1
|
Dong L, Hu S, Li X, Pei S, Jin L, Zhang L, Chen X, Min A, Yin M. SPP1 + TAM Regulates the Metastatic Colonization of CXCR4 + Metastasis-Associated Tumor Cells by Remodeling the Lymph Node Microenvironment. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400524. [PMID: 39236316 PMCID: PMC11600252 DOI: 10.1002/advs.202400524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 05/06/2024] [Indexed: 09/07/2024]
Abstract
Lymph node metastasis, the initial step in distant metastasis, represents a primary contributor to mortality in patients diagnosed with oral squamous cell carcinoma (OSCC). However, the underlying mechanisms of lymph node metastasis in OSCC remain incompletely understood. Here, the transcriptomes of 56 383 single cells derived from paired tissues of six OSCC patients are analyzed. This study founds that CXCR4+ epithelial cells, identified as highly malignant disseminated tumor cells (DTCs), exhibited a propensity for lymph node metastasis. Importantly, a distinct subset of tumor-associated macrophages (TAMs) characterized by exclusive expression of phosphoprotein 1 (SPP1) is discovered. These TAMs may remodel the metastatic lymph node microenvironment by potentially activating fibroblasts and promoting T cell exhaustion through SPP1-CD44 and CD155-CD226 ligand-receptor interactions, thereby facilitating colonization and proliferation of disseminated tumor cells. The research advanced the mechanistic understanding of metastatic tumor microenvironment (TME) and provided a foundation for the development of personalized treatments for OSCC patients with metastasis.
Collapse
Affiliation(s)
- Liang Dong
- Department of DermatologyHunan Engineering Research Center of Skin Health and DiseaseHunan Key Laboratory of Skin Cancer and PsoriasisXiangya HospitalCentral South UniversityChangshaHunan410008China
- Clinical Research Center (CRC)Medical Pathology Center (MPC)Cancer Early Detection and Treatment Center (CEDTC)Chongqing University Three Gorges HospitalChongqing UniversityChongqing404100China
- Translational Medicine Research Center (TMRC)School of Medicine Chongqing UniversityChongqing404100China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Shujun Hu
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008China
- Department of Oral and Maxillofacial SurgeryCenter of StomatologyXiangya HospitalCentral South UniversityChangshaHunan410008China
- Research Center of Oral and Maxillofacail TumorXiangya HospitalCentral South UniversityChangshaHunan410008China
- Insititute of Oral Cancer and Precancerous LesionsCentral South UniversityChangshaHunan410008China
| | - Xin Li
- Clinical Research Center (CRC)Medical Pathology Center (MPC)Cancer Early Detection and Treatment Center (CEDTC)Chongqing University Three Gorges HospitalChongqing UniversityChongqing404100China
- Translational Medicine Research Center (TMRC)School of Medicine Chongqing UniversityChongqing404100China
| | - Shiyao Pei
- Department of DermatologyHunan Engineering Research Center of Skin Health and DiseaseHunan Key Laboratory of Skin Cancer and PsoriasisXiangya HospitalCentral South UniversityChangshaHunan410008China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008China
- Department of DermatologyThird Xiangya HospitalCentral South UniversityChangsha410008China
| | - Liping Jin
- Department of DermatologyHunan Engineering Research Center of Skin Health and DiseaseHunan Key Laboratory of Skin Cancer and PsoriasisXiangya HospitalCentral South UniversityChangshaHunan410008China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Lining Zhang
- Clinical Research Center (CRC)Medical Pathology Center (MPC)Cancer Early Detection and Treatment Center (CEDTC)Chongqing University Three Gorges HospitalChongqing UniversityChongqing404100China
- Translational Medicine Research Center (TMRC)School of Medicine Chongqing UniversityChongqing404100China
| | - Xiang Chen
- Department of DermatologyHunan Engineering Research Center of Skin Health and DiseaseHunan Key Laboratory of Skin Cancer and PsoriasisXiangya HospitalCentral South UniversityChangshaHunan410008China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Anjie Min
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008China
- Department of Oral and Maxillofacial SurgeryCenter of StomatologyXiangya HospitalCentral South UniversityChangshaHunan410008China
- Research Center of Oral and Maxillofacail TumorXiangya HospitalCentral South UniversityChangshaHunan410008China
- Insititute of Oral Cancer and Precancerous LesionsCentral South UniversityChangshaHunan410008China
| | - Mingzhu Yin
- Department of DermatologyHunan Engineering Research Center of Skin Health and DiseaseHunan Key Laboratory of Skin Cancer and PsoriasisXiangya HospitalCentral South UniversityChangshaHunan410008China
- Clinical Research Center (CRC)Medical Pathology Center (MPC)Cancer Early Detection and Treatment Center (CEDTC)Chongqing University Three Gorges HospitalChongqing UniversityChongqing404100China
- Translational Medicine Research Center (TMRC)School of Medicine Chongqing UniversityChongqing404100China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008China
| |
Collapse
|
2
|
Göksu AY, Dirol H, Kocanci FG. Cromolyn sodium and masitinib combination inhibits fibroblast-myofibroblast transition and exerts additive cell-protective and antioxidant effects on a bleomycin-induced in vitro fibrosis model. Pharmacol Res Perspect 2024; 12:e70018. [PMID: 39360479 PMCID: PMC11447456 DOI: 10.1002/prp2.70018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 07/26/2024] [Accepted: 09/06/2024] [Indexed: 10/04/2024] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive and fatal fibrotic lung disease. While recent studies have suggested the potential efficacy of tyrosine kinase inhibitors in managing IPF, masitinib, a clinically used tyrosine kinase inhibitor, has not yet been investigated for its efficacy in fibrotic lung diseases. In a previous study on an in vitro neurodegenerative model, we demonstrated the synergistic antitoxic and antioxidant effects of masitinib combined with cromolyn sodium, an FDA-approved mast cell stabilizer. This study aims to investigate the anti-fibrotic and antioxidant effects of the masitinib-cromolyn sodium combination in an in vitro model of pulmonary fibrosis. Fibroblast cell cultures treated with bleomycin and/or hydrogen peroxide (H2O2) were subjected to masitinib and/or cromolyn sodium, followed by assessments of cell viability, morphological and apoptotic nuclear changes, triple-immunofluorescence labeling, and total oxidant/antioxidant capacities, besides ratio of Bax and Bcl-2 mRNA expressions as an indication of apoptosis. The combined treatment of masitinib and cromolyn sodium effectively prevented the fibroblast myofibroblast transition, a hallmark of fibrosis, and significantly reduced bleomycin / H2O2-induced apoptosis and oxidative stress. This study is the first to demonstrate the additive anti-fibrotic, cell-protective, and antioxidant effects of the masitinib-cromolyn sodium combination in an in vitro fibrosis model, suggesting its potential as an innovative therapeutic approach for pulmonary fibrosis. Combination therapy may be more advantageous in that both drugs could be administered in lower doses, exerting less side effects, and at the same time providing diverse mechanisms of action simultaneously.
Collapse
Affiliation(s)
- Azize Yasemin Göksu
- Department of Histology and EmbryologyAkdeniz University, School of MedicineAntalyaTurkey
- Department of Gene and Cell TherapyAkdeniz University, School of MedicineAntalyaTurkey
| | - Hulya Dirol
- Department of Chest DiseasesAkdeniz University, School of MedicineAntalyaTurkey
| | - Fatma Gonca Kocanci
- Vocational High School of Health Services, Department of Medical Laboratory TechniquesAlanya Alaaddin Keykubat UniversityAlanyaTurkey
| |
Collapse
|
3
|
Walker NM, Ibuki Y, McLinden AP, Misumi K, Mitchell DC, Kleer GG, Lock AM, Vittal R, Sonenberg N, Garner AL, Lama VN. MNK-driven eIF4E phosphorylation regulates the fibrogenic transformation of mesenchymal cells and chronic lung allograft dysfunction. J Clin Invest 2024; 134:e168393. [PMID: 39145446 PMCID: PMC11324311 DOI: 10.1172/jci168393] [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: 01/09/2023] [Accepted: 06/25/2024] [Indexed: 08/16/2024] Open
Abstract
Tissue fibrosis remains unamenable to meaningful therapeutic interventions and is the primary cause of chronic graft failure after organ transplantation. Eukaryotic translation initiation factor (eIF4E), a key translational regulator, serves as convergent target of multiple upstream profibrotic signaling pathways that contribute to mesenchymal cell (MC) activation. Here, we investigate the role of MAP kinase-interacting serine/threonine kinase-induced (MNK-induced) direct phosphorylation of eIF4E at serine 209 (Ser209) in maintaining fibrotic transformation of MCs and determine the contribution of the MNK/eIF4E pathway to the pathogenesis of chronic lung allograft dysfunction (CLAD). MCs from patients with CLAD demonstrated constitutively higher eIF4E phosphorylation at Ser209, and eIF4E phospho-Ser209 was found to be critical in regulating key fibrogenic protein autotaxin, leading to sustained β-catenin activation and profibrotic functions of CLAD MCs. MNK1 signaling was upregulated in CLAD MCs, and genetic or pharmacologic targeting of MNK1 activity inhibited eIF4E phospho-Ser209 and profibrotic functions of CLAD MCs in vitro. Treatment with an MNK1/2 inhibitor (eFT-508) abrogated allograft fibrosis in an orthotopic murine lung-transplant model. Together these studies identify what we believe is a previously unrecognized MNK/eIF4E/ATX/β-catenin signaling pathway of fibrotic transformation of MCs and present the first evidence, to our knowledge, for the utility of MNK inhibitors in fibrosis.
