51
|
Cadena-Suárez AR, Hernández-Hernández HA, Alvarado-Vásquez N, Rangel-Escareño C, Sommer B, Negrete-García MC. Role of MicroRNAs in Signaling Pathways Associated with the Pathogenesis of Idiopathic Pulmonary Fibrosis: A Focus on Epithelial-Mesenchymal Transition. Int J Mol Sci 2022; 23:ijms23126613. [PMID: 35743055 PMCID: PMC9224458 DOI: 10.3390/ijms23126613] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/05/2022] [Accepted: 06/08/2022] [Indexed: 12/15/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic and progressive disease with high mortality and unclear etiology. Previous evidence supports that the origin of this disease is associated with epigenetic alterations, age, and environmental factors. IPF initiates with chronic epithelial lung injuries, followed by basal membrane destruction, which promotes the activation of myofibroblasts and excessive synthesis of extracellular matrix (ECM) proteins, as well as epithelial-mesenchymal transition (EMT). Due to miRNAs’ role as regulators of apoptosis, proliferation, differentiation, and cell-cell interaction processes, some studies have involved miRNAs in the biogenesis and progression of IPF. In this context, the analysis and discussion of the probable association of miRNAs with the signaling pathways involved in the development of IPF would improve our knowledge of the associated molecular mechanisms, thereby facilitating its evaluation as a therapeutic target for this severe lung disease. In this work, the most recent publications evaluating the role of miRNAs as regulators or activators of signal pathways associated with the pathogenesis of IPF were analyzed. The search in Pubmed was made using the following terms: “miRNAs and idiopathic pulmonary fibrosis (IPF)”; “miRNAs and IPF and signaling pathways (SP)”; and “miRNAs and IPF and SP and IPF pathogenesis”. Additionally, we focus mainly on those works where the signaling pathways involved with EMT, fibroblast differentiation, and synthesis of ECM components were assessed. Finally, the importance and significance of miRNAs as potential therapeutic or diagnostic tools for the treatment of IPF are discussed.
Collapse
Affiliation(s)
- Ana Ruth Cadena-Suárez
- Laboratorio de Biología Molecular, Instituto Nacional de Enfermedades Respiratorias (INER) “Ismael Cosío Villegas”, Calz. Tlalpan 4502, Col. Sección XVI, Mexico City 14080, Mexico; (A.R.C.-S.); (H.A.H.-H.)
| | - Hilda Arely Hernández-Hernández
- Laboratorio de Biología Molecular, Instituto Nacional de Enfermedades Respiratorias (INER) “Ismael Cosío Villegas”, Calz. Tlalpan 4502, Col. Sección XVI, Mexico City 14080, Mexico; (A.R.C.-S.); (H.A.H.-H.)
| | - Noé Alvarado-Vásquez
- Departamento de Bioquímica, Instituto Nacional de Enfermedades Respiratorias (INER) “Ismael Cosío Villegas”, Calz. Tlalpan 4502, Col. Sección XVI, Mexico City 14080, Mexico;
| | - Claudia Rangel-Escareño
- Departamento de Genomica Computacional, Instituto Nacional de Medicina Genómica, Periférico Sur 4809, Col. Arenal Tepepan, Mexico City 14610, Mexico;
- Escuela de Ingenieria y Ciencias, Tecnológico de Monterrey, Epigmenio González 500, San Pablo 76130, Mexico
| | - Bettina Sommer
- Departamento de Investigación en Hiperreactividad Bronquial, Instituto Nacional de Enfermedades Respiratorias (INER) “Ismael Cosío Villegas”, Calz. Tlalpan 4502, Col. Sección XVI, Mexico City 14080, Mexico;
| | - María Cristina Negrete-García
- Laboratorio de Biología Molecular, Instituto Nacional de Enfermedades Respiratorias (INER) “Ismael Cosío Villegas”, Calz. Tlalpan 4502, Col. Sección XVI, Mexico City 14080, Mexico; (A.R.C.-S.); (H.A.H.-H.)
- Correspondence:
| |
Collapse
|
52
|
Si H, Yin C, Wang W, Davies P, Sanchez E, Suntravat M, Zawieja D, Cromer W. Effect of the snake venom component crotamine on lymphatic endothelial cell responses and lymph transport. Microcirculation 2022; 30:e12775. [PMID: 35689804 PMCID: PMC9850291 DOI: 10.1111/micc.12775] [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: 04/01/2022] [Revised: 05/16/2022] [Accepted: 06/06/2022] [Indexed: 01/21/2023]
Abstract
OBJECTIVE The pathology of snake envenomation is closely tied to the severity of edema in the tissue surrounding the area of the bite. Elucidating the mechanisms that promote the development of such severe edema is critical to a better understanding of how to treat this life-threatening injury. We focused on one of the most abundant venom components in North American viper venom, crotamine, and the effects it has on the cells and function of the lymphatic system. METHODS We used RT-PCR to identify the location and relative abundance of crotamine's cellular targets (Kvα channels) within the tissues and cells of the lymphatic system. We used calcium flux, nitrate production, and cell morphometry to determine the effects of crotamine on lymphatic endothelial cells. We used tracer transport, node morphometry, and node deposition to determine the effects of crotamine on lymph transport in vivo. RESULTS We found that genes that encode targets of crotamine are highly present in lymphatic tissues and cells and that there is a differential distribution of those genes that correlates with phasic contractile activity. We found that crotamine potentiates calcium flux in human dermal lymphatic endothelial cells in response to stimulation with histamine and sheer stress (but not alone) and that it alters the production of nitric oxide in response to shear as well as changes the level of F-actin polymerization of those same cells. Crotamine alters lymphatic transport of large molecular weight tracers to local lymph nodes and is deposited within the node mostly in the immediate subcapsular region. CONCLUSION This evidence suggests that snake venom components may have an impact on the function of the lymphatic system. This needs to be studied in greater detail as there are numerous venom components that may have effects on aspects of the lymphatic system. This would not only provide basic information on the pathobiology of snakebite but also provide targets for improved therapeutics to treat snakebite.
Collapse
Affiliation(s)
- Hongjiang Si
- Department of Medical Physiology, Texas A&M University Health Science Center
| | - Chunhiu Yin
- Center for Translational Cancer Research, Texas A&M Institute of Biosciences and Technology
| | - Wei Wang
- Department of Medical Physiology, Texas A&M University Health Science Center
| | - Peter Davies
- Center for Translational Cancer Research, Texas A&M Institute of Biosciences and Technology
| | - Elda Sanchez
- National Natural Toxins Research Center, Texas A&M Kingsville
| | | | - David Zawieja
- Department of Medical Physiology, Texas A&M University Health Science Center
| | - Walter Cromer
- Department of Medical Physiology, Texas A&M University Health Science Center
| |
Collapse
|
53
|
Sugita S, Hozaki M, Matsui TS, Nagayama K, Deguchi S, Nakamura M. Polarized light retardation analysis allows for the evaluation of tension in individual stress fibers. Biochem Biophys Res Commun 2022; 620:49-55. [DOI: 10.1016/j.bbrc.2022.06.066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 06/21/2022] [Indexed: 11/02/2022]
|
54
|
Khoonkari M, Liang D, Kamperman M, Kruyt FAE, van Rijn P. Physics of Brain Cancer: Multiscale Alterations of Glioblastoma Cells under Extracellular Matrix Stiffening. Pharmaceutics 2022; 14:pharmaceutics14051031. [PMID: 35631616 PMCID: PMC9145282 DOI: 10.3390/pharmaceutics14051031] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 04/27/2022] [Accepted: 05/06/2022] [Indexed: 12/12/2022] Open
Abstract
The biology and physics underlying glioblastoma is not yet completely understood, resulting in the limited efficacy of current clinical therapy. Recent studies have indicated the importance of mechanical stress on the development and malignancy of cancer. Various types of mechanical stress activate adaptive tumor cell responses that include alterations in the extracellular matrix (ECM) which have an impact on tumor malignancy. In this review, we describe and discuss the current knowledge of the effects of ECM alterations and mechanical stress on GBM aggressiveness. Gradual changes in the brain ECM have been connected to the biological and physical alterations of GBM cells. For example, increased expression of several ECM components such as glycosaminoglycans (GAGs), hyaluronic acid (HA), proteoglycans and fibrous proteins result in stiffening of the brain ECM, which alters inter- and intracellular signaling activity. Several mechanosensing signaling pathways have been identified that orchestrate adaptive responses, such as Hippo/YAP, CD44, and actin skeleton signaling, which remodel the cytoskeleton and affect cellular properties such as cell–cell/ECM interactions, growth, and migration/invasion of GBM cells. In vitro, hydrogels are used as a model to mimic the stiffening of the brain ECM and reconstruct its mechanics, which we also discuss. Overall, we provide an overview of the tumor microenvironmental landscape of GBM with a focus on ECM stiffening and its associated adaptive cellular signaling pathways and their possible therapeutic exploitation.
Collapse
Affiliation(s)
- Mohammad Khoonkari
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands; (M.K.); (D.L.)
- Polymer Science, Zernike Institute for Advanced Materials, University of Groningen, Nijenborgh 4, 9747 AG Groningen, The Netherlands;
| | - Dong Liang
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands; (M.K.); (D.L.)
| | - Marleen Kamperman
- Polymer Science, Zernike Institute for Advanced Materials, University of Groningen, Nijenborgh 4, 9747 AG Groningen, The Netherlands;
| | - Frank A. E. Kruyt
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands; (M.K.); (D.L.)
- Correspondence: (F.A.E.K.); (P.v.R.)
| | - Patrick van Rijn
- Department of Biomedical Engineering-FB40, University of Groningen, University Medical Center Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands
- W.J. Kolff Institute for Biomedical Engineering and Materials Science-FB41, University of Groningen, University Medical Center Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands
- Correspondence: (F.A.E.K.); (P.v.R.)
| |
Collapse
|
55
|
Bernal R, Van Hemelryck M, Gurchenkov B, Cuvelier D. Actin Stress Fibers Response and Adaptation under Stretch. Int J Mol Sci 2022; 23:ijms23095095. [PMID: 35563485 PMCID: PMC9101353 DOI: 10.3390/ijms23095095] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 04/11/2022] [Accepted: 04/14/2022] [Indexed: 02/04/2023] Open
Abstract
One of the many effects of soft tissues under mechanical solicitation in the cellular damage produced by highly localized strain. Here, we study the response of peripheral stress fibers (SFs) to external stretch in mammalian cells, plated onto deformable micropatterned substrates. A local fluorescence analysis reveals that an adaptation response is observed at the vicinity of the focal adhesion sites (FAs) due to its mechanosensor function. The response depends on the type of mechanical stress, from a Maxwell-type material in compression to a complex scenario in extension, where a mechanotransduction and a self-healing process takes place in order to prevent the induced severing of the SF. A model is proposed to take into account the effect of the applied stretch on the mechanics of the SF, from which relevant parameters of the healing process are obtained. In contrast, the repair of the actin bundle occurs at the weak point of the SF and depends on the amount of applied strain. As a result, the SFs display strain-softening features due to the incorporation of new actin material into the bundle. In contrast, the response under compression shows a reorganization with a constant actin material suggesting a gliding process of the SFs by the myosin II motors.
Collapse
Affiliation(s)
- Roberto Bernal
- Cellular Mechanics Laboratory, Physics Department, SMAT-C, Universidad de Santiago de Chile, Santiago 9170124, Chile;
- Correspondence: (R.B.); (D.C.)
| | - Milenka Van Hemelryck
- Cellular Mechanics Laboratory, Physics Department, SMAT-C, Universidad de Santiago de Chile, Santiago 9170124, Chile;
| | - Basile Gurchenkov
- Institut du Cerveau et de la Moelle Épinière, Hôpital Pitié Salpêtrière, 47 bd de l’Hôpital, 75013 Paris, France;
| | - Damien Cuvelier
- Sorbonne Université, Faculté des Sciences et Ingénierie, UFR 926 Chemistry, 75005 Paris, France
- Institut Pierre Gilles de Gennes, Paris Sciences et Lettres Research University, 75005 Paris, France
- Institut Curie, Paris Sciences et Lettres Research University, Centre National de la Recherche Scientifique, UMR 144, 75248 Paris, France
- Correspondence: (R.B.); (D.C.)
| |
Collapse
|
56
|
Morris T, Sue E, Geniesse C, Brieher WM, Tang VW. Synaptopodin stress fiber and contractomere at the epithelial junction. J Cell Biol 2022; 221:e202011162. [PMID: 35416930 PMCID: PMC9011326 DOI: 10.1083/jcb.202011162] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 10/07/2021] [Accepted: 02/09/2022] [Indexed: 12/27/2022] Open
Abstract
The apical junction of epithelial cells can generate force to control cell geometry and perform contractile processes while maintaining barrier function and adhesion. Yet, the structural basis for force generation at the apical junction is not fully understood. Here, we describe two synaptopodin-dependent actomyosin structures that are spatially, temporally, and structurally distinct. The first structure is formed by the retrograde flow of synaptopodin initiated at the apical junction, creating a sarcomeric stress fiber that lies parallel to the apical junction. Contraction of the apical stress fiber is associated with either clustering of membrane components or shortening of junctional length. Upon junction maturation, apical stress fibers are disassembled. In mature epithelial monolayer, a motorized "contractomere" capable of "walking the junction" is formed at the junctional vertex. Actomyosin activities at the contractomere produce a compressive force evident by actin filament buckling and measurement with a new α-actinin-4 force sensor. The motility of contractomeres can adjust junctional length and change cell packing geometry during cell extrusion and intercellular movement. We propose a model of epithelial homeostasis that utilizes contractomere motility to support junction rearrangement while preserving the permeability barrier.
