1
|
Midekssa FS, Davidson CD, Wieger ME, Kamen JL, Hanna KM, Jayco DKP, Hu MM, Friend NE, Putnam AJ, Helms AS, Shikanov A, Baker BM. Semi-synthetic fibrous fibrin composites promote 3D microvascular assembly, survival, and host integration of endothelial cells without mesenchymal cell support. Bioact Mater 2025; 49:652-669. [PMID: 40235652 PMCID: PMC11999628 DOI: 10.1016/j.bioactmat.2025.02.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 01/14/2025] [Accepted: 02/19/2025] [Indexed: 04/17/2025] Open
Abstract
Vasculogenic assembly of 3D capillary networks remains a promising approach to vascularizing tissue-engineered grafts, a significant outstanding challenge in tissue engineering and regenerative medicine. Current approaches for vasculogenic assembly rely on the inclusion of supporting mesenchymal cells alongside endothelial cells, co-encapsulated within vasculo-conducive materials such as low-density fibrin hydrogels. Here, we established a material-based approach to circumvent the need for supporting mesenchymal cells and report that the inclusion of synthetic matrix fibers in dense (>3 mg mL-1) 3D fibrin hydrogels can enhance vasculogenic assembly in endothelial cell monocultures. Surprisingly, we found that the addition of non-cell-adhesive synthetic matrix fibers compared to cell-adhesive synthetic fibers best encouraged vasculogenic assembly, proliferation, lumenogenesis, a vasculogenic transcriptional program, and additionally promoted cell-matrix interactions and intercellular force transmission. Implanting fiber-reinforced prevascularized constructs to assess graft-host vascular integration, we demonstrate additive effects of enhanced vascular network assembly during in vitro pre-culture, fiber-mediated improvements in endothelial cell survival and vascular maintenance post-implantation, and enhanced host cell infiltration that collectively enabled graft vessel integration with host circulation. This work establishes synthetic matrix fibers as an inexpensive alternative to sourcing and expanding secondary supporting cell types for the prevascularization of tissue constructs.
Collapse
Affiliation(s)
- Firaol S. Midekssa
- Department of Biomedical Engineering, University of Michigan Ann Arbor, MI, 48109, United States
| | - Christopher D. Davidson
- Department of Biomedical Engineering, University of Michigan Ann Arbor, MI, 48109, United States
| | - Megan E. Wieger
- Department of Biomedical Engineering, University of Michigan Ann Arbor, MI, 48109, United States
| | - Jordan L. Kamen
- Department of Biomedical Engineering, University of Michigan Ann Arbor, MI, 48109, United States
| | - Kaylin M. Hanna
- Department of Biomedical Engineering, University of Michigan Ann Arbor, MI, 48109, United States
| | - Danica Kristen P. Jayco
- Department of Biomedical Engineering, University of Michigan Ann Arbor, MI, 48109, United States
| | - Michael M. Hu
- Department of Biomedical Engineering, University of Michigan Ann Arbor, MI, 48109, United States
| | - Nicole E. Friend
- Department of Biomedical Engineering, University of Michigan Ann Arbor, MI, 48109, United States
| | - Andrew J. Putnam
- Department of Biomedical Engineering, University of Michigan Ann Arbor, MI, 48109, United States
| | - Adam S. Helms
- Division of Cardiovascular Medicine, University of Michigan Ann Arbor, MI 48109, United States
| | - Ariella Shikanov
- Department of Biomedical Engineering, University of Michigan Ann Arbor, MI, 48109, United States
- Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, MI, 48109, United States
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, 48109, United States
| | - Brendon M. Baker
- Department of Biomedical Engineering, University of Michigan Ann Arbor, MI, 48109, United States
| |
Collapse
|
2
|
Mulla SA, Patil A, Mali S, Jain AK, Jaiswal H, Sawant HR, Arvind R, Singh S. Unleashing the therapeutic role of cannabidiol in dentistry. J Oral Biol Craniofac Res 2024; 14:649-654. [PMID: 39296277 PMCID: PMC11409039 DOI: 10.1016/j.jobcr.2024.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Accepted: 09/02/2024] [Indexed: 09/21/2024] Open
Abstract
Cannabidiol (CBD) found in Cannabis sativa is a non-psychoactive compound which is capable of binding to CB1 and CB2 receptors. CBD has recently gained interest in dentistry although it has not been explored sufficiently yet. The therapeutic effects of CBD include anti-inflammatory, analgesic, antioxidant, biological and osteoinductive properties. The aim of this review is to highlight these effects with respect to various oral conditions and shed light on the current limitations and prospects for the use of CBD in maintaining oral health.
Collapse
Affiliation(s)
- Sayem Anwarhussain Mulla
- Department of Dentistry, Bharati Vidyapeeth (Deemed to be University), Dental College and Hospital, Navi Mumbai, Maharashtra, India, 400614
| | - Amit Patil
- Department of Conservative Dentistry and Endodontics, Bharati Vidyapeeth (Deemed to be University), Dental College and Hospital, Navi Mumbai, Maharashtra, India, 400614
| | - Sheetal Mali
- Department of Conservative Dentistry and Endodontics, Bharati Vidyapeeth (Deemed to be University), Dental College and Hospital, Navi Mumbai, Maharashtra, India, 400614
| | - Ashish K Jain
- Department of Conservative Dentistry and Endodontics, Bharati Vidyapeeth (Deemed to be University), Dental College and Hospital, Navi Mumbai, Maharashtra, India, 400614
| | - Himmat Jaiswal
- Department of Conservative Dentistry and Endodontics, Bharati Vidyapeeth (Deemed to be University), Dental College and Hospital, Navi Mumbai, Maharashtra, India, 400614
| | - Hitesh Ramdas Sawant
- Department of Orthodontics and Dentofacial Orthopaedics, Bharati Vidyapeeth (Deemed to be University), Dental College and Hospital, Navi Mumbai, Maharashtra, India, 400614
| | - Ritvi Arvind
- Department of Conservative Dentistry and Endodontics, Bharati Vidyapeeth (Deemed to be University), Dental College and Hospital, Navi Mumbai, Maharashtra, India, 400614
| | - Shruti Singh
- Department of Dentistry, Bharati Vidyapeeth (Deemed to be University), Dental College and Hospital, Navi Mumbai, Maharashtra, India, 400614
| |
Collapse
|
3
|
Tarchi SM, Pernia Marin M, Hossain MM, Salvatore M. Breast stiffness, a risk factor for cancer and the role of radiology for diagnosis. J Transl Med 2023; 21:582. [PMID: 37649088 PMCID: PMC10466778 DOI: 10.1186/s12967-023-04457-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 08/19/2023] [Indexed: 09/01/2023] Open
Abstract
Over the last five decades, breast density has been associated with increased risk of developing breast cancer. Mammographically dense breasts are considered those belonging to the heterogeneously dense breasts, and extremely dense breasts subgroups according to the American College of Radiology's Breast Imaging Reporting and Data System (BI-RADS). There is a statistically significant correlation between the increased mammographic density and the presence of more glandular tissue alone. However, the strength of this correlation is weak. Although the mechanisms driving breast density-related tumor initiation and progression are still unknown, there is evidence suggesting that certain molecular pathways participating in epithelial-stromal interactions may play a pivotal role in the deposition of fibrillar collagen, increased matrix stiffness, and cell migration that favor breast density and carcinogenesis. This article describes these molecular mechanisms as potential "landscapers" for breast density-related cancer. We also introduce the term "Breast Compactness" to reflect collagen density of breast tissue on chest CT scan and the use of breast stiffness measurements as imaging biomarkers for breast cancer screening and risk stratification.
Collapse
Affiliation(s)
- Sofia M Tarchi
- Department of Radiology, Columbia University Irving Medical Center, New York, NY, USA
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20072, Milan, Italy
| | - Monica Pernia Marin
- Department of Radiology, Columbia University Irving Medical Center, New York, NY, USA.
| | - Md Murad Hossain
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Mary Salvatore
- Department of Radiology, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
4
|
Bellocchio L, Patano A, Inchingolo AD, Inchingolo F, Dipalma G, Isacco CG, de Ruvo E, Rapone B, Mancini A, Lorusso F, Scarano A, Malcangi G, Inchingolo AM. Cannabidiol for Oral Health: A New Promising Therapeutical Tool in Dentistry. Int J Mol Sci 2023; 24:ijms24119693. [PMID: 37298644 DOI: 10.3390/ijms24119693] [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: 05/03/2023] [Revised: 05/30/2023] [Accepted: 06/01/2023] [Indexed: 06/12/2023] Open
Abstract
The medical use of cannabis has a very long history. Although many substances called cannabinoids are present in cannabis, Δ9tetrahydrocannabinol (Δ9-THC), cannabidiol (CBD) and cannabinol (CBN) are the three main cannabinoids that are most present and described. CBD itself is not responsible for the psychotropic effects of cannabis, since it does not produce the typical behavioral effects associated with the consumption of this drug. CBD has recently gained growing attention in modern society and seems to be increasingly explored in dentistry. Several subjective findings suggest some therapeutic effects of CBD that are strongly supported by research evidence. However, there is a plethora of data regarding CBD's mechanism of action and therapeutic potential, which are in many cases contradictory. We will first provide an overview of the scientific evidence on the molecular mechanism of CBD's action. Furthermore, we will map the recent developments regarding the possible oral benefits of CBD. In summary, we will highlight CBD's promising biological features for its application in dentistry, despite exiting patents that suggest the current compositions for oral care as the main interest of the industry.
Collapse
Affiliation(s)
- Luigi Bellocchio
- INSERM, U1215 NeuroCentre Magendie, Endocannabinoids and Neuroadaptation, University of Bordeaux, 33063 Bordeaux, France
| | - Assunta Patano
- Department of Interdisciplinary Medicine, University of Study "Aldo Moro", 70124 Bari, Italy
| | | | - Francesco Inchingolo
- Department of Interdisciplinary Medicine, University of Study "Aldo Moro", 70124 Bari, Italy
| | - Gianna Dipalma
- Department of Interdisciplinary Medicine, University of Study "Aldo Moro", 70124 Bari, Italy
| | - Ciro Gargiulo Isacco
- Department of Interdisciplinary Medicine, University of Study "Aldo Moro", 70124 Bari, Italy
| | - Elisabetta de Ruvo
- Department of Interdisciplinary Medicine, University of Study "Aldo Moro", 70124 Bari, Italy
| | - Biagio Rapone
- Department of Interdisciplinary Medicine, University of Study "Aldo Moro", 70124 Bari, Italy
| | - Antonio Mancini
- Department of Interdisciplinary Medicine, University of Study "Aldo Moro", 70124 Bari, Italy
| | - Felice Lorusso
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, 66100 Chieti, Italy
| | - Antonio Scarano
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, 66100 Chieti, Italy
| | - Giuseppina Malcangi
- Department of Interdisciplinary Medicine, University of Study "Aldo Moro", 70124 Bari, Italy
| | | |
Collapse
|
5
|
Berry CE, Downer M, Morgan AG, Griffin M, Liang NE, Kameni L, Laufey Parker JB, Guo J, Longaker MT, Wan DC. The effects of mechanical force on fibroblast behavior in cutaneous injury. Front Surg 2023; 10:1167067. [PMID: 37143767 PMCID: PMC10151708 DOI: 10.3389/fsurg.2023.1167067] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 03/29/2023] [Indexed: 05/06/2023] Open
Abstract
Wound healing results in the formation of scar tissue which can be associated with functional impairment, psychological stress, and significant socioeconomic cost which exceeds 20 billion dollars annually in the United States alone. Pathologic scarring is often associated with exaggerated action of fibroblasts and subsequent excessive accumulation of extracellular matrix proteins which results in fibrotic thickening of the dermis. In skin wounds, fibroblasts transition to myofibroblasts which contract the wound and contribute to remodeling of the extracellular matrix. Mechanical stress on wounds has long been clinically observed to result in increased pathologic scar formation, and studies over the past decade have begun to uncover the cellular mechanisms that underly this phenomenon. In this article, we will review the investigations which have identified proteins involved in mechano-sensing, such as focal adhesion kinase, as well as other important pathway components that relay the transcriptional effects of mechanical forces, such as RhoA/ROCK, the hippo pathway, YAP/TAZ, and Piezo1. Additionally, we will discuss findings in animal models which show the inhibition of these pathways to promote wound healing, reduce contracture, mitigate scar formation, and restore normal extracellular matrix architecture. Recent advances in single cell RNA sequencing and spatial transcriptomics and the resulting ability to further characterize mechanoresponsive fibroblast subpopulations and the genes that define them will be summarized. Given the importance of mechanical signaling in scar formation, several clinical treatments focused on reducing tension on the wound have been developed and are described here. Finally, we will look toward future research which may reveal novel cellular pathways and deepen our understanding of the pathogenesis of pathologic scarring. The past decade of scientific inquiry has drawn many lines connecting these cellular mechanisms that may lead to a map for the development of transitional treatments for patients on the path to scarless healing.
