1
|
Hesami Z, Sabzehali F, Khorsand B, Alipour S, Sadeghi A, Asri N, Pazienza V, Houri H. Microbiota as a state-of-the-art approach in precision medicine for pancreatic cancer management: A comprehensive systematic review. iScience 2025; 28:112314. [PMID: 40276756 PMCID: PMC12019022 DOI: 10.1016/j.isci.2025.112314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Revised: 12/22/2024] [Accepted: 03/25/2025] [Indexed: 04/26/2025] Open
Abstract
Emerging evidence suggests that harnessing the microbiome holds promise for innovative diagnostic and therapeutic strategies in the management of pancreatic cancer (PC). This study aims to systematically summarize the microbial markers associated with PC and assess their potential application in clinical outcome. Forty-one studies were included to assess the associations between microbial markers and PC. Among these, 13 were developed prediction models related to the microbiome in which the highest diagnostic and prognostic model belong to blood and intratumor markers, respectively. Notably, findings that utilize microbiotas from various body sites were elucidated, demonstrating their importance as unique signatures in biomarker discovery for diverse clinical applications. This review provides unique perspectives on overcoming challenges in PC by highlighting potential microbial-related markers as non-invasive approaches. Further clinical studies should evaluate the utility and accuracy of key indicators in the microbiome as a personalized tool for managing PC.
Collapse
Affiliation(s)
- Zeinab Hesami
- Student Research Committee, Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fattaneh Sabzehali
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Babak Khorsand
- Department of Neurology, University of California, Irvine, Irvine, CA, USA
| | - Samira Alipour
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amir Sadeghi
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nastaran Asri
- Celiac Disease and Gluten Related Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Valerio Pazienza
- Division of Gastroenterology, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Hamidreza Houri
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
2
|
Yang S, Hu Y, Cui M, Xu Q, Han X, Chang X, Zheng Q, Xiao J, Chen T, Li P, Dai M, Zhao Y. Microbiome, metabolome, and ionome profiling of cyst fluids reveals heterogeneity in pancreatic cystic neoplasms. Cancer Lett 2025; 623:217730. [PMID: 40252823 DOI: 10.1016/j.canlet.2025.217730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 04/11/2025] [Accepted: 04/17/2025] [Indexed: 04/21/2025]
Abstract
Pancreatic cystic neoplasms (PCNs) carry variable malignant potential, requiring precise clinical management. However, the heterogeneity and progression of PCNs remain poorly understood. This study analyzed the microbiome, metabolome, and ionome profiles of cyst fluids from 188 patients, including 165 with PCNs and 23 with other cyst types, using PacBio full-length 16S/ITS sequencing, LC-MS/MS, and ICP-MS. Bioinformatic analyses were performed, and metabolic enzyme and endoplasmic reticulum (ER) stress-related gene expression were examined using the PAAD TCGA dataset. PCNs were classified into distinct histopathological subtypes, including mucinous cystic lesions (MCLs) and serous cystic lesions (SCLs). MCLs demonstrated lower microbial diversity compared to SCLs, indicating microbial instability. Streptococcus and Staphylococcus were identified as key taxa in intraductal papillary mucinous neoplasms (IPMNs) and mucinous cystic neoplasms (MCNs), respectively. MCLs exhibited metabolic shifts towards lipid metabolism, while IPMNs showed distinct metabolic profiles potentially reflecting inflammation-related metabolic reprogramming. Ionic diversity varied among subtypes, with MCLs showing reduced diversity and IPMNs presenting broader ionic profiles. Palmitic acid (PA), a metabolite linked to Streptococcus, may contribute to pro-inflammatory metabolic alterations in IPMN. Our preliminary experiments demonstrated that co-culturing Streptococcus orails (S. orails) with ASAN-PaCa cells promoted their proliferation, accompanied by an elevation of PA levels in the supernatant. This integrative microbiome-metabolome-ionome analysis highlights histopathological heterogeneity among PCNs. While mechanistic associations remain to be fully defined, mucinous lesions may be more susceptible to microbe-driven metabolic disruption, with Streptococcus-associated lipid alterations as a potential contributing factor.
Collapse
Affiliation(s)
- Sen Yang
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science, and Peking Union Medical College, Beijing 100730, China
| | - Ya Hu
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science, and Peking Union Medical College, Beijing 100730, China
| | - Ming Cui
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science, and Peking Union Medical College, Beijing 100730, China
| | - Qiang Xu
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science, and Peking Union Medical College, Beijing 100730, China
| | - Xianlin Han
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science, and Peking Union Medical College, Beijing 100730, China
| | - Xiaoyan Chang
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Qingyuan Zheng
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science, and Peking Union Medical College, Beijing 100730, China
| | - Jinheng Xiao
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science, and Peking Union Medical College, Beijing 100730, China
| | - Tianqi Chen
- Medical Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Pengyu Li
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science, and Peking Union Medical College, Beijing 100730, China
| | - Menghua Dai
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science, and Peking Union Medical College, Beijing 100730, China.
| | - Yupei Zhao
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science, and Peking Union Medical College, Beijing 100730, China.
| |
Collapse
|
3
|
Stein-Thoeringer CK, Renz BW, De Castilhos J, von Ehrlich-Treuenstätt V, Wirth U, Tschaidse T, Hofmann FO, Koch DT, Beirith I, Ormanns S, Guba MO, Angele MK, Andrassy J, Niess H, D'Haese JG, Werner J, Ilmer M. Microbiome Dysbiosis With Enterococcus Presence in the Upper Gastrointestinal Tract Is a Risk Factor for Mortality in Patients Undergoing Surgery for Pancreatic Cancer. Ann Surg 2025; 281:615-623. [PMID: 38275104 DOI: 10.1097/sla.0000000000006210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2024]
Abstract
BACKGROUND Recent retrospective studies suggest a role for distinct microbiota in the perioperative morbidity and mortality of pancreatic head resections. OBJECTIVE We aimed to prospectively investigate the microbial colonization of critical operative sites of pancreatic head resections to identify microbial stratification factors for surgical and long-term oncologic outcomes. METHODS Prospective biomarker study applying 16S rRNA sequencing and microbial culturing to samples collected from various sites of the gastrointestinal tract and surgical sites of patients during pancreatic head resections at a German single high-volume pancreatic center. RESULTS A total of 101 patients were included {38 noncancer, 63 cancer patients [50 pancreatic ductal adenocarcinoma (PDAC) patients]} in the study. In a first data analysis series, 16S rRNA sequencing data were utilized from 96 patients to assess associations of microbiome profiles with clinical parameters and outcomes. In general, microbiome composition varied according to sampling site, cancer, age or preoperative endoscopic retrograde cholangiopancreatography (ERCP) intervention, notably for the bile microbiome. In the PDAC subcohort, the compositional variance of the bile or periampullary microbiome was significantly associated with postoperative complications such as intensive care unit admission; on a taxonomic level we observed Enterococcus spp. to be significantly more abundant in patients developing deep or organ-space surgical site infections (SSI). Elevated Enterococcus relative abundances in the upper gastrointestinal tract, in turn, were associated with 6 months mortality rates. In a second step, we focused on microbiological cultures collected from bile aspirates during surgery and investigated associations with perioperative complications and long-term survival. Notably, Enterococcus spp. were among the most prevalent pathobiont isolates observed in cancer patient bile specimens that were associated with severe SSIs, and thereby elevated mortality rates up to 24 months. Clinically relevant postoperative pancreatic fistulas or severe SSI were found as other major variables determining short-term mortality in this cancer patient cohort. In the context of adverse microbiological factors, a preoperative ERCP was also observed to segregate long-term survival, and it appeared to interact with the presence of Enterococcus spp. as highest mortality rates were observed in PDAC patients with both preoperative ERCP and presence of E. faecalis in bile aspirates. CONCLUSIONS The presence of Enterococcus spp. in bile ducts of PDAC patients undergoing pancreatic surgery represents a significant risk factor for perioperative infections and, thereby, elevated postoperative and long-term mortality. This finding supports previous data on the use of the antibiotic drug piperacillin-tazobactam as appropriate perioperative antibiotic prophylaxis for preventing adverse outcomes after pancreatoduodenectomy.
Collapse
Affiliation(s)
- Christoph K Stein-Thoeringer
- Laboratory of Translational Microbiome Science, Internal Medicine I, University Clinic Tuebingen, Germany
- CMFI Cluster of Excellence, University of Tuebingen, Germany
- DZIF (Deut. Zentrum für Infektionsforschung), HAARBI Partner Site Tuebingen, Germany
| | - Bernhard W Renz
- Department of General, Visceral and Transplantation Surgery, Hospital of the University of Munich, Ludwig-Maximilians-University Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, of the German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Juliana De Castilhos
- Laboratory of Translational Microbiome Science, Internal Medicine I, University Clinic Tuebingen, Germany
| | - Viktor von Ehrlich-Treuenstätt
- Department of General, Visceral and Transplantation Surgery, Hospital of the University of Munich, Ludwig-Maximilians-University Munich, Germany
| | - Ulrich Wirth
- Department of General, Visceral and Transplantation Surgery, Hospital of the University of Munich, Ludwig-Maximilians-University Munich, Germany
| | - Tengis Tschaidse
- Department of General, Visceral and Transplantation Surgery, Hospital of the University of Munich, Ludwig-Maximilians-University Munich, Germany
| | - Felix O Hofmann
- Department of General, Visceral and Transplantation Surgery, Hospital of the University of Munich, Ludwig-Maximilians-University Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, of the German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Dominik T Koch
- Department of General, Visceral and Transplantation Surgery, Hospital of the University of Munich, Ludwig-Maximilians-University Munich, Germany
| | - Iris Beirith
- Department of General, Visceral and Transplantation Surgery, Hospital of the University of Munich, Ludwig-Maximilians-University Munich, Germany
| | - Steffen Ormanns
- German Cancer Consortium (DKTK), Partner Site Munich, of the German Cancer Research Center (DKFZ), Heidelberg, Germany
- Institute of Pathology, Faculty of Medicine, Ludwig-Maximilians-University, Munich, Germany
| | - Markus O Guba
- Department of General, Visceral and Transplantation Surgery, Hospital of the University of Munich, Ludwig-Maximilians-University Munich, Germany
| | - Martin K Angele
- Department of General, Visceral and Transplantation Surgery, Hospital of the University of Munich, Ludwig-Maximilians-University Munich, Germany
| | - Joachim Andrassy
- Department of General, Visceral and Transplantation Surgery, Hospital of the University of Munich, Ludwig-Maximilians-University Munich, Germany
| | - Hanno Niess
- Department of General, Visceral and Transplantation Surgery, Hospital of the University of Munich, Ludwig-Maximilians-University Munich, Germany
| | - Jan G D'Haese
- Department of General, Visceral and Transplantation Surgery, Hospital of the University of Munich, Ludwig-Maximilians-University Munich, Germany
| | - Jens Werner
- Department of General, Visceral and Transplantation Surgery, Hospital of the University of Munich, Ludwig-Maximilians-University Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, of the German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Matthias Ilmer
- Department of General, Visceral and Transplantation Surgery, Hospital of the University of Munich, Ludwig-Maximilians-University Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, of the German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
4
|
Sulekha Suresh D, Jain T, Dudeja V, Iyer S, Dudeja V. From Microbiome to Malignancy: Unveiling the Gut Microbiome Dynamics in Pancreatic Carcinogenesis. Int J Mol Sci 2025; 26:3112. [PMID: 40243755 PMCID: PMC11988718 DOI: 10.3390/ijms26073112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Revised: 03/01/2025] [Accepted: 03/22/2025] [Indexed: 04/18/2025] Open
Abstract
Pancreatic cancer is a major cause of cancer-associated mortality globally, characterized by a poor prognosis and limited therapeutic response. The current approach for treating pancreatic cancer involves locoregional control with surgical resection and systemic therapy in the form of cytotoxic chemotherapy. However, despite standard-of-care treatment, the outcomes remain dismal. Emerging evidence suggests that the gut microbiota plays a significant role in pancreatic carcinogenesis through dysbiosis, chronic inflammation and immune modulation. Dysbiosis-driven alterations in the gut microbiota composition can disrupt intestinal homeostasis, promote systemic inflammation and create a tumor-permissive microenvironment in the pancreas. Moreover, the gut microbiota modulates the efficacy of systemic therapies, including chemotherapy and immunotherapy, by impacting drug metabolism and shaping the tumor immune landscape. This review is mainly focused on exploring the intricate interplay between the gut microbiota and pancreatic cancer, and also highlighting its dual role in carcinogenesis and the therapeutic response.
Collapse
Affiliation(s)
| | | | | | | | - Vikas Dudeja
- Division of Surgical Oncology, Department of Surgery, The University of Alabama at Birmingham, BDB 573 1808 7th Avenue South, Birmingham, AL 35294, USA; (D.S.S.); (T.J.); (V.D.); (S.I.)
| |
Collapse
|
5
|
Daniel N, Farinella R, Belluomini F, Fajkic A, Rizzato C, Souček P, Campa D, Hughes DJ. The relationship of the microbiome, associated metabolites and the gut barrier with pancreatic cancer. Semin Cancer Biol 2025; 112:43-57. [PMID: 40154652 DOI: 10.1016/j.semcancer.2025.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 02/26/2025] [Accepted: 03/19/2025] [Indexed: 04/01/2025]
Abstract
Pancreatic cancers have high mortality and rising incidence rates which may be related to unhealthy western-type dietary and lifestyle patterns as well as increasing body weights and obesity rates. Recent data also suggest a role for the gut microbiome in the development of pancreatic cancer. Here, we review the experimental and observational evidence for the roles of the oral, gut and intratumoural microbiomes, impaired gut barrier function and exposure to inflammatory compounds as well as metabolic dysfunction as contributors to pancreatic disease with a focus on pancreatic ductal adenocarcinoma (PDAC) initiation and progression. We also highlight some emerging gut microbiome editing techniques currently being investigated in the context of pancreatic disease. Notably, while the gut microbiome is significantly altered in PDAC and its precursor diseases, its utility as a diagnostic and prognostic tool is hindered by a lack of reproducibility and the potential for reverse causality in case-control cohorts. Future research should emphasise longitudinal and mechanistic studies as well as integrating lifestyle exposure and multi-omics data to unravel complex host-microbiome interactions. This will allow for deeper aetiologic and mechanistic insights that can inform treatments and guide public health recommendations.
Collapse
Affiliation(s)
- Neil Daniel
- Molecular Epidemiology of Cancer Group, UCD Conway Institute, School of Biomedical and Biomolecular Sciences, University College Dublin, Dublin, Ireland
| | | | | | - Almir Fajkic
- Department of Pathophysiology Faculty of Medicine, University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | | | - Pavel Souček
- Laboratory of Pharmacogenomics, Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic; Toxicogenomics Unit, National Institute of Public Health, Prague, Czech Republic
| | - Daniele Campa
- Department of Biology, University of Pisa, Pisa, Italy
| | - David J Hughes
- Molecular Epidemiology of Cancer Group, UCD Conway Institute, School of Biomedical and Biomolecular Sciences, University College Dublin, Dublin, Ireland.
| |
Collapse
|
6
|
Cheng H, Guo H, Wen C, Sun G, Tang F, Li Y. The dual role of gut microbiota in pancreatic cancer: new insights into onset and treatment. Ther Adv Med Oncol 2025; 17:17588359251324882. [PMID: 40093983 PMCID: PMC11909682 DOI: 10.1177/17588359251324882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 02/14/2025] [Indexed: 03/19/2025] Open
Abstract
Pancreatic cancer ranks among the most lethal digestive malignancies, exhibiting a steadily increasing incidence and mortality worldwide. Despite significant advances in cancer research, the 5-year survival rate remains below 10%, predominantly due to delayed diagnosis and limited therapeutic options. Concurrently, the gut microbiota-an integral component of host physiology-has emerged as a crucial player in the pathogenesis of pancreatic cancer. Mounting evidence indicates that alterations in gut microbial composition and function may influence tumor initiation, progression, and response to therapy. This review provides an in-depth examination of the intricate interplay between the gut microbiome and pancreatic cancer, highlighting potential diagnostic biomarkers and exploring microbiome-targeted therapeutic strategies to improve patient outcomes.
Collapse
Affiliation(s)
- Huijuan Cheng
- School of Life Sciences, Lanzhou University, Lanzhou, Gansu, P.R. China
- Gansu Provincial Key Laboratory of Environmental Oncology, Lanzhou University Second Hospital, Lanzhou, Gansu, P.R. China
| | - Hongkai Guo
- The Second School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, P.R. China
| | - Chengming Wen
- The Second School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, P.R. China
| | - Guodong Sun
- The Second School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, P.R. China
- Department of Medical Affairs, Lanzhou University First Hospital, Lanzhou, Gansu, P.R. China
| | - Futian Tang
- Gansu Provincial Key Laboratory of Environmental Oncology, Lanzhou University Second Hospital, Lanzhou, Gansu, P.R. China
| | - Yumin Li
- Gansu Provincial Key Laboratory of Environmental Oncology, Lanzhou University Second Hospital, No. 82, Cuiyingmen, Chengguan, Lanzhou, Gansu 730000, P.R. China
| |
Collapse
|
7
|
Németh K, Mezei E, Vörös J, Borka K, Pesti A, Kenessey I, Kiss A, Budai A. Subtype-associated complexity and prognostic significance of the NLRP3 inflammasome landscape in pancreatic neoplasms. J Pathol Clin Res 2025; 11:e70019. [PMID: 39969214 PMCID: PMC11837281 DOI: 10.1002/2056-4538.70019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 01/01/2025] [Accepted: 01/19/2025] [Indexed: 02/20/2025]
Abstract
Intraductal papillary mucinous neoplasm (IPMN) can progress into malignant pancreatic cancer, posing challenges in accurately assessing the risk of malignancy. While the nucleotide-binding oligomerization domain (NOD)-like receptor pyrin domain containing 3 (NLRP3) inflammasome pathway's role in pancreatic ductal adenocarcinoma (PDAC) has been extensively studied, its implications in IPMN remain unexplored. This study aimed to investigate the prognostic significance of NLRP3 inflammasome-related proteins across IPMN subtypes and their associations with tumor characteristics, with a secondary focus on comparing expression patterns in IPMN and PDAC. A cohort of 187 patients (100 IPMN and 87 PDAC) underwent high-dimensional histopathological imaging using the multiplexed immunohistochemical consecutive staining on single slide method and a semi-automated image analysis workflow. Expression levels of NLRP3, apoptosis-associated speck-like protein containing a caspase-recruitment domain (ASC), caspase-1, interleukin-1 beta, interleukin-18 (IL-18), interleukin-1 receptor antagonist, and interleukin-18 binding protein (IL-18BP) were evaluated and compared between IPMN and PDAC samples. The relationships between protein expression and tumor characteristics were examined. Principal component analysis distinguished between intestinal and nonintestinal clusters based on NLRP3-associated proteins. Lower IL-18 expression was linked to the intestinal subtype, while higher caspase-1 was linked to the pancreatobiliary subtype. Elevated caspase-1 and ASC expression were associated with invasiveness in IPMN. No significant correlation was found between the examined proteins and later-stage tumor characteristics in invasive cases. The IL-18/IL-18BP ratio was an independent prognostic factor in invasive IPMN. Our findings highlight the prognostic significance of IL-18 and the IL-18/IL-18BP ratio in invasive IPMNs. These results point to a complex regulation of NLRP3 inflammasome proteins, especially effector cytokines, in pancreatic neoplasms, which are strongly linked to subtype and prognosis.
