1
|
Bertola L, Pepe G, Dolce A, Lecchi C, Borroni EM, Savino B, Canesi S, Sala L, Moretti P, Giordano A, Ressel L, Scanziani E, Vegeto E, Recordati C. Sex-dependent modulation of caerulein-induced acute pancreatitis in C57BL/6J mice. Vet Pathol 2025; 62:382-396. [PMID: 39878085 DOI: 10.1177/03009858241312606] [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] [Indexed: 01/31/2025]
Abstract
Acute pancreatitis (AP) is a life-threatening condition, with a higher mortality rate in men than women and in which estrogens might play a protective role. This study aimed to investigate sex-dependent differences in a mouse model of caerulein-induced AP. Thirty-six C57BL/6J mice (19 females and 17 males) were treated intraperitoneally with phosphate-buffered saline or caerulein, and sacrificed 12 hours, 2 days, or 7 days after the last injection. Blood was collected for amylase, lipase, and glucose determination. Severity and extent of inflammation, apoptosis, and acinar to ductal metaplasia (ADM) in pancreatic tissue were scored histologically and total macrophages, major histocompatibility complex (MHC)-II+ cells, M2 macrophages, T and B cells, neutrophils, apoptosis, and ADM were marked immunohistochemically and quantified by digital image analysis. Serum amylase had a peak at 12 hours, without differences between the sexes. In females, pancreatitis reached a peak at 12 hours with a fast recovery while, in males, the peak was delayed to day 2 with residual apoptosis still present. Macrophages were the main inflammatory cell population, followed by T cells, B cells and neutrophils, without differences between sexes. In males, CD206+ cells and apoptosis were higher at both days 2 and 7, and cytokeratin-19+ (CK19+) ADM was higher at day 7 compared with females. The results of this study revealed a faster onset and resolution of caerulein-induced AP in female mice compared with male mice, supporting a sex-dependent modulation of acute pancreatitis.
Collapse
Affiliation(s)
- Luca Bertola
- University of Milan, Lodi, Italy
- Fondazione UNIMI, Milan, Italy
| | | | | | | | - Elena Monica Borroni
- University of Milan, Milan, Italy
- IRCCS Humanitas Research Hospital, Rozzano, MI, Italy
| | - Benedetta Savino
- University of Milan, Milan, Italy
- IRCCS Humanitas Research Hospital, Rozzano, MI, Italy
| | - Simone Canesi
- University of Milan, Lodi, Italy
- Fondazione UNIMI, Milan, Italy
| | - Laura Sala
- University of Milan, Lodi, Italy
- Fondazione UNIMI, Milan, Italy
| | | | | | | | | | | | | |
Collapse
|
2
|
Bal T. Scaffold-free endocrine tissue engineering: role of islet organization and implications in type 1 diabetes. BMC Endocr Disord 2025; 25:107. [PMID: 40259265 PMCID: PMC12010671 DOI: 10.1186/s12902-025-01919-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 01/17/2025] [Indexed: 04/23/2025] Open
Abstract
Type 1 diabetes (T1D) is a chronic hyperglycemia disorder emerging from beta-cell (insulin secreting cells of the pancreas) targeted autoimmunity. As the blood glucose levels significantly increase and the insulin secretion is gradually lost, the entire body suffers from the complications. Although various advances in the insulin analogs, blood glucose monitoring and insulin application practices have been achieved in the last few decades, a cure for the disease is not obtained. Alternatively, pancreas/islet transplantation is an attractive therapeutic approach based on the patient prognosis, yet this treatment is also limited mainly by donor shortage, life-long use of immunosuppressive drugs and risk of disease transmission. In research and clinics, such drawbacks are addressed by the endocrine tissue engineering of the pancreas. One arm of this engineering is scaffold-free models which often utilize highly developed cell-cell junctions, soluble factors and 3D arrangement of islets with the cellular heterogeneity to prepare the transplant formulations. In this review, taking T1D as a model autoimmune disease, techniques to produce so-called pseudoislets and their applications are studied in detail with the aim of understanding the role of mimicry and pointing out the promising efforts which can be translated from benchside to bedside to achieve exogenous insulin-free patient treatment. Likewise, these developments in the pseudoislet formation are tools for the research to elucidate underlying mechanisms in pancreas (patho)biology, as platforms to screen drugs and to introduce immunoisolation barrier-based hybrid strategies.
Collapse
Affiliation(s)
- Tugba Bal
- Department of Bioengineering, Faculty of Engineering and Natural Sciences, Uskudar University, Istanbul, 34662, Turkey.
| |
Collapse
|
3
|
Mititelu A, Grama A, Colceriu MC, Pop TL. Overview of the cellular and immune mechanisms involved in acute pancreatitis: In search of new prognosis biomarkers. Expert Rev Mol Med 2025; 27:e9. [PMID: 39757373 PMCID: PMC11879381 DOI: 10.1017/erm.2024.40] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 10/15/2024] [Accepted: 11/19/2024] [Indexed: 01/07/2025]
Abstract
Acute pancreatitis (AP) is an acute-onset gastrointestinal disease characterized by a significant inflammation of the pancreas. Most of the time, AP does not leave substantial changes in the pancreas after the resolution of the symptoms but the severe forms are associated with local or systemic complications. The pathogenesis of AP has long been investigated and, lately, the importance of intracellular mechanisms and the immune system has been described. The initial modifications in AP take place in the acinar cell. There are multiple mechanisms by which cellular homeostasis is impaired, one of the most important being calcium overload. Necrotic pancreatic cells initiate the inflammatory response by secreting inflammatory mediators and attracting immune cells. From this point on, the inflammation is sustained by the involvement of innate and adaptive immune systems. Multiple studies have demonstrated the importance of the first 48 h for identifying patients at risk for developing severe forms. For this reason, there is a need to find new, easy-to-use and reliable markers for accurate predictions of these forms. This review provides an overview of the main pathogenetic mechanisms involved in AP development and the most promising biomarkers for severity stratification.
Collapse
Affiliation(s)
- Alexandra Mititelu
- 2 Pediatric Discipline, Department of Mother and Child, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Alina Grama
- 2 Pediatric Discipline, Department of Mother and Child, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
- 2 Pediatric Clinic, Emergency Clinical Hospital for Children, Cluj-Napoca, Romania
| | - Marius-Cosmin Colceriu
- 2 Pediatric Discipline, Department of Mother and Child, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Tudor L. Pop
- 2 Pediatric Discipline, Department of Mother and Child, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
- 2 Pediatric Clinic, Emergency Clinical Hospital for Children, Cluj-Napoca, Romania
| |
Collapse
|
4
|
Giurini EF, Ralph O, Pappas SG, Gupta KH. Looking Beyond Checkpoint Inhibitor Monotherapy: Uncovering New Frontiers for Pancreatic Cancer Immunotherapy. Mol Cancer Ther 2025; 24:18-32. [PMID: 39311547 DOI: 10.1158/1535-7163.mct-24-0311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 08/01/2024] [Accepted: 09/09/2024] [Indexed: 01/03/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) stands out as one of the most aggressive and challenging tumors, characterized by a bleak prognosis with a mere 11% survival rate over 5 years in the United States. Its formidable nature is primarily attributed to its highly aggressive behavior and poor response to existing therapies. PDAC, being notably resistant to immune interventions, presents a significant obstacle in treatment strategies. While immune checkpoint inhibitor therapies have revolutionized outcomes for various cancers, their efficacy in PDAC remains exceedingly low, benefiting less than 1% of patients. The consistent failure of these therapies in PDAC has prompted intensive investigation, particularly at the preclinical level, to unravel the intricate mechanisms of resistance inherent in this cancer type. This pursuit aims to pave the way for the development of novel immunotherapeutic strategies tailored to the distinct characteristics of PDAC. This review endeavors to provide a comprehensive exploration of these emerging immunotherapy approaches in PDAC, with a specific emphasis on elucidating their underlying immunological mechanisms. Additionally, it sheds light on the recently identified factors driving resistance to immunotherapy and evasion of the immune system in PDAC, offering insights beyond the conventional drivers that have been extensively studied.
Collapse
Affiliation(s)
- Eileena F Giurini
- Division of Surgical Oncology, Department of Surgery, Rush University Medical Center, Chicago, Illinois
| | - Oliver Ralph
- Division of Surgical Oncology, Department of Surgery, Rush University Medical Center, Chicago, Illinois
| | - Sam G Pappas
- Division of Surgical Oncology, Department of Surgery, Rush University Medical Center, Chicago, Illinois
| | - Kajal H Gupta
- Division of Surgical Oncology, Department of Surgery, Rush University Medical Center, Chicago, Illinois
- Division of Pediatric Surgery, Department of Surgery, Rush University Medical Center, Chicago, Illinois
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, Illinois
| |
Collapse
|
5
|
Liu R, Wang K, Guo X, Wang Q, Zhang X, Peng K, Lu W, Chen Z, Cao F, Wang Z, Wen L. A causal relationship between distinct immune features and acute or chronic pancreatitis: results from a mendelian randomization analysis. Pancreatology 2024; 24:1219-1228. [PMID: 39419750 DOI: 10.1016/j.pan.2024.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 10/02/2024] [Accepted: 10/09/2024] [Indexed: 10/19/2024]
Abstract
OBJECTIVES This study aimed to thoroughly examining the causal link between immune traits and four types of pancreatitis, using mendelian randomization. METHODS Data on 731 immune traits were collected from the genome-wide association study (GWAS) database as exposure. Information regarding acute pancreatitis (AP), alcohol-induced acute pancreatitis (AAP), chronic pancreatitis (CP), and alcohol-induced chronic pancreatitis (ACP) were acquired from the FinnGen Consortium as outcomes. Mendelian randomization (MR) using inverse variance weighting (IVW) evaluated the links between immune traits and pancreatitis. We evaluated the robustness of the IVW results through sensitivity analyses and validated them using meta-analysis with AP and CP data from the UK Biobank in the GWAS catalog. RESULTS A total of 36 immune traits showed significant associations with susceptibility of four types of pancreatitis, including AP (7 traits), AAP (8 traits), CP (14 traits), and ACP (7 traits). Twenty characteristics were found to be potential risk factors for pancreatitis, identified in B Cells (5 traits), conventional dendritic cells (cDCs, 2 traits), maturation stage of T cells (2 traits), monocytes (2 traits), myeloid cells (2 traits), T cells, B cells, natural killer cells (TBNK, 2 traits), and regulatory T cells (Treg cells, 5 traits). Multiple sensitivity analyses confirmed the validity of the findings. Meta-analysis confirmed a solid causal relationship between CX3CR1 on CD14- CD16-of monocyte panel and the susceptibility of CP. CONCLUSIONS Our MR study identified immune traits causally linked to acute and chronic pancreatitis, offering new insights for early clinical intervention and immune cell-targeted therapies.
Collapse
Affiliation(s)
- Rujuan Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, Shanxi Province, China; Center for Biomarker Discovery and Validation, National Infrastructures for Translational Medicine (PUMCH) & State Key Laboratory of Complex, Severe, and Rare Diseases, Institute of Clinical Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Kui Wang
- Department of Gastroenterology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaoyu Guo
- Center for Biomarker Discovery and Validation, National Infrastructures for Translational Medicine (PUMCH) & State Key Laboratory of Complex, Severe, and Rare Diseases, Institute of Clinical Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Qiqi Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, Shanxi Province, China
| | - Xiuli Zhang
- Center for Biomarker Discovery and Validation, National Infrastructures for Translational Medicine (PUMCH) & State Key Laboratory of Complex, Severe, and Rare Diseases, Institute of Clinical Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Kaixin Peng
- Center for Biomarker Discovery and Validation, National Infrastructures for Translational Medicine (PUMCH) & State Key Laboratory of Complex, Severe, and Rare Diseases, Institute of Clinical Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Wanyi Lu
- Center for Biomarker Discovery and Validation, National Infrastructures for Translational Medicine (PUMCH) & State Key Laboratory of Complex, Severe, and Rare Diseases, Institute of Clinical Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Zhigao Chen
- Center for Biomarker Discovery and Validation, National Infrastructures for Translational Medicine (PUMCH) & State Key Laboratory of Complex, Severe, and Rare Diseases, Institute of Clinical Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Feng Cao
- Department of General Surgery, Xuanwu Hospital Capital Medical University, Beijing, China
| | - Zheng Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, Shanxi Province, China.
| | - Li Wen
- Center for Biomarker Discovery and Validation, National Infrastructures for Translational Medicine (PUMCH) & State Key Laboratory of Complex, Severe, and Rare Diseases, Institute of Clinical Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China.
| |
Collapse
|
6
|
Wei X, Weng Z, Xu X, Yao J. Exploration of a miRNA-mRNA network shared between acute pancreatitis and Epstein-Barr virus infection by integrated bioinformatics analysis. PLoS One 2024; 19:e0311130. [PMID: 39546499 PMCID: PMC11567522 DOI: 10.1371/journal.pone.0311130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 09/10/2024] [Indexed: 11/17/2024] Open
Abstract
Acute pancreatitis (AP) stands out as a primary cause of hospitalization within gastrointestinal ailments, attributed to diverse factors, including Epstein-Barr virus (EBV) infection. Nevertheless, the common miRNAs and genes shared between AP and EBV infection remain unclear. In the present study, four datasets GSE194331, GSE42455, GSE45918 and GSE109220 were selected and downloaded from the Gene Expression Omnibus (GEO) database. Differential expression analysis was performed to screen for differentially expressed genes (DEGs) and differentially expressed miRNAs (DEMs). Target genes of overlapping DEMs were predicted, and intersections with overlapping DEGs were used to construct a miRNA-mRNA network. In addition, the enrichment analysis, drug prediction, diagnostic accuracy assessment, competitive endogenous RNA (ceRNA) network construction, transcription factor (TF)-miRNA-mRNA network construction, and immune cell infiltration analysis were also carried out. We found a total of 111 genes and 8 miRNAs shared between AP and EBV infection. A miRNA-mRNA network was constructed, which comprised 5 miRNAs and 10 genes exhibiting robust diagnostic performance. Histone deacetylase (HDAC) inhibitor was identified as a novel therapeutic intervention from drug prediction analysis. The results of immune cell infiltration analysis revealed that a consistent and significant difference could be found on activated B cell in AP and EBV-infected individuals in comparison to the controls. Taken together, our work, for the first time, revealed a miRNA-mRNA network shared between AP and EBV infection, thereby enriching a deeper comprehension of the intricate molecular mechanisms and potential therapeutic targets entwined in these two pathological conditions.
Collapse
Affiliation(s)
- Xing Wei
- Department of Infectious Disease, The Nantong First People’s Hospital and The Affiliated Hospital 2 of Nantong University, Nantong, China
| | - Zhen Weng
- MOE Engineering Center of Hematological Disease, Soochow University, Suzhou, China
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Xia Xu
- Department of Gastroenterology, The Second People’s Hospital of Nantong and The Affiliated Rehabilitation Hospital of Nantong University, Nantong, China
| | - Jian Yao
- Department of Infectious Disease, The Nantong First People’s Hospital and The Affiliated Hospital 2 of Nantong University, Nantong, China
| |
Collapse
|
7
|
Liu Q, Zhu X, Guo S. From pancreas to lungs: The role of immune cells in severe acute pancreatitis and acute lung injury. Immun Inflamm Dis 2024; 12:e1351. [PMID: 39023414 PMCID: PMC11256889 DOI: 10.1002/iid3.1351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 06/25/2024] [Accepted: 07/08/2024] [Indexed: 07/20/2024] Open
Abstract
BACKGROUND Severe acute pancreatitis (SAP) is a potentially lethal inflammatory pancreatitis condition that is usually linked to multiple organ failure. When it comes to SAP, the lung is the main organ that is frequently involved. Many SAP patients experience respiratory failure following an acute lung injury (ALI). Clinicians provide insufficient care for compounded ALI since the underlying pathophysiology is unknown. The mortality rate of SAP patients is severely impacted by it. OBJECTIVE The study aims to provide insight into immune cells, specifically their roles and modifications during SAP and ALI, through a comprehensive literature review. The emphasis is on immune cells as a therapeutic approach for treating SAP and ALI. FINDINGS Immune cells play an important role in the complicated pathophysiology ofSAP and ALI by maintaining the right balance of pro- and anti-inflammatory responses. Immunomodulatory drugs now in the market have low thepeutic efficacy because they selectively target one immune cell while ignoring immune cell interactions. Accurate management of dysregulated immune responses is necessary. A critical initial step is precisely characterizing the activity of the immune cells during SAP and ALI. CONCLUSION Given the increasing incidence of SAP, immunotherapy is emerging as a potential treatment option for these patients. Interactions among immune cells improve our understanding of the intricacy of concurrent ALI in SAP patients. Acquiring expertise in these domains will stimulate the development of innovative immunomodulation therapies that will improve the outlook for patients with SAP and ALI.
