1
|
Cheng X, Shao P, Wang X, Jiang J, Chen J, Zhu J, Zhu W, Li Y, Zhang J, Chen J, Huang Z. Myeloid-Derived Suppressor Cell Accumulation Drives Intestinal Fibrosis through mCCL6/hCCL15 Chemokine-Mediated Fibroblast Activation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2411711. [PMID: 39739231 PMCID: PMC11848553 DOI: 10.1002/advs.202411711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Indexed: 01/02/2025]
Abstract
Intestinal fibrosis, a severe complication of Crohn's disease (CD), is linked to chronic inflammation, but the precise mechanism by which immune-driven intestinal inflammation leads to fibrosis development is not fully understood. This study investigates the role of myeloid-derived suppressor cells (MDSCs) in intestinal fibrosis in CD patients and a 2,4,6-trinitrobenzene sulfonic acid (TNBS)-induced mouse model. Elevated MDSCs are observed in inflamed intestinal tissues prior to fibrosis and their sustained presence in fibrotic tissues of both CD patients and murine models. Depletion of MDSCs significantly reduces fibrosis, highlighting their key role in the fibrotic process. Mechanistically, MDSC-derived mCCL6 activates fibroblasts via the CCR1-MAPK signaling, and interventions targeting this axis, including neutralizing antibodies, a CCR1 antagonist, or fibroblast-specific Ccr1 knockout mice reduce fibrosis. In CD patients with stenosis, human CCL15, analogous to mCCL6, is found to be elevated in MDSCs and activated fibroblasts. Additionally, CXCR2 and CCR2 ligands are identified as key mediators of MDSC recruitment in intestinal fibrosis. Blocking MDSC recruitment with CXCR2 and CCR2 antagonists alleviates intestinal fibrosis. These findings suggest that strategies targeting MDSC recruitment and mCCL6/hCCL15 signaling could offer therapeutic benefits for intestinal fibrosis.
Collapse
Affiliation(s)
- Xiaohui Cheng
- State Key Laboratory of Pharmaceutical BiotechnologySchool of Life SciencesNanjing UniversityNanjingJiangsu210023China
| | - Pingwen Shao
- State Key Laboratory of Pharmaceutical BiotechnologySchool of Life SciencesNanjing UniversityNanjingJiangsu210023China
| | - XinTong Wang
- State Key Laboratory of Pharmaceutical BiotechnologySchool of Life SciencesNanjing UniversityNanjingJiangsu210023China
| | - Juan Jiang
- State Key Laboratory of Pharmaceutical BiotechnologySchool of Life SciencesNanjing UniversityNanjingJiangsu210023China
| | - Jiahui Chen
- State Key Laboratory of Pharmaceutical BiotechnologySchool of Life SciencesNanjing UniversityNanjingJiangsu210023China
| | - Jie Zhu
- State Key Laboratory of Pharmaceutical BiotechnologySchool of Life SciencesNanjing UniversityNanjingJiangsu210023China
| | - Weiming Zhu
- Department of General SurgeryJinling HospitalSchool of MedicineNanjing UniversityNanjingJiangsu210002China
| | - Yi Li
- Department of General SurgeryJinling HospitalSchool of MedicineNanjing UniversityNanjingJiangsu210002China
| | - Junfeng Zhang
- State Key Laboratory of Pharmaceutical BiotechnologySchool of Life SciencesNanjing UniversityNanjingJiangsu210023China
| | - Jiangning Chen
- State Key Laboratory of Pharmaceutical BiotechnologySchool of Life SciencesNanjing UniversityNanjingJiangsu210023China
- State Key Laboratory of Analytical Chemistry for Life SciencesNanjing UniversityNanjingJiangsu210023China
| | - Zhen Huang
- State Key Laboratory of Pharmaceutical BiotechnologySchool of Life SciencesNanjing UniversityNanjingJiangsu210023China
- NJU Xishan Institute of Applied BiotechnologyXishan DistrictWuxiJiangsu214101China
| |
Collapse
|
2
|
Lawrence ALE, Berger RP, Hill DR, Huang S, Yadagiri VK, Bons B, Fields C, Knight JS, Wobus CE, Spence JR, Young VB, Abuaita BH, O'Riordan MX. Neutrophil prime unique transcriptional responses in intestinal organoids during infection with nontyphoidal Salmonella enterica serovars. mSphere 2024; 9:e0069324. [PMID: 39565098 DOI: 10.1128/msphere.00693-24] [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: 08/22/2024] [Accepted: 09/16/2024] [Indexed: 11/21/2024] Open
Abstract
Nontyphoidal strains of Salmonella enterica are a major cause of foodborne illnesses, and infection with these bacteria results in inflammatory gastroenteritis. Polymorphonuclear leukocytes (PMNs), also known as neutrophils, are a dominant immune cell type found at the site of infection in Salmonella-infected individuals, but how they regulate infection outcome is not well understood. Here, we used a co-culture model of primary human PMNs and human intestinal organoids to probe the role of PMNs during infection with two of the most prevalent Salmonella serovars: Salmonella enterica serovar Enteritidis and Typhimurium. Using a transcriptomics approach, we identified a dominant role for PMNs in mounting differential immune responses including production of pro-inflammatory cytokines, chemokines, and antimicrobial peptides. We also identified specific gene sets that were induced by PMNs in response to Enteritidis or Typhimurium infection. By comparing host responses to these serovars, we uncovered differential regulation of host metabolic pathways particularly induction of cholesterol biosynthetic pathways during Typhimurium infection and suppression of RNA metabolism during Enteritidis infection. Together, these findings provide insight into the role of human PMNs in modulating different host responses to pathogens that cause similar disease in humans.IMPORTANCENontyphoidal serovars of Salmonella enterica are known to induce robust recruitment of polymorphonuclear leukocytes (PMNs) in the gut during early stages of infection, but the specific role of PMNs in regulating infection outcome of different serovars is poorly understood. Due to differences in human infection progression compared to small animal models, characterizing the role of PMNs during infection has been challenging. Here, we used a co-culture model of human intestinal organoids with human primary PMNs to study the role of PMNs during infection of human intestinal epithelium. Using a transcriptomics approach, we define PMN-dependent reprogramming of the host response to Salmonella, establishing a clear role in amplifying pro-inflammatory gene expression. Additionally, the host response driven by PMNs differed between two similar nontyphoidal Salmonella serovars. These findings highlight the importance of building more physiological infection models to replicate human infection conditions to study host responses specific to individual pathogens.
Collapse
Affiliation(s)
- Anna-Lisa E Lawrence
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Ryan P Berger
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - David R Hill
- Department of Internal Medicine/Infectious Diseases Division, University of Michigan Medical School, Ann Arbor, Michigan, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Sha Huang
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Veda K Yadagiri
- Department of Internal Medicine/Infectious Diseases Division, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Brooke Bons
- Department of Internal Medicine/Infectious Diseases Division, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Courtney Fields
- Department of Internal Medicine/Infectious Diseases Division, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Jason S Knight
- Department of Internal Medicine/Infectious Diseases Division, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Christiane E Wobus
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Jason R Spence
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Vincent B Young
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
- Department of Internal Medicine/Infectious Diseases Division, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Basel H Abuaita
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Mary X O'Riordan
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| |
Collapse
|
3
|
Winge MCG, Nasrallah M, Jackrazi LV, Guo KQ, Fuhriman JM, Szafran R, Ramanathan M, Gurevich I, Nguyen NT, Siprashvili Z, Inayathullah M, Rajadas J, Porter DF, Khavari PA, Butte AJ, Marinkovich MP. Repurposing an epithelial sodium channel inhibitor as a therapy for murine and human skin inflammation. Sci Transl Med 2024; 16:eade5915. [PMID: 39661704 DOI: 10.1126/scitranslmed.ade5915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 04/12/2024] [Accepted: 11/18/2024] [Indexed: 12/13/2024]
Abstract
Inflammatory skin disease is characterized by a pathologic interplay between skin cells and immunocytes and can result in disfiguring cutaneous lesions and systemic inflammation. Immunosuppression is commonly used to target the inflammatory component; however, these drugs are often expensive and associated with side effects. To identify previously unidentified targets, we carried out a nonbiased informatics screen to identify drug compounds with an inverse transcriptional signature to keratinocyte inflammatory signals. Using psoriasis, a prototypic inflammatory skin disease, as a model, we used pharmacologic, transcriptomic, and proteomic characterization to find that benzamil, the benzyl derivative of the US Food and Drug Administration-approved diuretic amiloride, effectively reversed keratinocyte-driven inflammatory signaling. Through three independent mouse models of skin inflammation (Rac1G12V transgenic mice, topical imiquimod, and human skin xenografts from patients with psoriasis), we found that benzamil disrupted pathogenic interactions between the small GTPase Rac1 and its adaptor NCK1. This reduced STAT3 and NF-κB signaling and downstream cytokine production in keratinocytes. Genetic knockdown of sodium channels or pharmacological inhibition by benzamil prevented excess Rac1-NCK1 binding and limited proinflammatory signaling pathway activation in patient-derived keratinocytes without systemic immunosuppression. Both systemic and topical applications of benzamil were efficacious, suggesting that it may be a potential therapeutic avenue for treating skin inflammation.
Collapse
Affiliation(s)
- Mårten C G Winge
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Mazen Nasrallah
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Leandra V Jackrazi
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Konnie Q Guo
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jessica M Fuhriman
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Rebecca Szafran
- Unit of Dermatology, ME GHR, Karolinska University Hospital, SE-17176 Stockholm, Sweden
- Department of Medicine Solna, Karolinska Institutet, SE-17176 Stockholm, Sweden
| | - Muthukumar Ramanathan
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Irina Gurevich
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Ngon T Nguyen
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Zurab Siprashvili
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Mohammed Inayathullah
- Advanced Drug Delivery and Regenerative Biomaterials Laboratory, Cardiovascular Institute, Department of Medicine, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Jayakumar Rajadas
- Advanced Drug Delivery and Regenerative Biomaterials Laboratory, Cardiovascular Institute, Department of Medicine, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Douglas F Porter
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Paul A Khavari
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
- Dermatology Service, Veterans Affairs Medical Center, Palo Alto, CA 94304, USA
| | - Atul J Butte
- Bakar Computational Health Sciences Institute, University of California San Francisco, San Francisco, CA 94158, USA
| | - M Peter Marinkovich
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
- Dermatology Service, Veterans Affairs Medical Center, Palo Alto, CA 94304, USA
| |
Collapse
|
4
|
Nishiyama NC, Silverstein S, Darlington K, Kennedy Ng MM, Clough KM, Bauer M, Beasley C, Bharadwaj A, Ganesan R, Kapadia MR, Lau G, Lian G, Rahbar R, Sadiq TS, Schaner MR, Stem J, Friton J, Faubion WA, Sheikh SZ, Furey TS. eQTL in diseased colon tissue identifies novel target genes associated with IBD. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.14.618229. [PMID: 39464142 PMCID: PMC11507739 DOI: 10.1101/2024.10.14.618229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Genome-wide association studies (GWAS) have identified over 300 loci associated with the inflammatory bowel diseases (IBD), but putative causal genes for most are unknown. We conducted the largest disease-focused expression quantitative trait loci (eQTL) analysis using colon tissue from 252 IBD patients to determine genetic effects on gene expression and potential contribution to IBD. Combined with two non-IBD colon eQTL studies, we identified 194 potential target genes for 108 GWAS loci. eQTL in IBD tissue were enriched for IBD GWAS loci colocalizations, provided novel evidence for IBD-associated genes such as ABO and TNFRSF14, and identified additional target genes compared to non-IBD tissue eQTL. IBD-associated eQTL unique to diseased tissue had distinct regulatory and functional characteristics with increased effect sizes. Together, these highlight the importance of eQTL studies in diseased tissue for understanding functional consequences of genetic variants, and elucidating molecular mechanisms and regulation of key genes involved in IBD.
Collapse
Affiliation(s)
- Nina C. Nishiyama
- Curriculum in Bioinformatics and Computational Biology, Department of Genetics, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Center for Gastrointestinal Biology and Disease, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Sophie Silverstein
- Center for Gastrointestinal Biology and Disease, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Genetics, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Kimberly Darlington
- Center for Gastrointestinal Biology and Disease, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Division of Gastroenterology and Hepatology, Department of Medicine, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Meaghan M. Kennedy Ng
- Curriculum in Bioinformatics and Computational Biology, Department of Genetics, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Center for Gastrointestinal Biology and Disease, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Katelyn M. Clough
- Center for Gastrointestinal Biology and Disease, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Curriculum in Toxicology and Environmental Medicine, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Mikaela Bauer
- Center for Gastrointestinal Biology and Disease, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Caroline Beasley
- Center for Gastrointestinal Biology and Disease, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Genetics, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Akshatha Bharadwaj
- Department of Genetics, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Rajee Ganesan
- Department of Genetics, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Muneera R. Kapadia
- Department of Surgery, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Gwen Lau
- Center for Gastrointestinal Biology and Disease, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Grace Lian
- Center for Gastrointestinal Biology and Disease, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Reza Rahbar
- Department of Surgery, REX Healthcare of Wakefield, Raleigh, North Carolina, USA
| | - Timothy S. Sadiq
- Department of Surgery, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Matthew R. Schaner
- Center for Gastrointestinal Biology and Disease, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Jonathan Stem
- Department of Surgery, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Jessica Friton
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - William A. Faubion
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Shehzad Z. Sheikh
- Center for Gastrointestinal Biology and Disease, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Genetics, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Division of Gastroenterology and Hepatology, Department of Medicine, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Curriculum in Toxicology and Environmental Medicine, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Terrence S. Furey
- Curriculum in Bioinformatics and Computational Biology, Department of Genetics, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Center for Gastrointestinal Biology and Disease, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Genetics, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
5
|
Chae IG, Jung J, Kim DH, Choi JS, Chun KS. EP4 receptor agonist CAY10598 upregulates ROS-dependent Hsp90 cleavage in colorectal cancer cells. Free Radic Res 2024; 58:596-605. [PMID: 39258904 DOI: 10.1080/10715762.2024.2396909] [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: 05/23/2024] [Revised: 07/22/2024] [Accepted: 08/19/2024] [Indexed: 09/12/2024]
Abstract
Prostaglandin E2 (PGE2) interacts with four specific G protein-coupled receptors, namely EP1, EP2, EP3, and EP4, playing a pivotal role in determining the fate of cells. Our previous findings highlighted that stimulating the EP4 receptor with its agonist, CAY10598, triggers apoptosis in colon cancer HCT116 cells via the production of reactive oxygen species (ROS). This process also reduces the phosphorylation of the oncogenic protein JAK2 and leads to its degradation in these cells. In this study, our goal was to explore the pathways through which CAY10598 leads to JAK2 degradation. We focused on Hsp90, a heat shock protein family member known for its role as a molecular chaperone maintaining the stability of several key proteins including EGFR, MET, Akt, and JAK2. Our results show that CAY10598 decreases the levels of client proteins of Hsp90 in HCT116 cells, an effect reversible by pretreatment with the ROS scavenger N-acetyl cysteine (NAC) or the proteasome inhibitor MG132, indicating that the degradation is likely driven by ROS. Furthermore, we observed that CAY10598 cleaves both α and β isoforms of Hsp90, the process inhibited by NAC. Inhibition of EP4 with the antagonist GW627368x not only prevented the degradation of Hsp90 client proteins but also the cleavage of Hsp90 itself in CAY10598-treated HCT116 cells. Additionally, CAY10598 suppressed the growth of HCT116 cells implanted in mice. Our findings reveal that CAY10598 induces apoptosis in cancer cells by a novel mechanism involving the ROS-dependent cleavage of Hsp90, thereby inhibiting the function of crucial Hsp90 client proteins.
Collapse
Affiliation(s)
- In Gyung Chae
- College of Pharmacy, Keimyung University, Daegu, Republic of Korea
- Gyeongbuk Institute for Bio Industry (GIB), Gyeongbuk, Republic of Korea
| | - Joohee Jung
- College of Pharmacy, Duksung Women's University, Seoul, Republic of Korea
- Innovative Drug Center, Duksung Women's University, Seoul, Republic of Korea
| | - Do-Hee Kim
- Department of Chemistry, Kyonggi University, Suwon, Republic of Korea
| | - Joon-Seok Choi
- College of Pharmacy, Daegu Catholic University, Gyeongbuk, Republic of Korea
| | - Kyung-Soo Chun
- College of Pharmacy, Keimyung University, Daegu, Republic of Korea
| |
Collapse
|
6
|
Zhou T, Wu J, Khan A, Hu T, Wang Y, Salama ES, Su S, Han H, Jin W, Li X. A probiotic Limosilactobacillus fermentum GR-3 mitigates colitis-associated tumorigenesis in mice via modulating gut microbiome. NPJ Sci Food 2024; 8:61. [PMID: 39242568 PMCID: PMC11379937 DOI: 10.1038/s41538-024-00307-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 08/29/2024] [Indexed: 09/09/2024] Open
Abstract
Bacterial therapy for colorectal cancer (CRC) represents a burgeoning frontier. The probiotic Limosilactobacillus fermentum GR-3, derived from traditional food "Jiangshui", exhibited superior antioxidant capacity by producing indole derivatives ICA and IPA. In an AOM/DSS-induced CRC mouse model, GR-3 treatment alleviated weight loss, colon shortening, rectal bleeding and intestinal barrier disruption by reducing oxidative stress and inflammation. GR-3 colonization in distant colon induced apoptosis and reduced tumor incidence by 51.2%, outperforming the control strain and vitamin C. The beneficial effect of GR-3 on CRC was associated with gut microbiome modulation, increasing SCFA producer Lachnospiraceae NK4A136 group and suppressing pro-inflammatory strain Bacteroides. Metagenomic and metabolic analyses revealed that GR-3 intervention upregulated antioxidant genes (xseA, ALDH) and butyrate synthesis gene (bcd), while increasing beneficial metabolites (SCFAs, ICA, IPA, VB12 and VD3) and reducing harmful secondary bile acids. Overall, GR-3 emerges as a promising candidate in CRC therapy, offering effective gut microbiome remediation.
