1
|
Pollin G, Mathison AJ, de Assuncao TM, Thomas A, Zeighami A, Salmonson A, Liu H, Urrutia G, Vankayala P, Pandol SJ, Hong JC, Zimmermann MT, Iovanna J, Jin VX, Urrutia R, Lomberk G. Ehmt2 inactivation in pancreatic epithelial cells shapes the transcriptional landscape and inflammation response of the whole pancreas. Front Genet 2024; 15:1412767. [PMID: 38948355 PMCID: PMC11211573 DOI: 10.3389/fgene.2024.1412767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 05/17/2024] [Indexed: 07/02/2024] Open
Abstract
Introduction: The Euchromatic Histone Methyl Transferase Protein 2 (EHMT2), also known as G9a, deposits transcriptionally repressive chromatin marks that play pivotal roles in the maturation and homeostasis of multiple organs. Recently, we have shown that Ehmt2 inactivation in the mouse pancreas alters growth and immune gene expression networks, antagonizing Kras-mediated pancreatic cancer initiation and promotion. Here, we elucidate the essential role of Ehmt2 in maintaining a transcriptional landscape that protects organs from inflammation. Methods: Comparative RNA-seq studies between normal postnatal and young adult pancreatic tissue from Ehmt2 conditional knockout animals (Ehmt2 fl/fl ) targeted to the exocrine pancreatic epithelial cells (Pdx1-Cre and P48 Cre/+ ), reveal alterations in gene expression networks in the whole organ related to injury-inflammation-repair, suggesting an increased predisposition to damage. Thus, we induced an inflammation repair response in the Ehmt2 fl/fl pancreas and used a data science-based approach to integrate RNA-seq-derived pathways and networks, deconvolution digital cytology, and spatial transcriptomics. We also analyzed the tissue response to damage at the morphological, biochemical, and molecular pathology levels. Results and discussion: The Ehmt2 fl/fl pancreas displays an enhanced injury-inflammation-repair response, offering insights into fundamental molecular and cellular mechanisms involved in this process. More importantly, these data show that conditional Ehmt2 inactivation in exocrine cells reprograms the local environment to recruit mesenchymal and immunological cells needed to mount an increased inflammatory response. Mechanistically, this response is an enhanced injury-inflammation-repair reaction with a small contribution of specific Ehmt2-regulated transcripts. Thus, this new knowledge extends the mechanisms underlying the role of the Ehmt2-mediated pathway in suppressing pancreatic cancer initiation and modulating inflammatory pancreatic diseases.
Collapse
Affiliation(s)
- Gareth Pollin
- Linda T. and John A. Mellowes Center for Genomic Sciences and Precision Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
- Division of Research, Department of Surgery, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Angela J. Mathison
- Linda T. and John A. Mellowes Center for Genomic Sciences and Precision Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
- Division of Research, Department of Surgery, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Thiago M. de Assuncao
- Linda T. and John A. Mellowes Center for Genomic Sciences and Precision Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
- Division of Research, Department of Surgery, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Anju Thomas
- Linda T. and John A. Mellowes Center for Genomic Sciences and Precision Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
- Division of Research, Department of Surgery, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Atefeh Zeighami
- Linda T. and John A. Mellowes Center for Genomic Sciences and Precision Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Ann Salmonson
- Division of Research, Department of Surgery, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Hongfei Liu
- Linda T. and John A. Mellowes Center for Genomic Sciences and Precision Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Guillermo Urrutia
- Division of Research, Department of Surgery, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Pallavi Vankayala
- Linda T. and John A. Mellowes Center for Genomic Sciences and Precision Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Stephen J. Pandol
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Johnny C. Hong
- Division of Transplantation, Department of Surgery, College of Medicine, Pennsylvania State University, Hershey, PA, United States
| | - Michael T. Zimmermann
- Linda T. and John A. Mellowes Center for Genomic Sciences and Precision Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, United States
- Clinical and Translational Sciences Institute, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Juan Iovanna
- Centre de Recherche en Cancérologie de Marseille (CRCM), Institut National de la Santé et de la Recherche médicale (INSERM) U1068, CNRS UMR 7258, Parc Scientifique et Technologique de Luminy, Aix-Marseille Université and Institut Paoli-Calmettes, Marseille, France
| | - Victor X. Jin
- Linda T. and John A. Mellowes Center for Genomic Sciences and Precision Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
- Division of Biostatistics, Institute for Health and Equity, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Raul Urrutia
- Linda T. and John A. Mellowes Center for Genomic Sciences and Precision Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
- Division of Research, Department of Surgery, Medical College of Wisconsin, Milwaukee, WI, United States
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Gwen Lomberk
- Linda T. and John A. Mellowes Center for Genomic Sciences and Precision Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
- Division of Research, Department of Surgery, Medical College of Wisconsin, Milwaukee, WI, United States
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
2
|
Oh E, McCown EM, Ahn M, Garcia PA, Branciamore S, Tang S, Zeng DF, Roep BO, Thurmond DC. Syntaxin 4 Enrichment in β-Cells Prevents Conversion to Autoimmune Diabetes in Non-Obese Diabetic (NOD) Mice. Diabetes 2021; 70:2837-2849. [PMID: 34556496 PMCID: PMC8660989 DOI: 10.2337/db21-0170] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 09/07/2021] [Indexed: 11/13/2022]
Abstract
Syntaxin 4 (STX4), a plasma membrane-localized SNARE protein, regulates human islet β-cell insulin secretion and preservation of β-cell mass. We found that human type 1 diabetes (T1D) and NOD mouse islets show reduced β-cell STX4 expression, consistent with decreased STX4 expression, as a potential driver of T1D phenotypes. To test this hypothesis, we generated inducible β-cell-specific STX4-expressing NOD mice (NOD-iβSTX4). Of NOD-iβSTX4 mice, 73% had sustained normoglycemia vs. <20% of control NOD (NOD-Ctrl) mice by 25 weeks of age. At 12 weeks of age, before diabetes conversion, NOD-iβSTX4 mice demonstrated superior whole-body glucose tolerance and β-cell glucose responsiveness than NOD-Ctrl mice. Higher β-cell mass and reduced β-cell apoptosis were also detected in NOD-iβSTX4 pancreata compared with pancreata of NOD-Ctrl mice. Single-cell RNA sequencing revealed that islets from NOD-iβSTX4 had markedly reduced interferon-γ signaling and tumor necrosis factor-α signaling via nuclear factor-κB in islet β-cells, including reduced expression of the chemokine CCL5; CD4+ regulatory T cells were also enriched in NOD-iβSTX4 islets. These results provide a deeper mechanistic understanding of STX4 function in β-cell protection and warrant further investigation of STX4 enrichment as a strategy to reverse or prevent T1D in humans or protect β-cell grafts.
Collapse
Affiliation(s)
- Eunjin Oh
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Beckman Research Institute, Duarte, CA
| | - Erika M McCown
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Beckman Research Institute, Duarte, CA
| | - Miwon Ahn
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Beckman Research Institute, Duarte, CA
| | - Pablo A Garcia
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Beckman Research Institute, Duarte, CA
| | - Sergio Branciamore
- Department of Diabetes Complications and Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Beckman Research Institute, Duarte, CA
| | - Shanshan Tang
- Department of Immunology and Theranostics, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Beckman Research Institute, Duarte, CA
| | - De-Fu Zeng
- Department of Immunology and Theranostics, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Beckman Research Institute, Duarte, CA
| | - Bart O Roep
- Department of Diabetes Immunology, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Beckman Research Institute, Duarte, CA
| | - Debbie C Thurmond
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Beckman Research Institute, Duarte, CA
| |
Collapse
|
3
|
Lukes J, Wolthers BO, Altaf Raja R, Uhrinova K, Skvarova Kramarzova K, Hermanova I, Simcikova M, Kicko P, Zaliova M, Sramkova L, Stary J, Trka J, Schmiegelow K, Starkova J. Pancreatitis-associated protein as an early marker of asparaginase-associated pancreatitis. Leuk Lymphoma 2021; 62:3506-3510. [PMID: 34369235 DOI: 10.1080/10428194.2021.1961236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Julius Lukes
- CLIP - Childhood Leukaemia Investigation Prague, Prague, Czech Republic.,Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Benjamin O Wolthers
- Department of Pediatrics and Adolescent Medicine, University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Raheel Altaf Raja
- Department of Pediatrics and Adolescent Medicine, University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Karolina Uhrinova
- Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Karolina Skvarova Kramarzova
- CLIP - Childhood Leukaemia Investigation Prague, Prague, Czech Republic.,Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Ivana Hermanova
- CLIP - Childhood Leukaemia Investigation Prague, Prague, Czech Republic.,Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Marketa Simcikova
- Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Peter Kicko
- Department of Dermatovenereology, Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Marketa Zaliova
- CLIP - Childhood Leukaemia Investigation Prague, Prague, Czech Republic.,Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University, Prague, Czech Republic.,University Hospital Motol, Prague, Czech Republic
| | - Lucie Sramkova
- CLIP - Childhood Leukaemia Investigation Prague, Prague, Czech Republic.,Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University, Prague, Czech Republic.,University Hospital Motol, Prague, Czech Republic
| | - Jan Stary
- Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University, Prague, Czech Republic.,University Hospital Motol, Prague, Czech Republic
| | - Jan Trka
- CLIP - Childhood Leukaemia Investigation Prague, Prague, Czech Republic.,Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University, Prague, Czech Republic.,University Hospital Motol, Prague, Czech Republic
| | - Kjeld Schmiegelow
- Department of Pediatrics and Adolescent Medicine, University Hospital Rigshospitalet, Copenhagen, Denmark.,Institute of Clinical Medicine, Faculty of Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Julia Starkova
- CLIP - Childhood Leukaemia Investigation Prague, Prague, Czech Republic.,Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University, Prague, Czech Republic.,University Hospital Motol, Prague, Czech Republic
| |
Collapse
|
4
|
The Potential Role of REG Family Proteins in Inflammatory and Inflammation-Associated Diseases of the Gastrointestinal Tract. Int J Mol Sci 2021; 22:ijms22137196. [PMID: 34281249 PMCID: PMC8268738 DOI: 10.3390/ijms22137196] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 06/22/2021] [Accepted: 06/30/2021] [Indexed: 12/12/2022] Open
Abstract
Regenerating gene (REG) family proteins serve as multifunctional secretory molecules with trophic, antiapoptotic, anti-inflammatory, antimicrobial and probably immuno-regulatory effects. Since their discovery, accumulating evidence has clarified the potential roles of the REG family in the occurrence, progression and development of a wide range of inflammatory and inflammation-associated diseases of the gastrointestinal (GI) tract. However, significant gaps still exist due to the undefined nature of certain receptors, regulatory signaling pathways and possible interactions among distinct Reg members. In this narrative review, we first describe the structural features, distribution pattern and purported regulatory mechanisms of REG family proteins. Furthermore, we summarize the established and proposed roles of REG proteins in the pathogenesis of various inflammation-associated pathologies of the GI tract and the body as a whole, focusing particularly on carcinogenesis in the ulcerative colitis—colitic cancer sequence and gastric cancer. Finally, the clinical relevance of REG products in the context of diagnosis, treatment and prognostication are also discussed in detail. The current evidence suggests a need to better understanding the versatile roles of Reg family proteins in the pathogenesis of inflammatory-associated diseases, and their broadened future usage as therapeutic targets and prognostic biomarkers is anticipated.
Collapse
|
5
|
Park C, Oh J, Lee WM, Koh HR, Sohn UD, Ham SW, Oh K. Inhibition of NUPR1-Karyopherin β1 Binding Increases Anticancer Drug Sensitivity. Int J Mol Sci 2021; 22:ijms22062794. [PMID: 33801927 PMCID: PMC8000408 DOI: 10.3390/ijms22062794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 02/23/2021] [Accepted: 03/02/2021] [Indexed: 11/21/2022] Open
Abstract
Background: Nuclear protein-1 (NUPR1, also known as p8/Com-1) is a transcription factor involved in the regulation of cellular stress responses, including serum starvation and drug stimulation. Methods: We investigated the mechanism of NUPR1 nuclear translocation involving karyopherin β1 (KPNB1), using a single-molecule binding assay and confocal microscopy. The cellular effects associated with NUPR1–KPNB1 inhibition were investigated by gene expression profiling and cell cycle analysis. Results: The single-molecule binding assay revealed that KPNB1 bound to NUPR1 with a binding affinity of 0.75 nM and that this binding was blocked by the aminothiazole ATZ-502. Following doxorubicin-only treatment, NUPR1 was translocated to the nucleus in more than 90% and NUPR1 translocation was blocked by the ATZ-502 combination treatment in MDA-MB-231 with no change in NUPR1 expression, providing strong evidence that NUPR1 nuclear translocation was directly inhibited by the ATZ-502 treatment. Inhibition of KPNB1 and NUPR1 binding was associated with a synergistic anticancer effect (up to 19.6-fold) in various cancer cell lines. NUPR1-related genes were also downregulated following the doxorubicin–ATZ-502 combination treatment. Conclusion: Our current findings clearly demonstrate that NUPR1 translocation into the nucleus requires karyopherin β1 binding. Inhibition of the KPNB1 and NUPR1 interaction may constitute a new cancer therapeutic approach that can increase the drug efficacy while reducing the side effects.
Collapse
Affiliation(s)
- Chanhee Park
- Center for Metareceptome Research, College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak, Seoul 06974, Korea; (C.P.); (W.M.L.); (U.D.S.)
- Institute of Gastroenterology, College of Medicine, Yonsei University, 50-1 Yonsei-ro, Seodaemun, Seoul 03772, Korea
| | - Jiwon Oh
- Department of Chemistry, Chung-Ang University, 84 Heukseok-ro, Dongjak, Seoul 06974, Korea; (J.O.); (H.R.K.)
| | - Won Mo Lee
- Center for Metareceptome Research, College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak, Seoul 06974, Korea; (C.P.); (W.M.L.); (U.D.S.)
| | - Hye Ran Koh
- Department of Chemistry, Chung-Ang University, 84 Heukseok-ro, Dongjak, Seoul 06974, Korea; (J.O.); (H.R.K.)
| | - Uy Dong Sohn
- Center for Metareceptome Research, College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak, Seoul 06974, Korea; (C.P.); (W.M.L.); (U.D.S.)
| | - Seung Wook Ham
- Department of Chemistry, Chung-Ang University, 84 Heukseok-ro, Dongjak, Seoul 06974, Korea; (J.O.); (H.R.K.)
- Correspondence: (S.W.H.); (K.O.)
| | - Kyungsoo Oh
- Center for Metareceptome Research, College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak, Seoul 06974, Korea; (C.P.); (W.M.L.); (U.D.S.)