Collapse
Affiliation(s)
- Timothy Morris
- Department of Cell and Developmental Biology, University of Illinois, Urbana-Champaign, IL
| | - Eva Sue
- Department of Cell and Developmental Biology, University of Illinois, Urbana-Champaign, IL
| | - Caleb Geniesse
- Department of Cell and Developmental Biology, University of Illinois, Urbana-Champaign, IL
| | - William M Brieher
- Department of Cell and Developmental Biology, University of Illinois, Urbana-Champaign, IL
| | - Vivian W Tang
- Department of Cell and Developmental Biology, University of Illinois, Urbana-Champaign, IL
| |
Collapse
|
57
|
Best M, Gale ME, Wells CM. PAK-dependent regulation of actin dynamics in breast cancer cells. Int J Biochem Cell Biol 2022; 146:106207. [PMID: 35385780 PMCID: PMC9941713 DOI: 10.1016/j.biocel.2022.106207] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 03/24/2022] [Accepted: 03/29/2022] [Indexed: 11/24/2022]
Abstract
Metastatic Breast Cancer has a poor 25% survival rate and currently there are no clinical therapeutics which target metastasis. 'Migrastatics' are a new drug class which target migration pathway effector proteins in order to inhibit cancer cell invasion and metastasis. The p21-activated kinases (PAKs) are essential drivers of breast cancer cell migration and invasion through their regulation of actin cytoskeletal dynamics. Therefore, the PAKs present as attractive migrastatic candidates. Here we review how PAKs regulate distinct aspects of breast cancer actin dynamics focussing on cytoskeletal reorganisation, cell:matrix adhesion, actomyosin contractility and degradative invasion. Lastly, we discuss the introduction of PAK migrastatics into the well-honed breast cancer clinical pipeline.
Collapse
Affiliation(s)
- Marianne Best
- School of Cancer and Pharmaceutical Sciences, Kings College London, London UK.
| | - Madeline E. Gale
- School of Cancer and Pharmaceutical Sciences, Kings College London, London UK,North West Thames Regional Genetics Service, Northwick Park Hospital, London UK
| | - Claire M. Wells
- School of Cancer and Pharmaceutical Sciences, Kings College London, London UK,Corresponding author.
| |
Collapse
|
58
|
Che H, Selig M, Rolauffs B. Micro-patterned cell populations as advanced pharmaceutical drugs with precise functional control. Adv Drug Deliv Rev 2022; 184:114169. [PMID: 35217114 DOI: 10.1016/j.addr.2022.114169] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 02/14/2022] [Accepted: 02/15/2022] [Indexed: 11/29/2022]
Abstract
Human cells are both advanced pharmaceutical drugs and 'drug deliverers'. However, functional control prior to or after cell implantation remains challenging. Micro-patterning cells through geometrically defined adhesion sites allows controlling morphogenesis, polarity, cellular mechanics, proliferation, migration, differentiation, stemness, cell-cell interactions, collective cell behavior, and likely immuno-modulatory properties. Consequently, generating micro-patterned therapeutic cells is a promising idea that has not yet been realized and few if any steps have been undertaken in this direction. This review highlights potential therapeutic applications, summarizes comprehensively the many cell functions that have been successfully controlled through micro-patterning, details the established micro-pattern designs, introduces the available fabrication technologies to the non-specialized reader, and suggests a quality evaluation score. Such a broad review is not yet available but would facilitate the manufacturing of therapeutically patterned cell populations using micro-patterned cell-instructive biomaterials for improved functional control as drug delivery systems in the context of cells as pharmaceutical products.
Collapse
Affiliation(s)
- Hui Che
- G.E.R.N. Research Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center-Albert-Ludwigs-University of Freiburg, 79085 Freiburg im Breisgau, Germany; Orthopedics and Sports Medicine Center, Suzhou Municipal Hospital (North District), Nanjing Medical University Affiliated Suzhou Hospital, Suzhou 215006, China
| | - Mischa Selig
- G.E.R.N. Research Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center-Albert-Ludwigs-University of Freiburg, 79085 Freiburg im Breisgau, Germany; Faculty of Biology, University of Freiburg, Schaenzlestrasse 1, D-79104 Freiburg, Germany
| | - Bernd Rolauffs
- G.E.R.N. Research Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center-Albert-Ludwigs-University of Freiburg, 79085 Freiburg im Breisgau, Germany.
| |
Collapse
|
59
|
Sarkar P, Kumar GA, Shrivastava S, Chattopadhyay A. Chronic cholesterol depletion increases F-actin levels and induces cytoskeletal reorganization via a dual mechanism. J Lipid Res 2022; 63:100206. [PMID: 35390404 PMCID: PMC9096963 DOI: 10.1016/j.jlr.2022.100206] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 03/22/2022] [Accepted: 03/28/2022] [Indexed: 12/24/2022] Open
Abstract
Previous work from us and others has suggested that cholesterol is an important lipid in the context of the organization of the actin cytoskeleton. However, reorganization of the actin cytoskeleton upon modulation of membrane cholesterol is rarely addressed in the literature. In this work, we explored the signaling crosstalk between cholesterol and the actin cytoskeleton by using a high-resolution confocal microscopic approach to quantitatively measure changes in F-actin content upon cholesterol depletion. Our results show that F-actin content significantly increases upon chronic cholesterol depletion, but not during acute cholesterol depletion. In addition, utilizing inhibitors targeting the cholesterol biosynthetic pathway at different steps, we show that reorganization of the actin cytoskeleton could occur due to the synergistic effect of multiple pathways, including prenylated Rho GTPases and availability of membrane phosphatidylinositol 4,5-bisphosphate. These results constitute one of the first comprehensive dissections of the mechanistic basis underlying the interplay between cellular actin levels and cholesterol biosynthesis. We envision these results will be relevant for future understating of the remodeling of the actin cytoskeleton in pathological conditions with altered cholesterol.
Collapse
Affiliation(s)
- Parijat Sarkar
- CSIR-Centre for Cellular and Molecular Biology, Hyderabad, India
| | - G Aditya Kumar
- CSIR-Centre for Cellular and Molecular Biology, Hyderabad, India
| | | | | |
Collapse
|
60
|
Zhang Y, Lu Q, Li N, Xu M, Miyamoto T, Liu J. Sulforaphane suppresses metastasis of triple-negative breast cancer cells by targeting the RAF/MEK/ERK pathway. NPJ Breast Cancer 2022; 8:40. [PMID: 35332167 PMCID: PMC8948359 DOI: 10.1038/s41523-022-00402-4] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 02/22/2022] [Indexed: 12/15/2022] Open
Abstract
Breast cancer metastasis is the main cause of cancer death in women, so far, no effective treatment has inhibited breast cancer metastasis. Sulforaphane (SFN), a natural compound derived from broccoli, has shown potential health benefits in many cancers. However, research on breast cancer metastasis is still insufficient. Here, we showed that SFN, including its two isomers of R-SFN and S-SFN, significantly inhibited TGF-β1-induced migration and invasion in breast cancer cells. Proteomic and phosphoproteomic analysis showed that SFN affected the formation of the cytoskeleton. Subsequent experiments confirmed that SFN significantly inhibited TGF-β1-induced actin stress fiber formation and the expression of actin stress fiber formation-associated proteins, including paxillin, IQGAP1, FAK, PAK2, and ROCK. Additionally, SFN is directly bound to RAF family proteins (including ARAF, BRAF, and CRAF) and inhibited MEK and ERK phosphorylation. These in vitro results indicate that SFN targets the RAF/MEK/ERK signaling pathway to inhibit the formation of actin stress fibers, thereby inhibiting breast cancer cell metastasis.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Molecular and Cellular Physiology, Yamaguchi University, Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan.
| | - Qian Lu
- Department of Molecular and Cellular Physiology, Yamaguchi University, Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Nan Li
- Department of Molecular and Cellular Physiology, Yamaguchi University, Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Ming Xu
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University, Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Tatsuo Miyamoto
- Department of Molecular and Cellular Physiology, Yamaguchi University, Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Jing Liu
- College of Pharmacy, Dalian Medical University, No.9 West Section Lvshun South Road, Dalian, 116044, China.
| |
Collapse
|
61
|
Bolanta S, Malijauskaite S, McGourty K, O’Reilly EJ. Synthesis of Poly(acrylic acid)-Cysteine-Based Hydrogels with Highly Customizable Mechanical Properties for Advanced Cell Culture Applications. ACS OMEGA 2022; 7:9108-9117. [PMID: 35350353 PMCID: PMC8945188 DOI: 10.1021/acsomega.1c03408] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 12/10/2021] [Indexed: 05/15/2023]
Abstract
The fabrication of highly customizable scaffolds is a key enabling technology in the development of predictive in vitro cell models for applications in drug discovery, cancer research, and regenerative medicine. Naturally derived and synthetic hydrogels are good candidates for in vitro cell growth studies, owing to their soft and biocompatible nature; however, they are often hindered by limited ranges of stiffness and the requirement to modify the gel with additional extracellular matrix (ECM) proteins for cell adherence. Here, we report on the synthesis of a printable synthetic hydrogel based on cysteine-modified poly(acrylic acid) (PAA-Cys) with tuneable mechanical and swelling properties by incorporating acrylic acid into the PAA-Cys network and subsequent photoinitiated thiol-acrylate cross-linking. Control of the acrylic acid concentration and UV curing time produces a series of hydrogels with swelling ratios in excess of 100% and Young's modulus values ranging from ∼2 to ∼35 kPa, of which most soft tissues fall within. Biocompatibility studies with RPE1 cells showed excellent cell adhesion and cell viability without the need for further modification with ECM proteins, but still can be modified as needed. The versatility of the hydrogel tuneable properties is demonstrated by culturing with RPE1 cells, which in vivo perform an important function in the visual process and the dysfunction of which may lead to various retinal abnormalities, such as glaucoma.
Collapse
Affiliation(s)
- Sharon
O. Bolanta
- Department
of Chemical Sciences, Bernal Institute University
of Limerick, Limerick V94 T9PX, Ireland
| | - Sigita Malijauskaite
- Department
of Chemical Sciences, Bernal Institute University
of Limerick, Limerick V94 T9PX, Ireland
| | - Kieran McGourty
- Department
of Chemical Sciences, Bernal Institute University
of Limerick, Limerick V94 T9PX, Ireland
- Health
Research Institute (HRI), University of Limerick, Limerick V94 T9PX, Ireland
| | - Emmet J. O’Reilly
- Department
of Chemical Sciences, Bernal Institute University
of Limerick, Limerick V94 T9PX, Ireland
| |
Collapse
|
62
|
Harford TJ, Rezaee F, Dye BR, Fan J, Spence JR, Piedimonte G. RSV-induced changes in a 3-dimensional organoid model of human fetal lungs. PLoS One 2022; 17:e0265094. [PMID: 35263387 PMCID: PMC8906588 DOI: 10.1371/journal.pone.0265094] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 02/22/2022] [Indexed: 12/27/2022] Open
Abstract
We have shown that respiratory syncytial virus (RSV) can spread hematogenously from infected airways of a pregnant woman to the developing fetal lungs in utero. This study sought to measure RSV replication, cytopathic effects, and protein expression in human lung organoids (HLOs) reproducing architecture and transcriptional profiles of human fetal lungs during the 1st trimester of gestation. HLOs derived from human pluripotent stem cells were microinjected after 50 or 100 days in culture with medium or recombinant RSV-A2 expressing the red fluorescent protein gene (rrRSV). Infection was monitored by fluorescent microscopy and PCR. Immunohistochemistry and proteomic analysis were performed. RSV infected HLOs in a dose- and time-dependent manner. RSV-infected HLOs increased expression of CC10 (Club cells), but had sparse FOXJ1 (ciliated cells). Disruption of F-actin cytoskeleton was consistent with proteomic data showing a significant increase in Rho GTPases proteins. RSV upregulated the transient receptor potential vanilloid 1 (TRPV1) channel and, while β2 adrenergic receptor (β2AR) expression was decreased overall, its phosphorylated form increased. Our data suggest that prenatal RSV infection produces profound changes in fetal lungs' architecture and expression profiles and maybe an essential precursor of chronic airway dysfunction. expression profiles, and possibly be an important precursor of chronic airway dysfunction.
Collapse
Affiliation(s)
- Terri J. Harford
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, United States of America
| | - Fariba Rezaee
- Center for Pediatric Pulmonary Medicine, Cleveland Clinic Children’s, Department of Inflammation & Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, United States of America
| | - Briana R. Dye
- Departments of Internal Medicine and Cell & Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Jia Fan
- Department of Biochemistry and Molecular Biology, Center for Cellular and Molecular Diagnostics, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Jason R. Spence
- Departments of Internal Medicine and Cell & Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Giovanni Piedimonte
- Department of Biochemistry and Molecular Biology, Center for Cellular and Molecular Diagnostics, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
- Department of Pediatrics, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| |
Collapse
|
63
|
Chang CY, Dai ZX, Shih PJ. Modeling and simulation of cell migration on the basis of force equilibrium. INTERNATIONAL JOURNAL FOR NUMERICAL METHODS IN BIOMEDICAL ENGINEERING 2022; 38:e3550. [PMID: 34719116 DOI: 10.1002/cnm.3550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 10/26/2021] [Indexed: 06/13/2023]
Abstract
To study cell behavior, we developed a cell model to simulate cell movements and the interacting forces among cells and between cells and obstacles. The developed model simulates several cells simultaneously and examines correlations among characteristic parameters between cells and substrates during migration. We modified Odde's model to develop fundamental model, applied Gillespie's stochastic algorithm to design time during in the migration simulation, and employed Keren's membrane theory to analyze the equilibrium at the leading edges. Thus, the proposed model can analyze stresses due to substrate, the intracellular body, and the external interaction between cells and obstacles. Simulation results indicate that cell-cell interaction depends on the equilibrium between the forces at the leading edge of the membrane, namely the cell-substrate interaction force, cell-cell interaction forces, and the cell membrane force. These results also indicate that the migration direction is dependent on the resultant forces. The membrane force and substrate force directions are "low correlation," and the polymerization rate exhibits "little correlative" with the migration direction. We propose a modified cell migration model for simulating allocation and interaction among multiple cells. This model helps indicate the weightings of characteristic parameters that affect the cell migration direction and velocity.
