bratislava, Slovakia
| | - Anton Tazky
- Department of Pharmaceutical Analysis and Nuclear Pharmacy, Faculty of Pharmacy, Comenius University, Bratislava, Slovakia
- Toxicological and Antidoping Center, Faculty of Pharmacy, Comenius University, Bratislava, Slovakia
| | - Ludovit Skultety
- Biomedical Research Center of the Slovak Academy of Sciences, Institute of Virology, Bratislava, Slovakia
- Institute of Microbiology, Academy of Sciences of the Czech Republic, Prague, Czechia
| | - Lenka Minichova
- Biomedical Research Center of the Slovak Academy of Sciences, Institute of Virology, Bratislava, Slovakia
| | - Michal Chovanec
- 2nd Department of Oncology, Faculty of Medicine, Comenius University and National Cancer Institute, Bratislava, Slovakia
| | - Sona Ciernikova
- Biomedical Research Center of the Slovak Academy of Sciences, Cancer Research Institute, Bratislava, Slovakia
| | - Peter Mikus
- Department of Pharmaceutical Analysis and Nuclear Pharmacy, Faculty of Pharmacy, Comenius University, Bratislava, Slovakia
- Toxicological and Antidoping Center, Faculty of Pharmacy, Comenius University, Bratislava, Slovakia
| | - Juraj Piestansky
- Toxicological and Antidoping Center, Faculty of Pharmacy, Comenius University, Bratislava, Slovakia
- Department of Galenic Pharmacy, Faculty of Pharmacy, Comenius University, Bratislava, Slovakia
| |
Collapse
|
38
|
Shewry PR, Brouns F, Dunn J, Hood J, Burridge AJ, America AHP, Gilissen L, Proos-Huijsmans ZAM, van Straaten JP, Jonkers D, Lazzeri PA, Ward JL, Lovegrove A. Comparative compositions of grain of tritordeum, durum wheat and bread wheat grown in multi-environment trials. Food Chem 2023; 423:136312. [PMID: 37182491 DOI: 10.1016/j.foodchem.2023.136312] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/18/2023] [Revised: 05/02/2023] [Accepted: 05/03/2023] [Indexed: 05/16/2023]
Abstract
Three genotypes each of bread wheat, durum wheat and tritordeum were grown in randomized replicated field trials in Andalusia (Spain) for two years and wholemeal flours analysed for a range of components to identify differences in composition. The contents of all components that were determined varied widely between grain samples of the individual species and in most cases also overlapped between the three species. Nevertheless, statistically significant differences between the compositions of the three species were observed. Notably, tritordeum had significantly higher contents of protein, some minerals (magnesium and iron), total phenolics and methyl donors. Tritordeum also had higher levels of total amino acids (but not asparagine) and total sugars, including raffinose. By contrast, bread wheat and tritordeum had similar contents of the two major dietary fibre components in white flour, arabinoxylan and β-glucan, with significantly lower contents in durum wheat.
Collapse
Affiliation(s)
- Peter R Shewry
- Rothamsted Research, Harpenden, Hertfordshire AL5 2JQ, UK.
| | - Fred Brouns
- Department of Human Biology, Faculty of Health, Medicine and Life Sciences, School for Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, Netherlands
| | - Jack Dunn
- Rothamsted Research, Harpenden, Hertfordshire AL5 2JQ, UK
| | - Jessica Hood
- Rothamsted Research, Harpenden, Hertfordshire AL5 2JQ, UK
| | - Amanda J Burridge
- Life Sciences, University of Bristol, 24 Tyndall Avenue, Bristol BS8 1TQ, UK
| | - Antoine H P America
- BU Bioscience, Plant Sciences Group, Wageningen University & Research, Netherlands
| | - Luud Gilissen
- Plant Breeding, Wageningen University & Research, Wageningen, PoBox 16, 6700AA Wageningen, Netherlands
| | - Zsuzsan A M Proos-Huijsmans
- Nederlands Bakkerij Centrum, Agro Business Park 75-83, 6708 PV Wageningen, Postbus 360, 6700 AJ Wageningen, Netherlands
| | - Jan Philip van Straaten
- Nederlands Bakkerij Centrum, Agro Business Park 75-83, 6708 PV Wageningen, Postbus 360, 6700 AJ Wageningen, Netherlands
| | - Daisy Jonkers
- Division of Gastroenterology-Hepatology, Department of Internal Medicine and School for Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Centre, Maastricht, Netherlands
| | - Paul A Lazzeri
- Vivagran SL, Calle Calabria 35, SA2, 08015 Barcelona, Spain
| | - Jane L Ward
- Rothamsted Research, Harpenden, Hertfordshire AL5 2JQ, UK
| | | |
Collapse
|
39
|
Yang S, Hao S, Ye H, Zhang X. Global research on the crosstalk between intestinal microbiome and colorectal cancer: A visualization analysis. Front Cell Infect Microbiol 2023; 13:1083987. [PMID: 37009513 PMCID: PMC10050574 DOI: 10.3389/fcimb.2023.1083987] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/29/2022] [Accepted: 03/03/2023] [Indexed: 03/17/2023] Open
Abstract
BackgroundIncreasing evidence has shown that the intestinal microbiome (IM) is highly linked to colorectal cancer (CRC). To investigate scientific output, identify highly cited papers, and explore research hotspots and trends in the field of IM/CRC, we conducted a bibliometric and visualized analysis.MethodsA bibliographic search regarding IM/CRC research (2012-2021) was implemented on October 17, 2022. The terms attached to IM and CRC were searched for in the titles (TI), abstracts (AB), and author keywords (AK). The main information was extracted from the Web of Science Core Collection (WoSCC). Biblioshiny from R packages and VOSviewer were used for data visualization.ResultsA total of 1725 papers related to IM/CRC were retrieved. Publications on IM/CRC have grown rapidly from 2012 to 2021. China and the United States were in the leading position for publications in this field and made the most significant contributions to IM/CRC research. Shanghai Jiao Tong University and Harvard University were the most productive institutions. The high-yield authors were Yu Jun and Fang Jing Yuan. The International Journal of Molecular Sciences published the most papers, whereas Gut had the most citations. Historical citation analysis showed the evolution of IM/CRC research. Current status and hotspots were highlighted using keyword cluster analysis. The hot topics include the effect of IM on tumorigenesis, the effect of IM on CRC treatment, the role of IM in CRC screening, the mechanisms of IM involvement in CRC, and IM modulation for CRC management. Some topics, such as chemotherapy, immunotherapy, Fusobacterium nucleatum and short-chain fatty acids could be the focus of IM/CRC research in the coming years.ConclusionThis research evaluated the global scientific output of IM/CRC research and its quantitative features, identified some significant papers, and gathered information on the status and trends of IM/CRC research, which may shape future paths for academics and practitioners.
