1
|
Fuchs B, Mert S, Kuhlmann C, Birt A, Hofmann D, Wiggenhauser PS, Giunta RE, Chavez MN, Nickelsen J, Schenck TL, Moellhoff N. In Vivo Biocompatibility of Synechococcus sp. PCC 7002-Integrated Scaffolds for Skin Regeneration. J Funct Biomater 2024; 15:295. [PMID: 39452593 PMCID: PMC11508603 DOI: 10.3390/jfb15100295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 09/27/2024] [Accepted: 10/01/2024] [Indexed: 10/26/2024] Open
Abstract
Cyanobacteria, commonly known as blue-green algae, are prevalent in freshwater systems and have gained interest for their potential in medical applications, particularly in skin regeneration. Among these, Synechococcus sp. strain PCC 7002 stands out because of its rapid proliferation and capacity to be genetically modified to produce growth factors. This study investigates the safety of Synechococcus sp. PCC 7002 when used in scaffolds for skin regeneration, focusing on systemic inflammatory responses in a murine model. We evaluated the following three groups: scaffolds colonized with genetically engineered bacteria producing hyaluronic acid, scaffolds with wild-type bacteria, and control scaffolds without bacteria. After seven days, we assessed systemic inflammation by measuring changes in cytokine profiles and lymphatic organ sizes. The results showed no significant differences in spleen, thymus, and lymph node weights, indicating a lack of overt systemic toxicity. Blood cytokine analysis revealed elevated levels of IL-6 and IL-1β in scaffolds with bacteria, suggesting a systemic inflammatory response, while TNF-α levels remained unaffected. Proteome profiling identified distinct cytokine patterns associated with bacterial colonization, including elevated inflammatory proteins and products, indicative of acute inflammation. Conversely, control scaffolds exhibited protein profiles suggestive of a rejection response, characterized by increased levels of cytokines involved in T and B cell activation. Our findings suggest that Synechococcus sp. PCC 7002 does not appear to cause significant systemic toxicity, supporting its potential use in biomedical applications. Further research is necessary to explore the long-term effects and clinical implications of these responses.
Collapse
Affiliation(s)
- Benedikt Fuchs
- Division of Hand, Plastic and Aesthetic Surgery, LMU University Hospital, LMU Munich, 80336 Munich, Germany; (S.M.); (C.K.); (A.B.); (D.H.); (P.S.W.); (R.E.G.); (N.M.)
| | - Sinan Mert
- Division of Hand, Plastic and Aesthetic Surgery, LMU University Hospital, LMU Munich, 80336 Munich, Germany; (S.M.); (C.K.); (A.B.); (D.H.); (P.S.W.); (R.E.G.); (N.M.)
| | - Constanze Kuhlmann
- Division of Hand, Plastic and Aesthetic Surgery, LMU University Hospital, LMU Munich, 80336 Munich, Germany; (S.M.); (C.K.); (A.B.); (D.H.); (P.S.W.); (R.E.G.); (N.M.)
| | - Alexandra Birt
- Division of Hand, Plastic and Aesthetic Surgery, LMU University Hospital, LMU Munich, 80336 Munich, Germany; (S.M.); (C.K.); (A.B.); (D.H.); (P.S.W.); (R.E.G.); (N.M.)
| | - Daniel Hofmann
- Division of Hand, Plastic and Aesthetic Surgery, LMU University Hospital, LMU Munich, 80336 Munich, Germany; (S.M.); (C.K.); (A.B.); (D.H.); (P.S.W.); (R.E.G.); (N.M.)
| | - Paul Severin Wiggenhauser
- Division of Hand, Plastic and Aesthetic Surgery, LMU University Hospital, LMU Munich, 80336 Munich, Germany; (S.M.); (C.K.); (A.B.); (D.H.); (P.S.W.); (R.E.G.); (N.M.)
| | - Riccardo E. Giunta
- Division of Hand, Plastic and Aesthetic Surgery, LMU University Hospital, LMU Munich, 80336 Munich, Germany; (S.M.); (C.K.); (A.B.); (D.H.); (P.S.W.); (R.E.G.); (N.M.)
| | - Myra N. Chavez
- Institute of Anatomy, University of Bern, 3012 Bern, Switzerland;
| | - Jörg Nickelsen
- Molecular Plant Science, Department Biology I, LMU Munich, 80336 Munich, Germany;
| | | | - Nicholas Moellhoff
- Division of Hand, Plastic and Aesthetic Surgery, LMU University Hospital, LMU Munich, 80336 Munich, Germany; (S.M.); (C.K.); (A.B.); (D.H.); (P.S.W.); (R.E.G.); (N.M.)
| |
Collapse
|
2
|
Iyer S, Enman M, Sahay P, Dudeja V. Novel therapeutics to treat chronic pancreatitis: targeting pancreatic stellate cells and macrophages. Expert Rev Gastroenterol Hepatol 2024; 18:171-183. [PMID: 38761167 DOI: 10.1080/17474124.2024.2355969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 05/13/2024] [Indexed: 05/20/2024]
Abstract
INTRODUCTION Chronic pancreatitis (CP) is a persistent, recurrent, and progressive disorder that is characterized by chronic inflammation and irreversible fibrosis of the pancreas. It is associated with severe morbidity, resulting in intense abdominal pain, diabetes, exocrine and endocrine dysfunction, and an increased risk of pancreatic cancer. The etiological factors are diverse and the major risk factors include smoking, chronic alcoholism, as well as other environmental and genetic factors. The treatment and management of CP is challenging, and no definitive curative therapy is currently available. AREAS COVERED This review paper aims to provide an overview of the different cell types in the pancreas that is known to mediate disease progression and outline potential novel therapeutic approaches and drug targets that may be effective in treating and managing CP. The information presented in this review was obtained by conducting a NCBI PubMed database search, using relevant keywords. EXPERT OPINION In recent years, there has been an increased interest in the development of novel therapeutics for CP. A collaborative multi-disciplinary approach coupled with a consistent funding for research can expedite progress of translating the findings from bench to bedside.
Collapse
Affiliation(s)
- Srikanth Iyer
- Department of Surgery, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL, USA
| | - Macie Enman
- Department of Surgery, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL, USA
| | - Preeti Sahay
- Department of Surgery, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL, USA
| | - Vikas Dudeja
- Department of Surgery, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL, USA
| |
Collapse
|
3
|
Kong F, Pan Y, Wu D. Activation and Regulation of Pancreatic Stellate Cells in Chronic Pancreatic Fibrosis: A Potential Therapeutic Approach for Chronic Pancreatitis. Biomedicines 2024; 12:108. [PMID: 38255213 PMCID: PMC10813475 DOI: 10.3390/biomedicines12010108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 12/16/2023] [Accepted: 12/28/2023] [Indexed: 01/24/2024] Open
Abstract
In the complex progression of fibrosis in chronic pancreatitis, pancreatic stellate cells (PSCs) emerge as central figures. These cells, initially in a dormant state characterized by the storage of vitamin A lipid droplets within the chronic pancreatitis microenvironment, undergo a profound transformation into an activated state, typified by the secretion of an abundant extracellular matrix, including α-smooth muscle actin (α-SMA). This review delves into the myriad factors that trigger PSC activation within the context of chronic pancreatitis. These factors encompass alcohol, cigarette smoke, hyperglycemia, mechanical stress, acinar cell injury, and inflammatory cells, with a focus on elucidating their underlying mechanisms. Additionally, we explore the regulatory factors that play significant roles during PSC activation, such as TGF-β, CTGF, IL-10, PDGF, among others. The investigation into these regulatory factors and pathways involved in PSC activation holds promise in identifying potential therapeutic targets for ameliorating fibrosis in chronic pancreatitis. We provide a summary of recent research findings pertaining to the modulation of PSC activation, covering essential genes and innovative regulatory mediators designed to counteract PSC activation. We anticipate that this research will stimulate further insights into PSC activation and the mechanisms of pancreatic fibrosis, ultimately leading to the discovery of groundbreaking therapies targeting cellular and molecular responses within these processes.
Collapse
Affiliation(s)
- Fanyi Kong
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China; (F.K.); (Y.P.)
| | - Yingyu Pan
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China; (F.K.); (Y.P.)
| | - Dong Wu
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China; (F.K.); (Y.P.)
- Clinical Epidemiology Unit, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| |
Collapse
|
4
|
Thioredoxin-interacting protein deficiency protects against severe acute pancreatitis by suppressing apoptosis signal-regulating kinase 1. Cell Death Dis 2022; 13:914. [PMID: 36316322 PMCID: PMC9622726 DOI: 10.1038/s41419-022-05355-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 10/16/2022] [Accepted: 10/17/2022] [Indexed: 11/07/2022]
Abstract
Acute pancreatitis is a common acute inflammatory abdominal disease. When acute pancreatitis progresses to severe acute pancreatitis (SAP), it can lead to systemic inflammation and even multiple organ failure. Thioredoxin-interacting protein (TXNIP) is an important protein involved in redox reactions of the inflammatory response. However, the specific role of TXNIP in SAP remains unclear. In this study, we investigated the role of thioredoxin interacting protein (TXNIP) in acute pancreatitis when induced by high doses of arginine. We found that pancreatic damage and the inflammatory response associated with acute pancreatitis were largely restrained in TXNIP knock-out mice but were enhanced in mice overexpressing TXNIP. Interestingly, the phosphorylation of p38, JNK, and ASK1 diminished in TXNIP-KO mice with pancreatitis in comparison with wild-type mice. The role of oxidative stress in SAP was explored in two models: TXNIP and AVV-TXNIP. TXNIP knockdown or the inhibition of ASK1 by gs-4997 abrogated the increase in p-p38, p-JNK, and p-ASK1 in AR42J cells incubated with L-Arg. The administration of gs-4997 to mice with pancreatitis largely reduced the upregulation of IL-6, IL-1β, TNF-α, and MCP-1. Systemic inflammatory reactions and injury in the lungs and kidneys were assessed in TXNIP-KO and AVV-TXNIP mice with expected outcomes. In conclusion, TXNIP is a novel mediator of SAP and exerts action by regulating inflammatory responses and oxidative stress via the ASK1-dependent activation of the JNK/p38 pathways. Thus, targeting TXNIP may represent a promising approach to protect against SAP.
Collapse
|
5
|
Challagundla N, Shah D, Yadav S, Agrawal-Rajput R. Saga of monokines in shaping tumour-immune microenvironment: Origin to execution. Cytokine 2022; 157:155948. [PMID: 35764025 DOI: 10.1016/j.cyto.2022.155948] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 06/09/2022] [Accepted: 06/13/2022] [Indexed: 11/19/2022]
Abstract
Cellular communication mediated by cytokines is an important mechanism dictating immune responses, their cross talk and final immune output. Cytokines play a major role in dictating the immune outcome to cancer by regulating the events of development, differentiation and activation of innate immune cells. Cytokines are pleiotropic in nature, hence understanding their role individually or as member of network cytokines is critical to delineate their role in tumour immunity. Tumour systemically manipulates the immune system to evade and escape immune recognition for their uncontrollable growth and metastasis. The developing tumour comprise a large and diverse set of myeloid cells which are vulnerable to manipulation by the tumour-microenvironment. The innate immune cells of the monocytic lineage skew the fate of the adaptive immune cells and thus dictating cancer elimination or progression. Targeting cells at tumour cite is preposterous owing to their tight network, poor reach and abundance of immunosuppressive mechanisms. Monocytic lineage-derived cytokines (monokines) play crucial role in tumour regression or progression by either directly killing the tumour cells with TNFα or promoting its growth by TGFβ. In addition, the monokines like IL-12, IL-1β, IL-6, IL-10 and TGFβ direct the adaptive immune cells to secrete anti-tumour cytokines, TNFα, IFNγ, perforin and granzyme or pro-tumour cytokines, IL-10 and TGFβ. In this review, we elucidate the roles of monokines in dictating the fate of tumour by regulating responses at various stages of generation, differentiation and activation of immune cells along with the extensive cross talk. We have attempted to delineate the synergy and antagonism of major monokines among themselves or with tumour-derived or adaptive immune cytokines. The review provides an update on the possibilities of placing monokines to potential practical use as cytokine therapy against cancer.
