101
|
El-Mahdy NA, El-Sayad MES, El-Kadem AH, Abu-Risha SES. Targeting IL-10, ZO-1 gene expression and IL-6/STAT-3 trans-signaling by a combination of atorvastatin and mesalazine to enhance anti-inflammatory effects and attenuates progression of oxazolone-induced colitis. Fundam Clin Pharmacol 2020; 35:143-155. [PMID: 32383169 DOI: 10.1111/fcp.12563] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 04/30/2020] [Accepted: 05/04/2020] [Indexed: 12/13/2022]
Abstract
Ulcerative colitis (UC) is a chronic inflammatory disease characterized by diffused inflammation of the colon and rectum mucosa. The pathogenesis of UC is multifactorial, and the exact underlying mechanisms remain poorly understood. This study aims to investigate the effect of mesalazine and atorvastatin combination in enhancing anti-inflammatory effects and attenuates progression of oxazolone colitis in rats. In the present study, male albino rats (N = 60) were divided into six groups (10 rats each), the first two groups served as normal control and a control saline group. Colitis was induced by intra-rectal administration of oxazolone in the 5th and 7th days after pre-sensitization. Then, rats were divided into untreated group, groups treated with mesalazine or atorvastatin or their combination. Colitis was assessed by colon length, body weight, and incidence of diarrhea, rectal bleeding, and histopathology of colon tissue. Colon tissues were used for measuring interleukin 6 (IL-6), tumor necrosis factor alpha (TNF-α), IL-13, signal transducer and activator of transcription-3 (STAT-3), myeloperoxidase activity (MPO), reduced glutathione(GSH), and tissue expression of IL-10, tight junction protein zonula occludens (ZO-1), and caspase-3 genes. The combination therapy significantly attenuated progression of UC by decreasing incidence of diarrhea, rectal bleeding, weight loss, IL-13, IL-6, TNF-α, STAT-3, caspase-3, and MPO activity and significantly increased IL-10, ZO-1, colon length, and GSH content, and these effects were more superior to single drugs. These findings showed that combination therapy was able to ameliorate progression of UC and enhance anti-inflammatory effects possibly by restoring IL-10 and ZO-1 levels and limiting IL-6/STAT-3 trans-signaling.
Collapse
Affiliation(s)
- Nageh Ahmed El-Mahdy
- Faculty of Pharmacy, Department of Pharmacology and Toxicology, Tanta University, Tanta, 31527, Egypt
| | - Magda El-Sayed El-Sayad
- Faculty of Pharmacy, Department of Pharmacology and Toxicology, Tanta University, Tanta, 31527, Egypt
| | - Aya Hassan El-Kadem
- Faculty of Pharmacy, Department of Pharmacology and Toxicology, Tanta University, Tanta, 31527, Egypt
| | - Sally El-Sayed Abu-Risha
- Faculty of Pharmacy, Department of Pharmacology and Toxicology, Tanta University, Tanta, 31527, Egypt
| |
Collapse
|
102
|
Thilakasiri P, Huynh J, Poh AR, Tan CW, Nero TL, Tran K, Parslow AC, Afshar-Sterle S, Baloyan D, Hannan NJ, Buchert M, Scott AM, Griffin MD, Hollande F, Parker MW, Putoczki TL, Ernst M, Chand AL. Repurposing the selective estrogen receptor modulator bazedoxifene to suppress gastrointestinal cancer growth. EMBO Mol Med 2020; 11:emmm.201809539. [PMID: 30885958 PMCID: PMC6460354 DOI: 10.15252/emmm.201809539] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Excessive signaling through gp130, the shared receptor for the interleukin (IL)6 family of cytokines, is a common hallmark in solid malignancies and promotes their progression. Here, we established the in vivo utility of bazedoxifene, a steroid analog clinically approved for the treatment of osteoporosis, to suppress gp130‐dependent tumor growth of the gastrointestinal epithelium. Bazedoxifene administration reduced gastric tumor burden in gp130Y757F mice, where tumors arise exclusively through excessive gp130/STAT3 signaling in response to the IL6 family cytokine IL11. Likewise, in mouse models of sporadic colon and intestinal cancers, which arise from oncogenic mutations in the tumor suppressor gene Apc and the associated β‐catenin/canonical WNT pathway, bazedoxifene treatment reduces tumor burden. Consistent with the proposed orthogonal tumor‐promoting activity of IL11‐dependent gp130/STAT3 signaling, tumors of bazedoxifene‐treated Apc‐mutant mice retain excessive nuclear accumulation of β‐catenin and aberrant WNT pathway activation. Likewise, bazedoxifene treatment of human colon cancer cells harboring mutant APC did not reduce aberrant canonical WNT signaling, but suppressed IL11‐dependent STAT3 signaling. Our findings provide compelling proof of concept to support the repurposing of bazedoxifene for the treatment of gastrointestinal cancers in which IL11 plays a tumor‐promoting role.
Collapse
Affiliation(s)
- Pathum Thilakasiri
- Olivia Newton-John Cancer Research Institute, School of Cancer Medicine, La Trobe University, Heidelberg, Vic., Australia
| | - Jennifer Huynh
- Olivia Newton-John Cancer Research Institute, School of Cancer Medicine, La Trobe University, Heidelberg, Vic., Australia
| | - Ashleigh R Poh
- Olivia Newton-John Cancer Research Institute, School of Cancer Medicine, La Trobe University, Heidelberg, Vic., Australia
| | - Chin Wee Tan
- The Walter and Eliza Hall Institute, Melbourne, Vic., Australia
| | - Tracy L Nero
- ACRF Rational Drug Discovery Centre, St Vincent's Institute, Melbourne, Vic., Australia.,Department of Biochemistry and Molecular Biology, Bio21 Institute, University of Melbourne, Melbourne, Vic., Australia
| | - Kelly Tran
- Olivia Newton-John Cancer Research Institute, School of Cancer Medicine, La Trobe University, Heidelberg, Vic., Australia
| | - Adam C Parslow
- Olivia Newton-John Cancer Research Institute, School of Cancer Medicine, La Trobe University, Heidelberg, Vic., Australia.,Department of Molecular Imaging and Therapy, Austin Health, Melbourne, Vic., Australia
| | - Shoukat Afshar-Sterle
- Olivia Newton-John Cancer Research Institute, School of Cancer Medicine, La Trobe University, Heidelberg, Vic., Australia
| | - David Baloyan
- Olivia Newton-John Cancer Research Institute, School of Cancer Medicine, La Trobe University, Heidelberg, Vic., Australia
| | - Natalie J Hannan
- Department of Obstetrics and Gynaecology, University of Melbourne, Melbourne, Vic., Australia
| | - Michael Buchert
- Olivia Newton-John Cancer Research Institute, School of Cancer Medicine, La Trobe University, Heidelberg, Vic., Australia
| | - Andrew Mark Scott
- Olivia Newton-John Cancer Research Institute, School of Cancer Medicine, La Trobe University, Heidelberg, Vic., Australia.,Department of Molecular Imaging and Therapy, Austin Health, Melbourne, Vic., Australia.,Department of Medicine, University of Melbourne, Melbourne, Vic., Australia
| | - Michael Dw Griffin
- Department of Biochemistry and Molecular Biology, Bio21 Institute, University of Melbourne, Melbourne, Vic., Australia
| | - Frederic Hollande
- Department of Clinical Pathology, University of Melbourne Centre for Cancer Research, Victorian Comprehensive Cancer Centre, University of Melbourne, Melbourne, Vic., Australia
| | - Michael W Parker
- ACRF Rational Drug Discovery Centre, St Vincent's Institute, Melbourne, Vic., Australia.,Department of Biochemistry and Molecular Biology, Bio21 Institute, University of Melbourne, Melbourne, Vic., Australia
| | | | - Matthias Ernst
- Olivia Newton-John Cancer Research Institute, School of Cancer Medicine, La Trobe University, Heidelberg, Vic., Australia
| | - Ashwini L Chand
- Olivia Newton-John Cancer Research Institute, School of Cancer Medicine, La Trobe University, Heidelberg, Vic., Australia
| |
Collapse
|
103
|
Yu AI, Zhao L, Eaton KA, Ho S, Chen J, Poe S, Becker J, Gonzalez A, McKinstry D, Hasso M, Mendoza-Castrejon J, Whitfield J, Koumpouras C, Schloss PD, Martens EC, Chen GY. Gut Microbiota Modulate CD8 T Cell Responses to Influence Colitis-Associated Tumorigenesis. Cell Rep 2020; 31:107471. [PMID: 32268087 PMCID: PMC7934571 DOI: 10.1016/j.celrep.2020.03.035] [Citation(s) in RCA: 116] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 02/13/2020] [Accepted: 03/12/2020] [Indexed: 02/07/2023] Open
Abstract
There is increasing evidence that gut microbiome perturbations, also known as dysbiosis, can influence colorectal cancer development. To understand the mechanisms by which the gut microbiome modulates cancer susceptibility, we examine two wild-type mouse colonies with distinct gut microbial communities that develop significantly different tumor numbers using a mouse model of inflammation-associated tumorigenesis. We demonstrate that adaptive immune cells contribute to the different tumor susceptibilities associated with the two microbial communities. Mice that develop more tumors have increased colon lamina propria CD8+ IFNγ+ T cells before tumorigenesis but reduced CD8+ IFNγ+ T cells in tumors and adjacent tissues compared with mice that develop fewer tumors. Notably, intratumoral T cells in mice that develop more tumors exhibit increased exhaustion. Thus, these studies suggest that microbial dysbiosis can contribute to colon tumor susceptibility by hyperstimulating CD8 T cells to promote chronic inflammation and early T cell exhaustion, which can reduce anti-tumor immunity.
Collapse
Affiliation(s)
- Amy I Yu
- Graduate Program in Immunology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Lili Zhao
- Department of Biostatistics, University of Michigan, University of Michigan, Ann Arbor, MI 48109, USA
| | - Kathryn A Eaton
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Sharon Ho
- College of Literature, Science, and the Arts, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jiachen Chen
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Sara Poe
- Unit for Laboratory Animal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - James Becker
- Unit for Laboratory Animal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Allison Gonzalez
- College of Literature, Science, and the Arts, University of Michigan, Ann Arbor, MI 48109, USA
| | - Delaney McKinstry
- College of Literature, Science, and the Arts, University of Michigan, Ann Arbor, MI 48109, USA
| | - Muneer Hasso
- College of Literature, Science, and the Arts, University of Michigan, Ann Arbor, MI 48109, USA
| | | | - Joel Whitfield
- Cancer Center Immunology Core, University of Michigan, Ann Arbor, MI 48109, USA
| | - Charles Koumpouras
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Patrick D Schloss
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Eric C Martens
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Grace Y Chen
- Graduate Program in Immunology, University of Michigan, Ann Arbor, MI 48109, USA; Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
104
|
Morand S, Staats H, Creeden JF, Iqbal A, Kahaleh B, Stanbery L, Dworkin L, Nemunaitis J. Molecular mechanisms underlying rheumatoid arthritis and cancer development and treatment. Future Oncol 2020; 16:483-495. [PMID: 32100561 DOI: 10.2217/fon-2019-0722] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Given recent advances in cancer immune therapy, specifically use of checkpoint inhibitors, understanding the link between autoimmunity and cancer is essential. Rheumatoid arthritis (RA) affects about 1% of the population, and early diagnosis is key to prevent joint damage. Management consists of disease-modifying antirheumatic drugs that alter normal immunologic pathways, which could affect malignancy growth and survival. Prolonged immune dysregulation and the resulting inflammatory response associated with development of RA may also lead to increased cancer development risk. RA has long been associated with increased risk of non-Hodgkin's lymphoma [1] and further evidence supports relationship to lung cancer [2]. This review will address the mechanisms behind cancer development and progression in RA patients, biomarkers and assess cancer risk and early detection.
Collapse
Affiliation(s)
- Susan Morand
- Department of Medicine, University of Toledo College of Medicine & Life Sciences, Toledo, OH 43614, USA
| | - Hannah Staats
- Department of Medicine, University of Toledo College of Medicine & Life Sciences, Toledo, OH 43614, USA
| | - Justin Fortune Creeden
- Department of Medicine, University of Toledo College of Medicine & Life Sciences, Toledo, OH 43614, USA
| | - Azwar Iqbal
- Department of Medicine, University of Toledo College of Medicine & Life Sciences, Toledo, OH 43614, USA
| | - Bashar Kahaleh
- Department of Medicine, University of Toledo College of Medicine & Life Sciences, Toledo, OH 43614, USA
| | - Laura Stanbery
- Department of Medicine, University of Toledo College of Medicine & Life Sciences, Toledo, OH 43614, USA
| | - Lance Dworkin
- Department of Medicine, University of Toledo College of Medicine & Life Sciences, Toledo, OH 43614, USA
| | - John Nemunaitis
- Department of Medicine, University of Toledo College of Medicine & Life Sciences, Toledo, OH 43614, USA.,ProMedica Health System, Toledo, OH 43606, USA
| |
Collapse
|
105
|
Daou HN. Exercise as an anti-inflammatory therapy for cancer cachexia: a focus on interleukin-6 regulation. Am J Physiol Regul Integr Comp Physiol 2020; 318:R296-R310. [DOI: 10.1152/ajpregu.00147.2019] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cancer cachexia is a complicated disorder of extreme, progressive skeletal muscle wasting. It is directed by metabolic alterations and systemic inflammation dysregulation. Numerous studies have demonstrated that increased systemic inflammation promotes this type of cachexia and have suggested that cytokines are implicated in the skeletal muscle loss. Exercise is firmly established as an anti-inflammatory therapy that can attenuate or even reverse the process of muscle wasting in cancer cachexia. The interleukin IL-6 is generally considered to be a key player in the development of the microenvironment of malignancy; it promotes tumor growth and metastasis by acting as a bridge between chronic inflammation and cancerous tissue and it also induces skeletal muscle atrophy and protein breakdown. Paradoxically, a beneficial role for IL-6 has also been identified recently, and that is its status as a “founding member” of the myokine class of proteins. Skeletal muscle is an important source of circulating IL-6 in people who participate in exercise training. IL-6 acts as an anti-inflammatory myokine by inhibiting TNFα and improving glucose uptake through the stimulation of AMPK signaling. This review discusses the action of IL-6 in skeletal muscle tissue dysfunction and the role of IL-6 as an “exercise factor” that modulates the immune system. This review also sheds light on the main considerations related to the treatment of muscle wasting in cancer cachexia.
Collapse
|
106
|
Li C, Ma Y, Fei F, Zheng M, Li Z, Zhao Q, Du J, Liu K, Lu R, Zhang S. Critical role and its underlying molecular events of the plasminogen receptor, S100A10 in malignant tumor and non-tumor diseases. J Cancer 2020; 11:826-836. [PMID: 31949486 PMCID: PMC6959022 DOI: 10.7150/jca.36203] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 11/13/2019] [Indexed: 12/28/2022] Open
Abstract
S100A10 is a small molecular weight protein expressed in the cytoplasm of many cells and one of the members of the S100 protein family that binds calcium and forms the largest subgroup of EF-hand proteins. The regulatory processes of S100A10 are complicated. S100A10 participates in the regulation of a variety of tumor and non-tumor diseases through cascade reactions with multitudinous signaling molecules. In malignant tumors, such as acute promyelocytic leukemia (APL) and lung cancer, S100A10 is likely involved in their progression, including invasion and metastasis through the regulation of plasmin production and subsequent plasmin-dependent stimulation of other proteases, such as matrix metalloproteinase (MMP)-2 and -9. Both the plasmin and MMPs are capable of inducing degradation of the extracellular matrix (ECM) and basement membrane, which is a critical step for tumor progression. In non-tumor diseases, the distribution of S100A10 in the brain and its interaction with 5-hydroxytryptamine 1B (5-HT1B) receptor, an important mediator in the central nervous system that maintains a dynamic balance of the neurotransmitters, correlates with depression-like behavior. S100A10 also participates in inflammatory responses through the regulation of peripheral macrophage migration to the inflammatory sites, which depends on the generation of plasmin and other proteinases at the surface of macrophages. Considerable attention should be paid to understand the significant role of S100A10 in the modulation of malignant tumor and non-tumor diseases.
Collapse
Affiliation(s)
- Chunyuan Li
- Department of Pathology, Tianjin Union Medical Center, Tianjin, P.R. China
| | - Yi Ma
- Department of ophthalmology, Tianjin Union Medical Center, Tianjin, P.R. China
| | - Fei Fei
- Department of Pathology, Tianjin Union Medical Center, Tianjin, P.R. China
| | - Minying Zheng
- Department of Pathology, Tianjin Union Medical Center, Tianjin, P.R. China
| | - Zugui Li
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China
| | - Qi Zhao
- Tianjin Medical University, Tianjin, P.R. China
| | - Jiaxing Du
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China
| | - Kai Liu
- Tianjin Medical University, Tianjin, P.R. China
| | - Rui Lu
- Tianjin Medical University, Tianjin, P.R. China
| | - Shiwu Zhang
- Department of Pathology, Tianjin Union Medical Center, Tianjin, P.R. China
| |
Collapse
|
107
|
Peng M, Lee SH, Rahaman SO, Biswas D. Dietary probiotic and metabolites improve intestinal homeostasis and prevent colorectal cancer. Food Funct 2020; 11:10724-10735. [DOI: 10.1039/d0fo02652b] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Metabolites from Lactobacillus casei display substantial antioxidant and anti-inflammatory activities, inhibit colorectal cancer cell proliferation and growth, and modulate gut microfloral composition, specifically reducing sulfidogenic bacteria.
