1
|
Xiao H, Gong X, Jordan SN, Liang Z, Mak M. Viscosity regulates cell spreading and cell-extracellular matrix interactions. FEBS J 2024. [PMID: 39529371 DOI: 10.1111/febs.17306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 05/16/2024] [Accepted: 10/15/2024] [Indexed: 11/16/2024]
Abstract
Fluid viscosity and osmolarity are among some of the underappreciated mechanical stimuli that cells can detect. Abnormal changes of multiple fluidic factors such as viscosity and osmolarity have been linked with diseases such as cystic fibrosis, cancer, and coronary heart disease. Changes in viscosity have been recently suggested as a regulator of cell locomotion. These novel studies focus on cell migration and spreading on glass substrates and through microchannels, and it remains a question whether viscosity impacts the cellular remodeling of extracellular matrices (ECMs). Here, we demonstrate that elevated viscosity induces cellular remodeling of collagen substrates and enhances cell spreading on ECM-mimetic substrates. Our results expand on recent work showing that viscosity induces increased cellular forces and demonstrates that viscosity can drive local ECM densification. Our data further show that microtubules, Ras-related C3 botulinum toxin substrate 1 (Rac1), actin-related protein 2/3 (Arp2/3) complex, Rho-associated protein kinase 1 (ROCK), and myosin are important regulators of viscosity-induced ECM remodeling. In the context of viscosity-induced cell spreading, cells cultured on glass and collagen substrates exhibit markedly different responses to pharmacological treatments, indicating that microtubules, Rac1, and Arp2/3 play distinct roles in regulating cellular spreading depending on the substrate. In addition, our results demonstrate that high osmotic pressures override viscosity-induced cell spreading by suppressing membrane ruffling. Our results demonstrate viscosity as a regulator of ECM remodeling and cell spreading in a fibrillar microenvironment. We also reveal a complex interplay between viscosity and osmolarity. We anticipate that our research can pave the way for future investigations into the crucial roles played by viscosity in both physiological and pathological conditions.
Collapse
Affiliation(s)
- Hugh Xiao
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
- Department of Pharmacological Sciences, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Xiangyu Gong
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Seyma Nayir Jordan
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Zixie Liang
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Michael Mak
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
- Department of Pharmacological Sciences, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| |
Collapse
|
2
|
Yang Z, Liu X, Li X, Abbate M, Rui H, Guan M, Sun Z. The destruction of cytoplasmic skeleton leads to the change of nuclear structure and the looseness of lamin A submicroscopic network. Heliyon 2024; 10:e36583. [PMID: 39309767 PMCID: PMC11414493 DOI: 10.1016/j.heliyon.2024.e36583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 08/14/2024] [Accepted: 08/19/2024] [Indexed: 09/25/2024] Open
Abstract
The interaction between lamin A and the cytoplasmic skeleton plays a key role in maintaining nuclear mechanical properties. However, the effect of destruction of the cytoplasmic skeleton on the 3D submicroscopic structure of lamin A has not been elucidated. In this study, we developed an image quantization algorithm to quantify changes in the submicroscopic structure of the intact lamin A 3D network within the nucleus. We used blebbistatin or nocodazole to disrupt the fibrillar structure of F-actin or tubulin, respectively, and then quantified changes in the lamin A super-resolution network structure, the morphological and mechanical properties of the nucleus and the spatial distribution of chromosomes. Ultimately, we found for the first time that disruption of the cytoplasmic skeleton changes the lamin A submicroscopic network and nuclear structural characteristics. In summary, this study contributes to understanding the trans-nuclear membrane interaction characteristics of lamin A and the cytoplasmic skeleton.
Collapse
Affiliation(s)
- Zhenyu Yang
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, China
| | - Xianglong Liu
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, China
| | - Xiaoliang Li
- ZEISS Research Microscopy Solutions, Shanghai, China
| | | | - Han Rui
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, China
| | - Miao Guan
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, China
| | - Zhenglong Sun
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, China
- Shenzhen Bay Laboratory, Shenzhen, China
| |
Collapse
|
3
|
Yang F, Chen P, Jiang H, Xie T, Shao Y, Kim DH, Li B, Sun Y. Directional Cell Migration Guided by a Strain Gradient. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2302404. [PMID: 37735983 PMCID: PMC11467785 DOI: 10.1002/smll.202302404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 08/27/2023] [Indexed: 09/23/2023]
Abstract
Strain gradients widely exist in development and physiological activities. The directional movement of cells is essential for proper cell localization, and directional cell migration in responses to gradients of chemicals, rigidity, density, and topography of extracellular matrices have been well-established. However; it is unclear whether strain gradients imposed on cells are sufficient to drive directional cell migration. In this work, a programmable uniaxial cell stretch device is developed that creates controllable strain gradients without changing substrate stiffness or ligand distributions. It is demonstrated that over 60% of the single rat embryonic fibroblasts migrate toward the lower strain side in static and the 0.1 Hz cyclic stretch conditions at ≈4% per mm strain gradients. It is confirmed that such responses are distinct from durotaxis or haptotaxis. Focal adhesion analysis confirms higher rates of contact area and protrusion formation on the lower strain side of the cell. A 2D extended motor-clutch model is developed to demonstrate that the strain-introduced traction force determines integrin fibronectin pairs' catch-release dynamics, which drives such directional migration. Together, these results establish strain gradient as a novel cue to regulate directional cell migration and may provide new insights in development and tissue repairs.
Collapse
Affiliation(s)
- Feiyu Yang
- Department of Mechanical and Industrial Engineering, University of Massachusetts Amherst, Amherst, MA, 01003, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Pengcheng Chen
- Department of Engineering Mechanics, Tsinghua University, Beijing, 100084, China
| | - Han Jiang
- Department of Mechanical and Industrial Engineering, University of Massachusetts Amherst, Amherst, MA, 01003, USA
| | - Tianfa Xie
- Department of Mechanical and Industrial Engineering, University of Massachusetts Amherst, Amherst, MA, 01003, USA
| | - Yue Shao
- Department of Engineering Mechanics, Tsinghua University, Beijing, 100084, China
| | - Deok-Ho Kim
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Bo Li
- Department of Engineering Mechanics, Tsinghua University, Beijing, 100084, China
| | - Yubing Sun
- Department of Mechanical and Industrial Engineering, University of Massachusetts Amherst, Amherst, MA, 01003, USA
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst, MA, 01003, USA
| |
Collapse
|
4
|
Ouderkirk S, Sedley A, Ong M, Shifflet MR, Harkrider QC, Wright NT, Miller CJ. A Perspective on Developing Modeling and Image Analysis Tools to Investigate Mechanosensing Proteins. Integr Comp Biol 2023; 63:1532-1542. [PMID: 37558388 PMCID: PMC10755202 DOI: 10.1093/icb/icad107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 07/17/2023] [Accepted: 07/17/2023] [Indexed: 08/11/2023] Open
Abstract
The shift of funding organizations to prioritize interdisciplinary work points to the need for workflow models that better accommodate interdisciplinary studies. Most scientists are trained in a specific field and are often unaware of the kind of insights that other disciplines could contribute to solving various problems. In this paper, we present a perspective on how we developed an experimental pipeline between a microscopy and image analysis/bioengineering lab. Specifically, we connected microscopy observations about a putative mechanosensing protein, obscurin, to image analysis techniques that quantify cell changes. While the individual methods used are well established (fluorescence microscopy; ImageJ WEKA and mTrack2 programs; MATLAB), there are no existing best practices for how to integrate these techniques into a cohesive, interdisciplinary narrative. Here, we describe a broadly applicable workflow of how microscopists can more easily quantify cell properties (e.g., perimeter, velocity) from microscopy videos of eukaryotic (MDCK) adherent cells. Additionally, we give examples of how these foundational measurements can create more complex, customizable cell mechanics tools and models.
Collapse
Affiliation(s)
- Stephanie Ouderkirk
- Department of Chemistry, James Madison University, Harrisonburg, VA 22807, USA
| | - Alex Sedley
- Department of Engineering, James Madison University, Harrisonburg, VA 22807, USA
| | - Mason Ong
- Department of Engineering, James Madison University, Harrisonburg, VA 22807, USA
| | - Mary Ruth Shifflet
- Department of Chemistry, Bridgewater College, Bridgewater, VA 22812, USA
| | - Quinn C Harkrider
- Department of Chemistry, James Madison University, Harrisonburg, VA 22807, USA
| | - Nathan T Wright
- Department of Chemistry, James Madison University, Harrisonburg, VA 22807, USA
| | - Callie J Miller
- Department of Engineering, James Madison University, Harrisonburg, VA 22807, USA
| |
Collapse
|
5
|
Tagay Y, Kheirabadi S, Ataie Z, Singh RK, Prince O, Nguyen A, Zhovmer AS, Ma X, Sheikhi A, Tsygankov D, Tabdanov ED. Dynein-Powered Cell Locomotion Guides Metastasis of Breast Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2302229. [PMID: 37726225 PMCID: PMC10625109 DOI: 10.1002/advs.202302229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 08/20/2023] [Indexed: 09/21/2023]
Abstract
The principal cause of death in cancer patients is metastasis, which remains an unresolved problem. Conventionally, metastatic dissemination is linked to actomyosin-driven cell locomotion. However, the locomotion of cancer cells often does not strictly line up with the measured actomyosin forces. Here, a complementary mechanism of metastatic locomotion powered by dynein-generated forces is identified. These forces arise within a non-stretchable microtubule network and drive persistent contact guidance of migrating cancer cells along the biomimetic collagen fibers. It is also shown that the dynein-powered locomotion becomes indispensable during invasive 3D migration within a tissue-like luminal network formed by spatially confining granular hydrogel scaffolds (GHS) made up of microscale hydrogel particles (microgels). These results indicate that the complementary motricity mediated by dynein is always necessary and, in certain instances, sufficient for disseminating metastatic breast cancer cells. These findings advance the fundamental understanding of cell locomotion mechanisms and expand the spectrum of clinical targets against metastasis.
Collapse
Affiliation(s)
- Yerbol Tagay
- Department of PharmacologyPenn State College of MedicineThe Pennsylvania State UniversityHersheyPA17033USA
| | - Sina Kheirabadi
- Department of Chemical EngineeringThe Pennsylvania State UniversityUniversity ParkPA16802USA
| | - Zaman Ataie
- Department of Chemical EngineeringThe Pennsylvania State UniversityUniversity ParkPA16802USA
| | - Rakesh K. Singh
- Department of Obstetrics & GynecologyGynecology OncologyUniversity of Rochester Medical CenterRochesterNY14642USA
| | - Olivia Prince
- Center for Biologics Evaluation and ResearchU.S. Food and Drug AdministrationSilver SpringMD20903USA
| | - Ashley Nguyen
- Center for Biologics Evaluation and ResearchU.S. Food and Drug AdministrationSilver SpringMD20903USA
| | - Alexander S. Zhovmer
- Center for Biologics Evaluation and ResearchU.S. Food and Drug AdministrationSilver SpringMD20903USA
| | - Xuefei Ma
- Center for Biologics Evaluation and ResearchU.S. Food and Drug AdministrationSilver SpringMD20903USA
| | - Amir Sheikhi
- Department of Chemical EngineeringThe Pennsylvania State UniversityUniversity ParkPA16802USA
- Department of Biomedical EngineeringThe Pennsylvania State UniversityUniversity ParkPA16802USA
| | - Denis Tsygankov
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of Technology and Emory UniversityAtlantaGA30332USA
| | - Erdem D. Tabdanov
- Department of PharmacologyPenn State College of MedicineThe Pennsylvania State UniversityHersheyPA17033USA
- Penn State Cancer InstitutePenn State College of MedicineThe Pennsylvania State UniversityHersheyPA17033USA
| |
Collapse
|
6
|
Chen X, Xia Y, Du W, Liu H, Hou R, Song Y, Xu W, Mao Y, Chen J. Contact Guidance Drives Upward Cellular Migration at the Mesoscopic Scale. Cell Mol Bioeng 2023; 16:205-218. [PMID: 37456789 PMCID: PMC10338420 DOI: 10.1007/s12195-023-00766-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 04/05/2023] [Indexed: 07/18/2023] Open
Abstract
Introduction Cancer metastasis is associated with increased cancer incidence, recurrence, and mortality. The role of cell contact guidance behaviors in cancer metastasis has been recognized but has not been elucidated yet. Methods The contact guidance behavior of cancer cells in response to topographical constraints is identified using microgrooved substrates with varying dimensions at the mesoscopic scale. Then, the cell morphology is determined to quantitatively analyze the effects of substrate dimensions on cells contact guidance. Cell density and migrate velocity signatures within the cellular population are determined using time-lapse phase-contrast microscopy. The effect of soluble factors concentration is determined by culturing cells upside down. Then, the effect of cell-substrate interaction on cell migration is investigated using traction force microscopy. Results With increasing depth and decreasing groove width, cell elongation and alignment are enhanced, while cell spreading is inhibited. Moreover, cells display preferential distribution on the ridges, which is found to be more pronounced with increasing depth and groove width. Determinations of cell density and migration velocity signatures reveal that the preferential distribution on ridges is caused by cell upward migration. Combined with traction force measurement, we find that migration toward ridges is governed by different cell-substrate interactions between grooves and ridges caused by geometrical constraints. Interestingly, the upward migration of cells at the mesoscopic scale is driven by entropic maximization. Conclusions The mesoscopic cell contact guidance mechanism based on the entropic force driven theory provides basic support for the study of cell alignment and migration along healthy tissues with varying size, thereby aiding in the prediction of cancer metastasis. Graphical Abstract Supplementary Information The online version contains supplementary material available at 10.1007/s12195-023-00766-y.