Collapse
Affiliation(s)
- Kristóf Németh
- Department of Pathology, Forensics and Insurance MedicineSemmelweis UniversityBudapestHungary
| | - Eszter Mezei
- Department of Pathology, Forensics and Insurance MedicineSemmelweis UniversityBudapestHungary
| | - Justína Vörös
- Department of Pathology, Forensics and Insurance MedicineSemmelweis UniversityBudapestHungary
| | - Katalin Borka
- Department of Pathology, Forensics and Insurance MedicineSemmelweis UniversityBudapestHungary
| | - Adrián Pesti
- Department of Pathology, Forensics and Insurance MedicineSemmelweis UniversityBudapestHungary
| | - István Kenessey
- Department of Pathology, Forensics and Insurance MedicineSemmelweis UniversityBudapestHungary
- National Cancer Registry and Center for BiostatisticsNational Institute of OncologyBudapestHungary
| | - András Kiss
- Department of Pathology, Forensics and Insurance MedicineSemmelweis UniversityBudapestHungary
| | - András Budai
- Department of Pathology, Forensics and Insurance MedicineSemmelweis UniversityBudapestHungary
| |
Collapse
|
8
|
Zhao F, An R, Ma Y, Yu S, Gao Y, Wang Y, Yu H, Xie X, Zhang J. Integrated spatial multi-omics profiling of Fusobacterium nucleatum in breast cancer unveils its role in tumour microenvironment modulation and cancer progression. Clin Transl Med 2025; 15:e70273. [PMID: 40070022 PMCID: PMC11897063 DOI: 10.1002/ctm2.70273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 02/17/2025] [Accepted: 03/04/2025] [Indexed: 03/15/2025] Open
Abstract
Tumour-associated microbiota are integral components of the tumour microenvironment (TME). However, previous studies on intratumoral microbiota primarily rely on bulk tissue analysis, which may obscure their spatial distribution and localized effects. In this study, we applied in situ spatial-profiling technology to investigate the spatial distribution of intratumoral microbiota in breast cancer and their interactions with the local TME. Using 5R 16S rRNA gene sequencing and RNAscope FISH/CISH on patients' tissue, we identified significant spatial heterogeneity in intratumoral microbiota, with Fusobacterium nucleatum (F. nucleatum) predominantly localized in tumour cell-rich areas. GeoMx digital spatial profiling (DSP) revealed that regions colonized by F. nucleatum exhibit significant influence on the expression of RNAs and proteins involved in proliferation, migration and invasion. In vitro studies indicated that co-culture with F. nucleatum significantly stimulates the proliferation and migration of breast cancer cells. Integrative spatial multi-omics and co-culture transcriptomic analyses highlighted the MAPK signalling pathways as key altered pathways. By intersecting these datasets, VEGFD and PAK1 emerged as critical upregulated proteins in F. nucleatum-positive regions, showing strong positive correlations with MAPK pathway proteins. Moreover, the upregulation of VEGFD and PAK1 by F. nucleatum was confirmed in co-culture experiments, and their knockdown significantly reduced F. nucleatum-induced proliferation and migration. In conclusion, intratumoral microbiota in breast cancer exhibit significant spatial heterogeneity, with F. nucleatum colonization markedly altering tumour cell protein expression to promote progression and migration. These findings provide novel perspectives on the role of microbiota in breast cancer, identify potential therapeutic targets, and lay the foundation for future cancer treatments. KEY POINTS: Intratumoral Fusobacterium nucleatum exhibits significant spatial heterogeneity within breast cancer tissues. F. nucleatum colonization alters the expression of key proteins involved in tumour progression and migration. The MAPK signalling pathway is a critical mediator of F. nucleatum-induced breast cancer cell proliferation and migration. VEGFD and PAK1 are potential therapeutic targets to mitigate F. nucleatum-induced tumour progression.
Collapse
Affiliation(s)
- Feng Zhao
- Department of Clinical LaboratorySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouZhejiangPeople's Republic of China
- Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang ProvinceHangzhouZhejiangPeople's Republic of China
| | - Rui An
- Department of Clinical LaboratorySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouZhejiangPeople's Republic of China
- Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang ProvinceHangzhouZhejiangPeople's Republic of China
| | - Yilei Ma
- Department of Clinical LaboratorySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouZhejiangPeople's Republic of China
- Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang ProvinceHangzhouZhejiangPeople's Republic of China
| | - Shaobo Yu
- Department of Clinical LaboratorySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouZhejiangPeople's Republic of China
- Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang ProvinceHangzhouZhejiangPeople's Republic of China
| | - Yuzhen Gao
- Department of Clinical LaboratorySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouZhejiangPeople's Republic of China
- Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang ProvinceHangzhouZhejiangPeople's Republic of China
| | - Yanzhong Wang
- Department of Clinical LaboratorySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouZhejiangPeople's Republic of China
- Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang ProvinceHangzhouZhejiangPeople's Republic of China
| | - Haitao Yu
- Department of Clinical LaboratorySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouZhejiangPeople's Republic of China
- Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang ProvinceHangzhouZhejiangPeople's Republic of China
| | - Xinyou Xie
- Department of Clinical LaboratorySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouZhejiangPeople's Republic of China
- Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang ProvinceHangzhouZhejiangPeople's Republic of China
| | - Jun Zhang
- Department of Clinical LaboratorySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouZhejiangPeople's Republic of China
- Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang ProvinceHangzhouZhejiangPeople's Republic of China
| |
Collapse
|
9
|
Tavano F, Napoli A, Gioffreda D, Palmieri O, Latiano T, Tardio M, di Mola FF, Grottola T, Büchler MW, Gentile M, Latiano A, Mazza T, Perri F. Could the Microbial Profiling of Normal Pancreatic Tissue from Healthy Organ Donors Contribute to Understanding the Intratumoral Microbiota Signature in Pancreatic Ductal Adenocarcinoma? Microorganisms 2025; 13:452. [PMID: 40005817 PMCID: PMC11858623 DOI: 10.3390/microorganisms13020452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 02/14/2025] [Accepted: 02/14/2025] [Indexed: 02/27/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is associated with intratumoral microbiota changes. However, defining the normal pancreatic microbial composition remains a challenge. Herein, we tested the hypothesis that the microbial profiling of normal pancreatic tissue from healthy organ donors (HC) could help in determining the signature of microbiota in PDAC. Matched pairs of tumor and normal tissues from PDAC patients (n = 32) and normal pancreatic tissues from HC (n = 17) were analyzed by 16S rRNA gene sequencing. Dissimilarities in all the beta metrics emerged in both normal samples and tumor samples, compared to HC (Bray-Curtis dissimilarity and Jaccard distance: p = 0.002; weighted UniFrac distances: p = 0.42 and p = 0.012, respectively; unweighted UniFrac distance: p = 0.009); a trend toward a lower Faith's phylogenetic distance was found at the tumor level vs. HC (p = 0.08). Within PDAC, a lower Faith's phylogenetic distance (p = 0.003) and a significant unweighted UniFrac distance (p = 0.024) were observed in tumor samples vs. normal samples. We noted the presence of a decreased abundance of bacteria with potential beneficial effects (Jeotgalicoccus) and anticancer activity (Acinetobacter_guillouiae) in PDAC vs. HC; bacteria involved in immune homeostasis and suppression of tumor progression (Streptococcus_salivarius, Sphingomonas) were reduced, and those implicated in tumor initiation and development (Methylobacterium-Methylorubrum, g_Delftia) were enhanced in tumor samples vs. normal samples. Metagenomic functions involved in fatty acid synthesis were reduced in normal samples compared to HC, while peptidoglycan biosynthesis IV and L-rhamnose degradation were more abundant in tumor samples vs. normal samples. Future prospective studies on larger populations, also including patients in advanced tumor stages and considering all potential existing confounding factors, as well as further functional investigations, are needed to prove the role of microbiota-mediated pathogenicity in PDAC.
Collapse
Affiliation(s)
- Francesca Tavano
- Division of Gastroenterology and Endoscopy, Fondazione IRCCS “Casa Sollievo della Sofferenza” Hospital, Viale Cappuccini 1, 71013 San Giovanni Rotondo, FG, Italy
| | - Alessandro Napoli
- Bioinformatics Laboratory, Fondazione IRCCS “Casa Sollievo della Sofferenza” Hospital, Viale Cappuccini 1, 71013 San Giovanni Rotondo, FG, Italy
| | - Domenica Gioffreda
- Division of Gastroenterology and Endoscopy, Fondazione IRCCS “Casa Sollievo della Sofferenza” Hospital, Viale Cappuccini 1, 71013 San Giovanni Rotondo, FG, Italy
| | - Orazio Palmieri
- Division of Gastroenterology and Endoscopy, Fondazione IRCCS “Casa Sollievo della Sofferenza” Hospital, Viale Cappuccini 1, 71013 San Giovanni Rotondo, FG, Italy
| | - Tiziana Latiano
- Division of Gastroenterology and Endoscopy, Fondazione IRCCS “Casa Sollievo della Sofferenza” Hospital, Viale Cappuccini 1, 71013 San Giovanni Rotondo, FG, Italy
| | - Matteo Tardio
- Department of Surgery, Fondazione IRCCS “Casa Sollievo della Sofferenza” Hospital, Viale Cappuccini 1, 71013 San Giovanni Rotondo, FG, Italy
| | - Fabio Francesco di Mola
- Unit of Surgical Oncology, Casa di Cura Pierangeli, 65124 Pescara, PE, Italy
- Department of Medical, Oral and Biotechnological Sciences, University “G. d’Annunzio” Chieti-Pescara, 66100 Chieti, CH, Italy
| | - Tommaso Grottola
- Unit of Surgical Oncology, Casa di Cura Pierangeli, 65124 Pescara, PE, Italy
- Department of Innovative Technologies in Clinical Medicine and Dentistry, University “G. d’Annunzio” Chieti-Pescara, 66100 Chieti, CH, Italy
| | - Markus W. Büchler
- Botton-Champalimaud Pancreatic Cancer Center, Champalimaud Foundation, 1400-038 Lisbon, Portugal
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, 69120 Heidelberg, Germany
| | - Marco Gentile
- Division of Gastroenterology and Endoscopy, Fondazione IRCCS “Casa Sollievo della Sofferenza” Hospital, Viale Cappuccini 1, 71013 San Giovanni Rotondo, FG, Italy
| | - Anna Latiano
- Division of Gastroenterology and Endoscopy, Fondazione IRCCS “Casa Sollievo della Sofferenza” Hospital, Viale Cappuccini 1, 71013 San Giovanni Rotondo, FG, Italy
| | - Tommaso Mazza
- Bioinformatics Laboratory, Fondazione IRCCS “Casa Sollievo della Sofferenza” Hospital, Viale Cappuccini 1, 71013 San Giovanni Rotondo, FG, Italy
| | - Francesco Perri
- Division of Gastroenterology and Endoscopy, Fondazione IRCCS “Casa Sollievo della Sofferenza” Hospital, Viale Cappuccini 1, 71013 San Giovanni Rotondo, FG, Italy
| |
Collapse
|
10
|
Camisa PR, Caronni N, Crippa S. Targeting PGE2-IL-1β axis to impact future treatments for pancreatic ductal adenocarcinoma and its precursors. Expert Rev Anticancer Ther 2025; 25:5-8. [PMID: 39754391 DOI: 10.1080/14737140.2025.2450235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 12/26/2024] [Accepted: 01/03/2025] [Indexed: 01/06/2025]
Affiliation(s)
- Paolo Riccardo Camisa
- Division of Pancreatic Surgery, Pancreas Translational & Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Nicoletta Caronni
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Stefano Crippa
- Division of Pancreatic Surgery, Pancreas Translational & Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
11
|
D’Antonio DL, Zenoniani A, Umme S, Piattelli A, Curia MC. Intratumoral Fusobacterium nucleatum in Pancreatic Cancer: Current and Future Perspectives. Pathogens 2024; 14:2. [PMID: 39860963 PMCID: PMC11768203 DOI: 10.3390/pathogens14010002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 12/22/2024] [Accepted: 12/24/2024] [Indexed: 01/27/2025] Open
Abstract
The intratumoral microbiome plays a significant role in many cancers, such as lung, pancreatic, and colorectal cancer. Pancreatic cancer (PC) is one of the most lethal malignancies and is often diagnosed at advanced stages. Fusobacterium nucleatum (Fn), an anaerobic Gram-negative bacterium primarily residing in the oral cavity, has garnered significant attention for its emerging role in several extra-oral human diseases and, lately, in pancreatic cancer progression and prognosis. It is now recognized as oncobacterium. Fn engages in pancreatic tumorigenesis and metastasis through multifaceted mechanisms, including immune response modulation, virulence factors, control of cell proliferation, intestinal metabolite interactions, DNA damage, and epithelial-mesenchymal transition. Additionally, compelling research suggests that Fn may exert detrimental effects on cancer treatment outcomes. This paper extends the perspective to pancreatic cancer associated with Fn. The central focus is to unravel the oncogenomic changes driven by Fn in colonization, initiation, and promotion of pancreatic cancer development. The presence of Fusobacterium species can be considered a prognostic marker of PC, and it is also correlated to chemoresistance. Furthermore, this review underscores the clinical research significance of Fn as a potential tumor biomarker and therapeutic target, offering a novel outlook on its applicability in cancer detection and prognostic assessment. It is thought that given the role of Fn in tumor formation and metastasis processes via its FadA, FapA, Fap2, and RadD, new therapies for tumor treatment targeting Fn will be developed.
Collapse
Affiliation(s)
- Domenica Lucia D’Antonio
- Department of Medical, Oral and Biotechnological Sciences, “G. d’Annunzio” University of Chieti-Pescara, Via dei Vestini, 66100 Chieti, Italy; (D.L.D.); (A.Z.); (S.U.)
| | - Anna Zenoniani
- Department of Medical, Oral and Biotechnological Sciences, “G. d’Annunzio” University of Chieti-Pescara, Via dei Vestini, 66100 Chieti, Italy; (D.L.D.); (A.Z.); (S.U.)
| | - Samia Umme
- Department of Medical, Oral and Biotechnological Sciences, “G. d’Annunzio” University of Chieti-Pescara, Via dei Vestini, 66100 Chieti, Italy; (D.L.D.); (A.Z.); (S.U.)
- Department of Neuroscience, Imaging and Clinical Sciences, “G. d’Annunzio” University of Chieti-Pescara, Via dei Vestini, 66100 Chieti, Italy
| | - Adriano Piattelli
- School of Dentistry, Saint Camillus International University of Health and Medical Sciences (UniCamillus), 00131 Rome, Italy;
- Facultad de Medicina, UCAM Universidad Católica San Antonio de Murcia, 30107 Murcia, Spain
| | - Maria Cristina Curia
- Department of Medical, Oral and Biotechnological Sciences, “G. d’Annunzio” University of Chieti-Pescara, Via dei Vestini, 66100 Chieti, Italy; (D.L.D.); (A.Z.); (S.U.)
| |
Collapse
|
12
|
Dai H, Chen X, Yang J, Loiola RA, Lu A, Cheung KCP. Insights and therapeutic advances in pancreatic cancer: the role of electron microscopy in decoding the tumor microenvironment. Front Cell Dev Biol 2024; 12:1460544. [PMID: 39744013 PMCID: PMC11688199 DOI: 10.3389/fcell.2024.1460544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Accepted: 09/23/2024] [Indexed: 01/04/2025] Open
Abstract
Pancreatic cancer is one of the most lethal cancers, with a 5-year overall survival rate of less than 10%. Despite the development of novel therapies in recent decades, current chemotherapeutic strategies offer limited clinical benefits due to the high heterogeneity and desmoplastic tumor microenvironment (TME) of pancreatic cancer as well as inefficient drug penetration. Antibody- and nucleic acid-based targeting therapies have emerged as strong contenders in pancreatic cancer drug discovery. Numerous studies have shown that these strategies can significantly enhance drug accumulation in tumors while reducing systemic toxicity. Additionally, electron microscopy (EM) has been a critical tool for high-resolution analysis of the TME, providing insights into the ultrastructural changes associated with pancreatic cancer progression and treatment responses. This review traces the current and technological advances in EM, particularly the development of ultramicrotomy and improvements in sample preparation that have facilitated the detailed visualization of cellular and extracellular components of the TME. This review highlights the contribution of EM in assessing the efficacy of therapeutic agents, from revealing apoptotic changes to characterizing the effects of novel compounds like ionophore antibiotic gramicidin A on cellular ultrastructures. Moreover, the review delves into the potential of EM in studying the interactions between the tumor microbiome and cancer cell migration, as well as in aiding the development of targeted therapies like antibody-drug conjugates (ADCs) and aptamer-drug conjugates (ApDCs).
