1
|
Basilico B, Palamà IE, D’Amone S, Lauro C, Rosito M, Grieco M, Ratano P, Cordella F, Sanchini C, Di Angelantonio S, Ragozzino D, Cascione M, Gigli G, Cortese B. Substrate stiffness effect on molecular crosstalk of epithelial-mesenchymal transition mediators of human glioblastoma cells. Front Oncol 2022; 12:983507. [PMID: 36091138 PMCID: PMC9454310 DOI: 10.3389/fonc.2022.983507] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 08/04/2022] [Indexed: 11/13/2022] Open
Abstract
The complexity of the microenvironment effects on cell response, show accumulating evidence that glioblastoma (GBM) migration and invasiveness are influenced by the mechanical rigidity of their surroundings. The epithelial–mesenchymal transition (EMT) is a well-recognized driving force of the invasive behavior of cancer. However, the primary mechanisms of EMT initiation and progression remain unclear. We have previously showed that certain substrate stiffness can selectively stimulate human GBM U251-MG and GL15 glioblastoma cell lines motility. The present study unifies several known EMT mediators to uncover the reason of the regulation and response to these stiffnesses. Our results revealed that changing the rigidity of the mechanical environment tuned the response of both cell lines through change in morphological features, epithelial-mesenchymal markers (E-, N-Cadherin), EGFR and ROS expressions in an interrelated manner. Specifically, a stiffer microenvironment induced a mesenchymal cell shape, a more fragmented morphology, higher intracellular cytosolic ROS expression and lower mitochondrial ROS. Finally, we observed that cells more motile showed a more depolarized mitochondrial membrane potential. Unravelling the process that regulates GBM cells’ infiltrative behavior could provide new opportunities for identification of new targets and less invasive approaches for treatment.
Collapse
Affiliation(s)
| | - Ilaria Elena Palamà
- National Research Council-Nanotechnology Institute (CNR Nanotec), Lecce, Italy
| | - Stefania D’Amone
- National Research Council-Nanotechnology Institute (CNR Nanotec), Lecce, Italy
| | - Clotilde Lauro
- Department of Physiology and Pharmacology, Sapienza University, Rome, Italy
| | - Maria Rosito
- Department of Physiology and Pharmacology, Sapienza University, Rome, Italy
- Center for Life Nanoscience, Italian Institute of Technology (IIT), Rome, Italy
| | - Maddalena Grieco
- National Research Council-Nanotechnology Institute (CNR Nanotec), Lecce, Italy
| | - Patrizia Ratano
- National Research Council-Nanotechnology Institute (CNR Nanotec), Rome, Italy
| | - Federica Cordella
- Center for Life Nanoscience, Italian Institute of Technology (IIT), Rome, Italy
| | - Caterina Sanchini
- Center for Life Nanoscience, Italian Institute of Technology (IIT), Rome, Italy
| | - Silvia Di Angelantonio
- Department of Physiology and Pharmacology, Sapienza University, Rome, Italy
- Center for Life Nanoscience, Italian Institute of Technology (IIT), Rome, Italy
| | - Davide Ragozzino
- Department of Physiology and Pharmacology, Sapienza University, Rome, Italy
| | | | - Giuseppe Gigli
- Department of Physiology and Pharmacology, Sapienza University, Rome, Italy
- Department of Mathematics and Physics “Ennio De Giorgi” University of Salento, Lecce, Italy
| | - Barbara Cortese
- National Research Council-Nanotechnology Institute (CNR Nanotec), Rome, Italy
- *Correspondence: Barbara Cortese,
| |
Collapse
|
2
|
Zhao SN, Qi RQ, Gao XH, Chen HD. Sporothrix schenckii regulates macrophage inflammatory responses via the c-JUN-induced Dab2 transcription. Exp Dermatol 2022; 31:1330-1340. [PMID: 35441732 DOI: 10.1111/exd.14580] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 03/24/2022] [Accepted: 04/13/2022] [Indexed: 11/30/2022]
Abstract
Macrophages, which serve as a bridge between innate and adaptive immunity, play an important role in sporotrichosis. Sporothrix schenckii infections can produce immune responses such as macrophage polarization and inflammatory factor secretion. In the early stages of inflammation, the expression of DAB2 in macrophages is increased, which controls the secretion of inflammatory factors and affects the polarization of macrophages. However, the expressions and mechanisms of DAB2 in sporotrichosis are not clear. In this study, we examined the expression of DAB2 and its regulation of inflammatory factors under conditions of Sporothrix schenckii infection. Our results indicated that the Sporothrix schenckii infection increased the expression of DAB2 and revealed a mixed M1/M2-like type of gene expression in BMDMs with the inhibited Il6, Il1β and Arg1, and induced Tnfα, Il10 and Mgl1. The deficiency of Dab2 gene suspended the changes of cytokines. In addition, JNK activity in BMDMs was inhibited by Sporothrix schenckii infection, leading to an increase in c-JUN. We also identified c-JUN as a transcription factor for Dab2 through chromatin immunoprecipitation and luciferase reporter assays. In an in vivo mouse model, sporotrichosis induced skin lesions were accompanied with an upregulation of c-JUN and inhibition of JNK activity, which were in accord with findings from in vitro experiments. Taken together, these findings indicate that in the early stages of Sporothrix schenckii infection there is a promotion of DAB2 expression through the JNK/c-JUN pathway, effects which can then control the expression of inflammatory factors.