Collapse
Affiliation(s)
- Natalie M. Walker
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Yuta Ibuki
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - A. Patrick McLinden
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Keizo Misumi
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Dylan C. Mitchell
- Department of Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan, USA
| | - Gabriel G. Kleer
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Alison M. Lock
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Ragini Vittal
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Nahum Sonenberg
- Department of Biochemistry and McGill Cancer Center, McGill University, Montreal, Quebec, Canada
| | - Amanda L. Garner
- Department of Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan, USA
| | - Vibha N. Lama
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
4
|
Lian W, Zeng X, Li J, Zang Q, Liu Y, Lv H, Chen S, Huang S, Shen J, Tang L, Xu Y, Wu F, Zhang Q, Xu J. Single-cell sequencing reveals increased LAMB3-positive basal keratinocytes and ZNF90-positive fibroblasts in autologous cultured epithelium. Commun Biol 2024; 7:79. [PMID: 38200141 PMCID: PMC10781733 DOI: 10.1038/s42003-023-05747-5] [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: 01/22/2023] [Accepted: 12/27/2023] [Indexed: 01/12/2024] Open
Abstract
Autologous cultured epithelium grafting (ACEG) presents a promising treatment for refractory vitiligo, yet concerns regarding infections and immunological reactions hinder its surgical use due to serum and feeder dependencies. Addressing this, we culture autologous epithelium under serum- and feeder-free (SFF) conditions, comparing its safety and efficacy with serum- and feeder-dependent (SFD) conditions in stable vitiligo patients, and we discover no significant differences in repigmentation between the SFF and SFD grafts. Single-cell RNA transcriptomics on SFF- and SFD-cultured epithelium alongside healthy skin reveal increased populations of LAMB3+ basal keratinocytes and ZNF90+ fibroblasts in the SFF sheets. Functional analyses showcase active cellular metabolism in LAMB3+ basal keratinocytes, vital in extracellular matrix homeostasis, while ZNF90+ fibroblasts demonstrate increased differentiation, essential in collagen formation for cell adhesion. Importantly, these cell populations in SFF sheets exhibit enhanced interactions with melanocytes compared to SFD sheets. Further, knockdown experiments of LAMB3 in keratinocytes and ZNF90 in fibroblasts lead to a downregulation in melanocyte ligand-receptor-related genes. Overall, SFF sheets demonstrate comparable efficacy to SFD sheets, offering superior safety. LAMB3+ basal keratinocytes and ZNF90+ fibroblasts act as potential drivers behind repigmentation in ACEG under SFF conditions. This study provides translational insights into ACEG repigmentation and potential therapeutic targets for vitiligo.
Collapse
Affiliation(s)
- Weiling Lian
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai Institute of Dermatology, Shanghai, China
| | - Xuanhao Zeng
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai Institute of Dermatology, Shanghai, China
| | - Jian Li
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai Institute of Dermatology, Shanghai, China
| | - Qing Zang
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai Institute of Dermatology, Shanghai, China
| | - Yating Liu
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai Institute of Dermatology, Shanghai, China
| | - Haozhen Lv
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai Institute of Dermatology, Shanghai, China
| | - Shujun Chen
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai Institute of Dermatology, Shanghai, China
| | - Shiyi Huang
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai Institute of Dermatology, Shanghai, China
| | - Jiayi Shen
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai Institute of Dermatology, Shanghai, China
| | - Luyan Tang
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai Institute of Dermatology, Shanghai, China
| | - Yu Xu
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai Institute of Dermatology, Shanghai, China
| | - Fuyue Wu
- ReMed Regenerative Medicine Clinical Application Institute, Shanghai, China
| | - Qi Zhang
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai Institute of Dermatology, Shanghai, China.
| | - Jinhua Xu
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai Institute of Dermatology, Shanghai, China.
| |
Collapse
|
5
|
Chen S, Lastra RO, Paunesku T, Antipova O, Li L, Deng J, Luo Y, Wanzer MB, Popovic J, Li Y, Glasco AD, Jacobsen C, Vogt S, Woloschak GE. Development of Multi-Scale X-ray Fluorescence Tomography for Examination of Nanocomposite-Treated Biological Samples. Cancers (Basel) 2021; 13:cancers13174497. [PMID: 34503306 PMCID: PMC8430782 DOI: 10.3390/cancers13174497] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 08/25/2021] [Accepted: 08/26/2021] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Metal-oxide nanomaterials enter cancer and normal cells even when not specifically targeted, and often interact with specific cellular structures and biological molecules solely due to their innate physical-chemical properties. This raises concerns for the use of nanoparticles, which can be alleviated only with rigorous studies of nanoparticle–cell interactions, studies independent of post-interaction labeling of nanomaterials. X-ray fluorescence microscopy is an imaging technique that quantifies and maps all chemical elements from the periodic table solely based on their native fluorescence excited by the incoming X-ray. We used two different instruments to interrogate the same sample in 3D at two different resolutions and determine heterogeneity of cell-to-cell interactions with nanomaterials, as well as subcellular nanoparticle distribution. This is the first example of multi-scale 3D X-ray fluorescence imaging. This work begins a new era of study on how nanoparticle-based therapies can be developed to be more predictable and safer for use. Abstract Research in cancer nanotechnology is entering its third decade, and the need to study interactions between nanomaterials and cells remains urgent. Heterogeneity of nanoparticle uptake by different cells and subcellular compartments represent the greatest obstacles to a full understanding of the entire spectrum of nanomaterials’ effects. In this work, we used flow cytometry to evaluate changes in cell cycle associated with non-targeted nanocomposite uptake by individual cells and cell populations. Analogous single cell and cell population changes in nanocomposite uptake were explored by X-ray fluorescence microscopy (XFM). Very few nanoparticles are visible by optical imaging without labeling, but labeling increases nanoparticle complexity and the risk of modified cellular uptake. XFM can be used to evaluate heterogeneity of nanocomposite uptake by directly imaging the metal atoms present in the metal-oxide nanocomposites under investigation. While XFM mapping has been performed iteratively in 2D with the same sample at different resolutions, this study is the first example of serial tomographic imaging at two different resolutions. A cluster of cells exposed to non-targeted nanocomposites was imaged with a micron-sized beam in 3D. Next, the sample was sectioned for immunohistochemistry as well as a high resolution “zoomed in” X-ray fluorescence (XRF) tomography with 80 nm beam spot size. Multiscale XRF tomography will revolutionize our ability to explore cell-to-cell differences in nanomaterial uptake.
Collapse
Affiliation(s)
- Si Chen
- X-ray Science Division, Advanced Photon Source, Argonne National Laboratory, Lemont, IL 60439, USA; (S.C.); (O.A.); (L.L.); (J.D.); (Y.L.); (C.J.); (S.V.)
| | - Ruben Omar Lastra
- Department of Radiation Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; (R.O.L.); (T.P.); (M.B.W.); (J.P.); (Y.L.); (A.D.G.)
| | - Tatjana Paunesku
- Department of Radiation Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; (R.O.L.); (T.P.); (M.B.W.); (J.P.); (Y.L.); (A.D.G.)
| | - Olga Antipova
- X-ray Science Division, Advanced Photon Source, Argonne National Laboratory, Lemont, IL 60439, USA; (S.C.); (O.A.); (L.L.); (J.D.); (Y.L.); (C.J.); (S.V.)
| | - Luxi Li
- X-ray Science Division, Advanced Photon Source, Argonne National Laboratory, Lemont, IL 60439, USA; (S.C.); (O.A.); (L.L.); (J.D.); (Y.L.); (C.J.); (S.V.)
| | - Junjing Deng
- X-ray Science Division, Advanced Photon Source, Argonne National Laboratory, Lemont, IL 60439, USA; (S.C.); (O.A.); (L.L.); (J.D.); (Y.L.); (C.J.); (S.V.)
| | - Yanqi Luo
- X-ray Science Division, Advanced Photon Source, Argonne National Laboratory, Lemont, IL 60439, USA; (S.C.); (O.A.); (L.L.); (J.D.); (Y.L.); (C.J.); (S.V.)
| | - Michael Beau Wanzer
- Department of Radiation Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; (R.O.L.); (T.P.); (M.B.W.); (J.P.); (Y.L.); (A.D.G.)
| | - Jelena Popovic
- Department of Radiation Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; (R.O.L.); (T.P.); (M.B.W.); (J.P.); (Y.L.); (A.D.G.)
| | - Ya Li
- Department of Radiation Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; (R.O.L.); (T.P.); (M.B.W.); (J.P.); (Y.L.); (A.D.G.)
| | - Alexander D. Glasco
- Department of Radiation Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; (R.O.L.); (T.P.); (M.B.W.); (J.P.); (Y.L.); (A.D.G.)
| | - Chris Jacobsen
- X-ray Science Division, Advanced Photon Source, Argonne National Laboratory, Lemont, IL 60439, USA; (S.C.); (O.A.); (L.L.); (J.D.); (Y.L.); (C.J.); (S.V.)
