1
|
Wood TR, Kucinski I, Voiculescu O. Distinct molecular profile of the chick organizer as a stem zone during axial elongation. Open Biol 2024; 14:240139. [PMID: 38955223 PMCID: PMC11519809 DOI: 10.1098/rsob.240139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 06/04/2024] [Accepted: 06/07/2024] [Indexed: 07/04/2024] Open
Abstract
The vertebrate organizer plays a crucial role in building the main (antero-posterior) axis of the embryo: it neuralizes the surrounding ectoderm, and is the site of emigration for cells making axial and paraxial mesendoderm during elongation. The chick organizer becomes a stem zone at the onset of elongation; it stops recruiting cells from the neighbouring ectoderm and generates all its derivatives from the small number of resident cells it contains at the end of gastrulation stages. Nothing is known about the molecular identity of this stem zone. Here, we specifically labelled long-term resident cells of the organizer and compared their RNA-seq profile to that of the neighbouring cell populations. Screening by reverse transcription-polymerase chain reaction and in situ hybridization identified four genes (WIF1, PTGDS, ThPO and UCKL1) that are upregulated only in the organizer region when it becomes a stem zone and remain expressed there during axial elongation. In experiments specifically labelling the resident cells of the mature organizer, we show that only these cells express these genes. These findings molecularly define the organizer as a stem zone and offer a key to understanding how this zone is set up, the molecular control of its cells' behaviour and the evolution of axial growth zones.
Collapse
Affiliation(s)
- Timothy R. Wood
- Department of Physiology, Development and Neuroscience, University of Cambridge, CambridgeCB2 3DY, UK
| | - Iwo Kucinski
- Department of Physiology, Development and Neuroscience, University of Cambridge, CambridgeCB2 3DY, UK
| | - Octavian Voiculescu
- Department of Physiology, Development and Neuroscience, University of Cambridge, CambridgeCB2 3DY, UK
| |
Collapse
|
2
|
Li J, Liu Y, Zhang R, Yang Q, Xiong W, He Y, Ye Q. Insights into the role of mesenchymal stem cells in cutaneous medical aesthetics: from basics to clinics. Stem Cell Res Ther 2024; 15:169. [PMID: 38886773 PMCID: PMC11184751 DOI: 10.1186/s13287-024-03774-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 05/27/2024] [Indexed: 06/20/2024] Open
Abstract
With the development of the economy and the increasing prevalence of skin problems, cutaneous medical aesthetics are gaining more and more attention. Skin disorders like poor wound healing, aging, and pigmentation have an impact not only on appearance but also on patients with physical and psychological issues, and even impose a significant financial burden on families and society. However, due to the complexities of its occurrence, present treatment options cannot produce optimal outcomes, indicating a dire need for new and effective treatments. Mesenchymal stem cells (MSCs) and their secretomics treatment is a new regenerative medicine therapy that promotes and regulates endogenous stem cell populations and/or replenishes cell pools to achieve tissue homeostasis and regeneration. It has demonstrated remarkable advantages in several skin-related in vivo and in vitro investigations, aiding in the improvement of skin conditions and the promotion of skin aesthetics. As a result, this review gives a complete description of recent scientific breakthroughs in MSCs for skin aesthetics and the limitations of their clinical applications, aiming to provide new ideas for future research and clinical transformation.
Collapse
Affiliation(s)
- Junyi Li
- Center of Regenerative Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Ye Liu
- Center of Regenerative Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Rui Zhang
- Center of Regenerative Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Qianyu Yang
- Center of Regenerative Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Wei Xiong
- Center of Regenerative Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Yan He
- Institute of Regenerative and Translational Medicine, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, 430030, China.
| | - Qingsong Ye
- Center of Regenerative Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
3
|
Humenik F, Danko J, Krešáková L, Vdoviaková K, Vrabec V, Vasilová E, Giretová M, Tóth Š, Fagová Z, Babík J, Medvecký Ľ. A Chitosan-Based Biomaterial Combined with Mesenchymal Stem Cell-Conditioned Medium for Wound Healing and Skin Regeneration. Int J Mol Sci 2023; 24:16080. [PMID: 38003269 PMCID: PMC10671656 DOI: 10.3390/ijms242216080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 11/03/2023] [Accepted: 11/06/2023] [Indexed: 11/26/2023] Open
Abstract
The aim of this study was to provide a beneficial treatment effect of novel chitosan bio-polymeric material enriched with mesenchymal stem cell products derived from the canine adipose tissue (AT-MSC) on the artificial skin defect in a rabbit model. For the objectivity of the regeneration evaluation, we used histological analysis and a scoring system created by us, taking into account all the attributes of regeneration, such as inflammatory reaction, necrosis, granulation, formation of individual skin layers and hair follicles. We observed an acceleration and improvement in the healing of an artificially created skin defect after eight and ten weeks in comparison with negative control (spontaneous healing without biomaterial). Moreover, we were able to described hair follicles and epidermis layer in histological skin samples treated with a chitosan-based biomaterial on the eighth week after grafting.
Collapse
Affiliation(s)
- Filip Humenik
- Department of Morphological Sciences, University of Veterinary Medicine and Pharmacy in Košice, 041 81 Košice, Slovakia
| | - Ján Danko
- Department of Morphological Sciences, University of Veterinary Medicine and Pharmacy in Košice, 041 81 Košice, Slovakia
- Educational, Scientific and Research Institute AGEL, 811 06 Bratislava, Slovakia
| | - Lenka Krešáková
- Department of Morphological Sciences, University of Veterinary Medicine and Pharmacy in Košice, 041 81 Košice, Slovakia
| | - Katarína Vdoviaková
- Department of Morphological Sciences, University of Veterinary Medicine and Pharmacy in Košice, 041 81 Košice, Slovakia
| | - Vladimír Vrabec
- Clinic of Birds, Exotic and Free Living Animals, University of Veterinary Medicine and Pharmacy in Košice, 041 81 Košice, Slovakia
| | - Emília Vasilová
- Clinic of Birds, Exotic and Free Living Animals, University of Veterinary Medicine and Pharmacy in Košice, 041 81 Košice, Slovakia
| | - Mária Giretová
- Division of Functional and Hybrid Systems, Institute of Materials Research of SAS, 040 01 Košice, Slovakia
| | - Štefan Tóth
- Department of Histology and Embryology, University of Pavol Jozef Šafárik, 041 80 Košice, Slovakia
| | - Zuzana Fagová
- Department of Histology and Embryology, University of Pavol Jozef Šafárik, 041 80 Košice, Slovakia
| | - Ján Babík
- Clinic of Burns and Reconstructive Medicine, AGEL Hospital, 040 15 Košice-Šaca, Slovakia
| | - Ľubomír Medvecký
- Department of Morphological Sciences, University of Veterinary Medicine and Pharmacy in Košice, 041 81 Košice, Slovakia
- Division of Functional and Hybrid Systems, Institute of Materials Research of SAS, 040 01 Košice, Slovakia
| |
Collapse
|
4
|
Andalib E, Kashfi M, Mahmoudvand G, Rezaei E, Mahjoor M, Torki A, Afkhami H. Application of hypoxia-mesenchymal stem cells in treatment of anaerobic bacterial wound infection: wound healing and infection recovery. Front Microbiol 2023; 14:1251956. [PMID: 37869672 PMCID: PMC10586055 DOI: 10.3389/fmicb.2023.1251956] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 09/18/2023] [Indexed: 10/24/2023] Open
Abstract
Mesenchymal stromal cells, commonly referred to as MSCs, are a type of multipotent stem cells that are typically extracted from adipose tissue and bone marrow. In the field of tissue engineering and regenerative medicine, MSCs and their exosomes have emerged as revolutionary tools. Researchers are now devoting greater attention to MSCs because of their ability to generate skin cells like fibroblasts and keratinocytes, as well as their distinctive potential to decrease inflammation and emit pro-angiogenic molecules at the site of wounds. More recent investigations revealed that MSCs can exert numerous direct and indirect antimicrobial effects that are immunologically mediated. Collectively, these antimicrobial properties can remove bacterial infections when the MSCs are delivered in a therapeutic setting. Regardless of the positive therapeutic potential of MSCs for a multitude of conditions, transplanted MSC cell retention continues to be a major challenge. Since MSCs are typically administered into naturally hypoxic tissues, understanding the impact of hypoxia on the functioning of MSCs is crucial. Hypoxia has been postulated to be among the factors determining the differentiation of MSCs, resulting in the production of inflammatory cytokines throughout the process of tissue regeneration and wound repair. This has opened new horizons in developing MSC-based systems as a potent therapeutic tool in oxygen-deprived regions, including anaerobic wound infection sites. This review sheds light on the role of hypoxia-MSCs in the treatment of anaerobic bacterial wound infection in terms of both their regenerative and antimicrobial activities.
Collapse
Affiliation(s)
- Elahe Andalib
- Department of Microbiology, School of Basic Sciences, Islamic Azad University Science and Research Branch, Tehran, Iran
| | - Mojtaba Kashfi
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
- Department of Medical Microbiology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Golnaz Mahmoudvand
- Student Research Committee, USERN Office, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Elaheh Rezaei
- Department of Microbiology, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Mohamad Mahjoor
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
- Department of Immunology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Alireza Torki
- Department of Medical Microbiology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Medical Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Hamed Afkhami
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
- Department of Medical Microbiology, Faculty of Medicine, Shahed University, Tehran, Iran
| |
Collapse
|
5
|
Balamurugan K, Poria DK, Sehareen SW, Krishnamurthy S, Tang W, McKennett L, Padmanaban V, Czarra K, Ewald AJ, Ueno NT, Ambs S, Sharan S, Sterneck E. Stabilization of E-cadherin adhesions by COX-2/GSK3β signaling is a targetable pathway in metastatic breast cancer. JCI Insight 2023; 8:156057. [PMID: 36757813 PMCID: PMC10070121 DOI: 10.1172/jci.insight.156057] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 02/07/2023] [Indexed: 02/10/2023] Open
Abstract
Metastatic progression of epithelial cancers can be associated with epithelial-mesenchymal transition (EMT) including transcriptional inhibition of E-cadherin (CDH1) expression. Recently, EM plasticity (EMP) and E-cadherin-mediated, cluster-based metastasis and treatment resistance have become more appreciated. However, the mechanisms that maintain E-cadherin expression in this context are less understood. Through studies of inflammatory breast cancer (IBC) and a 3D tumor cell "emboli" culture paradigm, we discovered that cyclooxygenase 2 (COX-2; PTGS2), a target gene of C/EBPδ (CEBPD), or its metabolite prostaglandin E2 (PGE2) promotes protein stability of E-cadherin, β-catenin, and p120 catenin through inhibition of GSK3β. The COX-2 inhibitor celecoxib downregulated E-cadherin complex proteins and caused cell death. Coexpression of E-cadherin and COX-2 was seen in breast cancer tissues from patients with poor outcome and, along with inhibitory GSK3β phosphorylation, in patient-derived xenografts (PDX) including triple negative breast cancer (TNBC).Celecoxib alone decreased E-cadherin protein expression within xenograft tumors, though CDH1 mRNA levels increased, and reduced circulating tumor cell (CTC) clusters. In combination with paclitaxel, celecoxib attenuated or regressed lung metastases. This study has uncovered a mechanism by which metastatic breast cancer cells can maintain E-cadherin-mediated cell-to-cell adhesions and cell survival, suggesting that some patients with COX-2+/E-cadherin+ breast cancer may benefit from targeting of the PGE2 signaling pathway.
Collapse
Affiliation(s)
- Kuppusamy Balamurugan
- Laboratory of Cell and Developmental Signaling, Center for Cancer Research (CCR), National Cancer Institute (NCI), Frederick, Maryland, USA
| | - Dipak K Poria
- Laboratory of Cell and Developmental Signaling, Center for Cancer Research (CCR), National Cancer Institute (NCI), Frederick, Maryland, USA
| | - Saadiya W Sehareen
- Laboratory of Cell and Developmental Signaling, Center for Cancer Research (CCR), National Cancer Institute (NCI), Frederick, Maryland, USA
| | - Savitri Krishnamurthy
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Wei Tang
- Laboratory of Human Carcinogenesis, CCR, NCI, Bethesda, Maryland, USA
| | - Lois McKennett
- Laboratory Animal Sciences Program, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Veena Padmanaban
- Departments of Cell Biology and Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kelli Czarra
- Laboratory Animal Sciences Program, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Andrew J Ewald
- Departments of Cell Biology and Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Naoto T Ueno
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Stefan Ambs
- Laboratory of Human Carcinogenesis, CCR, NCI, Bethesda, Maryland, USA
| | - Shikha Sharan
- Laboratory of Cell and Developmental Signaling, Center for Cancer Research (CCR), National Cancer Institute (NCI), Frederick, Maryland, USA
| | - Esta Sterneck
- Laboratory of Cell and Developmental Signaling, Center for Cancer Research (CCR), National Cancer Institute (NCI), Frederick, Maryland, USA
| |
Collapse
|
6
|
Huang Q, Liu X, Guo SW. Higher fibrotic content of endometriotic lesions is associated with diminished prostaglandin E2 signaling. Reprod Med Biol 2021; 21:e12423. [PMID: 34938147 PMCID: PMC8656679 DOI: 10.1002/rmb2.12423] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 10/12/2021] [Accepted: 10/21/2021] [Indexed: 12/12/2022] Open
Abstract
Purpose While the prevailing view holds that the prostaglandin E2 (PGE2) signaling plays a vital role in endometriosis, PGE2 also is known to be anti-fibrotic. We investigated the immunostaining of COX-2, EP2, and EP4, along with fibrotic content in ovarian endometrioma (OE) and deep endometriosis (DE) lesions, and in OE lesions from adolescent and adult patients. In addition, we evaluated the effect of substrate stiffness on the expression of COX-2, EP2, and EP4 in endometrial stromal cells. Methods Immunohistochemistry analysis of COX-2, EP2, and EP4, along with the quantification of lesional fibrosis, was conducted for OE and DE lesion samples and also OE lesion samples from adolescent and adult patients. The effect of substrate rigidity on fibroblast-to-myofibroblast transdifferentiation (FMT) and the expression of COX-2, EP2, and EP4, with or without TGF-β1 stimulation, were investigated. Results The immunostaining of COX-2, EP2, and EP4 was substantially reduced in endometriotic lesions as lesions became more fibrotic. Both TGF-β1 stimulation and stiff substrates induced FMT and reduced the expression of COX-2, EP2, and EP4. Conclusions Since fibrosis is a common feature of endometriosis, our results thus cast doubts on the use of therapeutics that suppresses the PGE2 signaling pathway, either by inhibiting COX-2 or EP2/EP4.