Collapse
Affiliation(s)
- Tuoyu Zhou
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou, Gansu, 730000, China
- State Key Laboratory of Grassland Agro-ecosystems, College of Pastoral Agricultural Science and Technology, Lanzhou University, Lanzhou, China
| | - Jingyuan Wu
- The First Clinical Medical College of Lanzhou University, Lanzhou University, Lanzhou, 730000, China
| | - Aman Khan
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou, Gansu, 730000, China
| | - Tianxiang Hu
- Georgia Cancer Center, Augusta University, 1410 Laney Walker Blvd, Augusta, GA, 30912, USA
| | - Yiqing Wang
- The First Clinical Medical College of Lanzhou University, Lanzhou University, Lanzhou, 730000, China
| | - El-Sayed Salama
- Department of Occupational and Environmental Health, School of Public Health, Lanzhou University, Lanzhou, 730000, Gansu Province, PR China
| | - Shaochen Su
- Healthy Examination & Management Center, First Hospital of Lanzhou University, Lanzhou, 730000, China
| | - Huawen Han
- State Key Laboratory of Grassland Agro-ecosystems, College of Pastoral Agricultural Science and Technology, Lanzhou University, Lanzhou, China.
| | - Weilin Jin
- Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, Lanzhou, China.
| | - Xiangkai Li
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou, Gansu, 730000, China.
| |
Collapse
|
7
|
Tang W, Lo CWS, Ma W, Chu ATW, Tong AHY, Chung BHY. Revealing the role of SPP1 + macrophages in glioma prognosis and therapeutic targeting by investigating tumor-associated macrophage landscape in grade 2 and 3 gliomas. Cell Biosci 2024; 14:37. [PMID: 38515213 PMCID: PMC10956315 DOI: 10.1186/s13578-024-01218-4] [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: 10/27/2023] [Accepted: 03/13/2024] [Indexed: 03/23/2024] Open
Abstract
BACKGROUND Glioma is a highly heterogeneous brain tumor categorized into World Health Organization (WHO) grades 1-4 based on its malignancy. The suppressive immune microenvironment of glioma contributes significantly to unfavourable patient outcomes. However, the cellular composition and their complex interplays within the glioma environment remain poorly understood, and reliable prognostic markers remain elusive. Therefore, in-depth exploration of the tumor microenvironment (TME) and identification of predictive markers are crucial for improving the clinical management of glioma patients. RESULTS Our analysis of single-cell RNA-sequencing data from glioma samples unveiled the immunosuppressive role of tumor-associated macrophages (TAMs), mediated through intricate interactions with tumor cells and lymphocytes. We also discovered the heterogeneity within TAMs, among which a group of suppressive TAMs named TAM-SPP1 demonstrated a significant association with Epidermal Growth Factor Receptor (EGFR) amplification, impaired T cell response and unfavourable patient survival outcomes. Furthermore, by leveraging genomic and transcriptomic data from The Cancer Genome Atlas (TCGA) dataset, two distinct molecular subtypes with a different constitution of TAMs, EGFR status and clinical outcomes were identified. Exploiting the molecular differences between these two subtypes, we developed a four-gene-based prognostic model. This model displayed strong associations with an elevated level of suppressive TAMs and could be used to predict anti-tumor immune response and prognosis in glioma patients. CONCLUSION Our findings illuminated the molecular and cellular mechanisms that shape the immunosuppressive microenvironment in gliomas, providing novel insights into potential therapeutic targets. Furthermore, the developed prognostic model holds promise for predicting immunotherapy response and assisting in more precise risk stratification for glioma patients.
Collapse
Affiliation(s)
- Wenshu Tang
- Hong Kong Genome Institute, 2/F, Building 20E, Hong Kong Science Park, Hong Kong, China
| | - Cario W S Lo
- Hong Kong Genome Institute, 2/F, Building 20E, Hong Kong Science Park, Hong Kong, China
| | - Wei Ma
- Hong Kong Genome Institute, 2/F, Building 20E, Hong Kong Science Park, Hong Kong, China
| | - Annie T W Chu
- Hong Kong Genome Institute, 2/F, Building 20E, Hong Kong Science Park, Hong Kong, China
| | - Amy H Y Tong
- Hong Kong Genome Institute, 2/F, Building 20E, Hong Kong Science Park, Hong Kong, China
| | - Brian H Y Chung
- Hong Kong Genome Institute, 2/F, Building 20E, Hong Kong Science Park, Hong Kong, China.
- Department of Pediatrics and Adolescent Medicine, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
8
|
Jiao X, Liu B, Dong X, Wang S, Cai X, Zhang H, Qin Z. Exploring PLGA-OH-CATH30 Microspheres for Oral Therapy of Escherichia coli-Induced Enteritis. Biomolecules 2024; 14:86. [PMID: 38254686 PMCID: PMC10813405 DOI: 10.3390/biom14010086] [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: 11/14/2023] [Revised: 12/20/2023] [Accepted: 01/08/2024] [Indexed: 01/24/2024] Open
Abstract
Antibiotic therapy effectively addresses Escherichia coli-induced enteric diseases, but its excessive utilization results in microbial imbalance and heightened resistance. This study evaluates the therapeutic efficacy of orally administered poly (lactic-co-glycolic acid) (PLGA)-loaded antimicrobial peptide OH-CATH30 microspheres in murine bacterial enteritis. Mice were categorized into the healthy control group (CG), untreated model group (MG), OH-CATH30 treatment group (OC), PLGA-OH-CATH30 treatment group (POC), and gentamicin sulfate treatment group (GS). Except for the control group, all other experimental groups underwent Escherichia coli-induced enteritis, followed by a 5-day treatment period. The evaluation encompassed clinical symptoms, intestinal morphology, blood parameters, inflammatory response, and gut microbiota. PLGA-OH-CATH30 microspheres significantly alleviated weight loss and intestinal damage while also reducing the infection-induced increase in spleen index. Furthermore, these microspheres normalized white blood cell count and neutrophil ratio, suppressed inflammatory factors (IL-1β, IL-6, and TNF-α), and elevated the anti-inflammatory factor IL-10. Analysis of 16S rRNA sequencing results demonstrated that microsphere treatment increased the abundance of beneficial bacteria, including Phocaeicola vulgatus, in the intestinal tract while concurrently decreasing the abundance of pathogenic bacteria, such as Escherichia. In conclusion, PLGA-OH-CATH30 microspheres have the potential to ameliorate intestinal damage and modulate the intestinal microbiota, making them a promising alternative to antibiotics for treating enteric diseases induced by Escherichia coli.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Zhihua Qin
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao 266109, China; (X.J.); (B.L.); (X.D.); (S.W.); (X.C.); (H.Z.)
| |
Collapse
|
9
|
Li XJ, Nie P, Herdewijn P, Sun JG. Unlocking the synthetic approaches and clinical application of approved small-molecule drugs for gastrointestinal cancer treatment: A comprehensive exploration. Eur J Med Chem 2023; 262:115928. [PMID: 37944387 DOI: 10.1016/j.ejmech.2023.115928] [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: 10/15/2023] [Revised: 10/29/2023] [Accepted: 10/31/2023] [Indexed: 11/12/2023]
Abstract
Gastrointestinal (GI) cancers encompass a group of malignancies affecting the digestive system, including the stomach, esophagus, liver, colon, rectum and pancreas. These cancers represent a significant global health burden, necessitating effective treatment strategies. Small-molecule drugs have emerged as crucial therapeutic options in the fight against GI cancers due to their oral bioavailability, targeted mechanisms of action, and well-established safety profiles. The review then elucidates the clinical applications and synthetic methods of clinically approved small-molecule drugs for the treatment of GI cancer, shedding light on their mechanisms of action and their potential in mitigating GI cancer progression. The review also discusses future prospects and the evolving landscape of small-molecule drug development in GI oncology, highlighting the potential for personalized medicine. In summary, this review provides valuable insights into cutting-edge strategies for harnessing clinically approved small-molecule drugs to combat GI cancer effectively.
Collapse
Affiliation(s)
- Xiao-Jing Li
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Peng Nie
- Medicinal Chemistry, Rega Institute of Medical Research, KU Leuven, Herestraat 49, 3000, Leuven, Belgium.
| | - Piet Herdewijn
- Medicinal Chemistry, Rega Institute of Medical Research, KU Leuven, Herestraat 49, 3000, Leuven, Belgium.
| | - Jian-Gang Sun
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China.
| |
Collapse
|
10
|
Zhao H, Li W, Zhou X, Pan L, Feng Y, Gao P, Ji J, Zhang H, Zhao K, Wang C, Lu Z. C-X-C Motif Chemokine Ligand 1 Promotes Colitis by Modulating the Gut Microbiota. J Innate Immun 2023; 16:33-44. [PMID: 38071977 PMCID: PMC10776137 DOI: 10.1159/000535637] [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: 07/17/2023] [Accepted: 12/01/2023] [Indexed: 01/11/2024] Open
Abstract
INTRODUCTION C-X-C motif chemokine ligand 1 (CXCL1) is a potent neutrophil chemoattractant that plays a pivotal role in recruiting neutrophils during inflammatory conditions. This study explored the role of CXCL1 in modulating the gut microbiota, influencing neutrophil infiltration, and contributing to the development of colitis. METHODS We employed quantitative PCR to assess CXCL1 expression in colon samples. A mouse model of dextran sulfate sodium (DSS)-induced colitis was utilized to explore the progression of colitis in wild-type (WT) and CXCL1-deficient (CXCL1-/-) mice. RESULTS Colitis attenuation was evident in CXCL1-/- mice. Significant alterations were observed in the gut microbiome, as revealed by 16S rRNA gene sequencing. Furthermore, CXCL1-/- mice exhibited reduced gut permeability and diminished endotoxin levels in peripheral blood following DSS treatment compared to WT mice. In response to DSS treatment, WT mice showed a clear increase in neutrophil infiltration, while CXCL1-/- mice exhibited lower levels of infiltration. Fecal microbiota transplantation (FMT) using stools from CXCL1-/- mice alleviated DSS-induced colitis. Interestingly, FMT from patients with colitis increased CXCL1 and Ly6G expression in the colons of gut-sterilized mice. Clinical data analysis revealed elevated CXCL1 and CD15 expression in patients with colitis, with a positive correlation between the severity of colitis and the expression of CXCL1 and CD15. CONCLUSION These findings shed light on the pivotal role of CXCL1 in promoting colitis by modulating the gut microbiota.
Collapse
Affiliation(s)
- Hang Zhao
- Department of Gastroenterology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai, China
| | - Wenhua Li
- Department of Gastroenterology, Jiading Branch of Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xin Zhou
- Department of Gastroenterology, Jintan Hospital, Jiangsu University, Changzhou, China
| | - Liang Pan
- Department of Gastroenterology, Jintan Hospital, Jiangsu University, Changzhou, China
| | - Yun Feng
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Pingyu Gao
- Pathology Center, Jintan Hospital, Jiangsu University, Zhenjiang, China
| | - Jie Ji
- Clinical Laboratory, Jintan Hospital, Jiangsu University, Zhenjiang, China
| | - Huanyan Zhang
- Clinical Laboratory, Jintan Hospital, Jiangsu University, Zhenjiang, China
| | - Kai Zhao
- Department of Gastroenterology, Jintan Hospital, Jiangsu University, Changzhou, China
| | - Chi Wang
- Department of Precision Mechanical Engineering, Shanghai University, Shanghai, China
| | - Zhanjun Lu
- Department of Gastroenterology, Jintan Hospital, Jiangsu University, Changzhou, China
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
11
|
Chang CM, Chang CC, Lam HYP, Peng SY, Lai YH, Hsiang BD, Liao YY, Hsu HJ, Jiang SJ. Therapeutic Peptide RF16 Derived from CXCL8 Inhibits MDA-MB-231 Cell Invasion and Metastasis. Int J Mol Sci 2023; 24:14029. [PMID: 37762330 PMCID: PMC10531501 DOI: 10.3390/ijms241814029] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/08/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023] Open
Abstract
Interleukin (IL)-8 plays a vital role in regulating inflammation and breast cancer formation by activating CXCR1/2. We previously designed an antagonist peptide, (RF16), to inhibits the activation of downstream signaling pathways by competing with IL-8 in binding to CXCR1/2, thereby inhibiting IL-8-induced chemoattractant monocyte binding. To evaluate the effect of the RF16 peptide on breast cancer progression, triple-negative MDA-MB-231 and ER-positive MCF-7 breast cancer cells were used to investigate whether RF16 can inhibit the IL-8-induced breast cancer metastasis. Using growth, proliferation, and invasiveness assays, the results revealed that RF16 reduced cell proliferation, migration, and invasiveness in MDA-MB-231 cells. The RF16 peptide also regulated the protein and mRNA expressions of epithelial-mesenchymal transition (EMT) markers in IL-8-stimulated MDA-MB-231 cells. It also inhibited downstream IL-8 signaling and the IL-8-induced inflammatory response via the mitogen-activated protein kinase (MAPK) and Phosphoinositide 3-kinase (PI3K) pathways. In the xenograft tumor mouse model, RF16 synergistically reinforces the antitumor efficacy of docetaxel by improving mouse survival and retarding tumor growth. Our results indicate that RF16 significantly inhibited IL-8-stimulated cell growth, migration, and invasion in MDA-MB-231 breast cancer cells by blocking the activation of p38 and AKT cascades. It indicated that the RF16 peptide may serve as a new supplementary drug for breast cancer.
Collapse
Affiliation(s)
- Chun-Ming Chang
- Department of General Surgery, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 97004, Taiwan;
- Institute of Medical Sciences, Tzu Chi University, Hualien 97004, Taiwan
| | - Chun-Chun Chang
- Department of Laboratory Medicine, Hualien Tzu Chi Hospital, Hualien 97004, Taiwan;
- Department of Laboratory Medicine and Biotechnology, College of Medicine, Tzu Chi University, Hualien 97004, Taiwan
| | - Ho Yin Pekkle Lam
- Department of Biochemistry, School of Medicine, Tzu Chi University, Hualien 97004, Taiwan; (H.Y.P.L.); (S.-Y.P.); (Y.-H.L.)
- Master Program in Biomedical Sciences, School of Medicine, Tzu Chi University, Hualien 97004, Taiwan;
| | - Shih-Yi Peng
- Department of Biochemistry, School of Medicine, Tzu Chi University, Hualien 97004, Taiwan; (H.Y.P.L.); (S.-Y.P.); (Y.-H.L.)
- Master Program in Biomedical Sciences, School of Medicine, Tzu Chi University, Hualien 97004, Taiwan;
| | - Yi-Hsuan Lai
- Department of Biochemistry, School of Medicine, Tzu Chi University, Hualien 97004, Taiwan; (H.Y.P.L.); (S.-Y.P.); (Y.-H.L.)
| | - Bi-Da Hsiang
- Department of Molecular Biology and Human Genetics, School of Medicine, Tzu Chi University, Hualien 97004, Taiwan;
| | - Yu-Yi Liao
- Master Program in Biomedical Sciences, School of Medicine, Tzu Chi University, Hualien 97004, Taiwan;
| | - Hao-Jen Hsu
- Department of Biomedical Sciences and Engineering, College of Medicine, Tzu Chi University, Hualien 97004, Taiwan;
| | - Shinn-Jong Jiang
- Department of Biochemistry, School of Medicine, Tzu Chi University, Hualien 97004, Taiwan; (H.Y.P.L.); (S.-Y.P.); (Y.-H.L.)
- Master Program in Biomedical Sciences, School of Medicine, Tzu Chi University, Hualien 97004, Taiwan;
| |
Collapse
|
12
|
Lê A, Selle A, Aubert P, Durand T, Brosseau C, Bordron P, Delage E, Chaffron S, Petitfils C, Cenac N, Neunlist M, Bodinier M, Rolli-Derkinderen M. Maternal prebiotic supplementation impacts colitis development in offspring mice. Front Nutr 2023; 9:988529. [PMID: 36687706 PMCID: PMC9849907 DOI: 10.3389/fnut.2022.988529] [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: 07/07/2022] [Accepted: 11/11/2022] [Indexed: 01/07/2023] Open
Abstract
Background and aims Maternal diet plays a key role in preventing or contributing to the development of chronic diseases, such as obesity, allergy, and brain disorders. Supplementation of maternal diet with prebiotics has been shown to reduce the risk of food allergies and affect the intestinal permeability in offspring later in life. However, its role in modulating the development of other intestinal disorders, such as colitis, remains unknown. Therefore, we investigated the effects of prebiotic supplementation in pregnant mice on the occurrence of colitis in their offspring. Materials and methods Offspring from mothers, who were administered prebiotic galacto-oligosaccharides and inulin during gestation or fed a control diet, were subjected to three cycles of dextran sulphate sodium (DSS) treatment to induce chronic colitis, and their intestinal function and disease activity were evaluated. Colonic remodelling, gut microbiota composition, and lipidomic and transcriptomic profiles were also assessed. Results DSS-treated offspring from prebiotic-fed mothers presented a higher disease score, increased weight loss, and increased faecal humidity than those from standard diet-fed mothers. DSS-treated offspring from prebiotic-fed mothers also showed increased number of colonic mucosal lymphocytes and macrophages than the control group, associated with the increased colonic concentrations of resolvin D5, protectin DX, and 14-hydroxydocosahexaenoic acid, and modulation of colonic gene expression. In addition, maternal prebiotic supplementation induced an overabundance of eight bacterial families and a decrease in the butyrate caecal concentration in DSS-treated offspring. Conclusion Maternal prebiotic exposure modified the microbiota composition and function, lipid content, and transcriptome of the colon of the offspring. These modifications did not protect against colitis, but rather sensitised the mice to colitis development.
