1
|
Graham AJ, Khoo MW, Srivastava V, Viragova S, Kim H, Parekh K, Hennick KM, Bird M, Goldhammer N, Yu JZ, Morley CD, Lebel P, Kumar S, Rosenbluth JM, Nowakowski TJ, Klein O, Gómez-Sjöberg R, Gartner ZJ. MAGIC matrices: freeform bioprinting materials to support complex and reproducible organoid morphogenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.01.578324. [PMID: 38370663 PMCID: PMC10871257 DOI: 10.1101/2024.02.01.578324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
Organoids are powerful models of tissue physiology, yet their applications remain limited due to their relatively simple morphology and high organoid-to-organoid structural variability. To address these limitations we developed a soft, composite yield-stress extracellular matrix that supports optimal organoid morphogenesis following freeform 3D bioprinting of cell slurries at tissue-like densities. The material is designed with two temperature regimes: at 4 °C it exhibits reversible yield-stress behavior to support long printing times without compromising cell viability. When transferred to cell culture at 37 °C, the material cross-links and exhibits similar viscoelasticity and plasticity to basement membrane extracts such as Matrigel. We first characterize the rheological properties of MAGIC matrices that optimize organoid morphogenesis, including low stiffness and high stress relaxation. Next, we combine this material with a custom piezoelectric printhead that allows more reproducible and robust self-organization from uniform and spatially organized tissue "seeds." We apply MAGIC matrix bioprinting for high-throughput generation of intestinal, mammary, vascular, salivary gland, and brain organoid arrays that are structurally similar to those grown in pure Matrigel, but exhibit dramatically improved homogeneity in organoid size, shape, maturation time, and efficiency of morphogenesis. The flexibility of this method and material enabled fabrication of fully 3D microphysiological systems, including perfusable organoid tubes that experience cyclic 3D strain in response to pressurization. Furthermore, the reproducibility of organoid structure increased the statistical power of a drug response assay by up to 8 orders-of-magnitude for a given number of comparisons. Combined, these advances lay the foundation for the efficient fabrication of complex tissue morphologies by canalizing their self-organization in both space and time.
Collapse
Affiliation(s)
- Austin J. Graham
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA
- Chan Zuckerberg Biohub SF, San Francisco, CA
| | | | - Vasudha Srivastava
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA
| | - Sara Viragova
- Department of Orofacial Sciences, University of California San Francisco, San Francisco, CA
| | - Honesty Kim
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA
- Chan Zuckerberg Biohub SF, San Francisco, CA
| | - Kavita Parekh
- Department of Bioengineering, University of California Berkeley, Berkeley, CA
| | - Kelsey M. Hennick
- Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA
| | - Malia Bird
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA
| | - Nadine Goldhammer
- Department of Medicine, University of California San Francisco, San Francisco, CA
| | - Jie Zeng Yu
- Department of Medicine, University of California San Francisco, San Francisco, CA
| | - Cameron D. Morley
- Department of Bioengineering, University of California Berkeley, Berkeley, CA
| | - Paul Lebel
- Chan Zuckerberg Biohub SF, San Francisco, CA
| | - Sanjay Kumar
- Department of Bioengineering, University of California Berkeley, Berkeley, CA
- Department of Chemical and Biomolecular Engineering, University of California Berkeley, Berkeley, CA
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA
| | - Jennifer M. Rosenbluth
- Chan Zuckerberg Biohub SF, San Francisco, CA
- Department of Medicine, University of California San Francisco, San Francisco, CA
| | - Tomasz J. Nowakowski
- Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA
| | - Ophir Klein
- Department of Orofacial Sciences, University of California San Francisco, San Francisco, CA
- Department of Pediatrics, Cedars-Sinai Guerin Children’s, Los Angeles, CA
| | | | - Zev J. Gartner
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA
- Chan Zuckerberg Biohub SF, San Francisco, CA
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA
- Center for Cellular Construction, University of California San Francisco, San Francisco, CA
| |
Collapse
|
2
|
Pan B, Wu F, Lu S, Lu W, Cao J, Cheng F, Ou M, Chen Y, Zhang F, Wu G, Mei L. Luteolin-Loaded Hyaluronidase Nanoparticles with Deep Tissue Penetration Capability for Idiopathic Pulmonary Fibrosis Treatment. SMALL METHODS 2024:e2400980. [PMID: 39370583 DOI: 10.1002/smtd.202400980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 09/20/2024] [Indexed: 10/08/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive interstitial lung disease characterized by sustained fibrotic lesions. Orally administered drugs usually fail to efficiently penetrate the interstitial tissue and reach the lesions, resulting in low treatment efficiency. Luteolin (Lut) is a natural flavonoid, active metabolites of which possess antioxidant, anti-inflammatory, anti-fibrotic, and anti-apoptotic properties. In this study, a nano-formulation is developed by loading Lut into hyaluronidase nanoparticles (Lut@HAase). These Lut@HAase nanoparticles (NPs) exhibit small size and good stability, suitable for noninvasive inhalation and accumulation in the lungs, and hyaluronidase at the site of lesions can degrade hyaluronic acid in the interstitial tissue, enabling efficient penetration of Lut. Lut's therapeutic effect, when administered via NPs, is studied both in vitro (using MRC5 cells) and in vivo (using IPF mice models), and its anti-fibrotic properties are found to inhibit inflammation and eliminate reactive oxygen species. Conclusively, this study demonstrates that Lut@HAase can improve lung function and enhance survival rates while reducing lung damage with few abnormalities during IPF treatment.
Collapse
Affiliation(s)
- Bo Pan
- The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, China
| | - Fangping Wu
- The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, China
| | - Shanming Lu
- Department of Pathology, Longgang Central Hospital, Shenzhen, Guangdong, 518100, China
| | - Wenwen Lu
- The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, China
| | - Jiahui Cao
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, China
| | - Fei Cheng
- The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, China
| | - Meitong Ou
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, China
| | - Youyi Chen
- The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, China
| | - Fan Zhang
- Department of Pathology, Longgang Central Hospital, Shenzhen, Guangdong, 518100, China
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, China
| | - Guolin Wu
- The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, China
| | - Lin Mei
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, China
| |
Collapse
|
3
|
Wang H, Wu J, Yang L, Liu S, Sui X, Guo Q, Chen M, Xia Y. Cell-free decellularized skin matrix scaffolds: A promising approach for meniscus regeneration in a rabbit meniscectomy model. Acta Biomater 2024; 187:66-81. [PMID: 39168422 DOI: 10.1016/j.actbio.2024.08.014] [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: 03/22/2024] [Revised: 08/06/2024] [Accepted: 08/11/2024] [Indexed: 08/23/2024]
Abstract
Tissue engineering presents a promising approach for the treatment of meniscal injuries, yet the development of meniscal scaffolds that exhibit both superior biomechanical properties and biocompatibility remains a considerable challenge. In this study, decellularized skin matrix (DSM) scaffolds were first prepared using porcine skin through decellularization and freeze-drying techniques. The DSM scaffold has favorable porosity, hydrophilicity, and biocompatibility. Importantly, the collagen content and tensile modulus of the scaffold are comparable to those of native meniscus (44.13 ± 2.396 mg/g vs. 42.41 ± 2.40 mg/g and 103.30 ± 2.98 MPa vs. 128.80 ± 9.115 MPa). Subsequently, the peptide PFSSTKT (PFS) with mesenchymal stem cells (MSCs) recruitment capability was used to modify DSM to construct DSM-PFS scaffolds. Compared to the DSM scaffold, the optimized DSM-PFS scaffold enhanced in vitro collagen and glycosaminoglycan (GAG) production and upregulated the expression of cartilage-specific genes. Furthermore, the DSM-PFS scaffold was more effective in recruiting MSCs in vitro. In vivo studies in rabbit models showed that the DSM-PFS scaffold successfully promoted meniscus tissue regeneration. Three months post-implantation, meniscus tissue formation can be observable, and after six months, the neo-meniscus exhibited tissue structure and tensile properties similar to the native meniscus. Notably, the DSM-PFS scaffold exhibited significant chondroprotective effects, slowing osteoarthritis (OA) progression. In conclusion, the DSM-PFS scaffold may represent a promising candidate for future applications in meniscus tissue engineering. STATEMENT OF SIGNIFICANCE: We developed a decellularized skin matrix (DSM) meniscus scaffold using whole-layer porcine skin, demonstrating superior biomechanical strength and biocompatibility. Following modification with the stem cell-recruiting peptide PFS, the optimized DSM-PFS scaffold outperformed the DSM scaffold in cell attraction, collagen and glycosaminoglycan production, and cartilage-specific gene expression. Implanted into rabbit knee joints, the cell-free DSM-PFS scaffold induced meniscal tissue formation within three months, achieving the histological structure and tensile strength of the native meniscus by six months. Moreover, it significantly protected the cartilage. Our findings provide new insights into the fabrication of scaffolds for meniscal tissue engineering, with the DSM-PFS scaffold emerging as an ideal candidate for future applications.
Collapse
Affiliation(s)
- Hao Wang
- Department of Orthopedics, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, Gansu, China; Department of Orthopaedic Surgery, Beijing Jishuitan Hospital, Capital Medical University, No.31 Xinjiekou East Street, Xicheng District, Beijing 100035, China; Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Jie Wu
- Department of Orthopedics, Eighth Medical Center, General Hospital of Chinese PLA, Beijing 100853, China
| | - Liupu Yang
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Shuyun Liu
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Xiang Sui
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Quanyi Guo
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing 100853, China.
| | - Mingxue Chen
- Department of Orthopaedic Surgery, Beijing Jishuitan Hospital, Capital Medical University, No.31 Xinjiekou East Street, Xicheng District, Beijing 100035, China.
| | - Yayi Xia
- Department of Orthopedics, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, Gansu, China.
| |
Collapse
|
4
|
Parikh A, Krogman W, Walker J. The impact of volatile anesthetics and propofol on phosphatidylinositol 4,5-bisphosphate signaling. Arch Biochem Biophys 2024; 757:110045. [PMID: 38801966 DOI: 10.1016/j.abb.2024.110045] [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: 02/14/2024] [Revised: 04/29/2024] [Accepted: 05/24/2024] [Indexed: 05/29/2024]
Abstract
Phosphatidylinositol 4,5-bisphosphate (PIP2), as well as other anionic phospholipids, play a pivotal role in various cellular processes, including ion channel regulation, receptor trafficking, and intracellular signaling pathways. The binding of volatile anesthetics and propofol to PIP2 leads to alterations in PIP2-mediated signaling causing modulation of ion channels such as ɣ-aminobutyric acid type A (GABAA) receptors, voltage-gated calcium channels, and potassium channels through various mechanisms. Additionally, the interaction between anionic phospholipids and G protein-coupled receptors plays a critical role in various anesthetic pathways, with these anesthetic-induced changes impacting PIP2 levels which cause cascading effects on receptor trafficking, including GABAA receptor internalization. This comprehensive review of various mechanisms of interaction provides insights into the intricate interplay between PIP2 signaling and anesthetic-induced changes, shedding light on the molecular mechanisms underlying anesthesia.