Collapse
Affiliation(s)
- Hong Dai
- Department of Chemistry, The Hong Kong University of Science and Technology, Kowloon, Hong Kong SAR, China
| | - Xingxuan Chen
- Phenome Research Center, School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong SAR, China
| | - Jiawen Yang
- School of Life Science, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | | | - Aiping Lu
- Phenome Research Center, School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong SAR, China
| | - Kenneth C. P. Cheung
- Phenome Research Center, School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong SAR, China
| |
Collapse
|
13
|
Huang L, Jiang C, Yan M, Wan W, Li S, Xiang Z, Wu J. The oral-gut microbiome axis in breast cancer: from basic research to therapeutic applications. Front Cell Infect Microbiol 2024; 14:1413266. [PMID: 39639864 PMCID: PMC11617537 DOI: 10.3389/fcimb.2024.1413266] [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] [Scholar Register] [Received: 04/07/2024] [Accepted: 11/01/2024] [Indexed: 12/07/2024] Open
Abstract
As a complicated and heterogeneous condition, breast cancer (BC) has posed a tremendous public health challenge across the world. Recent studies have uncovered the crucial effect of human microbiota on various perspectives of health and disease, which include cancer. The oral-gut microbiome axis, particularly, have been implicated in the occurrence and development of colorectal cancer through their intricate interactions with host immune system and modulation of systemic inflammation. However, the research concerning the impact of oral-gut microbiome axis on BC remains scarce. This study focused on comprehensively reviewing and summarizing the latest ideas about the potential bidirectional relation of the gut with oral microbiota in BC, emphasizing their potential impact on tumorigenesis, treatment response, and overall patient outcomes. This review can reveal the prospect of tumor microecology and propose a novel viewpoint that the oral-gut microbiome axis can be a breakthrough point in future BC studies.
Collapse
Affiliation(s)
- Lan Huang
- Department of Clinical Laboratory, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, China
| | - Chun Jiang
- Department of Clinical Laboratory, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, China
| | - Meina Yan
- Department of Clinical Laboratory, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, China
| | - Weimin Wan
- Department of Clinical Laboratory, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, China
| | - Shuxiang Li
- Department of Clinical Laboratory, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, China
| | - Ze Xiang
- Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jian Wu
- Department of Clinical Laboratory, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, China
| |
Collapse
|
14
|
Li P, Zhang H, Dai M. Current status and prospect of gut and oral microbiome in pancreatic cancer: Clinical and translational perspectives. Cancer Lett 2024; 604:217274. [PMID: 39307411 DOI: 10.1016/j.canlet.2024.217274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 09/18/2024] [Accepted: 09/19/2024] [Indexed: 09/27/2024]
Abstract
Pancreatic cancer is a highly lethal malignancy, and its diagnosis and treatment continue to pose significant challenges. Despite advancements in surgical and comprehensive treatment methods, the five-year survival rate remains below 12 %. With the rapid development of microbiome science, the gut and oral microbiota, which are readily accessible and can be sampled non-invasively, have emerged as a novel area of interest in pancreatic cancer research. Dysbiosis in these microbial communities can induce persistent inflammatory responses and affect the host's immune system, promoting cancer development and impacting the efficacy of treatments like chemotherapy and immunotherapy. This review provides an up-to-date overview of the roles of both gut and oral microbiota in the onset, progression, diagnosis, and treatment of pancreatic cancer. It analyzes the potential of utilizing these microbiomes as biomarkers and therapeutic targets from a clinical application perspective. Furthermore, it discusses future research directions aimed at harnessing these insights to advance the diagnosis and treatment strategies for pancreatic cancer. By focusing on the microbiome's role in clinical and translational medicine, this review offers insights into improving pancreatic cancer diagnosis and treatment outcomes.
Collapse
Affiliation(s)
- Pengyu Li
- Department of General Surgery, Peking Union Medical College Hospital (PUMCH), Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Hanyu Zhang
- Department of General Surgery, Peking Union Medical College Hospital (PUMCH), Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China; Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Menghua Dai
- Department of General Surgery, Peking Union Medical College Hospital (PUMCH), Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.
| |
Collapse
|
15
|
van Eijck CWF, Ju J, van 't Land FR, Verheij M, Li Y, Stubbs A, Doukas M, Lila K, Heij LR, Wiltberger G, Alonso L, Malats N, Groot Koerkamp B, Vietsch EE, van Eijck CHJ. The tumor immune microenvironment in resected treatment-naive pancreatic cancer patients with long-term survival. Pancreatology 2024; 24:1057-1065. [PMID: 39218754 DOI: 10.1016/j.pan.2024.08.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 07/27/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal cancers worldwide. Presently, only a fraction of patients undergo successful surgical resection, the most effective treatment. Enhancing treatment strategies necessitates a deep comprehension of the factors underlying extended survival after surgical resection in patients. METHODS This study aims to identify the important factors of PDAC patients' long-term survival with metatranscriptomics and multiplex immunofluorescence (IF) staining analyses. Specifically, differences in tumor immune microenvironment (TIME) were investigated between treatment-naïve PDAC short-term survivors (STS, overall survival <6 months) and long-term survivors (LTS, overall survival >5 years). RESULTS As a result, we detected 589 over-expressed genes, including HOXB9, CDA, and HOXB8, and 507 under-expressed genes, including AMY2B, SCARA5, and SLC2A2 in LTS. Most of the Reactome overbiological pathways enriched in our data were over-expressed in LTS, such as RHO GTPase Effectors and Cell Cycle Checkpoints. Eleven microbiomes significantly differed between LTS and STS, including Sphingopyxis and Capnocytophaga. Importantly, we demonstrate that the TIME profile with an increased abundance of memory B cells and the reduction of M0 and pro-tumoral M2 macrophages are associated with a good prognosis in PDAC. CONCLUSIONS In this study, we delved into the TIME with metatranscriptomics and IF staining analyses to understand the prerequisite of prolonged survival in PDAC patients. In LTS, several biological pathways were overexpressed, and specific microbiomes were identified. Furthermore, apparent differences in driven immune factors were found that provide valuable insights into developing new treatment strategies.
Collapse
Affiliation(s)
- Casper W F van Eijck
- Erasmus MC Cancer Institute, Department of Surgery, University Medical Center Rotterdam, the Netherlands; Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain.
| | - Jie Ju
- Department of Pathology and Clinical Bioinformatics, Erasmus University Medical Center Rotterdam, the Netherlands
| | - Freek R van 't Land
- Erasmus MC Cancer Institute, Department of Surgery, University Medical Center Rotterdam, the Netherlands
| | - Maaike Verheij
- Erasmus MC Cancer Institute, Department of Surgery, University Medical Center Rotterdam, the Netherlands
| | - Yunlei Li
- Department of Pathology and Clinical Bioinformatics, Erasmus University Medical Center Rotterdam, the Netherlands
| | - Andrew Stubbs
- Department of Pathology and Clinical Bioinformatics, Erasmus University Medical Center Rotterdam, the Netherlands
| | - Michael Doukas
- Department of Pathology and Clinical Bioinformatics, Erasmus University Medical Center Rotterdam, the Netherlands
| | - Karishma Lila
- Department of Pathology and Clinical Bioinformatics, Erasmus University Medical Center Rotterdam, the Netherlands
| | - Lara R Heij
- Institute of Pathology, Medical Center University Duisburg-Essen, Essen, Germany; Department of Surgery and Transplantation, University Hospital Essen, Germany; Department of General, Gastrointestinal, Hepatobiliary and Transplant Surgery, RWTH Aachen University Hospital, Aachen, Germany
| | - Georg Wiltberger
- Department of General, Gastrointestinal, Hepatobiliary and Transplant Surgery, RWTH Aachen University Hospital, Aachen, Germany
| | - Lola Alonso
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Núria Malats
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Bas Groot Koerkamp
- Erasmus MC Cancer Institute, Department of Surgery, University Medical Center Rotterdam, the Netherlands
| | - Eveline E Vietsch
- Erasmus MC Cancer Institute, Department of Surgery, University Medical Center Rotterdam, the Netherlands
| | - Casper H J van Eijck
- Erasmus MC Cancer Institute, Department of Surgery, University Medical Center Rotterdam, the Netherlands; Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| |
Collapse
|
16
|
Lombardo C, Fazio R, Sinagra M, Gattuso G, Longo F, Lombardo C, Salmeri M, Zanghì GN, Loreto CAE. Intratumoral Microbiota: Insights from Anatomical, Molecular, and Clinical Perspectives. J Pers Med 2024; 14:1083. [PMID: 39590575 PMCID: PMC11595780 DOI: 10.3390/jpm14111083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 10/26/2024] [Accepted: 10/29/2024] [Indexed: 11/28/2024] Open
Abstract
The human microbiota represents a heterogeneous microbial community composed of several commensal, symbiotic, and even pathogenic microorganisms colonizing both the external and internal body surfaces. Despite the term "microbiota" being commonly used to identify microorganisms inhabiting the gut, several pieces of evidence suggest the presence of different microbiota physiologically colonizing other organs. In this context, several studies have also confirmed that microbes are integral components of tumor tissue in different types of cancer, constituting the so-called "intratumoral microbiota". The intratumoral microbiota is closely related to the occurrence and development of cancer as well as to the efficacy of anticancer treatments. Indeed, intratumoral microbiota can contribute to carcinogenesis and metastasis formation as some microbes can directly cause DNA damage, while others can induce the activation of proinflammatory responses or oncogenic pathways and alter the tumor microenvironment (TME). All these characteristics make the intratumoral microbiota an interesting topic to investigate for both diagnostic and prognostic purposes in order to improve the management of cancer patients. This review aims to gather the most recent data on the role of the intratumoral microbiota in cancer development, progression, and response to treatment, as well as its potential diagnostic and prognostic value.
Collapse
Affiliation(s)
- Claudia Lombardo
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (C.L.); (R.F.); (M.S.); (C.L.); (M.S.); (C.A.E.L.)
| | - Rosanna Fazio
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (C.L.); (R.F.); (M.S.); (C.L.); (M.S.); (C.A.E.L.)
| | - Marta Sinagra
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (C.L.); (R.F.); (M.S.); (C.L.); (M.S.); (C.A.E.L.)
| | - Giuseppe Gattuso
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (C.L.); (R.F.); (M.S.); (C.L.); (M.S.); (C.A.E.L.)
| | - Federica Longo
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (C.L.); (R.F.); (M.S.); (C.L.); (M.S.); (C.A.E.L.)
| | - Cinzia Lombardo
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (C.L.); (R.F.); (M.S.); (C.L.); (M.S.); (C.A.E.L.)
| | - Mario Salmeri
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (C.L.); (R.F.); (M.S.); (C.L.); (M.S.); (C.A.E.L.)
| | - Guido Nicola Zanghì
- Department of General Surgery and Medical-Surgical Specialties, Policlinico-Vittorio Emanuele Hospital, University of Catania, 95123 Catania, Italy;
| | - Carla Agata Erika Loreto
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (C.L.); (R.F.); (M.S.); (C.L.); (M.S.); (C.A.E.L.)
| |
Collapse
|
17
|
Mi S, Cai S, Lou G, Xue M. Two-sample Mendelian randomization analysis of the relationship between periodontitis and risk of upper gastrointestinal cancers. Postgrad Med J 2024; 100:820-826. [PMID: 38840504 DOI: 10.1093/postmj/qgae069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 05/02/2024] [Accepted: 05/23/2024] [Indexed: 06/07/2024]
Abstract
PURPOSE The aim of the present study is to explore the possible association between periodontitis and upper gastrointestinal (UGI) cancers, including esophageal and gastric cancers, utilizing the Mendelian randomization method. METHODS In this research, we utilized the Mendelian randomization method to examine the causal association between periodontitis and UGI cancers. Genome-wide association studies data for periodontitis were obtained from the Gene-Lifestyle Interactions in Dental Endpoints consortium, while UGI cancers' data were accessed from FinnGen's Biobank. After rigorously screening instrumental variables for periodontitis, we analyzed them with UGI cancers primarily using the inverse variance weighted. Finally, to identify outliers, the results were subjected to a leave-one-out sensitivity analysis. RESULTS Inverse variance weighted (fixed effect) results revealed that periodontitis is a risk factor for gastric cancer (OR = 1.7735, 95% CI: 1.1576 to 2.7170, P = 0.0085). As for esophageal cancer, no statistically significant correlation was observed. Furthermore, no outliers were detected in any of the results. CONCLUSION Our two-sample Mendelian randomization study obviously demonstrates a significant positive association between periodontitis and gastric cancer, while no statistically significant correlation was found for esophageal cancer.
Collapse
Affiliation(s)
- Shuyi Mi
- Department of Gastroenterology, The Second Affiliated Hospital of Zhejiang University School of Medicine, No.88 Jiefang Road, Shangcheng District, Hangzhou, Zhejiang, 310009, China
- Institute of Gastroenterology, Zhejiang University, Hangzhou, Zhejiang, 310009, China
| | - Shangwen Cai
- Department of Gastroenterology, The Second Affiliated Hospital of Zhejiang University School of Medicine, No.88 Jiefang Road, Shangcheng District, Hangzhou, Zhejiang, 310009, China
- Institute of Gastroenterology, Zhejiang University, Hangzhou, Zhejiang, 310009, China
| | - Guochun Lou
- Department of Gastroenterology, The Second Affiliated Hospital of Zhejiang University School of Medicine, No.88 Jiefang Road, Shangcheng District, Hangzhou, Zhejiang, 310009, China
- Institute of Gastroenterology, Zhejiang University, Hangzhou, Zhejiang, 310009, China
| | - Meng Xue
- Department of Gastroenterology, The Second Affiliated Hospital of Zhejiang University School of Medicine, No.88 Jiefang Road, Shangcheng District, Hangzhou, Zhejiang, 310009, China
- Institute of Gastroenterology, Zhejiang University, Hangzhou, Zhejiang, 310009, China
| |
Collapse
|
18
|
Wu J, Li J, Yan M, Xiang Z. Gut and oral microbiota in gynecological cancers: interaction, mechanism, and therapeutic value. NPJ Biofilms Microbiomes 2024; 10:104. [PMID: 39389989 PMCID: PMC11467339 DOI: 10.1038/s41522-024-00577-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 09/29/2024] [Indexed: 10/12/2024] Open
Abstract
Gynecologic cancers develop from the female reproductive organs. Microbial dysbiosis in the gut and oral cavity can communicate with each other through various ways, leading to mucosal destruction, inflammatory response, genomic instability, and ultimately inducing cancer and worsening. Here, we introduce the mechanisms of interactions between gut and oral microbiota and their changes in the development of gynecologic tumors. In addition, new therapeutic approaches based on microbiota modulation are discussed.
Collapse
Affiliation(s)
- Jian Wu
- Department of Clinical Laboratory, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, China.
| | - Jiarui Li
- Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Meina Yan
- Department of Clinical Laboratory, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, China
| | - Ze Xiang
- Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| |
Collapse
|
19
|
Merali N, Chouari T, Sweeney C, Halle-Smith J, Jessel MD, Wang B, O’ Brien J, Suyama S, Jiménez JI, Roberts KJ, Velliou E, Sivakumar S, Rockall TA, Demirkan A, Pedicord V, Deng D, Giovannetti E, Annels NE, Frampton AE. The microbial composition of pancreatic ductal adenocarcinoma: a systematic review of 16S rRNA gene sequencing. Int J Surg 2024; 110:6771-6799. [PMID: 38874485 PMCID: PMC11487005 DOI: 10.1097/js9.0000000000001762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 05/24/2024] [Indexed: 06/15/2024]
Abstract
BACKGROUND Pancreatic cancer, specifically pancreatic ductal adenocarcinoma (PDAC), continues to pose a significant clinical and scientific challenge. The most significant finding of recent years is that PDAC tumours harbour their specific microbiome, which differs amongst tumour entities and is distinct from healthy tissue. This review aims to evaluate and summarise all PDAC studies that have used the next-generation technique, 16S rRNA gene amplicon sequencing within each bodily compartment. As well as establishing a causal relationship between PDAC and the microbiome. MATERIALS AND METHODS This systematic review was carried out according to the Preferred Reporting Items for Systematic Reviews and Meta-analysis (PRISMA) guidelines. A comprehensive search strategy was designed, and 1727 studies were analysed. RESULTS In total, 38 studies were selected for qualitative analysis and summarised significant PDAC bacterial signatures. Despite the growing amount of data provided, we are not able to state a universal 16S rRNA gene microbial signature that can be used for PDAC screening. This is most certainly due to the heterogeneity of the presentation of results, lack of available datasets, and the intrinsic selection bias between studies. CONCLUSION Several key studies have begun to shed light on causality and the influence the microbiome constituents and their produced metabolites could play in tumorigenesis and influencing outcomes. The challenge in this field is to shape the available microbial data into targetable signatures. Making sequenced data readily available is critical, coupled with the coordinated standardisation of data and the need for consensus guidelines in studies investigating the microbiome in PDAC.