- Correspondence: (S.W.H.); (K.O.)
| |
Collapse
|
6
|
Teresa Borrello M, Rita Emma M, Listi A, Rubis M, Coslet S, Augello G, Cusimano A, Cabibi D, Porcasi R, Giannitrapani L, Soresi M, Pantuso G, Blyth K, Montalto G, Pin C, Cervello M, Iovanna J. NUPR1 protects liver from lipotoxic injury by improving the endoplasmic reticulum stress response. FASEB J 2021; 35:e21395. [PMID: 33566371 DOI: 10.1096/fj.202002413rr] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 01/08/2021] [Accepted: 01/08/2021] [Indexed: 12/12/2022]
Abstract
Non-alcoholic fatty liver (NAFL) and related syndromes affect one-third of the adult population in industrialized and developing countries. Lifestyle and caloric oversupply are the main causes of such array of disorders, but the molecular mechanisms underlying their etiology remain elusive. Nuclear Protein 1 (NUPR1) expression increases upon cell injury in all organs including liver. Recently, we reported NUPR1 actively participates in the activation of the Unfolded Protein Response (UPR). The UPR typically maintains protein homeostasis, but downstream mediators of the pathway regulate metabolic functions including lipid metabolism. As increases in UPR and NUPR1 in obesity and liver disease have been well documented, the goal of this study was to investigate the roles of NUPR1 in this context. To establish whether NUPR1 is involved in these liver conditions we used patient-derived liver biopsies and in vitro and in vivo NUPR1 loss of functions models. First, we analyzed NUPR1 expression in a cohort of morbidly obese patients (MOPs), with simple fatty liver (NAFL) or more severe steatohepatitis (NASH). Next, we explored the metabolic roles of NUPR1 in wild-type (Nupr1+/+ ) or Nupr1 knockout mice (Nupr1-/- ) fed with a high-fat diet (HFD) for 15 weeks. Immunohistochemical and mRNA analysis revealed NUPR1 expression is inversely correlated to hepatic steatosis progression. Mechanistically, we found NUPR1 participates in the activation of PPAR-α signaling via UPR. As PPAR-α signaling is controlled by UPR, collectively, these findings suggest a novel function for NUPR1 in protecting liver from metabolic distress by controlling lipid homeostasis, possibly through the UPR.
Collapse
Affiliation(s)
- Maria Teresa Borrello
- Centre de Recherche en Cancérologie de Marseille, INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Marseille, France
| | - Maria Rita Emma
- Istituto per la Ricerca e l'Innovazione Biomedicale (IRIB), Consiglio Nazionale Delle Ricerche, Palermo, Italy
| | - Angela Listi
- Centre de Recherche en Cancérologie de Marseille, INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Marseille, France
| | - Marion Rubis
- Centre de Recherche en Cancérologie de Marseille, INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Marseille, France
| | - Sergiu Coslet
- MI-mAbs, Aix-Marseille University, Parc Scientifique et Technologique de Luminy, Marseille, France
| | - Giuseppa Augello
- Istituto per la Ricerca e l'Innovazione Biomedicale (IRIB), Consiglio Nazionale Delle Ricerche, Palermo, Italy
| | - Antonella Cusimano
- Istituto per la Ricerca e l'Innovazione Biomedicale (IRIB), Consiglio Nazionale Delle Ricerche, Palermo, Italy
| | - Daniela Cabibi
- Dipartimento di Promozione della Salute, Materno-Infantile, Medicina Interna e Specialistica di Eccellenza (PROMISE), University of Palermo, Palermo, Italy
| | - Rossana Porcasi
- Dipartimento di Promozione della Salute, Materno-Infantile, Medicina Interna e Specialistica di Eccellenza (PROMISE), University of Palermo, Palermo, Italy
| | - Lydia Giannitrapani
- Dipartimento di Promozione della Salute, Materno-Infantile, Medicina Interna e Specialistica di Eccellenza (PROMISE), University of Palermo, Palermo, Italy
| | - Maurizio Soresi
- Dipartimento di Promozione della Salute, Materno-Infantile, Medicina Interna e Specialistica di Eccellenza (PROMISE), University of Palermo, Palermo, Italy
| | - Gianni Pantuso
- Department of Surgical Oncological and Oral Sciences, Division of General and Oncological Surgery, University of Palermo, Palermo, Italy
| | - Karen Blyth
- Cancer Research UK Beatson Institute, Glasgow, UK
| | - Giuseppe Montalto
- Dipartimento di Promozione della Salute, Materno-Infantile, Medicina Interna e Specialistica di Eccellenza (PROMISE), University of Palermo, Palermo, Italy
| | - Christopher Pin
- Children's Health Research Institute, The University of Western Ontario, London, ON, Canada
| | - Melchiorre Cervello
- Istituto per la Ricerca e l'Innovazione Biomedicale (IRIB), Consiglio Nazionale Delle Ricerche, Palermo, Italy
| | - Juan Iovanna
- Centre de Recherche en Cancérologie de Marseille, INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Marseille, France
| |
Collapse
|
7
|
Cao Y, Tian Y, Liu Y, Su Z. Reg3β: A Potential Therapeutic Target for Tissue Injury and Inflammation-Associated Disorders. Int Rev Immunol 2021; 41:160-170. [PMID: 33426979 DOI: 10.1080/08830185.2020.1869731] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Since regenerating islet-derived 3β (Reg3β) was first reported, various studies have been conducted to explore the involvement of Reg3β in a gamut of maladies, such as diabetes, pancreatitis, pancreatic ductal adenocarcinoma, and extrapancreatic maladies such as inflammatory bowel disease, acute liver failure, and myocardial infarction. Surprisingly, there is currently no systematic review of Reg3β. Therefore, we summarize the structural characteristics, transcriptional regulation, putative receptors, and signaling pathways of Reg3β. The exact functional roles in various diseases, especially gastrointestinal and liver diseases, are also discussed. Reg3β plays multiple roles in promoting proliferation, inducing differentiation, preventing apoptosis, and resisting bacteria. The present review may provide new directions for the diagnosis and treatment of gastrointestinal, liver, and pancreatic diseases.
Collapse
Affiliation(s)
- Yuwen Cao
- International Genome Center, Jiangsu University, Zhenjiang, China.,Department of Immunology, Jiangsu University, Zhenjiang, China
| | - Yu Tian
- International Genome Center, Jiangsu University, Zhenjiang, China.,Department of Immunology, Jiangsu University, Zhenjiang, China
| | - Yueqin Liu
- Laboratory Center, the Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Zhaoliang Su
- International Genome Center, Jiangsu University, Zhenjiang, China.,Department of Immunology, Jiangsu University, Zhenjiang, China.,Laboratory Center, the Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, China
| |
Collapse
|
8
|
Päth G, Mehana AE, Pilz IH, Alt M, Baumann J, Sommerer I, Hoffmeister A, Seufert J. NUPR1 preserves insulin secretion of pancreatic β-cells during inflammatory stress by multiple low-dose streptozotocin and high-fat diet. Am J Physiol Endocrinol Metab 2020; 319:E338-E344. [PMID: 32574111 PMCID: PMC7473916 DOI: 10.1152/ajpendo.00088.2020] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Obesity is associated with dyslipidemia and subclinical inflammation that promotes metabolic disturbances including insulin resistance and pancreatic β-cell dysfunction. The nuclear protein, transcriptional regulator 1 (NUPR1) responds to cellular stresses and features tissue protective properties. To characterize the role of NUPR1 in endocrine pancreatic islets during inflammatory stress, we generated transgenic mice with β-cell-specific Nupr1 overexpression (βNUPR1). Under normal conditions, βNUPR1 mice did not differ from wild type (WT) littermates and display normal glucose homeostasis and β-cell mass. For induction of inflammatory conditions, mice were treated with multiple low-dose streptozotocin (mld-STZ) and/or fed a high-fat diet (HFD). All treatments significantly worsened glycaemia in WT mice, while βNUPR1 mice substantially preserved insulin secretion and glucose tolerance. HFD increased β-cell mass in all animals, with βNUPR1 mice tending to show higher values. The improved outcome of βNUPR1 mice was accompanied by decreased NF-κB activation and lymphocyte infiltration in response to mld-STZ. In vitro, isolated βNUPR1 islets preserved insulin secretion and content with insignificantly low apoptosis during culture stress and IL-1β exposure. These findings suggest that NUPR1 plays a vital role in the protection of β-cells from apoptosis, related degradation of insulin storages and subsequent secretion during inflammatory and obesity-related tissue stress.
Collapse
Affiliation(s)
- Günter Päth
- Division of Endocrinology and Diabetology, Department of Medicine II, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Amir E Mehana
- Division of Endocrinology and Diabetology, Department of Medicine II, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Faculty of Science, Department of Zoology, Suez Canal University, Ismailia, Egypt
| | - Ingo H Pilz
- Division of Endocrinology and Diabetology, Department of Medicine II, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Marcus Alt
- Division of Endocrinology and Diabetology, Department of Medicine II, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Johannes Baumann
- Division of Endocrinology and Diabetology, Department of Medicine II, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Ines Sommerer
- Division of Gastroenterology and Rheumatology, Neurology and Dermatology, Department of Medicine, University of Leipzig, Germany
| | - Albrecht Hoffmeister
- Division of Gastroenterology and Rheumatology, Neurology and Dermatology, Department of Medicine, University of Leipzig, Germany
| | - Jochen Seufert
- Division of Endocrinology and Diabetology, Department of Medicine II, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
9
|
Edwards JA, Tan N, Toussaint N, Ou P, Mueller C, Stanek A, Zinsou V, Roudnitsky S, Sagal M, Dresner L, Schwartzman A, Huan C. Role of regenerating islet-derived proteins in inflammatory bowel disease. World J Gastroenterol 2020; 26:2702-2714. [PMID: 32550748 PMCID: PMC7284176 DOI: 10.3748/wjg.v26.i21.2702] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 03/26/2020] [Accepted: 05/13/2020] [Indexed: 02/06/2023] Open
Abstract
Inflammatory bowel disease (IBD) is an inflammatory disorder of the gastrointestinal tract that affects millions of patients worldwide. It has a complex and multifactorial etiology leading to excessive exposure of intestinal epithelium to microbial antigens, inappropriate activation of the immune system and ultimately to the damage of intestinal tissues. Although numerous efforts have been made to improve the disease management, IBD remains persistently recurring and beyond cure. This is due largely to the gaps in our understanding of the pathogenesis of IBD that hamper the development of timely diagnoses and effective treatment. However, some recent discoveries, including the beneficial effects of interleukin-22 (IL-22) on the inflamed intestine, have shed light on a self-protective mechanism in IBD. Regenerating islet-derived (REG/Reg) proteins are small secretory proteins which function as IL-22's downstream effectors. Mounting studies have demonstrated that IBD patients have significantly increased REG expressions in the injured intestine, but with undefined mechanisms and roles. The reported functions of REG/Reg proteins in intestinal homeostasis, such as those of antibacterial, anti-inflammatory and tissue repair, lead us to discuss their potential mechanisms and clinical relevance in IBD in order to advance IBD research and management.
Collapse
Affiliation(s)
- Jodi-Ann Edwards
- Department of Surgery, State University of New York, Downstate Health Sciences University, Brooklyn, NY 11203, United States
| | - Nicholas Tan
- College of Medicine, State University of New York, Downstate Health Sciences University, Brooklyn, NY 11203, United States
| | - Nadlie Toussaint
- College of Medicine, State University of New York, Downstate Health Sciences University, Brooklyn, NY 11203, United States
| | - Peiqi Ou
- MCB program, School of Graduate Studies, State University of New York, Downstate Health Sciences University, Brooklyn, NY 11203, United States
| | - Cathy Mueller
- Department of Surgery, State University of New York, Downstate Health Sciences University, Brooklyn, NY 11203, United States
| | - Albert Stanek
- Department of Surgery, State University of New York, Downstate Health Sciences University, Brooklyn, NY 11203, United States
| | - Vladimir Zinsou
- College of Medicine, State University of New York, Downstate Health Sciences University, Brooklyn, NY 11203, United States
| | - Sean Roudnitsky
- Department of Surgery, State University of New York, Downstate Health Sciences University, Brooklyn, NY 11203, United States
| | - Michelle Sagal
- Department of Surgery, State University of New York, Downstate Health Sciences University, Brooklyn, NY 11203, United States
| | - Lisa Dresner
- Department of Surgery, State University of New York, Downstate Health Sciences University, Brooklyn, NY 11203, United States
| | - Alexander Schwartzman
- Department of Surgery, State University of New York, Downstate Health Sciences University, Brooklyn, NY 11203, United States
| | - Chongmin Huan
- Department of Surgery and Cell Biology, State University of New York, Downstate Health Sciences University, Brooklyn, NY 11203, United States
| |
Collapse
|
10
|
Hepatocarcinoma-intestine-pancreas/pancreatitis-associated protein (HIP/PAP) confers protection against hepatic fibrosis through downregulation of transforming growth factor β receptor II. J Transl Med 2020; 100:466-482. [PMID: 31641222 DOI: 10.1038/s41374-019-0314-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 07/23/2019] [Accepted: 07/30/2019] [Indexed: 01/18/2023] Open
Abstract
Hepatocarcinoma-intestine-pancreas/pancreatitis-associated protein (HIP/PAP) has antimicrobial, antioxidant, anti-inflammatory, mitogenic, and antiapoptotic effects and thus exerts important functions in the maintenance of integrity and homeostasis of several organs, such as the gastrointestinal tract, pancreas, and liver. Although the potent hepatoprotective effect of HIP/PAP has been validated, its impact on liver fibrosis has not been reported. In this study, we evaluated the role of HIP/PAP on hepatic fibrosis and explored the possible underlying mechanisms. We found that the expression of HIP/PAP and its mouse counterpart, Reg3B, was markedly upregulated in fibrotic human or mouse livers. Intraperitoneal (i.p.) interleukin (IL)-10, IL-6, and TNF-α but not TGF-β1 significantly induced hepatic overexpression of Reg3B in mice. In both CCl4 and BDL liver fibrosis models, adenovirus-mediated ectopic expression of HIP/PAP markedly alleviated liver injury, inflammation, collagen deposition, hepatic stellate cell activation, and the overexpression of profibrotic cytokines, including transforming growth factor β1 (TGF-β1), platelet-derived growth factor (PDGF)-A, B, connective tissue growth factor (CTGF), and plasminogen activator inhibitor-1 (PAI-1), in mice. In vitro experiments demonstrated that, in addition to suppressing hepatic stellate cell proliferation and accelerating hepatocyte proliferation, HIP/PAP mitigated TGF-β1-induced hepatic stellate cell activation, hepatocyte epithelial-mesenchymal transition (EMT) and upregulated expression of profibrotic cytokines in both hepatic stellate cells and hepatocytes. Moreover, HIP/PAP attenuated the overexpression of TGF-β receptor II (TGF-βRII) in fibrotic mouse livers and decreased the basal expression of TGF-βRII in nonfibrotic mouse livers as well as in cultured hepatocytes and hepatic stellate cells, which is at least partly attributable to the TGF-β1-antagonizing function of HIP/PAP. This study indicates that increased expression of hepatic HIP/PAP serves as a countermeasure against liver injury and fibrosis. Exogenous supplementation of HIP/PAP might be a promising therapeutic agent for hepatic fibrosis as well as liver injury.