Collapse
Affiliation(s)
- Qi Liu
- Emergency Medicine Clinical Research Center, Beijing Chao‐Yang HospitalCapital Medical UniversityBeijingChina
- Beijing Key Laboratory of Cardiopulmonary Cerebral ResuscitationBeijingChina
| | - Xiaomei Zhu
- Emergency Medicine Clinical Research Center, Beijing Chao‐Yang HospitalCapital Medical UniversityBeijingChina
- Beijing Key Laboratory of Cardiopulmonary Cerebral ResuscitationBeijingChina
| | - Shubin Guo
- Emergency Medicine Clinical Research Center, Beijing Chao‐Yang HospitalCapital Medical UniversityBeijingChina
- Beijing Key Laboratory of Cardiopulmonary Cerebral ResuscitationBeijingChina
| |
Collapse
|
8
|
Mahadevan KK, Dyevoich AM, Chen Y, Li B, Sugimoto H, Sockwell AM, McAndrews KM, Sthanam LK, Wang H, Shalapour S, Watowich SS, Kalluri R. Type I conventional dendritic cells facilitate immunotherapy in pancreatic cancer. Science 2024; 384:eadh4567. [PMID: 38935717 PMCID: PMC11841451 DOI: 10.1126/science.adh4567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Accepted: 04/23/2024] [Indexed: 06/29/2024]
Abstract
Inflammation and tissue damage associated with pancreatitis can precede or occur concurrently with pancreatic ductal adenocarcinoma (PDAC). We demonstrate that in PDAC coupled with pancreatitis (ptPDAC), antigen-presenting type I conventional dendritic cells (cDC1s) are specifically activated. Immune checkpoint blockade therapy (iCBT) leads to cytotoxic CD8+ T cell activation and elimination of ptPDAC with restoration of life span even upon PDAC rechallenge. Using PDAC antigen-loaded cDC1s as a vaccine, immunotherapy-resistant PDAC was rendered sensitive to iCBT with elimination of tumors. cDC1 vaccination coupled with iCBT identified specific CDR3 sequences in the tumor-infiltrating CD8+ T cells with potential therapeutic importance. This study identifies a fundamental difference in the immune microenvironment in PDAC concurrent with, or without, pancreatitis and provides a rationale for combining cDC1 vaccination with iCBT as a potential treatment option.
Collapse
Affiliation(s)
- Krishnan K. Mahadevan
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Allison M. Dyevoich
- Department of Immunology, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Yang Chen
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Bingrui Li
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Hikaru Sugimoto
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Amari M. Sockwell
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Kathleen M. McAndrews
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Lakshmi Kavitha Sthanam
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Huamin Wang
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Shabnam Shalapour
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Stephanie S. Watowich
- Department of Immunology, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Raghu Kalluri
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, TX
- Department of Bioengineering, Rice University, Houston, TX
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX
| |
Collapse
|
9
|
Aney KJ, Jeong WJ, Vallejo AF, Burdziak C, Chen E, Wang A, Koak P, Wise K, Jensen K, Pe'er D, Dougan SK, Martelotto L, Nissim S. Novel Approach for Pancreas Transcriptomics Reveals the Cellular Landscape in Homeostasis and Acute Pancreatitis. Gastroenterology 2024; 166:1100-1113. [PMID: 38325760 PMCID: PMC11102849 DOI: 10.1053/j.gastro.2024.01.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 01/27/2024] [Accepted: 01/30/2024] [Indexed: 02/09/2024]
Abstract
BACKGROUND & AIMS Acinar cells produce digestive enzymes that impede transcriptomic characterization of the exocrine pancreas. Thus, single-cell RNA-sequencing studies of the pancreas underrepresent acinar cells relative to histological expectations, and a robust approach to capture pancreatic cell responses in disease states is needed. We sought to innovate a method that overcomes these challenges to accelerate study of the pancreas in health and disease. METHODS We leverage FixNCut, a single-cell RNA-sequencing approach in which tissue is reversibly fixed with dithiobis(succinimidyl propionate) before dissociation and single-cell preparation. We apply FixNCut to an established mouse model of acute pancreatitis, validate findings using GeoMx whole transcriptome atlas profiling, and integrate our data with prior studies to compare our method in both mouse and human pancreas datasets. RESULTS FixNCut achieves unprecedented definition of challenging pancreatic cells, including acinar and immune populations in homeostasis and acute pancreatitis, and identifies changes in all major cell types during injury and recovery. We define the acinar transcriptome during homeostasis and acinar-to-ductal metaplasia and establish a unique gene set to measure deviation from normal acinar identity. We characterize pancreatic immune cells, and analysis of T-cell subsets reveals a polarization of the homeostatic pancreas toward type-2 immunity. We report immune responses during acute pancreatitis and recovery, including early neutrophil infiltration, expansion of dendritic cell subsets, and a substantial shift in the transcriptome of macrophages due to both resident macrophage activation and monocyte infiltration. CONCLUSIONS FixNCut preserves pancreatic transcriptomes to uncover novel cell states during homeostasis and following pancreatitis, establishing a broadly applicable approach and reference atlas for study of pancreas biology and disease.
Collapse
Affiliation(s)
- Katherine J Aney
- Biological and Biomedical Sciences Program, Harvard Medical School, Boston, Massachusetts; Health Sciences & Technology Program, Harvard-MIT, Boston, Massachusetts; Genetics Division, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts; Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Woo-Jeong Jeong
- Genetics Division, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts; Dana-Farber Cancer Institute, Boston, Massachusetts
| | | | - Cassandra Burdziak
- Computational and Systems Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ethan Chen
- Genetics Division, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts; Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Austin Wang
- Harvard University, Cambridge, Massachusetts
| | - Pal Koak
- Genetics Division, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts; Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Kellie Wise
- Adelaide Centre for Epigenetics (ACE), University of Adelaide, South Australia, Australia; South Australian immunoGENomics Cancer Institute (SAiGENCI), University of Adelaide, South Australia, Australia
| | - Kirk Jensen
- Adelaide Centre for Epigenetics (ACE), University of Adelaide, South Australia, Australia; South Australian immunoGENomics Cancer Institute (SAiGENCI), University of Adelaide, South Australia, Australia; Australian Genome Research Facility, Melbourne, Victoria, Australia
| | - Dana Pe'er
- Computational and Systems Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York; Howard Hughes Medical Institute, Chevy Chase, Maryland
| | - Stephanie K Dougan
- Dana-Farber Cancer Institute, Boston, Massachusetts; Department of Immunology, Harvard Medical School, Boston, Massachusetts
| | - Luciano Martelotto
- Adelaide Centre for Epigenetics (ACE), University of Adelaide, South Australia, Australia; South Australian immunoGENomics Cancer Institute (SAiGENCI), University of Adelaide, South Australia, Australia.
| | - Sahar Nissim
- Biological and Biomedical Sciences Program, Harvard Medical School, Boston, Massachusetts; Health Sciences & Technology Program, Harvard-MIT, Boston, Massachusetts; Genetics Division, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts; Dana-Farber Cancer Institute, Boston, Massachusetts; Gastroenterology Division, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
10
|
Milivojcevic Bevc I, Tasic-Uros D, Stojanovic BS, Jovanovic I, Dimitrijevic Stojanovic M, Gajovic N, Jurisevic M, Radosavljevic G, Pantic J, Stojanovic B. Redefining Immune Dynamics in Acute Pancreatitis: The Protective Role of Galectin-3 Deletion and Treg Cell Enhancement. Biomolecules 2024; 14:642. [PMID: 38927046 PMCID: PMC11201657 DOI: 10.3390/biom14060642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 05/22/2024] [Accepted: 05/28/2024] [Indexed: 06/28/2024] Open
Abstract
Acute pancreatitis (AP) is a complex inflammatory condition that can lead to systemic inflammatory responses and multiple organ dysfunction. This study investigates the role of Galectin-3 (Gal-3), a β-galactoside-binding lectin, in modulating acquired immune responses in AP. Acute pancreatitis was induced by ligation of the bile-pancreatic duct in wild-type and Galectin-3-deficient C57BL/6 mice. We determined the phenotypic and molecular features of inflammatory cells, serum concentrations of amylase, pancreatic trypsin activity, and pancreatic and lung pathology. Galectin-3 deficiency decreased the total number of CD3+CD49- T cells and CD4+ T helper cells, downregulated the production of inflammatory cytokine and IFN-γ, and increased the accumulation of IL-10-producing Foxp3+ T regulatory cells and regulatory CD4+ T cells in the pancreata of diseased animals. The deletion of Galectin-3 ameliorates acute pancreatitis characterized by lowering serum amylase concentration and pancreatic trypsin activity, and attenuating of the histopathology of the lung. These findings shed light on the role of Galectin-3 in acquired immune response in acute pancreatitis and identify Galectin-3 as an attractive target for investigation of the immunopathogenesis of disease and for consideration as a potential therapeutic target for patients with acute inflammatory disease of the pancreas.
Collapse
Affiliation(s)
| | - Danijela Tasic-Uros
- City Medical Emergency Department, 11000 Belgrade, Serbia; (I.M.B.); (D.T.-U.)
| | - Bojana S. Stojanovic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia; (I.J.); (N.G.); (M.J.); (G.R.); (J.P.); (B.S.)
- Department of Pathophysiology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| | - Ivan Jovanovic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia; (I.J.); (N.G.); (M.J.); (G.R.); (J.P.); (B.S.)
| | - Milica Dimitrijevic Stojanovic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia; (I.J.); (N.G.); (M.J.); (G.R.); (J.P.); (B.S.)
- Department of Pathology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| | - Nevena Gajovic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia; (I.J.); (N.G.); (M.J.); (G.R.); (J.P.); (B.S.)
| | - Milena Jurisevic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia; (I.J.); (N.G.); (M.J.); (G.R.); (J.P.); (B.S.)
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| | - Gordana Radosavljevic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia; (I.J.); (N.G.); (M.J.); (G.R.); (J.P.); (B.S.)
| | - Jelena Pantic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia; (I.J.); (N.G.); (M.J.); (G.R.); (J.P.); (B.S.)
| | - Bojan Stojanovic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia; (I.J.); (N.G.); (M.J.); (G.R.); (J.P.); (B.S.)
- Department of Surgery, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| |
Collapse
|
11
|
Waddell H, Stevenson TJ, Mole DJ. The role of the circadian rhythms in critical illness with a focus on acute pancreatitis. Heliyon 2023; 9:e15335. [PMID: 37089281 PMCID: PMC10119767 DOI: 10.1016/j.heliyon.2023.e15335] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 01/20/2023] [Accepted: 04/03/2023] [Indexed: 04/25/2023] Open
Abstract
Circadian rhythms are responsible for governing various physiological processes, including hormone secretion, immune responses, metabolism, and the sleep/wake cycle. In critical illnesses such as acute pancreatitis (AP), circadian rhythms can become dysregulated due to disease. Evidence suggests that time of onset of disease, coupled with peripheral inflammation brought about by AP will impact on the circadian rhythms generated in the central pacemaker and peripheral tissues. Cells of the innate and adaptive immune system are governed by circadian rhythms and the diurnal pattern of expression can be disrupted during disease. Peak circadian immune cell release and gene expression can coincide with AP onset, that may increase pancreatic injury, tissue damage and the potential for systemic inflammation and multiple organ failure to develop. Here, we provide an overview of the role of circadian rhythms in AP and the underpinning inflammatory mechanisms to contextualise ongoing research into the chronobiology and chronotherapeutics of AP.
Collapse
Affiliation(s)
- Heather Waddell
- Medical Research Council Centre for Inflammation Research, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, EH16 4TJ, UK
| | - Tyler J. Stevenson
- Institute of Biodiversity and Animal Health and Comparative Medicine, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, G61 1QH, UK
| | - Damian J. Mole
- Medical Research Council Centre for Inflammation Research, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, EH16 4TJ, UK
- Clinical Surgery, School of Clinical Sciences and Community Health, The University of Edinburgh, Edinburgh, EH16 4SB, UK
| |
Collapse
|
12
|
Mahadevan KK, Dyevoich AM, Chen Y, Li B, Sugimoto H, Sockwell AM, McAndrews KM, Wang H, Shalapour S, Watowich SS, Kalluri R. Antigen-presenting type-I conventional dendritic cells facilitate curative checkpoint blockade immunotherapy in pancreatic cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.05.531191. [PMID: 36945457 PMCID: PMC10028824 DOI: 10.1101/2023.03.05.531191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/23/2023]
Abstract
Inflammation and tissue damage associated with pancreatitis can precede or occur concurrently with pancreatic ductal adenocarcinoma (PDAC). We demonstrate that in PDAC coupled with pancreatitis (ptPDAC), antigen-presenting type-I conventional dendritic cells (cDC1s) are specifically activated. Immune checkpoint blockade therapy (iCBT) leads to cytotoxic CD8 + T cell activation and eradication of ptPDAC with restoration of lifespan even upon PDAC re-challenge. Such eradication of ptPDAC was reversed following specific depletion of dendritic cells. Employing PDAC antigen-loaded cDC1s as a vaccine, immunotherapy-resistant PDAC was rendered sensitive to iCBT with a curative outcome. Analysis of the T-cell receptor (TCR) sequences in the tumor infiltrating CD8 + T cells following cDC1 vaccination coupled with iCBT identified unique CDR3 sequences with potential therapeutic significance. Our findings identify a fundamental difference in the immune microenvironment and adaptive immune response in PDAC concurrent with, or without pancreatitis, and provides a rationale for combining cDC1 vaccination with iCBT as a potential treatment option.
Collapse
|
13
|
Liu S, Szatmary P, Lin JW, Wang Q, Sutton R, Chen L, Liu T, Huang W, Xia Q. Circulating monocytes in acute pancreatitis. Front Immunol 2022; 13:1062849. [PMID: 36578487 PMCID: PMC9791207 DOI: 10.3389/fimmu.2022.1062849] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 11/28/2022] [Indexed: 12/14/2022] Open
Abstract
Acute pancreatitis is a common gastrointestinal disease characterized by inflammation of the exocrine pancreas and manifesting itself through acute onset of abdominal pain. It is frequently associated with organ failure, pancreatic necrosis, and death. Mounting evidence describes monocytes - phagocytic, antigen presenting, and regulatory cells of the innate immune system - as key contributors and regulators of the inflammatory response and subsequent organ failure in acute pancreatitis. This review highlights the recent advances of dynamic change of numbers, phenotypes, and functions of circulating monocytes as well as their underling regulatory mechanisms with a special focus on the role of lipid modulation during acute pancreatitis.