Collapse
Affiliation(s)
- Charlotte E. Berry
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Mauricio Downer
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Annah G. Morgan
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Michelle Griffin
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Norah E. Liang
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Lionel Kameni
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Jennifer B. Laufey Parker
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Jason Guo
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Michael T. Longaker
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, United States
| | - Derrick C. Wan
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States
- Correspondence: Derrick C. Wan
| |
Collapse
|
6
|
Ghuloum FI, Johnson CA, Riobo-Del Galdo NA, Amer MH. From mesenchymal niches to engineered in vitro model systems: Exploring and exploiting biomechanical regulation of vertebrate hedgehog signalling. Mater Today Bio 2022; 17:100502. [PMID: 36457847 PMCID: PMC9707069 DOI: 10.1016/j.mtbio.2022.100502] [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: 09/23/2022] [Revised: 11/08/2022] [Accepted: 11/20/2022] [Indexed: 11/23/2022] Open
Abstract
Tissue patterning is the result of complex interactions between transcriptional programs and various mechanical cues that modulate cell behaviour and drive morphogenesis. Vertebrate Hedgehog signalling plays key roles in embryogenesis and adult tissue homeostasis, and is central to skeletal development and the osteogenic differentiation of mesenchymal stem cells. The expression of several components of the Hedgehog signalling pathway have been reported to be mechanically regulated in mesodermal tissue patterning and osteogenic differentiation in response to external stimulation. Since a number of bone developmental defects and skeletal diseases, such as osteoporosis, are directly linked to aberrant Hedgehog signalling, a better knowledge of the regulation of Hedgehog signalling in the mechanosensitive bone marrow-residing mesenchymal stromal cells will present novel avenues for modelling these diseases and uncover novel opportunities for extracellular matrix-targeted therapies. In this review, we present a brief overview of the key molecular players involved in Hedgehog signalling and the basic concepts of mechanobiology, with a focus on bone development and regeneration. We also highlight the correlation between the activation of the Hedgehog signalling pathway in response to mechanical cues and osteogenesis in bone marrow-derived mesenchymal stromal cells. Finally, we propose different tissue engineering strategies to apply the expanding knowledge of 3D material-cell interactions in the modulation of Hedgehog signalling in vitro for fundamental and translational research applications.
Collapse
Affiliation(s)
- Fatmah I. Ghuloum
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
- Department of Biological Sciences, Faculty of Science, Kuwait University, Kuwait City, Kuwait
| | - Colin A. Johnson
- Leeds Institute of Medical Research, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Natalia A. Riobo-Del Galdo
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
- Leeds Institute of Medical Research, Faculty of Medicine and Health, University of Leeds, Leeds, UK
- Astbury Centre for Structural Molecular Biology, University of Leeds, UK
| | - Mahetab H. Amer
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
7
|
Du R, Li D, Huang Y, Xiao H, Xue J, Ji J, Feng Y, Fan Y. Effect of mechanical stretching and substrate stiffness on the morphology, cytoskeleton and nuclear shape of corneal endothelial cells. MEDICINE IN NOVEL TECHNOLOGY AND DEVICES 2022. [DOI: 10.1016/j.medntd.2022.100180] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
8
|
Mechanical Stretch Induced Skin Regeneration: Molecular and Cellular Mechanism in Skin Soft Tissue Expansion. Int J Mol Sci 2022; 23:ijms23179622. [PMID: 36077018 PMCID: PMC9455829 DOI: 10.3390/ijms23179622] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 08/16/2022] [Accepted: 08/23/2022] [Indexed: 11/17/2022] Open
Abstract
Skin soft tissue expansion is one of the most basic and commonly used techniques in plastic surgery to obtain excess skin for a variety of medical uses. However, skin soft tissue expansion is faced with many problems, such as long treatment process, poor skin quality, high retraction rate, and complications. Therefore, a deeper understanding of the mechanisms of skin soft tissue expansion is needed. The key to skin soft tissue expansion lies in the mechanical stretch applied to the skin by an inflatable expander. Mechanical stimulation activates multiple signaling pathways through cellular adhesion molecules and regulates gene expression profiles in cells. Meanwhile, various types of cells contribute to skin expansion, including keratinocytes, dermal fibroblasts, and mesenchymal stem cells, which are also regulated by mechanical stretch. This article reviews the molecular and cellular mechanisms of skin regeneration induced by mechanical stretch during skin soft tissue expansion.
Collapse
|
9
|
The Distraction Osteogenesis Callus: a Review of the Literature. Clin Rev Bone Miner Metab 2022. [DOI: 10.1007/s12018-021-09282-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
10
|
Dobrokhotov O, Sunagawa M, Torii T, Mii S, Kawauchi K, Enomoto A, Sokabe M, Hirata H. Anti-Malignant Effect of Tensile Loading to Adherens Junctions in Cutaneous Squamous Cell Carcinoma Cells. Front Cell Dev Biol 2021; 9:728383. [PMID: 34858971 PMCID: PMC8632149 DOI: 10.3389/fcell.2021.728383] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 10/19/2021] [Indexed: 11/13/2022] Open
Abstract
Actomyosin contractility regulates various cellular processes including proliferation and differentiation while dysregulation of actomyosin activity contributes to cancer development and progression. Previously, we have reported that actomyosin-generated tension at adherens junctions is required for cell density-dependent inhibition of proliferation of normal skin keratinocytes. However, it remains unclear how actomyosin contractility affects the hyperproliferation ability of cutaneous squamous cell carcinoma (cSCC) cells. In this study, we find that actomyosin activity is impaired in cSCC cells both in vitro and in vivo. External application of tensile loads to adherens junctions by sustained mechanical stretch attenuates the proliferation of cSCC cells, which depends on intact adherens junctions. Forced activation of actomyosin of cSCC cells also inhibits their proliferation in a cell-cell contact-dependent manner. Furthermore, the cell cycle arrest induced by tensile loading to adherens junctions is accompanied by epidermal differentiation in cSCC cells. Our results show that the degree of malignant properties of cSCC cells can be reduced by applying tensile loads to adherens junctions, which implies that the mechanical status of adherens junctions may serve as a novel therapeutic target for cSCC.
Collapse
Affiliation(s)
- Oleg Dobrokhotov
- Mechanobiology Laboratory, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masaki Sunagawa
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Division of Surgical Oncology, Department of Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takeru Torii
- Frontiers of Innovative Research in Science and Technology, Konan University, Kobe, Japan
| | - Shinji Mii
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Keiko Kawauchi
- Frontiers of Innovative Research in Science and Technology, Konan University, Kobe, Japan
| | - Atsushi Enomoto
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masahiro Sokabe
- Mechanobiology Laboratory, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroaki Hirata
- Mechanobiology Laboratory, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
11
|
Anbar HS, Isa Z, Elounais JJ, Jameel MA, Zib JH, Samer AM, Jawad AF, El-Gamal MI. Steroid sulfatase inhibitors: the current landscape. Expert Opin Ther Pat 2021; 31:453-472. [PMID: 33783295 DOI: 10.1080/13543776.2021.1910237] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Introduction: Steroid sulfatase (STS) enzyme is responsible for transforming the inactive sulfate metabolites of steroid sex hormones into the active free steroids. Both the deficiency and the over-expression of STS are associated with the pathophysiology of certain diseases. This article provides the readership with a comprehensive review about STS enzyme and its recently reported inhibitors.Areas covered: In the present article, we reviewed the structure, location, and substrates of STS enzyme, physiological functions of STS, and disease states related to over-expression or deficiency of STS enzyme. STS inhibitors reported during the last five years (2016-present) have been reviewed as well.Expert opinion: Irosustat is the most successful STS inhibitor drug candidate so far. It is currently under investigation in clinical trials for treatment of estrogen-dependent breast cancer. Non-steroidal sulfamate is the most favorable scaffold for STS inhibitor design. They can be beneficial for the treatment of hormone-dependent cancers and neurodegenerative disorders without significant estrogenic side effects. Moreover, dual-acting molecules (inhibitors of STS + another synergistic mechanism) can be therapeutically efficient.
Collapse
Affiliation(s)
- Hanan S Anbar
- Department of Clinical Pharmacy and Pharmacotherapeutics, Dubai Pharmacy College for Girls, Dubai, United Arab Emirates
| | - Zahraa Isa
- Department of Clinical Pharmacy and Pharmacotherapeutics, Dubai Pharmacy College for Girls, Dubai, United Arab Emirates
| | - Jana J Elounais
- Department of Clinical Pharmacy and Pharmacotherapeutics, Dubai Pharmacy College for Girls, Dubai, United Arab Emirates
| | - Mariam A Jameel
- Department of Clinical Pharmacy and Pharmacotherapeutics, Dubai Pharmacy College for Girls, Dubai, United Arab Emirates
| | - Joudi H Zib
- Department of Clinical Pharmacy and Pharmacotherapeutics, Dubai Pharmacy College for Girls, Dubai, United Arab Emirates
| | - Aya M Samer
- Department of Clinical Pharmacy and Pharmacotherapeutics, Dubai Pharmacy College for Girls, Dubai, United Arab Emirates
| | - Aya F Jawad
- Department of Clinical Pharmacy and Pharmacotherapeutics, Dubai Pharmacy College for Girls, Dubai, United Arab Emirates
| | - Mohammed I El-Gamal
- Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates.,Department of Medicinal Chemistry, Faculty of Pharmacy, University of Mansoura, Mansoura, Egypt
| |
Collapse
|
12
|
Deguchi S, Kato A, Wu P, Hakamada M, Mabuchi M. Heterogeneous role of integrins in fibroblast response to small cyclic mechanical stimulus generated by a nanoporous gold actuator. Acta Biomater 2021; 121:418-430. [PMID: 33326880 DOI: 10.1016/j.actbio.2020.12.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 12/07/2020] [Accepted: 12/08/2020] [Indexed: 02/07/2023]
Abstract
It is important to understand the effects of mechanical stimulation on cell behaviors for homeostasis. Many studies have been performed on cell responses to mechanical stimuli, but the mechanosensing mechanism is still under debate. In the present study, experiments employing molecular dynamics (MD) simulations concerning the effects of cyclic mechanical stimulus on cell proliferation were performed based on the hypothesis that mechanosensing depends on integrin types. We used a nanoporous gold (NPG) actuator to prevent transfer of a mechanical stimulus via molecules other than integrins. Surprisingly, a small cyclic strain of only 0.5% enhanced the proliferation of fibroblasts. α5β1 and αvβ3 integrins showed high sensitivity to the mechanical stimulus, whereas α1β1 and α2β1 integrins exhibited low mechanosensitivity. The MD simulations showed that different conformational changes of the integrin headpiece induced by binding to the ECM led to a difference in mechanosensitivity between αI and αI-less integrin types. Thus, the present study provides evidence to support the hypothesis and suggests the mechanism for the heterogeneous roles of integrins in mechanosensing.
Collapse
Affiliation(s)
- Soichiro Deguchi
- Graduate School of Energy Science, Kyoto University, Yoshidahonmachi, Sakyo, Kyoto 606-8501, Japan.
| | - Atsushi Kato
- Graduate School of Energy Science, Kyoto University, Yoshidahonmachi, Sakyo, Kyoto 606-8501, Japan
| | - Peizheng Wu
- Graduate School of Energy Science, Kyoto University, Yoshidahonmachi, Sakyo, Kyoto 606-8501, Japan
| | - Masataka Hakamada
- Graduate School of Energy Science, Kyoto University, Yoshidahonmachi, Sakyo, Kyoto 606-8501, Japan
| | - Mamoru Mabuchi
- Graduate School of Energy Science, Kyoto University, Yoshidahonmachi, Sakyo, Kyoto 606-8501, Japan
| |
Collapse
|
13
|
Quantitative Phosphoproteomics Reveals Cell Alignment and Mitochondrial Length Change under Cyclic Stretching in Lung Cells. Int J Mol Sci 2020; 21:ijms21114074. [PMID: 32517296 PMCID: PMC7312583 DOI: 10.3390/ijms21114074] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 06/04/2020] [Accepted: 06/05/2020] [Indexed: 12/14/2022] Open
Abstract
Lung cancer is a leading cause of death. Most previous studies have been based on traditional cell-culturing methods. However, lung cells are periodically subjected to mechanical forces during breathing. Understanding the mechanisms underlying the cyclic stretching induced in lung cells may be important for lung cancer therapy. Here, we applied cyclic stretching to stimulate the continual contraction that is present under physiological conditions in lung cells. We first uncovered the stretching-induced phosphoproteome in lung cancer cell line A549 and fibroblast cell line IMR-90. We identified 2048 and 2604 phosphosites corresponding to 837 and 1008 phosphoproteins in A549 and IMR-90, respectively. Furthermore, we combined our phosphoproteomics and public gene expression data to identify the biological functions in response to cyclic stretching. Interestingly, cytoskeletal and mitochondrial reorganization were enriched. We further used cell imaging analysis to validate the profiling results and found that this physical force changed cell alignment and mitochondrial length. This study not only reveals the molecular mechanism of cyclic stretching but also provides evidence that cell stretching causes cellular rearrangement and mitochondrial length change.
Collapse
|
14
|
Miller AE, Hu P, Barker TH. Feeling Things Out: Bidirectional Signaling of the Cell-ECM Interface, Implications in the Mechanobiology of Cell Spreading, Migration, Proliferation, and Differentiation. Adv Healthc Mater 2020; 9:e1901445. [PMID: 32037719 PMCID: PMC7274903 DOI: 10.1002/adhm.201901445] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 01/10/2020] [Indexed: 12/16/2022]
Abstract
Biophysical cues stemming from the extracellular environment are rapidly transduced into discernible chemical messages (mechanotransduction) that direct cellular activities-placing the extracellular matrix (ECM) as a potent regulator of cell behavior. Dynamic reciprocity between the cell and its associated matrix is essential to the maintenance of tissue homeostasis and dysregulation of both ECM mechanical signaling, via pathological ECM turnover, and internal mechanotransduction pathways contribute to disease progression. This review covers the current understandings of the key modes of signaling used by both the cell and ECM to coregulate one another. By taking an outside-in approach, the inherent complexities and regulatory processes at each level of signaling (ECM, plasma membrane, focal adhesion, and cytoplasm) are captured to give a comprehensive picture of the internal and external mechanoregulatory environment. Specific emphasis is placed on the focal adhesion complex which acts as a central hub of mechanical signaling, regulating cell spreading, migration, proliferation, and differentiation. In addition, a wealth of available knowledge on mechanotransduction is curated to generate an integrated signaling network encompassing the central components of the focal adhesion, cytoplasm and nucleus that act in concert to promote durotaxis, proliferation, and differentiation in a stiffness-dependent manner.
