151
|
Woodland DC, Liu W, Leong J, Sears ML, Luo P, Chen X. Short-term high-fat feeding induces islet macrophage infiltration and β-cell replication independently of insulin resistance in mice. Am J Physiol Endocrinol Metab 2016; 311:E763-E771. [PMID: 27577853 PMCID: PMC5241555 DOI: 10.1152/ajpendo.00092.2016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 08/22/2016] [Indexed: 02/06/2023]
Abstract
Short-term high-fat consumption stimulates mouse islet β-cell replication through unknown mechanisms. Resident macrophages (MΦs) are capable of secreting various factors involved in islet development and tissue remodeling. We hypothesized that a short-term high-fat diet (HFD) promotes MΦ infiltration in pancreatic islets and that MΦs serve as a regulator of β-cell replication. To test these hypotheses and dissect mechanisms involved in HFD-induced β-cell replication, adult C57BL/6J mice were fed a HFD for 7 days with or without administration of clodronate-containing liposomes, an MΦ-depleting agent. Mouse body and epididymal fat pad weights, and nonfasting blood glucose and fasting serum insulin levels were measured, and pancreatic islet β-cell replication, oxidative stress, and MΦ infiltration were examined. Short-term HFD promoted an increase in body and epididymal fat pad weight and blood glucose levels, along with an increased fasting serum insulin concentration. β-Cell replication, islet MΦ infiltration, and the percentage of inducible NO synthase positive MΦs in the islets increased significantly in mice fed the HFD. Immunofluorescence staining for 8-oxo-2'-deoxyguanosine or activated caspase-3 revealed no significant induction of DNA damage or apoptosis, respectively. In addition, no change in stromal-derived factor 1-expressing cells was found induced by HFD. Despite continuous elevation of nonfasting blood glucose and fasting serum insulin levels, depletion of MΦs through treatments of clodronate abrogated HFD-induced β-cell replication. These findings demonstrated that HFD-induced MΦ infiltration is responsible for β-cell replication. This study suggests the existence of MΦ-mediated mechanisms in β-cell replication that are independent of insulin resistance.
Collapse
Affiliation(s)
- David C Woodland
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, New York; Department of Surgery, Columbia University Medical Center, New York, New York
| | - Wei Liu
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, New York; Department of Surgery, Columbia University Medical Center, New York, New York; The Second Clinical Medicine College, Jilin University, Changchun, Jilin Province, China
| | - Jacky Leong
- Touro College of Osteopathic Medicine, New York, New York; and
| | - Mallory L Sears
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, New York; Department of Surgery, Columbia University Medical Center, New York, New York
| | - Ping Luo
- The Second Clinical Medicine College, Jilin University, Changchun, Jilin Province, China
| | - Xiaojuan Chen
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, New York; Department of Surgery, Columbia University Medical Center, New York, New York;
| |
Collapse
|
152
|
Prasadan K, Shiota C, Xiangwei X, Ricks D, Fusco J, Gittes G. A synopsis of factors regulating beta cell development and beta cell mass. Cell Mol Life Sci 2016; 73:3623-37. [PMID: 27105622 PMCID: PMC5002366 DOI: 10.1007/s00018-016-2231-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 03/24/2016] [Accepted: 04/14/2016] [Indexed: 12/29/2022]
Abstract
The insulin-secreting beta cells in the endocrine pancreas regulate blood glucose levels, and loss of functional beta cells leads to insulin deficiency, hyperglycemia (high blood glucose) and diabetes mellitus. Current treatment strategies for type-1 (autoimmune) diabetes are islet transplantation, which has significant risks and limitations, or normalization of blood glucose with insulin injections, which is clearly not ideal. The type-1 patients can lack insulin counter-regulatory mechanism; therefore, hypoglycemia is a potential risk. Hence, a cell-based therapy offers a better alternative for the treatment of diabetes. Past research was focused on attempting to generate replacement beta cells from stem cells; however, recently there has been an increasing interest in identifying mechanisms that will lead to the conversion of pre-existing differentiated endocrine cells into beta cells. The goal of this review is to provide an overview of several of the key factors that regulate new beta cell formation (neogenesis) and beta cell proliferation.
Collapse
Affiliation(s)
- Krishna Prasadan
- Rangos Research Center, Children's Hospital of University of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA, 15224, USA
| | - Chiyo Shiota
- Rangos Research Center, Children's Hospital of University of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA, 15224, USA
| | - Xiao Xiangwei
- Rangos Research Center, Children's Hospital of University of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA, 15224, USA
| | - David Ricks
- Rangos Research Center, Children's Hospital of University of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA, 15224, USA
| | - Joseph Fusco
- Rangos Research Center, Children's Hospital of University of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA, 15224, USA
| | - George Gittes
- Rangos Research Center, Children's Hospital of University of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA, 15224, USA.
| |
Collapse
|
153
|
De Groef S, Staels W, Van Gassen N, Lemper M, Yuchi Y, Sojoodi M, Bussche L, Heremans Y, Leuckx G, De Leu N, Van de Casteele M, Baeyens L, Heimberg H. Sources of beta cells inside the pancreas. Diabetologia 2016; 59:1834-7. [PMID: 27053238 DOI: 10.1007/s00125-016-3879-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Accepted: 12/16/2015] [Indexed: 11/29/2022]
Abstract
The generation of beta(-like) cells to compensate for their absolute or relative shortage in type 1 and type 2 diabetes is an obvious therapeutic strategy. Patients first received grafts of donor islet cells over 25 years ago, but this procedure has not become routine in clinical practice because of a donor cell shortage and (auto)immune problems. Transplantation of differentiated embryonic and induced pluripotent stem cells may overcome some but not all the current limitations. Reprogramming exocrine cells towards functional beta(-like) cells would offer an alternative abundant and autologous source of beta(-like) cells. This review focuses on work by our research group towards achieving such a source of cells. It summarises a presentation given at the 'Can we make a better beta cell?' symposium at the 2015 annual meeting of the EASD. It is accompanied by two other reviews on topics from this symposium (by Amin Ardestani and Kathrin Maedler, DOI: 10.1007/s00125-016-3892-9 , and by Heiko Lickert and colleagues, DOI: 10.1007/s00125-016-3949-9 ) and a commentary by the Session Chair, Shanta Persaud (DOI: 10.1007/s00125-016-3870-2 ).
Collapse
Affiliation(s)
- Sofie De Groef
- Diabetes Research Center, Vrije Universiteit Brussel, Laarbeeklaan 103, B1090, Brussels, Belgium
| | - Willem Staels
- Diabetes Research Center, Vrije Universiteit Brussel, Laarbeeklaan 103, B1090, Brussels, Belgium
| | - Naomi Van Gassen
- Diabetes Research Center, Vrije Universiteit Brussel, Laarbeeklaan 103, B1090, Brussels, Belgium
| | - Marie Lemper
- Diabetes Research Center, Vrije Universiteit Brussel, Laarbeeklaan 103, B1090, Brussels, Belgium
| | - Yixing Yuchi
- Diabetes Research Center, Vrije Universiteit Brussel, Laarbeeklaan 103, B1090, Brussels, Belgium
| | - Mozhdeh Sojoodi
- Diabetes Research Center, Vrije Universiteit Brussel, Laarbeeklaan 103, B1090, Brussels, Belgium
| | - Leen Bussche
- Diabetes Research Center, Vrije Universiteit Brussel, Laarbeeklaan 103, B1090, Brussels, Belgium
| | - Yves Heremans
- Diabetes Research Center, Vrije Universiteit Brussel, Laarbeeklaan 103, B1090, Brussels, Belgium
| | - Gunter Leuckx
- Diabetes Research Center, Vrije Universiteit Brussel, Laarbeeklaan 103, B1090, Brussels, Belgium
| | - Nico De Leu
- Diabetes Research Center, Vrije Universiteit Brussel, Laarbeeklaan 103, B1090, Brussels, Belgium
| | - Mark Van de Casteele
- Diabetes Research Center, Vrije Universiteit Brussel, Laarbeeklaan 103, B1090, Brussels, Belgium
| | - Luc Baeyens
- Diabetes Research Center, Vrije Universiteit Brussel, Laarbeeklaan 103, B1090, Brussels, Belgium
| | - Harry Heimberg
- Diabetes Research Center, Vrije Universiteit Brussel, Laarbeeklaan 103, B1090, Brussels, Belgium.
| |
Collapse
|
154
|
Staels W, De Groef S, Bussche L, Leuckx G, Van de Casteele M, De Leu N, Baeyens L, Heremans Y, Heimberg H. Making β(-like)-cells from exocrine pancreas. Diabetes Obes Metab 2016; 18 Suppl 1:144-51. [PMID: 27615144 DOI: 10.1111/dom.12725] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 04/26/2016] [Indexed: 12/13/2022]
Abstract
Creating an abundant source of β(-like)-cells has been a major goal in diabetes research for many decades. The concept of cell plasticity has inspired many strategies towards regenerative medicine, but its successes have been limited until very recently. Today, most cell types in the pancreas are considered candidates for the generation of β(-like)-cells through transdifferentiation. While β(-like)-cells that are in vitro differentiated from human embryonic stem cells are already being grafted in patients, β(-like)-cells generated by transdifferentiation are not yet ready for clinical application. These cells would however offer several advantages over the current β(-like)-cells generated by directed differentiation, especially concerning safety issues. In addition, perfect control of the transdifferentiation efficiency would through targeted drug delivery support a non-invasive cell therapy for diabetes. Lastly, focusing on the exocrine pancreas as prime candidate makes sense in view of their abundance and high plasticity. Keeping these hopeful perspectives in mind, it is worth to continue focused research on the mechanisms that control transdifferentiation from pancreas exocrine to β-cells.
Collapse
Affiliation(s)
- W Staels
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
- Department of Pediatrics, Division of Pediatric Endocrinology, Ghent University Hospital and Department of Pediatrics and Genetics, Ghent University, Ghent, Belgium
| | - S De Groef
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - L Bussche
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - G Leuckx
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - M Van de Casteele
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - N De Leu
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
- Departments of Endocrinology, UZ Brussel, Brussels, Belgium
- ASZ Aalst, Aalst, Belgium
| | - L Baeyens
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Y Heremans
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - H Heimberg
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium.
| |
Collapse
|
155
|
Derr A, Yang C, Zilionis R, Sergushichev A, Blodgett DM, Redick S, Bortell R, Luban J, Harlan DM, Kadener S, Greiner DL, Klein A, Artyomov MN, Garber M. End Sequence Analysis Toolkit (ESAT) expands the extractable information from single-cell RNA-seq data. Genome Res 2016; 26:1397-1410. [PMID: 27470110 PMCID: PMC5052061 DOI: 10.1101/gr.207902.116] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 07/27/2016] [Indexed: 12/27/2022]
Abstract
RNA-seq protocols that focus on transcript termini are well suited for applications in which template quantity is limiting. Here we show that, when applied to end-sequencing data, analytical methods designed for global RNA-seq produce computational artifacts. To remedy this, we created the End Sequence Analysis Toolkit (ESAT). As a test, we first compared end-sequencing and bulk RNA-seq using RNA from dendritic cells stimulated with lipopolysaccharide (LPS). As predicted by the telescripting model for transcriptional bursts, ESAT detected an LPS-stimulated shift to shorter 3′-isoforms that was not evident by conventional computational methods. Then, droplet-based microfluidics was used to generate 1000 cDNA libraries, each from an individual pancreatic islet cell. ESAT identified nine distinct cell types, three distinct β-cell types, and a complex interplay between hormone secretion and vascularization. ESAT, then, offers a much-needed and generally applicable computational pipeline for either bulk or single-cell RNA end-sequencing.
Collapse
Affiliation(s)
- Alan Derr
- Program in Bioinformatics and Integrative Biology, University of Massachusetts Medical School, Worcester, Massachusetts 01655, USA
| | - Chaoxing Yang
- Program in Molecular Medicine, Diabetes Center of Excellence, University of Massachusetts Medical School, Worcester, Massachusetts 01655, USA
| | - Rapolas Zilionis
- Department of System Biology, Harvard Medical School, Boston, Massachusetts 02115, USA; Institute of Biotechnology, Vilnius University, LT 02241 Vilnius, Lithuania
| | - Alexey Sergushichev
- Computer Technologies Department, ITMO University, Saint Petersburg, 197101, Russia; Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, Missouri 63110, USA
| | - David M Blodgett
- Department of Medicine, Diabetes Center of Excellence, University of Massachusetts Medical School, Worcester, Massachusetts 01655, USA
| | - Sambra Redick
- Program in Molecular Medicine, Diabetes Center of Excellence, University of Massachusetts Medical School, Worcester, Massachusetts 01655, USA
| | - Rita Bortell
- Program in Molecular Medicine, Diabetes Center of Excellence, University of Massachusetts Medical School, Worcester, Massachusetts 01655, USA
| | - Jeremy Luban
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01655, USA
| | - David M Harlan
- Department of Medicine, Diabetes Center of Excellence, University of Massachusetts Medical School, Worcester, Massachusetts 01655, USA
| | - Sebastian Kadener
- Biological Chemistry Department, Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, 91904, Israel
| | - Dale L Greiner
- Program in Molecular Medicine, Diabetes Center of Excellence, University of Massachusetts Medical School, Worcester, Massachusetts 01655, USA; Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01655, USA
| | - Allon Klein
- Department of System Biology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Maxim N Artyomov
- Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, Missouri 63110, USA
| | - Manuel Garber
- Program in Bioinformatics and Integrative Biology, University of Massachusetts Medical School, Worcester, Massachusetts 01655, USA; Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01655, USA
| |
Collapse
|
156
|
Hu J, Xu JF, Ge WL. MiR-497 enhances metastasis of oral squamous cell carcinoma through SMAD7 suppression. Am J Transl Res 2016; 8:3023-3031. [PMID: 27508022 PMCID: PMC4969438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2016] [Accepted: 02/29/2016] [Indexed: 06/06/2023]
Abstract
SMAD7 is a key inhibitor of transforming growth factor β (TGFβ) receptor signaling, which regulates the alteration of cancer cell invasiveness through epithelial-mesenchymal cell conversion. Since microRNAs (miRNAs) play a potential role in the tumorigenesis, cancer cell growth and metastases of oral squamous cell carcinoma (OSCC), determination of the involved miRNAs that may regulate SMAD7-mediated OSCC cell invasion appears to be one important question. Here, we found that the levels of miR-497 were significantly increased and the levels of SMAD7 were significantly decreased in OSCC specimens, compared to the paired adjacent non-tumor tissue. Moreover, miR-497 and SMAD7 inversely correlated in OSCC specimens. The 5-year survival of the patients with higher miR-497 levels in the resected OSCC was worse than those high miR-497 levels. Bioinformatics analyses showed that miR-497 targeted the 3'-UTR of SMAD7 mRNA to inhibit its translation, which was proved by luciferase reporter assay. Furthermore, miR-497 overexpression increased SMAD7-suppressed cell invasion, while miR-497 depletion decreased SMAD7-suppressed cell invasion in OSCC cells, in both a transwell cell invasion assay and a scratch would healing assay. Together, our data suggest that suppression of miR-497 in OSCC cells may promote cancer cell invasion via suppression of SMAD7, and highlight miR-497 as an intriguing therapeutic target to prevent OSCC metastases.