Collapse
Affiliation(s)
- S N Zhao
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, China.,Key Laboratory of Immunodermatology, Ministry of Health and Ministry of Education, China.,National Engineering Research Center for Immunodermatoloigcal Theranostics, Shenyang, China
| | - R Q Qi
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, China.,Key Laboratory of Immunodermatology, Ministry of Health and Ministry of Education, China.,National Engineering Research Center for Immunodermatoloigcal Theranostics, Shenyang, China
| | - X H Gao
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, China.,Key Laboratory of Immunodermatology, Ministry of Health and Ministry of Education, China.,National Engineering Research Center for Immunodermatoloigcal Theranostics, Shenyang, China
| | - H D Chen
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, China.,Key Laboratory of Immunodermatology, Ministry of Health and Ministry of Education, China.,National Engineering Research Center for Immunodermatoloigcal Theranostics, Shenyang, China
| |
Collapse
|
3
|
Rizaldy D, Toriyama M, Kato H, Fukui R, Fujita F, Nakamura M, Okada F, Morita A, Ishii KJ. Increase in primary cilia in the epidermis of patients with atopic dermatitis and psoriasis. Exp Dermatol 2021; 30:792-803. [PMID: 33455013 DOI: 10.1111/exd.14285] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Revised: 01/09/2021] [Accepted: 01/11/2021] [Indexed: 12/19/2022]
Abstract
Primary cilia influence cell activity, and thus have a unique role in maintaining cell proliferation and differentiation. In atopic dermatitis (AD) and psoriasis, areas of skin inflammation exhibit dysregulated keratinocyte homeostasis. The role of primary cilia in these conditions remains unclear. The objectives of this study is to elucidate the incidence of primary cilia in skin inflammation and the potential mechanism underlying the dysregulation of keratinocytes. Primary cilia were observed using immunofluorescence staining. Normal skin samples were compared with skin samples from patients with AD or psoriasis in terms of cilia numbers and length. The effect of cytokine stimulation on ciliogenesis in keratinocytes was analysed using a primary keratinocyte culture. IFT88, an important ciliary intraflagellar protein, was blocked in Th2 and Th17 cytokines-stimulated keratinocytes. These effects were analysed with quantitative polymerase chain reaction and Western blot. Significant increases in ciliated cells were observed in AD and psoriasis skin samples compared with normal skin samples. The stimulation of keratinocytes using Th2 and Th17 cytokines modulated the formation of primary cilia. The amount of IFT88 in the primary cilia associated with the phosphorylation of JNK, but not p38, in keratinocytes stimulated with interleukin-13, 17A and 22. An increase of ciliated cells in the epidermis may impair keratinocyte differentiation under stress conditions caused by inflammation in both AD and psoriasis patients.
Collapse
Affiliation(s)
- Defri Rizaldy
- Laboratory of Advanced Cosmetic Science, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan.,Laboratory of Mock up Vaccine, Center for Vaccine and Adjuvant Research (CVAR), National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan.,Department of Pharmaceutical Biology, School of Pharmacy, Bandung Institute of Technology, Bandung, Indonesia
| | - Manami Toriyama
- Laboratory of Advanced Cosmetic Science, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan.,Laboratory of Mock up Vaccine, Center for Vaccine and Adjuvant Research (CVAR), National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan.,Laboratory for Molecular Signal Transduction, Graduate School of Science and Technology, Nara Institute of Science and Technology, Nara, Japan
| | - Hiroko Kato
- Laboratory of Advanced Cosmetic Science, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan.,Laboratory of Mock up Vaccine, Center for Vaccine and Adjuvant Research (CVAR), National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
| | - Runa Fukui
- Laboratory of Advanced Cosmetic Science, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan.,Laboratory of Mock up Vaccine, Center for Vaccine and Adjuvant Research (CVAR), National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
| | - Fumitaka Fujita
- Laboratory of Advanced Cosmetic Science, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan.,Laboratory of Mock up Vaccine, Center for Vaccine and Adjuvant Research (CVAR), National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan.,Mandom Corporation, Osaka, Japan
| | - Motoki Nakamura
- Laboratory of Advanced Cosmetic Science, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan.,Department of Geriatric and Environmental Dermatology, Graduate School of Medical Sciences, Nagoya City University, Nagoya, Japan
| | - Fumihiro Okada
- Laboratory of Advanced Cosmetic Science, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan.,Mandom Corporation, Osaka, Japan
| | - Akimichi Morita
- Department of Geriatric and Environmental Dermatology, Graduate School of Medical Sciences, Nagoya City University, Nagoya, Japan
| | - Ken J Ishii
- Laboratory of Mock up Vaccine, Center for Vaccine and Adjuvant Research (CVAR), National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan.,Laboratory of Vaccine Science, WPI Immunology Frontier Research Center (IFReC), Osaka University, Osaka, Japan.,Division of Vaccine Science, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
4
|
Wang K, Man K, Liu J, Liu Y, Chen Q, Zhou Y, Yang Y. Microphysiological Systems: Design, Fabrication, and Applications. ACS Biomater Sci Eng 2020; 6:3231-3257. [PMID: 33204830 PMCID: PMC7668566 DOI: 10.1021/acsbiomaterials.9b01667] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Microphysiological systems, including organoids, 3-D printed tissue constructs and organ-on-a-chips (organ chips), are physiologically relevant in vitro models and have experienced explosive growth in the past decades. Different from conventional, tissue culture plastic-based in vitro models or animal models, microphysiological systems recapitulate key microenvironmental characteristics of human organs and mimic their primary functions. The advent of microphysiological systems is attributed to evolving biomaterials, micro-/nanotechnologies and stem cell biology, which enable the precise control over the matrix properties and the interactions between cells, tissues and organs in physiological conditions. As such, microphysiological systems have been developed to model a broad spectrum of organs from microvasculature, eye, to lung and many others to understand human organ development and disease pathology and facilitate drug discovery. Multiorgans-on-a-chip systems have also been developed by integrating multiple associated organ chips in a single platform, which allows to study and employ the organ function in a systematic approach. Here we first discuss the design principles of microphysiological systems with a focus on the anatomy and physiology of organs, and then review the commonly used fabrication techniques and biomaterials for microphysiological systems. Subsequently, we discuss the recent development of microphysiological systems, and provide our perspectives on advancing microphysiological systems for preclinical investigation and drug discovery of human disease.