Collapse
|
11
|
Sun X, Yan P, Zou C, Wong YK, Shu Y, Lee YM, Zhang C, Yang ND, Wang J, Zhang J. Targeting autophagy enhances the anticancer effect of artemisinin and its derivatives. Med Res Rev 2019; 39:2172-2193. [PMID: 30972803 DOI: 10.1002/med.21580] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 03/07/2019] [Accepted: 03/16/2019] [Indexed: 12/12/2022]
Abstract
Artemisinin and its derivatives, with their outstanding clinical efficacy and safety, represent the most effective and impactful antimalarial drugs. Apart from its antimalarial effect, artemisinin has also been shown to exhibit selective anticancer properties against multiple cancer types both in vitro and in vivo. Specifically, our previous studies highlighted the therapeutic effects of artemisinin on autophagy regulation. Autophagy is a well-conserved degradative process that recycles cytoplasmic contents and organelles in lysosomes to maintain cellular homeostasis. The deregulation of autophagy is often observed in cancer cells, where it contributes to tumor adaptation to nutrient-deficient tumor microenvironments. This review discusses recent advances in the anticancer properties of artemisinin and its derivatives via their regulation of autophagy, mitophagy, and ferritinophagy. In particular, we will discuss the mechanisms of artemisinin activation in cancer and novel findings regarding the role of artemisinin in regulating autophagy, which involves changes in multiple signaling pathways. More importantly, with increasing failure rates and the high cost of the development of novel anticancer drugs, the strategy of repurposing traditional therapeutic Chinese medicinal agents such as artemisinin to treat cancer provides a more attractive alternative. We believe that the topics covered here will be important in demonstrating the potential of artemisinin and its derivatives as safe and potent anticancer agents.
Collapse
Affiliation(s)
- Xin Sun
- Department of Oncology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Peiyi Yan
- Department of Clinical Laboratory, Shanghai Putuo District People's Hospital, Shanghai, China
| | - Chang Zou
- Clinical Medical Research Center, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University, Shenzhen Public Service Platform on Tumor Precision Medicine and Molecular Diagnosis, Shenzhen People's Hospital, Shenzhen, China
| | - Yin-Kwan Wong
- Department of Pharmacology, Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Yuhan Shu
- College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou, China
| | - Yew Mun Lee
- Department of Pharmacology, Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Chongjing Zhang
- Institute of Material Medical, Peking Union Medical College, Beijing, China
| | - Nai-Di Yang
- Department of Pharmacology, Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Jigang Wang
- Clinical Medical Research Center, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University, Shenzhen Public Service Platform on Tumor Precision Medicine and Molecular Diagnosis, Shenzhen People's Hospital, Shenzhen, China.,Department of Pharmacology, Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Artemisinin Research Center, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China.,Key Laboratory of Cardio-Cerebrovascular Disease Prevention & Therapy, Gannan Medical University, Ganzhou, China
| | - Jianbin Zhang
- Department of Oncology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China
| |
Collapse
|
12
|
Inactivation of NUPR1 promotes cell death by coupling ER-stress responses with necrosis. Sci Rep 2018; 8:16999. [PMID: 30451898 PMCID: PMC6242935 DOI: 10.1038/s41598-018-35020-3] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 10/30/2018] [Indexed: 12/16/2022] Open
Abstract
It was already described that genetic inhibition of NUPR1 induces tumor growth arrest. In this paper we studied the metabolism changes after NUPR1 downregulation in pancreatic cancer cells, which results in a significant decrease of OXPHOS activity with a concomitant lower ATP production which precedes the necrotic cell death. We demonstrated that NUPR1 downregulation induces a mitochondrial failure with a loss of the mitochondrial membrane potential, a strong increase in ROS production and a concomitant relocalization of mitochondria to the vicinity of the endoplasmic reticulum (ER). In addition, the transcriptomic analysis of NUPR1-deficient cells shows a decrease in the expression of some ER stress response-associated genes. Indeed, in ER stressors-treated cells with thapsigargin, brefeldin A or tunicamycin, a greater increase in necrosis and decrease of ATP content was observed in NUPR1-defficent cells. Finally, in vivo experiments, using acute pancreatitis which induces ER stress as well as NUPR1 activation, we observed that NUPR1 expression protects acinar cells from necrosis in mice. Importantly, we also report that the cell death observed after knocking-down NUPR1 expression is completely reversed by incubation with Necrostatin-1, but not by inhibiting caspase activity with Z-VAD-FMK. Altogether, these data enable us to describe a model in which inactivation of NUPR1 in pancreatic cancer cells results in an ER stress that induces a mitochondrial malfunction, a deficient ATP production and, as consequence, the cell death mediated by a programmed necrosis.
Collapse
|
13
|
Hollenbach M, Klöting N, Sommerer I, Lorenz J, Heindl M, Kern M, Mössner J, Blüher M, Hoffmeister A. p8 deficiency leads to elevated pancreatic beta cell mass but does not contribute to insulin resistance in mice fed with high-fat diet. PLoS One 2018; 13:e0201159. [PMID: 30040846 PMCID: PMC6057664 DOI: 10.1371/journal.pone.0201159] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 07/10/2018] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND p8 was initially described as being overexpressed in acute pancreatitis and encoding a ubiquitous stress protein. Analysis of insulin sensitivity and glucose tolerance in p8-knockout and haplodeficient mice revealed counterintuitive results. Thus, we determined glycemic control of p8 in mice fed with standard (SD) and high-fat diet (HFD). METHODS p8-/- and wild type (p8+/+) mice were used for analysis of glucagon (immunohistochemistry), insulin levels (ELISA) and beta cell mass. Hyperinsulinemic- euglycemic glucose clamp technique, i.p. glucose tolerance test (ipGTT), i.p. insulin tolerance test (ipITT) and metabolic chamber analysis were performed in SD (4% fat) and HFD (55% fat) groups. RESULTS p8-/- mice showed no differences in glucagon or insulin content but higher insulin secretion from pancreatic islets upon glucose stimulation. p8 deficiency resulted in elevated beta cell mass but was not associated with increased insulin resistance in ipGTT or ipITT. Glucose clamp tests also revealed no evidence of association of p8 deficiency with insulin resistance. Metabolic chamber analysis showed equal energy expenditure in p8-/- mice and wild type animals. CONCLUSION p8 depletion may contribute to glucose metabolism via stress-induced insulin production and elevated beta cell mass. Nevertheless, p8 knockout showed no impact on insulin resistance in SD and HFD-fed mice.
Collapse
Affiliation(s)
- Marcus Hollenbach
- Department of Medicine, Neurology and Dermatology, Division of Gastroenterology and Rheumatology, University of Leipzig, Leipzig, Germany
- * E-mail:
| | - Nora Klöting
- IFB Adiposity Disease, University of Leipzig, Leipzig, Germany
| | - Ines Sommerer
- Department of Medicine, Neurology and Dermatology, Division of Gastroenterology and Rheumatology, University of Leipzig, Leipzig, Germany
| | - Jana Lorenz
- Department of Medicine, Neurology and Dermatology, Division of Gastroenterology and Rheumatology, University of Leipzig, Leipzig, Germany
| | - Mario Heindl
- Department of Medicine, Neurology and Dermatology, Division of Gastroenterology and Rheumatology, University of Leipzig, Leipzig, Germany
| | - Matthias Kern
- German Diabetes Center Leipzig, University of Leipzig, Leipzig, Germany
| | - Joachim Mössner
- Department of Medicine, Neurology and Dermatology, Division of Gastroenterology and Rheumatology, University of Leipzig, Leipzig, Germany
| | - Matthias Blüher
- Department of Medicine, Neurology and Dermatology, Division of Endocrinology and Nephrology, University of Leipzig, Leipzig, Germany
| | - Albrecht Hoffmeister
- Department of Medicine, Neurology and Dermatology, Division of Gastroenterology and Rheumatology, University of Leipzig, Leipzig, Germany
| |
Collapse
|
14
|
Lepage D, Bruneau J, Brouillard G, Jones C, Lussier CR, Rémillard A, Lemieux É, Asselin C, Boudreau F. Identification of GATA-4 as a novel transcriptional regulatory component of regenerating islet-derived family members. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2015; 1849:1411-22. [PMID: 26477491 DOI: 10.1016/j.bbagrm.2015.10.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Revised: 10/08/2015] [Accepted: 10/13/2015] [Indexed: 01/30/2023]
Abstract
Intestinal epithelial cells are exposed to luminal bacterial threat and require adequate defense mechanisms to ensure host protection and epithelium regeneration against possible deleterious damage. Differentiated intestinal epithelial cells produce antimicrobial and regenerative components that protect against such challenges. Few intestinal specific transcription factors have been identified to control the switching from repression to activation of this class of gene. Herein, we show that gene transcription of some regenerating islet-derived (REG) family members is dependent on the transcription factor GATA-4. Silencing of GATA-4 expression in cultured intestinal epithelial cells identified Reg3β as a target gene using an unbiased approach of gene expression profiling. Co-transfection and RNA interference assays identified complex GATA-4-interactive transcriptional components required for the activation or repression of Reg3β gene activity. Conditional deletion of Gata4 in the mouse intestinal epithelium supported its regulatory role for Reg1, Reg3α, Reg3β and Reg3γ genes. Reg1 dramatic down-modulation of expression in Gata4 conditional null mice was associated with a significant decrease in intestinal epithelial cell migration. Altogether, these results identify a novel and complex role for GATA-4 in the regulation of REG family members gene expression.
Collapse
Affiliation(s)
- David Lepage
- Département d'anatomie et biologie cellulaire, Faculté de médecine et des sciences de la santé, Pavillon de recherche appliquée sur le cancer, Université de Sherbrooke, Sherbrooke, QC, J1E 4K8, Canada
| | - Joannie Bruneau
- Département d'anatomie et biologie cellulaire, Faculté de médecine et des sciences de la santé, Pavillon de recherche appliquée sur le cancer, Université de Sherbrooke, Sherbrooke, QC, J1E 4K8, Canada
| | - Geneviève Brouillard
- Département d'anatomie et biologie cellulaire, Faculté de médecine et des sciences de la santé, Pavillon de recherche appliquée sur le cancer, Université de Sherbrooke, Sherbrooke, QC, J1E 4K8, Canada
| | - Christine Jones
- Département d'anatomie et biologie cellulaire, Faculté de médecine et des sciences de la santé, Pavillon de recherche appliquée sur le cancer, Université de Sherbrooke, Sherbrooke, QC, J1E 4K8, Canada
| | - Carine R Lussier
- Département d'anatomie et biologie cellulaire, Faculté de médecine et des sciences de la santé, Pavillon de recherche appliquée sur le cancer, Université de Sherbrooke, Sherbrooke, QC, J1E 4K8, Canada
| | - Anthony Rémillard
- Département d'anatomie et biologie cellulaire, Faculté de médecine et des sciences de la santé, Pavillon de recherche appliquée sur le cancer, Université de Sherbrooke, Sherbrooke, QC, J1E 4K8, Canada
| | - Étienne Lemieux
- Département d'anatomie et biologie cellulaire, Faculté de médecine et des sciences de la santé, Pavillon de recherche appliquée sur le cancer, Université de Sherbrooke, Sherbrooke, QC, J1E 4K8, Canada
| | - Claude Asselin
- Département d'anatomie et biologie cellulaire, Faculté de médecine et des sciences de la santé, Pavillon de recherche appliquée sur le cancer, Université de Sherbrooke, Sherbrooke, QC, J1E 4K8, Canada
| | - François Boudreau
- Département d'anatomie et biologie cellulaire, Faculté de médecine et des sciences de la santé, Pavillon de recherche appliquée sur le cancer, Université de Sherbrooke, Sherbrooke, QC, J1E 4K8, Canada.
| |
Collapse
|
15
|
Dakshinamurti K, Bagchi RA, Abrenica B, Czubryt MP. Microarray analysis of pancreatic gene expression during biotin repletion in biotin-deficient rats. Can J Physiol Pharmacol 2015; 93:1103-10. [PMID: 26312779 DOI: 10.1139/cjpp-2014-0517] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Biotin is a B vitamin involved in multiple metabolic pathways. In humans, biotin deficiency is relatively rare but can cause dermatitis, alopecia, and perosis. Low biotin levels occur in individuals with type-2 diabetes, and supplementation with biotin plus chromium may improve blood sugar control. The acute effect on pancreatic gene expression of biotin repletion following chronic deficiency is unclear, therefore we induced biotin deficiency in adult male rats by feeding them a 20% raw egg white diet for 6 weeks. Animals were then randomized into 2 groups: one group received a single biotin supplement and returned to normal chow lacking egg white, while the second group remained on the depletion diet. After 1 week, pancreata were removed from biotin-deficient (BD) and biotin-repleted (BR) animals and RNA was isolated for microarray analysis. Biotin depletion altered gene expression in a manner indicative of inflammation, fibrosis, and defective pancreatic function. Conversely, biotin repletion activated numerous repair and anti-inflammatory pathways, reduced fibrotic gene expression, and induced multiple genes involved in pancreatic endocrine and exocrine function. A subset of the results was confirmed by quantitative real-time PCR analysis, as well as by treatment of pancreatic AR42J cells with biotin. The results indicate that biotin repletion, even after lengthy deficiency, results in the rapid induction of repair processes in the pancreas.