Collapse
Affiliation(s)
- Qingqing Huang
- Shanghai OB/GYN Hospital Fudan University Shanghai China.,The Third Affiliated Hospital of Guangzhou Medical University Guangzhou Guangdong China
| | - Xishi Liu
- Shanghai OB/GYN Hospital Fudan University Shanghai China.,Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases Fudan University Shanghai China
| | - Sun-Wei Guo
- Shanghai OB/GYN Hospital Fudan University Shanghai China.,Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases Fudan University Shanghai China
| |
Collapse
|
7
|
The Role of MSC in Wound Healing, Scarring and Regeneration. Cells 2021; 10:cells10071729. [PMID: 34359898 PMCID: PMC8305394 DOI: 10.3390/cells10071729] [Citation(s) in RCA: 199] [Impact Index Per Article: 49.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/02/2021] [Accepted: 07/03/2021] [Indexed: 02/06/2023] Open
Abstract
Tissue repair and regeneration after damage is not completely understood, and current therapies to support this process are limited. The wound healing process is associated with cell migration and proliferation, extracellular matrix remodeling, angiogenesis and re-epithelialization. In normal conditions, a wound will lead to healing, resulting in reparation of the tissue. Several risk factors, chronic inflammation, and some diseases lead to a deficient wound closure, producing a scar that can finish with a pathological fibrosis. Mesenchymal stem/stromal cells (MSCs) are widely used for their regenerative capacity and their possible therapeutically potential. Derived products of MSCs, such as exosomes or extravesicles, have shown a therapeutic potential similar to MSCs, and these cell-free products may be interesting in clinics. MSCs or their derivative products have shown paracrine beneficial effects, regulating inflammation, modifying the fibroblast activation and production of collagen and promoting neovascularization and re-epithelialization. This review describes the effects of MSCs and their derived products in each step of the wound repair process. As well, it reviews the pre-clinical and clinical use of MSCs to benefit in skin wound healing in diabetic associated wounds and in pathophysiological fibrosis.
Collapse
|
8
|
Colangelo MT, Galli C, Guizzardi S. The effects of polydeoxyribonucleotide on wound healing and tissue regeneration: a systematic review of the literature. Regen Med 2020; 15:1801-1821. [PMID: 32757710 DOI: 10.2217/rme-2019-0118] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Aim: The present study evaluated the effects of polydeoxyribonucleotide (PDRN) on tissue regeneration, paying special attention to the molecular mechanisms that underlie its tissue remodeling actions to better identify its effective therapeutic potential in wound healing. Materials & methods: Strategic searches were conducted through MEDLINE/PubMed, Google Scholar, Scopus, Web of Science and the Cochrane Central Register of Controlled Trials, from their earliest available dates to March 2020. The studies were included with the following eligibility criteria: studies evaluating tissue regeneration, and being an in vitro, in vivo and clinical study. Results: Out of more than 90 articles, 34 fulfilled the eligibility criteria. All data obtained proved the ability of PDRN in promoting a physiological tissue repair through salvage pathway and adenosine A2A receptor activation. Conclusion: Up to date PDRN has proved promising results in term of wound regeneration, healing time and absence of side effects.
Collapse
Affiliation(s)
- Maria T Colangelo
- Department of Medicine & Surgery, Histology & Embryology Lab, University of Parma, Parma, Italy
| | - Carlo Galli
- Department of Medicine & Surgery, University of Parma, Parma, Italy
| | - Stefano Guizzardi
- Department of Medicine & Surgery, Histology & Embryology Lab, University of Parma, Parma, Italy
| |
Collapse
|
9
|
Yamamoto S, Miyama T, Komoda T, Sugawara M, Nonomura M, Nakanishi J. A Facile Assay of Epithelial-mesenchymal Transition Based on Cooperativity Quantification of Cellular Autonomous Motions. ANAL SCI 2020; 36:263-267. [PMID: 31588066 DOI: 10.2116/analsci.19p233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Epithelial-mesenchymal transition (EMT), a qualitative change in cell migration behavior during cancer invasion and metastasis, is becoming a new target for anticancer drugs. Therefore, it is crucial to develop in vitro assays for the evaluation of the abilities of drug candidates to control EMT progression. We herein report on a method for the quantification of the EMT based on particle image velocimetry and correlation functions. The exponential fitting of the correlation curve gives an index (λ), which represents transforming growth factor (TGF)-β1-induced EMT progression and its suppression by inhibitors. Moreover, real-time monitoring of the λ value illustrates a time-dependent EMT progressing process, which occurs earlier than the bio-chemical changes in an EMT marker protein expression. The results demonstrate the usefulness of the present method for kinetic studies of EMT progression as well as EMT inhibitor screening.
Collapse
Affiliation(s)
- Shota Yamamoto
- International Center for Materials Nanoarchitectonics (WPI-MANA), National Institute for Materials Science (NIMS)
| | - Tatsuya Miyama
- International Center for Materials Nanoarchitectonics (WPI-MANA), National Institute for Materials Science (NIMS).,Department of Mathematical Information Engineering, College of Industrial Technology, Nihon University
| | - Takafumi Komoda
- Department of Mechanical Engineering, Graduate School of Engineering, Chiba University
| | - Michiko Sugawara
- Department of Mechanical Engineering, Graduate School of Engineering, Chiba University
| | - Makiko Nonomura
- Department of Mathematical Information Engineering, College of Industrial Technology, Nihon University
| | - Jun Nakanishi
- International Center for Materials Nanoarchitectonics (WPI-MANA), National Institute for Materials Science (NIMS).,Graduate School of Advanced Science and Engineering, Waseda University
| |
Collapse
|
10
|
Lee CH. Reversal of Epithelial-Mesenchymal Transition by Natural Anti-Inflammatory and Pro-Resolving Lipids. Cancers (Basel) 2019; 11:E1841. [PMID: 31766574 PMCID: PMC6966475 DOI: 10.3390/cancers11121841] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 11/16/2019] [Accepted: 11/19/2019] [Indexed: 02/08/2023] Open
Abstract
Epithelial mesenchymal transition (EMT) is a key process in the progression of malignant cancer. Therefore, blocking the EMT can be a critical fast track for the development of anticancer drugs. In this paper, we update recent research output of EMT and we explore suppression of EMT by natural anti-inflammatory compounds and pro-resolving lipids.
Collapse
Affiliation(s)
- Chang Hoon Lee
- College of Pharmacy, Dongguk University, Seoul 100-715, Korea
| |
Collapse
|
11
|
Chen X, Ge W, Hu J, Dong T, Yao H, Chen L, Geng B, Zhou H. Inhibition of prostaglandin E2 receptor 4 by lnc000908 to promote the endothelial-mesenchymal transition participation in cardiac remodelling. J Cell Mol Med 2019; 23:6355-6367. [PMID: 31297954 PMCID: PMC6714495 DOI: 10.1111/jcmm.14524] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 05/25/2019] [Accepted: 06/12/2019] [Indexed: 12/19/2022] Open
Abstract
Long non‐coding RNAs (lncRNAs) have emerged as potent regulators of cardiac disease; however, the role of lncRNA in cardiac fibrosis remains partially understood. In this study, we identified a cardiac endothelial‐enriched lncRNA‐lnc000908, which was markedly up‐regulated in rats with cardiac fibrosis. In addition, the expression of prostaglandin E2 receptor 4 (EP4) was decreased in cardiac fibrosis. In vivo lnc000908 silencing by lentivirus increased the EP4 level, decreased endothelial‐mesenchymal transition (EndMT) and improved cardiac fibrosis and cardiac function. Consistently, the lnc000908 knockdown also up‐regulated EP4 and suppressed transforming growth factor‐beta (TGF‐β)‐induced EndMT in cardiac microvascular endothelial cells. In contrast, the lnc000908 overexpression by lentivirus decreased the EP4 level and induced EndMT. Of note, these pro‐ or anti‐EndMT effects were reversed by the EP4 overexpression or the EP4 antagonist AH‐23848, respectively. This study demonstrates that lnc000908 is a novel regulator of cardiac fibrosis by modulating the EP4 expression and EndMT.
Collapse
Affiliation(s)
- Xingxing Chen
- Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Wenhua Ge
- Stomatological Hospital, College of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - Jie Hu
- Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Tiancheng Dong
- Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Hui Yao
- Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Lingzhi Chen
- Department of Clinical Laboratory, Wenzhou Central Hospital, Wenzhou, China
| | - Bin Geng
- Hypertension Center of Fuwai Hospital, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hao Zhou
- Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
12
|
Jung J, Lee YJ, Choi YH, Park EM, Kim HS, Kang JL. Gas6 Prevents Epithelial-Mesenchymal Transition in Alveolar Epithelial Cells via Production of PGE 2, PGD 2 and Their Receptors. Cells 2019; 8:cells8070643. [PMID: 31247991 PMCID: PMC6678614 DOI: 10.3390/cells8070643] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 06/21/2019] [Accepted: 06/25/2019] [Indexed: 12/22/2022] Open
Abstract
The epithelial-mesenchymal transition (EMT) is important in organ fibrosis. We hypothesized that growth arrest-specific protein 6 (Gas6) and its underlying mechanisms play roles in the prevention of EMT in alveolar epithelial cells (ECs). In this study, to determine whether Gas6 prevents TGF-β1-induced EMT in LA-4 and primary alveolar type II ECs, real-time PCR and immunoblotting in cell lysates and ELISA in culture supernatants were performed. Migration and invasion assays were performed using Transwell chambers. Pretreatment of ECs with Gas6 inhibited TGF-β1-induced EMT based on cell morphology, changes in EMT marker expression, and induction of EMT-activating transcription factors. Gas6 enhanced the levels of cyclooxygenase-2 (COX-2)-derived prostaglandin E2 (PGE2) and PGD2 as well as of their receptors. COX-2 inhibitors and antagonists of PGE2 and PGD2 receptors reversed the inhibition of TGF-β1-induced EMT, migration, and invasion by Gas6. Moreover, knockdown of Axl or Mer reversed the enhancement of PGE2 and PGD2 and suppression of EMT, migration and invasion by Gas6. Our data suggest Gas6-Axl or -Mer signalling events may reprogram ECs to resist EMT via the production of PGE2, PGD2, and their receptors.
Collapse
Affiliation(s)
- Jihye Jung
- Department of Physiology, School of Medicine, Ewha Womans University, Seoul 07804, Korea.
- Tissue Injury Defense Research Center, School of Medicine, Ewha Womans University, Seoul 07804, Korea.
| | - Ye-Ji Lee
- Department of Physiology, School of Medicine, Ewha Womans University, Seoul 07804, Korea.
- Tissue Injury Defense Research Center, School of Medicine, Ewha Womans University, Seoul 07804, Korea.
| | - Youn-Hee Choi
- Department of Physiology, School of Medicine, Ewha Womans University, Seoul 07804, Korea.
- Tissue Injury Defense Research Center, School of Medicine, Ewha Womans University, Seoul 07804, Korea.
| | - Eun-Mi Park
- Tissue Injury Defense Research Center, School of Medicine, Ewha Womans University, Seoul 07804, Korea.
- Department of Pharmacology, School of Medicine, Ewha Womans University, Seoul 07804, Korea.
| | - Hee-Sun Kim
- Tissue Injury Defense Research Center, School of Medicine, Ewha Womans University, Seoul 07804, Korea.
- Department of Molecular Medicine, School of Medicine, Ewha Womans University, Seoul 07804, Korea.
| | - Jihee L Kang
- Department of Physiology, School of Medicine, Ewha Womans University, Seoul 07804, Korea.
- Tissue Injury Defense Research Center, School of Medicine, Ewha Womans University, Seoul 07804, Korea.
| |
Collapse
|
13
|
Abstract
Bioactive lipids are essential components of human cells and tissues. As discussed in this review, the cancer lipidome is diverse and malleable, with the ability to promote or inhibit cancer pathogenesis. Targeting lipids within the tumor and surrounding microenvironment may be a novel therapeutic approach for treating cancer patients. Additionally, the emergence of a novel super-family of lipid mediators termed specialized pro-resolving mediators (SPMs) has revealed a new role for bioactive lipid mediators in the resolution of inflammation in cancer biology. The role of SPMs in cancer holds great promise in our understanding of cancer pathogenesis and can ultimately be used in future cancer diagnostics and therapy.