Collapse
Affiliation(s)
- Kai Wang
- Department of Biomedical Engineering, University of North Texas, Denton, Texas 76207, United States
| | - Kun Man
- Department of Biomedical Engineering, University of North Texas, Denton, Texas 76207, United States
| | - Jiafeng Liu
- Department of Biomedical Engineering, University of North Texas, Denton, Texas 76207, United States
| | - Yang Liu
- North Texas Eye Research Institute, Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, Fort Worth, Texas 76107, United States
| | - Qi Chen
- The Key Laboratory of Innate Immune Biology of Fujian Province, Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University, Fuzhou 350117, China
| | - Yong Zhou
- Department of Emergency, Xinqiao Hospital, Chongqing 400037, China
| | - Yong Yang
- Department of Biomedical Engineering, University of North Texas, Denton, Texas 76207, United States
| |
Collapse
|
5
|
Substrate softness promotes terminal differentiation of human keratinocytes without altering their ability to proliferate back into a rigid environment. Arch Dermatol Res 2019; 311:741-751. [DOI: 10.1007/s00403-019-01962-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 05/13/2019] [Accepted: 06/15/2019] [Indexed: 12/20/2022]
|
6
|
Wood RA, Barbour MJ, Gould GW, Cunningham MR, Plevin RJ. Conflicting evidence for the role of JNK as a target in breast cancer cell proliferation: Comparisons between pharmacological inhibition and selective shRNA knockdown approaches. Pharmacol Res Perspect 2018; 6. [PMID: 29417765 PMCID: PMC5817830 DOI: 10.1002/prp2.376] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 11/01/2017] [Indexed: 11/11/2022] Open
Abstract
As a target, the JNK pathway has been implicated in roles including cell death, proliferation, and inflammation in variety of contexts which span cardiovascular disease, neurodegenerative pathologies, and cancer. JNK1 and JNK2 have recently been demonstrated to function independently, highlighting a new parameter in the study of the JNK pathway. In order for JNK1 and JNK2-specific roles to be defined, better tools need to be employed. Previous studies have relied upon the broad spectrum JNK inhibitor, SP600125, to characterize the role of JNK signaling in a number of cell lines, including the breast cancer cell line MCF-7. In line with previous literature, our study has demonstrated that SP600125 treatment inhibited c-Jun and JNK phosphorylation and MCF-7 proliferation. However, in addition to targeting JNK1, JNK2, and JNK3, SP600125 has been previously demonstrated to suppress the activity of a number of other serine/threonine kinases, making SP600125 an inadequate tool for JNK isoform-specific roles to be determined. In this study, lentiviral shRNA was employed to selectively knockdown JNK1, JNK2, and JNK1/2 in MCF-7 cells. Using this approach, JNK phosphorylation was fully inhibited following stable knockdown of respective JNK isoforms. Interestingly, despite suppression of JNK phosphorylation, MCF-7 cell proliferation, cell cycle progression, or cell death remained unaffected. These findings raise the question of whether JNK phosphorylation really is pivotal in MCF-7 cell growth and death or if suppression of these events is a result of one of the many off-targets cited for SP600125.
Collapse
Affiliation(s)
- Rachel A Wood
- Strathclyde Institute for Pharmacy and Biomedical Sciences (SIPBS), University of Strathclyde, Glasgow, UK
| | - Mark J Barbour
- Strathclyde Institute for Pharmacy and Biomedical Sciences (SIPBS), University of Strathclyde, Glasgow, UK
| | - Gwyn W Gould
- Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Margaret R Cunningham
- Strathclyde Institute for Pharmacy and Biomedical Sciences (SIPBS), University of Strathclyde, Glasgow, UK
| | - Robin J Plevin
- Strathclyde Institute for Pharmacy and Biomedical Sciences (SIPBS), University of Strathclyde, Glasgow, UK
| |
Collapse
|
7
|
Lu H, Liu S, Zhang G, Wu B, Zhu Y, Frederick DT, Hu Y, Zhong W, Randell S, Sadek N, Zhang W, Chen G, Cheng C, Zeng J, Wu LW, Zhang J, Liu X, Xu W, Krepler C, Sproesser K, Xiao M, Miao B, Liu J, Song CD, Liu JY, Karakousis GC, Schuchter LM, Lu Y, Mills G, Cong Y, Chernoff J, Guo J, Boland GM, Sullivan RJ, Wei Z, Field J, Amaravadi RK, Flaherty KT, Herlyn M, Xu X, Guo W. PAK signalling drives acquired drug resistance to MAPK inhibitors in BRAF-mutant melanomas. Nature 2017; 550:133-136. [PMID: 28953887 PMCID: PMC5891348 DOI: 10.1038/nature24040] [Citation(s) in RCA: 128] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 08/24/2017] [Indexed: 12/18/2022]
Abstract
Targeted BRAF inhibition (BRAFi) and combined BRAF and MEK inhibition (BRAFi and MEKi) therapies have markedly improved the clinical outcomes of patients with metastatic melanoma. Unfortunately, the efficacy of these treatments is often countered by the acquisition of drug resistance. Here we investigated the molecular mechanisms that underlie acquired resistance to BRAFi and to the combined therapy. Consistent with previous studies, we show that resistance to BRAFi is mediated by ERK pathway reactivation. Resistance to the combined therapy, however, is mediated by mechanisms independent of reactivation of ERK in many resistant cell lines and clinical samples. p21-activated kinases (PAKs) become activated in cells with acquired drug resistance and have a pivotal role in mediating resistance. Our screening, using a reverse-phase protein array, revealed distinct mechanisms by which PAKs mediate resistance to BRAFi and the combined therapy. In BRAFi-resistant cells, PAKs phosphorylate CRAF and MEK to reactivate ERK. In cells that are resistant to the combined therapy, PAKs regulate JNK and β-catenin phosphorylation and mTOR pathway activation, and inhibit apoptosis, thereby bypassing ERK. Together, our results provide insights into the molecular mechanisms underlying acquired drug resistance to current targeted therapies, and may help to direct novel drug development efforts to overcome acquired drug resistance.