Collapse
Affiliation(s)
- Krishnamurti Dakshinamurti
- Institute of Cardiovascular Sciences, St. Boniface Hospital Research Centre, 351 Tache Avenue, Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R2H 2A6, Canada.,Institute of Cardiovascular Sciences, St. Boniface Hospital Research Centre, 351 Tache Avenue, Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R2H 2A6, Canada
| | - Rushita A Bagchi
- Institute of Cardiovascular Sciences, St. Boniface Hospital Research Centre, 351 Tache Avenue, Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R2H 2A6, Canada.,Institute of Cardiovascular Sciences, St. Boniface Hospital Research Centre, 351 Tache Avenue, Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R2H 2A6, Canada
| | - Bernard Abrenica
- Institute of Cardiovascular Sciences, St. Boniface Hospital Research Centre, 351 Tache Avenue, Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R2H 2A6, Canada.,Institute of Cardiovascular Sciences, St. Boniface Hospital Research Centre, 351 Tache Avenue, Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R2H 2A6, Canada
| | - Michael P Czubryt
- Institute of Cardiovascular Sciences, St. Boniface Hospital Research Centre, 351 Tache Avenue, Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R2H 2A6, Canada.,Institute of Cardiovascular Sciences, St. Boniface Hospital Research Centre, 351 Tache Avenue, Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R2H 2A6, Canada
| |
Collapse
|
16
|
Cendrowski J, Lobo VJSA, Sendler M, Salas A, Kühn JP, Molero X, Fukunaga R, Mayerle J, Lerch MM, Real FX. Mnk1 is a novel acinar cell-specific kinase required for exocrine pancreatic secretion and response to pancreatitis in mice. Gut 2015; 64:937-47. [PMID: 25037190 DOI: 10.1136/gutjnl-2013-306068] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Accepted: 06/25/2014] [Indexed: 01/11/2023]
Abstract
OBJECTIVE Pancreatic acinar cell maturation is dependent on the activity of the pancreas transcription factor 1 (PTF1) complex. Induction of pancreatitis leads to MAP kinase activation and transient suppression of the acinar differentiation programme. We investigated the role of MAP kinase-interacting kinase 1 (Mnk1) in mouse exocrine pancreas development and in the response to secretagogue-induced pancreatitis. DESIGN Mnk1 expression was analysed using immunohistochemistry, RT-qPCR and western blotting. Ptf1a binding to Mnk1 was assessed by chromatin immunoprecipitation and qPCR. Acute pancreatitis was induced in wild type and Mnk1(-/-) mice by 7 h intraperitoneal injections of caerulein. In vitro amylase secretion and trypsinogen activation were assessed using freshly isolated acinar cells. In vivo secretion was quantified by secretin-stimulated MRI. RESULTS Mnk1 is expressed at the highest levels in pancreatic acinar cells and is a direct PTF1 target. Mnk1 is activated upon induction of pancreatitis and is indispensable for eIF4E phosphorylation. The pancreas of Mnk1(-/-) mice is histologically normal. Digestive enzyme content is significantly increased and c-Myc and Ccnd1 levels are reduced in Mnk1(-/-) mice. Upon induction of acute pancreatitis, Mnk1(-/-) mice show impaired eIF4E phosphorylation, activation of c-Myc and downregulation of zymogen content. Acinar cells show defective relocalisation of digestive enzymes, polarity defects and impaired secretory response in vitro and in vivo. CONCLUSIONS Mnk1 is a novel pancreatic acinar cell-specific stress response kinase that regulates digestive enzyme abundance and eIF4E phosphorylation. It is required for the physiological secretory response of acinar cells and for the homeostatic response to caerulein administration during acute pancreatitis.
Collapse
Affiliation(s)
- Jaroslaw Cendrowski
- Epithelial Carcinogenesis Group, Spanish National Cancer Research Center-CNIO, Madrid, Spain
| | | | - Matthias Sendler
- Department of Medicine A, University Medicine, Ernst-Moritz-Arndt University, Greifswald, Germany
| | - Antonio Salas
- Servei d'Anatomia Patològica, Hospital Mútua Terrassa, Barcelona, Spain
| | - Jens-Peter Kühn
- Institute of Radiology, University Medicine, Ernst-Moritz-University, Greifswald, Germany
| | - Xavier Molero
- Exocrine Pancreas Research Unit, Hospital Universitari Vall d'Hebron, Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, CIBEREHD, Barcelona, Spain
| | | | - Julia Mayerle
- Department of Medicine A, University Medicine, Ernst-Moritz-Arndt University, Greifswald, Germany
| | - Markus M Lerch
- Department of Medicine A, University Medicine, Ernst-Moritz-Arndt University, Greifswald, Germany
| | - Francisco X Real
- Epithelial Carcinogenesis Group, Spanish National Cancer Research Center-CNIO, Madrid, Spain Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, Barcelona, Spain
| |
Collapse
|
17
|
Chen SS, Hu W, Wang Z, Lou XE, Zhou HJ. p8 attenuates the apoptosis induced by dihydroartemisinin in cancer cells through promoting autophagy. Cancer Biol Ther 2015; 16:770-9. [PMID: 25891535 DOI: 10.1080/15384047.2015.1026477] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Dihydroartemisinin (DHA) exhibits anticancer activities in a variety of cancer cells, but DHA alone are not effective enough for cancer therapy. In this study we found the stress-regulated protein p8 was obviously increased after DHA treatment in several cancer cells, which further to induce autophagy by the upregulation of endoplasmic reticulum stress-related protein ATF4 and CHOP. Furthermore, when we silenced p8 by siRNA in cancer cells, the apoptosis induced by DHA were notably increased, whereas the overexpression of p8 in cancer cells leaded to the resistance to DHA-induced apoptosis. Moreover, we found the inhibition of autophagy with chloroquine (CQ) can enhance the anticancer effect of DHA both in vitro and in vivo. In conclusion, we found that p8-mediated autophagy attenuates DHA-induced apoptosis in cancer cells, which provides evidence to support the use p8 as a cancer therapeutic target, and suggests that the combination treatment with DHA and autophagy inhibitor might be an effective cancer therapeutic strategy.
Collapse
Affiliation(s)
- Sang-Sang Chen
- a Institute of Pharmacology and Toxicology; College of Pharmaceutical Sciences; Zhejiang University ; Hangzhou , PR China
| | | | | | | | | |
Collapse
|
18
|
Chen XY, Yang YS, Chen K, Chen LS, Xie WR, Wang H. JAK-STAT signaling pathway and acute pancreatitis. Shijie Huaren Xiaohua Zazhi 2015; 23:932-937. [DOI: 10.11569/wcjd.v23.i6.932] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [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
The pathogenesis of acute pancreatitis has long been an important research topic. In acute pancreatitis, cytokines and growth factors bind to Janus kinase (JAK) related receptors, and activate JAKs. The activated JAKs phosphorylate the tyrosine residues of the receptor. The downstream signal transducers and activators of transcription (STAT) then bind to the specific site of the phosphorylated JAK receptor complexes, leading to the activation of STATs. The activated STATs detach from the receptor complexes and translocate to the nucleus to regulate the expression of Bcl-2, Bcl-X(L), Mcl-1 and other genes, thereby participating in the pathogenesis of pancreatitis. Such signal transduction can be terminated by the dephosphorylation of STATs. At present, more and more clinical experiments and animal studies have shown that the JAK-STAT pathway is closely related with acute pancreatitis. In this article, we will review the structure, distribution, and function of JAK-STAT signaling pathway as well as the role of JAK-STAT signaling pathway in the pathogenesis of acute pancreatitis.
Collapse
|
19
|
Abstract
Acute pancreatitis is an inflammatory process of the pancreatic gland that eventually may lead to a severe systemic inflammatory response. A key event in pancreatic damage is the intracellular activation of NF-κB and zymogens, involving also calcium, cathepsins, pH disorders, autophagy, and cell death, particularly necrosis. This review focuses on the new role of redox signaling in acute pancreatitis. Oxidative stress and redox status are involved in the onset of acute pancreatitis and also in the development of the systemic inflammatory response, being glutathione depletion, xanthine oxidase activation, and thiol oxidation in proteins critical features of the disease in the pancreas. On the other hand, the release of extracellular hemoglobin into the circulation from the ascitic fluid in severe necrotizing pancreatitis enhances lipid peroxidation in plasma and the inflammatory infiltrate into the lung and up-regulates the HIF-VEGF pathway, contributing to the systemic inflammatory response. Therefore, redox signaling and oxidative stress contribute to the local and systemic inflammatory response during acute pancreatitis.
Collapse
|
20
|
Weis S, Bielow T, Sommerer I, Iovanna J, Malicet C, Mössner J, Hoffmeister A. P8 deficiency increases cellular ROS and induces HO-1. Arch Biochem Biophys 2014; 565:89-94. [PMID: 25475530 DOI: 10.1016/j.abb.2014.11.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 11/07/2014] [Accepted: 11/18/2014] [Indexed: 10/24/2022]
Abstract
The gene p8 encodes for a small cytoprotective protein with no apparent enzymatic activity being proposed to act as co-transcription factor whose expression is increased during inflammation. Recent data from astrocytes demonstrates that p8 suppression leads to induction of heme oxygenase 1 (HO-1). Here, we assessed the cross-talk between p8 and HO-1 in mouse embryonic fibroblasts (MEF) observing an increased expression of HO-1 in p8-deficient (p8(-/-)) MEFs in non-treated and treated conditions. This effect was independent of the cell cycle. Our findings revealed that generation of reactive oxygen species (ROS) was higher in p8(-/-) MEFs. Mitochondria and NADPH oxidases were not the origin of ROS. This observation was not restricted to MEF as suppression of p8 gene transcription in MiaPaCa-2 cells also led to increased intracellular ROS. Additionally, p8 deficiency did not affect the Rac1 dependant NADPH oxidase complex. Our data shows that p8 deficiency increases ROS and subsequently the expression of anti-oxidative enzymes, such as HO-1, suggesting an involvement in the anti-oxidative defense. Moreover, we suggest that the severity of AP observed in p8(-/-) mice is induced by an impaired anti oxidative capacity of the pancreas, which is caused by increased generation of ROS.
Collapse
Affiliation(s)
- Sebastian Weis
- Division of Gastroenterology and Rheumatology, Department of Internal Medicine, Neurology and Dermatology, University Hospital Leipzig, Germany; Center for Sepsis Control & Care, Jena University Hospital, Jena, Germany; Center for Infectious Diseases and Infection Control, Jena University Hospital, Jena, Germany.
| | - Tobias Bielow
- Division of Gastroenterology and Rheumatology, Department of Internal Medicine, Neurology and Dermatology, University Hospital Leipzig, Germany
| | - Ines Sommerer
- Division of Gastroenterology and Rheumatology, Department of Internal Medicine, Neurology and Dermatology, University Hospital Leipzig, Germany
| | - Juan Iovanna
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Marseille, France
| | | | - Joachim Mössner
- Division of Gastroenterology and Rheumatology, Department of Internal Medicine, Neurology and Dermatology, University Hospital Leipzig, Germany
| | - Albrecht Hoffmeister
- Division of Gastroenterology and Rheumatology, Department of Internal Medicine, Neurology and Dermatology, University Hospital Leipzig, Germany
| |
Collapse
|
21
|
Abstract
OBJECTIVES The gene p8 was initially described in pancreatic tissue during acute experimental pancreatitis, a disease that is characterized by a systemic immune response. Although early reports suggested that p8 affects leukocyte migration during acute pancreatitis (AP), no studies revealing its immune-modulatory effects have been performed. METHODS We investigated the composition of the cellular immune system in naive p8 knockout (p8(−/−)) mice and compared with matched wild-type mice during pancreatitis. RESULTS In young mice, there were no relevant differences in the composition of peripheral and splenic CD3(+), CD3(+)CD4(+), CD3(+)CD8(+), CD11b(+)Gr-1(-), and Gr-1 cells. In mature p8(−/−) mice, increased splenic CD4CD25FoxP3 cells, spleen siderosis, and increased marginal zones in the splenic white pulp were found. During AP, peripheral and splenic CD3(+) and CD3CD4 declined stronger in the p8(−/−) mice. The spleen of the p8(−/−) mice showed severe hypoplasia of the white pulp and mild hyperplasia of the red pulp. This was associated with a significantly increased rate of apoptosis. CONCLUSIONS We conclude that p8 has no impact on the cellular composition of the adaptive and innate immune systems in noninflammatory conditions. However, it may limit apoptosis and maintain homeostasis of the immune reaction during AP.
Collapse
|
22
|
Ma SC, Yao JF, Guo Y, Cui DL, Yang H, Han JL. Relationship between Reg proteins and intestinal mucosa barrier damage in rats with severe acute pancreatitis. Shijie Huaren Xiaohua Zazhi 2014; 22:3744-3752. [DOI: 10.11569/wcjd.v22.i25.3744] [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 detect the expression of regenerating islet-derived proteins (Reg)Ⅰand Ⅲ in the intestinal mucosa of rats with severe acute pancreatitis (SAP), and to evaluate the relationship between the levels of RegⅠand Ⅲ and intestinal mucosal barrier damage.
METHODS: Seventy-two adult SD rats were randomly divided into three groups: a normal control (N) group, an SAP (S) group, and a pyrrolidine dithiocarbamate (PDTC, 10 mg/kg) pretreatment (P) group. Each group was further divided into two subgroups for testing at different time points (12 and 24 h), with 12 rats in each subgroup. The rats in the S group were given 20% L-arginine (L-Arg, 2.5 g/kg) by intraperitoneal injection twice at one-hour interval to induce SAP. The N group was given equal volume of normal saline. The P group was given PDTC 10 mg/kg by intraperitoneal injection 1 h before the first injection of L-Arg. All rats were killed 12 h or 24 h after L-Arg injection to collect blood, pancreatic and intestinal tissue samples. The pathological changes in pancreatic and intestinal tissues were observed and graded under an optical microscope. ELISA was used to detect the levels of serum interleukin 22 (IL-22), tumor necrosis factor-α (TNF-α) and intestinal fatty acid binding protein (I-FABP). The expression of RegⅠand Ⅲ mRNAs in intestinal tissue was evaluated by RT-PCR. The levels of RegⅠ, Ⅲ and nuclear-factor κB (NF-κB) proteins in intestinal tissue were detected by Western blot.
RESULTS: In the SAP group, the scores of pancreatic changes (12 h: 8.92 ± 1.130; 24 h: 11.31 ± 1.609) and intestinal mucosal changes (12 h: 3.79 ± 0.689, 24 h: 4.33 ± 0.354), and the levels of IL-22 (12 h: 712.46 ng/mL ± 81.549 ng/mL, 24 h: 751.02 ng/mL ± 104.054 ng/mL), TNF-α (12 h: 138.08 ng/mL ± 20.369 ng/mL, 24 h: 159.43 ng/mL ± 24.46 ng/mL), I-FABP (12 h: 338.04 IU/mL ± 61.876 IU/mL, 24 h: 395.26 IU/mL ± 58.547 IU/mL), intestinal NF-κB p65 (12 h: 0.51 ± 0.065, 24 h: 0.60 ± 0.066), RegⅠprotein (12 h: 0.45 ± 0.047, 24 h: 0.56 ± 0.033), and Reg Ⅲ protein (12 h: 0.70 ± 0.084, 24 h: 0.92 ± 0.163) were significantly higher (P < 0.05) than those in the control group. Compared with the S group, pretreatment with different doses of PDTC significantly decreased the above parameters (P < 0.05), although the levels of these parameters were still significantly higher than those in the N group (P < 0.05). There were positive correlations among RegⅠand Ⅲ protein expression, intestinal mucosal pathological score, IL-22, I-FABP, TNF-α, and NF-κB p65 expression.
CONCLUSION: RegⅠand Ⅲ protein expression is upregulated in SAP, which is possibly associated with intestinal mucosa damage and NF-κB signaling pathway activation.