Collapse
Affiliation(s)
- Andrew E Miller
- Department of Biomedical Engineering, University of Virginia, 415 Lane Rd. MR5 1225, Charlottesville, VA, 22903, USA
| | - Ping Hu
- Department of Biomedical Engineering, University of Virginia, 415 Lane Rd. MR5 1225, Charlottesville, VA, 22903, USA
| | - Thomas H Barker
- Department of Biomedical Engineering, University of Virginia, 415 Lane Rd. MR5 1225, Charlottesville, VA, 22903, USA
| |
Collapse
|
15
|
Enhancement of FAK alleviates ventilator-induced alveolar epithelial cell injury. Sci Rep 2020; 10:419. [PMID: 31942012 PMCID: PMC6962166 DOI: 10.1038/s41598-019-57350-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 12/16/2019] [Indexed: 01/19/2023] Open
Abstract
Mechanical ventilation induces lung injury by damaging alveolar epithelial cells (AECs), but the pathogenesis remains unknown. Focal adhesion kinase (FAK) is a cytoplasmic protein tyrosine kinase that is involved in cell growth and intracellular signal transduction pathways. This study explored the potential role of FAK in AECs during lung injury induced by mechanical ventilation. High-volume mechanical ventilation (HMV) was used to create a mouse lung injury model, which was validated by analysis of lung weight, bronchoalveolar lavage fluid and histological investigation. The expression of FAK and Akt in AECs were evaluated. In addition, recombinant FAK was administered to mice via the tail vein, and then the extent of lung injury was assessed. Mouse AECs were cultured in vitro, and FAK expression in cells under stretch was investigated. The effects of FAK on cell proliferation, migration and apoptosis were investigated. The results showed that HMV decreased FAK expression in AECs of mice, while FAK supplementation attenuated lung injury, reduced protein levels/cell counts in the bronchoalveolar lavage fluid and decreased histological lung injury and oedema. The protective effect of FAK promoted AEC proliferation and migration and prevented cells from undergoing apoptosis, which restored the integrity of the alveoli through Akt pathway. Therefore, the decrease in FAK expression by HMV is essential for injury to epithelial cells and the disruption of alveolar integrity. FAK supplementation can reduce AEC injury associated with HMV.
Collapse
|
16
|
Wang R, Thayer P, Goldstein A, Wagner WD. Interaction of material stiffness and negative pressure to enhance differentiation of bone marrow-derived stem cells and osteoblast proliferation. J Tissue Eng Regen Med 2020; 14:295-305. [PMID: 31845531 DOI: 10.1002/term.2993] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 10/31/2019] [Accepted: 11/27/2019] [Indexed: 01/03/2023]
Abstract
Negative pressure wound therapy (NPWT) results in improved wound repair and the combined use of NPWT with elastomeric materials may further stimulate and accelerate tissue repair. No firmly established treatment modalities using both NPWT and biomaterials exist for orthopedic application. The goal of this study was to investigate the response of osteoblasts and bone marrow-derived mesenchymal stem cells to negative pressure and to determine whether a newly developed elastic osteomimetic bone repair material (BRM), a blend of type I collagen, chondroitin 6-sulfate, and poly (octanediol citrate) could enhance the osteoblastic phenotype. The results indicate that proliferation and alkaline phosphatase activity of hFOB1.19 osteoblasts were significantly increased with exposure to 12 hr of negative pressure (-125 mmHg). Follow-on studies with rat and human mesenchymal stem cells confirmed that negative pressure enhanced osteoblastic maturation. In addition, a significant interaction of negative pressure and electrospun BRM resulted in increased mRNA expression of alkaline phosphatase, osteopontin, collagen1α2, and HIF1α, whereas little or no effect on these genes was observed on electrospun collagen or tissue culture plastic. Together, these results suggest that the use of this novel biomaterial, BRM, with NPWT may ultimately translate into a safe and cost-effective clinical application to accelerate bone repair.
Collapse
Affiliation(s)
- Rui Wang
- Plastic and Reconstructive Surgery, Wake Forest University School of Medicine, Winston-Salem, North Carolina.,Biomedical Engineering and Science, Virginia Tech-Wake Forest University School of Biomedical Engineering and Science, Winston-Salem, North Carolina
| | - Patrick Thayer
- Biomedical Engineering and Science, Virginia Tech-Wake Forest University School of Biomedical Engineering and Science, Winston-Salem, North Carolina
| | - Aaron Goldstein
- Biomedical Engineering and Science, Virginia Tech-Wake Forest University School of Biomedical Engineering and Science, Winston-Salem, North Carolina.,Department of Chemical Engineering, Virginia Tech, Blacksburg, Virginia
| | - William D Wagner
- Plastic and Reconstructive Surgery, Wake Forest University School of Medicine, Winston-Salem, North Carolina.,Biomedical Engineering and Science, Virginia Tech-Wake Forest University School of Biomedical Engineering and Science, Winston-Salem, North Carolina
| |
Collapse
|
17
|
Fang Y, Wu D, Birukov KG. Mechanosensing and Mechanoregulation of Endothelial Cell Functions. Compr Physiol 2019; 9:873-904. [PMID: 30873580 PMCID: PMC6697421 DOI: 10.1002/cphy.c180020] [Citation(s) in RCA: 125] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Vascular endothelial cells (ECs) form a semiselective barrier for macromolecules and cell elements regulated by dynamic interactions between cytoskeletal elements and cell adhesion complexes. ECs also participate in many other vital processes including innate immune reactions, vascular repair, secretion, and metabolism of bioactive molecules. Moreover, vascular ECs represent a unique cell type exposed to continuous, time-dependent mechanical forces: different patterns of shear stress imposed by blood flow in macrovasculature and by rolling blood cells in the microvasculature; circumferential cyclic stretch experienced by the arterial vascular bed caused by heart propulsions; mechanical stretch of lung microvascular endothelium at different magnitudes due to spontaneous respiration or mechanical ventilation in critically ill patients. Accumulating evidence suggests that vascular ECs contain mechanosensory complexes, which rapidly react to changes in mechanical loading, process the signal, and develop context-specific adaptive responses to rebalance the cell homeostatic state. The significance of the interactions between specific mechanical forces in the EC microenvironment together with circulating bioactive molecules in the progression and resolution of vascular pathologies including vascular injury, atherosclerosis, pulmonary edema, and acute respiratory distress syndrome has been only recently recognized. This review will summarize the current understanding of EC mechanosensory mechanisms, modulation of EC responses to humoral factors by surrounding mechanical forces (particularly the cyclic stretch), and discuss recent findings of magnitude-specific regulation of EC functions by transcriptional, posttranscriptional and epigenetic mechanisms using -omics approaches. We also discuss ongoing challenges and future opportunities in developing new therapies targeting dysregulated mechanosensing mechanisms to treat vascular diseases. © 2019 American Physiological Society. Compr Physiol 9:873-904, 2019.
Collapse
Affiliation(s)
- Yun Fang
- Department of Medicine, University of Chicago, Chicago, Illinois, USA,Correspondence to
| | - David Wu
- Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | - Konstantin G. Birukov
- Department of Anesthesiology, University of Maryland Baltimore School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
18
|
Jansen K, Atherton P, Ballestrem C. Mechanotransduction at the cell-matrix interface. Semin Cell Dev Biol 2017; 71:75-83. [DOI: 10.1016/j.semcdb.2017.07.027] [Citation(s) in RCA: 155] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 07/19/2017] [Accepted: 07/19/2017] [Indexed: 01/09/2023]
|
19
|
Zhang N, Ma D, Wang L, Zhu X, Pan Q, Zhao Y, Zhu W, Zhou J, Wang L, Chai Z, Ao J, Sun H, Tang Z. Insufficient Radiofrequency Ablation Treated Hepatocellular Carcinoma Cells Promote Metastasis by Up-Regulation ITGB3. J Cancer 2017; 8:3742-3754. [PMID: 29151962 PMCID: PMC5688928 DOI: 10.7150/jca.20816] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Accepted: 08/16/2017] [Indexed: 12/27/2022] Open
Abstract
Radiofrequency ablation (RFA) is one of the standards of care for early stage hepatocellular carcinoma (HCC). However, rapid progression of residual tumor after RFA has been confirmed. The aim of this study was to investigate the underlying mechanism of this phenomenon. Human HCC cell lines HCCLM3 and HepG2 were employed to establish insufficient RFA models in vivo and in vitro, respectively. The effects of insufficient RFA on metastatic potential of residual tumors were evaluated. The molecular changes after insufficient RFA were evaluated by PCR array, western blot, immunofluorescence, and immunohistochemistry. Results showed that insufficient RFA significantly promoted lung and intrahepatic residual tumor cells in vivo, and heat intervention promoted migration and invasion of hepatoma cells in vitro. PCR array revealed that the expression of integrin β3 (ITGB3) and MMP2 were up-regulated in the residual tumors of HCCLM3 xenograft model. The up-regulation of ITGB3 was confirmed by qRT-PCR, Western blot and immunohistochemistry. Knockdown ITGB3 expression in HCCLM3 cells by shRNA significantly lowered the pro-metastatic effects of insufficient RFA. Mechanism studies indicated that ITGB3 mediated the expression of MMP2 by activing FAK/PI3K/AKT signaling pathway. The up-regulation of ITGB3 contributed to enhanced metastatic potential of residual cancer in HCCLM3 model after insufficient RFA. Targeting ITGB3 expression may further improve the clinical effects of RFA.
Collapse
Affiliation(s)
- Ning Zhang
- Department of Liver Surgery, Fudan University Shanghai Cancer Center, Cancer Hospital, Shanghai, China
| | - Dening Ma
- Department of Colorectal Cancer Surgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Lu Wang
- Department of Liver Surgery, Fudan University Shanghai Cancer Center, Cancer Hospital, Shanghai, China
| | - Xiaodong Zhu
- Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Ministry of Education, Shanghai, China
| | - Qi Pan
- Department of Liver Surgery, Fudan University Shanghai Cancer Center, Cancer Hospital, Shanghai, China
| | - Yiming Zhao
- Department of Liver Surgery, Fudan University Shanghai Cancer Center, Cancer Hospital, Shanghai, China
| | - Weiping Zhu
- Department of Liver Surgery, Fudan University Shanghai Cancer Center, Cancer Hospital, Shanghai, China
| | - Jiamin Zhou
- Department of Liver Surgery, Fudan University Shanghai Cancer Center, Cancer Hospital, Shanghai, China
| | - Longrong Wang
- Department of Liver Surgery, Fudan University Shanghai Cancer Center, Cancer Hospital, Shanghai, China
| | - Zongtao Chai
- Department of Hepatic Surgery VI, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Jianyang Ao
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Huichuan Sun
- Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Ministry of Education, Shanghai, China
| | - Zhaoyou Tang
- Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Ministry of Education, Shanghai, China
| |
Collapse
|
20
|
Felsen D, Diaz BJ, Chen J, Gonzalez J, Kristensen MLV, Bohn AB, Roth BT, Poppas DP, Nørregaard R. Pressure and stretch differentially affect proliferation of renal proximal tubular cells. Physiol Rep 2017; 5:e13346. [PMID: 28904080 PMCID: PMC5599855 DOI: 10.14814/phy2.13346] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 06/14/2017] [Indexed: 12/17/2022] Open
Abstract
Renal obstruction is frequently found in adults and children. Mechanical stimuli, including pressure and stretch in the obstructed kidney, contribute to damage; animal models of obstruction are characterized by increased cellular proliferation. We were interested in the direct effects of pressure and stretch on renal tubular cell proliferation. Human HKC-8 or rat NRK-52E proximal tubule cells were subjected to either pressure [0, 60 or 90 mmHg] or static stretch [0 or 20%] for 24 or 48 h. Cell proliferation was measured by cell counting, cell cycle analyzed by flow cytometry, and PCNA and Skp2 expression were determined by qPCR or western blot. Blood gases were determined in an iSTAT system. Proliferation was also assessed in vivo after 24 h of ureteral obstruction. There was a significant increase in HKC-8 cell number after 48 h of exposure to either 60 or 90 mmHg pressure. Western blot and qPCR confirmed increased expression of PCNA and Skp2 in pressurized cells. Cell cycle measurements demonstrated an increase in HKC-8 in S phase. Mechanical stretching increased PCNA protein expression in HKC-8 cells after 48 h while no effect was observed on Skp2 and cell cycle measurements. Increased PCNA expression was found at 24 h after ureteral obstruction. We demonstrate direct transduction of pressure into a proliferative response in HKC-8 and NRK-52E cells, measured by cell number, PCNA and Skp2 expression and increase in cells in S phase, whereas stretch had a less robust effect on proliferation.