Collapse
Affiliation(s)
- Hezhe Lu
- Department of Biology, the Hospital of the University of Pennsylvania, and Department of Dermatology, Perelman School of Medicine, University of Pennsylvania
| | - Shujing Liu
- Department of Pathology and Laboratory Medicine, the Hospital of the University of Pennsylvania, and Department of Dermatology, Perelman School of Medicine, University of Pennsylvania
| | - Gao Zhang
- Molecular and Cellular Oncogenesis Program and Melanoma Research Center, The Wistar Institute, Philadelphia, PA19104, U.S.A
| | - Bin Wu
- Department of Biology, the Hospital of the University of Pennsylvania, and Department of Dermatology, Perelman School of Medicine, University of Pennsylvania
| | - Yueyao Zhu
- Department of Biology, the Hospital of the University of Pennsylvania, and Department of Dermatology, Perelman School of Medicine, University of Pennsylvania
| | | | - Yi Hu
- Department of Biology, Drexel University, Philadelphia, PA19104, U.S.A
| | - Wenqun Zhong
- Department of Biology, the Hospital of the University of Pennsylvania, and Department of Dermatology, Perelman School of Medicine, University of Pennsylvania
| | - Sergio Randell
- Molecular and Cellular Oncogenesis Program and Melanoma Research Center, The Wistar Institute, Philadelphia, PA19104, U.S.A
| | - Norah Sadek
- Molecular and Cellular Oncogenesis Program and Melanoma Research Center, The Wistar Institute, Philadelphia, PA19104, U.S.A
| | - Wei Zhang
- Department of Biology, the Hospital of the University of Pennsylvania, and Department of Dermatology, Perelman School of Medicine, University of Pennsylvania
| | - Gang Chen
- Department of Biology, the Hospital of the University of Pennsylvania, and Department of Dermatology, Perelman School of Medicine, University of Pennsylvania
| | - Chaoran Cheng
- Department of Computer Science, New Jersey Institute of Technology, Newark, NJ07102, U.S.A
| | - Jingwen Zeng
- Department of Biology, the Hospital of the University of Pennsylvania, and Department of Dermatology, Perelman School of Medicine, University of Pennsylvania
| | - Lawrence W. Wu
- Molecular and Cellular Oncogenesis Program and Melanoma Research Center, The Wistar Institute, Philadelphia, PA19104, U.S.A
| | - Jie Zhang
- Department of Computer Science, New Jersey Institute of Technology, Newark, NJ07102, U.S.A
| | - Xiaoming Liu
- Department of Pathology and Laboratory Medicine, the Hospital of the University of Pennsylvania, and Department of Dermatology, Perelman School of Medicine, University of Pennsylvania
| | - Wei Xu
- Abramson Cancer Center and Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, U.S.A
| | - Clemens Krepler
- Molecular and Cellular Oncogenesis Program and Melanoma Research Center, The Wistar Institute, Philadelphia, PA19104, U.S.A
| | - Katrin Sproesser
- Molecular and Cellular Oncogenesis Program and Melanoma Research Center, The Wistar Institute, Philadelphia, PA19104, U.S.A
| | - Min Xiao
- Molecular and Cellular Oncogenesis Program and Melanoma Research Center, The Wistar Institute, Philadelphia, PA19104, U.S.A
| | - Benchun Miao
- Massachusetts General Hospital Cancer Center, Boston, MA02114, U.S.A
| | - Jianglan Liu
- Molecular and Cellular Oncogenesis Program and Melanoma Research Center, The Wistar Institute, Philadelphia, PA19104, U.S.A
| | - Claire D. Song
- Department of Biology, the Hospital of the University of Pennsylvania, and Department of Dermatology, Perelman School of Medicine, University of Pennsylvania
| | - Jephrey Y. Liu
- Abramson Cancer Center and Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, U.S.A
| | - Giorgos C. Karakousis
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, PA19104, U.S.A
| | - Lynn M. Schuchter
- Abramson Cancer Center and Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, U.S.A
| | - Yiling Lu
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX77054, USA
| | - Gordon Mills
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX77054, USA
| | - Yusheng Cong
- Institute of Aging Research, Hangzhou Normal University School of Medicine, Hangzhou, China
| | - Jonathan Chernoff
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, PA19111, U.S.A
| | - Jun Guo
- Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing, 100036, China
| | - Genevieve M. Boland
- Department of Surgical Oncology, Massachusetts General Hospital, Boston, Massachusetts 02114, U.S.A
| | - Ryan J. Sullivan
- Massachusetts General Hospital Cancer Center, Boston, MA02114, U.S.A
| | - Zhi Wei
- Department of Computer Science, New Jersey Institute of Technology, Newark, NJ07102, U.S.A
| | - Jeffrey Field
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104, U.S.A
| | - Ravi K. Amaravadi
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, PA19104, U.S.A
| | - Keith T. Flaherty
- Massachusetts General Hospital Cancer Center, Boston, MA02114, U.S.A
| | - Meenhard Herlyn
- Molecular and Cellular Oncogenesis Program and Melanoma Research Center, The Wistar Institute, Philadelphia, PA19104, U.S.A
| | - Xiaowei Xu
- Department of Pathology and Laboratory Medicine, the Hospital of the University of Pennsylvania, and Department of Dermatology, Perelman School of Medicine, University of Pennsylvania
| | - Wei Guo
- Department of Biology, the Hospital of the University of Pennsylvania, and Department of Dermatology, Perelman School of Medicine, University of Pennsylvania
| |
Collapse
|
8
|
Yao F, Zhang M, Chen L. Adipose Tissue-Specialized Immunologic Features Might Be the Potential Therapeutic Target of Prospective Medicines for Obesity. J Diabetes Res 2017; 2017:4504612. [PMID: 28466023 PMCID: PMC5390594 DOI: 10.1155/2017/4504612] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 03/03/2017] [Accepted: 03/13/2017] [Indexed: 12/19/2022] Open
Abstract
Excessive lipid accumulation in adipose tissue is either the source of obesity or the cause and result of chronic local inflammation, and recent studies indicate that the accumulation may induce many other specialized immunologic features with macrophages and epidemic diseases. We analyze the effective stages of immune cells in adipose tissue, including macrophage recruitment, macrophage polarization, and macrophage-like phenotype preadipocyte possession to find optimal sites as drug targets. Subsequently, some main signaling pathways are summarized in this review, including the AMP-activated protein kinase (AMPK) pathway, the JNK signaling pathway, and a novel one, the Notch signaling pathway. We illustrate all these points in order to determine the general pathogenesis of chronic low-grade local inflammation in adipose tissue and the related signaling pathways. In addition, signal-associated prospective compounds, such as berberine, are summarized and discussed with potential targets in pathogenesis. This might provide some possible thoughts and novel therapies for studying chronic inflammatory diseases, such as insulin resistance and type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Fan Yao
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Ming Zhang
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Li Chen
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
- School of Nursing, Jilin University, Changchun 130021, China
- *Li Chen:
| |
Collapse
|
9
|
Niu W, Wang Y, Wang Z, Xin Q, Wang Y, Feng L, Zhao L, Wen J, Zhang H, Wang C, Xia G. JNK signaling regulates E-cadherin junctions in germline cysts and determines primordial follicle formation in mice. Development 2016; 143:1778-87. [PMID: 27013242 DOI: 10.1242/dev.132175] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2015] [Accepted: 03/11/2016] [Indexed: 12/23/2022]
Abstract
Physiologically, the size of the primordial follicle pool determines the reproductive lifespan of female mammals, while its establishment largely depends on a process of germline cyst breakdown during the perinatal period. The mechanisms regulating this process are poorly understood. Here we demonstrate that c-Jun amino-terminal kinase (JNK) signaling is crucial for germline cyst breakdown and primordial follicle formation. JNK was specifically localized in oocytes and its activity increased as germline cyst breakdown progressed. Importantly, disruption of JNK signaling with a specific inhibitor (SP600125) or knockdown technology (Lenti-JNK-shRNAs) resulted in significantly suppressed cyst breakdown and primordial follicle formation in cultured mouse ovaries. Our results show that E-cadherin is intensely expressed in germline cysts, and that its decline is necessary for oocyte release from the cyst. However, inhibition of JNK signaling leads to aberrantly enhanced localization of E-cadherin at oocyte-oocyte contact sites. WNT4 expression is upregulated after SP600125 treatment. Additionally, similar to the effect of SP600125 treatment, WNT4 overexpression delays cyst breakdown and is accompanied by abnormal E-cadherin expression patterns. In conclusion, our results suggest that JNK signaling, which is inversely correlated with WNT4, plays an important role in perinatal germline cyst breakdown and primordial follicle formation by regulating E-cadherin junctions between oocytes in mouse ovaries.