Collapse
Affiliation(s)
- Xiaoxiao Chen
- School of Advanced Manufacturing, Nanchang University, Nanchang, 330031 Jiangxi China
- Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, Hefei, 230027 Anhui China
| | - Youjun Xia
- Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, Hefei, 230027 Anhui China
- Key Laboratory of Precision Scientific Instrumentation of Anhui Higher Education Institutes, University of Science and Technology of China, Hefei, 230027 Anhui China
| | - Wenqiang Du
- Biomedical Engineering Department, University of Connecticut Health Center, Farmington, CT 06030 USA
| | - Han Liu
- School of Advanced Manufacturing, Nanchang University, Nanchang, 330031 Jiangxi China
| | - Ran Hou
- School of Advanced Manufacturing, Nanchang University, Nanchang, 330031 Jiangxi China
| | - Yiyu Song
- School of Advanced Manufacturing, Nanchang University, Nanchang, 330031 Jiangxi China
| | - Wenhu Xu
- School of Advanced Manufacturing, Nanchang University, Nanchang, 330031 Jiangxi China
| | - Yuxin Mao
- School of Biomedical Engineering, Anhui Medical University, Hefei, 230032 Anhui China
| | - Jianfeng Chen
- School of Advanced Manufacturing, Nanchang University, Nanchang, 330031 Jiangxi China
| |
Collapse
|
7
|
Tagay Y, Kheirabadi S, Ataie Z, Singh RK, Prince O, Nguyen A, Zhovmer AS, Ma X, Sheikhi A, Tsygankov D, Tabdanov ED. Dynein-Powered Cell Locomotion Guides Metastasis of Breast Cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.04.535605. [PMID: 37066378 PMCID: PMC10104034 DOI: 10.1101/2023.04.04.535605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
Metastasis is a principal cause of death in cancer patients, which remains an unresolved fundamental and clinical problem. Conventionally, metastatic dissemination is linked to the actomyosin-driven cell locomotion. However, locomotion of cancer cells often does not strictly line up with the measured actomyosin forces. Here, we identify a complementary mechanism of metastatic locomotion powered by the dynein-generated forces. These forces that arise within a non-stretchable microtubule network drive persistent contact guidance of migrating cancer cells along the biomimetic collagen fibers. We also show that dynein-powered locomotion becomes indispensable during invasive 3D migration within a tissue-like luminal network between spatially confining hydrogel microspheres. Our results indicate that the complementary contractile system of dynein motors and microtubules is always necessary and in certain instances completely sufficient for dissemination of metastatic breast cancer cells. These findings advance fundamental understanding of cell locomotion mechanisms and expand the spectrum of clinical targets against metastasis.
Collapse
Affiliation(s)
- Yerbol Tagay
- Department of Pharmacology, Penn State College of Medicine, The Pennsylvania State University, Hershey, PA, 17033, USA
| | - Sina Kheirabadi
- Department of Chemical Engineering, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Zaman Ataie
- Department of Chemical Engineering, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Rakesh K. Singh
- Department of Obstetrics & Gynecology, University of Rochester Medical Center, Rochester, NY, USA
| | - Olivia Prince
- Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, 20903, USA
| | - Ashley Nguyen
- Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, 20903, USA
| | - Alexander S. Zhovmer
- Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, 20903, USA
| | - Xuefei Ma
- Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, 20903, USA
| | - Amir Sheikhi
- Department of Chemical Engineering, The Pennsylvania State University, University Park, PA, 16802, USA
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Denis Tsygankov
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA
| | - Erdem D. Tabdanov
- Department of Pharmacology, Penn State College of Medicine, The Pennsylvania State University, Hershey, PA, 17033, USA
- Penn State Cancer Institute, Penn State College of Medicine, The Pennsylvania State University, Hershey, PA, 17033, USA
| |
Collapse
|
8
|
Ueda Y, Deguchi S. Emergence of multiple set-points of cellular homeostatic tension. J Biomech 2023; 151:111543. [PMID: 36931176 DOI: 10.1016/j.jbiomech.2023.111543] [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: 07/21/2022] [Revised: 02/01/2023] [Accepted: 03/07/2023] [Indexed: 03/13/2023]
Abstract
Stress fibers (SFs), a contractile actin bundle in nonmuscle mesenchymal cells, are known to intrinsically sustain a constant level of tension or tensional stress, a process called cellular tensional homeostasis. Malfunction in this homeostatic process has been implicated in many diseases such atherosclerosis, but its mechanisms remain incompletely understood. Interestingly, the homeostatic stress in individual SFs is altered upon recruitment of α-smooth muscle actin in particular cellular contexts to reinforce the preexisting SFs. While this transition of the set-point stress is somewhat a universal process observed across different cell types, no clear explanation has been provided as to why cells end up possessing different stable stresses. To address the underlying physics, here we describe that imposing a realistic assumption on the nature of SFs yields the presence of multiple set-points of the homeostatic stress, which transition among them depending on the magnitude of the cellular tension. We analytically derive non-dimensional parameters that characterize the extent of the transition and predict that SFs tend to acquire secondary stable stresses if they are subject to as large a change in stiffness as possible or to as immediate a transition as possible upon increasing the tension. This is a minimal and simple explanation, but given the frequent emergence of force-dependent transformation of various subcellular structures in addition to that of SFs, the theoretical concept presented here would offer an essential guide to addressing potential common mechanisms governing complicated cellular mechanobiological responses.
Collapse
Affiliation(s)
- Yuika Ueda
- Division of Bioengineering, Graduate School of Engineering Science, Osaka University, Japan
| | - Shinji Deguchi
- Division of Bioengineering, Graduate School of Engineering Science, Osaka University, Japan.
| |
Collapse
|
9
|
Onishi K, Ishihara S, Takahashi M, Sakai A, Enomoto A, Suzuki K, Haga H. Substrate stiffness induces nuclear localization of myosin regulatory light chain to suppress apoptosis. FEBS Lett 2023; 597:643-656. [PMID: 36723402 DOI: 10.1002/1873-3468.14592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/24/2022] [Accepted: 01/09/2023] [Indexed: 02/02/2023]
Abstract
Stiffness of the extracellular matrix regulates various biological responses, but the response mechanisms are poorly understood. Here, we found that the nuclear diphosphorylated myosin regulatory light chain (2P-MRLC) is a critical mechanomediator that suppresses apoptosis in response to substrate stiffness. Stiff substrates promoted the nuclear localization of 2P-MRLC. Zipper-interacting protein kinase [ZIPK; also known as death-associated protein kinase 3 (DAPK3)], a kinase for MRLC, was localized in the nucleus in response to stiff substrates and promoted the nuclear localization of 2P-MRLC. Moreover, actin fiber formation induced by substrate stiffness promoted the nuclear localization of 2P-MRLC via ZIPK. 2P-MRLC in response to substrate stiffness suppressed the expression of MAF bZIP transcription factor B (MafB) and repressed apoptosis. These findings reveal a newly identified role of MRLC in mechanotransduction.
Collapse
Affiliation(s)
- Katsuya Onishi
- Division of Soft Matter, Graduate School of Life Science, Hokkaido University, Sapporo, Japan
| | - Seiichiro Ishihara
- Department of Advanced Transdisciplinary Sciences, Faculty of Advanced Life Science, Hokkaido University, Sapporo, Japan
| | - Masayuki Takahashi
- Department of Chemistry, Faculty of Science, Hokkaido University, Sapporo, Japan
| | - Akihiro Sakai
- Department of Pathology, Nagoya University Graduate School of Medicine, Japan
| | - Atsushi Enomoto
- Department of Pathology, Nagoya University Graduate School of Medicine, Japan
| | - Kentaro Suzuki
- Department of Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University, Japan
| | - Hisashi Haga
- Department of Advanced Transdisciplinary Sciences, Faculty of Advanced Life Science, Hokkaido University, Sapporo, Japan
| |
Collapse
|
10
|
Saito T, Matsunaga D, Deguchi S. Long-term molecular turnover of actin stress fibers revealed by advection-reaction analysis in fluorescence recovery after photobleaching. PLoS One 2022; 17:e0276909. [PMCID: PMC9639824 DOI: 10.1371/journal.pone.0276909] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Accepted: 10/15/2022] [Indexed: 11/09/2022] Open
Abstract
Fluorescence recovery after photobleaching (FRAP) is a versatile technique to evaluate the intracellular molecular exchange called turnover. Mechanochemical models of FRAP typically consider the molecular diffusion and chemical reaction that simultaneously occur on a time scale of seconds to minutes. Particularly for long-term measurements, however, a mechanical advection effect can no longer be ignored, which transports the proteins in specific directions within the cells and accordingly shifts the spatial distribution of the local chemical equilibrium. Nevertheless, existing FRAP models have not considered the spatial shift, and as such, the turnover rate is often analyzed without considering the spatiotemporally updated chemical equilibrium. Here we develop a new FRAP model aimed at long-term measurements to quantitatively determine the two distinct effects of the advection and chemical reaction, i.e., the different major sources of the change in fluorescence intensity. To validate this approach, we carried out FRAP experiments on actin in stress fibers over a time period of more than 900 s, and the advection rate was shown to be comparable in magnitude to the chemical dissociation rate. We further found that the actin–myosin interaction and actin polymerization differently affect the advection and chemical dissociation. Our results suggest that the distinction between the two effects is indispensable to extract the intrinsic chemical properties of the actin cytoskeleton from the observations of complicated turnover in cells.
Collapse
Affiliation(s)
- Takumi Saito
- Division of Bioengineering, Graduate School of Engineering Science, Osaka University, Osaka, Japan
- Graduate School of Biomedical Engineering, Tohoku University, Tohoku, Japan
- JSPS Research Fellowship for Young Scientists, Tokyo, Japan
| | - Daiki Matsunaga
- Division of Bioengineering, Graduate School of Engineering Science, Osaka University, Osaka, Japan
| | - Shinji Deguchi
- Division of Bioengineering, Graduate School of Engineering Science, Osaka University, Osaka, Japan
- * E-mail:
| |
Collapse
|
11
|
Can Blebbistatin block the hypertrophy status in the zebrafish exvivo cardiac model? Biochim Biophys Acta Mol Basis Dis 2022; 1868:166471. [PMID: 35750268 DOI: 10.1016/j.bbadis.2022.166471] [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: 02/22/2022] [Revised: 05/31/2022] [Accepted: 06/16/2022] [Indexed: 11/23/2022]
Abstract
Ex-vivo simple models are powered tools to study cardiac hypertrophy. It is possible to control the activation of critical genes and thus test the effects of drug therapies before the in vivo tests. A zebrafish cardiac hypertrophy developed by 500 μM phenylephrine (PE) treatment in ex vivo culture has been demonstrated to activate the essential expression of the embryonal genes. These genes are the same as those described in several previous pieces of research on hypertrophic pathology in humans. The efficacy of the chemical drug Blebbistatin (BL) on hypertrophy induced ex vivo cultured hearts is studied in this research. BL can inhibit the myosins and the calcium wave in counteracting the hypertrophy status caused by PE. Samples treated with PE, BL and PE simultaneously, or pre/post-treatment with BL, have been analysed for the embryonal gene activation concerning the hypertrophy status. The qRTPCR has shown an inhibitory effect of BL treatments on the microRNAs downregulation with the consequent low expression of essential embryonal genes. In particular, BL seems to be effective in blocking the hyperplasia of the epicardium but less effective in myocardium hypertrophy. The model can make it possible to obtain knowledge on the transduction pathways activated by BL and investigate the potential use of this drug in treating cardiac hypertrophy in humans.
Collapse
|
12
|
Nanmo A, Yan L, Asaba T, Wan L, Kageyama T, Fukuda J. Bioprinting of hair follicle germs for hair regenerative medicine. Acta Biomater 2022:S1742-7061(22)00360-9. [PMID: 35718100 DOI: 10.1016/j.actbio.2022.06.021] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 05/31/2022] [Accepted: 06/08/2022] [Indexed: 11/18/2022]
Abstract
Hair regenerative medicine is a promising approach to treat hair loss. The replication of in vivo tissue configurations and microenvironments, such as hair follicle germs, has been studied to prepare tissue grafts for hair regenerative medicine. However, such approaches should be scalable, because a single patient with alopecia requires thousands of tissue grafts. In this paper, we propose an approach for the scalable and automated preparation of highly hair-inductive tissue grafts using a bioprinter. Two collagen droplets (2 µL each) containing mesenchymal and epithelial cells were placed adjacent to each other to fabricate hair-follicle-germ-like grafts. During three days of culture, the pairs of microgel beads were spontaneously contracted by cell traction forces, whereas the two cell types remained separated, where the densities of the cells and collagen were enriched more than 10 times. This approach allowed us to fabricate submillimeter objects printed with millimeter-order accuracy, facilitating scalable and automated tissue graft preparation. Because of mesenchymal-epithelial interactions, hair microgels (HMGs, i.e., collagen- and cell-enriched microgels) efficiently regenerate hair follicles and shafts when transplanted into the back skin of mice. However, the generated hair shafts mostly remain under the skin. Therefore, we printed microgel beads onto surgical suture guides arrayed on a stage. The microgel beads were contracted along with the suture guides in culture prior to transplantation. The guide-inserted HMGs significantly improved hair-shaft sprouting through the skin, owing to the control of the orientation of the HMGs transplanted into the skin. This approach is a promising strategy to advance hair regenerative medicine. STATEMENT OF SIGNIFICANCE: This study proposes an approach for the scalable and automated preparation of highly hair-inductive grafts using a bioprinter. Two collagen droplets containing mesenchymal and epithelial cells were placed adjacently. Cell traction forces caused the pairs of microgel beads to spontaneously contract in culture. Because of mesenchymal-epithelial interactions, hair microgels (HMGs) efficiently regenerated hair follicles on the back skin of mice. However, the generated hair shafts remained mostly beneath the skin. Therefore, we printed microgel beads onto surgical suture guides arrayed on a stage. The guide-inserted HMGs significantly improved hair-shaft sprouting through the skin owing to the control of the orientation of the HMGs in the skin. This approach represents a promising strategy for advancing hair regenerative medicine.