Collapse
Affiliation(s)
- Nabeel Merali
- Minimal Access Therapy Training Unit (MATTU), Royal Surrey Hospital NHS Foundation Trust
- Department of Hepato-Pancreato-Biliary (HPB) Surgery, Royal Surrey County Hospital NHS Foundation Trust
- Section of Oncology, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Science, University of Surrey
| | - Tarak Chouari
- Minimal Access Therapy Training Unit (MATTU), Royal Surrey Hospital NHS Foundation Trust
- Section of Oncology, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Science, University of Surrey
| | - Casie Sweeney
- Minimal Access Therapy Training Unit (MATTU), Royal Surrey Hospital NHS Foundation Trust
| | - James Halle-Smith
- Hepatobiliary and Pancreatic Surgery Unit, Queen Elizabeth Hospital Birmingham, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Maria-Danae Jessel
- Section of Oncology, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Science, University of Surrey
| | - Bing Wang
- Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam
| | - James O’ Brien
- Minimal Access Therapy Training Unit (MATTU), Royal Surrey Hospital NHS Foundation Trust
| | - Satoshi Suyama
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge
| | | | - Keith J. Roberts
- Hepatobiliary and Pancreatic Surgery Unit, Queen Elizabeth Hospital Birmingham, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Eirini Velliou
- Centre for 3D Models of Health and Disease, Division of Surgery and Interventional Science, University College London (UCL), London
| | - Shivan Sivakumar
- Oncology Department and Institute of Immunology and Immunotherapy, Birmingham Medical School, University of Birmingham
| | - Timothy A. Rockall
- Minimal Access Therapy Training Unit (MATTU), Royal Surrey Hospital NHS Foundation Trust
| | - Ayse Demirkan
- Section of Statistical Multi-Omics, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Science, University of Surrey
- Surrey Institute for People-Centred AI, University of Surrey, Guildford, Surrey
| | - Virginia Pedicord
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge
| | - Dongmei Deng
- Department of Preventive Dentistry, Academic Center for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Elisa Giovannetti
- Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam
- Fondazione Pisa per la Scienza, San Giuliano, Italy
| | - Nicola E. Annels
- Section of Oncology, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Science, University of Surrey
| | - Adam E. Frampton
- Minimal Access Therapy Training Unit (MATTU), Royal Surrey Hospital NHS Foundation Trust
- Department of Hepato-Pancreato-Biliary (HPB) Surgery, Royal Surrey County Hospital NHS Foundation Trust
- Section of Oncology, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Science, University of Surrey
| |
Collapse
|
20
|
Baima G, Minoli M, Michaud DS, Aimetti M, Sanz M, Loos BG, Romandini M. Periodontitis and risk of cancer: Mechanistic evidence. Periodontol 2000 2024; 96:83-94. [PMID: 38102837 PMCID: PMC11579815 DOI: 10.1111/prd.12540] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 10/24/2023] [Accepted: 11/01/2023] [Indexed: 12/17/2023]
Abstract
This review aims to critically analyze the pathways of interaction and the pathogenic mechanisms linking periodontitis and oral bacteria with the initiation/progression of cancer at different body compartments. A higher risk of head and neck cancer has been consistently associated with periodontitis. This relationship has been explained by the local promotion of dysbiosis, chronic inflammation, immune evasion, and direct (epi)genetic damage to epithelial cells by periodontal pathobionts and their toxins. Epidemiological reports have also studied a possible link between periodontitis and the incidence of other malignancies at distant sites, such as lung, breast, prostate, and digestive tract cancers. Mechanistically, different pathways have been involved, including the induction of a chronic systemic inflammatory state and the spreading of oral pathobionts with carcinogenic potential. Indeed, periodontitis may promote low-grade systemic inflammation and phenotypic changes in the mononuclear cells, leading to the release of free radicals and cytokines, as well as extracellular matrix degradation, which are all mechanisms involved in carcinogenic and metastatic processes. Moreover, the transient hematogenous spill out or micro-aspiration/swallowing of periodontal bacteria and their virulence factors (i.e., lipopolysaccharides, fimbriae), may lead to non-indigenous bacterial colonization of multiple microenvironments. These events may in turn replenish the tumor-associated microbiome and thus influence the molecular hallmarks of cancer. Particularly, specific strains of oral pathobionts (e.g., Porphyromonas gingivalis and Fusobacterium nucleatum) may translocate through the hematogenous and enteral routes, being implicated in esophageal, gastric, pancreatic, and colorectal tumorigenesis through the modulation of the gastrointestinal antitumor immune system (i.e., tumor-infiltrating T cells) and the increased expression of pro-inflammatory/oncogenic genes. Ultimately, the potential influence of common risk factors, relevant comorbidities, and upstream drivers, such as gerovulnerability to multiple diseases, in explaining the relationship cannot be disregarded. The evidence analyzed here emphasizes the possible relevance of periodontitis in cancer initiation/progression and stimulates future research endeavors.
Collapse
Affiliation(s)
- Giacomo Baima
- Department of Surgical Sciences, C.I.R. Dental SchoolUniversity of TurinTurinItaly
| | - Margherita Minoli
- Department of PeriodontologyUniversità Vita‐Salute San RaffaeleMilanItaly
| | - Dominique S. Michaud
- Department of Public Health and Community MedicineTufts University School of MedicineBostonMassachusettsUSA
| | - Mario Aimetti
- Department of Surgical Sciences, C.I.R. Dental SchoolUniversity of TurinTurinItaly
| | - Mariano Sanz
- Faculty of OdontologyUniversity ComplutenseMadridSpain
- Department of Periodontology, Faculty of DentistryUniversity of OsloOsloNorway
| | - Bruno G. Loos
- Department of Periodontology, ACTA ‐ Academic Centre for Dentistry AmsterdamUniversity of Amsterdam and Vrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Mario Romandini
- Department of Periodontology, Faculty of DentistryUniversity of OsloOsloNorway
| |
Collapse
|
21
|
Parveen S, Alqahtani AS, Aljabri MY, Dawood T, Khan SS, Gupta B, Vempalli S, Hassan AAHAA, Elamin NMH. Exploring the Interplay: Oral–Gut Microbiome Connection and the Impact of Diet and Nutrition. EUROPEAN JOURNAL OF GENERAL DENTISTRY 2024; 13:165-176. [DOI: 10.1055/s-0044-1786154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
AbstractThe intricate interplay between the oral and intestinal microbiota holds increasing fascination within the context of health and nutrition. Serving as the gateway to the gastrointestinal tract, the oral microbiota hosts a diverse array of microbial species that significantly influence well-being or contribute to various diseases. Dysbiosis in the oral microbiota has been linked to conditions such as dental caries, periodontal diseases, and systemic disorders, including diabetes, cardiovascular disease, obesity, rheumatoid arthritis, Alzheimer's disease, and colorectal cancer. This review aims to comprehend the nuanced relationship between oral and intestinal microbiotas, exploring the pivotal role of diet in developing strategies for wellness promotion and disease prevention. Drawing insights from a myriad of studies encompassing both animals and humans, we examine the implications of microbial dysbiosis and its impact on health. A bibliographic search of 78 scientific articles was conducted across PubMed Central, Web of Science, Scopus, Google Scholar, and the Saudi digital library from January 2000 to August 2023. Following a rigorous screening process, the full texts of selected articles were critically reviewed to extract relevant information. Articles not meeting the inclusion criteria—specifically focused on oral–intestinal microbiota interaction and diet and nutrition—were meticulously excluded. Diet emerges as a key player in influencing both oral and intestinal microbiotas. Various dietary components, such as fiber, prebiotics, probiotics, and bioactive compounds, have demonstrated significant effects on the diversity and function of microorganisms in these ecosystems. Conversely, diets high in processed foods, added sugars, and saturated fats correlate with dysbiosis and an elevated risk of oral and gastrointestinal diseases. Understanding the intricacies of this interaction is paramount for the development of innovative approaches fostering a balanced oral–gut microbiota axis and improving overall human health. The implications extend to preventive and therapeutic interventions, emphasizing the practical importance of unraveling these complexities for public health and clinical practice. This comprehensive review delves into the intricate relationship between gut and oral microbiota, shedding light on their roles in various diseases, particularly focusing on oral diseases. Key findings are summarized, and implications for future research and clinical practice are discussed. In conclusion, the review underscores the urgent need for special attention to key microbiota in developing targeted interventions for promoting oral and gut health.
Collapse
Affiliation(s)
- Sameena Parveen
- Department of Maxillofacial Surgery and Diagnostic Sciences, College of Dentistry, Jazan University, Jazan, Kingdom of Saudi Arabia
| | - Ahmed Shaher Alqahtani
- Department of Maxillofacial Surgery and Diagnostic Sciences, College of Dentistry, Jazan University, Jazan, Kingdom of Saudi Arabia
| | - Mohammed Y. Aljabri
- Department of Maxillofacial Surgery and Diagnostic Sciences, College of Dentistry, Jazan University, Jazan, Kingdom of Saudi Arabia
| | - Tazeen Dawood
- Department of Preventive Dental Sciences, College of Dentistry, Jazan University, Jazan, Kingdom of Saudi Arabia
| | - Samar Saeed Khan
- Department of Maxillofacial Surgery and Diagnostic Sciences, College of Dentistry, Jazan University, Jazan, Kingdom of Saudi Arabia
| | - Bharti Gupta
- Department of Maxillofacial Surgery and Diagnostic Sciences, College of Dentistry, Jazan University, Jazan, Kingdom of Saudi Arabia
| | - Swetha Vempalli
- Department of Maxillofacial Surgery and Diagnostic Sciences, College of Dentistry, Jazan University, Jazan, Kingdom of Saudi Arabia
| | | | - Nahid Mahmoud Hassan Elamin
- Department of Maxillofacial Surgery and Diagnostic Sciences, College of Dentistry, Jazan University, Jazan, Kingdom of Saudi Arabia
| |
Collapse
|
22
|
Sheng D, Jin C, Yue K, Yue M, Liang Y, Xue X, Li P, Zhao G, Zhang L. Pan-cancer atlas of tumor-resident microbiome, immunity and prognosis. Cancer Lett 2024; 598:217077. [PMID: 38908541 DOI: 10.1016/j.canlet.2024.217077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 05/23/2024] [Accepted: 06/14/2024] [Indexed: 06/24/2024]
Abstract
The existence of microbiome in human tumors has been determined widely, but evaluating the contribution of intratumoral bacteria and fungi to tumor immunity and prognosis from a pan-cancer perspective remains absent. We designed an improved microbial analysis pipeline to reduce interference from host sequences, complemented with integration analysis of intratumoral microbiota at species level with clinical indicators, tumor microenvironment, and prognosis across cancer types. We found that intratumoral microbiota is associated with immunophenotyping, with high-immunity subtypes showing greater bacterial and fungal richness compared to low-immunity groups. We also noted that the combination of fungi and bacteria demonstrated promising prognostic value across cancer types. We, thus, present The Cancer Microbiota (TCMbio), an interactive platform that provides the intratumoral bacteria and fungi data, and a comprehensive analysis module for 33 types of cancers. This led to the discovery of clinical and prognostic significance of intratumoral microbes.
Collapse
Affiliation(s)
- Dashuang Sheng
- Microbiome-X, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China; State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| | - Chuandi Jin
- Microbiome-X, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Kaile Yue
- Microbiome-X, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Min Yue
- Microbiome-X, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yijia Liang
- Microbiome-X, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xinxin Xue
- Microbiome-X, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Pingfu Li
- Shandong Huxley Medical Technology Co.,Ltd., Jinan, China
| | - Guoping Zhao
- Microbiome-X, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China; State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China; CAS Key Laboratory of Computational Biology, Bio-Med Big Data Center, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China.
| | - Lei Zhang
- Microbiome-X, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China; State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China.
| |
Collapse
|
23
|
Lou F, Li M, Chu T, Duan H, Liu H, Zhang J, Duan K, Liu H, Wei F. Comprehensive analysis of clinical data and radiomic features from contrast enhanced CT for differentiating benign and malignant pancreatic intraductal papillary mucinous neoplasms. Sci Rep 2024; 14:17218. [PMID: 39060387 PMCID: PMC11282090 DOI: 10.1038/s41598-024-68067-6] [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] [Scholar Register] [Received: 04/19/2024] [Accepted: 07/19/2024] [Indexed: 07/28/2024] Open
Abstract
The primary aim of this investigation was to leverage radiomics features derived from contrast-enhanced abdominal computed tomography (CT) scans to devise a predictive model to discern the benign and malignant nature of intraductal papillary mucinous neoplasms (IPMNs). Radiomic signatures were meticulously crafted to delineate benign from malignant IPMNs by extracting pertinent features from contrast-enhanced CT images within a designated training cohort (n = 84). Subsequent validation was conducted with data from an independent test cohort (n = 37). The discriminative ability of the model was quantitatively evaluated through receiver operating characteristic (ROC) curve analysis, with the integration of carefully selected clinical features to improve the comparative analysis. Arterial-phase images were utilized to construct a model comprising 8 features for distinguishing between benign and malignant cases. The model achieved an accuracy of 0.891 [95% confidence interval (95% CI), 0.816-0.996] in the cross-validation set and 0.553 (95% CI 0.360-0.745) in the test set. Conversely, employing 9 features from the venous-phase resulted in a model with a cross-validation accuracy of 0.862 (95%CI 0.777-0.946) and a test set accuracy of 0.801 (95% CI 0.653-0.950).Integrating the identified clinical features with imaging features yielded a model with a cross-validation accuracy of 0.934 (95% CI 0.879-0.990) and a test set accuracy of 0.904 (95% CI 0.808-0.999), thereby further improving its discriminatory ability. Our findings distinctly illustrate that venous-phase radiomics features eclipse arterial-phase radiomic features in terms of predictive accuracy regarding the nature of IPMNs. Furthermore, the synthesis and meticulous screening of clinical features with radiomic data significantly increased the diagnostic efficacy of our model, underscoring the pivotal importance of a comprehensive and integrated approach for accurate risk stratification in IPMN management.
Collapse
Affiliation(s)
- Fengxiang Lou
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, 130000, China
| | - Mingyang Li
- Department of Radiology, The First Hospital of Jilin University, Changchun, 130000, China
| | - Tongjia Chu
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, 130000, China
| | - Haoyu Duan
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, 130000, China
| | - Huan Liu
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, 130000, China
| | - Jian Zhang
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, 130000, China
| | - Kehang Duan
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, 130000, China
| | - Han Liu
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, 130000, China
| | - Feng Wei
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, 130000, China.
| |
Collapse
|
24
|
Hering M, Madi A, Sandhoff R, Ma S, Wu J, Mieg A, Richter K, Mohr K, Knabe N, Stichling D, Poschet G, Bestvater F, Frank L, Utikal J, Umansky V, Cui G. Sphinganine recruits TLR4 adaptors in macrophages and promotes inflammation in murine models of sepsis and melanoma. Nat Commun 2024; 15:6067. [PMID: 39025856 PMCID: PMC11258287 DOI: 10.1038/s41467-024-50341-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Accepted: 07/08/2024] [Indexed: 07/20/2024] Open
Abstract
After recognizing its ligand lipopolysaccharide, Toll-like receptor 4 (TLR4) recruits adaptor proteins to the cell membrane, thereby initiating downstream signaling and triggering inflammation. Whether this recruitment of adaptor proteins is dependent solely on protein-protein interactions is unknown. Here, we report that the sphingolipid sphinganine physically interacts with the adaptor proteins MyD88 and TIRAP and promotes MyD88 recruitment in macrophages. Myeloid cell-specific deficiency in serine palmitoyltransferase long chain base subunit 2, which encodes the key enzyme catalyzing sphingolipid biosynthesis, decreases the membrane recruitment of MyD88 and inhibits inflammatory responses in in vitro bone marrow-derived macrophage and in vivo sepsis models. In a melanoma mouse model, serine palmitoyltransferase long chain base subunit 2 deficiency decreases anti-tumor myeloid cell responses and increases tumor growth. Therefore, sphinganine biosynthesis is required for the initiation of TLR4 signal transduction and serves as a checkpoint for macrophage pattern recognition in sepsis and melanoma mouse models.
Collapse
Affiliation(s)
- Marvin Hering
- T Cell Metabolism Group (D192), German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany.
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim (UMM), Ruprecht-Karls University of Heidelberg, Mannheim, Germany.
- DKFZ Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany.
| | - Alaa Madi
- T Cell Metabolism Group (D192), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Roger Sandhoff
- Lipid Pathobiochemistry Group (A411), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sicong Ma
- T Cell Metabolism Group (D192), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, 230601, China
| | - Jingxia Wu
- T Cell Metabolism Group (D192), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, 230601, China
| | - Alessa Mieg
- T Cell Metabolism Group (D192), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Karsten Richter
- Electron Microscopy Core Facility (W230), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Kerstin Mohr
- T Cell Metabolism Group (D192), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Nora Knabe
- T Cell Metabolism Group (D192), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Helmholtz Institute for Translational Oncology, Mainz (HI-TRON Mainz)-A Helmholtz Institute of the DKFZ, Mainz, Germany
| | - Diana Stichling
- T Cell Metabolism Group (D192), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Gernot Poschet
- Metabolomics Core Technology Platform, Centre for Organismal Studies (COS), Heidelberg University, Heidelberg, Germany
| | - Felix Bestvater
- Light Microscopy Core Facility (W210), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Larissa Frank
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
- Division of Cellular Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jochen Utikal
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim (UMM), Ruprecht-Karls University of Heidelberg, Mannheim, Germany
- DKFZ Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany
| | - Viktor Umansky
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim (UMM), Ruprecht-Karls University of Heidelberg, Mannheim, Germany
- DKFZ Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany
- Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Guoliang Cui
- T Cell Metabolism Group (D192), German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany.
- Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, 230601, China.
- Helmholtz Institute for Translational Oncology, Mainz (HI-TRON Mainz)-A Helmholtz Institute of the DKFZ, Mainz, Germany.
- Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| |
Collapse
|
25
|
Zhang P, Shi H, Guo R, Li L, Guo X, Yang H, Chang D, Cheng Y, Zhao G, Li S, Zhong Q, Zhang H, Zhao P, Fu C, Song Y, Yang L, Wang Y, Zhang Y, Jiang J, Wang T, Zhao J, Li Y, Wang B, Chen F, Zhao H, Wang Y, Wang J, Ma S. Metagenomic analysis reveals altered gut virome and diagnostic potential in pancreatic cancer. J Med Virol 2024; 96:e29809. [PMID: 39016466 DOI: 10.1002/jmv.29809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 06/15/2024] [Accepted: 07/08/2024] [Indexed: 07/18/2024]
Abstract
Pancreatic cancer (PC) is a highly aggressive malignancy with a poor prognosis, making early diagnosis crucial for improving patient outcomes. While the gut microbiome, including bacteria and viruses, is believed to be essential in cancer pathogenicity, the potential contribution of the gut virome to PC remains largely unexplored. In this study, we conducted a comparative analysis of the gut viral compositional and functional profiles between PC patients and healthy controls, based on fecal metagenomes from two publicly available data sets comprising a total of 101 patients and 82 healthy controls. Our results revealed a decreasing trend in the gut virome diversity of PC patients with disease severity. We identified significant alterations in the overall viral structure of PC patients, with a meta-analysis revealing 219 viral operational taxonomic units (vOTUs) showing significant differences in relative abundance between patients and healthy controls. Among these, 65 vOTUs were enriched in PC patients, and 154 were reduced. Host prediction revealed that PC-enriched vOTUs preferentially infected bacterial members of Veillonellaceae, Enterobacteriaceae, Fusobacteriaceae, and Streptococcaceae, while PC-reduced vOTUs were more likely to infect Ruminococcaceae, Lachnospiraceae, Clostridiaceae, Oscillospiraceae, and Peptostreptococcaceae. Furthermore, we constructed random forest models based on the PC-associated vOTUs, achieving an optimal average area under the curve (AUC) of up to 0.879 for distinguishing patients from controls. Through additional 10 public cohorts, we demonstrated the reproducibility and high specificity of these viral signatures. Our study suggests that the gut virome may play a role in PC development and could serve as a promising target for PC diagnosis and therapeutic intervention. Future studies should further explore the underlying mechanisms of gut virus-bacteria interactions and validate the diagnostic models in larger and more diverse populations.