Collapse
Affiliation(s)
- Amélie Lê
- The Enteric Nervous System in Gut and Brain Disorders, Institut des Maladies de l’Appareil Digestif, Institut National Pour la Santé et la Recherche Médicale, Nantes Université, Nantes, France
| | - Amandine Selle
- Unité de Recherche 1268 Biopolymères Interactions Assemblages, Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement, Nantes, France
| | - Philippe Aubert
- The Enteric Nervous System in Gut and Brain Disorders, Institut des Maladies de l’Appareil Digestif, Institut National Pour la Santé et la Recherche Médicale, Nantes Université, Nantes, France
| | - Tony Durand
- The Enteric Nervous System in Gut and Brain Disorders, Institut des Maladies de l’Appareil Digestif, Institut National Pour la Santé et la Recherche Médicale, Nantes Université, Nantes, France
| | - Carole Brosseau
- Unité de Recherche 1268 Biopolymères Interactions Assemblages, Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement, Nantes, France
| | - Philippe Bordron
- The Enteric Nervous System in Gut and Brain Disorders, Institut des Maladies de l’Appareil Digestif, Institut National Pour la Santé et la Recherche Médicale, Nantes Université, Nantes, France
| | - Erwan Delage
- UMR 6004, LS2N, Nantes Université, Ecole Centrale Nantes, CNRS, Nantes, France
| | - Samuel Chaffron
- UMR 6004, LS2N, Nantes Université, Ecole Centrale Nantes, CNRS, Nantes, France
| | - Camille Petitfils
- UMR 1220, Institut de Recherche en Santé Digestive, Toulouse, France
| | - Nicolas Cenac
- UMR 1220, Institut de Recherche en Santé Digestive, Toulouse, France
| | - Michel Neunlist
- The Enteric Nervous System in Gut and Brain Disorders, Institut des Maladies de l’Appareil Digestif, Institut National Pour la Santé et la Recherche Médicale, Nantes Université, Nantes, France
| | - Marie Bodinier
- Unité de Recherche 1268 Biopolymères Interactions Assemblages, Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement, Nantes, France
| | - Malvyne Rolli-Derkinderen
- The Enteric Nervous System in Gut and Brain Disorders, Institut des Maladies de l’Appareil Digestif, Institut National Pour la Santé et la Recherche Médicale, Nantes Université, Nantes, France,*Correspondence: Malvyne Rolli-Derkinderen,
| |
Collapse
|
13
|
Wang Z, Liu H, Gong Y, Cheng Y. Establishment and validation of an aging-related risk signature associated with prognosis and tumor immune microenvironment in breast cancer. Eur J Med Res 2022; 27:317. [PMID: 36581948 PMCID: PMC9798726 DOI: 10.1186/s40001-022-00924-4] [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: 09/26/2022] [Accepted: 12/01/2022] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Breast cancer (BC) is a highly malignant and heterogeneous tumor which is currently the cancer with the highest incidence and seriously endangers the survival and prognosis of patients. Aging, as a research hotspot in recent years, is widely considered to be involved in the occurrence and development of a variety of tumors. However, the relationship between aging-related genes (ARGs) and BC has not yet been fully elucidated. MATERIALS AND METHODS The expression profiles and clinicopathological data were acquired in the Cancer Genome Atlas (TCGA) and the gene expression omnibus (GEO) database. Firstly, the differentially expressed ARGs in BC and normal breast tissues were investigated. Based on these differential genes, a risk model was constructed composed of 11 ARGs via univariate and multivariate Cox analysis. Subsequently, survival analysis, independent prognostic analysis, time-dependent receiver operating characteristic (ROC) analysis and nomogram were performed to assess its ability to sensitively and specifically predict the survival and prognosis of patients, which was also verified in the validation set. In addition, functional enrichment analysis and immune infiltration analysis were applied to reveal the relationship between the risk scores and tumor immune microenvironment, immune status and immunotherapy. Finally, multiple datasets and real-time polymerase chain reaction (RT-PCR) were utilized to verify the expression level of the key genes. RESULTS An 11-gene signature (including FABP7, IGHD, SPIB, CTSW, IGKC, SEZ6, S100B, CXCL1, IGLV6-57, CPLX2 and CCL19) was established to predict the survival of BC patients, which was validated by the GEO cohort. Based on the risk model, the BC patients were divided into high- and low-risk groups, and the high-risk patients showed worse survival. Stepwise ROC analysis and Cox analyses demonstrated the good performance and independence of the model. Moreover, a nomogram combined with the risk score and clinical parameters was built for prognostic prediction. Functional enrichment analysis revealed the robust relationship between the risk model with immune-related functions and pathways. Subsequent immune microenvironment analysis, immunotherapy, etc., indicated that the immune status of patients in the high-risk group decreased, and the anti-tumor immune function was impaired, which was significantly different with those in the low-risk group. Eventually, the expression level of FABP7, IGHD, SPIB, CTSW, IGKC, SEZ6, S100B, CXCL1, IGLV6-57 and CCL19 was identified as down-regulated in tumor cell line, while CPLX2 up-regulated, which was mostly similar with the results in TCGA and Human Protein Atlas (HPA) via RT-PCR. CONCLUSIONS In summary, our study constructed a risk model composed of ARGs, which could be used as a solid model for predicting the survival and prognosis of BC patients. Moreover, this model also played an important role in tumor immunity, providing a new direction for patient immune status assessment and immunotherapy selection.
Collapse
Affiliation(s)
- Zitao Wang
- grid.412632.00000 0004 1758 2270Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei China
| | - Hua Liu
- grid.412632.00000 0004 1758 2270Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei China
| | - Yiping Gong
- grid.412632.00000 0004 1758 2270Department of Breast Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei China
| | - Yanxiang Cheng
- grid.412632.00000 0004 1758 2270Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei China
| |
Collapse
|
14
|
Deng Q, Yao Y, Yang J, Khoshaba R, Shen Y, Wang X, Cao D. AKR1B8 deficiency drives severe DSS-induced acute colitis through invasion of luminal bacteria and activation of innate immunity. Front Immunol 2022; 13:1042549. [PMID: 36518763 PMCID: PMC9742539 DOI: 10.3389/fimmu.2022.1042549] [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: 09/12/2022] [Accepted: 11/07/2022] [Indexed: 11/29/2022] Open
Abstract
Background Dysfunction of intestinal epithelial cells (IECs) promotes inflammatory bowel disease (IBD) and associated colorectal cancer (CRC). AKR1B8 deficiency impairs the IEC barrier function, leading to susceptibility to chronic colitis induced by dextran sulfate sodium (DSS), yet it remains unclear how acute colitic response is in AKR1B8 deficient mice. Methods AKR1B8 knockout (KO) and littermate wild type mice were exposed to oral 1.5% DSS in drinking water for 6 days. Disease activity index and histopathological inflammation scores by H&E staining were calculated for colitic severity; permeability was assessed by fluorescein isothiocyanate dextran (FITC-Dextran) probes and bacterial invasion and transmission were detected by in situ hybridization in mucosa or by culture in blood agar plates. Immunofluorescent staining and flow cytometry were applied for immune cell quantification. Toll-like receptor 4 (TLR4) and target gene expression was analyzed by Western blotting and qRT-PCR. Results AKR1B8 KO mice developed severe acute colitis at a low dose (1.5%) of DSS in drinking water compared to wild type controls. In AKR1B8 KO mice, FITC-dextran was penetrated easily and luminal bacteria invaded to the surface of IEC layer on day 3, and excessive bacteria translocated into the colonic mucosa, mesenteric lymph nodes (MLNs) and liver on day 6, which was much mild in wild type mice. Hyper-infiltration of neutrophils and basophils occurred in AKR1B8 KO mice, and monocytes in spleen and macrophages in colonic mucosa increased markedly compared to wild type mice. TLR4 signaling in colonic epithelial cells of AKR1B8 KO mice was activated to promote great IL-1β and IL-6 expression compared to wild type mice. Conclusions AKR1B8 deficiency in IECs drives severe acute colitis induced by DSS at a low dose through activation of the innate immunity, being a novel pathogenic factor of colitis.
Collapse
Affiliation(s)
- Qiulin Deng
- Department of Proctology, The Affiliated Nanhua Hospital, University of South China Hengyang Medical School, Hengyang, Hunan, China
| | - Yichen Yao
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Jing Yang
- Department of Gastroenterology, The First Affiliated Hospital, University of South China Hengyang Medical School, Hengyang, Hunan, China
| | - Ramina Khoshaba
- Department of Medical Microbiology, Immunology, and Cell Biology, Southern Illinois University, School of Medicine, Springfield, IL, United States,Department of Biotechnology, College of Science, University of Baghdad, Baghdad, Iraq
| | - Yi Shen
- Department of Medical Microbiology, Immunology, and Cell Biology, Southern Illinois University, School of Medicine, Springfield, IL, United States
| | - Xin Wang
- Department of Medical Microbiology, Immunology, and Cell Biology, Southern Illinois University, School of Medicine, Springfield, IL, United States,Department of Medicine, Harvard Medical School, Boston, MA, United States,Jeff and Penny Vinik Center for Translational Immunology Research, Division of Allergy and Clinical Immunology, Brigham and Women’s Hospital, Boston, MA, United States,*Correspondence: Xin Wang, ; Deliang Cao,
| | - Deliang Cao
- Department of Gastroenterology, The First Affiliated Hospital, University of South China Hengyang Medical School, Hengyang, Hunan, China,Department of Medical Microbiology, Immunology, and Cell Biology, Southern Illinois University, School of Medicine, Springfield, IL, United States,*Correspondence: Xin Wang, ; Deliang Cao,
| |
Collapse
|
15
|
Sun YT, Li GX. Ferroptosis and chronic gastritis and gastric cancer. Shijie Huaren Xiaohua Zazhi 2022; 30:859-864. [DOI: 10.11569/wcjd.v30.i19.859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
As a unique form of cell death in the context of redox, ferroptosis has become a hot research topic in recent years for its potential anti-tumor effect. Digestive system diseases are a common group of human diseases. Increasing studies have addressed the important role of ferroptosis in digestive system diseases. Here, we discuss the mechanism of ferroptosis, and analyze the role of ferroptosis in chronic gastritis and gastric cancer. Targeting ferroptosis may be one of the directions for the treatment of digestive system diseases.
Collapse
Affiliation(s)
- Yi-Tian Sun
- Department of Gastroenterology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou 310015, Zhejiang Province, China
| | - Guo-Xiong Li
- Department of Gastroenterology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou 310015, Zhejiang Province, China
| |
Collapse
|
16
|
Hassan SU, Chua EG, Kaur P, Paz EA, Tay CY, Greeff JC, Liu S, Martin GB. Contribution of the Immune Response in the Ileum to the Development of Diarrhoea caused by Helminth Infection: Studies with the Sheep Model. Funct Integr Genomics 2022; 22:865-877. [PMID: 35576023 PMCID: PMC9550700 DOI: 10.1007/s10142-022-00864-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 04/18/2022] [Accepted: 04/20/2022] [Indexed: 11/04/2022]
Abstract
Gastrointestinal helminths are a global health issue, for humans as well as domestic animals. Most studies focus on the tissues that are infected with the parasite, but here we studied the ileum, a tissue that is rarely infected by helminths. We tested whether inflammation in the ileum contributes to the development and severity of diarrhoea, by comparing sheep that are susceptible (n = 4) or resistant (n = 4) to the disease. We analyzed the ileum transcriptome using RNASeq sequencing approach and various bioinformatics tools including FastQC, STAR, featureCounts, DESeq2, DAVID, clusterProfiler, Cytoscape (ClusterONE) and EnrichR. We identified 243 differentially expressed genes (DEGs), of which 118 were up-regulated and 125 were down-regulated DEGs in the diarrhoea-susceptible animals compared to the diarrhoea-resistant animals. The resulting DEGs were functionally enriched for biological processes, pathways and gene set enrichment analysis. The up-regulated DEGs suggested that an inflammatory immune response was coupled with genes involved in 'Th2 immune response' and 'anti-inflammatory response'. The down-regulated DEGs were related to ion transport, muscle contraction and pathways preventing inflammation. We conclude that i) susceptibility to helminth-induced diarrhoea involves an inflammatory response at a non-infectious site; ii) down-regulation of pathways preventing inflammation can contribute to the severity of diarrhoea; and iii) genes involved in anti-inflammatory responses can reduce the inflammation and diarrhoea.
Collapse
Affiliation(s)
- Shamshad Ul Hassan
- UWA School of Agriculture and Environment, The University of Western Australia, Crawley, WA, 6009, Australia
- Helicobacter Research Laboratory, The Marshall Centre for Infectious Disease Research and Training, School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia
| | - Eng Guan Chua
- Helicobacter Research Laboratory, The Marshall Centre for Infectious Disease Research and Training, School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia
| | - Parwinder Kaur
- UWA School of Agriculture and Environment, The University of Western Australia, Crawley, WA, 6009, Australia
| | - Erwin A Paz
- UWA School of Agriculture and Environment, The University of Western Australia, Crawley, WA, 6009, Australia
- Helicobacter Research Laboratory, The Marshall Centre for Infectious Disease Research and Training, School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia
| | - Chin Yen Tay
- Helicobacter Research Laboratory, The Marshall Centre for Infectious Disease Research and Training, School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia
| | - Johan C Greeff
- UWA School of Agriculture and Environment, The University of Western Australia, Crawley, WA, 6009, Australia
- Department of Primary Industries and Regional Development, Western Australia, 3 Baron Hay Court, South Perth, WA, 6151, Australia
| | - Shimin Liu
- UWA School of Agriculture and Environment, The University of Western Australia, Crawley, WA, 6009, Australia
| | - Graeme B Martin
- UWA School of Agriculture and Environment, The University of Western Australia, Crawley, WA, 6009, Australia.
| |
Collapse
|
17
|
Sandys O, Te Velde A. Raising the Alarm: Environmental Factors in the Onset and Maintenance of Chronic (Low-Grade) Inflammation in the Gastrointestinal Tract. Dig Dis Sci 2022; 67:4355-4368. [PMID: 34981314 DOI: 10.1007/s10620-021-07327-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 11/10/2021] [Indexed: 12/12/2022]
Abstract
Chronic inflammatory disease of the gastrointestinal (GI) tract is defined by several pathophysiological characteristics, such as dysbiosis of the microbiota, epithelial barrier hyperpermeability, systemic dissemination of endotoxins and chronic inflammation. In addition to well-reported environmental factors in non-communicable disease, such as smoking, diet, and exercise, humans are frequently exposed to myriads more environmental factors, from pesticides to food additives. Such factors are ubiquitous across both our diet and indoor/outdoor environments. A major route of human exposure to these factors is ingestion, which frequently occurs due to their intentional addition (intentional food additives) and/or unintentional contamination (unintentional food contaminants) of food products-often linked to environmental pollution. Understanding how this persistent, diverse exposure impacts GI health is of paramount importance, as deterioration of the GI barrier is proposed to be the first step towards systemic inflammation and chronic disease. Therefore, we aim to evaluate the impact of ingestion of environmental factors on inflammatory processes in the GI tract. In this review, we highlight human exposure to intentional food additives (e.g. emulsifiers, bulking agents) and unintentional food contaminants (e.g. persistent organic pollutants, pesticides, microplastics), then present evidence for their association with chronic disease, modification of the GI microbiota, increased permeability of the GI barrier, systemic dissemination of endotoxins, local (and distal) pro-inflammatory signalling, and induction of oxidative stress and/or endoplasmic reticulum stress. We also propose a link to NLRP3-inflammasome activation. These findings highlight the contribution of common environmental factors towards deterioration of GI health and the induction of pathophysiology associated with onset and maintenance of chronic inflammation in the GI tract.
Collapse
Affiliation(s)
- Oliver Sandys
- Amsterdam UMC, University of Amsterdam, Tytgat Institute for Liver and Intestinal Research, AGEM, Amsterdam, The Netherlands
- Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Anje Te Velde
- Amsterdam UMC, University of Amsterdam, Tytgat Institute for Liver and Intestinal Research, AGEM, Amsterdam, The Netherlands.
| |
Collapse
|
18
|
Wang T, Wang S, Dong S, Zhang Y, Ismael M, Wang S, Shi C, Yang J, Wang X, Lü X. Interaction of Companilactobacillus crustorum MN047-derived bacteriocins with gut microbiota. Food Chem 2022; 396:133730. [PMID: 35878442 DOI: 10.1016/j.foodchem.2022.133730] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 06/17/2022] [Accepted: 07/15/2022] [Indexed: 11/04/2022]
Abstract
Companilactobacillus crustorum MN047-derived bacteriocins (CCDB) have inhibitory effects on the growth of pathogens. In this study, a pectin/zein beads delivery system was used to investigate the effects of CCDB on the dextran sulfate sodium-induced colitis in mice. The focus was given on aspects linked with the gut microbiota, intestinal epithelial barrier, oxidative stress, and inflammation. Results suggested that CCDB alleviated the pathological symptoms of colitis, including increased disease activity index and shortened colon length. CCDB strengthened the gut barrier by increasing goblet cells and promoting the expressions of MUC2 and tight junctions-related proteins. CCDB decreased oxidative mediators and increased antioxidant mediators in serum or colon tissue. Furthermore, CCDB reduced harmful bacteria and enriched beneficial bacteria, which further decreased serum LPS and increased fecal butyric acid. In addition, CCDB inhibited the overexpressions of proinflammatory cytokines, chemokines, and pathogens/LPS-activated TLR4/NF-κB pathway. Therefore, CCDB is a potential dietary supplement to relieve colitis.
Collapse
Affiliation(s)
- Tao Wang
- College of Food Science and Engineering, Northwest Agriculture and Forestry University, Yangling, Shaanxi 712100, China
| | - Shuxuan Wang
- College of Food Science and Engineering, Northwest Agriculture and Forestry University, Yangling, Shaanxi 712100, China
| | - Shuchen Dong
- College of Food Science and Engineering, Northwest Agriculture and Forestry University, Yangling, Shaanxi 712100, China
| | - Yu Zhang
- College of Food Science and Engineering, Northwest Agriculture and Forestry University, Yangling, Shaanxi 712100, China
| | - Mohamedelfatieh Ismael
- College of Food Science and Engineering, Northwest Agriculture and Forestry University, Yangling, Shaanxi 712100, China
| | - Shuang Wang
- College of Food Science and Engineering, Northwest Agriculture and Forestry University, Yangling, Shaanxi 712100, China
| | - Caihong Shi
- College of Food Science and Engineering, Northwest Agriculture and Forestry University, Yangling, Shaanxi 712100, China
| | - Jie Yang
- School of Life Science and Technology, Xinjiang University, Urumqi, Xinjiang 830046, China
| | - Xin Wang
- College of Food Science and Engineering, Northwest Agriculture and Forestry University, Yangling, Shaanxi 712100, China.
| | - Xin Lü
- College of Food Science and Engineering, Northwest Agriculture and Forestry University, Yangling, Shaanxi 712100, China.
| |
Collapse
|
19
|
Lartey NL, Vargas-Robles H, Guerrero-Fonseca IM, Nava P, Kumatia EK, Ocloo A, Schnoor M. Annickia polycarpa extract attenuates inflammation, neutrophil recruitment, and colon damage during colitis. Immunol Lett 2022; 248:99-108. [PMID: 35841974 DOI: 10.1016/j.imlet.2022.07.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 06/08/2022] [Accepted: 07/11/2022] [Indexed: 11/09/2022]
Abstract
Inflammatory bowel diseases (IBD) including Crohn's disease (CD) and ulcerative colitis (UC) are complex inflammatory disorders of the digestive tract. Dysfunctional intestinal epithelial barrier, uncontrolled neutrophil recruitment into the colon, and oxidative stress are major features of IBD. IBD cannot be cured, but symptoms can be alleviated with anti-inflammatory drugs, which often show adverse effects. Thus, safer alternative treatment options are needed. Given the known anti-inflammatory properties of Annickia polycarpa extract (APE), we hypothesized that APE improves the outcome of the inflammatory response during colitis. We assessed APE effects on colon histology, epithelial barrier function and neutrophil recruitment during DSS-induced colitis in mice treated with APE. APE treatment significantly reduced the disease activity index and prevented DSS-induced colon damage as evidenced by reduced colon shortening, ulcerations, crypt dysplasia, edema formation, and leukocyte infiltration. Expression of the pro-inflammatory cytokines TNF-α, IL-6, and IL-1β were significantly diminished in APE-treated mice. Importantly, APE administration reduced neutrophil infiltration into the lamina propria leading to reduced oxidative stress, tight junction disruption and epithelial permeability in the colon. Thus, we propose APE as additional treatment strategy to attenuate colitis symptoms and enhance life quality of individuals with IBD.