Collapse
Affiliation(s)
- Wanbao Niu
- State Key Laboratory of Agrobiotechnology, College of Biological Science, China Agricultural University, Beijing 100193, China
| | - Ye Wang
- State Key Laboratory of Agrobiotechnology, College of Biological Science, China Agricultural University, Beijing 100193, China
| | - Zhengpin Wang
- Laboratory of Cellular and Developmental Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Qiliang Xin
- State Key Laboratory of Agrobiotechnology, College of Biological Science, China Agricultural University, Beijing 100193, China
| | - Yijing Wang
- National Institute of Biological Sciences, Zhongguancun Life Science Park, Changping, Beijing 102206, China
| | - Lizhao Feng
- State Key Laboratory of Agrobiotechnology, College of Biological Science, China Agricultural University, Beijing 100193, China
| | - Lihua Zhao
- State Key Laboratory of Agrobiotechnology, College of Biological Science, China Agricultural University, Beijing 100193, China
| | - Jia Wen
- State Key Laboratory of Agrobiotechnology, College of Biological Science, China Agricultural University, Beijing 100193, China
| | - Hua Zhang
- State Key Laboratory of Agrobiotechnology, College of Biological Science, China Agricultural University, Beijing 100193, China
| | - Chao Wang
- State Key Laboratory of Agrobiotechnology, College of Biological Science, China Agricultural University, Beijing 100193, China
| | - Guoliang Xia
- State Key Laboratory of Agrobiotechnology, College of Biological Science, China Agricultural University, Beijing 100193, China
| |
Collapse
|
10
|
Neganova I, Shmeleva E, Munkley J, Chichagova V, Anyfantis G, Anderson R, Passos J, Elliott DJ, Armstrong L, Lako M. JNK/SAPK Signaling Is Essential for Efficient Reprogramming of Human Fibroblasts to Induced Pluripotent Stem Cells. Stem Cells 2016; 34:1198-212. [PMID: 26867034 PMCID: PMC4982072 DOI: 10.1002/stem.2327] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 01/12/2016] [Indexed: 12/22/2022]
Abstract
Reprogramming of somatic cells to the phenotypic state termed “induced pluripotency” is thought to occur through three consecutive stages: initiation, maturation, and stabilisation. The initiation phase is stochastic but nevertheless very important as it sets the gene expression pattern that permits completion of reprogramming; hence a better understanding of this phase and how this is regulated may provide the molecular cues for improving the reprogramming process. c‐Jun N‐terminal kinase (JNK)/stress‐activated protein kinase (SAPKs) are stress activated MAPK kinases that play an essential role in several processes known to be important for successful completion of the initiation phase such as cellular proliferation, mesenchymal to epithelial transition (MET) and cell cycle regulation. In view of this, we postulated that manipulation of this pathway would have significant impacts on reprogramming of human fibroblasts to induced pluripotent stem cells. Accordingly, we found that key components of the JNK/SAPK signaling pathway increase expression as early as day 3 of the reprogramming process and continue to rise in reprogrammed cells throughout the initiation and maturation stages. Using both chemical inhibitors and RNA interference of MKK4, MKK7 and JNK1, we tested the role of JNK/SAPK signaling during the initiation stage of neonatal and adult fibroblast reprogramming. These resulted in complete abrogation of fully reprogrammed colonies and the emergence of partially reprogrammed colonies which disaggregated and were lost from culture during the maturation stage. Inhibition of JNK/SAPK signaling resulted in reduced cell proliferation, disruption of MET and loss of the pluripotent phenotype, which either singly or in combination prevented establishment of pluripotent colonies. Together these data provide new evidence for an indispensable role for JNK/SAPK signaling to overcome the well‐established molecular barriers in human somatic cell induced reprogramming. Stem Cells2016;34:1198–1212
Collapse
Affiliation(s)
- Irina Neganova
- Institute of Genetic Medicine, International Centre for Life and Centre for Integrated Systems Biology of Ageing and Nutrition
| | - Evgenija Shmeleva
- Institute of Genetic Medicine, International Centre for Life and Centre for Integrated Systems Biology of Ageing and Nutrition
| | - Jennifer Munkley
- Institute of Genetic Medicine, International Centre for Life and Centre for Integrated Systems Biology of Ageing and Nutrition
| | - Valeria Chichagova
- Institute of Genetic Medicine, International Centre for Life and Centre for Integrated Systems Biology of Ageing and Nutrition
| | - George Anyfantis
- Institute of Genetic Medicine, International Centre for Life and Centre for Integrated Systems Biology of Ageing and Nutrition
| | - Rhys Anderson
- Institute for Ageing and Health, Newcastle University
| | - Joao Passos
- Institute for Ageing and Health, Newcastle University
| | - David J Elliott
- Institute of Genetic Medicine, International Centre for Life and Centre for Integrated Systems Biology of Ageing and Nutrition
| | - Lyle Armstrong
- Institute of Genetic Medicine, International Centre for Life and Centre for Integrated Systems Biology of Ageing and Nutrition
| | - Majlinda Lako
- Institute of Genetic Medicine, International Centre for Life and Centre for Integrated Systems Biology of Ageing and Nutrition
| |
Collapse
|
11
|
Lee G, Kim H, Kim HM. RhoA-JNK Regulates the E-Cadherin Junctions of Human Gingival Epithelial Cells. J Dent Res 2015; 95:284-91. [DOI: 10.1177/0022034515619375] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