Collapse
Affiliation(s)
- Mengfei Peng
- Department of Animal and Avian Sciences
- University of Maryland
- College Park
- USA
- Biological Sciences Program
| | - Seong-Ho Lee
- Department of Nutrition and Food Science
- University of Maryland
- College Park
- USA
| | - Shaik O. Rahaman
- Department of Nutrition and Food Science
- University of Maryland
- College Park
- USA
| | - Debabrata Biswas
- Department of Animal and Avian Sciences
- University of Maryland
- College Park
- USA
- Biological Sciences Program
| |
Collapse
|
108
|
Abstract
Despite advanced clinical treatments, mortality in patients with metastatic colorectal cancer (CRC) remains high. Three critical determinants in CRC progression include the epithelial proliferation checkpoints, epithelial-to-mesenchymal transition (EMT) and inflammatory cytokines in the tumour microenvironment. Genes involved in these three processes are regulated at the transcriptional and post-transcriptional level. Recent studies revealed previously unappreciated roles of non-coding ribonucleic acids (ncRNAs) in modulating the proliferation checkpoints, EMT, and inflammatory gene expression in CRC. In this review, we will discuss the mechanisms underlying the roles of ncRNAs in CRC as well as examine future perspectives in this field. Better understanding of ncRNA biology will provide novel targets for future therapeutic development.
Collapse
Affiliation(s)
- Shengyun Ma
- Cellular and Molecular Medicine, University of California , San Diego, USA
| | - Tianyun Long
- Cellular and Molecular Medicine, University of California , San Diego, USA
| | | |
Collapse
|
109
|
Greten FR, Grivennikov SI. Inflammation and Cancer: Triggers, Mechanisms, and Consequences. Immunity 2019; 51:27-41. [PMID: 31315034 DOI: 10.1016/j.immuni.2019.06.025] [Citation(s) in RCA: 2367] [Impact Index Per Article: 394.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 06/21/2019] [Accepted: 06/25/2019] [Indexed: 02/07/2023]
Abstract
Inflammation predisposes to the development of cancer and promotes all stages of tumorigenesis. Cancer cells, as well as surrounding stromal and inflammatory cells, engage in well-orchestrated reciprocal interactions to form an inflammatory tumor microenvironment (TME). Cells within the TME are highly plastic, continuously changing their phenotypic and functional characteristics. Here, we review the origins of inflammation in tumors, and the mechanisms whereby inflammation drives tumor initiation, growth, progression, and metastasis. We discuss how tumor-promoting inflammation closely resembles inflammatory processes typically found during development, immunity, maintenance of tissue homeostasis, or tissue repair and illuminate the distinctions between tissue-protective and pro-tumorigenic inflammation, including spatiotemporal considerations. Defining the cornerstone rules of engagement governing molecular and cellular mechanisms of tumor-promoting inflammation will be essential for further development of anti-cancer therapies.
Collapse
Affiliation(s)
- Florian R Greten
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, 60596 Frankfurt/Main, Germany; Frankfurt Cancer Institute, Goethe University Frankfurt, 60596 Frankfurt/Main, Germany; German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
| | - Sergei I Grivennikov
- Cancer Prevention and Control Program, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA.
| |
Collapse
|
110
|
Chen J, Ye X, Pitmon E, Lu M, Wan J, Jellison ER, Adler AJ, Vella AT, Wang K. IL-17 inhibits CXCL9/10-mediated recruitment of CD8 + cytotoxic T cells and regulatory T cells to colorectal tumors. J Immunother Cancer 2019; 7:324. [PMID: 31775909 PMCID: PMC6880503 DOI: 10.1186/s40425-019-0757-z] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 09/23/2019] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND The IL-17 family cytokines are potent drivers of colorectal cancer (CRC) development. We and others have shown that IL-17 mainly signals to tumor cells to promote CRC, but the underlying mechanism remains unclear. IL-17 also dampens Th1-armed anti-tumor immunity, in part by attracting myeloid cells to tumor. Whether IL-17 controls the activity of adaptive immune cells in a more direct manner, however, is unknown. METHODS Using mouse models of sporadic or inducible colorectal cancers, we ablated IL-17RA in the whole body or specifically in colorectal tumor cells. We also performed adoptive bone marrow reconstitution to knockout CXCR3 in hematopoietic cells. Histological and immunological experimental methods were used to reveal the link among IL-17, chemokine production, and CRC development. RESULTS Loss of IL-17 signaling in mouse CRC resulted in marked increase in the recruitment of CD8+ cytotoxic T lymphocytes (CTLs) and regulatory T cells (Tregs), starting from early stage CRC lesions. This is accompanied by the increased expression of anti-inflammatory cytokines IL-10 and TGF-β. IL-17 signaling also inhibits the production of T cell attracting chemokines CXCL9 and CXCL10 by tumor cells. Conversely, the inability of hematopoietic cells to respond to CXCL9/10 resulted in decreased tumor infiltration by CTLs and Tregs, decreased levels of IL-10 and TGF-β, and increased numbers of tumor lesions. Blockade of IL-17 signaling resulted in increased expression of immune checkpoint markers. On the other hand, treatment of mouse CRC with anti-CTLA-4 antibody led to increased expression of pro-tumor IL-17. CONCLUSION IL-17 signals to colorectal tumor cells and inhibits their production of CXCL9/10 chemokines. By doing so, IL-17 inhibits the infiltration of CD8+ CTLs and Tregs to CRC, thus promoting CRC development. Cancer immunotherapy may be benefited by the use of anti-IL-17 agents as adjuvant therapies, which serve to block both IL-17-mediated tumor promotion and T cell exclusion.
Collapse
MESH Headings
- Animals
- Biomarkers, Tumor
- Cell Line, Tumor
- Chemokine CXCL10/genetics
- Chemokine CXCL10/metabolism
- Chemokine CXCL9/metabolism
- Colorectal Neoplasms/immunology
- Colorectal Neoplasms/metabolism
- Colorectal Neoplasms/pathology
- Colorectal Neoplasms/therapy
- Cytokines/biosynthesis
- Gene Expression
- Humans
- Interleukin-17/metabolism
- Lymphocytes, Tumor-Infiltrating/drug effects
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Mice
- Mice, Knockout
- Models, Biological
- Neoplasm Staging
- Signal Transduction/drug effects
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/metabolism
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
Collapse
Affiliation(s)
- Ju Chen
- Department of Immunology, School of Medicine, UConn Health, 263 Farmington Ave, Farmington, CT, 06030, USA
| | - Xiaoyang Ye
- Department of Immunology, School of Medicine, UConn Health, 263 Farmington Ave, Farmington, CT, 06030, USA
- Shenzhen Key Laboratory for Neuronal Structural Biology, Biomedical Research Institute, Shenzhen Peking University - The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Elise Pitmon
- Department of Immunology, School of Medicine, UConn Health, 263 Farmington Ave, Farmington, CT, 06030, USA
| | - Mengqian Lu
- Department of Immunology, School of Medicine, UConn Health, 263 Farmington Ave, Farmington, CT, 06030, USA
- School of Acupuncture-moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Jun Wan
- Shenzhen Key Laboratory for Neuronal Structural Biology, Biomedical Research Institute, Shenzhen Peking University - The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Evan R Jellison
- Department of Immunology, School of Medicine, UConn Health, 263 Farmington Ave, Farmington, CT, 06030, USA
| | - Adam J Adler
- Department of Immunology, School of Medicine, UConn Health, 263 Farmington Ave, Farmington, CT, 06030, USA
| | - Anthony T Vella
- Department of Immunology, School of Medicine, UConn Health, 263 Farmington Ave, Farmington, CT, 06030, USA
| | - Kepeng Wang
- Department of Immunology, School of Medicine, UConn Health, 263 Farmington Ave, Farmington, CT, 06030, USA.
| |
Collapse
|
111
|
Yin A, Luo Y, Chen W, He M, Deng JH, Zhao N, Cao L, Wang L. FAM96A Protects Mice From Dextran Sulfate Sodium (DSS)-Induced Colitis by Preventing Microbial Dysbiosis. Front Cell Infect Microbiol 2019; 9:381. [PMID: 31803631 PMCID: PMC6876263 DOI: 10.3389/fcimb.2019.00381] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 10/24/2019] [Indexed: 12/19/2022] Open
Abstract
Family with sequence similarity 96 member A (FAM96A) is an evolutionarily conserved intracellular protein that is involved in the maturation of the Fe/S protein, iron regulatory protein 1 (IRP1), and the mitochondria-related apoptosis of gastrointestinal stromal tumor cells. In this study, we used a mouse model of chemically induced colitis to investigate the physiological role of FAM96A in intestinal homeostasis and inflammation. At baseline, colons from Fam96a−/− mice exhibited microbial dysbiosis, dysregulated epithelial cell turnover, an increased number of goblet cells, and disordered tight junctions with functional deficits affecting intestinal permeability. After cohousing, the differences between wild-type and Fam96a−/− colons were abrogated, suggesting that FAM96A affects colonic epithelial cells in a microbiota-dependent manner. Fam96a deficiency in mice resulted in increased susceptibility to dextran sulfate sodium (DSS)-induced colitis. Importantly, the colitogenic activity of Fam96a−/− intestinal microbiota was transferable to wild-type littermate mice via fecal microbial transplantation (FMT), leading to exacerbation of DSS-induced colitis. Taken together, our data indicate that FAM96A helps to maintain colonic homeostasis and protect against DSS-induced colitis by preventing gut microbial dysbiosis. This study used gene knockout animals to help to understand the in vivo effects of the Fam96a gene for the first time and provides new evidence regarding host–microbiota interactions.
Collapse
Affiliation(s)
- Ang Yin
- Department of Immunology, Center for Human Disease Genomics, Health Science Center, School of Basic Medical Sciences, Peking University, Beijing, China.,Key Laboratory of Medical Immunology, School of Basic Medical Science, Peking University, Ministry of Health, Beijing, China
| | - Yang Luo
- Department of Immunology, Center for Human Disease Genomics, Health Science Center, School of Basic Medical Sciences, Peking University, Beijing, China.,Key Laboratory of Medical Immunology, School of Basic Medical Science, Peking University, Ministry of Health, Beijing, China
| | - Wei Chen
- Department of Immunology, Center for Human Disease Genomics, Health Science Center, School of Basic Medical Sciences, Peking University, Beijing, China.,Key Laboratory of Medical Immunology, School of Basic Medical Science, Peking University, Ministry of Health, Beijing, China
| | - Minwei He
- Department of Immunology, Center for Human Disease Genomics, Health Science Center, School of Basic Medical Sciences, Peking University, Beijing, China.,Key Laboratory of Medical Immunology, School of Basic Medical Science, Peking University, Ministry of Health, Beijing, China
| | - Jin Hai Deng
- Department of Immunology, Center for Human Disease Genomics, Health Science Center, School of Basic Medical Sciences, Peking University, Beijing, China.,Key Laboratory of Medical Immunology, School of Basic Medical Science, Peking University, Ministry of Health, Beijing, China
| | - Ning Zhao
- Department of Immunology, Center for Human Disease Genomics, Health Science Center, School of Basic Medical Sciences, Peking University, Beijing, China.,Key Laboratory of Medical Immunology, School of Basic Medical Science, Peking University, Ministry of Health, Beijing, China
| | - Lulu Cao
- Department of Immunology, Center for Human Disease Genomics, Health Science Center, School of Basic Medical Sciences, Peking University, Beijing, China.,Key Laboratory of Medical Immunology, School of Basic Medical Science, Peking University, Ministry of Health, Beijing, China
| | - Lu Wang
- Department of Immunology, Center for Human Disease Genomics, Health Science Center, School of Basic Medical Sciences, Peking University, Beijing, China.,Key Laboratory of Medical Immunology, School of Basic Medical Science, Peking University, Ministry of Health, Beijing, China
| |
Collapse
|
112
|
Thilakasiri PS, Dmello RS, Nero TL, Parker MW, Ernst M, Chand AL. Repurposing of drugs as STAT3 inhibitors for cancer therapy. Semin Cancer Biol 2019; 68:31-46. [PMID: 31711994 DOI: 10.1016/j.semcancer.2019.09.022] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 09/20/2019] [Accepted: 09/24/2019] [Indexed: 02/06/2023]
Abstract
Drug repurposing is a valuable approach in delivering new cancer therapeutics rapidly into the clinic. Existing safety and patient tolerability data for drugs already in clinical use represent an untapped resource in terms of identifying therapeutic agents for off-label protein targets. The multicellular effects of STAT3 mediated by a range of various upstream signaling pathways make it an attractive therapeutic target with utility in a range of diseases including cancer, and has led to the development of a variety of STAT3 inhibitors. Moreover, heightened STAT3 transcriptional activation in tumor cells and within the cells of the tumor microenvironment contribute to disease progression. Consequently, there are many STAT3 inhibitors in preclinical development or under evaluation in clinical trials for their therapeutic efficacy predominantly in inflammatory diseases and cancer. Despite these advances, many challenges remain in ultimately providing STAT3 inhibitors to patients as cancer treatments, highlighting the need not only for a better understanding of the mechanisms associated with STAT3 activation, but also how various pharmaceutical agents suppress STAT3 activity in various cancers. In this review we discuss the importance of STAT3-dependent functions in cancer, review the status of compounds designed as direct-acting STAT3 inhibitors, and describe some of the strategies for repurposing of drugs as STAT3 inhibitors for cancer therapy.
Collapse
Affiliation(s)
- Pathum S Thilakasiri
- Cancer and Inflammation Program, Olivia Newton-John Cancer Research Institute, School of Cancer Medicine, La Trobe University, Heidelberg, Vic., Australia
| | - Rhynelle S Dmello
- Cancer and Inflammation Program, Olivia Newton-John Cancer Research Institute, School of Cancer Medicine, La Trobe University, Heidelberg, Vic., Australia
| | - Tracy L Nero
- ACRF Rational Drug Discovery Centre, St Vincent's Institute, Melbourne, Vic., Australia; Department of Biochemistry and Molecular Biology, Bio21 Institute, University of Melbourne, Melbourne, Vic., Australia
| | - Michael W Parker
- ACRF Rational Drug Discovery Centre, St Vincent's Institute, Melbourne, Vic., Australia; Department of Biochemistry and Molecular Biology, Bio21 Institute, University of Melbourne, Melbourne, Vic., Australia
| | - Matthias Ernst
- Cancer and Inflammation Program, Olivia Newton-John Cancer Research Institute, School of Cancer Medicine, La Trobe University, Heidelberg, Vic., Australia
| | - Ashwini L Chand
- Cancer and Inflammation Program, Olivia Newton-John Cancer Research Institute, School of Cancer Medicine, La Trobe University, Heidelberg, Vic., Australia.
| |
Collapse
|
113
|
Koh SJ, Kim JW, Kim BG, Lee KL, Kim DW, Kim JS. Matricellular protein periostin promotes colitis-associated colon tumorigenesis in mice. Carcinogenesis 2019; 40:102-111. [PMID: 30204842 DOI: 10.1093/carcin/bgy120] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 08/27/2018] [Accepted: 09/07/2018] [Indexed: 12/28/2022] Open
Abstract
Periostin is expressed in inflamed colonic mucosa and colon cancer tissue; however, its role in the development of colitis-associated colon cancer (CAC) remains unclear. Wild-type and periostin-deficient (Postn-/-) mice were given a single intraperitoneal injection of azoxymethane at 12.5 mg/kg on day 0. Seven days later, 2% dextran sulfate sodium (DSS) was administered via drinking water for 5 days, followed by untreated, free water consumption for 16 days. This cycle was repeated three times. In vitro assays were performed using COLO205 and HCT116 cells. Small interfering RNA was used to inhibit Postn gene translation. Periostin expression was determined using colon samples from patients with CAC. Postn-/- mice exhibited lower tumor burden compared with wild-type mice. Exposure to azoxymethane/DSS resulted in extensive epithelial apoptosis in Postn-/- mice compared with that in wild-type mice. In addition, immunoreactivity for IκB kinase, β-catenin and COX2 was markedly reduced in Postn-/- mice. Expression of interleukin (IL)-1β and tumor necrosis factor α (TNF-α) significantly decreased, whereas that of IL-10 and transforming growth factor β (TGF-β) increased in peritoneal macrophages isolated from Postn-/- mice. Silencing of the Postn gene resulted in reduced cell viability, which was associated with caspase-3 activation, and this was reversed by treatment with recombinant periostin. Knockdown of Postn downregulated bcl-2, cIAP1, cFLIP-L, VEGF, Axin 2 and cyclin D1, and upregulated bak expression. Periostin expression was significantly increased in patients with CAC. Periostin aggravates CAC development, which suggests that periostin is a potential therapeutic target for the prevention of CAC in patients with inflammatory bowel disease.