Collapse
Affiliation(s)
- Shiyu Liu
- West China Centre of Excellence for Pancreatitis, Institute of Integrated Traditional Chinese and Western Medicine, West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu, China
| | - Peter Szatmary
- Liverpool Pancreatitis Research Group, Liverpool University Hospitals NHS Foundation Trust and Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Jing-wen Lin
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Qiqi Wang
- West China Centre of Excellence for Pancreatitis, Institute of Integrated Traditional Chinese and Western Medicine, West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu, China
| | - Robert Sutton
- Liverpool Pancreatitis Research Group, Liverpool University Hospitals NHS Foundation Trust and Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Lu Chen
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Tingting Liu
- West China Centre of Excellence for Pancreatitis, Institute of Integrated Traditional Chinese and Western Medicine, West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu, China,*Correspondence: Tingting Liu, ; Wei Huang, ; Qing Xia,
| | - Wei Huang
- West China Centre of Excellence for Pancreatitis, Institute of Integrated Traditional Chinese and Western Medicine, West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu, China,Institutes for Systems Genetics & Immunology and Inflammation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China,*Correspondence: Tingting Liu, ; Wei Huang, ; Qing Xia,
| | - Qing Xia
- West China Centre of Excellence for Pancreatitis, Institute of Integrated Traditional Chinese and Western Medicine, West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu, China,*Correspondence: Tingting Liu, ; Wei Huang, ; Qing Xia,
| |
Collapse
|
14
|
Takauji S, Konishi H, Fujiya M, Ueno N, Tanaka H, Sato H, Isozaki S, Kashima S, Moriichi K, Mizukami Y, Okumura T. Polyphosphate, Derived from Lactobacillus brevis, Modulates the Intestinal Microbiome and Attenuates Acute Pancreatitis. Dig Dis Sci 2021; 66:3872-3884. [PMID: 33492535 DOI: 10.1007/s10620-020-06747-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 11/23/2020] [Indexed: 02/05/2023]
Abstract
BACKGROUND We previously showed that Lactobacillus brevis-derived polyphosphate (poly P) exerts a curative effect on intestinal inflammation. However, whether or not poly P improves the inflammation and injury of distant organs remains unclear. AIMS We aimed to investigate the change in the intestinal microbiome and to evaluate the protective effect of poly P on injuries in a cerulein-induced acute pancreatitis (AP) mouse. METHODS Poly P was orally administered to BALB/C mice every day for 24 days, and then mice were intraperitoneally injected with cerulein. Before cerulein injection, stool samples were collected and analyzed by 16S rRNA gene sequencing. Mice were sacrificed at 24 h after the last cerulein injection; subsequently, the serum, pancreas, and colon were collected. RESULTS The microbial profile differed markedly between poly P and control group. Notably, the levels of beneficial bacteria, including Alistipes and Candidatus_Saccharimonas, were significantly increased, while those of the virulent bacteria Desulfovibrio were decreased in the poly P group. The elevations of the serum amylase and lipase levels by cerulein treatment were suppressed by the pre-administration of poly P for 24 days, but not for 7 days. The numbers of cells MPO-positive by immunohistology were decreased and the levels of MCP-1 significantly reduced in the AP + Poly P group. An immunofluorescence analysis showed that the ZO-1 and occludin in the colon was strongly augmented in the epithelial cell membrane layer in the AP + Poly P group. CONCLUSIONS Poly P attenuates AP through both modification of the intestinal microbiome and enhancement of the intestinal barrier integrity.
Collapse
Affiliation(s)
- Shuhei Takauji
- Department of Emergency Medicine, Asahikawa Medical University, 2-1, Midorigaoka Higashi, Asahikawa, 078-8510, Japan
- Gastroenterology and Endoscopy, Division of Metabolism and Biosystemic Science, Gastroenterology, and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, 2-1, Midorigaoka Higashi, Asahikawa, 078-8510, Japan
| | - Hiroaki Konishi
- Department of Gastroenterology and Advanced Medical Sciences, Asahikawa Medical University, 2-1, Midorigaoka Higashi, Asahikawa, 078-8510, Japan
| | - Mikihiro Fujiya
- Gastroenterology and Endoscopy, Division of Metabolism and Biosystemic Science, Gastroenterology, and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, 2-1, Midorigaoka Higashi, Asahikawa, 078-8510, Japan.
- Department of Gastroenterology and Advanced Medical Sciences, Asahikawa Medical University, 2-1, Midorigaoka Higashi, Asahikawa, 078-8510, Japan.
| | - Nobuhiro Ueno
- Gastroenterology and Endoscopy, Division of Metabolism and Biosystemic Science, Gastroenterology, and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, 2-1, Midorigaoka Higashi, Asahikawa, 078-8510, Japan
| | - Hiroki Tanaka
- Division of Tumor Pathology, Department of Pathology, Asahikawa Medical University, 2-1, Midorigaoka Higashi, Asahikawa, 078-8510, Japan
| | - Hiroki Sato
- Gastroenterology and Endoscopy, Division of Metabolism and Biosystemic Science, Gastroenterology, and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, 2-1, Midorigaoka Higashi, Asahikawa, 078-8510, Japan
| | - Shotaro Isozaki
- Gastroenterology and Endoscopy, Division of Metabolism and Biosystemic Science, Gastroenterology, and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, 2-1, Midorigaoka Higashi, Asahikawa, 078-8510, Japan
| | - Shin Kashima
- Gastroenterology and Endoscopy, Division of Metabolism and Biosystemic Science, Gastroenterology, and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, 2-1, Midorigaoka Higashi, Asahikawa, 078-8510, Japan
| | - Kentaro Moriichi
- Gastroenterology and Endoscopy, Division of Metabolism and Biosystemic Science, Gastroenterology, and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, 2-1, Midorigaoka Higashi, Asahikawa, 078-8510, Japan
| | - Yusuke Mizukami
- Cancer Genetics, Division of Metabolism and Biosystemic Science, Gastroenterology, and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, 2-1, Midorigaoka Higashi, Asahikawa, 078-8510, Japan
| | - Toshikatsu Okumura
- Division of Metabolism and Biosystemic Science, Gastroenterology, and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, 2-1, Midorigaoka Higashi, Asahikawa, 078-8510, Japan
| |
Collapse
|
15
|
Li G, Chen H, Liu L, Xiao P, Xie Y, Geng X, Zhang T, Zhang Y, Lu T, Tan H, Li L, Sun B. Role of Interleukin-17 in Acute Pancreatitis. Front Immunol 2021; 12:674803. [PMID: 34594321 PMCID: PMC8476864 DOI: 10.3389/fimmu.2021.674803] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 08/31/2021] [Indexed: 11/24/2022] Open
Abstract
Acute pancreatitis (AP) is a leading cause of death and is commonly accompanied by systemic manifestations that are generally associated with a poor prognosis. Many cytokines contribute to pancreatic tissue damage and cause systemic injury. Interleukin-17 (IL-17) is a cytokine that may play a vital role in AP. Specifically, IL-17 has important effects on the immune response and causes interactions between different inflammatory mediators in the AP-related microenvironment. In this literature review, we will discuss the existing academic understanding of IL-17 and the impacts of IL-17 in different cells (especially in acinar cells and immune system cells) in AP pathogenesis. The clinical significance and potential mechanisms of IL-17 on AP deterioration are emphasized. The evidence suggests that inhibiting the IL-17 cytokine family could alleviate the pathogenic process of AP, and we highlight therapeutic strategies that directly or indirectly target IL-17 cytokines in acute pancreatitis.
Collapse
Affiliation(s)
- Guanqun Li
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hongze Chen
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Liwei Liu
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Peng Xiao
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yu Xie
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xinglong Geng
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Tao Zhang
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yang Zhang
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Tianqi Lu
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hongtao Tan
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Le Li
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Bei Sun
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, China
| |
Collapse
|
16
|
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: 33] [Impact Index Per Article: 8.3] [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
|
17
|
Hidalgo-Sastre A, Kuebelsbeck LA, Jochheim LS, Staufer LM, Altmayr F, Johannes W, Steiger K, Ronderos M, Hartmann D, Hüser N, Schmid RM, Holzmann B, von Figura G. Toll-like receptor 3 expression in myeloid cells is essential for efficient regeneration after acute pancreatitis in mice. Eur J Immunol 2021; 51:1182-1194. [PMID: 33521935 DOI: 10.1002/eji.202048771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 11/12/2020] [Accepted: 01/29/2021] [Indexed: 11/09/2022]
Abstract
Stringent regulation of the inflammatory response is crucial for normal tissue regeneration. Here, we analyzed the role of Toll-like receptor 3 (TLR3) in pancreatic regeneration after acute pancreatitis (AP). AP was induced by caerulein treatment in mice with global TLR3 deficiency (TLR3OFF ) or in mice re-expressing TLR3 exclusively in the myeloid cell lineage (TLR3Mye ). Compared to WT mice, TLR3OFF mice had a markedly increased formation of acinar-to-ductal metaplasia (ADM) that persisted until day 7 after initiation of AP. Pancreatic tissue of WT mice was completely regenerated after 5 days with no detectable ADM structures. The enhancing effect of TLR3-deficiency on ADM formation was closely linked with an increased and prolonged accumulation of macrophages in pancreata of TLR3OFF mice. Importantly, the phenotype of TLR3OFF mice was rescued in TLR3Mye mice, demonstrating the causative role of myeloid cell selective TLR3 signaling. Moreover, in vitro stimulation of macrophages through TLR3 initiated cell death by a caspase-8-associated mechanism. Therefore, these findings provide evidence that TLR3 signaling in myeloid cells is sufficient to limit inflammation and ADM formation and to promote regeneration after AP. Notably, resolution of inflammation after AP was associated with macrophage sensitivity to TLR3-mediated cell death.
Collapse
Affiliation(s)
- Ana Hidalgo-Sastre
- School of Medicine, Medizinische Klinik und Poliklinik II, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Ludwig A Kuebelsbeck
- School of Medicine, Medizinische Klinik und Poliklinik II, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Leonie S Jochheim
- School of Medicine, Medizinische Klinik und Poliklinik II, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Lina M Staufer
- School of Medicine, Medizinische Klinik und Poliklinik II, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Felicitas Altmayr
- Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Department of Surgery, Munich, Germany
| | - Widya Johannes
- Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Department of Surgery, Munich, Germany
| | - Katja Steiger
- Technical University of Munich, School of Medicine, Department of Pathology, Munich, Germany
| | - Monica Ronderos
- Technical University of Munich, School of Medicine, Department of Pathology, Munich, Germany
| | - Daniel Hartmann
- Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Department of Surgery, Munich, Germany
| | - Norbert Hüser
- Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Department of Surgery, Munich, Germany
| | - Roland M Schmid
- School of Medicine, Medizinische Klinik und Poliklinik II, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Bernhard Holzmann
- Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Department of Surgery, Munich, Germany
| | - Guido von Figura
- School of Medicine, Medizinische Klinik und Poliklinik II, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| |
Collapse
|
18
|
Yang B, Davis JM, Gomez TH, Younes M, Zhao X, Shen Q, Wang R, Ko TC, Cao Y. Characteristic pancreatic and splenic immune cell infiltration patterns in mouse acute pancreatitis. Cell Biosci 2021; 11:28. [PMID: 33531047 PMCID: PMC7852096 DOI: 10.1186/s13578-021-00544-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 01/21/2021] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND A systemic evaluation of immune cell infiltration patterns in experimental acute pancreatitis (AP) is lacking. Using multi-dimensional flow cytometry, this study profiled infiltrating immune cell types in multiple AP mouse models. METHODS Three AP models were generated in C57BL/6 mice via cerulein (CAE) injection, alcohol and palmitoleic acid (EtOH + POA) injection, and alcohol diet feeding and cerulein (EtOH + CAE) injection. Primary pancreatic cells and splenocytes were prepared, and multi-dimensional flow cytometry was performed and analyzed by manual gating and computerized PhenoGraph, followed by visualization with t-distributed stochastic neighbor embedding (t-SNE). RESULTS CAE treatment induced a time-dependent increase of major innate immune cells and a decrease of follicular B cells, and TCD4+ cells and the subtypes in the pancreas, whereas elicited a reversed pattern in the spleen. EtOH + POA treatment resulted in weaker effects than CAE treatment. EtOH feeding enhanced CAE-induced amylase secretion, but unexpectedly attenuated CAE-induced immune cell regulation. In comparison with manual gating analysis, computerized analysis demonstrated a remarkable time efficiency and reproducibility on the innate immune cells and B cells. CONCLUSIONS The reverse pattern of increased innate and decreased adaptive immune cells was consistent in the pancreas in CAE and EtOH + POA treatments. Alcohol feeding opposed the CAE effect on immune cell regulation. Together, the immune profiling approach utilized in this study provides a better understanding of overall immune responses in AP, which may facilitate the identification of intervention windows and new therapeutic strategies. Computerized analysis is superior to manual gating by dramatically reducing analysis time.
Collapse
Affiliation(s)
- Baibing Yang
- Department of Surgery, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Joy M Davis
- Department of Surgery, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Thomas H Gomez
- Center of Laboratory Animal Medicine and Care, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Mamoun Younes
- Department of Pathology & Laboratory Medicine, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
- Department of Pathology, George Washington University School of Medicine and Health Sciences, George Washington University Hospital, Washington, DC, 20037, USA
| | - Xiurong Zhao
- Department of Neurology, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Qiang Shen
- Department of Genetics, Louisiana State University Health Sciences Center, New Orleans, LA, 70112, USA
| | - Run Wang
- Department of Surgery, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Tien C Ko
- Department of Surgery, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA.
| | - Yanna Cao
- Department of Surgery, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA.
| |
Collapse
|
19
|
Peng C, Li Z, Yu X. The Role of Pancreatic Infiltrating Innate Immune Cells in Acute Pancreatitis. Int J Med Sci 2021; 18:534-545. [PMID: 33390823 PMCID: PMC7757151 DOI: 10.7150/ijms.51618] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 11/13/2020] [Indexed: 12/13/2022] Open
Abstract
Acute pancreatitis (AP) is a leading cause of gastrointestinal-related hospital admissions with significant morbidity and mortality. Although the underlying pathophysiology of AP is rather complex, which greatly limits the treatment options, more and more studies have revealed that infiltrating immune cells play a critical role in the pathogenesis of AP and determine disease severity. Thus, immunomodulatory therapy targeting immune cells and related inflammatory mediators is expected to be a novel treatment modality for AP which may improve the prognosis of patients. Cells of the innate immune system, including macrophages, neutrophils, dendritic cells, and mast cells, represent the majority of infiltrating cells during AP. In this review, an overview of different populations of innate immune cells and their role during AP will be discussed, with a special focus on neutrophils and macrophages.
Collapse
Affiliation(s)
- Cheng Peng
- Department of Hepatopancreatobiliary Surgery, Third Xiangya Hospital, Central South University, Changsha 410013, Hunan, China
| | - Zhiqiang Li
- Department of Hepatopancreatobiliary Surgery, Third Xiangya Hospital, Central South University, Changsha 410013, Hunan, China
| | - Xiao Yu
- Department of Hepatopancreatobiliary Surgery, Third Xiangya Hospital, Central South University, Changsha 410013, Hunan, China
| |
Collapse
|
20
|
Alcohol and Smoking Mediated Modulations in Adaptive Immunity in Pancreatitis. Cells 2020; 9:cells9081880. [PMID: 32796685 PMCID: PMC7463831 DOI: 10.3390/cells9081880] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 08/05/2020] [Accepted: 08/06/2020] [Indexed: 12/12/2022] Open
Abstract
Pancreatitis is a condition of pancreatic inflammation driven by injury to the pancreatic parenchyma. The extent of acinar insult, intensity, and type of immune response determines the severity of the disease. Smoking, alcohol and autoimmune pancreatitis are some of the predominant risk factors that increase the risk of pancreatitis by differentially influencing the adaptive immune system. The overall decrease in peripheral lymphocyte (T-, B- and (natural killer T-) NKT-cell) count and increased infiltration into the damaged pancreatic tissue highlight the contribution of adaptive immunity in the disease pathology. Smoking and alcohol modulate the responsiveness and apoptosis of T- and B-cells during pancreatic insult. Acute pancreatitis worsens with smoking and alcohol, leading to the development of systemic inflammatory response syndrome and compensatory anti-inflammatory response syndrome, suggesting the critical role of adaptive immunity in fatal outcomes such as multiple organ dysfunction. The presence of CD4+ and CD8+ T-lymphocytes and perforin-expressing cells in the fibrotic tissue in chronic pancreatitis modulate the severity of the disease. Due to their important role in altering the severity of the disease, attempts to target adaptive immune mediators will be critical for the development of novel therapeutic interventions.