Collapse
Affiliation(s)
- Diane Felsen
- Department of Urology, Institute for Pediatric Urology, Komansky Center for Children's Health Weill Cornell Medicine, New York, New York
| | - Bianca J Diaz
- Department of Urology, Institute for Pediatric Urology, Komansky Center for Children's Health Weill Cornell Medicine, New York, New York
| | - Jie Chen
- Department of Urology, Institute for Pediatric Urology, Komansky Center for Children's Health Weill Cornell Medicine, New York, New York
| | - Juana Gonzalez
- Center for Clinical and Translational Science Rockefeller University, New York, New York
| | | | - Anja B Bohn
- Department of Clinical Medicine, Aarhus University, Aarhus C., Denmark
| | - Brendan T Roth
- Department of Urology, Institute for Pediatric Urology, Komansky Center for Children's Health Weill Cornell Medicine, New York, New York
| | - Dix P Poppas
- Department of Urology, Institute for Pediatric Urology, Komansky Center for Children's Health Weill Cornell Medicine, New York, New York
| | - Rikke Nørregaard
- Department of Clinical Medicine, Aarhus University, Aarhus C., Denmark
| |
Collapse
|
21
|
Ye DJ, Kwon YJ, Shin S, Baek HS, Shin DW, Chun YJ. Induction of Integrin Signaling by Steroid Sulfatase in Human Cervical Cancer Cells. Biomol Ther (Seoul) 2017; 25:321-328. [PMID: 27956712 PMCID: PMC5424643 DOI: 10.4062/biomolther.2016.155] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2016] [Revised: 09/02/2016] [Accepted: 10/04/2016] [Indexed: 12/19/2022] Open
Abstract
Steroid sulfatase (STS) is an enzyme responsible for the hydrolysis of aryl and alkyl sulfates. STS plays a pivotal role in the regulation of estrogens and androgens that promote the growth of hormone-dependent tumors, such as those of breast or prostate cancer. However, the molecular function of STS in tumor growth is still not clear. To elucidate the role of STS in cancer cell proliferation, we investigated whether STS is able to regulate the integrin signaling pathway. We found that overexpression of STS in HeLa cells increases the protein and mRNA levels of integrin β1 and fibronectin, a ligand of integrin α5β1. Dehydroepiandrosterone (DHEA), one of the main metabolites of STS, also increases mRNA and protein expression of integrin β1 and fibronectin. Further, STS expression and DHEA treatment enhanced phosphorylation of focal adhesion kinase (FAK) at the Tyr 925 residue. Moreover, increased phosphorylation of ERK at Thr 202 and Tyr 204 residues by STS indicates that STS activates the MAPK/ERK pathway. In conclusion, these results suggest that STS expression and DHEA treatment may enhance MAPK/ERK signaling through up-regulation of integrin β1 and activation of FAK.
Collapse
Affiliation(s)
- Dong-Jin Ye
- College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Yeo-Jung Kwon
- College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Sangyun Shin
- College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Hyoung-Seok Baek
- College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Dong-Won Shin
- College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Young-Jin Chun
- College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea
| |
Collapse
|
22
|
Cell culture: complications due to mechanical release of ATP and activation of purinoceptors. Cell Tissue Res 2017; 370:1-11. [PMID: 28434079 PMCID: PMC5610203 DOI: 10.1007/s00441-017-2618-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 03/21/2017] [Indexed: 12/11/2022]
Abstract
There is abundant evidence that ATP (adenosine 5′-triphosphate) is released from a variety of cultured cells in response to mechanical stimulation. The release mechanism involved appears to be a combination of vesicular exocytosis and connexin and pannexin hemichannels. Purinergic receptors on cultured cells mediate both short-term purinergic signalling of secretion and long-term (trophic) signalling such as proliferation, migration, differentiation and apoptosis. We aim in this review to bring to the attention of non-purinergic researchers using tissue culture that the release of ATP in response to mechanical stress evoked by the unavoidable movement of the cells acting on functional purinergic receptors on the culture cells is likely to complicate the interpretation of their data.
Collapse
|
23
|
Sager HB, Hulsmans M, Lavine KJ, Moreira MB, Heidt T, Courties G, Sun Y, Iwamoto Y, Tricot B, Khan OF, Dahlman JE, Borodovsky A, Fitzgerald K, Anderson DG, Weissleder R, Libby P, Swirski FK, Nahrendorf M. Proliferation and Recruitment Contribute to Myocardial Macrophage Expansion in Chronic Heart Failure. Circ Res 2016; 119:853-64. [PMID: 27444755 DOI: 10.1161/circresaha.116.309001] [Citation(s) in RCA: 346] [Impact Index Per Article: 38.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 07/21/2016] [Indexed: 12/12/2022]
Abstract
RATIONALE Macrophages reside in the healthy myocardium, participate in ischemic heart disease, and modulate myocardial infarction (MI) healing. Their origin and roles in post-MI remodeling of nonischemic remote myocardium, however, remain unclear. OBJECTIVE This study investigated the number, origin, phenotype, and function of remote cardiac macrophages residing in the nonischemic myocardium in mice with chronic heart failure after coronary ligation. METHODS AND RESULTS Eight weeks post MI, fate mapping and flow cytometry revealed that a 2.9-fold increase in remote macrophages results from both increased local macrophage proliferation and monocyte recruitment. Heart failure produced by extensive MI, through activation of the sympathetic nervous system, expanded medullary and extramedullary hematopoiesis. Circulating Ly6C(high) monocytes rose from 64±5 to 108±9 per microliter of blood (P<0.05). Cardiac monocyte recruitment declined in Ccr2(-/-) mice, reducing macrophage numbers in the failing myocardium. Mechanical strain of primary murine and human macrophage cultures promoted cell cycle entry, suggesting that the increased wall tension in post-MI heart failure stimulates local macrophage proliferation. Strained cells activated the mitogen-activated protein kinase pathway, whereas specific inhibitors of this pathway reduced macrophage proliferation in strained cell cultures and in the failing myocardium (P<0.05). Steady-state cardiac macrophages, monocyte-derived macrophages, and locally sourced macrophages isolated from failing myocardium expressed different genes in a pattern distinct from the M1/M2 macrophage polarization paradigm. In vivo silencing of endothelial cell adhesion molecules curbed post-MI monocyte recruitment to the remote myocardium and preserved ejection fraction (27.4±2.4 versus 19.1±2%; P<0.05). CONCLUSIONS Myocardial failure is influenced by an altered myeloid cell repertoire.
Collapse
Affiliation(s)
- Hendrik B Sager
- From the Center for Systems Biology, Department of Imaging (H.B.S., M.H., T.H., G.C., Y.S., Y.I., B.T., R.W., F.K.S., M.N.) and Cardiovascular Research Center (M.N.), Massachusetts General Hospital and Harvard Medical School, Boston; Center for Cardiovascular Research, Washington University School of Medicine, St Louis, MS (K.J.L.); Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA (M.B.M., P.L.); Department of Cardiology and Angiology I, Heart Center Freiburg University, Germany (T.H.); Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA (O.F.K., J.E.D., D.G.A.); David H. Koch Institute for Integrative Cancer Research (O.F.K., J.E.D., D.G.A.) and Department of Chemical Engineering (D.G.A.), Massachusetts Institute of Technology, Cambridge; Alnylam Pharmaceuticals, Cambridge, MA (A.B., K.F.); and Department of Systems Biology, Harvard Medical School, Boston, MA (R.W.).
| | - Maarten Hulsmans
- From the Center for Systems Biology, Department of Imaging (H.B.S., M.H., T.H., G.C., Y.S., Y.I., B.T., R.W., F.K.S., M.N.) and Cardiovascular Research Center (M.N.), Massachusetts General Hospital and Harvard Medical School, Boston; Center for Cardiovascular Research, Washington University School of Medicine, St Louis, MS (K.J.L.); Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA (M.B.M., P.L.); Department of Cardiology and Angiology I, Heart Center Freiburg University, Germany (T.H.); Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA (O.F.K., J.E.D., D.G.A.); David H. Koch Institute for Integrative Cancer Research (O.F.K., J.E.D., D.G.A.) and Department of Chemical Engineering (D.G.A.), Massachusetts Institute of Technology, Cambridge; Alnylam Pharmaceuticals, Cambridge, MA (A.B., K.F.); and Department of Systems Biology, Harvard Medical School, Boston, MA (R.W.)
| | - Kory J Lavine
- From the Center for Systems Biology, Department of Imaging (H.B.S., M.H., T.H., G.C., Y.S., Y.I., B.T., R.W., F.K.S., M.N.) and Cardiovascular Research Center (M.N.), Massachusetts General Hospital and Harvard Medical School, Boston; Center for Cardiovascular Research, Washington University School of Medicine, St Louis, MS (K.J.L.); Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA (M.B.M., P.L.); Department of Cardiology and Angiology I, Heart Center Freiburg University, Germany (T.H.); Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA (O.F.K., J.E.D., D.G.A.); David H. Koch Institute for Integrative Cancer Research (O.F.K., J.E.D., D.G.A.) and Department of Chemical Engineering (D.G.A.), Massachusetts Institute of Technology, Cambridge; Alnylam Pharmaceuticals, Cambridge, MA (A.B., K.F.); and Department of Systems Biology, Harvard Medical School, Boston, MA (R.W.)
| | - Marina B Moreira
- From the Center for Systems Biology, Department of Imaging (H.B.S., M.H., T.H., G.C., Y.S., Y.I., B.T., R.W., F.K.S., M.N.) and Cardiovascular Research Center (M.N.), Massachusetts General Hospital and Harvard Medical School, Boston; Center for Cardiovascular Research, Washington University School of Medicine, St Louis, MS (K.J.L.); Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA (M.B.M., P.L.); Department of Cardiology and Angiology I, Heart Center Freiburg University, Germany (T.H.); Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA (O.F.K., J.E.D., D.G.A.); David H. Koch Institute for Integrative Cancer Research (O.F.K., J.E.D., D.G.A.) and Department of Chemical Engineering (D.G.A.), Massachusetts Institute of Technology, Cambridge; Alnylam Pharmaceuticals, Cambridge, MA (A.B., K.F.); and Department of Systems Biology, Harvard Medical School, Boston, MA (R.W.)
| | - Timo Heidt
- From the Center for Systems Biology, Department of Imaging (H.B.S., M.H., T.H., G.C., Y.S., Y.I., B.T., R.W., F.K.S., M.N.) and Cardiovascular Research Center (M.N.), Massachusetts General Hospital and Harvard Medical School, Boston; Center for Cardiovascular Research, Washington University School of Medicine, St Louis, MS (K.J.L.); Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA (M.B.M., P.L.); Department of Cardiology and Angiology I, Heart Center Freiburg University, Germany (T.H.); Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA (O.F.K., J.E.D., D.G.A.); David H. Koch Institute for Integrative Cancer Research (O.F.K., J.E.D., D.G.A.) and Department of Chemical Engineering (D.G.A.), Massachusetts Institute of Technology, Cambridge; Alnylam Pharmaceuticals, Cambridge, MA (A.B., K.F.); and Department of Systems Biology, Harvard Medical School, Boston, MA (R.W.)
| | - Gabriel Courties
- From the Center for Systems Biology, Department of Imaging (H.B.S., M.H., T.H., G.C., Y.S., Y.I., B.T., R.W., F.K.S., M.N.) and Cardiovascular Research Center (M.N.), Massachusetts General Hospital and Harvard Medical School, Boston; Center for Cardiovascular Research, Washington University School of Medicine, St Louis, MS (K.J.L.); Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA (M.B.M., P.L.); Department of Cardiology and Angiology I, Heart Center Freiburg University, Germany (T.H.); Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA (O.F.K., J.E.D., D.G.A.); David H. Koch Institute for Integrative Cancer Research (O.F.K., J.E.D., D.G.A.) and Department of Chemical Engineering (D.G.A.), Massachusetts Institute of Technology, Cambridge; Alnylam Pharmaceuticals, Cambridge, MA (A.B., K.F.); and Department of Systems Biology, Harvard Medical School, Boston, MA (R.W.)
| | - Yuan Sun
- From the Center for Systems Biology, Department of Imaging (H.B.S., M.H., T.H., G.C., Y.S., Y.I., B.T., R.W., F.K.S., M.N.) and Cardiovascular Research Center (M.N.), Massachusetts General Hospital and Harvard Medical School, Boston; Center for Cardiovascular Research, Washington University School of Medicine, St Louis, MS (K.J.L.); Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA (M.B.M., P.L.); Department of Cardiology and Angiology I, Heart Center Freiburg University, Germany (T.H.); Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA (O.F.K., J.E.D., D.G.A.); David H. Koch Institute for Integrative Cancer Research (O.F.K., J.E.D., D.G.A.) and Department of Chemical Engineering (D.G.A.), Massachusetts Institute of Technology, Cambridge; Alnylam Pharmaceuticals, Cambridge, MA (A.B., K.F.); and Department of Systems Biology, Harvard Medical School, Boston, MA (R.W.)
| | - Yoshiko Iwamoto
- From the Center for Systems Biology, Department of Imaging (H.B.S., M.H., T.H., G.C., Y.S., Y.I., B.T., R.W., F.K.S., M.N.) and Cardiovascular Research Center (M.N.), Massachusetts General Hospital and Harvard Medical School, Boston; Center for Cardiovascular Research, Washington University School of Medicine, St Louis, MS (K.J.L.); Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA (M.B.M., P.L.); Department of Cardiology and Angiology I, Heart Center Freiburg University, Germany (T.H.); Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA (O.F.K., J.E.D., D.G.A.); David H. Koch Institute for Integrative Cancer Research (O.F.K., J.E.D., D.G.A.) and Department of Chemical Engineering (D.G.A.), Massachusetts Institute of Technology, Cambridge; Alnylam Pharmaceuticals, Cambridge, MA (A.B., K.F.); and Department of Systems Biology, Harvard Medical School, Boston, MA (R.W.)