Collapse
Affiliation(s)
- Nathaniel L Lartey
- Department of Molecular Biomedicine, CINVESTAV-IPN, Avenida IPN 2508, 07360 Mexico-City, Mexico; Department of Health and Allied Sciences, Baldwin University College, Osu-Accra, Ghana
| | - Hilda Vargas-Robles
- Department of Molecular Biomedicine, CINVESTAV-IPN, Avenida IPN 2508, 07360 Mexico-City, Mexico
| | | | - Porfirio Nava
- Department of Physiology, Biophysics and Neurosciences, CINVESTAV-IPN, Avenida IPN 2508, 07360 Mexico-City, Mexico
| | - Emmanuel K Kumatia
- Department of Phytochemistry, Centre for Plant Medicine Research. Akuapem-Mampong, Ghana
| | - Augustine Ocloo
- Department of Biochemistry, Cell, and Molecular Biology, University of Ghana, LG 54, Legon, Ghana
| | - Michael Schnoor
- Department of Molecular Biomedicine, CINVESTAV-IPN, Avenida IPN 2508, 07360 Mexico-City, Mexico.
| |
Collapse
|
20
|
Wang Q, Chen Y, Gao W, Feng H, Zhang B, Wang H, Lu H, Tan Y, Dong Y, Xu M. Identification and Validation of a Four-Gene Ferroptosis Signature for Predicting Overall Survival of Lung Squamous Cell Carcinoma. Front Oncol 2022; 12:933925. [PMID: 35912252 PMCID: PMC9330609 DOI: 10.3389/fonc.2022.933925] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 05/27/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundLung squamous cell carcinoma (LUSC) represents 30% of all non-small cell lung carcinoma. Targeted therapy is not sufficient for LUSC patients because of the low frequency of targeted-effective mutation in LUSC whereas immunotherapy offers more options for patients with LUSC. We explored a ferroptosis-related prognostic signature that can potentially assess the prognosis and immunotherapy efficacy of LUSC patients.MethodsA total of 502 LUSC patients were downloaded from The Cancer Genome Atlas (TCGA). The external validation data were obtained from the Gene Expression Omnibus (GEO): GSE73403. Then, we identified the candidate genes and constructed the prognostic signature through the Cox survival regression analyses and least absolute shrinkage and selection operator (LASSO). Risk score plot, Kaplan–Meier curve, and ROC curve were used to assess the prognostic power and performance of the model. The CIBERSORT algorithm estimated the fraction of immune cell types. TIDE was utilized to predict the response to immunotherapy. IMvigor210 was used to explore the association between the risk scores and immunotherapy outcomes. A nomogram combined selected clinical characteristics, and the risk scores were constructed.ResultsWe screened 132 differentially expressed ferroptosis-related genes. According to KEGG and GO pathway analyses, these genes were mainly engaged in the positive regulation of cytokine production, cytokine metabolic process, and oxidoreductase activity. We then constructed a prognostic model via LASSO regression. The proportions of CD8+ T cells, CD4+ activated T cells, and follicular helper T cells were significantly different between low-risk and high-risk groups. TIDE algorithm indicated that low-risk LUSC patients might profit more from immune checkpoint inhibitors. The predictive value of the ferroptosis gene model in immunotherapy response was further confirmed in IMvigor210. Finally, we combined the clinical characteristics with a LASSO regression model to construct a nomogram that could be easily applied in clinical practice.ConclusionWe identified a prognostic model that provides an accurate and objective basis for guiding individualized treatment decisions for LUSC.
Collapse
Affiliation(s)
- Qi Wang
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
- *Correspondence: Qi Wang,
| | - Yaokun Chen
- Breast Disease Diagnosis and Treatment Center, Qingdao Center Medical Group, Qingdao, China
| | - Wen Gao
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hui Feng
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Biyuan Zhang
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Haiji Wang
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Haijun Lu
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ye Tan
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yinying Dong
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Mingjin Xu
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
21
|
Xu M, Leskinen K, Gritti T, Groma V, Arola J, Lepistö A, Sipponen T, Saavalainen P, Söderlund-Venermo M. Prevalence, Cell Tropism, and Clinical Impact of Human Parvovirus Persistence in Adenomatous, Cancerous, Inflamed, and Healthy Intestinal Mucosa. Front Microbiol 2022; 13:914181. [PMID: 35685923 PMCID: PMC9171052 DOI: 10.3389/fmicb.2022.914181] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 04/25/2022] [Indexed: 11/18/2022] Open
Abstract
Parvoviruses are single-stranded DNA viruses, infecting many animals from insects to humans. Human parvovirus B19 (B19V) causes erythema infectiosum, arthropathy, anemia, and fetal death, and human bocavirus (HBoV) 1 causes respiratory tract infections, while HBoV2-4 are enteric. Parvoviral genomes can persist in diverse non-permissive tissues after acute infection, but the host-cell tropism and the impact of their tissue persistence are poorly studied. We searched for parvoviral DNA in a total of 427 intestinal biopsy specimens, as paired disease-affected and healthy mucosa, obtained from 130 patients with malignancy, ulcerative colitis (UC), or adenomas, and in similar intestinal segments from 55 healthy subjects. Only three (1.6%) individuals exhibited intestinal HBoV DNA (one each of HBoV1, 2, and 3). Conversely, B19V DNA persisted frequently in the intestine, with 50, 47, 31, and 27% detection rates in the patients with malignancy, UC, or adenomas, and in the healthy subjects, respectively. Intra-individually, B19V DNA persisted significantly more often in the healthy intestinal segments than in the inflamed colons of UC patients. The highest loads of B19V DNA were seen in the ileum and colon specimens of two healthy individuals. With dual-RNAscope in situ hybridization and immunohistochemistry assays, we located the B19V persistence sites of these intestines in mucosal B cells of lymphoid follicles and vascular endothelial cells. Viral messenger RNA transcription remained, however, undetected. RNA sequencing (RNA-seq) identified 272 differentially expressed cellular genes between B19V DNA-positive and -negative healthy ileum biopsy specimens. Pathway enrichment analysis revealed that B19V persistence activated the intestinal cell viability and inhibited apoptosis. Lifelong B19V DNA persistence thus modulates host gene expression, which may lead to clinical outcomes.
Collapse
Affiliation(s)
- Man Xu
- Department of Virology, University of Helsinki, Helsinki, Finland
| | - Katarzyna Leskinen
- Research Programs Unit, Department of Immunobiology, University of Helsinki, Helsinki, Finland
| | - Tommaso Gritti
- Department of Virology, University of Helsinki, Helsinki, Finland
| | - Valerija Groma
- Joint Laboratory of Electron Microscopy, Riga Stradin,s University, Riga, Latvia
| | - Johanna Arola
- Department of Pathology, University of Helsinki, Helsinki, Finland
- HUS Diagnostic Centre, Helsinki University Hospital, Helsinki, Finland
| | - Anna Lepistö
- Department of Colorectal Surgery, Helsinki University Hospital, Helsinki, Finland
- Applied Tumor Genomics, Research Programs Unit, University of Helsinki, Helsinki, Finland
| | - Taina Sipponen
- HUCH Abdominal Center, Helsinki University Hospital, Helsinki, Finland
| | - Päivi Saavalainen
- Research Programs Unit, Department of Immunobiology, University of Helsinki, Helsinki, Finland
- Folkhälsan Research Center, Helsinki, Finland
| | - Maria Söderlund-Venermo
- Department of Virology, University of Helsinki, Helsinki, Finland
- *Correspondence: Maria Söderlund-Venermo,
| |
Collapse
|
22
|
Aljabban J, Rohr M, Borkowski VJ, Nemer M, Cohen E, Hashi N, Aljabban H, Boateng E, Syed S, Mohammed M, Mukhtar A, Hadley D, Panahiazar M. Probing predilection to Crohn's disease and Crohn's disease flares: A crowd-sourced bioinformatics approach. J Pathol Inform 2022; 13:100094. [PMID: 36268056 PMCID: PMC9576970 DOI: 10.1016/j.jpi.2022.100094] [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] [Indexed: 12/15/2022] Open
Abstract
Background Crohn's Disease (CD) is an inflammatory disease of the gastrointestinal tract that affects millions of patients. While great strides have been made in treatment, namely in biologic therapy such as anti-TNF drugs, CD remains a significant health burden. Method We conducted two meta-analyses using our STARGEO platform to tag samples from Gene Expression Omnibus. One analysis compares inactive colonic biopsies from CD patients to colonic biopsies from healthy patients as a control and the other compares colonic biopsies from active CD lesions to inactive lesions. Separate tags were created to tag colonic samples from inflamed biopsies (total of 65 samples) and quiescent tissue in CD patients (total of 39 samples), and healthy tissue from non-CD patients (total of 30 samples). Results from the two meta-analyses were analyzed using Ingenuity Pathway Analysis. Results For the inactive CD vs healthy tissue analysis, we noted FXR/RXR and LXR/RXR activation, superpathway of citrulline metabolism, and atherosclerosis signaling as top canonical pathways. The top upstream regulators include genes implicated in innate immunity, such as TLR3 and HNRNPA2B1, and sterol regulation through SREBF2. In addition, the sterol regulator SREBF2, lipid metabolism was the top disease network identified in IPA (Fig. 1). Top upregulated genes hold implications in innate immunity (DUOX2, REG1A/1B/3A) and cellular transport and absorption (ABCG5, NPC1L1, FOLH1, and SLC6A14). Top downregulated genes largely held roles in cell adhesion and integrity, including claudin 8, PAQR5, and PRKACB.For the active vs inactive CD analysis, we found immune cell adhesion and diapedesis, hepatic fibrosis/hepatic stellate cell activation, LPS/IL-1 inhibition of RXR function, and atherosclerosis as top canonical pathways. Top upstream regulators included inflammatory mediators LPS, TNF, IL1B, and TGFB1. Top upregulated genes function in the immune response such as IL6, CXCL1, CXCR2, MMP1/7/12, and PTGS2. Downregulated genes dealt with cellular metabolism and transport such as CPO, RBP2, G6PC, PCK1, GSTA1, and MEP1B. Conclusion Our results build off established and recently described research in the field of CD. We demonstrate the use of our user-friendly platform, STARGEO, in investigating disease and finding therapeutic avenues.
Collapse
Affiliation(s)
- Jihad Aljabban
- University of Wisconsin Hospitals and Clinics, Madison, WI, United States,Corresponding author.
| | - Michael Rohr
- University of Central Florida College of Medicine, Orlando, FL, United States
| | | | - Mary Nemer
- University of Wisconsin Hospitals and Clinics, Madison, WI, United States
| | - Eli Cohen
- Vanderbilt University Medical Center, Nashville, TN, United States
| | - Naima Hashi
- Mayo Clinic Minnesota, Rochester, MN, United States
| | | | - Emmanuel Boateng
- Vanderbilt University Medical Center, Nashville, TN, United States
| | - Saad Syed
- Northwestern Memorial Hospital, Chicago, IL, United States
| | | | - Ali Mukhtar
- Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States
| | - Dexter Hadley
- University of Central Florida College of Medicine, Orlando, FL, United States
| | - Maryam Panahiazar
- University of California San Francisco, San Francisco, CA, United States
| |
Collapse
|
23
|
Ghalandary M, Li Y, Fröhlich T, Magg T, Liu Y, Rohlfs M, Hollizeck S, Conca R, Schwerd T, Uhlig HH, Bufler P, Koletzko S, Muise AM, Snapper SB, Hauck F, Klein C, Kotlarz D. Valosin-containing protein-regulated endoplasmic reticulum stress causes NOD2-dependent inflammatory responses. Sci Rep 2022; 12:3906. [PMID: 35273242 PMCID: PMC8913691 DOI: 10.1038/s41598-022-07804-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 02/24/2022] [Indexed: 11/24/2022] Open
Abstract
NOD2 polymorphisms may affect sensing of the bacterial muramyl dipeptide (MDP) and trigger perturbed inflammatory responses. Genetic screening of a patient with immunodeficiency and enteropathy revealed a rare homozygous missense mutation in the first CARD domain of NOD2 (ENST00000300589; c.160G > A, p.E54K). Biochemical assays confirmed impaired NOD2-dependent signaling and proinflammatory cytokine production in patient's cells and heterologous cellular models with overexpression of the NOD2 mutant. Immunoprecipitation-coupled mass spectrometry unveiled the ATPase valosin-containing protein (VCP) as novel interaction partner of wildtype NOD2, while the binding to the NOD2 variant p.E54K was abrogated. Knockdown of VCP in coloncarcinoma cells led to impaired NF-κB activity and IL8 expression upon MDP stimulation. In contrast, tunicamycin-induced ER stress resulted in increased IL8, CXCL1, and CXCL2 production in cells with knockdown of VCP, while enhanced expression of these proinflammatory molecules was abolished upon knockout of NOD2. Taken together, these data suggest that VCP-mediated inflammatory responses upon ER stress are NOD2-dependent.
Collapse
Affiliation(s)
- Maryam Ghalandary
- Dr. von Hauner Children's Hospital, Department of Pediatrics, University Hospital, Ludwig-Maximilians-Universität (LMU) Munich, 80337, Munich, Germany
| | - Yue Li
- Dr. von Hauner Children's Hospital, Department of Pediatrics, University Hospital, Ludwig-Maximilians-Universität (LMU) Munich, 80337, Munich, Germany
| | - Thomas Fröhlich
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, LMU Munich, Munich, Germany
| | - Thomas Magg
- Dr. von Hauner Children's Hospital, Department of Pediatrics, University Hospital, Ludwig-Maximilians-Universität (LMU) Munich, 80337, Munich, Germany
| | - Yanshan Liu
- Dr. von Hauner Children's Hospital, Department of Pediatrics, University Hospital, Ludwig-Maximilians-Universität (LMU) Munich, 80337, Munich, Germany
| | - Meino Rohlfs
- Dr. von Hauner Children's Hospital, Department of Pediatrics, University Hospital, Ludwig-Maximilians-Universität (LMU) Munich, 80337, Munich, Germany
| | - Sebastian Hollizeck
- Dr. von Hauner Children's Hospital, Department of Pediatrics, University Hospital, Ludwig-Maximilians-Universität (LMU) Munich, 80337, Munich, Germany
| | - Raffaele Conca
- Dr. von Hauner Children's Hospital, Department of Pediatrics, University Hospital, Ludwig-Maximilians-Universität (LMU) Munich, 80337, Munich, Germany
| | - Tobias Schwerd
- Dr. von Hauner Children's Hospital, Department of Pediatrics, University Hospital, Ludwig-Maximilians-Universität (LMU) Munich, 80337, Munich, Germany
| | - Holm H Uhlig
- Translational Gastroenterology Unit and Department of Pediatrics, and Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Philip Bufler
- Dr. von Hauner Children's Hospital, Department of Pediatrics, University Hospital, Ludwig-Maximilians-Universität (LMU) Munich, 80337, Munich, Germany
- Department of Pediatric Gastroenterology, Nephrology and Metabolic Diseases, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, Berlin, Germany
| | - Sibylle Koletzko
- Dr. von Hauner Children's Hospital, Department of Pediatrics, University Hospital, Ludwig-Maximilians-Universität (LMU) Munich, 80337, Munich, Germany
- Department of Pediatrics, School of Medicine Collegium, Medicum University of Warmia and Mazury, Olsztyn, Poland
| | - Aleixo M Muise
- SickKids Inflammatory Bowel Disease Center, Research Institute, Hospital for Sick Children, Toronto, ON, M5G1X8, Canada
- Cell Biology Program, Research Institute, Hospital for Sick Children, Toronto, ON, M5G1X8, Canada
- VEO-IBD Consortium, University Hospital, LMU Munich, 80337, Munich, Germany
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Hospital for Sick Children, University of Toronto, Toronto, ON, M5G1X8, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON, M5G1A8, Canada
| | - Scott B Snapper
- VEO-IBD Consortium, University Hospital, LMU Munich, 80337, Munich, Germany
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
- Division of Gastroenterology, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Fabian Hauck
- Dr. von Hauner Children's Hospital, Department of Pediatrics, University Hospital, Ludwig-Maximilians-Universität (LMU) Munich, 80337, Munich, Germany
| | - Christoph Klein
- Dr. von Hauner Children's Hospital, Department of Pediatrics, University Hospital, Ludwig-Maximilians-Universität (LMU) Munich, 80337, Munich, Germany
- VEO-IBD Consortium, University Hospital, LMU Munich, 80337, Munich, Germany
- Gene Center, LMU Munich, Munich, Germany
- Deutsche Zentrum für Infektionsforschung (DZIF), Inhoffenstraße 7, 38124 Braunschweig, Germany
| | - Daniel Kotlarz
- Dr. von Hauner Children's Hospital, Department of Pediatrics, University Hospital, Ludwig-Maximilians-Universität (LMU) Munich, 80337, Munich, Germany.
- VEO-IBD Consortium, University Hospital, LMU Munich, 80337, Munich, Germany.
| |
Collapse
|
24
|
Yin H, Miao Z, Wang L, Su B, Liu C, Jin Y, Wu B, Han H, Yuan X. Fusobacterium nucleatum promotes liver metastasis in colorectal cancer by regulating the hepatic immune niche and altering gut microbiota. Aging (Albany NY) 2022; 14:1941-1958. [PMID: 35212644 PMCID: PMC8908934 DOI: 10.18632/aging.203914] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 12/11/2021] [Indexed: 11/25/2022]
Abstract
Liver metastasis is the major cause of death in colorectal cancer (CRC) patients. Nevertheless, the underlying mechanisms remain unknown. Gut microbiota intricately affect the initiation and progression of CRC by instigating immune response through the secretion of pro-inflammatory cytokines. In this study, we investigated the contribution of Fusobacterium nucleatum (F.nucleatum) to the microbiota-liver axis of CRC in mice, focusing on the correlation between liver immunity and gut microbiota alterations. When F. nucleatum was orally administered to mice, CRC liver metastasis was evidently exaggerated and accompanied by noticeable deleterious effects on body weight, cecum weight, and overall survival time. Further evaluation of the immune response and cytokine profiles revealed a substantial increase in the levels of pro-inflammatory cytokines such as IL6, IL12, IL9, IL17A, CXCL1, MCP-1, TNF-α, and IFN-γ in the plasma of mice treated with F. nucleatum as compared to that in the untreated control mice. Besides, hepatic immune response was also modulated by recruitment of myeloid-derived suppressor cells, reduction in the infiltration of natural killer (NK) and T helper-17 (Th17) cells, as well as increase in regulatory T cell accumulation in the liver. Additionally, sustained F. nucleatum exposure abridged the murine gut microbiota diversity, inducing an imbalanced and restructured intestinal microflora. In particular, the abundance of CRC-promoting bacteria such as Enterococcus and Escherichia/Shigella was evidently elevated post F. nucleatum treatment. Thus, our findings suggest that F. nucleatum might be an important factor involved in promoting CRC liver metastasis by triggering of liver immunity through the regulation of gut microbiota structure and composition.