Collapse
Affiliation(s)
- Megan L Sulciner
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| | - Allison Gartung
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Molly M Gilligan
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Charles N Serhan
- Department of Anesthesiology, Center for Experimental Therapeutics and Reperfusion Injury, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Dipak Panigrahy
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
14
|
Ding H, Bai F, Cao H, Xu J, Fang L, Wu J, Yuan Q, Zhou Y, Sun Q, He W, Dai C, Zen K, Jiang L, Yang J. PDE/cAMP/Epac/C/EBP-β Signaling Cascade Regulates Mitochondria Biogenesis of Tubular Epithelial Cells in Renal Fibrosis. Antioxid Redox Signal 2018; 29:637-652. [PMID: 29216750 DOI: 10.1089/ars.2017.7041] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
AIMS Cyclic adenosine 3'5'-monophosphate (cAMP) is a universal second messenger that plays an important role in intracellular signal transduction. cAMP is synthesized by adenylate cyclases from adenosine triphosphate and terminated by the phosphodiesterases (PDEs). In the present study, we investigated the role of the cAMP pathway in tubular epithelial cell mitochondrial biogenesis in the pathogenesis of renal fibrosis. RESULTS We found that the cAMP levels were decreased in fibrotic kidney tissues, and replenishing cAMP could ameliorate tubular atrophy and extracellular matrix deposition. The downregulation of cAMP was mainly attributed to the increased PDE4 expression in tubular epithelial cells. The inhibition of PDE4 by PDE4 siRNA or the specific inhibitor, rolipram, attenuated unilateral ureteral obstruction-induced renal interstitial fibrosis and transforming growth factor (TGF)-β1-stimulated primary tubular epithelial cell (PTC) damage. The Epac1/Rap1 pathway contributed to the main effect of cAMP on renal fibrosis. Rolipram could restore C/EBP-β and PGC-1α expression and protect the mitochondrial function and structure of PTCs under TGF-β1 stimulation. The antifibrotic role of rolipram in renal fibrosis relies on C/EBP-β and PGC-1α expression in tubular epithelial cells. Innovation and Conclusion: The results of the present study indicate that cAMP signaling regulates the mitochondrial biogenesis of tubular epithelial cells in renal fibrosis. Restoring cAMP by the PDE4 inhibitor rolipram may ameliorate renal fibrosis by targeting C/EBP-β/PGC1-α and mitochondrial biogenesis. Antioxid. Redox Signal. 29, 637-652.
Collapse
Affiliation(s)
- Hao Ding
- 1 Center for Kidney Disease, Second Affiliated Hospital, Nanjing Medical University , Nanjing, China
| | - Feng Bai
- 1 Center for Kidney Disease, Second Affiliated Hospital, Nanjing Medical University , Nanjing, China .,2 Department of Endocrinology and Metabolism, Huai'an Hospital Affiliated to Xuzhou Medical University and Huai'an Second People's Hospital , Huai'an, China
| | - Hongdi Cao
- 1 Center for Kidney Disease, Second Affiliated Hospital, Nanjing Medical University , Nanjing, China
| | - Jing Xu
- 1 Center for Kidney Disease, Second Affiliated Hospital, Nanjing Medical University , Nanjing, China
| | - Li Fang
- 3 Department of Nephrology, Affiliated Hospital of Nantong University , Nantong, China
| | - Jining Wu
- 1 Center for Kidney Disease, Second Affiliated Hospital, Nanjing Medical University , Nanjing, China
| | - Qi Yuan
- 1 Center for Kidney Disease, Second Affiliated Hospital, Nanjing Medical University , Nanjing, China
| | - Yang Zhou
- 1 Center for Kidney Disease, Second Affiliated Hospital, Nanjing Medical University , Nanjing, China
| | - Qi Sun
- 1 Center for Kidney Disease, Second Affiliated Hospital, Nanjing Medical University , Nanjing, China
| | - Weichun He
- 1 Center for Kidney Disease, Second Affiliated Hospital, Nanjing Medical University , Nanjing, China
| | - Chunsun Dai
- 1 Center for Kidney Disease, Second Affiliated Hospital, Nanjing Medical University , Nanjing, China
| | - Ke Zen
- 4 State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University Advanced Institute of Life Sciences , Nanjing, China
| | - Lei Jiang
- 1 Center for Kidney Disease, Second Affiliated Hospital, Nanjing Medical University , Nanjing, China
| | - Junwei Yang
- 1 Center for Kidney Disease, Second Affiliated Hospital, Nanjing Medical University , Nanjing, China
| |
Collapse
|
15
|
Stone Ii R, Natesan S, Kowalczewski CJ, Mangum LH, Clay NE, Clohessy RM, Carlsson AH, Tassin DH, Chan RK, Rizzo JA, Christy RJ. Advancements in Regenerative Strategies Through the Continuum of Burn Care. Front Pharmacol 2018; 9:672. [PMID: 30038569 PMCID: PMC6046385 DOI: 10.3389/fphar.2018.00672] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 06/05/2018] [Indexed: 01/09/2023] Open
Abstract
Burns are caused by several mechanisms including flame, scald, chemical, electrical, and ionizing and non-ionizing radiation. Approximately half a million burn cases are registered annually, of which 40 thousand patients are hospitalized and receive definitive treatment. Burn care is very resource intensive as the treatment regimens and length of hospitalization are substantial. Burn wounds are classified based on depth as superficial (first degree), partial-thickness (second degree), or full-thickness (third degree), which determines the treatment necessary for successful healing. The goal of burn wound care is to fully restore the barrier function of the tissue as quickly as possible while minimizing infection, scarring, and contracture. The aim of this review is to highlight how tissue engineering and regenerative medicine strategies are being used to address the unique challenges of burn wound healing and define the current gaps in care for both partial- and full-thickness burn injuries. This review will present the current standard of care (SOC) and provide information on various treatment options that have been tested pre-clinically or are currently in clinical trials. Due to the complexity of burn wound healing compared to other skin injuries, burn specific treatment regimens must be developed. Recently, tissue engineering and regenerative medicine strategies have been developed to improve skin regeneration that can restore normal skin physiology and limit adverse outcomes, such as infection, delayed re-epithelialization, and scarring. Our emphasis will be centered on how current clinical and pre-clinical research of pharmacological agents, biomaterials, and cellular-based therapies can be applied throughout the continuum of burn care by targeting the stages of wound healing: hemostasis, inflammation, cell proliferation, and matrix remodeling.
Collapse
Affiliation(s)
- Randolph Stone Ii
- Combat Trauma and Burn Injury Research, US Army Institute of Surgical Research San Antonio, TX, United States
| | - Shanmugasundaram Natesan
- Combat Trauma and Burn Injury Research, US Army Institute of Surgical Research San Antonio, TX, United States
| | - Christine J Kowalczewski
- Combat Trauma and Burn Injury Research, US Army Institute of Surgical Research San Antonio, TX, United States
| | - Lauren H Mangum
- Combat Trauma and Burn Injury Research, US Army Institute of Surgical Research San Antonio, TX, United States.,Extremity Trauma and Regenerative Medicine, US Army Institute of Surgical Research San Antonio, TX, United States
| | - Nicholas E Clay
- Combat Trauma and Burn Injury Research, US Army Institute of Surgical Research San Antonio, TX, United States
| | - Ryan M Clohessy
- Combat Trauma and Burn Injury Research, US Army Institute of Surgical Research San Antonio, TX, United States
| | - Anders H Carlsson
- Dental and Craniofacial Trauma Research, US Army Institute of Surgical Research San Antonio, TX, United States
| | - David H Tassin
- Dental and Craniofacial Trauma Research, US Army Institute of Surgical Research San Antonio, TX, United States
| | - Rodney K Chan
- Dental and Craniofacial Trauma Research, US Army Institute of Surgical Research San Antonio, TX, United States
| | - Julie A Rizzo
- Burn Flight Team, US Army Institute of Surgical Research San Antonio, TX, United States
| | - Robert J Christy
- Combat Trauma and Burn Injury Research, US Army Institute of Surgical Research San Antonio, TX, United States
| |
Collapse
|
16
|
Teh SW, Mok PL, Abd Rashid M, Bastion MLC, Ibrahim N, Higuchi A, Murugan K, Mariappan R, Subbiah SK. Recent Updates on Treatment of Ocular Microbial Infections by Stem Cell Therapy: A Review. Int J Mol Sci 2018; 19:ijms19020558. [PMID: 29438279 PMCID: PMC5855780 DOI: 10.3390/ijms19020558] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 12/03/2017] [Accepted: 12/12/2017] [Indexed: 02/06/2023] Open
Abstract
Ocular microbial infection has emerged as a major public health crisis during the past two decades. A variety of causative agents can cause ocular microbial infections; which are characterized by persistent and destructive inflammation of the ocular tissue; progressive visual disturbance; and may result in loss of visual function in patients if early and effective treatments are not received. The conventional therapeutic approaches to treat vision impairment and blindness resulting from microbial infections involve antimicrobial therapy to eliminate the offending pathogens or in severe cases; by surgical methods and retinal prosthesis replacing of the infected area. In cases where there is concurrent inflammation, once infection is controlled, anti-inflammatory agents are indicated to reduce ocular damage from inflammation which ensues. Despite advances in medical research; progress in the control of ocular microbial infections remains slow. The varying level of ocular tissue recovery in individuals and the incomplete visual functional restoration indicate the chief limitations of current strategies. The development of a more extensive therapy is needed to help in healing to regain vision in patients. Stem cells are multipotent stromal cells that can give rise to a vast variety of cell types following proper differentiation protocol. Stem cell therapy shows promise in reducing inflammation and repairing tissue damage on the eye caused by microbial infections by its ability to modulate immune response and promote tissue regeneration. This article reviews a selected list of common infectious agents affecting the eye; which include fungi; viruses; parasites and bacteria with the aim of discussing the current antimicrobial treatments and the associated therapeutic challenges. We also provide recent updates of the advances in stem cells studies on sepsis therapy as a suggestion of optimum treatment regime for ocular microbial infections.
Collapse
Affiliation(s)
- Seoh Wei Teh
- Department of Biomedical Science, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia.
| | - Pooi Ling Mok
- Department of Biomedical Science, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia.
- Genetics and Regenerative Medicine Research Centre, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia.
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Aljouf University, 72442 Sakaka, Aljouf Province, Saudi Arabia.
| | - Munirah Abd Rashid
- Department of Ophthalmology, Faculty of Medicine, UKM Medical Center, 56000 Cheras, Kuala Lumpur, Malaysia.
| | - Mae-Lynn Catherine Bastion
- Department of Ophthalmology, Faculty of Medicine, UKM Medical Center, 56000 Cheras, Kuala Lumpur, Malaysia.
| | - Normala Ibrahim
- Department of Psychiatry, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia.
| | - Akon Higuchi
- Department of Chemical and Materials Engineering, National Central University, No. 300, Jhongda RD., Jhongli, 32001 Taoyuan, Taiwan.
| | - Kadarkarai Murugan
- Department of Zoology, Thiruvalluvar University, Serkkadu, 632 115 Vellore, India.
| | - Rajan Mariappan
- Biomaterials in Medicinal Chemistry Laboratory, Department of Natural Products Chemistry, School of Chemistry, Madurai Kamaraj University, Madurai, 625 021 Tamil Nadu, India.
| | - Suresh Kumar Subbiah
- Genetics and Regenerative Medicine Research Centre, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia.
- Department of Medical Microbiology and Parasitology, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia.
| |
Collapse
|
17
|
Giacomelli C, Daniele S, Romei C, Tavanti L, Neri T, Piano I, Celi A, Martini C, Trincavelli ML. The A 2B Adenosine Receptor Modulates the Epithelial- Mesenchymal Transition through the Balance of cAMP/PKA and MAPK/ERK Pathway Activation in Human Epithelial Lung Cells. Front Pharmacol 2018; 9:54. [PMID: 29445342 PMCID: PMC5797802 DOI: 10.3389/fphar.2018.00054] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Accepted: 01/15/2018] [Indexed: 12/12/2022] Open
Abstract
The epithelial-mesenchymal transition (EMT) is a complex process in which cell phenotype switches from the epithelial to mesenchymal one. The deregulations of this process have been related with the occurrence of different diseases such as lung cancer and fibrosis. In the last decade, several efforts have been devoted in understanding the mechanisms that trigger and sustain this transition process. Adenosine is a purinergic signaling molecule that has been involved in the onset and progression of chronic lung diseases and cancer through the A2B adenosine receptor subtype activation, too. However, the relationship between A2BAR and EMT has not been investigated, yet. Herein, the A2BAR characterization was carried out in human epithelial lung cells. Moreover, the effects of receptor activation on EMT were investigated in the absence and presence of transforming growth factor-beta (TGF-β1), which has been known to promote the transition. The A2BAR activation alone decreased and increased the expression of epithelial markers (E-cadherin) and the mesenchymal one (Vimentin, N-cadherin), respectively, nevertheless a complete EMT was not observed. Surprisingly, the receptor activation counteracted the EMT induced by TGF-β1. Several intracellular pathways regulate the EMT: high levels of cAMP and ERK1/2 phosphorylation has been demonstrated to counteract and promote the transition, respectively. The A2BAR stimulation was able to modulated these two pathways, cAMP/PKA and MAPK/ERK, shifting the fine balance toward activation or inhibition of EMT. In fact, using a selective PKA inhibitor, which blocks the cAMP pathway, the A2BAR-mediated EMT promotion were exacerbated, and conversely the selective inhibition of MAPK/ERK counteracted the receptor-induced transition. These results highlighted the A2BAR as one of the receptors involved in the modulation of EMT process. Nevertheless, its activation is not enough to trigger a complete transition, its ability to affect different intracellular pathways could represent a mechanism at the basis of EMT maintenance/inhibition based on the extracellular microenvironment. Despite further investigations are needed, herein for the first time the A2BAR has been related to the EMT process, and therefore to the different EMT-related pathologies.