| | - Benoit Tricot
- From the Center for Systems Biology, Department of Imaging (H.B.S., M.H., T.H., G.C., Y.S., Y.I., B.T., R.W., F.K.S., M.N.) and Cardiovascular Research Center (M.N.), Massachusetts General Hospital and Harvard Medical School, Boston; Center for Cardiovascular Research, Washington University School of Medicine, St Louis, MS (K.J.L.); Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA (M.B.M., P.L.); Department of Cardiology and Angiology I, Heart Center Freiburg University, Germany (T.H.); Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA (O.F.K., J.E.D., D.G.A.); David H. Koch Institute for Integrative Cancer Research (O.F.K., J.E.D., D.G.A.) and Department of Chemical Engineering (D.G.A.), Massachusetts Institute of Technology, Cambridge; Alnylam Pharmaceuticals, Cambridge, MA (A.B., K.F.); and Department of Systems Biology, Harvard Medical School, Boston, MA (R.W.)
| | - Omar F Khan
- From the Center for Systems Biology, Department of Imaging (H.B.S., M.H., T.H., G.C., Y.S., Y.I., B.T., R.W., F.K.S., M.N.) and Cardiovascular Research Center (M.N.), Massachusetts General Hospital and Harvard Medical School, Boston; Center for Cardiovascular Research, Washington University School of Medicine, St Louis, MS (K.J.L.); Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA (M.B.M., P.L.); Department of Cardiology and Angiology I, Heart Center Freiburg University, Germany (T.H.); Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA (O.F.K., J.E.D., D.G.A.); David H. Koch Institute for Integrative Cancer Research (O.F.K., J.E.D., D.G.A.) and Department of Chemical Engineering (D.G.A.), Massachusetts Institute of Technology, Cambridge; Alnylam Pharmaceuticals, Cambridge, MA (A.B., K.F.); and Department of Systems Biology, Harvard Medical School, Boston, MA (R.W.)
| | - James E Dahlman
- From the Center for Systems Biology, Department of Imaging (H.B.S., M.H., T.H., G.C., Y.S., Y.I., B.T., R.W., F.K.S., M.N.) and Cardiovascular Research Center (M.N.), Massachusetts General Hospital and Harvard Medical School, Boston; Center for Cardiovascular Research, Washington University School of Medicine, St Louis, MS (K.J.L.); Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA (M.B.M., P.L.); Department of Cardiology and Angiology I, Heart Center Freiburg University, Germany (T.H.); Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA (O.F.K., J.E.D., D.G.A.); David H. Koch Institute for Integrative Cancer Research (O.F.K., J.E.D., D.G.A.) and Department of Chemical Engineering (D.G.A.), Massachusetts Institute of Technology, Cambridge; Alnylam Pharmaceuticals, Cambridge, MA (A.B., K.F.); and Department of Systems Biology, Harvard Medical School, Boston, MA (R.W.)
| | - Anna Borodovsky
- From the Center for Systems Biology, Department of Imaging (H.B.S., M.H., T.H., G.C., Y.S., Y.I., B.T., R.W., F.K.S., M.N.) and Cardiovascular Research Center (M.N.), Massachusetts General Hospital and Harvard Medical School, Boston; Center for Cardiovascular Research, Washington University School of Medicine, St Louis, MS (K.J.L.); Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA (M.B.M., P.L.); Department of Cardiology and Angiology I, Heart Center Freiburg University, Germany (T.H.); Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA (O.F.K., J.E.D., D.G.A.); David H. Koch Institute for Integrative Cancer Research (O.F.K., J.E.D., D.G.A.) and Department of Chemical Engineering (D.G.A.), Massachusetts Institute of Technology, Cambridge; Alnylam Pharmaceuticals, Cambridge, MA (A.B., K.F.); and Department of Systems Biology, Harvard Medical School, Boston, MA (R.W.)
| | - Kevin Fitzgerald
- From the Center for Systems Biology, Department of Imaging (H.B.S., M.H., T.H., G.C., Y.S., Y.I., B.T., R.W., F.K.S., M.N.) and Cardiovascular Research Center (M.N.), Massachusetts General Hospital and Harvard Medical School, Boston; Center for Cardiovascular Research, Washington University School of Medicine, St Louis, MS (K.J.L.); Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA (M.B.M., P.L.); Department of Cardiology and Angiology I, Heart Center Freiburg University, Germany (T.H.); Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA (O.F.K., J.E.D., D.G.A.); David H. Koch Institute for Integrative Cancer Research (O.F.K., J.E.D., D.G.A.) and Department of Chemical Engineering (D.G.A.), Massachusetts Institute of Technology, Cambridge; Alnylam Pharmaceuticals, Cambridge, MA (A.B., K.F.); and Department of Systems Biology, Harvard Medical School, Boston, MA (R.W.)
| | - Daniel G Anderson
- From the Center for Systems Biology, Department of Imaging (H.B.S., M.H., T.H., G.C., Y.S., Y.I., B.T., R.W., F.K.S., M.N.) and Cardiovascular Research Center (M.N.), Massachusetts General Hospital and Harvard Medical School, Boston; Center for Cardiovascular Research, Washington University School of Medicine, St Louis, MS (K.J.L.); Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA (M.B.M., P.L.); Department of Cardiology and Angiology I, Heart Center Freiburg University, Germany (T.H.); Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA (O.F.K., J.E.D., D.G.A.); David H. Koch Institute for Integrative Cancer Research (O.F.K., J.E.D., D.G.A.) and Department of Chemical Engineering (D.G.A.), Massachusetts Institute of Technology, Cambridge; Alnylam Pharmaceuticals, Cambridge, MA (A.B., K.F.); and Department of Systems Biology, Harvard Medical School, Boston, MA (R.W.)
| | - Ralph Weissleder
- From the Center for Systems Biology, Department of Imaging (H.B.S., M.H., T.H., G.C., Y.S., Y.I., B.T., R.W., F.K.S., M.N.) and Cardiovascular Research Center (M.N.), Massachusetts General Hospital and Harvard Medical School, Boston; Center for Cardiovascular Research, Washington University School of Medicine, St Louis, MS (K.J.L.); Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA (M.B.M., P.L.); Department of Cardiology and Angiology I, Heart Center Freiburg University, Germany (T.H.); Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA (O.F.K., J.E.D., D.G.A.); David H. Koch Institute for Integrative Cancer Research (O.F.K., J.E.D., D.G.A.) and Department of Chemical Engineering (D.G.A.), Massachusetts Institute of Technology, Cambridge; Alnylam Pharmaceuticals, Cambridge, MA (A.B., K.F.); and Department of Systems Biology, Harvard Medical School, Boston, MA (R.W.)
| | - Peter Libby
- From the Center for Systems Biology, Department of Imaging (H.B.S., M.H., T.H., G.C., Y.S., Y.I., B.T., R.W., F.K.S., M.N.) and Cardiovascular Research Center (M.N.), Massachusetts General Hospital and Harvard Medical School, Boston; Center for Cardiovascular Research, Washington University School of Medicine, St Louis, MS (K.J.L.); Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA (M.B.M., P.L.); Department of Cardiology and Angiology I, Heart Center Freiburg University, Germany (T.H.); Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA (O.F.K., J.E.D., D.G.A.); David H. Koch Institute for Integrative Cancer Research (O.F.K., J.E.D., D.G.A.) and Department of Chemical Engineering (D.G.A.), Massachusetts Institute of Technology, Cambridge; Alnylam Pharmaceuticals, Cambridge, MA (A.B., K.F.); and Department of Systems Biology, Harvard Medical School, Boston, MA (R.W.)
| | - Filip K Swirski
- From the Center for Systems Biology, Department of Imaging (H.B.S., M.H., T.H., G.C., Y.S., Y.I., B.T., R.W., F.K.S., M.N.) and Cardiovascular Research Center (M.N.), Massachusetts General Hospital and Harvard Medical School, Boston; Center for Cardiovascular Research, Washington University School of Medicine, St Louis, MS (K.J.L.); Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA (M.B.M., P.L.); Department of Cardiology and Angiology I, Heart Center Freiburg University, Germany (T.H.); Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA (O.F.K., J.E.D., D.G.A.); David H. Koch Institute for Integrative Cancer Research (O.F.K., J.E.D., D.G.A.) and Department of Chemical Engineering (D.G.A.), Massachusetts Institute of Technology, Cambridge; Alnylam Pharmaceuticals, Cambridge, MA (A.B., K.F.); and Department of Systems Biology, Harvard Medical School, Boston, MA (R.W.)
| | - Matthias Nahrendorf
- From the Center for Systems Biology, Department of Imaging (H.B.S., M.H., T.H., G.C., Y.S., Y.I., B.T., R.W., F.K.S., M.N.) and Cardiovascular Research Center (M.N.), Massachusetts General Hospital and Harvard Medical School, Boston; Center for Cardiovascular Research, Washington University School of Medicine, St Louis, MS (K.J.L.); Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA (M.B.M., P.L.); Department of Cardiology and Angiology I, Heart Center Freiburg University, Germany (T.H.); Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA (O.F.K., J.E.D., D.G.A.); David H. Koch Institute for Integrative Cancer Research (O.F.K., J.E.D., D.G.A.) and Department of Chemical Engineering (D.G.A.), Massachusetts Institute of Technology, Cambridge; Alnylam Pharmaceuticals, Cambridge, MA (A.B., K.F.); and Department of Systems Biology, Harvard Medical School, Boston, MA (R.W.).
| |
Collapse
|
24
|
Gupta P, Hari Narayana S. N. G, Kasiviswanathan U, Agarwal T, K. S, Mukhopadhyay D, Pal K, Giri S, Maiti TK, Banerjee I. Substrate stiffness does affect the fate of human keratinocytes. RSC Adv 2016. [DOI: 10.1039/c5ra19947f] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Epithelial cells response to the varying stiffness of polydimethyl siloxane (PDMS) substrate.
Collapse
Affiliation(s)
- Prerak Gupta
- Department of Biotechnology & Medical Engineering
- National Institute of Technology
- Rourkela-769008
- India
| | | | - Uvanesh Kasiviswanathan
- Department of Biotechnology & Medical Engineering
- National Institute of Technology
- Rourkela-769008
- India
| | - Tarun Agarwal
- Department of Biotechnology & Medical Engineering
- National Institute of Technology
- Rourkela-769008
- India
- Department of Biotechnology
| | - Senthilguru K.
- Department of Biotechnology & Medical Engineering
- National Institute of Technology
- Rourkela-769008
- India
| | - Devdeep Mukhopadhyay
- Department of Biotechnology
- Indian Institute of Technology Kharagpur
- Kharagpur-721302
- India
| | - Kunal Pal
- Department of Biotechnology & Medical Engineering
- National Institute of Technology
- Rourkela-769008
- India
| | - Supratim Giri
- Department of Chemistry
- National Institute of Technology
- Rourkela-769008
- India
| | - Tapas K. Maiti
- Department of Biotechnology
- Indian Institute of Technology Kharagpur
- Kharagpur-721302
- India
| | - Indranil Banerjee
- Department of Biotechnology & Medical Engineering
- National Institute of Technology
- Rourkela-769008
- India
| |
Collapse
|
25
|
Morrissey JB, Cheng RY, Davoudi S, Gilbert PM. Biomechanical Origins of Muscle Stem Cell Signal Transduction. J Mol Biol 2015; 428:1441-54. [PMID: 26004541 DOI: 10.1016/j.jmb.2015.05.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 05/03/2015] [Accepted: 05/06/2015] [Indexed: 10/23/2022]
Abstract
Skeletal muscle, the most abundant and widespread tissue in the human body, contracts upon receiving electrochemical signals from the nervous system to support essential functions such as thermoregulation, limb movement, blinking, swallowing and breathing. Reconstruction of adult muscle tissue relies on a pool of mononucleate, resident muscle stem cells, known as "satellite cells", expressing the paired-box transcription factor Pax7 necessary for their specification during embryonic development and long-term maintenance during adult life. Satellite cells are located around the myofibres in a niche at the interface of the basal lamina and the host fibre plasma membrane (i.e., sarcolemma), at a very low frequency. Upon damage to the myofibres, quiescent satellite cells are activated and give rise to a population of transient amplifying myogenic progenitor cells, which eventually exit the cell cycle permanently and fuse to form new myofibres and regenerate the tissue. A subpopulation of satellite cells self-renew and repopulate the niche, poised to respond to future demands. Harnessing the potential of satellite cells relies on a complete understanding of the molecular mechanisms guiding their regulation in vivo. Over the past several decades, studies revealed many signal transduction pathways responsible for satellite cell fate decisions, but the niche cues driving the activation and silencing of these pathways are less clear. Here we explore the scintillating possibility that considering the dynamic changes in the biophysical properties of the skeletal muscle, namely stiffness, and the stretch and shear forces to which a myofibre can be subjected to may provide missing information necessary to gain a full understanding of satellite cell niche regulation.