Collapse
Affiliation(s)
- Jun Hu
- Department of Prosthodontics, Hospital of Stomatology Affiliated to Zhejiang UniversityHangzhou 310006, China
| | - Jun-Feng Xu
- Department of Stomatology, Tongde Hospital of Zhejiang ProvinceHangzhou 310012, China
| | - Wei-Li Ge
- Department of Maxillofacial Surgery, Hospital of Stomatology Affiliated to Zhejiang UniversityHangzhou 310006, China
| |
Collapse
|
157
|
Yan X, Cen Y, Wang Q. Mesenchymal stem cells alleviate experimental rheumatoid arthritis through microRNA-regulated IκB expression. Sci Rep 2016; 6:28915. [PMID: 27354158 PMCID: PMC4926108 DOI: 10.1038/srep28915] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 06/10/2016] [Indexed: 02/08/2023] Open
Abstract
Previous studies have demonstrated that mesenchymal stem cell (MSC) transplantation reduces the severity of collagen-induced arthritis (CIA) in mice, which is a model for rheumatoid arthritis (RA) in humans. However, the underlying molecular mechanisms remain ill-defined. Here, we showed that MSC transplantation reduced the activities of NF-κB signaling and decreased microRNA-548e (miR-548e) levels in the joint tissue in CIA-mice, seemingly through activation of transforming growth factor β receptor signaling. Bioinformatics analyses revealed that miR-548e inhibited protein translation of the NF-κB inhibitor, IκB, through binding to the 3′-UTR of the IκB mRNA. MSCs co-transplanted with adeno-associated virus (AAV) carrying miR-548e abolished the therapeutic effects of MSCs on CIA. On the other hand, transplantation of AAV carrying antisense of miR-548e (as-miR-548e) partially mimicked the effects of MSC transplantation on CIA. Together, these data suggest that MSC transplantation may alleviate experimental RA partially through suppressing miR-548e-mediated IκB inhibition.
Collapse
Affiliation(s)
- Xin Yan
- Department of Rheumatology, Shanxi University affiliated the First Hospital, Taiyuan 030001, China
| | - Yurong Cen
- Department of Nephrology and Rheumatology, Shanghai Jiaotong University affiliated Sixth People's Hospital, South Campus, Shanghai 201400, China
| | - Qin Wang
- Department of Nephrology and Rheumatology, Shanghai Jiaotong University affiliated Sixth People's Hospital, South Campus, Shanghai 201400, China.,Department of Nephrology and Rheumatology, Nanfang Medical University affiliated Fengxian Hospital, South Campus, Shanghai 201400, China
| |
Collapse
|
158
|
Saunders D, Powers AC. Replicative capacity of β-cells and type 1 diabetes. J Autoimmun 2016; 71:59-68. [PMID: 27133598 DOI: 10.1016/j.jaut.2016.03.014] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Accepted: 03/28/2016] [Indexed: 12/16/2022]
Abstract
Efforts to restore β-cell number or mass in type 1 diabetes (T1D) must combine an intervention to stimulate proliferation of remaining β-cells and an intervention to mitigate or control the β-cell-directed autoimmunity. This review highlights features of the β-cell, including it being part of a pancreatic islet, a mini-organ that is highly vascularized and highly innervated, and efforts to promote β-cell proliferation. In addition, the β-cell in T1D exists in a microenvironment with interactions and input from other islet cell types, extracellular matrix, vascular endothelial cells, neuronal projections, and immune cells, all of which likely influence the β-cell's capacity for replication. Physiologic β-cell proliferation occurs in human and rodents in the neonatal period and early in life, after which there is an age-dependent decline in β-cell proliferation, and also as part of the β-cell's compensatory response to the metabolic challenges of pregnancy and insulin resistance. This review reviews the molecular pathways involved in this β-cell proliferation and highlights recent work in two areas: 1) Investigators, using high-throughput screening to discover small molecules that promote human β-cell proliferation, are now focusing on the dual-specificity tyrosine-regulated kinase-1a and cell cycle-dependent kinase inhibitors CDKN2C/p18 or CDKN1A/p21as targets of compounds to stimulate adult human β-cell proliferation. 2) Local inflammation, macrophages, and the local β-cell microenvironment promote β-cell proliferation. Future efforts to harness the responsible mechanisms may lead to new approaches to promote β-cell proliferation in T1D.
Collapse
Affiliation(s)
- Diane Saunders
- Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Alvin C Powers
- Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, TN, United States; Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States; VA Tennessee Valley Healthcare System, Nashville, TN, United States.
| |
Collapse
|
159
|
Sun X, Deng QF, Liang ZF, Zhang ZQ, Zhao L, Geng H, Xie DD, Wang Y, Yu DX, Zhong CY. Curcumin reverses benzidine-induced cell proliferation by suppressing ERK1/2 pathway in human bladder cancer T24 cells. EXPERIMENTAL AND TOXICOLOGIC PATHOLOGY : OFFICIAL JOURNAL OF THE GESELLSCHAFT FUR TOXIKOLOGISCHE PATHOLOGIE 2016; 68:215-222. [PMID: 26776764 DOI: 10.1016/j.etp.2015.12.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Revised: 12/01/2015] [Accepted: 12/21/2015] [Indexed: 02/07/2023]
Abstract
Bladder cancer is one of the leading causes of cancer-related death in the world. Prolonged exposure to benzidine is a known cause of bladder cancer. Curcumin has been clinically used in chemoprevention and treatment of cancer. However, it remains unknown whether mitogen-activated protein kinase (MAPK) pathways are involved in curcumin-mediated protection from benzidine-associated promotive effects on bladder cancer. In our study, we found that benzidine increased the proliferation of human bladder cancer T24 cells, triggered transition of the cells from G1 to S phase, elevated the expression of cyclin D1 and proliferating cell nuclear antigen (PCNA) and decreased p21 expression. Meanwhile, exposure of T24 cells to benzidine resulted in activation of extracellular regulated protein kinases 1 and 2 (ERK1/2) pathway as well as activator protein 1 (AP-1) proteins. Treatment with ERK1/2 inhibitor U0126 or curcumin effectively abrogated benzidine-triggered cell proliferation and ERK1/2/AP-1 activation. These results suggested for the first time that curcumin in low concentrations played a protective role in benzidine-induced ERK1/2/AP-1 activation and proliferation of bladder cancer cells, therefore providing new insights into the pathogenesis and chemoprevention of benzidine-associated bladder cancer.
Collapse
Affiliation(s)
- Xin Sun
- Department of Urology, the Second Affiliated Hospital of Anhui Medical University, Hefei 230032, China
| | - Qi-Fei Deng
- Department of Urology, the Second Affiliated Hospital of Anhui Medical University, Hefei 230032, China
| | - Zhao-Feng Liang
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Zhi-Qiang Zhang
- Department of Urology, the Second Affiliated Hospital of Anhui Medical University, Hefei 230032, China
| | - Li Zhao
- Department of Urology, the Second Affiliated Hospital of Anhui Medical University, Hefei 230032, China
| | - Hao Geng
- Department of Urology, the Second Affiliated Hospital of Anhui Medical University, Hefei 230032, China
| | - Dong-Dong Xie
- Department of Urology, the Second Affiliated Hospital of Anhui Medical University, Hefei 230032, China
| | - Yi Wang
- Department of Urology, the Second Affiliated Hospital of Anhui Medical University, Hefei 230032, China
| | - De-Xin Yu
- Department of Urology, the Second Affiliated Hospital of Anhui Medical University, Hefei 230032, China.
| | - Cai-Yun Zhong
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing 211166, China.
| |
Collapse
|
160
|
Xiao X, Fischbach S, Song Z, Gaffar I, Zimmerman R, Wiersch J, Prasadan K, Shiota C, Guo P, Ramachandran S, Witkowski P, Gittes GK. Transient Suppression of TGFβ Receptor Signaling Facilitates Human Islet Transplantation. Endocrinology 2016; 157:1348-1356. [PMID: 26872091 PMCID: PMC4816736 DOI: 10.1210/en.2015-1986] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 02/09/2016] [Indexed: 11/19/2022]
Abstract
Although islet transplantation is an effective treatment for severe diabetes, its broad application is greatly limited due to a shortage of donor islets. Suppression of TGFβ receptor signaling in β-cells has been shown to increase β-cell proliferation in mice, but has not been rigorously examined in humans. Here, treatment of human islets with a TGFβ receptor I inhibitor, SB-431542 (SB), significantly improved C-peptide secretion by β-cells, and significantly increased β-cell number by increasing β-cell proliferation. In addition, SB increased cell-cycle activators and decreased cell-cycle suppressors in human β-cells. Transplantation of SB-treated human islets into diabetic immune-deficient mice resulted in significant improvement in blood glucose control, significantly higher serum and graft insulin content, and significantly greater increases in β-cell proliferation in the graft, compared with controls. Thus, our data suggest that transient suppression of TGFβ receptor signaling may improve the outcome of human islet transplantation, seemingly through increasing β-cell number and function.
Collapse
Affiliation(s)
| | | | | | | | - Ray Zimmerman
- Division of Pediatric Surgery (X.X., S.F., Z.S., I.G., R.Z., J.W., K.P., C.S., P.G., G.K.G.), Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224; Department of General Surgery (Z.S.), The Third Xiangya Hospital of Central S University, Changsha 410013, China; and Department of Surgery (S.R., P.W.), University of Chicago, Chicago, Illinois 60637
| | - John Wiersch
- Division of Pediatric Surgery (X.X., S.F., Z.S., I.G., R.Z., J.W., K.P., C.S., P.G., G.K.G.), Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224; Department of General Surgery (Z.S.), The Third Xiangya Hospital of Central S University, Changsha 410013, China; and Department of Surgery (S.R., P.W.), University of Chicago, Chicago, Illinois 60637
| | - Krishna Prasadan
- Division of Pediatric Surgery (X.X., S.F., Z.S., I.G., R.Z., J.W., K.P., C.S., P.G., G.K.G.), Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224; Department of General Surgery (Z.S.), The Third Xiangya Hospital of Central S University, Changsha 410013, China; and Department of Surgery (S.R., P.W.), University of Chicago, Chicago, Illinois 60637
| | - Chiyo Shiota
- Division of Pediatric Surgery (X.X., S.F., Z.S., I.G., R.Z., J.W., K.P., C.S., P.G., G.K.G.), Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224; Department of General Surgery (Z.S.), The Third Xiangya Hospital of Central S University, Changsha 410013, China; and Department of Surgery (S.R., P.W.), University of Chicago, Chicago, Illinois 60637
| | - Ping Guo
- Division of Pediatric Surgery (X.X., S.F., Z.S., I.G., R.Z., J.W., K.P., C.S., P.G., G.K.G.), Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224; Department of General Surgery (Z.S.), The Third Xiangya Hospital of Central S University, Changsha 410013, China; and Department of Surgery (S.R., P.W.), University of Chicago, Chicago, Illinois 60637
| | - Sabarinathan Ramachandran
- Division of Pediatric Surgery (X.X., S.F., Z.S., I.G., R.Z., J.W., K.P., C.S., P.G., G.K.G.), Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224; Department of General Surgery (Z.S.), The Third Xiangya Hospital of Central S University, Changsha 410013, China; and Department of Surgery (S.R., P.W.), University of Chicago, Chicago, Illinois 60637
| | - Piotr Witkowski
- Division of Pediatric Surgery (X.X., S.F., Z.S., I.G., R.Z., J.W., K.P., C.S., P.G., G.K.G.), Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224; Department of General Surgery (Z.S.), The Third Xiangya Hospital of Central S University, Changsha 410013, China; and Department of Surgery (S.R., P.W.), University of Chicago, Chicago, Illinois 60637
| | - George K. Gittes
- Division of Pediatric Surgery (X.X., S.F., Z.S., I.G., R.Z., J.W., K.P., C.S., P.G., G.K.G.), Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224; Department of General Surgery (Z.S.), The Third Xiangya Hospital of Central S University, Changsha 410013, China; and Department of Surgery (S.R., P.W.), University of Chicago, Chicago, Illinois 60637
| |
Collapse
|
161
|
Fan Y, Xu LL, Shi CY, Wei W, Wang DS, Cai DF. MicroRNA-454 regulates stromal cell derived factor-1 in the control of the growth of pancreatic ductal adenocarcinoma. Sci Rep 2016; 6:22793. [PMID: 26976451 PMCID: PMC4792164 DOI: 10.1038/srep22793] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 02/10/2016] [Indexed: 12/19/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly malignant carcinoma with an extremely high lethality. We recently reported that hypoxia-inducible factor 1 (HIF-1) targets quiescin sulfhydryl oxidase 1 to facilitate PDAC cell growth and invasion. Here, we analyzed the control of another HIF-1 target, stromal cell derived factor-1 (SDF-1), in PDAC cells. We detected significantly more CD68+ macrophages in the PDAC, compared to normal human pancreas (NT). Since macrophages are recruited to the tissue through their expression of CXCR4 in response to SDF-1, we thus examined the SDF-1 levels in the PDAC specimens. Surprisingly, the SDF-1 protein but not mRNA significantly increased in PDAC, compared to NT. Moreover, a SDF-1-targeting microRNA, miR-454, was found to decrease in PDAC. Promoter luciferase assay confirmed that bindings of miR-454 to 3'-UTR of SDF-1 mRNAs inhibited SDF-1 protein translation. Co-culture of bone marrow derived macrophages and miR-454-modified PDAC cells in a transwell migration experiment showed that macrophages migrated less towards miR-454-overexpressing PDAC cells, and migrated more towards miR-454-depleted cells. Implanted miR-454-depleted PDAC cells grew significantly faster than control, while implanted miR-454-overexpressing PDAC cells grew significantly slower than control. Together, our data suggest that miR-454 may regulate SDF-1 in the control of the growth of PDAC.