The junctional epithelium (JE) is unique with regard to its wide intercellular spaces and sparsely developed intercellular junctions. Thus, knowledge of the molecular mechanisms that regulate the formation of the intercellular junctions of the junctional epithelium may be essential to understand the pathophysiology of the JE. HOK-16B cells, a normal human gingival epithelial cell line, were used to identify the molecules involved in the regulation of the formation of intercellular E-cadherin junctions between human gingival epithelial cells. Activation of c-Jun N-terminal kinase (JNK) disrupted the intercellular junctions through the dissociation of E-cadherin. The role of JNK in the formation of these E-cadherin junctions was further confirmed by demonstrating that JNK inhibition induced the formation of intercellular E-cadherin junctions. The upstream signaling of JNK was also examined. Activation of the small GTPase RhoA disrupted the formation of E-cadherin junctions between HOK-16B cells, which was accompanied by JNK activation. Disruption of these intercellular junctions upon RhoA activation was prevented when JNK activity was inhibited. In contrast, RhoA inactivation led to HOK-16B cell aggregation and the formation of intercellular junctions, even under conditions in which the cellular junctions were naturally disrupted by growth on a strongly adhesive surface. Furthermore, the JE of mouse molars had high JNK activity associated with low E-cadherin expression, which was reversed in the other gingival epithelia, including the sulcular epithelium. Interestingly, JNK activity was increased in cells grown on a solid surface, where cells showed higher RhoA activity than those grown on soft surfaces. Together, these results indicate that the decreased formation of intercellular E-cadherin junctions within the JE may be coupled to high JNK activity, which is activated by the upregulation of RhoA on solid tooth surfaces.
Collapse
Affiliation(s)
- G. Lee
- Laboratory for the Study of Molecular Biointerfaces, Department of Oral Histology and Developmental Biology, Program of Cell and Developmental Biology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Republic of Korea
| | - H.J. Kim
- Laboratory for the Study of Molecular Biointerfaces, Department of Oral Histology and Developmental Biology, Program of Cell and Developmental Biology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Republic of Korea
| | - H.-M. Kim
- Laboratory for the Study of Molecular Biointerfaces, Department of Oral Histology and Developmental Biology, Program of Cell and Developmental Biology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
12
|
Zimmermann M, Kugler SJ, Schulz A, Nagel AC. Loss of putzig Activity Results in Apoptosis during Wing Imaginal Development in Drosophila. PLoS One 2015; 10:e0124652. [PMID: 25894556 PMCID: PMC4403878 DOI: 10.1371/journal.pone.0124652] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Accepted: 03/17/2015] [Indexed: 12/22/2022] Open
Abstract
The Drosophila gene putzig (pzg) encodes a nuclear protein that is an integral component of the Trf2/Dref complex involved in the transcription of proliferation-related genes. Moreover, Pzg is found in a complex together with the nucleosome remodeling factor NURF, where it promotes Notch target gene activation. Here we show that downregulation of pzg activity in the developing wing imaginal discs induces an apoptotic response, accompanied by the induction of the pro-apoptotic gene reaper, repression of Drosophila inhibitor of apoptosis protein accumulation and the activation of the caspases Drice, Caspase3 and Dcp1. As a further consequence ‘Apoptosis induced Proliferation’ (AiP) and ‘Apoptosis induced Apoptosis’ (AiA) are triggered. As expected, the activity of the stress kinase Jun N-terminal kinase (JNK), proposed to mediate both processes, is ectopically induced in response to pzg loss. In addition, the expression of the mitogen wingless (wg) but not of decapentaplegic (dpp) is observed. We present evidence that downregulation of Notch activates Dcp1 caspase and JNK signaling, however, neither induces ectopic wg nor dpp expression. In contrast, the consequences of Dref-RNAi were largely indistinguishable from pzg-RNAi with regard to apoptosis induction. Moreover, overexpression of Dref ameliorated the downregulation of pzg compatible with the notion that the two are required together to maintain cell and tissue homeostasis in Drosophila.
Collapse
Affiliation(s)
- Mirjam Zimmermann
- Institute of Genetics, University of Hohenheim, 70599 Stuttgart, Germany
| | - Sabrina J. Kugler
- Institute of Genetics, University of Hohenheim, 70599 Stuttgart, Germany
| | - Adriana Schulz
- Institute of Genetics, University of Hohenheim, 70599 Stuttgart, Germany
| | - Anja C. Nagel
- Institute of Genetics, University of Hohenheim, 70599 Stuttgart, Germany
- * E-mail:
| |
Collapse
|
13
|
Alimperti S, Andreadis ST. CDH2 and CDH11 act as regulators of stem cell fate decisions. Stem Cell Res 2015; 14:270-82. [PMID: 25771201 DOI: 10.1016/j.scr.2015.02.002] [Citation(s) in RCA: 115] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Revised: 01/24/2015] [Accepted: 02/10/2015] [Indexed: 12/14/2022] Open
Abstract
Accumulating evidence suggests that the mechanical and biochemical signals originating from cell-cell adhesion are critical for stem cell lineage specification. In this review, we focus on the role of cadherin mediated signaling in development and stem cell differentiation, with emphasis on two well-known cadherins, cadherin-2 (CDH2) (N-cadherin) and cadherin-11 (CDH11) (OB-cadherin). We summarize the existing knowledge regarding the role of CDH2 and CDH11 during development and differentiation in vivo and in vitro. We also discuss engineering strategies to control stem cell fate decisions by fine-tuning the extent of cell-cell adhesion through surface chemistry and microtopology. These studies may be greatly facilitated by novel strategies that enable monitoring of stem cell specification in real time. We expect that better understanding of how intercellular adhesion signaling affects lineage specification may impact biomaterial and scaffold design to control stem cell fate decisions in three-dimensional context with potential implications for tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Stella Alimperti
- Bioengineering Laboratory, Department of Chemical and Biological Engineering, University at Buffalo, State University of New York, Amherst, NY 14260-4200, USA
| | - Stelios T Andreadis
- Bioengineering Laboratory, Department of Chemical and Biological Engineering, University at Buffalo, State University of New York, Amherst, NY 14260-4200, USA; Center of Excellence in Bioinformatics and Life Sciences, Buffalo, NY 14203, USA.