Collapse
Affiliation(s)
| | | | - Chiara Romei
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, Pisa, Italy.,Radiology Unit, University Hospital of Pisa, Pisa, Italy
| | - Laura Tavanti
- Pneumology Unit, Cardio-Thoracic Department, University Hospital of Pisa, Pisa, Italy
| | - Tommaso Neri
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, Pisa, Italy
| | - Ilaria Piano
- Department of Pharmacy, University of Pisa, Pisa, Italy
| | - Alessandro Celi
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, Pisa, Italy
| | | | | |
Collapse
|
18
|
Zuccarini M, Giuliani P, Buccella S, Di Liberto V, Mudò G, Belluardo N, Carluccio M, Rossini M, Condorelli DF, Rathbone MP, Caciagli F, Ciccarelli R, Di Iorio P. Modulation of the TGF-β1-induced epithelial to mesenchymal transition (EMT) mediated by P1 and P2 purine receptors in MDCK cells. Purinergic Signal 2017; 13:429-442. [PMID: 28616713 PMCID: PMC5714834 DOI: 10.1007/s11302-017-9571-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 05/29/2017] [Indexed: 12/17/2022] Open
Abstract
Epithelial to mesenchymal transition (EMT) occurs during embryogenesis or under pathological conditions such as hypoxia, injury, chronic inflammation, or tissue fibrosis. In renal tubular epithelial cells (MDCK), TGF-β1 induces EMT by reducing or increasing epithelial or mesenchymal marker expression, respectively. In this study, we confirmed that the cAMP analogues, 8-CPT-cAMP or N6-Ph-cAMP, inhibited the TGF-β1-driven overexpression of the mesenchymal markers ZEB-1, Slug, Fibronectin, and α-SMA. Furthermore, we showed that A1, A2A, P2Y1, P2Y11, and P2X7 purine receptor agonists modulated the TGF-β1-induced EMT through the involvement of PKA and/or MAPK/ERK signaling. The stimulation of A2A receptor reduced the overexpression of the EMT-related markers, mainly through the cAMP-dependent PKA pathway, as confirmed by cell pre-treatment with Myr-PKI. Both A1 and P2Y1 receptor stimulation exacerbated the TGF-β1-driven effects, which were reduced by cell pre-treatment with the MAPK inhibitor PD98059, according to the increased ERK1/2 phosphorylation upon receptor activation. The effects induced by P2Y11 receptor activation were oppositely modulated by PKA or MAPK inhibition, in line with the dual nature of the Gs- and Gq-coupled receptor. Differently, P2X7 receptor induced, per se, similar and not additive effects compared to TGF-β1, after prolonged cell exposure to BzATP. These results suggest a putative role of purine receptors as target for anti-fibrotic agents.
Collapse
Affiliation(s)
- Mariachiara Zuccarini
- Department of Medical and Oral Sciences and Biotechnologies, "G.d'Annunzio" University of Chieti-Pescara, Chieti, Italy.
| | - Patricia Giuliani
- Department of Medical and Oral Sciences and Biotechnologies, "G.d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Silvana Buccella
- Department of Medical and Oral Sciences and Biotechnologies, "G.d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Valentina Di Liberto
- Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo, Palermo, Italy
| | - Giuseppa Mudò
- Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo, Palermo, Italy
| | - Natale Belluardo
- Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo, Palermo, Italy
| | - Marzia Carluccio
- Department of Medical and Oral Sciences and Biotechnologies, "G.d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Margherita Rossini
- Department of Medical and Oral Sciences and Biotechnologies, "G.d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | | | - Michel Piers Rathbone
- Department of Medicine, Division of Neurology, McMaster University, Hamilton, ON, Canada
| | - Francesco Caciagli
- Department of Medical and Oral Sciences and Biotechnologies, "G.d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Renata Ciccarelli
- Department of Medical and Oral Sciences and Biotechnologies, "G.d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Patrizia Di Iorio
- Department of Medical and Oral Sciences and Biotechnologies, "G.d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| |
Collapse
|
19
|
Kim YR, Park MK, Kang GJ, Kim HJ, Kim EJ, Byun HJ, Lee MY, Lee CH. Leukotriene B4 induces EMT and vimentin expression in PANC-1 pancreatic cancer cells: Involvement of BLT2 via ERK2 activation. Prostaglandins Leukot Essent Fatty Acids 2016; 115:67-76. [PMID: 27914516 DOI: 10.1016/j.plefa.2016.10.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 10/22/2016] [Accepted: 10/24/2016] [Indexed: 12/19/2022]
Abstract
Leukotriene B4 (LTB4) is a leukocyte chemoattractant and plays a major role controlling inflammatory responses including pancreatitis. LTB4 is known to be correlated with cancer progression. LTB4 induces keratin phosphorylation and reorganization by activating extracellular regulated kinase (ERK) in PANC-1 pancreatic cancer cell lines. However, the role of LTB4 in epithelial mesenchymal transition (EMT) and vimentin expression in pancreatic cancer cells is unknown. We examined whether LTB4 induces EMT and vimentin expression by Western blot, si-RNA, and RT-PCR. LTB4 induced morphological change, decreased E-cadherin expression and increased N-cadherin and vimentin expression. LTB4 increased migration and invasion of PANC-1 cancer cells. LTB4 dose-dependently upregulated expression of vimentin in PANC-1 cancer cells. LTB4-induced vimentin expression was suppressed by LY255283 (BLT2 antagonist). Comp A, a BLT2 agonist, further increased vimentin expression. Gene silencing of BLT2 suppressed LTB4-or Comp A-induced vimentin expression in PANC-1 cells. The MEK inhibitor, PD98059 suppressed Comp A-induced vimentin expression. Comp A or transfection of plasmid containing BLT2 cDNA (pCBLT2) activated ERK, and BLT2 gene silencing suppressed Comp A-induced ERK activation. ERK2 siRNA abrogated Comp A-induced vimentin expression and ERK2 overexpression enhanced vimentin expression. One of well-known cause of ras mutation, cigarette smoke extracts increased BLT2 expression in PANC-1 cancer cells. Taken together, these results suggest that BLT2 is involved in LTB4-induced vimentin expression through ERK2 in PANC-1 cells.
Collapse
Affiliation(s)
- You Ri Kim
- College of Pharmacy, Dongguk University, Seoul 100-715, Republic of Korea
| | - Mi Kyung Park
- College of Pharmacy, Dongguk University, Seoul 100-715, Republic of Korea
| | - Gyeong Jin Kang
- College of Pharmacy, Dongguk University, Seoul 100-715, Republic of Korea
| | - Hyun Ji Kim
- College of Pharmacy, Dongguk University, Seoul 100-715, Republic of Korea
| | - Eun Ji Kim
- College of Pharmacy, Dongguk University, Seoul 100-715, Republic of Korea
| | - Hyun Jung Byun
- College of Pharmacy, Dongguk University, Seoul 100-715, Republic of Korea
| | - Moo-Yeol Lee
- College of Pharmacy, Dongguk University, Seoul 100-715, Republic of Korea
| | - Chang Hoon Lee
- College of Pharmacy, Dongguk University, Seoul 100-715, Republic of Korea.
| |
Collapse
|
20
|
Montford JR, Lehman AMB, Scobey MS, Weiser-Evans MCM, Nemenoff RA, Furgeson SB. Cytosolic phospholipase A 2α increases proliferation and de-differentiation of human renal tubular epithelial cells. Prostaglandins Other Lipid Mediat 2016; 126:1-8. [PMID: 27554058 DOI: 10.1016/j.prostaglandins.2016.08.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Revised: 07/14/2016] [Accepted: 08/19/2016] [Indexed: 01/03/2023]
Abstract
The group IVA calcium-dependent cytosolic phospholipase A2 (cPLA2α) enzyme controls the release of arachidonic acid from membrane bound phospholipids and is the rate-limiting step in production of eicosanoids. A variety of different kidney injuries activate cPLA2α, therefore we hypothesized that cPLA2α activity would regulate pathologic processes in HK-2 cells, a human renal tubular epithelial cell line, by regulating cell phenotype and proliferation. In two lentiviral cPLA2α-silenced knockdowns, we observed decreased proliferation and increased apoptosis compared to control HK-2 cells. cPLA2α-silenced cells also demonstrated an altered morphology, had increased expression E-cadherin, and decreased expression of Ncadherin. Increased levels of E-cadherin were associated with increased promoter activity and decreased levels of SNAIL1, SNAIL2, and ZEB1, transcriptional repressors of E-cadherin expression. Addition of exogenous arachidonic acid, but not PGE2, reversed the phenotypic changes in cPLA2α-silenced cells. These data suggest that cPLA2α may play a key role in renal repair after injury through a PGE2-independent mechanism.
Collapse
Affiliation(s)
- John R Montford
- Division of Renal Diseases and Hypertension, Department of Medicine, University of Colorado-Anschutz Medical Campus, Aurora, CO, United States.
| | - Allison M B Lehman
- Division of Renal Diseases and Hypertension, Department of Medicine, University of Colorado-Anschutz Medical Campus, Aurora, CO, United States
| | - Micah S Scobey
- Division of Renal Diseases and Hypertension, Department of Medicine, University of Colorado-Anschutz Medical Campus, Aurora, CO, United States
| | - Mary C M Weiser-Evans
- Division of Renal Diseases and Hypertension, Department of Medicine, University of Colorado-Anschutz Medical Campus, Aurora, CO, United States
| | - Raphael A Nemenoff
- Division of Renal Diseases and Hypertension, Department of Medicine, University of Colorado-Anschutz Medical Campus, Aurora, CO, United States
| | - Seth B Furgeson
- Division of Renal Diseases and Hypertension, Department of Medicine, University of Colorado-Anschutz Medical Campus, Aurora, CO, United States; Denver Health and Hospitals, Denver, CO, United States
| |
Collapse
|
21
|
Macrophages programmed by apoptotic cells inhibit epithelial-mesenchymal transition in lung alveolar epithelial cells via PGE2, PGD2, and HGF. Sci Rep 2016; 6:20992. [PMID: 26875548 PMCID: PMC4753481 DOI: 10.1038/srep20992] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 01/15/2016] [Indexed: 12/20/2022] Open
Abstract
Apoptotic cell clearance results in the release of growth factors and the action of signaling molecules involved in tissue homeostasis maintenance. Here, we investigated whether and how macrophages programmed by apoptotic cells inhibit the TGF-β1-induced Epithelial-mesenchymal transition (EMT) process in lung alveolar epithelial cells. Treatment with conditioned medium derived from macrophages exposed to apoptotic cells, but not viable or necrotic cells, inhibited TGF-β1-induced EMT, including loss of E-cadherin, synthesis of N-cadherin and α-smooth muscle actin, and induction of EMT-activating transcription factors, such as Snail1/2, Zeb1/2, and Twist1. Exposure of macrophages to cyclooxygenase (COX-2) inhibitors (NS-398 and COX-2 siRNA) or RhoA/Rho kinase inhibitors (Y-27632 and RhoA siRNA) and LA-4 cells to antagonists of prostaglandin E2 (PGE2) receptor (EP4 [AH-23848]), PGD2 receptors (DP1 [BW-A868C] and DP2 [BAY-u3405]), or the hepatocyte growth factor (HGF) receptor c-Met (PHA-665752), reversed EMT inhibition by the conditioned medium. Additionally, we found that apoptotic cell instillation inhibited bleomycin-mediated EMT in primary mouse alveolar type II epithelial cells in vivo. Our data suggest a new model for epithelial cell homeostasis, by which the anti-EMT programming of macrophages by apoptotic cells may control the progressive fibrotic reaction via the production of potent paracrine EMT inhibitors.
Collapse
|
22
|
Chu C, Bottaro DP, Betenbaugh MJ, Shiloach J. Stable Ectopic Expression of ST6GALNAC5 Induces Autocrine MET Activation and Anchorage-Independence in MDCK Cells. PLoS One 2016; 11:e0148075. [PMID: 26848584 PMCID: PMC4743853 DOI: 10.1371/journal.pone.0148075] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Accepted: 01/12/2016] [Indexed: 11/19/2022] Open
Abstract
The epithelial-mesenchymal transition (EMT) is a complex cancer progression that can boost the metastatic potential of transformed cells by inducing migration, loss of cell adhesion, and promoting proliferation under anchorage-independent conditions. A DNA microarray analysis was performed comparing parental anchorage-dependent MDCK cells and anchorage-independent MDCK cells that were engineered to express human siat7e (ST6GALNAC5). The comparison identified several genes involved in the EMT process that were differentially expressed between the anchorage-dependent and the anchorage-independent MDCK cell lines. The hepatocyte growth factor gene (hgf) was found to be over-expressed in the engineered MDCK-siat7e cells at both transcription and protein expression levels. Phosphorylation analysis of the MET receptor tyrosine kinase confirmed the activation of an autocrine loop of the HGF/ MET signaling pathway in the MDCK-siat7e cells. When MET activities were suppressed by using the small-molecular inhibitor drug PF-02341066 (Crizotinib), the anchorage-independent MDCK-siat7e cells reverted to the cellular morphology of the parental anchorage-dependent MDCK cells. These observations indicate that the MET receptor plays a central role in the growth properties of the MDCK cells and its phosphorylation status is likely dependent on sialylation. Further investigation of the downstream signaling targets in the MET network showed that the degree of MDCK cell adhesion correlated with secretion levels of a matrix metalloproteinase, MMP1, suggesting a role of metalloproteinases in the EMT process. These results demonstrate that in addition to its application in biotechnology processes, MDCK-siat7e may serve as a model cell for metastasis studies to decipher the sequence of events leading up to the activation of EMT.