- Department of Physics and Astronomy, Weinberg College of Arts and Sciences, Northwestern University, Evanston, IL 60208, USA
| | - Stefan Vogt
- X-ray Science Division, Advanced Photon Source, Argonne National Laboratory, Lemont, IL 60439, USA; (S.C.); (O.A.); (L.L.); (J.D.); (Y.L.); (C.J.); (S.V.)
| | - Gayle E. Woloschak
- Department of Radiation Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; (R.O.L.); (T.P.); (M.B.W.); (J.P.); (Y.L.); (A.D.G.)
- Correspondence: ; Tel.: +1-312-503-4322
| |
Collapse
|
6
|
Wang M, Dai M, Wang D, Xiong W, Zeng Z, Guo C. The regulatory networks of the Hippo signaling pathway in cancer development. J Cancer 2021; 12:6216-6230. [PMID: 34539895 PMCID: PMC8425214 DOI: 10.7150/jca.62402] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 08/15/2021] [Indexed: 01/14/2023] Open
Abstract
The Hippo signaling pathway is a relatively young tumor-related signaling pathway. Although it was discovered lately, research on it developed rapidly. The Hippo signaling pathway is closely relevant to the occurrence and development of tumors and the maintenance of organ size and other biological processes. This manuscript focuses on YAP, the core molecule of the Hippo signaling pathway, and discussion the upstream and downstream regulatory networks of the Hippo signaling pathway during tumorigenesis and development. It also summarizes the relevant drugs involved in this signaling pathway, which may be helpful to the development of targeted drugs for cancer therapy.
Collapse
Affiliation(s)
- Maonan Wang
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Manli Dai
- Hunan Food and Drug Vocational College, Changsha 410036, China
| | - Dan Wang
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Wei Xiong
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Zhaoyang Zeng
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Can Guo
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| |
Collapse
|
7
|
Deciphering the role of cartilage protein 1 in human dermal fibroblasts: a transcriptomic approach. Funct Integr Genomics 2021; 21:503-511. [PMID: 34269961 DOI: 10.1007/s10142-021-00792-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 05/23/2021] [Accepted: 06/01/2021] [Indexed: 10/20/2022]
Abstract
Cartilage acidic protein 1A (hCRTAC1-A) is an extracellular matrix protein (ECM) of human hard and soft tissue that is associated with matrix disorders. The central role of fibroblasts in tissue integrity and ECM health made primary human dermal fibroblasts (NHDF) the model for the present study, which aimed to provide new insight into the molecular function of hCRTAC1-A. Specifically, we explored the differential expression patterns of specific genes associated with the presence of hCRTAC1-A by RNA-seq and RT-qPCR analysis. Functional enrichment analysis demonstrated, for the very first time, that hCRTAC1-A is involved in extracellular matrix organization and development, through its regulatory effect on asporin, decorin, and complement activity, in cell proliferation, regeneration, wound healing, and collagen degradation. This work provides a better understanding of putative hCRTAC1-A actions in human fibroblasts and a fundamental insight into its function in tissue biology.
Collapse
|
8
|
Blokland K, Pouwels S, Schuliga M, Knight D, Burgess J. Regulation of cellular senescence by extracellular matrix during chronic fibrotic diseases. Clin Sci (Lond) 2020; 134:2681-2706. [PMID: 33084883 PMCID: PMC7578566 DOI: 10.1042/cs20190893] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 10/05/2020] [Accepted: 10/06/2020] [Indexed: 02/07/2023]
Abstract
The extracellular matrix (ECM) is a complex network of macromolecules surrounding cells providing structural support and stability to tissues. The understanding of the ECM and the diverse roles it plays in development, homoeostasis and injury have greatly advanced in the last three decades. The ECM is crucial for maintaining tissue homoeostasis but also many pathological conditions arise from aberrant matrix remodelling during ageing. Ageing is characterised as functional decline of tissue over time ultimately leading to tissue dysfunction, and is a risk factor in many diseases including cardiovascular disease, diabetes, cancer, dementia, glaucoma, chronic obstructive pulmonary disease (COPD) and fibrosis. ECM changes are recognised as a major driver of aberrant cell responses. Mesenchymal cells in aged tissue show signs of growth arrest and resistance to apoptosis, which are indicative of cellular senescence. It was recently postulated that cellular senescence contributes to the pathogenesis of chronic fibrotic diseases in the heart, kidney, liver and lung. Senescent cells negatively impact tissue regeneration while creating a pro-inflammatory environment as part of the senescence-associated secretory phenotype (SASP) favouring disease progression. In this review, we explore and summarise the current knowledge around how aberrant ECM potentially influences the senescent phenotype in chronic fibrotic diseases. Lastly, we will explore the possibility for interventions in the ECM-senescence regulatory pathways for therapeutic potential in chronic fibrotic diseases.
Collapse
Affiliation(s)
- Kaj E.C. Blokland
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD, Groningen, The Netherlands
- University of Newcastle, School of Biomedical Sciences and Pharmacy, Callaghan, NSW, Australia
- National Health and Medical Research Council Centre of Research Excellence in Pulmonary Fibrosis, Sydney, NSW, Australia
| | - Simon D. Pouwels
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD, Groningen, The Netherlands
- Department of Lung Diseases, University Medical Center Groningen, Groningen, The Netherlands
| | - Michael Schuliga
- University of Newcastle, School of Biomedical Sciences and Pharmacy, Callaghan, NSW, Australia
| | - Darryl A. Knight
- University of Newcastle, School of Biomedical Sciences and Pharmacy, Callaghan, NSW, Australia
- National Health and Medical Research Council Centre of Research Excellence in Pulmonary Fibrosis, Sydney, NSW, Australia
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Providence Health Care Research Institute, Vancouver, BC, Canada
| | - Janette K. Burgess
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD, Groningen, The Netherlands
| |
Collapse
|
9
|
Huth S, Huth L, Marquardt Y, Fietkau K, Dahl E, Esser PR, Martin SF, Heise R, Merk HF, Baron JM. Inter-α-Trypsin Inhibitor Heavy Chain 5 (ITIH5) Is a Natural Stabilizer of Hyaluronan That Modulates Biological Processes in the Skin. Skin Pharmacol Physiol 2020; 33:198-206. [PMID: 32799206 DOI: 10.1159/000509371] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 06/11/2020] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Hyaluronan (HA) is a major component of the skin that exerts a variety of biological functions. Inter-α-trypsin inhibitor heavy chain (ITIH) proteins comprise a family of hyaladherins of which ITIH5 has recently been described in skin, where it plays a functional role in skin morphology and inflammatory skin diseases including allergic contact dermatitis (ACD). OBJECTIVE The current study focused on the ITIH5-HA interaction and its potential clinical and functional impact in extracellular matrix (ECM) stabilization. METHODS Studying the molecular effects of ITIH5 in skin, we established skin models comprising murine skin cells of Itih5 knockout mice and corresponding wild-type controls. In addition, human dermal fibroblasts with an ITIH5 knockdown as well as a murine recombinant Itih5 protein were established to examine the interaction between ITIH5 and HA using in vitro adhesion and HA degradation assays. To understand more precisely the role of ITIH5 in inflammatory skin diseases such as ACD, we generated ITIH5 knockout cells of the KeratinoSens® cell line. RESULTS Using murine skin models, ITIH5 knockdown fibroblasts, and a reactive oxygen species (ROS)-mediated HA degradation assay, we proved that ITIH5 binds to HA, thereby acting as a stabilizer of HA. Moreover, microarray profiling revealed the impact of ITIH5 on biological processes such as skin development and ECM homeostasis. Performing the in vitro KeratinoSens skin sensitization assay, we detected that ITIH5 decreases the sensitizing potential of moderate and strong contact sensitizers. CONCLUSION Taken together, our experiments revealed that ITIH5 forms complexes with HA, thereby on the one hand stabilizing HA and facilitating the formation of ECM structures and on the other hand modulating inflammatory responses.
Collapse
Affiliation(s)
- Sebastian Huth
- Department of Dermatology and Allergology, Medical Faculty, RWTH Aachen University, Aachen, Germany,
| | - Laura Huth
- Department of Dermatology and Allergology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Yvonne Marquardt
- Department of Dermatology and Allergology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Katharina Fietkau
- Department of Dermatology and Allergology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Edgar Dahl
- Molecular Oncology Group, Institute of Pathology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Philipp R Esser
- Allergy Research Group, Department of Dermatology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Stefan F Martin
- Allergy Research Group, Department of Dermatology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Ruth Heise
- Department of Dermatology and Allergology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Hans F Merk
- Department of Dermatology and Allergology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Jens Malte Baron
- Department of Dermatology and Allergology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
10
|
Ooki T, Hatakeyama M. Hyaluronan Degradation Promotes Cancer via Hippo-YAP Signaling: An Intervention Point for Cancer Therapy. Bioessays 2020; 42:e2000005. [PMID: 32449813 DOI: 10.1002/bies.202000005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 03/16/2020] [Indexed: 12/14/2022]
Abstract
High-molecular-weight hyaluronan acts as a ligand of the tumor-suppressive Hippo signal, whereas degradation of hyaluronan from a high-molecular-weight form to a low-molecular-weight forms by hyaluronidase 2 inhibits Hippo signal activation and thereby activates the pro-oncogenic transcriptional coactivator yes-associated protein (YAP), which creates a cancer-predisposing microenvironment and drives neoplastic transformation of cells through both cell-autonomous and non-cell-autonomous mechanisms. In fact, accumulation of low-molecular-weight hyaluronan in tissue stroma is observed in many types of cancers. Since inhibition of YAP activity suppresses tumor growth in vivo, pharmacological intervention of the Hippo-YAP signal is an attractive approach for future drug development. In this review, pharmacological intervention of excessive hyaluronan degradation as a novel approach for inhibition of the Hippo-YAP signal is also discussed. Development of hyaluronidase inhibitors may provide novel therapeutic strategies for human malignant tumors.