Collapse
Affiliation(s)
- Seong-Joon Koh
- Department of Internal Medicine, Division of Gastroenteology, Seoul National University Boramae Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Ji Won Kim
- Department of Internal Medicine, Division of Gastroenteology, Seoul National University Boramae Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Byeong Gwan Kim
- Department of Internal Medicine, Division of Gastroenteology, Seoul National University Boramae Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Kook Lae Lee
- Department of Internal Medicine, Division of Gastroenteology, Seoul National University Boramae Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Dae Woo Kim
- Department of Otorhinolaryngology-Head and Neck Surgery, Seoul National University Boramae Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Joo Sung Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea.,Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
114
|
ADAM17 Activity and IL-6 Trans-Signaling in Inflammation and Cancer. Cancers (Basel) 2019; 11:cancers11111736. [PMID: 31694340 PMCID: PMC6895846 DOI: 10.3390/cancers11111736] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 10/25/2019] [Accepted: 11/02/2019] [Indexed: 02/08/2023] Open
Abstract
All ligands of the epidermal growth factor receptor (EGF-R) are transmembrane proteins, which need to be proteolytically cleaved in order to be systemically active. The major protease responsible for this cleavage is the membrane metalloprotease ADAM17, which also has been implicated in cleavage of TNFα and interleukin-6 (IL-6) receptor. It has been recently shown that in the absence of ADAM17, the main protease for EGF-R ligand processing, colon cancer formation is largely abrogated. Intriguingly, colon cancer formation depends on EGF-R activity on myeloid cells rather than on intestinal epithelial cells. A major activity of EGF-R on myeloid cells is the stimulation of IL-6 synthesis. Subsequently, IL-6 together with the ADAM17 shed soluble IL-6 receptor acts on intestinal epithelial cells via IL-6 trans-signaling to induce colon cancer formation, which can be blocked by the inhibitor of IL-6 trans-signaling, sgp130Fc. Blockade of IL-6 trans-signaling therefore offers a new therapeutic window downstream of the EGF-R for the treatment of colon cancer and possibly of other EGF-R related neoplastic diseases.
Collapse
|
115
|
Epithelial EP4 plays an essential role in maintaining homeostasis in colon. Sci Rep 2019; 9:15244. [PMID: 31645712 PMCID: PMC6811535 DOI: 10.1038/s41598-019-51639-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 10/04/2019] [Indexed: 12/15/2022] Open
Abstract
Colonic epithelial cells comprise the mucosal barrier, and their dysfunction promotes microbial invasion from the gut lumen and induces the development of intestinal inflammation. The EP4 receptor is known to mediate the protective effect of prostaglandin (PG) E2 in the gastrointestinal tract; however, the exact role of epithelial EP4 in intestinal pathophysiology remains unknown. In the present study, we aimed to investigate the role of epithelial EP4 in maintaining colonic homeostasis by characterizing the intestinal epithelial cell-specific EP4 knockout (EP4 cKO) mice. Mice harboring the epithelial EP4 deletion showed significantly lower colonic crypt depth and lower numbers of secretory cell lineages, as well as impaired epithelial cells in the colon. Interestingly, EP4-deficient colon epithelia showed a higher number of apoptotic cells. Consistent with the defect in mucosal barrier function of colonic epithelia and secretory cell lineages, EP4 cKO colon stroma showed enhanced immune cell infiltration, which was accompanied by increased production of inflammatory cytokines. Furthermore, EP4-deficient colons were susceptible to dextran sulfate sodium (DSS)-induced colitis. Our study is the first to demonstrate that epithelial EP4 loss resulted in potential “inflammatory” status under physiological conditions. These findings provided insights into the crucial role of epithelial PGE2/EP4 axis in maintaining intestinal homeostasis.
Collapse
|
116
|
Transforming Growth Factor-β Signaling in Immunity and Cancer. Immunity 2019; 50:924-940. [PMID: 30995507 DOI: 10.1016/j.immuni.2019.03.024] [Citation(s) in RCA: 1601] [Impact Index Per Article: 266.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 03/19/2019] [Accepted: 03/25/2019] [Indexed: 12/18/2022]
Abstract
Transforming growth factor (TGF)-β is a crucial enforcer of immune homeostasis and tolerance, inhibiting the expansion and function of many components of the immune system. Perturbations in TGF-β signaling underlie inflammatory diseases and promote tumor emergence. TGF-β is also central to immune suppression within the tumor microenvironment, and recent studies have revealed roles in tumor immune evasion and poor responses to cancer immunotherapy. Here, we present an overview of the complex biology of the TGF-β family and its context-dependent nature. Then, focusing on cancer, we discuss the roles of TGF-β signaling in distinct immune cell types and how this knowledge is being leveraged to unleash the immune system against the tumor.
Collapse
|
117
|
Aryappalli P, Shabbiri K, Masad RJ, Al-Marri RH, Haneefa SM, Mohamed YA, Arafat K, Attoub S, Cabral-Marques O, Ramadi KB, Fernandez-Cabezudo MJ, Al-Ramadi BK. Inhibition of Tyrosine-Phosphorylated STAT3 in Human Breast and Lung Cancer Cells by Manuka Honey is Mediated by Selective Antagonism of the IL-6 Receptor. Int J Mol Sci 2019; 20:E4340. [PMID: 31491838 PMCID: PMC6769459 DOI: 10.3390/ijms20184340] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 08/26/2019] [Accepted: 09/01/2019] [Indexed: 12/30/2022] Open
Abstract
Aberrantly high levels of tyrosine-phosphorylated signal transducer and activator of transcription 3 (p-STAT3) are found constitutively in ~50% of human lung and breast cancers, acting as an oncogenic transcription factor. We previously demonstrated that Manuka honey (MH) inhibits p-STAT3 in breast cancer cells, but the exact mechanism remained unknown. Herein, we show that MH-mediated inhibition of p-STAT3 in breast (MDA-MB-231) and lung (A549) cancer cell lines is accompanied by decreased levels of gp130 and p-JAK2, two upstream components of the IL-6 receptor (IL-6R) signaling pathway. Using an ELISA-based assay, we demonstrate that MH binds directly to IL-6Rα, significantly inhibiting (~60%) its binding to the IL-6 ligand. Importantly, no evidence of MH binding to two other cytokine receptors, IL-11Rα and IL-8R, was found. Moreover, MH did not alter the levels of tyrosine-phosphorylated or total Src family kinases, which are also constitutively activated in cancer cells, suggesting that signaling via other growth factor receptors is unaffected by MH. Binding of five major MH flavonoids (luteolin, quercetin, galangin, pinocembrin, and chrysin) was also tested, and all but pinocembrin could demonstrably bind IL-6Rα, partially (30-35%) blocking IL-6 binding at the highest concentration (50 μM) used. In agreement, each flavonoid inhibited p-STAT3 in a dose-dependent manner, with estimated IC50 values in the 3.5-70 μM range. Finally, docking analysis confirmed the capacity of each flavonoid to bind in an energetically favorable configuration to IL-6Rα at a site predicted to interfere with ligand binding. Taken together, our findings identify IL-6Rα as a direct target of MH and its flavonoids, highlighting IL-6R blockade as a mechanism for the anti-tumor activity of MH, as well as a viable therapeutic target in IL-6-dependent cancers.
Collapse
Affiliation(s)
- Priyanka Aryappalli
- Department of Medical Microbiology & Immunology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Khadija Shabbiri
- Department of Biochemistry, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Razan J Masad
- Department of Medical Microbiology & Immunology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Roadha H Al-Marri
- Department of Medical Microbiology & Immunology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Shoja M Haneefa
- Department of Medical Microbiology & Immunology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Yassir A Mohamed
- Department of Medical Microbiology & Immunology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Kholoud Arafat
- Department of Pharmacology & Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Samir Attoub
- Department of Pharmacology & Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Otavio Cabral-Marques
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP 05508-000, Brazil
| | - Khalil B Ramadi
- Harvard-MIT Health Sciences and Technology Division, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Maria J Fernandez-Cabezudo
- Department of Biochemistry, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates.
| | - Basel K Al-Ramadi
- Department of Medical Microbiology & Immunology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates.
| |
Collapse
|
118
|
Croes L, Fransen E, Hylebos M, Buys K, Hermans C, Broeckx G, Peeters M, Pauwels P, Op de Beeck K, Van Camp G. Determination of the Potential Tumor-Suppressive Effects of Gsdme in a Chemically Induced and in a Genetically Modified Intestinal Cancer Mouse Model. Cancers (Basel) 2019; 11:cancers11081214. [PMID: 31434357 PMCID: PMC6721630 DOI: 10.3390/cancers11081214] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 08/06/2019] [Accepted: 08/13/2019] [Indexed: 02/06/2023] Open
Abstract
Gasdermin E (GSDME), also known as deafness autosomal dominant 5 (DFNA5) and previously identified to be an inducer of regulated cell death, is frequently epigenetically inactivated in different cancer types, suggesting that GSDME is a tumor suppressor gene. In this study, we aimed to evaluate the tumor-suppressive effects of GSDME in two intestinal cancer mouse models. To mimic the silencing of GSDME by methylation as observed in human cancers, a Gsdme knockout (KO) mouse was developed. The effect of GSDME on tumorigenesis was studied both in a chemically induced and in a genetic intestinal cancer mouse model, as strong evidence shows that GSDME plays a role in human colorectal cancer and representative mouse models for intestinal cancer are available. Azoxymethane (AOM) was used to induce colorectal tumors in the chemically induced intestinal cancer model (n = 100). For the genetic intestinal cancer model, Apc1638N/+ mice were used (n = 37). In both experiments, the number of mice bearing microscopic proliferative lesions, the number and type of lesions per mouse and the histopathological features of the adenocarcinomas were compared between Gsdme KO and wild type (WT) mice. Unfortunately, we found no major differences between Gsdme KO and WT mice, neither for the number of affected mice nor for the multiplicity of proliferative lesions in the mice. However, recent breakthroughs on gasdermin function indicate that GSDME is an executioner of necrotic cell death. Therefore, it is possible that GSDME may be important for creating an inflammatory microenvironment around the tumor. This is in line with the trend towards more severe inflammation in WT compared to Gsdme KO mice, that we observed in our study. We conclude that the effect of GSDME in tumor biology is probably more subtle than previously thought.
Collapse
Affiliation(s)
- Lieselot Croes
- Center of Medical Genetics, University of Antwerp, Prins Boudewijnlaan 43/6, Edegem, BE-2650 Antwerp, Belgium
- Center for Oncological Research, University of Antwerp, Universiteitsplein 1, Wilrijk, BE-2610 Antwerp, Belgium
| | - Erik Fransen
- Center of Medical Genetics, University of Antwerp, Prins Boudewijnlaan 43/6, Edegem, BE-2650 Antwerp, Belgium
- StatUa Center for Statistics, University of Antwerp, Prinsstraat 13, BE-2000 Antwerp, Belgium
| | - Marieke Hylebos
- Center of Medical Genetics, University of Antwerp, Prins Boudewijnlaan 43/6, Edegem, BE-2650 Antwerp, Belgium
- Center for Oncological Research, University of Antwerp, Universiteitsplein 1, Wilrijk, BE-2610 Antwerp, Belgium
| | - Kimberly Buys
- Center of Medical Genetics, University of Antwerp, Prins Boudewijnlaan 43/6, Edegem, BE-2650 Antwerp, Belgium
- Center for Oncological Research, University of Antwerp, Universiteitsplein 1, Wilrijk, BE-2610 Antwerp, Belgium
| | - Christophe Hermans
- Center for Oncological Research, University of Antwerp, Universiteitsplein 1, Wilrijk, BE-2610 Antwerp, Belgium
| | - Glenn Broeckx
- Department of Pathology, Antwerp University Hospital, Wilrijkstraat 10, Edegem, BE-2650 Antwerp, Belgium
| | - Marc Peeters
- Center for Oncological Research, University of Antwerp, Universiteitsplein 1, Wilrijk, BE-2610 Antwerp, Belgium
- Department of Medical Oncology, Antwerp University Hospital, Wilrijkstraat 10, Edegem, BE-2650 Antwerp, Belgium
| | - Patrick Pauwels
- Center for Oncological Research, University of Antwerp, Universiteitsplein 1, Wilrijk, BE-2610 Antwerp, Belgium
- Department of Pathology, Antwerp University Hospital, Wilrijkstraat 10, Edegem, BE-2650 Antwerp, Belgium
| | - Ken Op de Beeck
- Center of Medical Genetics, University of Antwerp, Prins Boudewijnlaan 43/6, Edegem, BE-2650 Antwerp, Belgium
- Center for Oncological Research, University of Antwerp, Universiteitsplein 1, Wilrijk, BE-2610 Antwerp, Belgium
| | - Guy Van Camp
- Center of Medical Genetics, University of Antwerp, Prins Boudewijnlaan 43/6, Edegem, BE-2650 Antwerp, Belgium.
- Center for Oncological Research, University of Antwerp, Universiteitsplein 1, Wilrijk, BE-2610 Antwerp, Belgium.
| |
Collapse
|
119
|
Wang D, Zheng X, Fu B, Nian Z, Qian Y, Sun R, Tian Z, Wei H. Hepatectomy promotes recurrence of liver cancer by enhancing IL-11-STAT3 signaling. EBioMedicine 2019; 46:119-132. [PMID: 31375423 PMCID: PMC6711863 DOI: 10.1016/j.ebiom.2019.07.058] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 07/18/2019] [Accepted: 07/22/2019] [Indexed: 02/07/2023] Open
Abstract
Background Patients undergoing surgical resection of hepatocellular carcinoma (HCC) are at risk of recurrence; however, the underlying mechanism remains poorly understood. Methods Through the analysis of gene expression profiles in tumour and matched normal tissues from patients with hepatocellular carcinoma (HCC), we identified differences in interleukin-11 (IL-11) expression. Further, we used genetic mouse, orthotopic tumour, chemically induced, and orthotopic allograft models to study the correlation between IL-11 and postsurgical recurrence. Additionally, we conducted a series of experiments, including histology and immunohistochemistry analysis, three-dimensional culture, immunofluorescence, western blotting, enzyme-linked immunosorbent assay (ELISA) and flow cytometry to investigate the role of IL-11-signal transducer and activator of transcription 3 (STAT3) signaling in HCC recurrence. Findings We demonstrate that IL-11 levels increase after surgery, triggering HCC outgrowth. Accordingly, pharmacological blocking of IL-11-STAT3 signaling in model systems significantly alleviates tumour cell proliferation and suppresses postsurgical recurrence of HCC tumours. Interpretation These data demonstrate that IL-11 has a central role in postsurgical HCC recurrence, and that inhibition of IL-11-STAT3 signaling is a potential therapeutic strategy to prevent recurrence. Fund Natural Science Foundation of China.
Collapse
Affiliation(s)
- Dongyao Wang
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China; Institute of Immunology, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Xiaohu Zheng
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China; Institute of Immunology, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Binqing Fu
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China; Institute of Immunology, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Zhigang Nian
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China; Institute of Immunology, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Yeben Qian
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230027, China
| | - Rui Sun
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China; Institute of Immunology, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Zhigang Tian
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China; Institute of Immunology, University of Science and Technology of China, Hefei, Anhui 230027, China.
| | - Haiming Wei
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China; Institute of Immunology, University of Science and Technology of China, Hefei, Anhui 230027, China.
| |
Collapse
|
120
|
Targeting immune cell circuits and trafficking in inflammatory bowel disease. Nat Immunol 2019; 20:970-979. [PMID: 31235952 DOI: 10.1038/s41590-019-0415-0] [Citation(s) in RCA: 450] [Impact Index Per Article: 75.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 05/01/2019] [Indexed: 12/20/2022]
Abstract
Inflammatory bowel diseases (IBDs) such as Crohn's disease and ulcerative colitis are characterized by uncontrolled activation of intestinal immune cells in a genetically susceptible host. Due to the progressive and destructive nature of the inflammatory process in IBD, complications such as fibrosis, stenosis or cancer are frequently observed, which highlights the need for effective anti-inflammatory therapy. Studies have identified altered trafficking of immune cells and pathogenic immune cell circuits as crucial drivers of mucosal inflammation and tissue destruction in IBD. A defective gut barrier and microbial dysbiosis induce such accumulation and local activation of immune cells, which results in a pro-inflammatory cytokine loop that overrides anti-inflammatory signals and causes chronic intestinal inflammation. This Review discusses pathogenic cytokine responses of immune cells as well as immune cell trafficking as a rational basis for new translational therapies in IBD.
Collapse
|
121
|
Liu YT, Xu L, Bennett L, Hooks JC, Liu J, Zhou Q, Liem P, Zheng Y, Skapek SX. Identification of De Novo Enhancers Activated by TGFβ to Drive Expression of CDKN2A and B in HeLa Cells. Mol Cancer Res 2019; 17:1854-1866. [PMID: 31189690 DOI: 10.1158/1541-7786.mcr-19-0289] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 05/01/2019] [Accepted: 06/07/2019] [Indexed: 02/06/2023]
Abstract
Disruption of the CDKN2A (INK4A/ARF) and B (INK4B) genes, which encode three function-independent tumor suppressors, is one of the most common events in human cancer. Because their relative importance in tumor prevention appears to be species- and context-specific, studying their regulation can shed light on mechanisms by which they are bypassed in malignant transformation. We previously unveiled a new pathway in which TGFβ selectively induces Arf at mouse Cdkn2a in eye development and cultured fibroblasts. As TGFβ signaling is often derailed in cancer development or progression, we investigated its control of CDKN2A/B in human cancer. Computational analyses of sequencing and array data from nearly 11,000 patients with cancer in TCGA showed discordant expression of ARF and INK4A in most cancer subtypes, with gene copy-number loss and promoter methylation involved in only a subset. Using HeLa cells as a model, we found that exogenous TGFβ induced ARF mRNA and protein, and ARF knockdown limited TGFβ-mediated growth suppression. TGFβ-mediated ARF mRNA induction required SMAD2/3, p38MAPK, and SP1, and ARF mRNA was induced without added RNAPII recruitment. Chromatin immunoprecipitation unveiled a remote enhancer element engaged by TGFβ by a mechanism that partially depended on p38MAPK. CRISPR-based editing of this enhancer limited induction of ARF and INK4B by TGFβ, but not by oncogenic RAS. IMPLICATIONS: Our findings reveal new molecular mechanisms by which CDKN2A/B regulation is coupled to external cues, and those findings represent entry points to further explore pharmacologic strategies to restore their expression in cancer.