Collapse
Affiliation(s)
- Ayaka Nanmo
- Faculty of Engineering, Yokohama National University, 79-5 Tokiwadai, Hodogaya-ku, Yokohama, Kanagawa 240-8501, Japan
| | - Lei Yan
- Faculty of Engineering, Yokohama National University, 79-5 Tokiwadai, Hodogaya-ku, Yokohama, Kanagawa 240-8501, Japan
| | - Tomoki Asaba
- Faculty of Engineering, Yokohama National University, 79-5 Tokiwadai, Hodogaya-ku, Yokohama, Kanagawa 240-8501, Japan
| | - Licheng Wan
- Faculty of Engineering, Yokohama National University, 79-5 Tokiwadai, Hodogaya-ku, Yokohama, Kanagawa 240-8501, Japan
| | - Tatsuto Kageyama
- Faculty of Engineering, Yokohama National University, 79-5 Tokiwadai, Hodogaya-ku, Yokohama, Kanagawa 240-8501, Japan; Kanagawa Institute of Industrial Science and Technology, 3-2-1 Sakado Takatsu-ku, Kawasaki, Kanagawa 213-0012, Japan; Japan Science and Technology Agency (JST)-PRESTO, 4-1-8 Honcho, Kawaguchi, Saitama 332-0012, Japan
| | - Junji Fukuda
- Faculty of Engineering, Yokohama National University, 79-5 Tokiwadai, Hodogaya-ku, Yokohama, Kanagawa 240-8501, Japan; Kanagawa Institute of Industrial Science and Technology, 3-2-1 Sakado Takatsu-ku, Kawasaki, Kanagawa 213-0012, Japan.
| |
Collapse
|
13
|
Sakurai M, Weber P, Wolff G, Wieder A, Szendroedi J, Herzig S, Ekim Üstünel B. TSC22D4 promotes TGFβ1-induced activation of hepatic stellate cells. Biochem Biophys Res Commun 2022; 618:46-53. [PMID: 35714570 DOI: 10.1016/j.bbrc.2022.05.100] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 05/19/2022] [Accepted: 05/31/2022] [Indexed: 11/17/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD), non-alcoholic steatohepatitis (NASH) and liver fibrosis emerge as progressive liver diseases that accompany metabolic syndrome usually characterized by obesity, insulin resistance and type 2 diabetes. Currently no FDA approved treatments exist for the treatment of NASH and liver fibrosis, which requires a better knowledge of the underlying molecular mechanisms. TSC22D4 belongs to the TSC-22 protein family, the members of which are regulated by inflammatory and stress signals. Interestingly, patients with type 2 diabetes, with NAFLD as well as with NASH all have elevated levels of hepatic TSC22D4 expression. Previous studies with targeted deletion of TSC22D4 specifically in hepatocytes showed that TSC22D4 not only acts as a critical controller of diabetic hyperglycemia, but also contributes to NAFLD/NASH progression. To gain better insight into the development of progressive liver diseases, here we studied the function of TSC22D4 in hepatic stellate cells (HSCs), which play a key role in the pathogenesis of liver fibrosis. Our results indicated that TSC22D4 contributes to TGFβ1-mediated activation of HSCs and promotes their proliferation and migration. RNA-Sequencing analysis revealed that TSC22D4 initiates transcriptional events associated with HSC activation. Overall, our findings establish TSC22D4 as a key hub in the development of liver fibrosis, acting across different cellular compartments. Combinatorial TSC22D4 targeting in both hepatocytes and HSC may thus show superior efficacy against progressive liver disease.
Collapse
Affiliation(s)
- Minako Sakurai
- Institute for Diabetes and Cancer (IDC), Helmholtz Diabetes Center, Helmholtz Center, Munich, Neuherberg, Germany; Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine 1, Heidelberg University Hospital, Heidelberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Peter Weber
- Institute for Diabetes and Cancer (IDC), Helmholtz Diabetes Center, Helmholtz Center, Munich, Neuherberg, Germany; Research Unit Radiation Cytogenetics, Helmholtz Zentrum München Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH), Neuherberg, Germany; Clinical Cooperation Group "Personalized Radiotherapy in Head and Neck Cancer," Helmholtz Zentrum München Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH), Neuherberg, Germany
| | - Gretchen Wolff
- Institute for Diabetes and Cancer (IDC), Helmholtz Diabetes Center, Helmholtz Center, Munich, Neuherberg, Germany; Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine 1, Heidelberg University Hospital, Heidelberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Annika Wieder
- Institute for Diabetes and Cancer (IDC), Helmholtz Diabetes Center, Helmholtz Center, Munich, Neuherberg, Germany; Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine 1, Heidelberg University Hospital, Heidelberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Julia Szendroedi
- Institute for Diabetes and Cancer (IDC), Helmholtz Diabetes Center, Helmholtz Center, Munich, Neuherberg, Germany; Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine 1, Heidelberg University Hospital, Heidelberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Stephan Herzig
- Institute for Diabetes and Cancer (IDC), Helmholtz Diabetes Center, Helmholtz Center, Munich, Neuherberg, Germany; Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine 1, Heidelberg University Hospital, Heidelberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Bilgen Ekim Üstünel
- Institute for Diabetes and Cancer (IDC), Helmholtz Diabetes Center, Helmholtz Center, Munich, Neuherberg, Germany; Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine 1, Heidelberg University Hospital, Heidelberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany.
| |
Collapse
|
14
|
Sun J, Guo Y, Chen T, Jin T, Ma L, Ai L, Guo J, Niu Z, Yang R, Wang Q, Yu X, Gao H, Zhang Y, Su W, Song X, Ji W, Zhang Q, Huang M, Fan X, Du Z, Liang H. Systematic analyses identify the anti-fibrotic role of lncRNA TP53TG1 in IPF. Cell Death Dis 2022; 13:525. [PMID: 35661695 PMCID: PMC9166247 DOI: 10.1038/s41419-022-04975-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 05/18/2022] [Accepted: 05/25/2022] [Indexed: 01/21/2023]
Abstract
Long non-coding RNA (lncRNA) was reported to be a critical regulator of cellular homeostasis, but poorly understood in idiopathic pulmonary fibrosis (IPF). Here, we systematically identified a crucial lncRNA, p53-induced long non-coding RNA TP53 target 1 (TP53TG1), which was the dysregulated hub gene in IPF regulatory network and one of the top degree genes and down-regulated in IPF-drived fibroblasts. Functional experiments revealed that overexpression of TP53TG1 attenuated the increased expression of fibronectin 1 (Fn1), Collagen 1α1, Collagen 3α1, ACTA2 mRNA, Fn1, and Collagen I protein level, excessive fibroblasts proliferation, migration and differentiation induced by TGF-β1 in MRC-5 as well as PMLFs. In vivo assays identified that forced expression of TP53TG1 by adeno-associated virus 5 (AAV5) not only prevented BLM-induced experimental fibrosis but also reversed established lung fibrosis in the murine model. Mechanistically, TP53TG1 was found to bind to amount of tight junction proteins. Importantly, we found that TP53TG1 binds to the Myosin Heavy Chain 9 (MYH9) to inhibit its protein expression and thus the MYH9-mediated activation of fibroblasts. Collectively, we identified the TP53TG1 as a master suppressor of fibroblast activation and IPF, which could be a potential hub for targeting treatment of the disease.
Collapse
Affiliation(s)
- Jian Sun
- grid.258164.c0000 0004 1790 3548Zhuhai People’s Hospital, Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Hospital Affiliated With Jinan University, Jinan University, Zhuhai, 519000 Guangdong China ,grid.410736.70000 0001 2204 9268Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081 China
| | - Yingying Guo
- grid.410736.70000 0001 2204 9268Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081 China
| | - Tingting Chen
- grid.410736.70000 0001 2204 9268Department of Systems Biology, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081 China
| | - Tongzhu Jin
- grid.410736.70000 0001 2204 9268Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081 China
| | - Lu Ma
- grid.410736.70000 0001 2204 9268Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081 China
| | - Liqiang Ai
- grid.410736.70000 0001 2204 9268Department of Systems Biology, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081 China
| | - Jiayu Guo
- grid.410736.70000 0001 2204 9268Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081 China
| | - Zhihui Niu
- grid.410736.70000 0001 2204 9268Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081 China
| | - Ruoxuan Yang
- grid.410736.70000 0001 2204 9268Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081 China
| | - Qianqian Wang
- grid.410736.70000 0001 2204 9268Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081 China
| | - Xiaojiang Yu
- grid.410736.70000 0001 2204 9268Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081 China
| | - Huiying Gao
- grid.410736.70000 0001 2204 9268Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081 China
| | - Yuhan Zhang
- grid.410736.70000 0001 2204 9268Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081 China
| | - Wei Su
- grid.410736.70000 0001 2204 9268Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081 China
| | - Xiaoying Song
- grid.410736.70000 0001 2204 9268Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081 China
| | - Weihang Ji
- grid.410736.70000 0001 2204 9268Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081 China
| | - Qing Zhang
- grid.410736.70000 0001 2204 9268Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081 China
| | - Mengqin Huang
- grid.410736.70000 0001 2204 9268Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081 China
| | - Xingxing Fan
- grid.259384.10000 0000 8945 4455State Key Laboratory of Quality Research in Chinese Medicine/Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau (SAR), China
| | - Zhimin Du
- grid.258164.c0000 0004 1790 3548Zhuhai People’s Hospital, Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Hospital Affiliated With Jinan University, Jinan University, Zhuhai, 519000 Guangdong China ,grid.410736.70000 0001 2204 9268Institute of Clinical Pharmacy, the 2nd Affiliated Hospital, Harbin Medical University, Harbin, 150081 China
| | - Haihai Liang
- grid.258164.c0000 0004 1790 3548Zhuhai People’s Hospital, Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Hospital Affiliated With Jinan University, Jinan University, Zhuhai, 519000 Guangdong China ,grid.410736.70000 0001 2204 9268Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081 China ,Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, 150081 China
| |
Collapse
|
15
|
Matozo T, Kogachi L, de Alencar BC. Myosin motors on the pathway of viral infections. Cytoskeleton (Hoboken) 2022; 79:41-63. [PMID: 35842902 DOI: 10.1002/cm.21718] [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: 04/27/2022] [Revised: 06/25/2022] [Accepted: 07/07/2022] [Indexed: 01/30/2023]
Abstract
Molecular motors are microscopic machines that use energy from adenosine triphosphate (ATP) hydrolysis to generate movement. While kinesins and dynein are molecular motors associated with microtubule tracks, myosins bind to and move on actin filaments. Mammalian cells express several myosin motors. They power cellular processes such as endo- and exocytosis, intracellular trafficking, transcription, migration, and cytokinesis. As viruses navigate through cells, they may take advantage or be hindered by host components and machinery, including the cytoskeleton. This review delves into myosins' cell roles and compares them to their reported functions in viral infections. In most cases, the previously described myosin functions align with their reported role in viral infections, although not in all cases. This opens the possibility that knowledge obtained from studying myosins in viral infections might shed light on new physiological roles for myosins in cells. However, given the high number of myosins expressed and the variety of viruses investigated in the different studies, it is challenging to infer whether the interactions found are specific to a single virus or can be applied to other viruses with the same characteristics. We conclude that the participation of myosins in viral cycles is still a largely unexplored area, especially concerning unconventional myosins.
Collapse
Affiliation(s)
- Tais Matozo
- Departamento de Imunologia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Leticia Kogachi
- Departamento de Imunologia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Bruna Cunha de Alencar
- Departamento de Imunologia, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
16
|
Yu X, Elfimova N, Müller M, Bachurski D, Koitzsch U, Drebber U, Mahabir E, Hansen HP, Friedman SL, Klein S, Dienes HP, Hösel M, Buettner R, Trebicka J, Kondylis V, Mannaerts I, Odenthal M. Autophagy-Related Activation of Hepatic Stellate Cells Reduces Cellular miR-29a by Promoting Its Vesicular Secretion. Cell Mol Gastroenterol Hepatol 2022; 13:1701-1716. [PMID: 35219894 PMCID: PMC9046234 DOI: 10.1016/j.jcmgh.2022.02.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 02/16/2022] [Accepted: 02/17/2022] [Indexed: 01/07/2023]
Abstract
BACKGROUND & AIMS Liver fibrosis arises from long-term chronic liver injury, accompanied by an accelerated wound healing response with interstitial accumulation of extracellular matrix (ECM). Activated hepatic stellate cells (HSC) are the main source for ECM production. MicroRNA29a (miR-29a) is a crucial antifibrotic miRNA that is repressed during fibrosis, resulting in up-regulation of collagen synthesis. METHODS Intracellular and extracellular miRNA levels of primary and immortalized myofibroblastic HSC in response to profibrogenic stimulation by transforming growth factor β (TGFβ) or platelet-derived growth factor-BB (PDGF-BB) or upon inhibition of vesicular transport and autophagy processes were determined by quantitative polymerase chain reaction. Autophagy flux was studied by electron microscopy, flow cytometry, immunoblotting, and immunocytochemistry. Hepatic and serum miR-29a levels were quantified by using both liver tissue and serum samples from a cohort of chronic hepatitis C virus patients and a murine CCl4 induced liver fibrosis model. RESULTS In our study, we show that TGFβ and PDGF-BB resulted in decrease of intracellular miR-29a and a pronounced increase of vesicular miR-29a release into the supernatant. Strikingly, miR-29a vesicular release was accompanied by enhanced autophagic activity and up-regulation of the autophagy marker protein LC3. Moreover, autophagy inhibition strongly prevented miR-29a secretion and repressed its targets' expression such as Col1A1. Consistently, hepatic miR-29a loss and increased LC3 expression in myofibroblastic HSC were associated with increased serum miR-29a levels in CCl4-treated murine liver fibrosis and specimens of hepatitis C virus patients with chronic liver disease. CONCLUSIONS We provide evidence that activation-associated autophagy in HSC induces release of miR-29a, whereas inhibition of autophagy represses fibrogenic gene expression in part through attenuated miR-29a secretion.