Collapse
Affiliation(s)
- Takuya Ooki
- Division of Microbiology, Graduate School of Medicine, the University of Tokyo, Tokyo, 113-0033, Japan
| | - Masanori Hatakeyama
- Division of Microbiology, Graduate School of Medicine, the University of Tokyo, Tokyo, 113-0033, Japan
| |
Collapse
|
11
|
Jiménez-Meléndez A, Fernández-Álvarez M, Calle A, Ramírez MÁ, Diezma-Díaz C, Vázquez-Arbaizar P, Ortega-Mora LM, Álvarez-García G. Lytic cycle of Besnoitia besnoiti tachyzoites displays similar features in primary bovine endothelial cells and fibroblasts. Parasit Vectors 2019; 12:517. [PMID: 31685001 PMCID: PMC6829937 DOI: 10.1186/s13071-019-3777-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 10/29/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Bovine besnoitiosis, caused by the cyst-forming apicomplexan parasite Besnoitia besnoiti, is a chronic and debilitating cattle disease that continues to spread in Europe in the absence of control tools. In this scenario, in vitro culture systems are valuable tools to carry out drug screenings and to unravel host-parasite interactions. However, studies performed in bovine target cells are scarce. METHODS The objective of the present study was to obtain primary bovine aortic endothelial cells (BAECs) and fibroblast cell cultures, target cells during the acute and the chronic stage of the disease, respectively, from healthy bovine donors. Afterwards, expression of surface (CD31, CD34 and CD44) and intracellular markers (vimentin and cytokeratin) was studied to characterize cell populations by flow cytometry. Next, the lytic cycle of B. besnoiti tachyzoites was studied in both target cells. Invasion rates (IRs) were determined by immunofluorescence at several time points post-infection, and proliferation kinetics were studied by quantitative PCR (qPCR). Finally, the influence of bovine viral diarrhea virus (BVDV) co-infection on the host cell machinery, and consequently on B. besnoiti invasion and proliferation, was investigated in BAECs. RESULTS Morphology and cytometry results confirmed the endothelial and fibroblast origins. CD31 was the surface marker that best discriminated between BAECs and fibroblasts, since fibroblasts lacked CD31 labelling. Expression of CD34 was weak in low-passage BAECs and absent in high-passage BAECs and fibroblasts. Positive labelling for CD44, vimentin and cytokeratin was observed in both BAECs and fibroblasts. Regarding the lytic cycle of the parasite, although low invasion rates (approximately 3-4%) were found in both cell culture systems, more invasion was observed in BAECs at 24 and 72 hpi. The proliferation kinetics did not differ between BAECs and fibroblasts. BVDV infection favoured early Besnoitia invasion but there was no difference in tachyzoite yields observed in BVDV-BAECs compared to BAECs. CONCLUSIONS We have generated and characterized two novel standardized in vitro models for Besnoitia besnoiti infection based on bovine primary target BAECs and fibroblasts, and have shown the relevance of BVDV coinfections, which should be considered in further studies with other cattle pathogens.
Collapse
Affiliation(s)
- Alejandro Jiménez-Meléndez
- SALUVET, Animal Health Department, Faculty of Veterinary Sciences, Complutense University of Madrid, Ciudad Universitaria s/n, 28040 Madrid, Spain
| | - María Fernández-Álvarez
- SALUVET, Animal Health Department, Faculty of Veterinary Sciences, Complutense University of Madrid, Ciudad Universitaria s/n, 28040 Madrid, Spain
| | - Alexandra Calle
- Departamento de Reproducción Animal, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Avenida Puerta de Hierro 12, local 10, 28040 Madrid, Spain
| | - Miguel Ángel Ramírez
- Departamento de Reproducción Animal, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Avenida Puerta de Hierro 12, local 10, 28040 Madrid, Spain
| | - Carlos Diezma-Díaz
- SALUVET, Animal Health Department, Faculty of Veterinary Sciences, Complutense University of Madrid, Ciudad Universitaria s/n, 28040 Madrid, Spain
| | - Patricia Vázquez-Arbaizar
- SALUVET, Animal Health Department, Faculty of Veterinary Sciences, Complutense University of Madrid, Ciudad Universitaria s/n, 28040 Madrid, Spain
| | - Luis Miguel Ortega-Mora
- SALUVET, Animal Health Department, Faculty of Veterinary Sciences, Complutense University of Madrid, Ciudad Universitaria s/n, 28040 Madrid, Spain
| | - Gema Álvarez-García
- SALUVET, Animal Health Department, Faculty of Veterinary Sciences, Complutense University of Madrid, Ciudad Universitaria s/n, 28040 Madrid, Spain
| |
Collapse
|
12
|
Swaminathan S, Cranston AN, Clyne AM. A Three-Dimensional In Vitro Coculture Model to Quantify Breast Epithelial Cell Adhesion to Endothelial Cells. Tissue Eng Part C Methods 2019; 25:609-618. [PMID: 31441384 PMCID: PMC7718851 DOI: 10.1089/ten.tec.2019.0122] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 08/20/2019] [Indexed: 12/12/2022] Open
Abstract
Three-dimensional (3D) in vitro culture models better recapitulate the tissue microenvironment, and therefore may provide a better platform to evaluate therapeutic effects on adhesive cell-cell interactions. The objective of this study was to determine if AD-01, a peptide derivative of FK506-binding protein like that is reported to bind to the adhesion receptor CD44, would induce a greater reduction in breast epithelial spheroid adhesion to endothelial tube-like networks in our 3D coculture model system compared to two-dimensional (2D) culture. MCF10A, MCF10A-NeuN, MDA-MB-231, and MCF7 breast epithelial cells were pretreated with AD-01 either as single cells or as spheroids. Breast epithelial cell adhesion to 2D tissue culture substrates was first measured, followed by spheroid formation (breast cell-cell adhesion) and spheroid adhesion to Matrigel or endothelial networks. Finally, CD44 expression was quantified in breast epithelial cells in 2D and 3D culture. Our results show that AD-01 had the largest effect on spheroid formation, specifically in breast cancer cell lines. AD-01 also inhibited breast cancer spheroid adhesion to and migration along endothelial networks. The different breast epithelial cell lines expressed more CD44 when cultured as 3D spheroids, but this did not universally translate into higher protein levels. This study shows that 3D coculture models can enable unique insights into cell adhesion, migration, and cell-cell interactions, thereby enhancing understanding of basic biological mechanisms. Furthermore, such 3D coculture systems may also represent a more relevant testing platform for understanding the mechanism-of-action of new therapeutic agents. Impact Statement Cell adhesion is inherently different in two dimensional (2D) compared to three dimensional (3D) culture; yet, most adhesion assays in academia and industry are still conducted in 2D because few simple, yet effective, adhesion models exist in 3D. Recently we developed a 3D in vitro coculture model to examine breast epithelial spheroid interactions with endothelial tubes. We now show that this 3D coculture model can effectively be used to interrogate and quantify drug-induced differences in breast epithelial cell adhesion that are unique to 3D cocultures. This 3D coculture adhesion model can furthermore be modified for use with other cell types to better predict drug effects on cell-vasculature adhesion.
Collapse
Affiliation(s)
- Swathi Swaminathan
- Mechanical Engineering and Mechanics, Drexel University, Philadelphia, Pennsylvania
| | - Aaron N. Cranston
- Centre for Precision Therapeutics, Health Sciences Building, Almac Discovery Ltd., Belfast, United Kingdom
| | - Alisa Morss Clyne
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland
| |
Collapse
|
13
|
High-Molecular-Weight Hyaluronan Is a Hippo Pathway Ligand Directing Cell Density-Dependent Growth Inhibition via PAR1b. Dev Cell 2019; 49:590-604.e9. [DOI: 10.1016/j.devcel.2019.04.018] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 01/11/2019] [Accepted: 04/13/2019] [Indexed: 02/06/2023]
|
14
|
Wang J, Wu X, Zheng Y, Wen H, Ji H, Zhao Y, Guan W. Isolation and biological characterization of mesenchymal stem cells from goose dermis. Poult Sci 2018; 97:3236-3247. [PMID: 29790972 DOI: 10.3382/ps/pey178] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2018] [Accepted: 05/09/2018] [Indexed: 11/20/2022] Open
Abstract
The skin is a natural target of stem cell research because of its large size and easy accessibility. Cutaneous mesenchymal stem cells have shown to be a promising source of various adult stem cell or progenitor cell populations, which provide an important source of stem cell-based investigation. Nowadays, much work has been done on dermal-derived mesenchymal stem cells (DMSCs) from humans, mice, sheep, and other mammals, but the literature on avian species has been rarely reported. As an animal model, the goose is an endemic species abounding in dermal tissues which is important in the global economy. In this study, we isolated and established the mesenchymal stem cell line from dermis tissue of goose, which were subcultured to passage 21 in vitro without loss of their functional integrity in terms of morphology, renewal capacity, and presence of mesenchymal stem cell markers. Cryopreservation and resuscitation were also observed in different passages. To investigate the biological characteristics of goose DMSCs, immunofluorescence, reverse transcription-polymerase chain reaction, and flow cytometry were used to detect the characteristic surface markers. Growth curves and the capacity of colony forming were performed to test the self-renew and proliferative ability. Furthermore, the DMSCs are induced to osteoblasts, adipocytes, and chondrocytes in vitro. Our results suggest that DMSCs isolated from goose embryos possess similar biological characteristics to those from other species. The methods in establishment and cultivation of goose DMSCs line demonstrated a good self-renew and expansion potential in vitro, which provided a technological platform for preserving the valuable genetic resources of poultry and a great inspiration for in vitro investigation of avian MSCs.