Collapse
|
23
|
Grasso D, Garcia MN, Hamidi T, Cano C, Calvo E, Lomberk G, Urrutia R, Iovanna JL. Genetic inactivation of the pancreatitis-inducible gene Nupr1 impairs PanIN formation by modulating Kras(G12D)-induced senescence. Cell Death Differ 2014; 21:1633-41. [PMID: 24902898 PMCID: PMC4158688 DOI: 10.1038/cdd.2014.74] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Revised: 04/01/2014] [Accepted: 04/30/2014] [Indexed: 12/29/2022] Open
Abstract
Nuclear protein 1 (Nupr1), a small chromatin protein, has a critical role in cancer development, progression and resistance to therapy. Previously, we had demonstrated that Nupr1 cooperates with KrasG12D to induce pancreas intraepithelial neoplasias (PanIN) formation and pancreatic ductal adenocarcinoma development in mice. However, the molecular mechanisms by which Nupr1 influences Kras-mediated preneoplastic growth remain to be fully characterized. In the current study, we report evidence supporting a role for Nupr1 as a gene modifier of KrasG12D-induced senescence, which must be overcome to promote PanIN formation. We found that genetic inactivation of Nupr1 in mice impairs Kras-induced PanIN, leading to an increase in β-galactosidase-positive cells and an upregulation of surrogate marker genes for senescence. More importantly, both of these cellular and molecular changes are recapitulated by the results of mechanistic experiments using RNAi-based inactivation of Nupr1 in human pancreatic cancer cell models. In addition, the senescent phenotype, which results from Nupr1 inactivation, is accompanied by activation of the FoxO3a-Skp2-p27Kip1-pRb-E2F pathway in vivo and in vitro. Thus, combined, these results show, for the first time, that Nupr1 aids oncogenic Kras to bypass senescence in a manner that cooperatively promotes PanIN formation. Besides its mechanistic importance, this new knowledge bears medical relevance as it delineates early pathobiological events that may be targeted in the future as a means to interfere with the formation of preneoplastic lesions early during pancreatic carcinogenesis.
Collapse
Affiliation(s)
- D Grasso
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Marseille, France
| | - M N Garcia
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Marseille, France
| | - T Hamidi
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Marseille, France
| | - C Cano
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Marseille, France
| | - E Calvo
- Molecular Endocrinology and Oncology Research Center, CHUL Research Center, Quebec City, QC, Canada
| | - G Lomberk
- Laboratory of Epigenetics and Chromatin Dynamics, Gastroenterology Research Unit, Departments of Biochemistry and Molecular Biology, Biophysics, and Medicine, Mayo Clinic, Rochester, MN, USA
| | - R Urrutia
- Laboratory of Epigenetics and Chromatin Dynamics, Gastroenterology Research Unit, Departments of Biochemistry and Molecular Biology, Biophysics, and Medicine, Mayo Clinic, Rochester, MN, USA
| | - J L Iovanna
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Marseille, France
| |
Collapse
|
24
|
Cano CE, Hamidi T, Garcia MN, Grasso D, Loncle C, Garcia S, Calvo E, Lomberk G, Dusetti N, Bartholin L, Urrutia R, Iovanna JL. Genetic inactivation of Nupr1 acts as a dominant suppressor event in a two-hit model of pancreatic carcinogenesis. Gut 2014; 63:984-95. [PMID: 24026351 DOI: 10.1136/gutjnl-2013-305221] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Nuclear protein 1 (Nupr1) is a major factor in the cell stress response required for Kras(G12D)-driven formation of pancreatic intraepithelial neoplastic lesions (PanINs). We evaluated the relevance of Nupr1 in the development of pancreatic cancer. METHODS We investigated the role of Nupr1 in pancreatic ductal adenocarcinoma (PDAC) progression beyond PanINs in Pdx1-cre;LSL-Kras(G12D);Ink4a/Arf(fl/fl)(KIC) mice. RESULTS Even in the context of the second tumorigenic hit of Ink4a/Arf deletion, Nupr1 deficiency led to suppression of malignant transformation involving caspase 3 activation in premalignant cells of KIC pancreas. Only half of Nupr1-deficient;KIC mice achieved PDAC development, and incident cases survived longer than Nupr1(wt);KIC mice. This was associated with the development of well-differentiated PDACs in Nupr1-deficient;KIC mice, which displayed enrichment of genes characteristic of the recently identified human classical PDAC subtype. Nupr1-deficient;KIC PDACs also shared with human classical PDACs the overexpression of the Kras-activation gene signature. In contrast, Nupr1(wt);KIC mice developed invasive PDACs with enriched gene signature of human quasi-mesenchymal (QM) PDACs. Cells derived from Nupr1-deficient;KIC PDACs growth in an anchorage-independent manner in vitro had higher aldehyde dehydrogenase activity and overexpressed nanog, Oct-4 and Sox2 transcripts compared with Nupr1(wt);KIC cells. Moreover, Nupr1-deficient and Nurpr1(wt);KIC cells differed in their sensitivity to the nucleoside analogues Ly101-4b and WJQ63. Together, these findings show the pivotal role of Nupr1 in both the initiation and late stages of PDAC in vivo, with a potential impact on PDAC cell stemness. CONCLUSIONS According to Nupr1 status, KIC mice develop tumours that phenocopy human classical or QM-PDAC, respectively, and present differential drug sensitivity, thus becoming attractive models for preclinical drug trials.
Collapse
Affiliation(s)
- Carla E Cano
- Centre de Recherche en Carcérologie de Marseille (CRCM), INSERM UMR 1068, CNRS UMR 7258, Aix-Marseille University and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, , Marseille, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Morris JP, Greer R, Russ HA, von Figura G, Kim GE, Busch A, Lee J, Hertel KJ, Kim S, Mcmanus M, Hebrok M. Dicer regulates differentiation and viability during mouse pancreatic cancer initiation. PLoS One 2014; 9:e95486. [PMID: 24788257 PMCID: PMC4006805 DOI: 10.1371/journal.pone.0095486] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Accepted: 03/26/2014] [Indexed: 12/13/2022] Open
Abstract
miRNA levels are altered in pancreatic ductal adenocarcinoma (PDA), the most common and lethal pancreatic malignancy, and intact miRNA processing is essential for lineage specification during pancreatic development. However, the role of miRNA processing in PDA has not been explored. Here we study the role of miRNA biogenesis in PDA development by deleting the miRNA processing enzyme Dicer in a PDA mouse model driven by oncogenic Kras. We find that loss of Dicer accelerates Kras driven acinar dedifferentiation and acinar to ductal metaplasia (ADM), a process that has been shown to precede and promote the specification of PDA precursors. However, unconstrained ADM also displays high levels of apoptosis. Dicer loss does not accelerate development of Kras driven PDA precursors or PDA, but surprisingly, we observe that mouse PDA can develop without Dicer, although at the expense of proliferative capacity. Our data suggest that intact miRNA processing is involved in both constraining pro-tumorigenic changes in pancreatic differentiation as well as maintaining viability during PDA initiation.
Collapse
Affiliation(s)
- John P. Morris
- Diabetes Center, Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - Renee Greer
- Diabetes Center, Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - Holger A. Russ
- Diabetes Center, Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - Guido von Figura
- Diabetes Center, Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - Grace E. Kim
- Department of Pathology, University of California San Francisco, San Francisco, California, United States of America
| | - Anke Busch
- Department of Microbiology and Molecular Genetics, University of California Irvine, Irvine, California, United States of America
| | - Jonghyeob Lee
- Department of Developmental Biology, Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, California, United States of America
| | - Klemens J. Hertel
- Department of Microbiology and Molecular Genetics, University of California Irvine, Irvine, California, United States of America
| | - Seung Kim
- Department of Developmental Biology, Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, California, United States of America
| | - Michael Mcmanus
- Diabetes Center, Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - Matthias Hebrok
- Diabetes Center, Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
- * E-mail:
| |
Collapse
|
26
|
Calderari S, Irminger JC, Giroix MH, Ehses JA, Gangnerau MN, Coulaud J, Rickenbach K, Gauguier D, Halban P, Serradas P, Homo-Delarche F. Regenerating 1 and 3b gene expression in the pancreas of type 2 diabetic Goto-Kakizaki (GK) rats. PLoS One 2014; 9:e90045. [PMID: 24587207 PMCID: PMC3936001 DOI: 10.1371/journal.pone.0090045] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Accepted: 01/27/2014] [Indexed: 02/06/2023] Open
Abstract
Regenerating (REG) proteins are associated with islet development, β-cell damage, diabetes and pancreatitis. Particularly, REG-1 and REG-3-beta are involved in cell growth/survival and/or inflammation and the Reg1 promoter contains interleukin-6 (IL-6)-responsive elements. We showed by transcriptome analysis that islets of Goto-Kakizaki (GK) rats, a model of spontaneous type 2 diabetes, overexpress Reg1, 3α, 3β and 3γ, vs Wistar islets. Goto-Kakizaki rat islets also exhibit increased cytokine/chemokine expression/release, particularly IL-6. Here we analyzed Reg1 and Reg3β expression and REG-1 immuno-localization in the GK rat pancreas in relationship with inflammation. Isolated pancreatic islets and acinar tissue from male adult Wistar and diabetic GK rats were used for quantitative RT-PCR analysis. REG-1 immunohistochemistry was performed on paraffin sections with a monoclonal anti-rat REG-1 antibody. Islet cytokine/chemokine release was measured after 48 h-culture. Islet macrophage-positive area was quantified on cryostat sections using anti-CD68 and major histocompatibility complex (MHC) class II antibodies. Pancreatic exocrine-to-endocrine Reg1 and Reg3β mRNA ratios were markedly increased in Wistar vs GK rats. Conversely, both genes were upregulated in isolated GK rat islets. These findings were unexpected, because Reg genes are expressed in the pancreatic acinar tissue. However, we observed REG-1 protein labeling in acinar peri-ductal tissue close to islets and around large, often disorganized, GK rat islets, which may retain acinar cells due to their irregular shape. These large islets also showed peri-islet macrophage infiltration and increased release of various cytokines/chemokines, particularly IL-6. Thus, IL-6 might potentially trigger acinar REG-1 expression and secretion in the vicinity of large diabetic GK rat islets. This increased acinar REG-1 expression might reflect an adaptive though unsuccessful response to deleterious microenvironment.
Collapse
MESH Headings
- Animals
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/metabolism
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Chemokines/blood
- Chemokines/metabolism
- Diabetes Mellitus, Type 2/blood
- Diabetes Mellitus, Type 2/genetics
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/pathology
- Gene Expression Regulation
- Islets of Langerhans/metabolism
- Lectins, C-Type/genetics
- Lectins, C-Type/metabolism
- Lithostathine/genetics
- Lithostathine/metabolism
- Macrophages/metabolism
- Male
- Pancreatitis-Associated Proteins
- Rats
- Rats, Wistar
Collapse
Affiliation(s)
- Sophie Calderari
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMRS 872, Team 6, Centre de Recherche des Cordeliers (CRC), Université Pierre et Marie Curie, Paris 6, Paris, France
| | - Jean-Claude Irminger
- Department of Genetic Medicine and Development, University of Geneva, Geneva, Switzerland
| | - Marie-Hélène Giroix
- Equipe associée au Centre National de la Recherche Scientifique (CNRS) 4413-Unité de Biologie Fonctionnelle et Adaptative (BFA), Team 1 (Biologie et Pathologie du Pancréas Endocrine (B2PE)), Université Paris-Diderot Sorbonne-Paris-Cité, Paris, France
| | - Jan A. Ehses
- Department of Surgery, Faculty of Medicine, University of British Columbia and Child and Family Research Institute, Vancouver, BC, Canada
| | - Marie-Noëlle Gangnerau
- Equipe associée au Centre National de la Recherche Scientifique (CNRS) 4413-Unité de Biologie Fonctionnelle et Adaptative (BFA), Team 1 (Biologie et Pathologie du Pancréas Endocrine (B2PE)), Université Paris-Diderot Sorbonne-Paris-Cité, Paris, France
| | - Josiane Coulaud
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMRS 872, Team 6, Centre de Recherche des Cordeliers (CRC), Université Pierre et Marie Curie, Paris 6, Paris, France
| | - Katharina Rickenbach
- Department of Genetic Medicine and Development, University of Geneva, Geneva, Switzerland
| | - Dominique Gauguier
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMRS 872, Team 6, Centre de Recherche des Cordeliers (CRC), Université Pierre et Marie Curie, Paris 6, Paris, France
| | - Philippe Halban
- Department of Genetic Medicine and Development, University of Geneva, Geneva, Switzerland
| | - Patricia Serradas
- INSERM UMRS 872, Team 9, CRC, Université Pierre et Marie Curie, Paris 6, Paris, France
| | - Françoise Homo-Delarche
- Institut National de la Santé et de la Recherche Médicale (INSERM) UMRS 872, Team 6, Centre de Recherche des Cordeliers (CRC), Université Pierre et Marie Curie, Paris 6, Paris, France
- * E-mail:
| |
Collapse
|
27
|
Zhang R, Yao F, Cheng C, Chen Y, Lv Y, Li Z, Zhao N, Wang T, Xin W, Zou X, Hou L. Expression and roles of As-NUPR1 protein from Artemia sinica during embryo development and in response to salinity stress. Mol Biol Rep 2014; 41:3465-73. [PMID: 24510410 DOI: 10.1007/s11033-014-3208-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2013] [Accepted: 01/27/2014] [Indexed: 11/24/2022]
Abstract
As-NUPR1, a stress-related protein, plays an important role in post-diapause during embryonic development in the brine shrimp Artemia sinica. In the present study, successful expression of As-NUPR1 from the cDNA sequence isolated from A. sinica was demonstrated using a prokaryotic expression system. The recombinant protein consisted of 132 amino acids with a molecular weight of 15 kDa, and a predicted isoelectric point of 7.17. As-NUPR1 polyclonal antibodies were prepared by immunization of Balb/c mice with purified recombinant As-NUPR1 protein as an antigen, and immunological studies were carried out. Expression of As-NUPR1 during different developmental stages of the embryo and in response to salinity stress was analyzed in A. sinica using Western blots. The experimental results showed that the expression of As-NUPR1 is widely distributed at different developmental stages in A. sinica, and there was no tissue or organ specificity. Expression of As-NUPR1 decreased gradually during the diapause termination stage of embryo development, after which there was a general increase in expression after breaking the shell. In addition, As-NUPR1 expression was highly upregulated under conditions of high salinity. These results suggest that the As-NUPR1 protein is a stress-related protein that plays a role in protecting embryos from high salt damage in different embryonic developmental stages, especially during the post-diapause period.