Collapse
Affiliation(s)
- Yen-Ting Liu
- Division of Hematology/Oncology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Lin Xu
- Division of Hematology/Oncology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas.,Quantitative Biomedical Research Center, Department of Clinical Sciences, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Lynda Bennett
- Division of Hematology/Oncology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Jared C Hooks
- Division of Hematology/Oncology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Jing Liu
- Division of Hematology/Oncology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Qinbo Zhou
- Quantitative Biomedical Research Center, Department of Clinical Sciences, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Priscilla Liem
- Division of Hematology/Oncology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Yanbin Zheng
- Division of Hematology/Oncology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Stephen X Skapek
- Division of Hematology/Oncology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas. .,Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
122
|
Walker WH, Borniger JC. Molecular Mechanisms of Cancer-Induced Sleep Disruption. Int J Mol Sci 2019; 20:E2780. [PMID: 31174326 PMCID: PMC6600154 DOI: 10.3390/ijms20112780] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 06/03/2019] [Accepted: 06/04/2019] [Indexed: 02/07/2023] Open
Abstract
Sleep is essential for health. Indeed, poor sleep is consistently linked to the development of systemic disease, including depression, metabolic syndrome, and cognitive impairments. Further evidence has accumulated suggesting the role of sleep in cancer initiation and progression (primarily breast cancer). Indeed, patients with cancer and cancer survivors frequently experience poor sleep, manifesting as insomnia, circadian misalignment, hypersomnia, somnolence syndrome, hot flushes, and nightmares. These problems are associated with a reduction in the patients' quality of life and increased mortality. Due to the heterogeneity among cancers, treatment regimens, patient populations and lifestyle factors, the etiology of cancer-induced sleep disruption is largely unknown. Here, we discuss recent advances in understanding the pathways linking cancer and the brain and how this leads to altered sleep patterns. We describe a conceptual framework where tumors disrupt normal homeostatic processes, resulting in aberrant changes in physiology and behavior that are detrimental to health. Finally, we discuss how this knowledge can be leveraged to develop novel therapeutic approaches for cancer-associated sleep disruption, with special emphasis on host-tumor interactions.
Collapse
Affiliation(s)
- William H Walker
- Department of Neuroscience, West Virginia University, Morgantown, WV 26506, USA.
| | - Jeremy C Borniger
- Department of Psychiatry & Behavioral Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
123
|
Li ZW, Sun B, Gong T, Guo S, Zhang J, Wang J, Sugawara A, Jiang M, Yan J, Gurary A, Zheng X, Gao B, Xiao SY, Chen W, Ma C, Farrar C, Zhu C, Chan OTM, Xin C, Winnicki A, Winnicki J, Tang M, Park R, Winnicki M, Diener K, Wang Z, Liu Q, Chu CH, Arter ZL, Yue P, Alpert L, Hui GS, Fei P, Turkson J, Yang W, Wu G, Tao A, Ramos JW, Moisyadi S, Holcombe RF, Jia W, Birnbaumer L, Zhou X, Chu WM. GNAI1 and GNAI3 Reduce Colitis-Associated Tumorigenesis in Mice by Blocking IL6 Signaling and Down-regulating Expression of GNAI2. Gastroenterology 2019; 156:2297-2312. [PMID: 30836096 PMCID: PMC6628260 DOI: 10.1053/j.gastro.2019.02.040] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 02/06/2019] [Accepted: 02/28/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS Interleukin 6 (IL6) and tumor necrosis factor contribute to the development of colitis-associated cancer (CAC). We investigated these signaling pathways and the involvement of G protein subunit alpha i1 (GNAI1), GNAI2, and GNAI3 in the development of CAC in mice and humans. METHODS B6;129 wild-type (control) or mice with disruption of Gnai1, Gnai2, and/or Gnai3 or conditional disruption of Gnai2 in CD11c+ or epithelial cells were given dextran sulfate sodium (DSS) to induce colitis followed by azoxymethane (AOM) to induce carcinogenesis; some mice were given an antibody against IL6. Feces were collected from mice, and the compositions of microbiomes were analyzed by polymerase chain reactions. Dendritic cells (DCs) and myeloid-derived suppressor cells (MDSCs) isolated from spleen and colon tissues were analyzed by flow cytometry. We performed immunoprecipitation and immunoblot analyses of colon tumor tissues, MDSCs, and mouse embryonic fibroblasts to study the expression levels of GNAI1, GNAI2, and GNAI3 and the interactions of GNAI1 and GNAI3 with proteins in the IL6 signaling pathway. We analyzed the expression of Gnai2 messenger RNA by CD11c+ cells in the colonic lamina propria by PrimeFlow, expression of IL6 in DCs by flow cytometry, and secretion of cytokines in sera and colon tissues by enzyme-linked immunosorbent assay. We obtained colon tumor and matched nontumor tissues from 83 patients with colorectal cancer having surgery in China and 35 patients with CAC in the United States. Mouse and human colon tissues were analyzed by histology, immunoblot, immunohistochemistry, and/or RNA-sequencing analyses. RESULTS GNAI1 and GNAI3 (GNAI1;3) double-knockout (DKO) mice developed more severe colitis after administration of DSS and significantly more colonic tumors than control mice after administration of AOM plus DSS. Development of increased tumors in DKO mice was not associated with changes in fecal microbiomes but was associated with activation of nuclear factor (NF) κB and signal transducer and activator of transcription (STAT) 3; increased levels of GNAI2, nitric oxide synthase 2, and IL6; increased numbers of CD4+ DCs and MDSCs; and decreased numbers of CD8+ DCs. IL6 was mainly produced by CD4+/CD11b+, but not CD8+, DCs in DKO mice. Injection of DKO mice with a blocking antibody against IL6 reduced the expansion of MDSCs and the number of tumors that developed after CAC induction. Incubation of MDSCs or mouse embryonic fibroblasts with IL6 induced activation of either NF-κB by a JAK2-TRAF6-TAK1-CHUK/IKKB signaling pathway or STAT3 by JAK2. This activation resulted in expression of GNAI2, IL6 signal transducer (IL6ST, also called GP130) and nitric oxide synthase 2, and expansion of MDSCs; the expression levels of these proteins and expansion of MDSCs were further increased by the absence of GNAI1;3 in cells and mice. Conditional disruption of Gnai2 in CD11c+ cells of DKO mice prevented activation of NF-κB and STAT3 and changes in numbers of DCs and MDSCs. Colon tumor tissues from patients with CAC had reduced levels of GNAI1 and GNAI3 and increased levels of GNAI2 compared with normal tissues. Further analysis of a public human colorectal tumor DNA microarray database (GSE39582) showed that low Gani1 and Gnai3 messenger RNA expression and high Gnai2 messenger RNA expression were significantly associated with decreased relapse-free survival. CONCLUSIONS GNAI1;3 suppresses DSS-plus-AOM-induced colon tumor development in mice, whereas expression of GNAI2 in CD11c+ cells and IL6 in CD4+/CD11b+ DCs appears to promote these effects. Strategies to induce GNAI1;3, or block GNAI2 and IL6, might be developed for the prevention or therapy of CAC in patients.
Collapse
Affiliation(s)
- Zhi-Wei Li
- Cancer Biology Program, University of Hawaii Cancer Center, Honolulu, Hawaii
| | - Beicheng Sun
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Ting Gong
- Cancer Biology Program, University of Hawaii Cancer Center, Honolulu, Hawaii
| | - Sheng Guo
- Cancer Biology Program, University of Hawaii Cancer Center, Honolulu, Hawaii; Department of Endocrine, Genetics and Metabolism, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Jianhua Zhang
- Cancer Biology Program, University of Hawaii Cancer Center, Honolulu, Hawaii; Department of Pediatrics, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Junlong Wang
- Cancer Biology Program, University of Hawaii Cancer Center, Honolulu, Hawaii
| | - Atsushi Sugawara
- Institute for Biogenesis Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii
| | - Meisheng Jiang
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, California
| | - Junjun Yan
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Alexandra Gurary
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii
| | - Xin Zheng
- Cancer Biology Program, University of Hawaii Cancer Center, Honolulu, Hawaii
| | - Bifeng Gao
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Shu-Yuan Xiao
- Department of Pathology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China; Department of Pathology, University of Chicago, Chicago, Illinois
| | - Wenlian Chen
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, Hawaii
| | - Chi Ma
- Cancer Biology Program, University of Hawaii Cancer Center, Honolulu, Hawaii
| | - Christine Farrar
- The Microscopy, Imaging, and Flow Cytometry Shared Resource, University of Hawaii Cancer Center, Honolulu, Hawaii
| | - Chenjun Zhu
- Cancer Biology Program, University of Hawaii Cancer Center, Honolulu, Hawaii
| | - Owen T M Chan
- Pathology Core, University of Hawaii Cancer Center, Honolulu, Hawaii
| | - Can Xin
- Cancer Biology Program, University of Hawaii Cancer Center, Honolulu, Hawaii
| | - Andrew Winnicki
- Cancer Biology Program, University of Hawaii Cancer Center, Honolulu, Hawaii
| | - John Winnicki
- Cancer Biology Program, University of Hawaii Cancer Center, Honolulu, Hawaii
| | - Mingxin Tang
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii
| | - Ryan Park
- Cancer Biology Program, University of Hawaii Cancer Center, Honolulu, Hawaii
| | - Mary Winnicki
- Cancer Biology Program, University of Hawaii Cancer Center, Honolulu, Hawaii
| | - Katrina Diener
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Zhanwei Wang
- Cancer Biology Program, University of Hawaii Cancer Center, Honolulu, Hawaii
| | - Qicai Liu
- Cancer Biology Program, University of Hawaii Cancer Center, Honolulu, Hawaii; Department of Cardiology and Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Catherine H Chu
- Cancer Biology Program, University of Hawaii Cancer Center, Honolulu, Hawaii
| | - Zhaohui L Arter
- Cancer Biology Program, University of Hawaii Cancer Center, Honolulu, Hawaii
| | - Peibin Yue
- Cancer Biology Program, University of Hawaii Cancer Center, Honolulu, Hawaii
| | - Lindsay Alpert
- Department of Pathology, University of Chicago, Chicago, Illinois
| | - George S Hui
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii
| | - Peiwen Fei
- Cancer Biology Program, University of Hawaii Cancer Center, Honolulu, Hawaii
| | - James Turkson
- Cancer Biology Program, University of Hawaii Cancer Center, Honolulu, Hawaii
| | - Wentian Yang
- Department of Orthopedics, Rhode Island Hospital, Brown University Alpert Medical School, Providence, Rhode Island
| | - Guangyu Wu
- Department of Pharmacology and Toxicology, Augusta University, Augusta, Georgia
| | - Ailin Tao
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy and Clinical Immunology, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Joe W Ramos
- Cancer Biology Program, University of Hawaii Cancer Center, Honolulu, Hawaii
| | - Stefan Moisyadi
- Institute for Biogenesis Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii
| | - Randall F Holcombe
- Cancer Biology Program, University of Hawaii Cancer Center, Honolulu, Hawaii
| | - Wei Jia
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, Hawaii
| | - Lutz Birnbaumer
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina; Institute for Biomedical Research (BIOMED), Universidad Católica Argentina, Buenos Aires, Argentina
| | - Xiqiao Zhou
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Wen-Ming Chu
- Cancer Biology Program, University of Hawaii Cancer Center, Honolulu, Hawaii; The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy and Clinical Immunology, Guangzhou Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
124
|
Avci AB, Feist E, Burmester GR. Targeting IL-6 or IL-6 Receptor in Rheumatoid Arthritis: What's the Difference? BioDrugs 2019; 32:531-546. [PMID: 30488231 DOI: 10.1007/s40259-018-0320-3] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Interleukin-6 (IL-6) signaling is a critical target in inflammatory pathways. Today, tocilizumab (TCZ) and sarilumab (SAR), two IL-6 receptor-inhibiting monoclonal antibodies, are widely used in the treatment of rheumatoid arthritis (RA), with a favorable efficacy/safety profile. Successful introduction of such agents in the treatment of RA has encouraged the development of other agents targeting different points of the pathway. Sirukumab (SRK), a human anti-IL-6 monoclonal antibody, has been evaluated in clinical trials and showed largely similar clinical efficacy compared with TCZ and other IL-6 pathway-targeting agents. Furthermore, the drug safety profile seemed to reflect the profile of adverse effects and laboratory abnormalities seen in other inhibitors of the IL-6 pathway. However, increased death rates under SRK treatment compared with placebo raised safety concerns, which led to the decision by the FDA to decline the approval of SRK in August 2017. However, during the 18-week true placebo-controlled period, mortality rates were identical in the placebo- and SRK-treated patients. Comparisons after week 18 may be confounded by some factors, and also the 'crossover' design resulted in various treatment groups with varying drug exposure periods. The limited placebo exposure relative to SRK exposure makes interpretation of mortality rates difficult. We do not know whether the imbalance in mortality rates seen for SRK is a true safety signal or a result of bias due to the study design. Therefore, further long-term clinical data as well as basic research is needed to allow deeper insight into IL-6 signaling.
Collapse
MESH Headings
- Antibodies, Monoclonal/pharmacology
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Monoclonal, Humanized/pharmacology
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antirheumatic Agents/pharmacology
- Antirheumatic Agents/therapeutic use
- Arthritis, Rheumatoid/drug therapy
- Arthritis, Rheumatoid/immunology
- Arthritis, Rheumatoid/mortality
- Clinical Trials as Topic
- Drug Approval
- Humans
- Interleukin-6/antagonists & inhibitors
- Interleukin-6/immunology
- Receptors, Interleukin-6/antagonists & inhibitors
- Receptors, Interleukin-6/immunology
- Signal Transduction/drug effects
- Signal Transduction/immunology
- Survival Rate
- Treatment Outcome
Collapse
Affiliation(s)
- Ali Berkant Avci
- Department of Internal Medicine, Rheumatology, Life Hospital, Antalya, Turkey
| | - Eugen Feist
- Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin, Berlin, Germany.
| | - Gerd Rüdiger Burmester
- Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin, Berlin, Germany
| |
Collapse
|
125
|
Degos C, Heinemann M, Barrou J, Boucherit N, Lambaudie E, Savina A, Gorvel L, Olive D. Endometrial Tumor Microenvironment Alters Human NK Cell Recruitment, and Resident NK Cell Phenotype and Function. Front Immunol 2019; 10:877. [PMID: 31105699 PMCID: PMC6498896 DOI: 10.3389/fimmu.2019.00877] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 04/05/2019] [Indexed: 12/21/2022] Open
Abstract
Endometrial Cancer is the most common cancer in the female genital tract in developed countries, and with its increasing incidence due to risk factors such as aging and obesity tends to become a public health issue. However, its immune environment has been less characterized than in other tumors such as breast cancers. NK cells are cytotoxic innate lymphoid cells that are considered as a major anti-tumoral effector cell type which function is drastically altered in tumors which participates to tumor progression. Here we characterize tumor NK cells both phenotypically and functionally in the tumor microenvironment of endometrial cancer. For that, we gathered endometrial tumors, tumor adjacent healthy tissue, blood from matching patients and healthy donor blood to perform comparative analysis of NK cells. First we found that NK cells were impoverished in the tumor infiltrate. We then compared the phenotype of NK cells in the tumor and found that tumor resident CD103+ NK cells exhibited more co-inhibitory molecules such as Tigit, and TIM-3 compared to recruited CD103− NK cells and that the expression of these molecules increased with the severity of the disease. We showed that both chemokines (CXCL12, IP-10, and CCL27) and cytokines profiles (IL-1β and IL-6) were altered in the tumor microenvironment and might reduce NK cell function and recruitment to the tumor site. This led to hypothesize that the tumor microenvironment reduces resident NK cells cytotoxicity which we confirmed by measuring cytotoxic effector production and degranulation. Taken together, our results show that the tumor microenvironment reshapes NK cell phenotype and function to promote tumor progression.