Collapse
Affiliation(s)
- Han Yin
- Department of Oncology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Zhuangzhuang Miao
- Department of Neurosurgery, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Lu Wang
- Department of Oncology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Beibei Su
- Department of Oncology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Chaofan Liu
- Department of Oncology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Jin
- Department of Oncology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Bili Wu
- Department of Oncology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Hu Han
- Department of Oncology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Xianglin Yuan
- Department of Oncology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
25
|
Wang T, Shi C, Wang S, Zhang Y, Wang S, Ismael M, Zhang J, Wang X, Lü X. Protective Effects of Companilactobacillus crustorum MN047 against Dextran Sulfate Sodium-Induced Ulcerative Colitis: A Fecal Microbiota Transplantation Study. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:1547-1561. [PMID: 35077172 DOI: 10.1021/acs.jafc.1c07316] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Gut microbiota dysbiosis could aggravate the development of ulcerative colitis (UC). Companilactobacillus crustorum MN047 (CCMN) is a potential gut microbiota-regulating probiotic that could produce multiple novel bacteriocins. In this study, fecal microbiota transplantation (FMT) was used to verify whether CCMN could alleviate dextran sulfate sodium-induced UC by regulating gut microbiota. Results showed that both CCMN and FMT ameliorated the symptoms of UC, including attenuating the increased disease activity index, shortened colon length, gut barrier damage, and inflammation. Briefly, CCMN and FMT upregulated the expressions of MUCs and tight junctions, downregulated the expressions of proinflammatory cytokines and chemokines, increased fecal short-chain fatty acids, and lowered serum lipopolysaccharides, which were associated with the regulation of gut microbiota (e.g., increased Akkermansia, Blautia, and Ruminococcus levels). These results demonstrated that CCMN could ameliorate UC by modulating gut microbiota and inhibiting the TLR4/NF-κB pathway. Therefore, CCMN could be considered as a potential probiotic supplement for ameliorating UC.
Collapse
Affiliation(s)
- Tao Wang
- College of Food Science and Engineering, Northwest Agriculture and Forestry University, Yangling, Shaanxi 712100, China
| | - Caihong Shi
- College of Food Science and Engineering, Northwest Agriculture and Forestry University, Yangling, Shaanxi 712100, China
| | - Shuxuan Wang
- College of Food Science and Engineering, Northwest Agriculture and Forestry University, Yangling, Shaanxi 712100, China
| | - Yu Zhang
- College of Food Science and Engineering, Northwest Agriculture and Forestry University, Yangling, Shaanxi 712100, China
| | - Shuang Wang
- College of Food Science and Engineering, Northwest Agriculture and Forestry University, Yangling, Shaanxi 712100, China
| | - Mohamedelfaieh Ismael
- College of Food Science and Engineering, Northwest Agriculture and Forestry University, Yangling, Shaanxi 712100, China
| | - Jing Zhang
- College of Food Science and Engineering, Northwest Agriculture and Forestry University, Yangling, Shaanxi 712100, China
| | - Xin Wang
- College of Food Science and Engineering, Northwest Agriculture and Forestry University, Yangling, Shaanxi 712100, China
| | - Xin Lü
- College of Food Science and Engineering, Northwest Agriculture and Forestry University, Yangling, Shaanxi 712100, China
| |
Collapse
|
26
|
Arifa RDN, Brito CB, de Paula TP, Lima RL, Menezes‐Garcia Z, Cassini‐Vieira P, Vilas Boas FA, Queiroz‐Junior CM, da Silva JM, da Silva TA, Barcelos LS, Fagundes CT, Teixeira MM, Souza DG. Eosinophil plays a crucial role in intestinal mucositis induced by antineoplastic chemotherapy. Immunology 2021; 165:355-368. [DOI: 10.1111/imm.13442] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 12/01/2021] [Accepted: 12/15/2021] [Indexed: 12/01/2022] Open
Affiliation(s)
- Raquel D N Arifa
- Laboratory of Microorganism‐Host Interaction Department of Microbiology
| | - Camila B Brito
- Laboratory of Microorganism‐Host Interaction Department of Microbiology
| | - Talles P de Paula
- Laboratory of Microorganism‐Host Interaction Department of Microbiology
| | - Renata L Lima
- Laboratory of Microorganism‐Host Interaction Department of Microbiology
| | | | | | | | - Celso M Queiroz‐Junior
- Department of Oral Pathology and Surgery Faculty of Dentistry Universidade Federal de Minas Gerais Belo Horizonte, Minas Gerais Brazil
| | - Janine M da Silva
- Department of Oral Pathology and Surgery Faculty of Dentistry Universidade Federal de Minas Gerais Belo Horizonte, Minas Gerais Brazil
| | - Tarcília A da Silva
- Department of Oral Pathology and Surgery Faculty of Dentistry Universidade Federal de Minas Gerais Belo Horizonte, Minas Gerais Brazil
| | | | - Caio T. Fagundes
- Laboratory of Microorganism‐Host Interaction Department of Microbiology
- Center for Drug Research and Development of Pharmaceuticals
| | - Mauro M Teixeira
- Center for Drug Research and Development of Pharmaceuticals
- Department of Biochemistry and Immunology Institute of Biological Sciences
| | - Daniele G. Souza
- Laboratory of Microorganism‐Host Interaction Department of Microbiology
| |
Collapse
|
27
|
Wang T, Wang P, Ge W, Shi C, Xiao G, Wang X, Lü X. Protective effect of a multi-strain probiotics mixture on azoxymethane/dextran sulfate sodium-induced colon carcinogenesis. FOOD BIOSCI 2021. [DOI: 10.1016/j.fbio.2021.101346] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
28
|
Wang T, Wang P, Ge W, Shi C, Xiao G, Wang X, Lü X. The probiotic Companilactobacillus crustorum MN047 alleviates colitis-associated tumorigenesis via modulating the intestinal microenvironment. Food Funct 2021; 12:11331-11342. [PMID: 34668003 DOI: 10.1039/d1fo01531a] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Supplementation of probiotics is a promising method to alleviate colorectal cancer (CRC) via modulating the intestinal microenvironment. This study aimed to assess the potential anti-CRC effect of Companilactobacillus crustorum MN047 on an azoxymethane and dextran sulfate sodium-induced colitis-associated (CA)-CRC mouse model. Mice were gavaged with C. crustorum MN047 once daily (∼1 × 109 CFU per mouse). The CA-CRC ameliorating effect of this strain was investigated based on the gut microbiota, inflammation and intestinal barrier integrity. Results showed that C. crustorum MN047 could significantly attenuate tumorigenesis and inflammation via suppressing the TLR4/NF-κB pathway. Moreover, this probiotic could improve the intestinal barrier integrity by increasing the mRNA level of some tight junction-related proteins and reducing goblet cell loss. In addition, C. crustorum MN047 administration led to an increase in beneficial bacteria and a decrease in harmful bacteria, thereby increasing SCFAs and reducing LPS levels. These results suggested that C. crustorum MN047 could partially ameliorate the formation of CA-CRC by modulating the gut microbiota, attenuating inflammation and enhancing the intestinal barrier integrity. Therefore, C. crustorum MN047 was a promising probiotic supplement for attenuating CA-CRC.
Collapse
Affiliation(s)
- Tao Wang
- College of Food Science and Engineering, Northwest Agriculture and Forestry University, Yangling, Shaanxi 712100, China.
| | - Panpan Wang
- College of Food Science and Engineering, Northwest Agriculture and Forestry University, Yangling, Shaanxi 712100, China.
| | - Wupeng Ge
- College of Food Science and Engineering, Northwest Agriculture and Forestry University, Yangling, Shaanxi 712100, China.
| | - Chao Shi
- College of Food Science and Engineering, Northwest Agriculture and Forestry University, Yangling, Shaanxi 712100, China.
| | - Gongnian Xiao
- Zhejiang Provincial Key Lab for Biological and Chemical Processing Technologies of Farm Products, School of Biological and Chemical Engineering, Zhejiang University of Science and Technology, Hangzhou, Zhejiang 310023, China
| | - Xin Wang
- College of Food Science and Engineering, Northwest Agriculture and Forestry University, Yangling, Shaanxi 712100, China.
| | - Xin Lü
- College of Food Science and Engineering, Northwest Agriculture and Forestry University, Yangling, Shaanxi 712100, China.
| |
Collapse
|
29
|
Rehman S, Gora AH, Siriyappagouder P, Brugman S, Fernandes JMO, Dias J, Kiron V. Zebrafish intestinal transcriptome highlights subdued inflammatory responses to dietary soya bean and efficacy of yeast β-glucan. JOURNAL OF FISH DISEASES 2021; 44:1619-1637. [PMID: 34237181 DOI: 10.1111/jfd.13484] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 06/11/2021] [Accepted: 06/14/2021] [Indexed: 06/13/2023]
Abstract
Anti-nutritional factors in dietary components can have a negative impact on the intestinal barrier. Here, we present soya bean-induced changes in the intestine of juvenile zebrafish and the effect of yeast β-glucan through a transcriptomic approach. The inclusion of soya bean meal affected the expression of several intestinal barrier function-related genes like arl4ca, rab25b, rhoub, muc5ac, muc5d, clcn2c and cltb in zebrafish. Several metabolic genes like cyp2x10.2, cyp2aa2, aldh3a2b, crata, elovl4, elovl6, slc51a, gpat2 and ATP-dependent peptidase activity (lonrf, clpxb) were altered in the intestinal tissue. The expression of immune-related genes like nlrc3, nlrp12, gimap8, prdm1 and tph1a, and genes related to cell cycle, DNA damage and DNA repair (e.g. spo11, rad21l1, nabp1b, spata22, tdrd9) were also affected in the soya bean fed group. Furthermore, our study suggests the plausible effect of yeast β-glucan through the modulation of several genes that regulate immune responses and barrier integrity. Our findings indicate a subdued inflammation in juvenile zebrafish fed soya bean meal and the efficacy of β-glucan to counter these subtle inflammatory responses.
Collapse
Affiliation(s)
- Saima Rehman
- Faculty of Biosciences and Aquaculture, Nord University, Bodø, Norway
| | - Adnan H Gora
- Faculty of Biosciences and Aquaculture, Nord University, Bodø, Norway
| | | | - Sylvia Brugman
- Department of Animal Sciences, Wageningen University, Wageningen, The Netherlands
| | | | | | - Viswanath Kiron
- Faculty of Biosciences and Aquaculture, Nord University, Bodø, Norway
| |
Collapse
|
30
|
Chan BD, Wong WY, Lee MML, Leung TW, Shum TY, Cho WCS, Chen S, Tai WCS. Centipeda minima Extract Attenuates Dextran Sodium Sulfate-Induced Acute Colitis in Mice by Inhibiting Macrophage Activation and Monocyte Chemotaxis. Front Pharmacol 2021; 12:738139. [PMID: 34616300 PMCID: PMC8489405 DOI: 10.3389/fphar.2021.738139] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 09/07/2021] [Indexed: 01/18/2023] Open
Abstract
Inflammatory bowel disease (IBD) is an idiopathic inflammatory disease affecting the gastrointestinal tract. IBD is characterized by courses of relapse and remission, and remains incurable. Although multiple factors are related to the pathogenesis of IBD, disruption of intestinal mucosa homeostasis has been proposed to be a major contributor to IBD, and abnormal activation of immune cells is key for initiation of the inflammatory response. Macrophages are the most abundant immune cells in the intestine. Once activated, they are responsible for secretion of pro-inflammatory cytokines and chemokines to attract circulating monocytes to inflammatory sites, exacerbating the inflammatory response, and leading to tissue damage. Therefore, the suppression of activated macrophages, cytokine/chemokine production, and subsequent monocyte chemotaxis possesses great potential for the treatment of IBD. In our study, we have demonstrated the inhibitory effect of Centipeda minima total extract (CME) on the activation of NF-κB, STAT3, and MAPK signaling in LPS-stimulated RAW264.7 macrophages. In addition, we identified the significant suppressive effect of CME on CCL8 expression in activated macrophages, which potentially contributed to inhibition of monocyte chemotaxis. In the DSS-induced acute colitis mouse model, we have demonstrated the suppressive effect of CME on intestinal macrophage infiltration and its ameliorative effect in IBD. Altogether, we have provided evidence of the therapeutic effect of CME in IBD and the potential of CME for the treatment of IBD.
Collapse
Affiliation(s)
- Brandon Dow Chan
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, SAR China
| | - Wing-Yan Wong
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, SAR China
| | - Magnolia Muk-Lan Lee
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, SAR China
| | - Tsz-Wing Leung
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, SAR China
| | - Tan-Yu Shum
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, SAR China
| | - William Chi-Shing Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Kowloon, Hong Kong, SAR China
| | - Sibao Chen
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, SAR China
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China
| | - William Chi-Shing Tai
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, SAR China
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China
| |
Collapse
|
31
|
Qin YF, Li GM, Wang G, Kong DJ, Wang HD, Zhao YM, Hao JP, Qin H, Sun DQ, Wang H. Identification of Hub Gene TIMP1 and Relative ceRNAs Regulatory Network in Colorectal Cancer. Ther Clin Risk Manag 2021; 17:889-901. [PMID: 34475758 PMCID: PMC8407779 DOI: 10.2147/tcrm.s321101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 08/16/2021] [Indexed: 11/23/2022] Open
Abstract
Objective This study aimed to discover the ceRNAs network in the pathophysiological development of human colorectal cancer (CRC) and to screen biomarkers for target therapy and prognosis by using integrated bioinformatics analysis. Methods Data on gene expressions of mRNAs, miRNAs, and circRNAs and clinical information were downloaded from The Cancer Genome Atlas and Gene Expression Omnibus databases, respectively. Differentially expressed mRNAs (DEmRNAs) were identified by using the DESeq2 package of R software. Functional enrichment analysis was conducted using the ClusterProfiler package of R software. The protein–protein interaction (PPI) network was shown by the STRING website. Survival analysis of hub genes was performed using the survival package in R software. Interactions among hub genes, differentially expressed miRNAs (DEmiRNAs), and differentially expressed circRNAs (DEcircRNAs) were used to construct the ceRNAs network. Results A total of 412 DEmRNAs including 82 upregulated and 330 downregulated genes were screened out between 473 CRC and 41 normal samples. Two hundred and sixty DEcircRNAs including 253 upregulated and 7 downregulated genes were altered between 23 CRC and 23 normal samples. One hundred and ninety DEmiRNAs including 82 upregulated and 108 downregulated genes were obtained between 450 CRC and 8 normal samples. A ceRNAs and PPI network were successfully constructed, and TIMP1 associated with prognosis was employed. Conclusion The present study identified a novel circRNAs-miRNAs-mRNA ceRNAs network, which implied that TIMP1 and related miRNAs, circRNAs were potential biomarkers underlying the development of CRC, providing new insights for survival predictions and therapeutic targets.
Collapse
Affiliation(s)
- Ya-Fei Qin
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, People's Republic of China.,Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
| | - Guang-Ming Li
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, People's Republic of China.,Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
| | - Grace Wang
- Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - De-Jun Kong
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, People's Republic of China.,Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
| | - Hong-Da Wang
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, People's Republic of China.,Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
| | - Yi-Ming Zhao
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, People's Republic of China.,Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
| | - Jing-Peng Hao
- Department of Anorectal Surgery, The Second Hospital of Tianjin Medical University, Tianjin, People's Republic of China
| | - Hong Qin
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, People's Republic of China.,Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
| | - Da-Qing Sun
- Department of Pediatric Surgery, Tianjin Medical University, Tianjin, People's Republic of China
| | - Hao Wang
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, People's Republic of China.,Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
| |
Collapse
|
32
|
Kim JT, Napier DL, Kim J, Li C, Lee EY, Weiss HL, Wang Q, Evers BM. Ketogenesis alleviates TNFα-induced apoptosis and inflammatory responses in intestinal cells. Free Radic Biol Med 2021; 172:90-100. [PMID: 34087430 PMCID: PMC8355065 DOI: 10.1016/j.freeradbiomed.2021.05.032] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 05/21/2021] [Accepted: 05/22/2021] [Indexed: 12/20/2022]
Abstract
The disturbance of strictly regulated self-regeneration in mammalian intestinal epithelium is associated with various intestinal disorders, particularly inflammatory bowel diseases (IBDs). TNFα, which plays a critical role in the pathogenesis of IBDs, has been reported to inhibit production of ketone bodies such as β-hydroxybutyrate (βHB). However, the role of ketogenesis in the TNFα-mediated pathological process is not entirely known. Here, we showed the regulation and role of HMGCS2, the rate-limiting enzyme of ketogenesis, in TNFα-induced apoptotic and inflammatory responses in intestinal epithelial cells. Treatment with TNFα dose-dependently decreased protein and mRNA expression of HMGCS2 and its product, βHB production in human colon cancer cell lines HT29 and Caco2 cells and mouse small intestinal organoids. Moreover, the repressed level of HMGCS2 protein was found in intestinal epithelium of IBD patients with Crohn's disease and ulcerative colitis as compared with normal tissues. Furthermore, knockdown of HMGCS2 enhanced and in contrast, HMGCS2 overexpression attenuated, the TNFα-induced apoptosis and expression of pro-inflammatory chemokines (CXCL1-3) in HT29, Caco2 cells and DLD1 cells, respectively. Treatment with βHB or rosiglitazone, an agonist of PPARγ, which increases ketogenesis, attenuated TNFα-induced apoptosis in the intestinal epithelial cells. Finally, HMGCS2 knockdown enhanced TNFα-induced reactive oxygen species (ROS) generation. In addition, hydrogen peroxide, the major ROS contributing to intestine injury, decreased HMGCS2 expression and βHB production in the intestinal cells and mouse organoids. Our findings demonstrate that increased ketogenesis attenuates TNFα-induced apoptosis and inflammation in intestinal cells, suggesting a protective role for ketogenesis in TNFα-induced intestinal pathologies.