Collapse
Affiliation(s)
- Naveen Challagundla
- Immunology Lab, Indian Institute of Advanced Research, Koba Institutional Area, Gandhinagar, Gujarat 382426, India
| | - Dhruvi Shah
- Immunology Lab, Indian Institute of Advanced Research, Koba Institutional Area, Gandhinagar, Gujarat 382426, India
| | - Shivani Yadav
- Immunology Lab, Indian Institute of Advanced Research, Koba Institutional Area, Gandhinagar, Gujarat 382426, India
| | - Reena Agrawal-Rajput
- Immunology Lab, Indian Institute of Advanced Research, Koba Institutional Area, Gandhinagar, Gujarat 382426, India.
| |
Collapse
|
6
|
Jiang T, Wei F, Xie K. Clinical significance of pancreatic ductal metaplasia. J Pathol 2022; 257:125-139. [PMID: 35170758 DOI: 10.1002/path.5883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 02/06/2022] [Accepted: 02/14/2022] [Indexed: 11/08/2022]
Abstract
Pancreatic ductal metaplasia (PDM) is the stepwise replacement of differentiated somatic cells with ductal or ductal-like cells in the pancreas. PDM is usually triggered by cellular and environmental insults. PDM development may involve all cell lineages of the pancreas, and acinar cells with the highest plasticity are the major source of PDM. Pancreatic progenitor cells are also involved as cells of origin or transitional intermediates. PDM is heterogeneous at the histological, cellular, and molecular levels and only certain subsets of PDM develop further into pancreatic intraepithelial neoplasia (PanIN) and then pancreatic ductal adenocarcinoma (PDAC). The formation and evolution of PDM is regulated at the cellular and molecular levels through a complex network of signaling pathways. The key molecular mechanisms that drive PDM formation and its progression into PanIN/PDAC remain unclear, but represent key targets for reversing or inhibiting PDM. Alternatively, PDM could be a source of pancreas regeneration, including both exocrine and endocrine components. Cellular aging and apoptosis are obstacles to PDM-to-PanIN progression or pancreas regeneration. Functional identification of the cellular and molecular events driving senescence and apoptosis in PDM and its progression would help not only to restrict the development of PDM into PanIN/PDAC, but may also facilitate pancreatic regeneration. This review systematically assesses recent advances in the understanding of PDM physiology and pathology, with a focus on its implications for enhancing regeneration and prevention of cancer. © 2022 The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Tingting Jiang
- Center for Pancreatic Cancer Research, The South China University of Technology School of Medicine, Guangzhou, PR China
- Department of Pathology, The South China University of Technology School of Medicine, Guangzhou, PR China
| | - Fang Wei
- Institute of Digestive Diseases Research, The South China University of Technology School of Medicine, Guangzhou, PR China
| | - Keping Xie
- Center for Pancreatic Cancer Research, The South China University of Technology School of Medicine, Guangzhou, PR China
- Department of Pathology, The South China University of Technology School of Medicine, Guangzhou, PR China
| |
Collapse
|
7
|
Chen J, Wang T, Xu H, Zhu Y, Du Y, Liu B, Zhao Q, Zhang Y, Liu L, Yuan L, Fang J, Xie Y, Liu S, Wu R, Shao D, Song X, He B, Brunekreef B, Huang W. An extended analysis of cardiovascular benefits of indoor air filtration intervention among elderly: a randomized crossover trial (Beijing indoor air purifier study, BIAPSY). GLOBAL HEALTH JOURNAL 2022. [DOI: 10.1016/j.glohj.2022.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
8
|
Xu H, Liu S, Wang Y, Wu R, Yi T, Wang T, Zhu Y, Fang J, Xie Y, Zhao Q, Song X, Chen J, Rajagopaplan S, Brook RD, Li J, Cao J, Huang W. The mediating role of vascular inflammation in traffic-related air pollution associated changes in insulin resistance in healthy adults. Int J Hyg Environ Health 2021; 239:113878. [PMID: 34757311 DOI: 10.1016/j.ijheh.2021.113878] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 10/22/2021] [Accepted: 10/25/2021] [Indexed: 02/06/2023]
Abstract
AIM The precise pathophysiologic pathway linking traffic-related air pollution (TRAP) to diabetes mellitus is not well elucidated. We aimed to investigate whether activation of vascular inflammation can be a mechanistic linkage between ambient TRAP and insulin resistance. METHODS Study outcomes were determined by assessing a series of circulating biomarkers indicative of insulin resistance and vascular inflammation among 73 healthy adults who underwent repeated clinical visits in Beijing, China, 2014-2016. Concomitantly, concentrations of ambient TRAP indices, including particulate matter in diameter <2.5 μm (PM2.5), particles in size fractions of 5-560 nm, black carbon, carbon monoxide, nitrogen dioxide, and oxides of nitrogen, were continuously monitored. RESULTS Participants experienced extremely high levels of TRAP exposures, with mean (standard deviation) PM2.5 concentrations of 91.8 (48.3) μg/m3, throughout the study. We found that interquartile range increases in exposure to moving average concentrations of various TRAP indices at prior up to 7 days were associated with significant elevations of 8.9-49.6% in insulin levels. Higher pollutant levels were also related to worsening metrics of insulin resistance (soluble insulin receptor ectodomain, adipokines, and homeostasis model assessment of insulin resistance) and heightened vascular inflammatory responses, particularly disruptions of the receptor activator of nuclear factor κB ligand/osteoprotegerin system balance and elevations of monocyte/macrophage and T cell activation markers. Mediation analyses showed that activation of vascular inflammation could explain up to 66% of the alterations in metrics of insulin resistance attributable to air pollution. CONCLUSION Our results suggest that ambient traffic pollution exposure was capable of promoting insulin resistance possibly via generating vascular inflammation.
Collapse
Affiliation(s)
- Hongbing Xu
- Department of Occupational and Environmental Health Sciences, Peking University School of Public Health, And Peking University Institute of Environmental Medicine, Beijing, China; Key Laboratory of Molecular Cardiovascular Sciences of the Ministry of Education, Beijing, China
| | - Shengcong Liu
- Division of Cardiology, Peking University First Hospital, Beijing, China; Key Laboratory of Molecular Cardiovascular Sciences of the Ministry of Education, Beijing, China
| | - Yang Wang
- Department of Prevention and Health Care, Hospital of Health Science Center, Peking University, Beijing, China
| | - Rongshan Wu
- Department of Occupational and Environmental Health Sciences, Peking University School of Public Health, And Peking University Institute of Environmental Medicine, Beijing, China; State Key Laboratory of Environmental Criteria and Risk Assessment, State Environmental Protection Key Laboratory of Ecological Effect and Risk Assessment of Chemicals, Chinese Research Academy of Environmental Sciences, Beijing, China
| | - Tieci Yi
- Division of Cardiology, Peking University First Hospital, Beijing, China; Key Laboratory of Molecular Cardiovascular Sciences of the Ministry of Education, Beijing, China
| | - Tong Wang
- Department of Occupational and Environmental Health Sciences, Peking University School of Public Health, And Peking University Institute of Environmental Medicine, Beijing, China; Key Laboratory of Molecular Cardiovascular Sciences of the Ministry of Education, Beijing, China
| | - Yutong Zhu
- Department of Occupational and Environmental Health Sciences, Peking University School of Public Health, And Peking University Institute of Environmental Medicine, Beijing, China; Key Laboratory of Molecular Cardiovascular Sciences of the Ministry of Education, Beijing, China
| | - Jiakun Fang
- Department of Occupational and Environmental Health Sciences, Peking University School of Public Health, And Peking University Institute of Environmental Medicine, Beijing, China; Key Laboratory of Molecular Cardiovascular Sciences of the Ministry of Education, Beijing, China
| | - Yunfei Xie
- Department of Occupational and Environmental Health Sciences, Peking University School of Public Health, And Peking University Institute of Environmental Medicine, Beijing, China; Key Laboratory of Molecular Cardiovascular Sciences of the Ministry of Education, Beijing, China
| | - Qian Zhao
- Department of Occupational and Environmental Health Sciences, Peking University School of Public Health, And Peking University Institute of Environmental Medicine, Beijing, China; Key Laboratory of Molecular Cardiovascular Sciences of the Ministry of Education, Beijing, China
| | - Xiaoming Song
- Department of Occupational and Environmental Health Sciences, Peking University School of Public Health, And Peking University Institute of Environmental Medicine, Beijing, China; Key Laboratory of Molecular Cardiovascular Sciences of the Ministry of Education, Beijing, China
| | - Jie Chen
- Department of Occupational and Environmental Health Sciences, Peking University School of Public Health, And Peking University Institute of Environmental Medicine, Beijing, China; Institute for Risk Assessment Sciences, University Medical Centre Utrecht, University of Utrecht, the Netherlands
| | - Sanjay Rajagopaplan
- Division of Cardiovascular Medicine, Case Western Reserve University, Ohio, USA
| | - Robert D Brook
- Division of Cardiovascular Medicine, University of Michigan, Michigan, USA
| | - Jianping Li
- Division of Cardiology, Peking University First Hospital, Beijing, China; Key Laboratory of Molecular Cardiovascular Sciences of the Ministry of Education, Beijing, China
| | - Junji Cao
- Institute of Atmospheric Physics Chinese Academy of Sciences, Beijing, China.
| | - Wei Huang
- Department of Occupational and Environmental Health Sciences, Peking University School of Public Health, And Peking University Institute of Environmental Medicine, Beijing, China; Key Laboratory of Molecular Cardiovascular Sciences of the Ministry of Education, Beijing, China.
| |
Collapse
|
9
|
Kandikattu HK, Venkateshaiah SU, Mishra A. Chronic Pancreatitis and the Development of Pancreatic Cancer. Endocr Metab Immune Disord Drug Targets 2021; 20:1182-1210. [PMID: 32324526 DOI: 10.2174/1871530320666200423095700] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 12/31/2019] [Accepted: 01/20/2020] [Indexed: 02/07/2023]
Abstract
Pancreatitis is a fibro-inflammatory disorder of the pancreas that can occur acutely or chronically as a result of the activation of digestive enzymes that damage pancreatic cells, which promotes inflammation. Chronic pancreatitis with persistent fibro-inflammation of the pancreas progresses to pancreatic cancer, which is the fourth leading cause of cancer deaths across the globe. Pancreatic cancer involves cross-talk of inflammatory, proliferative, migratory, and fibrotic mechanisms. In this review, we discuss the role of cytokines in the inflammatory cell storm in pancreatitis and pancreatic cancer and their role in the activation of SDF1α/CXCR4, SOCS3, inflammasome, and NF-κB signaling. The aberrant immune reactions contribute to pathological damage of acinar and ductal cells, and the activation of pancreatic stellate cells to a myofibroblast-like phenotype. We summarize several aspects involved in the promotion of pancreatic cancer by inflammation and include a number of regulatory molecules that inhibit that process.
Collapse
Affiliation(s)
- Hemanth K Kandikattu
- Department of Medicine, Tulane Eosinophilic Disorders Centre (TEDC), Section of Pulmonary Diseases, Tulane University School of Medicine, New Orleans, LA 70112, United States
| | - Sathisha U Venkateshaiah
- Department of Medicine, Tulane Eosinophilic Disorders Centre (TEDC), Section of Pulmonary Diseases, Tulane University School of Medicine, New Orleans, LA 70112, United States
| | - Anil Mishra
- Department of Medicine, Tulane Eosinophilic Disorders Centre (TEDC), Section of Pulmonary Diseases, Tulane University School of Medicine, New Orleans, LA 70112, United States
| |
Collapse
|
10
|
Exosomes as Pleiotropic Players in Pancreatic Cancer. Biomedicines 2021; 9:biomedicines9030275. [PMID: 33803470 PMCID: PMC8002012 DOI: 10.3390/biomedicines9030275] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 03/03/2021] [Accepted: 03/05/2021] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer (PC) incidence is rising and due to late diagnosis, combined with unsatisfactory response to current therapeutic approaches, this tumor has an extremely high mortality rate. A better understanding of the mechanisms underlying pancreatic carcinogenesis is of paramount importance for rational diagnostic and therapeutic approaches. Multiple lines of evidence have showed that exosomes are actively involved in intercellular communication by transferring their cargos of bioactive molecules to recipient cells within the tumor microenvironment and systemically. Intriguingly, exosomes may exert both protumor and antitumor effects, supporting or hampering processes that play a role in the pathogenesis and progression of PC, including shifts in tumor metabolism, proliferation, invasion, metastasis, and chemoresistance. They also have a dual role in PC immunomodulation, exerting immunosuppressive or immune enhancement effects through several mechanisms. PC-derived exosomes also induce systemic metabolic alterations, leading to the onset of diabetes and weight loss. Moreover, exosomes have been described as promising diagnostic and prognostic biomarkers for PC. Their potential application in PC therapy as drug carriers and therapeutic targets is under investigation. In this review, we provide an overview of the multiple roles played by exosomes in PC biology through their specific cargo biomolecules and of their potential exploitation in early diagnosis and treatment of PC.
Collapse
|
11
|
Wu N, Xu XF, Xin JQ, Fan JW, Wei YY, Peng QX, Duan LF, Wang W, Zhang H. The effects of nuclear factor-kappa B in pancreatic stellate cells on inflammation and fibrosis of chronic pancreatitis. J Cell Mol Med 2020; 25:2213-2227. [PMID: 33377616 PMCID: PMC7882951 DOI: 10.1111/jcmm.16213] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 11/13/2020] [Accepted: 11/20/2020] [Indexed: 02/06/2023] Open
Abstract
The activation of pancreatic stellate cells (PSCs) plays a critical role in the progression of pancreatic fibrosis. Nuclear factor‐kappa B (NF‐κB) is associated with chronic pancreatitis (CP). Previous evidence indicated that NF‐κB in acinar cells played a double‐edged role upon pancreatic injury, whereas NF‐κB in inflammatory cells promoted the progression of CP. However, the effects of NF‐κB in PSCs have not been studied. In the present study, using two CP models and RNAi strategy of p65 in cultured PSCs, we found that the macrophage infiltration and MCP‐1 expression were increased, and the NF‐κBp65 protein level was elevated. NF‐κBp65 was co‐expressed with PSCs. In vitro, TGF‐β1 induced overexpression of the TGF‐β receptor 1, phosphorylated TGF‐β1–activated kinase 1 (p‐TAK1) and NF‐κB in the PSCs. Moreover, the concentration of MCP‐1 in the supernatant of activated PSCs was elevated. The migration of BMDMs was promoted by the supernatant of activated PSCs. Further knockdown of NF‐κBp65 in PSCs resulted in a decline of BMDM migration, accompanied by a lower production of MCP‐1. These findings indicate that TGF‐β1 can induce the activation of NF‐κB pathway in PSCs by regulating p‐TAK1, and the NF‐κB pathway in PSCs may be a target of chronic inflammation and fibrosis.