Collapse
Affiliation(s)
- Ayaan Parikh
- Wichita Collegiate School, Wichita, KS. 9115 E 13th St N, Wichita, KS, 67206, USA.
| | - William Krogman
- University of Kansas School of Medicine-Wichita, Wichita, KS, USA; Department of Anesthesiology, 929 N St Francis, Room 8079, Wichita, KS, 67214, USA
| | - James Walker
- University of Kansas School of Medicine-Wichita, Wichita, KS, USA; Department of Anesthesiology, 929 N St Francis, Room 8079, Wichita, KS, 67214, USA
| |
Collapse
|
5
|
Chan DD, Guilak F, Sah RL, Calve S. Mechanobiology of Hyaluronan: Connecting Biomechanics and Bioactivity in Musculoskeletal Tissues. Annu Rev Biomed Eng 2024; 26:25-47. [PMID: 38166186 DOI: 10.1146/annurev-bioeng-073123-120541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2024]
Abstract
Hyaluronan (HA) plays well-recognized mechanical and biological roles in articular cartilage and synovial fluid, where it contributes to tissue structure and lubrication. An understanding of how HA contributes to the structure of other musculoskeletal tissues, including muscle, bone, tendon, and intervertebral discs, is growing. In addition, the use of HA-based therapies to restore damaged tissue is becoming more prevalent. Nevertheless, the relationship between biomechanical stimuli and HA synthesis, degradation, and signaling in musculoskeletal tissues remains understudied, limiting the utility of HA in regenerative medicine. In this review, we discuss the various roles and significance of endogenous HA in musculoskeletal tissues. We use what is known and unknown to motivate new lines of inquiry into HA biology within musculoskeletal tissues and in the mechanobiology governing HA metabolism by suggesting questions that remain regarding the relationship and interaction between biological and mechanical roles of HA in musculoskeletal health and disease.
Collapse
Affiliation(s)
- Deva D Chan
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, USA;
- School of Mechanical Engineering, Purdue University, West Lafayette, Indiana, USA
| | - Farshid Guilak
- Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, Missouri, USA
- Shriners Hospitals for Children-St. Louis, St. Louis, Missouri, USA
| | - Robert L Sah
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego, La Jolla, California, USA
| | - Sarah Calve
- Paul M. Rady Department of Mechanical Engineering, University of Colorado Boulder, Boulder, Colorado, USA
| |
Collapse
|
6
|
Byfield FJ, Eftekhari B, Kaymak-Loveless K, Mandal K, Li D, Wells RG, Chen W, Brujic J, Bergamaschi G, Wuite GJL, Patteson AE, Janmey PA. Metabolically intact nuclei are fluidized by the activity of the chromatin remodeling motor BRG1. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.12.589275. [PMID: 38659735 PMCID: PMC11042217 DOI: 10.1101/2024.04.12.589275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
The structure and dynamics of the cell nucleus regulate nearly every facet of the cell. Changes in nuclear shape limit cell motility and gene expression. Although the nucleus is generally seen as the stiffest organelle in the cell, cells can nevertheless deform the nucleus to large strains by small mechanical stresses. Here, we show that the mechanical response of the cell nucleus exhibits active fluidization that is driven by the BRG 1 motor of the SWI/SNF/BAF chromatin-remodeling complex. Atomic force microscopy measurements show that the nucleus alters stiffness in response to the cell substrate stiffness, which is retained after the nucleus is isolated and that the work of nuclear compression is mostly dissipated rather than elastically stored. Inhibiting BRG 1 stiffens the nucleus and eliminates dissipation and nuclear remodeling both in isolated nuclei and in intact cells. These findings demonstrate a novel link between nuclear motor activity and global nuclear mechanics.
Collapse
|
7
|
Ji S, Li Y, Xiang L, Liu M, Xiong M, Cui W, Fu X, Sun X. Cocktail Cell-Reprogrammed Hydrogel Microspheres Achieving Scarless Hair Follicle Regeneration. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2306305. [PMID: 38225741 DOI: 10.1002/advs.202306305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 10/24/2023] [Indexed: 01/17/2024]
Abstract
The scar repair inevitably causes damage of skin function and loss of skin appendages such as hair follicles (HF). It is of great challenge in wound repair that how to intervene in scar formation while simultaneously remodeling HF niche and inducing in situ HF regeneration. Here, chemical reprogramming techniques are used to identify a clinically chemical cocktail (Tideglusib and Tamibarotene) that can drive fibroblasts toward dermal papilla cell (DPC) fate. Considering the advantage of biomaterials in tissue repair and their regulation in cell behavior that may contributes to cellular reprogramming, the artificial HF seeding (AHFS) hydrogel microspheres, inspired by the natural processes of "seeding and harvest", are constructed via using a combination of liposome nanoparticle drug delivery system, photoresponsive hydrogel shell, positively charged polyamide modification, microfluidic and photocrosslinking techniques. The identified chemical cocktail is as the core nucleus of AHFS. In vitro and in vivo studies show that AHFS can regulate fibroblast fate, induce fibroblast-to-DPC reprogramming by activating the PI3K/AKT pathway, finally promoting wound healing and in situ HF regeneration while inhibiting scar formation in a two-pronged translational approach. In conclusion, AHFS provides a new and effective strategy for functional repair of skin wounds.
Collapse
Affiliation(s)
- Shuaifei Ji
- Research Center for Tissue Repair and Regeneration affiliated to the Medical Innovation Research Department, PLA General Hospital and PLA Medical College, PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, P. R. China
| | - Yingying Li
- Research Center for Tissue Repair and Regeneration affiliated to the Medical Innovation Research Department, PLA General Hospital and PLA Medical College, PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, P. R. China
| | - Lei Xiang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| | - Mingyue Liu
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| | - Mingchen Xiong
- Research Center for Tissue Repair and Regeneration affiliated to the Medical Innovation Research Department, PLA General Hospital and PLA Medical College, PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, P. R. China
| | - Wenguo Cui
- Research Center for Tissue Repair and Regeneration affiliated to the Medical Innovation Research Department, PLA General Hospital and PLA Medical College, PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, P. R. China
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| | - Xiaobing Fu
- Research Center for Tissue Repair and Regeneration affiliated to the Medical Innovation Research Department, PLA General Hospital and PLA Medical College, PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, P. R. China
| | - Xiaoyan Sun
- Research Center for Tissue Repair and Regeneration affiliated to the Medical Innovation Research Department, PLA General Hospital and PLA Medical College, PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, P. R. China
| |
Collapse
|
8
|
Chen G, Gao X, Chen J, Peng L, Chen S, Tang C, Dai Y, Wei Q, Luo D. Actomyosin Activity and Piezo1 Activity Synergistically Drive Urinary System Fibroblast Activation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2303369. [PMID: 37867255 PMCID: PMC10667826 DOI: 10.1002/advs.202303369] [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: 05/24/2023] [Revised: 08/11/2023] [Indexed: 10/24/2023]
Abstract
Mechanical cues play a crucial role in activating myofibroblasts from quiescent fibroblasts during fibrosis, and the stiffness of the extracellular matrix is of significant importance in this process. While intracellular force mediated by myosin II and calcium influx regulated by Piezo1 are the primary mechanisms by which cells sense and respond to mechanical forces, their intercellular mechanical interaction remains to be elucidated. Here, hydrogels with tunable substrate are used to systematically investigate the crosstalk of myosin II and Piezo1 in fibroblast to myofibroblast transition (FMT). The findings reveal that the two distinct signaling pathways are integrated to convert mechanical stiffness signals into biochemical signals during bladder-specific FMT. Moreover, it is demonstrated that the crosstalk between myosin II and Piezo1 sensing mechanisms synergistically establishes a sustained feed-forward loop that contributes to chromatin remodeling, induces the expression of downstream target genes, and ultimately exacerbates FMT, in which the intracellular force activates Piezo1 by PI3K/PIP3 pathway-mediated membrane tension and the Piezo1-regulated calcium influx enhances intracellular force by the classical FAK/RhoA/ROCK pathway. Finally, the multifunctional Piezo1 in the complex feedback circuit of FMT drives to further identify that targeting Piezo1 as a therapeutic option for ameliorating bladder fibrosis and dysfunction.
Collapse
Affiliation(s)
- Guo Chen
- Department of UrologyInstitute of Urology (Laboratory of Reconstructive Urology)West China HospitalSichuan UniversityChengduSichuan610041P. R. China
- Department of Urology and Pelvic surgeryWest China School of Public Health and West China Fourth HospitalSichuan UniversityChengduSichuan610041P. R. China
| | - Xiaoshuai Gao
- Department of UrologyInstitute of Urology (Laboratory of Reconstructive Urology)West China HospitalSichuan UniversityChengduSichuan610041P. R. China
| | - Jiawei Chen
- Department of UrologyInstitute of Urology (Laboratory of Reconstructive Urology)West China HospitalSichuan UniversityChengduSichuan610041P. R. China
| | - Liao Peng
- Department of UrologyInstitute of Urology (Laboratory of Reconstructive Urology)West China HospitalSichuan UniversityChengduSichuan610041P. R. China
| | - Shuang Chen
- Department of UrologyInstitute of Urology (Laboratory of Reconstructive Urology)West China HospitalSichuan UniversityChengduSichuan610041P. R. China
| | - Cai Tang
- Department of UrologyInstitute of Urology (Laboratory of Reconstructive Urology)West China HospitalSichuan UniversityChengduSichuan610041P. R. China
| | - Yi Dai
- Department of Urology and Pelvic surgeryWest China School of Public Health and West China Fourth HospitalSichuan UniversityChengduSichuan610041P. R. China
| | - Qiang Wei
- College of Polymer Science and EngineeringState Key Laboratory of Polymer Materials and EngineeringSichuan UniversityChengduSichuan610065P. R. China
| | - Deyi Luo
- Department of UrologyInstitute of Urology (Laboratory of Reconstructive Urology)West China HospitalSichuan UniversityChengduSichuan610041P. R. China
| |
Collapse
|
9
|
Sauer F, Grosser S, Shahryari M, Hayn A, Guo J, Braun J, Briest S, Wolf B, Aktas B, Horn L, Sack I, Käs JA. Changes in Tissue Fluidity Predict Tumor Aggressiveness In Vivo. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2303523. [PMID: 37553780 PMCID: PMC10502644 DOI: 10.1002/advs.202303523] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Indexed: 08/10/2023]
Abstract
Cancer progression is caused by genetic changes and associated with various alterations in cell properties, which also affect a tumor's mechanical state. While an increased stiffness has been well known for long for solid tumors, it has limited prognostic power. It is hypothesized that cancer progression is accompanied by tissue fluidization, where portions of the tissue can change position across different length scales. Supported by tabletop magnetic resonance elastography (MRE) on stroma mimicking collagen gels and microscopic analysis of live cells inside patient derived tumor explants, an overview is provided of how cancer associated mechanisms, including cellular unjamming, proliferation, microenvironment composition, and remodeling can alter a tissue's fluidity and stiffness. In vivo, state-of-the-art multifrequency MRE can distinguish tumors from their surrounding host tissue by their rheological fingerprints. Most importantly, a meta-analysis on the currently available clinical studies is conducted and universal trends are identified. The results and conclusions are condensed into a gedankenexperiment about how a tumor can grow and eventually metastasize into its environment from a physics perspective to deduce corresponding mechanical properties. Based on stiffness, fluidity, spatial heterogeneity, and texture of the tumor front a roadmap for a prognosis of a tumor's aggressiveness and metastatic potential is presented.