Collapse
Affiliation(s)
- Rui Zhang
- College of Life Sciences, Liaoning Normal University, No. 1, Liushu South Street, Ganjingzi District, Dalian, 116081, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Barbosa-Sampaio HC, Liu B, Drynda R, Rodriguez de Ledesma AM, King AJ, Bowe JE, Malicet C, Iovanna JL, Jones PM, Persaud SJ, Muller DS. Nupr1 deletion protects against glucose intolerance by increasing beta cell mass. Diabetologia 2013; 56:2477-86. [PMID: 23900510 DOI: 10.1007/s00125-013-3006-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Accepted: 07/05/2013] [Indexed: 01/08/2023]
Abstract
AIMS/HYPOTHESIS The stress-activated nuclear protein transcription regulator 1 (NUPR1) is induced in response to glucose and TNF-α, both of which are elevated in type 2 diabetes, and Nupr1 has been implicated in cell proliferation and apoptosis cascades. We used Nupr1(-/-) mice to study the role of Nupr1 in glucose homeostasis under normal conditions and following maintenance on a high-fat diet (HFD). METHODS Glucose homeostasis in vivo was determined by measuring glucose tolerance, insulin sensitivity and insulin secretion. Islet number, morphology and beta cell area were assessed by immunofluorescence and morphometric analysis, and islet cell proliferation was quantified by analysis of BrdU incorporation. Islet gene expression was measured by gene arrays and quantitative RT-PCR, and gene promoter activities were monitored by measuring luciferase activity. RESULTS Nupr1(-/-) mice had increased beta cell mass as a consequence of enhanced islet cell proliferation. Nupr1-dependent suppression of beta cell Ccna2 and Tcf19 promoter activities was identified as a mechanism through which Nupr1 may regulate beta cell cycle progression. Nupr1(-/-) mice maintained on a normal diet were mildly insulin resistant, but were normoglycaemic with normal glucose tolerance because of compensatory increases in basal and glucose-induced insulin secretion. Nupr1 deletion was protective against HFD-induced obesity, insulin resistance and glucose intolerance. CONCLUSIONS/INTERPRETATION Inhibition of NUPR1 expression or activity has the potential to protect against the metabolic defects associated with obesity and type 2 diabetes.
Collapse
Affiliation(s)
- Helena C Barbosa-Sampaio
- Diabetes Research Group, Division of Diabetes and Nutritional Sciences, School of Medicine, King's College London, 2.9N Hodgkin Building, Guy's Campus, London, SE1 1UL, UK
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Gironella M, Calvo C, Fernández A, Closa D, Iovanna JL, Rosello-Catafau J, Folch-Puy E. Reg3β deficiency impairs pancreatic tumor growth by skewing macrophage polarization. Cancer Res 2013; 73:5682-94. [PMID: 23867474 DOI: 10.1158/0008-5472.can-12-3057] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The lectin Reg3β provides crucial protection to various tissues against inflammation, a potential risk factor for pancreatic ductal adenocarcinoma. Reg3β is also overexpressed in serum and pancreatic juice from patients with this cancer, but its function in this context remains to be elucidated. In this study, we investigated the role of Reg3β in tumor development in an orthotopic mouse model of pancreatic cancer. Reg3β deletion in mice drastically impaired pancreatic tumor growth, correlating with decreased angiogenesis and increased apoptosis of tumor cells. Moreover, Reg3β deficiency resulted in an alteration of the tumoral immune microenvironment, reflected by a decrease in the M2/M1 ratio of tumor-associated macrophages and an upregulation of CD3(+) cell infiltration. Addition of Reg3β to prestimulated RAW 264.7 or primary macrophages enhanced M2 polarization through the activation of STAT3 signaling pathway. Conditioned media from Reg3β-M2-polarized primary macrophages inhibited apoptosis and prolonged the viability of Panc02 tumor cells. Our studies reveal a novel role for Reg3β as a tumor promoter in pancreatic adenocarcinoma through the regulation of tumor stroma. Thus, inhibition of this protein may be a useful strategy in treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Meritxell Gironella
- Authors' Affiliations: Gastroenterology Department, Hospital Clinic of Barcelona, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), IDIBAPS, Barcelona, Catalonia; Experimental Pathology Department, IIBB-CSIC, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Barcelona, Catalonia, Spain; and Centre de Recherche en Cancérologie de Marseille (CRCM), Unité 1068, Marseille, France
| | | | | | | | | | | | | |
Collapse
|
30
|
Shigekawa M, Hikita H, Kodama T, Shimizu S, Li W, Uemura A, Miyagi T, Hosui A, Kanto T, Hiramatsu N, Tatsumi T, Takeda K, Akira S, Takehara T. Pancreatic STAT3 protects mice against caerulein-induced pancreatitis via PAP1 induction. THE AMERICAN JOURNAL OF PATHOLOGY 2012; 181:2105-13. [PMID: 23064197 DOI: 10.1016/j.ajpath.2012.08.038] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2012] [Revised: 08/13/2012] [Accepted: 08/28/2012] [Indexed: 02/07/2023]
Abstract
The signal transducer and activator of transcription 3 (STAT3) is a transcription factor that controls expressions of several genes involved in cell survival, proliferation and differentiation, and tissue inflammation. However, the significance of pancreatic STAT3 in acute pancreatitis remains unclear. We generated conditional STAT3 knockout (stat3(Δ/Δ)) mice by crossing stat3(flox/flox) mice with Pdx1-promoter Cre transgenic mice. Caerulein administration activated pancreatic STAT3 and induced acute pancreatitis as early as 3 hours in wild-type mice, and full recovery from the induced pancreatic injury was observed within 7 days. The levels of serum amylase and lipase and histologic scores of pancreatic necrosis and inflammatory cell infiltration were significantly higher at 3 hours in stat3(Δ/Δ) mice than in stat3(flox/flox) mice. Pancreatic recovery after pancreatitis was significantly delayed in stat3(Δ/Δ) mice compared with stat3(flox/flox) mice. Although stat3(flox/flox) mice had marked production in the pancreas of pancreatitis-associated protein 1 (PAP1), a serum acute phase protein, this induction was completely abrogated in stat3(Δ/Δ) mice. Enforced production of PAP1 by a hydrodynamic procedure in the liver significantly suppressed pancreatic necrosis and inflammation and also promoted pancreatic regeneration and recovery in stat3(Δ/Δ) mice to levels similar to those observed in stat3(flox/flox) mice. In conclusion, pancreatic STAT3 is indispensable for PAP1 production, and this STAT3/PAP1 pathway plays a protective role in caerulein-induced pancreatitis.
Collapse
Affiliation(s)
- Minoru Shigekawa
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Suita, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Lv Y, Yang X, Huo Y, Tian H, Li S, Yin Y, Hao Z. Adenovirus-mediated hepatocarcinoma-intestine-pancreas/pancreatitis-associated protein suppresses dextran sulfate sodium-induced acute ulcerative colitis in rats. Inflamm Bowel Dis 2012; 18:1950-60. [PMID: 22419609 DOI: 10.1002/ibd.22887] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2011] [Accepted: 12/28/2011] [Indexed: 01/04/2023]
Abstract
BACKGROUND Although increased expression of hepatocarcinoma-intestine-pancreas/pancreatitis-associated protein (HIP/PAP) has been reported in ulcerative colitis (UC), its role in UC remains unclear. This study was designed to assess the function of HIP/PAP in experimental UC and further to explore its underlying mechanisms. METHODS Recombinant adenovirus was prepared to mediate ectopic expression of HIP/PAP in the colon of rats. The effect of HIP/PAP on dextran sodium sulfate (DSS)-induced colitis was assessed by disease activity index (DAI), macroscopic, and histological evaluations. Superoxide dismutase (SOD) and myeloperoxidase (MPO) activities, malondialdehyde (MDA) content, and tumor necrosis factor α (TNF-α) and interleukin-6 (IL-6) production were determined in colonic mucosa. Proliferation cell nuclear antigen (PCNA) was immunostained to reflect the proliferation of colonic epithelia. The effects of HIP/PAP on proliferation and H(2)O(2) -induced apoptosis of SW480 and LoVo colonic adenocarcinoma cells were also determined. Gene expression profiles in SW480 after HIP/PAP overexpression were analyzed by microarray analysis. RESULTS The protective effect of HIP/PAP against DSS-induced colitis in rats was confirmed. Ectopic expression of HIP/PAP resulted in attenuation of oxidative damage, reduction of TNF-α and IL-6 expression, and elevation of epithelial proliferation in colonic mucosa and led to decreased apoptosis and increased proliferation in colonic adenocarcinoma cells. Microarray analysis revealed altered expression of inflammation-related molecules, growth factors, proliferation-related molecules, and antioxidant enzymes under overexpression of HIP/PAP. CONCLUSIONS HIP/PAP has a protective effect against DSS-induced colitis in rats via inhibiting inflammation, alleviating oxidative damage, and promoting colonic epithelium regeneration. HIP/PAP might represent a new promising therapeutic strategy in UC.
Collapse
Affiliation(s)
- Yifei Lv
- Department of Gastroenterology, First Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, China
| | | | | | | | | | | | | |
Collapse
|
32
|
Vincent AJ, Ren S, Harris LG, Devine DJ, Samant RS, Fodstad O, Shevde LA. Cytoplasmic translocation of p21 mediates NUPR1-induced chemoresistance: NUPR1 and p21 in chemoresistance. FEBS Lett 2012; 586:3429-34. [PMID: 22858377 DOI: 10.1016/j.febslet.2012.07.063] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2012] [Revised: 07/09/2012] [Accepted: 07/22/2012] [Indexed: 12/22/2022]
Abstract
The expression of Nuclear Protein 1 (NUPR1) is associated with chemoresistance in multiple malignancies. We previously reported that NUPR1 functions as a transcriptional cofactor for the p300-p53 complex and transcriptionally regulates p21 expression. In the present study we investigated the activity of NUPR1 in p53-deficient, triple-negative, inflammatory SUM159 breast cancer cells. Our studies reveal that NUPR1 confers growth benefit and chemoresistance by causing Akt-mediated phosphorylation and subsequent cytoplasmic re-localization of p21 and activation of the anti-apoptotic Bcl-xL protein. Our findings elucidate a NUPR1-PI-3-K/Akt-phospho-p21 axis that functions in p53-negative, inflammatory breast cancer cells to enhance chemoresistance in breast cancer.
Collapse
Affiliation(s)
- Andrew J Vincent
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL, United States
| | | | | | | | | | | | | |
Collapse
|
33
|
Cano CE, Sandí MJ, Hamidi T, Calvo EL, Turrini O, Bartholin L, Loncle C, Secq V, Garcia S, Lomberk G, Kroemer G, Urrutia R, Iovanna JL. Homotypic cell cannibalism, a cell-death process regulated by the nuclear protein 1, opposes to metastasis in pancreatic cancer. EMBO Mol Med 2012; 4:964-79. [PMID: 22821859 PMCID: PMC3491828 DOI: 10.1002/emmm.201201255] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2012] [Revised: 06/15/2012] [Accepted: 06/20/2012] [Indexed: 01/06/2023] Open
Abstract
Pancreatic adenocarcinoma (PDAC) is an extremely deadly disease for which all treatments available have failed to improve life expectancy significantly. This may be explained by the high metastatic potential of PDAC cells, which results from their dedifferentiation towards a mesenchymal phenotype. Some PDAC present cell-in-cell structures whose origin and significance are currently unknown. We show here that cell-in-cells form after homotypic cell cannibalism (HoCC). We found PDAC patients whose tumours display HoCC develop less metastasis than those without. In vitro, HoCC was promoted by inactivation of the nuclear protein 1 (Nupr1), and was enhanced by treatment with transforming growth factor β. HoCC ends with death of PDAC cells, consistent with a metastasis suppressor role for this phenomenon. Hence, our data indicates a protective role for HoCC in PDAC and identifies Nupr1 as a molecular regulator of this process.
Collapse
|
34
|
Role of heat-stable enterotoxins in the induction of early immune responses in piglets after infection with enterotoxigenic Escherichia coli. PLoS One 2012; 7:e41041. [PMID: 22815904 PMCID: PMC3398878 DOI: 10.1371/journal.pone.0041041] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2011] [Accepted: 06/21/2012] [Indexed: 01/01/2023] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) strains that produce heat-stable (ST) and/or heat - labile (LT) enterotoxins are cause of post – weaning diarrhea in piglets. However, the relative importance of the different enterotoxins in host immune responses against ETEC infection has been poorly defined. In the present study, several isogenic mutant strains of an O149:F4ac+, LT+ STa+ STb+ ETEC strain were constructed that lack the expression of LT in combination with one or both types of ST enterotoxins (STa and/or STb). The small intestinal segment perfusion (SISP) technique and microarray analysis were used to study host early immune responses induced by these mutant strains 4 h after infection in comparison to the wild type strain and a PBS control. Simultaneously, net fluid absorption of pig small intestinal mucosa was measured 4 h after infection, allowing us to correlate enterotoxin secretion with gene regulation. Microarray analysis showed on the one hand a non-toxin related general antibacterial response comprising genes such as PAP, MMP1 and IL8. On the other hand, results suggest a dominant role for STb in small intestinal secretion early after post-weaning infection, as well as in the induced innate immune response through differential regulation of immune mediators like interleukin 1 and interleukin 17.
Collapse
|
35
|
Fu S, Stanek A, Mueller CM, Brown NA, Huan C, Bluth MH, Zenilman ME. Acute pancreatitis in aging animals: Loss of pancreatitis-associated protein protection? World J Gastroenterol 2012; 18:3379-88. [PMID: 22807607 PMCID: PMC3396190 DOI: 10.3748/wjg.v18.i26.3379] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2011] [Revised: 07/28/2011] [Accepted: 05/12/2012] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the effect of age on severity of acute pancreatitis (AP) using biochemical markers, histology and expression of the protective pancreatitis-associated proteins (PAPs).
METHODS: AP was induced via intraductal injection of 4% sodium taurocholate in young and old rats. Sera and pancreata were assayed at 24 h for the parameters listed above; we also employed a novel molecular technique to assess bacterial infiltration using polymerase chain reaction to measure bacterial genomic ribosomal RNA.
RESULTS: At 24 h after induction of AP, the pancreata of older animals had less edema (mean ± SE histologic score of young vs old: 3.11 ± 0.16 vs 2.50 ± -0.11, P < 0.05), decreased local inflammatory response (histologic score of stromal infiltrate: 3.11 ± 0.27 vs 2.00 ± 0.17, P < 0.05) and increased bacterial infiltration (174% ± 52% increase from sham vs 377% ± 4%, P < 0.05). A decreased expression of PAP1 and PAP2 was demonstrated by Western blotting analysis and immunohistochemical staining. There were no differences in serum amylase and lipase activity, or tissue myeloperoxidase or monocyte chemotactic protein-1 levels. However, in the most-aged group, serum C-reactive protein levels were higher (young vs old: 0.249 ± 0.04 mg/dL vs 2.45 ± 0.68 mg/dL, P < 0.05).
CONCLUSION: In older animals, there is depressed PAP expression related to a blunted inflammatory response in AP which is associated with worsened bacterial infiltration and higher C-reactive protein level; this may explain the more aggressive clinical course.