| |
Collapse
|
14
|
Li N, Li Y, Duan X. Heat shock protein 72 confers protection in retinal ganglion cells and lateral geniculate nucleus neurons via blockade of the SAPK/JNK pathway in a chronic ocular-hypertensive rat model. Neural Regen Res 2014; 9:1395-401. [PMID: 25221598 PMCID: PMC4160872 DOI: 10.4103/1673-5374.137595] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/28/2014] [Indexed: 01/03/2023] Open
Abstract
Optic nerve transection increased the expression of heat shock protein 72 (HSP72) in the lateral geniculate body, indicating that this protein is involved in the prevention of neuronal injury. Zinc sulfate and quercetin induced and inhibited the expression of HSP72, respectively. Intraperitoneal injections of zinc sulfate, SP600125 (c-Jun N-terminal kinase inhibitor), or quercetin were performed on retinal ganglion cells in a Wistar rat model of chronic ocular hypertension. Our results showed that compared with the control group, the expression of HSP72 in retinal ganglion cells and the lateral geniculate body was increased after the injection of zinc sulfate, but was decreased after the injection of quercetin. The expression of phosphorylated c-Jun N-terminal kinases and phosphorylated c-Jun were visible 3 days after injection in the control group, and reached a peak at 7 days. Zinc sulfate and SP600125 significantly decreased the expression of p-c-Jun, whereas quercetin significantly enhanced the expression of this protein. These results suggest that HSP72 protects retinal ganglion cells and lateral geniculate body in a rat model of chronic ocular hypertension from injury by blocking the activation of the stress-activated kinase/c-Jun N-terminal kinase apoptotic pathway.
Collapse
Affiliation(s)
- Ning Li
- Department of Ophthalmology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
| | - Yuehua Li
- Department of Ophthalmology, Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Xuanchu Duan
- Department of Ophthalmology, Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| |
Collapse
|
15
|
Efficient Keratinocyte Differentiation Strictly Depends on JNK-Induced Soluble Factors in Fibroblasts. J Invest Dermatol 2014; 134:1332-1341. [DOI: 10.1038/jid.2013.535] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Revised: 11/14/2013] [Accepted: 11/16/2013] [Indexed: 12/21/2022]
|
16
|
Zhou D, Huang C, Lin Z, Zhan S, Kong L, Fang C, Li J. Macrophage polarization and function with emphasis on the evolving roles of coordinated regulation of cellular signaling pathways. Cell Signal 2014; 26:192-7. [DOI: 10.1016/j.cellsig.2013.11.004] [Citation(s) in RCA: 393] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Accepted: 11/01/2013] [Indexed: 02/06/2023]
|
17
|
Lisanti MP, Tsirigos A, Pavlides S, Reeves KJ, Peiris-Pagès M, Chadwick AL, Sanchez-Alvarez R, Lamb R, Howell A, Martinez-Outschoorn UE, Sotgia F. JNK1 stress signaling is hyper-activated in high breast density and the tumor stroma: connecting fibrosis, inflammation, and stemness for cancer prevention. Cell Cycle 2013; 13:580-99. [PMID: 24434780 DOI: 10.4161/cc.27379] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Mammography is an important screening modality for the early detection of DCIS and breast cancer lesions. More specifically, high mammographic density is associated with an increased risk of breast cancer. However, the biological processes underlying this phenomenon remain largely unknown. Here, we re-interrogated genome-wide transcriptional profiling data obtained from low-density (LD) mammary fibroblasts (n = 6 patients) and high-density (HD) mammary fibroblasts (n = 7 patients) derived from a series of 13 female patients. We used these raw data to generate a "breast density" gene signature consisting of>1250 transcripts that were significantly increased in HD fibroblasts, relative to LD fibroblasts. We then focused on the genes that were increased by ≥ 1.5-fold (P<0.05) and performed gene set enrichment analysis (GSEA), using the molecular signatures database (MSigDB). Our results indicate that HD fibroblasts show the upregulation and/or hyper-activation of several key cellular processes, including the stress response, inflammation, stemness, and signal transduction. The transcriptional profiles of HD fibroblasts also showed striking similarities to human tumors, including head and neck, liver, thyroid, lung, and breast cancers. This may reflect functional similarities between cancer-associated fibroblasts (CAFs) and HD fibroblasts. This is consistent with the idea that the presence of HD fibroblasts may be a hallmark of a pre-cancerous phenotype. In these biological processes, GSEA predicts that several key signaling pathways may be involved, including JNK1, iNOS, Rho GTPase(s), FGF-R, EGF-R, and PDGF-R-mediated signal transduction, thereby creating a pro-inflammatory, pro-proliferative, cytokine, and chemokine-rich microenvironment. HD fibroblasts also showed significant overlap with gene profiles derived from smooth muscle cells under stress (JNK1) and activated/infected macrophages (iNOS). Thus, HD fibroblasts may behave like activated myofibroblasts and macrophages, to create and maintain a fibrotic and inflammatory microenvironment. Finally, comparisons between the HD fibroblast gene signature and breast cancer tumor stroma revealed that JNK1 stress signaling is the single most significant biological process that is shared between these 2 data sets (with P values between 5.40E-09 and 1.02E-14), and is specifically associated with tumor recurrence. These results implicate "stromal JNK1 signaling" in the pathogenesis of human breast cancers and the transition to malignancy. Augmented TGF-β signaling also emerged as a common feature linking high breast density with tumor stroma and breast cancer recurrence (P = 5.23E-05). Similarities between the HD fibroblast gene signature, wound healing, and the cancer-associated fibroblast phenotype were also noted. Thus, this unbiased informatics analysis of high breast density provides a novel framework for additional experimental exploration and new hypothesis-driven breast cancer research, with a focus on cancer prevention and personalized medicine.