Collapse
Affiliation(s)
- Pan Zhang
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Shaanxi Key Laboratory of Gastrointestinal Motility Disorders, Xi'an, Shaanxi, China
- Shaanxi Provincial Clinical Research Center for Gastrointestinal Diseases, Xi'an, Shaanxi, China
- Digestive Disease Quality Control Center of Shaanxi Province, Xi'an, Shaanxi, China
| | - Haitao Shi
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Shaanxi Key Laboratory of Gastrointestinal Motility Disorders, Xi'an, Shaanxi, China
- Shaanxi Provincial Clinical Research Center for Gastrointestinal Diseases, Xi'an, Shaanxi, China
- Digestive Disease Quality Control Center of Shaanxi Province, Xi'an, Shaanxi, China
| | - Ruochun Guo
- Puensum Genetech Institute, Wuhan, Hubei, China
| | - Lu Li
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Shaanxi Key Laboratory of Gastrointestinal Motility Disorders, Xi'an, Shaanxi, China
- Shaanxi Provincial Clinical Research Center for Gastrointestinal Diseases, Xi'an, Shaanxi, China
- Digestive Disease Quality Control Center of Shaanxi Province, Xi'an, Shaanxi, China
| | - Xiaoyan Guo
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Shaanxi Key Laboratory of Gastrointestinal Motility Disorders, Xi'an, Shaanxi, China
- Shaanxi Provincial Clinical Research Center for Gastrointestinal Diseases, Xi'an, Shaanxi, China
- Digestive Disease Quality Control Center of Shaanxi Province, Xi'an, Shaanxi, China
| | - Hui Yang
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Shaanxi Key Laboratory of Gastrointestinal Motility Disorders, Xi'an, Shaanxi, China
- Shaanxi Provincial Clinical Research Center for Gastrointestinal Diseases, Xi'an, Shaanxi, China
- Digestive Disease Quality Control Center of Shaanxi Province, Xi'an, Shaanxi, China
| | - Danyan Chang
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Shaanxi Key Laboratory of Gastrointestinal Motility Disorders, Xi'an, Shaanxi, China
- Shaanxi Provincial Clinical Research Center for Gastrointestinal Diseases, Xi'an, Shaanxi, China
- Digestive Disease Quality Control Center of Shaanxi Province, Xi'an, Shaanxi, China
| | - Yan Cheng
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Shaanxi Key Laboratory of Gastrointestinal Motility Disorders, Xi'an, Shaanxi, China
- Shaanxi Provincial Clinical Research Center for Gastrointestinal Diseases, Xi'an, Shaanxi, China
- Digestive Disease Quality Control Center of Shaanxi Province, Xi'an, Shaanxi, China
| | - Gang Zhao
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Shaanxi Key Laboratory of Gastrointestinal Motility Disorders, Xi'an, Shaanxi, China
- Shaanxi Provincial Clinical Research Center for Gastrointestinal Diseases, Xi'an, Shaanxi, China
- Digestive Disease Quality Control Center of Shaanxi Province, Xi'an, Shaanxi, China
| | - Shenghui Li
- Puensum Genetech Institute, Wuhan, Hubei, China
| | - Qingling Zhong
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Shaanxi Key Laboratory of Gastrointestinal Motility Disorders, Xi'an, Shaanxi, China
- Shaanxi Provincial Clinical Research Center for Gastrointestinal Diseases, Xi'an, Shaanxi, China
- Digestive Disease Quality Control Center of Shaanxi Province, Xi'an, Shaanxi, China
| | - Huan Zhang
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Shaanxi Key Laboratory of Gastrointestinal Motility Disorders, Xi'an, Shaanxi, China
- Shaanxi Provincial Clinical Research Center for Gastrointestinal Diseases, Xi'an, Shaanxi, China
- Digestive Disease Quality Control Center of Shaanxi Province, Xi'an, Shaanxi, China
| | - Ping Zhao
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Shaanxi Key Laboratory of Gastrointestinal Motility Disorders, Xi'an, Shaanxi, China
- Shaanxi Provincial Clinical Research Center for Gastrointestinal Diseases, Xi'an, Shaanxi, China
- Digestive Disease Quality Control Center of Shaanxi Province, Xi'an, Shaanxi, China
| | - Cui Fu
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Shaanxi Key Laboratory of Gastrointestinal Motility Disorders, Xi'an, Shaanxi, China
- Shaanxi Provincial Clinical Research Center for Gastrointestinal Diseases, Xi'an, Shaanxi, China
- Digestive Disease Quality Control Center of Shaanxi Province, Xi'an, Shaanxi, China
| | - Yahua Song
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Shaanxi Key Laboratory of Gastrointestinal Motility Disorders, Xi'an, Shaanxi, China
- Shaanxi Provincial Clinical Research Center for Gastrointestinal Diseases, Xi'an, Shaanxi, China
- Digestive Disease Quality Control Center of Shaanxi Province, Xi'an, Shaanxi, China
| | - Longbao Yang
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Shaanxi Key Laboratory of Gastrointestinal Motility Disorders, Xi'an, Shaanxi, China
- Shaanxi Provincial Clinical Research Center for Gastrointestinal Diseases, Xi'an, Shaanxi, China
- Digestive Disease Quality Control Center of Shaanxi Province, Xi'an, Shaanxi, China
| | - Yan Wang
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Shaanxi Key Laboratory of Gastrointestinal Motility Disorders, Xi'an, Shaanxi, China
- Shaanxi Provincial Clinical Research Center for Gastrointestinal Diseases, Xi'an, Shaanxi, China
- Digestive Disease Quality Control Center of Shaanxi Province, Xi'an, Shaanxi, China
| | - Yue Zhang
- Puensum Genetech Institute, Wuhan, Hubei, China
| | - Jiong Jiang
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Shaanxi Key Laboratory of Gastrointestinal Motility Disorders, Xi'an, Shaanxi, China
- Shaanxi Provincial Clinical Research Center for Gastrointestinal Diseases, Xi'an, Shaanxi, China
- Digestive Disease Quality Control Center of Shaanxi Province, Xi'an, Shaanxi, China
| | - Ting Wang
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Shaanxi Key Laboratory of Gastrointestinal Motility Disorders, Xi'an, Shaanxi, China
- Shaanxi Provincial Clinical Research Center for Gastrointestinal Diseases, Xi'an, Shaanxi, China
- Digestive Disease Quality Control Center of Shaanxi Province, Xi'an, Shaanxi, China
| | - Juhui Zhao
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Shaanxi Key Laboratory of Gastrointestinal Motility Disorders, Xi'an, Shaanxi, China
- Shaanxi Provincial Clinical Research Center for Gastrointestinal Diseases, Xi'an, Shaanxi, China
- Digestive Disease Quality Control Center of Shaanxi Province, Xi'an, Shaanxi, China
| | - Yong Li
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Shaanxi Key Laboratory of Gastrointestinal Motility Disorders, Xi'an, Shaanxi, China
- Shaanxi Provincial Clinical Research Center for Gastrointestinal Diseases, Xi'an, Shaanxi, China
- Digestive Disease Quality Control Center of Shaanxi Province, Xi'an, Shaanxi, China
| | - Biyuan Wang
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Shaanxi Key Laboratory of Gastrointestinal Motility Disorders, Xi'an, Shaanxi, China
- Shaanxi Provincial Clinical Research Center for Gastrointestinal Diseases, Xi'an, Shaanxi, China
- Digestive Disease Quality Control Center of Shaanxi Province, Xi'an, Shaanxi, China
| | - Fenrong Chen
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Shaanxi Key Laboratory of Gastrointestinal Motility Disorders, Xi'an, Shaanxi, China
- Shaanxi Provincial Clinical Research Center for Gastrointestinal Diseases, Xi'an, Shaanxi, China
- Digestive Disease Quality Control Center of Shaanxi Province, Xi'an, Shaanxi, China
| | - Hongli Zhao
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Shaanxi Key Laboratory of Gastrointestinal Motility Disorders, Xi'an, Shaanxi, China
- Shaanxi Provincial Clinical Research Center for Gastrointestinal Diseases, Xi'an, Shaanxi, China
- Digestive Disease Quality Control Center of Shaanxi Province, Xi'an, Shaanxi, China
| | - Yonghua Wang
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Shaanxi Key Laboratory of Gastrointestinal Motility Disorders, Xi'an, Shaanxi, China
- Shaanxi Provincial Clinical Research Center for Gastrointestinal Diseases, Xi'an, Shaanxi, China
- Digestive Disease Quality Control Center of Shaanxi Province, Xi'an, Shaanxi, China
| | - Jinhai Wang
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Shaanxi Key Laboratory of Gastrointestinal Motility Disorders, Xi'an, Shaanxi, China
- Shaanxi Provincial Clinical Research Center for Gastrointestinal Diseases, Xi'an, Shaanxi, China
- Digestive Disease Quality Control Center of Shaanxi Province, Xi'an, Shaanxi, China
| | - Shiyang Ma
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Shaanxi Key Laboratory of Gastrointestinal Motility Disorders, Xi'an, Shaanxi, China
- Shaanxi Provincial Clinical Research Center for Gastrointestinal Diseases, Xi'an, Shaanxi, China
- Digestive Disease Quality Control Center of Shaanxi Province, Xi'an, Shaanxi, China
| |
Collapse
|
26
|
Pust MM, Rocha Castellanos DM, Rzasa K, Dame A, Pishchany G, Assawasirisin C, Liss A, Fernandez-Del Castillo C, Xavier RJ. Absence of a pancreatic microbiome in intraductal papillary mucinous neoplasm. Gut 2024; 73:1131-1141. [PMID: 38429112 PMCID: PMC11187374 DOI: 10.1136/gutjnl-2023-331012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 02/06/2024] [Indexed: 03/03/2024]
Abstract
OBJECTIVE This study aims to validate the existence of a microbiome within intraductal papillary mucinous neoplasm (IPMN) that can be differentiated from the taxonomically diverse DNA background of next-generation sequencing procedures. DESIGN We generated 16S rRNA amplicon sequencing data to analyse 338 cyst fluid samples from 190 patients and 19 negative controls, the latter collected directly from sterile syringes in the operating room. A subset of samples (n=20) and blanks (n=5) were spiked with known concentrations of bacterial cells alien to the human microbiome to infer absolute abundances of microbial traces. All cyst fluid samples were obtained intraoperatively and included IPMNs with various degrees of dysplasia as well as other cystic neoplasms. Follow-up culturing experiments were conducted to assess bacterial growth for microbiologically significant signals. RESULTS Microbiome signatures of cyst fluid samples were inseparable from those of negative controls, with no difference in taxonomic diversity, and microbial community composition. In a patient subgroup that had recently undergone invasive procedures, a bacterial signal was evident. This outlier signal was not characterised by higher taxonomic diversity but by an increased dominance index of a gut-associated microbe, leading to lower taxonomic evenness compared with the background signal. CONCLUSION The 'microbiome' of IPMNs and other pancreatic cystic neoplasms does not deviate from the background signature of negative controls, supporting the concept of a sterile environment. Outlier signals may appear in a small fraction of patients following recent invasive endoscopic procedures. No associations between microbial patterns and clinical or cyst parameters were apparent.
Collapse
Affiliation(s)
- Marie-Madlen Pust
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
- Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | | | - Kara Rzasa
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Andrea Dame
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Gleb Pishchany
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, USA
| | - Charnwit Assawasirisin
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Andrew Liss
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Ramnik J Xavier
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
27
|
Nie F, Zhang J, Tian H, Zhao J, Gong P, Wang H, Wang S, Yang P, Yang C. The role of CXCL2-mediated crosstalk between tumor cells and macrophages in Fusobacterium nucleatum-promoted oral squamous cell carcinoma progression. Cell Death Dis 2024; 15:277. [PMID: 38637499 PMCID: PMC11026399 DOI: 10.1038/s41419-024-06640-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 03/28/2024] [Accepted: 04/02/2024] [Indexed: 04/20/2024]
Abstract
Dysbiosis of the oral microbiota is related to chronic inflammation and carcinogenesis. Fusobacterium nucleatum (Fn), a significant component of the oral microbiota, can perturb the immune system and form an inflammatory microenvironment for promoting the occurrence and progression of oral squamous cell carcinoma (OSCC). However, the underlying mechanisms remain elusive. Here, we investigated the impacts of Fn on OSCC cells and the crosstalk between OSCC cells and macrophages. 16 s rDNA sequencing and fluorescence in situ hybridization verified that Fn was notably enriched in clinical OSCC tissues compared to paracancerous tissues. The conditioned medium co-culture model validated that Fn and macrophages exhibited tumor-promoting properties by facilitating OSCC cell proliferation, migration, and invasion. Besides, Fn and OSCC cells can recruit macrophages and facilitate their M2 polarization. This crosstalk between OSCC cells and macrophages was further enhanced by Fn, thereby amplifying this positive feedback loop between them. The production of CXCL2 in response to Fn stimulation was a significant mediator. Suppression of CXCL2 in OSCC cells weakened Fn's promoting effects on OSCC cell proliferation, migration, macrophage recruitment, and M2 polarization. Conversely, knocking down CXCL2 in macrophages reversed the Fn-induced feedback effect of macrophages on the highly invasive phenotype of OSCC cells. Mechanistically, Fn activated the NF-κB pathway in both OSCC cells and macrophages, leading to the upregulation of CXCL2 expression. In addition, the SCC7 subcutaneous tumor-bearing model in C3H mice also substantiated Fn's ability to enhance tumor progression by facilitating cell proliferation, activating NF-κB signaling, up-regulating CXCL2 expression, and inducing M2 macrophage infiltration. However, these effects were reversed by the CXCL2-CXCR2 inhibitor SB225002. In summary, this study suggests that Fn contributes to OSCC progression by promoting tumor cell proliferation, macrophage recruitment, and M2 polarization. Simultaneously, the enhanced CXCL2-mediated crosstalk between OSCC cells and macrophages plays a vital role in the pro-cancer effect of Fn.
Collapse
Affiliation(s)
- Fujiao Nie
- Department of Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong, China
| | - Jie Zhang
- Advanced Medical Research Institute, Shandong University, Jinan, Shandong, China
| | - Haoyang Tian
- Department of Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong, China
| | - Jingjing Zhao
- Department of Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong, China
| | - Pizhang Gong
- Department of Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong, China
| | - Huiru Wang
- Department of Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong, China
| | - Suli Wang
- Department of Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong, China
| | - Pishan Yang
- Department of Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong, China.
| | - Chengzhe Yang
- Department of Oral and Maxillofacial Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China.
| |
Collapse
|
28
|
Eckhoff AM, Fletcher AA, Kelly MS, Dohlman A, McIntyre CA, Shen X, Iyer MK, Nussbaum DP, Allen PJ. Comprehensive Assessment of the Intrinsic Pancreatic Microbiome. Ann Surg 2024:00000658-990000000-00843. [PMID: 38623754 PMCID: PMC11480254 DOI: 10.1097/sla.0000000000006299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/17/2024]
Abstract
OBJECTIVE We sought to comprehensively profile tissue and cyst fluid in patients with benign, precancerous, and cancerous conditions of the pancreas to characterize the intrinsic pancreatic microbiome. SUMMARY BACKGROUND DATA Small studies in pancreatic ductal adenocarcinoma (PDAC) and intraductal papillary mucinous neoplasm (IPMN) have suggested that intra-pancreatic microbial dysbiosis may drive malignant transformation. METHODS Pancreatic samples were collected at the time of resection from 109 patients. Samples included tumor tissue (control, n=20; IPMN, n=20; PDAC, n=19) and pancreatic cyst fluid (IPMN, n=30; SCA, n=10; MCN, n=10). Assessment of bacterial DNA by quantitative PCR and 16S ribosomal RNA gene sequencing was performed. Downstream analyses determined the relative abundances of individual taxa between groups and compared intergroup diversity. Whole-genome sequencing data from 140 patients with PDAC in the National Cancer Institute's Clinical Proteomic Tumor Analysis Consortium (CPTAC) were analyzed to validate findings. RESULTS Sequencing of pancreatic tissue yielded few microbial reads regardless of diagnosis, and analysis of pancreatic tissue showed no difference in the abundance and composition of bacterial taxa between normal pancreas, IPMN, or PDAC groups. Low-grade dysplasia (LGD) and high-grade dysplasia (HGD) IPMN were characterized by low bacterial abundances with no difference in tissue composition and a slight increase in Pseudomonas and Sediminibacterium in HGD cyst fluid. Decontamination analysis using the CPTAC database confirmed a low-biomass, low-diversity intrinsic pancreatic microbiome that did not differ by pathology. CONCLUSIONS Our analysis of the pancreatic microbiome demonstrated very low intrinsic biomass that is relatively conserved across diverse neoplastic conditions and thus unlikely to drive malignant transformation.
Collapse
Affiliation(s)
- Austin M Eckhoff
- Department of Surgery, Duke University; Durham, North Carolina, USA
| | | | - Matthew S Kelly
- Department of Pediatrics, Duke University; Durham, North Carolina, USA
- Department of Molecular Genetics and Microbiology; Durham, North Carolina, USA
| | - Anders Dohlman
- Department of Biomedical Engineering, Duke Microbiome Center, Duke University; Durham, North Carolina, USA
| | - Caitlin A McIntyre
- Department of Surgery, Memorial Sloan Kettering, New York, New York, USA
| | - Xiling Shen
- Department of Surgery, Memorial Sloan Kettering, New York, New York, USA
- Terasaki Institute, Los Angeles, California, USA
| | - Matthew K Iyer
- Department of Surgery, Duke University; Durham, North Carolina, USA
| | | | - Peter J Allen
- Department of Surgery, Duke University; Durham, North Carolina, USA
| |
Collapse
|
29
|
Saba E, Farhat M, Daoud A, Khashan A, Forkush E, Menahem NH, Makkawi H, Pandi K, Angabo S, Kawasaki H, Plaschkes I, Parnas O, Zamir G, Atlan K, Elkin M, Katz L, Nussbaum G. Oral bacteria accelerate pancreatic cancer development in mice. Gut 2024; 73:770-786. [PMID: 38233197 DOI: 10.1136/gutjnl-2023-330941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 12/28/2023] [Indexed: 01/19/2024]
Abstract
OBJECTIVE Epidemiological studies highlight an association between pancreatic ductal adenocarcinoma (PDAC) and oral carriage of the anaerobic bacterium Porphyromonas gingivalis, a species highly linked to periodontal disease. We analysed the potential for P. gingivalis to promote pancreatic cancer development in an animal model and probed underlying mechanisms. DESIGN We tracked P. gingivalis bacterial translocation from the oral cavity to the pancreas following administration to mice. To dissect the role of P. gingivalis in PDAC development, we administered bacteria to a genetically engineered mouse PDAC model consisting of inducible acinar cell expression of mutant Kras (Kras +/LSL-G12D; Ptf1a-CreER, iKC mice). These mice were used to study the cooperative effects of Kras mutation and P. gingivalis on the progression of pancreatic intraepithelial neoplasia (PanIN) to PDAC. The direct effects of P. gingivalis on acinar cells and PDAC cell lines were studied in vitro. RESULTS P. gingivalis migrated from the oral cavity to the pancreas in mice and can be detected in human PanIN lesions. Repetitive P. gingivalis administration to wild-type mice induced pancreatic acinar-to-ductal metaplasia (ADM), and altered the composition of the intrapancreatic microbiome. In iKC mice, P. gingivalis accelerated PanIN to PDAC progression. In vitro, P. gingivalis infection induced acinar cell ADM markers SOX9 and CK19, and intracellular bacteria protected PDAC cells from reactive oxygen species-mediated cell death resulting from nutrient stress. CONCLUSION Taken together, our findings demonstrate a causal role for P. gingivalis in pancreatic cancer development in mice.