Collapse
Affiliation(s)
- Jingjing Wang
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China.,School of kinesiology and health, Harbin Institute of Physical Education, Harbin, Heilongjiang province 150008, China
| | - Xulun Wu
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Yanjie Zheng
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China.,School of Life Sciences, Jiamusi University, Jiamusi, Heilongjiang province 154007, China
| | - Hebao Wen
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China.,School of sports science, Mudanjiang Normal University, Mudanjiang, Heilongjiang province 157011, China
| | - Hongda Ji
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China.,School of kinesiology and health, Harbin Institute of Physical Education, Harbin, Heilongjiang province 150008, China
| | - Yuhua Zhao
- School of kinesiology and health, Harbin Institute of Physical Education, Harbin, Heilongjiang province 150008, China
| | - Weijun Guan
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| |
Collapse
|
15
|
The role of CD44, hyaluronan and NHE1 in cardiac remodeling. Life Sci 2018; 209:197-201. [PMID: 30089233 DOI: 10.1016/j.lfs.2018.08.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 07/16/2018] [Accepted: 08/04/2018] [Indexed: 12/12/2022]
Abstract
Cardiac remodeling, characterized by excessive extracellular matrix (ECM) remodeling, predisposes the heart to failure if left unresolved. Understanding the signaling mechanisms involved in excessive extracellular matrix (ECM) remodeling is necessary to identify the means to regress the development of cardiac remodeling and heart failure. Recently, hyaluronan (HA), a ubiquitously expressed glycosaminoglycan in the ECM, was shown to participate in tissue fibrosis and myofibroblast proliferation through interacting with its ubiquitously expressed cell-surface receptor, CD44. CD44 is a multifunctional transmembrane glycoprotein that serves as a cell-surface receptor for a number of ECM proteins. The mechanism by which the interaction between CD44-HA contributes to ECM and cardiac remodeling remains unknown. A previous study performed on a non-cardiac model showed that CD44-HA enhances Na+/H+ exchanger isoform-1 (NHE1) activity, causing ECM remodeling, HA metabolism and tumor invasion. Interestingly, NHE1 has been demonstrated to be involved in cardiac remodeling and myocardial fibrosis. In addition, it has previously been demonstrated that CD44 is upregulated in transgenic mouse hearts expressing active NHE-1. The role of CD44, HA and NHE1 and the cellular interplay of these factors in the ECM and cardiac remodeling is the focus of this review.
Collapse
|
16
|
Fan Z, Xia H, Xu H, Ma J, Zhou S, Hou W, Tang Q, Gong Q, Nie Y, Bi F. Standard CD44 modulates YAP1 through a positive feedback loop in hepatocellular carcinoma. Biomed Pharmacother 2018; 103:147-156. [PMID: 29649630 DOI: 10.1016/j.biopha.2018.03.042] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 03/09/2018] [Accepted: 03/09/2018] [Indexed: 02/07/2023] Open
Abstract
High expression levels of CD44 and YAP have been identified as poor prognostic factors in hepatocellular carcinoma (HCC). However, the mechanistic relationship between CD44 and YAP during HCC tumorigenesis remains largely unknown. To investigate the mutual regulation between standard CD44 (CD44S) and YAP1 in HCC cell lines and tissue samples, CD44S and YAP1 expression in 40 pairs of tumor samples and matched distal normal tissues from HCC patients was examined by immunohistochemical staining. High expression of either CD44S or YAP1 was associated with a younger age and worse pathology grade. In addition, high levels of CD44S and YAP1 were associated with increased vascular invasion and more severe liver cirrhosis, respectively. CD44S expression was positively correlated with YAP1 expression in these HCC tissues. In vitro experiments suggested that CD44S could positively regulate the expression of YAP1 and its target genes via the PI3K/Akt pathway in HCC cells. Moreover, CD44S is regulated by the YAP1/TEAD axis. These results reveal a novel positive feedback loop involving CD44S and YAP1, in which CD44S functions as both an upstream regulator and a downstream effector of YAP1 in HCC. This feedback loop might constitute a broadly conserved module for regulating cell proliferation and invasion during HCC tumorigenesis. Blocking this positive feedback loop that involves CD44S and YAP1 might represent a new approach for HCC treatment.
Collapse
Affiliation(s)
- Zhenhai Fan
- Department of Medical Oncology and Laboratory of Molecular Targeted Therapy in Oncology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, PR China; Key Laboratory of Cell Engineering of Guizhou, The Affiliated Hospital of Zunyi Medical College, Zunyi, Guizhou, 573003, PR China
| | - Hongwei Xia
- Department of Medical Oncology and Laboratory of Molecular Targeted Therapy in Oncology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, PR China
| | - Huanji Xu
- Department of Medical Oncology and Laboratory of Molecular Targeted Therapy in Oncology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, PR China
| | - Ji Ma
- Department of Medical Oncology and Laboratory of Molecular Targeted Therapy in Oncology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, PR China; Department of Breast Surgery, Lanzhou General Hospital of PLA, Lanzhou, Gansu, 730000, PR China
| | - Sheng Zhou
- Department of Medical Oncology and Laboratory of Molecular Targeted Therapy in Oncology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, PR China
| | - Wanting Hou
- Department of Medical Oncology and Laboratory of Molecular Targeted Therapy in Oncology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, PR China
| | - Qiulin Tang
- Department of Medical Oncology and Laboratory of Molecular Targeted Therapy in Oncology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, PR China
| | - Qiyong Gong
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, PR China
| | - Yongzhan Nie
- State Key Laboratory of Cancer Biology & Xijing Hospital of Digest Diseases, Fourth Military Medical University, Xi'an, Shanxi, 710032, PR China
| | - Feng Bi
- Department of Medical Oncology and Laboratory of Molecular Targeted Therapy in Oncology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, PR China.
| |
Collapse
|
17
|
Ruiter MS, Pesce M. Mechanotransduction in Coronary Vein Graft Disease. Front Cardiovasc Med 2018; 5:20. [PMID: 29594150 PMCID: PMC5861212 DOI: 10.3389/fcvm.2018.00020] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 02/22/2018] [Indexed: 12/19/2022] Open
Abstract
Autologous saphenous veins are the most commonly used conduits in revascularization of the ischemic heart by coronary artery bypass graft surgery, but are subject to vein graft failure. The current mini review aims to provide an overview of the role of mechanotransduction signalling underlying vein graft failure to further our understanding of the disease progression and to improve future clinical treatment. Firstly, limitation of damage during vein harvest and engraftment can improve outcome. In addition, cell cycle inhibition, stimulation of Nur77 and external grafting could form interesting therapeutic options. Moreover, the Hippo pathway, with the YAP/TAZ complex as the main effector, is emerging as an important node controlling conversion of mechanical signals into cellular responses. This includes endothelial cell inflammation, smooth muscle cell proliferation/migration, and monocyte attachment/infiltration. The combined effects of expression levels and nuclear/cytoplasmic translocation make YAP/TAZ interesting novel targets in the prevention and treatment of vein graft disease. Pharmacological, molecular and/or mechanical conditioning of saphenous vein segments between harvest and grafting may potentiate targeted and specific treatment to improve long-term outcome.
Collapse
Affiliation(s)
- Matthijs Steven Ruiter
- Cardiovascular Tissue Engineering Unit, Centro Cardiologico Monzino (IRCCS), Milan, Italy
| | - Maurizio Pesce
- Cardiovascular Tissue Engineering Unit, Centro Cardiologico Monzino (IRCCS), Milan, Italy
| |
Collapse
|
18
|
Stiffness-dependent motility and proliferation uncoupled by deletion of CD44. Sci Rep 2017; 7:16499. [PMID: 29184125 PMCID: PMC5705666 DOI: 10.1038/s41598-017-16486-z] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 11/14/2017] [Indexed: 01/16/2023] Open
Abstract
Information in the microenvironment guides complex cellular decisions such as whether or not to proliferate and migrate. The effects of soluble extracellular signals on these cellular functions are fairly well understood, but relatively little is known about how the extracellular matrix (ECM), and particularly the mechanical information in the ECM, guides these cellular decisions. Here, we show that CD44, a major receptor for the glycosaminoglycan ECM component hyaluronan, coordinates the motility and proliferative responses to ECM stiffening. We analyzed these cellular responses on fibronectin-coated polyacrylamide hydrogels prepared at a physiologic range of ECM stiffness and found that stiffening of the ECM leads to both cell cycling and cell motility in serum-stimulated primary mouse dermal fibroblasts. Remarkably, deletion of CD44 impaired stiffness-stimulated motility of the primary cells without affecting other hallmark cellular responses to ECM stiffening including cell spread area, stress fiber formation, focal adhesion maturation, and intracellular stiffening. Even stiffness-mediated cell proliferation was unaffected by deletion of CD44. Our results reveal a novel effect of CD44, which is imposed downstream of ECM-mechanosensing and determines if cells couple or uncouple their proliferative and motility responses to ECM stiffness.