Collapse
Affiliation(s)
- Xiaojie Yu
- Institute for Pathology, Medical Faculty and University Hospital of Cologne, University of Cologne, Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Natalia Elfimova
- Institute for Pathology, Medical Faculty and University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Marion Müller
- Institute for Pathology, Medical Faculty and University Hospital of Cologne, University of Cologne, Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Daniel Bachurski
- Department I of Internal Medicine, Medical Faculty and University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Ulrike Koitzsch
- Institute for Pathology, Medical Faculty and University Hospital of Cologne, University of Cologne, Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Uta Drebber
- Institute for Pathology, Medical Faculty and University Hospital of Cologne, University of Cologne, Cologne, Germany; Center of Integrative Oncology, University Clinic of Cologne and Bonn, Germany
| | - Esther Mahabir
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Hinrich P Hansen
- Department I of Internal Medicine, Medical Faculty and University Hospital of Cologne, University of Cologne, Cologne, Germany; Center of Integrative Oncology, University Clinic of Cologne and Bonn, Germany
| | - Scott L Friedman
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Sabine Klein
- Department of Internal Medicine I, University Hospital Frankfurt, Frankfurt, Germany
| | - Hans Peter Dienes
- Institute for Pathology, Medical Faculty and University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Marianna Hösel
- Institute for Pathology, Medical Faculty and University Hospital of Cologne, University of Cologne, Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Reinhard Buettner
- Institute for Pathology, Medical Faculty and University Hospital of Cologne, University of Cologne, Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany; Center of Integrative Oncology, University Clinic of Cologne and Bonn, Germany
| | - Jonel Trebicka
- Department of Internal Medicine I, University Hospital Frankfurt, Frankfurt, Germany; European Foundation for the Study of Chronic Liver Failure - EF CLIF, Barcelona, Spain
| | - Vangelis Kondylis
- Institute for Pathology, Medical Faculty and University Hospital of Cologne, University of Cologne, Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Inge Mannaerts
- Liver Cell Biology Research Group, Vrije Universiteit Brussel, Brussel, Belgium.
| | - Margarete Odenthal
- Institute for Pathology, Medical Faculty and University Hospital of Cologne, University of Cologne, Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany; Center of Integrative Oncology, University Clinic of Cologne and Bonn, Germany.
| |
Collapse
|
17
|
He ZQ, Yuan XW, Lu ZB, Li YH, Li YF, Liu X, Wang L, Zhang Y, Zhou Q, Li W. Pharmacological regulation of tissue fibrosis by targeting the mechanical contraction of myofibroblasts. FUNDAMENTAL RESEARCH 2022; 2:37-47. [PMID: 38933917 PMCID: PMC11197686 DOI: 10.1016/j.fmre.2021.11.033] [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: 09/17/2021] [Revised: 11/26/2021] [Accepted: 11/30/2021] [Indexed: 11/22/2022] Open
Abstract
Fibrosis can occur in almost all tissues and organs and affects normal physiological function, which may have serious consequences, such as organ failure. However, there are currently no effective, broad-spectrum drugs suitable for clinical application. Revealing the process of fibrosis is an important prerequisite for the development of new therapeutic targets and drugs. Studies have shown that the limiting of myofibroblast activation or the promoting of their elimination can ameliorate fibrosis. However, it has not been reported whether a direct decrease in cell contraction can inhibit fibrosis in vivo. Here, we have shown that (-)-blebbistatin (Ble), a non-muscle myosin Ⅱ inhibitor, displayed significant inhibition of liver fibrosis in different chronic injury mouse models in vivo. We found that Ble reduced the stiffness of fibrotic tissues from the early stage, which reduced the extent of myofibroblast activation induced by a stiffer extracellular matrix (ECM). Moreover, Ble also reduced the activation of myofibroblasts induced by TGF-β1, which is the most potent pro-fibrotic cytokine. Mechanistically, Ble reduced mechanical contraction, which inhibited the assembly of stress fibers, decreased the F/G-actin ratio, and led to the exnucleation of YAP1 and MRTF-A. Finally, we verified its broad-spectrum antifibrotic effect in multiple models of organ fibrosis. Our results highlighted the important role of mechanical contraction in myofibroblast activation and maintenance, rather than just a characteristic of activation, suggesting that it may be a potential target to explore broad-spectrum drugs for the treatment of fibrotic diseases.
Collapse
Affiliation(s)
- Zheng-Quan He
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Institute for Stem Cell and Regenerative Medicine, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Chinese Academy of Sciences, Beijing 100101, China
| | - Xue-Wei Yuan
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Institute for Stem Cell and Regenerative Medicine, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Chinese Academy of Sciences, Beijing 100101, China
| | - Zong-Bao Lu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Institute for Stem Cell and Regenerative Medicine, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yu-Huan Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- The First Hospital of Jilin University, Changchun Jilin 130021, China
| | - Yu-Fei Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Institute for Stem Cell and Regenerative Medicine, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Chinese Academy of Sciences, Beijing 100101, China
| | - Xin Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Institute for Stem Cell and Regenerative Medicine, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Liu Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Institute for Stem Cell and Regenerative Medicine, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ying Zhang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Institute for Stem Cell and Regenerative Medicine, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qi Zhou
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Institute for Stem Cell and Regenerative Medicine, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wei Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Institute for Stem Cell and Regenerative Medicine, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
18
|
Ehlinger C, Mathieu E, Rabineau M, Ball V, Lavalle P, Haikel Y, Vautier D, Kocgozlu L. Insensitivity of dental pulp stem cells migration to substrate stiffness. Biomaterials 2021; 275:120969. [PMID: 34157563 DOI: 10.1016/j.biomaterials.2021.120969] [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] [Received: 04/06/2021] [Revised: 05/26/2021] [Accepted: 06/09/2021] [Indexed: 12/16/2022]
Abstract
Dental pulp stem cells (DPSCs) are a promising cell source for regeneration of dental pulp. Migration is a key event but influence of the microenvironment rigidity (5 kPa at the center of dental pulp to 20 GPa for the dentin) is largely unknown. Mechanical signals are transmitted from the extracellular matrix to the cytoskeleton, to the nuclei, and to the chromatin, potentially regulating gene expression. To identify the microenvironmental influence on migration, we analyzed motility on PDMS substrates with stiffness increasing from 1.5 kPa up to 2.5 MPa. We found that migration speed slightly increases as substrate stiffness decreases in correlation with decreasing focal adhesion size. Motility is relatively insensitive to substrate stiffness, even on a bi-rigidity PDMS substrate where DPSCs migrate without preferential direction. Migration is independent of both myosin II activity and YAP translocation after myosin II inhibition. Additionally, inhibition of Arp2/3 complex leads to significant speed decrease for all rigidities, suggesting contribution of the lamellipodia in the migration. Interestingly, the chromatin architecture remains stable after a 7-days exposure on the PDMS substrates for all rigidity. To design scaffold mimicking dental pulp environment, similar DPSCs migration for all rigidity, leaves field open to choose this mechanical parameter.
Collapse
Affiliation(s)
- Claire Ehlinger
- Inserm UMR-S1121, Centre de Recherche en Biomédecine de Strasbourg (CRBS), 1 rue Eugène Boeckel, 67084, Strasbourg, France; Université de Strasbourg, Faculté de Chirurgie Dentaire, 8 rue Sainte Elisabeth, 67000, Strasbourg, France; Fédération de Médecine Translationnelle, Strasbourg, France
| | - Eric Mathieu
- Inserm UMR-S1121, Centre de Recherche en Biomédecine de Strasbourg (CRBS), 1 rue Eugène Boeckel, 67084, Strasbourg, France; Université de Strasbourg, Faculté de Chirurgie Dentaire, 8 rue Sainte Elisabeth, 67000, Strasbourg, France; Fédération de Médecine Translationnelle, Strasbourg, France
| | - Morgane Rabineau
- Inserm UMR-S1121, Centre de Recherche en Biomédecine de Strasbourg (CRBS), 1 rue Eugène Boeckel, 67084, Strasbourg, France; Université de Strasbourg, Faculté de Chirurgie Dentaire, 8 rue Sainte Elisabeth, 67000, Strasbourg, France; Fédération de Médecine Translationnelle, Strasbourg, France
| | - Vincent Ball
- Inserm UMR-S1121, Centre de Recherche en Biomédecine de Strasbourg (CRBS), 1 rue Eugène Boeckel, 67084, Strasbourg, France; Université de Strasbourg, Faculté de Chirurgie Dentaire, 8 rue Sainte Elisabeth, 67000, Strasbourg, France; Fédération de Médecine Translationnelle, Strasbourg, France
| | - Philippe Lavalle
- Inserm UMR-S1121, Centre de Recherche en Biomédecine de Strasbourg (CRBS), 1 rue Eugène Boeckel, 67084, Strasbourg, France; Université de Strasbourg, Faculté de Chirurgie Dentaire, 8 rue Sainte Elisabeth, 67000, Strasbourg, France; Fédération de Médecine Translationnelle, Strasbourg, France
| | - Youssef Haikel
- Inserm UMR-S1121, Centre de Recherche en Biomédecine de Strasbourg (CRBS), 1 rue Eugène Boeckel, 67084, Strasbourg, France; Université de Strasbourg, Faculté de Chirurgie Dentaire, 8 rue Sainte Elisabeth, 67000, Strasbourg, France; Fédération de Médecine Translationnelle, Strasbourg, France
| | - Dominique Vautier
- Inserm UMR-S1121, Centre de Recherche en Biomédecine de Strasbourg (CRBS), 1 rue Eugène Boeckel, 67084, Strasbourg, France; Université de Strasbourg, Faculté de Chirurgie Dentaire, 8 rue Sainte Elisabeth, 67000, Strasbourg, France; Fédération de Médecine Translationnelle, Strasbourg, France.
| | - Leyla Kocgozlu
- Inserm UMR-S1121, Centre de Recherche en Biomédecine de Strasbourg (CRBS), 1 rue Eugène Boeckel, 67084, Strasbourg, France; Université de Strasbourg, Faculté de Chirurgie Dentaire, 8 rue Sainte Elisabeth, 67000, Strasbourg, France; Fédération de Médecine Translationnelle, Strasbourg, France.
| |
Collapse
|
19
|
Li Z, Bratlie KM. Fibroblasts treated with macrophage conditioned medium results in phenotypic shifts and changes in collagen organization. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 122:111915. [PMID: 33641908 DOI: 10.1016/j.msec.2021.111915] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 12/30/2020] [Accepted: 01/23/2021] [Indexed: 01/08/2023]
Abstract
In tissue regeneration, the goal is to regenerate tissue similar to what was damaged or missing while preventing fibrotic scarring, which may lead to decreased mechanical strength and dissimilar tissue characteristics compared to native tissue. We believe collagen orientation plays a critical role in wound contraction and scarring and that it is modulated by myofibroblasts. We used macrophage conditioned medium to simulate complex events that can influence the fibroblast phenotype during the wound healing process. In addition to examining the effect of macrophage phenotype on fibroblasts, we inhibited focal adhesion kinase (FAK), Rho-associated protein kinase (ROCK), and myosin II for fibroblasts cultured on both tissue culture plastic and methacrylated gellan gum to understand how different pathways and materials influence fibroblast responses. Collagen orientation, α-SMA expression, focal adhesion area, and cell migration were altered by inhibition of FAK, ROCK, or myosin II and macrophage phenotype, along with the substrate. An increase in either focal adhesion area or α-smooth muscle actin (α-SMA) expression correlated with an aligned collagen orientation. Gellan gum hydrogels upregulated α-SMA expression in ROCK inhibited conditioned media and downregulated the FAK area in FAK and ROCK inhibited conditioned media. Myosin II had no impact on the α-SMA expression on the substrate compared to coverslip except for M2 conditioned medium. Gellan gum hydrogel significantly increased cell migration under FAK and Myosin II mediated conditioned media and unconditioned media. Collectively, our study examined how macrophage phenotype influences fibroblast response, which would be beneficial in controlling scar tissue formation.
Collapse
Affiliation(s)
- Zhuqing Li
- Department of Materials Science & Engineering, Iowa State University, Ames, IA 50011, USA
| | - Kaitlin M Bratlie
- Department of Materials Science & Engineering, Iowa State University, Ames, IA 50011, USA; Department of Chemical & Biological Engineering, Iowa State University, Ames, IA 50011, USA.
| |
Collapse
|
20
|
Sun X, Zhu M, Chen X, Jiang X. MYH9 Inhibition Suppresses TGF-β1-Stimulated Lung Fibroblast-to-Myofibroblast Differentiation. Front Pharmacol 2021; 11:573524. [PMID: 33519439 PMCID: PMC7838063 DOI: 10.3389/fphar.2020.573524] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 11/05/2020] [Indexed: 12/04/2022] Open
Abstract
Previous cDNA microarray results showed that MYH9 gene expression levels are increased in TGF-β1-stimulated lung fibroblast. Recently, our proteomic results revealed that the expression levels of MYH9 protein are notably upregulated in lung tissues of bleomycin-treated rats. However, whether MYH9 plays a critical role in the differentiation of fibroblast remains unclear. Herein, we demonstrated that TGF-β1 increased MYH9 expression, and siRNA-mediated knockdown of MYH9 and pharmacological inhibition of MYH9 ATPase activity remarkably repressed TGF-β1-induced lung fibroblast-to-myofibroblast differentiation. TGF-β1-stimulated MYH9 induction might be via ALK5/Smad2/3 pathway but not through noncanonical pathways, including p38 mitogen-activated kinase, and Akt pathways in lung fibroblasts. Our results showed that MYH9 inhibition suppressed TGF-β1-induced lung fibroblast-to-myofibroblast differentiation, which provides valuable information for illuminating the pathological mechanisms of lung fibroblast differentiation, and gives clues for finding new potential target for pulmonary fibrosis treatment.
Collapse
Affiliation(s)
- Xionghua Sun
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Mei Zhu
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Xihua Chen
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Xiaogang Jiang
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| |
Collapse
|
21
|
Pedrosa AT, Murphy KN, Nogueira AT, Brinkworth AJ, Thwaites TR, Aaron J, Chew TL, Carabeo RA. A post-invasion role for Chlamydia type III effector TarP in modulating the dynamics and organization of host cell focal adhesions. J Biol Chem 2020; 295:14763-14779. [PMID: 32843479 PMCID: PMC7586217 DOI: 10.1074/jbc.ra120.015219] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 08/12/2020] [Indexed: 01/09/2023] Open
Abstract
The human pathogen Chlamydia trachomatis targets epithelial cells lining the genital mucosa. We observed that infection of various cell types, including fibroblasts and epithelial cells resulted in the formation of unusually stable and mature focal adhesions that resisted disassembly induced by the myosin II inhibitor, blebbistatin. Superresolution microscopy revealed in infected cells the vertical displacement of paxillin and focal adhesion kinase from the signaling layer of focal adhesions, whereas vinculin remained in its normal position within the force transduction layer. The candidate type III effector TarP, which localized to focal adhesions during infection and when expressed ectopically, was sufficient to mimic both the reorganization and blebbistatin-resistant phenotypes. These effects of TarP, including its localization to focal adhesions, required a post-invasion interaction with the host protein vinculin through a specific domain at the C terminus of TarP. This interaction is repurposed from an actin-recruiting and -remodeling complex to one that mediates nanoarchitectural and dynamic changes of focal adhesions. The consequence of Chlamydia-stabilized focal adhesions was restricted cell motility and enhanced attachment to the extracellular matrix. Thus, via a novel mechanism, Chlamydia inserts TarP within focal adhesions to alter their organization and stability.