Collapse
Affiliation(s)
- Michael P Lisanti
- Breakthrough Breast Cancer Research Unit and the Manchester Breast Centre; Institute of Cancer Sciences; University of Manchester; Manchester, UK; Manchester Centre for Cellular Metabolism (MCCM); University of Manchester; Manchester, UK
| | - Aristotelis Tsirigos
- Computational Biology Center; IBM T.J. Watson Research Center; Yorktown Heights, NY USA
| | - Stephanos Pavlides
- Postgraduate Research Institute of Science, Technology, Environment and Medicine (PRI); Limassol, Cyprus
| | - Kimberley Jayne Reeves
- Breakthrough Breast Cancer Research Unit and the Manchester Breast Centre; Institute of Cancer Sciences; University of Manchester; Manchester, UK; Manchester Centre for Cellular Metabolism (MCCM); University of Manchester; Manchester, UK
| | - Maria Peiris-Pagès
- Breakthrough Breast Cancer Research Unit and the Manchester Breast Centre; Institute of Cancer Sciences; University of Manchester; Manchester, UK; Manchester Centre for Cellular Metabolism (MCCM); University of Manchester; Manchester, UK
| | - Amy L Chadwick
- Breakthrough Breast Cancer Research Unit and the Manchester Breast Centre; Institute of Cancer Sciences; University of Manchester; Manchester, UK; Manchester Centre for Cellular Metabolism (MCCM); University of Manchester; Manchester, UK
| | - Rosa Sanchez-Alvarez
- Breakthrough Breast Cancer Research Unit and the Manchester Breast Centre; Institute of Cancer Sciences; University of Manchester; Manchester, UK; Manchester Centre for Cellular Metabolism (MCCM); University of Manchester; Manchester, UK
| | - Rebecca Lamb
- Breakthrough Breast Cancer Research Unit and the Manchester Breast Centre; Institute of Cancer Sciences; University of Manchester; Manchester, UK; Manchester Centre for Cellular Metabolism (MCCM); Institute of Human Development; University of Manchester; Manchester, UK
| | - Anthony Howell
- Breakthrough Breast Cancer Research Unit and the Manchester Breast Centre; Institute of Cancer Sciences; University of Manchester; Manchester, UK
| | | | - Federica Sotgia
- Breakthrough Breast Cancer Research Unit and the Manchester Breast Centre; Institute of Cancer Sciences; University of Manchester; Manchester, UK; Manchester Centre for Cellular Metabolism (MCCM); University of Manchester; Manchester, UK
| |
Collapse
|
18
|
You H, Lei P, Andreadis ST. JNK is a novel regulator of intercellular adhesion. Tissue Barriers 2013; 1:e26845. [PMID: 24868495 PMCID: PMC3942331 DOI: 10.4161/tisb.26845] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Revised: 10/16/2013] [Accepted: 10/16/2013] [Indexed: 12/11/2022] Open
Abstract
c-Jun N-terminal Kinase (JNK) is a family of protein kinases, which are activated by stress stimuli such as inflammation, heat stress and osmotic stress, and regulate diverse cellular processes including proliferation, survival and apoptosis. In this review, we focus on a recently discovered function of JNK as a regulator of intercellular adhesion. We summarize the existing knowledge regarding the role of JNK during the formation of cell-cell junctions. The potential mechanisms and implications for processes requiring dynamic formation and dissolution of cell-cell junctions including wound healing, migration, cancer metastasis and stem cell differentiation are also discussed.
Collapse
Affiliation(s)
- Hui You
- Bioengineering Laboratory; Department of Chemical and Biological Engineering; University at Buffalo; The State University of New York; Amherst, NY USA
| | - Pedro Lei
- Bioengineering Laboratory; Department of Chemical and Biological Engineering; University at Buffalo; The State University of New York; Amherst, NY USA
| | - Stelios T Andreadis
- Bioengineering Laboratory; Department of Chemical and Biological Engineering; University at Buffalo; The State University of New York; Amherst, NY USA ; Department of Biomedical Engineering; University at Buffalo; The State University of New York; Amherst, NY USA ; Center for Excellence in Bioinformatics and Life Sciences; University at Buffalo; The State University of New York; Amherst, NY USA
| |
Collapse
|
19
|
Mishra J, Verma RK, Alpini G, Meng F, Kumar N. Role of Janus kinase 3 in mucosal differentiation and predisposition to colitis. J Biol Chem 2013; 288:31795-806. [PMID: 24045942 DOI: 10.1074/jbc.m113.504126] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Janus kinase 3 (Jak3) is a nonreceptor tyrosine kinase expressed in both hematopoietic and nonhematopoietic cells. Previously, we characterized the functions of Jak3 in cytoskeletal remodeling, epithelial wound healing, and mucosal homeostasis. However, the role of Jak3 in mucosal differentiation and inflammatory bowel disease was not known. In this report, we characterize the role of Jak3 in mucosal differentiation, basal colonic inflammation, and predisposition toward colitis. Using the Jak3 knock-out (KO) mouse model, we show that Jak3 is expressed in colonic mucosa of mice, and the loss of mucosal expression of Jak3 resulted in reduced expression of differentiation markers for the cells of both enterocytic and secretory lineages. Jak3 KO mice showed reduced expression of colonic villin, carbonic anhydrase, secretory mucin muc2, and increased basal colonic inflammation reflected by increased levels of pro-inflammatory cytokines IL-6 and IL-17A in colon along with increased colonic myeloperoxidase activity. The inflammations in KO mice were associated with shortening of colon length, reduced cecum length, decreased crypt heights, and increased severity toward dextran sulfate sodium-induced colitis. In differentiated human colonic epithelial cells, Jak3 redistributed to basolateral surfaces and interacted with adherens junction (AJ) protein β-catenin. Jak3 expression in these cells was essential for AJ localization of β-catenin and maintenance of epithelial barrier functions. Collectively, these results demonstrate the essential role of Jak3 in the colon where it facilitated mucosal differentiation by promoting the expression of differentiation markers and enhanced colonic barrier functions through AJ localization of β-catenin.
Collapse
Affiliation(s)
- Jayshree Mishra
- From the Department of Pharmaceutical Sciences, College of Pharmacy, Texas A&M University System Health Science Center, Kingsville, Texas 78363 and
| | | | | | | | | |
Collapse
|