Collapse
Affiliation(s)
- James B Morrissey
- Institute of Biomaterials and Biomedical Engineering, Toronto, ON, Canada M5S3G9; Donnelly Centre for Cellular and Biomolecular Research, Toronto, ON, Canada M5S3E1
| | - Richard Y Cheng
- Institute of Biomaterials and Biomedical Engineering, Toronto, ON, Canada M5S3G9; Donnelly Centre for Cellular and Biomolecular Research, Toronto, ON, Canada M5S3E1
| | - Sadegh Davoudi
- Institute of Biomaterials and Biomedical Engineering, Toronto, ON, Canada M5S3G9; Donnelly Centre for Cellular and Biomolecular Research, Toronto, ON, Canada M5S3E1
| | - Penney M Gilbert
- Institute of Biomaterials and Biomedical Engineering, Toronto, ON, Canada M5S3G9; Donnelly Centre for Cellular and Biomolecular Research, Toronto, ON, Canada M5S3E1.
| |
Collapse
|
26
|
Codelia VA, Sun G, Irvine KD. Regulation of YAP by mechanical strain through Jnk and Hippo signaling. Curr Biol 2014; 24:2012-7. [PMID: 25127217 PMCID: PMC4160395 DOI: 10.1016/j.cub.2014.07.034] [Citation(s) in RCA: 183] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Revised: 06/04/2014] [Accepted: 07/14/2014] [Indexed: 12/31/2022]
Abstract
Mechanical forces affect all the tissues of our bodies. Experiments conducted mainly on cultured cells have established that altering these forces influences cell behaviors, including migration, differentiation, apoptosis, and proliferation [1, 2]. The transcriptional coactivator YAP has been identified as a nuclear relay of mechanical signals, but the molecular mechanisms that lead to YAP activation were not identified [3]. YAP is the main transcriptional effector of the Hippo signaling pathway, a major growth regulatory pathway within metazoa [4], but at least in some instances, the influence of mechanical strain on YAP was reported to be independent of Hippo signaling [5, 6]. Here, we identify a molecular pathway that can promote the proliferation of cultured mammary epithelial cells in response to cyclic or static stretch. These mechanical stimuli are associated with increased activity of the transcriptional coactivator YAP, which is due at least in part to inhibition of Hippo pathway activity. Much of this influence on Hippo signaling can be accounted for by the activation of c-Jun N-terminal kinase (JNK) activity by mechanical strain and subsequent inhibition of Hippo signaling by JNK. LATS1 is a key negative regulator of YAP within the Hippo pathway, and we further show that cyclic stretch is associated with a JNK-dependent increase in binding of a LATS inhibitor, LIMD1, to the LATS1 kinase and that reduction of LIMD1 expression suppresses the activation of YAP by cyclic stretch. Together, these observations establish a pathway for mechanical regulation of cell proliferation via JNK-mediated inhibition of Hippo signaling.
Collapse
Affiliation(s)
- Veronica A Codelia
- Howard Hughes Medical Institute, Waksman Institute, and Department of Molecular Biology and Biochemistry, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Gongping Sun
- Howard Hughes Medical Institute, Waksman Institute, and Department of Molecular Biology and Biochemistry, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Kenneth D Irvine
- Howard Hughes Medical Institute, Waksman Institute, and Department of Molecular Biology and Biochemistry, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA.
| |
Collapse
|
27
|
In vitro experimental study for the determination of cellular axial strain threshold and preferential axial strain from cell orientation behavior in a non-uniform deformation field. Cell Biochem Biophys 2014; 67:1249-59. [PMID: 23703662 DOI: 10.1007/s12013-013-9643-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Cells within connective tissues are routinely subjected to a wide range of non-uniform mechanical loads that regulate many cell behaviors. In the present study, the relationship between cell orientation angle and strain value of the membrane was comprehensively investigated using an inhomogeneous strain field. Additionally, the cellular axial strain threshold, which corresponds to the launching of cell reorientation response, was elucidated. Human bone marrow mesenchymal stem cells were used for these experiments. In this study, an inhomogeneous strain distribution was easily created by removing one side holes of an elastic chamber in a commonly used uniaxial stretching device. The strains of 2D stretched membranes were quantified on a position-by-position basis using the digital image correlation method. The normal strain in the direction of stretch was changed continuously from 2.0 to 15.0%. A 3D histogram of the cell frequency, which was correlated with the cell orientation angle and normal strain of the membrane, made it possible to determine the axial strain threshold accurately. The value of the axial strain threshold was 4.4 ± 0.3%, which was reasonable compared with previous studies based on cyclic uniaxial stretch stimulation (homogeneous strain field). Additionally, preferential axial strain of cells, which was a cell property firstly introduced, was also achieved and the value was -2.0 ± 0.1%. This study is novel in three respects: (i) it precisely and easily determined the axial strain threshold of cells; (ii) it is the first to suggest preferential axial strain of cells; and (iii) it methodically investigated cell behavior in an inhomogeneous strain field.
Collapse
|
28
|
Tomakidi P, Schulz S, Proksch S, Weber W, Steinberg T. Focal adhesion kinase (FAK) perspectives in mechanobiology: implications for cell behaviour. Cell Tissue Res 2014; 357:515-26. [PMID: 24988914 DOI: 10.1007/s00441-014-1945-2] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Accepted: 06/04/2014] [Indexed: 11/24/2022]
Abstract
Mechanobiology is a scientific interface discipline emerging from engineering and biology. With regard to tissue-regenerative cell-based strategies, mechanobiological concepts, including biomechanics as a target for cell and human mesenchymal stem cell behaviour, are on the march. Based on the periodontium as a paradigm, this mini-review discusses the key role of focal-adhesion kinase (FAK) in mechanobiology, since it is involved in mediating the transformation of environmental biomechanical signals into cell behavioural responses via mechanotransducing signalling cascades. These processes enable cells to adjust quickly to environmental cues, whereas adjustment itself relies on the specific intramolecular phosphorylation of FAK tyrosine residues and the multiple interactions of FAK with distinct partners. Furthermore, interaction-triggered mechanotransducing pathways govern the dynamics of focal adhesion sites and cell behaviour. Facets of behaviour not only include cell spreading and motility, but also proliferation, differentiation and apoptosis. In translational terms, identified and characterized biomechanical parameters can be incorporated into innovative concepts of cell- and tissue-tailored clinically applied biomaterials controlling cell behaviour as desired.
Collapse
Affiliation(s)
- Pascal Tomakidi
- Department of Oral Biotechnology, University Hospital Freiburg, Hugstetter Strasse 55, 79106, Freiburg, Germany,
| | | | | | | | | |
Collapse
|
29
|
Bonakdar N, Schilling A, Lennert P, Spörrer M, Gerum RC, Alonso JL, Goldmann WH. Measuring mechanical properties in cells: three easy methods for biologists. Cell Biol Int 2014; 38:1227-32. [PMID: 24803101 DOI: 10.1002/cbin.10303] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Accepted: 04/14/2014] [Indexed: 11/08/2022]
Affiliation(s)
- Navid Bonakdar
- Department of Physics, Biophysics, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | | | | | | | | | | | | |
Collapse
|
30
|
Yang X, Zhang Y, Zhang L, He T, Zhang J, Li C. Prion protein and cancers. Acta Biochim Biophys Sin (Shanghai) 2014; 46:431-40. [PMID: 24681883 DOI: 10.1093/abbs/gmu019] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The normal cellular prion protein, PrP(C) is a highly conserved and widely expressed cell surface glycoprotein in all mammals. The expression of PrP is pivotal in the pathogenesis of prion diseases; however, the normal physiological functions of PrP(C) remain incompletely understood. Based on the studies in cell models, a plethora of functions have been attributed to PrP(C). In this paper, we reviewed the potential roles that PrP(C) plays in cell physiology and focused on its contribution to tumorigenesis.
Collapse
Affiliation(s)
- Xiaowen Yang
- Department of the First Abdominal Surgery, Jiangxi Tumor Hospital, Nanchang 330029, China
| | - Yan Zhang
- Department of Molecular Endocrinology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Lihua Zhang
- Department of Pathology, Zhongda Hospital, Southeast University, Nanjing 210009, China
| | - Tianlin He
- Department of General Surgery, Changhai Hospital of Second Military Medical University, Shanghai 200433, China
| | - Jie Zhang
- Department of Stomatology, The First Affiliated Hospital of Shihezi University Medical College, Shihezi 832000, China
| | - Chaoyang Li
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| |
Collapse
|
31
|
Duscher D, Maan ZN, Wong VW, Rennert RC, Januszyk M, Rodrigues M, Hu M, Whitmore AJ, Whittam AJ, Longaker MT, Gurtner GC. Mechanotransduction and fibrosis. J Biomech 2014; 47:1997-2005. [PMID: 24709567 DOI: 10.1016/j.jbiomech.2014.03.031] [Citation(s) in RCA: 145] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Revised: 03/18/2014] [Accepted: 03/18/2014] [Indexed: 01/06/2023]
Abstract
Scarring and tissue fibrosis represent a significant source of morbidity in the United States. Despite considerable research focused on elucidating the mechanisms underlying cutaneous scar formation, effective clinical therapies are still in the early stages of development. A thorough understanding of the various signaling pathways involved is essential to formulate strategies to combat fibrosis and scarring. While initial efforts focused primarily on the biochemical mechanisms involved in scar formation, more recent research has revealed a central role for mechanical forces in modulating these pathways. Mechanotransduction, which refers to the mechanisms by which mechanical forces are converted to biochemical stimuli, has been closely linked to inflammation and fibrosis and is believed to play a critical role in scarring. This review provides an overview of our current understanding of the mechanisms underlying scar formation, with an emphasis on the relationship between mechanotransduction pathways and their therapeutic implications.
Collapse
Affiliation(s)
- Dominik Duscher
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Zeshaan N Maan
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Victor W Wong
- Department of Plastic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Robert C Rennert
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Michael Januszyk
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Melanie Rodrigues
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Michael Hu
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Arnetha J Whitmore
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Alexander J Whittam
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Michael T Longaker
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Geoffrey C Gurtner
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
32
|
Cyr61 induces the expression of monocyte chemoattractant protein-1 via the integrin ανβ3, FAK, PI3K/Akt, and NF-κB pathways in retinal vascular endothelial cells. Cell Signal 2014; 26:133-40. [DOI: 10.1016/j.cellsig.2013.08.026] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2013] [Revised: 08/04/2013] [Accepted: 08/27/2013] [Indexed: 11/23/2022]
|
33
|
Mhanna R, Öztürk E, Schlink P, Zenobi-Wong M. Probing the microenvironmental conditions for induction of superficial zone protein expression. Osteoarthritis Cartilage 2013; 21:1924-32. [PMID: 23978656 DOI: 10.1016/j.joca.2013.08.017] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2013] [Revised: 08/09/2013] [Accepted: 08/14/2013] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To determine the in vitro conditions which promote expression of superficial zone protein (SZP). METHODS Chondrocytes from 6-month-old calves were expanded in monolayer culture and the expression of SZP in alginate bead and monolayer culture was quantified with quantitative real time-polymerase chain reaction (qRT-PCR) and immunostaining. The effect of oxygen tension on SZP expression was determined by qRT-PRC analysis of cells cultured in two dimension (2D) and three dimension (3D) under hypoxic (1% pO2) or normoxic (21% pO2) conditions. Finally, to examine the effect of cyclic tensile strain on expression of SZP in 2D and 3D cultures, chondrocytes encapsulated in alginate beams or seeded on type I collagen coated polydimethylsiloxane (PDMS) chambers were subjected to 5% strain at 1 Hz, 2 h/day for 4 days or 2 h at the fourth day of culture and mRNA levels were quantified. RESULTS Bovine chondrocytes in monolayer showed a drastic decrease in SZP expression, similar in trend to the commonly reported downregulation of type II collagen (Col2). Chondrocytes embedded in alginate beads for 4 days re-expressed SZP but not Col2. SZP expression was higher under normoxic conditions whereas Col2 was upregulated only in alginate beads under hypoxic conditions. Cyclic mechanical strain showed a tendency to upregulate mRNA levels of SZP. CONCLUSIONS A microenvironment encompassing a soft encapsulation material and 21% oxygen is sufficient for fibroblastic chondrocytes to re-express SZP. These results serve as a guideline for the design of stratified engineered articular cartilage and suggest that microenvironmental cues (oxygen tension level) strongly influence the pattern of SZP expression in vivo.
Collapse
Affiliation(s)
- R Mhanna
- Cartilage Engineering + Regeneration Laboratory, ETHZ, Schafmattstrasse 22, 8093 Zürich, Switzerland
| | | | | | | |
Collapse
|
34
|
Abstract
Increased endothelial permeability and reduction of alveolar liquid clearance capacity are two leading pathogenic mechanisms of pulmonary edema, which is a major complication of acute lung injury, severe pneumonia, and acute respiratory distress syndrome, the pathologies characterized by unacceptably high rates of morbidity and mortality. Besides the success in protective ventilation strategies, no efficient pharmacological approaches exist to treat this devastating condition. Understanding of fundamental mechanisms involved in regulation of endothelial permeability is essential for development of barrier protective therapeutic strategies. Ongoing studies characterized specific barrier protective mechanisms and identified intracellular targets directly involved in regulation of endothelial permeability. Growing evidence suggests that, although each protective agonist triggers a unique pattern of signaling pathways, selected common mechanisms contributing to endothelial barrier protection may be shared by different barrier protective agents. Therefore, understanding of basic barrier protective mechanisms in pulmonary endothelium is essential for selection of optimal treatment of pulmonary edema of different etiology. This article focuses on mechanisms of lung vascular permeability, reviews major intracellular signaling cascades involved in endothelial monolayer barrier preservation and summarizes a current knowledge regarding recently identified compounds which either reduce pulmonary endothelial barrier disruption and hyperpermeability, or reverse preexisting lung vascular barrier compromise induced by pathologic insults.