Collapse
Affiliation(s)
- António T Pedrosa
- Bacteriology Section, Programme in Microbiology, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Korinn N Murphy
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska, USA; School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, Washington, USA
| | - Ana T Nogueira
- Bacteriology Section, Programme in Microbiology, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Amanda J Brinkworth
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska, USA; School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, Washington, USA
| | - Tristan R Thwaites
- Bacteriology Section, Programme in Microbiology, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Jesse Aaron
- Advanced Imaging Center, Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, Virginia, USA
| | - Teng-Leong Chew
- Advanced Imaging Center, Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, Virginia, USA
| | - Rey A Carabeo
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska, USA.
| |
Collapse
|
22
|
Kuntze A, Goetsch O, Fels B, Najder K, Unger A, Wilhelmi M, Sargin S, Schimmelpfennig S, Neumann I, Schwab A, Pethő Z. Protonation of Piezo1 Impairs Cell-Matrix Interactions of Pancreatic Stellate Cells. Front Physiol 2020; 11:89. [PMID: 32116794 PMCID: PMC7033545 DOI: 10.3389/fphys.2020.00089] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 01/27/2020] [Indexed: 12/12/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is characterized by an acidic and fibrotic stroma. The extracellular matrix (ECM) causing the fibrosis is primarily formed by pancreatic stellate cells (PSCs). The effects of the altered biomechanics and pH landscape in the pathogenesis of PDAC, however, are poorly understood. Mechanotransduction in cells has been linked to the function of mechanosensitive ion channels such as Piezo1. Here, we tested whether this channel plays crucial roles in transducing mechanical signals in the acidic PDAC microenvironment. We performed immunofluorescence, Ca2+ influx and intracellular pH measurements in PSCs and complemented them by live-cell imaging migration experiments in order to assess the function of Piezo1 channels in PSCs. We evaluated whether Piezo1 responds to changes of extracellular and/or intracellular pH in the pathophysiological range (pH 6.6 and pH 6.9, respectively). We validated our results using Piezo1-transfected HEK293 cells as a model system. Indeed, acidification of the intracellular space severely inhibits Piezo1-mediated Ca2+ influx into PSCs. In addition, stimulation of Piezo1 channels with its activator Yoda1 accelerates migration of PSCs on a two-dimensional ECM as well as in a 3D setting. Furthermore, Yoda1-activated PSCs transmit more force to the surrounding ECM under physiological pH, as revealed by measuring the dislocation of microbeads embedded in the surrounding matrix. This is paralleled by an enhanced phosphorylation of myosin light chain isoform 9 after Piezo1 stimulation. Intriguingly, upon acidification, Piezo1 activation leads to the initiation of cell death and disruption of PSC spheroids. In summary, stimulating Piezo1 activates PSCs by inducing Ca2+ influx which in turn alters the cytoskeletal architecture. This results in increased cellular motility and ECM traction, which can be useful for the cells to invade the surroundings and to detach from the tissue. However, in the presence of an acidic extracellular pH, although net Ca2+ influx is reduced, Piezo1 activation leads to severe cell stress also limiting cellular viability. In conclusion, our results indicate a strong interdependence between environmental pH, the mechanical output of PSCs and stromal mechanics, which promotes early local invasion of PDAC cells.
Collapse
Affiliation(s)
- Anna Kuntze
- Institute of Physiology II, University of Münster, Münster, Germany
| | - Ole Goetsch
- Institute of Physiology II, University of Münster, Münster, Germany
| | - Benedikt Fels
- Institute of Physiology, University of Lübeck, Lübeck, Germany
| | - Karolina Najder
- Institute of Physiology II, University of Münster, Münster, Germany
| | - Andreas Unger
- Institute of Physiology II, University of Münster, Münster, Germany
| | | | - Sarah Sargin
- Institute of Physiology II, University of Münster, Münster, Germany
| | | | - Ilka Neumann
- Institute of Physiology II, University of Münster, Münster, Germany
| | - Albrecht Schwab
- Institute of Physiology II, University of Münster, Münster, Germany
| | - Zoltan Pethő
- Institute of Physiology II, University of Münster, Münster, Germany
| |
Collapse
|
23
|
Multi-well plate cell contraction assay detects negatively correlated cellular responses to pharmacological inhibitors in contractility and migration. Biochem Biophys Res Commun 2020; 521:527-532. [DOI: 10.1016/j.bbrc.2019.10.160] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 10/22/2019] [Indexed: 12/28/2022]
|
24
|
Thüroff F, Goychuk A, Reiter M, Frey E. Bridging the gap between single-cell migration and collective dynamics. eLife 2019; 8:e46842. [PMID: 31808744 PMCID: PMC6992385 DOI: 10.7554/elife.46842] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 12/06/2019] [Indexed: 11/13/2022] Open
Abstract
Motivated by the wealth of experimental data recently available, we present a cellular-automaton-based modeling framework focussing on high-level cell functions and their concerted effect on cellular migration patterns. Specifically, we formulate a coarse-grained description of cell polarity through self-regulated actin organization and its response to mechanical cues. Furthermore, we address the impact of cell adhesion on collective migration in cell cohorts. The model faithfully reproduces typical cell shapes and movements down to the level of single cells, yet allows for the efficient simulation of confluent tissues. In confined circular geometries, we find that specific properties of individual cells (polarizability; contractility) influence the emerging collective motion of small cell cohorts. Finally, we study the properties of expanding cellular monolayers (front morphology; stress and velocity distributions) at the level of extended tissues.
Collapse
Affiliation(s)
- Florian Thüroff
- Arnold Sommerfeld Center for Theoretical Physics and Center for NanoScience, Department of PhysicsLudwig-Maximilians-Universität MünchenMunichGermany
| | - Andriy Goychuk
- Arnold Sommerfeld Center for Theoretical Physics and Center for NanoScience, Department of PhysicsLudwig-Maximilians-Universität MünchenMunichGermany
| | - Matthias Reiter
- Arnold Sommerfeld Center for Theoretical Physics and Center for NanoScience, Department of PhysicsLudwig-Maximilians-Universität MünchenMunichGermany
| | - Erwin Frey
- Arnold Sommerfeld Center for Theoretical Physics and Center for NanoScience, Department of PhysicsLudwig-Maximilians-Universität MünchenMunichGermany
| |
Collapse
|
25
|
Wang C, Zhang F, Shan B, Liu J, Zhu L. [Real-time measurement of cell contractile force during activation of human hepatic stellate cell line LX-2]. SHENG WU YI XUE GONG CHENG XUE ZA ZHI = JOURNAL OF BIOMEDICAL ENGINEERING = SHENGWU YIXUE GONGCHENGXUE ZAZHI 2019; 36:841-849. [PMID: 31631634 PMCID: PMC9935154 DOI: 10.7507/1001-5515.201810048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Indexed: 11/03/2022]
Abstract
The contractile force of hepatic stellate cells plays a very important role in liver damage, hepatitis and fibrosis. In this paper, a method based on polydimethylsiloxane (PDMS) thin micropillar arrays is proposed to measure the contractile force of human hepatic stellate cell line LX-2, which enables dynamic measurement of the subcellular distribution of force magnitude and direction. First, thin micropillar arrays on glass bottom dish were fabricated using two-step casting process in order to meet the working distance requirement of 100× objective lens. After hydrophilic treatment and protein imprint, cells were seeded on the micropillar arrays. LX-2 cells, which were quiesced by growth in serum-free medium, were activated by adding fetal bovine serum (FBS). The deflections of the micropillars were achieved by image processing technique, and then the contractile force of cells exerted on the micropillars was calculated according to mechanical simulation results, and was analyzed under both quiescent and activated conditions. The experimental results show that the average traction force of quiescent cells is about 20 nN, while the contractile force of activated cells increased to 110 nN upon adding FBS. This method can quantify the contractile force of LX-2 cell on subcellular scale in both quiescent and activated states, which may benefit pathology study and drug screen for chronic liver diseases resulted from liver fibrosis.
Collapse
Affiliation(s)
- Chunran Wang
- Key Laboratory of the Ministry of Education for Optoelectronic Measurement Technology and Instrument, Beijing Information Science & Technology University, Beijing 100192, P.R.China
| | - Fan Zhang
- Key Laboratory of the Ministry of Education for Optoelectronic Measurement Technology and Instrument, Beijing Information Science & Technology University, Beijing 100192, P.R.China;Overseas Expertise Introduction Center for Discipline Innovation, Beijing Information Science & Technology University, Beijing 100192,
| | - Baojuan Shan
- Key Laboratory of the Ministry of Education for Optoelectronic Measurement Technology and Instrument, Beijing Information Science & Technology University, Beijing 100192, P.R.China
| | - Jiaqi Liu
- Key Laboratory of the Ministry of Education for Optoelectronic Measurement Technology and Instrument, Beijing Information Science & Technology University, Beijing 100192, P.R.China
| | - Lianqing Zhu
- Key Laboratory of the Ministry of Education for Optoelectronic Measurement Technology and Instrument, Beijing Information Science & Technology University, Beijing 100192, P.R.China;Overseas Expertise Introduction Center for Discipline Innovation, Beijing Information Science & Technology University, Beijing 100192, P.R.China
| |
Collapse
|
26
|
Duarte S, Viedma-Poyatos Á, Navarro-Carrasco E, Martínez AE, Pajares MA, Pérez-Sala D. Vimentin filaments interact with the actin cortex in mitosis allowing normal cell division. Nat Commun 2019; 10:4200. [PMID: 31519880 PMCID: PMC6744490 DOI: 10.1038/s41467-019-12029-4] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 08/09/2019] [Indexed: 01/27/2023] Open
Abstract
The vimentin network displays remarkable plasticity to support basic cellular functions and reorganizes during cell division. Here, we show that in several cell types vimentin filaments redistribute to the cell cortex during mitosis, forming a robust framework interwoven with cortical actin and affecting its organization. Importantly, the intrinsically disordered tail domain of vimentin is essential for this redistribution, which allows normal mitotic progression. A tailless vimentin mutant forms curly bundles, which remain entangled with dividing chromosomes leading to mitotic catastrophes or asymmetric partitions. Serial deletions of vimentin tail domain gradually impair cortical association and mitosis progression. Disruption of f-actin, but not of microtubules, causes vimentin bundling near the chromosomes. Pathophysiological stimuli, including HIV-protease and lipoxidation, induce similar alterations. Interestingly, full filament formation is dispensable for cortical association, which also occurs in vimentin particles. These results unveil implications of vimentin dynamics in cell division through its interplay with the actin cortex. The intermediate filament vimentin reorganizes during mitosis, but its molecular regulation and impact on the cell during cell division is unclear. Here, the authors show that vimentin filaments redistribute to the cell cortex during mitosis intertwining with and affecting actin organization.
Collapse
Affiliation(s)
- Sofia Duarte
- Department of Structural and Chemical Biology, Centro de Investigaciones Biológicas (CSIC), Ramiro de Maeztu 9, 28040, Madrid, Spain
| | - Álvaro Viedma-Poyatos
- Department of Structural and Chemical Biology, Centro de Investigaciones Biológicas (CSIC), Ramiro de Maeztu 9, 28040, Madrid, Spain
| | - Elena Navarro-Carrasco
- Department of Structural and Chemical Biology, Centro de Investigaciones Biológicas (CSIC), Ramiro de Maeztu 9, 28040, Madrid, Spain
| | - Alma E Martínez
- Department of Structural and Chemical Biology, Centro de Investigaciones Biológicas (CSIC), Ramiro de Maeztu 9, 28040, Madrid, Spain
| | - María A Pajares
- Department of Structural and Chemical Biology, Centro de Investigaciones Biológicas (CSIC), Ramiro de Maeztu 9, 28040, Madrid, Spain
| | - Dolores Pérez-Sala
- Department of Structural and Chemical Biology, Centro de Investigaciones Biológicas (CSIC), Ramiro de Maeztu 9, 28040, Madrid, Spain.
| |
Collapse
|
27
|
Nandagopal N, Santat LA, Elowitz MB. Cis-activation in the Notch signaling pathway. eLife 2019; 8:37880. [PMID: 30628888 PMCID: PMC6345567 DOI: 10.7554/elife.37880] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 01/09/2019] [Indexed: 12/31/2022] Open
Abstract
The Notch signaling pathway consists of transmembrane ligands and receptors that can interact both within the same cell (cis) and across cell boundaries (trans). Previous work has shown that cis-interactions act to inhibit productive signaling. Here, by analyzing Notch activation in single cells while controlling cell density and ligand expression level, we show that cis-ligands can also activate Notch receptors. This cis-activation process resembles trans-activation in its ligand level dependence, susceptibility to cis-inhibition, and sensitivity to Fringe modification. Cis-activation occurred for multiple ligand-receptor pairs, in diverse cell types, and affected survival in neural stem cells. Finally, mathematical modeling shows how cis-activation could potentially expand the capabilities of Notch signaling, for example enabling ‘negative’ (repressive) signaling. These results establish cis-activation as an additional mode of signaling in the Notch pathway, and should contribute to a more complete understanding of how Notch signaling functions in developmental, physiological, and biomedical contexts.