Collapse
Affiliation(s)
- Frank Sauer
- Soft Matter Physics DivisionPeter‐Debye‐Institute for Soft Matter Physics04103LeipzigGermany
| | - Steffen Grosser
- Soft Matter Physics DivisionPeter‐Debye‐Institute for Soft Matter Physics04103LeipzigGermany
- Institute for Bioengineering of CataloniaThe Barcelona Institute for Science and Technology (BIST)Barcelona08028Spain
| | - Mehrgan Shahryari
- Department of RadiologyCharité‐Universitätsmedizin10117BerlinGermany
| | - Alexander Hayn
- Department of HepatologyLeipzig University Hospital04103LeipzigGermany
| | - Jing Guo
- Department of RadiologyCharité‐Universitätsmedizin10117BerlinGermany
| | - Jürgen Braun
- Institute of Medical InformaticsCharité‐Universitätsmedizin10117BerlinGermany
| | - Susanne Briest
- Department of GynecologyLeipzig University Hospital04103LeipzigGermany
| | - Benjamin Wolf
- Department of GynecologyLeipzig University Hospital04103LeipzigGermany
| | - Bahriye Aktas
- Department of GynecologyLeipzig University Hospital04103LeipzigGermany
| | - Lars‐Christian Horn
- Division of Breast, Urogenital and Perinatal PathologyLeipzig University Hospital04103LeipzigGermany
| | - Ingolf Sack
- Department of RadiologyCharité‐Universitätsmedizin10117BerlinGermany
| | - Josef A. Käs
- Soft Matter Physics DivisionPeter‐Debye‐Institute for Soft Matter Physics04103LeipzigGermany
| |
Collapse
|
10
|
Makarova N, Lekka M, Gnanachandran K, Sokolov I. Mechanical Way To Study Molecular Structure of Pericellular Layer. ACS APPLIED MATERIALS & INTERFACES 2023; 15:35962-35972. [PMID: 37489588 PMCID: PMC10401571 DOI: 10.1021/acsami.3c06341] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 07/13/2023] [Indexed: 07/26/2023]
Abstract
Atomic force microscopy (AFM) has been used to study the mechanical properties of cells, in particular, malignant cells. Softening of various cancer cells compared to their nonmalignant counterparts has been reported for various cell types. However, in most AFM studies, the pericellular layer was ignored. As was shown, it could substantially change the measured cell rigidity and miss important information on the physical properties of the pericellular layer. Here we take into account the pericellular layer by using the brush model to do the AFM indentation study of bladder epithelial bladder nonmalignant (HCV29) and cancerous (TCCSUP) cells. It allows us to measure not only the quasistatic Young's modulus of the cell body but also the physical properties of the pericellular layer (the equilibrium length and grafting density). We found that the inner pericellular brush was longer for cancer cells, but its grafting density was similar to that found for nonmalignant cells. The outer brush was much shorter and less dense for cancer cells. Furthermore, we demonstrate a method to convert the obtained physical properties of the pericellular layer into biochemical language better known to the cell biology community. It is done by using heparinase I and neuraminidase enzymatic treatments that remove specific molecular parts of the pericellular layer. The presented here approach can also be used to decipher the molecular composition of not only pericellular but also other molecular layers.
Collapse
Affiliation(s)
- Nadezda Makarova
- Department
of Mechanical Engineering, Tufts University, Medford, Massachusetts 02155, United States
| | - Małgorzata Lekka
- Department
of Biophysical Microstructures, Institute
of Nuclear Physics PAN, PL-31342 Kraków, Poland
| | - Kajangi Gnanachandran
- Department
of Biophysical Microstructures, Institute
of Nuclear Physics PAN, PL-31342 Kraków, Poland
| | - Igor Sokolov
- Department
of Mechanical Engineering, Tufts University, Medford, Massachusetts 02155, United States
- Department
of Physics, Tufts University, Medford, Massachusetts 02155, United States
| |
Collapse
|
11
|
Bucki R, Iwamoto DV, Shi X, Kerr KE, Byfield FJ, Suprewicz Ł, Skłodowski K, Sutaria J, Misiak P, Wilczewska AZ, Ramachandran S, Wolfe A, Thanh MTH, Whalen E, Patteson AE, Janmey PA. Extracellular vimentin is sufficient to promote cell attachment, spreading, and motility by a mechanism involving N-acetyl glucosamine-containing structures. J Biol Chem 2023; 299:104963. [PMID: 37356720 PMCID: PMC10392088 DOI: 10.1016/j.jbc.2023.104963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 05/29/2023] [Accepted: 06/06/2023] [Indexed: 06/27/2023] Open
Abstract
Vimentin intermediate filaments form part of the cytoskeleton of mesenchymal cells, but under pathological conditions often associated with inflammation, vimentin filaments depolymerize as the result of phosphorylation or citrullination, and vimentin oligomers are secreted or released into the extracellular environment. In the extracellular space, vimentin can bind surfaces of cells and the extracellular matrix, and the interaction between extracellular vimentin and cells can trigger changes in cellular functions, such as activation of fibroblasts to a fibrotic phenotype. The mechanism by which extracellular vimentin binds external cell membranes and whether vimentin alone can act as an adhesive anchor for cells is largely uncharacterized. Here, we show that various cell types (normal and vimentin null fibroblasts, mesenchymal stem cells, and A549 lung carcinoma cells) attach to and spread on polyacrylamide hydrogel substrates covalently linked to vimentin. Using traction force microscopy and spheroid expansion assays, we characterize how different cell types respond to extracellular vimentin. Cell attachment to and spreading on vimentin-coated surfaces is inhibited by hyaluronic acid degrading enzymes, hyaluronic acid synthase inhibitors, soluble heparin or N-acetyl glucosamine, all of which are treatments that have little or no effect on the same cell types binding to collagen-coated hydrogels. These studies highlight the effectiveness of substrate-bound vimentin as a ligand for cells and suggest that carbohydrate structures, including the glycocalyx and glycosylated cell surface proteins that contain N-acetyl glucosamine, form a novel class of adhesion receptors for extracellular vimentin that can either directly support cell adhesion to a substrate or fine-tune the glycocalyx adhesive properties.
Collapse
Affiliation(s)
- Robert Bucki
- Department of Physiology, Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA; Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Białystok, Białystok, Poland.
| | - Daniel V Iwamoto
- Department of Physiology, Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Xuechen Shi
- Department of Physiology, Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Katherine E Kerr
- Department of Physiology, Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Fitzroy J Byfield
- Department of Physiology, Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Łukasz Suprewicz
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Białystok, Białystok, Poland
| | - Karol Skłodowski
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Białystok, Białystok, Poland
| | - Julian Sutaria
- Department of Physiology, Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Paweł Misiak
- Faculty of Chemistry, University of Białystok, Białystok, Poland
| | | | | | - Aaron Wolfe
- Ichor Life Sciences, Inc, LaFayette, New York, USA; Lewis School of Health Sciences, Clarkson University, Potsdam, New York, USA
| | - Minh-Tri Ho Thanh
- Physics Department, BioInspired Institute, Syracuse University, Syracuse, New York, USA
| | - Eli Whalen
- Physics Department, BioInspired Institute, Syracuse University, Syracuse, New York, USA
| | - Alison E Patteson
- Physics Department, BioInspired Institute, Syracuse University, Syracuse, New York, USA.
| | - Paul A Janmey
- Department of Physiology, Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
12
|
Rajendran AK, Sankar D, Amirthalingam S, Kim HD, Rangasamy J, Hwang NS. Trends in mechanobiology guided tissue engineering and tools to study cell-substrate interactions: a brief review. Biomater Res 2023; 27:55. [PMID: 37264479 DOI: 10.1186/s40824-023-00393-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 05/09/2023] [Indexed: 06/03/2023] Open
Abstract
Sensing the mechanical properties of the substrates or the matrix by the cells and the tissues, the subsequent downstream responses at the cellular, nuclear and epigenetic levels and the outcomes are beginning to get unraveled more recently. There have been various instances where researchers have established the underlying connection between the cellular mechanosignalling pathways and cellular physiology, cellular differentiation, and also tissue pathology. It has been now accepted that mechanosignalling, alone or in combination with classical pathways, could play a significant role in fate determination, development, and organization of cells and tissues. Furthermore, as mechanobiology is gaining traction, so do the various techniques to ponder and gain insights into the still unraveled pathways. This review would briefly discuss some of the interesting works wherein it has been shown that specific alteration of the mechanical properties of the substrates would lead to fate determination of stem cells into various differentiated cells such as osteoblasts, adipocytes, tenocytes, cardiomyocytes, and neurons, and how these properties are being utilized for the development of organoids. This review would also cover various techniques that have been developed and employed to explore the effects of mechanosignalling, including imaging of mechanosensing proteins, atomic force microscopy (AFM), quartz crystal microbalance with dissipation measurements (QCMD), traction force microscopy (TFM), microdevice arrays, Spatio-temporal image analysis, optical tweezer force measurements, mechanoscanning ion conductance microscopy (mSICM), acoustofluidic interferometric device (AID) and so forth. This review would provide insights to the researchers who work on exploiting various mechanical properties of substrates to control the cellular and tissue functions for tissue engineering and regenerative applications, and also will shed light on the advancements of various techniques that could be utilized to unravel the unknown in the field of cellular mechanobiology.