Collapse
|
21
|
Zhao Y, Wu Z, Chanal M, Guillaumond F, Goehrig D, Bachy S, Principe M, Ziverec A, Flaman JM, Collin G, Tomasini R, Pasternack A, Ritvos O, Vasseur S, Bernard D, Hennino A, Bertolino P. Oncogene-Induced Senescence Limits the Progression of Pancreatic Neoplasia through Production of Activin A. Cancer Res 2020; 80:3359-3371. [PMID: 32554750 DOI: 10.1158/0008-5472.can-19-3763] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 04/08/2020] [Accepted: 06/12/2020] [Indexed: 11/16/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a deadly and aggressive cancer. Understanding mechanisms that drive preneoplastic pancreatic lesions is necessary to improve early diagnostic and therapeutic strategies. Mutations and inactivation of activin-like kinase (ALK4) have been demonstrated to favor PDAC onset. Surprisingly, little is known regarding the ligands that drive ALK4 signaling in pancreatic cancer or how this signaling pathway limits the initiation of neoplastic lesions. In this study, data mining and histologic analyses performed on human and mouse tumor tissues revealed that activin A is the major ALK4 ligand that drives PDAC initiation. Activin A, which is absent in normal acinar cells, was strongly induced during acinar-to-ductal metaplasia (ADM), which was promoted by pancreatitis or the activation of KrasG12D in mice. Activin A expression during ADM was associated with the cellular senescence program that is induced in precursor lesions. Blocking activin A signaling through the use of a soluble form of activin receptor IIB (sActRIIB-Fc) and ALK4 knockout in mice expressing KrasG12D resulted in reduced senescence associated with decreased expression of p21, reduced phosphorylation of H2A histone family member X (H2AX), and increased proliferation. Thus, this study indicates that activin A acts as a protective senescence-associated secretory phenotype factor produced by Kras-induced senescent cells during ADM, which limits the expansion and proliferation of pancreatic neoplastic lesions. SIGNIFICANCE: This study identifies activin A to be a beneficial, senescence-secreted factor induced in pancreatic preneoplastic lesions, which limits their proliferation and ultimately slows progression into pancreatic cancers.
Collapse
Affiliation(s)
- Yajie Zhao
- Cancer Research Centre of Lyon, INSERM U1052, CNRS UMR5286, Claude Bernard University, Lyon, France.,Department of Geriatrics, Ruijin Hospital, School of Medicine, Shanghai Jia Tong University, Shanghai, China
| | - Zhichong Wu
- Cancer Research Centre of Lyon, INSERM U1052, CNRS UMR5286, Claude Bernard University, Lyon, France
| | - Marie Chanal
- Cancer Research Centre of Lyon, INSERM U1052, CNRS UMR5286, Claude Bernard University, Lyon, France
| | - Fabienne Guillaumond
- Centre de Recherche en Cancérologie de Marseille, Unité 1068, Institut National de la Santé et de la Recherche Médicale, Marseille, France.,Institut Paoli-Calmettes, Marseille, France.,Unité Mixte de Recherche (UMR 7258), Centre national de la Recherche Scientifique, Marseille, France.,Université Aix-Marseille, Marseille, France
| | - Delphine Goehrig
- Cancer Research Centre of Lyon, INSERM U1052, CNRS UMR5286, Claude Bernard University, Lyon, France
| | - Sophie Bachy
- Cancer Research Centre of Lyon, INSERM U1052, CNRS UMR5286, Claude Bernard University, Lyon, France
| | - Moitza Principe
- Cancer Research Centre of Lyon, INSERM U1052, CNRS UMR5286, Claude Bernard University, Lyon, France
| | - Audrey Ziverec
- Cancer Research Centre of Lyon, INSERM U1052, CNRS UMR5286, Claude Bernard University, Lyon, France
| | - Jean-Michel Flaman
- Cancer Research Centre of Lyon, INSERM U1052, CNRS UMR5286, Claude Bernard University, Lyon, France
| | - Guillaume Collin
- Cancer Research Centre of Lyon, INSERM U1052, CNRS UMR5286, Claude Bernard University, Lyon, France
| | - Richard Tomasini
- Centre de Recherche en Cancérologie de Marseille, Unité 1068, Institut National de la Santé et de la Recherche Médicale, Marseille, France.,Institut Paoli-Calmettes, Marseille, France.,Unité Mixte de Recherche (UMR 7258), Centre national de la Recherche Scientifique, Marseille, France.,Université Aix-Marseille, Marseille, France
| | - Arja Pasternack
- Department of Physiology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Olli Ritvos
- Department of Physiology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Sophie Vasseur
- Centre de Recherche en Cancérologie de Marseille, Unité 1068, Institut National de la Santé et de la Recherche Médicale, Marseille, France.,Institut Paoli-Calmettes, Marseille, France.,Unité Mixte de Recherche (UMR 7258), Centre national de la Recherche Scientifique, Marseille, France.,Université Aix-Marseille, Marseille, France
| | - David Bernard
- Cancer Research Centre of Lyon, INSERM U1052, CNRS UMR5286, Claude Bernard University, Lyon, France
| | - Ana Hennino
- Cancer Research Centre of Lyon, INSERM U1052, CNRS UMR5286, Claude Bernard University, Lyon, France
| | - Philippe Bertolino
- Cancer Research Centre of Lyon, INSERM U1052, CNRS UMR5286, Claude Bernard University, Lyon, France.
| |
Collapse
|
22
|
Hegde S, Krisnawan VE, Herzog BH, Zuo C, Breden MA, Knolhoff BL, Hogg GD, Tang JP, Baer JM, Mpoy C, Lee KB, Alexander KA, Rogers BE, Murphy KM, Hawkins WG, Fields RC, DeSelm CJ, Schwarz JK, DeNardo DG. Dendritic Cell Paucity Leads to Dysfunctional Immune Surveillance in Pancreatic Cancer. Cancer Cell 2020; 37:289-307.e9. [PMID: 32183949 PMCID: PMC7181337 DOI: 10.1016/j.ccell.2020.02.008] [Citation(s) in RCA: 297] [Impact Index Per Article: 59.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 12/04/2019] [Accepted: 02/14/2020] [Indexed: 12/26/2022]
Abstract
Here, we utilized spontaneous models of pancreatic and lung cancer to examine how neoantigenicity shapes tumor immunity and progression. As expected, neoantigen expression during lung adenocarcinoma development leads to T cell-mediated immunity and disease restraint. By contrast, neoantigen expression in pancreatic ductal adenocarcinoma (PDAC) results in exacerbation of a fibro-inflammatory microenvironment that drives disease progression and metastasis. Pathogenic TH17 responses are responsible for this neoantigen-induced tumor progression in PDAC. Underlying these divergent T cell responses in pancreas and lung cancer are differences in infiltrating conventional dendritic cells (cDCs). Overcoming cDC deficiency in early-stage PDAC leads to disease restraint, while restoration of cDC function in advanced PDAC restores tumor-restraining immunity and enhances responsiveness to radiation therapy.
Collapse
Affiliation(s)
- Samarth Hegde
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Varintra E Krisnawan
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Brett H Herzog
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Chong Zuo
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Marcus A Breden
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Brett L Knolhoff
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Graham D Hogg
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jack P Tang
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - John M Baer
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Cedric Mpoy
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Kyung Bae Lee
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Katherine A Alexander
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Buck E Rogers
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO 63110, USA; Alvin J. Siteman Comprehensive Cancer Center, St. Louis, MO 63110, USA
| | - Kenneth M Murphy
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA; Howard Hughes Medical Institute, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - William G Hawkins
- Department of Surgery, Barnes-Jewish Hospital, St. Louis, MO 63110, USA; Alvin J. Siteman Comprehensive Cancer Center, St. Louis, MO 63110, USA
| | - Ryan C Fields
- Department of Surgery, Barnes-Jewish Hospital, St. Louis, MO 63110, USA; Alvin J. Siteman Comprehensive Cancer Center, St. Louis, MO 63110, USA
| | - Carl J DeSelm
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO 63110, USA; Alvin J. Siteman Comprehensive Cancer Center, St. Louis, MO 63110, USA
| | - Julie K Schwarz
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO 63110, USA; Alvin J. Siteman Comprehensive Cancer Center, St. Louis, MO 63110, USA
| | - David G DeNardo
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA; Alvin J. Siteman Comprehensive Cancer Center, St. Louis, MO 63110, USA.
| |
Collapse
|
23
|
PD-L1 engagement on T cells promotes self-tolerance and suppression of neighboring macrophages and effector T cells in cancer. Nat Immunol 2020; 21:442-454. [PMID: 32152508 DOI: 10.1038/s41590-020-0620-x] [Citation(s) in RCA: 271] [Impact Index Per Article: 54.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 01/24/2020] [Indexed: 12/13/2022]
Abstract
Programmed cell death protein 1 (PD-1) ligation delimits immunogenic responses in T cells. However, the consequences of programmed cell death 1 ligand 1 (PD-L1) ligation in T cells are uncertain. We found that T cell expression of PD-L1 in cancer was regulated by tumor antigen and sterile inflammatory cues. PD-L1+ T cells exerted tumor-promoting tolerance via three distinct mechanisms: (1) binding of PD-L1 induced STAT3-dependent 'back-signaling' in CD4+ T cells, which prevented activation, reduced TH1-polarization and directed TH17-differentiation. PD-L1 signaling also induced an anergic T-bet-IFN-γ- phenotype in CD8+ T cells and was equally suppressive compared to PD-1 signaling; (2) PD-L1+ T cells restrained effector T cells via the canonical PD-L1-PD-1 axis and were sufficient to accelerate tumorigenesis, even in the absence of endogenous PD-L1; (3) PD-L1+ T cells engaged PD-1+ macrophages, inducing an alternative M2-like program, which had crippling effects on adaptive antitumor immunity. Collectively, we demonstrate that PD-L1+ T cells have diverse tolerogenic effects on tumor immunity.
Collapse
|
24
|
Munir F, Jamshed MB, Shahid N, Hussain HM, Muhammad SA, Mamun AA, Zhang Q. Advances in immunomodulatory therapy for severe acute pancreatitis. Immunol Lett 2020; 217:72-76. [DOI: 10.1016/j.imlet.2019.11.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 10/24/2019] [Accepted: 11/06/2019] [Indexed: 02/08/2023]
|
25
|
Pan LL, Niu W, Fang X, Liang W, Li H, Chen W, Zhang H, Bhatia M, Sun J. Clostridium butyricum Strains Suppress Experimental Acute Pancreatitis by Maintaining Intestinal Homeostasis. Mol Nutr Food Res 2019; 63:e1801419. [PMID: 31034143 DOI: 10.1002/mnfr.201801419] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 04/13/2019] [Indexed: 01/24/2023]
Abstract
SCOPE Acute pancreatitis (AP) is a common abdominal inflammatory disease. Disturbed gut homeostasis secondary to pancreatic inflammation aggravates the condition retroactively. The current study investigates potential beneficial effects of Clostridium butyricum (C. butyricum) strains on AP and underlying mechanisms. METHODS AND RESULTS C. butyricum strains MIYAIRI 588 (CBM588) and CGMCC0313.1 (CB0313.1) were supplemented to mice for three weeks before experimental AP or SAP induction. Both CBM588 and CB0313.1 protected against AP, as evidenced by reduced serum amylase and lipase levels, pancreatic edema, and myeloperoxidase activity. Amelioration of both experimental AP and SAP by CB0313.1 indicated a non-model-specific effect. Moreover, C. butyricum inhibited pancreatic neutrophil and dendritic cell infiltration, nucleotide-binding domain leucine-rich repeat-containing family, pyrin domain-containing 3 inflammasome activation, and pro-inflammatory pathways. Additionally in the gut, C. butyricum strains attenuated AP-associated intestinal inflammation and barrier dysfunction, accompanied with reduced pathogenic bacteria Escherichia coli and Enterococcus penetration into pancreas. Gut microbiome analyses further revealed that beneficial effects of C. butyricum on pancreatic-gut homeostasis were correlated with improved dysbiosis. In particular, relative abundance of Desulfovibrionaceae decreased, and Verrucomicrobiaceae Clostridiaceae and Lactobacillaceae increased. CONCLUSIONS For the first time, a protective effect of C. butyricum in AP by modulating intestinal homeostasis is demonstrated.
Collapse
Affiliation(s)
- Li-Long Pan
- School of Medicine, Jiangnan University, Wuxi, 214122, P. R. China
| | - Wenying Niu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, P. R. China
| | - Xin Fang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, P. R. China
| | - Wenjie Liang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, P. R. China
| | - Hongli Li
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, P. R. China
| | - Wei Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, P. R. China
| | - Hao Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, P. R. China
| | - Madhav Bhatia
- Inflammation Research Group, Department of Pathology, University of Otago, Christchurch, 8140, New Zealand
| | - Jia Sun
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, P. R. China
| |
Collapse
|
26
|
Liu ZY, Zheng M, Li YM, Fan XY, Wang JC, Li ZC, Yang HJ, Yu JM, Cui J, Jiang JL, Tang J, Chen ZN. RIP3 promotes colitis-associated colorectal cancer by controlling tumor cell proliferation and CXCL1-induced immune suppression. Am J Cancer Res 2019; 9:3659-3673. [PMID: 31281505 PMCID: PMC6587173 DOI: 10.7150/thno.32126] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Accepted: 05/12/2019] [Indexed: 12/19/2022] Open
Abstract
Rationale: Necroptosis is a programmed form of non-apoptotic cell death that requires receptor-interacting protein 3 (RIP3). RIP3 has been shown to be relevant in multiple tumor types and has differential impact on tumor progression. We investigated whether RIP3 is involved in the progression of colitis-associated cancer (CAC) in mice. Methods: Tissues from colorectal cancer patients were examined for RIP3 expression. CAC was induced using azoxymethane (AOM) injection followed by dextran sodium sulfate (DSS) treatment in RIP3-deficient or wild-type mice. Colon tissues were collected and analyzed by Western blotting and gene expression profile analyses. Immune cell infiltration and CXCL1 expression were examined by flow cytometry and Real-time PCR, respectively. Results: RIP3 expression was upregulated in mouse CAC and human colon cancer. RIP3-deficient mice showed significantly attenuated colitis-associated tumorigenesis. Bone marrow transplantation experiments suggested that RIP3's function in hematopoietic cells primarily contributes to the phenotype. RIP3 supported epithelial proliferation and tumor growth via JNK signaling but had no effect on apoptosis. RIP3 deletion increased T cell accumulation and reduced infiltration by immunosuppressive subsets of myeloid cells during acute colitis and CAC. The immune-suppressive tumor microenvironment was dependent on RIP3-induced expression of the chemokine attractant CXCL1, and administration of recombinant CXCL1 during CAC restored tumorigenesis in Rip3-/- mice. Conclusion: Our results reveal an unexpected function of RIP3 in enhancing the proliferation of premalignant intestinal epithelial cells (IECs) and promoting myeloid cell-induced adaptive immune suppression. These two distinct mechanisms of RIP3-induced JNK and CXCL1 signalling contribute to CAC progression.
Collapse
|
27
|
Pan LL, Deng YY, Wang R, Wu C, Li J, Niu W, Yang Q, Bhatia M, Gudmundsson GH, Agerberth B, Diana J, Sun J. Lactose Induces Phenotypic and Functional Changes of Neutrophils and Macrophages to Alleviate Acute Pancreatitis in Mice. Front Immunol 2018; 9:751. [PMID: 29719535 PMCID: PMC5913286 DOI: 10.3389/fimmu.2018.00751] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 03/26/2018] [Indexed: 12/12/2022] Open
Abstract
Acute pancreatitis (AP) is one common clinical acute abdominal disease, for which specific pharmacological or nutritional therapies remain elusive. Lactose, a macronutrient and an inducer of host innate immune responses, possesses immune modulatory functions. The current study aimed to investigate potential modulatory effects of lactose and the interplay between the nutrient and pancreatic immunity during experimentally induced AP in mice. We found that either prophylactic or therapeutic treatment of lactose time-dependently reduced the severity of AP, as evidenced by reduced pancreatic edema, serum amylase levels, and pancreatic myeloperoxidase activities, as well as by histological examination of pancreatic damage. Overall, lactose promoted a regulatory cytokine milieu in the pancreas and reduced infiltration of inflammatory neutrophils and macrophages. On acinar cells, lactose was able to suppress caerulein-induced inflammatory signaling pathways and to suppress chemoattractant tumor necrosis factor (TNF)-α and monocyte chemotactic protein-1 production. Additionally, lactose acted on pancreas-infiltrated macrophages, increasing interleukin-10 and decreasing tumor necrosis factor alpha production. Notably, lactose treatment reversed AP-associated infiltration of activated neutrophils. Last, the effect of lactose on neutrophil infiltration was mimicked by a galectin-3 antagonist, suggesting a potential endogenous target of lactose. Together, the current study demonstrates an immune regulatory effect of lactose to alleviate AP and suggests its potential as a convenient, value-added therapeutic macronutrient to control AP, and lower the risk of its systemic complications.