Collapse
Affiliation(s)
- Elias Saba
- Institute of Biomedical and Oral Research, Hebrew University-Hadassah, Jerusalem, Israel
| | - Maria Farhat
- Institute of Biomedical and Oral Research, Hebrew University-Hadassah, Jerusalem, Israel
| | - Alaa Daoud
- Institute of Biomedical and Oral Research, Hebrew University-Hadassah, Jerusalem, Israel
| | - Arin Khashan
- Institute of Biomedical and Oral Research, Hebrew University-Hadassah, Jerusalem, Israel
| | - Esther Forkush
- Gastroenterology, Hadassah Medical Center, Jerusalem, Israel
| | - Noam Hallel Menahem
- Institute of Biomedical and Oral Research, Hebrew University-Hadassah, Jerusalem, Israel
| | - Hasnaa Makkawi
- Institute of Biomedical and Oral Research, Hebrew University-Hadassah, Jerusalem, Israel
| | - Karthikeyan Pandi
- Institute of Biomedical and Oral Research, Hebrew University-Hadassah, Jerusalem, Israel
| | - Sarah Angabo
- Institute of Biomedical and Oral Research, Hebrew University-Hadassah, Jerusalem, Israel
| | - Hiromichi Kawasaki
- Institute of Biomedical and Oral Research, Hebrew University-Hadassah, Jerusalem, Israel
- Central Research Institute, Wakunaga Pharmaceutical Co Ltd, Koda-cho, Akitakata-shi, Hiroshima, Japan
| | - Inbar Plaschkes
- Info-CORE, Bioinformatics Unit of the I-CORE, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Oren Parnas
- Immunology and Cancer Research, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Gideon Zamir
- Experimental Surgery, Hebrew University Hadassah Medical School, Jerusalem, Israel
| | | | - Michael Elkin
- Oncology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Lior Katz
- Department of Gastroenterology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Gabriel Nussbaum
- Institute of Biomedical and Oral Research, Hebrew University-Hadassah, Jerusalem, Israel
| |
Collapse
|
30
|
Fu Y, Li J, Cai W, Huang Y, Liu X, Ma Z, Tang Z, Bian X, Zheng J, Jiang J, Li C. The emerging tumor microbe microenvironment: From delineation to multidisciplinary approach-based interventions. Acta Pharm Sin B 2024; 14:1560-1591. [PMID: 38572104 PMCID: PMC10985043 DOI: 10.1016/j.apsb.2023.11.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 10/20/2023] [Accepted: 11/03/2023] [Indexed: 04/05/2024] Open
Abstract
Intratumoral microbiota has become research hotspots, and emerges as a non-negligent new component of tumor microenvironments (TME), due to its powerful influence on tumor initiation, metastasis, immunosurveillance and prognosis despite in low-biomass. The accumulations of microbes, and their related components and metabolites within tumor tissues, endow TME with additional pluralistic features which are distinct from the conventional one. Therefore, it's definitely necessary to comprehensively delineate the sophisticated landscapes of tumor microbe microenvironment, as well as their functions and related underlying mechanisms. Herein, in this review, we focused on the fields of tumor microbe microenvironment, including the heterogeneity of intratumor microbiota in different types of tumors, the controversial roles of intratumoral microbiota, the basic features of tumor microbe microenvironment (i.e., pathogen-associated molecular patterns (PAMPs), typical microbial metabolites, autophagy, inflammation, multi-faceted immunomodulation and chemoresistance), as well as the multidisciplinary approach-based intervention of tumor microbiome for cancer therapy by applying wild-type or engineered live microbes, microbiota metabolites, antibiotics, synthetic biology and rationally designed biomaterials. We hope our work will provide valuable insight to deeply understand the interplay of cancer-immune-microbial, and facilitate the development of microbes-based tumor-specific treatments.
Collapse
Affiliation(s)
- Yu Fu
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Jia Li
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China
| | - Wenyun Cai
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Yulan Huang
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Xinlong Liu
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Zhongyi Ma
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Zhongjie Tang
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Xufei Bian
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Ji Zheng
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China
| | - Jiayun Jiang
- Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Chong Li
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| |
Collapse
|
31
|
Hunter C, Dia K, Boykins J, Perry K, Banerjee N, Cuffee J, Armstrong E, Morgan G, Banerjee HN, Banerjee A, Bhattacharya S. An investigation for phylogenetic characterization of human Pancreatic cancer microbiome by 16SrDNA Sequencing and Bioinformatics techniques. RESEARCH SQUARE 2024:rs.3.rs-4140368. [PMID: 38585738 PMCID: PMC10996791 DOI: 10.21203/rs.3.rs-4140368/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Pancreatic cancer is a significant public health concern, with increasing incidence rates and limited treatment options. Recent studies have highlighted the role of the human microbiome, particularly the gut microbiota, in the development and progression of this disease. Microbial dysbiosis, characterized by alterations in the composition and function of the gut microbiota, has been implicated in pancreatic carcinogenesis through mechanisms involving chronic inflammation, immune dysregulation, and metabolic disturbances. Researchers have identified specific microbial signatures associated with pancreatic cancer, offering potential biomarkers for early detection and prognostication. By leveraging advanced sequencing and bioinformatics tools, scientists have delineated differences in the gut microbiota between pancreatic cancer patients and healthy individuals, providing insights into disease pathogenesis and potential diagnostic strategies. Moreover, the microbiome holds promise as a therapeutic target in pancreatic cancer treatment. Interventions aimed at modulating the microbiome, such as probiotics, prebiotics, and fecal microbiota transplantation, have demonstrated potential in enhancing the efficacy of existing cancer therapies, including chemotherapy and immunotherapy. These approaches can influence immune responses, alter tumor microenvironments, and sensitize tumors to treatment, offering new avenues for improving patient outcomes and overcoming therapeutic resistance. Overall, understanding the complex interplay between the microbiome and pancreatic cancer is crucial for advancing our knowledge of disease mechanisms and identifying innovative therapeutic strategies. Here we report phylogenetic analysis of the 16S microbial sequences of the pancreatic cancer mice microbiome and corresponding age matched healthy mice microbiome. We successfully identified differentially abundance of microbiota in the pancreatic cancer.
Collapse
Affiliation(s)
- Colby Hunter
- Elizabeth City State University campus of The University of North Carolina
| | - Khadimou Dia
- Elizabeth City State University campus of The University of North Carolina
| | - Julia Boykins
- Elizabeth City State University campus of The University of North Carolina
| | - Karrington Perry
- Elizabeth City State University campus of The University of North Carolina
| | - Narendra Banerjee
- Elizabeth City State University campus of The University of North Carolina
| | - Jazmine Cuffee
- Elizabeth City State University campus of The University of North Carolina
| | - Erik Armstrong
- Elizabeth City State University campus of The University of North Carolina
| | - Gabrielle Morgan
- Elizabeth City State University campus of The University of North Carolina
| | | | | | | |
Collapse
|
32
|
Hu Y, Jones D, Esnakula AK, Krishna SG, Chen W. Molecular Pathology of Pancreatic Cystic Lesions with a Focus on Malignant Progression. Cancers (Basel) 2024; 16:1183. [PMID: 38539517 PMCID: PMC10969285 DOI: 10.3390/cancers16061183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 03/12/2024] [Accepted: 03/14/2024] [Indexed: 11/11/2024] Open
Abstract
The malignant progression of pancreatic cystic lesions (PCLs) remains understudied with a knowledge gap, yet its exploration is pivotal for effectively stratifying patient risk and detecting cancer at its earliest stages. Within this review, we delve into the latest discoveries on the molecular level, revealing insights into the IPMN molecular landscape and revised progression model, associated histologic subtypes, and the role of inflammation in the pathogenesis and malignant progression of IPMN. Low-grade PCLs, particularly IPMNs, can develop into high-grade lesions or invasive carcinoma, underscoring the need for long-term surveillance of these lesions if they are not resected. Although KRAS and GNAS remain the primary oncogenic drivers of neoplastic development in IPMNs, additional genes that are important in tumorigenesis have been recently identified by whole exome sequencing. A more complete understanding of the genes involved in the molecular progression of IPMN is critical for effective monitoring to minimize the risk of malignant progression. Complicating these strategies, IPMNs are also frequently multifocal and multiclonal, as demonstrated by comparative molecular analysis. Algorithms for preoperative cyst sampling and improved radiomic techniques are emerging to model this spatial and temporal genetic heterogeneity better. Here, we review the molecular pathology of PCLs, focusing on changes associated with malignant progression. Developing models of molecular risk stratification in PCLs which can complement radiologic and clinical features, facilitate the early detection of pancreatic cancer, and enable the development of more personalized surveillance and management strategies are summarized.
Collapse
Affiliation(s)
- Yan Hu
- James Molecular Laboratory, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA; (Y.H.); (D.J.)
| | - Dan Jones
- James Molecular Laboratory, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA; (Y.H.); (D.J.)
| | - Ashwini K. Esnakula
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA;
| | - Somashekar G. Krishna
- Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA;
| | - Wei Chen
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA;
| |
Collapse
|
33
|
Zou L, Mo S, Jia C, Pang J, Chang X, Chen J. The tumoral microbiome of pancreatic intraductal papillary mucinous neoplasm: A single-center retrospective cohort study. J Gastroenterol Hepatol 2024; 39:496-505. [PMID: 38111357 DOI: 10.1111/jgh.16437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 10/18/2023] [Accepted: 11/14/2023] [Indexed: 12/20/2023]
Abstract
BACKGROUND AND AIM Pancreatic intraductal papillary mucinous neoplasm (IPMN) is one of the most common precancerous lesions of pancreatic carcinoma. Studies have found that the tumoral microbiome has an important influence on pancreatic carcinoma. However, the tumoral microbiome of IPMNs has rarely been explored. METHODS Tumoral microbiome gene sequencing was carried out using 16 specimens of IPMN and 45 specimens of IPMN with associated invasive carcinoma (IPMN-IC) by 2bRAD sequencing for microbiome. The profile of the tumoral microbiome was summarized. Associations of the tumoral microbiome with disease grade, histological subtype, and prognosis were analyzed. RESULTS A total of 598 species of microbes were identified, comprising 228 genera, 109 families, 60 orders, 29 classes, 14 phyla, and 2 kingdoms. The genus Pseudomonas was detected more frequently and had higher relative abundance in IPMN-ICs; Alcaligenes faecalis was detected with higher relative abundance in IPMNs. Bifidobacterium pseudolongum had a higher relative abundance in the IPMN-IC group, regardless of histological subtype. Moreover, among patients with IPMN-ICs, those with a high relative abundance of B. pseudolongum had better overall survival than those with a low relative abundance. Patients who were positive for Staphylococcus aureus or Mycolicibacillus koreensis had shorter survival. The presence of S. aureus was an independent risk factor for poor prognosis. CONCLUSIONS There are enriching tumoral microbes in IPMN. The tumoral microbiome of IPMN is different from that of IPMN-IC.
Collapse
Affiliation(s)
- Long Zou
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shengwei Mo
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Congwei Jia
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Junyi Pang
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaoyan Chang
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jie Chen
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
34
|
Wang B, Deng J, Donati V, Merali N, Frampton AE, Giovannetti E, Deng D. The Roles and Interactions of Porphyromonas gingivalis and Fusobacterium nucleatum in Oral and Gastrointestinal Carcinogenesis: A Narrative Review. Pathogens 2024; 13:93. [PMID: 38276166 PMCID: PMC10820765 DOI: 10.3390/pathogens13010093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 01/12/2024] [Accepted: 01/15/2024] [Indexed: 01/27/2024] Open
Abstract
Epidemiological studies have spotlighted the intricate relationship between individual oral bacteria and tumor occurrence. Porphyromonas gingivalis and Fusobacteria nucleatum, which are known periodontal pathogens, have emerged as extensively studied participants with potential pathogenic abilities in carcinogenesis. However, the complex dynamics arising from interactions between these two pathogens were less addressed. This narrative review aims to summarize the current knowledge on the prevalence and mechanism implications of P. gingivalis and F. nucleatum in the carcinogenesis of oral squamous cell carcinoma (OSCC), colorectal cancer (CRC), and pancreatic ductal adenocarcinoma (PDAC). In particular, it explores the clinical and experimental evidence on the interplay between P. gingivalis and F. nucleatum in affecting oral and gastrointestinal carcinogenesis. P. gingivalis and F. nucleatum, which are recognized as keystone or bridging bacteria, were identified in multiple clinical studies simultaneously. The prevalence of both bacteria species correlated with cancer development progression, emphasizing the potential impact of the collaboration. Regrettably, there was insufficient experimental evidence to demonstrate the synergistic function. We further propose a hypothesis to elucidate the underlying mechanisms, offering a promising avenue for future research in this dynamic and evolving field.
Collapse
Affiliation(s)
- Bing Wang
- Department of Medical Oncology, Amsterdam University Medical Center, Cancer Center Amsterdam, Vrije Universiteit, 1081 HV Amsterdam, The Netherlands; (B.W.); (J.D.); (V.D.); (E.G.)
| | - Juan Deng
- Department of Medical Oncology, Amsterdam University Medical Center, Cancer Center Amsterdam, Vrije Universiteit, 1081 HV Amsterdam, The Netherlands; (B.W.); (J.D.); (V.D.); (E.G.)
| | - Valentina Donati
- Department of Medical Oncology, Amsterdam University Medical Center, Cancer Center Amsterdam, Vrije Universiteit, 1081 HV Amsterdam, The Netherlands; (B.W.); (J.D.); (V.D.); (E.G.)
- Unit of Pathological Anatomy 2, Azienda Ospedaliero-Universitaria Pisana, 56100 Pisa, Italy
| | - Nabeel Merali
- Minimal Access Therapy Training Unit (MATTU), Royal Surrey County Hospital, NHS Foundation Trust, Egerton Road, Guildford GU2 7XX, UK; (N.M.); (A.E.F.)
- Department of Hepato-Pancreato-Biliary (HPB) Surgery, Royal Surrey County Hospital, NHS Foundation Trust, Egerton Road, Guildford GU2 7XX, UK
- Section of Oncology, Department of Clinical and Experimental Medicine, Faculty of Health Medical Science, University of Surrey, Guilford GU2 7WG, UK
| | - Adam E. Frampton
- Minimal Access Therapy Training Unit (MATTU), Royal Surrey County Hospital, NHS Foundation Trust, Egerton Road, Guildford GU2 7XX, UK; (N.M.); (A.E.F.)
- Department of Hepato-Pancreato-Biliary (HPB) Surgery, Royal Surrey County Hospital, NHS Foundation Trust, Egerton Road, Guildford GU2 7XX, UK
- Section of Oncology, Department of Clinical and Experimental Medicine, Faculty of Health Medical Science, University of Surrey, Guilford GU2 7WG, UK
| | - Elisa Giovannetti
- Department of Medical Oncology, Amsterdam University Medical Center, Cancer Center Amsterdam, Vrije Universiteit, 1081 HV Amsterdam, The Netherlands; (B.W.); (J.D.); (V.D.); (E.G.)
- Fondazione Pisana per la Scienza, 56100 Pisa, Italy
| | - Dongmei Deng
- Department of Prevention Dentistry, Academic Center for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universitreit Amsterdam, 1081 LA Amsterdam, The Netherlands
| |
Collapse
|
35
|
León-Letelier RA, Dou R, Vykoukal J, Yip-Schneider MT, Maitra A, Irajizad E, Wu R, Dennison JB, Do KA, Zhang J, Schmidt CM, Hanash S, Fahrmann JF. Contributions of the Microbiome-Derived Metabolome for Risk Assessment and Prognostication of Pancreatic Cancer. Clin Chem 2024; 70:102-115. [PMID: 38175578 PMCID: PMC11836914 DOI: 10.1093/clinchem/hvad186] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 10/16/2023] [Indexed: 01/05/2024]
Abstract
BACKGROUND Increasing evidence implicates microbiome involvement in the development and progression of pancreatic ductal adenocarcinoma (PDAC). Studies suggest that reflux of gut or oral microbiota can lead to colonization in the pancreas, resulting in dysbiosis that culminates in release of microbial toxins and metabolites that potentiate an inflammatory response and increase susceptibility to PDAC. Moreover, microbe-derived metabolites can exert direct effector functions on precursors and cancer cells, as well as other cell types, to either promote or attenuate tumor development and modulate treatment response. CONTENT The occurrence of microbial metabolites in biofluids thereby enables risk assessment and prognostication of PDAC, as well as having potential for design of interception strategies. In this review, we first highlight the relevance of the microbiome for progression of precancerous lesions in the pancreas and, using liquid chromatography-mass spectrometry, provide supporting evidence that microbe-derived metabolites manifest in pancreatic cystic fluid and are associated with malignant progression of intraductal papillary mucinous neoplasm(s). We secondly summarize the biomarker potential of microbe-derived metabolite signatures for (a) identifying individuals at high risk of developing or harboring PDAC and (b) predicting response to treatment and disease outcomes. SUMMARY The microbiome-derived metabolome holds considerable promise for risk assessment and prognostication of PDAC.