Collapse
Affiliation(s)
- Chia-Yu Chang
- Department of Mechanical Engineering, National Taiwan University, Taipei city, Taiwan
| | - Zhi-Xuan Dai
- Department of Mechanical Engineering, National Taiwan University, Taipei city, Taiwan
| | - Po-Jen Shih
- Department of Biomedical Engineering, National Taiwan University, Taipei city, Taiwan
| |
Collapse
|
64
|
Kim Y, Cho AY, Kim HC, Ryu D, Jo SA, Jung YS. Effects of Natural Polyphenols on Oxidative Stress-Mediated Blood–Brain Barrier Dysfunction. Antioxidants (Basel) 2022; 11:antiox11020197. [PMID: 35204080 PMCID: PMC8868362 DOI: 10.3390/antiox11020197] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 01/14/2022] [Accepted: 01/17/2022] [Indexed: 02/01/2023] Open
Abstract
The blood-brain barrier (BBB), which consists mainly of brain microvascular endothelial cells and astrocytes connected by tight junctions (TJs) and adhesion molecules (AMs), maintains the homeostatic balance between brain parenchyma and extracellular fluid. Accumulating evidence shows that BBB dysfunction is a common feature of neurodegenerative diseases, including stroke, traumatic brain injury, and Alzheimer’s disease. Among the various pathological pathways of BBB dysfunction, reactive oxygen species (ROS) are known to play a key role in inducing BBB disruption mediated via TJ modification, AM induction, cytoskeletal reorganization, and matrix metalloproteinase activation. Thus, antioxidants have been suggested to exert beneficial effects on BBB dysfunction-associated brain diseases. In this review, we summarized the sources of ROS production in multiple cells that constitute or surround the BBB, such as BBB endothelial cells, astrocytes, microglia, and neutrophils. We also reviewed various pathological mechanisms by which BBB disruption is caused by ROS in these cells. Finally, we summarized the effects of various natural polyphenols on BBB dysfunction to suggest a therapeutic strategy for BBB disruption-related brain diseases.
Collapse
Affiliation(s)
- Yeonjae Kim
- College of Pharmacy, Ajou University, Suwon 16499, Korea; (Y.K.); (A.Y.C.); (H.C.K.); (D.R.)
- Research Institute of Pharmaceutical Sciences and Technology, Ajou University, Suwon 16499, Korea
| | - A Yeon Cho
- College of Pharmacy, Ajou University, Suwon 16499, Korea; (Y.K.); (A.Y.C.); (H.C.K.); (D.R.)
| | - Hong Cheol Kim
- College of Pharmacy, Ajou University, Suwon 16499, Korea; (Y.K.); (A.Y.C.); (H.C.K.); (D.R.)
| | - Dajung Ryu
- College of Pharmacy, Ajou University, Suwon 16499, Korea; (Y.K.); (A.Y.C.); (H.C.K.); (D.R.)
- Research Institute of Pharmaceutical Sciences and Technology, Ajou University, Suwon 16499, Korea
| | - Sangmee Ahn Jo
- Department of Nanobiomedical Science & BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Korea;
- Department of Pharmacology, College of Pharmacy, Dankook University, Cheonan 31116, Korea
| | - Yi-Sook Jung
- College of Pharmacy, Ajou University, Suwon 16499, Korea; (Y.K.); (A.Y.C.); (H.C.K.); (D.R.)
- Research Institute of Pharmaceutical Sciences and Technology, Ajou University, Suwon 16499, Korea
- Correspondence: ; Tel.: +82-31-219-3444
| |
Collapse
|
65
|
Scandling BW, Gou J, Thomas J, Xuan J, Xue C, Gooch KJ. A Mechanistic Motor-Clutch Model That Explains Cell Shape Dynamics to Cyclic Stretch. Mol Biol Cell 2022; 33:ar27. [PMID: 35020463 PMCID: PMC9250388 DOI: 10.1091/mbc.e20-01-0087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Many cells in the body experience cyclic mechanical loading, which can impact cellular processes and morphology. In vitro studies often report that cells reorient in response to cyclic stretch of their substrate. To explore cellular mechanisms involved in this reorientation, a computational model was developed by adapting previous computational models of the actin–myosin–integrin motor-clutch system developed by others. The computational model predicts that under most conditions, actin bundles align perpendicular to the direction of applied cyclic stretch, but under specific conditions, such as low substrate stiffness, actin bundles align parallel to the direction of stretch. The model also predicts that stretch frequency impacts the rate of reorientation and that proper myosin function is critical in the reorientation response. These computational predictions are consistent with reports from the literature and new experimental results presented here. The model suggests that the impact of different stretching conditions (stretch type, amplitude, frequency, substrate stiffness, etc.) on the direction of cell alignment can largely be understood by considering their impact on cell–substrate detachment events, specifically whether detachments preferentially occur during stretching or relaxing of the substrate.
Collapse
Affiliation(s)
- Benjamin W Scandling
- Department of Biomedical Engineering, The Ohio State University.,The Frick Center for Heart Failure and Arrhythmia, Davis Heart Lung Research Institute, The Ohio State University
| | - Jia Gou
- Department of Mathematics, University of Minnesota.,Current Affiliation: Department of Mathematics, University of California, Riverside
| | - Jessica Thomas
- Department of Biomedical Engineering, The Ohio State University
| | - Jacqueline Xuan
- Department of Biomedical Engineering, The Ohio State University
| | - Chuan Xue
- School of Mathematics, University of Minnesota
| | - Keith J Gooch
- Department of Biomedical Engineering, The Ohio State University.,The Frick Center for Heart Failure and Arrhythmia, Davis Heart Lung Research Institute, The Ohio State University
| |
Collapse
|
66
|
Cagigas ML, Bryce NS, Ariotti N, Brayford S, Gunning PW, Hardeman EC. Correlative cryo-ET identifies actin/tropomyosin filaments that mediate cell-substrate adhesion in cancer cells and mechanosensitivity of cell proliferation. NATURE MATERIALS 2022; 21:120-128. [PMID: 34518666 DOI: 10.1038/s41563-021-01087-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 07/23/2021] [Indexed: 05/26/2023]
Abstract
The actin cytoskeleton is the primary driver of cellular adhesion and mechanosensing due to its ability to generate force and sense the stiffness of the environment. At the cell's leading edge, severing of the protruding Arp2/3 actin network generates a specific actin/tropomyosin (Tpm) filament population that controls lamellipodial persistence. The interaction between these filaments and adhesion to the environment is unknown. Using cellular cryo-electron tomography we resolve the ultrastructure of the Tpm/actin copolymers and show that they specifically anchor to nascent adhesions and are essential for focal adhesion assembly. Re-expression of Tpm1.8/1.9 in transformed and cancer cells is sufficient to restore cell-substrate adhesions. We demonstrate that knock-out of Tpm1.8/1.9 disrupts the formation of dorsal actin bundles, hindering the recruitment of α-actinin and non-muscle myosin IIa, critical mechanosensors. This loss causes a force-generation and proliferation defect that is notably reversed when cells are grown on soft surfaces. We conclude that Tpm1.8/1.9 suppress the metastatic phenotype, which may explain why transformed cells naturally downregulate this Tpm subset during malignant transformation.
Collapse
Affiliation(s)
- Maria Lastra Cagigas
- School of Medical Sciences, Faculty of Medicine and Health, UNSW Sydney, Sydney, New South Wales, Australia
| | - Nicole S Bryce
- School of Medical Sciences, Faculty of Medicine and Health, UNSW Sydney, Sydney, New South Wales, Australia
| | - Nicholas Ariotti
- School of Medical Sciences, Faculty of Medicine and Health, UNSW Sydney, Sydney, New South Wales, Australia
- Electron Microscope Unit, Mark Wainwright Analytical Centre, UNSW Sydney, Sydney, New South Wales, Australia
| | - Simon Brayford
- School of Medical Sciences, Faculty of Medicine and Health, UNSW Sydney, Sydney, New South Wales, Australia
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, New South Wales, Australia
| | - Peter W Gunning
- School of Medical Sciences, Faculty of Medicine and Health, UNSW Sydney, Sydney, New South Wales, Australia.
| | - Edna C Hardeman
- School of Medical Sciences, Faculty of Medicine and Health, UNSW Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
67
|
Fujiya T, Asanuma K, Koike T, Okata T, Saito M, Asano N, Imatani A, Masamune A. Nitric oxide could promote development of Barrett's esophagus by S-nitrosylation-induced inhibition of Rho-ROCK signaling in esophageal fibroblasts. Am J Physiol Gastrointest Liver Physiol 2022; 322:G107-G116. [PMID: 34786954 DOI: 10.1152/ajpgi.00124.2021] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 09/21/2021] [Indexed: 01/31/2023]
Abstract
Barrett's esophagus arises in the process of wound healing in distal esophageal epithelium damaged by gastroesophageal reflux disease. Differentiation of fibroblast into myofibroblasts, a smooth muscle cell-like phenotype and tissue contraction are crucial processes in wound healing. No study has evaluated mechanism by which luminal esophageal nitric oxide (NO) affect Rho-associated coiled coil-forming protein kinase (Rho-ROCK) signaling pathway, a key factor of tissue contraction, in stromal fibroblasts to develop Barrett's esophagus. Using esophageal fibroblasts, we performed collagen-based cell contraction assays and evaluated influence of Rho-ROCK signaling in the exposure to acidic bile salts and NOC-9, which is an NO donor. We found that enhanced cell contraction induced by acidic bile salts was inhibited by NO, accompanied by decrease in phosphorylated myosin light chain expression and stress fiber formation. NO directly S-nitrosylated GTP-RhoA and consequently blocked Rho-ROCK signaling. Moreover, exposure to NO and Y27632, a Rho-ROCK signaling inhibitor, decreased α-SMA expression and increased bone morphogenetic protein-4 (BMP4) expression and secretion. These findings could account for the increased expression of BMP4 in the columnar epithelial cells and stromal fibroblasts in human Barrett's esophagus. NO could impair wound contraction by blocking the Rho-ROCK signaling pathway and promote the development of Barrett's esophagus.NEW & NOTEWORTHY Barrett's esophagus is the condition where esophageal epithelium damaged by gastroesophageal reflux disease (GERD) is abnormally healed via replacing of metaplastic columnar epithelium, but very few studies have conducted focusing wound healing in the development of Barrett's esophagus. Esophageal luminal nitric oxide inhibits Rho-ROCK signaling pathway in esophageal fibroblasts, which leads to delay tissue contraction, a pivotal step in proper wound healing. Moreover, this inhibition increases tissue BMP4 expression. Impaired wound healing could be related to Barrett's esophagus.
Collapse
Affiliation(s)
- Taku Fujiya
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kiyotaka Asanuma
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tomoyuki Koike
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tomoki Okata
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Masahiro Saito
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Naoki Asano
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Akira Imatani
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Atsushi Masamune
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
68
|
Tsuda H, Tominaga SI, Ohtsuki M, Komine M. Nuclear IL-33 regulates cytokinesis and cell motility in normal human epidermal keratinocytes. J Dermatol Sci 2022; 105:113-120. [DOI: 10.1016/j.jdermsci.2022.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 01/03/2022] [Accepted: 01/24/2022] [Indexed: 11/25/2022]
|
69
|
LIM Kinases in Osteosarcoma Development. Cells 2021; 10:cells10123542. [PMID: 34944050 PMCID: PMC8699892 DOI: 10.3390/cells10123542] [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: 10/29/2021] [Revised: 12/06/2021] [Accepted: 12/10/2021] [Indexed: 12/17/2022] Open
Abstract
Tumorigenesis is a long-term and multistage process that often leads to the formation of metastases. During this pathological course, two major events appear to be crucial: primary tumour growth and metastatic expansion. In this context, despite research and clinical advances during the past decades, bone cancers remain a leading cause of death worldwide among paediatric cancer patients. Osteosarcomas are the most common malignant bone tumours in children and adolescents. Notwithstanding advances in therapeutic treatments, many patients succumb to these diseases. In particular, less than 30% of patients who demonstrate metastases at diagnosis or are poor responders to chemotherapy survive 5 years after initial diagnosis. LIM kinases (LIMKs), comprising LIMK1 and LIMK2, are common downstream effectors of several signalization pathways, and function as a signalling node that controls cytoskeleton dynamics through the phosphorylation of the cofilin family proteins. In recent decades, several reports have indicated that the functions of LIMKs are mainly implicated in the regulation of actin microfilament and the control of microtubule dynamics. Previous studies have thus identified LIMKs as cancer-promoting regulators in multiple organ cancers, such as breast cancer or prostate cancer. This review updates the current understanding of LIMK involvement in osteosarcoma progression.
Collapse
|
70
|
Patil A, Nandi S, Kale N, Bobade C, Banerjee S, Patil Y, Khandare J. Designing 3D-nanosubstrates mimicking biological cell growth: pitfalls of using 2D substrates in the evaluation of anticancer efficiency. NANOSCALE 2021; 13:17473-17485. [PMID: 34651162 DOI: 10.1039/d1nr03816h] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Designing nano-substrates (NS) that support three-dimensional (3D) cell growth using physico-chemical interventions mimicking the cellular microenvironment is highly challenging. Here we report NS that assist 3D cell development (3D NS) using multi-components on a glass substrate (2D GS), which mimics the ex vivo tissue microenvironment and promotes 3D cell growth superior to conventional 2D cell culturing methodologies. 3D NS were chemically fabricated by linking the combination of advanced materials imparting different physico-chemical traits, for example, multiwalled carbon nanotubes (CNT), graphene (G), bovine serum albumin (BSA), and iron oxide magnetic nanoparticles (MNP). We compared cell-substrate interactions resulting in cellular morphological changes, influence on the cell circularity index (CI), nuclear-cytoplasmic ratios (N/C), and nuclear compression or derangements using human colorectal carcinoma cells (HCT116) and cervical cancer (HeLa) cells. We observed the increase in N/C, extended on the 3D NS micro-environment as indicative of cellular adaptation and the transformation. HCT116 and HeLa cells on 2D GS showed an N/C ratio <0.3, and 3D NS cultured cells exhibited a higher N/C ratio (>0.5). The most significant increase in the ratio, relative to arrested cell spreading, was observed with G-3D NS. Furthermore, 3D NS were evaluated for the cell viability differentiations using the anticancer drug doxorubicin (Dox). The drug-treated cells on 3D NS demonstrated far-displaced N/C ratios compared to 2D GS. In conclusion, 3D NS systems implicate an 'in vitro to in vivo' relevance for the outcome in cell biology, cell proliferation and migration, and in anticancer drug efficacy evaluation.