Collapse
Affiliation(s)
- Konstantin G Birukov
- Lung Injury Center, Section of Pulmonary and Critical Care, Department of Medicine, University of Chicago, Chicago, Illinois, USA.
| | | | | |
Collapse
|
35
|
Ito M, Arakawa T, Okayama M, Shitara A, Mizoguchi I, Takuma T. Gravity loading induces adenosine triphosphate release and phosphorylation of extracellular signal-regulated kinases in human periodontal ligament cells. ACTA ACUST UNITED AC 2013; 5:266-74. [PMID: 23798356 DOI: 10.1111/jicd.12049] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Accepted: 02/23/2013] [Indexed: 01/24/2023]
Abstract
AIM The periodontal ligament (PDL) receives mechanical stress (MS) from dental occlusion or orthodontic tooth movement. Mechanical stress is thought to be a trigger for remodeling of the PDL and alveolar bone, although its signaling mechanism is still unclear. So we investigated the effect of MS on adenosine triphosphate (ATP) release and extracellular signal-regulated kinases (ERK) phosphorylation in PDL cells. METHODS Mechanical stress was applied to human PDL cells as centrifugation-mediated gravity loading. Apyrase, Ca(2+)-free medium and purinergic receptor agonists and antagonists were utilized to analyze the contribution of purinergic receptors to ERK phosphorylation. RESULTS Gravity loading and ATP increased ERK phosphorylation by 5 and 2.5 times, respectively. Gravity loading induced ATP release from PDL cells by tenfold. Apyrase and suramin diminished ERK phosphorylation induced by both gravity loading and ATP. Under Ca(2+)-free conditions the phosphorylation by gravity loading was partially decreased, whereas ATP-induced phosphorylation was unaffected. Receptors P2Y4 and P2Y6 were prominently expressed in the PDL cells. CONCLUSION Gravity loading induced ATP release and ERK phosphorylation in PDL fibroblasts, and ATP signaling via P2Y receptors was partially involved in this phosphorylation, which in turn would enhance gene expression for the remodeling of PDL tissue during orthodontic tooth movement.
Collapse
Affiliation(s)
- Mai Ito
- Department of Biochemistry, School of Dentistry, Health Sciences University of Hokkaido, Hokkaido, Japan; Department of Orthodontics, School of Dentistry, Health Sciences University of Hokkaido, Hokkaido, Japan
| | | | | | | | | | | |
Collapse
|
36
|
Rustad KC, Wong VW, Gurtner GC. The role of focal adhesion complexes in fibroblast mechanotransduction during scar formation. Differentiation 2013; 86:87-91. [PMID: 23623400 DOI: 10.1016/j.diff.2013.02.003] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2013] [Accepted: 02/14/2013] [Indexed: 11/17/2022]
Abstract
Historically, great efforts have been made to elucidate the biochemical pathways that direct the complex process of wound healing; however only recently has there been recognition of the importance that mechanical signals play in the process of tissue repair and scar formation. The body's physiologic response to injury involves a dynamic interplay between mechanical forces and biochemical cues which directs a cascade of signals leading ultimately to the formation of fibrotic scar. Fibroblasts are a highly mechanosensitive cell type and are also largely responsible for the generation of the fibrotic matrix during scar formation and are thus a critical player in the process of mechanotransduction during tissue repair. Mechanotransduction is initiated at the interface between the cell membrane and the extracellular matrix where mechanical signals are first translated into a biochemical response. Focal adhesions are dynamic multi-protein complexes through which the extracellular matrix links to the intracellular cytoskeleton. These focal adhesion complexes play an integral role in the propagation of this initial mechanical cue into an extensive network of biochemical signals leading to widespread downstream effects including the influx of inflammatory cells, stimulation of angiogenesis, keratinocyte migration, fibroblast proliferation and collagen synthesis. Increasing evidence has demonstrated the importance of the biomechanical milieu in healing wounds and suggests that an integrated approach to the discovery of targets to decrease scar formation may prove more clinically efficacious than previous purely biochemical strategies.
Collapse
Affiliation(s)
- Kristine C Rustad
- Department of Surgery, Stanford University, Stanford, California, USA
| | | | | |
Collapse
|
37
|
Arya N, Arora A, Vasu KS, Sood AK, Katti DS. Combination of single walled carbon nanotubes/graphene oxide with paclitaxel: a reactive oxygen species mediated synergism for treatment of lung cancer. NANOSCALE 2013; 5:2818-29. [PMID: 23443459 DOI: 10.1039/c3nr33190c] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Heterogeneity in tumors has led to the development of combination therapies that enable enhanced cell death. Previously explored combination therapies mostly involved the use of bioactive molecules. In this work, we explored a non-conventional strategy of using carbon nanostructures (CNs) [single walled carbon nanotube (SWNT) and graphene oxide (GO)] for potentiating the efficacy of a bioactive molecule [paclitaxel (Tx)] for the treatment of lung cancer. The results demonstrated enhanced cell death following combination treatment of SWNT/GO and Tx indicating a synergistic effect. In addition, synergism was abrogated in the presence of an anti-oxidant, N-acetyl cysteine (NAC), and was therefore shown to be reactive oxygen species (ROS) dependent. It was further demonstrated using bromodeoxyuridine (BrdU) incorporation assay that treatment with CNs was associated with enhanced mitogen associated protein kinase (MAPK) activation that was ROS mediated. Hence, these results for the first time demonstrated the potential of SWNT/GO as co-therapeutic agents with Tx for the treatment of lung cancer.
Collapse
Affiliation(s)
- Neha Arya
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology - Kanpur, Kanpur-208016, Uttar Pradesh, India
| | | | | | | | | |
Collapse
|
38
|
Hata M, Naruse K, Ozawa S, Kobayashi Y, Nakamura N, Kojima N, Omi M, Katanosaka Y, Nishikawa T, Naruse K, Tanaka Y, Matsubara T. Mechanical stretch increases the proliferation while inhibiting the osteogenic differentiation in dental pulp stem cells. Tissue Eng Part A 2012; 19:625-33. [PMID: 23153222 DOI: 10.1089/ten.tea.2012.0099] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Dental pulp stem cells (DPSCs), which can differentiate into several types of cells, are subjected to mechanical stress by jaw movement and occlusal forces. In this study, we evaluated how the uniaxial mechanical stretch influences proliferation and differentiation of DPSCs. DPSCs were isolated and cultured from male Sprague-Dawley rats. Cultured DPSCs were identified by surface markers and the differentiation capabilities as adipocytes or osteoblasts. To examine the response to mechanical stress, uniaxial stretch was exposed to cultured DPSCs. We evaluated the impact of stretch on the intracellular signaling, proliferation, osteogenic differentiation, and gene expressions of DPSCs. Stretch increased the phosphorylation of Akt, ERK1/2, and p38 MAP kinase as well as the proliferation of DPSCs. The stretch-induced proliferation of DPSCs was abolished by the inhibition of the ERK pathway. On the other hand, stretch significantly decreased the osteogenic differentiation of DPSCs, but did not affect the adipogenic differentiation. We also confirmed mRNA expressions of osteocalcin and osteopontin were significantly suppressed by stretch. In conclusion, uniaxial stretch increased the proliferation of DPSCs, while suppressing osteogenic differentiation. These results suggest a crucial role of mechanical stretch in the preservation of DPSCs in dentin. Furthermore, mechanical stretch may be a useful tool for increasing the quantity of DPSCs in vitro for regenerative medicine.
Collapse
Affiliation(s)
- Masaki Hata
- Department of Removable Prosthodontics, School of Dentistry, Aichi-Gakuin University, Nagoya, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
High and low mammographic density human breast tissues maintain histological differential in murine tissue engineering chambers. Breast Cancer Res Treat 2012; 135:177-87. [DOI: 10.1007/s10549-012-2128-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2012] [Accepted: 06/04/2012] [Indexed: 01/11/2023]
|
40
|
Richards JM, Farrar EJ, Kornreich BG, Moïse NS, Butcher JT. The mechanobiology of mitral valve function, degeneration, and repair. J Vet Cardiol 2012; 14:47-58. [PMID: 22366572 PMCID: PMC3586284 DOI: 10.1016/j.jvc.2012.01.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2011] [Revised: 12/24/2011] [Accepted: 01/17/2012] [Indexed: 12/28/2022]
Abstract
In degenerative valve disease, the highly organized mitral valve leaflet matrix stratification is progressively destroyed and replaced with proteoglycan rich, mechanically inadequate tissue. This is driven by the actions of originally quiescent valve interstitial cells that become active contractile and migratory myofibroblasts. While treatment for myxomatous mitral valve disease in humans ranges from repair to total replacement, therapies in dogs focus on treating the consequences of the resulting mitral regurgitation. The fundamental gap in our understanding is how the resident valve cells respond to altered mechanical signals to drive tissue remodeling. Despite the pathological similarities and high clinical occurrence, surprisingly little mechanistic insight has been gleaned from the dog. This review presents what is known about mitral valve mechanobiology from clinical, in vivo, and in vitro data. There are a number of experimental strategies already available to pursue this significant opportunity, but success requires the collaboration between veterinary clinicians, scientists, and engineers.
Collapse
Affiliation(s)
| | - Emily J. Farrar
- Department of Biomedical Engineering, Cornell University, Ithaca NY, USA
| | - Bruce G. Kornreich
- Department of Clinical Sciences, Section of Cardiology, College of Veterinary Medicine, Cornell University, Ithaca NY, USA
| | - N. Sydney Moïse
- Department of Clinical Sciences, Section of Cardiology, College of Veterinary Medicine, Cornell University, Ithaca NY, USA
| | | |
Collapse
|
41
|
Abstract
Benign and malignant fibroproliferative disorders (FPDs) include idiopathic pulmonary fibrosis, hepatic cirrhosis, myelofibrosis, systemic sclerosis, Dupuytren's contracture, hypertrophic scars, and keloids. They are characterized by excessive connective tissue accumulation and slow but continuous tissue contraction that lead to progressive deterioration in the normal structure and function of affected organs. In recent years, research in diverse fields has increasingly highlighted the potential role of mechanobiology in the molecular mechanisms of fibroproliferation. Mechanobiology, the heart of which is mechanotransduction, is the process whereby cells sense mechanical forces and transduce them, thereby changing the intracellular biochemistry and gene expression. Understanding mechanosignaling may provide new insights into the convergent roles played by interrelated molecules and overlapping signaling pathways during the inflammatory, proliferative, and fibrotic cellular activities that are the hallmarks of fibroproliferation. The main cellular players in FPDs are fibroblasts and myofibroblasts. Consequently, this article discusses integrins and the roles they play in cellular-extracellular matrix interactions. Also described are the signaling pathways that are known to participate in mechanosignaling: these include the transforming growth factor-β/Smad, mitogen-activated protein kinase, RhoA/ROCK, Wnt/β-catenin, and tumor necrosis factor-α/nuclear factor kappa-light-chain-enhancer of activated B cells pathways. Also outlined is the progress in our understanding of the cellular-extracellular matrix interactions that are associated with fibroproliferative mechanosignaling through matricellular proteins. The tensegrity and tensional homeostasis models are also discussed. A better understanding of the mechanosignaling pathways in the FPD microenvironment will almost certainly lead to the development of novel interventions that can prevent, reduce, or even reverse FPD formation and/or progression.
Collapse
Affiliation(s)
- Chenyu Huang
- Department of Plastic, Reconstructive and Aesthetic Surgery, Nippon Medical School, Tokyo, Japan
| | | |
Collapse
|
42
|
Provenzano PP, Keely PJ. Mechanical signaling through the cytoskeleton regulates cell proliferation by coordinated focal adhesion and Rho GTPase signaling. J Cell Sci 2011; 124:1195-205. [PMID: 21444750 DOI: 10.1242/jcs.067009] [Citation(s) in RCA: 387] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The notion that cell shape and spreading can regulate cell proliferation has evolved over several years, but only recently has this been linked to forces from within and upon the cell. This emerging area of mechanical signaling is proving to be wide-spread and important for all cell types. The microenvironment that surrounds cells provides a complex spectrum of different, simultaneously active, biochemical, structural and mechanical stimuli. In this milieu, cells probe the stiffness of their microenvironment by pulling on the extracellular matrix (ECM) and/or adjacent cells. This process is dependent on transcellular cell-ECM or cell-cell adhesions, as well as cell contractility mediated by Rho GTPases, to provide a functional linkage through which forces are transmitted through the cytoskeleton by intracellular force-generating proteins. This Commentary covers recent advances in the underlying mechanisms that control cell proliferation by mechanical signaling, with an emphasis on the role of 3D microenvironments and in vivo extracellular matrices. Moreover, as there is much recent interest in the tumor-stromal interaction, we will pay particular attention to exciting new data describing the role of mechanical signaling in the progression of breast cancer.