Collapse
Affiliation(s)
- Ji Tae Kim
- Markey Cancer Center, Lexington, KY, 40536, USA
| | | | - Jinhwan Kim
- Markey Cancer Center, Lexington, KY, 40536, USA
| | - Chang Li
- Markey Cancer Center, Lexington, KY, 40536, USA
| | - Eun Y Lee
- Department of Pathology and Laboratory Medicine, Department of Surgery, Lexington, KY, 40536, USA
| | | | - Qingding Wang
- Markey Cancer Center, Lexington, KY, 40536, USA; Department of Surgery, University of Kentucky, Lexington, KY, 40536, USA.
| | - B Mark Evers
- Markey Cancer Center, Lexington, KY, 40536, USA; Department of Surgery, University of Kentucky, Lexington, KY, 40536, USA.
| |
Collapse
|
33
|
Wang T, Zhang L, Wang P, Liu Y, Wang G, Shan Y, Yi Y, Zhou Y, Liu B, Wang X, Lü X. Lactobacillus coryniformis MXJ32 administration ameliorates azoxymethane/dextran sulfate sodium-induced colitis-associated colorectal cancer via reshaping intestinal microenvironment and alleviating inflammatory response. Eur J Nutr 2021; 61:85-99. [PMID: 34185157 DOI: 10.1007/s00394-021-02627-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 06/24/2021] [Indexed: 12/24/2022]
Abstract
PURPOSE Gut microbiota has been reported to contribute to either prevent or promote colorectal cancer (CRC), and treatment with probiotics might be a promising intervention method. The present study aimed to evaluate the potential anti-CRC effects of Lactobacillus coryniformis MXJ32 on a colitis-associated (CA)-CRC mouse model. METHODS The CA-CRC mouse model was induced by a single intraperitoneal injection of 10 mg/kg azoxymethane and followed by three 7-day cycles of 2% dextran sulfate sodium in drinking water with a 14-day recovery period. Mice were supplemented with L. coryniformis MXJ32 by oral gavage (1 × 109 CFU/day/mouse). The CA-CRC attenuating effects of this probiotic were assessed via intestinal barrier integrity, inflammation, and gut microenvironment. RESULTS Treatment with L. coryniformis MXJ32 could significantly inhibit the total number of tumors and the average tumor diameter. This probiotic administration prevented the damage of intestinal barrier function by enhancing the expression of tight junction proteins (Occludin, Claudin-1, and ZO-1) and recovering the loss of goblet cells. Moreover, L. coryniformis MXJ32 alleviated intestinal inflammation via down-regulating the expression of inflammatory cytokines (TNF-α, IL-1β, IL-6, IL-γ, and IL-17a) and chemokines (Cxcl1, Cxcl2, Cxcl3, Cxcl5, and Ccl7). In addition, L. coryniformis MXJ32 supplementation increased the abundance of some beneficial bacteria (such as SCFAs-producing bacteria, Lactobacillus, Bifidobacterium, Akkermansia, and Faecalibaculum) and decreased the abundance of some harmful bacteria (such as pro-inflammatory bacteria, Desulfovibrio and Helicobacter), which in turn attenuated the overexpression of inflammation. CONCLUSION Lactobacillus coryniformis MXJ32 could effectively ameliorate CA-CRC via regulating intestinal microenvironment, alleviating inflammation, and intestinal barrier damage, which further suggested that L. coryniformis MXJ32 could be considered as a functional food ingredient for the alleviation of CA-CRC.
Collapse
Affiliation(s)
- Tao Wang
- College of Food Science and Engineering, Northwest Agriculture and Forestry University, No. 22 Xinong Road, Yangling District, Xianyang, 712100, Shaanxi, China
| | - Leshan Zhang
- College of Food Science and Engineering, Northwest Agriculture and Forestry University, No. 22 Xinong Road, Yangling District, Xianyang, 712100, Shaanxi, China
| | - Panpan Wang
- College of Food Science and Engineering, Northwest Agriculture and Forestry University, No. 22 Xinong Road, Yangling District, Xianyang, 712100, Shaanxi, China
| | - Yilin Liu
- College of Food Science and Engineering, Northwest Agriculture and Forestry University, No. 22 Xinong Road, Yangling District, Xianyang, 712100, Shaanxi, China
| | - Gangtu Wang
- College of Food Science and Engineering, Northwest Agriculture and Forestry University, No. 22 Xinong Road, Yangling District, Xianyang, 712100, Shaanxi, China
| | - Yuanyuan Shan
- College of Food Science and Engineering, Northwest Agriculture and Forestry University, No. 22 Xinong Road, Yangling District, Xianyang, 712100, Shaanxi, China
| | - Yanglei Yi
- College of Food Science and Engineering, Northwest Agriculture and Forestry University, No. 22 Xinong Road, Yangling District, Xianyang, 712100, Shaanxi, China
| | - Yuan Zhou
- College of Food Science and Engineering, Northwest Agriculture and Forestry University, No. 22 Xinong Road, Yangling District, Xianyang, 712100, Shaanxi, China
| | - Bianfang Liu
- College of Food Science and Engineering, Northwest Agriculture and Forestry University, No. 22 Xinong Road, Yangling District, Xianyang, 712100, Shaanxi, China.
| | - Xin Wang
- College of Food Science and Engineering, Northwest Agriculture and Forestry University, No. 22 Xinong Road, Yangling District, Xianyang, 712100, Shaanxi, China.
| | - Xin Lü
- College of Food Science and Engineering, Northwest Agriculture and Forestry University, No. 22 Xinong Road, Yangling District, Xianyang, 712100, Shaanxi, China.
| |
Collapse
|
34
|
Grifka-Walk HM, Jenkins BR, Kominsky DJ. Amino Acid Trp: The Far Out Impacts of Host and Commensal Tryptophan Metabolism. Front Immunol 2021; 12:653208. [PMID: 34149693 PMCID: PMC8213022 DOI: 10.3389/fimmu.2021.653208] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 05/11/2021] [Indexed: 12/12/2022] Open
Abstract
Tryptophan (Trp) is an essential amino acid primarily derived from the diet for use by the host for protein synthesis. The intestinal tract is lined with cells, both host and microbial, that uptake and metabolize Trp to also generate important signaling molecules. Serotonin (5-HT), kynurenine and its downstream metabolites, and to a lesser extent other neurotransmitters are generated by the host to signal onto host receptors and elicit physiological effects. 5-HT production by neurons in the CNS regulates sleep, mood, and appetite; 5-HT production in the intestinal tract by enterochromaffin cells regulates gastric motility and inflammation in the periphery. Kynurenine can signal onto the aryl hydrocarbon receptor (AHR) to elicit pleiotropic responses from several cell types including epithelial and immune cells, or can be further metabolized into bioactive molecules to influence neurodegenerative disease. There is a remarkable amount of cross-talk with the microbiome with regard to tryptophan metabolites as well. The gut microbiome can regulate the production of host tryptophan metabolites and can use dietary or recycled trp to generate bioactive metabolites themselves. Trp derivatives like indole are able to signal onto xenobiotic receptors, including AHR, to elicit tolerogenic effects. Here, we review studies that demonstrate that tryptophan represents a key intra-kingdom signaling molecule.
Collapse
Affiliation(s)
| | | | - Douglas J. Kominsky
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT, United States
| |
Collapse
|
35
|
Daskalaki MG, Axarlis K, Aspevik T, Orfanakis M, Kolliniati O, Lapi I, Tzardi M, Dermitzaki E, Venihaki M, Kousoulaki K, Tsatsanis C. Fish Sidestream-Derived Protein Hydrolysates Suppress DSS-Induced Colitis by Modulating Intestinal Inflammation in Mice. Mar Drugs 2021; 19:312. [PMID: 34071180 PMCID: PMC8228426 DOI: 10.3390/md19060312] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 05/25/2021] [Accepted: 05/26/2021] [Indexed: 12/17/2022] Open
Abstract
Inflammatory bowel disease is characterized by extensive intestinal inflammation, and therapies against the disease target suppression of the inflammatory cascade. Nutrition has been closely linked to the development and suppression of inflammatory bowel disease, which to a large extent is attributed to the complex immunomodulatory properties of nutrients. Diets containing fish have been suggested to promote health and suppress inflammatory diseases. Even though most of the health-promoting properties of fish-derived nutrients are attributed to fish oil, the potential health-promoting properties of fish protein have not been investigated. Fish sidestreams contain large amounts of proteins, currently unexploited, with potential anti-inflammatory properties, and may possess additional benefits through bioactive peptides and free amino acids. In this project, we utilized fish protein hydrolysates, based on mackerel and salmon heads and backbones, as well as flounder skin collagen. Mice fed with a diet supplemented with different fish sidestream-derived protein hydrolysates (5% w/w) were exposed to the model of DSS-induced colitis. The results show that dietary supplements containing protein hydrolysates from salmon heads suppressed chemically-induced colitis development as determined by colon length and pro-inflammatory cytokine production. To evaluate colitis severity, we measured the expression of different pro-inflammatory cytokines and chemokines and found that the same supplement suppressed the pro-inflammatory cytokines IL-6 and TNFα and the chemokines Cxcl1 and Ccl3. We also assessed the levels of the anti-inflammatory cytokines IL-10 and Tgfb and found that selected protein hydrolysates induced their expression. Our findings demonstrate that protein hydrolysates derived from fish sidestreams possess anti-inflammatory properties in the model of DSS-induced colitis, providing a novel underexplored source of health-promoting dietary supplements.
Collapse
Affiliation(s)
- Maria G. Daskalaki
- Laboratory of Clinical Chemistry, Medical School, University of Crete, 70013 Heraklion, Greece; (M.G.D.); (K.A.); (M.O.); (O.K.); (I.L.); (E.D.); (M.V.)
- Institute of Molecular Biology and Biotechnology, FORTH, 71100 Heraklion, Greece
| | - Konstantinos Axarlis
- Laboratory of Clinical Chemistry, Medical School, University of Crete, 70013 Heraklion, Greece; (M.G.D.); (K.A.); (M.O.); (O.K.); (I.L.); (E.D.); (M.V.)
- Institute of Molecular Biology and Biotechnology, FORTH, 71100 Heraklion, Greece
| | - Tone Aspevik
- Department of Nutrition and Feed Technology, Nofima AS, 5141 Bergen, Norway; (T.A.); (K.K.)
| | - Michail Orfanakis
- Laboratory of Clinical Chemistry, Medical School, University of Crete, 70013 Heraklion, Greece; (M.G.D.); (K.A.); (M.O.); (O.K.); (I.L.); (E.D.); (M.V.)
- Institute of Molecular Biology and Biotechnology, FORTH, 71100 Heraklion, Greece
| | - Ourania Kolliniati
- Laboratory of Clinical Chemistry, Medical School, University of Crete, 70013 Heraklion, Greece; (M.G.D.); (K.A.); (M.O.); (O.K.); (I.L.); (E.D.); (M.V.)
- Institute of Molecular Biology and Biotechnology, FORTH, 71100 Heraklion, Greece
| | - Ioanna Lapi
- Laboratory of Clinical Chemistry, Medical School, University of Crete, 70013 Heraklion, Greece; (M.G.D.); (K.A.); (M.O.); (O.K.); (I.L.); (E.D.); (M.V.)
- Institute of Molecular Biology and Biotechnology, FORTH, 71100 Heraklion, Greece
| | - Maria Tzardi
- Laboratory of Pathology, School of Medicine, University of Crete, 70013 Heraklion, Greece;
| | - Eirini Dermitzaki
- Laboratory of Clinical Chemistry, Medical School, University of Crete, 70013 Heraklion, Greece; (M.G.D.); (K.A.); (M.O.); (O.K.); (I.L.); (E.D.); (M.V.)
| | - Maria Venihaki
- Laboratory of Clinical Chemistry, Medical School, University of Crete, 70013 Heraklion, Greece; (M.G.D.); (K.A.); (M.O.); (O.K.); (I.L.); (E.D.); (M.V.)
| | - Katerina Kousoulaki
- Department of Nutrition and Feed Technology, Nofima AS, 5141 Bergen, Norway; (T.A.); (K.K.)
| | - Christos Tsatsanis
- Laboratory of Clinical Chemistry, Medical School, University of Crete, 70013 Heraklion, Greece; (M.G.D.); (K.A.); (M.O.); (O.K.); (I.L.); (E.D.); (M.V.)
- Institute of Molecular Biology and Biotechnology, FORTH, 71100 Heraklion, Greece
| |
Collapse
|
36
|
Boleij A, Fathi P, Dalton W, Park B, Wu X, Huso D, Allen J, Besharati S, Anders RA, Housseau F, Mackenzie AE, Jenkins L, Milligan G, Wu S, Sears CL. G-protein coupled receptor 35 (GPR35) regulates the colonic epithelial cell response to enterotoxigenic Bacteroides fragilis. Commun Biol 2021; 4:585. [PMID: 33990686 PMCID: PMC8121840 DOI: 10.1038/s42003-021-02014-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 03/18/2021] [Indexed: 02/03/2023] Open
Abstract
G protein-coupled receptor (GPR)35 is highly expressed in the gastro-intestinal tract, predominantly in colon epithelial cells (CEC), and has been associated with inflammatory bowel diseases (IBD), suggesting a role in gastrointestinal inflammation. The enterotoxigenic Bacteroides fragilis (ETBF) toxin (BFT) is an important virulence factor causing gut inflammation in humans and animal models. We identified that BFT signals through GPR35. Blocking GPR35 function in CECs using the GPR35 antagonist ML145, in conjunction with shRNA knock-down and CRISPRcas-mediated knock-out, resulted in reduced CEC-response to BFT as measured by E-cadherin cleavage, beta-arrestin recruitment and IL-8 secretion. Importantly, GPR35 is required for the rapid onset of ETBF-induced colitis in mouse models. GPR35-deficient mice showed reduced death and disease severity compared to wild-type C57Bl6 mice. Our data support a role for GPR35 in the CEC and mucosal response to BFT and underscore the importance of this molecule for sensing ETBF in the colon.
Collapse
Affiliation(s)
- Annemarie Boleij
- Johns Hopkins University, Department of Medicine, Division of Infectious Diseases, Baltimore, MD, USA.
- Radboud University Medical Center (Radboudumc), Department of Pathology, Radboud Institute for Molecular Life sciences (RIMLS), Nijmegen, The Netherlands.
| | - Payam Fathi
- Johns Hopkins University, Department of Medicine, Division of Infectious Diseases, Baltimore, MD, USA
| | - William Dalton
- Johns Hopkins University, Department of Oncology Center-Hematologic Malignancies, Baltimore, MD, USA
| | - Ben Park
- Johns Hopkins University, Department of Oncology Center-Hematologic Malignancies, Baltimore, MD, USA
- Vanderbilt University Medical Center, Department of Medicine, Division of Hematology and Oncology, Nashville, Tenessee, USA
| | - Xinqun Wu
- Johns Hopkins University, Department of Medicine, Division of Infectious Diseases, Baltimore, MD, USA
| | - David Huso
- Johns Hopkins University, Department of Molecular and Comparative Pathobiology, Baltimore, MD, USA
| | - Jawara Allen
- Johns Hopkins University, Department of Medicine, Division of Infectious Diseases, Baltimore, MD, USA
| | - Sepideh Besharati
- Johns Hopkins University, Department of Pathobiology, Baltimore, MD, USA
| | - Robert A Anders
- Johns Hopkins University, Department of Pathobiology, Baltimore, MD, USA
| | - Franck Housseau
- Johns Hopkins University, Department of Oncology Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA
| | - Amanda E Mackenzie
- Centre for Translational Pharmacology, Institute of Molecular, Cell, and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, Scotland, UK
| | - Laura Jenkins
- Centre for Translational Pharmacology, Institute of Molecular, Cell, and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, Scotland, UK
| | - Graeme Milligan
- Centre for Translational Pharmacology, Institute of Molecular, Cell, and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, Scotland, UK
| | - Shaoguang Wu
- Johns Hopkins University, Department of Medicine, Division of Infectious Diseases, Baltimore, MD, USA
| | - Cynthia L Sears
- Johns Hopkins University, Department of Medicine, Division of Infectious Diseases, Baltimore, MD, USA
| |
Collapse
|
37
|
Niccolai E, Russo E, Baldi S, Ricci F, Nannini G, Pedone M, Stingo FC, Taddei A, Ringressi MN, Bechi P, Mengoni A, Fani R, Bacci G, Fagorzi C, Chiellini C, Prisco D, Ramazzotti M, Amedei A. Significant and Conflicting Correlation of IL-9 With Prevotella and Bacteroides in Human Colorectal Cancer. Front Immunol 2021; 11:573158. [PMID: 33488574 PMCID: PMC7820867 DOI: 10.3389/fimmu.2020.573158] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 11/19/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND AND AIM Gut microbiota (GM) can support colorectal cancer (CRC) progression by modulating immune responses through the production of both immunostimulatory and/or immunosuppressive cytokines. The role of IL-9 is paradigmatic because it can either promote tumor progression in hematological malignancies or inhibit tumorigenesis in solid cancers. Therefore, we investigate the microbiota-immunity axis in healthy and tumor mucosa, focusing on the correlation between cytokine profile and GM signature. METHODS In this observational study, we collected tumor (CRC) and healthy (CRC-S) mucosa samples from 45 CRC patients, who were undergoing surgery in 2018 at the Careggi University Hospital (Florence, Italy). First, we characterized the tissue infiltrating lymphocyte subset profile and the GM composition. Subsequently, we evaluated the CRC and CRC-S molecular inflammatory response and correlated this profile with GM composition, using Dirichlet multinomial regression. RESULTS CRC samples displayed higher percentages of Th17, Th2, and Tregs. Moreover, CRC tissues showed significantly higher levels of MIP-1α, IL-1α, IL-1β, IL-2, IP-10, IL-6, IL-8, IL-17A, IFN-γ, TNF-α, MCP-1, P-selectin, and IL-9. Compared to CRC-S, CRC samples also showed significantly higher levels of the following genera: Fusobacteria, Proteobacteria, Fusobacterium, Ruminococcus2, and Ruminococcus. Finally, the abundance of Prevotella spp. in CRC samples negatively correlated with IL-17A and positively with IL-9. On the contrary, Bacteroides spp. presence negatively correlated with IL-9. CONCLUSIONS Our data consolidate antitumor immunity impairment and the presence of a distinct microbiota profile in the tumor microenvironment compared with the healthy mucosa counterpart. Relating the CRC cytokine profile with GM composition, we confirm the presence of bidirectional crosstalk between the immune response and the host's commensal microorganisms. Indeed, we document, for the first time, that Prevotella spp. and Bacteroides spp. are, respectively, positively and negatively correlated with IL-9, whose role in CRC development is still under debate.