Collapse
Affiliation(s)
- Ricardo A. León-Letelier
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA, 77030
| | - Rongzhang Dou
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA, 77030
| | - Jody Vykoukal
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA, 77030
| | | | - Anirban Maitra
- Department of Translational Molecular Pathology and Sheikh Ahmed Center for Pancreatic Cancer Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA, 77030
| | - Ehsan Irajizad
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA, 77030
| | - Ranran Wu
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA, 77030
| | - Jennifer B. Dennison
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA, 77030
| | - Kim-An Do
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA, 77030
| | - Jianjun Zhang
- Department of Epidemiology, Richard M. Fairbanks School of Public Health, Indiana University, Indianapolis, Indiana, USA
| | - C Max Schmidt
- Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Samir Hanash
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA, 77030
| | - Johannes F. Fahrmann
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA, 77030
| |
Collapse
|
36
|
Cruz MS, Tintelnot J, Gagliani N. Roles of microbiota in pancreatic cancer development and treatment. Gut Microbes 2024; 16:2320280. [PMID: 38411395 PMCID: PMC10900280 DOI: 10.1080/19490976.2024.2320280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 02/14/2024] [Indexed: 02/28/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive disease with poor prognosis. This is due to the fact that most cases are only diagnosed at an advanced and palliative disease stage, and there is a high incidence of therapy resistance. Despite ongoing efforts, to date, the mechanisms underlying PDAC oncogenesis and its poor responses to treatment are still largely unclear. As the study of the microbiome in cancer progresses, growing evidence suggests that bacteria or fungi might be key players both in PDAC oncogenesis as well as in its resistance to chemo- and immunotherapy, for instance through modulation of the tumor microenvironment and reshaping of the host immune response. Here, we review how the microbiota exerts these effects directly or indirectly via microbial-derived metabolites. Finally, we further discuss the potential of modulating the microbiota composition as a therapy in PDAC.
Collapse
Affiliation(s)
- Mariana Santos Cruz
- II. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), Hamburg, Germany
| | - Joseph Tintelnot
- II. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), Hamburg, Germany
| | - Nicola Gagliani
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), Hamburg, Germany
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
37
|
Krieger M, Guo M, Merritt J. Reexamining the role of Fusobacterium nucleatum subspecies in clinical and experimental studies. Gut Microbes 2024; 16:2415490. [PMID: 39394990 PMCID: PMC11486156 DOI: 10.1080/19490976.2024.2415490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 09/17/2024] [Accepted: 10/04/2024] [Indexed: 10/14/2024] Open
Abstract
The Gram-negative anaerobic species Fusobacterium nucleatum was originally described as a commensal organism from the human oral microbiome. However, it is now widely recognized as a key inflammophilic pathobiont associated with a wide variety of oral and extraoral diseases. Historically, F. nucleatum has been classified into four subspecies that have been generally considered as functionally interchangeable in their pathogenic potential. Recent studies have challenged this notion, as clinical data reveal a highly biased distribution of F. nucleatum subspecies within disease sites of both inflammatory oral diseases and various malignancies. This review details the historical basis for the F. nucleatum subspecies designations and summarizes our current understanding of the similarities and distinctions between these organisms to provide important context for future clinical and laboratory studies of F. nucleatum.
Collapse
Affiliation(s)
- Madeline Krieger
- Division of Biomaterial and Biomedical Sciences, School of Dentistry, Oregon Health & Science University (OHSU), Portland, OR, USA
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health & Science University (OHSU), Portland, OR, USA
| | - Mingzhe Guo
- Division of Biomaterial and Biomedical Sciences, School of Dentistry, Oregon Health & Science University (OHSU), Portland, OR, USA
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University (OHSU), Portland, OR, USA
| | - Justin Merritt
- Division of Biomaterial and Biomedical Sciences, School of Dentistry, Oregon Health & Science University (OHSU), Portland, OR, USA
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University (OHSU), Portland, OR, USA
| |
Collapse
|
38
|
Shirai H, Tsukada K. Understanding bacterial infiltration of the pancreas through a deformable pancreatic duct. J Biomech 2024; 162:111883. [PMID: 38064997 DOI: 10.1016/j.jbiomech.2023.111883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 11/20/2023] [Accepted: 11/22/2023] [Indexed: 01/16/2024]
Abstract
Tiny amount of bacteria are found in the pancreas in pancreatitis and cancer, which seemed involved in inflammation and carcinogenesis. However, bacterial infiltration from the duodenum is inhibited by the physical defense mechanisms such as bile flow and the sphincter of Oddi. To understand how the bacteria possibly infiltrate the pancreas through a deformable pancreatic duct, influenced by the periodic contractions of the sphincter of Oddi, a mathematical model of bacterial infiltration is developed that considered large deformation, fluid flow, and bacterial transport in a deformable pancreatic duct. In addition, the sphincter's contraction wave is modeled by including its propagation from the pancreas toward the duodenum. Simulated structure of the deformed duct with the relaxed sphincter and simulated bile distribution agreed reasonably well with the literature, validating the model. Bacterial infiltration from the duodenum in a deformable pancreatic duct, following the sphincter's contraction, is counteracted by a gradual peristalsis-like deformation of the pancreatic duct, due to an antegrade contraction wave propagation from the pancreas to the duodenum, Parametric sensitivity analysis demonstrated that bacterial infiltration is increased with lower bile and pancreatic juice flow rate, greater contraction amplitude and frequency, thinner wall thickness, and retrograde contraction wave propagation. Since contraction waves following retrograde propagation are increased in patients with common bile duct stones and pancreatitis, they may possibly be factors for continuum inflammation of pancreas. (224 words).
Collapse
Affiliation(s)
- Hiroaki Shirai
- Graduate School of Science and Technology, Keio University, 3-14-1 Hiyoshi Kohoku-ku, Yokohama-shi, Kanagawa 223-8522, Japan.
| | - Kosuke Tsukada
- Graduate School of Science and Technology, Keio University, 3-14-1 Hiyoshi Kohoku-ku, Yokohama-shi, Kanagawa 223-8522, Japan; Faculty of Science and Technology, Keio University, 3-14-1 Hiyoshi Kohoku-ku, Yokohama-shi, Kanagawa 223-8522, Japan
| |
Collapse
|
39
|
Sivam HGP, Chin BY, Gan SY, Ng JH, Gwenhure A, Chan EWL. Lipopolysaccharide (LPS) stimulation of Pancreatic Ductal Adenocarcinoma (PDAC) and macrophages activates the NLRP3 inflammasome that influences the levels of pro-inflammatory cytokines in a co-culture model. Cancer Biol Ther 2023; 24:2284857. [PMID: 38018872 PMCID: PMC10783839 DOI: 10.1080/15384047.2023.2284857] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 11/14/2023] [Indexed: 11/30/2023] Open
Abstract
Modified macrophages, tumor-associated macrophages (TAMs), are key contributors to the survival, growth, and metastatic behavior of pancreatic ductal adenocarcinoma (PDAC) cells. Central to the role of inflammation and TAMs lies the NLRP3 inflammasome. This study investigated the effects of LPS-stimulated inflammation on cell proliferation, levels of pro-inflammatory cytokines, and the NLRP3 inflammasome pathway in a co-culture model using PDAC cells and macrophages in the presence or absence of MCC950, a NLRP3-specific inhibitor. The effects of LPS-stimulated inflammation were tested on two PDAC cell lines (Panc 10.05 and SW 1990) co-cultured with RAW 264.7 macrophages. Cell proliferation was determined using the MTT assay. Levels of pro-inflammatory cytokines, IL-1β, and TNF-α were determined by ELISA. Western blot analyses were used to examine the expression of NLRP3 in both PDAC cells and macrophages. The co-culture and interaction between PDAC cell lines and macrophages led to pro-inflammatory microenvironment under LPS stimulation as evidenced by high levels of secreted IL-1β and TNF-α. Inhibition of the NLRP3 inflammasome by MCC950 counteracted the effects of LPS stimulation on the regulation of the NLRP3 inflammasome and pro-inflammatory cytokines in PDAC and macrophages. However, MCC950 differentially modified the viability of the metastatic vs primary PDAC cell lines. LPS stimulation increased PDAC cell viability by regulating the NLRP3 inflammasome and pro-inflammatory cytokines in the tumor microenvironment of PDAC cells/macrophages co-cultures. The specific inhibition of the NLRP inflammasome by MCC950 effectively counteracted the LPS-stimulated inflammation.
Collapse
Affiliation(s)
| | - Beek Yoke Chin
- Institute for Research, Development and Innovation, International Medical University, Kuala Lumpur, Malaysia
| | - Sook Yee Gan
- Department of Life Science, School of Pharmacy, International Medical University, Kuala Lumpur, Malaysia
| | - Jia Hao Ng
- School of Postgraduate Studies, International Medical University, Kuala Lumpur, Malaysia
| | - Agnes Gwenhure
- School of Postgraduate Studies, International Medical University, Kuala Lumpur, Malaysia
| | - Elaine Wan Ling Chan
- Institute for Research, Development and Innovation, International Medical University, Kuala Lumpur, Malaysia
| |
Collapse
|
40
|
He S, Bai X, Xu Y. Structural insight into the role of thiolase from Fusobacterium nucleatum. Biochem Biophys Res Commun 2023; 688:149151. [PMID: 37951156 DOI: 10.1016/j.bbrc.2023.149151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 10/22/2023] [Accepted: 10/23/2023] [Indexed: 11/13/2023]
Abstract
Fusobacterium nucleatum (F. nucleatum) is an anaerobic gram-negative bacterium that was previously thought to be related to the progression of colorectal cancer. In F. nucleatum, thiolase participates in fatty acid metabolism, and it can catalyse the transfer of an acetyl group from acetyl-CoA to another molecule, typically a fatty acid or another molecule in the synthesis of lipids. To gain deeper insight into the molecular mechanism governing the function of thiolase in F. nucleatum (Fn0495), we herein report the structure of Fn0495. The monomer of Fn0495 consists of three subdomains, namely, the N-terminal domain (residues 1-117 and 252-270), the C-terminal domain (residues 273-393), and the loop domain (residues 118-251). Fn0495 shows a unique difference in the charge and structure of the substrate binding pocket compared with homologous proteins. This research found three conserved residues (Cys88, His357, and Cys387) in Fn0495 arranged near a potential substrate binding pocket. In this study, the conformational changes between the covering loop, catalytic cysteine loop, regulatory determinant region, and homologous protein were compared. These results will enhance our understanding of the molecular characteristics and roles of the thiolase family.
Collapse
Affiliation(s)
- Shanru He
- Department of Bioengineering, College of Life Science, Dalian Minzu University, Dalian, 116600, Liaoning, China; Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of Education, College of Life Science, Dalian Minzu University, China
| | - Xue Bai
- Department of Bioengineering, College of Life Science, Dalian Minzu University, Dalian, 116600, Liaoning, China; Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of Education, College of Life Science, Dalian Minzu University, China
| | - Yongbin Xu
- Department of Bioengineering, College of Life Science, Dalian Minzu University, Dalian, 116600, Liaoning, China; Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of Education, College of Life Science, Dalian Minzu University, China; CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, Liaoning, China.
| |
Collapse
|
41
|
Asili P, Mirahmad M, Rezaei P, Mahdavi M, Larijani B, Tavangar SM. The Association of Oral Microbiome Dysbiosis with Gastrointestinal Cancers and Its Diagnostic Efficacy. J Gastrointest Cancer 2023; 54:1082-1101. [PMID: 36600023 DOI: 10.1007/s12029-022-00901-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/15/2022] [Indexed: 01/06/2023]
Abstract
BACKGROUND The second leading mortality cause in the world is cancer, making it a critical issue that impacts human health. As a result, scientists are looking for novel biomarkers for cancer detection. The oral microbiome, made up of approximately 700 species-level taxa, is a significant source for discovering novel biomarkers. In this review, we aimed to prepare a summary of research that has investigated the association between the oral microbiome and gastrointestinal cancers. METHODS We searched online scientific datasets including Web of Science, PubMed, Scopus, and Google Scholar. Eligibility criteria included human studies that reported abundances of the oral microbiome, or its diagnostic/prognostic performance in patients with gastrointestinal cancers. RESULTS Some phyla of the oral microbiome have a relationship with cancers. Some particular phyla of the oral microbiome that may be related to gastrointestinal cancers consist of Firmicutes, Actinobacteria, Bacteroidetes, Proteobacteria, and Fusobacteria. Changes in the abundances of Porphyromonas, Fusobacterium, Prevotella, and Veillonella are correlated with carcinogenesis, and may be used for distinguishing cancer patients from healthy subjects. Oral, colorectal, pancreatic, and esophageal cancers are the most important cancers related to the oral microbiome. CONCLUSION The results of this study may help future research to select bacteria as an early diagnostic or prognostic biomarker of gastrointestinal cancer. Given the current state of our knowledge, additional research is required to comprehend the multiplex processes underlying the role of bacterial microbiota upon cancer progression and to characterize the complex microbiota-host interaction network.
Collapse
Affiliation(s)
- Pooria Asili
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Mirahmad
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Parisa Rezaei
- Department of Nutrition, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Mohammad Mahdavi
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Mohammad Tavangar
- Chronic Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.
- Department of Pathology, Dr. Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
42
|
Meng Q, Zhou Q, Shi S, Xiao J, Ma Q, Yu J, Chen J, Kang Y. VTwins: inferring causative microbial features from metagenomic data of limited samples. Sci Bull (Beijing) 2023; 68:2806-2816. [PMID: 37919157 DOI: 10.1016/j.scib.2023.10.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 07/19/2023] [Accepted: 10/23/2023] [Indexed: 11/04/2023]
Abstract
It is difficult to infer causality from high-dimension metagenomic data due to interference from numerous confounders. By imitating the twin studies in genetic research, we develop a straightforward method-virtual twins (VTwins)-to eliminate the confounder effects by transforming the original cohort into a paired cohort of "Twin" samples with distinct phenotypes but matched taxonomic profiles. The results show that VTwins outperforms the conventional approach in the sensitivity of identifying causative features and only requires a 10-fold reduced sample size for recalling disease-associated microbes or pathways, as tested by simulated and empirical data. Benchmark test with other 16 kinds of software further validates the power and applicability of VTwins for handling high-dimension compositional datasets and mining causalities in metagenomic research. In conclusion, VTwins is straightforward and effective in handling high-diversity, high-dimension compositional data, promising applications in mining causalities for metagenomic and potentially other omics data. VTwins is open access and available at https://github.com/mengqingren/VTwins.
Collapse
Affiliation(s)
- Qingren Meng
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China; School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China; National Clinical Research Center for Infectious Diseases, The Third People's Hospital of Shenzhen, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen 518100, China
| | - Qian Zhou
- International Cancer Center, Shenzhen University Medical School, Shenzhen 518055, China
| | - Shuo Shi
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
| | - Jingfa Xiao
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
| | - Qin Ma
- Department of Biomedical Informatics, The Ohio State University, Columbus OH 43210, USA
| | - Jun Yu
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100190, China
| | - Jun Chen
- National Clinical Research Center for Infectious Diseases, The Third People's Hospital of Shenzhen, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen 518100, China
| | - Yu Kang
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100190, China.
| |
Collapse
|
43
|
Nasiri K, Amiri Moghaddam M, Etajuri EA, Badkoobeh A, Tavakol O, Rafinejad M, Forutan Mirhosseini A, Fathi A. Periodontitis and progression of gastrointestinal cancer: current knowledge and future perspective. Clin Transl Oncol 2023; 25:2801-2811. [PMID: 37036595 DOI: 10.1007/s12094-023-03162-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 02/26/2023] [Indexed: 04/11/2023]
Abstract
Periodontitis is a polymicrobial disorder caused by dysbiosis. Porphyromonas gingivalis (P.gingivalis) and Fusobacterium nucleatum (F.nucleatum) are pathobiont related to periodontitis pathogenesis and were found to be abundant in the intestinal mucosa of inflammatory bowel disease (IBD) and colorectal cancer (CRC) patients. Besides, periodontal infections have been found in a variety of tissues and organs, indicating that periodontitis is not just an inflammation limited to the oral cavity. Considering the possible translocation of pathobiont from the oral cavity to the gastrointestinal (GI) tract, this study aimed to review the published articles in this field to provide a comprehensive view of the existing knowledge about the relationship between periodontitis and GI malignancies by focusing on the oral/gut axis.
Collapse
Affiliation(s)
- Kamyar Nasiri
- Department of Dentistry, Islamic Azad University, Tehran, Iran
| | - Masoud Amiri Moghaddam
- Department of Periodontics, Dental Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Enas Abdalla Etajuri
- Department of Restorative Dentistry, Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia
| | - Ashkan Badkoobeh
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Qom University of Medical Sciences, Qom, Iran
| | - Omid Tavakol
- Department of Prosthodontics, Islamic Azad University, Shiraz, Iran
| | | | | | - Amirhossein Fathi
- Department of Prosthodontics, Dental Materials Research Center, Dental Research Institute, School of Dentistry, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
44
|
Xue X, Li R, Chen Z, Li G, Liu B, Guo S, Yue Q, Yang S, Xie L, Zhang Y, Zhao J, Tan R. The role of the symbiotic microecosystem in cancer: gut microbiota, metabolome, and host immunome. Front Immunol 2023; 14:1235827. [PMID: 37691931 PMCID: PMC10484231 DOI: 10.3389/fimmu.2023.1235827] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 07/12/2023] [Indexed: 09/12/2023] Open
Abstract
The gut microbiota is not just a simple nutritional symbiosis that parasitizes the host; it is a complex and dynamic ecosystem that coevolves actively with the host and is involved in a variety of biological activities such as circadian rhythm regulation, energy metabolism, and immune response. The development of the immune system and immunological functions are significantly influenced by the interaction between the host and the microbiota. The interactions between gut microbiota and cancer are of a complex nature. The critical role that the gut microbiota plays in tumor occurrence, progression, and treatment is not clear despite the already done research. The development of precision medicine and cancer immunotherapy further emphasizes the importance and significance of the question of how the microbiota takes part in cancer development, progression, and treatment. This review summarizes recent literature on the relationship between the gut microbiome and cancer immunology. The findings suggest the existence of a "symbiotic microecosystem" formed by gut microbiota, metabolome, and host immunome that is fundamental for the pathogenesis analysis and the development of therapeutic strategies for cancer.