Collapse
|
36
|
Hamidi T, Algül H, Cano CE, Sandi MJ, Molejon MI, Riemann M, Calvo EL, Lomberk G, Dagorn JC, Weih F, Urrutia R, Schmid RM, Iovanna JL. Nuclear protein 1 promotes pancreatic cancer development and protects cells from stress by inhibiting apoptosis. J Clin Invest 2012; 122:2092-103. [PMID: 22565310 DOI: 10.1172/jci60144] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2011] [Accepted: 03/14/2012] [Indexed: 12/13/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has the lowest survival rate of all cancers and shows remarkable resistance to cell stress. Nuclear protein 1 (Nupr1), which mediates stress response in the pancreas, is frequently upregulated in pancreatic cancer. Here, we report that Nupr1 plays an essential role in pancreatic tumorigenesis. In a mouse model of pancreatic cancer with constitutively expressed oncogenic Kras(G12D), we found that loss of Nupr1 protected from the development of pancreatic intraepithelial neoplasias (PanINs). Further, in cultured pancreatic cells, nutrient deprivation activated Nupr1 expression, which we found to be required for cell survival. We found that Nupr1 protected cells from stress-induced death by inhibiting apoptosis through a pathway dependent on transcription factor RelB and immediate early response 3 (IER3). NUPR1, RELB, and IER3 proteins were coexpressed in mouse PanINs from Kras(G12D)-expressing pancreas. Moreover, pancreas-specific deletion of Relb in a Kras(G12D) background resulted in delayed in PanIN development associated with a lack of IER3 expression. Thus, efficient PanIN formation was dependent on the expression of Nupr1 and Relb, with likely involvement of IER3. Finally, in patients with PDAC, expression of NUPR1, RELB, and IER3 was significantly correlated with a poor prognosis. Cumulatively, these results reveal a NUPR1/RELB/IER3 stress-related pathway that is required for oncogenic Kras(G12D)-dependent transformation of the pancreas.
Collapse
Affiliation(s)
- Tewfik Hamidi
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM UMR 1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Marseille, France
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Zechner D, Spitzner M, Bobrowski A, Knapp N, Kuhla A, Vollmar B. Diabetes aggravates acute pancreatitis and inhibits pancreas regeneration in mice. Diabetologia 2012; 55:1526-34. [PMID: 22327285 DOI: 10.1007/s00125-012-2479-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2011] [Accepted: 01/11/2012] [Indexed: 12/18/2022]
Abstract
AIMS/HYPOTHESIS It is well established that acute pancreatitis often causes diabetes and that a high blood glucose level associated with pancreatitis is a marker of poor prognosis. The aim of this study was to evaluate if diabetes merely reflects the severity of pancreatitis or whether it can also aggravate the progression of this disease in a vicious circle. METHODS Reversible acute oedematous pancreatitis was induced in untreated and streptozotocin-treated diabetic mice by injection of cerulein. Progression of pancreatitis was studied by immunohistochemistry, ELISA and various other enzyme assays. The production of regenerating islet-derived 3β (REG3β) was determined by western blot and immunohistochemistry. RESULTS While cerulein treatment in non-diabetic mice resulted in acute pancreatitis followed by regeneration of the pancreas within 7 days, diabetes aggravated pancreatitis, inhibited the regeneration of the exocrine tissue and led to strong atrophy of the pancreas. The aggravation of pancreatitis by diabetes was characterised by decreased production of the anti-inflammatory protein REG3β, increased inflammation, augmented oedema formation and increased cell death during the acute phase of pancreatitis (p < 0.05). During the regenerative phase, diabetes augmented inflammation, increased cell death, reduced acinar cell expansion and increased the expansion of duct as well as interstitial cells, resulting in the formation of tubular complexes (p < 0.05). Administration of insulin reversed the observed phenotype in diabetic mice. CONCLUSIONS/INTERPRETATION Diabetes aggravates acute pancreatitis and suppresses regeneration of the exocrine tissue. Thus, diabetes is not just a concomitant phenomenon of pancreatitis, but can have a fundamental influence on the progression of acute pancreatitis.
Collapse
Affiliation(s)
- D Zechner
- Institute for Experimental Surgery, University of Rostock, Schillingallee 69a, 18057 Rostock, Germany
| | | | | | | | | | | |
Collapse
|
38
|
Escobar J, Pereda J, López-Rodas G, Sastre J. Redox signaling and histone acetylation in acute pancreatitis. Free Radic Biol Med 2012; 52:819-37. [PMID: 22178977 DOI: 10.1016/j.freeradbiomed.2011.11.009] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2011] [Revised: 11/08/2011] [Accepted: 11/09/2011] [Indexed: 12/22/2022]
Abstract
Histone acetylation via CBP/p300 coordinates the expression of proinflammatory cytokines in the activation phase of inflammation, particularly through mitogen-activated protein kinases (MAPKs), nuclear factor-κB (NF-κB), and signal transducers and activators of transcription (STAT) pathways. In contrast, histone deacetylases (HDACs) and protein phosphatases are mainly involved in the attenuation phase of inflammation. The role of reactive oxygen species (ROS) in the inflammatory cascade is much more important than expected. Mitochondrial ROS act as signal-transducing molecules that trigger proinflammatory cytokine production via inflammasome-independent and inflammasome-dependent pathways. The major source of ROS in acute inflammation seems to be NADPH oxidases, whereas NF-κB, protein phosphatases, and HDACs are the major targets of ROS and redox signaling in this process. There is a cross-talk between oxidative stress and proinflammatory cytokines through serine/threonine protein phosphatases, tyrosine protein phosphatases, and MAPKs that greatly contributes to amplification of the uncontrolled inflammatory cascade and tissue injury in acute pancreatitis. Chromatin remodeling during induction of proinflammatory genes would depend primarily on phosphorylation of transcription factors and their binding to gene promoters together with recruitment of histone acetyltransferases. PP2A should be considered a key modulator of the inflammatory cascade in acute pancreatitis through the ERK/NF-κB pathway and histone acetylation.
Collapse
Affiliation(s)
- Javier Escobar
- Department of Physiology, School of Pharmacy, University of Valencia, Burjasot, Valencia, Spain
| | | | | | | |
Collapse
|
39
|
Yang X, Jin H, Liu K, Gu Q, Xu X. A novel peptide derived from human pancreatitis-associated protein inhibits inflammation in vivo and in vitro and blocks NF-kappa B signaling pathway. PLoS One 2011; 6:e29155. [PMID: 22195011 PMCID: PMC3237597 DOI: 10.1371/journal.pone.0029155] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2011] [Accepted: 11/21/2011] [Indexed: 12/14/2022] Open
Abstract
Background Pancreatitis-associated protein (PAP) is a pancreatic secretory protein belongs to the group VII of C-type lectin family. Emerging evidence suggests that PAP plays a protective effect in inflammatory diseases. In the present study, we newly identified a 16-amino-acid peptide (named PAPep) derived from C-type lectin-like domain (CTLD) of human PAP with potent anti-inflammatory activity using both in vivo and in vitro assays. Methodology/Principal Findings We assessed the anti-inflammatory effect of PAPep on endotoxin-induced uveitis (EIU) in rats and demonstrated that intravitreal pretreatment of PAPep concentration-dependently attenuated clinical manifestation of EIU rats, reduced protein leakage and cell infiltration into the aqueous humor (AqH), suppressed tumor necrosis factor (TNF)-α, interleukin (IL)-6, intercellular adhesion molecule-1 (ICAM-1) and monocyte chemoattractant protein (MCP)-1 production in ocular tissues, and improved histopathologic manifestation of EIU. Furthermore, PAPep suppressed the LPS-induced mRNA expression of TNF-α and IL-6 in RAW 264.7 cells, inhibited protein expression of ICAM-1 in TNF-α-stimulated human umbilical vein endothelial cells (HUVECs) as well as U937 cells adhesion to HUVECs. Western blot analysis in ocular tissues and different cell lines revealed that the possible mechanism for this anti-inflammatory effect of PAPep may depend on its ability to inhibit the activation of NF-kB signaling pathway. Conclusions/Significance Our studies provide the first evidence that the sequence of PAPep is within the critically active region for the anti-inflammatory function of PAP and the peptide may be a promising candidate for the management of ocular inflammatory diseases.
Collapse
Affiliation(s)
- Xiaolu Yang
- Department of Ophthalmology, Shanghai First People's Hospital, Shanghai Jiaotong University, Shanghai, People's Republic of China
- Shanghai Key Laboratory of Fundus Disease, Shanghai, People's Republic of China
| | - Huiyi Jin
- Department of Ophthalmology, Shanghai First People's Hospital, Shanghai Jiaotong University, Shanghai, People's Republic of China
- Shanghai Key Laboratory of Fundus Disease, Shanghai, People's Republic of China
| | - Kun Liu
- Department of Ophthalmology, Shanghai First People's Hospital, Shanghai Jiaotong University, Shanghai, People's Republic of China
- Shanghai Key Laboratory of Fundus Disease, Shanghai, People's Republic of China
| | - Qing Gu
- Department of Ophthalmology, Shanghai First People's Hospital, Shanghai Jiaotong University, Shanghai, People's Republic of China
- Shanghai Key Laboratory of Fundus Disease, Shanghai, People's Republic of China
| | - Xun Xu
- Department of Ophthalmology, Shanghai First People's Hospital, Shanghai Jiaotong University, Shanghai, People's Republic of China
- Shanghai Key Laboratory of Fundus Disease, Shanghai, People's Republic of China
- * E-mail:
| |
Collapse
|
40
|
Parikh A, Stephan AF, Tzanakakis ES. Regenerating proteins and their expression, regulation and signaling. Biomol Concepts 2011; 3:57-70. [PMID: 22582090 DOI: 10.1515/bmc.2011.055] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The regenerating (Reg) protein family comprises C-type lectin-like proteins discovered independently during pancreatitis and pancreatic islet regeneration. However, an increasing number of studies provide evidence of participation of Reg proteins in the proliferation and differentiation of diverse cell types. Moreover, Reg family members are associated with various pathologies, including diabetes and forms of gastrointestinal cancer. These findings have led to the emergence of key roles for Reg proteins as anti-inflammatory, antiapoptotic and mitogenic agents in multiple physiologic and disease contexts. Yet, there are significant gaps in our knowledge regarding the regulation of expression of different Reg genes. In addition, the pathways relaying Reg-triggered signals, their targets and potential cross-talk with other cascades are still largely unknown. In this review, the expression patterns of different Reg members in the pancreas and extrapancreatic tissues are described. Moreover, factors known to modulate Reg levels in different cell types are discussed. Several signaling pathways, which have been implicated in conferring the effects of Reg ligands to date, are also delineated. Further efforts are necessary for elucidating the biological processes underlying the action of Reg proteins and their involvement in various maladies. Better understanding of the function of Reg genes and proteins will be beneficial in the design and development of therapies utilizing or targeting this protein group.
Collapse
Affiliation(s)
- Abhirath Parikh
- Department of Chemical and Biological Engineering, State University of New York at Buffalo, Buffalo, NY 14260
| | | | | |
Collapse
|
41
|
Pancreatitis-associated proteins' regulation of inflammation is correlated with their ability to aggregate. Pancreas 2011; 40:1151-3. [PMID: 21926556 DOI: 10.1097/mpa.0b013e3182218006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
|
42
|
Xu C, Zhang X, Wang G, Chang C, Zhang L, Cheng Q, Lu A. Role of the autonomic nervous system in rat liver regeneration. Cell Mol Neurobiol 2011; 31:527-40. [PMID: 21264506 DOI: 10.1007/s10571-011-9646-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2010] [Accepted: 01/04/2011] [Indexed: 01/12/2023]
Abstract
To study the regulatory role of autonomic nervous system in rat regenerating liver, surgical operations of rat partial hepatectomy (PH) and its operation control (OC), sympathectomy combining partial hepatectomy (SPH), vagotomy combining partial hepatectomy (VPH), and total liver denervation combining partial hepatectomy (TDPH) were performed, then expression profiles of regenerating livers at 2 h after operation were detected using Rat Genome 230 2.0 array. It was shown that the expressions of 97 genes in OC, 230 genes in PH, 253 genes in SPH, 187 genes in VPH, and 177 genes in TDPH were significantly changed in biology. The relevance analysis showed that in SPH, genes involved in stimulus response, immunity response, amino acids and K(+) transport, amino acid catabolism, cell adhesion, cell proliferation mediated by JAK-STAT, Ca(+), and platelet-derived growth factor receptor, cell growth and differentiation through JAK-STAT were up-regulated, while the genes involved in chromatin assembly and disassembly, and cell apoptosis mediated by MAPK were down-regulated. In VPH, the genes associated with chromosome modification-related transcription factor, oxygen transport, and cell apoptosis mediated by MAPK pathway were up-regulated, but the genes associated with amino acid catabolism, histone acetylation-related transcription factor, and cell differentiation mediated by Wnt pathway were down-regulated. In TDPH, the genes related to immunity response, growth and development of regenerating liver, cell growth by MAPK pathway were up-regulated. Our data suggested that splanchnic and vagal nerves could regulate the expressions of liver regeneration-related genes.
Collapse
Affiliation(s)
- Cunshuan Xu
- College of Life Science, Henan Normal University, No. 46, Construction East Road, Xinxiang, 453007, Henan, China.
| | | | | | | | | | | | | |
Collapse
|
43
|
Xiong X, Wang X, Li B, Chowdhury S, Lu Y, Srikant CB, Ning G, Liu JL. Pancreatic islet-specific overexpression of Reg3β protein induced the expression of pro-islet genes and protected the mice against streptozotocin-induced diabetes mellitus. Am J Physiol Endocrinol Metab 2011; 300:E669-80. [PMID: 21245462 DOI: 10.1152/ajpendo.00600.2010] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Reg family proteins have been implicated in islet β-cell proliferation, survival, and regeneration. The expression of Reg3β (pancreatitis-associated protein) is highly induced in experimental diabetes and acute pancreatitis, but its precise role has not been established. Through knockout studies, this protein was shown to be mitogenic, antiapoptotic, and anti-inflammatory in the liver and pancreatic acinars. To test whether it can promote islet cell growth or survival against experimental damage, we developed β-cell-specific overexpression using rat insulin I promoter, evaluated the changes in normal islet function, gene expression profile, and the response to streptozotocin-induced diabetes. Significant and specific overexpression of Reg3β was achieved in the pancreatic islets of RIP-I/Reg3β mice, which exhibited normal islet histology, β-cell mass, and in vivo and in vitro insulin secretion in response to high glucose yet were slightly hyperglycemic and low in islet GLUT2 level. Upon streptozotocin treatment, in contrast to wild-type littermates that became hyperglycemic in 3 days and lost 15% of their weight, RIP-I/Reg3β mice were significantly protected from hyperglycemia and weight loss. To identify specific targets affected by Reg3β overexpression, a whole genome DNA microarray on islet RNA isolated from the transgenic mice revealed more than 45 genes significantly either up- or downregulated. Among them, islet-protective osteopontin/SPP1 and acute responsive nuclear protein p8/NUPR1 were significantly induced, a result further confirmed by real-time PCR, Western blots, and immunohistochemistry. Our results suggest that Reg3β is unlikely an islet growth factor but a putative protector that prevents streptozotocin-induced damage by inducing the expression of specific genes.