Collapse
Affiliation(s)
- Nan Wu
- Department of Pathophysiology, Shaanxi University of Chinese Medicine, Xi'an, China
| | - Xiao-Fan Xu
- Medical Experiment Center, Shaanxi University of Chinese Medicine, Xi'an, China.,Ningxia Medical University, Yinchuan, China
| | - Jia-Qi Xin
- Department of Pathophysiology, Shaanxi University of Chinese Medicine, Xi'an, China
| | - Jian-Wei Fan
- Department of Pathophysiology, Shaanxi University of Chinese Medicine, Xi'an, China
| | - Yuan-Yuan Wei
- Department of Pathophysiology, Shaanxi University of Chinese Medicine, Xi'an, China
| | - Qing-Xia Peng
- Department of Pathophysiology, Shaanxi University of Chinese Medicine, Xi'an, China
| | - Li-Fang Duan
- Department of Pathophysiology, Shaanxi University of Chinese Medicine, Xi'an, China
| | - Wei Wang
- Department of General Surgery & Research Institute of Pancreatic Disease, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hong Zhang
- Department of Pathophysiology, Shaanxi University of Chinese Medicine, Xi'an, China
| |
Collapse
|
12
|
Aujla PK, Kassiri Z. Diverse origins and activation of fibroblasts in cardiac fibrosis. Cell Signal 2020; 78:109869. [PMID: 33278559 DOI: 10.1016/j.cellsig.2020.109869] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 11/30/2020] [Accepted: 12/01/2020] [Indexed: 12/21/2022]
Abstract
Cardiac fibroblasts (cFBs) have emerged as a heterogenous cell population. Fibroblasts are considered the main cell source for synthesis of the extracellular matrix (ECM) and as such a dysregulation in cFB function, activity, or viability can lead to disrupted ECM structure or fibrosis. Fibrosis can be initiated in response to different injuries and stimuli, and can be reparative (beneficial) or reactive (damaging). FBs need to be activated to myofibroblasts (MyoFBs) which have augmented capacity in synthesizing ECM proteins, causing fibrosis. In addition to the resident FBs in the myocardium, a number of other cells (pericytes, fibrocytes, mesenchymal, and hematopoietic cells) can transform into MyoFBs, further driving the fibrotic response. Multiple molecules including hormones, cytokines, and growth factors stimulate this process leading to generation of activated MyoFBs. Contribution of different cell types to cFBs and MyoFBs can result in an exponential increase in the number of MyoFBs and an accelerated pro-fibrotic response. Given the diversity of the cell sources, and the array of interconnected signalling pathways that lead to formation of MyoFBs and subsequently fibrosis, identifying a single target to limit the fibrotic response in the myocardium has been challenging. This review article will delineate the importance and relevance of fibroblast heterogeneity in mediating fibrosis in different models of heart failure and will highlight important signalling pathways implicated in myofibroblast activation.
Collapse
Affiliation(s)
- Preetinder K Aujla
- Department of Physiology, Cardiovascular Research Center, University of Alberta, Edmonton, Alberta, Canada
| | - Zamaneh Kassiri
- Department of Physiology, Cardiovascular Research Center, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
13
|
Roife D, Sarcar B, Fleming JB. Stellate Cells in the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1263:67-84. [PMID: 32588324 DOI: 10.1007/978-3-030-44518-8_6] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
As tumor microenvironments share many of the same qualities as chronic wounds, attention is turning to the wound-repair cells that support the growth of cancerous cells. Stellate cells are star-shaped cells that were first discovered in the perisinusoidal spaces in the liver and have been found to support wound healing by the secretion of growth factors and extracellular matrix. They have since been also found to serve a similar function in the pancreas. In both organs, the wound-healing process may become dysregulated and lead to pathological fibrosis (also known as cirrhosis in the liver). In recent years there has been increasing attention paid to the role of these cells in tumor formation and progression. They may be a factor in initiating the first steps of carcinogenesis such as with liver cirrhosis and hepatocellular carcinoma and also contribute to continued tumor growth, invasion, metastasis, evasion of the immune system, and resistance to chemotherapy, in cancers of both the liver and pancreas. In this chapter we aim to review the structure and function of hepatic and pancreatic stellate cells and their contributions to the tumor microenvironment in their respective cancers and also discuss potential new targets for cancer therapy based on our new understanding of these vital components of the tumor stroma.
Collapse
Affiliation(s)
- David Roife
- Department of Surgery, University of South Florida Morsani College of Medicine, Tampa, FL, USA.,Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Bhaswati Sarcar
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Jason B Fleming
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center, Tampa, FL, USA.
| |
Collapse
|
14
|
Polet M, Laloux L, Cambier S, Ziebel J, Gutleb AC, Schneider YJ. Soluble silver ions from silver nanoparticles induce a polarised secretion of interleukin-8 in differentiated Caco-2 cells. Toxicol Lett 2020; 325:14-24. [PMID: 32062016 DOI: 10.1016/j.toxlet.2020.02.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 02/02/2020] [Accepted: 02/04/2020] [Indexed: 12/18/2022]
Abstract
Because of their antimicrobial properties, silver nanoparticles are increasingly incorporated in food-related and hygiene products, which thereby could lead to their ingestion. Although their cytotoxicity mediated by oxidative stress has been largely studied, their effects on inflammation remain controversial. Moreover, the involvement of silver ions (originating from Ag0 oxidation) in their mode of action is still unclear. In this context, the present study aims at assessing the impact of silver nanoparticles on the secretion of the pro-inflammatory chemokine interleukin-8 by Caco-2 cells forming an in vitro model of the intestinal mucosal barrier. Silver nanoparticles induced a vectorized secretion of interleukin-8 towards the apical compartment, which is found in the medium 21 h after the incubation. This secretion seems mediated by Nrf2 signalling pathway that orchestrates cellular defense against oxidative stress. The soluble silver fraction of silver nanoparticles suspensions led to a similar amount of secreted interleukin-8 than silver nanoparticles, suggesting an involvement of silver ions in this interleukin-8 secretion.
Collapse
Affiliation(s)
- Madeleine Polet
- Louvain Institute of Biomolecular Science and Technology (LIBST), Université catholique de Louvain (UCLouvain), Louvain-la-Neuve, Belgium
| | - Laurie Laloux
- Louvain Institute of Biomolecular Science and Technology (LIBST), Université catholique de Louvain (UCLouvain), Louvain-la-Neuve, Belgium
| | - Sébastien Cambier
- Environmental Research and Innovation (ERIN) Department, Luxembourg Institute of Science and Technology (LIST), Esch/Alzette, Luxembourg
| | - Johanna Ziebel
- Environmental Research and Innovation (ERIN) Department, Luxembourg Institute of Science and Technology (LIST), Esch/Alzette, Luxembourg
| | - Arno C Gutleb
- Environmental Research and Innovation (ERIN) Department, Luxembourg Institute of Science and Technology (LIST), Esch/Alzette, Luxembourg
| | - Yves-Jacques Schneider
- Louvain Institute of Biomolecular Science and Technology (LIBST), Université catholique de Louvain (UCLouvain), Louvain-la-Neuve, Belgium.
| |
Collapse
|
15
|
CpG-ODN Induces a Dose-Dependent Enrichment of Immunological Niches in the Spleen and Lungs of Neonatal Chicks That Correlates with the Protective Immunity against Escherichia coli. J Immunol Res 2020; 2020:2704728. [PMID: 32411791 PMCID: PMC7201825 DOI: 10.1155/2020/2704728] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 12/05/2019] [Indexed: 01/25/2023] Open
Abstract
Immunoprotective function of oligodeoxynucleotides containing CpG motifs (CpG-ODN) has been demonstrated in neonatal chickens against common bacterial pathogens such as E.coli and Salmonella sp. Our recent study reported that CpG-ODN administration enriches immune compartments in neonatal chicks. However, a causal relationship between CpG-ODN-induced immune enrichment and protective mechanisms remains unestablished. In this study, we investigated in ovo administered CpG-ODN-mediated immune cell recruitment in the immunological niches in lymphoid (spleen) and nonlymphoid (lungs) organs using various doses of CpG-ODN and examined whether the immunological profiles have any correlation with immunoprotection against E.coli infection. Eighteen-day-old embryonated eggs were injected with either 5, 10, 25, and 50 μg of CpG-ODN or saline (n = ~40 per group). On the day of hatch (72 hr after CpG-ODN treatment), we collected the spleen and lungs (n = 3‐4 per group) and examined the recruitment of macrophages/monocytes, their expression of MHCII and CD40, and the number of CD4+ and CD8+ T-cell subsets in the immunological niches in the spleen and lungs using flow cytometry. We observed the dose-dependent recruitment of immune cells, wherein 25 μg and 50 μg of CpG-ODN induced significant enrichment of immunological niches in both the spleen and the lungs. Four days after the CpG-ODN treatment (1-day after hatch), chicks were challenged with a virulent strain of E. coli (1 × 104 or 1 × 105 cfu, subcutaneously). Clinical outcome and mortality were monitored for 8 days postchallenge. We found that both 25 μg and 50 μg of CpG-ODN provided significant protection and reduced clinical scores compared to saline controls against E. coli infection. Overall, the present study revealed that CpG-ODNs orchestrate immunological niches in neonatal chickens in a dose-dependent manner that resulted in differential protection against E. coli infection, thus supporting a cause and effect relationship between CpG-ODN-induced immune enrichment and the antibacterial immunity.
Collapse
|
16
|
Fan J, Duan L, Wu N, Xu X, Xin J, Jiang S, Zhang C, Zhang H. Baicalin Ameliorates Pancreatic Fibrosis by Inhibiting the Activation of Pancreatic Stellate Cells in Mice with Chronic Pancreatitis. Front Pharmacol 2020; 11:607133. [PMID: 33536916 PMCID: PMC7848203 DOI: 10.3389/fphar.2020.607133] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 12/02/2020] [Indexed: 02/05/2023] Open
Abstract
Pancreatic inflammation and fibrosis are typical pathological features in chronic pancreatitis (CP). Activated pancreatic stellate cells (PSCs) have been regarded as the core event in the development of pancreatic fibrosis and are considered to be the key target for treatment of CP. Baicalin (C21H18O11), the main chemical composition of Baikal skullcap in the traditional Chinese medicines Dachaihu decoction (DCHD) and Xiaochaihu decoction (XCHD), has shown significant effects in the treatment of pancreatic fibrosis in CP mice; however, whether baicalin can inhibit the activation of PSCs and its underlying mechanism remain unclear. In this study, the influence of baicalin on activated PSCs in vitro and in vivo was investigated, and the results showed that Baicalin could significantly ameliorate the degree of pancreatic inflammation and fibrosis, while decreasing the levels of alpha-smooth muscle actin (α-SMA), F4/80 (surface markers of mouse macrophages), nuclear factor kappa-B (NF-κB), monocyte chemotactic protein 1 (MCP-1), and collagen type I alpha 1 (COL1A1)in the pancreas. Moreover, NF-κB and α-SMA were co-expressed in the pancreas of CP mice. Baicalin treatment markedly reduced the expression of co-location of α-SMA and NF-κB. In vitro, the protein expression levels of transforming growth factor-β receptor 1 (TGF-βR1), phosphorylated TGF-β activated kinase 1 p-TAK 1, and NF-κBp65 in PSCs were all remarkably reduced after treatment with baicalin. In addition, baicalin could inhibit MCP-1 mRNA expression in supernatant of activated PSCs, as well as the excessive migration of macrophages. Taken together, our findings indicated that baicalin could inhibit the TGF-β1/TGF-βR1/TAK1/NF-κB signaling pathway of activated PSCs, reduce the secretion of MCP-1, and further decrease the infiltration of macrophages and inflammation cells of the local microenvironment of the pancreas. Thus, this study provides a reliable experimental basis for baicalin in the prevention and treatment of CP.
Collapse
Affiliation(s)
- Jianwei Fan
- Basic Medical Academy, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Lifang Duan
- Basic Medical Academy, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Nan Wu
- Basic Medical Academy, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Xiaofan Xu
- Medical Experiment Center, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Jiaqi Xin
- Basic Medical Academy, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Shengnan Jiang
- Basic Medical Academy, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Cheng Zhang
- Department of Hepatobiliary Surgery, Xianyang Central Hospital, Xianyang, China
- *Correspondence: Cheng Zhang, ; Hong Zhang,
| | - Hong Zhang
- Basic Medical Academy, Shaanxi University of Chinese Medicine, Xianyang, China
- Medical Experiment Center, Shaanxi University of Chinese Medicine, Xianyang, China
- *Correspondence: Cheng Zhang, ; Hong Zhang,
| |
Collapse
|
17
|
Pandey V, Storz P. Targeting the tumor microenvironment in pancreatic ductal adenocarcinoma. Expert Rev Anticancer Ther 2019; 19:473-482. [PMID: 31148495 PMCID: PMC6548630 DOI: 10.1080/14737140.2019.1622417] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 05/20/2019] [Indexed: 12/18/2022]
Abstract
Introduction: The dismally slow improvement in patient survival over the years for pancreatic cancer patients is mainly due to two factors: the late diagnosis, at which point the disease is spread to distant organs; and the fact that tumor cells are surrounded by a dense, highly immunosuppressive microenvironment. The tumor microenvironment not only shields pancreatic cancer cells from chemotherapy but also leaves it unsusceptible to various immunotherapeutic strategies that have been proven successful in other types of cancer. Areas covered: This review highlights the main components of the pancreatic tumor microenvironment, how they cross-talk with each other to generate stroma and promote tumor growth. Additionally, we discuss the most promising treatment targets in the microenvironment whose modulation can be robustly tested in combination with standard of care chemotherapy. Currently, active clinical trials for pancreatic cancer involving components of the microenvironment are also listed. Expert opinion: Although immunotherapeutic approaches involving checkpoint inhibition are being pursued enthusiastically, there is still more work to be done with several other emerging immune targets that could provide therapeutic benefit.