Collapse
Affiliation(s)
- Xiaoyu Xue
- School of Pharmacy, Southwest Medical University, Luzhou, China
- Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medical Sciences, State Key Laboratory of Quality Evaluation of Traditional Chinese Medicine, Sichuan Engineering Technology Research Center of Genuine Regional Drug, Sichuan Provincial Engineering Research Center of Formation Principle and Quality Evaluation of Genuine Medicinal Materials, Translational Chinese Medicine Key Laboratory of Sichuan Province, Chengdu, China
| | - Rui Li
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Zhenni Chen
- Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medical Sciences, State Key Laboratory of Quality Evaluation of Traditional Chinese Medicine, Sichuan Engineering Technology Research Center of Genuine Regional Drug, Sichuan Provincial Engineering Research Center of Formation Principle and Quality Evaluation of Genuine Medicinal Materials, Translational Chinese Medicine Key Laboratory of Sichuan Province, Chengdu, China
- College of Food and Biological Engineering, Chengdu University, Chengdu, China
| | - Guiyu Li
- Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medical Sciences, State Key Laboratory of Quality Evaluation of Traditional Chinese Medicine, Sichuan Engineering Technology Research Center of Genuine Regional Drug, Sichuan Provincial Engineering Research Center of Formation Principle and Quality Evaluation of Genuine Medicinal Materials, Translational Chinese Medicine Key Laboratory of Sichuan Province, Chengdu, China
| | - Bisheng Liu
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Shanshan Guo
- Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medical Sciences, State Key Laboratory of Quality Evaluation of Traditional Chinese Medicine, Sichuan Engineering Technology Research Center of Genuine Regional Drug, Sichuan Provincial Engineering Research Center of Formation Principle and Quality Evaluation of Genuine Medicinal Materials, Translational Chinese Medicine Key Laboratory of Sichuan Province, Chengdu, China
| | - Qianhua Yue
- Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medical Sciences, State Key Laboratory of Quality Evaluation of Traditional Chinese Medicine, Sichuan Engineering Technology Research Center of Genuine Regional Drug, Sichuan Provincial Engineering Research Center of Formation Principle and Quality Evaluation of Genuine Medicinal Materials, Translational Chinese Medicine Key Laboratory of Sichuan Province, Chengdu, China
| | - Siye Yang
- Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medical Sciences, State Key Laboratory of Quality Evaluation of Traditional Chinese Medicine, Sichuan Engineering Technology Research Center of Genuine Regional Drug, Sichuan Provincial Engineering Research Center of Formation Principle and Quality Evaluation of Genuine Medicinal Materials, Translational Chinese Medicine Key Laboratory of Sichuan Province, Chengdu, China
| | - Linlin Xie
- Traditional Chinese Medicine Hospital Affiliated to Southwest Medical University, Classical Chinese Medicine Diagnosis and Treatment Center, Luzhou, China
| | - Yiguan Zhang
- School of Pharmacy, Southwest Medical University, Luzhou, China
- Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medical Sciences, State Key Laboratory of Quality Evaluation of Traditional Chinese Medicine, Sichuan Engineering Technology Research Center of Genuine Regional Drug, Sichuan Provincial Engineering Research Center of Formation Principle and Quality Evaluation of Genuine Medicinal Materials, Translational Chinese Medicine Key Laboratory of Sichuan Province, Chengdu, China
| | - Junning Zhao
- Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medical Sciences, State Key Laboratory of Quality Evaluation of Traditional Chinese Medicine, Sichuan Engineering Technology Research Center of Genuine Regional Drug, Sichuan Provincial Engineering Research Center of Formation Principle and Quality Evaluation of Genuine Medicinal Materials, Translational Chinese Medicine Key Laboratory of Sichuan Province, Chengdu, China
| | - Ruirong Tan
- Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medical Sciences, State Key Laboratory of Quality Evaluation of Traditional Chinese Medicine, Sichuan Engineering Technology Research Center of Genuine Regional Drug, Sichuan Provincial Engineering Research Center of Formation Principle and Quality Evaluation of Genuine Medicinal Materials, Translational Chinese Medicine Key Laboratory of Sichuan Province, Chengdu, China
| |
Collapse
|
45
|
Bai L, Yan X, Lv J, Qi P, Song X, Zhang L. Intestinal Flora in Chemotherapy Resistance of Biliary Pancreatic Cancer. BIOLOGY 2023; 12:1151. [PMID: 37627035 PMCID: PMC10452461 DOI: 10.3390/biology12081151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 08/18/2023] [Accepted: 08/18/2023] [Indexed: 08/27/2023]
Abstract
Biliary pancreatic malignancy has an occultic onset, a high degree of malignancy, and a poor prognosis. Most clinical patients miss the opportunity for surgical resection of the tumor. Systemic chemotherapy is still one of the important methods for the treatment of biliary pancreatic malignancies. Many chemotherapy regimens are available, but their efficacy is not satisfactory, and the occurrence of chemotherapy resistance is a major reason leading to poor prognosis. With the advancement of studies on intestinal flora, it has been found that intestinal flora is correlated with and plays an important role in chemotherapy resistance. The application of probiotics and other ways to regulate intestinal flora can improve this problem. This paper aims to review and analyze the research progress of intestinal flora in the chemotherapy resistance of biliary pancreatic malignancies to provide new ideas for treatment.
Collapse
Affiliation(s)
- Liuhui Bai
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, China; (L.B.); (X.Y.); (J.L.); (P.Q.); (X.S.)
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, China
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, The First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Xiangdong Yan
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, China; (L.B.); (X.Y.); (J.L.); (P.Q.); (X.S.)
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, China
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, The First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Jin Lv
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, China; (L.B.); (X.Y.); (J.L.); (P.Q.); (X.S.)
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, China
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, The First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Ping Qi
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, China; (L.B.); (X.Y.); (J.L.); (P.Q.); (X.S.)
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, China
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, The First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Xiaojing Song
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, China; (L.B.); (X.Y.); (J.L.); (P.Q.); (X.S.)
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, China
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, The First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Lei Zhang
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, China; (L.B.); (X.Y.); (J.L.); (P.Q.); (X.S.)
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, China
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, The First Hospital of Lanzhou University, Lanzhou 730000, China
| |
Collapse
|
46
|
Liu C, Zhang H, Li T, Jiang Z, Yuan Y, Chen X. Fusobacterium nucleatum Promotes Megakaryocyte Maturation in Patients with Gastric Cancer via Inducing the Production of Extracellular Vesicles Containing 14-3-3ε. Infect Immun 2023; 91:e0010223. [PMID: 37404144 PMCID: PMC10429653 DOI: 10.1128/iai.00102-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 06/11/2023] [Indexed: 07/06/2023] Open
Abstract
Fusobacterium nucleatum colonization contributes to the occurrence of portal vein thrombosis in patients with gastric cancer (GC). However, the underlying mechanism by which F. nucleatum promotes thrombosis remains unclear. In this study, we recruited a total of 91 patients with GC and examined the presence of F. nucleatum in tumor and adjacent non-tumor tissues by fluorescence in situ hybridization and quantitative PCR. Neutrophil extracellular traps (NETs) were detected by immunohistochemistry. Extracellular vesicles (EVs) were extracted from the peripheral blood and proteins in the EVs were identified by mass spectrometry (MS). HL-60 cells differentiated into neutrophils were used to package engineered EVs to imitate the EVs released from NETs. Hematopoietic progenitor cells (HPCs) and K562 cells were used for megakaryocyte (MK) in vitro differentiation and maturation to examine the function of EVs. We observed that F. nucleatum-positive patients had increased NET and platelet counts. EVs from F. nucleatum-positive patients could promote the differentiation and maturation of MKs and had upregulated 14-3-3 proteins, especially 14-3-3ε. 14-3-3ε upregulation promoted MK differentiation and maturation in vitro. HPCs and K562 cells could receive 14-3-3ε from the EVs, which interacted with GP1BA and 14-3-3ζ to trigger PI3K-Akt signaling. In conclusion, we identified for the first time that F. nucleatum infection promotes NET formation, which releases EVs containing 14-3-3ε. These EVs could deliver 14-3-3ε to HPCs and promote their differentiation into MKs via activation of PI3K-Akt signaling.
Collapse
Affiliation(s)
- Chang Liu
- Department of Critical Care Medicine, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan, China
- Department of Cardiovascular Medicine, The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - He Zhang
- Department of Pathology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Tiepeng Li
- Department of Immunology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Zhiqiang Jiang
- Department of General Surgery, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Yiqiang Yuan
- Department of Cardiovascular Medicine, The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
- Department of Cardiovascular Medicine, The 7th People’s Hospital of Zhengzhou, Henan Cardiovascular Hospital Affiliated to Southern Medical University/The Second School of Clinical Medicine, Southern Medical University, Zhengzhou, Henan, China
- Department of Cardiovascular Medicine, Henan Provincial Chest Hospital, Zhengzhou, Henan, China
| | - Xiaobing Chen
- Department of Gastrointestinal Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan, China
| |
Collapse
|
47
|
Mekapogu AR, Xu Z, Pothula S, Perera C, Pang T, Hosen SMZ, Damalanka V, Janetka J, Goldstein D, Pirola R, Wilson J, Apte M. HGF/c-Met pathway inhibition combined with chemotherapy increases cytotoxic T-cell infiltration and inhibits pancreatic tumour growth and metastasis. Cancer Lett 2023; 568:216286. [PMID: 37354984 DOI: 10.1016/j.canlet.2023.216286] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 06/17/2023] [Accepted: 06/20/2023] [Indexed: 06/26/2023]
Abstract
Pancreatic cancer (PC) is a deadly cancer with a high mortality rate. The unique characteristics of PC, including desmoplasia and immunosuppression, have made it difficult to develop effective treatment strategies. Pancreatic stellate cells (PSCs) play a crucial role in the progression of the disease by interacting with cancer cells. One of the key mediators of PSC - cancer cell interactions is the hepatocyte growth factor (HGF)/c-MET pathway. Using an immunocompetent in vivo model of PC as well as in vitro experiments, this study has shown that a combined approach using HGF/c-MET inhibitors to target stromal-tumour interactions and chemotherapy (gemcitabine) to target cancer cells effectively decreases tumour volume, EMT, and stemness, and importantly, eliminates metastasis. Notably, HGF/c-MET inhibition decreases TGF-β secretion by cancer cells, resulting in an increase in cytotoxic T-cell infiltration, thus contributing to cancer cell death in tumours. HGF/c-MET inhibition + chemotherapy was also found to normalise the gut microbiome and improve gut microbial diversity. These findings provide a strong platform for assessment of this triple therapy (HGF/c-MET inhibition + chemotherapy) approach in the clinical setting.
Collapse
Affiliation(s)
- Alpha Raj Mekapogu
- Pancreatic Research Group, South West Sydney Clinical Campuses, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Australia; Ingham Institute for Applied Medical Research, Sydney, Australia
| | - Zhihong Xu
- Pancreatic Research Group, South West Sydney Clinical Campuses, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Australia; Ingham Institute for Applied Medical Research, Sydney, Australia
| | - Srinivasa Pothula
- Pancreatic Research Group, South West Sydney Clinical Campuses, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Australia; AbCellera, Beaconsfield, New South Wales, United Kingdom
| | - Chamini Perera
- Pancreatic Research Group, South West Sydney Clinical Campuses, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Australia; Ingham Institute for Applied Medical Research, Sydney, Australia
| | - Tony Pang
- Pancreatic Research Group, South West Sydney Clinical Campuses, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Australia; Surgical Innovations Unit, Westmead Hospital, Sydney, Australia; Westmead Clinical School, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - S M Zahid Hosen
- Pancreatic Research Group, South West Sydney Clinical Campuses, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Australia; Ingham Institute for Applied Medical Research, Sydney, Australia
| | - Vishnu Damalanka
- Department of Biochemistry and Molecular Biophysics, Washington University, St. Louis, USA
| | - James Janetka
- Department of Biochemistry and Molecular Biophysics, Washington University, St. Louis, USA
| | - David Goldstein
- Pancreatic Research Group, South West Sydney Clinical Campuses, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Australia
| | - Romano Pirola
- Pancreatic Research Group, South West Sydney Clinical Campuses, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Australia
| | - Jeremy Wilson
- Pancreatic Research Group, South West Sydney Clinical Campuses, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Australia; Ingham Institute for Applied Medical Research, Sydney, Australia
| | - Minoti Apte
- Pancreatic Research Group, South West Sydney Clinical Campuses, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Australia; Ingham Institute for Applied Medical Research, Sydney, Australia.
| |
Collapse
|
48
|
Bruno JS, Fregnani ER. Oral microbiome as a new research-target for supportive care and precision oncology. Curr Opin Oncol 2023; 35:276-281. [PMID: 37222190 DOI: 10.1097/cco.0000000000000947] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
PURPOSE OF REVIEW A growing number of studies demonstrate the oral bacterial shift in cancer patients and the enrichment of oral bacteria in distant tumours. During the oncological treatment, opportunistic oral bacteria correlate with oral toxicities. This review focused on the most recent studies to identify which genera are the most mentioned and deserved further investigation. RECENT FINDINGS This review evaluated bacterial changes in patients with head and neck, colorectal, lung and breast cancer. Greater composition of disease-related genera (e.g., Fusobacterium , Porphyromonas , Lactobacillus , Streptococcus , and Parvimonas ) are present in the oral cavity of these groups of patients. The tumour specimen characterisation of head and neck, pancreatic and colorectal cancer also describes the presence of oral taxa. No evidence indicates that commensal oral bacteria have protective roles in distant tumours. Regardless, oral care is critical to prevent the growth of oral pathogens and reduce infection foci. SUMMARY Recent evidence suggests that oral microbiota is a potential biomarker for oncological clinical outcomes and oral toxicities. Currently, the literature presents a remarkable methodological variety - from the sample collection site to the preference of the data analysis tools. For the oral microbiome to achieve the stage of being used as a clinical tool in the oncological context, more studies are necessary.
Collapse
|
49
|
Reuss JM, Alonso-Gamo L, Garcia-Aranda M, Reuss D, Albi M, Albi B, Vilaboa D, Vilaboa B. Oral Mucosa in Cancer Patients-Putting the Pieces Together: A Narrative Review and New Perspectives. Cancers (Basel) 2023; 15:3295. [PMID: 37444405 DOI: 10.3390/cancers15133295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 06/15/2023] [Accepted: 06/18/2023] [Indexed: 07/15/2023] Open
Abstract
The oral mucosa is a key player in cancer patients and during cancer treatment. The increasing prevalence of cancer and cancer-therapy-associated side effects are behind the major role that oral mucosa plays in oncological patients. Oral mucositis is a debilitating severe complication caused by the early toxicity of chemo and/or radiotherapy that can restrict treatment outcome possibilities, even challenging a patient's survival. It has been referred to as the most feared cancer treatment complication. Predictive variables as to who will be affected, and to what extent, are still unclear. Additionally, oral mucositis is one of the sources of the increasing economic burden of cancer, not only for patients and their families but also for institutions and governments. All efforts should be implemented in the search for new approaches to minimize the apparently ineluctable outburst of oral mucositis during cancer treatment. New perspectives derived from different approaches to explaining the interrelation between oral mucositis and the oral microbiome or the similarities with genitourinary mucosa may help elucidate the biomolecular pathways and mechanisms behind oral mucosa cancer-therapy-related toxicity, and what is more important is its management in order to minimize treatment side effects and provide enhanced cancer support.
Collapse
Affiliation(s)
- Jose Manuel Reuss
- Department of Postgraduate Prosthodontics, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Laura Alonso-Gamo
- Department of Pediatrics, Hospital Infanta Cristina, 28981 Madrid, Spain
| | - Mariola Garcia-Aranda
- Centro Integral Oncológico Clara Campal, Department of Oncologic Radiotherapy, Hospital Universitario Sanchinarro, 28050 Madrid, Spain
| | - Debora Reuss
- Lecturer Dental School, Universidad San Pablo CEU, 28003 Madrid, Spain
| | - Manuel Albi
- Department of Gynecology and Obstetrics, Quironsalud Group Public Hospitals, 28223 Madrid, Spain
| | - Beatriz Albi
- Department of Gynecology and Obstetrics, Hospital Universitario Fundación Jiménez Díaz, 28040 Madrid, Spain
| | - Debora Vilaboa
- Aesthetic Dentistry Department, Universidad San Pablo CEU, 28003 Madrid, Spain
| | | |
Collapse
|
50
|
Kaune T, Griesmann H, Theuerkorn K, Hämmerle M, Laumen H, Krug S, Plumeier I, Kahl S, Junca H, Gustavo dos Anjos Borges L, Michl P, Pieper DH, Rosendahl J. Gender-specific changes of the gut microbiome correlate with tumor development in murine models of pancreatic cancer. iScience 2023; 26:106841. [PMID: 37255660 PMCID: PMC10225934 DOI: 10.1016/j.isci.2023.106841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 02/14/2023] [Accepted: 05/04/2023] [Indexed: 06/01/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a devastating disease with a dismal outcome. To improve understanding of sequential microbiome changes during PDAC development we analyzed mouse models of pancreatic carcinogenesis (KC mice recapitulating pre-invasive PanIN formation, as well as KPC mice recapitulating invasive PDAC) during early tumor development and subsequent tumor progression. Diversity and community composition were analyzed depending on genotype, age, and gender. Both mouse models demonstrated concordant abundance changes of several genera influenced by one or more of the investigated factors. Abundance was significantly impacted by gender, highlighting the need to further elucidate the impact of gender differences. The findings underline the importance of the microbiome in PDAC development and indicate that microbiological screening of patients at risk and targeting the microbiome in PDAC development may be feasible in future.
Collapse
Affiliation(s)
- Tom Kaune
- Department of Internal Medicine I, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Heidi Griesmann
- Department of Internal Medicine I, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Katharina Theuerkorn
- Department of Internal Medicine I, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Monika Hämmerle
- Institute of Pathology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Helmut Laumen
- Department of Internal Medicine I, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Sebastian Krug
- Department of Internal Medicine I, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
- Klinik für Innere Medizin IV, Universitätsklinikum Heidelberg, Heidelberg, Germany
| | - Iris Plumeier
- Microbial Interactions and Processes Research Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Silke Kahl
- Microbial Interactions and Processes Research Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Howard Junca
- Microbial Interactions and Processes Research Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | | | - Patrick Michl
- Department of Internal Medicine I, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
- Klinik für Innere Medizin IV, Universitätsklinikum Heidelberg, Heidelberg, Germany
| | - Dietmar H. Pieper
- Microbial Interactions and Processes Research Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Jonas Rosendahl
- Department of Internal Medicine I, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| |
Collapse
|