Collapse
Affiliation(s)
- Clara Degos
- Tumor Immunology Team, IBISA Immunomonitoring Platform, Cancer Research Center of Marseillle, INSERM U1068, CNRS U7258, Institut Paoli-Calmettes, Aix-Marseille University, Marseille, France
| | - Mellie Heinemann
- Department of Surgical Oncology 2, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Aix-Marseille University, Marseille, France
| | - Julien Barrou
- Department of Surgical Oncology 2, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Aix-Marseille University, Marseille, France
| | - Nicolas Boucherit
- Tumor Immunology Team, IBISA Immunomonitoring Platform, Cancer Research Center of Marseillle, INSERM U1068, CNRS U7258, Institut Paoli-Calmettes, Aix-Marseille University, Marseille, France
| | - Eric Lambaudie
- Department of Surgical Oncology 2, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Aix-Marseille University, Marseille, France
| | | | - Laurent Gorvel
- Tumor Immunology Team, IBISA Immunomonitoring Platform, Cancer Research Center of Marseillle, INSERM U1068, CNRS U7258, Institut Paoli-Calmettes, Aix-Marseille University, Marseille, France
| | - Daniel Olive
- Tumor Immunology Team, IBISA Immunomonitoring Platform, Cancer Research Center of Marseillle, INSERM U1068, CNRS U7258, Institut Paoli-Calmettes, Aix-Marseille University, Marseille, France
| |
Collapse
|
126
|
Horst K, Höfler J, Martin L, Greven J, Schürholz T, Simon TP, Marx G, Hildebrand F. Geriatric Polytrauma-Cardiovascular and Immunologic Response in a Murine Two-Hit Model of Trauma. J Surg Res 2019; 241:87-94. [PMID: 31018170 DOI: 10.1016/j.jss.2019.03.053] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Revised: 03/07/2019] [Accepted: 03/22/2019] [Indexed: 02/03/2023]
Abstract
BACKGROUND The aims of the present study were to establish a clinically relevant two-hit model with trauma/hemorrhage followed by sepsis in older mice and investigate age-dependent cardiovascular and immunologic specificities under these conditions. MATERIALS AND METHODS In aged mice (12, 18, and 24 mo old), a femur fracture followed by hemorrhage was induced. After resuscitation, animals were monitored for 72 h before sepsis was induced. Vital signs were monitored during shock. Systemic interleukin (IL)-6 levels were measured daily. Expression of sarcoplasmic or endoplasmic reticulum calcium ATPase (SERCA) and IL-6 receptor were analyzed in heart, lung, and liver tissues. RESULTS After induction of shock, mean arterial pressure decreased significantly in all groups (12 mo, P < 0.001; 18 mo, P < 0.001; 24 mo, P = 0.013). Compared with younger animals, 24-mo old mice were not able to adequately compensate for hypovolemia by an increase of heart rate (P = 0.711). Expression of SERCA2 (P = 0.002) and IL-6 receptor on myocytes (P = 0.037), lung (P = 0.005), and liver (P = 0.009) tissues were also lowest in this group. Systemic IL-6 values showed the most distinct posttraumatic response in 24-mo-old mice (P = 0.016). Survival rate decreased significantly with increased age (P = 0.005). CONCLUSIONS The increased mortality rate in older animals was associated with a limited compensatory physiological response and a more distinct immunologic reaction after trauma and sepsis. A decreased SERCA2 expression and missing feedback loops due to a reduced density of organ bound immune receptors might represent possible explanations for the observed age-dependent differences.
Collapse
Affiliation(s)
- Klemens Horst
- Department of Orthopaedic Trauma, RWTH Aachen University, Aachen, Germany.
| | - Johannes Höfler
- Department of Orthopaedic Trauma, RWTH Aachen University, Aachen, Germany
| | - Lukas Martin
- Department of Intensive Care and Intermediate Care, RWTH Aachen University, Aachen, Germany
| | - Johannes Greven
- Department of Orthopaedic Trauma, RWTH Aachen University, Aachen, Germany
| | - Tobias Schürholz
- Department of Anaesthesia and Intensive Care, University of Rostock, Rostock, Germany
| | - Tim P Simon
- Department of Intensive Care and Intermediate Care, RWTH Aachen University, Aachen, Germany
| | - Gernot Marx
- Department of Intensive Care and Intermediate Care, RWTH Aachen University, Aachen, Germany
| | - Frank Hildebrand
- Department of Orthopaedic Trauma, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
127
|
Grill JI, Neumann J, Ofner A, Marschall MK, Zierahn H, Herbst A, Wolf E, Kolligs FT. Dro1/Ccdc80 inactivation promotes AOM/DSS-induced colorectal carcinogenesis and aggravates colitis by DSS in mice. Carcinogenesis 2019; 39:1176-1184. [PMID: 29901779 DOI: 10.1093/carcin/bgy077] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 06/11/2018] [Indexed: 12/17/2022] Open
Abstract
Colorectal carcinogenesis is a progressive multistep process involving the sequential accumulation of genetic alterations in tumor suppressor genes and oncogenes. Downregulated by oncogenes 1 (Dro1/Ccdc80) has been shown to be a potent tumor suppressor of colorectal carcinogenesis in the genetic ApcMin/+ mouse model. In ApcMin/+ mice, loss of DRO1 strongly increases colonic tumor multiplicity and leads to the regular formation of adenocarcinoma in the colon. To investigate DRO1's role in chemically induced as well as inflammation-associated colorectal carcinogenesis, the effect of Dro1 inactivation was studied in mice subjected to the carcinogen azoxymethane (AOM) and upon combined treatment with AOM and the proinflammatory agent dextran sodium sulfate (DSS), respectively. Loss of DRO1 increases multiplicity of preneoplastic aberrant crypt foci and colonic tumors upon administration of AOM. Combined treatment with AOM and DSS leads to increased colonic tumor number and promotes formation of adenocarcinoma in the colon. Moreover, Dro1 inactivation aggravates histological signs of acute and chronic DSS-induced colitis, strongly enlarges the size of ulcerative lesions in the intestinal lining, and exacerbates clinical signs and morbidity by DSS. Our results demonstrate DRO1 to be a strong tumor suppressor in the chemically induced colon carcinogenic mouse model. Additionally, we demonstrate DRO1 to inhibit colitis-associated colon cancer formation and uncover a novel putative role for DRO1 in inflammatory bowel disease.
Collapse
Affiliation(s)
- Jessica I Grill
- Department of Medicine II, University of Munich, Munich, Germany.,Institute of Molecular Animal Breeding and Biotechnology, Gene Center, Munich, Germany
| | - Jens Neumann
- Institute of Pathology, University of Munich, Munich, Germany
| | - Andrea Ofner
- Department of Medicine II, University of Munich, Munich, Germany
| | | | - Heike Zierahn
- Institute of Molecular Animal Breeding and Biotechnology, Gene Center, Munich, Germany
| | - Andreas Herbst
- Department of Medicine II, University of Munich, Munich, Germany
| | - Eckhard Wolf
- Institute of Molecular Animal Breeding and Biotechnology, Gene Center, Munich, Germany
| | - Frank T Kolligs
- Department of Medicine II, University of Munich, Munich, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Internal Medicine and Gastroenterology, HELIOS Klinikum Berlin-Buch, Berlin, Germany
| |
Collapse
|
128
|
Dawidowska M, Jaksik R, Drobna M, Szarzyńska-Zawadzka B, Kosmalska M, Sędek Ł, Machowska L, Lalik A, Lejman M, Ussowicz M, Kałwak K, Kowalczyk JR, Szczepański T, Witt M. Comprehensive Investigation of miRNome Identifies Novel Candidate miRNA-mRNA Interactions Implicated in T-Cell Acute Lymphoblastic Leukemia. Neoplasia 2019; 21:294-310. [PMID: 30763910 PMCID: PMC6372882 DOI: 10.1016/j.neo.2019.01.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 01/15/2019] [Accepted: 01/17/2019] [Indexed: 02/08/2023]
Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive malignancy originating from T-cell precursors. The genetic landscape of T-ALL has been largely characterized by next-generation sequencing. Yet, the transcriptome of miRNAs (miRNome) of T-ALL has been less extensively studied. Using small RNA sequencing, we characterized the miRNome of 34 pediatric T-ALL samples, including the expression of isomiRs and the identification of candidate novel miRNAs (not previously annotated in miRBase). For the first time, we show that immunophenotypic subtypes of T-ALL present different miRNA expression profiles. To extend miRNome characteristics in T-ALL (to 82 T-ALL cases), we combined our small RNA-seq results with data available in Gene Expression Omnibus. We report on miRNAs most abundantly expressed in pediatric T-ALL and miRNAs differentially expressed in T-ALL versus normal mature T-lymphocytes and thymocytes, representing candidate oncogenic and tumor suppressor miRNAs. Using eight target prediction algorithms and pathway enrichment analysis, we identified differentially expressed miRNAs and their predicted targets implicated in processes (defined in Gene Ontology and Kyoto Encyclopedia of Genes and Genomes) of potential importance in pathogenesis of T-ALL, including interleukin-6-mediated signaling, mTOR signaling, and regulation of apoptosis. We finally focused on hsa-mir-106a-363 cluster and functionally validated direct interactions of hsa-miR-20b-5p and hsa-miR-363-3p with 3' untranslated regions of their predicted targets (PTEN, SOS1, LATS2), overrepresented in regulation of apoptosis. hsa-mir-106a-363 is a paralogue of prototypic oncogenic hsa-mir-17-92 cluster with yet unestablished role in the pathogenesis of T-ALL. Our study provides a firm basis and data resource for functional analyses on the role of miRNA-mRNA interactions in T-ALL.
Collapse
Key Words
- all, acute lymphoblastic leukemia
- egil, european group for immunological classification of leukemias
- geo, gene expression omnibus
- go, gene ontology
- isomir, isoform of mirna
- kegg, kyoto encyclopedia of genes and genomes
- mirnome, transcriptome of mirnas
- mre, mirna response element
- or, odds ratio
- rt-qpcr, quantitative reverse transcription polymerase chain reaction
- small rna-seq, next-generation sequencing of small rnas
- t-all, t-cell acute lymphoblastic leukemia
- 3′utr, 3′ untranslated region
Collapse
Affiliation(s)
- Małgorzata Dawidowska
- Institute of Human Genetics, Polish Academy of Sciences, Strzeszyńska 32, 60-479 Poznań, Poland.
| | - Roman Jaksik
- Silesian University of Technology, Akademicka 16, 44-100 Gliwice, Poland.
| | - Monika Drobna
- Institute of Human Genetics, Polish Academy of Sciences, Strzeszyńska 32, 60-479 Poznań, Poland.
| | - Bronisława Szarzyńska-Zawadzka
- Institute of Human Genetics, Polish Academy of Sciences, Strzeszyńska 32, 60-479 Poznań, Poland; Institute of Human Genetics, Polish Academy of Sciences, Strzeszyńska 32, 60-479 Poznań, Poland.
| | - Maria Kosmalska
- Institute of Human Genetics, Polish Academy of Sciences, Strzeszyńska 32, 60-479 Poznań, Poland.
| | - Łukasz Sędek
- Department of Microbiology and Immunology, Medical University of Silesia in Katowice, Jordana 19, 41-808 Zabrze, Poland.
| | - Ludomiła Machowska
- Clinic of Pediatric Oncology Hematology and Transplantology, Poznań University of Medical Sciences, Szpitalna 27/33, 60-572 Poznań, Poland.
| | - Anna Lalik
- Silesian University of Technology, Akademicka 16, 44-100 Gliwice, Poland.
| | - Monika Lejman
- Laboratory of Genetic Diagnostics, Medical University of Lublin, Children's University Hospital, Gębali 6, 20-093 Lublin, Poland.
| | - Marek Ussowicz
- Department of Pediatric Bone Marrow Transplantation, Oncology, and Hematology, Wroclaw Medical University, Borowska 213, 50-556 Wroclaw, Poland.
| | - Krzysztof Kałwak
- Department of Pediatric Bone Marrow Transplantation, Oncology, and Hematology, Wroclaw Medical University, Borowska 213, 50-556 Wroclaw, Poland.
| | - Jerzy R Kowalczyk
- Department of Pediatric Hematology, Oncology and Transplantology, Medical University of Lublin, Gębali 6, 20-093 Lublin, Poland.
| | - Tomasz Szczepański
- Department of Pediatric Hematology and Oncology, Zabrze, Medical University of Silesia in Katowice, 3 Maja 13-15, 41-800 Zabrze, Poland.
| | - Michał Witt
- Institute of Human Genetics, Polish Academy of Sciences, Strzeszyńska 32, 60-479 Poznań, Poland.
| |
Collapse
|
129
|
Cancer-induced inflammation and inflammation-induced cancer in colon: a role for S1P lyase. Oncogene 2019; 38:4788-4803. [PMID: 30816345 DOI: 10.1038/s41388-019-0758-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 01/03/2019] [Accepted: 01/05/2019] [Indexed: 12/14/2022]
Abstract
A role of sphingolipids for inflammatory bowel disease and cancer is evident. However, the relative and separate contribution of sphingolipid deterioration in inflammation versus carcinogenesis for the pathophysiology of colitis-associated colon cancer (CAC) was unknown and therefore examined in this study. We performed isogenic bone marrow transplantation of inducible sphingosine-1-phosphate (S1P) lyase knockout mice to specifically modulate sphingolipids and associated genes and proteins in a compartment-specific way in a DSS/AOM mediated CAC model. 3D organoid cultures were used in vitro. S1P lyase (SGPL1) knockout in either immune cells or tissue, caused local sphingolipid accumulation leading to a dichotomic development of CAC: Immune cell SGPL1 knockout (I-SGPL-/-) augmented massive immune cell infiltration initiating colitis with lesions and calprotectin increase. Pathological crypt remodeling plus extracellular S1P-signaling caused delayed tumor formation characterized by S1P receptor 1, STAT3 mRNA increase, as well as programmed cell death ligand 1 expression, accompanied by a putatively counter regulatory STAT1S727 phosphorylation. In contrast, tissue SGPL1 knockout (T-SGPL-/-) provoked immediate occurrence of epithelial-driven tumors with upregulated sphingosine kinase 1, S1P receptor 2 and epidermal growth factor receptor. Here, progressing carcinogenesis was accompanied by an IL-12 to IL-23 shift with a consecutive development of a Th2/GATA3-driven, tumor-favoring microenvironment. Moreover, the knockout models showed distinct lymphopenia and neutrophilia, different from the full SGPL1 knockout. This study shows that depending on the initiating cellular S1P source, the pathophysiology of inflammation-induced cancer versus cancer-induced inflammation develops through separate, discernible molecular steps.
Collapse
|
130
|
Jones SA, Takeuchi T, Aletaha D, Smolen J, Choy EH, McInnes I. Interleukin 6: The biology behind the therapy. ACTA ACUST UNITED AC 2019. [DOI: 10.1136/conmed-2018-000005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The cytokine interleukin (IL)−6 performs a diverse portfolio of functions in normal physiology and disease. These functions extend beyond the typical role for an inflammatory cytokine, and IL-6 often displays hormone-like properties that affect metabolic processes associated with lipid metabolism, insulin resistance, and the neuroendocrine system. Consequently, the biology of IL-6 is complex. Recent advances in the field have led to novel interpretations of how IL-6 delivers immune homeostasis in health and yet drives disease pathology during infection, autoimmunity, and cancer. Various biological drugs that target IL-6 are in clinical practice or emerging in clinical trials and pre-clinical development programmes. The challenge is knowing how and when to apply these therapies. In this review, we will explore the biology behind IL-6 directed therapies and identify some key hurdles for future investigation.
Collapse
|
131
|
Neurath MF. IL-23 in inflammatory bowel diseases and colon cancer. Cytokine Growth Factor Rev 2018; 45:1-8. [PMID: 30563755 DOI: 10.1016/j.cytogfr.2018.12.002] [Citation(s) in RCA: 145] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 12/11/2018] [Indexed: 02/07/2023]
Abstract
Studies in recent years have identified a pivotal role of the cytokine IL-23 in the pathogenesis of inflammatory bowel diseases (IBD: Crohn´s disease, ulcerative colitis) and colitis-associated colon cancer. Genetic studies revealed that subgroups of IBD patients have single nucleotide polymorphisms in the IL-23R gene suggesting that IL-23R signaling affects disease susceptibility. Furthermore, increased production of IL-23 by macrophages, dendritic cells or granulocytes has been observed in various mouse models of colitis, colitis-associated cancer and IBD patients. Moreover, in several murine models of colitis, suppression of IL-12/IL-23 p40, IL-23 p19 or IL-23R function led to marked suppression of gut inflammation. This finding was associated with reduced activation of IL-23 target cells such as T helper 17 cells, innate lymphoid cells type 3, granulocytes and natural killer cells as well as with impaired production of proinflammatory cytokines. Based on these findings, targeting of IL-23 emerges as important concept for suppression of gut inflammation and inflammation-associated cancer growth. Consistently, neutralizing antibodies against IL-12/IL-23 p40 and IL-23 p19 have been successfully used in clinical trials for therapy of Crohn´s disease and pilot studies in ulcerative colitis are ongoing. These findings underline the crucial regulatory role of IL-23 in chronic intestinal inflammation and colitis-associated cancer and indicate that therapeutic strategies aiming at IL-23 blockade may be of key relevance for future therapy of IBD patients.