Collapse
Affiliation(s)
- Veethika Pandey
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, USA
| | - Peter Storz
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, USA
| |
Collapse
|
18
|
Gunawardana T, Ahmed KA, Goonewardene K, Popowich S, Kurukulasuriya S, Karunarathna R, Gupta A, Lockerbie B, Foldvari M, Tikoo SK, Willson P, Gomis S. Synthetic CpG-ODN rapidly enriches immune compartments in neonatal chicks to induce protective immunity against bacterial infections. Sci Rep 2019; 9:341. [PMID: 30674918 PMCID: PMC6344490 DOI: 10.1038/s41598-018-36588-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 11/19/2018] [Indexed: 12/13/2022] Open
Abstract
Oligodeoxynucleotides containing CpG motifs (CpG-ODN) induce innate immunity against bacterial infections. Despite recent advances, how CpG-ODN alone protects against bacterial infections remained elusive. Here, we report for the first time, to our knowledge, that CpG-ODN orchestrates anti-microbial protective immunity by inducing a rapid enrichment of various immune compartments in chickens. In this study, eighteen-day-old embryonated eggs were injected with either 50 µg of CpG-ODN or saline (~n = 90 per group). In the first experiment, four days after CpG-ODN treatment, chicks were challenged subcutaneously with a virulent strain of Escherichia coli (E. coli) and mortality was monitored for 8 days. We found significant protection, and reduced clinical scores in CpG-ODN treated chicks. To gain insights into mechanisms of protection induced by CpG-ODN, first we investigated cytokine expression kinetics elicited by CpG-ODN. The spleen and lung were collected from embryos or chicks (n = 3-4 per group) at 10 time points post-CpG-ODN inoculation. Multiplex gene analysis (interleukin (IL)-1, IL-4, IL-6, IL-10, IL-18, interferon (IFN)-γ, IFN-α, and lipopolysaccharide induced tumor necrosis factor (LITAF), revealed a significantly higher expression of pro-inflammatory cytokines following CpG-ODN treatment compared to the saline controls. In our study, LITAF stands out in the cytokine profiles of spleen and lungs, underscoring its role in CpG-ODN-induced protection. The third experiment was designed to examine the effects of CpG-ODN on immune cell populations in spleen, lungs, and thymus. Flow cytometry analysis was conducted at 24, 48 and 72 hrs (thymus only collected at 72 hr) after CpG-ODN administration to examine the changes in CD4+ and CD8+ T-cell subsets, monocyte/macrophage cell populations and their expression of maturation markers (CD40 and CD86). Flow cytometry data indicated a significant enrichment of macrophages, CD4+ and CD8+ T-cell subsets in both spleen and lungs of CpG-ODN treated embryos and chicks. Macrophages in spleen and lungs showed an upregulation of CD40 but not CD86, whereas thymocytes revealed significantly high CD4 and CD8 expression. Overall, the present study has demonstrated that CpG-ODN provides protection in neonatal chicks against E. coli infection not only by eliciting cytokine responses and stimulating immune cells but also through enriching immunological niches in spleen and lungs.
Collapse
Affiliation(s)
- Thushari Gunawardana
- Department of Veterinary Pathology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK, S7N 5B4, Canada
| | - Khawaja Ashfaque Ahmed
- Department of Veterinary Pathology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK, S7N 5B4, Canada.
| | - Kalhari Goonewardene
- Department of Veterinary Pathology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK, S7N 5B4, Canada
| | - Shelly Popowich
- Department of Veterinary Pathology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK, S7N 5B4, Canada
| | - Shanika Kurukulasuriya
- Department of Veterinary Pathology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK, S7N 5B4, Canada
| | - Ruwani Karunarathna
- Department of Veterinary Pathology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK, S7N 5B4, Canada
| | - Ashish Gupta
- Department of Veterinary Pathology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK, S7N 5B4, Canada
| | - Betty Lockerbie
- Department of Veterinary Pathology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK, S7N 5B4, Canada
| | - Marianna Foldvari
- School of Pharmacy, University of Waterloo, 200 University Avenue West, Waterloo, ON, N2L 3G1, Canada
| | - Suresh K Tikoo
- Vaccinology and Immunotherapy, School of Public Health, University of Saskatchewan, Saskatoon, SK, S7N 5E3, Canada
| | - Philip Willson
- Canadian Centre for Health and Safety in Agriculture, University of Saskatchewan, Saskatoon, SK, S7N 5E5, Canada
| | - Susantha Gomis
- Department of Veterinary Pathology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK, S7N 5B4, Canada.
| |
Collapse
|
19
|
Cullis J, Das S, Bar-Sagi D. Kras and Tumor Immunity: Friend or Foe? Cold Spring Harb Perspect Med 2018; 8:cshperspect.a031849. [PMID: 29229670 DOI: 10.1101/cshperspect.a031849] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
With the recent breakthroughs in immunotherapy as curative treatments in certain tumor types, there has been renewed interest in the relationship between immunity and tumor growth. Although we are gaining a greater understanding of the complex interplay of immune modulating components in the tumor microenvironment, the specific role that tumor cells play in shaping the immune milieu is still not well characterized. In this review, we focus on how mutant Kras tumor cells contribute to tumor immunity, with a specific focus on processes induced directly or indirectly by the oncogene.
Collapse
Affiliation(s)
- Jane Cullis
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, New York 10016
| | - Shipra Das
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, New York 10016
| | - Dafna Bar-Sagi
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, New York 10016
| |
Collapse
|
20
|
Vazquez Rodriguez G, Abrahamsson A, Jensen LDE, Dabrosin C. Adipocytes Promote Early Steps of Breast Cancer Cell Dissemination via Interleukin-8. Front Immunol 2018; 9:1767. [PMID: 30105032 PMCID: PMC6077262 DOI: 10.3389/fimmu.2018.01767] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 07/17/2018] [Indexed: 12/21/2022] Open
Abstract
Fat is a major tissue component in human breast cancer (BC). Whether breast adipocytes (BAd) affect early stages of BC metastasis is yet unknown. BC progression is dependent on angiogenesis and inflammation, and interleukin-8 (IL-8) and vascular endothelial growth factor (VEGF) are key regulators of these events. Here, we show that BAd increased the dissemination of estrogen receptor positive BC cells (BCC) in vivo in the zebrafish model of metastasis, while dissemination of the more aggressive and metastatic BCC such as estrogen receptor negative was unaffected. While anti-VEGF and anti-IL-8 exhibited equal inhibition of angiogenesis at the primary tumor site, anti-IL-8 reduced BCC dissemination whereas anti-VEGF had minor effects on this early metastatic event. Mechanistically, overexpression of cell-adhesion molecules in BCC and neutrophils via IL-8 increased the dissemination of BCC. Importantly, the extracellular in vivo levels of IL-8 were 40-fold higher than those of VEGF in human BC. Our results suggest that IL-8 is a clinical relevant and promising therapeutic target for human BC.
Collapse
Affiliation(s)
- Gabriela Vazquez Rodriguez
- Department of Oncology, Linköping University, Linköping, Sweden.,Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Annelie Abrahamsson
- Department of Oncology, Linköping University, Linköping, Sweden.,Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Lasse Dahl Ejby Jensen
- Department of Medical and Health Sciences, Division of Cardiovascular Medicine, Linköping University, Linköping, Sweden
| | - Charlotta Dabrosin
- Department of Oncology, Linköping University, Linköping, Sweden.,Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| |
Collapse
|
21
|
Abstract
Stellate cells are resident lipid-storing cells of the pancreas and liver that transdifferentiate to a myofibroblastic state in the context of tissue injury. Beyond having roles in tissue homeostasis, stellate cells are increasingly implicated in pathological fibrogenic and inflammatory programs that contribute to tissue fibrosis and that constitute a growth-permissive tumor microenvironment. Although the capacity of stellate cells for extracellular matrix production and remodeling has long been appreciated, recent research efforts have demonstrated diverse roles for stellate cells in regulation of epithelial cell fate, immune modulation, and tissue health. Our present understanding of stellate cell biology in health and disease is discussed here, as are emerging means to target these multifaceted cells for therapeutic benefit.
Collapse
Affiliation(s)
- Mara H Sherman
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, Oregon 97201, USA;
| |
Collapse
|
22
|
Tay C, Liu YH, Kanellakis P, Kallies A, Li Y, Cao A, Hosseini H, Tipping P, Toh BH, Bobik A, Kyaw T. Follicular B Cells Promote Atherosclerosis via T Cell–Mediated Differentiation Into Plasma Cells and Secreting Pathogenic Immunoglobulin G. Arterioscler Thromb Vasc Biol 2018; 38:e71-e84. [DOI: 10.1161/atvbaha.117.310678] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 03/14/2018] [Indexed: 12/20/2022]
Abstract
Objective—
B cells promote or protect development of atherosclerosis. In this study, we examined the role of MHCII (major histocompatibility II), CD40 (cluster of differentiation 40), and Blimp-1 (B-lymphocyte–induced maturation protein) expression by follicular B (FO B) cells in development of atherosclerosis together with the effects of IgG purified from atherosclerotic mice.
Approach and Results—
Using mixed chimeric
Ldlr
−/−
mice whose B cells are deficient in MHCII or CD40, we demonstrate that these molecules are critical for the proatherogenic actions of FO B cells. During development of atherosclerosis, these deficiencies affected T–B cell interactions, germinal center B cells, plasma cells, and IgG. As FO B cells differentiating into plasma cells require Blimp-1, we also assessed its role in the development of atherosclerosis. Blimp-1-deficient B cells greatly attenuated atherosclerosis and immunoglobulin—including IgG production, preventing IgG accumulation in atherosclerotic lesions; Blimp-1 deletion also attenuated lesion proinflammatory cytokines, apoptotic cell numbers, and necrotic core. To determine the importance of IgG for atherosclerosis, we purified IgG from atherosclerotic mice. Their transfer but not IgG from nonatherosclerotic mice into
Ldlr
−/−
mice whose B cells are Blimp-1-deficient increased atherosclerosis; transfer was associated with IgG accumulating in atherosclerotic lesions, increased lesion inflammatory cytokines, apoptotic cell numbers, and necrotic core size.
Conclusions—
The mechanism by which FO B cells promote atherosclerosis is highly dependent on their expression of MHCII, CD40, and Blimp-1. FO B cell differentiation into IgG-producing plasma cells also is critical for their proatherogenic actions. Targeting B–T cell interactions and pathogenic IgG may provide novel therapeutic strategies to prevent atherosclerosis and its adverse cardiovascular complications.
Collapse
Affiliation(s)
- Christopher Tay
- From the Vascular Biology and Atherosclerosis Lab, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (C.T., Y.-H.L., P.K., Y.L., A.C., H.H., A.B., T.K.)
| | - Yu-Han Liu
- From the Vascular Biology and Atherosclerosis Lab, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (C.T., Y.-H.L., P.K., Y.L., A.C., H.H., A.B., T.K.)
| | - Peter Kanellakis
- From the Vascular Biology and Atherosclerosis Lab, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (C.T., Y.-H.L., P.K., Y.L., A.C., H.H., A.B., T.K.)
| | - Axel Kallies
- Walter and Eliza Hall Institute, Parkville, Victoria, Australia (A.K.)
| | - Yi Li
- From the Vascular Biology and Atherosclerosis Lab, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (C.T., Y.-H.L., P.K., Y.L., A.C., H.H., A.B., T.K.)
| | - Anh Cao
- From the Vascular Biology and Atherosclerosis Lab, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (C.T., Y.-H.L., P.K., Y.L., A.C., H.H., A.B., T.K.)
| | - Hamid Hosseini
- From the Vascular Biology and Atherosclerosis Lab, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (C.T., Y.-H.L., P.K., Y.L., A.C., H.H., A.B., T.K.)
| | - Peter Tipping
- Department of Medicine, Centre for Inflammatory Diseases (P.T., B.-H.T., T.K)
| | - Ban-Hock Toh
- Department of Medicine, Centre for Inflammatory Diseases (P.T., B.-H.T., T.K)
| | - Alex Bobik
- From the Vascular Biology and Atherosclerosis Lab, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (C.T., Y.-H.L., P.K., Y.L., A.C., H.H., A.B., T.K.)
- Department of Immunology (A.B.), Monash University, Melbourne, Victoria, Australia
| | - Tin Kyaw
- From the Vascular Biology and Atherosclerosis Lab, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (C.T., Y.-H.L., P.K., Y.L., A.C., H.H., A.B., T.K.)
- Department of Medicine, Centre for Inflammatory Diseases (P.T., B.-H.T., T.K)
| |
Collapse
|
23
|
Um J, Jung N, Kim D, Choi S, Lee SH, Son Y, Park KS. Substance P preserves pancreatic β-cells in type 1 and type 2 diabetic mice. Biochem Biophys Res Commun 2018; 499:960-966. [PMID: 29626466 DOI: 10.1016/j.bbrc.2018.04.028] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 04/03/2018] [Indexed: 01/09/2023]
Abstract
Preservation of pancreatic β-cells is required for the development of therapies for type 1 and type 2 diabetes (T1D and T2D, respectively). Our previous study demonstrated that substance P (SP) preserves β-cell populations in mice with streptozotocin-induced T1D. Here, we demonstrated that chronic systemic treatment with SP restored the mass of β-cells both in nonobese diabetic (NOD) mice with T1D or db/db mice with T2D. SP delayed the onset of T1D in NOD mice via immune modulation. SP inhibited immune infiltration into islets and the salivary glands of NOD mice. In db/db mice, SP treatment rescued glucose intolerance. Moreover, SP inhibited apoptosis, as well as the activation of pancreatic stellate cells in pancreatic islets of db/db mice. SP downregulated the number of α-smooth muscle actin (α-SMA) expressing cells in db/db pancreatic islets. Cleaved-caspase-3 expression was reduced in islets of SP-treated db/db mice compared to that in the control. Therefore, these results suggested that SP may preserve pancreatic β-cells through immune modulation and protection from the stimulated activation of pancreatic stellate cells and apoptosis in T1D and T2D, respectively.