Collapse
Affiliation(s)
- Ashwini Patil
- MAEER's Maharashtra Institute of Pharmacy, Kothrud, Pune 411038, India
| | - Semonti Nandi
- MAEER's Maharashtra Institute of Pharmacy, Kothrud, Pune 411038, India
| | - Narendra Kale
- MAEER's Maharashtra Institute of Pharmacy, Kothrud, Pune 411038, India
| | | | - Shashwat Banerjee
- School of Pharmacy, Dr Vishwananth Karad MIT-World Peace University, Kothrud, Pune 411038, India.
| | - Yuvraj Patil
- School of Pharmacy, Dr Vishwananth Karad MIT-World Peace University, Kothrud, Pune 411038, India.
| | - Jayant Khandare
- School of Consciousness, Dr Vishwananth Karad MIT-World Peace University, Kothrud, Pune 411038, India.
- School of Consciousness, MIT-WPU, Kothrud, Pune 411038, India
| |
Collapse
|
71
|
Castle EL, Robinson CA, Douglas P, Rinker KD, Corcoran JA. Viral Manipulation of a Mechanoresponsive Signaling Axis Disassembles Processing Bodies. Mol Cell Biol 2021; 41:e0039921. [PMID: 34516278 PMCID: PMC8547432 DOI: 10.1128/mcb.00399-21] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 08/28/2021] [Accepted: 09/04/2021] [Indexed: 12/23/2022] Open
Abstract
Processing bodies (PBs) are ribonucleoprotein granules important for cytokine mRNA decay that are targeted for disassembly by many viruses. Kaposi's sarcoma-associated herpesvirus is the etiological agent of the inflammatory endothelial cancer, Kaposi's sarcoma, and a PB-regulating virus. The virus encodes kaposin B (KapB), which induces actin stress fibers (SFs) and cell spindling as well as PB disassembly. We now show that KapB-mediated PB disassembly requires actin rearrangements, RhoA effectors, and the mechanoresponsive transcription activator, YAP. Moreover, ectopic expression of active YAP or exposure of ECs to mechanical forces caused PB disassembly in the absence of KapB. We propose that the viral protein KapB activates a mechanoresponsive signaling axis and links changes in cell shape and cytoskeletal structures to enhanced inflammatory molecule expression using PB disassembly. Our work implies that cytoskeletal changes in other pathologies may similarly impact the inflammatory environment.
Collapse
Affiliation(s)
- Elizabeth L. Castle
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Carolyn-Ann Robinson
- Charbonneau Cancer Institute, University of Calgary, Calgary, Alberta, Canada
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Pauline Douglas
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
- Charbonneau Cancer Institute, University of Calgary, Calgary, Alberta, Canada
| | - Kristina D. Rinker
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
- Department of Chemical and Petroleum Engineering and Centre for Bioengineering Research and Education, University of Calgary, Calgary, Alberta, Canada
- Charbonneau Cancer Institute, University of Calgary, Calgary, Alberta, Canada
| | - Jennifer A. Corcoran
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
- Charbonneau Cancer Institute, University of Calgary, Calgary, Alberta, Canada
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
72
|
Ulloa LS, Perissinotto F, Rago I, Goldoni A, Santoro R, Pesce M, Casalis L, Scaini D. Carbon Nanotubes Substrates Alleviate Pro-Calcific Evolution in Porcine Valve Interstitial Cells. NANOMATERIALS 2021; 11:nano11102724. [PMID: 34685165 PMCID: PMC8538037 DOI: 10.3390/nano11102724] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 10/11/2021] [Accepted: 10/12/2021] [Indexed: 01/19/2023]
Abstract
The quest for surfaces able to interface cells and modulate their functionality has raised, in recent years, the development of biomaterials endowed with nanocues capable of mimicking the natural extracellular matrix (ECM), especially for tissue regeneration purposes. In this context, carbon nanotubes (CNTs) are optimal candidates, showing dimensions and a morphology comparable to fibril ECM constituents. Moreover, when immobilized onto surfaces, they demonstrated outstanding cytocompatibility and ease of chemical modification with ad hoc functionalities. In this study, we interface porcine aortic valve interstitial cells (pVICs) to multi-walled carbon nanotube (MWNT) carpets, investigating the impact of surface nano-morphology on cell properties. The results obtained indicate that CNTs significantly affect cell behavior in terms of cell morphology, cytoskeleton organization, and mechanical properties. We discovered that CNT carpets appear to maintain interfaced pVICs in a sort of “quiescent state”, hampering cell activation into a myofibroblasts-like phenotype morphology, a cellular evolution prodromal to Calcific Aortic Valve Disease (CAVD) and characterized by valve interstitial tissue stiffening. We found that this phenomenon is linked to CNTs’ ability to alter cell tensional homeostasis, interacting with cell plasma membranes, stabilizing focal adhesions and enabling a better strain distribution within cells. Our discovery contributes to shedding new light on the ECM contribution in modulating cell behavior and will open the door to new criteria for designing nanostructured scaffolds to drive cell functionality for tissue engineering applications.
Collapse
Affiliation(s)
- Luisa Severino Ulloa
- Dipartimento di Fisica, Università di Trieste, Piazzale Europa 1, 34127 Trieste, Italy; (L.S.U.); (F.P.); (I.R.)
| | - Fabio Perissinotto
- Dipartimento di Fisica, Università di Trieste, Piazzale Europa 1, 34127 Trieste, Italy; (L.S.U.); (F.P.); (I.R.)
| | - Ilaria Rago
- Dipartimento di Fisica, Università di Trieste, Piazzale Europa 1, 34127 Trieste, Italy; (L.S.U.); (F.P.); (I.R.)
| | - Andrea Goldoni
- Elettra-Sincrotrone Trieste S.C.p.A., Basovizza, 34149 Trieste, Italy;
| | - Rosaria Santoro
- Unità di Ingegneria Tissutale Cardiovascolare, Centro Cardiologico Monzino, IRCCS, 20138 Milan, Italy; (R.S.); (M.P.)
| | - Maurizio Pesce
- Unità di Ingegneria Tissutale Cardiovascolare, Centro Cardiologico Monzino, IRCCS, 20138 Milan, Italy; (R.S.); (M.P.)
| | - Loredana Casalis
- Elettra-Sincrotrone Trieste S.C.p.A., Basovizza, 34149 Trieste, Italy;
- Correspondence: (L.C.); (D.S.)
| | - Denis Scaini
- Area di Neuroscienze, Scuola Internazionale Superiore di Studi Avanzati, Via Bonomea 265, 34136 Trieste, Italy
- Faculty of Medicine, Imperial College London, London W12 0NN, UK
- Correspondence: (L.C.); (D.S.)
| |
Collapse
|
73
|
Cook BL, Chao CJ, Alford PW. Architecture-Dependent Mechano-Adaptation in Single Vascular Smooth Muscle Cells. J Biomech Eng 2021; 143:101002. [PMID: 33972987 DOI: 10.1115/1.4051117] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Indexed: 01/03/2023]
Abstract
Arteries grow and remodel following mechanical perturbation. Vascular smooth muscle cells (VSMCs) within the artery undergo hyperplasia, hypertrophy, or change their contractility following sustained changes in loading. Experimental evidence in vivo and in vitro suggests that VSMCs grow and remodel to maintain a constant transmural stress, or "target" stress. This behavior is often described using a stress-dependent finite growth framework. Typically, computational models of arterial growth and remodeling account for VSMC behavior in a constrained mixture formulation that incorporates behavior of each component of the artery. However, these models do not account for differential VSMC architecture observed in situ, which may significantly influence growth and remodeling behavior. Here, we used cellular microbiaxial stretching (CμBS) to characterize how VSMCs with different cytoskeletal architectures respond to a sustained step change in strain. We find that VSMC F-actin architecture becomes more aligned following stretch and retains this alignment after 24 h. Further, we find that VSMC stress magnitude depends on cellular architecture. Qualitatively, however, stress behavior following stretch is consistent across cell architectures-stress increases following stretch and returns to prestretch magnitudes after 24 h. Finally, we formulated an architecture-dependent targeted growth law that accounts for experimentally measured cytoskeletal alignment and attributes stress evolution to individual fiber growth and find that this model robustly captures long-term stress evolution in single VSMCs. These results suggest that VSMC mechano-adaptation depends on cellular architecture, which has implications for growth and remodeling in regions of arteries with differential architecture, such as at bifurcations.
Collapse
Affiliation(s)
- Bernard L Cook
- Department of Biomedical Engineering, University of Minnesota, Nils Hasselmo Hall, Room 7-105 312 Church Street SE, Minneapolis, MN 55455
| | - Christina J Chao
- Department of Biomedical Engineering, University of Minnesota, Nils Hasselmo Hall, Room 7-105 312 Church Street SE, Minneapolis, MN 55455
| | - Patrick W Alford
- Department of Biomedical Engineering, University of Minnesota, Nils Hasselmo Hall, Room 7-105 312 Church Street SE, Minneapolis, MN 55455
| |
Collapse
|
74
|
Calamaras TD, Pande S, Baumgartner RA, Kim SK, McCarthy JC, Martin GL, Tam K, McLaughlin AL, Wang GR, Aronovitz MJ, Lin W, Aguirre JI, Baca P, Liu P, Richards DA, Davis RJ, Karas RH, Jaffe IZ, Blanton RM. MLK3 mediates impact of PKG1α on cardiac function and controls blood pressure through separate mechanisms. JCI Insight 2021; 6:e149075. [PMID: 34324442 PMCID: PMC8492323 DOI: 10.1172/jci.insight.149075] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 07/28/2021] [Indexed: 11/17/2022] Open
Abstract
cGMP-dependent protein kinase 1α (PKG1α) promotes left ventricle (LV) compensation after pressure overload. PKG1-activating drugs improve heart failure (HF) outcomes but are limited by vasodilation-induced hypotension. Signaling molecules that mediate PKG1α cardiac therapeutic effects but do not promote PKG1α-induced hypotension could therefore represent improved therapeutic targets. We investigated roles of mixed lineage kinase 3 (MLK3) in mediating PKG1α effects on LV function after pressure overload and in regulating BP. In a transaortic constriction HF model, PKG activation with sildenafil preserved LV function in MLK3+/+ but not MLK3-/- littermates. MLK3 coimmunoprecipitated with PKG1α. MLK3-PKG1α cointeraction decreased in failing LVs. PKG1α phosphorylated MLK3 on Thr277/Ser281 sites required for kinase activation. MLK3-/- mice displayed hypertension and increased arterial stiffness, though PKG stimulation with sildenafil or the soluble guanylate cyclase (sGC) stimulator BAY41-2272 still reduced BP in MLK3-/- mice. MLK3 kinase inhibition with URMC-099 did not affect BP but induced LV dysfunction in mice. These data reveal MLK3 as a PKG1α substrate mediating PKG1α preservation of LV function but not acute PKG1α BP effects. Mechanistically, MLK3 kinase-dependent effects preserved LV function, whereas MLK3 kinase-independent signaling regulated BP. These findings suggest augmenting MLK3 kinase activity could preserve LV function in HF but avoid hypotension from PKG1α activation.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Kelly Tam
- Molecular Cardiology Research Institute and
| | | | | | | | - Weiyu Lin
- Molecular Cardiology Research Institute and
- Graduate School of Biomedical Sciences, Tufts University, Boston, Massachusetts, USA
| | | | - Paulina Baca
- Molecular Cardiology Research Institute and
- Graduate School of Biomedical Sciences, Tufts University, Boston, Massachusetts, USA
| | - Peiwen Liu
- Molecular Cardiology Research Institute and
- Graduate School of Biomedical Sciences, Tufts University, Boston, Massachusetts, USA
| | | | - Roger J. Davis
- University of Massachusetts School of Medicine, Worchester, Massachusetts, USA
| | | | - Iris Z. Jaffe
- Molecular Cardiology Research Institute and
- Graduate School of Biomedical Sciences, Tufts University, Boston, Massachusetts, USA
| | - Robert M. Blanton
- Molecular Cardiology Research Institute and
- Graduate School of Biomedical Sciences, Tufts University, Boston, Massachusetts, USA
| |
Collapse
|
75
|
Hauke L, Narasimhan S, Primeßnig A, Kaverina I, Rehfeldt F. A Focal Adhesion Filament Cross-correlation Kit for fast, automated segmentation and correlation of focal adhesions and actin stress fibers in cells. PLoS One 2021; 16:e0250749. [PMID: 34506490 PMCID: PMC8432882 DOI: 10.1371/journal.pone.0250749] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 08/23/2021] [Indexed: 11/18/2022] Open
Abstract
Focal adhesions (FAs) and associated actin stress fibers (SFs) form a complex mechanical system that mediates bidirectional interactions between cells and their environment. This linked network is essential for mechanosensing, force production and force transduction, thus directly governing cellular processes like polarization, migration and extracellular matrix remodeling. We introduce a tool for fast and robust coupled analysis of both FAs and SFs named the Focal Adhesion Filament Cross-correlation Kit (FAFCK). Our software can detect and record location, axes lengths, area, orientation, and aspect ratio of focal adhesion structures as well as the location, length, width and orientation of actin stress fibers. This enables users to automate analysis of the correlation of FAs and SFs and study the stress fiber system in a higher degree, pivotal to accurately evaluate transmission of mechanocellular forces between a cell and its surroundings. The FAFCK is particularly suited for unbiased and systematic quantitative analysis of FAs and SFs necessary for novel approaches of traction force microscopy that uses the additional data from the cellular side to calculate the stress distribution in the substrate. For validation and comparison with other tools, we provide datasets of cells of varying quality that are labelled by a human expert. Datasets and FAFCK are freely available as open source under the GNU General Public License.