Collapse
Affiliation(s)
- Markus F Neurath
- Department of Medicine 1, University of Erlangen-Nürnberg, Kussmaul Research Campus & Ludwig Demling Endoscopy Center of Excellence, Erlangen, Germany; Deutsches Zentrum für Immuntherapie (DZI), University of Erlangen-Nürnberg, Germany.
| |
Collapse
|
132
|
Öner MG, Rokavec M, Kaller M, Bouznad N, Horst D, Kirchner T, Hermeking H. Combined Inactivation of TP53 and MIR34A Promotes Colorectal Cancer Development and Progression in Mice Via Increasing Levels of IL6R and PAI1. Gastroenterology 2018; 155:1868-1882. [PMID: 30099074 DOI: 10.1053/j.gastro.2018.08.011] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Revised: 07/30/2018] [Accepted: 08/03/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Combined inactivation of the microRNA 34a gene (MIR34A, by methylation) and the TP53 gene (by mutation or deletion) is observed in 50% of colorectal tumors that progress to distant metastases. We studied mice with intestinal disruption of Mir34a and Tp53 to investigate mechanisms of colorectal carcinogenesis and identify strategies to block these processes. METHODS Mice with disruption of Mir34a and/or Tp53 specifically in intestinal epithelial cells (IECs) (Mir34aΔIEC mice, Tp53ΔIEC mice, and Mir34aΔIEC/Tp53ΔIEC mice) and controls (Mir34aFl/Fl/Tp53Fl/Fl) were given azoxymethane to induce colorectal carcinogenesis. Some mice were given intraperitoneal injections of an antibody against mouse interleukin 6 receptor (IL6R), or received an inhibitor of PAI1 (tiplaxtinin) in their chow. Intestinal tissues were collected and analyzed by immunohistochemistry; gene expression profiles were analyzed by RNA sequencing. We determined the expression and localization of PAI1 in 61 human primary colon cancers and compared them to MIR34A methylation and inactivating mutations in TP53. Data on mRNA levels, methylation, and clinical features of 628 colon and rectal adenocarcinomas were obtained from The Cancer Genome Atlas portal. RESULTS Mir34aΔIEC/Tp53ΔIEC mice developed larger and more colorectal tumors, with increased invasion of surrounding tissue and metastasis to lymph nodes, than control mice or mice with disruption of either gene alone. Cells in tumors from the Mir34aΔIEC/Tp53ΔIEC mice had decreased apoptosis and increased proliferation compared to tumor cells from control mice, and expressed higher levels of genes, that regulate inflammation (including Il6r and Stat3) and epithelial-mesenchymal transition. The gene expression pattern of the tumors from Mir34aΔIEC/Tp53ΔIEC mice was similar to that of human colorectal tumor consensus molecular subtype 4 (mesenchymal, invasive). We identified the Pai1 messenger RNA as a target of Mir34a; levels of PAI1 protein were increased in primary colon cancer samples, that displayed methylation of MIR34A and mutational inactivation of TP53. Administration of tiplaxtinin or anti-IL6R antibody to Mir34aΔIEC/Tp53ΔIEC mice decreased proliferation of cancer cells, and reduced colorectal tumor invasion and metastasis. CONCLUSIONS In mice, we demonstrated that combined inactivation of Mir34a and Tp53 promotes azoxymethane-induced colorectal carcinogenesis and tumor progression and metastasis by increasing levels of IL6R and PAI1. Strategies to inhibit these processes might be developed to slow progression of colorectal cancer.
Collapse
Affiliation(s)
- Meryem Gülfem Öner
- Experimental and Molecular Pathology, Institute of Pathology, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Matjaz Rokavec
- Experimental and Molecular Pathology, Institute of Pathology, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Markus Kaller
- Experimental and Molecular Pathology, Institute of Pathology, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Nassim Bouznad
- Experimental and Molecular Pathology, Institute of Pathology, Ludwig-Maximilians-University Munich, Munich, Germany
| | - David Horst
- Institute of Pathology, Ludwig-Maximilians-University Munich, Munich, Germany; Institute of Pathology, Charité-Universitätsmedizin Berlin, Berlin, Germany; German Cancer Consortium, Heidelberg, Germany; German Cancer Research Center, Heidelberg, Germany
| | - Thomas Kirchner
- Institute of Pathology, Ludwig-Maximilians-University Munich, Munich, Germany; German Cancer Consortium, Heidelberg, Germany; German Cancer Research Center, Heidelberg, Germany
| | - Heiko Hermeking
- Experimental and Molecular Pathology, Institute of Pathology, Ludwig-Maximilians-University Munich, Munich, Germany; German Cancer Consortium, Heidelberg, Germany; German Cancer Research Center, Heidelberg, Germany.
| |
Collapse
|
133
|
Mathies F, Steffens N, Kleinschmidt D, Stuhlmann F, Huber FJ, Roy U, Meyer T, Luetgehetmann M, von Petersdorff M, Seiz O, Herkel J, Schramm C, Flavell RA, Gagliani N, Krebs C, Panzer U, Abdullah Z, Strowig T, Bedke T, Huber S. Colitis Promotes a Pathological Condition of the Liver in the Absence of Foxp3 + Regulatory T Cells. THE JOURNAL OF IMMUNOLOGY 2018; 201:3558-3568. [PMID: 30446566 DOI: 10.4049/jimmunol.1800711] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 10/16/2018] [Indexed: 02/06/2023]
Abstract
Inflammatory bowel disease is associated with extraintestinal diseases such as primary sclerosing cholangitis in the liver. Interestingly, it is known that an imbalance between Foxp3+ regulatory T cells (Treg) and Th17 cells is involved in inflammatory bowel disease and also in primary sclerosing cholangitis. To explain these associations, one hypothesis is that intestinal inflammation and barrier defects promote liver disease because of the influx of bacteria and inflammatory cells to the liver. However, whether and how this is linked to the Treg and Th17 cell imbalance is unclear. To address this, we used dextran sodium sulfate (DSS) and T cell transfer colitis mouse models. We analyzed the pathological conditions of the intestine and liver on histological, cellular, and molecular levels. We observed bacterial translocation and an influx of inflammatory cells, in particular Th17 cells, to the liver during colitis. In the DSS colitis model, in which Treg were concomitantly increased in the liver, we did not observe an overt pathological condition of the liver. In contrast, the T cell-mediated colitis model, in which Treg are not abundant, was associated with marked liver inflammation and a pathological condition. Of note, upon depletion of Treg in DEREG mice, DSS colitis promotes accumulation of Th17 cells and a pathological condition of the liver. Finally, we studied immune cell migration using KAEDE mice and found that some of these cells had migrated directly from the inflamed intestine into the liver. Overall, these data indicate that colitis can promote a pathological condition of the liver and highlight an important role of Treg in controlling colitis-associated liver inflammation.
Collapse
Affiliation(s)
- Franziska Mathies
- I. Medizinische Klinik und Poliklinik, Universitätsklinikum Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Niklas Steffens
- I. Medizinische Klinik und Poliklinik, Universitätsklinikum Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Doerte Kleinschmidt
- I. Medizinische Klinik und Poliklinik, Universitätsklinikum Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Friederike Stuhlmann
- I. Medizinische Klinik und Poliklinik, Universitätsklinikum Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Francis J Huber
- I. Medizinische Klinik und Poliklinik, Universitätsklinikum Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Urmi Roy
- Microbial Immune Regulation Research Group, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Thomas Meyer
- Institut für Medizinische Mikrobiologie, Virologie und Hygiene, Universitätsklinikum Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Marc Luetgehetmann
- Institut für Medizinische Mikrobiologie, Virologie und Hygiene, Universitätsklinikum Hamburg-Eppendorf, 20246 Hamburg, Germany
| | | | - Oliver Seiz
- I. Medizinische Klinik und Poliklinik, Universitätsklinikum Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Johannes Herkel
- I. Medizinische Klinik und Poliklinik, Universitätsklinikum Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Christoph Schramm
- I. Medizinische Klinik und Poliklinik, Universitätsklinikum Hamburg-Eppendorf, 20246 Hamburg, Germany.,Martin Zeitz Centrum für Seltene Erkrankungen, Universitätsklinikum Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Richard A Flavell
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520.,Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06520
| | - Nicola Gagliani
- I. Medizinische Klinik und Poliklinik, Universitätsklinikum Hamburg-Eppendorf, 20246 Hamburg, Germany.,Klinik und Poliklinik für Allgemein-, Viszeral- und Thoraxchirurgie, Universitätsklinikum Hamburg-Eppendorf, 20246 Hamburg, Germany.,Immunology and Allergy Unit, Department of Medicine Solna, Karolinska Institute, 17176 Stockholm, Sweden
| | - Christian Krebs
- III. Medizinische Klinik und Poliklinik, Universitätsklinikum Hamburg-Eppendorf, 20246 Hamburg, Germany; and
| | - Ulf Panzer
- III. Medizinische Klinik und Poliklinik, Universitätsklinikum Hamburg-Eppendorf, 20246 Hamburg, Germany; and
| | - Zeinab Abdullah
- Institute of Experimental Immunology, University Hospital Bonn, 53127 Bonn, Germany
| | - Till Strowig
- Microbial Immune Regulation Research Group, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Tanja Bedke
- I. Medizinische Klinik und Poliklinik, Universitätsklinikum Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Samuel Huber
- I. Medizinische Klinik und Poliklinik, Universitätsklinikum Hamburg-Eppendorf, 20246 Hamburg, Germany;
| |
Collapse
|
134
|
Ruder B, Murtadak V, Stürzl M, Wirtz S, Distler U, Tenzer S, Mahapatro M, Greten FR, Hu Y, Neurath MF, Cesarman E, Ballon G, Günther C, Becker C. Chronic intestinal inflammation in mice expressing viral Flip in epithelial cells. Mucosal Immunol 2018; 11:1621-1629. [PMID: 30104627 PMCID: PMC8063487 DOI: 10.1038/s41385-018-0068-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 07/04/2018] [Accepted: 07/10/2018] [Indexed: 02/04/2023]
Abstract
Viruses are present in the intestinal microflora and are currently discussed as a potential causative mechanism for the development of inflammatory bowel disease. A number of viruses, such as Human Herpesvirus-8, express homologs to cellular FLIPs, which are major contributors for the regulation of epithelial cell death. In this study we analyzed the consequences of constitutive expression of HHV8-viral FLIP in intestinal epithelial cells (IECs) in mice. Surprisingly, expression of vFlip disrupts tissue homeostasis and induces severe intestinal inflammation. Moreover vFlipIEC-tg mice showed reduced Paneth cell numbers, associated with excessive necrotic cell death. On a molecular level vFlip expression altered classical and alternative NFκB activation. Blocking of alternative NFκB signaling by deletion of Ikka in vivo largely protected mice from inflammation and Paneth cell loss induced by vFLIP. Collectively, our data provide functional evidence that expression of a single viral protein in IECs can be sufficient to disrupt epithelial homeostasis and to initiate chronic intestinal inflammation.
Collapse
Affiliation(s)
- Barbara Ruder
- Department of Medicine 1, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Vinay Murtadak
- Division of Molecular and Experimental Surgery, Department of Surgery, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Michael Stürzl
- Division of Molecular and Experimental Surgery, Department of Surgery, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Stefan Wirtz
- Department of Medicine 1, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Ute Distler
- Institute for Immunology, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Stefan Tenzer
- Institute for Immunology, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Mousumi Mahapatro
- Department of Medicine 1, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Florian R. Greten
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt am Main, Germany
| | - Yinling Hu
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - Markus F. Neurath
- Department of Medicine 1, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Ethel Cesarman
- Department of Pathology and Laboratory Medicine, Weill Medical College of Cornell University, New York, NY, USA
| | - Gianna Ballon
- Department of Pathology and Laboratory Medicine, Northwell Health, Lake Success, NY, USA
| | - Claudia Günther
- Department of Medicine 1, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Christoph Becker
- Department of Medicine 1, University of Erlangen-Nuremberg, Erlangen, Germany
| |
Collapse
|
135
|
Chen L, Jiang B, Zhong C, Guo J, Zhang L, Mu T, Zhang Q, Bi X. Chemoprevention of colorectal cancer by black raspberry anthocyanins involved the modulation of gut microbiota and SFRP2 demethylation. Carcinogenesis 2018; 39:471-481. [PMID: 29361151 DOI: 10.1093/carcin/bgy009] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2017] [Accepted: 01/13/2018] [Indexed: 12/12/2022] Open
Abstract
Freeze-dried black raspberry (BRB) powder is considered as a potential cancer chemopreventive agent. In this study, we fed azoxymethane (AOM)/dextran sodium sulfate (DSS)-treated C57BL/6J mice with a diet containing BRB anthocyanins for 12 weeks, and this led to a reduction in colon carcinogenesis. These animals had consistently lower tumor multiplicity compared with AOM/DSS-treated mice not receiving BRB anthocyanins. In AOM/DSS-treated mice, the number of pathogenic bacteria, including Desulfovibrio sp. and Enterococcus spp., was increased significantly, whereas probiotics such as Eubacterium rectale, Faecalibacterium prausnitzii and Lactobacillus were dramatically decreased, but BRB anthocyanins supplement could reverse this imbalance in gut microbiota. BRB anthocyanins also caused the demethylation of the SFRP2 gene promoter, resulting in increased expression of SFRP2, both at the mRNA and protein levels. Furthermore, the expression levels of DNMT31 and DNMT3B, as well as of p-STAT3 were downregulated by BRB anthocyanins in these animals. Taken together, these results suggested that BRB anthocyanins could modulate the composition of gut commensal microbiota, and changes in inflammation and the methylation status of the SFRP2 gene may play a central role in the chemoprevention of CRC.
Collapse
Affiliation(s)
- Lili Chen
- Department of Biotechnology, College of Life Science, Liaoning University, Shenyang, China
| | - Bowen Jiang
- Department of Biotechnology, College of Life Science, Liaoning University, Shenyang, China
| | - Chunge Zhong
- Department of Biotechnology, College of Life Science, Liaoning University, Shenyang, China
| | - Jun Guo
- Department of Biotechnology, College of Life Science, Liaoning University, Shenyang, China
| | - Lihao Zhang
- Department of Biotechnology, College of Life Science, Liaoning University, Shenyang, China
| | - Teng Mu
- Department of Biotechnology, College of Life Science, Liaoning University, Shenyang, China
| | - Qiuhua Zhang
- Department of Pharmacology, Liaoning University of Traditional Chinese Medicine, Shenyang, PR China
| | - Xiuli Bi
- Department of Biotechnology, College of Life Science, Liaoning University, Shenyang, China.,Department of Biotechnology, Research Center for Computer Simulating and Information Processing of Bio-macromolecules of Liaoning Province, Liaoning University, Shenyang, China
| |
Collapse
|
136
|
NLRP3 inflammasome in colitis and colitis-associated colorectal cancer. Mamm Genome 2018; 29:817-830. [DOI: 10.1007/s00335-018-9783-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 09/04/2018] [Indexed: 12/21/2022]
|
137
|
Xue M, Shi L, Wang W, Chen S, Wang L. An Overview of Molecular Profiles in Ulcerative Colitis-Related Cancer. Inflamm Bowel Dis 2018; 24:1883-1894. [PMID: 29945208 DOI: 10.1093/ibd/izy221] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Indexed: 12/16/2022]
Abstract
Ulcerative colitis (UC) is an independent risk factor of colorectal cancer (CRC). Both genetic and epigentic events induce a unique molecular profile during the development from UC to UC-related CRC (UCRC). These molecular changes play varied roles in DNA repair, immune response, cell metabolism, and interaction with the microbiota during the carcinogenesis process. This review will systmatically discuss the molecular characteristics of UCRC and point out the future perspectives in this research field.
Collapse
Affiliation(s)
- Meng Xue
- Department of Gastroenterology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.,Institute of Gastroenterology, Zhejiang University, Hangzhou, Zhejiang, China
| | - Liuhong Shi
- Department of Ultrasound, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Weijia Wang
- Department of Cardiology, School of Medicine, the Johns Hopkins Hospital, Baltimore, Maryland
| | - Shujie Chen
- Department of Gastroenterology, Sir Runrun Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Liangjing Wang
- Department of Gastroenterology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.,Institute of Gastroenterology, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
138
|
Stanilova S, Stanilov N, Julianov A, Manolova I, Miteva L. Transforming growth factor-β1 gene promoter -509C/T polymorphism in association with expression affects colorectal cancer development and depends on gender. PLoS One 2018; 13:e0201775. [PMID: 30071009 PMCID: PMC6072135 DOI: 10.1371/journal.pone.0201775] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 07/20/2018] [Indexed: 12/12/2022] Open
Abstract
It is widely known that sporadic colorectal cancer (CRC) is age-related diseases with higher incidence rate among men. Transforming growth factor-β1 (TGF-β1) is a major immune regulatory cytokine with a great impact and dual role in gastrointestinal carcinogenesis. In this context, the aim of the study was to explore the role of circulating TGF-β1 and the -509C/T functional promoter polymorphism (rs1800469) within the TGF-β1 gene (TGFB1) in the susceptibility, progression, and prognosis of CRC among Bulgarian male and female patients. Patients with sporadic CRC and healthy controls were genotyped by polymerase-chain reaction–restriction fragment length polymorphism. Serum TGF-β1 levels before and after curative surgery were determined by ELISA. Total RNA was extracted from paired tumor, normal mucosa and distant metastasis samples and was used for quantitative detection of TGFB1 mRNA by TaqMan qPCR.We observed that TGF-β1 serum levels depend on the -509C/T genotype in combination with gender. TGF-β1 serum levels in CRC patients were decreased compared to controls, but statistical significance was reached only for men. In the stratified analysis by gender and genotype, a significant association was found for the CC genotype. Overall, our results indicate that the -509C allele increased the cancer risk, particularly for advanced stages (OR = 1.477; p = 0.029). The results from the relative mRNA quantification showed a significant upregulation of TGFB1 in distant metastases compared to primary tumor tissues and higher TGFB1 mRNA levels in men (RQ = 4.959; p = 0.022). In conclusion, we present data that diminished circulating TGF-β1 due to the CC genotype could be a possible risk factor for tumor susceptibility and progression. This association is more pronounced in males than in females. Colorectal cancer tissue expression of TGFB1 gene mRNA correlates with tumor progression and metastasis.