Collapse
Affiliation(s)
- Jihyun Um
- Graduate School of Biotechnology, Kyung Hee University, Yongin 17104, South Korea
| | - Nunggum Jung
- Graduate School of Biotechnology, Kyung Hee University, Yongin 17104, South Korea
| | - Dongjin Kim
- Graduate School of Biotechnology, Kyung Hee University, Yongin 17104, South Korea
| | - Sanghyuk Choi
- Graduate School of Biotechnology, Kyung Hee University, Yongin 17104, South Korea
| | - Sang-Ho Lee
- Kyung Hee University Hospital at Gangdong, Seoul 05278, South Korea
| | - Youngsook Son
- Graduate School of Biotechnology, Kyung Hee University, Yongin 17104, South Korea
| | - Ki-Sook Park
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, Seoul 02447, South Korea; East-West Medical Research Institute, Kyung Hee University, Seoul 02447, South Korea; College of Medicine, Kyung Hee University, Seoul 02447, South Korea; Kyung Hee University Medical Center, Seoul 02447, South Korea.
| |
Collapse
|
24
|
Zhao W, Ajani JA, Sushovan G, Ochi N, Hwang R, Hafley M, Johnson RL, Bresalier RS, Logsdon CD, Zhang Z, Song S. Galectin-3 Mediates Tumor Cell-Stroma Interactions by Activating Pancreatic Stellate Cells to Produce Cytokines via Integrin Signaling. Gastroenterology 2018; 154:1524-1537.e6. [PMID: 29274868 DOI: 10.1053/j.gastro.2017.12.014] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 11/22/2017] [Accepted: 12/18/2017] [Indexed: 12/19/2022]
Abstract
BACKGROUND & AIMS Pancreatic ductal adenocarcinoma (PDAC) is characterized by activated pancreatic stellate cells (PSCs), abundance of extracellular matrix (ECM), and production of cytokines and chemokines. Galectin 3 (GAL3), a β-galactoside-specific lectin, contributes to PDAC development but its effects on the stroma and cytokine production are unclear. METHODS The effect of recombinant human GAL3 (rGAL3) on activation of PSCs, production of cytokines, and ECM proteins was determined by proliferation, invasion, cytokine array, and quantitative polymerase chain reaction. We assessed co-cultures of PDAC cells with GAL3 genetic alterations with PSCs. Production of interleukin 8 (IL8) and activities of nuclear factor (NF)-κB were determined by enzyme-linked immunosorbent assay and luciferase reporter analyses. We studied the effects of inhibitors of NF-κB and integrin-linked kinase (ILK) on pathways activated by rGAL3. RESULTS In analyses of the Gene Expression Omnibus database and our dataset, we observed higher levels of GAL3, IL8, and other cytokines in PDAC than in nontumor tissues. Production of IL8, granulocyte-macrophage colony-stimulating factor, chemokine ligand 1, and C-C motif chemokine ligand 2 increased in PSCs exposed to rGAL3 compared with controls. Culture of PSCs with PDAC cells that express different levels of GAL3 resulted in proliferation and invasion of PSCs that increased with level of GAL3. GAL3 stimulated transcription of IL8 through integrin subunit beta 1 (ITGB1) on PSCs, which activates NF-κB through ILK. Inhibitors of ILK or NF-κB or a neutralizing antibody against ITGB1 blocked transcription and production of IL8 from PSCs induced by rGAL3. The GAL3 inhibitor significantly reduced growth and metastases of orthotopic tumors that formed from PDAC and PSC cells co-implanted in mice. CONCLUSION GAL3 activates PSC cells to produce inflammatory cytokines via ITGB1signaling to ILK and activation of NF-κB. Inhibition of this pathway reduced growth and metastases of pancreatic orthotopic tumors in mice.
Collapse
Affiliation(s)
- Wei Zhao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education) and Department of Cell Biology, Peking University Cancer Hospital and Institute, Beijing, People's Republic of China; Department of Gastrointestinal Medical Oncology, University of Texas M.D. Anderson Cancer Center, Houston, Texas.
| | - Jaffer A Ajani
- Department of Gastrointestinal Medical Oncology, University of Texas M.D. Anderson Cancer Center, Houston, Texas.
| | - Guha Sushovan
- Department of Gastroenterology, Hepatology, and Nutrition, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Nobuo Ochi
- Department of Gastroenterology, Hepatology, and Nutrition, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Rosa Hwang
- Department of Breast Surgical Oncology, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Margarete Hafley
- Department of Gastroenterology, Hepatology, and Nutrition, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Randy L Johnson
- Department of Cancer Biology, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Robert S Bresalier
- Department of Gastroenterology, Hepatology, and Nutrition, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Craig D Logsdon
- Department of Cancer Biology, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Zhiqian Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education) and Department of Cell Biology, Peking University Cancer Hospital and Institute, Beijing, People's Republic of China
| | - Shumei Song
- Department of Gastrointestinal Medical Oncology, University of Texas M.D. Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
25
|
Song H, Zhang Y. Regulation of pancreatic stellate cell activation by Notch3. BMC Cancer 2018; 18:36. [PMID: 29304760 PMCID: PMC5756326 DOI: 10.1186/s12885-017-3957-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 12/21/2017] [Indexed: 01/18/2023] Open
Abstract
Background Activated pancreatic stellate cells (PaSCs) are the key cellular source of cancer-associated fibroblasts in the pancreatic stroma of patients with pancreatic ductal adenocarcinoma (PDAC), however, the activation mechanism of PaSCs is not yet known. The Notch signaling pathway, components of which are expressed in stromal cells, is involved in the fibrosis of several organs, including the lung and liver. In the current study, we investigated whether Notch signal transduction is involved in PaSC activation in PDAC. Methods The expression of Notch signaling pathway components in human PDAC was examined via immunohistochemical staining and assessed in mouse PaSCs using RT-qPCR and western blotting. Notch3 expression in both PDAC stromal cells and activated mouse PaSCs was evaluated using immunofluorescence, RT-qPCR and western blotting. The impact of siRNA-mediated Notch3 knockdown on PaSC activation was detected with RT-qPCR and western blotting, and the impact on PaSC proliferation and migration was detected using CCK-8 assays and scratch experiments. The effect of conditioned medium from PaSCs activated with Notch3 siRNA on pancreatic cancer (LTPA) cells was also detected with CCK-8 assays and scratch experiments. The data were analyzed for statistical significance using Student’s t-test. Results Notch3 was overexpressed in both human PDAC stromal cells and activated mouse PaSCs, and Notch3 knockdown with Notch3 siRNA decreased the proliferation and migration of mouse PaSCs. The levels of markers related to PaSC activation, such as α-smooth muscle actin (α-SMA), collagen I and fibronectin, decreased in response to Notch3 knockdown, indicating that Notch3 plays an important role in PaSC activation. Furthermore, we confirmed that inhibition of PaSC activation via Notch3 siRNA reduced the proliferation and migration of PaSC-induced mouse pancreatic cancer (LTPA) cells. Conclusions Notch3 inhibition in PaSCs can inhibit the activation, proliferation and migration of PaSCs and reduce the PaSC-induced pro-tumorigenic effect. Therefore, Notch3 silencing in PaSCs is a potential novel therapeutic option for patients with PDAC. Electronic supplementary material The online version of this article (10.1186/s12885-017-3957-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Haiyan Song
- Department of Biochemistry and Molecular Biology, Cancer Institute, Beijing Key Laboratory for Cancer Invasion and Metastasis Research, Capital Medical University, No. 10 Xitoutiao, You An Men, Fengtai District, Beijing, 100069, People's Republic of China
| | - Yuxiang Zhang
- Department of Biochemistry and Molecular Biology, Cancer Institute, Beijing Key Laboratory for Cancer Invasion and Metastasis Research, Capital Medical University, No. 10 Xitoutiao, You An Men, Fengtai District, Beijing, 100069, People's Republic of China.
| |
Collapse
|
26
|
Wils LJ, Bijlsma MF. Epigenetic regulation of the Hedgehog and Wnt pathways in cancer. Crit Rev Oncol Hematol 2018; 121:23-44. [DOI: 10.1016/j.critrevonc.2017.11.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 11/17/2017] [Accepted: 11/17/2017] [Indexed: 12/14/2022] Open
|
27
|
Melstrom LG, Salazar MD, Diamond DJ. The pancreatic cancer microenvironment: A true double agent. J Surg Oncol 2017; 116:7-15. [PMID: 28605029 DOI: 10.1002/jso.24643] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 03/17/2017] [Indexed: 12/18/2022]
Abstract
The tumor microenvironment in pancreatic cancer is a complex balance of pro- and anti-tumor components. The dense desmoplasia consists of immune cells, extracellular matrix, growth factors, cytokines, and cancer associated fibroblasts (CAF) or pancreatic stellate cells (PSC). There are a multitude of targets including hyaluronan, angiogenesis, focal adhesion kinase (FAK), connective tissue growth factor (CTGF), CD40, chemokine (C-X-C motif) receptor 4 (CXCR-4), immunotherapy, and Vitamin D. The developing clinical therapeutics will be reviewed.
Collapse
Affiliation(s)
- Laleh G Melstrom
- Department of Surgery and Experimental Therapeutics, City of Hope National Medical Center, Duarte, California
| | - Marcela D Salazar
- Department of Experimental Therapeutics, City of Hope National Medical Center, Duarte, California
| | - Don J Diamond
- Department of Experimental Therapeutics, City of Hope National Medical Center, Duarte, California
| |
Collapse
|
28
|
Abstract
Chronic pancreatitis (CP) is a progressive inflammatory disease of the pancreas, leading to its fibrotic destruction. There are currently no drugs that can stop or slow the progression of the disease. The etiology of the disease is multifactorial, whereas recurrent attacks of acute pancreatitis are thought to precede the development of CP. A better understanding of the pathology of CP is needed to facilitate improved diagnosis and treatment strategies for this disease. The present paper develops a mathematical model of CP based on a dynamic network that includes macrophages, pancreatic stellate cells, and prominent cytokines that are present at high levels in the CP microenvironment. The model is represented by a system of partial differential equations. The model is used to explore in silico potential drugs that could slow the progression of the disease, for example infliximab (anti-TNF-[Formula: see text]) and tocilizumab or siltuximab (anti-IL-6/IL-6R).
Collapse
|
29
|
Ferdek PE, Jakubowska MA. Biology of pancreatic stellate cells-more than just pancreatic cancer. Pflugers Arch 2017; 469:1039-1050. [PMID: 28382480 PMCID: PMC5554282 DOI: 10.1007/s00424-017-1968-0] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 03/13/2017] [Accepted: 03/16/2017] [Indexed: 01/18/2023]
Abstract
Pancreatic stellate cells, normally quiescent, are capable of remarkable transition into their activated myofibroblast-like phenotype. It is now commonly accepted that these cells play a pivotal role in the desmoplastic reaction present in severe pancreatic disorders. In recent years, enormous scientific effort has been devoted to understanding their roles in pancreatic cancer, which continues to remain one of the most deadly diseases. Therefore, it is not surprising that considerably less attention has been given to studying physiological functions of pancreatic stellate cells. Here, we review recent advances not only in the field of pancreatic stellate cell pathophysiology but also emphasise their roles in physiological processes.
Collapse
Affiliation(s)
- Pawel E Ferdek
- Medical Research Council Group, Cardiff School of Biosciences, Cardiff University, Cardiff, Wales, CF10 3AX, UK.
| | - Monika A Jakubowska
- Medical Research Council Group, Cardiff School of Biosciences, Cardiff University, Cardiff, Wales, CF10 3AX, UK
| |
Collapse
|
30
|
Interleukin-36α Induces Inflammatory Mediators From Human Pancreatic Myofibroblasts Via a MyD88 Dependent Pathway. Pancreas 2017; 46:539-548. [PMID: 28099250 DOI: 10.1097/mpa.0000000000000765] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVES Interleukin-36 (IL-36) is a recently described proinflammatory cytokine, characterized by the induction of inflammatory mediators. In the present study, we investigated the biological activity and the signal transduction of IL-36α in human pancreatic myofibroblasts. METHODS The mRNA and protein expression of inflammatory mediators was evaluated using real-time polymerase chain reaction and enzyme-linked immunosorbent assay, respectively. The expression of IL-36α and its receptor in the pancreatic tissue was evaluated using immunohistochemical technique. Intracellular signaling pathways were evaluated using immunoblotting and specific small interference RNA-transfected cells. RESULTS Interleukin-36α and its receptor complex IL-36R/IL-1RAcP were detected in fibrotic tissue of chronic pancreatitis. Interleukin-36α dose- and time-dependently induced the mRNA expression and protein secretion of CXCL1, CXCL8, MMP-1, and MMP-3 from human pancreatic myofibroblasts. Interleukin-36α assembled MyD88 adaptor proteins (MyD88, TRAF6, IRAK1, and TAK1) into a complex. Furthermore, IL-36α induced the phosphorylation of mitogen-activated protein kinases and the activation of nuclear factor κB and activator protein 1. Mitogen-activated protein kinase inhibitors and small interference RNAs specific for nuclear factor κB and activator protein 1 significantly suppressed the protein secretion of inflammatory mediators induced by IL-36α stimulation. CONCLUSIONS It was suggested that IL-36α plays an important role in the pathophysiology of inflammation and fibrosis in the pancreas via an autocrine function.
Collapse
|
31
|
Bynigeri RR, Jakkampudi A, Jangala R, Subramanyam C, Sasikala M, Rao GV, Reddy DN, Talukdar R. Pancreatic stellate cell: Pandora's box for pancreatic disease biology. World J Gastroenterol 2017; 23:382-405. [PMID: 28210075 PMCID: PMC5291844 DOI: 10.3748/wjg.v23.i3.382] [Citation(s) in RCA: 129] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 11/09/2016] [Accepted: 12/19/2016] [Indexed: 02/06/2023] Open
Abstract
Pancreatic stellate cells (PSCs) were identified in the early 1980s, but received much attention after 1998 when the methods to isolate and culture them from murine and human sources were developed. PSCs contribute to a small proportion of all pancreatic cells under physiological condition, but are essential for maintaining the normal pancreatic architecture. Quiescent PSCs are characterized by the presence of vitamin A laden lipid droplets. Upon PSC activation, these perinuclear lipid droplets disappear from the cytosol, attain a myofibroblast like phenotype and expresses the activation marker, alpha smooth muscle actin. PSCs maintain their activated phenotype via an autocrine loop involving different cytokines and contribute to progressive fibrosis in chronic pancreatitis (CP) and pancreatic ductal adenocarcinoma (PDAC). Several pathways (e.g., JAK-STAT, Smad, Wnt signaling, Hedgehog etc.), transcription factors and miRNAs have been implicated in the inflammatory and profibrogenic function of PSCs. The role of PSCs goes much beyond fibrosis/desmoplasia in PDAC. It is now shown that PSCs are involved in significant crosstalk between the pancreatic cancer cells and the cancer stroma. These interactions result in tumour progression, metastasis, tumour hypoxia, immune evasion and drug resistance. This is the rationale for therapeutic preclinical and clinical trials that have targeted PSCs and the cancer stroma.