Collapse
Affiliation(s)
- Elena Niccolai
- Department of Clinical and Experimental Medicine, University of Florence, Florence, Italy
| | - Edda Russo
- Department of Clinical and Experimental Medicine, University of Florence, Florence, Italy
| | - Simone Baldi
- Department of Clinical and Experimental Medicine, University of Florence, Florence, Italy
| | - Federica Ricci
- Department of Biomedical, Experimental and Clinical Sciences “Mario Serio” University of Florence, Florence, Italy
| | - Giulia Nannini
- Department of Clinical and Experimental Medicine, University of Florence, Florence, Italy
| | - Matteo Pedone
- Department of Statistics, Computer Science, Applications “G. Parenti”, Florence, Italy
| | | | - Antonio Taddei
- Department of Clinical and Experimental Medicine, University of Florence, Florence, Italy
| | | | - Paolo Bechi
- Department of Clinical and Experimental Medicine, University of Florence, Florence, Italy
| | - Alessio Mengoni
- Department of Biology, University of Florence, Florence, Italy
| | - Renato Fani
- Department of Biology, University of Florence, Florence, Italy
| | - Giovanni Bacci
- Department of Biology, University of Florence, Florence, Italy
| | - Camilla Fagorzi
- Department of Biology, University of Florence, Florence, Italy
| | | | - Domenico Prisco
- Department of Clinical and Experimental Medicine, University of Florence, Florence, Italy
- SOD of Interdisciplinary Internal Medicine, Azienda Ospedaliera Universitaria Careggi (AOUC), Florence, Italy
| | - Matteo Ramazzotti
- Department of Biomedical, Experimental and Clinical Sciences “Mario Serio” University of Florence, Florence, Italy
| | - Amedeo Amedei
- Department of Clinical and Experimental Medicine, University of Florence, Florence, Italy
- SOD of Interdisciplinary Internal Medicine, Azienda Ospedaliera Universitaria Careggi (AOUC), Florence, Italy
| |
Collapse
|
38
|
Hu J, Zhao Q, Kong LY, Wang J, Yan J, Xia X, Jia Z, Heimberger AB, Li S. Regulation of tumor immune suppression and cancer cell survival by CXCL1/2 elevation in glioblastoma multiforme. SCIENCE ADVANCES 2021; 7:eabc2511. [PMID: 33571109 PMCID: PMC7840139 DOI: 10.1126/sciadv.abc2511] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 12/04/2020] [Indexed: 05/17/2023]
Abstract
The invasiveness and high immune suppression of glioblastoma multiforme (GBM) produce poor survival of afflicted patients. Unfortunately, in the past decades, no therapeutic approach has remarkably improved the survival time of patients with GBM. Our analysis of the TCGA database and brain tumor tissue arrays indicated that CXCL1 and CXCL2 overexpression is closely associated with GBM's aggressiveness. Our results showed that elevation of CXCL1 or CXCL2 facilitated myeloid cell migration and simultaneously disrupted CD8+ T cell accumulation at tumor sites, causing accelerated tumor progression. Yet, blockade of CXCL1/2 significantly prevented myeloid-derived suppressor cell migration and thereby increased CD8+ T cell accumulation in vitro and in vivo. CXCL1/2 also promoted the paracrine factor S100A9 and further activated Erk1/2 and p70S60k, whereas blocking CXCL1/2 down-regulated these prosurvival factors. The combination of targeting CXCL1/2 and standard temozolomide chemotherapy improved upon the antitumor efficacy of chemotherapy alone, extending the overall survival time in GBM.
Collapse
Affiliation(s)
- Jiemiao Hu
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Qingnan Zhao
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ling-Yuan Kong
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jian Wang
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jun Yan
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xueqing Xia
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Zhiliang Jia
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Amy B Heimberger
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Shulin Li
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
39
|
Zhou Y, Yang D, Yang Q, Lv X, Huang W, Zhou Z, Wang Y, Zhang Z, Yuan T, Ding X, Tang L, Zhang J, Yin J, Huang Y, Yu W, Wang Y, Zhou C, Su Y, He A, Sun Y, Shen Z, Qian B, Meng W, Fei J, Yao Y, Pan X, Chen P, Hu H. Single-cell RNA landscape of intratumoral heterogeneity and immunosuppressive microenvironment in advanced osteosarcoma. Nat Commun 2020; 11:6322. [PMID: 33303760 PMCID: PMC7730477 DOI: 10.1038/s41467-020-20059-6] [Citation(s) in RCA: 333] [Impact Index Per Article: 66.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Accepted: 11/06/2020] [Indexed: 02/07/2023] Open
Abstract
Osteosarcoma is the most frequent primary bone tumor with poor prognosis. Through RNA-sequencing of 100,987 individual cells from 7 primary, 2 recurrent, and 2 lung metastatic osteosarcoma lesions, 11 major cell clusters are identified based on unbiased clustering of gene expression profiles and canonical markers. The transcriptomic properties, regulators and dynamics of osteosarcoma malignant cells together with their tumor microenvironment particularly stromal and immune cells are characterized. The transdifferentiation of malignant osteoblastic cells from malignant chondroblastic cells is revealed by analyses of inferred copy-number variation and trajectory. A proinflammatory FABP4+ macrophages infiltration is noticed in lung metastatic osteosarcoma lesions. Lower osteoclasts infiltration is observed in chondroblastic, recurrent and lung metastatic osteosarcoma lesions compared to primary osteoblastic osteosarcoma lesions. Importantly, TIGIT blockade enhances the cytotoxicity effects of the primary CD3+ T cells with high proportion of TIGIT+ cells against osteosarcoma. These results present a single-cell atlas, explore intratumor heterogeneity, and provide potential therapeutic targets for osteosarcoma.
Collapse
Affiliation(s)
- Yan Zhou
- Oncology Department of Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Dong Yang
- Orthopaedic Department of Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Qingcheng Yang
- Orthopaedic Department of Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Xiaobin Lv
- Central Laboratory of the First Hospital of Nanchang, Nanchang, 330008, China
| | - Wentao Huang
- Pathology Department of Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Zhenhua Zhou
- Department of Orthopedic Oncology, Changzheng Hospital of Naval Military Medical University, Shanghai, 200003, China
| | - Yaling Wang
- Oncology Department of Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Zhichang Zhang
- Orthopaedic Department of Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Ting Yuan
- Orthopaedic Department of Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Xiaomin Ding
- Oncology Department of Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Lina Tang
- Oncology Department of Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Jianjun Zhang
- Oncology Department of Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Junyi Yin
- Oncology Department of Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Yujing Huang
- Oncology Department of Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Wenxi Yu
- Oncology Department of Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Yonggang Wang
- Oncology Department of Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Chenliang Zhou
- Oncology Department of Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Yang Su
- Oncology Department of Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Aina He
- Oncology Department of Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Yuanjue Sun
- Oncology Department of Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Zan Shen
- Oncology Department of Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Binzhi Qian
- MRC Centre for Reproductive Health & Edinburgh Cancer Research UK Centre, Queen's Medical Research Institute, EH16 4TJ, Edinburgh, United Kingdom
| | - Wei Meng
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
- Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Guangzhou, 510515, China
| | - Jia Fei
- Department of Biochemistry and Molecular Biology, Medical College of Jinan University, 601 Western Huangpu Avenue, Guangzhou, 510632, China
| | - Yang Yao
- Oncology Department of Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China.
| | - Xinghua Pan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.
- Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Guangzhou, 510515, China.
| | - Peizhan Chen
- Clinical Research Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201821, China.
| | - Haiyan Hu
- Oncology Department of Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China.
| |
Collapse
|
40
|
Daghestani MH, H. Hakami H, Ambreen K, Saleem AM, Aleisa NA, Al Neghery LM, Amin MH. Assessment of tumor-suppressive inflammatory tendency of Echis coloratus venom against colon carcinogenesis, via up-regulation of anti-tumorigenic inflammatory cells and down-regulation of pro-tumorigenic inflammatory cells, in colon cancer cell lines. TOXIN REV 2020. [DOI: 10.1080/15569543.2020.1818107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Affiliation(s)
- Maha H. Daghestani
- Department of Zoology, College of Science, Centre for Scientific and Medical Female Colleges, King Saud University, Riyadh, Saudi Arabia
| | - Hana H. Hakami
- Department of Zoology, College of Science, Centre for Scientific and Medical Female Colleges, King Saud University, Riyadh, Saudi Arabia
| | - Khushboo Ambreen
- Department of Biotechnology, Integral University, Lucknow, India
| | - Abdulaziz M. Saleem
- Department of Surgery, Medical College, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Nadia A. Aleisa
- Department of Zoology, College of Science, Centre for Scientific and Medical Female Colleges, King Saud University, Riyadh, Saudi Arabia
| | - Lina M. Al Neghery
- Department of Biology, College of Science, Al Imam Mohammad Ibn Saud Islamic University, Riyadh, Saudi Arabia
| | | |
Collapse
|
41
|
Ding P, Li L, Li L, Lv X, Zhou D, Wang Q, Chen J, Yang C, Xu E, Dai W, Zhang X, Wang N, Wang Q, Zhang W, Zhang L, Zhou Y, Gu H, Lei Q, Zhou X, Hu W. C5aR1 is a master regulator in Colorectal Tumorigenesis via Immune modulation. Theranostics 2020; 10:8619-8632. [PMID: 32754267 PMCID: PMC7392014 DOI: 10.7150/thno.45058] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 06/24/2020] [Indexed: 02/07/2023] Open
Abstract
Numerous factors have been claimed to play important roles in colorectal cancer (CRC) tumorigenesis, including myeloid-derived suppressor cells (MDSCs) and other immune cells, cytokines, and chemokines; however, the precise mechanisms of colorectal tumorigenesis remain elusive, and there is a lack of effective preventive treatments. Here, we investigated the role of complement system, a key regulator of immune surveillance and homeostasis, in colorectal tumorigenesis. Methods: The prototypical CRC model was induced by combined administration of azoxymethane (AOM)/ dextran sulfate sodium (DSS) in Wild-type (WT), C3-, C5-, C5ar1-, and C5ar2-deficient mice. Using flow cytometry, immunohistochemical staining and multiplex bead assay, we profiled the immune cells, cytokines and chemokines. Bone marrow transplantation was employed to determine the contribution of immune cells in colorectal tumorigenesis. Further, we used C5aR1 antagonist PMX205 to investigate the protective role in colorectal tumorigenesis. Results: Complement was extensively activated in inflamed tissues of AOM/DSS-induced murine CRC model, leading to multifaceted consequences. The deficiency of complement C5 or especially C5ar1, but not C3 almost completely prevented CRC tumorigenesis. C5a/C5aR1 signaling recruited MDSCs into the inflamed colorectum to impair CD8+ T cells, and modulated the production of critical cytokines and chemokines, thus initiating CRC. Moreover, the C5aR1 antagonist PMX205 strongly impeded colorectal tumorigenesis. Bone marrow transplantation further revealed that C5aR1 expression by immune cells was critical for colorectal tumorigenesis. Conclusion: Our study identifies C5a/C5aR1 signaling as a vital immunomodulatory program in CRC tumorigenesis and suggests a feasible preventive strategy.
Collapse
|
42
|
Ma C, Yang D, Wang B, Wu C, Wu Y, Li S, Liu X, Lassen K, Dai L, Yang S. Gasdermin D in macrophages restrains colitis by controlling cGAS-mediated inflammation. SCIENCE ADVANCES 2020; 6:eaaz6717. [PMID: 32671214 PMCID: PMC7314554 DOI: 10.1126/sciadv.aaz6717] [Citation(s) in RCA: 116] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Accepted: 03/09/2020] [Indexed: 05/13/2023]
Abstract
The functional relevance and mechanistic basis of the effects of the pyroptosis executioner Gasdermin D (GSDMD) on colitis remain unclear. In this study, we observed that GSDMD protein was activated during intestinal inflammation in a model of chemically induced colitis. GSDMD deficiency exacerbated experimental colitis independent of changes in the microbiota and without affecting the production of antimicrobial peptides. GSDMD deficiency in macrophages, but not epithelial cells, was sufficient to drive this exacerbated experimental colitis. We further demonstrate that GSDMD functions in macrophages as a negative regulator to control cyclic GMP-AMP synthase (cGAS)-dependent inflammation, thereby protecting against colitis. Moreover, the administration of cGAS inhibitor can rescue the colitogenic phenotype in GSDMD-deficient mice. Collectively, these findings provide the first demonstration of GSDMD's role in controlling colitis and a detailed delineation of the underlying mechanism.
Collapse
Affiliation(s)
- Chunmei Ma
- Department of Immunology, Key Laboratory of Immunological Environment and Disease, State Key Laboratory of Reproductive Medicine, Center for Global Health, Nanjing Medical University, Nanjing, China
| | - Dongxue Yang
- Department of Immunology, Key Laboratory of Immunological Environment and Disease, State Key Laboratory of Reproductive Medicine, Center for Global Health, Nanjing Medical University, Nanjing, China
| | - Bingwei Wang
- Department of Pharmacology, Nanjing University of Chinese Medicine, Nanjing, China
| | - Chunyan Wu
- Department of Immunology, Key Laboratory of Immunological Environment and Disease, State Key Laboratory of Reproductive Medicine, Center for Global Health, Nanjing Medical University, Nanjing, China
| | - Yuqing Wu
- Department of Immunology, Key Laboratory of Immunological Environment and Disease, State Key Laboratory of Reproductive Medicine, Center for Global Health, Nanjing Medical University, Nanjing, China
| | - Sheng Li
- Department of Immunology, Key Laboratory of Immunological Environment and Disease, State Key Laboratory of Reproductive Medicine, Center for Global Health, Nanjing Medical University, Nanjing, China
| | - Xue Liu
- Department of Immunology, Key Laboratory of Immunological Environment and Disease, State Key Laboratory of Reproductive Medicine, Center for Global Health, Nanjing Medical University, Nanjing, China
| | - Kara Lassen
- Roche Innovation Center Basel, Basel, Switzerland
| | - Lue Dai
- Roche Innovation Center Shanghai, Shanghai, China
| | - Shuo Yang
- Department of Immunology, Key Laboratory of Immunological Environment and Disease, State Key Laboratory of Reproductive Medicine, Center for Global Health, Nanjing Medical University, Nanjing, China
| |
Collapse
|
43
|
Lee H, Son YS, Lee MO, Ryu JW, Park K, Kwon O, Jung KB, Kim K, Ryu TY, Baek A, Kim J, Jung CR, Ryu CM, Park YJ, Han TS, Kim DS, Cho HS, Son MY. Low-dose interleukin-2 alleviates dextran sodium sulfate-induced colitis in mice by recovering intestinal integrity and inhibiting AKT-dependent pathways. Theranostics 2020; 10:5048-5063. [PMID: 32308767 PMCID: PMC7163458 DOI: 10.7150/thno.41534] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Accepted: 03/23/2020] [Indexed: 12/14/2022] Open
Abstract
Several phase 1/2 clinical trials showed that low-dose interleukin-2 (IL-2) treatment is a safe and effective strategy for the treatment of chronic graft-versus-host disease, hepatitis C virus-induced vasculitis, and type 1 diabetes. Ulcerative colitis (UC) is a chronic inflammatory condition of the colon that lacks satisfactory treatment. In this study, we aimed to determine the effects of low-dose IL-2 as a therapeutic for UC on dextran sulfate sodium (DSS)-induced colitis. Methods: Mice with DSS-induced colitis were intraperitoneally injected with low-dose IL-2. Survival, body weight, disease activity index, colon length, histopathological score, myeloperoxidase activity and inflammatory cytokine levels as well as intestinal barrier integrity were examined. Differential gene expression after low-dose IL-2 treatment was analyzed by RNA-sequencing. Results: Low-dose IL-2 significantly improved the symptoms of DSS-induced colitis in mice and attenuated pro-inflammatory cytokine production and immune cell infiltration. The most effective dose range of IL-2 was 16K-32K IU/day. Importantly, low-dose IL-2 was effective in ameliorating the disruption of epithelial barrier integrity in DSS-induced colitis tissues by restoring tight junction proteins and mucin production and suppressing apoptosis. The colon tissue of DSS-induced mice exposed to low-dose IL-2 mimic gene expression patterns in the colons of control mice. Furthermore, we identified the crucial role of the PI3K-AKT pathway in exerting the therapeutic effect of low-dose IL-2. Conclusions: The results of our study suggest that low-dose IL-2 has therapeutic effects on DSS-induced colitis and potential clinical value in treating UC.
Collapse
|
44
|
Alafate W, Li X, Zuo J, Zhang H, Xiang J, Wu W, Xie W, Bai X, Wang M, Wang J. Elevation of CXCL1 indicates poor prognosis and radioresistance by inducing mesenchymal transition in glioblastoma. CNS Neurosci Ther 2020; 26:475-485. [PMID: 32187449 PMCID: PMC7080429 DOI: 10.1111/cns.13297] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 10/14/2019] [Accepted: 10/18/2019] [Indexed: 12/13/2022] Open
Abstract
INTRODUCTION Glioblastoma (GBM) is identified as a lethal malignant tumor derived from the nervous system. Despite the standard clinical strategy including maximum surgical resection, temozolomide (TMZ) chemotherapy, and radiotherapy, the median survival of GBM patients remains <15 months. Accumulating evidence indicates that rapid-acquired radioresistance is one of the most common reasons for GBM recurrence. Therefore, developing novel therapeutic targets for radioresistant GBM could yield long-term cures. AIMS To investigate the functional role of CXCL1 in the acquired radioresistance and identify the molecular pathway correlated to CXCL1. RESULTS In this study, we identified that CXCL1 is highly expressed in GBM and the elevation of CXCL1 is involved in radioresistance and poor prognosis in GBM patients. Additionally, silencing CXCL1 attenuated the proliferation and radioresistance of GBM cells. Furthermore, we demonstrated that CXCL1-overexpression induced radioresistance through mesenchymal transition of GBM via the activation of nuclear factor-kappa B (NF-κB) signaling. CONCLUSION CXCL1 was highly enriched in GBM and positively correlated with poor prognosis in GBM patients. Additionally, elevated CXCL1 induced radioresistance in GBM through regulation of NF-κB signaling by promoting mesenchymal transition in GBM.