Collapse
Affiliation(s)
- Lara Hauke
- Third Institute of Physics—Biophysics, Georg-August-University Göttingen, Göttingen, Germany
| | - Shwetha Narasimhan
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, United States of America
| | - Andreas Primeßnig
- Third Institute of Physics—Biophysics, Georg-August-University Göttingen, Göttingen, Germany
| | - Irina Kaverina
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, United States of America
- * E-mail: (IK); (FR)
| | - Florian Rehfeldt
- Third Institute of Physics—Biophysics, Georg-August-University Göttingen, Göttingen, Germany
- Experimental Physics I, University of Bayreuth, Bayreuth, Germany
- * E-mail: (IK); (FR)
| |
Collapse
|
76
|
Sohrabi Kashani A, Packirisamy M. Cancer-Nano-Interaction: From Cellular Uptake to Mechanobiological Responses. Int J Mol Sci 2021; 22:9587. [PMID: 34502495 PMCID: PMC8431109 DOI: 10.3390/ijms22179587] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/25/2021] [Accepted: 08/28/2021] [Indexed: 12/12/2022] Open
Abstract
With the advancement of nanotechnology, the nano-bio-interaction field has emerged. It is essential to enhance our understanding of nano-bio-interaction in different aspects to design nanomedicines and improve their efficacy for therapeutic and diagnostic applications. Many researchers have extensively studied the toxicological responses of cancer cells to nano-bio-interaction, while their mechanobiological responses have been less investigated. The mechanobiological properties of cells such as elasticity and adhesion play vital roles in cellular functions and cancer progression. Many studies have noticed the impacts of cellular uptake on the structural organization of cells and, in return, the mechanobiology of human cells. Mechanobiological changes induced by the interactions of nanomaterials and cells could alter cellular functions and influence cancer progression. Hence, in addition to biological responses, the possible mechanobiological responses of treated cells should be monitored as a standard methodology to evaluate the efficiency of nanomedicines. Studying the cancer-nano-interaction in the context of cell mechanics takes our knowledge one step closer to designing safe and intelligent nanomedicines. In this review, we briefly discuss how the characteristic properties of nanoparticles influence cellular uptake. Then, we provide insight into the mechanobiological responses that may occur during the nano-bio-interactions, and finally, the important measurement techniques for the mechanobiological characterizations of cells are summarized and compared. Understanding the unknown mechanobiological responses to nano-bio-interaction will help with developing the application of nanoparticles to modulate cell mechanics for controlling cancer progression.
Collapse
Affiliation(s)
| | - Muthukumaran Packirisamy
- Optical Bio-Microsystem Lab, Micro-Nano-Bio-Integration Centre, Department of Mechanical, Industrial and Aerospace Engineering, Concordia University, 1455 De Maisonneuve Blvd. W., Montreal, QC H3G 1M8, Canada;
| |
Collapse
|
77
|
Nishimura Y, Shi S, Li Q, Bershadsky AD, Viasnoff V. Crosstalk between myosin II and formin functions in the regulation of force generation and actomyosin dynamics in stress fibers. Cells Dev 2021; 168:203736. [PMID: 34455135 DOI: 10.1016/j.cdev.2021.203736] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 07/23/2021] [Accepted: 08/16/2021] [Indexed: 10/20/2022]
Abstract
REF52 fibroblasts have a well-developed contractile machinery, the most prominent elements of which are actomyosin stress fibers with highly ordered organization of actin and myosin IIA filaments. The relationship between contractile activity and turnover dynamics of stress fibers is not sufficiently understood. Here, we simultaneously measured the forces exerted by stress fibers (using traction force microscopy or micropillar array sensors) and the dynamics of actin and myosin (using photoconversion-based monitoring of actin incorporation and high-resolution fluorescence microscopy of myosin II light chain). Our data revealed new features of the crosstalk between myosin II-driven contractility and stress fiber dynamics. During normal stress fiber turnover, actin incorporated all along the stress fibers and not only at focal adhesions. Incorporation of actin into stress fibers/focal adhesions, as well as actin and myosin II filaments flow along stress fibers, strongly depends on myosin II activity. Myosin II-dependent generation of traction forces does not depend on incorporation of actin into stress fibers per se, but still requires formin activity. This previously overlooked function of formins in maintenance of the actin cytoskeleton connectivity could be the main mechanism of formin involvement in traction force generation.
Collapse
Affiliation(s)
- Yukako Nishimura
- Mechanobiology Institute, National University of Singapore, T-Lab, 5A Engineering Drive 1, 117411, Singapore; Division of Developmental Physiology, Institute for Genetic Medicine, Hokkaido University, Kita-15, Nishi-7, Kita-ku, Sapporo 060-0815, Japan
| | - Shidong Shi
- Mechanobiology Institute, National University of Singapore, T-Lab, 5A Engineering Drive 1, 117411, Singapore
| | - Qingsen Li
- IFOM-FIRC Institute of Molecular Oncology, Via Adamello, 16, 20139 Milan, Italy
| | - Alexander D Bershadsky
- Mechanobiology Institute, National University of Singapore, T-Lab, 5A Engineering Drive 1, 117411, Singapore; Department of Molecular Cell Biology, Weizmann Institute of Science, 234 Herzl Street, POB 26, Rehovot 7610001, Israel.
| | - Virgile Viasnoff
- Mechanobiology Institute, National University of Singapore, T-Lab, 5A Engineering Drive 1, 117411, Singapore; CNRS UMI 3639, Singapore; Department of Biological Sciences, National university of Singapore, S3 #05-01, 16 Science Drive 4, 117558, Singapore.
| |
Collapse
|
78
|
Protein friction and filament bending facilitate contraction of disordered actomyosin networks. Biophys J 2021; 120:4029-4040. [PMID: 34390686 DOI: 10.1016/j.bpj.2021.08.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 06/21/2021] [Accepted: 08/06/2021] [Indexed: 12/23/2022] Open
Abstract
We use mathematical modeling and computation to investigate how protein friction facilitates contraction of disordered actomyosin networks. We simulate two-dimensional networks using an agent-based model, consisting of a system of force-balance equations for myosin motor proteins and semiflexible actin filaments. A major advantage of our approach is that it enables direct calculation of the network stress tensor, which provides a quantitative measure of contractility. Exploiting this, we use repeated simulations of disordered networks to confirm that both protein friction and actin filament bending are required for contraction. We then use simulations of elementary two-filament systems to show that filament bending flexibility can facilitate contraction on the microscopic scale. Finally, we show that actin filament turnover is necessary to sustain contraction and prevent filament aggregation. Simulations with and without turnover also exhibit contractile pulses. However, these pulses are aperiodic, suggesting that periodic pulsation can only arise because of additional regulatory mechanisms or more complex mechanical behavior.
Collapse
|
79
|
Oo Y, Nealiga JQL, Suwanborirux K, Chamni S, Ecoy GAU, Pongrakhananon V, Chanvorachote P, Chaotham C. 22-O-(N-Boc-L-glycine) ester of renieramycin M inhibits migratory activity and suppresses epithelial-mesenchymal transition in human lung cancer cells. J Nat Med 2021; 75:949-966. [PMID: 34287745 DOI: 10.1007/s11418-021-01549-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 07/04/2021] [Indexed: 12/19/2022]
Abstract
The incidence of metastasis stage crucially contributes to high recurrence and mortality rate in lung cancer patients. Unfortunately, no available treatment inhibits migration, a key metastasis process in lung cancer. In this study, the effect of 22-O-(N-Boc-L-glycine) ester of renieramycin M (22-Boc-Gly-RM), a semi-synthetic amino ester derivative of bistetrahydroisoquinolinequinone alkaloid isolated from Xestospongia sp., on migratory behavior of human lung cancer cells was investigated. Following 24 h of treatment, 22-Boc-Gly-RM at non-toxic concentrations (0.5-1 μM) effectively restrained motility of human lung cancer H460 cells assessed through wound healing, transwell migration, and multicellular spheroid models. The capability to invade through matrix component was also repressed in H460 cells cultured with 0.1-1 µM 22-Boc-Gly-RM. The dose-dependent reduction of phalloidin-stained actin stress fibers corresponded with the downregulated Rac1-GTP level presented via western blot analysis in 22-Boc-Gly-RM-treated cells. Treatment with 0.1-1 μM of 22-Boc-Gly-RM obviously caused suppression of p-FAK/p-Akt signal and consequent inhibition of epithelial-to-mesenchymal transition (EMT), which was evidenced with augmented level of E-cadherin and reduction of N-cadherin expression. The alteration of invasion-related proteins in 22-Boc-Gly-RM-treated H460 cells was indicated by the diminution of matrix metalloproteinases (MT1-MMP, MMP-2, MMP-7, and MMP-9), as well as the upregulation of tissue inhibitors of metalloproteinases (TIMP), TIMP2, and TIMP3. Thus, 22-Boc-Gly-RM is a promising candidate for anti-metastasis treatment in lung cancer through inhibition of migratory features associated with suppression on EMT.
Collapse
Affiliation(s)
- Yamin Oo
- Department of Biochemistry and Microbiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Justin Quiel Lasam Nealiga
- Department of Biochemistry and Microbiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Khanit Suwanborirux
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Supakarn Chamni
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand.,Natural Products and Nanoparticles Research Unit (NP2), Chulalongkorn University, Bangkok, 10330, Thailand
| | - Gea Abigail Uy Ecoy
- Department of Biochemistry and Microbiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand.,Department of Pharmacy, School of Health Care Professions, University of San Carlos, 6000, Cebu, Philippines
| | - Varisa Pongrakhananon
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Pithi Chanvorachote
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand.,Cell-Based Drug and Health Products Development Research Unit, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Chatchai Chaotham
- Department of Biochemistry and Microbiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand. .,Cell-Based Drug and Health Products Development Research Unit, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand.
| |
Collapse
|
80
|
Harmoush B, Tsikolia N, Viebahn C. Epiblast and trophoblast morphogenesis in the pre-gastrulation blastocyst of the pig. A light- and electron-microscopical study. J Morphol 2021; 282:1339-1361. [PMID: 34176156 DOI: 10.1002/jmor.21389] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 05/31/2021] [Accepted: 06/15/2021] [Indexed: 01/18/2023]
Abstract
The epiblast of the amniote embryo is of paramount importance during early development as it gives rise to all tissues of the embryo proper. In mammals, it emerges through segregation of the hypoblast from the inner cell mass and subsequently undergoes transformation into an epithelial sheet to create the embryonic disc. In rodents and man, the epiblast cell layer is covered by the polar trophoblast which forms the placenta. In mammalian model organisms (rabbit, pig, several non-human primates), however, the placenta is formed by mural trophoblast whereas the polar trophoblast disintegrates prior to gastrulation and thus exposes the epiblast to the microenvironment of the uterine cavity. Both, polar trophoblast disintegration and epiblast epithelialization, thus pose special cell-biological requirements but these are still rather ill-understood when compared to those of gastrulation morphogenesis. This study therefore applied high-resolution light and transmission electron microscopy and three-dimensional (3D) reconstruction to 8- to 10-days-old pig embryos and defines the following steps of epiblast transformation: (1) rosette formation in the center of the ball-shaped epiblast, (2) extracellular cavity formation in the rosette center, (3) epiblast segregation into two subpopulations - addressed here as dorsal and ventral epiblast - separated by a "pro-amniotic" cavity. Ventral epiblast cells form between them a special type of desmosomes with a characteristic dense felt of microfilaments and are destined to generate the definitive epiblast. The dorsal epiblast remains a mass of non-polarized cells and closely associates with the disintegrating polar trophoblast, which shows morphological features of both apoptosis and autophagocytosis. Morphogenesis of the definitive epiblast in the pig may thus exclude a large portion of bona fide epiblast cells from contributing to the embryo proper and establishes contact de novo with the mural trophoblast at the junction between the two newly defined epiblast cell populations.
Collapse
Affiliation(s)
- Braah Harmoush
- Institute of Anatomy and Embryology, University Medical Centre Göttingen, Göttingen, Germany
| | - Nikoloz Tsikolia
- Institute of Anatomy and Embryology, University Medical Centre Göttingen, Göttingen, Germany
| | - Christoph Viebahn
- Institute of Anatomy and Embryology, University Medical Centre Göttingen, Göttingen, Germany
| |
Collapse
|
81
|
Peng JM, Hsieh SY, Cheng JH, Luo JW, Su YL, Luo HL. Confirming whether KLHL23 deficiency potentiates migration in urothelial carcinoma. CHINESE J PHYSIOL 2021; 64:142-149. [PMID: 34169920 DOI: 10.4103/cjp.cjp_110_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Epithelial-mesenchymal transition (EMT) is associated with malignant tumors. In a previous study, we found that KLHL23 is a tumor suppressor gene that inhibits EMT and cancer dissemination. However, the correlation between its expression and cancer progression in urothelial carcinoma (UC) remains unknown. This study showed that the deficiency of KLHL23 in the invasive leading cancer cells is important for improving cell migration in UC. Currently, little is known about the underlying mechanisms of KLHL23-mediated cytoskeleton remodeling in the metastatic leading cells of tumors. Our findings showed that silencing of KLHL23 promotes cell migration in UC by regulating the translocation of focal adhesion proteins. Lack of KLHL23 causes abnormal formation of lamellipodia and increases the EMT phenotype and migration. Wound healing assay revealed that KLHL23 potentiates the actin bundles and intracellular focal adhesion protein formation in the invasive leading cells. Knockdown of KLHL23 abolishes the formation of actin stress fibers and translocalizes vinculin to the perimembrane, which enhances the mobility of cancer cells. To elucidate the mechanism, we found that during migration, KLHL23 appears in the leading cells in large numbers and binds to the actin stress fibers. A large amount of vinculin accumulated at both ends of the KLHL23/actin fibers, indicating an increase in cell anchorage. Thus, KLHL23 might play a critical role in enhancing actin fibers and promoting focal adhesion complex formation in the invasive leading cells. Analysis of the overall survival revealed that low KLHL23 is associated with poor survival in patients with bladder UC, indicating its clinical significance. We hypothesize that KLHL23 is involved in the formation of actin stress fibers and focal adhesion complexes in the invasive leading cells and may be associated with EMT progression and prognosis in UC patients.