Collapse
Affiliation(s)
- Chia Chu
- Biotechnology Core Laboratory, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Donald P. Bottaro
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Michael J. Betenbaugh
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Joseph Shiloach
- Biotechnology Core Laboratory, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
23
|
Mao S, Zhang J. The emerging role of hepatocyte growth factor in renal diseases. J Recept Signal Transduct Res 2015; 36:303-9. [DOI: 10.3109/10799893.2015.1080275] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
24
|
Loeffler I, Wolf G. Epithelial-to-Mesenchymal Transition in Diabetic Nephropathy: Fact or Fiction? Cells 2015; 4:631-52. [PMID: 26473930 PMCID: PMC4695850 DOI: 10.3390/cells4040631] [Citation(s) in RCA: 193] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Revised: 09/29/2015] [Accepted: 09/30/2015] [Indexed: 12/17/2022] Open
Abstract
The pathophysiology of diabetic nephropathy (DN), one of the most serious complications in diabetic patients and the leading cause of end-stage renal disease worldwide, is complex and not fully elucidated. A typical hallmark of DN is the excessive deposition of extracellular matrix (ECM) proteins in the glomerulus and in the renal tubulointerstitium, eventually leading to glomerulosclerosis and interstitial fibrosis. Although it is obvious that myofibroblasts play a major role in the synthesis and secretion of ECM, the origin of myofibroblasts in DN remains the subject of controversial debates. A number of studies have focused on epithelial-to-mesenchymal transition (EMT) as one source of matrix-generating fibroblasts in the diseased kidney. EMT is characterized by the acquisition of mesenchymal properties by epithelial cells, preferentially proximal tubular cells and podocytes. In this review we comprehensively review the literature and discuss arguments both for and against a function of EMT in renal fibrosis in DN. While the precise extent of the contribution to nephrotic fibrosis is certainly arduous to quantify, the picture that emerges from this extensive body of literature suggests EMT as a major source of myofibroblasts in DN.
Collapse
Affiliation(s)
- Ivonne Loeffler
- Department of Internal Medicine III, University Hospital, University of Jena, Erlanger Allee 101, D-07747 Jena, Germany
| | - Gunter Wolf
- Department of Internal Medicine III, University Hospital, University of Jena, Erlanger Allee 101, D-07747 Jena, Germany.
| |
Collapse
|
25
|
Yoon YS, Kim SY, Kim MJ, Lim JH, Cho MS, Kang JL. PPARγ activation following apoptotic cell instillation promotes resolution of lung inflammation and fibrosis via regulation of efferocytosis and proresolving cytokines. Mucosal Immunol 2015; 8:1031-46. [PMID: 25586556 PMCID: PMC4762910 DOI: 10.1038/mi.2014.130] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Accepted: 12/05/2014] [Indexed: 02/04/2023]
Abstract
Changes in macrophage phenotype have been implicated in apoptotic cell-mediated immune modulation via induction of peroxisome proliferator-activated receptor-γ (PPARγ). In this study, we characterized PPARγ induction by apoptotic cell instillation over the course of bleomycin-induced lung injury in C57BL/6 mice. Next, the role of PPARγ activation in resolving lung inflammation and fibrosis was investigated. Our data demonstrate that apoptotic cell instillation after bleomycin results in immediate and prolonged enhancement of PPARγ mRNA and protein in alveolar macrophages and lung. Moreover, PPARγ activity and expression of its target molecules, including CD36, macrophage mannose receptor, and arginase 1, were persistently enhanced following apoptotic cell instillation. Coadministration of the PPARγ antagonist, GW9662, reversed the enhanced efferocytosis, and the reduced proinflammatory cytokine expression, neutrophil recruitment, myeloperoxidase activity, hydroxyproline contents, and fibrosis markers, including type 1 collagen α2, fibronectin and α-smooth muscle actin (α-SMA), in the lung by apoptotic cell instillation. In addition, inhibition of PPARγ activity reversed the expression of transforming growth factor-β (TGF-β), interleukin (IL)-10, and hepatocyte growth factor (HGF). These findings indicate that one-time apoptotic cell instillation contributes to anti-inflammatory and antifibrotic responses via upregulation of PPARγ expression and subsequent activation, leading to regulation of efferocytosis and production of proresolving cytokines.
Collapse
Affiliation(s)
- Y-S Yoon
- Department of Physiology, School of Medicine, Ewha Womans University, Yangcheon-ku, Seoul, Korea
- Tissue Injury Defense Research Center, School of Medicine, Ewha Womans University, Yangcheon-ku, Seoul, Korea
| | - S-Y Kim
- Department of Physiology, School of Medicine, Ewha Womans University, Yangcheon-ku, Seoul, Korea
- Tissue Injury Defense Research Center, School of Medicine, Ewha Womans University, Yangcheon-ku, Seoul, Korea
| | - M-J Kim
- Department of Physiology, School of Medicine, Ewha Womans University, Yangcheon-ku, Seoul, Korea
- Tissue Injury Defense Research Center, School of Medicine, Ewha Womans University, Yangcheon-ku, Seoul, Korea
| | - J-H Lim
- Department of Microbiology, School of Medicine, Ewha Womans University, Yangcheon-ku, Seoul, Korea
| | - M-S Cho
- Department of Pathology, School of Medicine, Ewha Womans University, Yangcheon-ku, Seoul, Korea
| | - J L Kang
- Department of Physiology, School of Medicine, Ewha Womans University, Yangcheon-ku, Seoul, Korea
- Tissue Injury Defense Research Center, School of Medicine, Ewha Womans University, Yangcheon-ku, Seoul, Korea
- Global Top 5 Research Program, School of Medicine, Ewha Womans University, Yangcheon-ku, Seoul, Korea
| |
Collapse
|
26
|
Li M, Zhao Y, Hao H, Han W, Fu X. Mesenchymal stem cell-based therapy for nonhealing wounds: today and tomorrow. Wound Repair Regen 2015; 23:465-82. [PMID: 25877885 DOI: 10.1111/wrr.12304] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Revised: 03/09/2015] [Accepted: 03/30/2015] [Indexed: 12/31/2022]
Abstract
Although advancements have been made with traditional therapies, the treatment of chronic nonhealing wounds still remains a tough challenge. In the past two decades, mesenchymal stem cell (MSC)-based therapy has emerged as a promising therapeutic strategy for nonhealing wounds because of their characteristics including self-renewal and a multidirectional differentiation ability and their easy collection and weak immunogenicity. There is a growing body of basic scientific studies that shed light on the functional mechanism of MSCs in modulating nonhealing wounds. Furthermore, critical advances have been achieved using MSC-based therapy in preclinical animal models as well as in clinics trials. In this present review, we summarize the mechanisms of MSCs and highlight the important preclinical and clinical trials of MSC therapy for nonhealing wounds. In particular, the combination of MSCs transplantation and tissue-engineered skin is addressed as a new strategy to optimize the delivery efficiency and therapeutic potential. Additionally, the current drawbacks of MSC therapy and the potential to further optimize the use of MSCs are implied.
Collapse
Affiliation(s)
- Meirong Li
- Wound Healing and Cell Biology Laboratory, Institute of Basic Medical Science, Chinese PLA General Hospital, Beijing, Peoples Republic of China.,Trauma Treatment Center, Central Laboratory, Chinese PLA General Hospital Hainan Branch, Sanya, Peoples Republic of China
| | - Yali Zhao
- Wound Healing and Cell Biology Laboratory, Institute of Basic Medical Science, Chinese PLA General Hospital, Beijing, Peoples Republic of China.,Trauma Treatment Center, Central Laboratory, Chinese PLA General Hospital Hainan Branch, Sanya, Peoples Republic of China
| | - Haojie Hao
- Wound Healing and Cell Biology Laboratory, Institute of Basic Medical Science, Chinese PLA General Hospital, Beijing, Peoples Republic of China
| | - Weidong Han
- Wound Healing and Cell Biology Laboratory, Institute of Basic Medical Science, Chinese PLA General Hospital, Beijing, Peoples Republic of China
| | - Xiaobing Fu
- Wound Healing and Cell Biology Laboratory, Institute of Basic Medical Science, Chinese PLA General Hospital, Beijing, Peoples Republic of China
| |
Collapse
|
27
|
Yeo CD, Kim JW, Ha JH, Kim SJ, Lee SH, Kim IK, Kim YK. Chemopreventive effect of phosphodieasterase-4 inhibition in benzo(a)pyrene-induced murine lung cancer model. Exp Lung Res 2014; 40:500-6. [DOI: 10.3109/01902148.2014.950769] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
28
|
Nasrallah R, Hassouneh R, Hébert RL. Chronic kidney disease: targeting prostaglandin E2 receptors. Am J Physiol Renal Physiol 2014; 307:F243-50. [PMID: 24966087 DOI: 10.1152/ajprenal.00224.2014] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Chronic kidney disease is a leading cause of morbidity and mortality in the world. A better understanding of disease mechanisms has been gained in recent years, but the current management strategies are ineffective at preventing disease progression. A widespread focus of research is placed on elucidating the specific processes implicated to find more effective therapeutic options. PGE2, acting on its four EP receptors, regulates many renal disease processes; thus EP receptors could prove to be important targets for kidney disease intervention strategies. This review summarizes the major pathogenic mechanisms contributing to initiation and progression of chronic kidney disease, emphasizing the role of hyperglycemia, hypertension, inflammation, and oxidative stress. We have long recognized the multifaceted role of PGs in both the initiation and progression of chronic kidney disease, yet studies are only now seriously contemplating specific EP receptors as targets for therapy. Given the plethora of renal complications attributed to PG involvement in the kidney, this review highlights these pathogenic events and emphasizes the PGE2 receptor targets as options available to complement current therapeutic strategies.
Collapse
Affiliation(s)
- Rania Nasrallah
- Department of Cellular and Molecular Medicine, and Kidney Research Centre, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Ramzi Hassouneh
- Department of Cellular and Molecular Medicine, and Kidney Research Centre, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Richard L Hébert
- Department of Cellular and Molecular Medicine, and Kidney Research Centre, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
29
|
Milara J, Peiró T, Serrano A, Guijarro R, Zaragozá C, Tenor H, Cortijo J. Roflumilast N-oxide inhibits bronchial epithelial to mesenchymal transition induced by cigarette smoke in smokers with COPD. Pulm Pharmacol Ther 2014; 28:138-48. [PMID: 24525294 DOI: 10.1016/j.pupt.2014.02.001] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Revised: 01/02/2014] [Accepted: 02/02/2014] [Indexed: 12/20/2022]
Abstract
BACKGROUND Epithelial to mesenchymal transition (EMT) is under discussion as a potential mechanism of small airway remodelling in COPD. In bronchial epithelium of COPD and smokers markers of EMT were described. In vitro, EMT may be reproduced by exposing well-differentiated human bronchial epithelial cells (WD-HBEC) to cigarette smoke extract (CSE). EMT may be mitigated by an increase in cellular cAMP. OBJECTIVE This study explored the effects of roflumilast N-oxide, a PDE4 inhibitor on CSE-induced EMT in WD-HBEC and in primary bronchial epithelial cells from smokers and COPD in vitro. METHODS WD-HBEC from normal donors were stimulated with CSE (2.5%) for 72 h in presence of roflumilast N-oxide (2 nM or 1 μM) or vehicle. mRNA and protein of EMT markers αSMA, vimentin, collagen-1, E-cadherin, ZO-1, KRT5 as well as NOX4 were quantified by real-time quantitative PCR or protein array, respectively. Phosphorylated and total ERK1/2 and Smad3 were assessed by protein array. cAMP and TGFβ1 were measured by ELISA. Reactive oxygen species (ROS) were determined by DCF fluorescence, after 30 min CSE (2.5%). Apoptosis was measured with Annexin V/PI labelling. In some experiments, EMT markers were determined in monolayers of bronchial epithelial cells from smokers, COPD versus controls. RESULTS Roflumilast N-oxide protected from CSE-induced EMT in WD-HBEC. The PDE4 inhibitor reversed both the increase in mesenchymal and the loss in epithelial EMT markers. Roflumilast N-oxide restored the loss in cellular cAMP following CSE, reduced ROS, NOX4 expression, the increase in TGFβ1 release, phospho ERK1/2 and Smad3. The PDE4 inhibitor partly protected from the increment in apoptosis with CSE. Finally the PDE4 inhibitor decreased mesenchymal yet increased epithelial phenotype markers in HBEC of COPD and smokers. CONCLUSIONS Roflumilast N-oxide may mitigate epithelial-mesenchymal transition in bronchial epithelial cells in vitro.