Collapse
Affiliation(s)
- Xiaoquan Xiong
- Fraser Laboratories for Diabetes Research, Department of Medicine, McGill University Health Centre, Montreal, Quebec, Canada
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Tamizhselvi R, Shrivastava P, Koh YH, Zhang H, Bhatia M. Preprotachykinin-A gene deletion regulates hydrogen sulfide-induced toll-like receptor 4 signaling pathway in cerulein-treated pancreatic acinar cells. Pancreas 2011; 40:444-452. [PMID: 21289528 DOI: 10.1097/mpa.0b013e31820720e6] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVE This study aimed to determine the effect of hydrogen sulfide (H2S) on Toll-like receptor 4 (TLR4)-mediated innate immune signaling in acute pancreatitis (AP) via substance P. METHODS Male Swiss mice were treated with hourly intraperitoneal injections of cerulein (50 μg/kg) for 10 hours. dl-propargylglycine ([PAG] 100 mg/kg, intraperitoneally), an inhibitor of H2S formation, was administered 1 hour after the induction of AP. Pancreatic acinar cells from male preprotachykinin-A gene-knockout mice (PPTA) and their wild-type counterparts were incubated with or without cerulein (10 M for 60 minutes). To better understand the effect of H2S in inflammation, acinar cells were stimulated with cerulein after addition of H2S donor, sodium hydrosulfide. In addition, cerulein-treated pancreatic acinar cells were pretreated with PAG (30 μM) for 1 hour. RESULTS The H2S inhibitor PAG eliminated TLR4, interleukin 1 receptor-associated kinase 4, tumor necrosis factor receptor-associated factor 6, and nuclear factor-κB (NF-κB) levels in in vitro and in vivo models of cerulein-induced AP. PPTA gene deletion reduced TLR4, myeloid differentiation factor 88, interleukin 1 receptor-associated kinase 4, tumor necrosis factor receptor-associated factor 6, and NF-κB in cerulein-treated pancreatic acinar cells, whereas administration of sodium hydrosulfide resulted in a further rise in TLR4 and NF-κB levels in cerulein-treated pancreatic acinar cells. CONCLUSION The present findings show for the first time that in AP, H2S may up-regulate the TLR4 pathway and NF-κB via substance P.
Collapse
|
45
|
Kawahara S, Konishi H, Morino M, Ohata K, Kiyama H. Pancreatitis-associated protein-I and pancreatitis-associated protein-III expression in a rat model of kainic acid-induced seizure. Neuroscience 2010; 175:273-80. [PMID: 21093549 DOI: 10.1016/j.neuroscience.2010.11.020] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2010] [Revised: 11/11/2010] [Accepted: 11/12/2010] [Indexed: 11/30/2022]
Abstract
The pancreatitis-associated protein (PAP) family (also known as the regenerating gene (Reg) family) is a group of 16 kDa secretory proteins structurally classified as the calcium dependent-type lectin superfamily. Some PAP family members are expressed in neurons following peripheral nerve injury and traumatic brain injury. To determine whether PAP family members are expressed in non-traumatic brain injury, expressions were analyzed following kainic acid (KA)-induced seizure. PAP-I (also known as Reg2 in rat and RegIII-beta in mouse) and pancreatitis associated protein-III (PAP-III; RegIII-gamma in mouse) messenger ribonucleic acid (mRNA) was transiently expressed in some restricted areas, such as the hippocampus and parahippocampal area; expression was observed immediately at a maximal level 1 day after seizure. Expression disappeared within 3 days after seizure. In situ hybridization (ISH) and immunohistochemistry revealed neuronal PAP-I and PAP-III expression in the hippocampal dentate gyrus, perirhinal and entorhinal cortices, and the posterior cortical nucleus of the amygdala. The number of PAP-III mRNA-positive neurons was significantly greater than PAP-I mRNA-positive neurons. The majority of positive neurons co-localized with c-Jun, but not with glutamic acid decarboxylase (GAD). These results may suggest that PAP-I and PAP-III induction in non-GABAergic neurons would protect neurons against damage following seizure.
Collapse
Affiliation(s)
- S Kawahara
- Department of Anatomy and Neurobiology, Osaka City University, Graduate School of Medicine, Osaka 545-8585, Japan
| | | | | | | | | |
Collapse
|
46
|
Löhr JM, Faissner R, Koczan D, Bewerunge P, Bassi C, Brors B, Eils R, Frulloni L, Funk A, Halangk W, Jesenofsky R, Kaderali L, Kleeff J, Krüger B, Lerch MM, Lösel R, Magnani M, Neumaier M, Nittka S, Sahin-Tóth M, Sänger J, Serafini S, Schnölzer M, Thierse HJ, Wandschneider S, Zamboni G, Klöppel G, Klöppel G. Autoantibodies against the exocrine pancreas in autoimmune pancreatitis: gene and protein expression profiling and immunoassays identify pancreatic enzymes as a major target of the inflammatory process. Am J Gastroenterol 2010; 105:2060-71. [PMID: 20407433 PMCID: PMC3099227 DOI: 10.1038/ajg.2010.141] [Citation(s) in RCA: 102] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVES Autoimmune pancreatitis (AIP) is thought to be an immune-mediated inflammatory process, directed against the epithelial components of the pancreas. The objective was to identify novel markers of disease and to unravel the pathogenesis of AIP. METHODS To explore key targets of the inflammatory process, we analyzed the expression of proteins at the RNA and protein level using genomics and proteomics, immunohistochemistry, western blot, and immunoassay. An animal model of AIP with LP-BM5 murine leukemia virus-infected mice was studied in parallel. RNA microarrays of pancreatic tissue from 12 patients with AIP were compared with those of 8 patients with non-AIP chronic pancreatitis. RESULTS Expression profiling showed 272 upregulated genes, including those encoding for immunoglobulins, chemokines and their receptors, and 86 downregulated genes, including those for pancreatic proteases such as three trypsinogen isoforms. Protein profiling showed that the expression of trypsinogens and other pancreatic enzymes was greatly reduced. Immunohistochemistry showed a near-loss of trypsin-positive acinar cells, which was also confirmed by western blotting. The serum of AIP patients contained high titers of autoantibodies against the trypsinogens PRSS1 and PRSS2 but not against PRSS3. In addition, there were autoantibodies against the trypsin inhibitor PSTI (the product of the SPINK1 gene). In the pancreas of AIP animals, we found similar protein patterns and a reduction in trypsinogen. CONCLUSIONS These data indicate that the immune-mediated process characterizing AIP involves pancreatic acinar cells and their secretory enzymes such as trypsin isoforms. Demonstration of trypsinogen autoantibodies may be helpful for the diagnosis of AIP.
Collapse
Affiliation(s)
- J.-Matthias Löhr
- Molecular Gastroenterology, German Cancer Research Center, Heidelberg, Germany,Department of Medicine II, University of Heidelberg, Germany,Department of Surgical Gastroenterology, Karolinska Institute, Stockholm, Sweden,Author for correspondence: Matthias Löhr, MD Professor of Gastroenterology & Hepatology Karolinska Institutet CLINTEC, K53 Dept. of Surgical Gastroenterology Hälsovägen SE-141 86 Stockholm Phone: +46 8 5858-2431 Fax: +46 8 5858-2340
| | - Ralf Faissner
- Molecular Gastroenterology, German Cancer Research Center, Heidelberg, Germany
| | - Dirk Koczan
- Department of Immunology, University of Rostock, Germany
| | - Peter Bewerunge
- Department of Theoretical Bioinformatics, German Cancer Research Center, Heidelberg, Germany
| | - Claudio Bassi
- Department of Biomedical and Surgical Sciences, University of Verona, Italy
| | - Benedikt Brors
- Department of Theoretical Bioinformatics, German Cancer Research Center, Heidelberg, Germany
| | - Roland Eils
- Department of Theoretical Bioinformatics, German Cancer Research Center, Heidelberg, Germany
| | - Luca Frulloni
- Department of Biomedical and Surgical Sciences, University of Verona, Italy
| | - Anette Funk
- Molecular Gastroenterology, German Cancer Research Center, Heidelberg, Germany,Department of Medicine II, University of Heidelberg, Germany
| | | | - Ralf Jesenofsky
- Molecular Gastroenterology, German Cancer Research Center, Heidelberg, Germany
| | - Lars Kaderali
- Department of Theoretical Bioinformatics, German Cancer Research Center, Heidelberg, Germany
| | - Jörg Kleeff
- Department of Surgery, University of Heidelberg, Germany
| | | | | | - Ralf Lösel
- Department of Clinical Pharmacology, University of Heidelberg, Germany
| | - Mauro Magnani
- Institute of Biological Chemistry, University of Urbino, Italy
| | - Michael Neumaier
- Department of Clinical Chemistry, University of Heidelberg, Germany
| | - Stephanie Nittka
- Department of Clinical Chemistry, University of Heidelberg, Germany
| | - Miklós Sahin-Tóth
- Department of Molecular and Cell Biology, Boston University Henry M. Goldman School of Dental Medicine, Massachusetts, USA
| | - Julian Sänger
- Molecular Gastroenterology, German Cancer Research Center, Heidelberg, Germany
| | - Sonja Serafini
- Institute of Biological Chemistry, University of Urbino, Italy
| | - Martina Schnölzer
- Functional Proteome Analysis, German Cancer Research Center, Heidelberg, Germany
| | - Hermann-Josef Thierse
- Department of Dermatology, Medical Faculty Mannheim, University of Heidelberg, Germany
| | - Silke Wandschneider
- Molecular Gastroenterology, German Cancer Research Center, Heidelberg, Germany,Functional Proteome Analysis, German Cancer Research Center, Heidelberg, Germany
| | | | | | | |
Collapse
|
47
|
Xu YH, Chen K, Cui SL, Wang H. Advances in understanding the role of inflammatory and antiinflammatory factors in the pathogenesis of acute pancreatitis. Shijie Huaren Xiaohua Zazhi 2010; 18:1912-1918. [DOI: 10.11569/wcjd.v18.i18.1912] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [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
Acute pancreatitis (AP) is a common clinical disorder presenting as acute abdomen. As AP is often complicated with many other serious diseases, the overall mortality rate is high in AP patients. At present, the research on the pathogenesis of AP has attracted wide attention though it has not been fully clarified yet. Many theories, such as "self-digestion of the pancreas", "inflammatory response" and "intestinal bacterial translocation", have been proposed to explain the pathogenesis of AP. Extensive research is being conducted to investigate the relationship of inflammatory and antiinflammatory factors with AP. In this article, we will review the recent advances in understanding the role of inflammatory and antiinflammatory factors in the pathogenesis of AP.
Collapse
|
48
|
Cloning and characterization of p8 homolog cDNA in the Atlantic halibut (Hippoglossus hippoglossus). Biochem Genet 2010; 48:504-15. [PMID: 20454949 DOI: 10.1007/s10528-010-9334-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2009] [Accepted: 01/25/2010] [Indexed: 10/19/2022]
Abstract
The p8 gene encodes a transcription factor known to modulate cell growth, division, and apoptosis and influences gene expression. In this study, an Atlantic halibut (Hippoglossus hippoglossus) homolog of the p8 gene was cloned, sequenced, and characterized. The full-length p8 cDNA consists of 601 bp and encodes 76 amino acids with a molecular mass of 9 kD. The bHLH region is well conserved between Atlantic halibut and other animals. Analysis by RT-PCR showed that the p8 transcript is constitutively expressed in 9 of the 12 tissues tested: pancreas, intestine, stomach, gill, head kidney, heart, liver, ovary, and spleen. A predicted microRNA target site was found in the 3'UTR of Atlantic halibut p8 mRNA. We speculate that the target site may pair to microRNA molecules because the target site resides in a big loop, a space large enough for the binding of microRNA molecules.
Collapse
|
49
|
He SQ, Yao JR, Zhang FX, Wang Q, Bao L, Zhang X. Inflammation and nerve injury induce expression of pancreatitis-associated protein-II in primary sensory neurons. Mol Pain 2010; 6:23. [PMID: 20420691 PMCID: PMC2873504 DOI: 10.1186/1744-8069-6-23] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2010] [Accepted: 04/26/2010] [Indexed: 01/18/2023] Open
Abstract
Pancreatitis-associated protein (PAP)-I and -II, lectin-related secretory proteins, are members of the regenerating gene (Reg) family. Although expression of PAP-I was found in the dorsal root ganglion (DRG) neurons following peripheral nerve injury and cystitis, whether PAP-II could be expressed in DRG neurons in chronic pain models remains unclear. The present study shows an inflammation- and nerve injury-triggered expression of PAP-II in rat DRG neurons. In situ hybridization showed that only a few DRG neurons normally contained PAP-I and -II mRNAs. After peripheral inflammation, PAP-I and -II mRNAs were present in over half of small DRG neurons. Such an elevated expression of PAP-I and -II reached the peak level on the second day. Immunostaining showed that the expression of PAP-II was mostly increased in the isolectin B4-positive subset of small DRG neurons after inflammation. Furthermore, the expression of PAP-II was also induced in DRG neurons after peripheral nerve injury. Interestingly, PAP-II expression was shifted from small neurons on day 2 to large DRG neurons that expressed neuropeptide Y during the later post-injury days. These results suggest that PAP-II may play potential roles in the modulation of spinal sensory pathways in pathological pain states.
Collapse
Affiliation(s)
- Shao-Qiu He
- Institute of Neuroscience and State Key Laboratory of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai 200031, China
| | | | | | | | | | | |
Collapse
|
50
|
Ampo KI, Suzuki A, Konishi H, Kiyama H. Induction of pancreatitis-associated protein (PAP) family members in neurons after traumatic brain injury. J Neurotrauma 2010; 26:1683-93. [PMID: 19351265 DOI: 10.1089/neu.2008.0847] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
The pancreatitis-associated protein (PAP) family is a group of 16-kDa secretory proteins initially identified in the pancreas in rats with acute pancreatitis. Although induction of PAP family genes was reported in peripheral nerve injury models, the expression in the central nervous system after traumatic injury has not been examined. In the present study, we examined the expression of PAP family members (PAP-I, PAP-II, and PAP-III) in the rat brain following traumatic brain injury (TBI) induced by weight drop. There was a significant upregulation of PAP-I and PAP-III mRNA in the injured cortex beginning at 1 day after TBI. Immunohistochemical double-staining indicated that PAP-I and PAP-III staining was localized in a subpopulation of neurons in the peri-injured region. Expression of both PAP-I and PAP-III mRNA was observed following a transient increase in inflammatory cytokines, including TNF-alpha, IL-6, and IL-1beta mRNA. The results of the present study suggest that expression of PAP family members in response to traumatic and inflammatory stimuli are not restricted to the pancreas, intestine, and peripheral nervous system, and are likely a more general cellular response, including the central nervous system in the rat. Thus, PAP family members may have an anti-inflammatory role, and this may contribute to the protection of injured neurons.
Collapse
Affiliation(s)
- Kei-Ichi Ampo
- Department of Anatomy and Neurobiology, Osaka City University, Graduate School of Medicine, Abeno-ku, Osaka, Japan
| | | | | | | |
Collapse
|