Collapse
Affiliation(s)
- Li-Long Pan
- School of Medicine, Jiangnan University, Wuxi, China
| | - Yuan-Yuan Deng
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
- Nutrition and Immunology Laboratory, School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Ruxing Wang
- School of Medicine, Jiangnan University, Wuxi, China
- Wuxi People’s Hospital Affiliated to Nanjing Medical University, Wuxi, China
| | - Chengfei Wu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
- Nutrition and Immunology Laboratory, School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Jiahong Li
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
- Nutrition and Immunology Laboratory, School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Wenying Niu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
- Nutrition and Immunology Laboratory, School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Qin Yang
- School of Medicine, Jiangnan University, Wuxi, China
| | - Madhav Bhatia
- Inflammation Research Group, Department of Pathology, University of Otago, Christchurch, New Zealand
| | | | - Birgitta Agerberth
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Julien Diana
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unité 1151, Institute Necker-Enfants Malades (INEM), Centre National de la Recherche Scienctifique, Unité 8253, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Jia Sun
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
- Nutrition and Immunology Laboratory, School of Food Science and Technology, Jiangnan University, Wuxi, China
| |
Collapse
|
28
|
Gukovskaya AS, Gukovsky I, Algül H, Habtezion A. Autophagy, Inflammation, and Immune Dysfunction in the Pathogenesis of Pancreatitis. Gastroenterology 2017; 153:1212-1226. [PMID: 28918190 PMCID: PMC6338477 DOI: 10.1053/j.gastro.2017.08.071] [Citation(s) in RCA: 243] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 08/09/2017] [Accepted: 08/17/2017] [Indexed: 12/13/2022]
Abstract
Pancreatitis is a common disorder with significant morbidity and mortality, yet little is known about its pathogenesis, and there is no specific or effective treatment. Its development involves dysregulated autophagy and unresolved inflammation, demonstrated by studies in genetic and experimental mouse models. Disease severity depends on whether the inflammatory response resolves or amplifies, leading to multi-organ failure. Dysregulated autophagy might promote the inflammatory response in the pancreas. We discuss the roles of autophagy and inflammation in pancreatitis, mechanisms of deregulation, and connections among disordered pathways. We identify gaps in our knowledge and delineate perspective directions for research. Elucidation of pathogenic mechanisms could lead to new targets for treating or reducing the severity of pancreatitis.
Collapse
Affiliation(s)
- Anna S Gukovskaya
- Department of Medicine, David Geffen School of Medicine, University of California at Los Angeles, California; Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California.
| | - Ilya Gukovsky
- Department of Medicine, David Geffen School of Medicine, University of California at Los Angeles, California; Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California
| | - Hana Algül
- II Medizinische Klinik, Klinikum Rechts der Isar, Technische Universität München, Munich, Germany
| | - Aida Habtezion
- Division of Gastroenterology and Hepatology, Stanford University School of Medicine, Stanford, California
| |
Collapse
|
29
|
He Y, Wu C, Li J, Li H, Sun Z, Zhang H, de Vos P, Pan LL, Sun J. Inulin-Type Fructans Modulates Pancreatic-Gut Innate Immune Responses and Gut Barrier Integrity during Experimental Acute Pancreatitis in a Chain Length-Dependent Manner. Front Immunol 2017; 8:1209. [PMID: 29018453 PMCID: PMC5622924 DOI: 10.3389/fimmu.2017.01209] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 09/12/2017] [Indexed: 12/11/2022] Open
Abstract
Acute pancreatitis (AP) is a common abdominal inflammatory disorder and one of the leading causes of hospital admission for gastrointestinal disorders. No specific pharmacological or nutritional therapy is available but highly needed. Inulin-type fructans (ITFs) are capable of modifying gut immune and barrier homeostasis in a chemistry-dependent manner and hence potentially applicable for managing AP, but their efficacy in AP has not been demonstrated yet. The current study aimed to examine and compare modulatory effects of ITFs with different degrees of fermentability on pancreatic-gut immunity and barrier function during experimentally induced AP in mice. BALB/c mice were fed short (I)- or long (IV)-chain ITFs supplemented diets for up to 3 days before AP induction by caerulein. Attenuating effects on AP development were stronger with ITF IV than with ITF I. We found that long-chain ITF IV attenuated the severity of AP, as evidenced by reduced serum amylase levels, lipase levels, pancreatic myeloperoxidase activity, pancreatic edema, and histological examination demonstrating reduced pancreatic damage. Short-chain ITF I demonstrated only partial protective effects. Both ITF IV and ITF I modulated AP-associated systemic cytokine levels. ITF IV but not ITF I restored AP-associated intestinal barrier dysfunction by upregulating colonic tight junction modulatory proteins, antimicrobial peptides, and improved general colonic histology. Additionally, differential modulatory effects of ITF IV and ITF I were observed on pancreatic and gut immunity: ITF IV supplementation prevented innate immune cell infiltration in the pancreas and colon and tissue cytokine production. Similar effects were only observed in the gut with ITF I and not in the pancreas. Lastly, ITF IV but not ITF I downregulated AP-triggered upregulation of IL-1 receptor-associated kinase 4 (IRAK-4) and phosphor-c-Jun N-terminal kinase (p-JNK), and a net decrease of phosphor-nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) p65 (p-NF-κB p65) nuclear translocation and activation in the pancreas. Our findings demonstrate a clear chain length-dependent effect of inulin on AP. The attenuating effects are caused by modulating effects of long-chain inulin on the pancreatic-gut immunity via the pancreatic IRAK-4/p-JNK/p-NF-κBp65 signaling pathway and on prevention of disruption of the gut barrier.
Collapse
Affiliation(s)
- Yue He
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Chengfei Wu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Jiahong Li
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Hongli Li
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Zhenghua Sun
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Hao Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Paul de Vos
- Division of Medical Biology, Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Li-Long Pan
- School of Medicine, Jiangnan University, Wuxi, China
| | - Jia Sun
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
- School of Medicine, Jiangnan University, Wuxi, China
| |
Collapse
|
30
|
Daley D, Mani VR, Mohan N, Akkad N, Ochi A, Heindel DW, Lee KB, Zambirinis CP, Pandian GSB, Savadkar S, Torres-Hernandez A, Nayak S, Wang D, Hundeyin M, Diskin B, Aykut B, Werba G, Barilla RM, Rodriguez R, Chang S, Gardner L, Mahal LK, Ueberheide B, Miller G. Dectin 1 activation on macrophages by galectin 9 promotes pancreatic carcinoma and peritumoral immune tolerance. Nat Med 2017; 23:556-567. [PMID: 28394331 PMCID: PMC5419876 DOI: 10.1038/nm.4314] [Citation(s) in RCA: 277] [Impact Index Per Article: 34.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 03/01/2017] [Indexed: 12/29/2022]
Abstract
The progression of pancreatic oncogenesis requires immune-suppressive inflammation in cooperation with oncogenic mutations. However, the drivers of intratumoral immune tolerance are uncertain. Dectin 1 is an innate immune receptor crucial for anti-fungal immunity, but its role in sterile inflammation and oncogenesis has not been well defined. Furthermore, non-pathogen-derived ligands for dectin 1 have not been characterized. We found that dectin 1 is highly expressed on macrophages in pancreatic ductal adenocarcinoma (PDA). Dectin 1 ligation accelerated the progression of PDA in mice, whereas deletion of Clec7a-the gene encoding dectin 1-or blockade of dectin 1 downstream signaling was protective. We found that dectin 1 can ligate the lectin galectin 9 in mouse and human PDA, which results in tolerogenic macrophage programming and adaptive immune suppression. Upon disruption of the dectin 1-galectin 9 axis, CD4+ and CD8+ T cells, which are dispensable for PDA progression in hosts with an intact signaling axis, become reprogrammed into indispensable mediators of anti-tumor immunity. These data suggest that targeting dectin 1 signaling is an attractive strategy for developing an immunotherapy for PDA.
Collapse
Affiliation(s)
- Donnele Daley
- S.A. Localio Laboratory, Department of Surgery, New York University School of Medicine, 430 East 29th Street, New York, NY 10016
| | - Vishnu R. Mani
- S.A. Localio Laboratory, Department of Surgery, New York University School of Medicine, 430 East 29th Street, New York, NY 10016
| | - Navyatha Mohan
- S.A. Localio Laboratory, Department of Surgery, New York University School of Medicine, 430 East 29th Street, New York, NY 10016
| | - Neha Akkad
- S.A. Localio Laboratory, Department of Surgery, New York University School of Medicine, 430 East 29th Street, New York, NY 10016
| | - Atsuo Ochi
- S.A. Localio Laboratory, Department of Surgery, New York University School of Medicine, 430 East 29th Street, New York, NY 10016
| | - Daniel W. Heindel
- Department of Chemistry, 100 Washington Square East, New York University, New York, NY 10003
| | - Ki Buom Lee
- S.A. Localio Laboratory, Department of Surgery, New York University School of Medicine, 430 East 29th Street, New York, NY 10016
| | - Constantinos P. Zambirinis
- S.A. Localio Laboratory, Department of Surgery, New York University School of Medicine, 430 East 29th Street, New York, NY 10016
| | | | - Shivraj Savadkar
- S.A. Localio Laboratory, Department of Surgery, New York University School of Medicine, 430 East 29th Street, New York, NY 10016
| | - Alejandro Torres-Hernandez
- S.A. Localio Laboratory, Department of Surgery, New York University School of Medicine, 430 East 29th Street, New York, NY 10016
| | - Shruti Nayak
- S.A. Localio Laboratory, Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, 430 East 29th Street, New York, NY 10016
| | - Ding Wang
- S.A. Localio Laboratory, Department of Medicine, New York University School of Medicine, 430 East 29th Street, New York, NY 10016
| | - Mautin Hundeyin
- S.A. Localio Laboratory, Department of Surgery, New York University School of Medicine, 430 East 29th Street, New York, NY 10016
| | - Brian Diskin
- S.A. Localio Laboratory, Department of Surgery, New York University School of Medicine, 430 East 29th Street, New York, NY 10016
| | - Berk Aykut
- S.A. Localio Laboratory, Department of Surgery, New York University School of Medicine, 430 East 29th Street, New York, NY 10016
| | - Gregor Werba
- S.A. Localio Laboratory, Department of Surgery, New York University School of Medicine, 430 East 29th Street, New York, NY 10016
| | - Rocky M. Barilla
- S.A. Localio Laboratory, Department of Surgery, New York University School of Medicine, 430 East 29th Street, New York, NY 10016
| | - Robert Rodriguez
- S.A. Localio Laboratory, Department of Surgery, New York University School of Medicine, 430 East 29th Street, New York, NY 10016
| | - Steven Chang
- S.A. Localio Laboratory, Department of Surgery, New York University School of Medicine, 430 East 29th Street, New York, NY 10016
| | - Lawrence Gardner
- S.A. Localio Laboratory, Department of Medicine, New York University School of Medicine, 430 East 29th Street, New York, NY 10016
| | - Lara K. Mahal
- Department of Chemistry, 100 Washington Square East, New York University, New York, NY 10003
| | - Beatrix Ueberheide
- S.A. Localio Laboratory, Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, 430 East 29th Street, New York, NY 10016
| | - George Miller
- S.A. Localio Laboratory, Department of Surgery, New York University School of Medicine, 430 East 29th Street, New York, NY 10016
- S.A. Localio Laboratory, Department of Cell Biology, New York University School of Medicine, 430 East 29th Street, New York, NY 10016
| |
Collapse
|
31
|
Abstract
OBJECTIVES This study was conducted to assess the preventive/therapeutic effects of combined administration of resveratrol and guggulsterone on cerulein-induced acute pancreatitis in mice. METHODS Acute pancreatitis was induced by intraperitoneal injection of cerulein in mice. Serum amylase assay and histology were performed to measure the severity of pancreatitis. Western blotting and multiplex cytokine/chemokine analysis were conducted to understand the action mechanisms of the reagents. RESULTS Serum amylase assay and histology revealed that the severity of acute pancreatitis was reduced by the combinatory treatment with resveratrol and guggulsterone, but the ratio of the band intensity implied that reduced nuclear factor-κB activation is primarily responsible for the effect. The reduced amounts of keratinocyte chemoattractant (chemokine [C-X-C motif] ligand 1), interferon gamma-induced protein 10 (C-X-C motif chemokine 10) and interleukin 6 expression in the sera could be involved in attenuated immune cell migration and reduced inflammation by these reagents. CONCLUSIONS Combinatory treatment with resveratrol and guggulsterone marginally reduced cerulein-induced mild acute pancreatitis in mice.
Collapse
|
32
|
Immunopathogenesis of pancreatitis. Mucosal Immunol 2017; 10:283-298. [PMID: 27848953 DOI: 10.1038/mi.2016.101] [Citation(s) in RCA: 140] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 10/06/2016] [Indexed: 02/04/2023]
Abstract
The conventional view of the pathogenesis of acute and chronic pancreatitis is that it is due to a genetic- or environment-based abnormality of intracellular acinar trypsinogen activation and thus to the induction of acinar cell injury that, in turn, sets in motion an intra-pancreatic inflammatory process. More recent studies, reviewed here, present strong evidence that while such trypsinogen activation is likely a necessary first step in the inflammatory cascade underlying pancreatitis, sustained pancreatic inflammation is dependent on damage-associated molecular patterns-mediated cytokine activation causing the translocation of commensal (gut) organisms into the circulation and their induction of innate immune responses in acinar cells. Quite unexpectedly, these recent studies reveal that the innate responses involve activation of responses by an innate factor, nucleotide-binding oligomerization domain 1 (NOD1), and that such NOD1 responses have a critical role in the activation/production of nuclear factor-kappa B and type I interferon. In addition, they reveal that chronic inflammation and its accompanying fibrosis are dependent on the generation of IL-33 by injured acinar cells and its downstream induction of T cells producing IL-13. These recent studies thus establish that pancreatitis is quite a unique form of inflammation and one susceptible to newer, more innovative therapy.
Collapse
|
33
|
Abstract
PURPOSE OF REVIEW Acute pancreatitis is a major cause of gastrointestinal morbidity for which specific therapy is greatly needed to prevent progression to and induce resolution of severe disease. RECENT FINDINGS Innate immune components and metabolite signaling are recently identified as strong determinants of disease severity and resolution in acute pancreatitis and this work will be discussed herein. SUMMARY Targeting innate immune cell populations and metabolite signaling pathways in acute pancreatitis may result in broader and ultimately more efficacious re-direction of the inflammatory programme toward disease resolution and improved clinical outcomes.
Collapse
|
34
|
Daley D, Zambirinis CP, Seifert L, Akkad N, Mohan N, Werba G, Barilla R, Torres-Hernandez A, Hundeyin M, Mani VRK, Avanzi A, Tippens D, Narayanan R, Jang JE, Newman E, Pillarisetty VG, Dustin ML, Bar-Sagi D, Hajdu C, Miller G. γδ T Cells Support Pancreatic Oncogenesis by Restraining αβ T Cell Activation. Cell 2016; 166:1485-1499.e15. [PMID: 27569912 PMCID: PMC5017923 DOI: 10.1016/j.cell.2016.07.046] [Citation(s) in RCA: 271] [Impact Index Per Article: 30.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 02/16/2016] [Accepted: 07/27/2016] [Indexed: 02/07/2023]
Abstract
Inflammation is paramount in pancreatic oncogenesis. We identified a uniquely activated γδT cell population, which constituted ∼40% of tumor-infiltrating T cells in human pancreatic ductal adenocarcinoma (PDA). Recruitment and activation of γδT cells was contingent on diverse chemokine signals. Deletion, depletion, or blockade of γδT cell recruitment was protective against PDA and resulted in increased infiltration, activation, and Th1 polarization of αβT cells. Although αβT cells were dispensable to outcome in PDA, they became indispensable mediators of tumor protection upon γδT cell ablation. PDA-infiltrating γδT cells expressed high levels of exhaustion ligands and thereby negated adaptive anti-tumor immunity. Blockade of PD-L1 in γδT cells enhanced CD4(+) and CD8(+) T cell infiltration and immunogenicity and induced tumor protection suggesting that γδT cells are critical sources of immune-suppressive checkpoint ligands in PDA. We describe γδT cells as central regulators of effector T cell activation in cancer via novel cross-talk.