Collapse
|
32
|
Kota J, Hancock J, Kwon J, Korc M. Pancreatic cancer: Stroma and its current and emerging targeted therapies. Cancer Lett 2017; 391:38-49. [PMID: 28093284 DOI: 10.1016/j.canlet.2016.12.035] [Citation(s) in RCA: 126] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 12/22/2016] [Accepted: 12/23/2016] [Indexed: 12/20/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal human malignancies with a 5-year survival rate of 8%. Dense, fibrotic stroma associated with pancreatic tumors is a major obstacle for drug delivery to the tumor bed and plays a crucial role in pancreatic cancer progression. Targeting stroma is considered as a potential therapeutic strategy to improve anti-cancer drug efficacy and patient survival. Although numerous stromal depletion therapies have reached the clinic, they add little to overall survival and are often associated with toxicity. Furthermore, increasing evidence suggests the anti-tumor properties of stroma. Its complete ablation enhanced tumor progression and reduced survival. Consequently, efforts are now focused on developing stromal-targeted therapies that normalize the reactive stroma and avoid the extremes: stromal abundance vs. complete depletion. In this review, we summarized the state of current and emerging anti-stromal targeted therapies, with major emphasis on the role of miRNAs in PDAC stroma and their potential use as novel therapeutic agents to modulate PDAC tumor-stromal interactions.
Collapse
Affiliation(s)
- Janaiah Kota
- Department of Medical and Molecular Genetics, Indiana University School of Medicine (IUSM), Indianapolis, IN, USA; The Melvin and Bren Simon Cancer Center, IUSM, Indianapolis, IN, USA; Center for Pancreatic Cancer Research, Indiana University and Purdue University-Indianapolis (IUPUI), Indianapolis, IN, USA.
| | - Julie Hancock
- Department of Medical and Molecular Genetics, Indiana University School of Medicine (IUSM), Indianapolis, IN, USA
| | - Jason Kwon
- Department of Medical and Molecular Genetics, Indiana University School of Medicine (IUSM), Indianapolis, IN, USA
| | - Murray Korc
- The Melvin and Bren Simon Cancer Center, IUSM, Indianapolis, IN, USA; Center for Pancreatic Cancer Research, Indiana University and Purdue University-Indianapolis (IUPUI), Indianapolis, IN, USA; Department of Biochemistry and Molecular Biology, IUSM, Indianapolis, IN, USA; Department of Medicine, IUSM, Indianapolis, IN, USA
| |
Collapse
|
33
|
Storck H, Hild B, Schimmelpfennig S, Sargin S, Nielsen N, Zaccagnino A, Budde T, Novak I, Kalthoff H, Schwab A. Ion channels in control of pancreatic stellate cell migration. Oncotarget 2017; 8:769-784. [PMID: 27903970 PMCID: PMC5352195 DOI: 10.18632/oncotarget.13647] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 11/07/2016] [Indexed: 12/30/2022] Open
Abstract
Pancreatic stellate cells (PSCs) play a critical role in the progression of pancreatic ductal adenocarcinoma (PDAC). Once activated, PSCs support proliferation and metastasis of carcinoma cells. PSCs even co-metastasise with carcinoma cells. This requires the ability of PSCs to migrate. In recent years, it has been established that almost all "hallmarks of cancer" such as proliferation or migration/invasion also rely on the expression and function of ion channels. So far, there is only very limited information about the function of ion channels in PSCs. Yet, there is growing evidence that ion channels in stromal cells also contribute to tumor progression. Here we investigated the function of KCa3.1 channels in PSCs. KCa3.1 channels are also found in many tumor cells of different origin. We revealed the functional expression of KCa3.1 channels by means of Western blot, immunofluorescence and patch clamp analysis. The impact of KCa3.1 channel activity on PSC function was determined with live-cell imaging and by measuring the intracellular Ca2+ concentration ([Ca2+]i). KCa3.1 channel blockade or knockout prevents the stimulation of PSC migration and chemotaxis by reducing the [Ca2+]i and calpain activity. KCa3.1 channels functionally cooperate with TRPC3 channels that are upregulated in PDAC stroma. Knockdown of TRPC3 channels largely abolishes the impact of KCa3.1 channels on PSC migration. In summary, our results clearly show that ion channels are crucial players in PSC physiology and pathophysiology.
Collapse
Affiliation(s)
| | | | | | - Sarah Sargin
- Institut für Physiologie II, 48149 Münster, Gemany
| | | | - Angela Zaccagnino
- UKSH, Campus Kiel, Institut für Experimentelle Tumorforschung (IET), Sektion Molekulare Onkologie, D-24105 Kiel, Germany
| | - Thomas Budde
- Institut für Physiologie I, 48149 Münster, Gemany
| | - Ivana Novak
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, DK 2100 Copenhagen, Denmark
| | - Holger Kalthoff
- UKSH, Campus Kiel, Institut für Experimentelle Tumorforschung (IET), Sektion Molekulare Onkologie, D-24105 Kiel, Germany
| | | |
Collapse
|
34
|
Slaats J, ten Oever J, van de Veerdonk FL, Netea MG. IL-1β/IL-6/CRP and IL-18/ferritin: Distinct Inflammatory Programs in Infections. PLoS Pathog 2016; 12:e1005973. [PMID: 27977798 PMCID: PMC5158075 DOI: 10.1371/journal.ppat.1005973] [Citation(s) in RCA: 209] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The host inflammatory response against infections is characterized by the release of pro-inflammatory cytokines and acute-phase proteins, driving both innate and adaptive arms of the immune response. Distinct patterns of circulating cytokines and acute-phase responses have proven indispensable for guiding the diagnosis and management of infectious diseases. This review discusses the profiles of acute-phase proteins and circulating cytokines encountered in viral and bacterial infections. We also propose a model in which the inflammatory response to viral (IL-18/ferritin) and bacterial (IL-6/CRP) infections presents with specific plasma patterns of immune biomarkers.
Collapse
Affiliation(s)
- Jeroen Slaats
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
- * E-mail:
| | - Jaap ten Oever
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Frank L. van de Veerdonk
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Mihai G. Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
35
|
Fernandez‐Patron C, Kassiri Z, Leung D. Modulation of Systemic Metabolism by MMP‐2: From MMP‐2 Deficiency in Mice to MMP‐2 Deficiency in Patients. Compr Physiol 2016; 6:1935-1949. [DOI: 10.1002/cphy.c160010] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
36
|
Poormasjedi-Meibod MS, Salimi Elizei S, Leung V, Baradar Jalili R, Ko F, Ghahary A. Kynurenine Modulates MMP-1 and Type-I Collagen Expression Via Aryl Hydrocarbon Receptor Activation in Dermal Fibroblasts. J Cell Physiol 2016; 231:2749-60. [PMID: 26992058 DOI: 10.1002/jcp.25383] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2015] [Accepted: 03/15/2016] [Indexed: 01/13/2023]
Abstract
Dermal fibrosis is characterized by a high deposition of extracellular matrix (ECM) and tissue cellularity. Unfortunately all means of treating this condition are unsatisfactory. We have previously reported the anti-fibrotic effects of Kynurenine (Kyn), a tryptophan metabolite, in fibrotic rabbit ear model. Here, we report the mechanism by which Kyn modulates the expression of key ECM components in dermal fibroblasts. The results showed that Kyn activates aryl hydrocarbon receptor (AHR) nuclear translocation and up-regulates cytochrome-P450 (CYP1A-1) expression, the AHR target gene. A specific AHR antagonist, 6,2',4'-trimethoxyflavone, inhibited the Kyn-dependent modulation of CYP1A-1, MMP-1, and type-I collagen expression. Establishing the anti-fibrogenic effect of Kyn and its mechanism of action, we then developed nano-fibrous Kyn slow-releasing dressings and examined their anti-fibrotic efficacy in vitro and in a rat model. Our results showed the feasibility of incorporating Kyn into PVA/PLGA nanofibers, prolonging the Kyn release up to 4 days tested. Application of medicated-dressings significantly improved the dermal fibrosis indicated by MMP-1 induction, alpha-smooth muscle actin and type-I collagen suppression, and reduced tissue cellularity, T-cells and myofibroblasts. This study clarifies the mechanism by which Kyn modulates ECM expression and reports the development of a new slow-releasing anti-fibrogenic dressing. J. Cell. Physiol. 231: 2749-2760, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
| | - Sanam Salimi Elizei
- Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada
| | - Victor Leung
- Department of Materials Engineering, University of British Columbia, Vancouver, British Columbia, Canada
| | - Reza Baradar Jalili
- Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada
| | - Frank Ko
- Department of Materials Engineering, University of British Columbia, Vancouver, British Columbia, Canada
| | - Aziz Ghahary
- Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
37
|
Robinson SM, Fan L, White SA, Charnley RM, Mann J. The role of exosomes in the pathogenesis of pancreatic ductal adenocarcinoma. Int J Biochem Cell Biol 2016; 75:131-9. [PMID: 27017975 DOI: 10.1016/j.biocel.2016.03.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Revised: 03/21/2016] [Accepted: 03/21/2016] [Indexed: 02/07/2023]
Abstract
Exosomes are small membrane bound vesicles secreted by cancer cells that have a cytosol rich in proteins and nucleic acids which are capable of modulating the phenotype of neighbouring cells which take them up. In this review we explore the mechanisms through which exosomes are able to impact on the pathogenesis of pancreatic ductal cancer through the modulation of tumour formation and development and exploitation of the tumour microenvironment to modulate both the adaptive and innate immune response. In addition we highlight the potential utility of exosomes not only as biomarkers of disease but also as tools to be used in the therapeutic armamentarium against this disease.
Collapse
Affiliation(s)
- Stuart M Robinson
- Fibrosis Research Group, Institute of Cellular Medicine, Newcastle University, Framlington Place, Newcastle upon Tyne NE2 4HH, United Kingdom; Department of HPB Surgery, Freeman Hospital, Freeman Road, High Heaton, Newcastle upon Tyne, NE7 7DN, United Kingdom.
| | - Lavender Fan
- Fibrosis Research Group, Institute of Cellular Medicine, Newcastle University, Framlington Place, Newcastle upon Tyne NE2 4HH, United Kingdom
| | - Steven A White
- Department of HPB Surgery, Freeman Hospital, Freeman Road, High Heaton, Newcastle upon Tyne, NE7 7DN, United Kingdom
| | - Richard M Charnley
- Department of HPB Surgery, Freeman Hospital, Freeman Road, High Heaton, Newcastle upon Tyne, NE7 7DN, United Kingdom
| | - Jelena Mann
- Fibrosis Research Group, Institute of Cellular Medicine, Newcastle University, Framlington Place, Newcastle upon Tyne NE2 4HH, United Kingdom
| |
Collapse
|
38
|
Toste PA, Nguyen AH, Kadera BE, Duong M, Wu N, Gawlas I, Tran LM, Bikhchandani M, Li L, Patel SG, Dawson DW, Donahue TR. Chemotherapy-Induced Inflammatory Gene Signature and Protumorigenic Phenotype in Pancreatic CAFs via Stress-Associated MAPK. Mol Cancer Res 2016; 14:437-47. [PMID: 26979711 DOI: 10.1158/1541-7786.mcr-15-0348] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 03/01/2016] [Indexed: 12/18/2022]
Abstract
UNLABELLED Pancreatic ductal adenocarcinoma (PDAC) has a characteristically dense stroma comprised predominantly of cancer-associated fibroblasts (CAF). CAFs promote tumor growth, metastasis, and treatment resistance. This study aimed to investigate the molecular changes and functional consequences associated with chemotherapy treatment of PDAC CAFs. Chemoresistant immortalized CAFs (R-CAF) were generated by continuous incubation in gemcitabine. Gene expression differences between treatment-naïve CAFs (N-CAF) and R-CAFs were compared by array analysis. Functionally, tumor cells (TC) were exposed to N-CAF- or R-CAF-conditioned media and assayed for migration, invasion, and viability in vitro Furthermore, a coinjection (TC and CAF) model was used to compare tumor growth in vivo R-CAFs increased TC viability, migration, and invasion compared with N-CAFs. In vivo, TCs coinjected with R-CAFs grew larger than those accompanied by N-CAFs. Genomic analysis demonstrated that R-CAFs had increased expression of various inflammatory mediators, similar to the previously described senescence-associated secretory phenotype (SASP). In addition, SASP mediators were found to be upregulated in response to short duration treatment with gemcitabine in both immortalized and primary CAFs. Inhibition of stress-associated MAPK signaling (P38 MAPK or JNK) attenuated SASP induction as well as the tumor-supportive functions of chemotherapy-treated CAFs in vitro and in vivo These results identify a negative consequence of chemotherapy on the PDAC microenvironment that could be targeted to improve the efficacy of current therapeutic regimens. IMPLICATIONS Chemotherapy treatment of pancreatic cancer-associated fibroblasts results in a proinflammatory response driven by stress-associated MAPK signaling that enhances tumor cell growth and invasiveness. Mol Cancer Res; 14(5); 437-47. ©2016 AACR.