Collapse
Affiliation(s)
- Yue Fan
- Department of Integrated TCM & Western Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Li-Li Xu
- Department of Integrated TCM & Western Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Chen-Ye Shi
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Wei Wei
- Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Institute for Nutritional Sciences, Shanghai 200032, China
| | - Dan-Song Wang
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Ding-Fang Cai
- Department of Integrated TCM & Western Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| |
Collapse
|
162
|
Wynn TA, Vannella KM. Macrophages in Tissue Repair, Regeneration, and Fibrosis. Immunity 2016; 44:450-462. [PMID: 26982353 PMCID: PMC4794754 DOI: 10.1016/j.immuni.2016.02.015] [Citation(s) in RCA: 2882] [Impact Index Per Article: 320.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 01/28/2016] [Accepted: 02/17/2016] [Indexed: 11/18/2022]
Abstract
Inflammatory monocytes and tissue-resident macrophages are key regulators of tissue repair, regeneration, and fibrosis. After tissue injury, monocytes and macrophages undergo marked phenotypic and functional changes to play critical roles during the initiation, maintenance, and resolution phases of tissue repair. Disturbances in macrophage function can lead to aberrant repair, such that uncontrolled production of inflammatory mediators and growth factors, deficient generation of anti-inflammatory macrophages, or failed communication between macrophages and epithelial cells, endothelial cells, fibroblasts, and stem or tissue progenitor cells all contribute to a state of persistent injury, and this could lead to the development of pathological fibrosis. In this review, we discuss the mechanisms that instruct macrophages to adopt pro-inflammatory, pro-wound-healing, pro-fibrotic, anti-inflammatory, anti-fibrotic, pro-resolving, and tissue-regenerating phenotypes after injury, and we highlight how some of these mechanisms and macrophage activation states could be exploited therapeutically.
Collapse
Affiliation(s)
- Thomas A Wynn
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA.
| | - Kevin M Vannella
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA
| |
Collapse
|
163
|
Xiao X, Fischbach S, Fusco J, Zimmerman R, Song Z, Nebres P, Ricks DM, Prasadan K, Shiota C, Husain SZ, Gittes GK. PNA lectin for purifying mouse acinar cells from the inflamed pancreas. Sci Rep 2016; 6:21127. [PMID: 26884345 PMCID: PMC4756371 DOI: 10.1038/srep21127] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 01/18/2016] [Indexed: 12/14/2022] Open
Abstract
Better methods for purifying human or mouse acinar cells without the need for genetic modification are needed. Such techniques would be advantageous for the specific study of certain mechanisms, such as acinar-to-beta-cell reprogramming and pancreatitis. Ulex Europaeus Agglutinin I (UEA-I) lectin has been used to label and isolate acinar cells from the pancreas. However, the purity of the UEA-I-positive cell fraction has not been fully evaluated. Here, we screened 20 widely used lectins for their binding specificity for major pancreatic cell types, and found that UEA-I and Peanut agglutinin (PNA) have a specific affinity for acinar cells in the mouse pancreas, with minimal affinity for other major pancreatic cell types including endocrine cells, duct cells and endothelial cells. Moreover, PNA-purified acinar cells were less contaminated with mesenchymal and inflammatory cells, compared to UEA-I purified acinar cells. Thus, UEA-I and PNA appear to be excellent lectins for pancreatic acinar cell purification. PNA may be a better choice in situations where mesenchymal cells or inflammatory cells are significantly increased in the pancreas, such as type 1 diabetes, pancreatitis and pancreatic cancer.
Collapse
Affiliation(s)
- Xiangwei Xiao
- Division of Pediatric Surgery, Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, 4401 Penn Ave, Pittsburgh, PA15224, USA
| | - Shane Fischbach
- Division of Pediatric Surgery, Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, 4401 Penn Ave, Pittsburgh, PA15224, USA
| | - Joseph Fusco
- Division of Pediatric Surgery, Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, 4401 Penn Ave, Pittsburgh, PA15224, USA
| | - Ray Zimmerman
- Division of Pediatric Surgery, Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, 4401 Penn Ave, Pittsburgh, PA15224, USA
| | - Zewen Song
- Division of Pediatric Surgery, Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, 4401 Penn Ave, Pittsburgh, PA15224, USA
| | - Philip Nebres
- Division of Pediatric Surgery, Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, 4401 Penn Ave, Pittsburgh, PA15224, USA
| | - David Matthew Ricks
- Division of Pediatric Surgery, Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, 4401 Penn Ave, Pittsburgh, PA15224, USA
| | - Krishna Prasadan
- Division of Pediatric Surgery, Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, 4401 Penn Ave, Pittsburgh, PA15224, USA
| | - Chiyo Shiota
- Division of Pediatric Surgery, Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, 4401 Penn Ave, Pittsburgh, PA15224, USA
| | - Sohail Z. Husain
- Division of Pediatrics, Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, 4401 Penn Ave, Pittsburgh, PA15224, USA
| | - George K. Gittes
- Division of Pediatric Surgery, Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, 4401 Penn Ave, Pittsburgh, PA15224, USA
| |
Collapse
|
164
|
Wang L, Li Y, Qin H, Xing D, Su J, Hu Z. Crosstalk between ACE2 and PLGF regulates vascular permeability during acute lung injury. Am J Transl Res 2016; 8:1246-52. [PMID: 27158411 PMCID: PMC4846968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2016] [Accepted: 01/29/2016] [Indexed: 06/05/2023]
Abstract
Angiotensin converting enzyme 2 (ACE2) treatment suppresses the severity of acute lung injury (ALI), through antagonizing hydrolyzing angiotensin II (AngII) and the ALI-induced apoptosis of pulmonary endothelial cells. Nevertheless, the effects of ACE2 on vessel permeability and its relationship with placental growth factor (PLGF) remain ill-defined. In the current study, we examined the relationship between ACE2 and PLGF in ALI model in mice. We used a previously published bleomycin method to induce ALI in mice, and treated the mice with ACE2. We analyzed the levels of PLGF in these mice. The mouse lung vessel permeability was determined by a fluorescence pharmacokinetic assay following i.v. injection of 62.5 µg/kg Visudyne. PLGF pump or soluble Flt-1 (sFlt-1) pump was given to augment or suppress PLGF effects, respectively. The long-term effects on lung function were determined by measurement of lung resistance using methacholine. We found that ACE2 treatment did not alter PLGF levels in lung, but antagonized the effects of PLGF on increases of lung vessel permeability. Ectogenic PLGF abolished the antagonizing effects of ACE2 on the vessel permeability against PLGF. On the other hand, suppression of PLGF signaling mimicked the effects of ACE2 on the vessel permeability against PLGF. The suppression of vessel permeability resulted in improvement of lung function after ALI. Thus, ACE2 may antagonize the PLGF-mediated increases in lung vessel permeability during ALI, resulting in improvement of lung function after ALI.
Collapse
Affiliation(s)
- Lantao Wang
- Department of Emergency, Fourth Hospital of Hebei Medical UniversityShijiazhuang 050011, China
| | - Yong Li
- Department of Surgery, Fourth Hospital of Hebei Medical UniversityShijiazhuang 050011, China
| | - Hao Qin
- Department of Emergency, Fourth Hospital of Hebei Medical UniversityShijiazhuang 050011, China
| | - Dong Xing
- Department of Emergency, Fourth Hospital of Hebei Medical UniversityShijiazhuang 050011, China
| | - Jie Su
- Department of Emergency, Fourth Hospital of Hebei Medical UniversityShijiazhuang 050011, China
| | - Zhenjie Hu
- Department of Emergency, Fourth Hospital of Hebei Medical UniversityShijiazhuang 050011, China
| |
Collapse
|
165
|
Shi C, Li G, Tong Y, Deng Y, Fan J. Role of CTGF gene promoter methylation in the development of hepatic fibrosis. Am J Transl Res 2016; 8:125-132. [PMID: 27069546 PMCID: PMC4759422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2015] [Accepted: 12/04/2015] [Indexed: 06/05/2023]
Abstract
Connective tissue growth factor (CTGF) plays a critical role in the hepatic stellate cells (HSCs)-mediated development of hepatic fibrosis. Nevertheless, the effects of CTGF gene promoter methylation in the pathogenesis of hepatic fibrosis remain largely unknown. In the current study, we isolated and overexpressed CTGF in primary HSCs. We analyzed the CTGF gene promoter methylation inHSCs that undergo a phenotypic change into myofibroblast-like cellsthat express α-smooth muscle actin (α-SMA) in vitro and in vivo in a CCl4-induced rat hepatic fibrosis model. We found that CTGF promoted the phenotypic changes of HSCs into myofibroblasts in vitro, while inhibition of CTGF promoter methylation augmented the process, suggesting that CTGF gene promoter methylation may negatively regulate hepatic fibrosis. In vivo, CCl4 induced hepatic fibrosis in rats, and the severity of hepatic fibrosis inversely correlated with the levels of CTGF gene promoter methylation in HSCs. Together, our data demonstrate that CTGF gene promoter methylation may prevent the development of hepatic fibrosis, and low level of CTGF gene promoter methylation in HSCs may be a predisposing factor for developing liver fibrotic disease.
Collapse
Affiliation(s)
- Cuicui Shi
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine Shanghai 200092, China
| | - Guangming Li
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine Shanghai 200092, China
| | - Yanyan Tong
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine Shanghai 200092, China
| | - Yilin Deng
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine Shanghai 200092, China
| | - Jiangao Fan
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine Shanghai 200092, China
| |
Collapse
|
166
|
Gittes GK. Multiple roles for TGFβ receptor type II in regulating the pancreatic response in acute pancreatitis. J Pathol 2016; 238:603-5. [DOI: 10.1002/path.4676] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 11/24/2015] [Indexed: 12/18/2022]
Affiliation(s)
- George K Gittes
- Children's Hospital of Pittsburgh and the Division of Pediatric Surgery; University of Pittsburgh School of Medicine; Pittsburgh PA USA
| |
Collapse
|
167
|
Yu K, Fischbach S, Xiao X. Beta Cell Regeneration in Adult Mice: Controversy Over the Involvement of Stem Cells. Curr Stem Cell Res Ther 2016; 11:542-546. [PMID: 25429702 PMCID: PMC5078597 DOI: 10.2174/1574888x10666141126113110] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Revised: 10/17/2014] [Accepted: 11/24/2014] [Indexed: 01/06/2023]
Abstract
Islet transplantation is an effective therapy for severe diabetes. Nevertheless, the short supply of donor pancreases constitutes a formidable obstacle to its extensive clinical application. This shortage heightens the need for alternative sources of insulin-producing beta cells. Since mature beta cells have a very slow proliferation rate, which further declines with age, great efforts have been made to identify beta cell progenitors in the adult pancreas. However, the question whether facultative beta cell progenitors indeed exist in the adult pancreas remains largely unresolved. In the current review, we discuss the problems in past studies and review the milestone studies and recent publications.
Collapse
Affiliation(s)
- Ke Yu
- Beijing Key Laboratory of Diabetes Prevention and Care, Department of Endocrinology, Lu He Hospital, Capital Medical University, Beijing, China
| | - Shane Fischbach
- Division of Pediatric Surgery, Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh,USA
- Division of Biology and Medicine, Brown University, Providence,USA
| | - Xiangwei Xiao
- Division of Pediatric Surgery, Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh,USA
| |
Collapse
|
168
|
Affiliation(s)
- Xiangwei Xiao
- Department of Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224
| |
Collapse
|
169
|
Mesenchymal stem cells alleviate experimental asthma by inducing polarization of alveolar macrophages. Inflammation 2015; 38:485-92. [PMID: 24958014 DOI: 10.1007/s10753-014-9954-6] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
The reparative and immunoregulatory properties of mesenchymal stromal cells (MSCs) have made them attractive candidates for cellular therapy. However, the underlying mechanism of the effects of transplanted MSCs on allergic asthma remains elusive. Here, we show that administration of MSCs isolated from human bone marrow provoked a pronounced polarization in alveolar macrophages to M2 subtypes, rather than induced an increase in the total macrophage number, and efficiently inhibited hallmark features of asthma, including airway hyperresponsiveness and eosinophilic accumulation. Moreover, transforming growth factor beta (TGF-β) signaling pathway appeared to mediate the effects of MSCs on macrophage polarization and subsequently the inhibition of hallmark features of asthma. Inhibition of TGF-β signaling was sufficient to inhibit the macrophage polarization in response to MSCs and consequently reserved the inhibitory effects of macrophage polarization on hallmark features of asthma. Collectively, our data demonstrate that human MSCs have immunosuppressive activity on asthma, which is mediated by TGF-β-signaling-dependent alveolar macrophage polarization.
Collapse
|
170
|
Li R, Zhang Y, Mohamed MA, Wei X, Cheng C. Macrophages play an essential role in the long effects of low-dose photodynamic therapy on vessel permeability. Int J Biochem Cell Biol 2015; 71:55-61. [PMID: 26667270 DOI: 10.1016/j.biocel.2015.12.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 11/25/2015] [Accepted: 12/02/2015] [Indexed: 12/16/2022]
Abstract
Low-dose photodynamic therapy (L-PDT) has been used to transiently increase the permeability of tumor vessels to improve the delivery of chemotherapeutic drugs to lung tumors. However, the effects of L-PDT in a long-term on delivery of chemotherapeutic drugs are unknown. In this study, we studied this question as well as the underlying mechanisms. We found that the effects of L-PDT on tumor vessel permeability appeared to be prolonged. Moreover, L-PDT significantly increased the number of tumor associated macrophages, and appeared to induce macrophage polarization to a M1 phenotype. Further analyses showed that L-PDT upregulated stromal cell-derived factor 1 (SDF-1) in tumor to recruit macrophages through a SDF-1/Chemokine (C-X-C Motif) Receptor 4 (CXCR4) axis, which accounted for the prolonged effects of L-PDT on vessel permeability. Application of a specific CXCR4 inhibitor significantly suppressed the L-PDT-induced macrophage recruitment, resulting in abolishment of the prolonged effects of L-PDT on vessel permeability. Furthermore, the inhibitory effects of Liporubicin™ on the growth of the implanted tumor in L-PDT-treated mice were significantly attenuated by CXCR4 inhibition. Thus, our data demonstrate a previously unappreciated long-lasting effect of L-PDT on vessel permeability, and suggest that this long-lasting effects of L-PDT treatment on vessel permeability may result from modulation of macrophage recruitment and polarization. Hence, L-PDT may be a promising method to assist chemotherapeutic approaches.