Collapse
Affiliation(s)
- Donnele Daley
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, 430 East 29th Street, New York, NY 10016, USA; Department of Cell Biology, New York University School of Medicine, 430 East 29th Street, New York, NY 10016, USA
| | - Constantinos Pantelis Zambirinis
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, 430 East 29th Street, New York, NY 10016, USA; Department of Cell Biology, New York University School of Medicine, 430 East 29th Street, New York, NY 10016, USA
| | - Lena Seifert
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, 430 East 29th Street, New York, NY 10016, USA; Department of Cell Biology, New York University School of Medicine, 430 East 29th Street, New York, NY 10016, USA
| | - Neha Akkad
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, 430 East 29th Street, New York, NY 10016, USA
| | - Navyatha Mohan
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, 430 East 29th Street, New York, NY 10016, USA
| | - Gregor Werba
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, 430 East 29th Street, New York, NY 10016, USA
| | - Rocky Barilla
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, 430 East 29th Street, New York, NY 10016, USA
| | - Alejandro Torres-Hernandez
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, 430 East 29th Street, New York, NY 10016, USA
| | - Mautin Hundeyin
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, 430 East 29th Street, New York, NY 10016, USA
| | - Vishnu Raj Kumar Mani
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, 430 East 29th Street, New York, NY 10016, USA
| | - Antonina Avanzi
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, 430 East 29th Street, New York, NY 10016, USA
| | - Daniel Tippens
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, 430 East 29th Street, New York, NY 10016, USA
| | - Rajkishen Narayanan
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, 430 East 29th Street, New York, NY 10016, USA
| | - Jung-Eun Jang
- Department of Biochemistry, New York University School of Medicine, 430 East 29th Street, New York, NY 10016, USA; Department of Pathology, New York University School of Medicine, 430 East 29th Street, New York, NY 10016, USA
| | - Elliot Newman
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, 430 East 29th Street, New York, NY 10016, USA
| | - Venu Gopal Pillarisetty
- Department of Surgery, University of Washington School of Medicine, 1959 NE Pacific Street, Seattle, WA 98195, USA
| | - Michael Loran Dustin
- Department of Pathology, New York University School of Medicine, 430 East 29th Street, New York, NY 10016, USA; The Kennedy Institute of Rheumatology, University of Oxford, Roosevelt Drive, Headington Oxford OX3 7FY, UK
| | - Dafna Bar-Sagi
- Department of Biochemistry, New York University School of Medicine, 430 East 29th Street, New York, NY 10016, USA
| | - Cristina Hajdu
- Department of Pathology, New York University School of Medicine, 430 East 29th Street, New York, NY 10016, USA
| | - George Miller
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, 430 East 29th Street, New York, NY 10016, USA; Department of Cell Biology, New York University School of Medicine, 430 East 29th Street, New York, NY 10016, USA.
| |
Collapse
|
35
|
Greco SH, Torres-Hernandez A, Kalabin A, Whiteman C, Rokosh R, Ravirala S, Ochi A, Gutierrez J, Salyana MA, Mani VR, Nagaraj SV, Deutsch M, Seifert L, Daley D, Barilla R, Hundeyin M, Nikifrov Y, Tejada K, Gelb BE, Katz SC, Miller G. Mincle Signaling Promotes Con A Hepatitis. THE JOURNAL OF IMMUNOLOGY 2016; 197:2816-27. [PMID: 27559045 DOI: 10.4049/jimmunol.1600598] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 07/21/2016] [Indexed: 12/12/2022]
Abstract
Con A hepatitis is regarded as a T cell-mediated model of acute liver injury. Mincle is a C-type lectin receptor that is critical in the immune response to mycobacteria and fungi but does not have a well-defined role in preclinical models of non-pathogen-mediated inflammation. Because Mincle can ligate the cell death ligand SAP130, we postulated that Mincle signaling drives intrahepatic inflammation and liver injury in Con A hepatitis. Acute liver injury was assessed in the murine Con A hepatitis model using C57BL/6, Mincle(-/-), and Dectin-1(-/-) mice. The role of C/EBPβ and hypoxia-inducible factor-1α (HIF-1α) signaling was assessed using selective inhibitors. We found that Mincle was highly expressed in hepatic innate inflammatory cells and endothelial cells in both mice and humans. Furthermore, sterile Mincle ligands and Mincle signaling intermediates were increased in the murine liver in Con A hepatitis. Most significantly, Mincle deletion or blockade protected against Con A hepatitis, whereas Mincle ligation exacerbated disease. Bone marrow chimeric and adoptive transfer experiments suggested that Mincle signaling in infiltrating myeloid cells dictates disease phenotype. Conversely, signaling via other C-type lectin receptors did not alter disease course. Mechanistically, we found that Mincle blockade decreased the NF-κβ-related signaling intermediates C/EBPβ and HIF-1α, both of which are necessary in macrophage-mediated inflammatory responses. Accordingly, Mincle deletion lowered production of nitrites in Con A hepatitis and inhibition of both C/EBPβ and HIF-1α reduced the severity of liver disease. Our work implicates a novel innate immune driver of Con A hepatitis and, more broadly, suggests a potential role for Mincle in diseases governed by sterile inflammation.
Collapse
Affiliation(s)
- Stephanie H Greco
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, New York, NY 10016
| | - Alejandro Torres-Hernandez
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, New York, NY 10016
| | - Aleksandr Kalabin
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, New York, NY 10016
| | - Clint Whiteman
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, New York, NY 10016
| | - Rae Rokosh
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, New York, NY 10016
| | - Sushma Ravirala
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, New York, NY 10016
| | - Atsuo Ochi
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, New York, NY 10016
| | - Johana Gutierrez
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, New York, NY 10016
| | - Muhammad Atif Salyana
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, New York, NY 10016
| | - Vishnu R Mani
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, New York, NY 10016
| | - Savitha V Nagaraj
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, New York, NY 10016
| | - Michael Deutsch
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, New York, NY 10016
| | - Lena Seifert
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, New York, NY 10016
| | - Donnele Daley
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, New York, NY 10016
| | - Rocky Barilla
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, New York, NY 10016
| | - Mautin Hundeyin
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, New York, NY 10016
| | - Yuriy Nikifrov
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, New York, NY 10016
| | - Karla Tejada
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, New York, NY 10016
| | - Bruce E Gelb
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, New York, NY 10016
| | - Steven C Katz
- Immunotherapy Program, Roger Williams Medical Center, Providence, RI 02908; and
| | - George Miller
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, New York, NY 10016; Department of Cell Biology, New York University School of Medicine, New York, NY 10016
| |
Collapse
|
36
|
Seifert L, Werba G, Tiwari S, Giao Ly NN, Alothman S, Alqunaibit D, Avanzi A, Barilla R, Daley D, Greco SH, Torres-Hernandez A, Pergamo M, Ochi A, Zambirinis CP, Pansari M, Rendon M, Tippens D, Hundeyin M, Mani VR, Hajdu C, Engle D, Miller G. The necrosome promotes pancreatic oncogenesis via CXCL1 and Mincle-induced immune suppression. Nature 2016; 532:245-9. [PMID: 27049944 PMCID: PMC4833566 DOI: 10.1038/nature17403] [Citation(s) in RCA: 474] [Impact Index Per Article: 52.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 02/05/2016] [Indexed: 12/11/2022]
Abstract
Neoplastic pancreatic epithelial cells are believed to die through caspase 8-dependent apoptotic cell death, and chemotherapy is thought to promote tumour apoptosis. Conversely, cancer cells often disrupt apoptosis to survive. Another type of programmed cell death is necroptosis (programmed necrosis), but its role in pancreatic ductal adenocarcinoma (PDA) is unclear. There are many potential inducers of necroptosis in PDA, including ligation of tumour necrosis factor receptor 1 (TNFR1), CD95, TNF-related apoptosis-inducing ligand (TRAIL) receptors, Toll-like receptors, reactive oxygen species, and chemotherapeutic drugs. Here we report that the principal components of the necrosome, receptor-interacting protein (RIP)1 and RIP3, are highly expressed in PDA and are further upregulated by the chemotherapy drug gemcitabine. Blockade of the necrosome in vitro promoted cancer cell proliferation and induced an aggressive oncogenic phenotype. By contrast, in vivo deletion of RIP3 or inhibition of RIP1 protected against oncogenic progression in mice and was associated with the development of a highly immunogenic myeloid and T cell infiltrate. The immune-suppressive tumour microenvironment associated with intact RIP1/RIP3 signalling depended in part on necroptosis-induced expression of the chemokine attractant CXCL1, and CXCL1 blockade protected against PDA. Moreover, cytoplasmic SAP130 (a subunit of the histone deacetylase complex) was expressed in PDA in a RIP1/RIP3-dependent manner, and Mincle--its cognate receptor--was upregulated in tumour-infiltrating myeloid cells. Ligation of Mincle by SAP130 promoted oncogenesis, whereas deletion of Mincle protected against oncogenesis and phenocopied the immunogenic reprogramming of the tumour microenvironment that was induced by RIP3 deletion. Cellular depletion suggested that whereas inhibitory macrophages promote tumorigenesis in PDA, they lose their immune-suppressive effects when RIP3 or Mincle is deleted. Accordingly, T cells, which are not protective against PDA progression in mice with intact RIP3 or Mincle signalling, are reprogrammed into indispensable mediators of anti-tumour immunity in the absence of RIP3 or Mincle. Our work describes parallel networks of necroptosis-induced CXCL1 and Mincle signalling that promote macrophage-induced adaptive immune suppression and thereby enable PDA progression.
Collapse
Affiliation(s)
- Lena Seifert
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, 550 First Avenue, New York, NY 10016
| | - Gregor Werba
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, 550 First Avenue, New York, NY 10016
| | - Shaun Tiwari
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, 550 First Avenue, New York, NY 10016
| | - Nancy Ngoc Giao Ly
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, 550 First Avenue, New York, NY 10016
| | - Sara Alothman
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, 550 First Avenue, New York, NY 10016
| | - Dalia Alqunaibit
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, 550 First Avenue, New York, NY 10016
| | - Antonina Avanzi
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, 550 First Avenue, New York, NY 10016
| | - Rocky Barilla
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, 550 First Avenue, New York, NY 10016
| | - Donnele Daley
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, 550 First Avenue, New York, NY 10016
| | - Stephanie H. Greco
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, 550 First Avenue, New York, NY 10016
| | - Alejandro Torres-Hernandez
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, 550 First Avenue, New York, NY 10016
| | - Matthew Pergamo
- S. Arthur Localio Laboratory, Department of Cell Biology, New York University School of Medicine, 550 First Avenue, New York, NY 10016
| | - Atsuo Ochi
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, 550 First Avenue, New York, NY 10016
| | - Constantinos P. Zambirinis
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, 550 First Avenue, New York, NY 10016
| | - Mridul Pansari
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, 550 First Avenue, New York, NY 10016
| | - Mauricio Rendon
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, 550 First Avenue, New York, NY 10016
| | - Daniel Tippens
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, 550 First Avenue, New York, NY 10016
| | - Mautin Hundeyin
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, 550 First Avenue, New York, NY 10016
| | - Vishnu R. Mani
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, 550 First Avenue, New York, NY 10016
| | - Cristina Hajdu
- S. Arthur Localio Laboratory, Department of Pathology, New York University School of Medicine, 550 First Avenue, New York, NY 10016
| | - Dannielle Engle
- Cold Spring Harbor Laboratories, Cold Spring Harbor, NY 11724
| | - George Miller
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, 550 First Avenue, New York, NY 10016
- S. Arthur Localio Laboratory, Department of Cell Biology, New York University School of Medicine, 550 First Avenue, New York, NY 10016
| |
Collapse
|
37
|
Shamoon M, Deng Y, Chen YQ, Bhatia M, Sun J. Therapeutic implications of innate immune system in acute pancreatitis. Expert Opin Ther Targets 2015; 20:73-87. [PMID: 26565751 DOI: 10.1517/14728222.2015.1077227] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Acute pancreatitis (AP) is an inflammatory disorder of the pancreas encompassing a cascade of cellular and molecular events. It starts from premature activation of zymogens with the involvement of innate immune system to a potential systemic inflammatory response and multiple organ failure. Leukocytes are the major cell population that participate in the propagation of the disease. Current understanding of the course of AP is still far from complete, limiting treatment options mostly to conservative supportive care. Emerging evidence has pointed to modulation of the immune system for strategic therapeutic development, by mitigating the inflammatory response and severity of AP. In the current review, we have focused on the role of innate immunity in the condition and highlighted therapeutics targeting it for treatment of this challenging disease. AREAS COVERED The current review has aimed to elaborate in-depth understanding of specific roles of innate immune cells, derived mediators and inflammatory pathways that are involved in AP. Summarizing the recent therapeutics and approaches applied experimentally that target immune responses to attenuate AP. EXPERT OPINION The current state of knowledge on AP, limitations of presently available therapeutic approaches and the promise of therapeutic implications of innate immune system in AP are discussed.
Collapse
Affiliation(s)
- Muhammad Shamoon
- a 1 Jiangnan University, School of Food Science and Technology, The Synergetic Innovation Center of Food Safety and Nutrition, State Key Laboratory of Food Science and Technology , Wuxi, Jiangsu, China
| | - Yuanyuan Deng
- a 1 Jiangnan University, School of Food Science and Technology, The Synergetic Innovation Center of Food Safety and Nutrition, State Key Laboratory of Food Science and Technology , Wuxi, Jiangsu, China
| | - Yong Q Chen
- a 1 Jiangnan University, School of Food Science and Technology, The Synergetic Innovation Center of Food Safety and Nutrition, State Key Laboratory of Food Science and Technology , Wuxi, Jiangsu, China
| | - Madhav Bhatia
- b 2 University of Otago, Inflammation Research Group, Department of Pathology , Christchurch, 2 Riccarton Avenue, P.O. Box 4345, Christchurch 8140, New Zealand
| | - Jia Sun
- a 1 Jiangnan University, School of Food Science and Technology, The Synergetic Innovation Center of Food Safety and Nutrition, State Key Laboratory of Food Science and Technology , Wuxi, Jiangsu, China
| |
Collapse
|
38
|
Greco SH, Tomkötter L, Vahle AK, Rokosh R, Avanzi A, Mahmood SK, Deutsch M, Alothman S, Alqunaibit D, Ochi A, Zambirinis C, Mohaimin T, Rendon M, Levie E, Pansari M, Torres-Hernandez A, Daley D, Barilla R, Pachter HL, Tippens D, Malik H, Boutajangout A, Wisniewski T, Miller G. TGF-β Blockade Reduces Mortality and Metabolic Changes in a Validated Murine Model of Pancreatic Cancer Cachexia. PLoS One 2015; 10:e0132786. [PMID: 26172047 PMCID: PMC4501823 DOI: 10.1371/journal.pone.0132786] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Accepted: 06/18/2015] [Indexed: 01/05/2023] Open
Abstract
Cancer cachexia is a debilitating condition characterized by a combination of anorexia, muscle wasting, weight loss, and malnutrition. This condition affects an overwhelming majority of patients with pancreatic cancer and is a primary cause of cancer-related death. However, few, if any, effective therapies exist for both treatment and prevention of this syndrome. In order to develop novel therapeutic strategies for pancreatic cancer cachexia, appropriate animal models are necessary. In this study, we developed and validated a syngeneic, metastatic, murine model of pancreatic cancer cachexia. Using our model, we investigated the ability of transforming growth factor beta (TGF-β) blockade to mitigate the metabolic changes associated with cachexia. We found that TGF-β inhibition using the anti-TGF-β antibody 1D11.16.8 significantly improved overall mortality, weight loss, fat mass, lean body mass, bone mineral density, and skeletal muscle proteolysis in mice harboring advanced pancreatic cancer. Other immunotherapeutic strategies we employed were not effective. Collectively, we validated a simplified but useful model of pancreatic cancer cachexia to investigate immunologic treatment strategies. In addition, we showed that TGF-β inhibition can decrease the metabolic changes associated with cancer cachexia and improve overall survival.