Collapse
Affiliation(s)
- Arun Kumar Rajendran
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
| | - Deepthi Sankar
- Polymeric Biomaterials Lab, School of Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, 682041, India
| | - Sivashanmugam Amirthalingam
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
- Institute of Engineering Research, Seoul National University, Seoul, 08826, Republic of Korea
| | - Hwan D Kim
- Department of Polymer Science and Engineering, Korea National University of Transportation, Chungju, 27469, Republic of Korea
- Department of Biomedical Engineering, Korea National University of Transportation, Chungju, 27469, Republic of Korea
| | - Jayakumar Rangasamy
- Polymeric Biomaterials Lab, School of Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, 682041, India.
| | - Nathaniel S Hwang
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea.
- Institute of Engineering Research, Seoul National University, Seoul, 08826, Republic of Korea.
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul, 08826, Republic of Korea.
- Bio-MAX/N-Bio Institute, Institute of Bio-Engineering, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
13
|
Mandal K, Sangabathuni S, Haghniaz R, Kawakita S, Mecwan M, Nakayama A, Zhang X, Edalati M, Huang W, Lopez Hernandez A, Jucaud V, Dokmeci MR, Khademhosseini A. Oxygen-generating microparticles downregulate HIF-1α expression, increase cardiac contractility, and mitigate ischemic injury. Acta Biomater 2023; 159:211-225. [PMID: 36669549 PMCID: PMC9992239 DOI: 10.1016/j.actbio.2023.01.030] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 12/20/2022] [Accepted: 01/11/2023] [Indexed: 01/19/2023]
Abstract
Myocardial hypoxia is the low oxygen tension in the heart tissue implicated in many diseases, including ischemia, cardiac dysfunction, or after heart procurement for transplantation. Oxygen-generating microparticles have recently emerged as a potential strategy for supplying oxygen to sustain cell survival, growth, and tissue functionality in hypoxia. Here, we prepared oxygen-generating microparticles with poly D,L-lactic-co-glycolic acid, and calcium peroxide (CPO), which yielded a continuous morphology capable of sustained oxygen release for up to 24 h. We demonstrated that CPO microparticles increased primary rat cardiomyocyte metabolic activity while not affecting cell viability during hypoxia. Moreover, hypoxia-inducible factor (HIF)-1α, which is upregulated during hypoxia, can be downregulated by delivering oxygen using CPO microparticles. Single-cell traction force microscopy data demonstrated that the reduced energy generated by hypoxic cells could be restored using CPO microparticles. We engineered cardiac tissues that showed higher contractility in the presence of CPO microparticles compared to hypoxic cells. Finally, we observed reduced myocardial injuries in ex vivo rabbit hearts treated with CPO microparticles. In contrast, an acute early myocardial injury was observed for the hearts treated with control saline solution in hypoxia. In conclusion, CPO microparticles improved cell and tissue contractility and gene expression while reducing hypoxia-induced myocardial injuries in the heart. STATEMENT OF SIGNIFICANCE: Oxygen-releasing microparticles can reduce myocardial ischemia, allograft rejection, or irregular heartbeats after heart transplantation. Here we present biodegradable oxygen-releasing microparticles that are capable of sustained oxygen release for more than 24 hrs. We then studied the impact of sustained oxygen release from microparticles on gene expresseion and cardiac cell and tissue function. Previous studies have not measured cardiac tissue or cell mechanics during hypoxia, which is important for understanding proper cardiac function and beating. Using traction force microscopy and an engineered tissue-on-a-chip, we demonstrated that our oxygen-releasing microparticles improve cell and tissue contractility during hypoxia while downregulating the HIF-1α expression level. Finally, using the microparticles, we showed reduced myocardial injuries in rabbit heart tissue, confirming the potential of the particles to be used for organ transplantation or tissue engineering.
Collapse
Affiliation(s)
- Kalpana Mandal
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, USA.
| | - Sivakoti Sangabathuni
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, USA; California NanoSystems Institute, University of California, Los Angeles, California 90095, USA
| | - Reihaneh Haghniaz
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, USA; California NanoSystems Institute, University of California, Los Angeles, California 90095, USA; Department of Bioengineering, University of California, Los Angeles, California 90095, USA
| | - Satoru Kawakita
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, USA
| | - Marvin Mecwan
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, USA
| | - Aya Nakayama
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, USA
| | - Xuexiang Zhang
- Department of Bioengineering, University of California, Los Angeles, California 90095, USA
| | - Masoud Edalati
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, USA
| | - Wei Huang
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, USA
| | - Ana Lopez Hernandez
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, USA
| | - Vadim Jucaud
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, USA
| | - Mehmet R Dokmeci
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, USA.
| | - Ali Khademhosseini
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, USA.
| |
Collapse
|
14
|
Xie W, Wei X, Kang H, Jiang H, Chu Z, Lin Y, Hou Y, Wei Q. Static and Dynamic: Evolving Biomaterial Mechanical Properties to Control Cellular Mechanotransduction. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2204594. [PMID: 36658771 PMCID: PMC10037983 DOI: 10.1002/advs.202204594] [Citation(s) in RCA: 38] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 12/28/2022] [Indexed: 06/17/2023]
Abstract
The extracellular matrix (ECM) is a highly dynamic system that constantly offers physical, biological, and chemical signals to embraced cells. Increasing evidence suggests that mechanical signals derived from the dynamic cellular microenvironment are essential controllers of cell behaviors. Conventional cell culture biomaterials, with static mechanical properties such as chemistry, topography, and stiffness, have offered a fundamental understanding of various vital biochemical and biophysical processes, such as cell adhesion, spreading, migration, growth, and differentiation. At present, novel biomaterials that can spatiotemporally impart biophysical cues to manipulate cell fate are emerging. The dynamic properties and adaptive traits of new materials endow them with the ability to adapt to cell requirements and enhance cell functions. In this review, an introductory overview of the key players essential to mechanobiology is provided. A biophysical perspective on the state-of-the-art manipulation techniques and novel materials in designing static and dynamic ECM-mimicking biomaterials is taken. In particular, different static and dynamic mechanical cues in regulating cellular mechanosensing and functions are compared. This review to benefit the development of engineering biomechanical systems regulating cell functions is expected.
Collapse
Affiliation(s)
- Wenyan Xie
- Department of BiotherapyState Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengduSichuan610065China
| | - Xi Wei
- Department of Mechanical EngineeringThe University of Hong KongHong KongChina
| | - Heemin Kang
- Department of Materials Science and EngineeringKorea UniversitySeoul02841South Korea
| | - Hong Jiang
- Department of BiotherapyState Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengduSichuan610065China
| | - Zhiqin Chu
- Department of Electrical and Electronic Engineering (Joint Appointment with School of Biomedical Sciences)The University of Hong KongHong KongChina
| | - Yuan Lin
- Department of Mechanical EngineeringThe University of Hong KongHong KongChina
| | - Yong Hou
- Department of Electrical and Electronic EngineeringThe University of Hong KongHong KongChina
- Institut für Chemie und BiochemieFreie Universität BerlinTakustrasse 314195BerlinGermany
| | - Qiang Wei
- College of Polymer Science and EngineeringState Key Laboratory of Polymer Materials and EngineeringSichuan UniversityChengdu610065China
| |
Collapse
|
15
|
Hara S, Inoue Y, Aoki S, Tanaka K, Shirasuna K, Iwata H. Beneficial Effect of Polysaccharide Gel Made of Xanthan Gum and Locust Bean Gum on Bovine Oocytes. Int J Mol Sci 2023; 24:ijms24043508. [PMID: 36834915 PMCID: PMC9963600 DOI: 10.3390/ijms24043508] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 02/06/2023] [Accepted: 02/08/2023] [Indexed: 02/12/2023] Open
Abstract
The present study examined the effect of polysaccharides gels made of xanthan gum and locust bean gum (gel culture system) on oocyte maturation and explored the molecules causing the beneficial effect of the gel culture system. Oocytes and cumulus cells complexes were collected from slaughterhouse-derived ovaries and cultured on a plastic plate or gel. The gel culture system improved the rate of development to the blastocyst stage. The oocytes that matured on the gel contained high lipid contents and F-actin formation, and the resultant 8-cell stage embryos had low DNA methylation levels compared to their plate counterparts. RNA sequencing of the oocytes and embryos revealed the differentially expressed genes between the gel and plate culture systems, and upstream regulator analysis revealed estradiol and TGFB1 as top activated upstream molecules. The medium of the gel culture system contained higher concentrations of estradiol and TGFB1 than that of the plate cultures system. Supplementation of the maturation medium with either estradiol or TGFB1 resulted in high lipid content in oocytes. In addition, TGFB1 improved the developmental ability of the oocytes and increased F-actin content while reducing DNA methylation levels in the 8-cell stage embryos. In conclusion, the gel culture system is useful for embryo production, potentially through the upregulation of TGFB1.
Collapse
|
16
|
Wu Y, Li X, Sun Y, Tan X, Wang C, Wang Z, Ye L. Multiscale design of stiffening and ROS scavenging hydrogels for the augmentation of mandibular bone regeneration. Bioact Mater 2023; 20:111-125. [PMID: 35663335 PMCID: PMC9133584 DOI: 10.1016/j.bioactmat.2022.05.021] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 04/19/2022] [Accepted: 05/10/2022] [Indexed: 12/13/2022] Open
Abstract
Although biomimetic hydrogels play an essential role in guiding bone remodeling, reconstructing large bone defects is still a significant challenge since bioinspired gels often lack osteoconductive capacity, robust mechanical properties and suitable antioxidant ability for bone regeneration. To address these challenges, we first engineered molecular design of hydrogels (gelatin/polyethylene glycol diacrylate/2-(dimethylamino)ethyl methacrylate, GPEGD), where their mechanical properties were significantly enhanced via introducing trace amounts of additives (0.5 wt%). The novel hybrid hydrogels show high compressive strength (>700 kPa), stiff modulus (>170 kPa) and strong ROS-scavenging ability. Furthermore, to endow the GPEGD hydrogels excellent osteoinductions, novel biocompatible, antioxidant and BMP-2 loaded polydopamine/heparin nanoparticles (BPDAH) were developed for functionalization of the GPEGD gels (BPDAH-GPEGD). In vitro results indicate that the antioxidant BPDAH-GPEGD is able to deplete elevated ROS levels to protect cells viability against ROS damage. More importantly, the BPDAH-GPEGD hydrogels have good biocompatibility and promote the osteo differentiation of preosteoblasts and bone regenerations. At 4 and 8 weeks after implantation of the hydrogels in a mandibular bone defect, Micro-computed tomography and histology results show greater bone volume and enhancements in the quality and rate of bone regeneration in the BPDAH-GPEGD hydrogels. Thus, the multiscale design of stiffening and ROS scavenging hydrogels could serve as a promising material for bone regeneration applications. Trace additives of DMAEMA markedly enhanced the mechanical performances of the gelatin-based hydrogels through molecular induced multiple crosslinking structures. Molecular design strategy combined with bioactive nanocomposites have a synergistically effects on promoting ROS scavenging ability and osteoactivity of the biomimetic hydrogels.