Collapse
Affiliation(s)
- Rui Li
- Department of Cardiothoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yi Zhang
- Department of Surgery, the Third Affiliated Hospital of Jianghan University, 259 Baixiu Street, Wuhan 430030, China
| | - Mohamed Abdulkadir Mohamed
- Department of Cardiothoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiang Wei
- Department of Cardiothoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Cai Cheng
- Department of Cardiothoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
171
|
Filipek A, Czerwińska ME, Kiss AK, Wrzosek M, Naruszewicz M. Oleacein enhances anti-inflammatory activity of human macrophages by increasing CD163 receptor expression. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2015; 22:1255-1261. [PMID: 26655408 DOI: 10.1016/j.phymed.2015.10.005] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Revised: 10/16/2015] [Accepted: 10/20/2015] [Indexed: 06/05/2023]
Abstract
BACKGROUND Oleacein (dialdehydic form of decarboxymethyl elenolic acid linked to hydroxytyrosol; 3,4-DHPEA-EDA) have been proven to possess antioxidant and anti-inflammatory activity. PURPOSE In this study, we examined whether oleacein could increase CD163 and IL-10 receptor expression as well as HO-1 intracellular secretion in human macrophages. METHODS Effect of oleacein (10 and 20 μmol/l) or oleacein together with complexes of haemoglobin (Hb) and haptoglobin 1-1 (Hp11) or haptoglobin 2-2 (Hp22) on expression of IL-10 and CD163 receptor was determined by Flow Cytometry. Expression of CD163mRNA was measured by real-time quantitative RT-PCR. Heme oxygenase 1 (HO-1) intracellular secretion in macrophages was investigated by enzyme-linked immunosorbent assay (ELISA). RESULTS Oleacein (OC) together with complexes HbHp11 or HbHp22 stimulated the expression of CD163 (30-100-fold), IL-10 (170-300-fold) and HO-1 secretion (60-130-fold) after 5 days of coincubation. The 2-fold (24 h), 4-fold (48 h) increase of CD163 mRNA level and its final (72 h) decrease was also observed. CONCLUSION Our results suggested that oleacein enhances anti-inflammatory activity of complexes haemoglobin with haptoglobin 1-1 and 2-2 and could play a potential role in the prevention of inflammatory disease related to atherosclerosis.
Collapse
Affiliation(s)
- Agnieszka Filipek
- Department of Pharmacognosy and Molecular Basis of Phytotherapy, Faculty of Pharmacy, Medical University of Warsaw, Banacha 1, 02-097 Warsaw, Poland
| | - Monika E Czerwińska
- Department of Pharmacognosy and Molecular Basis of Phytotherapy, Faculty of Pharmacy, Medical University of Warsaw, Banacha 1, 02-097 Warsaw, Poland
| | - Anna K Kiss
- Department of Pharmacognosy and Molecular Basis of Phytotherapy, Faculty of Pharmacy, Medical University of Warsaw, Banacha 1, 02-097 Warsaw, Poland
| | - Małgorzata Wrzosek
- Department of Pharmacogenomics, Division of Biochemistry and Clinical Chemistry, Medical University of Warsaw, Banacha 1, 02-097 Warsaw, Poland
| | - Marek Naruszewicz
- Department of Pharmacognosy and Molecular Basis of Phytotherapy, Faculty of Pharmacy, Medical University of Warsaw, Banacha 1, 02-097 Warsaw, Poland.
| |
Collapse
|
172
|
Grabliauskaite K, Saponara E, Reding T, Bombardo M, Seleznik GM, Malagola E, Zabel A, Faso C, Sonda S, Graf R. Inactivation of TGFβ receptor II signalling in pancreatic epithelial cells promotes acinar cell proliferation, acinar-to-ductal metaplasia and fibrosis during pancreatitis. J Pathol 2015; 238:434-45. [PMID: 26510396 DOI: 10.1002/path.4666] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Revised: 09/28/2015] [Accepted: 10/20/2015] [Indexed: 12/14/2022]
Abstract
Determining signalling pathways that regulate pancreatic regeneration following pancreatitis is critical for implementing therapeutic interventions. In this study we elucidated the molecular mechanisms underlying the effects of transforming growth factor-β (TGFβ) in pancreatic epithelial cells during tissue regeneration. To this end, we conditionally inactivated TGFβ receptor II (TGFβ-RII) using a Cre-LoxP system under the control of pancreas transcription factor 1a (PTF1a) promoter, specific for the pancreatic epithelium, and evaluated the molecular and cellular changes in a mouse model of cerulein-induced pancreatitis. We show that TGFβ-RII signalling does not mediate the initial acinar cell damage observed at the onset of pancreatitis. However, TGFβ-RII signalling not only restricts acinar cell replication during the regenerative phase of the disease but also limits ADM formation in vivo and in vitro in a cell-autonomous manner. Analyses of molecular mechanisms underlying the observed phenotype revealed that TGFβ-RII signalling stimulates the expression of cyclin-dependent kinase inhibitors and intersects with the EGFR signalling axis. Finally, TGFβ-RII ablation in epithelial cells resulted in increased infiltration of inflammatory cells in the early phases of pancreatitis and increased activation of pancreatic stellate cells in the later stages of pancreatitis, thus highlighting a TGFβ-based crosstalk between epithelial and stromal cells regulating the development of pancreatic inflammation and fibrosis. Collectively, our data not only contribute to clarifying the cellular processes governing pancreatic tissue regeneration, but also emphasize the conserved role of TGFβ as a tumour suppressor, both in the regenerative process following pancreatitis and in the initial phases of pancreatic cancer.
Collapse
Affiliation(s)
- Kamile Grabliauskaite
- Swiss Hepato-Pancreato-Biliary Centre, Department of Visceral and Transplantation Surgery, University Hospital, Zurich, and Zurich Centre for Integrative Human Physiology (ZIHP), University of Zurich, Switzerland
| | - Enrica Saponara
- Swiss Hepato-Pancreato-Biliary Centre, Department of Visceral and Transplantation Surgery, University Hospital, Zurich, and Zurich Centre for Integrative Human Physiology (ZIHP), University of Zurich, Switzerland
| | - Theresia Reding
- Swiss Hepato-Pancreato-Biliary Centre, Department of Visceral and Transplantation Surgery, University Hospital, Zurich, and Zurich Centre for Integrative Human Physiology (ZIHP), University of Zurich, Switzerland
| | - Marta Bombardo
- Swiss Hepato-Pancreato-Biliary Centre, Department of Visceral and Transplantation Surgery, University Hospital, Zurich, and Zurich Centre for Integrative Human Physiology (ZIHP), University of Zurich, Switzerland
| | - Gitta M Seleznik
- Swiss Hepato-Pancreato-Biliary Centre, Department of Visceral and Transplantation Surgery, University Hospital, Zurich, and Zurich Centre for Integrative Human Physiology (ZIHP), University of Zurich, Switzerland
| | - Ermanno Malagola
- Swiss Hepato-Pancreato-Biliary Centre, Department of Visceral and Transplantation Surgery, University Hospital, Zurich, and Zurich Centre for Integrative Human Physiology (ZIHP), University of Zurich, Switzerland
| | - Anja Zabel
- Swiss Hepato-Pancreato-Biliary Centre, Department of Visceral and Transplantation Surgery, University Hospital, Zurich, and Zurich Centre for Integrative Human Physiology (ZIHP), University of Zurich, Switzerland
| | - Carmen Faso
- Institute of Parasitology, University of Zurich, Switzerland
| | - Sabrina Sonda
- Swiss Hepato-Pancreato-Biliary Centre, Department of Visceral and Transplantation Surgery, University Hospital, Zurich, and Zurich Centre for Integrative Human Physiology (ZIHP), University of Zurich, Switzerland
| | - Rolf Graf
- Swiss Hepato-Pancreato-Biliary Centre, Department of Visceral and Transplantation Surgery, University Hospital, Zurich, and Zurich Centre for Integrative Human Physiology (ZIHP), University of Zurich, Switzerland
| |
Collapse
|
173
|
Li X. TIPE2 regulates tumor-associated macrophages in skin squamous cell carcinoma. Tumour Biol 2015; 37:5585-90. [DOI: 10.1007/s13277-015-4388-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 11/04/2015] [Indexed: 01/10/2023] Open
|
174
|
Wan G, Wei B. Erythropoietin regulates Treg cells in asthma through TGFβ receptor signaling. Am J Transl Res 2015; 7:2305-2315. [PMID: 26807178 PMCID: PMC4697710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 10/11/2015] [Indexed: 06/05/2023]
Abstract
Asthma is a chronic inflammatory disorder of the airways, the development of which is suppressed by regulatory T cells (Treg). Erythropoietin (EPO) is originally defined as a hematopoietic growth factor. Recently, the anti-inflammatory effects of EPO in asthma have been acknowledged. However, the underlying mechanisms remain ill-defined. Here, we showed that EPO treatment significantly reduced the severity of an ovalbumin (OVA)-induced asthma in mice, seemingly through promoting Foxp3-mediated activation of Treg cells in OVA-treated mouse lung. The activation of Treg cells resulted from increases in transforming growth factor β1 (TGFβ1), which were mainly produced by M2 macrophages (M2M). In vitro, Co-culture with M2M increased Foxp3 levels in Treg cells and the Treg cell number, in a TGFβ receptor signaling dependent manner. Moreover, elimination of macrophages abolished the therapeutic effects of EPO in vivo. Together, our data suggest that EPO may increase M2M, which activate Treg cells through TGFβ receptor signaling to mitigate the severity of asthma.
Collapse
Affiliation(s)
- Guoshi Wan
- Third Department of Internal Medicine, Second Hospital of Yuhang District of Hangzhou311121, China
| | - Bing Wei
- Department of Pediatrics, General Hospital of Shenyang Military Area CommandShenyang 110016, China
| |
Collapse
|
175
|
Long Y, Liu X, Wang N, Zhou H, Zheng J. Chloroquine attenuates LPS-mediated macrophage activation through miR-669n-regulated SENP6 protein translation. Am J Transl Res 2015; 7:2335-2345. [PMID: 26807181 PMCID: PMC4697713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 10/12/2015] [Indexed: 06/05/2023]
Abstract
Chloroquine (CQ) has been shown to inhibit Toll-like receptor 4 (TLR4)-mediated monocyte and macrophage activation induced by lipopolysaccharide (LPS). However, the underlying mechanisms have not been completely elucidated. Recently, SUMO-specific protease 6 (SENP6) has been reported to suppress LPS-induced activation of macrophages through deSUMOlation of NF-κB essential modifier (NEMO). Here, we studied whether this molecular pathway may also be involved in CQ/LPS model. We found that CQ dose-dependently increased SENP6 protein, but not mRNA, in mouse macrophages, RAW264.7 cells. Overexpression of SENP6 in RAW264.7 cells significantly decreased the LPS-induced release of pro-inflammatory proteins, TNF-α, IL-6 and IFN-γ, while depletion of SENP6 in RAW264.7 cells significantly increased these proteins. Moreover, in LPS-treated RAW264.7 cells, CQ dose-dependently decreased the levels of microRNA-669n (miR-669n), which bound to 3'-UTR of SENP6 mRNA to inhibit its translation. Overexpression of miR-669n decreased SENP6, resulting in increased production of TNF-α, IL-6 and IFN-γ in RAW264.7 cells, while depletion of miR-669n increased SENP6, resulting in decreased production of TNF-α, IL-6 and IFN-γ in RAW264.7 cells. In vivo, delivery of miR-669n plasmids augmented the effects of LPS, while delivery of antisense of miR-669n (as-miR-669n) plasmids abolished the effects of LPS. Taken together, our data demonstrate a previously unappreciated molecular control of LPS-induced macrophage activation by CQ, through miR-669n-regulated SENP6 protein translation.
Collapse
Affiliation(s)
- Yupeng Long
- Medical Research Center, Southwestern Hospital, The Third Military Medical UniversityChongqing 400038, China
| | - Xin Liu
- Medical Research Center, Southwestern Hospital, The Third Military Medical UniversityChongqing 400038, China
| | - Ning Wang
- Medical Research Center, Southwestern Hospital, The Third Military Medical UniversityChongqing 400038, China
| | - Hong Zhou
- Department of Pharmacology, College of Pharmacy, The Third Military Medical UniversityChongqing 400038, China
| | - Jiang Zheng
- Medical Research Center, Southwestern Hospital, The Third Military Medical UniversityChongqing 400038, China
| |
Collapse
|
176
|
Husseini M, Wang GS, Patrick C, Crookshank JA, MacFarlane AJ, Noel JA, Strom A, Scott FW. Heme Oxygenase-1 Induction Prevents Autoimmune Diabetes in Association With Pancreatic Recruitment of M2-Like Macrophages, Mesenchymal Cells, and Fibrocytes. Endocrinology 2015; 156:3937-49. [PMID: 26252059 DOI: 10.1210/en.2015-1304] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Immunoregulatory and regenerative processes are activated in the pancreas during the development of type 1 diabetes (T1D) but are insufficient to prevent the disease. We hypothesized that the induction of cytoprotective heme oxygenase-1 (HO-1) by cobalt protophoryrin (CoPP) would prevent T1D by promoting anti-inflammatory and pro-repair processes. Diabetes-prone BioBreeding rats received ip CoPP or saline twice per week for 3 weeks, starting at 30 days and were monitored for T1D. Immunohistochemistry, confocal microscopy, quantitative RT-PCR, and microarrays were used to evaluate postinjection pancreatic changes at 51 days, when islet inflammation is first visible. T1D was prevented in CoPP-treated rats (29% vs 73%). Pancreatic Hmox1 was up-regulated along with islet-associated CD68(+)HO-1(+) cells, which were also observed in a striking peri-lobular interstitial infiltrate. Most interstitial cells expressed the mesenchymal marker vimentin and the hematopoietic marker CD34. Spindle-shaped, CD34(+)vimentin(+) cells coexpressed collagen V, characteristic of fibrocytes. M2 macrophage factors Krüppel-like factor 4, CD163, and CD206 were expressed by interstitial cells, consistent with pancreatic upregulation of several M2-associated genes. CoPP upregulated islet-regenerating REG genes and increased neogenic REG3β(+) and insulin(+) clusters. Thus, short-term induction of HO-1 promoted a protective M2-like milieu in the pancreas and recruited mesenchymal cells, M2 macrophages, and fibrocytes that imparted immunoregulatory and pro-repair effects, preventing T1D.