Collapse
Affiliation(s)
- Nagarajan Nandagopal
- Division of Biology and Biological Engineering, California Institute of Technology, Howard Hughes Medical Institute, Pasadena, United States
| | - Leah A Santat
- Division of Biology and Biological Engineering, California Institute of Technology, Howard Hughes Medical Institute, Pasadena, United States
| | - Michael B Elowitz
- Division of Biology and Biological Engineering, California Institute of Technology, Howard Hughes Medical Institute, Pasadena, United States
| |
Collapse
|
28
|
Pieuchot L, Marteau J, Guignandon A, Dos Santos T, Brigaud I, Chauvy PF, Cloatre T, Ponche A, Petithory T, Rougerie P, Vassaux M, Milan JL, Tusamda Wakhloo N, Spangenberg A, Bigerelle M, Anselme K. Curvotaxis directs cell migration through cell-scale curvature landscapes. Nat Commun 2018; 9:3995. [PMID: 30266986 PMCID: PMC6162274 DOI: 10.1038/s41467-018-06494-6] [Citation(s) in RCA: 151] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 09/07/2018] [Indexed: 11/27/2022] Open
Abstract
Cells have evolved multiple mechanisms to apprehend and adapt finely to their environment. Here we report a new cellular ability, which we term “curvotaxis” that enables the cells to respond to cell-scale curvature variations, a ubiquitous trait of cellular biotopes. We develop ultra-smooth sinusoidal surfaces presenting modulations of curvature in all directions, and monitor cell behavior on these topographic landscapes. We show that adherent cells avoid convex regions during their migration and position themselves in concave valleys. Live imaging combined with functional analysis shows that curvotaxis relies on a dynamic interplay between the nucleus and the cytoskeleton—the nucleus acting as a mechanical sensor that leads the migrating cell toward concave curvatures. Further analyses show that substratum curvature affects focal adhesions organization and dynamics, nuclear shape, and gene expression. Altogether, this work identifies curvotaxis as a new cellular guiding mechanism and promotes cell-scale curvature as an essential physical cue. The effect that microscale surface curvature has on cell migration has not been evaluated. Here the authors fabricate sinusoidal 3D surfaces and show that the cell nucleus and cytoskeleton cooperate to guide cells to concave valleys in a process they coin curvotaxis.
Collapse
Affiliation(s)
- Laurent Pieuchot
- Université de Haute-Alsace, CNRS, IS2M, UMR 7361, Mulhouse, F-68100, France. .,Université de Strasbourg, Strasbourg, F-67081, France.
| | - Julie Marteau
- Université de Valenciennes et du Hainaut Cambrésis, LAMIH, UMR-CNRS 8201, Le Mont Houy, Valenciennes, F-59313, France
| | - Alain Guignandon
- Univ Lyon, UJM-Saint-Etienne, INSERM, SAINBIOSE U1059, F-42023, Saint-Etienne, France
| | - Thomas Dos Santos
- Université de Haute-Alsace, CNRS, IS2M, UMR 7361, Mulhouse, F-68100, France.,Université de Strasbourg, Strasbourg, F-67081, France
| | - Isabelle Brigaud
- Université de Haute-Alsace, CNRS, IS2M, UMR 7361, Mulhouse, F-68100, France.,Université de Strasbourg, Strasbourg, F-67081, France
| | | | - Thomas Cloatre
- Université de Haute-Alsace, CNRS, IS2M, UMR 7361, Mulhouse, F-68100, France.,Université de Strasbourg, Strasbourg, F-67081, France
| | - Arnaud Ponche
- Université de Haute-Alsace, CNRS, IS2M, UMR 7361, Mulhouse, F-68100, France.,Université de Strasbourg, Strasbourg, F-67081, France
| | - Tatiana Petithory
- Université de Haute-Alsace, CNRS, IS2M, UMR 7361, Mulhouse, F-68100, France.,Université de Strasbourg, Strasbourg, F-67081, France
| | - Pablo Rougerie
- Laboratório de Biomineralização, Centro de Ciênça da Saúde, Federal University of Rio de Janeiro, Rio de Janeiro, 21941-902, Brazil
| | - Maxime Vassaux
- Aix Marseille Univ, CNRS, ISM, Inst Movement Sci, Marseille, F-13288, France
| | - Jean-Louis Milan
- Aix Marseille Univ, CNRS, ISM, Inst Movement Sci, Marseille, F-13288, France
| | - Nayana Tusamda Wakhloo
- Université de Haute-Alsace, CNRS, IS2M, UMR 7361, Mulhouse, F-68100, France.,Université de Strasbourg, Strasbourg, F-67081, France
| | - Arnaud Spangenberg
- Université de Haute-Alsace, CNRS, IS2M, UMR 7361, Mulhouse, F-68100, France.,Université de Strasbourg, Strasbourg, F-67081, France
| | - Maxence Bigerelle
- Université de Valenciennes et du Hainaut Cambrésis, LAMIH, UMR-CNRS 8201, Le Mont Houy, Valenciennes, F-59313, France
| | - Karine Anselme
- Université de Haute-Alsace, CNRS, IS2M, UMR 7361, Mulhouse, F-68100, France.,Université de Strasbourg, Strasbourg, F-67081, France
| |
Collapse
|
29
|
Rauscher AÁ, Gyimesi M, Kovács M, Málnási-Csizmadia A. Targeting Myosin by Blebbistatin Derivatives: Optimization and Pharmacological Potential. Trends Biochem Sci 2018; 43:700-713. [PMID: 30057142 DOI: 10.1016/j.tibs.2018.06.006] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 06/08/2018] [Accepted: 06/11/2018] [Indexed: 11/28/2022]
Abstract
Blebbistatin is a widely used inhibitor of myosin 2 that enables the study of a broad range of cytoskeleton-related processes. However, blebbistatin has several limitations hindering its applicability: it is fluorescent, poorly water soluble, cytotoxic, and prone to (photo)degradation. Despite these adverse effects, being the only available myosin 2-specific inhibitor, blebbistatin is rather a choice of necessity. Blebbistatin has been modified to improve its properties and some of the new compounds have proven to be useful replacements of the original molecule. This review summarizes recent results on blebbistatin development. We also discuss the pharmacological perspectives of these efforts, as myosins are becoming promising drug target candidates for a variety of conditions ranging from neurodegeneration to muscle disease, wound healing, and cancer metastasis.
Collapse
Affiliation(s)
- Anna Á Rauscher
- Motor Pharmacology Research Group, Department of Biochemistry, Eötvös Loránd University, H-1117 Budapest, Hungary
| | - Máté Gyimesi
- Motor Pharmacology Research Group, Department of Biochemistry, Eötvös Loránd University, H-1117 Budapest, Hungary
| | - Mihály Kovács
- Motor Pharmacology Research Group, Department of Biochemistry, Eötvös Loránd University, H-1117 Budapest, Hungary.
| | - András Málnási-Csizmadia
- Motor Pharmacology Research Group, Department of Biochemistry, Eötvös Loránd University, H-1117 Budapest, Hungary.
| |
Collapse
|
30
|
Roman BI, Verhasselt S, Mangodt CW, De Wever O, Stevens CV. Synthesis of C-ring-modified blebbistatin derivatives and evaluation of their myosin II ATPase inhibitory potency. Bioorg Med Chem Lett 2018; 28:2261-2264. [DOI: 10.1016/j.bmcl.2018.05.041] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 05/15/2018] [Accepted: 05/21/2018] [Indexed: 10/16/2022]
|
31
|
Abstract
( S)-Blebbistatin, a chiral tetrahydropyrroloquinolinone, is a widely used and well-characterized ATPase inhibitor selective for myosin II. The central role of myosin II in many normal and pathological biological processes has been revealed with the aid of this small molecule. The first part of this manuscript provides a summary of myosin II and ( S)-blebbistatin literature from a medicinal chemist's perspective. The second part of this perspective deals with the physicochemical deficiencies that trouble the use of ( S)-blebbistatin in advanced biological settings: low potency and solubility, fluorescence interference, (photo)toxicity, and stability issues. A large toolbox of analogues has been developed in which particular shortcomings have been addressed. This perspective provides a necessary overview of these developments and presents guidelines for selecting the best available analogue for a given application. As the unmet need for high-potency analogues remains, we also propose starting points for medicinal chemists in search of nanomolar myosin II inhibitors.
Collapse
|
32
|
Saxena N, Mogha P, Dash S, Majumder A, Jadhav S, Sen S. Matrix elasticity regulates mesenchymal stem cell chemotaxis. J Cell Sci 2018. [PMID: 29535208 DOI: 10.1242/jcs.211391] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Efficient homing of human mesenchymal stem cells (hMSCs) is likely to be dictated by a combination of physical and chemical factors present in the microenvironment. However, crosstalk between the physical and chemical cues remains incompletely understood. Here, we address this question by probing the efficiency of epidermal growth factor (EGF)-induced hMSC chemotaxis on substrates of varying stiffness (3, 30 and 600 kPa) inside a polydimethylsiloxane (PDMS) microfluidic device. Chemotactic speed was found to be the sum of a stiffness-dependent component and a chemokine concentration-dependent component. While the stiffness-dependent component scaled inversely with stiffness, the chemotactic component was independent of stiffness. Faster chemotaxis on the softest 3 kPa substrates is attributed to a combination of weaker adhesions and higher protrusion rate. While chemotaxis was mildly sensitive to contractility inhibitors, suppression of chemotaxis upon actin depolymerization demonstrates the role of actin-mediated protrusions in driving chemotaxis. In addition to highlighting the collective influence of physical and chemical cues in chemotactic migration, our results suggest that hMSC homing is more efficient on softer substrates.
Collapse
Affiliation(s)
- Neha Saxena
- Department of Chemical Engineering, IIT, Bombay, Maharashtra 400076, India
| | - Pankaj Mogha
- Department of Chemical Engineering, IIT, Bombay, Maharashtra 400076, India
| | - Silalipi Dash
- Department of Chemical Engineering, IIT, Bombay, Maharashtra 400076, India
| | - Abhijit Majumder
- Department of Chemical Engineering, IIT, Bombay, Maharashtra 400076, India
| | - Sameer Jadhav
- Department of Chemical Engineering, IIT, Bombay, Maharashtra 400076, India
| | - Shamik Sen
- Department of Bioscience and Bioengineering, IIT, Bombay, Maharashtra 400076, India
| |
Collapse
|
33
|
Lv Y, Fu L. The potential mechanism for Hydroxysafflor yellow A attenuating blood-brain barrier dysfunction via tight junction signaling pathways excavated by an integrated serial affinity chromatography and shotgun proteomics analysis approach. Neurochem Int 2017; 112:38-48. [PMID: 29107696 DOI: 10.1016/j.neuint.2017.10.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 10/19/2017] [Accepted: 10/25/2017] [Indexed: 12/15/2022]
Abstract
Our previous studies elucidated that hydroxysafflor yellow A (HSYA) exerted anti-inflammatory effects against ischemia stroke by inhibiting TLR4 pathway-mediated signaling transduction. However, only several targets were verified in that limited work. The integrated method of serial affinity chromatography (SAC) and shotgun proteomics analysis (SPA) might be an alternative approach for exploring a potential therapeutic role. SAC was induced to extract specific binding proteins in the brain tissue of 2 h of ischemia stroke mice via HSYA affinity matrices. SPA was conducted by nanoLC-MS/MS, while the identified proteins were mapped on to Gene Ontology and KEGG pathway components analysis. The protection of HSYA for blood-brain barrier in mice with ischemia stroke was assessed with the leakage of Evans Blue. The expression of tight junction proteins of blood-brain barrier: occludin, claudin-5, and ZO-1 were detected with ischemia boundary positive areas staining. The regulation of nonmuscle myosin heavy chain IIA (NMMHC IIA), TLR4-mediated PI3K/AKT/JNK1/2/14-3-3ε/NF-κB p65 signaling pathway were evaluated using western blot analysis. A total of 35 proteins with molecular eights ranging from 27,841.22 to 234,122.79 KD were identified. Gene Ontology annotation and KEGG pathways analysis of the identified proteins were conducted with tight junction and PI3K/AKT signaling pathways. HSYA could significantly reduce the leakage of Evans Blue in mice with ischemia stroke, while attenuating the expression of occludin, claudin-5, and ZO-1. Western blot demonstrated that regulation of NMMHC IIA, TLR4-mediated PI3K/AKT/JNK1/2/14-3-3ε/NF-κB p65 signaling pathway played an essential role in the protective effect of HSYA. The integrated method of SAC and SPA provides the promising explanations for exploring the mechanism underlying blood-brain barrier dysfunction via the tight junction pathway. HSYA could attenuate blood-brain barrier dysfunction in anti-inflammatory patterns in ischemia stroke mice via the tight junction pathway.
Collapse
Affiliation(s)
- Yanni Lv
- Pharmacy Department, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, China.
| | - Longsheng Fu
- Pharmacy Department, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, China
| |
Collapse
|
34
|
Zhai K, Zheng J, Tang Y, Li F, Lv Y, Zhang Y, Gao Z, Qi J, Yu B, Kou J. The saponin D39 blocks dissociation of non-muscular myosin heavy chain IIA from TNF receptor 2, suppressing tissue factor expression and venous thrombosis. Br J Pharmacol 2017; 174:2818-2831. [PMID: 28547925 PMCID: PMC5554322 DOI: 10.1111/bph.13885] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 04/27/2017] [Accepted: 05/08/2017] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND AND PURPOSE Non-muscular myosin heavy chain IIA (NMMHC IIA) plays a key role in tissue factor expression and venous thrombosis. Natural products might inhibit thrombosis through effects on NMMHC IIA. Here, we have shown that a natural saponin, D39, from Liriope muscari exerted anti-thrombotic activity in vivo, by targeting NMMHC IIA. EXPERIMENTAL APPROACH Expression and activity of tissue factor in endothelial cells were analysed in vitro by Western blot and simplified chromogenic assays. Interactions between D39 and NMMHC IIA were assessed by serial affinity chromatography and molecular docking analysis. D39-dependent interactions between NMMHC IIA and TNF receptor 2 (TNFR2) were measured by immunofluorescence, co-immunoprecipitation and proximity ligation assays. Anti-thrombotic activity of D39 in vivo was evaluated with a model of inferior vena cava ligation injury in mice. KEY RESULTS D39 inhibited tissue factor expression and procoagulant activities in HUVECs and decreased thrombus weight in inferior vena cava-ligated mice dose-dependently. Serial affinity chromatography and molecular docking analysis suggested that D39 bound to NMMHC IIA. In HEK293T cells, D39 inhibited tissue factor expression evoked by NMMHC IIA overexpression. This effect was blocked by NMMHC IIA knockdown in HUVECs. D39 inhibited dissociation of NMMHC IIA from TNFR2, which subsequently modulated the Akt/GSK3β-NF-κB signalling pathways. CONCLUSIONS AND IMPLICATIONS D39 inhibited tissue factor expression and thrombus formation by modulating the Akt/GSK3β and NF-κB signalling pathways through NMMHC IIA. We identified a new natural product that targeted NMMHC IIA, as a potential treatment for thrombotic disorders and other vasculopathies.