Collapse
Affiliation(s)
| | | | | | | | | | - Zhenming Wang
- Corresponding author. West China School of Stomatology, Sichuan University, No. 14, 3rd Section, South Renmin Road, Wuhou District, Chengdu, 610041, China.
| | - Ling Ye
- Corresponding author. West China School of Stomatology, Sichuan University, No. 14, 3rd Section, South Renmin Road, Wuhou District, Chengdu, 610041, China.
| |
Collapse
|
17
|
How do cells stiffen? Biochem J 2022; 479:1825-1842. [PMID: 36094371 DOI: 10.1042/bcj20210806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 08/21/2022] [Accepted: 08/24/2022] [Indexed: 11/17/2022]
Abstract
Cell stiffness is an important characteristic of cells and their response to external stimuli. In this review, we survey methods used to measure cell stiffness, summarize stimuli that alter cell stiffness, and discuss signaling pathways and mechanisms that control cell stiffness. Several pathological states are characterized by changes in cell stiffness, suggesting this property can serve as a potential diagnostic marker or therapeutic target. Therefore, we consider the effect of cell stiffness on signaling and growth processes required for homeostasis and dysfunction in healthy and pathological states. Specifically, the composition and structure of the cell membrane and cytoskeleton are major determinants of cell stiffness, and studies have identified signaling pathways that affect cytoskeletal dynamics both directly and by altered gene expression. We present the results of studies interrogating the effects of biophysical and biochemical stimuli on the cytoskeleton and other cellular components and how these factors determine the stiffness of both individual cells and multicellular structures. Overall, these studies represent an intersection of the fields of polymer physics, protein biochemistry, and mechanics, and identify specific mechanisms involved in mediating cell stiffness that can serve as therapeutic targets.
Collapse
|
18
|
Zhu JQ, Wu H, Li ZL, Xu XF, Xing H, Wang MD, Jia HD, Liang L, Li C, Sun LY, Wang YG, Shen F, Huang DS, Yang T. Responsive Hydrogels Based on Triggered Click Reactions for Liver Cancer. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2201651. [PMID: 35583434 DOI: 10.1002/adma.202201651] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 05/11/2022] [Indexed: 06/15/2023]
Abstract
Globally, liver cancer, which is one of the major cancers worldwide, has attracted the growing attention of technological researchers for its high mortality and limited treatment options. Hydrogels are soft 3D network materials containing a large number of hydrophilic monomers. By adding moieties such as nitrobenzyl groups to the network structure of a cross-linked nanocomposite hydrogel, the click reaction improves drug-release efficiency in vivo, which improves the survival rate and prolongs the survival time of liver cancer patients. The application of a nanocomposite hydrogel drug delivery system can not only enrich the drug concentration at the tumor site for a long time but also effectively prevents the distant metastasis of residual tumor cells. At present, a large number of researches have been working toward the construction of responsive nanocomposite hydrogel drug delivery systems, but there are few comprehensive articles to systematically summarize these discoveries. Here, this systematic review summarizes the synthesis methods and related applications of nanocomposite responsive hydrogels with actions to external or internal physiological stimuli. With different physical or chemical stimuli, the structural unit rearrangement and the controlled release of drugs can be used for responsive drug delivery in different states.
Collapse
Affiliation(s)
- Jia-Qi Zhu
- The Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College), Hangzhou, Zhejiang, 310014, China
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| | - Han Wu
- The Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College), Hangzhou, Zhejiang, 310014, China
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University (Naval Medical University), Shanghai, 200438, China
| | - Zhen-Li Li
- The Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College), Hangzhou, Zhejiang, 310014, China
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University (Naval Medical University), Shanghai, 200438, China
| | - Xin-Fei Xu
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University (Naval Medical University), Shanghai, 200438, China
| | - Hao Xing
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University (Naval Medical University), Shanghai, 200438, China
| | - Ming-Da Wang
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University (Naval Medical University), Shanghai, 200438, China
| | - Hang-Dong Jia
- The Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College), Hangzhou, Zhejiang, 310014, China
| | - Lei Liang
- The Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College), Hangzhou, Zhejiang, 310014, China
| | - Chao Li
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University (Naval Medical University), Shanghai, 200438, China
| | - Li-Yang Sun
- The Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College), Hangzhou, Zhejiang, 310014, China
| | - Yu-Guang Wang
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| | - Feng Shen
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University (Naval Medical University), Shanghai, 200438, China
| | - Dong-Sheng Huang
- The Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College), Hangzhou, Zhejiang, 310014, China
- School of Clinical Medicine, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Tian Yang
- The Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College), Hangzhou, Zhejiang, 310014, China
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University (Naval Medical University), Shanghai, 200438, China
| |
Collapse
|
19
|
Svarca A, Grava A, Dubnika A, Ramata-Stunda A, Narnickis R, Aunina K, Rieksta E, Boroduskis M, Jurgelane I, Locs J, Loca D. Calcium Phosphate/Hyaluronic Acid Composite Hydrogels for Local Antiosteoporotic Drug Delivery. Front Bioeng Biotechnol 2022; 10:917765. [PMID: 35866026 PMCID: PMC9294454 DOI: 10.3389/fbioe.2022.917765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 06/07/2022] [Indexed: 11/13/2022] Open
Abstract
Despite the bone ability of self-regeneration, large bone defects require surgical intervention. Likewise, when it comes to osteoporotic bone fractures, new approaches should be considered a supportive mechanism for the surgery. In recent years, more and more attention has been attracted to advanced drug delivery systems for local osteoporosis treatment, combining appropriate biomaterials with antiosteoporotic drugs, allowing simultaneously to regenerate the bone and locally treat the osteoporosis. Within the current research, hyaluronic acid/strontium ranelate (HA/SrRan), HA/calcium phosphate nanoparticles (HA/CaP NPs), and HA/CaP NPs/SrRan hydrogels were prepared. The effect of CaP and SrRan presence in the composites on the swelling behavior, gel fraction, molecular structure, microstructure, and SrRan and Sr2+ release, as well as in vitro cell viability was evaluated. Obtained results revealed that the route of CaP nanoparticle incorporation into the HA matrix had a significant effect on the hydrogel gel fraction, rheological properties, swelling behavior, and microstructure. Nevertheless, it had a negligible effect on the release kinetics of SrRan and Sr2+. The highest cell (3T3) viability (>80%) was observed for HA hydrogels, with and without SrRan. Moreover, the positive effect of SrRan on 3T3 cells was also demonstrated, showing a significant increase (up to 50%) in cell viability if the used concentrations of SrRan were in the range of 0.05-0.2 μg/ml.
Collapse
Affiliation(s)
- Alise Svarca
- Rudolfs Cimdins Riga Biomaterials Innovations and Development Centre of RTU, Institute of General Chemical Engineering, Faculty of Materials Science and Applied Chemistry, Riga Technical University, Riga, Latvia
- Baltic Biomaterials Centre of Excellence, Headquarters at Riga Technical University, Riga, Latvia
| | - Andra Grava
- Rudolfs Cimdins Riga Biomaterials Innovations and Development Centre of RTU, Institute of General Chemical Engineering, Faculty of Materials Science and Applied Chemistry, Riga Technical University, Riga, Latvia
- Baltic Biomaterials Centre of Excellence, Headquarters at Riga Technical University, Riga, Latvia
| | - Arita Dubnika
- Rudolfs Cimdins Riga Biomaterials Innovations and Development Centre of RTU, Institute of General Chemical Engineering, Faculty of Materials Science and Applied Chemistry, Riga Technical University, Riga, Latvia
- Baltic Biomaterials Centre of Excellence, Headquarters at Riga Technical University, Riga, Latvia
| | - Anna Ramata-Stunda
- Department of Microbiology and Biotechnology, Faculty of Biology, University of Latvia, Riga, Latvia
| | - Raimonds Narnickis
- Rudolfs Cimdins Riga Biomaterials Innovations and Development Centre of RTU, Institute of General Chemical Engineering, Faculty of Materials Science and Applied Chemistry, Riga Technical University, Riga, Latvia
| | - Kristine Aunina
- Rudolfs Cimdins Riga Biomaterials Innovations and Development Centre of RTU, Institute of General Chemical Engineering, Faculty of Materials Science and Applied Chemistry, Riga Technical University, Riga, Latvia
- Baltic Biomaterials Centre of Excellence, Headquarters at Riga Technical University, Riga, Latvia
| | - Eleonora Rieksta
- Rudolfs Cimdins Riga Biomaterials Innovations and Development Centre of RTU, Institute of General Chemical Engineering, Faculty of Materials Science and Applied Chemistry, Riga Technical University, Riga, Latvia
- Baltic Biomaterials Centre of Excellence, Headquarters at Riga Technical University, Riga, Latvia
| | - Martins Boroduskis
- Department of Microbiology and Biotechnology, Faculty of Biology, University of Latvia, Riga, Latvia
| | - Inga Jurgelane
- Rudolfs Cimdins Riga Biomaterials Innovations and Development Centre of RTU, Institute of General Chemical Engineering, Faculty of Materials Science and Applied Chemistry, Riga Technical University, Riga, Latvia
- Baltic Biomaterials Centre of Excellence, Headquarters at Riga Technical University, Riga, Latvia
| | - Janis Locs
- Rudolfs Cimdins Riga Biomaterials Innovations and Development Centre of RTU, Institute of General Chemical Engineering, Faculty of Materials Science and Applied Chemistry, Riga Technical University, Riga, Latvia
- Baltic Biomaterials Centre of Excellence, Headquarters at Riga Technical University, Riga, Latvia
| | - Dagnija Loca
- Rudolfs Cimdins Riga Biomaterials Innovations and Development Centre of RTU, Institute of General Chemical Engineering, Faculty of Materials Science and Applied Chemistry, Riga Technical University, Riga, Latvia
- Baltic Biomaterials Centre of Excellence, Headquarters at Riga Technical University, Riga, Latvia
| |
Collapse
|
20
|
Yang Y, Xu L, Wang J, Meng Q, Zhong S, Gao Y, Cui X. Recent advances in polysaccharide-based self-healing hydrogels for biomedical applications. Carbohydr Polym 2022; 283:119161. [DOI: 10.1016/j.carbpol.2022.119161] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 01/04/2022] [Accepted: 01/18/2022] [Indexed: 12/22/2022]
|
21
|
Li J, Shen J, Zhuang B, Wei M, Liu Y, Liu D, Yan W, Jia X, Jin Y. Light-triggered on-site rapid formation of antibacterial hydrogel dressings for accelerated healing of infected wounds. BIOMATERIALS ADVANCES 2022; 136:212784. [PMID: 35929299 DOI: 10.1016/j.bioadv.2022.212784] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 03/23/2022] [Accepted: 03/28/2022] [Indexed: 06/15/2023]
Abstract
An optimal wound dressing can seal variously shaped wounds and provide a complete barrier to resist bacterial invasion; more importantly, the dressing can be stretched or compressed when the wounds are subjected to external forces and quickly return to its original state after the forces are withdrawn. Here, we designed dressings with light-triggered on-site rapid formation of antibacterial hydrogel for the accelerated healing of infected wounds. The pro-hydrogel, composed of acrylamide (AM) and dopamine-hyaluronic acid-ε-poly-l-lysine (DA-HA-EPL), was filled into the Vibrio vulnificus-infected wound. A 405-nm blue light was exerted on the wound to rapidly photopolymerize AM to its polymer, i.e., polyacrylamide (PAM). A hydrogel network of PAM/DA-HA-EPL immediately formed on site within several seconds to insulate the wound. PAM/DA-HA-EPL possessed adhesion performance to adapt to changes in wound morphologies due to external forces. Moreover, it presented high antibacterial ability due to the presence of EPL, in vitro biocompatibility and the ability to promote cell migration. Vibrio vulnificus-infected wounds were established on full-thickness mouse skin, and the hydrogel dressing exhibited high healing efficiency in terms of skin tissue regeneration, collagen deposition, and angiogenesis. PAM/DA-HA-EPL is a promising hydrogel dressing for the accelerated healing of infected wounds.