Collapse
MESH Headings
- Animals
- Antigens, CD/metabolism
- Antigens, CD34/metabolism
- Antigens, Differentiation, Myelomonocytic/metabolism
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/metabolism
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Collagen Type V/metabolism
- Diabetes Mellitus, Type 1/genetics
- Diabetes Mellitus, Type 1/metabolism
- Diabetes Mellitus, Type 1/prevention & control
- Enzyme Induction/drug effects
- Female
- Gene Expression/drug effects
- Heme Oxygenase-1/biosynthesis
- Heme Oxygenase-1/genetics
- Insulin/genetics
- Insulin/metabolism
- Kruppel-Like Factor 4
- Lectins, C-Type/genetics
- Lectins, C-Type/metabolism
- Macrophages/drug effects
- Macrophages/metabolism
- Male
- Mannose Receptor
- Mannose-Binding Lectins/metabolism
- Mesenchymal Stem Cells/drug effects
- Mesenchymal Stem Cells/metabolism
- Microscopy, Confocal
- Pancreas/drug effects
- Pancreas/metabolism
- Pancreatitis-Associated Proteins
- Protoporphyrins/pharmacology
- Rats
- Receptors, Cell Surface/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Vimentin/metabolism
Collapse
Affiliation(s)
- Mahmoud Husseini
- Chronic Disease Program (M.H., G.-S.W., C.P., J.A.C., J.A.N., A.S., F.W.S.), Ottawa Hospital Research Institute, Ottawa, Ontario, Canada K1H 8L6 and Departments of Biochemistry, Microbiology, and Immunology (M.H., C.P., A.J.M., J.A.N., F.W.S.) and Medicine (F.W.S.), University of Ottawa, Ottawa, Ontario, Canada K1H 8L6; Food Directorate (A.J.M.), Health Products and Food Branch, Health Canada, Ottawa, Ontario, Canada K1A 0K9
| | - Gen-Sheng Wang
- Chronic Disease Program (M.H., G.-S.W., C.P., J.A.C., J.A.N., A.S., F.W.S.), Ottawa Hospital Research Institute, Ottawa, Ontario, Canada K1H 8L6 and Departments of Biochemistry, Microbiology, and Immunology (M.H., C.P., A.J.M., J.A.N., F.W.S.) and Medicine (F.W.S.), University of Ottawa, Ottawa, Ontario, Canada K1H 8L6; Food Directorate (A.J.M.), Health Products and Food Branch, Health Canada, Ottawa, Ontario, Canada K1A 0K9
| | - Christopher Patrick
- Chronic Disease Program (M.H., G.-S.W., C.P., J.A.C., J.A.N., A.S., F.W.S.), Ottawa Hospital Research Institute, Ottawa, Ontario, Canada K1H 8L6 and Departments of Biochemistry, Microbiology, and Immunology (M.H., C.P., A.J.M., J.A.N., F.W.S.) and Medicine (F.W.S.), University of Ottawa, Ottawa, Ontario, Canada K1H 8L6; Food Directorate (A.J.M.), Health Products and Food Branch, Health Canada, Ottawa, Ontario, Canada K1A 0K9
| | - Jennifer A Crookshank
- Chronic Disease Program (M.H., G.-S.W., C.P., J.A.C., J.A.N., A.S., F.W.S.), Ottawa Hospital Research Institute, Ottawa, Ontario, Canada K1H 8L6 and Departments of Biochemistry, Microbiology, and Immunology (M.H., C.P., A.J.M., J.A.N., F.W.S.) and Medicine (F.W.S.), University of Ottawa, Ottawa, Ontario, Canada K1H 8L6; Food Directorate (A.J.M.), Health Products and Food Branch, Health Canada, Ottawa, Ontario, Canada K1A 0K9
| | - Amanda J MacFarlane
- Chronic Disease Program (M.H., G.-S.W., C.P., J.A.C., J.A.N., A.S., F.W.S.), Ottawa Hospital Research Institute, Ottawa, Ontario, Canada K1H 8L6 and Departments of Biochemistry, Microbiology, and Immunology (M.H., C.P., A.J.M., J.A.N., F.W.S.) and Medicine (F.W.S.), University of Ottawa, Ottawa, Ontario, Canada K1H 8L6; Food Directorate (A.J.M.), Health Products and Food Branch, Health Canada, Ottawa, Ontario, Canada K1A 0K9
| | - J Ariana Noel
- Chronic Disease Program (M.H., G.-S.W., C.P., J.A.C., J.A.N., A.S., F.W.S.), Ottawa Hospital Research Institute, Ottawa, Ontario, Canada K1H 8L6 and Departments of Biochemistry, Microbiology, and Immunology (M.H., C.P., A.J.M., J.A.N., F.W.S.) and Medicine (F.W.S.), University of Ottawa, Ottawa, Ontario, Canada K1H 8L6; Food Directorate (A.J.M.), Health Products and Food Branch, Health Canada, Ottawa, Ontario, Canada K1A 0K9
| | - Alexander Strom
- Chronic Disease Program (M.H., G.-S.W., C.P., J.A.C., J.A.N., A.S., F.W.S.), Ottawa Hospital Research Institute, Ottawa, Ontario, Canada K1H 8L6 and Departments of Biochemistry, Microbiology, and Immunology (M.H., C.P., A.J.M., J.A.N., F.W.S.) and Medicine (F.W.S.), University of Ottawa, Ottawa, Ontario, Canada K1H 8L6; Food Directorate (A.J.M.), Health Products and Food Branch, Health Canada, Ottawa, Ontario, Canada K1A 0K9
| | - Fraser W Scott
- Chronic Disease Program (M.H., G.-S.W., C.P., J.A.C., J.A.N., A.S., F.W.S.), Ottawa Hospital Research Institute, Ottawa, Ontario, Canada K1H 8L6 and Departments of Biochemistry, Microbiology, and Immunology (M.H., C.P., A.J.M., J.A.N., F.W.S.) and Medicine (F.W.S.), University of Ottawa, Ottawa, Ontario, Canada K1H 8L6; Food Directorate (A.J.M.), Health Products and Food Branch, Health Canada, Ottawa, Ontario, Canada K1A 0K9
| |
Collapse
|
177
|
Reeves ARD, Spiller KL, Freytes DO, Vunjak-Novakovic G, Kaplan DL. Controlled release of cytokines using silk-biomaterials for macrophage polarization. Biomaterials 2015; 73:272-83. [PMID: 26421484 DOI: 10.1016/j.biomaterials.2015.09.027] [Citation(s) in RCA: 102] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2015] [Revised: 09/17/2015] [Accepted: 09/18/2015] [Indexed: 12/17/2022]
Abstract
Polarization of macrophages into an inflammatory (M1) or anti-inflammatory (M2) phenotype is important for clearing pathogens and wound repair, however chronic activation of either type of macrophage has been implicated in several diseases. Methods to locally control the polarization of macrophages is of great interest for biomedical implants and tissue engineering. To that end, silk protein was used to form biopolymer films that release either IFN-γ or IL-4 to control the polarization of macrophages. Modulation of the solubility of the silk films through regulation of β-sheet (crystalline) content enabled a short-term release (4-8 h) of either cytokine, with smaller amounts released out to 24 h. Altering the solubility of the films was accomplished by varying the time that the films were exposed to water vapor. The released IFN-γ or IL-4 induced polarization of THP-1 derived macrophages into the M1 or M2 phenotypes, respectively. The silk biomaterials were able to release enough IFN-γ or IL-4 to repolarize the macrophage from M1 to M2 and vice versa, demonstrating the well-established plasticity of macrophages. High β-sheet content films that are not soluble and do not release the trapped cytokines were also able to polarize macrophages that adhered to the surface through degradation of the silk protein. Chemically conjugating IFN-γ to silk films through disulfide bonds allowed for longer-term release to 10 days. The release of covalently attached IFN-γ from the films was also able to polarize M1 macrophages in vitro. Thus, the strategy described here offers new approaches to utilizing biomaterials for directing the polarization of macrophages.
Collapse
Affiliation(s)
- Andrew R D Reeves
- Department of Biomedical Engineering, Tufts University, Medford, MA, USA.
| | - Kara L Spiller
- School of Biomedical Engineering, Drexel University, Philadelphia, PA, USA.
| | - Donald O Freytes
- New York Stem Cell Foundation Research Institute, New York, NY, USA.
| | | | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, MA, USA.
| |
Collapse
|
178
|
Calderon B, Carrero JA, Ferris ST, Sojka DK, Moore L, Epelman S, Murphy KM, Yokoyama WM, Randolph GJ, Unanue ER. The pancreas anatomy conditions the origin and properties of resident macrophages. ACTA ACUST UNITED AC 2015; 212:1497-512. [PMID: 26347472 PMCID: PMC4577842 DOI: 10.1084/jem.20150496] [Citation(s) in RCA: 228] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 08/11/2015] [Indexed: 12/14/2022]
Abstract
Calderon et al. define the origin, turnover, and functional characteristics of pancreatic macrophages at both the exocrine and endocrine sites under noninflammatory conditions. We examine the features, origin, turnover, and gene expression of pancreatic macrophages under steady state. The data distinguish macrophages within distinct intrapancreatic microenvironments and suggest how macrophage phenotype is imprinted by the local milieu. Macrophages in islets of Langerhans and in the interacinar stroma are distinct in origin and phenotypic properties. In islets, macrophages are the only myeloid cells: they derive from definitive hematopoiesis, exchange to a minimum with blood cells, have a low level of self-replication, and depend on CSF-1. They express Il1b and Tnfa transcripts, indicating classical activation, M1, under steady state. The interacinar stroma contains two macrophage subsets. One is derived from primitive hematopoiesis, with no interchange by blood cells and alternative, M2, activation profile, whereas the second is derived from definitive hematopoiesis and exchanges with circulating myeloid cells but also shows an alternative activation profile. Complete replacement of islet and stromal macrophages by donor stem cells occurred after lethal irradiation with identical profiles as observed under steady state. The extraordinary plasticity of macrophages within the pancreatic organ and the distinct features imprinted by their anatomical localization sets the base for examining these cells in pathological conditions.
Collapse
Affiliation(s)
- Boris Calderon
- Department of Pathology and Immunology; Division of Rheumatology and Division of Cardiology, Department of Medicine; and Howard Hughes Medical Institute; Washington University School of Medicine in St. Louis, St. Louis, MO 63110
| | - Javier A Carrero
- Department of Pathology and Immunology; Division of Rheumatology and Division of Cardiology, Department of Medicine; and Howard Hughes Medical Institute; Washington University School of Medicine in St. Louis, St. Louis, MO 63110
| | - Stephen T Ferris
- Department of Pathology and Immunology; Division of Rheumatology and Division of Cardiology, Department of Medicine; and Howard Hughes Medical Institute; Washington University School of Medicine in St. Louis, St. Louis, MO 63110
| | - Dorothy K Sojka
- Department of Pathology and Immunology; Division of Rheumatology and Division of Cardiology, Department of Medicine; and Howard Hughes Medical Institute; Washington University School of Medicine in St. Louis, St. Louis, MO 63110
| | - Lindsay Moore
- Department of Pathology and Immunology; Division of Rheumatology and Division of Cardiology, Department of Medicine; and Howard Hughes Medical Institute; Washington University School of Medicine in St. Louis, St. Louis, MO 63110
| | - Slava Epelman
- Department of Pathology and Immunology; Division of Rheumatology and Division of Cardiology, Department of Medicine; and Howard Hughes Medical Institute; Washington University School of Medicine in St. Louis, St. Louis, MO 63110
| | - Kenneth M Murphy
- Department of Pathology and Immunology; Division of Rheumatology and Division of Cardiology, Department of Medicine; and Howard Hughes Medical Institute; Washington University School of Medicine in St. Louis, St. Louis, MO 63110 Department of Pathology and Immunology; Division of Rheumatology and Division of Cardiology, Department of Medicine; and Howard Hughes Medical Institute; Washington University School of Medicine in St. Louis, St. Louis, MO 63110
| | - Wayne M Yokoyama
- Department of Pathology and Immunology; Division of Rheumatology and Division of Cardiology, Department of Medicine; and Howard Hughes Medical Institute; Washington University School of Medicine in St. Louis, St. Louis, MO 63110 Department of Pathology and Immunology; Division of Rheumatology and Division of Cardiology, Department of Medicine; and Howard Hughes Medical Institute; Washington University School of Medicine in St. Louis, St. Louis, MO 63110 Department of Pathology and Immunology; Division of Rheumatology and Division of Cardiology, Department of Medicine; and Howard Hughes Medical Institute; Washington University School of Medicine in St. Louis, St. Louis, MO 63110
| | - Gwendalyn J Randolph
- Department of Pathology and Immunology; Division of Rheumatology and Division of Cardiology, Department of Medicine; and Howard Hughes Medical Institute; Washington University School of Medicine in St. Louis, St. Louis, MO 63110
| | - Emil R Unanue
- Department of Pathology and Immunology; Division of Rheumatology and Division of Cardiology, Department of Medicine; and Howard Hughes Medical Institute; Washington University School of Medicine in St. Louis, St. Louis, MO 63110
| |
Collapse
|
179
|
Yuchi Y, Cai Y, Legein B, De Groef S, Leuckx G, Coppens V, Van Overmeire E, Staels W, De Leu N, Martens G, Van Ginderachter JA, Heimberg H, Van de Casteele M. Estrogen Receptor α Regulates β-Cell Formation During Pancreas Development and Following Injury. Diabetes 2015; 64:3218-28. [PMID: 26015547 DOI: 10.2337/db14-1798] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 05/16/2015] [Indexed: 11/13/2022]
Abstract
Identifying pathways for β-cell generation is essential for cell therapy in diabetes. We investigated the potential of 17β-estradiol (E2) and estrogen receptor (ER) signaling for stimulating β-cell generation during embryonic development and in the severely injured adult pancreas. E2 concentration, ER activity, and number of ERα transcripts were enhanced in the pancreas injured by partial duct ligation (PDL) along with nuclear localization of ERα in β-cells. PDL-induced proliferation of β-cells depended on aromatase activity. The activation of Neurogenin3 (Ngn3) gene expression and β-cell growth in PDL pancreas were impaired when ERα was turned off chemically or genetically (ERα(-/-)), whereas in situ delivery of E2 promoted β-cell formation. In the embryonic pancreas, β-cell replication, number of Ngn3(+) progenitor cells, and expression of key transcription factors of the endocrine lineage were decreased by ERα inactivation. The current study reveals that E2 and ERα signaling can drive β-cell replication and formation in mouse pancreas.