Collapse
Affiliation(s)
- Paul A Toste
- Department of Surgery, Division of General Surgery, David Geffen School of Medicine at University of California Los Angeles (UCLA), Los Angeles, California
| | - Andrew H Nguyen
- Department of Surgery, Division of General Surgery, David Geffen School of Medicine at University of California Los Angeles (UCLA), Los Angeles, California
| | - Brian E Kadera
- Department of Surgery, Division of General Surgery, David Geffen School of Medicine at University of California Los Angeles (UCLA), Los Angeles, California
| | - Mindy Duong
- Department of Surgery, Division of General Surgery, David Geffen School of Medicine at University of California Los Angeles (UCLA), Los Angeles, California
| | - Nanping Wu
- Department of Surgery, Division of General Surgery, David Geffen School of Medicine at University of California Los Angeles (UCLA), Los Angeles, California
| | - Irmina Gawlas
- Department of Surgery, Division of General Surgery, David Geffen School of Medicine at University of California Los Angeles (UCLA), Los Angeles, California
| | - Linh M Tran
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at University of California Los Angeles (UCLA), Los Angeles, California
| | - Mihir Bikhchandani
- Department of Surgery, Division of General Surgery, David Geffen School of Medicine at University of California Los Angeles (UCLA), Los Angeles, California
| | - Luyi Li
- Department of Surgery, Division of General Surgery, David Geffen School of Medicine at University of California Los Angeles (UCLA), Los Angeles, California
| | - Sanjeet G Patel
- Department of Surgery, Division of General Surgery, David Geffen School of Medicine at University of California Los Angeles (UCLA), Los Angeles, California
| | - David W Dawson
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at University of California Los Angeles (UCLA), Los Angeles, California. Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at University of California Los Angeles (UCLA), Los Angeles, California
| | - Timothy R Donahue
- Department of Surgery, Division of General Surgery, David Geffen School of Medicine at University of California Los Angeles (UCLA), Los Angeles, California. Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at University of California Los Angeles (UCLA), Los Angeles, California. Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at University of California Los Angeles (UCLA), Los Angeles, California.
| |
Collapse
|
39
|
Wang M, Xiu L, Diao J, Wei L, Sun J. Sparstolonin B inhibits lipopolysaccharide-induced inflammation in 3T3-L1 adipocytes. Eur J Pharmacol 2015; 769:79-85. [PMID: 26522926 DOI: 10.1016/j.ejphar.2015.10.050] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2015] [Revised: 10/12/2015] [Accepted: 10/28/2015] [Indexed: 12/15/2022]
Abstract
Sparstolonin B (SsnB), an isocoumarin compound isolated from the tubers of both Sparganium stoloniferum and Scirpus yagara, has been reported to have anti-inflammatory effects. However, whether SsnB has anti-inflammatory effects on LPS-stimulated 3T3-L1 adipocytes remains unclear. In this study, we investigated the effects of SsnB on adipocyte inflammation in 3T3-L1 adipocytes and anti-obesity properties in high fat diet (HFD)-induced obese rats. 3T3-L1 adipocytes were pretreated with SsnB 1h before LPS treatment. The expression of MCP-1, IL-6, TNF-α, and IL-10 were measured by qRT-PCR and ELISA. The expression of PPAR-γ, TLR4 and NF-κB were detected by western blotting. SsnB was administered to HFD-induced obese rats to confirm its effects in vivo. Our results showed that SsnB dose-dependently inhibited LPS-induced MCP-1, IL-6, and TNF-α production. SsnB was found to inhibit LPS-induced TLR4 expression and NF-κB activition. Furthermore, SsnB was found to activate PPAR-γ and the inhibitory effects of SsnB on MCP-1, IL-6, and TNF-α production can be reversed by PPAR-γ antagonist GW9662. In vivo, SsnB was found to reduce the body weight of rats fed with HFD. SsnB also inhibited the levels of serum triglyceride (TG) and cholesterol (TC) induced by HFD. In conclusion, the results suggested that SsnB could reduce HFD-induced obesity in rats and inhibited LPS-induced cytokines production in 3T3-L1 adipocytes by activating PPAR-γ.
Collapse
Affiliation(s)
- Ming Wang
- Nephrology center of integrated traditional Chinese Medicine and Western Medicine, ZhuJiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Liangchang Xiu
- Department of Epidemiology and Medical Statistics, School of Public Health, Guangdong Medical College, Dongguan, Guangdong 523808, China
| | - Jianxin Diao
- The key laboratory of molecular biology, State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Lianbo Wei
- Nephrology center of integrated traditional Chinese Medicine and Western Medicine, ZhuJiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Jia Sun
- Department of Endocrinology, ZhuJiang Hospital, Southern Medical University, Guangzhou ,510280 China.
| |
Collapse
|
40
|
Sunflower Oil but Not Fish Oil Resembles Positive Effects of Virgin Olive Oil on Aged Pancreas after Life-Long Coenzyme Q Addition. Int J Mol Sci 2015; 16:23425-45. [PMID: 26426013 PMCID: PMC4632707 DOI: 10.3390/ijms161023425] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Revised: 09/21/2015] [Accepted: 09/22/2015] [Indexed: 12/15/2022] Open
Abstract
An adequate pancreatic structure is necessary for optimal organ function. Structural changes are critical in the development of age-related pancreatic disorders. In this context, it has been reported that different pancreatic compartments from rats were affected according to the fat composition consumed. Since there is a close relationship between mitochondria, oxidative stress and aging, an experimental approach has been developed to gain more insight into this process in the pancreas. A low dosage of coenzyme Q was administered life-long in rats in order to try to prevent pancreatic aging-related alterations associated to some dietary fat sources. According to that, three groups of rats were fed normocaloric diets containing Coenzyme Q (CoQ) for two years, where virgin olive, sunflower, or fish oil was included as unique fat source. Pancreatic samples for microscopy and blood samples were collected at the moment of euthanasia. The main finding is that CoQ supplementation gives different results according to fat used in diet. When sunflower oil was the main fat in the diet, CoQ supplementation seems to improve endocrine pancreas structure and in particular β-cell mass resembling positive effects of virgin olive oil. Conversely, CoQ intake does not seem to improve the structural alterations of exocrine compartment previously observed in fish oil fed rats. Therefore CoQ may improve pancreatic alterations associated to the chronic intake of some dietary fat sources.
Collapse
|
41
|
Fernandez-Patron C, Leung D. Emergence of a metalloproteinase / phospholipase A2 axis of systemic inflammation. ACTA ACUST UNITED AC 2015; 2:29-38. [PMID: 26491703 DOI: 10.2147/mnm.s48748] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
We review select aspects of the biology of matrix metalloproteinases (MMPs) with a focus on the modulation of inflammatory responses by MMP-2. MMP-2 is a zinc- and calcium-dependent endoprotease with substrates including extracellular matrix proteins, vasoactive peptides and chemokines. Humans and mice with MMP-2 deficiency exhibit a predominantly inflammatory phenotype. Recent research shows that MMP-2 deficient mice display elevated activity of a secreted phospholipase A2 in the heart. Additionally, MMP-2 deficient mice exhibit abnormally high prostaglandin E2 levels in various organs (i.e., the heart, brain and liver), signs of inflammation and exacerbated lipopolysaccharide-induced fever. We briefly review the biology of sPLA2 enzymes to propose the existence of a heart-centric MMP-2/sPLA2 axis of systemic inflammation. Moreover, we postulate that PLA2 activation is induced by chemokines, whose ability to signal inflammation is regulated in a tissue-specific fashion by MMPs. Thus, genetic and pharmacologically induced MMP-deficiencies can be expected to perturb PLA2-mediated inflammatory mechanisms.
Collapse
Affiliation(s)
- Carlos Fernandez-Patron
- Department of Biochemistry, Cardiovascular Research Group and Mazankowski Alberta Heart Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Dickson Leung
- Department of Biochemistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
42
|
Ferreira MJ, McKenna LB, Zhang J, Reichert M, Bakir B, Buza EL, Furth EE, Bogue CW, Rustgi AK, Kaestner KH. Spontaneous Pancreatitis Caused by Tissue-Specific Gene Ablation of Hhex in Mice. Cell Mol Gastroenterol Hepatol 2015; 1:550-569. [PMID: 26740970 PMCID: PMC4698881 DOI: 10.1016/j.jcmgh.2015.06.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
BACKGROUND & AIMS Perturbations in pancreatic ductal bicarbonate secretion cause chronic pancreatitis. The physiologic mechanism of ductal secretion is known, but its transcriptional control is not. We determine the role of the transcription factor hematopoietically expressed homeobox protein (Hhex) in ductal secretion and pancreatitis. METHODS We derived mice with pancreas-specific, Cremediated Hhex gene ablation to determine the requirement of Hhex in the pancreatic duct in early life and in adult stages. Histologic and immunostaining analyses were used to detect the presence of pathology. Pancreatic primary ductal cells were isolated to discover differentially expressed transcripts upon acute Hhex ablation on a cell autonomous level. RESULTS Hhex protein was detected throughout the embryonic and adult ductal trees. Ablation of Hhex in pancreatic progenitors resulted in postnatal ductal ectasia associated with acinar-to-ductal metaplasia, a progressive phenotype that ultimately resulted in chronic pancreatitis. Hhex ablation in adult mice, however, did not cause any detectable pathology. Ductal ectasia in young mice did not result from perturbation of expression of Hnf6, Hnf1β, or the primary cilia genes. RNA-seq analysis of Hhex-ablated pancreatic primary ductal cells showed mRNA levels of the G-protein coupled receptor natriuretic peptide receptor 3 (Npr3), implicated in paracrine signaling, up-regulated by 4.70-fold. CONCLUSIONS Although Hhex is dispensable for ductal cell function in the adult, ablation of Hhex in pancreatic progenitors results in pancreatitis. Our data highlight the critical role of Hhex in maintaining ductal homeostasis in early life and support ductal hypersecretion as a novel etiology of pediatric chronic pancreatitis.
Collapse
Affiliation(s)
- Mark J. Ferreira
- Department of Genetics and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Lindsay B. McKenna
- Department of Genetics and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jia Zhang
- Department of Genetics and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Maximilian Reichert
- Division of Gastroenterology, Department of Medicine, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Basil Bakir
- Division of Gastroenterology, Department of Medicine, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Elizabeth L. Buza
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Emma E. Furth
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Clifford W. Bogue
- Department of Pediatrics, Yale University School of Medicine, New Haven, Connecticut
| | - Anil K. Rustgi
- Division of Gastroenterology, Department of Medicine, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Klaus H. Kaestner
- Department of Genetics and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania,Correspondence Address correspondence to: Klaus H. Kaestner, PhD, Department of Genetics, Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, 12–126 Translational Research Center, 3400 Civic Center Boulevard, Philadelphia, Pennsylvania 19104. fax: 215-573-5892.
| |
Collapse
|
43
|
Abstract
We found that the secreted protein periostin (Postn) is highly induced after partial pancreatectomy in regenerating areas containing mesenchymal stroma and tubular complexes. Importantly, after partial pancreatectomy, Postn-deficient mice exhibit impaired mesenchymal formation and reduced regeneration specifically within the pancreatic β-cell compartment. Furthermore, Postn-deficient mice demonstrate an increased sensitivity to streptozotocin. Notably, injection of Postn directly into the pancreas stimulated proliferation of vimentin-expressing cells within 24 hours, and by 3 days, a mesenchymal stroma was present containing proliferating duct-like cells expressing the progenitor markers Ngn3 and Pdx1. Intraperitoneal injection of Postn resulted in increased numbers of islets and long-term glucoregulatory benefits with no adverse effects found in other tissues. Delivery of Postn throughout the pancreas via the common bile duct resulted in increased numbers of small insulin-expressing clusters and a significant improvement in glucose tolerance. Therefore, Postn is novel molecule capable of potentiating pancreatic β-cell regeneration.
Collapse
Affiliation(s)
- Johnathan K Smid
- Sprott Center for Stem Cell Research (J.K.S., S.F., M.A.R.), Ottawa Hospital Research Institute, Regenerative Medicine Program, and University of Ottawa (J.K.S., M.A.R.), Cellular and Molecular Medicine, Faculty of Medicine, Ottawa, Ontario, Canada K1H 8L6
| | | | | |
Collapse
|
44
|
Fang F, Pan J, Xu L, Su G, Li G, Wang J. Association between chemokine (C-C motif) ligand 2 gene -2518 A/G polymorphism and pancreatitis risk: a meta-analysis. Pancreatology 2014; 15:53-8. [PMID: 25499426 DOI: 10.1016/j.pan.2014.11.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Revised: 11/13/2014] [Accepted: 11/15/2014] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Many studies have focused on the relationship between chemokine (C-C motif) ligand 2 gene (CCL2) -2518 A/G polymorphism and pancreatitis risk, but the results remain inconsistent. Thus, a meta-analysis was carried out to derive a more precise estimation of the association between CCL2 -2518 A/G polymorphism and pancreatitis risk. METHODS Relevant publications were searched in several widely used databases and six studies were included in the meta-analysis. Pooled odds ratios (ORs) and 95% confidence intervals (CIs) were calculated to evaluate the strength of the association between CCL2 -2518 A/G polymorphism and pancreatitis risk. RESULTS Significant associations between CCL2 -2518 A/G polymorphism and pancreatitis risk were observed in both overall meta-analysis (OR = 0.62, 95% CI = 0.43-0.89 for AA versus AG + GG; OR = 0.71, 95% CI = 0.51-0.98 for A allele versus G allele), and acute pancreatitis subgroup (OR = 0.56, 95% CI = 0.31-0.99 for AA versus AG + GG), especially severe acute pancreatitis subgroup when compared with controls (OR = 0.48, 95% CI = 0.24-0.97 for AG versus GG; OR = 0.35, 95% CI = 0.18-0.70 for AA + AG versus GG). However, no significant pancreatitis risk variation was detected for all genetic models in the severe acute pancreatitis versus mild acute pancreatitis subgroup and the subgroup analysis based on ethnicity. CONCLUSIONS The CCL2 -2518 A/G polymorphism probably associates with pancreatitis risk, especially severe acute pancreatitis risk when compared with controls, with the G allele acting as a risk factor.