Collapse
Affiliation(s)
- Paolo P Provenzano
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.
| | | |
Collapse
|
43
|
Dalla Costa AP, Clemente CFMZ, Carvalho HF, Carvalheira JB, Nadruz W, Franchini KG. FAK mediates the activation of cardiac fibroblasts induced by mechanical stress through regulation of the mTOR complex. Cardiovasc Res 2010; 86:421-431. [PMID: 20038548 DOI: 10.1093/cvr/cvp416] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
AIMS Cardiac fibroblasts are activated by mechanical stress, but the underlying mechanisms involved remain poorly understood. In this study, we investigated whether focal adhesion kinase (FAK) plays a role in the activation of cardiac fibroblasts in response to cyclic stretch. METHODS AND RESULTS Neonatal (NF-P3/80--third passage, 80% confluence) and adult (AF-P1/80--first passage, 80% confluence) rat cardiac fibroblasts were exposed to cyclic stretch (biaxial, 1 Hz), which enhanced FAK phosphorylation at Tyr397. Proliferation (anti-5-bromo-2'-deoxyuridine and anti-Ki67 nuclear labelling), differentiation into myofibroblasts (expression of alpha-smooth muscle actin--alpha-SMA), and the activity of matrix metalloproteinase-2 were equally enhanced in stretched NF-P3/80 and AF-P1/80. Treatment with the integrin inhibitor RGD peptide impaired FAK phosphorylation and increased apoptosis (TUNEL) in non-stretched and stretched NF-P3/80, whereas FAK silencing induced by small interfering RNA modestly enhanced apoptosis only in stretched cells. RGD peptide or FAK silencing suppressed the activation of NF-P3/80 invoked by cyclic stretch. In addition, NF-P3/80 depleted of FAK were defective in AKT Ser473, TSC-2 Thr1462, and S6 kinase Thr389 phosphorylation induced by cyclic stretch. The activation of NF-P3/80 invoked by cyclic stretch was prevented by pre-treatment with the mammalian target of rapamycin (mTOR) inhibitor rapamycin, whereas supplementation with the amino acid, leucine, activated S6K and rescued the stretch-induced activation of NF-P3/80 depleted of FAK. CONCLUSIONS These findings demonstrate a critical role for the mTOR complex, downstream from FAK, in mediating the activation of cardiac fibroblasts in response to mechanical stress.
Collapse
Affiliation(s)
- Ana Paula Dalla Costa
- Department of Internal Medicine, School of Medicine and Department of Cell Biology, State University of Campinas, Campinas, SP, Brazil
| | | | | | | | | | | |
Collapse
|
44
|
Stops A, Heraty K, Browne M, O'Brien F, McHugh P. A prediction of cell differentiation and proliferation within a collagen–glycosaminoglycan scaffold subjected to mechanical strain and perfusive fluid flow. J Biomech 2010; 43:618-26. [DOI: 10.1016/j.jbiomech.2009.10.037] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2009] [Revised: 10/19/2009] [Accepted: 10/19/2009] [Indexed: 11/25/2022]
|
45
|
Applying controlled non-uniform deformation for in vitro studies of cell mechanobiology. Biomech Model Mechanobiol 2010; 9:329-44. [PMID: 20169395 DOI: 10.1007/s10237-009-0179-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2009] [Accepted: 11/05/2009] [Indexed: 10/19/2022]
Abstract
Cells within connective tissues routinely experience a wide range of non-uniform mechanical loads that regulate many cell behaviors. In this study, we developed an experimental system to produce complex strain patterns for the study of strain magnitude, anisotropy, and gradient effects on cells in culture. A standard equibiaxial cell stretching system was modified by affixing glass coverslips (5, 10, or 15 mm diameter) to the center of 35 mm diameter flexible-bottomed culture wells. Ring inserts were utilized to limit applied strain to different levels in each individual well at a given vacuum pressure thus enabling parallel experiments at different strain levels. Deformation fields were measured using high-density mapping for up to 6% applied strain. The addition of the rigid inclusion creates strong circumferential and radial strain gradients, with a continuous range of stretch anisotropy ranging from strip biaxial to equibiaxial strain and radial strains up to 24% near the inclusion. Dermal fibroblasts seeded within our 2D system (5 mm inclusions; 2% applied strain for 2 days at 0.2 Hz) demonstrated the characteristic orientation perpendicular to the direction of principal strain. Dermal fibroblasts seeded within fibrin gels (5 mm inclusions; 6% applied strain for 8 days at 0.2 Hz) oriented themselves similarly and compacted their surrounding matrix to an increasing extent with local strain magnitude. This study verifies how inhomogeneous strain fields can be produced in a tunable and simply constructed system and demonstrates the potential utility for studying gradients with a continuous spectrum of strain magnitudes and anisotropies.
Collapse
|
46
|
Koshman YE, Piano MR, Russell B, Schwertz DW. Signaling responses after exposure to 5 alpha-dihydrotestosterone or 17 beta-estradiol in norepinephrine-induced hypertrophy of neonatal rat ventricular myocytes. J Appl Physiol (1985) 2009; 108:686-96. [PMID: 20044473 DOI: 10.1152/japplphysiol.00994.2009] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Androgens appear to enhance, whereas estrogens mitigate, cardiac hypertrophy. However, signaling pathways in cells for short (3 min) and longer term (48 h) treatment with 17beta-estradiol (E2) or 5 alpha-dihydrotestosterone (DHT) are understudied. We compared the effect of adrenergic stimulation by norepinephrine (NE; 1 microM) alone or in combination with DHT (10 nM) or E2 (10 nM) treatment in neonatal rat ventricular myocytes (NRVMs) by cell area, protein synthesis, sarcomeric structure, gene expression, phosphorylation of extracellular signal-regulated (ERK), and focal adhesion kinases (FAK), and phospho-FAK nuclear localization. NE alone elicited the expected hypertrophy and strong sarcomeric organization, and DHT alone gave a similar but more modest response, whereas E2 did not alter cell size. Effects of NE dominated when used with either E2 or DHT with all combinations. Both sex hormones alone rapidly activated FAK but not ERK. Long-term or brief exposure to E2 attenuated NE-induced FAK phosphorylation, whereas DHT had no effect. Neither hormone altered NE-elicited ERK activation. Longer term exposure to E2 alone reduced FAK phosphorylation and reduced nuclear phospho-FAK, whereas its elevation was seen in the presence of NE with both sex hormones. The mitigating effects of E2 on the NE-elicited increase in cell size and the hypertrophic effect of DHT in NRVMs are in accordance with results observed in whole animal models. This is the first report of rapid, nongenomic sex hormone signaling via FAK activation and altered FAK trafficking to the nucleus in heart cells.
Collapse
Affiliation(s)
- Yevgeniya E Koshman
- Department of Physiology and Biophysics, University of Illinois at Chicago, MC 901, 835 South Wolcott Ave., Chicago, IL 60612-7342, USA
| | | | | | | |
Collapse
|
47
|
Provenzano PP, Inman DR, Eliceiri KW, Keely PJ. Matrix density-induced mechanoregulation of breast cell phenotype, signaling and gene expression through a FAK-ERK linkage. Oncogene 2009; 28:4326-43. [PMID: 19826415 PMCID: PMC2795025 DOI: 10.1038/onc.2009.299] [Citation(s) in RCA: 516] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2009] [Revised: 07/28/2009] [Accepted: 08/25/2009] [Indexed: 11/26/2022]
Abstract
Mammographically dense breast tissue is one of the greatest risk factors for developing breast carcinoma, yet the associated molecular mechanisms remain largely unknown. Importantly, regions of high breast density are associated with increased stromal collagen and epithelial cell content. We set out to determine whether increased collagen-matrix density, in the absence of stromal cells, was sufficient to promote proliferation and invasion characteristic of a malignant phenotype in non-transformed mammary epithelial cells. We demonstrate that increased collagen-matrix density increases matrix stiffness to promote an invasive phenotype. High matrix stiffness resulted in increased formation of activated three-dimensional (3D)-matrix adhesions and a chronically elevated outside-in/inside-out focal adhesion (FA) kinase (FAK)-Rho signaling loop, which was necessary to generate and maintain the invasive phenotype. Moreover, this signaling network resulted in hyperactivation of the Ras-mitogen-activated protein kinase (MAPK) pathway, which promoted growth of mammary epithelial cells in vitro and in vivo and activated a clinically relevant proliferation signature that predicts patient outcome. Hence, the current data provide compelling evidence for the importance of the mechanical features of the microenvironment, and suggest that mechanotransduction in these cells occurs through a FAK-Rho-ERK signaling network with extracellular signal-regulated kinase (ERK) as a bottleneck through which much of the response to mechanical stimuli is regulated. As such, we propose that increased matrix stiffness explains part of the mechanism behind increased epithelial proliferation and cancer risk in human patients with high breast tissue density.
Collapse
Affiliation(s)
- Paolo P. Provenzano
- Department of Pharmacology, University of Wisconsin, Madison, WI
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI
- Laboratory for Optical and Computational Instrumentation, University of Wisconsin, Madison, WI
- University of Wisconsin Paul P. Carbone Comprehensive Cancer Center, University of Wisconsin, Madison, WI
| | - David R. Inman
- Department of Pharmacology, University of Wisconsin, Madison, WI
- University of Wisconsin Paul P. Carbone Comprehensive Cancer Center, University of Wisconsin, Madison, WI
| | - Kevin W. Eliceiri
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI
- Laboratory for Optical and Computational Instrumentation, University of Wisconsin, Madison, WI
| | - Patricia J. Keely
- Department of Pharmacology, University of Wisconsin, Madison, WI
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI
- Laboratory for Optical and Computational Instrumentation, University of Wisconsin, Madison, WI
- University of Wisconsin Paul P. Carbone Comprehensive Cancer Center, University of Wisconsin, Madison, WI
| |
Collapse
|
48
|
Kiely PA, Baillie GS, Barrett R, Buckley DA, Adams DR, Houslay MD, O'Connor R. Phosphorylation of RACK1 on tyrosine 52 by c-Abl is required for insulin-like growth factor I-mediated regulation of focal adhesion kinase. J Biol Chem 2009; 284:20263-74. [PMID: 19423701 DOI: 10.1074/jbc.m109.017640] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Focal Adhesion Kinase (FAK) activity is controlled by growth factors and adhesion signals in tumor cells. The scaffolding protein RACK1 (receptor for activated C kinases) integrates insulin-like growth factor I (IGF-I) and integrin signaling, but whether RACK1 is required for FAK function is unknown. Here we show that association of FAK with RACK1 is required for both FAK phosphorylation and dephosphorylation in response to IGF-I. Suppression of RACK1 by small interfering RNA ablates FAK phosphorylation and reduces cell adhesion, cell spreading, and clonogenic growth. Peptide array and mutagenesis studies localize the FAK binding interface to blades I-III of the RACK1 beta-propeller and specifically identify a set of basic and hydrophobic amino acids (Arg-47, Tyr-52, Arg-57, Arg-60, Phe-65, Lys-127, and Lys-130) as key determinants for association with FAK. Mutation of tyrosine 52 alone is sufficient to disrupt interaction of RACK1 with FAK in cells where endogenous RACK1 is suppressed by small interfering RNA. Cells expressing a Y52F mutant RACK1 are impaired in adhesion, growth, and foci formation. Comparative analyses of homology models and crystal structures for RACK1 orthologues suggest a role for Tyr-52 as a site for phosphorylation that induces conformational change in RACK1, switching the protein into a FAK binding state. Tyrosine 52 is further shown to be phosphorylated by c-Abl kinase, and the c-Abl inhibitor STI571 disrupts FAK interaction with RACK1. We conclude that FAK association with RACK1 is regulated by phosphorylation of Tyr-52. Our data reveal a novel mechanism whereby IGF-I and c-Abl control RACK1 association with FAK to facilitate adhesion signaling.
Collapse
Affiliation(s)
- Patrick A Kiely
- From the Cell Biology Laboratory, Department of Biochemistry, BioSciences Institute, University College Cork, Cork, Ireland
| | | | | | | | | | | | | |
Collapse
|
49
|
Birukova AA, Rios A, Birukov KG. Long-term cyclic stretch controls pulmonary endothelial permeability at translational and post-translational levels. Exp Cell Res 2008; 314:3466-77. [PMID: 18824167 PMCID: PMC2638120 DOI: 10.1016/j.yexcr.2008.09.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2008] [Revised: 08/22/2008] [Accepted: 09/10/2008] [Indexed: 11/17/2022]
Abstract
We have previously described differential effects of physiologic (5%) and pathologic (18%) cyclic stretch (CS) on agonist-induced pulmonary endothelial permeability. This study examined acute and chronic effects of CS on agonist-induced intracellular signaling and cell morphology in the human lung macro- and microvascular endothelial cell (EC) monolayers. Endothelial permeability was assessed by analysis of morphological changes, parameters of cell contraction and measurements of transendothelial electrical resistance. Exposure of both microvascular and macrovascular EC to 18% CS for 2-96 h increased thrombin-induced permeability and monolayer disruption. Interestingly, the ability to promote thrombin responses was present in EC cultures exposed to 48-96 h of CS even after replating onto non-elastic substrates. In turn, physiologic CS preconditioning (72 h) attenuated thrombin-induced paracellular gap formation and MLC phosphorylation in replated EC cultures. Long-term preconditioning at 18% CS (72 h) increased the content of signaling and contractile proteins including Rho GTPase, MLC, MLC kinase, ZIP kinase, PAR1, caldesmon and HSP27 in the pulmonary microvascular and macrovascular cells. We conclude that short term CS regulates EC permeability via modulation of agonist-induced signaling, whereas long-term CS controls endothelial barrier at both post-translational level and via magnitude-dependent regulation of pulmonary EC phenotype, signaling and contractile protein expression.
Collapse
Affiliation(s)
- Anna A Birukova
- Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | | | | |
Collapse
|
50
|
HU X, CHENG D, ZHANG Y, JIANG F, YANG D, DING Y. Low-level mechanical strain induces extracellular signal-regulated kinase 1/2 activation in alveolar epithelial cells. Respirology 2008; 13:781-7. [DOI: 10.1111/j.1440-1843.2008.01347.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|