Collapse
Affiliation(s)
- Yixing Yuchi
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Ying Cai
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Bart Legein
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Sofie De Groef
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Gunter Leuckx
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Violette Coppens
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Eva Van Overmeire
- Myeloid Cell Immunology Laboratory, Vlaams Instituut voor Biotechnologie, Brussels, Belgium Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Willem Staels
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium Department of Pediatrics, Division of Pediatric Endocrinology, Ghent University Hospital, and Department of Pediatrics and Medical Genetics, Ghent University, Ghent, Belgium
| | - Nico De Leu
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium Department of Endocrinology, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Geert Martens
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Jo A Van Ginderachter
- Myeloid Cell Immunology Laboratory, Vlaams Instituut voor Biotechnologie, Brussels, Belgium Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Harry Heimberg
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | | |
Collapse
|
180
|
Li S, Liu X, Chen X, Zhang L, Wang X. Histone deacetylase 6 promotes growth of glioblastoma through inhibition of SMAD2 signaling. Tumour Biol 2015; 36:9661-5. [DOI: 10.1007/s13277-015-3747-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 10/29/2014] [Indexed: 11/28/2022] Open
|
181
|
Lahmar Q, Keirsse J, Laoui D, Movahedi K, Van Overmeire E, Van Ginderachter JA. Tissue-resident versus monocyte-derived macrophages in the tumor microenvironment. Biochim Biophys Acta Rev Cancer 2015; 1865:23-34. [PMID: 26145884 DOI: 10.1016/j.bbcan.2015.06.009] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Revised: 06/25/2015] [Accepted: 06/26/2015] [Indexed: 12/12/2022]
Abstract
The tumor-promoting role of macrophages has been firmly established in most cancer types. However, macrophage identity has been a matter of debate, since several levels of complexity result in considerable macrophage heterogeneity. Ontogenically, tissue-resident macrophages derive from yolk sac progenitors which either directly or via a fetal liver monocyte intermediate differentiate into distinct macrophage types during embryogenesis and are maintained throughout life, while a disruption of the steady state mobilizes monocytes and instructs the formation of monocyte-derived macrophages. Histologically, the macrophage phenotype is heavily influenced by the tissue microenvironment resulting in molecularly and functionally distinct macrophages in distinct organs. Finally, a change in the tissue microenvironment as a result of infectious or sterile inflammation instructs different modes of macrophage activation. These considerations are relevant in the context of tumors, which can be considered as sites of chronic sterile inflammation encompassing subregions with distinct environmental conditions (for example, hypoxic versus normoxic). Here, we discuss existing evidence on the role of macrophage subpopulations in steady state tissue and primary tumors of the breast, lung, pancreas, brain and liver.
Collapse
Affiliation(s)
- Qods Lahmar
- Myeloid Cell Immunology Lab, VIB, Brussels, Belgium; Lab of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Jiri Keirsse
- Myeloid Cell Immunology Lab, VIB, Brussels, Belgium; Lab of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Damya Laoui
- Myeloid Cell Immunology Lab, VIB, Brussels, Belgium; Lab of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Kiavash Movahedi
- Myeloid Cell Immunology Lab, VIB, Brussels, Belgium; Lab of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Eva Van Overmeire
- Myeloid Cell Immunology Lab, VIB, Brussels, Belgium; Lab of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Jo A Van Ginderachter
- Myeloid Cell Immunology Lab, VIB, Brussels, Belgium; Lab of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium.
| |
Collapse
|
182
|
Boyle ST, Faulkner JW, McColl SR, Kochetkova M. The chemokine receptor CCR6 facilitates the onset of mammary neoplasia in the MMTV-PyMT mouse model via recruitment of tumor-promoting macrophages. Mol Cancer 2015; 14:115. [PMID: 26047945 PMCID: PMC4464622 DOI: 10.1186/s12943-015-0394-1] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Accepted: 05/29/2015] [Indexed: 01/02/2023] Open
Abstract
Background The expression of the chemokine receptor CCR6 has been previously correlated with higher grades and stages of breast cancer and decreased relapse-free survival. Also, its cognate chemokine ligand CCL20 has been reported to induce proliferation of cultured human breast epithelial cells. Methods To establish if CCR6 plays a functional role in mammary tumorigenesis, a bigenic MMTV-PyMT CCR6-null mouse was generated and mammary tumor development was assessed. Levels of tumor-infiltrating immune cells within tumor-bearing mammary glands from MMTV-PyMT Ccr6WT and Ccr6−/− mice were also analyzed. Results Deletion of CCR6 delayed tumor onset, significantly reduced the extent of initial hyperplastic outgrowth, and decreased tumor incidence in PyMT transgenic mice. CCR6 was then shown to promote the recruitment of pro-tumorigenic macrophages to the tumor site, facilitating the onset of neoplasia. Conclusions This study delineated for the first time a role for CCR6 in the development of breast cancer, and demonstrated a critical function for this receptor in maintaining the pro-tumorigenic cancer microenvironment. Electronic supplementary material The online version of this article (doi:10.1186/s12943-015-0394-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sarah T Boyle
- Department of Molecular and Cellular Biology, School of Biological Sciences, University of Adelaide, Adelaide, South Australia, Australia
| | - Jessica W Faulkner
- Department of Molecular and Cellular Biology, School of Biological Sciences, University of Adelaide, Adelaide, South Australia, Australia
| | - Shaun R McColl
- Department of Molecular and Cellular Biology, School of Biological Sciences, University of Adelaide, Adelaide, South Australia, Australia.,Centre for Molecular Pathology, University of Adelaide, Adelaide, South Australia, Australia
| | - Marina Kochetkova
- Department of Molecular and Cellular Biology, School of Biological Sciences, University of Adelaide, Adelaide, South Australia, Australia.
| |
Collapse
|
183
|
Application of chemokine receptor antagonist with stents reduces local inflammation and suppresses cancer growth. Tumour Biol 2015; 36:8637-43. [DOI: 10.1007/s13277-015-3557-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 05/11/2015] [Indexed: 12/19/2022] Open
|
184
|
Zhao J, Kong Z, Xu F, Shen W. A role of MMP-14 in the regulation of invasiveness of nasopharyngeal carcinoma. Tumour Biol 2015; 36:8609-15. [PMID: 26040767 DOI: 10.1007/s13277-015-3558-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Accepted: 05/11/2015] [Indexed: 11/28/2022] Open
Abstract
Although matrix metalloproteinase 14 (MMP-14) has been shown to play a substantial role in the carcinogenesis of some types of cancer, its involvement in the pathogenesis of nasopharyngeal carcinoma (NPC) has not been reported. Here, we analyzed MMP-14 levels in the NPC specimens from patients and compared with the paired normal nasopharynx (NNP) tissues. We found that NPC had significantly higher messenger RNA (mRNA) and protein levels of MMP-14. Moreover, higher levels of MMP-14 correlated with more advanced status of clinical stage and lymphatic metastasis. In vitro, MMP-14 levels determined the potential of invasiveness of NPC cells, possibly through induction of EMT-associated genes. Our data thus highlight MMP-14 as a novel therapeutic target for NPC.
Collapse
Affiliation(s)
- Jian Zhao
- Department of Otorhinolaryngology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, No.164 Lanxi Road, Shanghai, 200062, China
| | - Zhongyu Kong
- Department of Otorhinolaryngology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, No.164 Lanxi Road, Shanghai, 200062, China
| | - Feng Xu
- Department of Otorhinolaryngology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, No.164 Lanxi Road, Shanghai, 200062, China
| | - Wei Shen
- Department of Otorhinolaryngology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, No.164 Lanxi Road, Shanghai, 200062, China.
| |
Collapse
|
185
|
Guo H, Xu Y, Fu Q. Curcumin inhibits growth of prostate carcinoma via miR-208-mediated CDKN1A activation. Tumour Biol 2015; 36:8511-7. [PMID: 26032092 DOI: 10.1007/s13277-015-3592-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Accepted: 05/19/2015] [Indexed: 01/22/2023] Open
Abstract
Prostate cancer (PC) is a prevalent cancer in aged men. Curcumin is an active ingredient that has been extracted from the rhizome of the plant Curcuma longa. Recently, a potential of Curcumin against PC has been reported in PC, whereas the underlying molecular mechanisms are not completely understood. Here, we studied the effects of low-dose Curcumin on PC cell growth. Curcumin (from 0.2 to 0.8 μmol/l) dose-dependently inhibited the proliferation of PC cells, without affecting cell apoptosis. Further analyses showed that Curcumin dose-dependently increased a cell cycle suppressor CDKN1A at protein levels, but not mRNA levels, in PC cells, suggesting that Curcumin may regulate the translation of CDKN1A, as well as a possible involvement of miRNA intervention. From all CDKN1A-3'-UTR-binding miRNAs, we found that miR-208 was specifically inhibited in PC cells dose-dependently by Curcumin. Moreover, miR-208 was found to bind CDKN1A to suppress its expression. In a loss-of-function experiment, PC cells that overexpressed miR-208 failed to decrease cell proliferation in response to Curcumin. Together, these data suggest that Curcumin inhibits growth of PC via miR-208-mediated CDKN1A activation.
Collapse
Affiliation(s)
- Hui Guo
- Department of Urology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Yuemin Xu
- Department of Urology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Qiang Fu
- Department of Urology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China.
| |
Collapse
|
186
|
Stewart AF, Hussain MA, García-Ocaña A, Vasavada RC, Bhushan A, Bernal-Mizrachi E, Kulkarni RN. Human β-cell proliferation and intracellular signaling: part 3. Diabetes 2015; 64:1872-85. [PMID: 25999530 PMCID: PMC4439562 DOI: 10.2337/db14-1843] [Citation(s) in RCA: 116] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
This is the third in a series of Perspectives on intracellular signaling pathways coupled to proliferation in pancreatic β-cells. We contrast the large knowledge base in rodent β-cells with the more limited human database. With the increasing incidence of type 1 diabetes and the recognition that type 2 diabetes is also due in part to a deficiency of functioning β-cells, there is great urgency to identify therapeutic approaches to expand human β-cell numbers. Therapeutic approaches might include stem cell differentiation, transdifferentiation, or expansion of cadaver islets or residual endogenous β-cells. In these Perspectives, we focus on β-cell proliferation. Past Perspectives reviewed fundamental cell cycle regulation and its upstream regulation by insulin/IGF signaling via phosphatidylinositol-3 kinase/mammalian target of rapamycin signaling, glucose, glycogen synthase kinase-3 and liver kinase B1, protein kinase Cζ, calcium-calcineurin-nuclear factor of activated T cells, epidermal growth factor/platelet-derived growth factor family members, Wnt/β-catenin, leptin, and estrogen and progesterone. Here, we emphasize Janus kinase/signal transducers and activators of transcription, Ras/Raf/extracellular signal-related kinase, cadherins and integrins, G-protein-coupled receptors, and transforming growth factor β signaling. We hope these three Perspectives will serve to introduce these pathways to new researchers and will encourage additional investigators to focus on understanding how to harness key intracellular signaling pathways for therapeutic human β-cell regeneration for diabetes.
Collapse
Affiliation(s)
- Andrew F Stewart
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Mehboob A Hussain
- Departments of Medicine and Pediatrics, Johns Hopkins School of Medicine, Baltimore, MD
| | - Adolfo García-Ocaña
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Rupangi C Vasavada
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Anil Bhushan
- Diabetes Center, University of California, San Francisco, San Francisco, CA
| | - Ernesto Bernal-Mizrachi
- Division of Metabolism, Endocrinology & Diabetes, University of Michigan, Ann Arbor, MI, and VA Ann Arbor Healthcare System, Ann Arbor, MI
| | - Rohit N Kulkarni
- Section of Islet Cell and Regenerative Biology, Joslin Diabetes Center, Harvard Medical School, Boston, MA
| |
Collapse
|
187
|
Xiao X, Gittes GK. Concise Review: New Insights Into the Role of Macrophages in β-Cell Proliferation. Stem Cells Transl Med 2015; 4:655-658. [PMID: 25900729 PMCID: PMC4449096 DOI: 10.5966/sctm.2014-0248] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 03/09/2015] [Indexed: 12/12/2022] Open
Abstract
UNLABELLED Diabetes mellitus can potentially be treated with islet transplantation, but additional sources of β cells are necessary to overcome the short supply of donor pancreases. Although controversy still exists, it is generally believed that the postnatal expansion of the β-cell mass is mainly through pre-existing β-cell replication. Thus, understanding the molecular mechanisms underlying the regulation of β-cell proliferation might lead to clinical strategies for increasing β-cell numbers, both in vitro and in vivo. Macrophages have a well-recognized role in the development of insulitis as part of the pathogenesis of type 1 diabetes. However, a potential role for macrophage polarization, triggered by specific environmental stimuli, in promoting β-cell proliferation has only recently been appreciated. In the present review, we discuss several independent studies, using different regeneration models, that demonstrate a substantial inductive role for macrophages in β-cell proliferation. Additional dissection of the involved cell-cell crosstalk through specific signal transduction pathways is expected to improve our understanding of β-cell proliferation and might facilitate the current β-cell replacement therapy. SIGNIFICANCE New independent findings from different β-cell regeneration models, contributed by different research groups, have provided compelling evidence to highlight a previously unappreciated role for macrophages in β-cell proliferation. Additional dissection of the underlying mechanisms and cell-cell crosstalk might shed new light on strategies to increase the functional β-cell mass in vivo and on β-cell replacement therapies.