Collapse
Affiliation(s)
- Jingfei Li
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Jintao Shen
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Bo Zhuang
- Department of Chemical Defense, Institute of NBC Defense, Beijing 102205, China
| | - Meng Wei
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Yan Liu
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Dongdong Liu
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Wenrui Yan
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Xueli Jia
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Yiguang Jin
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China.
| |
Collapse
|
22
|
Hang X, He S, Dong Z, Li Y, Huang Z, Zhang Y, Sun H, Lin L, Li H, Wang Y, Liu B, Wu N, Ren T, Fan Y, Lou J, Yang R, Jiang L, Chang L. High-Throughput DNA Tensioner Platform for Interrogating Mechanical Heterogeneity of Single Living Cells. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2106196. [PMID: 35322558 DOI: 10.1002/smll.202106196] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 12/24/2021] [Indexed: 06/14/2023]
Abstract
Cell mechanical forces play fundamental roles in regulating cellular responses to environmental stimulations. The shortcomings of conventional methods, including force resolution and cellular throughput, make them less accessible to mechanical heterogeneity at the single-cell level. Here, a DNA tensioner platform is introduced with high throughput (>10 000 cells per chip) and pN-level resolution. A microfluidic-based cell array is trapped on "hairpin-structured" DNA tensioners that enable transformation of the mechanical information of living cells into fluorescence signals. By using the platform, one can identify enhanced mechanical forces of drug-resistant cells as compared to their drug-sensitive counterparts, and mechanical differences between metastatic tumor cells in pleural effusion and nonmetastatic histiocytes. Further genetic analysis traces two genes, VEGFA and MINK1, that may play deterministic roles in regulating mechanical heterogeneities. In view of the ubiquity of cells' mechanical forces in the extracellular microenvironment (ECM), this platform shows wide potential to establish links of cellular mechanical heterogeneity to genetic heterogeneity.
Collapse
Affiliation(s)
- Xinxin Hang
- Key Laboratory of Biomechanics and Mechanobiology, Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, No. 37 Xueyuan Road, Haidian District, Beijing, 100083, China
| | - Shiqi He
- Key Laboratory of Biomechanics and Mechanobiology, Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, No. 37 Xueyuan Road, Haidian District, Beijing, 100083, China
| | - Zaizai Dong
- Key Laboratory of Biomechanics and Mechanobiology, Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, No. 37 Xueyuan Road, Haidian District, Beijing, 100083, China
| | - Yun Li
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, and China National Center for Bioinformation, Chinese Academy of Sciences, No. 1 Beichen West Road, Chaoyang District, Beijing, 100101, China
| | - Zheng Huang
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, and China National Center for Bioinformation, Chinese Academy of Sciences, No. 1 Beichen West Road, Chaoyang District, Beijing, 100101, China
| | - Yanruo Zhang
- Key Laboratory of RNA Biology, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, No. 15 Datun Road, Chaoyang District, Beijing, 100101, China
| | - Hong Sun
- Key Laboratory of Biomechanics and Mechanobiology, Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, No. 37 Xueyuan Road, Haidian District, Beijing, 100083, China
| | - Long Lin
- Key Laboratory of Biomechanics and Mechanobiology, Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, No. 37 Xueyuan Road, Haidian District, Beijing, 100083, China
| | - Hu Li
- Key Laboratory of Biomechanics and Mechanobiology, Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, No. 37 Xueyuan Road, Haidian District, Beijing, 100083, China
| | - Yang Wang
- Key Laboratory of Biomechanics and Mechanobiology, Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, No. 37 Xueyuan Road, Haidian District, Beijing, 100083, China
| | - Bing Liu
- Key Laboratory of Carcinogenesis and Translational Research of Ministry of Education, Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, No. 52 Fucheng Road, Haidian District, Beijing, 100142, China
| | - Nan Wu
- Key Laboratory of Carcinogenesis and Translational Research of Ministry of Education, Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, No. 52 Fucheng Road, Haidian District, Beijing, 100142, China
| | - Tianling Ren
- Beijing National Research Center for Information Science and Technology (BNRist), Institute of Microelectronics, Tsinghua University, No. 30 Shuangqing Road, Haidian District, Beijing, 100084, China
| | - Yubo Fan
- Key Laboratory of Biomechanics and Mechanobiology, Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, No. 37 Xueyuan Road, Haidian District, Beijing, 100083, China
| | - Jizhong Lou
- Key Laboratory of RNA Biology, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, No. 15 Datun Road, Chaoyang District, Beijing, 100101, China
| | - Ruiguo Yang
- Nebraska Center for Integrated Biomolecular Communication, Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| | - Lan Jiang
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, and China National Center for Bioinformation, Chinese Academy of Sciences, No. 1 Beichen West Road, Chaoyang District, Beijing, 100101, China
- College of Future Technology, and Sino-Danish College, University of Chinese Academy of Sciences, No. 19(A) Yuquan Road, Shijingshan District, Beijing, 100049, China
| | - Lingqian Chang
- Key Laboratory of Biomechanics and Mechanobiology, Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, No. 37 Xueyuan Road, Haidian District, Beijing, 100083, China
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Medical University, No. 81 Meishan Road, Hefei, 230032, China
| |
Collapse
|
23
|
Swoger M, Gupta S, Charrier EE, Bates M, Hehnly H, Patteson AE. Vimentin Intermediate Filaments Mediate Cell Morphology on Viscoelastic Substrates. ACS APPLIED BIO MATERIALS 2022; 5:552-561. [PMID: 34995457 PMCID: PMC8864613 DOI: 10.1021/acsabm.1c01046] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 12/27/2021] [Indexed: 12/12/2022]
Abstract
The ability of cells to take and change shape is a fundamental feature underlying development, wound repair, and tissue maintenance. Central to this process is physical and signaling interactions between the three cytoskeletal polymeric networks: F-actin, microtubules, and intermediate filaments (IFs). Vimentin is an IF protein that is essential to the mechanical resilience of cells and regulates cross-talk among the cytoskeleton, but its role in how cells sense and respond to the surrounding extracellular matrix is largely unclear. To investigate vimentin's role in substrate sensing, we designed polyacrylamide hydrogels that mimic the elastic and viscoelastic nature of in vivo tissues. Using wild-type and vimentin-null mouse embryonic fibroblasts, we show that vimentin enhances cell spreading on viscoelastic substrates, even though it has little effect in the limit of purely elastic substrates. Our results provide compelling evidence that vimentin modulates how cells sense and respond to their environment and thus plays a key role in cell mechanosensing.