Collapse
Affiliation(s)
- Stephanie H. Greco
- Department of Surgery, New York University School of Medicine, New York, New York, United States of America
| | - Lena Tomkötter
- Department of Surgery, New York University School of Medicine, New York, New York, United States of America
| | - Anne-Kristin Vahle
- Department of Surgery, New York University School of Medicine, New York, New York, United States of America
| | - Rae Rokosh
- Department of Surgery, New York University School of Medicine, New York, New York, United States of America
| | - Antonina Avanzi
- Department of Surgery, New York University School of Medicine, New York, New York, United States of America
| | - Syed Kashif Mahmood
- Department of Surgery, New York University School of Medicine, New York, New York, United States of America
| | - Michael Deutsch
- Department of Surgery, New York University School of Medicine, New York, New York, United States of America
| | - Sara Alothman
- Department of Surgery, New York University School of Medicine, New York, New York, United States of America
| | - Dalia Alqunaibit
- Department of Surgery, New York University School of Medicine, New York, New York, United States of America
| | - Atsuo Ochi
- Department of Surgery, New York University School of Medicine, New York, New York, United States of America
| | - Constantinos Zambirinis
- Department of Surgery, New York University School of Medicine, New York, New York, United States of America
| | - Tasnima Mohaimin
- Department of Surgery, New York University School of Medicine, New York, New York, United States of America
| | - Mauricio Rendon
- Department of Surgery, New York University School of Medicine, New York, New York, United States of America
| | - Elliot Levie
- Department of Surgery, New York University School of Medicine, New York, New York, United States of America
| | - Mridul Pansari
- Department of Surgery, New York University School of Medicine, New York, New York, United States of America
| | - Alejandro Torres-Hernandez
- Department of Surgery, New York University School of Medicine, New York, New York, United States of America
| | - Donnele Daley
- Department of Surgery, New York University School of Medicine, New York, New York, United States of America
| | - Rocky Barilla
- Department of Surgery, New York University School of Medicine, New York, New York, United States of America
| | - H. Leon Pachter
- Department of Surgery, New York University School of Medicine, New York, New York, United States of America
| | - Daniel Tippens
- Department of Surgery, New York University School of Medicine, New York, New York, United States of America
| | - Hassan Malik
- Department of Surgery, New York University School of Medicine, New York, New York, United States of America
| | - Allal Boutajangout
- Department of Neurology, New York University School of Medicine, New York, New York, United States of America
| | - Thomas Wisniewski
- Department of Neurology, New York University School of Medicine, New York, New York, United States of America
| | - George Miller
- Department of Surgery, New York University School of Medicine, New York, New York, United States of America
- Department of Cell Biology, New York University School of Medicine, New York, New York, United States of America
| |
Collapse
|
39
|
Krah NM, De La O JP, Swift GH, Hoang CQ, Willet SG, Chen Pan F, Cash GM, Bronner MP, Wright CV, MacDonald RJ, Murtaugh LC. The acinar differentiation determinant PTF1A inhibits initiation of pancreatic ductal adenocarcinoma. eLife 2015; 4. [PMID: 26151762 PMCID: PMC4536747 DOI: 10.7554/elife.07125] [Citation(s) in RCA: 120] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2015] [Accepted: 07/07/2015] [Indexed: 12/12/2022] Open
Abstract
Understanding the initiation and progression of pancreatic ductal adenocarcinoma (PDAC) may provide therapeutic strategies for this deadly disease. Recently, we and others made the surprising finding that PDAC and its preinvasive precursors, pancreatic intraepithelial neoplasia (PanIN), arise via reprogramming of mature acinar cells. We therefore hypothesized that the master regulator of acinar differentiation, PTF1A, could play a central role in suppressing PDAC initiation. In this study, we demonstrate that PTF1A expression is lost in both mouse and human PanINs, and that this downregulation is functionally imperative in mice for acinar reprogramming by oncogenic KRAS. Loss of Ptf1a alone is sufficient to induce acinar-to-ductal metaplasia, potentiate inflammation, and induce a KRAS-permissive, PDAC-like gene expression profile. As a result, Ptf1a-deficient acinar cells are dramatically sensitized to KRAS transformation, and reduced Ptf1a greatly accelerates development of invasive PDAC. Together, these data indicate that cell differentiation regulators constitute a new tumor suppressive mechanism in the pancreas.
Collapse
Affiliation(s)
- Nathan M Krah
- Department of Human Genetics, University of Utah, Salt Lake City, United States
| | - Jean-Paul De La O
- Department of Human Genetics, University of Utah, Salt Lake City, United States
| | - Galvin H Swift
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, United States
| | - Chinh Q Hoang
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, United States
| | - Spencer G Willet
- Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, United States
| | - Fong Chen Pan
- Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, United States
| | - Gabriela M Cash
- Department of Human Genetics, University of Utah, Salt Lake City, United States
| | - Mary P Bronner
- Department of Pathology, Huntsman Cancer Hospital, University of Utah, Salt Lake City, United States
| | - Christopher Ve Wright
- Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, United States
| | - Raymond J MacDonald
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, United States
| | - L Charles Murtaugh
- Department of Human Genetics, University of Utah, Salt Lake City, United States
| |
Collapse
|
40
|
Hoque R, Mehal WZ. Inflammasomes in pancreatic physiology and disease. Am J Physiol Gastrointest Liver Physiol 2015; 308:G643-51. [PMID: 25700081 PMCID: PMC4398840 DOI: 10.1152/ajpgi.00388.2014] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 02/10/2015] [Indexed: 01/31/2023]
Abstract
In this review we summarize the role of inflammasomes in pancreatic physiology and disease with a focus on acute pancreatitis where much recent progress has been made. New findings have identified inducers of and cell specificity of inflammasome component expression in the pancreas, the contribution of inflammasome-regulated effectors to pancreatitis, and metabolic regulation of inflammasome activation, which are strong determinants of injury in pancreatitis. New areas of pancreatic biology will be highlighted in the context of our evolving understanding of gut microbiome- and injury-induced inflammasome priming, pyroptosis, and innate immune-mediated regulation of cell metabolism.
Collapse
Affiliation(s)
- Rafaz Hoque
- 1Section of Digestive Diseases, Yale University School of Medicine, New Haven, Connecticut; and
| | - Wajahat Z. Mehal
- 1Section of Digestive Diseases, Yale University School of Medicine, New Haven, Connecticut; and ,2Section of Digestive Diseases, Department of Veterans Affairs Connecticut Healthcare, West Haven, Connecticut
| |
Collapse
|
41
|
Kui B, Balla Z, Vasas B, Végh ET, Pallagi P, Kormányos ES, Venglovecz V, Iványi B, Takács T, Hegyi P, Rakonczay Z. New insights into the methodology of L-arginine-induced acute pancreatitis. PLoS One 2015; 10:e0117588. [PMID: 25688985 PMCID: PMC4331527 DOI: 10.1371/journal.pone.0117588] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 12/29/2014] [Indexed: 02/06/2023] Open
Abstract
Animal models are ideal to study the pathomechanism and therapy of acute pancreatitis (AP). The use of L-arginine-induced AP model is nowadays becoming increasingly popular in mice. However, carefully looking through the literature, marked differences in disease severity could be observed. In fact, while setting up the L-arginine (2×4 g/kg i.p.)-induced AP model in BALB/c mice, we found a relatively low rate (around 15%) of pancreatic necrosis, whereas others have detected much higher rates (up to 55%). We suspected that this may be due to differences between mouse strains. We administered various concentrations (5-30%, pH = 7.4) and doses (2×4, 3×3, or 4×2.5 g/kg) of L-arginine-HCl in BALB/c, FVB/n and C57BL/6 mice. The potential gender-specific effect of L-arginine was investigated in C57BL/6 mice. The fate of mice in response to the i.p. injections of L arginine followed one of three courses. Some mice (1) developed severe AP or (2) remained AP-free by 72 h, whereas others (3) had to be euthanized (to avoid their death, which was caused by the high dose of L-arginine and not AP) within 12 h., In FVB/n and C57BL/6 mice, the pancreatic necrosis rate (about 50%) was significantly higher than that observed in BALB/c mice using 2×4 g/kg 10% L-arginine, but euthanasia was necessary in a large proportion of animals, The i.p. injection of lower L-arginine concentrations (e.g. 5-8%) in case of the 2×4 g/kg dose, or other L-arginine doses (3×3 or 4×2.5 g/kg, 10%) were better for inducing AP. We could not detect any significant differences between the AP severity of male and female mice. Taken together, when setting up the L-arginine-induced AP model, there are several important factors that are worth consideration such as the dose and concentration of the administered L arginine-HCl solution and also the strain of mice.
Collapse
Affiliation(s)
- Balázs Kui
- First Department of Medicine, University of Szeged, Szeged, Hungary
| | - Zsolt Balla
- First Department of Medicine, University of Szeged, Szeged, Hungary
| | - Béla Vasas
- Department of Pathology, University of Szeged, Szeged, Hungary
| | - Eszter T. Végh
- First Department of Medicine, University of Szeged, Szeged, Hungary
| | - Petra Pallagi
- First Department of Medicine, University of Szeged, Szeged, Hungary
| | | | - Viktória Venglovecz
- Department of Pharmacology and Pharmacotherapy, University of Szeged, Szeged, Hungary
| | - Béla Iványi
- Department of Pathology, University of Szeged, Szeged, Hungary
| | - Tamás Takács
- First Department of Medicine, University of Szeged, Szeged, Hungary
| | - Péter Hegyi
- First Department of Medicine, University of Szeged, Szeged, Hungary
- Hungarian Academy of Sciences-University of Szeged, Translational Gastroenterology Research Group, Szeged, Hungary
| | - Zoltán Rakonczay
- First Department of Medicine, University of Szeged, Szeged, Hungary
| |
Collapse
|
42
|
Abstract
Alcohol and gallstones are the most common etiologic factors in acute pancreatitis (AP). Recurrent AP can lead to chronic pancreatitis (CP). Although the underlying pathophysiology of the disease is complex, immune cells are critical in the pathogenesis of pancreatitis and determining disease severity. In this review, we discuss the role of innate and adaptive immune cells in both AP and CP, potential immune-based therapeutic targets, and animal models used to understand our knowledge of the disease. The relative difficulty of obtaining human pancreatic tissue during pancreatitis makes animal models necessary. Animal models of pancreatitis have been generated to understand disease pathogenesis, test therapeutic interventions, and investigate immune responses. Although current animal models do not recapitulate all aspects of human disease, until better models can be developed available models are useful in addressing key research questions. Differences between experimental and clinical pancreatitis need consideration, and when therapies are tested, models with established disease ought to be included.
Collapse
|
43
|
Abstract
Pancreatitis is caused by inflammatory injury to the exocrine pancreas, from which both humans and animal models appear to recover via regeneration of digestive enzyme-producing acinar cells. This regenerative process involves transient phases of inflammation, metaplasia, and redifferentiation, driven by cell-cell interactions between acinar cells, leukocytes, and resident fibroblasts. The NFκB signaling pathway is a critical determinant of pancreatic inflammation and metaplasia, whereas a number of developmental signals and transcription factors are devoted to promoting acinar redifferentiation after injury. Imbalances between these proinflammatory and prodifferentiation pathways contribute to chronic pancreatitis, characterized by persistent inflammation, fibrosis, and acinar dedifferentiation. Loss of acinar cell differentiation also drives pancreatic cancer initiation, providing a mechanistic link between pancreatitis and cancer risk. Unraveling the molecular bases of exocrine regeneration may identify new therapeutic targets for treatment and prevention of both of these deadly diseases.
Collapse
Affiliation(s)
- L Charles Murtaugh
- Department of Human Genetics, University of Utah School of Medicine, Salt Lake City, Utah 84112;
| | | |
Collapse
|
44
|
Zheng YQ, Huang J, Zeng FC, Zhou XY. Application of caerulein and lipopolysaccharides in creating mouse models of mild or severe acute pancreatitis. Shijie Huaren Xiaohua Zazhi 2014; 22:4068-4074. [DOI: 10.11569/wcjd.v22.i27.4068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To establish typical mouse models of mild or severe acute pancreatitis induced with caerulein (CAE) and/or lipopolysaccharides (LPS).
METHODS: Fifty healthy adult male C57 mice were randomly divided into five groups (with 10 mice in each group): a control group (CON group), the caerulein 7 group (CAE 7 group), a caerulein 7 plus LPS group (CAE 7 + LPS group), a caerulein 13 group (CAE 13 group), and a caerulein 13 plus LPS group (CAE 13 + LPS group). All the animals were killed three hours after the last intraperitoneal injection. The pancreas was carefully removed for microscopic examination and further observed under a transmission electron microscope (TEM). Serum amylase and lipase concentrations were assayed.
RESULTS: Enzyme levels and pathological score in all the experimental groups were significantly higher than those in the CON group (amylase lowest CAE 7 group: 27020 U/dL ± 3443 U/dL vs CON group: 2696 U/dL ± 400 U/dL, P < 0.01; lipase content lowest CAE 7 group: 1379 U/L ± 283 U/L vs CON group: 33 U/L ± 13 U/L, P < 0.01; pathological score lowest CAE 7 group: 5.8 ± 0.9 vs CON group: 0.1 ± 0.3, P < 0.01). Compared with the CAE 7 group, the enzyme levels and pathological score in the CAE 13 + LPS group increased more significantly (CAE 13 + LPS group amylase: 46969 U/dL ± 11852 U/dL vs CAE 7 group amylase: 27020 U/dL ± 3443 U/dL, P < 0.01; CAE13 + LPS group lipase: 1962 U/dL ± 496 U/dL vs CAE 7 group lipase: 1379 U/dL ± 283 U/dL, P < 0.05; CAE13 + LPS group pathological score : 11.1 ± 1.1 vs CAE 7 group pathological score : 5.8 ± 0.9, P < 0.05). The grade of pathological changes in the CAE 13 + LPS group was significantly higher than that in the CAE 13 group (CAE 13 + LPS group: 11.1 ± 1.1 vs CAE 13 group: 10.1 ± 0.99, P < 0.05). The ultrastructure of acinar cells was damaged in the CAE 7 group, and the rough endoplasmic reticulum and mitochondria were markedly swollen. However, in the CAE 13 + LPS group, the acinar cells were seriously damaged.
CONCLUSION: Caerulein alone by intraperitoneal injection 7 times can be used to prepare a typical model of acute edematous pancreatitis, and caerulein by intraperitoneal injection 13 times plus LPS at the last time can be used to produce typical acute necrotizing pancreatitis.
Collapse
|
45
|
Inman KS, Francis AA, Murray NR. Complex role for the immune system in initiation and progression of pancreatic cancer. World J Gastroenterol 2014; 20:11160-11181. [PMID: 25170202 PMCID: PMC4145756 DOI: 10.3748/wjg.v20.i32.11160] [Citation(s) in RCA: 107] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Revised: 01/27/2014] [Accepted: 04/16/2014] [Indexed: 02/06/2023] Open
Abstract
The immune system plays a complex role in the development and progression of pancreatic cancer. Inflammation can promote the formation of premalignant lesions and accelerate pancreatic cancer development. Conversely, pancreatic cancer is characterized by an immunosuppressive environment, which is thought to promote tumor progression and invasion. Here we review the current literature describing the role of the immune response in the progressive development of pancreatic cancer, with a focus on the mechanisms that drive recruitment and activation of immune cells at the tumor site, and our current understanding of the function of the immune cell types at the tumor. Recent clinical and preclinical data are reviewed, detailing the involvement of the immune response in pancreatitis and pancreatic cancer, including the role of specific cytokines and implications for disease outcome. Acute pancreatitis is characterized by a predominantly innate immune response, while chronic pancreatitis elicits an immune response that involves both innate and adaptive immune cells, and often results in profound systemic immune-suppression. Pancreatic adenocarcinoma is characterized by marked immune dysfunction driven by immunosuppressive cell types, tumor-promoting immune cells, and defective or absent inflammatory cells. Recent studies reveal that immune cells interact with cancer stem cells and tumor stromal cells, and these interactions have an impact on development and progression of pancreatic ductal adenocarcinoma (PDAC). Finally, current PDAC therapies are reviewed and the potential for harnessing the actions of the immune response to assist in targeting pancreatic cancer using immunotherapy is discussed.