Collapse
Affiliation(s)
- Ke‐feng Zhai
- State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Complex Prescription of TCMChina Pharmaceutical UniversityNanjingChina
- Institute of Pharmaceutical Biotechnology, School of Biological and Food EngineeringSuzhou UniversitySuzhouChina
| | - Jin‐rong Zheng
- State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Complex Prescription of TCMChina Pharmaceutical UniversityNanjingChina
| | - You‐mei Tang
- State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Complex Prescription of TCMChina Pharmaceutical UniversityNanjingChina
| | - Fang Li
- State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Complex Prescription of TCMChina Pharmaceutical UniversityNanjingChina
| | - Yan‐ni Lv
- State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Complex Prescription of TCMChina Pharmaceutical UniversityNanjingChina
| | - Yuan‐yuan Zhang
- State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Complex Prescription of TCMChina Pharmaceutical UniversityNanjingChina
| | - Zhen Gao
- Department of Medicine‐Ather&LipoBaylor Colledge of MedicineHoustonTXUSA
| | - Jin Qi
- State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Complex Prescription of TCMChina Pharmaceutical UniversityNanjingChina
| | - Bo‐yang Yu
- State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Complex Prescription of TCMChina Pharmaceutical UniversityNanjingChina
| | - Jun‐ping Kou
- State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Complex Prescription of TCMChina Pharmaceutical UniversityNanjingChina
| |
Collapse
|
35
|
Verhasselt S, Stevens CV, Van den broecke T, Bracke ME, Roman BI. Insights into the myosin II inhibitory potency of A-ring-modified ( S )-blebbistatin analogs. Bioorg Med Chem Lett 2017; 27:2986-2989. [DOI: 10.1016/j.bmcl.2017.05.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2017] [Revised: 05/02/2017] [Accepted: 05/03/2017] [Indexed: 11/30/2022]
|
36
|
Lee P, Wolgemuth CW. Physical Mechanisms of Cancer in the Transition to Metastasis. Biophys J 2017; 111:256-66. [PMID: 27410752 DOI: 10.1016/j.bpj.2016.05.046] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Revised: 05/20/2016] [Accepted: 05/31/2016] [Indexed: 10/21/2022] Open
Abstract
Whether a tumor is metastatic is one of the most significant factors that influence the prognosis for a cancer patient. The transition from a nonmetastatic tumor to a metastatic one is accompanied by a number of genetic and proteomic changes within the tumor cells. These protein-level changes conspire to produce behavioral changes in the cells: cells that had been relatively stationary begin to move, often as a group. In this study we ask the question of what cell-level biophysical changes are sufficient to initiate evasion away from an otherwise static tumor. We use a mathematical model developed to describe the biophysics of epithelial tissue to explore this problem. The model is first validated against in vitro wound healing experiments with cancer cell lines. Then we simulate the behavior of a group of mutated cells within a sea of healthy tissue. We find that moderate increases in adhesion between the cell and extracellular matrix (ECM) accompanied by a decrease in cell-cell adhesion and/or Rho family of small GTPase activation can cause a group of cells to break free from a tumor and spontaneously migrate. This result may explain why some metastatic cells have been observed to upregulate integrin, downregulate cadherin, and activate Rho family signaling.
Collapse
Affiliation(s)
- Pilhwa Lee
- Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | | |
Collapse
|
37
|
Tarle V, Gauquelin E, Vedula SRK, D'Alessandro J, Lim CT, Ladoux B, Gov NS. Modeling collective cell migration in geometric confinement. Phys Biol 2017; 14:035001. [PMID: 28467320 DOI: 10.1088/1478-3975/aa6591] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Monolayer expansion has generated great interest as a model system to study collective cell migration. During such an expansion the culture front often develops 'fingers', which we have recently modeled using a proposed feedback between the curvature of the monolayer's leading edge and the outward motility of the edge cells. We show that this model is able to explain the puzzling observed increase of collective cellular migration speed of a monolayer expanding into thin stripes, as well as describe the behavior within different confining geometries that were recently observed in experiments. These comparisons give support to the model and emphasize the role played by the edge cells and the edge shape during collective cell motion.
Collapse
Affiliation(s)
- Victoria Tarle
- Department of Chemical Physics, Weizmann Institute of Science, Rehovot 76100, Israel
| | | | | | | | | | | | | |
Collapse
|
38
|
Chronopoulos A, Robinson B, Sarper M, Cortes E, Auernheimer V, Lachowski D, Attwood S, García R, Ghassemi S, Fabry B, Del Río Hernández A. ATRA mechanically reprograms pancreatic stellate cells to suppress matrix remodelling and inhibit cancer cell invasion. Nat Commun 2016; 7:12630. [PMID: 27600527 PMCID: PMC5023948 DOI: 10.1038/ncomms12630] [Citation(s) in RCA: 195] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 07/18/2016] [Indexed: 12/13/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive malignancy with a dismal survival rate. Persistent activation of pancreatic stellate cells (PSCs) can perturb the biomechanical homoeostasis of the tumour microenvironment to favour cancer cell invasion. Here we report that ATRA, an active metabolite of vitamin A, restores mechanical quiescence in PSCs via a mechanism involving a retinoic acid receptor beta (RAR-β)-dependent downregulation of actomyosin (MLC-2) contractility. We show that ATRA reduces the ability of PSCs to generate high traction forces and adapt to extracellular mechanical cues (mechanosensing), as well as suppresses force-mediated extracellular matrix remodelling to inhibit local cancer cell invasion in 3D organotypic models. Our findings implicate a RAR-β/MLC-2 pathway in peritumoural stromal remodelling and mechanosensory-driven activation of PSCs, and further suggest that mechanical reprogramming of PSCs with retinoic acid derivatives might be a viable alternative to stromal ablation strategies for the treatment of PDAC. Persistent activation of pancreatic stellate cells (PSCs) can perturb the biomechanical homeostasis of the tumour microenvironment. Here the authors show that all-trans retinoic acid reduces retinoic acid receptor beta dependent-actomyosin contractility and restores mechanical quiescence in PSCs.
Collapse
Affiliation(s)
- Antonios Chronopoulos
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London SW7 2AZ, UK
| | - Benjamin Robinson
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London SW7 2AZ, UK
| | - Muge Sarper
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London SW7 2AZ, UK
| | - Ernesto Cortes
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London SW7 2AZ, UK
| | - Vera Auernheimer
- Department of Physics, Biophysics Group, University of Erlangen-Nuremberg, Erlangen 91054, Germany
| | - Dariusz Lachowski
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London SW7 2AZ, UK
| | - Simon Attwood
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London SW7 2AZ, UK
| | - Rebeca García
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London SW7 2AZ, UK
| | - Saba Ghassemi
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | - Ben Fabry
- Department of Physics, Biophysics Group, University of Erlangen-Nuremberg, Erlangen 91054, Germany
| | - Armando Del Río Hernández
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London SW7 2AZ, UK
| |
Collapse
|
39
|
Lu C, Xu W, Zhang F, Shao J, Zheng S. Nrf2 knockdown attenuates the ameliorative effects of ligustrazine on hepatic fibrosis by targeting hepatic stellate cell transdifferentiation. Toxicology 2016; 365:35-47. [DOI: 10.1016/j.tox.2016.07.018] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Revised: 07/25/2016] [Accepted: 07/26/2016] [Indexed: 12/23/2022]
|
40
|
Wei XH, Lin SS, Liu Y, Zhao RP, Khan GJ, Du HZ, Mao TT, Yu BY, Li RM, Yuan ST, Sun L. DT-13 attenuates human lung cancer metastasis via regulating NMIIA activity under hypoxia condition. Oncol Rep 2016; 36:991-9. [DOI: 10.3892/or.2016.4879] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2016] [Accepted: 03/08/2016] [Indexed: 11/05/2022] Open
|
41
|
Garbuzenko DV. [Aspects of pathogenetc pharmacotherapy for portal hypertension in liver cirrhosis]. TERAPEVT ARKH 2016; 88:101-108. [PMID: 27135108 DOI: 10.17116/terarkh2016888101-108] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The review of literature considers the principles of medical treatment for portal hypertension in liver cirrhosis, which are based on the current views of its development mechanisms. It describes both current pharmacotherapy methods for portal hypertension and drugs, the efficacy of which is being investigated.
Collapse
Affiliation(s)
- D V Garbuzenko
- South Ural State Medical University, Ministry of Health of Russia, Chelyabinsk, Russia
| |
Collapse
|
42
|
Jalan R, De Chiara F, Balasubramaniyan V, Andreola F, Khetan V, Malago M, Pinzani M, Mookerjee RP, Rombouts K. Ammonia produces pathological changes in human hepatic stellate cells and is a target for therapy of portal hypertension. J Hepatol 2016; 64:823-33. [PMID: 26654994 DOI: 10.1016/j.jhep.2015.11.019] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Revised: 11/09/2015] [Accepted: 11/11/2015] [Indexed: 12/14/2022]
Abstract
BACKGROUND & AIMS Hepatic stellate cells (HSCs) are vital to hepatocellular function and the liver response to injury. They share a phenotypic homology with astrocytes that are central in the pathogenesis of hepatic encephalopathy, a condition in which hyperammonemia plays a pathogenic role. This study tested the hypothesis that ammonia modulates human HSC activation in vitro and in vivo, and evaluated whether ammonia lowering, by using l-ornithine phenylacetate (OP), modifies HSC activation in vivo and reduces portal pressure in a bile duct ligation (BDL) model. METHODS Primary human HSCs were isolated and cultured. Proliferation (BrdU), metabolic activity (MTS), morphology (transmission electron, light and immunofluorescence microscopy), HSC activation markers, ability to contract, changes in oxidative status (ROS) and endoplasmic reticulum (ER) were evaluated to identify effects of ammonia challenge (50 μM, 100 μM, 300 μM) over 24-72 h. Changes in plasma ammonia levels, markers of HSC activation, portal pressure and hepatic eNOS activity were quantified in hyperammonemic BDL animals, and after OP treatment. RESULTS Pathophysiological ammonia concentrations caused significant and reversible changes in cell proliferation, metabolic activity and activation markers of hHSC in vitro. Ammonia also induced significant alterations in cellular morphology, characterised by cytoplasmic vacuolisation, ER enlargement, ROS production, hHSC contraction and changes in pro-inflammatory gene expression together with HSC-related activation markers such as α-SMA, myosin IIa, IIb, and PDGF-Rβ. Treatment with OP significantly reduced plasma ammonia (BDL 199.1 μmol/L±43.65 vs. BDL+OP 149.27 μmol/L±51.1, p<0.05) and portal pressure (BDL 14±0.6 vs. BDL+OP 11±0.3 mmHg, p<0.01), which was associated with increased eNOS activity and abrogation of HSC activation markers. CONCLUSIONS The results show for the first time that ammonia produces deleterious morphological and functional effects on HSCs in vitro. Targeting ammonia with the ammonia lowering drug OP reduces portal pressure and deactivates hHSC in vivo, highlighting the opportunity for evaluating ammonia lowering as a potential therapy in cirrhotic patients with portal hypertension.
Collapse
Affiliation(s)
- Rajiv Jalan
- Liver Failure Group, Institute for Liver & Digestive Health, University College of London, Royal Free, London, UK
| | - Francesco De Chiara
- Liver Failure Group, Institute for Liver & Digestive Health, University College of London, Royal Free, London, UK
| | - Vairappan Balasubramaniyan
- Liver Failure Group, Institute for Liver & Digestive Health, University College of London, Royal Free, London, UK
| | - Fausto Andreola
- Liver Failure Group, Institute for Liver & Digestive Health, University College of London, Royal Free, London, UK
| | - Varun Khetan
- Liver Failure Group, Institute for Liver & Digestive Health, University College of London, Royal Free, London, UK
| | - Massimo Malago
- Division of Surgery, University College London, Royal Free, London, UK
| | - Massimo Pinzani
- Regenerative Medicine & Fibrosis Group, Institute for Liver & Digestive Health, University College London, Royal Free, London, UK
| | - Rajeshwar P Mookerjee
- Liver Failure Group, Institute for Liver & Digestive Health, University College of London, Royal Free, London, UK.
| | - Krista Rombouts
- Regenerative Medicine & Fibrosis Group, Institute for Liver & Digestive Health, University College London, Royal Free, London, UK.