Collapse
Affiliation(s)
- Javier Milara
- Clinical Research Unit (UIC), University General Hospital Consortium, Valencia, Spain; Department of Biotechnology, Universidad Politécnica de Valencia, Spain; Research Foundation of General Hospital of Valencia, Spain.
| | - Teresa Peiró
- Research Foundation of General Hospital of Valencia, Spain; Department of Pharmacology, Faculty of Medicine, University of Valencia, Spain
| | - Adela Serrano
- Research Foundation of General Hospital of Valencia, Spain; CIBERES, Health Institute Carlos III, Valencia, Spain
| | - Ricardo Guijarro
- Department of Medicine, Faculty of Medicine, University of Valencia, Spain; Thoracic Surgery Unit, University General Hospital Consortium, Valencia, Spain
| | | | - Herman Tenor
- Takeda Pharmaceuticals International, Zürich, Switzerland
| | - Julio Cortijo
- Clinical Research Unit (UIC), University General Hospital Consortium, Valencia, Spain; Research Foundation of General Hospital of Valencia, Spain; Department of Pharmacology, Faculty of Medicine, University of Valencia, Spain; CIBERES, Health Institute Carlos III, Valencia, Spain
| |
Collapse
|
30
|
Kang HR, Lee JY, Lee CG. TGF-β1 as a therapeutic target for pulmonary fibrosis and COPD. Expert Rev Clin Pharmacol 2014; 1:547-58. [PMID: 24410556 DOI: 10.1586/17512433.1.4.547] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
TGF-β1 is a multifunctional molecule that is expressed in an exaggerated fashion during injury, inflammation and repair. Its expression is dysregulated in lung tissues from patients with pulmonary fibrosis and chronic obstructive pulmonary disease. In animal models, introduction of TGF-β1 expression in the lung causes prominent tissue fibrosis and alveolar destruction. On the other hand, the exaggerated production of TGF-β1, an inability to activate TGF-β1 or a block in TGF-β1 signaling have all been associated with the development of emphysematous pulmonary lesions. A number of studies have demonstrated that TGF-β1 is a major player in the pathogenesis of pulmonary fibrosis and emphysema. In this review, we discuss how TGF-β1 expression is regulated and mechanistically related to the development of tissue fibrosis and emphysema in experimental animal models and humans. We further highlight potential therapeutic options that control TGF-β1-associated genes or signals to restore extracellular matrix homeostasis in which TGF-β1 plays a central role.
Collapse
Affiliation(s)
- Hye-Ryun Kang
- Department of Internal Medicine, Hallym University School of Medicine, Anyang, Korea
| | | | | |
Collapse
|
31
|
Rampanelli E, Rouschop K, Teske GJD, Claessen N, Leemans JC, Florquin S. CD44v3-v10 reduces the profibrotic effects of TGF-β1 and attenuates tubular injury in the early stage of chronic obstructive nephropathy. Am J Physiol Renal Physiol 2013; 305:F1445-54. [PMID: 24026183 DOI: 10.1152/ajprenal.00340.2013] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
CD44 family members are cell surface glycoproteins, which are expressed on tubular epithelial cells (TEC) solely upon kidney injury and are involved in renal fibrosis development. Renal interstitial fibrosis is the final manifestation of chronic kidney diseases and is regulated by a complex network of cytokines, including the profibrotic factor transforming growth factor-β1 (TGF-β1) and the two antifibrotic cytokines bone morphogenic protein-7 (BMP-7) and hepatocyte growth factor (HGF). The present study investigates the potential role of CD44 standard (CD44s) and CD44v3-v10 (CD44v3) isoforms as modulators of the balance between TGF-β1 and HGF/BMP-7. CD44s is the shortest and most common isoform. CD44v3-v10 (CD44v3) has heparan sulfate moieties, which enable the binding to HGF/BMP-7, and hence, might exert renoprotective effects. Using transgenic mice overexpressing either CD44s or CD44v3 specifically on proximal TEC, we found that in vitro the overexpression of CD44v3 on primary TEC renders cells less susceptible to TGF-β1 profibrotic actions and more sensitive to BMP-7 and HGF compared with TEC overexpressing CD44s. One day after unilateral ureteric obstruction, obstructed kidneys from CD44v3 transgenic mice showed less tubular damage and myofibroblasts accumulation, which was associated with decreased TGF-β1 signaling and increased BMP-7 synthesis and signaling compared with kidneys from wild-type and CD44s transgenic mice. These data suggest that CD44v3 plays a renoprotective role in early stage of chronic obstructive nephropathy.
Collapse
Affiliation(s)
- Elena Rampanelli
- Dept. of Pathology, Rm. L2-112, Academic Medical Center, P.O. Box 22660, 1100 AZ, Amsterdam, The Netherlands.
| | | | | | | | | | | |
Collapse
|
32
|
Song JS, Kang CM, Park CK, Yoon HK. Thrombin induces epithelial-mesenchymal transition via PAR-1, PKC, and ERK1/2 pathways in A549 cells. Exp Lung Res 2013; 39:336-48. [PMID: 23919450 PMCID: PMC3793269 DOI: 10.3109/01902148.2013.820809] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Thrombin activates protease-activated receptor (PAR)-1 and induces a myofibroblast phenotype in normal lung fibroblasts. The origins of myofibroblasts are resident fibroblasts, fibrocytes, and epithelial-mesenchymal transition (EMT). We investigated the effects of thrombin, an important mediator of interstitial lung fibrosis, on EMT in A549 human alveolar epithelial cells. We show that thrombin induced EMT and collagen I secretion through the activation of PAR-1, and PKC and ERK1/2 phosphorylation in A549 cells. These effects were largely prevented by a specific PAR-1 antagonist, short interfering RNA (siRNA) directed against PAR-1, or specific PKCα/β, δ, and ε inhibitors. These data indicated that interaction with thrombin and alveolar epithelial cells might directly contribute to the pathogenesis of pulmonary fibrosis through EMT. Targeting PAR-1 on the pulmonary epithelium or specific inhibitors to PKCα/β, δ, and ε might stop the fibrotic processes in human idiopathic pulmonary fibrosis by preventing thrombin-induced EMT.
Collapse
Affiliation(s)
- Jeong Sup Song
- Department of Internal Medicine, Yeouido St Mary's Hospital, Catholic University Medical College , Seoul, Korea
| | | | | | | |
Collapse
|
33
|
Yoon YS, Lee YJ, Choi JY, Cho MS, Kang JL. Coordinated induction of cyclooxygenase-2/prostaglandin E2 and hepatocyte growth factor by apoptotic cells prevents lung fibrosis. J Leukoc Biol 2013; 94:1037-49. [PMID: 23922381 DOI: 10.1189/jlb.0513255] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Apoptotic cell instillation after bleomycin induces persistent HGF production and protects from pulmonary fibrosis, but the underlying mechanism remains unclear. We investigated immediate and prolonged effects of in vivo instillation of apoptotic cells into bleomycin-stimulated mouse lungs (2 days old) on COX-2 expression in lung tissue and alveolar macrophages and PGE2 production in BALF. Furthermore, functional interaction between these molecules and HGF, following apoptotic cell instillation in a bleomycin-induced lung fibrosis model, was assessed. Apoptotic cell instillation results in enhanced immediate and prolonged expression of COX-2 and PGE2 when compared with those from bleomycin-only-treated mice. Coadministration of the COX-2-selective inhibitor NS398 or the selective PGE2R EP2 inhibitor AH6809 inhibited the increase in HGF production. Inhibition of HGF signaling using PHA-665752 inhibited increases in COX-2 and PGE2. Long-term inhibition of COX-2, PGE2, or HGF reversed the reduction of TGF-β, apoptotic and MPO activities, protein levels, and hydroxyproline contents. Up-regulation of COX-2/PGE2 and HGF through a positive-feedback loop may be an important mechanism whereby apoptotic cell instillation exerts the net results of anti-inflammatory, antiapoptotic, and antifibrotic action.
Collapse
Affiliation(s)
- Young-So Yoon
- 1.Tissue Injury Defense Research Center, School of Medicine, Ewha Womans University, 911-1 Mok-6-dong, Yangcheon-ku, Seoul 158-056, Korea.
| | | | | | | | | |
Collapse
|
34
|
Shimokawa M, Haraguchi M, Kobayashi W, Higashi Y, Matsushita S, Kawai K, Kanekura T, Ozawa M. The transcription factor Snail expressed in cutaneous squamous cell carcinoma induces epithelial-mesenchymal transition and down-regulates COX-2. Biochem Biophys Res Commun 2013; 430:1078-82. [PMID: 23261444 DOI: 10.1016/j.bbrc.2012.12.035] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Accepted: 12/07/2012] [Indexed: 11/19/2022]
Abstract
Cutaneous spindle cell squamous cell carcinoma (SCC) is a rare, but highly malignant variant of SCC. The presence of spindle-shaped cells with a sarcomatous appearance, which are derived from squamous cells, suggests that these cells are produced as a result of epithelial-mesenchymal transition (EMT). EMT is a complex process in which epithelial cells lose their polarity and cell-cell contacts, while also acquiring increased motility and invasiveness. Snail regulates EMT by binding to proximal E-boxes in the promoter region of E-cadherin and repressing its transcription. When examining the expression of EMT markers and Snail in spindle cell SCCs, we found that cyclooxygenase-2 (COX-2) expression was down-regulated. Since it has been shown that COX-2 is constitutively overexpressed in a variety of malignancies, including colon, gastric, and lung carcinomas, the down-regulation of COX-2 expression was unexpected. The presence of E-box-like sequences in the promoter region of COX-2 prompted us to perform a more detailed analysis. We introduced a Snail expression vector into keratinocyte-derived cell lines (HaKaT, HSC5, and A431 cells), and isolated stable transfectants. We determined that COX-2 expression was down-regulated in cells expressing Snail. Consistent with these observations, reporter assays revealed that COX-2 promoter activity was repressed upon Snail overexpression. Thus Snail down-regulates COX-2 in these cells.
Collapse
Affiliation(s)
- Mitsuyoshi Shimokawa
- Department of Biochemistry and Molecular Biology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima 890-8544, Japan
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Insel PA, Murray F, Yokoyama U, Romano S, Yun H, Brown L, Snead A, Lu D, Aroonsakool N. cAMP and Epac in the regulation of tissue fibrosis. Br J Pharmacol 2012; 166:447-56. [PMID: 22233238 DOI: 10.1111/j.1476-5381.2012.01847.x] [Citation(s) in RCA: 112] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Fibrosis, the result of excess deposition of extracellular matrix (ECM), in particular collagen, leads to scarring and loss of function in tissues that include the heart, lung, kidney and liver. The second messenger cAMP can inhibit the formation and extent of ECM during this late phase of inflammation, but the mechanisms for these actions of cAMP and of agents that elevate tissue cAMP levels are not well understood. In this article, we review the fibrotic process and focus on two recently recognized aspects of actions of cAMP and its effector Epac (Exchange protein activated by cAMP): (a) blunting of epithelial-mesenchymal transformation (EMT) and (b) down-regulation of Epac expression by profibrotic agents (e.g. TGF-β, angiotensin II), which may promote tissue fibrosis by decreasing Epac-mediated antifibrotic actions. Pharmacological approaches that raise cAMP or blunt the decrease in Epac expression by profibrotic agents may thus be strategies to block or perhaps reverse tissue fibrosis. LINKED ARTICLES This article is part of a themed section on Novel cAMP Signalling Paradigms. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2012.166.issue-2.
Collapse
Affiliation(s)
- Paul A Insel
- Departments of Pharmacology Medicine, University of California San Diego, La Jolla, CA 92093, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
A comparison of epithelial-to-mesenchymal transition and re-epithelialization. Semin Cancer Biol 2012; 22:471-83. [PMID: 22863788 DOI: 10.1016/j.semcancer.2012.07.003] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2012] [Accepted: 07/20/2012] [Indexed: 12/21/2022]
Abstract
Wound healing and cancer metastasis share a common starting point, namely, a change in the phenotype of some cells from stationary to motile. The term, epithelial-to-mesenchymal transition (EMT) describes the changes in molecular biology and cellular physiology that allow a cell to transition from a sedentary cell to a motile cell, a process that is relevant not only for cancer and regeneration, but also for normal development of multicellular organisms. The present review compares the similarities and differences in cellular response at the molecular level as tumor cells enter EMT or as keratinocytes begin the process of re-epithelialization of a wound. Looking toward clinical interventions that might modulate these processes, the mechanisms and outcomes of current and potential therapies are reviewed for both anti-cancer and pro-wound healing treatments related to the pathways that are central to EMT. Taken together, the comparison of re-epithelialization and tumor EMT serves as a starting point for the development of therapies that can selectively modulate different forms of EMT.
Collapse
|
37
|
Jackson WM, Nesti LJ, Tuan RS. Mesenchymal stem cell therapy for attenuation of scar formation during wound healing. Stem Cell Res Ther 2012; 3:20. [PMID: 22668751 PMCID: PMC3392767 DOI: 10.1186/scrt111] [Citation(s) in RCA: 196] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Scars are a consequence of cutaneous wound healing that can be both unsightly and detrimental to the function of the tissue. Scar tissue is generated by excessive deposition of extracellular matrix tissue by wound healing fibroblasts and myofibroblasts, and although it is inferior to the uninjured skin, it is able to restore integrity to the boundary between the body and its environment. Scarring is not a necessary process to repair the dermal tissues. Rather, scar tissue forms due to specific mechanisms that occur during the adult wound healing process and are modulated primarily by the inflammatory response at the site of injury. Adult tissue-derived mesenchymal stem cells, which participate in normal wound healing, are trophic mediators of tissue repair. These cells participate in attenuating inflammation in the wound and reprogramming the resident immune and wound healing cells to favor tissue regeneration and inhibit fibrotic tissue formation. As a result, these cells have been considered and tested as a likely candidate for a cellular therapy to promote scar-less wound healing. This review identifies specific mechanisms by which mesenchymal stem cells can limit tissue fibrosis and summarizes recent in vivo studies where these cells have been used successfully to limit scar formation.