Collapse
Affiliation(s)
- Spaska Stanilova
- Department of Molecular Biology, Immunology and Medical Genetics, Medical Faculty, Trakia University, Stara Zagora, Bulgaria
- * E-mail:
| | - Noyko Stanilov
- Breast Oncoplastic Unit, University College London Hospital, London, United Kingdom
| | - Alexander Julianov
- Trakia Hospital, Stara Zagora, Bulgaria
- Department of Surgery, Medical Faculty, Trakia University, Stara Zagora, Bulgaria
| | - Irena Manolova
- Department of Molecular Biology, Immunology and Medical Genetics, Medical Faculty, Trakia University, Stara Zagora, Bulgaria
| | - Lyuba Miteva
- Department of Molecular Biology, Immunology and Medical Genetics, Medical Faculty, Trakia University, Stara Zagora, Bulgaria
| |
Collapse
|
139
|
Rolvering C, Zimmer AD, Ginolhac A, Margue C, Kirchmeyer M, Servais F, Hermanns HM, Hergovits S, Nazarov PV, Nicot N, Kreis S, Haan S, Behrmann I, Haan C. The PD-L1- and IL6-mediated dampening of the IL27/STAT1 anticancer responses are prevented by α-PD-L1 or α-IL6 antibodies. J Leukoc Biol 2018; 104:969-985. [PMID: 30040142 DOI: 10.1002/jlb.ma1217-495r] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 06/15/2018] [Accepted: 06/18/2018] [Indexed: 12/19/2022] Open
Abstract
Interleukin-27 (IL27) is a type-I cytokine of the IL6/IL12 family and is predominantly secreted by activated macrophages and dendritic cells. We show that IL27 induces STAT factor phosphorylation in cancerous cell lines of different tissue origin. IL27 leads to STAT1 phosphorylation and recapitulates an IFN-γ-like response in the microarray analyses, with up-regulation of genes involved in antiviral defense, antigen presentation, and immune suppression. Like IFN-γ, IL27 leads to an up-regulation of TAP2 and MHC-I proteins, which mediate increased tumor immune clearance. However, both cytokines also upregulate proteins such as PD-L1 (CD274) and IDO-1, which are associated with immune escape of cancer. Interestingly, differential expression of these genes was observed within the different cell lines and when comparing IL27 to IFN-γ. In coculture experiments of hepatocellular carcinoma (HCC) cells with peripheral blood mononuclear cells, pre-treatment of the HCC cells with IL27 resulted in lowered IL2 production by anti-CD3/-CD28 activated T-lymphocytes. Addition of anti-PD-L1 antibody, however, restored IL2 secretion. The levels of other TH 1 cytokines were also enhanced or restored upon administration of anti-PD-L1. In addition, we show that the suppression of IL27 signaling by IL6-type cytokine pre-stimulation-mimicking a situation occurring, for example, in IL6-secreting tumors or in tumor inflammation-induced cachexia-can be antagonized by antibodies against IL6-type cytokines or their receptors. Therapeutically, the antitumor effects of IL27 (mediated, e.g., by increased antigen presentation) might thus be increased by combining IL27 with blocking antibodies against PD-L1 or/and IL6-type cytokines.
Collapse
Affiliation(s)
- Catherine Rolvering
- University of Luxembourg, Life Sciences Research Unit-Signal Transduction Laboratory, Belvaux, Luxembourg
| | - Andreas D Zimmer
- University of Luxembourg, Life Sciences Research Unit-Signal Transduction Laboratory, Belvaux, Luxembourg
| | - Aurélien Ginolhac
- University of Luxembourg, Life Sciences Research Unit-Bioinformatics Core Facility, Belvaux, Luxembourg
| | - Christiane Margue
- University of Luxembourg, Life Sciences Research Unit-Signal Transduction Laboratory, Belvaux, Luxembourg
| | - Mélanie Kirchmeyer
- University of Luxembourg, Life Sciences Research Unit-Signal Transduction Laboratory, Belvaux, Luxembourg
| | - Florence Servais
- University of Luxembourg, Life Sciences Research Unit-Signal Transduction Laboratory, Belvaux, Luxembourg
| | - Heike M Hermanns
- University Hospital Würzburg, Medical Clinic II, Division of Hepatology, Würzburg, Germany
| | - Sabine Hergovits
- University Hospital Würzburg, Medical Clinic II, Division of Hepatology, Würzburg, Germany
| | - Petr V Nazarov
- Proteome and Genome Research Unit, Department of Oncology, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Nathalie Nicot
- Proteome and Genome Research Unit, Department of Oncology, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Stephanie Kreis
- University of Luxembourg, Life Sciences Research Unit-Signal Transduction Laboratory, Belvaux, Luxembourg
| | - Serge Haan
- University of Luxembourg, Life Sciences Research Unit-Molecular Disease Mechanisms Laboratory, Belvaux, Luxembourg
| | - Iris Behrmann
- University of Luxembourg, Life Sciences Research Unit-Signal Transduction Laboratory, Belvaux, Luxembourg
| | - Claude Haan
- University of Luxembourg, Life Sciences Research Unit-Signal Transduction Laboratory, Belvaux, Luxembourg
| |
Collapse
|
140
|
Rizzo A, Di Giovangiulio M, Stolfi C, Franzè E, Fehling HJ, Carsetti R, Giorda E, Colantoni A, Ortenzi A, Rugge M, Mescoli C, Monteleone G, Fantini MC. RORγt-Expressing Tregs Drive the Growth of Colitis-Associated Colorectal Cancer by Controlling IL6 in Dendritic Cells. Cancer Immunol Res 2018; 6:1082-1092. [PMID: 29991500 DOI: 10.1158/2326-6066.cir-17-0698] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2017] [Revised: 04/17/2018] [Accepted: 07/05/2018] [Indexed: 11/16/2022]
Abstract
Chronic inflammation drives colitis-associated colorectal cancer (CAC) in inflammatory bowel disease (IBD). FoxP3+ regulatory T cells (Treg) coexpressing the Th17-related transcription factor RORγt accumulate in the lamina propria of IBD patients, where they are thought to represent an intermediate stage of development toward a Th17 proinflammatory phenotype. However, the role of these cells in CAC is unknown. RORγt+FoxP3+ cells were investigated in human samples of CAC, and their phenotypic stability and function were investigated in an azoxymethane/dextran sulfate sodium model of CAC using Treg fate-mapping reporter and Treg-specific RORγt conditional knockout mice. Tumor development and the intratumoral inflammatory milieu were characterized in these mice. The functional role of CTLA-4 expressed by Tregs and FoxO3 in dendritic cells (DC) was studied in vitro and in vivo by siRNA-silencing experiments. RORγt expression identified a phenotypically stable population of tumor-infiltrating Tregs in humans and mice. Conditional RORγt knockout mice showed reduced tumor incidence, and dysplastic cells exhibited low Ki67 expression and STAT3 activation. Tumor-infiltrating DCs produced less IL6, a cytokine that triggers STAT3-dependent proliferative signals in neoplastic cells. RORγt-deficient Tregs isolated from tumors overexpressed CTLA-4 and induced DCs to have elevated expression of the transcription factor FoxO3, thus reducing IL6 expression. Finally, in vivo silencing of FoxO3 obtained by siRNA microinjection in the tumors of RORγt-deficient mice restored IL6 expression and tumor growth. These data demonstrate that RORγt expressed by tumor-infiltrating Tregs sustains tumor growth by leaving IL6 expression in DCs unchecked. Cancer Immunol Res; 6(9); 1082-92. ©2018 AACR.
Collapse
Affiliation(s)
- Angelamaria Rizzo
- Department of System Medicine, University of Rome Tor Vergata, Rome, Italy
| | | | - Carmine Stolfi
- Department of System Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Eleonora Franzè
- Department of System Medicine, University of Rome Tor Vergata, Rome, Italy
| | | | - Rita Carsetti
- Research Centre, Bambino Gesù Children's Hospital, Rome, Italy
| | - Ezio Giorda
- Research Centre, Bambino Gesù Children's Hospital, Rome, Italy
| | - Alfredo Colantoni
- Department of System Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Angela Ortenzi
- Department of System Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Massimo Rugge
- Department of Medicine DIMED Pathology and Cytopathology Unit, University of Padova, Padova, Italy
| | - Claudia Mescoli
- Department of Medicine DIMED Pathology and Cytopathology Unit, University of Padova, Padova, Italy
| | | | - Massimo C Fantini
- Department of System Medicine, University of Rome Tor Vergata, Rome, Italy.
| |
Collapse
|
141
|
Bartoli F, Bae S, Cometi L, Matucci Cerinic M, Furst DE. Sirukumab for the treatment of rheumatoid arthritis: update on sirukumab, 2018. Expert Rev Clin Immunol 2018; 14:539-547. [PMID: 29925278 DOI: 10.1080/1744666x.2018.1487291] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
INTRODUCTION Interleukin-6 (IL-6) is well-known for its pro-inflammatory properties, has been proven to target a wide range of cells in the joint, and has been implicated in extra-articular and articular manifestations in rheumatoid arthritis (RA). Tocilizumab (TCZ) is now widely used in patients with active RA and a number of additional agents that target the IL-6 pathways are under development, including sirukumab (SRK). Areas covered: SRK is an IgG1κ human anti-IL-6 monoclonal antibody which binds to IL-6 and prevents IL-6-mediated downstream effects. Initial trial results in phase-III studies in patients with RA seemed promising, showing improved results in patients with moderate-to-severe RA. Data derive from the phase-II study and the various SIRROUND studies (phase III). Expert commentary: The available data show that SRK50 mg every 4 weeks or 100 mg every 2 weeks will be effective in treating the RA population, with clinical improvements as early as week 2 and sustained over time. The adverse-event profile seems to be similar to TCZ, except for an increased mortality post open-label studies due to infections and cardiovascular events, our knowledge of which will be deepened with post-marketing surveillance and registry data.
Collapse
Affiliation(s)
- Francesca Bartoli
- a Department of Experimental and Clinical Medicine , University of Florence & Department of Geriatric Medicine, Division of Rheumatology, AOUC , Florence , Italy
| | - Sangmee Bae
- b Division of Rheumatology Fellow , Geffen School of Medicine at the University of California in Los Angeles , Los Angeles , California, USA
| | - Laura Cometi
- a Department of Experimental and Clinical Medicine , University of Florence & Department of Geriatric Medicine, Division of Rheumatology, AOUC , Florence , Italy
| | - Marco Matucci Cerinic
- a Department of Experimental and Clinical Medicine , University of Florence & Department of Geriatric Medicine, Division of Rheumatology, AOUC , Florence , Italy
| | - Daniel E Furst
- c Geffen School of Medicine at the University of California in Los Angeles (Emeritus), California, USA.,d Department of Experimental and Clinical Medicine , University of Florence , Florence , Italy.,e University of Washington , Seattle , Washington DC , USA.,f Pacific Arthritis , Los Angeles , California , USA.,g Seattle Rheumatology Associates , Seattle , Washington, DC, USA
| |
Collapse
|
142
|
Alessandra-Perini J, Rodrigues-Baptista KC, Machado DE, Nasciutti LE, Perini JA. Anticancer potential, molecular mechanisms and toxicity of Euterpe oleracea extract (açaí): A systematic review. PLoS One 2018; 13:e0200101. [PMID: 29966007 PMCID: PMC6028114 DOI: 10.1371/journal.pone.0200101] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Accepted: 06/19/2018] [Indexed: 12/18/2022] Open
Abstract
Cancer is an increasingly frequent malignancy worldwide, and despite the advances in drug development, it is still necessary to develop new plant-derived medicines. Euterpe oleracea (açaí) is abundant in South and Central America and has health benefits due to its high levels of phytochemicals, including lignans and polyphenols. The aim of this review was to systematically describe the safety and antitumor effects of açaí in preclinical models using rodents to provide a more comprehensive assessment of açaí for both therapeutic uses and the development of future clinical studies in cancer. Eligible studies were identified using four international databases (PubMed, Medline, Lilacs and SciELO) from their inception date through December 2017. The included studies were analyzed with methodological rigor (QATRS) to enable better quality control for these experimental studies. Sixty publications were identified in the databases, but only 9 articles were eligible: 6 evaluated the pharmacological effects of açaí in animal models of cancer (1 model each of esophageal cancer, urothelial cancer, melanoma and Walker-256 tumor and 2 models of colon cancer), and 3 were toxicological assays using preclinical models with rodents. Overall, 747 animals were analyzed. On a QATRS score scale of 0-20, the quality of the studies ranged from 16 to 20 points. Pulp was the main fraction of açaí administered, and an oral administration route was most common. The açaí dosage administered by gavage ranged from 30 mg/kg to 40,000 mg/kg, and açaí fed in the diet accounted for 2.5% to 5% of the diet. The anticarcinogenic and chemopreventive activities of açaí were observed in all experimental models of cancer and reduced the incidence, tumor cell proliferation, multiplicity and size of the tumors due to the antiinflammatory, antiproliferative and proapoptotic properties of açaí. No genotoxic effects were observed after açaí administration. The results of this review suggest that açaí is safe and can be used as a chemoprotective agent against cancer development. Açaí therapy may be a novel strategy for treating cancer.
Collapse
Affiliation(s)
- Jéssica Alessandra-Perini
- Morphological Science Program—PCM, Biomedical Sciences Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil
- Research Laboratory of Pharmaceutical Sciences—LAPESF, West Zone State University, Rio de Janeiro, Rio de Janeiro, Brazil
- * E-mail:
| | - Karina Cristina Rodrigues-Baptista
- Research Laboratory of Pharmaceutical Sciences—LAPESF, West Zone State University, Rio de Janeiro, Rio de Janeiro, Brazil
- Program of Post-graduation in Public Health and Environment—ENSP, National School of Public Health, Oswald Cruz Foundation, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Daniel Escorsim Machado
- Morphological Science Program—PCM, Biomedical Sciences Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil
- Research Laboratory of Pharmaceutical Sciences—LAPESF, West Zone State University, Rio de Janeiro, Rio de Janeiro, Brazil
- University Center IBMR, Laureate Universities, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luiz Eurico Nasciutti
- Morphological Science Program—PCM, Biomedical Sciences Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Jamila Alessandra Perini
- Research Laboratory of Pharmaceutical Sciences—LAPESF, West Zone State University, Rio de Janeiro, Rio de Janeiro, Brazil
- Program of Post-graduation in Public Health and Environment—ENSP, National School of Public Health, Oswald Cruz Foundation, Rio de Janeiro, Rio de Janeiro, Brazil
- Research Division, National Institute of Traumatology and Orthopedics—INTO, Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
143
|
NF-κB pathways in the development and progression of colorectal cancer. Transl Res 2018; 197:43-56. [PMID: 29550444 DOI: 10.1016/j.trsl.2018.02.002] [Citation(s) in RCA: 184] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 02/13/2018] [Accepted: 02/14/2018] [Indexed: 12/12/2022]
Abstract
Nuclear factor-κB (NF-κB) has been widely implicated in the development and progression of cancer. In colorectal cancer (CRC), NF-κB has a key role in cancer-related processes such as cell proliferation, apoptosis, angiogenesis, and metastasis. The role of NF-κB in CRC is complex, owed to the cross talk with other signaling pathways. Although there is sufficient evidence gained from cell lines and animal models that NF-κB is involved in cancer-related processes, because of a lack of studies in human tissue, the clinical evidence of its importance is limited in patients with CRC. This review summarizes evidence relating to how NF-κB is involved in the development and progression of CRC and comments on future work to be carried out.
Collapse
|
144
|
Li L, Shen A, Chu J, Sferra TJ, Sankararaman S, Ke X, Chen Y, Peng J. Pien Tze Huang ameliorates DSS‑induced colonic inflammation in a mouse colitis model through inhibition of the IL‑6/STAT3 pathway. Mol Med Rep 2018; 18:1113-1119. [PMID: 29845215 DOI: 10.3892/mmr.2018.9051] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 05/09/2018] [Indexed: 11/05/2022] Open
Abstract
Interleukin‑6 (IL‑6)/signal transducer and activator of transcription 3 (STAT3) pathway plays essential roles in the development of inflammatory diseases including ulcerative colitis (UC). Therefore, suppression of IL‑6/STAT3 signaling provides a promising therapeutic strategy in UC. Pien Tze Huang (PZH), a well‑known traditional Chinese formula, has been used in China and Southeast Asia for centuries as a folk remedy for various inflammatory diseases. However, the molecular mechanisms of its anti‑inflammatory effects remain to be elucidated. In the present study, we generated a mouse colitis model by using dextran sulfate sodium (DSS) and evaluated the therapeutic efficacy of PZH against UC by observing the clinical manifestations. We found that PZH obviously alleviated DSS‑induced colitis symptoms, including body weight loss, rectal bleeding and stool consistency. In addition, administration of PZH profoundly prevented DSS‑induced colon shortening, and ameliorated colonic histopathological changes such as mucosal ulceration, infiltration of inflammatory cells, crypt distortion and hyperplastic epithelium. Moreover, PZH markedly inhibited the serum level of the inflammatory biomarker serum amylase A (SAA) in UC mice. Furthermore, PZH treatment significantly inhibited DSS‑induced expression of IL‑6 in colon tissues. Finally, the increased phosphorylation level of STAT3, induced either by DSS in experimental mice or by IL‑6 in the differentiated human colorectal carcinoma cells, was significantly suppressed by PZH. These results suggest that the inhibition of IL‑6/STAT3 signaling is a potential mechanism by which PZH is used in the treatment of UC.