Collapse
|
46
|
Rao R, Graffeo CS, Gulati R, Jamal M, Narayan S, Zambirinis CP, Barilla R, Deutsch M, Greco SH, Ochi A, Tomkötter L, Blobstein R, Avanzi A, Tippens DM, Gelbstein Y, Van Heerden E, Miller G. Interleukin 17-producing γδT cells promote hepatic regeneration in mice. Gastroenterology 2014; 147:473-84.e2. [PMID: 24801349 PMCID: PMC4123443 DOI: 10.1053/j.gastro.2014.04.042] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Revised: 03/25/2014] [Accepted: 04/29/2014] [Indexed: 12/11/2022]
Abstract
BACKGROUND & AIMS Subsets of leukocytes synergize with regenerative growth factors to promote hepatic regeneration. γδT cells are early responders to inflammation-induced injury in a number of contexts. We investigated the role of γδT cells in hepatic regeneration using mice with disruptions in Tcrd (encodes the T-cell receptor δ chain) and Clec7a (encodes C-type lectin domain family 7 member a, also known as DECTIN1). METHODS We performed partial hepatectomies on wild-type C57BL/6, CD45.1, Tcrd(-/-), or Clec7a(-/-) mice. Cells were isolated from livers of patients and mice via mechanical and enzymatic digestion. γδT cells were purified by fluorescence-activated cell sorting. RESULTS In mice, partial hepatectomy up-regulated expression of CCL20 and ligands of Dectin-1, which was associated with recruitment and activation of γδT cells and their increased production of interleukin (IL)-17 family cytokines. Recruited γδT cells induced production of IL-6 by antigen-presenting cells and suppressed expression of interferon gamma by natural killer T cells, promoting hepatocyte proliferation. Absence of IL-17-producing γδT cells or deletion of Dectin-1 prevented development of regenerative phenotypes in subsets of innate immune cells. This slowed liver regeneration and was associated with reduced expression of regenerative growth factors and cell cycle regulators. Conversely, exogenous administration of IL-17 family cytokines or Dectin-1 ligands promoted regeneration. More broadly, we found that γδT cells are required for inflammatory responses mediated by IL-17 and Dectin-1. CONCLUSIONS γδT cells regulate hepatic regeneration by producing IL-22 and IL-17, which have direct mitogenic effects on hepatocytes and promote a regenerative phenotype in hepatic leukocytes, respectively. Dectin-1 ligation is required for γδT cells to promote hepatic regeneration.
Collapse
MESH Headings
- Animals
- Cell Proliferation
- Cells, Cultured
- Chemokine CCL20/metabolism
- Genotype
- Hepatectomy
- Hepatocytes/immunology
- Hepatocytes/metabolism
- Humans
- Inflammation Mediators/metabolism
- Interferon-gamma/metabolism
- Interleukin-17/metabolism
- Interleukin-6/metabolism
- Interleukins/metabolism
- Lectins, C-Type/deficiency
- Lectins, C-Type/genetics
- Lectins, C-Type/metabolism
- Liver/immunology
- Liver/metabolism
- Liver/surgery
- Liver Regeneration
- Lymphocyte Activation
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Phenotype
- Receptors, Antigen, T-Cell, gamma-delta/deficiency
- Receptors, Antigen, T-Cell, gamma-delta/genetics
- Receptors, Antigen, T-Cell, gamma-delta/metabolism
- Signal Transduction
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Time Factors
- Interleukin-22
Collapse
Affiliation(s)
- Raghavendra Rao
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, New York, New York
| | - Christopher S Graffeo
- S. Arthur Localio Laboratory, Department of Cell Biology, New York University School of Medicine, New York, New York
| | - Rishabh Gulati
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, New York, New York
| | - Mohsin Jamal
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, New York, New York
| | - Suchithra Narayan
- S. Arthur Localio Laboratory, Department of Cell Biology, New York University School of Medicine, New York, New York
| | - Constantinos P Zambirinis
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, New York, New York
| | - Rocky Barilla
- S. Arthur Localio Laboratory, Department of Cell Biology, New York University School of Medicine, New York, New York
| | - Michael Deutsch
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, New York, New York
| | - Stephanie H Greco
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, New York, New York
| | - Atsuo Ochi
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, New York, New York
| | - Lena Tomkötter
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, New York, New York
| | - Reuven Blobstein
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, New York, New York
| | - Antonina Avanzi
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, New York, New York
| | - Daniel M Tippens
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, New York, New York
| | - Yisroel Gelbstein
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, New York, New York
| | - Eliza Van Heerden
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, New York, New York
| | - George Miller
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, New York, New York; S. Arthur Localio Laboratory, Department of Cell Biology, New York University School of Medicine, New York, New York.
| |
Collapse
|
47
|
Cohen-Fredarow A, Tadmor A, Raz T, Meterani N, Addadi Y, Nevo N, Solomonov I, Sagi I, Mor G, Neeman M, Dekel N. Ovarian dendritic cells act as a double-edged pro-ovulatory and anti-inflammatory sword. Mol Endocrinol 2014; 28:1039-54. [PMID: 24825398 DOI: 10.1210/me.2013-1400] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Ovulation and inflammation share common attributes, including immune cell invasion into the ovary. The present study aims at deciphering the role of dendritic cells (DCs) in ovulation and corpus luteum formation. Using a CD11c-EYFP transgenic mouse model, ovarian transplantation experiments, and fluorescence-activated cell sorting analyses, we demonstrate that CD11c-positive, F4/80-negative cells, representing DCs, are recruited to the ovary under gonadotropin regulation. By conditional ablation of these cells in CD11c-DTR transgenic mice, we revealed that they are essential for expansion of the cumulus-oocyte complex, release of the ovum from the ovarian follicle, formation of a functional corpus luteum, and enhanced lymphangiogenesis. These experiments were complemented by allogeneic DC transplantation after conditional ablation of CD11c-positive cells that rescued ovulation. The pro-ovulatory effects of these cells were mediated by up-regulation of ovulation-essential genes. Interestingly, we detected a remarkable anti-inflammatory capacity of ovarian DCs, which seemingly serves to restrict the ovulatory-associated inflammation. In addition to discovering the role of DCs in ovulation, this study implies the extended capabilities of these cells, beyond their classic immunologic role, which is relevant also to other biological systems.
Collapse
Affiliation(s)
- Adva Cohen-Fredarow
- Department of Biological Regulation (A.C.-F., A.T., N.M., Y.A., N.N., I.So., I.Sa., M.N., N.D.), Weizmann Institute of Science, Rehovot 76100, Israel; Koret School of Veterinary Medicine (T.R.), The Hebrew University of Jerusalem, Rehovot 76100, Israel; B-nano Ltd (Y.A.), Rehovot 76326, Israel; and Department of Obstetrics Gynecology and Reproductive Science (G.M.), Reproductive Immunology Unit, Yale University School of Medicine, New Haven, Connecticut 06510
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Court H, Amoyel M, Hackman M, Lee KE, Xu R, Miller G, Bar-Sagi D, Bach EA, Bergö MO, Philips MR. Isoprenylcysteine carboxylmethyltransferase deficiency exacerbates KRAS-driven pancreatic neoplasia via Notch suppression. J Clin Invest 2014; 123:4681-94. [PMID: 24216479 DOI: 10.1172/jci65764] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Accepted: 08/08/2013] [Indexed: 12/20/2022] Open
Abstract
RAS is the most frequently mutated oncogene in human cancers. Despite decades of effort, anti-RAS therapies have remained elusive. Isoprenylcysteine carboxylmethyltransferase (ICMT) methylates RAS and other CaaX-containing proteins, but its potential as a target for cancer therapy has not been fully evaluated. We crossed a Pdx1-Cre;LSL-KrasG12D mouse, which is a model of pancreatic ductal adenocarcinoma (PDA), with a mouse harboring a floxed allele of Icmt. Surprisingly, we found that ICMT deficiency dramatically accelerated the development and progression of neoplasia. ICMT-deficient pancreatic ductal epithelial cells had a slight growth advantage and were resistant to premature senescence by a mechanism that involved suppression of cyclin-dependent kinase inhibitor 2A (p16INK4A) expression. ICMT deficiency precisely phenocopied Notch1 deficiency in the Pdx1-Cre;LSL-KrasG12D model by exacerbating pancreatic intraepithelial neoplasias, promoting facial papillomas, and derepressing Wnt signaling. Silencing ICMT in human osteosarcoma cells decreased Notch1 signaling in response to stimulation with cell-surface ligands. Additionally, targeted silencing of Ste14, the Drosophila homolog of Icmt, resulted in defects in wing development, consistent with Notch loss of function. Our data suggest that ICMT behaves like a tumor suppressor in PDA because it is required for Notch1 signaling.
Collapse
MESH Headings
- Animals
- Animals, Genetically Modified
- Carcinoma in Situ/genetics
- Carcinoma in Situ/metabolism
- Carcinoma in Situ/pathology
- Carcinoma, Pancreatic Ductal/genetics
- Carcinoma, Pancreatic Ductal/metabolism
- Carcinoma, Pancreatic Ductal/pathology
- Cell Line, Tumor
- Disease Models, Animal
- Drosophila melanogaster/genetics
- Drosophila melanogaster/growth & development
- Drosophila melanogaster/metabolism
- Female
- Genes, ras
- Humans
- Male
- Metaplasia
- Mice
- Mice, 129 Strain
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Mutant Strains
- Mice, Transgenic
- Mutation
- Pancreas/metabolism
- Pancreas/pathology
- Pancreatic Neoplasms/genetics
- Pancreatic Neoplasms/metabolism
- Pancreatic Neoplasms/pathology
- Protein Methyltransferases/deficiency
- Protein Methyltransferases/genetics
- Receptor, Notch1/metabolism
- Signal Transduction
Collapse
|
49
|
Kui B, Balla Z, Végh ET, Pallagi P, Venglovecz V, Iványi B, Takács T, Hegyi P, Rakonczay Z. Recent advances in the investigation of pancreatic inflammation induced by large doses of basic amino acids in rodents. J Transl Med 2014; 94:138-149. [PMID: 24365745 DOI: 10.1038/labinvest.2013.143] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2013] [Revised: 11/13/2013] [Accepted: 11/19/2013] [Indexed: 12/16/2022] Open
Abstract
It has been known for approximately 30 years that large doses of the semi-essential basic amino acid L-arginine induce severe pancreatic inflammation in rats. Recently, it has been demonstrated that L-arginine can also induce pancreatitis in mice. Moreover, other basic amino acids like L-ornithine and L-lysine can cause exocrine pancreatic damage without affecting the endocrine parenchyma and the ducts in rats. The utilization of these noninvasive severe basic amino acid-induced pancreatitis models is becoming increasingly popular and appreciated as these models nicely reproduce most laboratory and morphological features of human pancreatitis. Consequently, the investigation of basic amino acid-induced pancreatitis may offer us a better understanding of the pathogenesis and possible treatment options of the human disease.
Collapse
Affiliation(s)
- Balázs Kui
- First Department of Medicine, University of Szeged, Szeged, Hungary
| | - Zsolt Balla
- First Department of Medicine, University of Szeged, Szeged, Hungary
| | - Eszter T Végh
- First Department of Medicine, University of Szeged, Szeged, Hungary
| | - Petra Pallagi
- First Department of Medicine, University of Szeged, Szeged, Hungary
| | - Viktória Venglovecz
- Department of Pharmacology and Pharmacotherapy, University of Szeged, Szeged, Hungary
| | - Béla Iványi
- Department of Pathology, University of Szeged, Szeged, Hungary
| | - Tamás Takács
- First Department of Medicine, University of Szeged, Szeged, Hungary
| | - Péter Hegyi
- First Department of Medicine, University of Szeged, Szeged, Hungary
| | - Zoltán Rakonczay
- First Department of Medicine, University of Szeged, Szeged, Hungary
| |
Collapse
|
50
|
Henning JR, Graffeo CS, Rehman A, Fallon NC, Zambirinis CP, Ochi A, Barilla R, Jamal M, Deutsch M, Greco S, Ego-Osuala M, Saeed UB, Rao RS, Badar S, Quesada JP, Acehan D, Miller G. Dendritic cells limit fibroinflammatory injury in nonalcoholic steatohepatitis in mice. Hepatology 2013; 58:589-602. [PMID: 23322710 PMCID: PMC3638069 DOI: 10.1002/hep.26267] [Citation(s) in RCA: 139] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2012] [Accepted: 12/21/2012] [Indexed: 12/12/2022]
Abstract
UNLABELLED Nonalcoholic steatohepatitis (NASH) is the most common etiology of chronic liver dysfunction in the United States and can progress to cirrhosis and liver failure. Inflammatory insult resulting from fatty infiltration of the liver is central to disease pathogenesis. Dendritic cells (DCs) are antigen-presenting cells with an emerging role in hepatic inflammation. We postulated that DCs are important in the progression of NASH. We found that intrahepatic DCs expand and mature in NASH liver and assume an activated immune phenotype. However, rather than mitigating the severity of NASH, DC depletion markedly exacerbated intrahepatic fibroinflammation. Our mechanistic studies support a regulatory role for DCs in NASH by limiting sterile inflammation through their role in the clearance of apoptotic cells and necrotic debris. We found that DCs limit CD8(+) T-cell expansion and restrict Toll-like receptor expression and cytokine production in innate immune effector cells in NASH, including Kupffer cells, neutrophils, and inflammatory monocytes. Consistent with their regulatory role in NASH, during the recovery phase of disease, ablation of DC populations results in delayed resolution of intrahepatic inflammation and fibroplasia. CONCLUSION Our findings support a role for DCs in modulating NASH. Targeting DC functional properties may hold promise for therapeutic intervention in NASH.
Collapse
Affiliation(s)
- Justin R. Henning
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, 550 First Avenue, New York, NY 10016
| | - Christopher S. Graffeo
- S. Arthur Localio Laboratory, Department of Cell Biology, New York University School of Medicine, 550 First Avenue, New York, NY 10016
| | - Adeel Rehman
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, 550 First Avenue, New York, NY 10016
| | - Nina C. Fallon
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, 550 First Avenue, New York, NY 10016
| | - Constantinos P. Zambirinis
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, 550 First Avenue, New York, NY 10016
| | - Atsuo Ochi
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, 550 First Avenue, New York, NY 10016
| | - Rocky Barilla
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, 550 First Avenue, New York, NY 10016
| | - Mohsin Jamal
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, 550 First Avenue, New York, NY 10016
| | - Michael Deutsch
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, 550 First Avenue, New York, NY 10016
| | - Stephanie Greco
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, 550 First Avenue, New York, NY 10016
| | - Melvin Ego-Osuala
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, 550 First Avenue, New York, NY 10016
| | - Usama Bin Saeed
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, 550 First Avenue, New York, NY 10016
| | - Raghavendra S. Rao
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, 550 First Avenue, New York, NY 10016
| | - Sana Badar
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, 550 First Avenue, New York, NY 10016
| | - Juan P. Quesada
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, 550 First Avenue, New York, NY 10016
| | - Devrim Acehan
- S. Arthur Localio Laboratory, Department of Cell Biology, New York University School of Medicine, 550 First Avenue, New York, NY 10016
| | - George Miller
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, 550 First Avenue, New York, NY 10016,S. Arthur Localio Laboratory, Department of Cell Biology, New York University School of Medicine, 550 First Avenue, New York, NY 10016
| |
Collapse
|