Collapse
Affiliation(s)
- Maxx Swoger
- Physics
Department, Syracuse University, Syracuse, New York 13244, United States
- BioInspired
Institute, Syracuse University, Syracuse, New York 13244, United States
| | - Sarthak Gupta
- Physics
Department, Syracuse University, Syracuse, New York 13244, United States
- BioInspired
Institute, Syracuse University, Syracuse, New York 13244, United States
| | - Elisabeth E. Charrier
- Institute
of Medicine and Engineering, University
of Pennsylvania, Philadelphia, Pennsylvania 13210, United States
| | - Michael Bates
- Biology
Department, Syracuse University, Syracuse, New York 13244, United States
| | - Heidi Hehnly
- Biology
Department, Syracuse University, Syracuse, New York 13244, United States
| | - Alison E. Patteson
- Physics
Department, Syracuse University, Syracuse, New York 13244, United States
- BioInspired
Institute, Syracuse University, Syracuse, New York 13244, United States
| |
Collapse
|
24
|
Yang F, Liu S, Gu Y, Yan Y, Ding X, Zou L, Xu Z, Wang G. MicroRNA-22 promoted osteogenic differentiation of valvular interstitial cells by inhibiting CAB39 expression during aortic valve calcification. Cell Mol Life Sci 2022; 79:146. [PMID: 35190902 PMCID: PMC11073073 DOI: 10.1007/s00018-022-04177-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 12/30/2021] [Accepted: 01/29/2022] [Indexed: 12/15/2022]
Abstract
Calcific aortic valve disease (CAVD) is a common valve disease characterized by the fibro-calcific remodeling of the aortic valves, which is an actively regulated process involving osteogenic differentiation of valvular interstitial cells (VICs). MicroRNA (miRNA) is an essential regulator in diverse biological processes in cells. The present study aimed to explore the role and mechanism of miR-22 in the osteogenic differentiation of VICs. The expression profile of osteogenesis-related miRNAs was first detected in aortic valve tissue from CAVD patients (n = 33) and healthy controls (n = 12). miR-22 was highly expressed in calcified valve tissues (P < 0.01), and the expression was positively correlated with the expression of OPN (rs = 0.820, P < 0.01) and Runx2 (rs = 0.563, P < 0.01) in VICs isolated from mild or moderately calcified valves. The sustained high expression of miR-22 was also validated in an in-vitro VICs osteogenic model. Adenovirus-mediated gain-of-function and loss-of-function experiments were then performed. Overexpression of miR-22 significantly accelerated the calcification process of VICs, manifested by significant increases in calcium deposition, alkaline phosphate activity, and expression of osteoblastic differentiation markers. Conversely, inhibition of miR-22 significantly negated the calcification process. Subsequently, calcium-binding protein 39 (CAB39) was identified as a target of miR-22. Overexpression of miR-22 significantly reduced the expression of CAB39 in VICs, leading to decreased catalytic activity of the CAB39-LKB1-STRAD complex, which, in turn, exacerbated changes in the AMPK-mTOR signaling pathway, and ultimately accelerated the calcification process. In addition, ROS generation and autophagic activity during VIC calcification were also regulated by miR-22/CAB39 pathway. These results indicate that miR-22 is an important accelerator of the osteogenic differentiation of VICs, and a potential therapeutic target in CAVD.
Collapse
Affiliation(s)
- Fan Yang
- Department of Cardiovascular Surgery, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Suxuan Liu
- Department of Cardiology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Ying Gu
- Department of Cardiology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
- Department of Cardiology, Jinling Hospital, Nanjing University School of Medicine, Nanjing, 210002, China
| | - Yan Yan
- Department of Cardiovascular Surgery, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
- Department of Cardiothoracic Surgery, No.903 Hospital of Chinese People's Liberation Army, Hangzhou, Zhejiang, China
| | - Xueyan Ding
- Department of Cardiology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
- Department of Cardiology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, 310006, Zhejiang, China
| | - Liangjian Zou
- Department of Cardiovascular Surgery, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China.
| | - Zhiyun Xu
- Department of Cardiovascular Surgery, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China.
| | - Guokun Wang
- Department of Cardiovascular Surgery, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China.
| |
Collapse
|
25
|
Mierke CT. Viscoelasticity, Like Forces, Plays a Role in Mechanotransduction. Front Cell Dev Biol 2022; 10:789841. [PMID: 35223831 PMCID: PMC8864183 DOI: 10.3389/fcell.2022.789841] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 01/11/2022] [Indexed: 12/13/2022] Open
Abstract
Viscoelasticity and its alteration in time and space has turned out to act as a key element in fundamental biological processes in living systems, such as morphogenesis and motility. Based on experimental and theoretical findings it can be proposed that viscoelasticity of cells, spheroids and tissues seems to be a collective characteristic that demands macromolecular, intracellular component and intercellular interactions. A major challenge is to couple the alterations in the macroscopic structural or material characteristics of cells, spheroids and tissues, such as cell and tissue phase transitions, to the microscopic interferences of their elements. Therefore, the biophysical technologies need to be improved, advanced and connected to classical biological assays. In this review, the viscoelastic nature of cytoskeletal, extracellular and cellular networks is presented and discussed. Viscoelasticity is conceptualized as a major contributor to cell migration and invasion and it is discussed whether it can serve as a biomarker for the cells' migratory capacity in several biological contexts. It can be hypothesized that the statistical mechanics of intra- and extracellular networks may be applied in the future as a powerful tool to explore quantitatively the biomechanical foundation of viscoelasticity over a broad range of time and length scales. Finally, the importance of the cellular viscoelasticity is illustrated in identifying and characterizing multiple disorders, such as cancer, tissue injuries, acute or chronic inflammations or fibrotic diseases.
Collapse
Affiliation(s)
- Claudia Tanja Mierke
- Faculty of Physics and Earth Science, Peter Debye Institute of Soft Matter Physics, Biological Physics Division, University of Leipzig, Leipzig, Germany
| |
Collapse
|
26
|
Moretti L, Stalfort J, Barker TH, Abebayehu D. The interplay of fibroblasts, the extracellular matrix, and inflammation in scar formation. J Biol Chem 2022; 298:101530. [PMID: 34953859 PMCID: PMC8784641 DOI: 10.1016/j.jbc.2021.101530] [Citation(s) in RCA: 135] [Impact Index Per Article: 67.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 12/08/2021] [Indexed: 02/06/2023] Open
Abstract
Various forms of fibrosis, comprising tissue thickening and scarring, are involved in 40% of deaths across the world. Since the discovery of scarless functional healing in fetuses prior to a certain stage of development, scientists have attempted to replicate scarless wound healing in adults with little success. While the extracellular matrix (ECM), fibroblasts, and inflammatory mediators have been historically investigated as separate branches of biology, it has become increasingly necessary to consider them as parts of a complex and tightly regulated system that becomes dysregulated in fibrosis. With this new paradigm, revisiting fetal scarless wound healing provides a unique opportunity to better understand how this highly regulated system operates mechanistically. In the following review, we navigate the four stages of wound healing (hemostasis, inflammation, repair, and remodeling) against the backdrop of adult versus fetal wound healing, while also exploring the relationships between the ECM, effector cells, and signaling molecules. We conclude by singling out recent findings that offer promising leads to alter the dynamics between the ECM, fibroblasts, and inflammation to promote scarless healing. One factor that promises to be significant is fibroblast heterogeneity and how certain fibroblast subpopulations might be predisposed to scarless healing. Altogether, reconsidering fetal wound healing by examining the interplay of the various factors contributing to fibrosis provides new research directions that will hopefully help us better understand and address fibroproliferative diseases, such as idiopathic pulmonary fibrosis, liver cirrhosis, systemic sclerosis, progressive kidney disease, and cardiovascular fibrosis.
Collapse
Affiliation(s)
- Leandro Moretti
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
| | - Jack Stalfort
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
| | - Thomas Harrison Barker
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
| | - Daniel Abebayehu
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA.
| |
Collapse
|
27
|
Kreysing E, Hugh JM, Foster SK, Andresen K, Greenhalgh RD, Pillai EK, Dimitracopoulos A, Keyser UF, Franze K. Effective cell membrane tension is independent of polyacrylamide substrate stiffness. PNAS NEXUS 2022; 2:pgac299. [PMID: 36733291 PMCID: PMC9887938 DOI: 10.1093/pnasnexus/pgac299] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Accepted: 01/09/2023] [Indexed: 01/11/2023]
Abstract
Most animal cells are surrounded by a cell membrane and an underlying actomyosin cortex. Both structures are linked, and they are under tension. In-plane membrane tension and cortical tension both influence many cellular processes, including cell migration, division, and endocytosis. However, while actomyosin tension is regulated by substrate stiffness, how membrane tension responds to mechanical substrate properties is currently poorly understood. Here, we probed the effective membrane tension of neurons and fibroblasts cultured on glass and polyacrylamide substrates of varying stiffness using optical tweezers. In contrast to actomyosin-based traction forces, both peak forces and steady-state tether forces of cells cultured on hydrogels were independent of substrate stiffness and did not change after blocking myosin II activity using blebbistatin, indicating that tether and traction forces are not directly linked. Peak forces in fibroblasts on hydrogels were about twice as high as those in neurons, indicating stronger membrane-cortex adhesion in fibroblasts. Steady-state tether forces were generally higher in cells cultured on hydrogels than on glass, which we explain by a mechanical model. Our results provide new insights into the complex regulation of effective membrane tension and pave the way for a deeper understanding of the biological processes it instructs.
Collapse
Affiliation(s)
| | | | - Sarah K Foster
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3DY, UK,Systems Biology of Microbial Communities, Cluster of Excellence—CMFI, University of Tübingen, 72076 Tübingen, Germany
| | - Kurt Andresen
- Department of Physics, Gettysburg College, Gettysburg, PA 17325, USA
| | - Ryan D Greenhalgh
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3DY, UK
| | - Eva K Pillai
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3DY, UK
| | - Andrea Dimitracopoulos
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3DY, UK
| | | | | |
Collapse
|
28
|
Lei Y, Bortolin L, Benesch-Lee F, Oguntolu T, Dong Z, Bondah N, Billiar K. Hyaluronic acid regulates heart valve interstitial cell contraction in fibrin-based scaffolds. Acta Biomater 2021; 136:124-136. [PMID: 34592445 DOI: 10.1016/j.actbio.2021.09.046] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 09/23/2021] [Accepted: 09/23/2021] [Indexed: 11/30/2022]
Abstract
Heart valve disease is associated with high morbidity and mortality worldwide resulting in hundreds of thousands of heart valve replacements each year. Tissue engineered heart valves (TEHVs) have the potential to overcome the major limitations of traditional replacement valves; however, leaflet retraction has led to the failure of TEHVs in preclinical studies. As native unmodified hyaluronic acid (HA) is known to promote healthy tissue development in native heart valves, we hypothesize that adding unmodified HA to fibrin-based scaffolds common to tissue engineering will reduce retraction by increasing cell-scaffold interactions and density of the scaffolds. Using a custom high-throughput culture system, we found that incorporating HA into millimeter-scale fibrin-based cell-populated scaffolds increases initial fiber diameter and cell-scaffold interactions, causing a cascade of mechanical, morphological, and cellular responses. These changes lead to higher levels of scaffold compaction and stiffness, increased cell alignment, and less bundling of fibrin fibers by the cells during culture. These effects significantly reduce scaffold retraction and total contractile force each by around 25%. These findings increase our understanding of how HA alters tissue remodeling and could inform the design of the next generation of tissue engineered heart valves to help reduce retraction. STATEMENT OF SIGNIFICANCE: Tissue engineered heart valves (TEHVs) have the potential to overcome the major limitations of traditional replacement valves; however, leaflet retraction induced by excessive myofibroblast activation has led to failure in preclinical studies. Developing valves are rich in hyaluronic acid (HA), which helps maintain a physiological environment for tissue remodeling without retraction. We hypothesized that adding unmodified HA to TEHVs would reduce retraction by increasing cell-scaffold interactions and density of the scaffolds. Using a high-throughput tissue culture platform, we demonstrate that HA incorporation into a fibrin-based scaffold can significantly reduce tissue retraction and total contractile force by increasing fiber bundling and altering cell-mediated matrix remodeling, therefore increasing gel density and stiffness. These finding increase our knowledge of native HA's effects within the extracellular matrix, and provide a new tool for TEHV design.