Collapse
|
19
|
Kim Y, West GA, Ray G, Kessler SP, Petrey AC, Fiocchi C, McDonald C, Longworth MS, Nagy LE, de la Motte CA. Layilin is critical for mediating hyaluronan 35kDa-induced intestinal epithelial tight junction protein ZO-1 in vitro and in vivo. Matrix Biol 2017; 66:93-109. [PMID: 28978412 DOI: 10.1016/j.matbio.2017.09.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 09/14/2017] [Accepted: 09/16/2017] [Indexed: 02/06/2023]
Abstract
Tight junction proteins are critical in maintaining homeostatic intestinal permeability. Multiple intestinal inflammatory diseases are correlated with reduced expression of tight junction proteins. We have recently reported that oral treatment of mice with Hyaluronan 35kDa (HA35) increases colonic expression of tight junction protein zonula occludens-1 (ZO-1). Here, we investigate whether HA35 treatment enhances ZO-1 expression by direct interaction with intestinal epithelium in vitro and have identified the HA receptor responsible for HA35-mediated ZO-1 induction in colonic epithelium in vitro and in vivo. Our results reveal that HA35 treatment increases ZO-1 expression in mouse intestinal epithelial organoids, while large HA 2000kDa is not internalized into the cells. Our immunofluorescence data indicate that layilin, but neither toll-like receptor-4 (TLR-4) nor CD44, mediate the HA35-induced ZO-1 expression in colonic epithelium in vitro and in vivo. Additionally, using layilin null mice we have determined that layilin mediates HA35 induction of ZO-1 in healthy mice and during dextran sulfate sodium (DSS)-induced colitis. Furthermore, we find that while ZO-1 expression levels are reduced, layilin expression levels are equivalent in inflammatory bowel disease (IBD) patients and non-IBD controls. Together, our data suggest that layilin is an important HA receptor, that mediates the effect of oral HA35 treatment on intestinal epithelium. HA35 holds promise as a simple dietary supplement to strengthen gut barrier defense.
Collapse
Affiliation(s)
- Yeojung Kim
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Gail A West
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Greeshma Ray
- Department of Molecular Genetics, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Sean P Kessler
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Aaron C Petrey
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Claudio Fiocchi
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Christine McDonald
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Michelle S Longworth
- Department of Molecular Genetics, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Laura E Nagy
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Carol A de la Motte
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA.
| |
Collapse
|
20
|
Tsuneki M, Kinjo T, Mori T, Yoshida A, Kuyama K, Ohira A, Miyagi T, Takahashi K, Kawai A, Chuman H, Yamazaki N, Masuzawa M, Arakawa H. Survivin: A novel marker and potential therapeutic target for human angiosarcoma. Cancer Sci 2017; 108:2295-2305. [PMID: 28845553 PMCID: PMC5665764 DOI: 10.1111/cas.13379] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2017] [Revised: 08/10/2017] [Accepted: 08/14/2017] [Indexed: 12/20/2022] Open
Abstract
Human angiosarcoma is a rare malignant vascular tumor associated with extremely poor clinical outcome and generally arising in skin of the head and neck region. However, little is known about the molecular pathogeneses and useful immunohistochemical markers of angiosarcoma. To investigate the mechanisms of angiosarcoma progression, we collected 85 cases of human angiosarcoma specimens with clinical records and analyzed ISO-HAS-B patient-derived angiosarcoma cells. As control subjects, 54 cases of hemangioma and 34 of pyogenic granuloma were collected. Remarkably, consistent with our recent observations regarding the involvement of survivin expression following Hippo pathway inactivation in the neoplastic proliferation of murine hemangioendothelioma cells and human infantile hemangioma, nuclear survivin expression was observed in all cases of angiosarcoma but not in hemangiomas and pyogenic granulomas, and the Hippo pathway was inactivated in 90.3% of yes-associated protein (YAP) -positive angiosarcoma cases. However, survivin expression modes and YAP localization (Hippo pathway activation modes) were not correlated with survival. In addition, we confirmed that survivin small interference RNA (siRNA) transfection and YM155, an anti-survivin drug, elicited decreased nuclear survivin expression and cell proliferation in ISO-HAS-B cells which expressed survivin consistently. Conclusively, these findings support the importance of survivin as a good marker and critical regulator of cellular proliferation for human angiosarcoma and YM155 as a potential therapeutic agent.
Collapse
Affiliation(s)
- Masayuki Tsuneki
- Division of Cancer Biology, National Cancer Center Research Institute, Tokyo, Japan.,Division of Pathology, Department of Oral Diagnostic Sciences, School of Dentistry, Showa University, Tokyo, Japan
| | - Takao Kinjo
- Division of Morphological Pathology, Department of Basic Laboratory Sciences, School of Health Sciences, Faculty of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Taisuke Mori
- Departments of Pathology and Clinical Laboratories, National Cancer Center Hospital, Tokyo, Japan
| | - Akihiko Yoshida
- Departments of Pathology and Clinical Laboratories, National Cancer Center Hospital, Tokyo, Japan
| | - Kayo Kuyama
- Department of Oral Pathology, Nihon University School of Dentistry at Matsudo, Chiba, Japan
| | - Aoi Ohira
- Deparment of Dermatology, University of the Ryukyus, Okinawa, Japan
| | - Takuya Miyagi
- Deparment of Dermatology, University of the Ryukyus, Okinawa, Japan
| | - Kenzo Takahashi
- Deparment of Dermatology, University of the Ryukyus, Okinawa, Japan
| | - Akira Kawai
- Musculoskeletal Oncology and Rehabilitation, National Cancer Center Hospital, Tokyo, Japan
| | - Hirokazu Chuman
- Musculoskeletal Oncology and Rehabilitation, National Cancer Center Hospital, Tokyo, Japan
| | - Naoya Yamazaki
- Dermatologic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Mikio Masuzawa
- Department of Molecular Diagnostics, School of Allied Health Sciences, Kitasato University, Kanagawa, Japan
| | - Hirofumi Arakawa
- Division of Cancer Biology, National Cancer Center Research Institute, Tokyo, Japan
| |
Collapse
|
21
|
Synthetic peptide TEKKRRETVEREKE derived from ezrin induces differentiation of NIH/3T3 fibroblasts. Eur J Pharmacol 2017; 811:249-259. [DOI: 10.1016/j.ejphar.2017.06.033] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 06/22/2017] [Accepted: 06/26/2017] [Indexed: 01/02/2023]
|
22
|
Kuwahara G, Hashimoto T, Tsuneki M, Yamamoto K, Assi R, Foster TR, Hanisch JJ, Bai H, Hu H, Protack CD, Hall MR, Schardt JS, Jay SM, Madri JA, Kodama S, Dardik A. CD44 Promotes Inflammation and Extracellular Matrix Production During Arteriovenous Fistula Maturation. Arterioscler Thromb Vasc Biol 2017; 37:1147-1156. [PMID: 28450292 DOI: 10.1161/atvbaha.117.309385] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2016] [Accepted: 04/07/2017] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Arteriovenous fistulae (AVF) remain the optimal conduit for hemodialysis access but continue to demonstrate poor patency and poor rates of maturation. We hypothesized that CD44, a widely expressed cellular adhesion molecule that serves as a major receptor for extracellular matrix components, promotes wall thickening and extracellular matrix deposition during AVF maturation. APPROACH AND RESULTS AVF were created via needle puncture in wild-type C57BL/6J and CD44 knockout mice. CD44 mRNA and protein expression was increased in wild-type AVF. CD44 knockout mice showed no increase in AVF wall thickness (8.9 versus 26.8 μm; P=0.0114), collagen density, and hyaluronic acid density, but similar elastin density when compared with control AVF. CD44 knockout mice also showed no increase in vascular cell adhesion molecule-1 expression, intercellular adhesion molecule-1 expression, and monocyte chemoattractant protein-1 expression in the AVF compared with controls; there were also no increased M2 macrophage markers (transglutaminase-2: 81.5-fold, P=0.0015; interleukin-10: 7.6-fold, P=0.0450) in CD44 knockout mice. Delivery of monocyte chemoattractant protein-1 to CD44 knockout mice rescued the phenotype with thicker AVF walls (27.2 versus 14.7 μm; P=0.0306), increased collagen density (2.4-fold; P=0.0432), and increased number of M2 macrophages (2.1-fold; P=0.0335). CONCLUSIONS CD44 promotes accumulation of M2 macrophages, extracellular matrix deposition, and wall thickening during AVF maturation. These data show the association of M2 macrophages with wall thickening during AVF maturation and suggest that enhancing CD44 activity may be a strategy to increase AVF maturation.