Collapse
Affiliation(s)
- Shanshan Yang
- Department of Integrated Traditional Chinese and Western Medicine, Peking University First Hospital, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Shaodong Hao
- Spleen-Stomach Department, Fangshan Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Hui Ye
- Department of Integrated Traditional Chinese and Western Medicine, Peking University First Hospital, Beijing, China
- *Correspondence: Xuezhi Zhang, ; Hui Ye,
| | - Xuezhi Zhang
- Department of Integrated Traditional Chinese and Western Medicine, Peking University First Hospital, Beijing, China
- *Correspondence: Xuezhi Zhang, ; Hui Ye,
| |
Collapse
|
40
|
Kato I, Sun J. Microbiome and Diet in Colon Cancer Development and Treatment. Cancer J 2023; 29:89-97. [PMID: 36957979 PMCID: PMC10037538 DOI: 10.1097/ppo.0000000000000649] [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] [Academic Contribution Register] [Indexed: 03/25/2023]
Abstract
ABSTRACT Diet plays critical roles in defining our immune responses, microbiome, and progression of human diseases. With recent progress in sequencing and bioinformatic techniques, increasing evidence indicates the importance of diet-microbial interactions in cancer development and therapeutic outcome. Here, we focus on the epidemiological studies on diet-bacterial interactions in the colon cancer. We also review the progress of mechanistic studies using the experimental models. Finally, we discuss the limits and future directions in the research of microbiome and diet in cancer development and therapeutic outcome. Now, it is clear that microbes can influence the efficacy of cancer therapies. These research results open new possibilities for the diagnosis, prevention, and treatment of cancer. However, there are still big gaps to apply these new findings to the clinical practice.
Collapse
Affiliation(s)
- Ikuko Kato
- Department of Oncology, Wayne State University, Detroit Michigan, USA
- Department of Pathology, Wayne State University, Detroit Michigan, USA
| | - Jun Sun
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois Chicago, 840 S Wood Street, Room 704 CSB, MC716, Chicago, IL 60612, USA
- Department of Microbiology/Immunology, University of Illinois Chicago, Chicago, IL 60612, USA
- University of Illinois Cancer Center, 818 S Wolcott Avenue, Chicago, IL 60612, USA
- Jesse Brown VA Medical Center, 820 S. Damen Avenue, Chicago, IL 60612, USA
| |
Collapse
|
41
|
Dong Y, Zhang K, Wei J, Ding Y, Wang X, Hou H, Wu J, Liu T, Wang B, Cao H. Gut microbiota-derived short-chain fatty acids regulate gastrointestinal tumor immunity: a novel therapeutic strategy? Front Immunol 2023; 14:1158200. [PMID: 37122756 PMCID: PMC10140337 DOI: 10.3389/fimmu.2023.1158200] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/03/2023] [Accepted: 03/17/2023] [Indexed: 05/02/2023] Open
Abstract
Tumor immune microenvironment (TIME), a tumor-derived immune component, is proven to be closely related to the development, metastasis, and recurrence of tumors. Gut microbiota and its fermented-metabolites short-chain fatty acids (SCFAs) play a critical role in maintaining the immune homeostasis of gastrointestinal tumors. Consisting mainly of acetate, propionate, and butyrate, SCFAs can interact with G protein-coupled receptors 43 of T helper 1 cell or restrain histone deacetylases (HDACs) of cytotoxic T lymphocytes to exert immunotherapy effects. Studies have shed light on SCFAs can mediate the differentiation and function of regulatory T cells, as well as cytokine production in TIME. Additionally, SCFAs can alter epigenetic modification of CD8+ T cells by inhibiting HDACs to participate in the immune response process. In gastrointestinal tumors, the abundance of SCFAs and their producing bacteria is significantly reduced. Direct supplementation of dietary fiber and probiotics, or fecal microbiota transplantation to change the structure of gut microbiota can both increase the level of SCFAs and inhibit tumor development. The mechanism by which SCFAs modulate the progression of gastrointestinal tumors has been elucidated in this review, aiming to provide prospects for the development of novel immunotherapeutic strategies.
Collapse
|