Collapse
Affiliation(s)
- Wahafu Alafate
- Department of NeurosurgeryThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
- Center of Brain ScienceThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Xiaodong Li
- Department of NeurosurgeryThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Jie Zuo
- The Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Hua Zhang
- Department of NeurosurgeryThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
- Center of Brain ScienceThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Jianyang Xiang
- Department of NeurosurgeryThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Wei Wu
- Department of NeurosurgeryThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Wanfu Xie
- Department of NeurosurgeryThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Xiaobin Bai
- Department of NeurosurgeryThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Maode Wang
- Department of NeurosurgeryThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
- Center of Brain ScienceThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Jia Wang
- Department of NeurosurgeryThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
- Center of Brain ScienceThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| |
Collapse
|
45
|
Suh JH, Park MC, Goughnour PC, Min BS, Kim SB, Lee WY, Cho YB, Cheon JH, Lee KY, Nam DH, Kim S. Plasma Lysyl-tRNA Synthetase 1 (KARS1) as a Novel Diagnostic and Monitoring Biomarker for Colorectal Cancer. J Clin Med 2020; 9:533. [PMID: 32075312 PMCID: PMC7073917 DOI: 10.3390/jcm9020533] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 02/10/2020] [Accepted: 02/11/2020] [Indexed: 12/27/2022] Open
Abstract
Colorectal cancer (CRC) is one of the leading causes of world cancer deaths. To improve the survival rate of CRC, diagnosis and post-operative monitoring is necessary. Currently, biomarkers are used for CRC diagnosis and prognosis. However, these biomarkers have limitations of specificity and sensitivity. Levels of plasma lysyl-tRNA synthetase (KARS1), which was reported to be secreted from colon cancer cells by stimuli, along with other secreted aminoacyl-tRNA synthetases (ARSs), were analyzed in CRC and compared with the currently used biomarkers. The KARS1 levels of CRC patients (n = 164) plasma were shown to be higher than those of healthy volunteers (n = 32). The diagnostic values of plasma KARS1 were also evaluated by receiving operating characteristic (ROC) curve. Compared with other biomarkers and ARSs, KARS1 showed the best diagnostic value for CRC. The cancer specificity and burden correlation of plasma KARS1 level were validated using azoxymethane (AOM)/dextran sodium sulfate (DSS) model, and paired pre- and post-surgery CRC patient plasma. In the AOM/DSS model, the plasma level of KARS1 showed high correlation with number of polyps, but not for inflammation. Using paired pre- and post-surgery CRC plasma samples (n = 60), the plasma level of KARS1 was significantly decreased in post-surgery samples. Based on these evidence, KARS1, a surrogate biomarker reflecting CRC burden, can be used as a novel diagnostic and post-operative monitoring biomarker for CRC.
Collapse
Affiliation(s)
- Ji Hun Suh
- Medicinal Bioconvergence Research Center, College of Pharmacy, Seoul National University, Seoul 08826, Korea; (J.H.S.); (M.C.P.); (P.C.G.); (S.B.K.)
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Technology, Seoul National University, Seoul 08826, Korea
| | - Min Chul Park
- Medicinal Bioconvergence Research Center, College of Pharmacy, Seoul National University, Seoul 08826, Korea; (J.H.S.); (M.C.P.); (P.C.G.); (S.B.K.)
| | - Peter C. Goughnour
- Medicinal Bioconvergence Research Center, College of Pharmacy, Seoul National University, Seoul 08826, Korea; (J.H.S.); (M.C.P.); (P.C.G.); (S.B.K.)
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Technology, Seoul National University, Seoul 08826, Korea
| | - Byung Soh Min
- Seoul Republic of Korea Department of Surgery, Severance Hospital, Yonsei University College of Medicine, Seoul 03722, Korea; (B.S.M.); (K.Y.L.)
| | - Sang Bum Kim
- Medicinal Bioconvergence Research Center, College of Pharmacy, Seoul National University, Seoul 08826, Korea; (J.H.S.); (M.C.P.); (P.C.G.); (S.B.K.)
| | - Woo Yong Lee
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea; (W.Y.L.); (Y.B.C.)
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul 06351, Korea;
| | - Yong Beom Cho
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea; (W.Y.L.); (Y.B.C.)
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul 06351, Korea;
| | - Jae Hee Cheon
- Department of Internal Medicine and Institute of Gastroenterology, Yonsei University College of Medicine, Seoul 03722, Korea;
| | - Kang Young Lee
- Seoul Republic of Korea Department of Surgery, Severance Hospital, Yonsei University College of Medicine, Seoul 03722, Korea; (B.S.M.); (K.Y.L.)
| | - Do-Hyun Nam
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul 06351, Korea;
- Institute for Refractory Cancer Research, Samsung Medical Center, Seoul 06351, Korea
- Department of Neurosurgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
| | - Sunghoon Kim
- Medicinal Bioconvergence Research Center, College of Pharmacy, Seoul National University, Seoul 08826, Korea; (J.H.S.); (M.C.P.); (P.C.G.); (S.B.K.)
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Technology, Seoul National University, Seoul 08826, Korea
| |
Collapse
|
46
|
Epithelial RABGEF1 deficiency promotes intestinal inflammation by dysregulating intrinsic MYD88-dependent innate signaling. Mucosal Immunol 2020; 13:96-109. [PMID: 31628426 DOI: 10.1038/s41385-019-0211-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 09/18/2019] [Accepted: 09/28/2019] [Indexed: 02/06/2023]
Abstract
Intestinal epithelial cells (IECs) contribute to the regulation of intestinal homeostasis and inflammation through their interactions with the environment and host immune responses. Yet our understanding of IEC-intrinsic regulatory pathways remains incomplete. Here, we identify the guanine nucleotide exchange factor RABGEF1 as a regulator of intestinal homeostasis and innate pathways dependent on IECs. Mice with IEC-specific Rabgef1 deletion (called Rabgef1IEC-KO mice) developed a delayed spontaneous colitis associated with the local upregulation of IEC chemokine expression. In mouse models of colitis based on Interleukin-10 deficiency or dextran sodium sulfate (DSS) exposure, we found that IEC-intrinsic RABGEF1 deficiency exacerbated development of intestinal pathology and dysregulated IEC innate pathways and chemokine expression. Mechanistically, we showed that RABGEF1 deficiency in mouse IECs in vitro was associated with an impairment of early endocytic events, an increased activation of the p38 mitogen-activated protein kinase (MAPK)-dependent pathway, and increased chemokine secretion. Moreover, we provided evidence that the development of spontaneous colitis was dependent on microbiota-derived signals and intrinsic MYD88-dependent pathways in vivo. Our study identifies mouse RABGEF1 as an important regulator of intestinal inflammation, MYD88-dependent IEC-intrinsic signaling, and chemokine production. This suggests that RABGEF1-dependent pathways represent interesting therapeutic targets for inflammatory conditions in the gut.
Collapse
|
47
|
Krzystek-Korpacka M, Zawadzki M, Lewandowska P, Szufnarowski K, Bednarz-Misa I, Jacyna K, Witkiewicz W, Gamian A. Distinct Chemokine Dynamics in Early Postoperative Period after Open and Robotic Colorectal Surgery. J Clin Med 2019; 8:jcm8060879. [PMID: 31248170 PMCID: PMC6616914 DOI: 10.3390/jcm8060879] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 05/29/2019] [Accepted: 06/17/2019] [Indexed: 01/13/2023] Open
Abstract
Stress response to robot-assisted colorectal surgery is largely unknown. Therefore, we conducted a prospective comparative nonrandomized study evaluating the perioperative dynamics of chemokines: IL-8/CXCL8, MCP-1/CCL2, MIP-1α/CCL3, MIP-1β/CCL4, RANTES/CCL5, and eotaxin-1/CCL11 in 61 colorectal cancer patients following open colorectal surgery (OCS) or robot-assisted surgery (RACS) in reference to clinical data. Postoperative IL-8 and MCP-1 increase was reduced in RACS with a magnitude of blood loss, length of surgery, and concomitant up-regulation of IL-6 and TNFα as its independent predictors. RANTES at 8 h dropped in RACS and RANTES, and MIP1α/β at 24 h were more elevated in RACS than OCS. IL-8 and MCP-1 at 72 h remained higher in patients subsequently developing surgical site infections, in whom a 2.6- and 2.5-fold increase was observed. IL-8 up-regulation at 24 h in patients undergoing open procedure was predictive of anastomotic leak (AL; 94% accuracy). Changes in MCP-1 and RANTES were predictive of delayed restoration of bowel function. Chemokines behave differently depending on procedure. A robot-assisted approach may be beneficial in terms of chemokine dynamics by favoring Th1 immunity and attenuated angiogenic potential and postoperative ileus. Monitoring chemokine dynamics may prove useful for predicting adverse clinical events. Attenuated chemokine up-regulation results from less severe blood loss and diminished inflammatory response.
Collapse
Affiliation(s)
| | - Marek Zawadzki
- Department of Oncological Surgery, Regional Specialist Hospital, 51-124 Wroclaw, Poland.
| | - Paulina Lewandowska
- Department of Medical Biochemistry, Wroclaw Medical University, 50-368 Wroclaw, Poland.
| | | | - Iwona Bednarz-Misa
- Department of Medical Biochemistry, Wroclaw Medical University, 50-368 Wroclaw, Poland.
| | - Krzysztof Jacyna
- Department of Oncological Surgery, Regional Specialist Hospital, 51-124 Wroclaw, Poland.
| | - Wojciech Witkiewicz
- Department of Oncological Surgery, Regional Specialist Hospital, 51-124 Wroclaw, Poland.
- Research and Development Centre at Regional Specialist Hospital, 51-124 Wroclaw, Poland.
| | - Andrzej Gamian
- Department of Medical Biochemistry, Wroclaw Medical University, 50-368 Wroclaw, Poland.
- Laboratory of Medical Microbiology, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland.
| |
Collapse
|
48
|
Eichelmann F, Schulze MB, Wittenbecher C, Menzel J, Weikert C, di Giuseppe R, Biemann R, Isermann B, Fritsche A, Boeing H, Aleksandrova K. Association of Chemerin Plasma Concentration With Risk of Colorectal Cancer. JAMA Netw Open 2019; 2:e190896. [PMID: 30901045 PMCID: PMC6583278 DOI: 10.1001/jamanetworkopen.2019.0896] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
IMPORTANCE Inflammatory processes have been suggested to have an important role in colorectal cancer (CRC) etiology. Chemerin is a recently discovered inflammatory biomarker thought to exert chemotactic, adipogenic, and angiogenic functions. However, its potential link with CRC has not been sufficiently explored. OBJECTIVE To evaluate the prospective association of circulating plasma chemerin concentrations with incident CRC. DESIGN, SETTING, AND PARTICIPANTS Prospective case-cohort study based on 27 548 initially healthy participants from the European Prospective Investigation Into Cancer and Nutrition (EPIC)-Potsdam cohort who were followed for up to 16 years. Baseline study information and samples were collected between August 23, 1994, and September 25, 1998. Recruitment was according to random registry sampling from the geographical area of Potsdam, Germany, and surrounding municipalities. The last date of study follow-up was May 10, 2010. Statistical analysis was conducted in 2018. MAIN OUTCOMES AND MEASURES Incident CRC, colon cancer, and rectal cancer. Baseline chemerin plasma concentrations were measured by enzyme-linked immunosorbent assay. RESULTS A random subcohort of 221 incident CRC cases and 2329 participants free of CRC with available blood sample measurements were included in the analysis. The participants' mean (SD) age was 50 (9) years, 62.1% were female, and 16.5% had a body mass index greater than 30. In multivariable-adjusted Cox proportional hazards regression models taking into account established CRC risk factors, higher chemerin concentrations were associated with a greater risk of CRC, with a hazard ratio (HR) of 1.81 (95% CI, 1.08-3.05; P for trend = .007) for the highest chemerin quartile vs the lowest. Analyses by cancer subsite indicated a stronger association with colon cancer (HR, 2.27; 95% CI, 1.18-4.34 for the highest quartile vs the lowest; P for trend = .005) compared with rectal cancer (HR, 1.27; 95% CI, 0.57-2.85; P for trend = .35). The association was particularly strong for proximal colon cancer (HR, 3.97; 95% CI, 1.51-10.50; P for trend = .001). CONCLUSIONS AND RELEVANCE This study found that the association between chemerin concentration and the risk of incident CRC was linear and independent of established CRC risk factors. Further studies are warranted to evaluate chemerin as a novel immune-inflammatory agent in colorectal carcinogenesis.
Collapse
Affiliation(s)
- Fabian Eichelmann
- Senior Scientist Group Nutrition, Immunity and Metabolism, Department of Nutrition and Gerontology, German Institute of Human Nutrition Potsdam–Rehbrücke (DIfE), Nuthetal, Germany
| | - Matthias B. Schulze
- German Centre for Cardiovascular Research (DZHK), Berlin, Germany
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam–Rehbrücke (DIfE), Nuthetal, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute of Nutritional Science, University of Potsdam, Potsdam, Germany
| | - Clemens Wittenbecher
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam–Rehbrücke (DIfE), Nuthetal, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Juliane Menzel
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam–Rehbrücke (DIfE), Nuthetal, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Department of Food Safety, German Federal Institute for Risk Assessment, Berlin, Germany
| | - Cornelia Weikert
- Department of Food Safety, German Federal Institute for Risk Assessment, Berlin, Germany
- Institute for Social Medicine, Epidemiology and Health Economics, Charité University Medical Center, Berlin, Germany
| | - Romina di Giuseppe
- Institute of Epidemiology, Christian Albrechts University Kiel, Kiel, Germany
| | - Ronald Biemann
- Department of Clinical Chemistry and Pathobiochemistry, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Berend Isermann
- Department of Clinical Chemistry and Pathobiochemistry, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Andreas Fritsche
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Division of Endocrinology, Diabetology, Nephrology, Vascular Disease and Clinical Chemistry, Department of Internal Medicine, University of Tübingen, Tübingen, Germany
| | - Heiner Boeing
- Department of Epidemiology, German Institute of Human Nutrition Potsdam–Rehbrücke (DIfE), Nuthetal, Germany
| | - Krasimira Aleksandrova
- Senior Scientist Group Nutrition, Immunity and Metabolism, Department of Nutrition and Gerontology, German Institute of Human Nutrition Potsdam–Rehbrücke (DIfE), Nuthetal, Germany
- Institute of Nutritional Science, University of Potsdam, Potsdam, Germany
| |
Collapse
|
49
|
Rajarathnam K, Schnoor M, Richardson RM, Rajagopal S. How do chemokines navigate neutrophils to the target site: Dissecting the structural mechanisms and signaling pathways. Cell Signal 2019; 54:69-80. [PMID: 30465827 PMCID: PMC6664297 DOI: 10.1016/j.cellsig.2018.11.004] [Citation(s) in RCA: 171] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 11/07/2018] [Accepted: 11/08/2018] [Indexed: 12/12/2022]
Abstract
Chemokines play crucial roles in combating microbial infection and initiating tissue repair by recruiting neutrophils in a timely and coordinated manner. In humans, no less than seven chemokines (CXCL1, CXCL2, CXCL3, CXCL5, CXCL6, CXCL7, and CXCL8) and two receptors (CXCR1 and CXCR2) mediate neutrophil functions but in a context dependent manner. Neutrophil-activating chemokines reversibly exist as monomers and dimers, and their receptor binding triggers conformational changes that are coupled to G-protein and β-arrestin signaling pathways. G-protein signaling activates a variety of effectors including Ca2+ channels and phospholipase C. β-arrestin serves as a multifunctional adaptor and is coupled to several signaling hubs including MAP kinase and tyrosine kinase pathways. Both G-protein and β-arrestin signaling pathways play important non-overlapping roles in neutrophil trafficking and activation. Functional studies have established many similarities but distinct differences for a given chemokine and between chemokines at the level of monomer vs. dimer, CXCR1 vs. CXCR2 activation, and G-protein vs. β-arrestin pathways. We propose that two forms of the ligand binding two receptors and activating two signaling pathways enables fine-tuned neutrophil function compared to a single form, a single receptor, or a single pathway. We summarize the current knowledge on the molecular mechanisms by which chemokine monomers/dimers activate CXCR1/CXCR2 and how these interactions trigger G-protein/β-arrestin-coupled signaling pathways. We also discuss current challenges and knowledge gaps, and likely advances in the near future that will lead to a better understanding of the relationship between the chemokine-CXCR1/CXCR2-G-protein/β-arrestin axis and neutrophil function.
Collapse
Affiliation(s)
- Krishna Rajarathnam
- Department of Biochemistry and Molecular Biology, Department of Microbiology and Immunology, Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch, Galveston, TX, USA.
| | - Michael Schnoor
- Department for Molecular Biomedicine, Cinvestav-IPN, 07360 Mexico City, Mexico
| | - Ricardo M Richardson
- Julius L. Chambers Biomedical/Biotechnology Research Institute, North Carolina Central University, Durham, NC 27707, USA
| | | |
Collapse
|
50
|
Synbiotic combination of prebiotic grape pomace extract and probiotic Lactobacillus sp. reduced important intestinal inflammatory markers and in-depth signalling mediators in lipopolysaccharide-treated Caco-2 cells. Br J Nutr 2018; 121:291-305. [PMID: 30565527 DOI: 10.1017/s0007114518003410] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Inflammatory bowel diseases (IBD) are a major problem for public health, with an increased incidence and impact on life quality. The effect of pre- and probiotic combination has been less studied in IBD. Using genomic and proteomic array technologies, this study examined the efficacy of a new combination of natural alternatives: prebiotics (grape pomace extract, GP) and probiotics (lactobacilli mixture, Lb mix) on inflammation and intracellular signalling routes in a cellular model of inflammation. Caco-2 cells challenged with lipopolysaccharide (LPS) for 4 h were treated with GP extract (50 μg/ml gallic acid equivalent) and Lb combination (3 × 108 colony-forming units/ml total Lb) for 24 h. The profile expressions of forty key inflammatory markers and twenty-six signalling kinases were analysed. Other markers involved in inflammation were also investigated (NF-κB/RELA, Nrf2, aryl hydrocarbon receptor, Cyp1A1, Cyp1B1); 57·5 and 60 % of investigated genes and proteins, respectively, were down-regulated by the synbiotic combination. Relevant cytokines and chemokines involved in response to microbial infection and inflammation were reduced under the level induced by LPS treatment and toward the unchallenged control. As expected, the reduction effect seems to imply mitogen-activated protein kinase and NF-κB pathway. Most of the signalling molecules activated by LPS were decreased by GP extract and Lb mix. Our study indicates that the synbiotic combination of GP extract and Lactobacillus sp. mixture exerted anti-inflammatory properties, which are able to decrease the majority of inflammatory genes, their proteins and associated signalling markers. Due to protective role of GP compounds on lactobacilli probiotic, this synbiotic combination might serve as a promising adjunctive therapy in intestinal inflammations.
Collapse
|