Collapse
Affiliation(s)
- Go Kuwahara
- From the Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT (G.K., T.H., K.Y., R.A., T.R.F., J.J.H., H.B., H.H., C.D.P., M.R.H., J.A.M., A.D.); Department of Cardiovascular Surgery (G.K.) and Department of Regenerative Medicine and Transplantation (G.K., S.K.), Fukuoka University, Japan; Department of Surgery, Veterans Affairs Connecticut Healthcare Systems, West Haven (T.H., K.Y., H.B., H.H., A.D.); Division of Vascular Surgery, Department of Surgery, The University of Tokyo, Japan (T.H., K.Y.); Division of Pathology, Department of Oral Diagnostic Sciences, School of Dentistry, Showa University, Tokyo, Japan (M.T.); Department of Pathology (M.T., J.A.M.) and Department of Surgery (R.A., T.R.F., J.J.H., C.D.P., M.R.H., A.D.), Yale University School of Medicine, New Haven, CT; and Fischell Department of Bioengineering, University of Maryland, College Park (J.S.S., S.M.J.)
| | - Takuya Hashimoto
- From the Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT (G.K., T.H., K.Y., R.A., T.R.F., J.J.H., H.B., H.H., C.D.P., M.R.H., J.A.M., A.D.); Department of Cardiovascular Surgery (G.K.) and Department of Regenerative Medicine and Transplantation (G.K., S.K.), Fukuoka University, Japan; Department of Surgery, Veterans Affairs Connecticut Healthcare Systems, West Haven (T.H., K.Y., H.B., H.H., A.D.); Division of Vascular Surgery, Department of Surgery, The University of Tokyo, Japan (T.H., K.Y.); Division of Pathology, Department of Oral Diagnostic Sciences, School of Dentistry, Showa University, Tokyo, Japan (M.T.); Department of Pathology (M.T., J.A.M.) and Department of Surgery (R.A., T.R.F., J.J.H., C.D.P., M.R.H., A.D.), Yale University School of Medicine, New Haven, CT; and Fischell Department of Bioengineering, University of Maryland, College Park (J.S.S., S.M.J.)
| | - Masayuki Tsuneki
- From the Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT (G.K., T.H., K.Y., R.A., T.R.F., J.J.H., H.B., H.H., C.D.P., M.R.H., J.A.M., A.D.); Department of Cardiovascular Surgery (G.K.) and Department of Regenerative Medicine and Transplantation (G.K., S.K.), Fukuoka University, Japan; Department of Surgery, Veterans Affairs Connecticut Healthcare Systems, West Haven (T.H., K.Y., H.B., H.H., A.D.); Division of Vascular Surgery, Department of Surgery, The University of Tokyo, Japan (T.H., K.Y.); Division of Pathology, Department of Oral Diagnostic Sciences, School of Dentistry, Showa University, Tokyo, Japan (M.T.); Department of Pathology (M.T., J.A.M.) and Department of Surgery (R.A., T.R.F., J.J.H., C.D.P., M.R.H., A.D.), Yale University School of Medicine, New Haven, CT; and Fischell Department of Bioengineering, University of Maryland, College Park (J.S.S., S.M.J.)
| | - Kota Yamamoto
- From the Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT (G.K., T.H., K.Y., R.A., T.R.F., J.J.H., H.B., H.H., C.D.P., M.R.H., J.A.M., A.D.); Department of Cardiovascular Surgery (G.K.) and Department of Regenerative Medicine and Transplantation (G.K., S.K.), Fukuoka University, Japan; Department of Surgery, Veterans Affairs Connecticut Healthcare Systems, West Haven (T.H., K.Y., H.B., H.H., A.D.); Division of Vascular Surgery, Department of Surgery, The University of Tokyo, Japan (T.H., K.Y.); Division of Pathology, Department of Oral Diagnostic Sciences, School of Dentistry, Showa University, Tokyo, Japan (M.T.); Department of Pathology (M.T., J.A.M.) and Department of Surgery (R.A., T.R.F., J.J.H., C.D.P., M.R.H., A.D.), Yale University School of Medicine, New Haven, CT; and Fischell Department of Bioengineering, University of Maryland, College Park (J.S.S., S.M.J.)
| | - Roland Assi
- From the Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT (G.K., T.H., K.Y., R.A., T.R.F., J.J.H., H.B., H.H., C.D.P., M.R.H., J.A.M., A.D.); Department of Cardiovascular Surgery (G.K.) and Department of Regenerative Medicine and Transplantation (G.K., S.K.), Fukuoka University, Japan; Department of Surgery, Veterans Affairs Connecticut Healthcare Systems, West Haven (T.H., K.Y., H.B., H.H., A.D.); Division of Vascular Surgery, Department of Surgery, The University of Tokyo, Japan (T.H., K.Y.); Division of Pathology, Department of Oral Diagnostic Sciences, School of Dentistry, Showa University, Tokyo, Japan (M.T.); Department of Pathology (M.T., J.A.M.) and Department of Surgery (R.A., T.R.F., J.J.H., C.D.P., M.R.H., A.D.), Yale University School of Medicine, New Haven, CT; and Fischell Department of Bioengineering, University of Maryland, College Park (J.S.S., S.M.J.)
| | - Trenton R Foster
- From the Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT (G.K., T.H., K.Y., R.A., T.R.F., J.J.H., H.B., H.H., C.D.P., M.R.H., J.A.M., A.D.); Department of Cardiovascular Surgery (G.K.) and Department of Regenerative Medicine and Transplantation (G.K., S.K.), Fukuoka University, Japan; Department of Surgery, Veterans Affairs Connecticut Healthcare Systems, West Haven (T.H., K.Y., H.B., H.H., A.D.); Division of Vascular Surgery, Department of Surgery, The University of Tokyo, Japan (T.H., K.Y.); Division of Pathology, Department of Oral Diagnostic Sciences, School of Dentistry, Showa University, Tokyo, Japan (M.T.); Department of Pathology (M.T., J.A.M.) and Department of Surgery (R.A., T.R.F., J.J.H., C.D.P., M.R.H., A.D.), Yale University School of Medicine, New Haven, CT; and Fischell Department of Bioengineering, University of Maryland, College Park (J.S.S., S.M.J.)
| | - Jesse J Hanisch
- From the Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT (G.K., T.H., K.Y., R.A., T.R.F., J.J.H., H.B., H.H., C.D.P., M.R.H., J.A.M., A.D.); Department of Cardiovascular Surgery (G.K.) and Department of Regenerative Medicine and Transplantation (G.K., S.K.), Fukuoka University, Japan; Department of Surgery, Veterans Affairs Connecticut Healthcare Systems, West Haven (T.H., K.Y., H.B., H.H., A.D.); Division of Vascular Surgery, Department of Surgery, The University of Tokyo, Japan (T.H., K.Y.); Division of Pathology, Department of Oral Diagnostic Sciences, School of Dentistry, Showa University, Tokyo, Japan (M.T.); Department of Pathology (M.T., J.A.M.) and Department of Surgery (R.A., T.R.F., J.J.H., C.D.P., M.R.H., A.D.), Yale University School of Medicine, New Haven, CT; and Fischell Department of Bioengineering, University of Maryland, College Park (J.S.S., S.M.J.)
| | - Hualong Bai
- From the Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT (G.K., T.H., K.Y., R.A., T.R.F., J.J.H., H.B., H.H., C.D.P., M.R.H., J.A.M., A.D.); Department of Cardiovascular Surgery (G.K.) and Department of Regenerative Medicine and Transplantation (G.K., S.K.), Fukuoka University, Japan; Department of Surgery, Veterans Affairs Connecticut Healthcare Systems, West Haven (T.H., K.Y., H.B., H.H., A.D.); Division of Vascular Surgery, Department of Surgery, The University of Tokyo, Japan (T.H., K.Y.); Division of Pathology, Department of Oral Diagnostic Sciences, School of Dentistry, Showa University, Tokyo, Japan (M.T.); Department of Pathology (M.T., J.A.M.) and Department of Surgery (R.A., T.R.F., J.J.H., C.D.P., M.R.H., A.D.), Yale University School of Medicine, New Haven, CT; and Fischell Department of Bioengineering, University of Maryland, College Park (J.S.S., S.M.J.)
| | - Haidi Hu
- From the Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT (G.K., T.H., K.Y., R.A., T.R.F., J.J.H., H.B., H.H., C.D.P., M.R.H., J.A.M., A.D.); Department of Cardiovascular Surgery (G.K.) and Department of Regenerative Medicine and Transplantation (G.K., S.K.), Fukuoka University, Japan; Department of Surgery, Veterans Affairs Connecticut Healthcare Systems, West Haven (T.H., K.Y., H.B., H.H., A.D.); Division of Vascular Surgery, Department of Surgery, The University of Tokyo, Japan (T.H., K.Y.); Division of Pathology, Department of Oral Diagnostic Sciences, School of Dentistry, Showa University, Tokyo, Japan (M.T.); Department of Pathology (M.T., J.A.M.) and Department of Surgery (R.A., T.R.F., J.J.H., C.D.P., M.R.H., A.D.), Yale University School of Medicine, New Haven, CT; and Fischell Department of Bioengineering, University of Maryland, College Park (J.S.S., S.M.J.)