Collapse
Affiliation(s)
- Jei-Ming Peng
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Sen-Yung Hsieh
- Department of Gastroenterology and Hepatology, Chang Gung Memorial Hospital; Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Jai-Hong Cheng
- Center for Shockwave Medicine and Tissue Engineering, Medical Research, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine; Department of Leisure and Sports Management, Cheng Shiu University, Kaohsiung, Taiwan
| | - Jia-Wun Luo
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Yu-Li Su
- Division of Hematology Oncology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Taiwan
| | - Hao-Lun Luo
- Department of Urology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| |
Collapse
|
82
|
Eapen MS, Lu W, Hackett TL, Singhera GK, Mahmood MQ, Hardikar A, Ward C, Walters EH, Sohal SS. Increased myofibroblasts in the small airways, and relationship to remodelling and functional changes in smokers and COPD patients: potential role of epithelial-mesenchymal transition. ERJ Open Res 2021; 7:00876-2020. [PMID: 34109247 PMCID: PMC8181830 DOI: 10.1183/23120541.00876-2020] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 03/10/2021] [Indexed: 12/11/2022] Open
Abstract
Introduction Previous reports have shown epithelial-mesenchymal transition (EMT) as an active process that contributes to small airway fibrotic pathology. Myofibroblasts are highly active pro-fibrotic cells that secrete excessive and altered extracellular matrix (ECM). Here we relate small airway myofibroblast presence with airway remodelling, physiology and EMT activity in smokers and COPD patients. Methods Lung resections from nonsmoker controls, normal lung function smokers and COPD current and ex-smokers were stained with anti-human α-smooth muscle actin (SMA), collagen 1 and fibronectin. αSMA+ cells were computed in reticular basement membrane (Rbm), lamina propria and adventitia and presented per mm of Rbm and mm2 of lamina propria. Collagen-1 and fibronectin are presented as a percentage change from normal. All analyses including airway thickness were measured using Image-pro-plus 7.0. Results We found an increase in subepithelial lamina propria (especially) and adventitia thickness in all pathological groups compared to nonsmoker controls. Increases in αSMA+ myofibroblasts were observed in subepithelial Rbm, lamina propria and adventitia in both the smoker and COPD groups compared to nonsmoker controls. Furthermore, the increase in the myofibroblast population in the lamina propria was strongly associated with decrease in lung function, lamina propria thickening, increase in ECM protein deposition, and finally EMT activity in epithelial cells. Conclusions This is the first systematic characterisation of small airway myofibroblasts in COPD based on their localisation, with statistically significant correlations between them and other pan-airway structural, lung function and ECM protein changes. Finally, we suggest that EMT may be involved in such changes.
Collapse
Affiliation(s)
- Mathew Suji Eapen
- Respiratory Translational Research Group, Dept of Laboratory Medicine, School of Health Sciences, College of Health and Medicine, University of Tasmania, Launceston, Australia
| | - Wenying Lu
- Respiratory Translational Research Group, Dept of Laboratory Medicine, School of Health Sciences, College of Health and Medicine, University of Tasmania, Launceston, Australia
| | - Tillie L Hackett
- Dept of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, BC, Canada.,UBC Centre for Heart Lung Innovation, St Paul's Hospital, Vancouver, BC, Canada
| | - Gurpreet Kaur Singhera
- UBC Centre for Heart Lung Innovation, St Paul's Hospital, Vancouver, BC, Canada.,Dept of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Malik Q Mahmood
- School of Medicine, Deakin University, Waurn Ponds, Australia
| | - Ashutosh Hardikar
- Respiratory Translational Research Group, Dept of Laboratory Medicine, School of Health Sciences, College of Health and Medicine, University of Tasmania, Launceston, Australia.,Dept of Cardiothoracic Surgery, Royal Hobart Hospital, Hobart, Australia
| | - Chris Ward
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Eugene Haydn Walters
- School of Medicine, and Menzies Institute of Medical Research, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia.,These authors contributed equally
| | - Sukhwinder Singh Sohal
- Respiratory Translational Research Group, Dept of Laboratory Medicine, School of Health Sciences, College of Health and Medicine, University of Tasmania, Launceston, Australia.,These authors contributed equally
| |
Collapse
|
83
|
Kasai H, Ziv NE, Okazaki H, Yagishita S, Toyoizumi T. Spine dynamics in the brain, mental disorders and artificial neural networks. Nat Rev Neurosci 2021; 22:407-422. [PMID: 34050339 DOI: 10.1038/s41583-021-00467-3] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/14/2021] [Indexed: 12/15/2022]
Abstract
In the brain, most synapses are formed on minute protrusions known as dendritic spines. Unlike their artificial intelligence counterparts, spines are not merely tuneable memory elements: they also embody algorithms that implement the brain's ability to learn from experience and cope with new challenges. Importantly, they exhibit structural dynamics that depend on activity, excitatory input and inhibitory input (synaptic plasticity or 'extrinsic' dynamics) and dynamics independent of activity ('intrinsic' dynamics), both of which are subject to neuromodulatory influences and reinforcers such as dopamine. Here we succinctly review extrinsic and intrinsic dynamics, compare these with parallels in machine learning where they exist, describe the importance of intrinsic dynamics for memory management and adaptation, and speculate on how disruption of extrinsic and intrinsic dynamics may give rise to mental disorders. Throughout, we also highlight algorithmic features of spine dynamics that may be relevant to future artificial intelligence developments.
Collapse
Affiliation(s)
- Haruo Kasai
- Laboratory of Structural Physiology, Center for Disease Biology and Integrative Medicine, Faculty of Medicine, The University of Tokyo, Tokyo, Japan. .,International Research Center for Neurointelligence (WPI-IRCN), UTIAS, The University of Tokyo, Bunkyo-ku, Tokyo, Japan.
| | - Noam E Ziv
- Technion Faculty of Medicine and Network Biology Research Labs, Technion City, Haifa, Israel
| | - Hitoshi Okazaki
- Laboratory of Structural Physiology, Center for Disease Biology and Integrative Medicine, Faculty of Medicine, The University of Tokyo, Tokyo, Japan.,International Research Center for Neurointelligence (WPI-IRCN), UTIAS, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Sho Yagishita
- Laboratory of Structural Physiology, Center for Disease Biology and Integrative Medicine, Faculty of Medicine, The University of Tokyo, Tokyo, Japan.,International Research Center for Neurointelligence (WPI-IRCN), UTIAS, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Taro Toyoizumi
- Laboratory for Neural Computation and Adaptation, RIKEN Center for Brain Science, Saitama, Japan.,Department of Mathematical Informatics, Graduate School of Information Science and Technology, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
84
|
Anderson CA, Kovar DR, Gardel ML, Winkelman JD. LIM domain proteins in cell mechanobiology. Cytoskeleton (Hoboken) 2021; 78:303-311. [PMID: 34028199 DOI: 10.1002/cm.21677] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 05/06/2021] [Accepted: 05/07/2021] [Indexed: 12/13/2022]
Abstract
The actin cytoskeleton is important for maintaining mechanical homeostasis in adherent cells, largely through its regulation of adhesion and cortical tension. The LIM (Lin-11, Isl1, MEC-3) domain-containing proteins are involved in a myriad of cellular mechanosensitive pathways. Recent work has discovered that LIM domains bind to mechanically stressed actin filaments, suggesting a novel and widely conserved mechanism of mechanosensing. This review summarizes the current state of knowledge of LIM protein mechanosensitivity.
Collapse
Affiliation(s)
- Caitlin A Anderson
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, Illinois, USA
| | - David R Kovar
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, Illinois, USA.,Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, Illinois, USA
| | - Margaret L Gardel
- Institute for Biophysical Dynamics, University of Chicago, Chicago, Illinois, USA.,James Franck Institute, University of Chicago, Chicago, Illinois, USA.,Department of Physics, University of Chicago, Chicago, Illinois, USA.,Pritzker School of Molecular Engineering, University of Chicago, Chicago, Illinois, USA
| | - Jonathan D Winkelman
- Institute for Biophysical Dynamics, University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
85
|
Linfield DT, Gao N, Raduka A, Harford TJ, Piedimonte G, Rezaee F. RSV attenuates epithelial cell restitution by inhibiting actin cytoskeleton-dependent cell migration. Am J Physiol Lung Cell Mol Physiol 2021; 321:L189-L203. [PMID: 34010080 DOI: 10.1152/ajplung.00118.2021] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The airway epithelium's ability to repair itself after injury, known as epithelial restitution, is an essential mechanism enabling the respiratory tract's normal functions. Respiratory Syncytial Virus (RSV) is the leading cause of lower respiratory tract infections worldwide. We sought to determine whether RSV delays the airway epithelium wound repair process both in vitro and in vivo. We found that RSV infection attenuated epithelial cell migration, a step in wound repair, promoted stress fiber formation, and mediated assembly of large focal adhesions (FA). Inhibition of Rho kinase (ROCK), a master regulator of actin function, reversed these effects. There was increased RhoA and phospho-myosin light chain (pMLC2) following RSV infection. In vivo, mice were intraperitoneally inoculated with naphthalene to induce lung injury, followed by RSV infection. RSV infection delayed re-epithelialization. There were increased concentrations of pMLC2 in day 7 naphthalene plus RSV animals which normalized by day 14. This study suggests a key mechanism by which RSV infection delays wound healing.
Collapse
Affiliation(s)
| | - Nannan Gao
- Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, United States
| | - Andjela Raduka
- Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, United States
| | - Terri J Harford
- Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, United States
| | | | - Fariba Rezaee
- Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, United States.,Center for Pediatric Pulmonology, Cleveland Clinic Children's, Cleveland, Ohio, United States
| |
Collapse
|
86
|
Linklater ES, Duncan ED, Han KJ, Kaupinis A, Valius M, Lyons TR, Prekeris R. Rab40-Cullin5 complex regulates EPLIN and actin cytoskeleton dynamics during cell migration. J Cell Biol 2021; 220:212111. [PMID: 33999101 PMCID: PMC8129794 DOI: 10.1083/jcb.202008060] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 02/09/2021] [Accepted: 04/26/2021] [Indexed: 12/12/2022] Open
Abstract
Rab40b is a SOCS box–containing protein that regulates the secretion of MMPs to facilitate extracellular matrix remodeling during cell migration. Here, we show that Rab40b interacts with Cullin5 via the Rab40b SOCS domain. We demonstrate that loss of Rab40b–Cullin5 binding decreases cell motility and invasive potential and show that defective cell migration and invasion stem from alteration to the actin cytoskeleton, leading to decreased invadopodia formation, decreased actin dynamics at the leading edge, and an increase in stress fibers. We also show that these stress fibers anchor at less dynamic, more stable focal adhesions. Mechanistically, changes in the cytoskeleton and focal adhesion dynamics are mediated in part by EPLIN, which we demonstrate to be a binding partner of Rab40b and a target for Rab40b–Cullin5-dependent localized ubiquitylation and degradation. Thus, we propose a model where Rab40b–Cullin5-dependent ubiquitylation regulates EPLIN localization to promote cell migration and invasion by altering focal adhesion and cytoskeletal dynamics.
Collapse
Affiliation(s)
- Erik S Linklater
- Department of Cell and Developmental Biology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Emily D Duncan
- Department of Cell and Developmental Biology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Ke-Jun Han
- Department of Cell and Developmental Biology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Algirdas Kaupinis
- Proteomics Center, Institute of Biochemistry, Vilnius University Life Sciences Center, Vilnius, Lithuania
| | - Mindaugas Valius
- Proteomics Center, Institute of Biochemistry, Vilnius University Life Sciences Center, Vilnius, Lithuania
| | - Traci R Lyons
- Division of Medical Oncology, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO.,University of Colorado Cancer Center, Young Women's Breast Cancer Translational Program, Aurora, CO
| | - Rytis Prekeris
- Department of Cell and Developmental Biology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| |
Collapse
|
87
|
Articular Chondrocyte Phenotype Regulation through the Cytoskeleton and the Signaling Processes That Originate from or Converge on the Cytoskeleton: Towards a Novel Understanding of the Intersection between Actin Dynamics and Chondrogenic Function. Int J Mol Sci 2021; 22:ijms22063279. [PMID: 33807043 PMCID: PMC8004672 DOI: 10.3390/ijms22063279] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 03/16/2021] [Accepted: 03/17/2021] [Indexed: 02/08/2023] Open
Abstract
Numerous studies have assembled a complex picture, in which extracellular stimuli and intracellular signaling pathways modulate the chondrocyte phenotype. Because many diseases are mechanobiology-related, this review asked to what extent phenotype regulators control chondrocyte function through the cytoskeleton and cytoskeleton-regulating signaling processes. Such information would generate leverage for advanced articular cartilage repair. Serial passaging, pro-inflammatory cytokine signaling (TNF-α, IL-1α, IL-1β, IL-6, and IL-8), growth factors (TGF-α), and osteoarthritis not only induce dedifferentiation but also converge on RhoA/ROCK/Rac1/mDia1/mDia2/Cdc42 to promote actin polymerization/crosslinking for stress fiber (SF) formation. SF formation takes center stage in phenotype control, as both SF formation and SOX9 phosphorylation for COL2 expression are ROCK activity-dependent. Explaining how it is molecularly possible that dedifferentiation induces low COL2 expression but high SF formation, this review theorized that, in chondrocyte SOX9, phosphorylation by ROCK might effectively be sidelined in favor of other SF-promoting ROCK substrates, based on a differential ROCK affinity. In turn, actin depolymerization for redifferentiation would “free-up” ROCK to increase COL2 expression. Moreover, the actin cytoskeleton regulates COL1 expression, modulates COL2/aggrecan fragment generation, and mediates a fibrogenic/catabolic expression profile, highlighting that actin dynamics-regulating processes decisively control the chondrocyte phenotype. This suggests modulating the balance between actin polymerization/depolymerization for therapeutically controlling the chondrocyte phenotype.
Collapse
|
88
|
Naffa R, Padányi R, Ignácz A, Hegyi Z, Jezsó B, Tóth S, Varga K, Homolya L, Hegedűs L, Schlett K, Enyedi A. The Plasma Membrane Ca 2+ Pump PMCA4b Regulates Melanoma Cell Migration through Remodeling of the Actin Cytoskeleton. Cancers (Basel) 2021; 13:cancers13061354. [PMID: 33802790 PMCID: PMC8002435 DOI: 10.3390/cancers13061354] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/08/2021] [Accepted: 03/14/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Earlier we demonstrated that the plasma membrane Ca2+ pump PMCA4b inhibits migration and metastatic activity of BRAF mutant melanoma cells, however, the exact mechanism has not been fully understood. Here we demonstrate that PMCA4b acted through actin cytoskeleton remodeling in generating a low migratory melanoma cell phenotype resulting in increased cell–cell connections, lamellipodia and stress fiber formation. Both proper trafficking and calcium transporting activity of the pump were essential to complete these tasks indicating that controlling Ca2+ concentration levels at specific plasma membrane locations such as the cell front played a role. Our findings suggest that PMCA4b downregulation is likely one of the mechanisms that leads to the perturbed cancer cell cytoskeleton organization resulting in enhanced melanoma cell migration and metastasis. Abstract We demonstrated that the plasma membrane Ca2+ ATPase PMCA4b inhibits migration and metastatic activity of BRAF mutant melanoma cells. Actin dynamics are essential for cells to move, invade and metastasize, therefore, we hypothesized that PMCA4b affected cell migration through remodeling of the actin cytoskeleton. We found that expression of PMCA4b in A375 BRAF mutant melanoma cells induced a profound change in cell shape, cell culture morphology, and displayed a polarized migratory character. Along with these changes the cells became more rounded with increased cell–cell connections, lamellipodia and stress fiber formation. Silencing PMCA4b in MCF-7 breast cancer cells had a similar effect, resulting in a dramatic loss of stress fibers. In addition, the PMCA4b expressing A375 cells maintained front-to-rear Ca2+ concentration gradient with the actin severing protein cofilin localizing to the lamellipodia, and preserved the integrity of the actin cytoskeleton from a destructive Ca2+ overload. We showed that both PMCA4b activity and trafficking were essential for the observed morphology and motility changes. In conclusion, our data suggest that PMCA4b plays a critical role in adopting front-to-rear polarity in a normally spindle-shaped cell type through F-actin rearrangement resulting in a less aggressive melanoma cell phenotype.