Collapse
Affiliation(s)
- Xiangwei Xiao
- Division of Pediatric Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - George K Gittes
- Division of Pediatric Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
188
|
Morris DL. Minireview: Emerging Concepts in Islet Macrophage Biology in Type 2 Diabetes. Mol Endocrinol 2015; 29:946-62. [PMID: 26001058 DOI: 10.1210/me.2014-1393] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Chronic systemic inflammation is a hallmark feature of obesity and type 2 diabetes. Both resident and recruited islet macrophages contribute to the proinflammatory milieu of the diabetic islet. However, macrophages also appear to be critical for β-cell formation during development and support β-cell replication in experimental models of pancreas regeneration. In light of these findings, perhaps macrophages in the islet need to be viewed more as a fulcrum where deleterious inflammatory activation is balanced with beneficial tissue repair processes. Undoubtedly, defining the factors that contribute to the ontogeny, heterogeneity, and functionality of macrophages in normal, diseased, and regenerating islets will be necessary to determine whether that fulcrum can be moved to preserve functional β-cell mass in persons with diabetes. The intent of this review is to introduce the reader to emerging concepts of islet macrophage biology that may challenge the perception that macrophage accumulation in islets is merely a pathological feature of type 2 diabetes.
Collapse
Affiliation(s)
- David L Morris
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana 46202
| |
Collapse
|
189
|
Riley KG, Pasek RC, Maulis MF, Dunn JC, Bolus WR, Kendall PL, Hasty AH, Gannon M. Macrophages are essential for CTGF-mediated adult β-cell proliferation after injury. Mol Metab 2015; 4:584-91. [PMID: 26266091 PMCID: PMC4529497 DOI: 10.1016/j.molmet.2015.05.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Revised: 04/28/2015] [Accepted: 05/01/2015] [Indexed: 12/11/2022] Open
Abstract
Objective Promotion of endogenous β-cell mass expansion could facilitate regeneration in patients with diabetes. We discovered that the secreted protein CTGF (aka CCN2) promotes adult β-cell replication and mass regeneration after injury via increasing β-cell immaturity and shortening the replicative refractory period. However, the mechanism of CTGF-mediated β-cell proliferation is unknown. Here we focused on whether CTGF alters cells of the immune system to enhance β-cell replication. Methods Using mouse models for 50% β-cell ablation and conditional, β-cell-specific CTGF induction, we assessed changes in immune cell populations by performing immunolabeling and gene expression analyses. We tested the requirement for macrophages in CTGF-mediated β-cell proliferation via clodronate-based macrophage depletion. Results CTGF induction after 50% β-cell ablation increased both macrophages and T-cells in islets. An upregulation in the expression of several macrophage and T-cell chemoattractant genes was also observed in islets. Gene expression analyses suggest an increase in M1 and a decrease in M2 macrophage markers. Depletion of macrophages (without changes in T cell number) blocked CTGF-mediated β-cell proliferation and prevented the increase in β-cell immaturity. Conclusions Our data show that macrophages are critical for CTGF-mediated adult β-cell proliferation in the setting of partial β-cell ablation. This is the first study to link a specific β-cell proliferative factor with immune-mediated β-cell proliferation in a β-cell injury model. Following partial beta cell ablation, induction of CTGF in beta cells increases the number of pancreatic macrophages and T cells. Increased macrophages are found preferentially within islets. Expression of genes involved in macrophage and T cell chemoattraction is increased in islets following CTGF induction. Depletion of macrophages completely abrogates CTGF-mediated increases in beta cell proliferation after injury.
Collapse
Affiliation(s)
- Kimberly G. Riley
- Department of Cell and Developmental Biology at Vanderbilt University, Nashville, TN, USA
| | - Raymond C. Pasek
- Department of Medicine at Vanderbilt University, Nashville, TN, USA
| | | | - Jennifer C. Dunn
- Department of Medicine at Vanderbilt University, Nashville, TN, USA
| | - W. Reid Bolus
- Department of Veterans Affairs, Tennessee Valley Health Authority, Nashville, TN, USA
- Departments of Molecular Physiology and Biophysics at Vanderbilt University, Nashville, TN, USA
| | - Peggy L. Kendall
- Department of Medicine at Vanderbilt University, Nashville, TN, USA
| | - Alyssa H. Hasty
- Department of Veterans Affairs, Tennessee Valley Health Authority, Nashville, TN, USA
- Departments of Molecular Physiology and Biophysics at Vanderbilt University, Nashville, TN, USA
| | - Maureen Gannon
- Department of Veterans Affairs, Tennessee Valley Health Authority, Nashville, TN, USA
- Department of Cell and Developmental Biology at Vanderbilt University, Nashville, TN, USA
- Department of Medicine at Vanderbilt University, Nashville, TN, USA
- Departments of Molecular Physiology and Biophysics at Vanderbilt University, Nashville, TN, USA
- Corresponding author. Department of Medicine, Division of Diabetes, Endocrinology, and Metabolism, 2213 Garland Avenue 7465 MRBIV, Vanderbilt University, Nashville, Tennessee, TN 37232-0475, USA. Tel.: +1 615 936 2676; fax: +1 615 936 1667.
| |
Collapse
|
190
|
Larynx carcinoma regulates tumor-associated macrophages through PLGF signaling. Sci Rep 2015; 5:10071. [PMID: 25961789 PMCID: PMC4650800 DOI: 10.1038/srep10071] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Accepted: 03/30/2015] [Indexed: 12/22/2022] Open
Abstract
Cancer neovascularization plays an essential role in the metastasis of larynx carcinoma (LC). However, the underlying molecular mechanisms are not completely understood. Recently, we reported that placental growth factor (PLGF) regulates expression of matrix metalloproteinase 3 (MMP3) through ERK/MAPK signaling pathway in LC. Here, we show that MMP9 upregulated in LC, and appeared to be mainly produced by M2 macrophages (tumor-associated macrophages (TAM)). In a transwell co-culture system, PLGF secreted by LC cells triggered macrophage polarization to a TAM subtype that releases MMP9. Moreover, MMP9 was found to be activated in the PLGF-polarized TAM via transforming growth factor β (TGFβ) receptor signaling activation. Furthermore, PLGF in LC cells induced macrophage polarization in vivo, and significantly promoted the growth of LC. Thus, together with our previous work, our study highlights a pivotal role of cross-talk between TAM and LC in regulating the metastasis of LC.
Collapse
|
191
|
Zhao Z, Xi H, Xu D, Li C. Transforming growth factor β receptor signaling restrains growth of pancreatic carcinoma cells. Tumour Biol 2015; 36:7711-6. [PMID: 25934336 DOI: 10.1007/s13277-015-3466-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2015] [Accepted: 04/15/2015] [Indexed: 01/04/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is extremely malignant. Efficient control of cancer growth may substantially improve the survival of PDAC patients. However, no efficient treatments are so far available. Here, we inhibited transforming growth factor β (TGFβ) receptor signaling by overexpression of a key inhibitor of this pathway, SMAD7, in the mouse pancreas, using a recently developed intraductal infusion method. Overexpression of SMAD7 significantly increased growth of both implanted PDAC and PDAC by K-ras modification. Our data thus suggest that TGFβ receptor signaling restrains growth of PDAC, and modulation of TGFβ receptor signaling may be an effective treatment for PDAC.
Collapse
Affiliation(s)
- Zhiming Zhao
- Department of Surgical Oncology, Chinese PLA General Hospital, Beijing, 100853, China. .,Department of Surgical Oncology, Chinese PLA General Hospital, 28 Fuxing Road, Beijing, 100853, China.
| | - Hao Xi
- Department of Hepatobiliary Surgery, The Hospital of Shunyi District, Beijing, 101300, China
| | - Dabin Xu
- Department of Surgical Oncology, Chinese PLA General Hospital, Beijing, 100853, China
| | - Chenggang Li
- Department of Surgical Oncology, Chinese PLA General Hospital, Beijing, 100853, China
| |
Collapse
|
192
|
Lei C, Zhou X, Pang Y, Mao Y, Lu X, Li M, Zhang J. TGF-β signalling prevents pancreatic beta cell death after proliferation. Cell Prolif 2015; 48:356-62. [PMID: 25871744 DOI: 10.1111/cpr.12183] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 12/11/2014] [Indexed: 10/23/2022] Open
Abstract
OBJECTIVES Maintenance of functional beta cell mass is critical for prevention of diabetes. The transforming growth factor-beta (TGF-β) receptor signalling pathway plays an essential role in pancreatic development. However, its involvement in control of post-natal beta cell growth has only been recently reported. MATERIALS AND METHODS Here, we studied the role of TGF-β receptor signalling in beta cell proliferation after 50% partial pancreatectomy (PPx), using beta cell-specific TGF-β receptor II (TBR2)-mutated mice. RESULTS Consistent with previous reports, we found that inhibition of TGF-β receptor signalling in beta cells resulted in slightly higher beta cell mass 1 week after PPx, due to greater beta cell proliferation. However, beta cell mass in these beta cell-specific TBR2-mutated mice significantly decreased by 12 weeks after PPx, resulting from increase in beta cell apoptosis. CONCLUSIONS Our data thus suggest that TGF-β receptor signalling may be required for prevention of beta cell death after proliferation, and highlight this pathway as an essential regulator during post-natal beta cell homoeostasis.
Collapse
Affiliation(s)
- Chen Lei
- Xi'an Jiaotong University Medical School, Department of Physiology, Xian, 710061, China; Department of Endocrinology, The General Hospital of Ningxia Medical University, Yinchuan, 750004, China
| | | | | | | | | | | | | |
Collapse
|
193
|
Van Gassen N, Staels W, Van Overmeire E, De Groef S, Sojoodi M, Heremans Y, Leuckx G, Van de Casteele M, Van Ginderachter JA, Heimberg H, De Leu N. Concise Review: Macrophages: Versatile Gatekeepers During Pancreatic β-Cell Development, Injury, and Regeneration. Stem Cells Transl Med 2015; 4:555-63. [PMID: 25848123 DOI: 10.5966/sctm.2014-0272] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Accepted: 02/16/2015] [Indexed: 02/07/2023] Open
Abstract
UNLABELLED Macrophages are classically considered detrimental for pancreatic β-cell survival and function, thereby contributing to β-cell failure in both type 1 (T1D) and 2 (T2D) diabetes mellitus. In addition, adipose tissue macrophages negatively influence peripheral insulin signaling and promote obesity-induced insulin resistance in T2D. In contrast, recent data unexpectedly uncovered that macrophages are not only able to protect β cells during pancreatitis but also to orchestrate β-cell proliferation and regeneration after β-cell injury. Moreover, by altering their activation state, macrophages are able to improve insulin resistance in murine models of T2D. This review will elaborate on current insights in macrophage heterogeneity and on the evolving role of pancreas macrophages during organogenesis, tissue injury, and repair. Additional identification of macrophage subtypes and of their secreted factors might ultimately translate into novel therapeutic strategies for both T1D and T2D. SIGNIFICANCE Diabetes mellitus is a pandemic disease, characterized by severe acute and chronic complications. Macrophages have long been considered prime suspects in the pathogenesis of both type 1 and 2 diabetes mellitus. In this concise review, current insights in macrophage heterogeneity and on the, as yet, underappreciated role of alternatively activated macrophages in insulin sensing and β-cell development/repair are reported. Further identification of macrophage subtypes and of their secreted factors might ultimately translate into novel therapeutic strategies for diabetes mellitus.