Collapse
Affiliation(s)
- Wesley M Jackson
- Clinical and Experimental Orthopaedics Laboratory, Department of SurgeryUniformed Services University, Bethesda, MD 20814, USA
| | | | | |
Collapse
|
38
|
The intrinsic prostaglandin E2-EP4 system of the renal tubular epithelium limits the development of tubulointerstitial fibrosis in mice. Kidney Int 2012; 82:158-71. [PMID: 22513820 DOI: 10.1038/ki.2012.115] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Inflammatory responses in the kidney lead to tubulointerstitial fibrosis, a common feature of chronic kidney diseases. Here we examined the role of prostaglandin E(2) (PGE(2)) in the development of tubulointerstitial fibrosis. In the kidneys of wild-type mice, unilateral ureteral obstruction leads to progressive tubulointerstitial fibrosis with macrophage infiltration and myofibroblast proliferation. This was accompanied by an upregulation of COX-2 and PGE(2) receptor subtype EP(4) mRNAs. In the kidneys of EP(4) gene knockout mice, however, obstruction-induced histological alterations were significantly augmented. In contrast, an EP(4)-specific agonist significantly attenuated these alterations in the kidneys of wild-type mice. The mRNAs for macrophage chemokines and profibrotic growth factors were upregulated in the kidneys of wild-type mice after ureteral obstruction. This was significantly augmented in the kidneys of EP(4)-knockout mice and suppressed by the EP(4) agonist but only in the kidneys of wild-type mice. Notably, COX-2 and MCP-1 proteins, as well as EP(4) mRNA, were localized in renal tubular epithelial cells after ureteral obstruction. In cultured renal fibroblasts, another EP(4)-specific agonist significantly inhibited PDGF-induced proliferation and profibrotic connective tissue growth factor production. Hence, an endogenous PGE(2)-EP(4) system in the tubular epithelium limits the development of tubulointerstitial fibrosis by suppressing inflammatory responses.
Collapse
|
39
|
Swetha G, Chandra V, Phadnis S, Bhonde R. Glomerular parietal epithelial cells of adult murine kidney undergo EMT to generate cells with traits of renal progenitors. J Cell Mol Med 2012; 15:396-413. [PMID: 19840197 PMCID: PMC3822804 DOI: 10.1111/j.1582-4934.2009.00937.x] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Glomerular parietal epithelial cells (GPECs) are known to revert to embryonic phenotype in response to renal injury. However, the mechanism of de-differentiation in GPECs and the underlying cellular processes are not fully understood. In the present study, we show that cultured GPECs of adult murine kidney undergo epithelial-mesenchymal transition (EMT) to generate cells, which express CD24, CD44 and CD29 surface antigens. Characterization by qRT-PCR and immunostaining of these clonogenic cells demonstrate that they exhibit metastable phenotype with co-expression of both epithelial (cytokeratin-18) and mesenchymal (vimentin) markers. Transcript analysis by qRT-PCR revealed high expression of metanephric mesenchymal (Pax-2, WT-1, Six-1, Eya-1, GDNF) and uteric bud (Hoxb-7, C-Ret) genes in these cells, indicating their bipotent progenitor status. Incubation of GPECs with EMT blocker Prostaglandin E2, resulted in low expression of renal progenitor markers reflecting the correlation between EMT and acquired stemness in these cells. Additional in vitro renal commitment assays confirmed their functional staminality. When injected into E13.5 kidney rudiments, the cells incorporated into the developing kidney primordia and co-culture with E13.5 spinal cord resulted in branching and tubulogenesis in these cells. When implanted under renal capsule of unilaterally nephrectomized mice, these cells differentiated into immature glomeruli and vascular ducts. Our study demonstrates that EMT plays a major role in imparting plasticity to terminally differentiated GPECs by producing metastable cells with traits of kidney progenitors. The present study would improve our understanding on epithelial cell plasticity, furthering our knowledge of its role in renal repair and regeneration.
Collapse
Affiliation(s)
- G Swetha
- Tissue Engineering and Banking Laboratory, National Centre for Cell Science, Pune, India
| | | | | | | |
Collapse
|
40
|
Osada S, Imai H, Sasaki Y, Tanaka Y, Matsuhashi N, Okumura N, Nagase M, Nonaka K, Yoshida K. Therapeutic strategies for synchronous and multiple liver metastases from colorectal cancer. Oncol Rev 2012; 6:e9. [PMID: 25992211 PMCID: PMC4419631 DOI: 10.4081/oncol.2012.e9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2012] [Revised: 05/15/2012] [Accepted: 05/28/2012] [Indexed: 01/23/2023] Open
Abstract
Metastasis in the liver is one of the most critical factors in the prognosis of patients with colorectal cancer. The incidence of synchronous liver metastasis has been found to be approximately 20–25%, but the optimal timing of surgical resection remains controversial. Neoadjuvant chemotherapy has also been found to be beneficial not only for initially unresectable but also resectable synchronous metastases and traditional surgical strategies of hepatic resection with past chemotherapeutic regimens have been used less and less over the past several years. This review will discuss treatments in association with the recently developed chemotherapeutic regimens.
Collapse
Affiliation(s)
- Shinji Osada
- Surgical Oncology, Gifu University School of Medicine, Gifu, Japan
| | - Hisashi Imai
- Surgical Oncology, Gifu University School of Medicine, Gifu, Japan
| | - Yoshiyuki Sasaki
- Surgical Oncology, Gifu University School of Medicine, Gifu, Japan
| | - Yoshihiro Tanaka
- Surgical Oncology, Gifu University School of Medicine, Gifu, Japan
| | | | - Naoki Okumura
- Surgical Oncology, Gifu University School of Medicine, Gifu, Japan
| | - Michitaka Nagase
- Surgical Oncology, Gifu University School of Medicine, Gifu, Japan
| | - Kenichi Nonaka
- Surgical Oncology, Gifu University School of Medicine, Gifu, Japan
| | - Kazuhiro Yoshida
- Surgical Oncology, Gifu University School of Medicine, Gifu, Japan
| |
Collapse
|
41
|
Prostaglandins in cancer cell adhesion, migration, and invasion. Int J Cell Biol 2012; 2012:723419. [PMID: 22505934 PMCID: PMC3299390 DOI: 10.1155/2012/723419] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2011] [Accepted: 10/08/2011] [Indexed: 12/21/2022] Open
Abstract
Prostaglandins exert a profound influence over the adhesive, migratory, and invasive behavior of cells during the development and progression of cancer. Cyclooxygenase-2 (COX-2) and microsomal prostaglandin E2 synthase-1 (mPGES-1) are upregulated in inflammation and cancer. This results in the production of prostaglandin E2 (PGE2), which binds to and activates G-protein-coupled prostaglandin E1–4 receptors (EP1–4). Selectively targeting the COX-2/mPGES-1/PGE2/EP1–4 axis of the prostaglandin pathway can reduce the adhesion, migration, invasion, and angiogenesis. Once stimulated by prostaglandins, cadherin adhesive connections between epithelial or endothelial cells are lost. This enables cells to invade through the underlying basement membrane and extracellular matrix (ECM). Interactions with the ECM are mediated by cell surface integrins by “outside-in signaling” through Src and focal adhesion kinase (FAK) and/or “inside-out signaling” through talins and kindlins. Combining the use of COX-2/mPGES-1/PGE2/EP1–4 axis-targeted molecules with those targeting cell surface adhesion receptors or their downstream signaling molecules may enhance cancer therapy.
Collapse
|
42
|
Jackson WM, Nesti LJ, Tuan RS. Concise review: clinical translation of wound healing therapies based on mesenchymal stem cells. Stem Cells Transl Med 2011. [PMID: 23197639 DOI: 10.5966/sctm.2011-0024] [Citation(s) in RCA: 193] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
There is enormous worldwide demand for therapies to promote the efficient resolution of hard-to-heal wounds with minimal appearance of scarring. Recent in vitro studies with mesenchymal stem cells (MSCs) have identified numerous mechanisms by which these cells can promote the process of wound healing, and there is significant interest in the clinical translation of an MSC-based therapy to promote dermal regeneration. This review provides a systematic analysis of recent preclinical and clinical research to evaluate the use of MSCs in wound healing applications. These in vivo studies provide overwhelming evidence that MSCs can accelerate wound closure by modulating the inflammatory environment, promoting the formation of a well-vascularized granulation matrix, encouraging the migration of keratinocytes, and inhibiting apoptosis of wound healing cells. The trophic effects of MSC therapy also appear to augment wound healing in diabetic tissues, thereby preventing the formation of nonhealing ulcers. Finally, a number of delivery systems have been evaluated and indicate that MSCs could be the basis of a versatile therapy to fulfill the clinical needs for dermal regeneration. However, despite the apparent advantages of MSC-based therapies, there have been only limited clinical investigations of this type of therapy in humans. Thus, our review concludes with a discussion of the translational barriers that are limiting the widespread clinical use of MSCs to enhance wound healing.
Collapse
Affiliation(s)
- Wesley M Jackson
- Clinical and Experimental Orthopaedics Laboratory, Department of Surgery, Uniformed Services University, Bethesda, Maryland, USA
| | | | | |
Collapse
|
43
|
Salvianolic acid B reverses the epithelial-to-mesenchymal transition of HK-2 cells that is induced by transforming growth factor-β. Arch Pharm Res 2011; 34:477-83. [DOI: 10.1007/s12272-011-0317-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2010] [Revised: 08/23/2010] [Accepted: 08/26/2010] [Indexed: 11/29/2022]
|
44
|
Yamamoto E, Izawa T, Juniantito V, Kuwamura M, Yamate J. Relationship of Cell Proliferating Marker Expressions with PGE(2) Receptors in Regenerating Rat Renal Tubules after Cisplatin Injection. J Toxicol Pathol 2010; 23:271-5. [PMID: 22272038 PMCID: PMC3234632 DOI: 10.1293/tox.23.271] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2010] [Accepted: 07/05/2010] [Indexed: 11/20/2022] Open
Abstract
Cisplatin, an anticancer drug, is well known to have nephrotoxicity as an adverse effect. We investigated the expressions of cell cycle markers and prostaglandin E(2) (PGE(2)) receptors (EP) in the affected renal tubules in rats injected with a single dose (6 mg/kg body weight) of cisplatin. On days 1-3 after dosing, the affected renal epithelial cells were almost desquamated, showing necrosis. On day 5 onwards, the renal tubules were rimmed by flattened or cuboidal epithelial cells with basophilic cytoplasm; BrdU-immunopositive cells began to significantly increase, indicating regeneration. Simultaneously, TUNEL-positive apoptotic cells were also seen. On days 1-5, cyclin D1-immunopositive cells were decreased with an increased expression in p21 mRNA, indicating G(1) arrest in the cell cycle. The affected renal epithelial cells began to react to EP4 receptor, but not to EP2 receptor. Some EP4 receptor-reacting epithelial cells gave a positive reaction to BrdU or cyclin D1. Collectively, the affected renal tubules underwent various alterations such as necrosis, apoptosis, regeneration and G(1) arrest; the aspects might be influenced by endogenous PGE(2) through EP4 receptor.
Collapse
Affiliation(s)
- Emi Yamamoto
- Laboratory of Veterinary Pathology, Life and Environmental Sciences,
Osaka Prefecture University, 1–58 Rinku Ourai-Kita, Izumisano, Osaka 598-8531,
Japan
| | - Takeshi Izawa
- Laboratory of Veterinary Pathology, Life and Environmental Sciences,
Osaka Prefecture University, 1–58 Rinku Ourai-Kita, Izumisano, Osaka 598-8531,
Japan
| | - Vetnizah Juniantito
- Laboratory of Veterinary Pathology, Life and Environmental Sciences,
Osaka Prefecture University, 1–58 Rinku Ourai-Kita, Izumisano, Osaka 598-8531,
Japan
| | - Mitsuru Kuwamura
- Laboratory of Veterinary Pathology, Life and Environmental Sciences,
Osaka Prefecture University, 1–58 Rinku Ourai-Kita, Izumisano, Osaka 598-8531,
Japan
| | - Jyoji Yamate
- Laboratory of Veterinary Pathology, Life and Environmental Sciences,
Osaka Prefecture University, 1–58 Rinku Ourai-Kita, Izumisano, Osaka 598-8531,
Japan
| |
Collapse
|
45
|
Bauman KA, Wettlaufer SH, Okunishi K, Vannella KM, Stoolman JS, Huang SK, Courey AJ, White ES, Hogaboam CM, Simon RH, Toews GB, Sisson TH, Moore BB, Peters-Golden M. The antifibrotic effects of plasminogen activation occur via prostaglandin E2 synthesis in humans and mice. J Clin Invest 2010; 120:1950-60. [PMID: 20501949 DOI: 10.1172/jci38369] [Citation(s) in RCA: 119] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2010] [Accepted: 03/17/2010] [Indexed: 02/06/2023] Open
Abstract
Plasminogen activation to plasmin protects from lung fibrosis, but the mechanism underlying this antifibrotic effect remains unclear. We found that mice lacking plasminogen activation inhibitor-1 (PAI-1), which are protected from bleomycin-induced pulmonary fibrosis, exhibit lung overproduction of the antifibrotic lipid mediator prostaglandin E2 (PGE2). Plasminogen activation upregulated PGE2 synthesis in alveolar epithelial cells, lung fibroblasts, and lung fibrocytes from saline- and bleomycin-treated mice, as well as in normal fetal and adult primary human lung fibroblasts. This response was exaggerated in cells from Pai1-/- mice. Although enhanced PGE2 formation required the generation of plasmin, it was independent of proteinase-activated receptor 1 (PAR-1) and instead reflected proteolytic activation and release of HGF with subsequent induction of COX-2. That the HGF/COX-2/PGE2 axis mediates in vivo protection from fibrosis in Pai1-/- mice was demonstrated by experiments showing that a selective inhibitor of the HGF receptor c-Met increased lung collagen to WT levels while reducing COX-2 protein and PGE2 levels. Of clinical interest, fibroblasts from patients with idiopathic pulmonary fibrosis were found to be defective in their ability to induce COX-2 and, therefore, unable to upregulate PGE2 synthesis in response to plasmin or HGF. These studies demonstrate crosstalk between plasminogen activation and PGE2 generation in the lung and provide a mechanism for the well-known antifibrotic actions of the fibrinolytic pathway.