Collapse
Affiliation(s)
- Fang Fang
- Institute of Pediatric Research, Children's Hospital of Soochow University, 303 Jingde Road, Suzhou 215003, China
| | - Jian Pan
- Institute of Pediatric Research, Children's Hospital of Soochow University, 303 Jingde Road, Suzhou 215003, China
| | - Lixiao Xu
- Institute of Pediatric Research, Children's Hospital of Soochow University, 303 Jingde Road, Suzhou 215003, China
| | - Guanghao Su
- Institute of Pediatric Research, Children's Hospital of Soochow University, 303 Jingde Road, Suzhou 215003, China
| | - Gang Li
- Institute of Pediatric Research, Children's Hospital of Soochow University, 303 Jingde Road, Suzhou 215003, China
| | - Jian Wang
- Institute of Pediatric Research, Children's Hospital of Soochow University, 303 Jingde Road, Suzhou 215003, China.
| |
Collapse
|
45
|
Sobel K, Tham M, Stark HJ, Stammer H, Prätzel-Wunder S, Bickenbach JR, Boukamp P. Wnt-3a-activated human fibroblasts promote human keratinocyte proliferation and matrix destruction. Int J Cancer 2014; 136:2786-98. [PMID: 25403422 DOI: 10.1002/ijc.29336] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 11/03/2014] [Indexed: 12/20/2022]
Abstract
Aberrant Wnt regulation, detectable by nuclear translocation of beta-catenin, is a hallmark of many cancers including skin squamous cell carcinomas (SCCs). By analyzing primary human skin SCCs, we demonstrate that nuclear beta-catenin is not restricted to SCC cells but also detected in stromal fibroblasts, suggesting an important role for aberrant Wnt regulation also in the tumor microenvironment. When human keratinocytes and fibroblasts were treated with Wnt-3a, fibroblasts proved to be more responsive. Accordingly, Wnt-3a did not alter HaCaT cell functions in a cell-autonomous manner. However, when organotypic cultures (OTCs) were treated with Wnt-3a, HaCaT keratinocytes responded with increased proliferation. As nuclear beta-catenin was induced only in the fibroblasts, this argued for a Wnt-dependent, paracrine keratinocyte stimulation. Global gene expression analysis of Wnt-3a-stimulated fibroblasts identified genes encoding interleukin-8 (IL-8) and C-C motif chemokine 2 (CCL-2) as well as matrix metalloproteinase-1 (MMP-1) as Wnt-3a targets. In agreement, we show that IL-8 and CCL-2 were secreted in high amounts by Wnt-3a-stimulated fibroblasts also in OTCs. The functional role of IL-8 and CCL-2 as keratinocyte growth regulators was confirmed by directly stimulating HaCaT cell proliferation in conventional cultures. Most important, neutralizing antibodies against IL-8 and CCL-2 abolished the Wnt-dependent HaCaT cell hyperproliferation in OTCs. Additionally, MMP-1 was expressed in high amounts in Wnt-3a-stimulated OTCs and degraded the stromal matrix. Thus, our data show that Wnt-3a stimulates fibroblasts to secrete both keratinocyte proliferation-inducing cytokines and stroma-degrading metalloproteinases, thereby providing evidence for a novel Wnt deregulation in the tumor-stroma directly contributing to skin cancer progression.
Collapse
Affiliation(s)
- Katrin Sobel
- Genetics of Skin Carcinogenesis, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | | | | | | | | | | | | |
Collapse
|
46
|
Mathew E, Collins MA, Fernandez-Barrena MG, Holtz AM, Yan W, Hogan JO, Tata Z, Allen BL, Fernandez-Zapico ME, di Magliano MP. The transcription factor GLI1 modulates the inflammatory response during pancreatic tissue remodeling. J Biol Chem 2014; 289:27727-43. [PMID: 25104358 DOI: 10.1074/jbc.m114.556563] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Pancreatic cancer, one of the deadliest human malignancies, is almost uniformly associated with a mutant, constitutively active form of the oncogene Kras. Studies in genetically engineered mouse models have defined a requirement for oncogenic KRAS in both the formation of pancreatic intraepithelial neoplasias, the most common precursor lesions to pancreatic cancer, and in the maintenance and progression of these lesions. Previous work using an inducible model allowing tissue-specific and reversible expression of oncogenic Kras in the pancreas indicates that inactivation of this GTPase at the pancreatic intraepithelial neoplasia stage promotes pancreatic tissue repair. Here, we extend these findings to identify GLI1, a transcriptional effector of the Hedgehog pathway, as a central player in pancreatic tissue repair upon Kras inactivation. Deletion of a single allele of Gli1 results in improper stromal remodeling and perdurance of the inflammatory infiltrate characteristic of pancreatic tumorigenesis. Strikingly, this partial loss of Gli1 affects activated fibroblasts in the pancreas and the recruitment of immune cells that are vital for tissue recovery. Analysis of the mechanism using expression and chromatin immunoprecipitation assays identified a subset of cytokines, including IL-6, mIL-8, Mcp-1, and M-csf (Csf1), as direct GLI1 target genes potentially mediating this phenomenon. Finally, we demonstrate that canonical Hedgehog signaling, a known regulator of Gli1 activity, is required for pancreas recovery. Collectively, these data delineate a new pathway controlling tissue repair and highlight the importance of GLI1 in regulation of the pancreatic microenvironment during this cellular process.
Collapse
Affiliation(s)
- Esha Mathew
- From the Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, Michigan 48109
| | - Meredith A Collins
- From the Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, Michigan 48109
| | | | - Alexander M Holtz
- From the Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, Michigan 48109, the Medical Scientist Training Program, University of Michigan, Ann Arbor, Michigan 48109, Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan 48109
| | - Wei Yan
- the Department of Pathology, Michigan Center for Translational Pathology, and
| | | | | | - Benjamin L Allen
- From the Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, Michigan 48109, Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan 48109
| | | | - Marina Pasca di Magliano
- From the Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, Michigan 48109, the Medical Scientist Training Program, University of Michigan, Ann Arbor, Michigan 48109, Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan 48109 Departments of Surgery, and
| |
Collapse
|
47
|
Pancreatic stellate cells and CX3CR1: occurrence in normal pancreas and acute and chronic pancreatitis and effect of their activation by a CX3CR1 agonist. Pancreas 2014; 43:708-19. [PMID: 24681877 PMCID: PMC4315317 DOI: 10.1097/mpa.0000000000000109] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVES Numerous studies suggest important roles of the chemokine, fractalkine (CX3CL1), in acute/chronic pancreatitis; however, the possible mechanisms of the effects are unclear. Pancreatic stellate cells (PSCs) can play important roles in pancreatitis, secreting inflammatory cytokines/chemokines, as well as proliferation. Therefore, we investigated CX3CL1 receptor (CX3CR1) occurrence in normal pancreas and pancreatitis (acute/chronic) tissues and the effects of CX3CL1 on activated PSCs. METHODS CX3CR1 expression/localization in normal pancreas and pancreatitis (acute/chronic) tissues was evaluated with immunohistochemical analysis. CX3CR1 expression and effects of CX3CL1 on activated PSCs were examined with real-time polymerase chain reaction, BrdU (5-bromo-2-deoxyuridine) assays, and Western blotting. RESULTS In normal pancreas, acinar cells expressed CX3CR1 within granule-like formations in the cytoplasm, whereas in acute/chronic pancreatitis, acinar, ductal, and activated PSCs expressed CX3CR1 on cell membranes. With activation of normal PSCs, CX3CR1 is increased. CX3CL1 activated multiple signaling cascades in PSCs. CX3CL1 did not induce inflammatory genes expression in activated PSCs, but induced proliferation. CONCLUSIONS CX3CR1s are expressed in normal pancreas. Expression is increased in acute/chronic pancreatitis, and the CX3CR1s are activated. CX3CL1 induces proliferation of activated PSCs without increasing release of inflammatory mediators. These results suggest that CX3CR1 activation of PSCs could be important in their effects in pancreatitis, especially to PSC proliferation in pancreatitis where CX3CL1 levels are elevated.
Collapse
|
48
|
Regulation of chemokine CCL5 synthesis in human peritoneal fibroblasts: a key role of IFN-γ. Mediators Inflamm 2014; 2014:590654. [PMID: 24523572 PMCID: PMC3913084 DOI: 10.1155/2014/590654] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Revised: 12/08/2013] [Accepted: 12/12/2013] [Indexed: 01/11/2023] Open
Abstract
Peritonitis is characterized by a coordinated influx of various leukocyte subpopulations. The pattern of leukocyte recruitment is controlled by chemokines secreted primarily by peritoneal mesothelial cells and macrophages. We have previously demonstrated that some chemokines may be also produced by human peritoneal fibroblasts (HPFB). Aim of our study was to assess the potential of HPFB in culture to release CCL5, a potent chemoattractant for mononuclear leukocytes. Quiescent HPFB released constitutively no or trace amounts of CCL5. Stimulation of HPFB with IL-1β and TNF-α resulted in a time- (up to 96 h) and dose-dependent increase in CCL5 expression and release. IFN-γ alone did not induce CCL5 secretion over a wide range of concentrations (0.01–100 U/mL). However, it synergistically amplified the effects of TNF-α and IL-1β through upregulation of CCL5 mRNA. Moreover, pretreatment of cells with IFN-γ upregulated CD40 receptor, which enabled HPFB to respond to a recombinant ligand of CD40 (CD40L). Exposure of IFN-γ-treated HPFB, but not of control cells, to CD40L resulted in a dose-dependent induction of CCL5. These data demonstrate that HPFB synthesise CCL5 in response to inflammatory mediators present in the inflamed peritoneal cavity. HPFB-derived CCL5 may thus contribute to the intraperitoneal recruitment of mononuclear leukocytes during peritonitis.
Collapse
|
49
|
Nakamura T, Ito T, Uchida M, Hijioka M, Igarashi H, Oono T, Kato M, Nakamura K, Suzuki K, Jensen RT, Takayanagi R. PSCs and GLP-1R: occurrence in normal pancreas, acute/chronic pancreatitis and effect of their activation by a GLP-1R agonist. J Transl Med 2014; 94:63-78. [PMID: 24217090 PMCID: PMC3879597 DOI: 10.1038/labinvest.2013.133] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2013] [Revised: 10/02/2013] [Accepted: 10/21/2013] [Indexed: 12/20/2022] Open
Abstract
There is increasing concern about the development of pancreatitis in patients with diabetes mellitus who received long-term glucagon-like peptide-1 (GLP-1) analog treatment. Its pathogenesis is unknown. The effects of GLP-1 agonists on pancreatic endocrine cells are well studied; however, there is little information on effects on other pancreatic tissues that might be involved in inflammatory processes. Pancreatic stellate cells (PSCs) can have an important role in pancreatitis, secreting various inflammatory cytokines/chemokines, as well as collagen. In this study, we investigated GLP-1R occurrence in normal pancreas, acute pancreatitis (AP)/chronic pancreatitis (CP), and the effects of GLP-1 analog on normal PSCs, their ability to stimulate inflammatory mediator secretion or proliferation. GLP-1 receptor (GLP-1R) expression/localization in normal pancreas and pancreatitis (AP/CP) tissues were evaluated with histological/immunohistochemical analysis. PSCs were isolated from male Wistar rats. GLP-1R expression and effects of GLP-1 analog on activated PSCs was examined with real-time PCR, MTS assays and western blotting. In normal pancreas, pancreatic β cells expressed GLP-1R, with only low expression in acinar cells, whereas in AP or CP, acinar cells, ductal cells and activated PSCs expressed GLP-1R. With activation of normal PSCs, GLP-1R is markedly increased, as is multiple other incretin-related receptors. The GLP-1 analog, liraglutide, did not induce inflammatory genes expression in activated PSCs, but induced proliferation. Liraglutide activated multiple signaling cascades in PSCs, and the extracellular signal-regulated kinase pathway mediated the PSCs proliferation. GLP-1Rs are expressed in normal pancreas and there is marked enhanced expression in AP/CP. GLP-1-agonist induced cell proliferation of activated PSCs without increasing release of inflammatory mediators. These results suggest chronic treatment with GLP-1R agonists could lead to proliferation/chronic activation of PSCs, which may lead to important effects in the pancreas.
Collapse
Affiliation(s)
- Taichi Nakamura
- Department of Medicine and Bioregulatory Science, Kyushu University, Fukuoka, Japan
- Department of Cell Biology Section, NIDDK, National Institutes of Health, Bethesda, Maryland, United States
| | - Tetsuhide Ito
- Department of Medicine and Bioregulatory Science, Kyushu University, Fukuoka, Japan
| | - Masahiko Uchida
- Department of Medicine and Bioregulatory Science, Kyushu University, Fukuoka, Japan
| | - Masayuki Hijioka
- Department of Medicine and Bioregulatory Science, Kyushu University, Fukuoka, Japan
| | - Hisato Igarashi
- Department of Medicine and Bioregulatory Science, Kyushu University, Fukuoka, Japan
| | - Takamasa Oono
- Department of Medicine and Bioregulatory Science, Kyushu University, Fukuoka, Japan
| | - Masaki Kato
- Department of Medicine and Bioregulatory Science, Kyushu University, Fukuoka, Japan
| | - Kazuhiko Nakamura
- Department of Medicine and Bioregulatory Science, Kyushu University, Fukuoka, Japan
| | - Koichi Suzuki
- Department of Leprosy Research Center, National Institute of Infectious Diseases, Tokyo Japan
| | - Robert T. Jensen
- Department of Cell Biology Section, NIDDK, National Institutes of Health, Bethesda, Maryland, United States
| | - Ryoichi Takayanagi
- Department of Medicine and Bioregulatory Science, Kyushu University, Fukuoka, Japan
| |
Collapse
|
50
|
Brock C, Nielsen LM, Lelic D, Drewes AM. Pathophysiology of chronic pancreatitis. World J Gastroenterol 2013; 19:7231-7240. [PMID: 24259953 PMCID: PMC3831204 DOI: 10.3748/wjg.v19.i42.7231] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Revised: 07/09/2013] [Accepted: 08/29/2013] [Indexed: 02/06/2023] Open
Abstract
Chronic pancreatitis (CP) is an inflammatory disease of the pancreas characterized by progressive fibrotic destruction of the pancreatic secretory parenchyma. Despite the heterogeneity in pathogenesis and involved risk factors, processes such as necrosis/apoptosis, inflammation or duct obstruction are involved. This fibrosing process ultimately leads to progressive loss of the lobular morphology and structure of the pancreas, deformation of the large ducts and severe changes in the arrangement and composition of the islets. These conditions lead to irreversible morphological and structural changes resulting in impairment of both exocrine and endocrine functions. The prevalence of the disease is largely dependent on culture and geography. The etiological risk-factors associated with CP are multiple and involve both genetic and environmental factors. Throughout this review the M-ANNHEIM classification system will be used, comprising a detailed description of risk factors such as: alcohol-consumption, nicotine-consumption, nutritional factors, hereditary factors, efferent duct factors, immunological factors and miscellaneous and rare metabolic factors. Increased knowledge of the different etiological factors may encourage the use of further advanced diagnostic tools, which potentially will help clinicians to diagnose CP at an earlier stage. However, in view of the multi factorial disease and the complex clinical picture, it is not surprising that treatment of patients with CP is challenging and often unsuccessful.
Collapse
|