Collapse
Affiliation(s)
- Naomi Van Gassen
- Diabetes Research Center and Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium; Division of Pediatric Endocrinology, Department of Pediatrics, Ghent University Hospital, and Department of Pediatrics and Genetics, Ghent University, Ghent, Belgium; Myeloid Cell Immunology Laboratory, VIB, Brussels, Belgium; Department of Endocrinology, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Willem Staels
- Diabetes Research Center and Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium; Division of Pediatric Endocrinology, Department of Pediatrics, Ghent University Hospital, and Department of Pediatrics and Genetics, Ghent University, Ghent, Belgium; Myeloid Cell Immunology Laboratory, VIB, Brussels, Belgium; Department of Endocrinology, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Eva Van Overmeire
- Diabetes Research Center and Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium; Division of Pediatric Endocrinology, Department of Pediatrics, Ghent University Hospital, and Department of Pediatrics and Genetics, Ghent University, Ghent, Belgium; Myeloid Cell Immunology Laboratory, VIB, Brussels, Belgium; Department of Endocrinology, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Sofie De Groef
- Diabetes Research Center and Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium; Division of Pediatric Endocrinology, Department of Pediatrics, Ghent University Hospital, and Department of Pediatrics and Genetics, Ghent University, Ghent, Belgium; Myeloid Cell Immunology Laboratory, VIB, Brussels, Belgium; Department of Endocrinology, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Mozhdeh Sojoodi
- Diabetes Research Center and Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium; Division of Pediatric Endocrinology, Department of Pediatrics, Ghent University Hospital, and Department of Pediatrics and Genetics, Ghent University, Ghent, Belgium; Myeloid Cell Immunology Laboratory, VIB, Brussels, Belgium; Department of Endocrinology, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Yves Heremans
- Diabetes Research Center and Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium; Division of Pediatric Endocrinology, Department of Pediatrics, Ghent University Hospital, and Department of Pediatrics and Genetics, Ghent University, Ghent, Belgium; Myeloid Cell Immunology Laboratory, VIB, Brussels, Belgium; Department of Endocrinology, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Gunter Leuckx
- Diabetes Research Center and Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium; Division of Pediatric Endocrinology, Department of Pediatrics, Ghent University Hospital, and Department of Pediatrics and Genetics, Ghent University, Ghent, Belgium; Myeloid Cell Immunology Laboratory, VIB, Brussels, Belgium; Department of Endocrinology, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Mark Van de Casteele
- Diabetes Research Center and Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium; Division of Pediatric Endocrinology, Department of Pediatrics, Ghent University Hospital, and Department of Pediatrics and Genetics, Ghent University, Ghent, Belgium; Myeloid Cell Immunology Laboratory, VIB, Brussels, Belgium; Department of Endocrinology, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Jo A Van Ginderachter
- Diabetes Research Center and Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium; Division of Pediatric Endocrinology, Department of Pediatrics, Ghent University Hospital, and Department of Pediatrics and Genetics, Ghent University, Ghent, Belgium; Myeloid Cell Immunology Laboratory, VIB, Brussels, Belgium; Department of Endocrinology, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Harry Heimberg
- Diabetes Research Center and Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium; Division of Pediatric Endocrinology, Department of Pediatrics, Ghent University Hospital, and Department of Pediatrics and Genetics, Ghent University, Ghent, Belgium; Myeloid Cell Immunology Laboratory, VIB, Brussels, Belgium; Department of Endocrinology, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Nico De Leu
- Diabetes Research Center and Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium; Division of Pediatric Endocrinology, Department of Pediatrics, Ghent University Hospital, and Department of Pediatrics and Genetics, Ghent University, Ghent, Belgium; Myeloid Cell Immunology Laboratory, VIB, Brussels, Belgium; Department of Endocrinology, Universitair Ziekenhuis Brussel, Brussels, Belgium
| |
Collapse
|
194
|
Ginsenoside Rh2 inhibits metastasis of glioblastoma multiforme through Akt-regulated MMP13. Tumour Biol 2015; 36:6789-95. [PMID: 25835975 DOI: 10.1007/s13277-015-3387-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2015] [Accepted: 03/24/2015] [Indexed: 02/01/2023] Open
Abstract
Glioblastoma multiforme (GBM) is the most malignant type of primary brain tumor. Although the growth of the tumor cells in a relatively closed space may partially account for its malignancy, highly invasive nature of glioblastoma cells has been suggested to be the main reason for the failure of current therapeutic approaches. Ginsenoside Rh2 (GRh2) has recently been shown to significantly suppress the growth and survival of GBM through inhibiting epidermal growth factor receptor signaling, whereas its effects on the invasion and metastasis have not been examined. Here, we showed that GRh2 dose-dependently decreased GBM cell invasiveness in both scratch wound healing assay and Transwell cell migration assay. Moreover, the inhibitory effects of GRh2 on cell migration seemed to be conducted through decreased expression of matrix metalloproteinase (MMP)-13. Furthermore, using specific inhibitors, we found that GRh2 inhibited MMP13 through PI3k/Akt signaling pathway. Finally, high MMP13 levels were detected in GBM specimen from the patients. Together, these data suggest that GRh2 may suppress GBM migration through inhibiting Akt-mediated MMP13 activation. Thus, our data highlight a previous unappreciated role for GRh2 in suppressing GBM cell metastasis.
Collapse
|
195
|
Song L, Wang P, Tian Y, Chang D, Li K, Fan Y, Shen J, Du H, Mi R, Bian X, Tang X. Lung metastasis of pancreatic carcinoma is regulated by TGFβ signaling. Tumour Biol 2015; 36:2271-6. [PMID: 25409617 DOI: 10.1007/s13277-014-2834-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2014] [Accepted: 11/10/2014] [Indexed: 12/23/2022] Open
Abstract
The molecular regulation of the lung metastasis of pancreatic carcinoma (PCC) is not completely understood. Here, we show that the levels of phosphorylated SMAD3, ZEB1, ZEB2, Snail1, and Snail2 were significantly higher in PCC with lung metastasis than in PCC without lung metastasis. Overexpression of TGFβ1 enhanced the invasiveness of PCC cells, while inhibition of TGFβ1 decreased the invasiveness of PCC cells, which appeared to be conducted by activated TGFβ receptor signaling-induced upregulation of ZEB1, ZEB2, Snail1, and Snail2, suggesting a process of epithelial-mesenchymal transition (EMT). Taken together, our study provides evidence that TGFβ receptor signaling-induced EMT may be responsible for the increased PCC invasiveness to enhance its lung metastasis.
Collapse
Affiliation(s)
- Liqiang Song
- Department of Pathology, Institute of Basic Medical Sciences and School of Basic Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, 5 Dongdansantiao, 100005, Beijing, China,
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
196
|
El Khatib MM, Sakuma T, Tonne JM, Mohamed MS, Holditch SJ, Lu B, Kudva YC, Ikeda Y. β-Cell-targeted blockage of PD1 and CTLA4 pathways prevents development of autoimmune diabetes and acute allogeneic islets rejection. Gene Ther 2015; 22:430-8. [PMID: 25786871 DOI: 10.1038/gt.2015.18] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Revised: 01/07/2015] [Accepted: 02/12/2015] [Indexed: 12/21/2022]
Abstract
Protection of β cells from autoimmune destruction potentially cures type 1 diabetes mellitus (T1D). During antigen presentation, interactions between cytotoxic T-lymphocyte antigen-4 (CTLA4) and B7 molecules, or programmed death 1 (PD1) and its ligand PDL1, negatively regulate immune responses in a non-redundant manner. Here we employed β-cell-targeted adeno-associated virus serotype 8 (AAV8)-based vectors to overexpress an artificial PDL1-CTLA4Ig polyprotein or interleukin 10 (IL10). β-Cell-targeted expression of PDL1-CTLA4Ig or IL10 preserved β-cell mass and protected NOD mice from T1D development. When NOD mice were treated with vectors at early onset of hyperglycemia, PDL1-CTLA4Ig or IL10 alone failed to normalize the early onset of hyperglycemia. When drug-induced diabetic mice received major histocompatibility complex (MHC)-matched allo-islets, with or without pretreatment of the PDL1-CTLA4Ig-expressing vector, PDL1-CTLA4Ig-expressing islets were protected from rejection for at least 120 days. Similarly, transplantation of PDL1-CTLA4Ig-expressing MHC-matched islets into mice with established T1D resulted in protection of allo-islets from acute rejection, although islet grafts were eventually rejected. Thus the present study demonstrates the potent immuno-suppressive effects of β-cell-targeted PDL1-CTLA4Ig overexpression against T1D development and allo-islet rejection. The gene-based simultaneous inhibition of PD1 and CTLA4 pathways provides a unique strategy for immunosuppression-free tissue/organ transplantation, especially in the setting of no established autoimmunity.
Collapse
Affiliation(s)
- M M El Khatib
- Department of Molecular Medicine, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - T Sakuma
- Department of Molecular Medicine, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - J M Tonne
- Department of Molecular Medicine, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - M S Mohamed
- Department of Molecular Medicine, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - S J Holditch
- Department of Molecular Medicine, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - B Lu
- Department of Molecular Medicine, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Y C Kudva
- Division of Endocrinology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Y Ikeda
- Department of Molecular Medicine, Mayo Clinic College of Medicine, Rochester, MN, USA
| |
Collapse
|
197
|
Van Gassen N, Van Overmeire E, Leuckx G, Heremans Y, De Groef S, Cai Y, Elkrim Y, Gysemans C, Stijlemans B, Van de Casteele M, De Baetselier P, De Leu N, Heimberg H, Van Ginderachter JA. Macrophage dynamics are regulated by local macrophage proliferation and monocyte recruitment in injured pancreas. Eur J Immunol 2015; 45:1482-93. [PMID: 25645754 DOI: 10.1002/eji.201445013] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Revised: 01/13/2015] [Accepted: 01/27/2015] [Indexed: 12/24/2022]
Abstract
Pancreas injury by partial duct ligation (PDL) activates a healing response, encompassing β-cell neogenesis and proliferation. Macrophages (MΦs) were recently shown to promote β-cell proliferation after PDL, but they remain poorly characterized. We assessed myeloid cell diversity and the factors driving myeloid cell dynamics following acute pancreas injury by PDL. In naive and sham-operated pancreas, the myeloid cell compartment consisted mainly of two distinct tissue-resident MΦ types, designated MHC-II(lo) and MHC-II(hi) MΦs, the latter being predominant. MHC-II(lo) and MHC-II(hi) pancreas MΦs differed at the molecular level, with MHC-II(lo) MΦs being more M2-activated. After PDL, there was an early surge of Ly6C(hi) monocyte infiltration in the pancreas, followed by a transient MHC-II(lo) MΦ peak and ultimately a restoration of the MHC-II(hi) MΦ-dominated steady-state equilibrium. These intricate MΦ dynamics in PDL pancreas depended on monocyte recruitment by C-C chemokine receptor 2 and macrophage-colony stimulating factor receptor as well as on macrophage-colony stimulating factor receptor-dependent local MΦ proliferation. Functionally, MHC-II(lo) MΦs were more angiogenic. We further demonstrated that, at least in C-C chemokine receptor 2-KO mice, tissue MΦs, rather than Ly6C(hi) monocyte-derived MΦs, contributed to β-cell proliferation. Together, our study fully characterizes the MΦ subsets in the pancreas and clarifies the complex dynamics of MΦs after PDL injury.
Collapse
Affiliation(s)
- Naomi Van Gassen
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Eva Van Overmeire
- Myeloid Cell Immunology Laboratory, VIB, Brussels, Belgium.,Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Gunter Leuckx
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Yves Heremans
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Sofie De Groef
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Ying Cai
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Yvon Elkrim
- Myeloid Cell Immunology Laboratory, VIB, Brussels, Belgium.,Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Conny Gysemans
- Laboratory of Clinical and Experimental Endocrinology, Department of Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium
| | - Benoît Stijlemans
- Myeloid Cell Immunology Laboratory, VIB, Brussels, Belgium.,Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | | | - Patrick De Baetselier
- Myeloid Cell Immunology Laboratory, VIB, Brussels, Belgium.,Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Nico De Leu
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Harry Heimberg
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Jo A Van Ginderachter
- Myeloid Cell Immunology Laboratory, VIB, Brussels, Belgium.,Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
198
|
Wei Y, Tian Q, Zhao X, Wang X. Serum transforming growth factor beta 3 predicts future development of nonalcoholic fatty liver disease. Int J Clin Exp Med 2015; 8:4545-4550. [PMID: 26064382 PMCID: PMC4443216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2014] [Accepted: 02/03/2015] [Indexed: 06/04/2023]
Abstract
Transforming growth factor beta 3 (TGFb3) was mainly expressed by liver satellite cells in the normal liver, but it may be expressed by various liver cells during liver diseases, e.g. hepatitis and cirrhosis. However, whether TGFb3 level may be used to predict development of nonalcoholic fatty liver disease (NAFLD) has not been investigated before. Here we evaluated the relationship between TGFb3 and the susceptibility for developing NAFLD by comparing the incidence rates of developing NAFLD and serum TGFb3 levels in 1322 healthy subjects without other risk factors during a 4-year period. These healthy subjects were grouped into tertiles based on their serum TGFb3 levels that were measured in 2009. After 4 years, the odds ratios (ORs) of NAFLD development were analyzed based on the tertiles of TGFb3 levels in 2013. The cumulative incidence of NAFLD was 25.3% (334/1322) after four years. The NAFLD-developing group had higher serum TGFb3 levels in 2009 than those in the group that did not develop NAFLD (554±287 pg/ml vs. 285±173 pg/ml; P=0.002). When the serum TGFb3 levels in 2009 were grouped into tertiles, we found that the incidence of NAFLD in 2013 was significantly higher with increasing tertiles (6.3%, 38.0%, and 55.7%, respectively; P<0.05). Thus, our study demonstrate that higher serum TGFb3 levels in subjects devoid of NAFLD may have a higher chance of its future development, and highlight serum TGFb3 level as a novel predictor for development of NAFLD.
Collapse
Affiliation(s)
- Yongli Wei
- Department of Infectious Diseases, Linyi People’s HospitalLinyi 276000, Shandong Province, China
| | - Qing Tian
- Department of Infectious Diseases, Linyi People’s HospitalLinyi 276000, Shandong Province, China
| | - Xiuxia Zhao
- Department of Infectious Diseases, Linyi People’s HospitalLinyi 276000, Shandong Province, China
| | - Xingchun Wang
- Department of Internal Medicine, Linyi Tumor HospitalLinyi 276000, Shandong Province, China
| |
Collapse
|
199
|
Li J, Pan Y, Zhang B, Chen Q. Macrophages are needed in the progression of tuberculosis into lung cancer. Tumour Biol 2015; 36:6063-6. [DOI: 10.1007/s13277-015-3283-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Accepted: 02/18/2015] [Indexed: 11/30/2022] Open
|
200
|
Jian H, Zhao Y, Liu B, Lu S. SEMA4B inhibits growth of non-small cell lung cancer in vitro and in vivo. Cell Signal 2015; 27:1208-13. [PMID: 25746385 DOI: 10.1016/j.cellsig.2015.02.027] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Revised: 02/17/2015] [Accepted: 02/26/2015] [Indexed: 01/03/2023]
Abstract
We have recently shown that Semaphorin 4B (SEMA4B) inhibits the invasion of non-small cell lung cancer (NSCLC) through PI3K-dependent suppression of MMP9 activation. In the current study, we evaluated whether SEMA4B may also affect the growth of NSCLC. We thus used two human NSCLC lines, A549 and Calu-3, to examine our hypothesis. We found that overexpression of SEMA4B significantly decreased NSCLC cell growth, while SEMA4B inhibition significantly increased NSCLC cell growth, both in vitro and in vivo in an implanted NSCLC model. Adaptation of SEMA4B in NSCLC cells did not alter cell apoptosis, but changed the cell proliferation. Further analyses show that SEMA4B may induce FoxO1 nuclear retention through suppressing PI3K/Akt signaling pathway, which subsequently inhibited cell growth through the direct nuclear target of FoxO1, p21. Our study thus demonstrate a role of SEMA4B in suppressing NSCLC growth, besides its role in inhibiting cell metastasis, and highlights SEMA4B as a promising therapeutic target for NSCLC.
Collapse
Affiliation(s)
- Hong Jian
- Shanghai Lung Cancer Center, Shanghai Chest Hospital Affiliated to Shanghai Jiaotong University, Shanghai, China
| | - Yi Zhao
- Shanghai Lung Cancer Center, Shanghai Chest Hospital Affiliated to Shanghai Jiaotong University, Shanghai, China
| | - Bin Liu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Shun Lu
- Shanghai Lung Cancer Center, Shanghai Chest Hospital Affiliated to Shanghai Jiaotong University, Shanghai, China.
| |
Collapse
|