Collapse
Affiliation(s)
- Randa Naffa
- Department of Transfusiology, Semmelweis University, H-1089 Budapest, Hungary; (R.N.); (S.T.)
| | - Rita Padányi
- Department of Biophysics and Radiation Biology, Semmelweis University, H-1094 Budapest, Hungary;
| | - Attila Ignácz
- Department of Physiology and Neurobiology, Eötvös Loránd University, H-1117 Budapest, Hungary; (A.I.); (K.S.)
| | - Zoltán Hegyi
- Institute of Enzymology, Research Centre for Natural Sciences, Magyar Tudosok krt.2, H-1117 Budapest, Hungary; (Z.H.); (B.J.); (L.H.)
| | - Bálint Jezsó
- Institute of Enzymology, Research Centre for Natural Sciences, Magyar Tudosok krt.2, H-1117 Budapest, Hungary; (Z.H.); (B.J.); (L.H.)
| | - Sarolta Tóth
- Department of Transfusiology, Semmelweis University, H-1089 Budapest, Hungary; (R.N.); (S.T.)
- Department of Anatomy, Cell and Developmental Biology, Eötvös Loránd University, H-1117 Budapest, Hungary
| | | | - László Homolya
- Institute of Enzymology, Research Centre for Natural Sciences, Magyar Tudosok krt.2, H-1117 Budapest, Hungary; (Z.H.); (B.J.); (L.H.)
| | - Luca Hegedűs
- Department of Thoracic Surgery, Ruhrlandklinik, University Clinic Essen, 45239 Essen, Germany;
| | - Katalin Schlett
- Department of Physiology and Neurobiology, Eötvös Loránd University, H-1117 Budapest, Hungary; (A.I.); (K.S.)
| | - Agnes Enyedi
- Department of Transfusiology, Semmelweis University, H-1089 Budapest, Hungary; (R.N.); (S.T.)
- Correspondence:
| |
Collapse
|
89
|
Hetmanski JHR, Jones MC, Chunara F, Schwartz JM, Caswell PT. Combinatorial mathematical modelling approaches to interrogate rear retraction dynamics in 3D cell migration. PLoS Comput Biol 2021; 17:e1008213. [PMID: 33690598 PMCID: PMC7984637 DOI: 10.1371/journal.pcbi.1008213] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 03/22/2021] [Accepted: 02/05/2021] [Indexed: 12/17/2022] Open
Abstract
Cell migration in 3D microenvironments is a complex process which depends on the coordinated activity of leading edge protrusive force and rear retraction in a push-pull mechanism. While the potentiation of protrusions has been widely studied, the precise signalling and mechanical events that lead to retraction of the cell rear are much less well understood, particularly in physiological 3D extra-cellular matrix (ECM). We previously discovered that rear retraction in fast moving cells is a highly dynamic process involving the precise spatiotemporal interplay of mechanosensing by caveolae and signalling through RhoA. To further interrogate the dynamics of rear retraction, we have adopted three distinct mathematical modelling approaches here based on (i) Boolean logic, (ii) deterministic kinetic ordinary differential equations (ODEs) and (iii) stochastic simulations. The aims of this multi-faceted approach are twofold: firstly to derive new biological insight into cell rear dynamics via generation of testable hypotheses and predictions; and secondly to compare and contrast the distinct modelling approaches when used to describe the same, relatively under-studied system. Overall, our modelling approaches complement each other, suggesting that such a multi-faceted approach is more informative than methods based on a single modelling technique to interrogate biological systems. Whilst Boolean logic was not able to fully recapitulate the complexity of rear retraction signalling, an ODE model could make plausible population level predictions. Stochastic simulations added a further level of complexity by accurately mimicking previous experimental findings and acting as a single cell simulator. Our approach highlighted the unanticipated role for CDK1 in rear retraction, a prediction we confirmed experimentally. Moreover, our models led to a novel prediction regarding the potential existence of a 'set point' in local stiffness gradients that promotes polarisation and rapid rear retraction.
Collapse
Affiliation(s)
- Joseph H. R. Hetmanski
- Wellcome Trust Centre for Cell-Matrix Research, School of Biological Sciences, Faculty of Biology Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, United Kingdom
- * E-mail: (JHRH); (PTC)
| | - Matthew C. Jones
- Wellcome Trust Centre for Cell-Matrix Research, School of Biological Sciences, Faculty of Biology Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, United Kingdom
| | - Fatima Chunara
- Wellcome Trust Centre for Cell-Matrix Research, School of Biological Sciences, Faculty of Biology Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, United Kingdom
| | - Jean-Marc Schwartz
- Wellcome Trust Centre for Cell-Matrix Research, School of Biological Sciences, Faculty of Biology Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, United Kingdom
| | - Patrick T. Caswell
- Wellcome Trust Centre for Cell-Matrix Research, School of Biological Sciences, Faculty of Biology Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, United Kingdom
- * E-mail: (JHRH); (PTC)
| |
Collapse
|
90
|
Nrf2 regulates cell motility through RhoA-ROCK1 signalling in non-small-cell lung cancer cells. Sci Rep 2021; 11:1247. [PMID: 33441941 PMCID: PMC7806835 DOI: 10.1038/s41598-021-81021-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 12/30/2020] [Indexed: 02/06/2023] Open
Abstract
Nuclear factor-erythroid 2-related factor 2 (Nrf2) is a key transcriptional regulator of several antioxidant and anti-inflammatory enzymes. It binds to its endogenous inhibitor Kelch-like ECH-associated protein 1 (Keap1) in the cytoplasm under normal conditions. Various endogenous or environmental oxidative stresses can disrupt the Nrf2/Keap1 complex, allowing Nrf2 to translocate into the nucleus, where it induces the transcription of various cytoprotective enzymes by binding to antioxidant responsive elements. These enzymes have been reported to play a role in regulating tumour growth, angiogenesis, and chemoprevention. Invasion and migration are the most harmful aspects of cancer; they directly impacts the patients’ survival. Although the roles of Keap1/Nrf2 and their downstream genes in various cancers have been widely documented, their role in regulating cell motility still remains unclear, particularly in cancer cells. We observed that Nrf2 suppression following treatment with brusatol in non-small-cell lung cancer (NSCLC) cells with either exogenously introduced Keap1 or siNrf2 resulted in the inhibition of cell migration and invasion, with shrinking cell morphology due to decreased focal adhesions via inhibition of the RhoA–ROCK1 pathway. Nrf2 overexpression showed opposite results. Thus, the Nrf2/Keap1 pathway may affect cell motility by dysregulating the RhoA–ROCK1 signalling pathway in NSCLC.
Collapse
|
91
|
He L, Liu R, Yue H, Ren S, Zhu G, Guo Y, Qin C. Actin-granule formation is an additional step in cardiac myofibroblast differentiation. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:165. [PMID: 33569467 PMCID: PMC7867932 DOI: 10.21037/atm-20-8231] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Background Atrial fibrillation is the most common and long-lasting cardiac arrhythmia, and profoundly effects the daily lives of patients. The pathogenesis and persistence of atrial fibrillation is closely related to the cardiac fibroblast and its myofibroblast differentiation as increased collagen synthesis and migration capability. Thus better understanding of myofibroblast differentiation is essential for the prevention and treatment of atrial fibrillation. Methods Cardiac fibroblasts were isolated from neonatal rats and its actin structure was analyzed by immunofluorescence staining. Myofibroblast differentiation was induced by Angiotensin II (Ang II) and ROCK signaling related proteins were determined by western blot. Fasudil and Ricolinostat were employed to abrogate ROCK signaling and their effects on myofibroblast differentiation were assessed by IF microscopy and Celigo Image Cytometry. Results Stress actin fibers similar to actin filaments in myofibroblast differentiation are regulated by ROCK signaling, and our results also suggested Guanine nucleotide exchange factor-H1 (GEF-H1) phosphorylation could be induced by Ang II. In addition, Fasudil could down-regulate RhoA, GEF-H1, and phosphorylated GEF-H1 to inhibit ROCK signaling and further reduce Col I expression and the myofibroblast proportion. Conclusions An individual phase characterized by actin-granule formation was identified in cardiac myofibroblast differentiation. In the meanwhile, myofibroblast differentiation and its F-actin assembly could be detained in this phase by Fasudil abrogating the ROCK signaling pathway.
Collapse
Affiliation(s)
- Li He
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Ruiqi Liu
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Honghua Yue
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Shuofang Ren
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Guonian Zhu
- Research Core Facility of West China Hospital, Sichuan University, Chengdu, China
| | - Yingqiang Guo
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Chaoyi Qin
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
92
|
Tang H, Yi B, Wang X, Shen Y, Zhang Y. Understanding the cellular responses based on low-density electrospun fiber networks. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 119:111470. [PMID: 33321594 DOI: 10.1016/j.msec.2020.111470] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 08/14/2020] [Accepted: 08/25/2020] [Indexed: 01/12/2023]
Abstract
Fibers produced from electrospinning are well-known to be extremely fine with diameters ranging from tens of nanometers to a few microns. Such ultrafine fibers not only allow for engineering scaffolds resembling the ultrastructure of the native extracellular matrix, but also offer possibility to explore the remodeling behavior of cells in vitro, due to their mechanically 'adequate' softness endowed by their ultrafine fineness. However, the remodeling effect of cells on the biomimicking fibrous substrates remains to be understood, because the crisscrossing and entangling among nanofibers in those tightly packed fibrous mats ultimately lead to merely a topological phenomenon, similar to that of the nanofiber-like topography embossed on the surface of a solid matter. In this study, the effect of nanofiber density on cellular response behavior was investigated by reducing the density of electrospun fiber networks. Using polycaprolactone (PCL) as a model polymer, randomly oriented fiber networks with various densities, namely, 37.7 ± 16.3 μg/cm2 (D1), 103.8 ± 16.3 μg/cm2 (D2), 198.2 ± 40.0 μg/cm2 (D3), and 471.8 ± 32.7 μg/cm2 (D4), were prepared by electrospinning for varied collection durations (10 s, 50 s, 100 s, and 10 min, respectively). By examining the responsive behavior of the human induced pluripotent stem cell-derived mesenchymal stem cells (hiPS-MSCs) cultured on these nanofibrous networks, we showed that the fiber network with a moderate density (D2) is beneficial to the cell attachment, spreading, actin polymerization, contractility and migration. There also showed an increased tendency in nuclear localization of the Yes-associated protein (YAP) and subsequent activation of YAP responsive gene transcription, and cell proliferation and collagen synthesis were also enhanced on the D2. However, further increasing the fiber density (D3, D4) gave rise to weakened induction effect of fibers on the cellular responses. These results enrich our understanding on the effect of fiber density on cell behavior, and disclose the dependence of cellular responses on fiber density. This study paves the way to precisely design biomimetic fibrous scaffolds for achieving enhanced cell-scaffold interactions and tissue regeneration.
Collapse
Affiliation(s)
- Han Tang
- College of Chemistry, Chemical Engineering & Biotechnology, Donghua University, Shanghai 201620, China
| | - Bingcheng Yi
- College of Chemistry, Chemical Engineering & Biotechnology, Donghua University, Shanghai 201620, China
| | - Xianliu Wang
- College of Chemistry, Chemical Engineering & Biotechnology, Donghua University, Shanghai 201620, China
| | - Yanbing Shen
- College of Chemistry, Chemical Engineering & Biotechnology, Donghua University, Shanghai 201620, China
| | - Yanzhong Zhang
- College of Chemistry, Chemical Engineering & Biotechnology, Donghua University, Shanghai 201620, China; Key Lab of Science & Technology of Eco-Textile, Ministry of Education, Donghua University, Shanghai 201620, China; Shanghai Key Laboratory of Tissue Engineering, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; China Orthopaedic Regenerative Medicine Group (CORMed), Hangzhou 310058, China.
| |
Collapse
|
93
|
Strauss RE, Gourdie RG. Cx43 and the Actin Cytoskeleton: Novel Roles and Implications for Cell-Cell Junction-Based Barrier Function Regulation. Biomolecules 2020; 10:E1656. [PMID: 33321985 PMCID: PMC7764618 DOI: 10.3390/biom10121656] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 12/07/2020] [Accepted: 12/07/2020] [Indexed: 12/13/2022] Open
Abstract
Barrier function is a vital homeostatic mechanism employed by epithelial and endothelial tissue. Diseases across a wide range of tissue types involve dynamic changes in transcellular junctional complexes and the actin cytoskeleton in the regulation of substance exchange across tissue compartments. In this review, we focus on the contribution of the gap junction protein, Cx43, to the biophysical and biochemical regulation of barrier function. First, we introduce the structure and canonical channel-dependent functions of Cx43. Second, we define barrier function and examine the key molecular structures fundamental to its regulation. Third, we survey the literature on the channel-dependent roles of connexins in barrier function, with an emphasis on the role of Cx43 and the actin cytoskeleton. Lastly, we discuss findings on the channel-independent roles of Cx43 in its associations with the actin cytoskeleton and focal adhesion structures highlighted by PI3K signaling, in the potential modulation of cellular barriers. Mounting evidence of crosstalk between connexins, the cytoskeleton, focal adhesion complexes, and junctional structures has led to a growing appreciation of how barrier-modulating mechanisms may work together to effect solute and cellular flux across tissue boundaries. This new understanding could translate into improved therapeutic outcomes in the treatment of barrier-associated diseases.