Collapse
Affiliation(s)
- Kristy A Bauman
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
The preclinical pharmacology of roflumilast--a selective, oral phosphodiesterase 4 inhibitor in development for chronic obstructive pulmonary disease. Pulm Pharmacol Ther 2010; 23:235-56. [PMID: 20381629 DOI: 10.1016/j.pupt.2010.03.011] [Citation(s) in RCA: 241] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2009] [Revised: 02/18/2010] [Accepted: 03/30/2010] [Indexed: 01/02/2023]
Abstract
After more than two decades of research into phosphodiesterase 4 (PDE4) inhibitors, roflumilast (3-cyclopropylmethoxy-4-difluoromethoxy-N-[3,5-di-chloropyrid-4-yl]-benzamide) may become the first agent in this class to be approved for patient treatment worldwide. Within the PDE family of 11 known isoenzymes, roflumilast is selective for PDE4, showing balanced selectivity for subtypes A-D, and is of high subnanomolar potency. The active principle of roflumilast in man is its dichloropyridyl N-oxide metabolite, which has similar potency as a PDE4 inhibitor as the parent compound. The long half-life and high potency of this metabolite allows for once-daily, oral administration of a single, 500-microg tablet of roflumilast. The molecular mode of action of roflumilast--PDE4 inhibition and subsequent enhancement of cAMP levels--is well established. To further understand its functional mode of action in chronic obstructive pulmonary disease (COPD), for which roflumilast is being developed, a series of in vitro and in vivo preclinical studies has been performed. COPD is a progressive, devastating condition of the lung associated with an abnormal inflammatory response to noxious particles and gases, particularly tobacco smoke. In addition, according to the Global Initiative for Chronic Obstructive Lung Disease (GOLD), significant extrapulmonary effects, including comorbidities, may add to the severity of the disease in individual patients, and which may be addressed preferentially by orally administered remedies. COPD shows an increasing prevalence and mortality, and its treatment remains a high, unmet medical need. In vivo, roflumilast mitigates key COPD-related disease mechanisms such as tobacco smoke-induced lung inflammation, mucociliary malfunction, lung fibrotic and emphysematous remodelling, oxidative stress, pulmonary vascular remodelling and pulmonary hypertension. In vitro, roflumilast N-oxide has been demonstrated to affect the functions of many cell types, including neutrophils, monocytes/macrophages, CD4+ and CD8+ T-cells, endothelial cells, epithelial cells, smooth muscle cells and fibroblasts. These cellular effects are thought to be responsible for the beneficial effects of roflumilast on the disease mechanisms of COPD, which translate into reduced exacerbations and improved lung function. As a multicomponent disease, COPD requires a broad therapeutic approach that might be achieved by PDE4 inhibition. However, as a PDE4 inhibitor, roflumilast is not a direct bronchodilator. In summary, roflumilast may be the first-in-class PDE4 inhibitor for COPD therapy. In addition to being a non-steroid, anti-inflammatory drug designed to target pulmonary inflammation, the preclinical pharmacology described in this review points to a broad functional mode of action of roflumilast that putatively addresses additional COPD mechanisms. This enables roflumilast to offer effective, oral maintenance treatment for COPD, with an acceptable tolerability profile and the potential to favourably affect the extrapulmonary effects of the disease.
Collapse
|
47
|
Maher TM, Evans IC, Bottoms SE, Mercer PF, Thorley AJ, Nicholson AG, Laurent GJ, Tetley TD, Chambers RC, McAnulty RJ. Diminished prostaglandin E2 contributes to the apoptosis paradox in idiopathic pulmonary fibrosis. Am J Respir Crit Care Med 2010; 182:73-82. [PMID: 20203246 DOI: 10.1164/rccm.200905-0674oc] [Citation(s) in RCA: 143] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
RATIONALE Patients with idiopathic pulmonary fibrosis (IPF), a progressive disease with a dismal prognosis, exhibit an unexplained disparity of increased alveolar epithelial cell (AEC) apoptosis but reduced fibroblast apoptosis. OBJECTIVES To examine whether the failure of patients with IPF to up-regulate cyclooxygenase (COX)-2, and thus the antifibrotic mediator prostaglandin (PG)E(2), accounts for this imbalance. METHODS Fibroblasts and primary type II AECs were isolated from control and fibrotic human lung tissue. The effects of COX-2 inhibition and exogenous PGE(2) on fibroblast and AEC sensitivity to Fas ligand (FasL)-induced apoptosis were assessed. MEASUREMENTS AND MAIN RESULTS IPF lung fibroblasts are resistant to FasL-induced apoptosis compared with control lung fibroblasts. Inhibition of COX-2 in control lung fibroblasts resulted in an apoptosis-resistant phenotype. Administration of PGE(2) almost doubled the rate of FasL-induced apoptosis in fibrotic lung fibroblasts compared with FasL alone. Conversely, in primary fibrotic lung type II AECs, PGE(2) protected against FasL-induced apoptosis. In human control and, to a greater extent, fibrotic lung fibroblasts, PGE(2) inhibits the phosphorylation of Akt, suggesting that regulation of this prosurvival protein kinase is an important mechanism by which PGE(2) modulates cellular apoptotic responses. CONCLUSIONS The observation that PGE(2) deficiency results in increased AEC but reduced fibroblast sensitivity to apoptosis provides a novel pathogenic insight into the mechanisms driving persistent fibroproliferation in IPF.
Collapse
Affiliation(s)
- Toby M Maher
- Centre for Respiratory Research, University College London, Rayne Building, 5 University Street, London WC1E 6JJ, UK.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Yao G, Xu L, Wu X, Xu L, Yang J, Chen H. Preventive Effects of Salvianolic Acid B on Transforming Growth Factor-β1-Induced Epithelial-to-Mesenchymal Transition of Human Kidney Cells. Biol Pharm Bull 2009; 32:882-6. [DOI: 10.1248/bpb.32.882] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Gang Yao
- Department of Nephrology, 2nd Affiliated Hospital of Nanjing Medical University
| | - Lizhi Xu
- Jiangsu Key Laboratory of Molecular Medicine, School of Medicine, Nanjing University
| | - Xiaochun Wu
- Department of Nephrology, 2nd Affiliated Hospital of Nanjing Medical University
| | - Lingling Xu
- Department of Nephrology, 2nd Affiliated Hospital of Nanjing Medical University
| | - Junwei Yang
- Department of Nephrology, 2nd Affiliated Hospital of Nanjing Medical University
| | - Huimei Chen
- Jiangsu Key Laboratory of Molecular Medicine, School of Medicine, Nanjing University
| |
Collapse
|
49
|
Jia Z, Guo X, Zhang H, Wang MH, Dong Z, Yang T. Microsomal Prostaglandin Synthase-1–Derived Prostaglandin E
2
Protects Against Angiotensin II–Induced Hypertension via Inhibition of Oxidative Stress. Hypertension 2008; 52:952-9. [DOI: 10.1161/hypertensionaha.108.111229] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Prostaglandin (PG) E
2
has an established role in the regulation of vascular tone and reactivity. The present study examined the role and mechanism of microsomal PG synthase-1 (mPGES-1) in vascular response to angiotensin II (Ang II) infusion. A 7-day Ang II infusion at 0.35 mg/kg per day via osmotic minipump had no obvious effect on mean arterial blood pressure in mPGES-1
+/+
mice but induced a marked hypertensive response in mPGES-1
−/−
mice, associated with a parallel increase in urinary 8-isoprostane excretion and aortic NADPH oxidase activity and mRNA expression of p47
phox
, gp91
phox
, and Nox1. The hypertension in mPGES-1
−/−
mice was completely prevented by Tempol treatment and was fully restored on termination of the antioxidant. Apocynin induced a similar blood pressure–lowering effect as Tempol. The Ang II infusion induced mRNA expression of mPGES-1, as well as mPGES-2 and cytosolic PGE synthase in the aortas as assessed by real-time RT-PCR. Immunohistochemistry revealed remarkably enhanced immunoreactivity of mPGES-1 mostly in vascular smooth muscle cells. In cultured vascular smooth muscle cells, Ang II exerted a direct stimulatory effect on reactive oxygen species production, NADPH oxidase activity, and expression of p47
phox
, gp91
phox
, and Nox1 that were all inhibited by PGE
2
. The −/− mice also exhibited enhanced renal hemodynamic response to acute Ang II infusion at 150 nmol/kg per minute via a jugular vein over a period of 40 minutes. These results suggest that mPGES-1–derived PGE
2
buffers Ang II–induced vasoconstriction via inhibition of NADPH oxidase–dependent reactive oxygen species production.
Collapse
Affiliation(s)
- Zhanjun Jia
- From the Department of Internal Medicine (Z.J., X.G., H.Z., T.Y.), University of Utah and Veterans’ Affairs Medical Center, Salt Lake City; Departments of Physiology (M.-H.W.) and Cellular Biology and Anatomy (Z.D.), Medical College of Georgia, Augusta; and Medical Research Service (Z.D.), Veterans’ Affairs Medical Center, Augusta, Ga
| | - Xiaohua Guo
- From the Department of Internal Medicine (Z.J., X.G., H.Z., T.Y.), University of Utah and Veterans’ Affairs Medical Center, Salt Lake City; Departments of Physiology (M.-H.W.) and Cellular Biology and Anatomy (Z.D.), Medical College of Georgia, Augusta; and Medical Research Service (Z.D.), Veterans’ Affairs Medical Center, Augusta, Ga
| | - Hui Zhang
- From the Department of Internal Medicine (Z.J., X.G., H.Z., T.Y.), University of Utah and Veterans’ Affairs Medical Center, Salt Lake City; Departments of Physiology (M.-H.W.) and Cellular Biology and Anatomy (Z.D.), Medical College of Georgia, Augusta; and Medical Research Service (Z.D.), Veterans’ Affairs Medical Center, Augusta, Ga
| | - Mong-Heng Wang
- From the Department of Internal Medicine (Z.J., X.G., H.Z., T.Y.), University of Utah and Veterans’ Affairs Medical Center, Salt Lake City; Departments of Physiology (M.-H.W.) and Cellular Biology and Anatomy (Z.D.), Medical College of Georgia, Augusta; and Medical Research Service (Z.D.), Veterans’ Affairs Medical Center, Augusta, Ga
| | - Zheng Dong
- From the Department of Internal Medicine (Z.J., X.G., H.Z., T.Y.), University of Utah and Veterans’ Affairs Medical Center, Salt Lake City; Departments of Physiology (M.-H.W.) and Cellular Biology and Anatomy (Z.D.), Medical College of Georgia, Augusta; and Medical Research Service (Z.D.), Veterans’ Affairs Medical Center, Augusta, Ga
| | - Tianxin Yang
- From the Department of Internal Medicine (Z.J., X.G., H.Z., T.Y.), University of Utah and Veterans’ Affairs Medical Center, Salt Lake City; Departments of Physiology (M.-H.W.) and Cellular Biology and Anatomy (Z.D.), Medical College of Georgia, Augusta; and Medical Research Service (Z.D.), Veterans’ Affairs Medical Center, Augusta, Ga
| |
Collapse
|
50
|
Neil JR, Johnson KM, Nemenoff RA, Schiemann WP. Cox-2 inactivates Smad signaling and enhances EMT stimulated by TGF-beta through a PGE2-dependent mechanisms. Carcinogenesis 2008; 29:2227-35. [PMID: 18725385 DOI: 10.1093/carcin/bgn202] [Citation(s) in RCA: 128] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Although it is well established that mammary tumorigenesis converts transforming growth factor-beta (TGF-beta) from a tumor suppressor to a tumor promoter, the molecular, cellular and microenvironmental mechanisms underlying the dichotomous nature of TGF-beta in mammary epithelial cells (MECs) remains to be determined definitively. Aberrant upregulation of the inducible cyclooxygenase, Cox-2, occurs frequently in breast cancers and is associated with increasing disease severity and the acquisition of metastasis; however, the impact of Cox-2 expression on normal and malignant MEC response to TGF-beta remains unknown. We show here that TGF-beta induced Cox-2 expression in normal MECs during their acquisition of an epithelial-mesenchymal transition (EMT) phenotype. Moreover, stable Cox-2 expression in normal MECs stimulated their invasion, EMT and anchorage-independent growth and inhibited their activation of Smad2/3 by TGF-beta. Conversely, antagonizing TGF-beta signaling in malignant, metastatic MECs significantly reduced their expression of Cox-2 as well as enhanced their activation of Smad2/3 by TGF-beta. Along these lines, elevated Cox-2 expression elicited prostaglandin E(2) (PGE(2)) production and the autocrine activation of EP receptors, which antagonized Smad2/3 signaling in normal and malignant MECs. Importantly, rendering normal and malignant MECs Cox-2 deficient inhibited their production of PGE(2) and acquisition of an EMT morphology as well as potentiated their nuclear accumulation of Smad2/3 and transcription of plasminogen activator inhibitor-1 and p15 messenger RNA. Collectively, our findings establish Cox-2 as a novel antagonist of Smad2/3 signaling in normal and malignant MECs; they also suggest that chemotherapeutic targeting of Cox-2 may offer new inroads in restoring the tumor-suppressing activities of TGF-beta in malignant, metastatic breast cancers.
Collapse
Affiliation(s)
- Jason R Neil
- Department of Pharmacology, University of Colorado Health Sciences Center, Aurora, CO 80045, USA
| | | | | | | |
Collapse
|