| |
Collapse
|
43
|
Xu W, Lu C, Zhang F, Shao J, Zheng S. Dihydroartemisinin restricts hepatic stellate cell contraction via an FXR-S1PR2-dependent mechanism. IUBMB Life 2016; 68:376-87. [DOI: 10.1002/iub.1492] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 02/16/2016] [Indexed: 01/10/2023]
Affiliation(s)
- Wenxuan Xu
- Department of Pharmacology, School of Pharmacy; Nanjing University of Chinese Medicine; Nanjing Jiangsu Province China
| | - Chunfeng Lu
- Department of Pharmacology, School of Pharmacy; Nanjing University of Chinese Medicine; Nanjing Jiangsu Province China
| | - Feng Zhang
- Department of Pharmacology, School of Pharmacy; Nanjing University of Chinese Medicine; Nanjing Jiangsu Province China
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica; Nanjing University of Chinese Medicine; Nanjing Jiangsu Province China
| | - Jiangjuan Shao
- Department of Pharmacology, School of Pharmacy; Nanjing University of Chinese Medicine; Nanjing Jiangsu Province China
| | - Shizhong Zheng
- Department of Pharmacology, School of Pharmacy; Nanjing University of Chinese Medicine; Nanjing Jiangsu Province China
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica; Nanjing University of Chinese Medicine; Nanjing Jiangsu Province China
| |
Collapse
|
44
|
Southern BD, Grove LM, Rahaman SO, Abraham S, Scheraga RG, Niese KA, Sun H, Herzog EL, Liu F, Tschumperlin DJ, Egelhoff TT, Rosenfeld SS, Olman MA. Matrix-driven Myosin II Mediates the Pro-fibrotic Fibroblast Phenotype. J Biol Chem 2016; 291:6083-95. [PMID: 26763235 PMCID: PMC4813589 DOI: 10.1074/jbc.m115.712380] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Revised: 01/12/2016] [Indexed: 01/06/2023] Open
Abstract
Pro-fibrotic mesenchymal cells are known to be the key effector cells of fibroproliferative disease, but the specific matrix signals and the induced cellular responses that drive the fibrogenic phenotype remain to be elucidated. The key mediators of the fibroblast fibrogenic phenotype were characterized using a novel assay system that measures fibroblast behavior in response to actual normal and fibrotic lung tissue. Using this system, we demonstrate that normal lung promotes fibroblast motility and polarization, while fibrotic lung immobilizes the fibroblast and promotes myofibroblast differentiation. These context-specific phenotypes are surprisingly both mediated by myosin II. The role of myosin II is supported by the observation of an increase in myosin phosphorylation and a change in intracellular distribution in fibroblasts on fibrotic lung, as compared with normal lung. Moreover, loss of myosin II activity has opposing effects on protrusive activity in fibroblasts on normal and fibrotic lung. Loss of myosin II also selectively inhibits myofibroblast differentiation in fibroblasts on fibrotic lung. Importantly, these findings are recapitulated by varying the matrix stiffness of polyacrylamide gels in the range of normal and fibrotic lung tissue. Comparison of the effects of myosin inhibition on lung tissue with that of polyacrylamide gels suggests that matrix fiber organization drives the fibroblast phenotype under conditions of normal/soft lung, while matrix stiffness drives the phenotype under conditions of fibrotic/stiff lung. This work defines novel roles for myosin II as a key regulatory effector molecule of the pro-fibrotic phenotype, in response to biophysical properties of the matrix.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Huanxing Sun
- Yale ILD Center of Excellence, Yale School of Medicine, New Haven, Connecticut 06520
| | - Erica L Herzog
- Yale ILD Center of Excellence, Yale School of Medicine, New Haven, Connecticut 06520
| | - Fei Liu
- Department of Environmental Health, Harvard School of Public Health, Boston, Massachusetts 02115, and
| | - Daniel J Tschumperlin
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota 55905
| | | | - Steven S Rosenfeld
- Department of Cancer Biology, Cleveland Clinic, Lerner Research Institute, Cleveland, Ohio 44195
| | | |
Collapse
|
45
|
Jean L, Yang L, Majumdar D, Gao Y, Shi M, Brewer BM, Li D, Webb DJ. The Rho family GEF Asef2 regulates cell migration in three dimensional (3D) collagen matrices through myosin II. Cell Adh Migr 2015; 8:460-7. [PMID: 25517435 DOI: 10.4161/19336918.2014.983778] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Cell migration is fundamental to a variety of physiological processes, including tissue development, homeostasis, and regeneration. Migration has been extensively studied with cells on 2-dimensional (2D) substrates, but much less is known about cell migration in 3D environments. Tissues and organs are 3D, which is the native environment of cells in vivo, pointing to a need to understand migration and the mechanisms that regulate it in 3D environments. To investigate cell migration in 3D environments, we developed microfluidic devices that afford a controlled, reproducible platform for generating 3D matrices. Using these devices, we show that the Rho family guanine nucleotide exchange factor (GEF) Asef2 inhibits cell migration in 3D type I collagen (collagen I) matrices. Treatment of cells with the myosin II (MyoII) inhibitor blebbistatin abolished the decrease in migration by Asef2. Moreover, Asef2 enhanced MyoII activity as shown by increased phosphorylation of serine 19 (S19). Furthermore, Asef2 increased activation of Rac, which is a Rho family small GTPase, in 3D collagen I matrices. Inhibition of Rac activity by treatment with the Rac-specific inhibitor NSC23766 abrogated the Asef2-promoted increase in S19 MyoII phosphorylation. Thus, our results indicate that Asef2 regulates cell migration in 3D collagen I matrices through a Rac-MyoII-dependent mechanism.
Collapse
Key Words
- 2D, 2-dimensional
- 3D, 3-dimensional
- Collagen I, type I collagen
- DMEM, Dulbecco's Modified Eagle Medium
- ECM, extracellular matrix
- GEF, guanine nucleotide exchange factor
- MyoII, non-muscle myosin II
- PAK, p21-activated kinase
- PBD, p21-binding domain
- PBS, phosphate buffer saline
- PDMS, polydimethylsiloxane
- Rac
- Rho family GTPases
- UV, ultra-violet
- guanine nucleotide exchange factor
- microfluidics
- myosin II
- type I collagen
Collapse
Affiliation(s)
- Léolène Jean
- a Department of Biological Sciences and Vanderbilt Kennedy Center for Research on Human Development ; Vanderbilt University ; Nashville , TN USA
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Garbuzenko DV. Contemporary concepts of the medical therapy of portal hypertension under liver cirrhosis. World J Gastroenterol 2015; 21:6117-6126. [PMID: 26034348 PMCID: PMC4445090 DOI: 10.3748/wjg.v21.i20.6117] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Revised: 03/20/2015] [Accepted: 04/17/2015] [Indexed: 02/06/2023] Open
Abstract
Severe complications of liver cirrhosis are mostly related to portal hypertension. At the base of the pathogenesis of portal hypertension is the increase in hepatic vascular resistance to portal blood flow with subsequent development of hyperdynamic circulation, which, despite of the formation of collateral circulation, promotes progression of portal hypertension. An important role in its pathogenesis is played by the rearrangement of vascular bed and angiogenesis. As a result, strategic directions of the therapy of portal hypertension under liver cirrhosis include selectively decreasing hepatic vascular resistance with preserving or increasing portal blood flow, and correcting hyperdynamic circulation and pathological angiogenesis, while striving to reduce the hepatic venous pressure gradient to less than 12 mmHg or 20% of the baseline. Over the last years, substantial progress in understanding the pathophysiological mechanisms of hemodynamic disorders under liver cirrhosis has resulted in the development of new drugs for their correction. Although the majority of them have so far been investigated only in animal experiments, as well as at the molecular and cellular level, it might be expected that the introduction of the new methods in clinical practice will increase the efficacy of the conservative approach to the prophylaxis and treatment of portal hypertension complications. The purpose of the review is to describe the known methods of portal hypertension pharmacotherapy and discuss the drugs that may affect the basic pathogenetic mechanisms of its development.
Collapse
|
47
|
Zhai K, Tang Y, Zhang Y, Li F, Wang Y, Cao Z, Yu J, Kou J, Yu B. NMMHC IIA inhibition impedes tissue factor expression and venous thrombosis via Akt/GSK3β-NF-κB signalling pathways in the endothelium. Thromb Haemost 2015; 114:173-85. [PMID: 25881103 DOI: 10.1160/th14-10-0880] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Accepted: 02/19/2015] [Indexed: 01/29/2023]
Abstract
Non-muscle myosin heavy chain IIA (NMMHC IIA) has been shown to be involved in thrombus formation and inflammatory microparticle release in endothelial cells. However, the role of NMMHC IIA in regulating the expression of tissue factor (TF) and deep venous thrombosis remains to be elucidated. In the present study, endothelial cells were stimulated with tumour necrosis factor-α (TNF-α) to induce TF expression. Pretreatment with the NMMHC II inhibitor blebbistatin suppressed the mRNA and protein expressions as well as the procoagulant activity of TF in a dose-dependent manner. Blebbistatin enhanced Akt and GSK3β phosphorylation and inhibited NF-κB p65 nuclear translocation and IκBα degradation. These observations were similar to the effect of CHIR99021, a GSK3β inhibitor. TF downregulation by blebbistatin was antagonised by the PI3K inhibitor, wortmannin. Furthermore, siRNA knockdown of NMMHC IIA, but not IIB or IIC, inhibited TF expression, activated Akt/GSK3β and suppressed NF-κB signalling pathways, whereas the overexpression of NMMHC IIA increased TF expression. The binding of NMMHC IIA and TNF receptor 2 mediated signal internalisation in TNF-α-stimulated endothelial cells. Importantly, blebbistatin decreased endothelium NMMHC IIA and TF expression, deactivated GSK3β by inducing its phosphorylation, suppressed p65 nuclear translocation, and inhibited thrombus formation in a mouse deep venous thrombosis model.Our findings provide solid evidence that inhibition of NMMHC II, most likely NMMHC IIA, impedes TF expression and venous thrombosis via Akt/GSK3β-NF-κB signalling pathways in the endothelium both in vitro and in vivo. NMMHC IIA might be a potential novel target for the treatment of thrombotic disorders.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Jun Yu
- Dr. Jun Yu, Department of Internal Medicine, Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT 06519, USA, Tel.: +1 203 7372869, Fax: +1 203 7372290, E-mail:
| | - Junping Kou
- Dr. Junping Kou, State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Complex Prescription of TCM, China Pharmaceutical University, 639 Longmian Road, Nanjing, 211198, P. R. China, Tel./Fax: +86 25 86185158, E-mail:
| | | |
Collapse
|
48
|
Tarle V, Ravasio A, Hakim V, Gov NS. Modeling the finger instability in an expanding cell monolayer. Integr Biol (Camb) 2015; 7:1218-27. [DOI: 10.1039/c5ib00092k] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Curvature-controlled cellular forces at the edge of an expanding monolayer are sufficient for the initiation and growth of finger-like instability.
Collapse
Affiliation(s)
- Victoria Tarle
- Department of Chemical Physics
- Weizmann Institute of Science
- Rehovot 76100
- Israel
| | - Andrea Ravasio
- Mechanobiology Institute
- National University of Singapore
- Singapore
| | - Vincent Hakim
- Laboratoire de Physique Statistique
- CNRS
- Université P et M Curie
- Université Paris Diderot
- Ecole Normale Supérieure
| | - Nir S. Gov
- Department of Chemical Physics
- Weizmann Institute of Science
- Rehovot 76100
- Israel
| |
Collapse
|
49
|
Pelekanos RA, Ting MJ, Sardesai VS, Ryan JM, Lim YC, Chan JKY, Fisk NM. Intracellular trafficking and endocytosis of CXCR4 in fetal mesenchymal stem/stromal cells. BMC Cell Biol 2014; 15:15. [PMID: 24885150 PMCID: PMC4065074 DOI: 10.1186/1471-2121-15-15] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Accepted: 05/02/2014] [Indexed: 12/13/2022] Open
Abstract
Background Fetal mesenchymal stem/stromal cells (MSC) represent a developmentally-advantageous cell type with translational potential. To enhance adult MSC migration, studies have focussed on the role of the chemokine receptor CXCR4 and its ligand SDF-1 (CXCL12), but more recent work implicates an intricate system of CXCR4 receptor dimerization, intracellular localization, multiple ligands, splice variants and nuclear accumulation. We investigated the intracellular localization of CXCR4 in fetal bone marrow-derived MSC and role of intracellular trafficking in CXCR4 surface expression and function. Results We found that up to 4% of human fetal MSC have detectable surface-localized CXCR4. In the majority of cells, CXCR4 is located not at the cell surface, as would be required for ‘sensing’ migratory cues, but intracellularly. CXCR4 was identified in early endosomes, recycling endosomes, and lysosomes, indicating only a small percentage of CXCR4 travelling to the plasma membrane. Notably CXCR4 was also found in and around the nucleus, as detected with an anti-CXCR4 antibody directed specifically against CXCR4 isoform 2 differing only in N-terminal sequence. After demonstrating that endocytosis of CXCR4 is largely independent of endogenously-produced SDF-1, we next applied the cytoskeletal inhibitors blebbistatin and dynasore to inhibit endocytotic recycling. These increased the number of cells expressing surface CXCR4 by 10 and 5 fold respectively, and enhanced the number of cells migrating to SDF1 in vitro (up to 2.6 fold). These molecules had a transient effect on cell morphology and adhesion, which abated after the removal of the inhibitors, and did not alter functional stem cell properties. Conclusions We conclude that constitutive endocytosis is implicated in the regulation of CXCR4 membrane expression, and suggest a novel pharmacological strategy to enhance migration of systemically-transplanted cells.
Collapse
Affiliation(s)
- Rebecca A Pelekanos
- UQ Centre for Clinical Research, The University of Queensland, Herston QLD 4029, Australia.
| | | | | | | | | | | | | |
Collapse
|
50
|
The different roles of myosin IIA and myosin IIB in contraction of 3D collagen matrices by human fibroblasts. Exp Cell Res 2014; 326:295-306. [PMID: 24768700 DOI: 10.1016/j.yexcr.2014.04.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Revised: 04/09/2014] [Accepted: 04/14/2014] [Indexed: 02/06/2023]
Abstract
Contraction of 3D collagen matrices by fibroblasts frequently is used as an in vitro model of wound closure. Different iterations of the model - all conventionally referred to as "contraction" - involve different morphological patterns. During floating matrix contraction, cells initially are round without stress fibers and subsequently undergo spreading. During stressed matrix contraction, cells initially are spread with stress fibers and subsequently undergo shortening. In the current studies, we used siRNA silencing of myosin IIA (MyoIIA) and myosin IIB (MyoIIB) to test the roles of myosin II isoforms in fibroblast interactions with 3D collagen matrices and collagen matrix contraction. We found that MyoIIA but not MyoIIB was required for cellular global inward contractile force, formation of actin stress fibers, and morphogenic cell clustering. Stressed matrix contraction required MyoIIA but not MyoIIB. Either MyoIIA or MyoIIB was sufficient for floating matrix contraction (FMC) stimulated by platelet-derived growth factor. Neither MyoIIA or MyoIIB was necessary for FMC stimulated by serum. Our findings suggest that myosin II-dependent motor mechanisms for collagen translocation during extracellular matrix remodeling differ depending on cell tension and growth factor stimulation.
Collapse
|