| | - Clinton D Protack
- From the Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT (G.K., T.H., K.Y., R.A., T.R.F., J.J.H., H.B., H.H., C.D.P., M.R.H., J.A.M., A.D.); Department of Cardiovascular Surgery (G.K.) and Department of Regenerative Medicine and Transplantation (G.K., S.K.), Fukuoka University, Japan; Department of Surgery, Veterans Affairs Connecticut Healthcare Systems, West Haven (T.H., K.Y., H.B., H.H., A.D.); Division of Vascular Surgery, Department of Surgery, The University of Tokyo, Japan (T.H., K.Y.); Division of Pathology, Department of Oral Diagnostic Sciences, School of Dentistry, Showa University, Tokyo, Japan (M.T.); Department of Pathology (M.T., J.A.M.) and Department of Surgery (R.A., T.R.F., J.J.H., C.D.P., M.R.H., A.D.), Yale University School of Medicine, New Haven, CT; and Fischell Department of Bioengineering, University of Maryland, College Park (J.S.S., S.M.J.)
| | - Michael R Hall
- From the Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT (G.K., T.H., K.Y., R.A., T.R.F., J.J.H., H.B., H.H., C.D.P., M.R.H., J.A.M., A.D.); Department of Cardiovascular Surgery (G.K.) and Department of Regenerative Medicine and Transplantation (G.K., S.K.), Fukuoka University, Japan; Department of Surgery, Veterans Affairs Connecticut Healthcare Systems, West Haven (T.H., K.Y., H.B., H.H., A.D.); Division of Vascular Surgery, Department of Surgery, The University of Tokyo, Japan (T.H., K.Y.); Division of Pathology, Department of Oral Diagnostic Sciences, School of Dentistry, Showa University, Tokyo, Japan (M.T.); Department of Pathology (M.T., J.A.M.) and Department of Surgery (R.A., T.R.F., J.J.H., C.D.P., M.R.H., A.D.), Yale University School of Medicine, New Haven, CT; and Fischell Department of Bioengineering, University of Maryland, College Park (J.S.S., S.M.J.)
| | - John S Schardt
- From the Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT (G.K., T.H., K.Y., R.A., T.R.F., J.J.H., H.B., H.H., C.D.P., M.R.H., J.A.M., A.D.); Department of Cardiovascular Surgery (G.K.) and Department of Regenerative Medicine and Transplantation (G.K., S.K.), Fukuoka University, Japan; Department of Surgery, Veterans Affairs Connecticut Healthcare Systems, West Haven (T.H., K.Y., H.B., H.H., A.D.); Division of Vascular Surgery, Department of Surgery, The University of Tokyo, Japan (T.H., K.Y.); Division of Pathology, Department of Oral Diagnostic Sciences, School of Dentistry, Showa University, Tokyo, Japan (M.T.); Department of Pathology (M.T., J.A.M.) and Department of Surgery (R.A., T.R.F., J.J.H., C.D.P., M.R.H., A.D.), Yale University School of Medicine, New Haven, CT; and Fischell Department of Bioengineering, University of Maryland, College Park (J.S.S., S.M.J.)
| | - Steven M Jay
- From the Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT (G.K., T.H., K.Y., R.A., T.R.F., J.J.H., H.B., H.H., C.D.P., M.R.H., J.A.M., A.D.); Department of Cardiovascular Surgery (G.K.) and Department of Regenerative Medicine and Transplantation (G.K., S.K.), Fukuoka University, Japan; Department of Surgery, Veterans Affairs Connecticut Healthcare Systems, West Haven (T.H., K.Y., H.B., H.H., A.D.); Division of Vascular Surgery, Department of Surgery, The University of Tokyo, Japan (T.H., K.Y.); Division of Pathology, Department of Oral Diagnostic Sciences, School of Dentistry, Showa University, Tokyo, Japan (M.T.); Department of Pathology (M.T., J.A.M.) and Department of Surgery (R.A., T.R.F., J.J.H., C.D.P., M.R.H., A.D.), Yale University School of Medicine, New Haven, CT; and Fischell Department of Bioengineering, University of Maryland, College Park (J.S.S., S.M.J.)
| | - Joseph A Madri
- From the Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT (G.K., T.H., K.Y., R.A., T.R.F., J.J.H., H.B., H.H., C.D.P., M.R.H., J.A.M., A.D.); Department of Cardiovascular Surgery (G.K.) and Department of Regenerative Medicine and Transplantation (G.K., S.K.), Fukuoka University, Japan; Department of Surgery, Veterans Affairs Connecticut Healthcare Systems, West Haven (T.H., K.Y., H.B., H.H., A.D.); Division of Vascular Surgery, Department of Surgery, The University of Tokyo, Japan (T.H., K.Y.); Division of Pathology, Department of Oral Diagnostic Sciences, School of Dentistry, Showa University, Tokyo, Japan (M.T.); Department of Pathology (M.T., J.A.M.) and Department of Surgery (R.A., T.R.F., J.J.H., C.D.P., M.R.H., A.D.), Yale University School of Medicine, New Haven, CT; and Fischell Department of Bioengineering, University of Maryland, College Park (J.S.S., S.M.J.)
| | - Shohta Kodama
- From the Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT (G.K., T.H., K.Y., R.A., T.R.F., J.J.H., H.B., H.H., C.D.P., M.R.H., J.A.M., A.D.); Department of Cardiovascular Surgery (G.K.) and Department of Regenerative Medicine and Transplantation (G.K., S.K.), Fukuoka University, Japan; Department of Surgery, Veterans Affairs Connecticut Healthcare Systems, West Haven (T.H., K.Y., H.B., H.H., A.D.); Division of Vascular Surgery, Department of Surgery, The University of Tokyo, Japan (T.H., K.Y.); Division of Pathology, Department of Oral Diagnostic Sciences, School of Dentistry, Showa University, Tokyo, Japan (M.T.); Department of Pathology (M.T., J.A.M.) and Department of Surgery (R.A., T.R.F., J.J.H., C.D.P., M.R.H., A.D.), Yale University School of Medicine, New Haven, CT; and Fischell Department of Bioengineering, University of Maryland, College Park (J.S.S., S.M.J.)
| | - Alan Dardik
- From the Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT (G.K., T.H., K.Y., R.A., T.R.F., J.J.H., H.B., H.H., C.D.P., M.R.H., J.A.M., A.D.); Department of Cardiovascular Surgery (G.K.) and Department of Regenerative Medicine and Transplantation (G.K., S.K.), Fukuoka University, Japan; Department of Surgery, Veterans Affairs Connecticut Healthcare Systems, West Haven (T.H., K.Y., H.B., H.H., A.D.); Division of Vascular Surgery, Department of Surgery, The University of Tokyo, Japan (T.H., K.Y.); Division of Pathology, Department of Oral Diagnostic Sciences, School of Dentistry, Showa University, Tokyo, Japan (M.T.); Department of Pathology (M.T., J.A.M.) and Department of Surgery (R.A., T.R.F., J.J.H., C.D.P., M.R.H., A.D.), Yale University School of Medicine, New Haven, CT; and Fischell Department of Bioengineering, University of Maryland, College Park (J.S.S., S.M.J.).
| |
Collapse
|
23
|
Andl T, Zhou L, Yang K, Kadekaro AL, Zhang Y. YAP and WWTR1: New targets for skin cancer treatment. Cancer Lett 2017; 396:30-41. [PMID: 28279717 DOI: 10.1016/j.canlet.2017.03.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Revised: 01/11/2017] [Accepted: 03/01/2017] [Indexed: 12/26/2022]
Abstract
The core components of the Hippo signaling pathway are a cascade of kinases that govern the phosphorylation of downstream transcriptional co-activators, namely, YES-associated protein (YAP) and WW domain-containing transcription regulator protein 1 (WWTR1, also known as TAZ). The Hippo signaling pathway is considered an important tumor-suppressor pathway, and its dysregulation has been noted in a variety of human cancers, in which YAP/WWTR1 enable cancerous cells to overcome contact inhibition, and to grow and spread uncontrollably. Interestingly, however, recent studies have told a somewhat different but perhaps more intriguing YAP/WWTR1 story, as these studies found that YAP/WWTR1 function as a central hub that integrates signals from multiple upstream signaling pathways, cell-cell interactions and mechanical forces and then bind to and activate different downstream transcriptional factors to direct cell social behavior and cell-cell interactions. In this review, we present the latest findings on the role of YAP/WWTR1 in skin physiology, pathology and tumorigenesis and discuss the statuses of newly developed therapeutic interventions that target YAP/WWTR1 in human cancers, as well as their prospects for use as skin cancer treatments.
Collapse
Affiliation(s)
- Thomas Andl
- Burnett School of Biological Sciences, University of Central Florida, Orlando, FL 32816, USA
| | - Linli Zhou
- Division of Pharmaceutical Sciences, College of Pharmacy, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Kun Yang
- Division of Pharmaceutical Sciences, College of Pharmacy, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Ana Luisa Kadekaro
- Department of Dermatology, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Yuhang Zhang
- Division of Pharmaceutical Sciences, College of Pharmacy, University of Cincinnati, Cincinnati, OH 45267, USA.
| |
Collapse
|