Collapse
Affiliation(s)
- Randy E. Strauss
- Virginia Tech, Translational Biology Medicine and Health (TBMH) Program, Roanoke, VA 24016, USA
| | - Robert G. Gourdie
- Center for Heart and Reparative Medicine Research, Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA 24016, USA
- Virginia Tech Carilion School of Medicine, Roanoke, VA 24016, USA
- Department of Biomedical Engineering and Mechanics, Virginia Polytechnic Institute and State University, Blacksburg, VA 24060, USA
| |
Collapse
|
94
|
Boismal F, Serror K, Dobos G, Zuelgaray E, Bensussan A, Michel L. [Skin aging: Pathophysiology and innovative therapies]. Med Sci (Paris) 2020; 36:1163-1172. [PMID: 33296633 DOI: 10.1051/medsci/2020232] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
One of the major challenges of the 21st century is the fight against aging, defined as a set of physiological mechanisms altering the physical and intellectual capacities of human beings. Aging of the skin is only one visible part of this process. It is associated with major healing defects linked in part to the alteration of the biomechanical properties of skin cells, mainly dermal fibroblasts. The immune system, another key component in maintaining skin homeostasis and the efficient healing of wounds, also suffers the effects of time: the consequent skin immunosenescence would limit the anti-infectious and vaccine response, while promoting a pro-tumor environment. The main skin damages due to aging, whether intrinsic or extrinsic, will be detailed before listing the effective anti-aging strategies to combat age-related dermal and epidermal stigmas.
Collapse
Affiliation(s)
- Françoise Boismal
- Inserm U976 ; Centre de recherche sur la peau ; hôpital Saint-Louis, Paris, France
| | - Kevin Serror
- Service de chirurgie plastique et reconstructrice, hôpital Saint-Louis, Paris, France
| | - Gabor Dobos
- Inserm U976 ; Centre de recherche sur la peau ; hôpital Saint-Louis, Paris, France - Service de dermatologie, hôpital Saint Louis, Paris, France
| | - Elina Zuelgaray
- Inserm U976 ; Centre de recherche sur la peau ; hôpital Saint-Louis, Paris, France - Service de dermatologie, hôpital Saint Louis, Paris, France
| | - Armand Bensussan
- Inserm U976 ; Centre de recherche sur la peau ; hôpital Saint-Louis, Paris, France
| | - Laurence Michel
- Inserm U976 ; Centre de recherche sur la peau ; hôpital Saint-Louis, Paris, France - Service de dermatologie, hôpital Saint Louis, Paris, France
| |
Collapse
|
95
|
The Effects of Prolonged Storage on ARPE-19 Cells Stored at Three Different Storage Temperatures. Molecules 2020; 25:molecules25245809. [PMID: 33317020 PMCID: PMC7763992 DOI: 10.3390/molecules25245809] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 11/29/2020] [Accepted: 12/04/2020] [Indexed: 12/02/2022] Open
Abstract
This study aimed to investigate how prolonged storage of adult retinal pigment epithelial (ARPE-19) cell sheets affects cell metabolism, morphology, viability, and phenotype. ARPE-19 cell sheets were stored at three temperatures (4 °C, 16 °C, and 37 °C) for three weeks. Metabolic status and morphology of the cells were monitored by sampling medium and examining cells by phase-contrast microscopy, respectively, throughout the storage period. Cell viability was analyzed by flow cytometry, and phenotype was determined by epifluorescence microscopy after the storage. Lactate production and glucose consumption increased heavily, while pH dropped considerably, through storage at 37 °C compared to 4 °C and 16 °C. During storage, morphology started to deteriorate first at 4 °C, then at 37 °C, and was maintained the longest at 16 °C. Viability of the cells after three weeks of storage was best preserved at 16 °C, while cells stored at 4 °C and 37 °C had reduced viability. Dedifferentiation indicated by reduced expression of retinal pigment epithelium-specific protein 65 (RPE65), zonula occludens protein 1 (ZO-1), and occludin after three weeks of storage was noticed in all experimental groups compared to control. We conclude that storage temperature affects the metabolic status of ARPE-19 cells and that 16 °C reduces metabolic activity while protecting viability and morphology.
Collapse
|
96
|
Differentiated Daughter Cells Regulate Stem Cell Proliferation and Fate through Intra-tissue Tension. Cell Stem Cell 2020; 28:436-452.e5. [PMID: 33264636 DOI: 10.1016/j.stem.2020.11.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 09/30/2020] [Accepted: 11/04/2020] [Indexed: 02/06/2023]
Abstract
Basal stem cells fuel development, homeostasis, and regeneration of the epidermis. The proliferation and fate decisions of these cells are highly regulated by their microenvironment, including the basement membrane and underlying mesenchymal cells. Basal progenitors give rise to differentiated progeny that generate the epidermal barrier. Here, we present data that differentiated progeny also regulate the proliferation, differentiation, and migration of basal progenitor cells. Using two distinct mouse lines, we found that increasing contractility of differentiated cells resulted in non-cell-autonomous hyperproliferation of stem cells and prevented their commitment to a hair follicle lineage. This increased contractility also impaired movement of basal progenitors during hair placode morphogenesis and diminished migration of melanoblasts. These data suggest that intra-tissue tension regulates stem cell proliferation, fate decisions, and migration and that differentiated epidermal keratinocytes are a component of the stem cell niche that regulates development and homeostasis of the skin.
Collapse
|
97
|
Failler M, Giro-Perafita A, Owa M, Srivastava S, Yun C, Kahler DJ, Unutmaz D, Esteva FJ, Sánchez I, Dynlacht BD. Whole-genome screen identifies diverse pathways that negatively regulate ciliogenesis. Mol Biol Cell 2020; 32:169-185. [PMID: 33206585 PMCID: PMC8120696 DOI: 10.1091/mbc.e20-02-0111] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
We performed a high-throughput whole-genome RNAi screen to identify novel inhibitors of ciliogenesis in normal and basal breast cancer cells. Our screen uncovered a previously undisclosed, extensive network of genes linking integrin signaling and cellular adhesion to the extracellular matrix (ECM) with inhibition of ciliation in both normal and cancer cells. Surprisingly, a cohort of genes encoding ECM proteins was also identified. We characterized several ciliation inhibitory genes and showed that their silencing was accompanied by altered cytoskeletal organization and induction of ciliation, which restricts cell growth and migration in normal and breast cancer cells. Conversely, supplying an integrin ligand, vitronectin, to the ECM rescued the enhanced ciliation observed on silencing this gene. Aberrant ciliation could also be suppressed through hyperactivation of the YAP/TAZ pathway, indicating a potential mechanistic basis for our findings. Our findings suggest an unanticipated reciprocal relationship between ciliation and cellular adhesion to the ECM and provide a resource that could vastly expand our understanding of controls involving “outside-in” and “inside-out” signaling that restrain cilium assembly.
Collapse
Affiliation(s)
- Marion Failler
- Department of Pathology and Perlmutter Cancer Center, New York University School of Medicine, New York, NY 10016
| | - Ariadna Giro-Perafita
- Department of Pathology and Perlmutter Cancer Center, New York University School of Medicine, New York, NY 10016
| | - Mikito Owa
- Department of Pathology and Perlmutter Cancer Center, New York University School of Medicine, New York, NY 10016
| | - Shalini Srivastava
- Department of Pathology and Perlmutter Cancer Center, New York University School of Medicine, New York, NY 10016
| | - Chi Yun
- Department of Pathology and Perlmutter Cancer Center, New York University School of Medicine, New York, NY 10016
| | - David J Kahler
- Department of Pathology and Perlmutter Cancer Center, New York University School of Medicine, New York, NY 10016
| | - Derya Unutmaz
- Jackson Laboratory for Genomic Medicine and University of Connecticut School of Medicine, Farmington, CT 06031
| | - Francisco J Esteva
- Department of Pathology and Perlmutter Cancer Center, New York University School of Medicine, New York, NY 10016
| | - Irma Sánchez
- Department of Pathology and Perlmutter Cancer Center, New York University School of Medicine, New York, NY 10016
| | - Brian D Dynlacht
- Department of Pathology and Perlmutter Cancer Center, New York University School of Medicine, New York, NY 10016
| |
Collapse
|
98
|
In Vitro Evidences of Different Fibroblast Morpho-Functional Responses to Red, Near-Infrared and Violet-Blue Photobiomodulation: Clues for Addressing Wound Healing. APPLIED SCIENCES-BASEL 2020. [DOI: 10.3390/app10217878] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Although photobiomodulation (PBM) has proven promising to treat wounds, the lack of univocal guidelines and of a thorough understanding of light–tissue interactions hampers its mainstream adoption for wound healing promotion. This study compared murine and human fibroblast responses to PBM by red (635 ± 5 nm), near-infrared (NIR, 808 ± 1 nm), and violet-blue (405 ± 5 nm) light (0.4 J/cm2 energy density, 13 mW/cm2 power density). Cell viability was not altered by PBM treatments. Light and confocal laser scanning microscopy and biochemical analyses showed, in red PBM irradiated cells: F-actin assembly reduction, up-regulated expression of Ki67 proliferation marker and of vinculin in focal adhesions, type-1 collagen down-regulation, matrix metalloproteinase-2 and metalloproteinase-9 expression/functionality increase concomitant to their inhibitors (TIMP-1 and TIMP-2) decrease. Violet-blue and even more NIR PBM stimulated collagen expression/deposition and, likely, cell differentiation towards (proto)myofibroblast phenotype. Indeed, these cells exhibited a higher polygonal surface area, stress fiber-like structures, increased vinculin- and phospho-focal adhesion kinase-rich clusters and α-smooth muscle actin. This study may provide the experimental groundwork to support red, NIR, and violet-blue PBM as potential options to promote proliferative and matrix remodeling/maturation phases of wound healing, targeting fibroblasts, and to suggest the use of combined PBM treatments in the wound management setting.
Collapse
|
99
|
Lachowski D, Cortes E, Matellan C, Rice A, Lee DA, Thorpe SD, del Río Hernández AE. G Protein-Coupled Estrogen Receptor Regulates Actin Cytoskeleton Dynamics to Impair Cell Polarization. Front Cell Dev Biol 2020; 8:592628. [PMID: 33195261 PMCID: PMC7649801 DOI: 10.3389/fcell.2020.592628] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 09/24/2020] [Indexed: 12/30/2022] Open
Abstract
Mechanical forces regulate cell functions through multiple pathways. G protein-coupled estrogen receptor (GPER) is a seven-transmembrane receptor that is ubiquitously expressed across tissues and mediates the acute cellular response to estrogens. Here, we demonstrate an unidentified role of GPER as a cellular mechanoregulator. G protein-coupled estrogen receptor signaling controls the assembly of stress fibers, the dynamics of the associated focal adhesions, and cell polarization via RhoA GTPase (RhoA). G protein-coupled estrogen receptor activation inhibits F-actin polymerization and subsequently triggers a negative feedback that transcriptionally suppresses the expression of monomeric G-actin. Given the broad expression of GPER and the range of cytoskeletal changes modulated by this receptor, our findings position GPER as a key player in mechanotransduction.
Collapse
Affiliation(s)
- Dariusz Lachowski
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London, United Kingdom
| | - Ernesto Cortes
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London, United Kingdom
| | - Carlos Matellan
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London, United Kingdom
| | - Alistair Rice
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London, United Kingdom
| | - David A. Lee
- Institute of Bioengineering, School of Engineering and Material Science, Queen Mary University of London, London, United Kingdom
| | - Stephen D. Thorpe
- Institute of Bioengineering, School of Engineering and Material Science, Queen Mary University of London, London, United Kingdom
- UCD School of Medicine, UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Armando E. del Río Hernández
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London, United Kingdom
| |
Collapse
|
100
|
Yang X, Wang G, Huang X, Cheng M, Han Y. RNA-seq reveals the diverse effects of substrate stiffness on epidermal ovarian cancer cells. Aging (Albany NY) 2020; 12:20493-20511. [PMID: 33091877 PMCID: PMC7655203 DOI: 10.18632/aging.103906] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 07/21/2020] [Indexed: 12/27/2022]
Abstract
Background: Increasing evidence has confirmed that ovarian cancer is a mechanically responsive tumor both in vivo and in vitro. However, an understanding of the complete molecular mechanism involved in the response to substrate stiffness is lacking, as the associated transcriptome-wide effects have not been mapped. This limited understanding has restricted the identification of potential mechanically responsive targets in ovarian cancer. Results: To address these limitations, we used a polyacrylamide hydrogel system with a tunable Young’s modulus that broadly ranged from soft (1 kPa) to normal (6 kPa) and stiff (60 kPa) and investigated the effect of substrate rigidity on the morphology, spreading area, and cytoskeleton of SKOV-3 epidermal ovarian cancer (EOC) cells. RNA-seq analysis of these cells was then performed at appropriate timepoints to map the transcriptome-wide changes associated with stiffness sensing. We identified a large number of stiffness-sensing genes as well as many genes that were enriched in cancer-related pathways. Informed by these diverse expression results and based on bioinformatics analysis, we evaluated the hypothesis that PLEC and TNS2, which are located in focal adhesions and regulated by lnc-ZNF136, may play key roles in the EOC response to substrate stiffness. Conclusion: Overall, the results of the present study reveal previously unknown features of the EOC stiffness response and provide new insights into EOC metastasis in the clinic.
Collapse
Affiliation(s)
- Xiaoxu Yang
- School of Life Science and Technology, Weifang Medical University, Weifang 261053, Shandong, P.R. China
| | - Guohui Wang
- School of Life Science and Technology, Weifang Medical University, Weifang 261053, Shandong, P.R. China
| | - Xiaolei Huang
- School of Life Science and Technology, Weifang Medical University, Weifang 261053, Shandong, P.R. China
| | - Min Cheng
- Department of Physiology, Weifang Medical University, Weifang 261053, Shandong, P.R. China
| | - Yangyang Han
- School of Life Science and Technology, Weifang Medical University, Weifang 261053, Shandong, P.R. China
| |
Collapse
|