Collapse
Affiliation(s)
- Li Li
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Aling Shen
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Jianfeng Chu
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Thomas J Sferra
- Department of Gastroenterology, Rainbow Babies and Children's Hospital, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Senthilkumar Sankararaman
- Department of Gastroenterology, Rainbow Babies and Children's Hospital, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Xiao Ke
- Department of Gastroenterology, Second Affiliated Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350003, P.R. China
| | - Youqin Chen
- Department of Gastroenterology, Rainbow Babies and Children's Hospital, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Jun Peng
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| |
Collapse
|
145
|
Abstract
Patients with inflammatory bowel disease (IBD) are at significantly increased risk of colorectal cancer (CRC), principally resulting from the pro-neoplastic effects of chronic intestinal inflammation. Epidemiologic studies continue to highlight the increased risk of CRC in IBD. However, the incidence has declined over the past 30 years, attributed to both successful CRC-surveillance programs and improved control of mucosal inflammation. Risk factors that further increase the risk of IBD-related CRC include disease duration, extent and severity, the presence of inflammatory pseudopolyps, coexistent primary sclerosing cholangitis, and a family history of CRC. All major professional societies agree that IBD-CRC surveillance should occur more frequently than in the general population. Yet, guidelines and consensus statements differ on the surveillance schedule and preferred method of surveillance. Improved sensitivity to previously "invisible" flat dysplastic lesions using high definition and chromoendoscopy methods has resulted in many guidelines abandoning requirements for random untargeted biopsies of the colon. While colonic dysplasia remains a worrisome finding, and several clinical scenarios remain best addressed by total proctocolectomy due to concerns of synchronous undetected lesions and the unpredictable tempo of progression to malignancy, better detection techniques have also increased opportunities for endoscopic resection of dysplastic lesions that can be clearly delineated. Finally, the expanding armamentarium of medical options in IBD, including anti-tumor necrosis factor and anti-adhesion biologic therapies, have substantially improved our ability to control severe inflammation and likely reduce the risk of CRC over time.
Collapse
Affiliation(s)
- Ryan W. Stidham
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Peter D.R. Higgins
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
146
|
Eichenberger RM, Ryan S, Jones L, Buitrago G, Polster R, Montes de Oca M, Zuvelek J, Giacomin PR, Dent LA, Engwerda CR, Field MA, Sotillo J, Loukas A. Hookworm Secreted Extracellular Vesicles Interact With Host Cells and Prevent Inducible Colitis in Mice. Front Immunol 2018; 9:850. [PMID: 29760697 PMCID: PMC5936971 DOI: 10.3389/fimmu.2018.00850] [Citation(s) in RCA: 125] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 04/06/2018] [Indexed: 12/20/2022] Open
Abstract
Gastrointestinal (GI) parasites, hookworms in particular, have evolved to cause minimal harm to their hosts, allowing them to establish chronic infections. This is mediated by creating an immunoregulatory environment. Indeed, hookworms are such potent suppressors of inflammation that they have been used in clinical trials to treat inflammatory bowel diseases (IBD) and celiac disease. Since the recent description of helminths (worms) secreting extracellular vesicles (EVs), exosome-like EVs from different helminths have been characterized and their salient roles in parasite–host interactions have been highlighted. Here, we analyze EVs from the rodent parasite Nippostrongylus brasiliensis, which has been used as a model for human hookworm infection. N. brasiliensis EVs (Nb-EVs) are actively internalized by mouse gut organoids, indicating a role in driving parasitism. We used proteomics and RNA-Seq to profile the molecular composition of Nb-EVs. We identified 81 proteins, including proteins frequently present in exosomes (like tetraspanin, enolase, 14-3-3 protein, and heat shock proteins), and 27 sperm-coating protein-like extracellular proteins. RNA-Seq analysis revealed 52 miRNA species, many of which putatively map to mouse genes involved in regulation of inflammation. To determine whether GI nematode EVs had immunomodulatory properties, we assessed their potential to suppress GI inflammation in a mouse model of inducible chemical colitis. EVs from N. brasiliensis but not those from the whipworm Trichuris muris or control vesicles from grapes protected against colitic inflammation in the gut of mice that received a single intraperitoneal injection of EVs. Key cytokines associated with colitic pathology (IL-6, IL-1β, IFNγ, and IL-17a) were significantly suppressed in colon tissues from EV-treated mice. By contrast, high levels of the anti-inflammatory cytokine IL-10 were detected in Nb-EV-treated mice. Proteins and miRNAs contained within helminth EVs hold great potential application in development of drugs to treat helminth infections as well as chronic non-infectious diseases resulting from a dysregulated immune system, such as IBD.
Collapse
Affiliation(s)
- Ramon M Eichenberger
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| | - Stephanie Ryan
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| | - Linda Jones
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| | - Geraldine Buitrago
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| | - Ramona Polster
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| | - Marcela Montes de Oca
- Immunology and Infection Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Jennifer Zuvelek
- Pathology Queensland Cairns Laboratory, Queensland Health, Cairns, QLD, Australia
| | - Paul R Giacomin
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| | - Lindsay A Dent
- School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Christian R Engwerda
- Immunology and Infection Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Matthew A Field
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia.,Department of Immunology, John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
| | - Javier Sotillo
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| | - Alex Loukas
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| |
Collapse
|
147
|
Tian L, Xie XH, Zhu ZH. Calotropin regulates the apoptosis of non‑small cell cancer by regulating the cytotoxic T‑lymphocyte associated antigen 4‑mediated TGF‑β/ERK signaling pathway. Mol Med Rep 2018; 17:7683-7691. [PMID: 29620207 PMCID: PMC5983968 DOI: 10.3892/mmr.2018.8853] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 02/09/2018] [Indexed: 01/27/2023] Open
Abstract
Non-small-cell lung cancer (NSCLC) is one of the most common malignancies that is responsible for a high level of cancer-associated mortalities worldwide. Previous evidence has shown that Calotropin is an upstream activator of protein kinase B, which can further inhibit the growth and promote the apoptosis of NSCLC cells. In the present study, the efficacy of Calotropin on growth, aggressiveness and apoptosis of NSCLC cells was investigated, as well as the potential underlying mechanism. The results demonstrated that Calotropin inhibited H358 cell growth, migration and invasion. Flow cytometry assay showed that Calotropin promoted the apoptosis of H358 cells in vitro. Western blot analysis demonstrated that Calotropin inhibited fibronectin (FN), Vimentin (VIM) and E-cadherin (Eca) protein expression levels in H358 cells in vitro. In addition, Calotropin treatment upregulated pro-apoptosis gene expression, including caspase-3, caspase-8 and apoptotic protease activating factor-1, and downregulated anti-apoptosis gene expression, including P53, B-cell lymphoma (Bcl) 2 and Bcl-2-like protein 2 in H358 cells. The results also revealed that the expression levels of cytotoxic T-lymphocyte associated antigen 4 (CTLA-4) were decreased by Calotropin treatment in H358 cells. Analyses of the underlying mechanism indicated that Calotropin inhibited transforming growth factor-β (TGF-β) and extracellular signal-regulated kinase (ERK) expression. Overexpression of CTLA-4 inhibited Calotropin-mediated downregulation of TGF-β and ERK expression in H358 cells. In vivo assay revealed that Calotropin administration significantly inhibited tumor growth and prolonged animal survival over the 120-day observation period. Immunohistochemistry demonstrated that the number of apoptotic cells increased and the expression levels of CTLA-4 were decreased in the Calotropin-treated tumor group when compared with control. In addition, the expression levels of TGF-β and ERK were downregulated in the Calotropin-treated tumor group compared with control. In conclusion, the results of the present study indicated that Calotropin administration regulated NSCLC apoptosis by downregulating the CTLA-4-mediated TGF-β/ERK signaling pathway, suggesting that Calotropin may be a potential anti-cancer agent for the treatment of NSCLC.
Collapse
Affiliation(s)
- Lu Tian
- Department of Respiratory Medicine, The Fourth People's Hospital of Guiyang, Guiyang, Guizhou 550002, P.R. China
| | - Xiao-Hong Xie
- Department of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Institute of Respiratory Disease, Guangzhou, Guangdong 510120, P.R. China
| | - Ze-Hao Zhu
- Department of Respiratory Medicine, The Affiliated Zhoushan Hospital of Wenzhou Medical University, Zhoushan, Zhejiang 316000, P.R. China
| |
Collapse
|
148
|
Abstract
The IL-6/JAK/STAT3 pathway is aberrantly hyperactivated in many types of cancer, and such hyperactivation is generally associated with a poor clinical prognosis. In the tumour microenvironment, IL-6/JAK/STAT3 signalling acts to drive the proliferation, survival, invasiveness, and metastasis of tumour cells, while strongly suppressing the antitumour immune response. Thus, treatments that target the IL-6/JAK/STAT3 pathway in patients with cancer are poised to provide therapeutic benefit by directly inhibiting tumour cell growth and by stimulating antitumour immunity. Agents targeting IL-6, the IL-6 receptor, or JAKs have already received FDA approval for the treatment of inflammatory conditions or myeloproliferative neoplasms and for the management of certain adverse effects of chimeric antigen receptor T cells, and are being further evaluated in patients with haematopoietic malignancies and in those with solid tumours. Novel inhibitors of the IL-6/JAK/STAT3 pathway, including STAT3-selective inhibitors, are currently in development. Herein, we review the role of IL-6/JAK/STAT3 signalling in the tumour microenvironment and the status of preclinical and clinical investigations of agents targeting this pathway. We also discuss the potential of combining IL-6/JAK/STAT3 inhibitors with currently approved therapeutic agents directed against immune-checkpoint inhibitors.
Collapse
Affiliation(s)
- Daniel E. Johnson
- Department of Otolaryngology – Head and Neck Surgery, University of California, San Francisco, CA, USA
| | - Rachel A. O’Keefe
- Department of Otolaryngology – Head and Neck Surgery, University of California, San Francisco, CA, USA
| | - Jennifer R. Grandis
- Department of Otolaryngology – Head and Neck Surgery, University of California, San Francisco, CA, USA
| |
Collapse
|
149
|
Schmidt S, Schumacher N, Schwarz J, Tangermann S, Kenner L, Schlederer M, Sibilia M, Linder M, Altendorf-Hofmann A, Knösel T, Gruber ES, Oberhuber G, Bolik J, Rehman A, Sinha A, Lokau J, Arnold P, Cabron AS, Zunke F, Becker-Pauly C, Preaudet A, Nguyen P, Huynh J, Afshar-Sterle S, Chand AL, Westermann J, Dempsey PJ, Garbers C, Schmidt-Arras D, Rosenstiel P, Putoczki T, Ernst M, Rose-John S. ADAM17 is required for EGF-R-induced intestinal tumors via IL-6 trans-signaling. J Exp Med 2018; 215:1205-1225. [PMID: 29472497 PMCID: PMC5881468 DOI: 10.1084/jem.20171696] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 12/22/2017] [Accepted: 01/22/2018] [Indexed: 02/06/2023] Open
Abstract
Schmidt et al. show that loss of the membrane-bound metalloprotease ADAM17 led to impaired intestinal cancer development in the murine APCmin/+ model, which also depended on IL-6 trans-signaling via the soluble IL-6R and could be blocked by the specific IL-6 trans-signaling inhibitor sgp130Fc. Colorectal cancer is treated with antibodies blocking epidermal growth factor receptor (EGF-R), but therapeutic success is limited. EGF-R is stimulated by soluble ligands, which are derived from transmembrane precursors by ADAM17-mediated proteolytic cleavage. In mouse intestinal cancer models in the absence of ADAM17, tumorigenesis was almost completely inhibited, and the few remaining tumors were of low-grade dysplasia. RNA sequencing analysis demonstrated down-regulation of STAT3 and Wnt pathway components. Because EGF-R on myeloid cells, but not on intestinal epithelial cells, is required for intestinal cancer and because IL-6 is induced via EGF-R stimulation, we analyzed the role of IL-6 signaling. Tumor formation was equally impaired in IL-6−/− mice and sgp130Fc transgenic mice, in which only trans-signaling via soluble IL-6R is abrogated. ADAM17 is needed for EGF-R–mediated induction of IL-6 synthesis, which via IL-6 trans-signaling induces β-catenin–dependent tumorigenesis. Our data reveal the possibility of a novel strategy for treatment of colorectal cancer that could circumvent intrinsic and acquired resistance to EGF-R blockade.
Collapse
Affiliation(s)
- Stefanie Schmidt
- Biochemisches Institut, Christian Albrechts Universität Kiel, Kiel, Germany
| | - Neele Schumacher
- Biochemisches Institut, Christian Albrechts Universität Kiel, Kiel, Germany
| | - Jeanette Schwarz
- Biochemisches Institut, Christian Albrechts Universität Kiel, Kiel, Germany
| | - Simone Tangermann
- Unit of Laboratory Animal Pathology, University of Veterinary Medicine, Vienna, Austria
| | - Lukas Kenner
- Unit of Laboratory Animal Pathology, University of Veterinary Medicine, Vienna, Austria.,Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria.,Department of Experimental and Laboratory Animal Pathology, Medical University Vienna, Vienna, Austria
| | - Michaela Schlederer
- Department of Experimental and Laboratory Animal Pathology, Medical University Vienna, Vienna, Austria
| | - Maria Sibilia
- Institute of Cancer Research, Department of Medicine I, Medical University of Vienna, Comprehensive Cancer Center, Vienna, Austria
| | - Markus Linder
- Institute of Cancer Research, Department of Medicine I, Medical University of Vienna, Comprehensive Cancer Center, Vienna, Austria
| | | | - Thomas Knösel
- Institute of Pathology, Ludwig-Maximilians-University, Munich, Germany
| | - Elisabeth S Gruber
- Department of General Surgery, Division of Surgery and Comprehensive Cancer Center, Medical University Vienna, Vienna, Austria
| | - Georg Oberhuber
- Department of Experimental and Laboratory Animal Pathology, Medical University Vienna, Vienna, Austria
| | - Julia Bolik
- Biochemisches Institut, Christian Albrechts Universität Kiel, Kiel, Germany
| | - Ateequr Rehman
- Institute of Clinical Molecular Biology, Christian Albrechts Universität Kiel, Kiel, Germany
| | - Anupam Sinha
- Institute of Clinical Molecular Biology, Christian Albrechts Universität Kiel, Kiel, Germany
| | - Juliane Lokau
- Biochemisches Institut, Christian Albrechts Universität Kiel, Kiel, Germany
| | - Philipp Arnold
- Anatomisches Institut, Christian Albrechts Universität Kiel, Kiel, Germany
| | - Anne-Sophie Cabron
- Biochemisches Institut, Christian Albrechts Universität Kiel, Kiel, Germany
| | - Friederike Zunke
- Biochemisches Institut, Christian Albrechts Universität Kiel, Kiel, Germany
| | | | - Adele Preaudet
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia.,Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
| | - Paul Nguyen
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia.,Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
| | - Jennifer Huynh
- Olivia Newton-John Cancer Research Institute and La Trobe University School of Cancer Medicine, Heidelberg, VIC, Australia
| | - Shoukat Afshar-Sterle
- Olivia Newton-John Cancer Research Institute and La Trobe University School of Cancer Medicine, Heidelberg, VIC, Australia
| | - Ashwini L Chand
- Olivia Newton-John Cancer Research Institute and La Trobe University School of Cancer Medicine, Heidelberg, VIC, Australia
| | | | - Peter J Dempsey
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO
| | - Christoph Garbers
- Biochemisches Institut, Christian Albrechts Universität Kiel, Kiel, Germany
| | - Dirk Schmidt-Arras
- Biochemisches Institut, Christian Albrechts Universität Kiel, Kiel, Germany
| | - Philip Rosenstiel
- Institute of Clinical Molecular Biology, Christian Albrechts Universität Kiel, Kiel, Germany
| | - Tracy Putoczki
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia.,Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
| | - Matthias Ernst
- Olivia Newton-John Cancer Research Institute and La Trobe University School of Cancer Medicine, Heidelberg, VIC, Australia
| | - Stefan Rose-John
- Biochemisches Institut, Christian Albrechts Universität Kiel, Kiel, Germany
| |
Collapse
|
150
|
Avian leukosis virus subgroup J induces VEGF expression via NF-κB/PI3K-dependent IL-6 production. Oncotarget 2018; 7:80275-80287. [PMID: 27852059 PMCID: PMC5348319 DOI: 10.18632/oncotarget.13282] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 11/07/2016] [Indexed: 02/07/2023] Open
Abstract
Avian leukosis virus subgroup J (ALV-J) is an oncogenic virus causing hemangiomas and myeloid tumors in chickens. Interleukin-6 (IL-6) is a multifunctional pro-inflammatory interleukin involved in many types of cancer. We previously demonstrated that IL-6 expression was induced following ALV-J infection in chickens. The aim of this study is to characterize the mechanism by which ALV-J induces IL-6 expression, and the role of IL-6 in tumor development. Our results demonstrate that ALV-J infection increases IL-6 expression in chicken splenocytes, peripheral blood lymphocytes, and vascular endothelial cells. IL-6 production is induced by the ALV-J envelope protein gp85 and capsid protein p27 via PI3K- and NF-κB-mediated signaling. IL-6 in turn induced expression of vascular endothelial growth factor (VEGF)-A and its receptor, VEGFR-2, in vascular endothelial cells and embryonic vascular tissues. Suppression of IL-6 using siRNA inhibited the ALV-J induced VEGF-A and VEGFR-2 expression in vascular endothelial cells, indicating that the ALV-J-induced VEGF-A/VEGFR-2 expression is mediated by IL-6. As VEGF-A and VEGFR-2 are important factors in oncogenesis, our findings suggest that ALV-J hijacks IL-6 to promote tumorigenesis, and indicate that IL-6 could potentially serve as a therapeutic target in ALV-J infections.
Collapse
|