Collapse
Affiliation(s)
- Ying Lei
- Biomedical Engineering Department, Worcester Polytechnic Institute, Gateway Park 4008, 60 Prescott, Worcester, MA 01605, USA
| | - Luciano Bortolin
- Biomedical Engineering Department, Worcester Polytechnic Institute, Gateway Park 4008, 60 Prescott, Worcester, MA 01605, USA
| | - Frank Benesch-Lee
- Biomedical Engineering Department, Worcester Polytechnic Institute, Gateway Park 4008, 60 Prescott, Worcester, MA 01605, USA
| | - Teniola Oguntolu
- Biomedical Engineering Department, Worcester Polytechnic Institute, Gateway Park 4008, 60 Prescott, Worcester, MA 01605, USA
| | - Zhijie Dong
- Biomedical Engineering Department, Worcester Polytechnic Institute, Gateway Park 4008, 60 Prescott, Worcester, MA 01605, USA
| | - Narda Bondah
- Biomedical Engineering Department, Worcester Polytechnic Institute, Gateway Park 4008, 60 Prescott, Worcester, MA 01605, USA
| | - Kristen Billiar
- Biomedical Engineering Department, Worcester Polytechnic Institute, Gateway Park 4008, 60 Prescott, Worcester, MA 01605, USA.
| |
Collapse
|
29
|
Janmey PA, Hinz B, McCulloch CA. Physics and Physiology of Cell Spreading in Two and Three Dimensions. Physiology (Bethesda) 2021; 36:382-391. [PMID: 34704856 PMCID: PMC8560373 DOI: 10.1152/physiol.00020.2021] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 08/02/2021] [Accepted: 08/08/2021] [Indexed: 01/01/2023] Open
Abstract
Cells spread on surfaces and within three-dimensional (3-D) matrixes as they grow, divide, and move. Both chemical and physical signals orchestrate spreading during normal development, wound healing, and pathological states such as fibrosis and tumor growth. Diverse molecular mechanisms drive different forms of cell spreading. This article discusses mechanisms by which cells spread in 2-D and 3-D and illustrates new directions in studies of this aspect of cell function.
Collapse
Affiliation(s)
- Paul A Janmey
- Institute for Medicine and Engineering, Department of Physiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Boris Hinz
- Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | | |
Collapse
|
30
|
Zhou B, Gao Z, Liu W, Wu X, Wang W. Important role of mechanical microenvironment on macrophage dysfunction during keloid pathogenesis. Exp Dermatol 2021; 31:375-380. [PMID: 34665886 DOI: 10.1111/exd.14473] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 09/27/2021] [Accepted: 10/17/2021] [Indexed: 12/17/2022]
Abstract
Keloid is considered as a tumor-like skin disease with multiple aetiologies including immunological factors and mechanical microenvironment. Macrophages are plastic and diverse immune cells that play a critical role in maintaining tissue homeostasis by removing dead cells, debris, pathogens and repairing tissues after inflammation. The imbalance of M1/M2 macrophages and disturbances in macrophage functions can steer the progression of chronic inflammation and lead to the development of pathological fibrosis in keloid disease. Recently, it has been shown that macrophages are sensitive to mechanical signals, especially stretching tension and tissue stiffness, which can determine macrophage polarization and functions. Higher stretching tension is known to be an important pathogenic factor of keloid, and the formation of keloid will lead to an increase in tissue stiffness. As little is known about the underlying reasons of macrophages dysfunction in keloid, an understanding of how the mechanical microenvironment interacting with macrophages and affecting their behaviours may help provide mechanism insights into keloid pathogenesis. We thus hypothesize that the synergistic effect of stretching tension and matrix stiffness may contribute to the major pathophysiological niche attributes of macrophages' in vivo mechanical microenvironment in keloids. These mechanism insights of how macrophages sense and respond to their mechanical microenvironment would propel the development of novel strategies for keloid treatment.
Collapse
Affiliation(s)
- Boya Zhou
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, Shanghai, China
| | - Zhen Gao
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, Shanghai, China
| | - Wei Liu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, Shanghai, China.,National Tissue Engineering Center of China, Shanghai, China
| | - Xiaoli Wu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, Shanghai, China
| | - Wenbo Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, Shanghai, China
| |
Collapse
|
31
|
Lekka M, Gnanachandran K, Kubiak A, Zieliński T, Zemła J. Traction force microscopy - Measuring the forces exerted by cells. Micron 2021; 150:103138. [PMID: 34416532 DOI: 10.1016/j.micron.2021.103138] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 07/15/2021] [Accepted: 08/09/2021] [Indexed: 12/23/2022]
Abstract
Cells generate mechanical forces (traction forces, TFs) while interacting with the extracellular matrix or neighbouring cells. Forces are generated by both cells and extracellular matrix (ECM) and transmitted within the cell-ECM or cell-cell contacts involving focal adhesions or adherens junctions. Within more than two decades, substantial progress has been achieved in techniques that measure TFs. One of the techniques is traction force microscopy (TFM). This review discusses the TFM and its advances in measuring TFs exerted by cells (single cells and multicellular systems) at cell-ECM and cell-cell junctional intracellular interfaces. The answers to how cells sense, adapt and respond to mechanical forces unravel their role in controlling and regulating cell behaviour in normal and pathological conditions.
Collapse
Affiliation(s)
- Małgorzata Lekka
- Institute of Nuclear Physics, Polish Academy of Sciences, PL-31342, Cracow, Poland.
| | | | - Andrzej Kubiak
- Institute of Nuclear Physics, Polish Academy of Sciences, PL-31342, Cracow, Poland
| | - Tomasz Zieliński
- Institute of Nuclear Physics, Polish Academy of Sciences, PL-31342, Cracow, Poland
| | - Joanna Zemła
- Institute of Nuclear Physics, Polish Academy of Sciences, PL-31342, Cracow, Poland
| |
Collapse
|
32
|
Lei R, Akins EA, Wong KCY, Repina NA, Wolf KJ, Dempsey GE, Schaffer DV, Stahl A, Kumar S. Multiwell Combinatorial Hydrogel Array for High-Throughput Analysis of Cell-ECM Interactions. ACS Biomater Sci Eng 2021; 7:2453-2465. [PMID: 34028263 DOI: 10.1021/acsbiomaterials.1c00065] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Biophysical cues in the extracellular matrix (ECM) regulate cell behavior in a complex, nonlinear, and interdependent manner. To quantify these important regulatory relationships and gain a comprehensive understanding of mechanotransduction, there is a need for high-throughput matrix platforms that enable parallel culture and analysis of cells in various matrix conditions. Here we describe a multiwell hyaluronic acid (HA) platform in which cells are cultured on combinatorial arrays of hydrogels spanning a range of elasticities and adhesivities. Our strategy utilizes orthogonal photopatterning of stiffness and adhesivity gradients, with the stiffness gradient implemented by a programmable light illumination system. The resulting platform allows individual treatment and analysis of each matrix environment while eliminating contributions of haptotaxis and durotaxis. In human mesenchymal stem cells, our platform recapitulates expected relationships between matrix stiffness, adhesivity, and cell mechanosensing. We further applied the platform to show that as integrin ligand density falls, cell adhesion and migration depend more strongly on CD44-mediated interactions with the HA backbone. We anticipate that our system could bear great value for mechanistic discovery and screening where matrix mechanics and adhesivity are expected to influence phenotype.
Collapse
Affiliation(s)
- Ruoxing Lei
- Department of Chemistry, Latimer Hall, University of California, Berkeley, Berkeley, California 94720, United States.,Department of Bioengineering, Stanley Hall, University of California, Berkeley, Berkeley, California 94720, United States
| | - Erin A Akins
- Department of Bioengineering, Stanley Hall, University of California, Berkeley, Berkeley, California 94720, United States.,University of California, Berkeley - University of California, San Francisco Graduate Program in Bioengineering, Stanley Hall, Berkeley, California 94720, United States
| | - Kelly C Y Wong
- Department of Bioengineering, Stanley Hall, University of California, Berkeley, Berkeley, California 94720, United States
| | - Nicole A Repina
- Department of Bioengineering, Stanley Hall, University of California, Berkeley, Berkeley, California 94720, United States.,University of California, Berkeley - University of California, San Francisco Graduate Program in Bioengineering, Stanley Hall, Berkeley, California 94720, United States
| | - Kayla J Wolf
- Department of Bioengineering, Stanley Hall, University of California, Berkeley, Berkeley, California 94720, United States.,University of California, Berkeley - University of California, San Francisco Graduate Program in Bioengineering, Stanley Hall, Berkeley, California 94720, United States
| | - Garrett E Dempsey
- Department of Nutritional Sciences and Toxicology, Morgan Hall, University of California, Berkeley, California 94720, United States
| | - David V Schaffer
- Department of Bioengineering, Stanley Hall, University of California, Berkeley, Berkeley, California 94720, United States.,University of California, Berkeley - University of California, San Francisco Graduate Program in Bioengineering, Stanley Hall, Berkeley, California 94720, United States.,Department of Molecular and Cell Biology, Life Sciences Addition, University of California, Berkeley, California 94720, United States.,Department of Chemical and Biomolecular Engineering, Gilman Hall, University of California, Berkeley, Berkeley, California 94720, United States
| | - Andreas Stahl
- University of California, Berkeley - University of California, San Francisco Graduate Program in Bioengineering, Stanley Hall, Berkeley, California 94720, United States.,Department of Nutritional Sciences and Toxicology, Morgan Hall, University of California, Berkeley, California 94720, United States
| | - Sanjay Kumar
- Department of Bioengineering, Stanley Hall, University of California, Berkeley, Berkeley, California 94720, United States.,University of California, Berkeley - University of California, San Francisco Graduate Program in Bioengineering, Stanley Hall, Berkeley, California 94720, United States.,Department of Chemical and Biomolecular Engineering, Gilman Hall, University of California, Berkeley, Berkeley, California 94720, United States.,Department of Bioengineering and Therapeutic Sciences, Byers Hall, University of California, San Francisco, San Francisco, California 94143, United States
| |
Collapse
|
33
|
Mishra YG, Manavathi B. Focal adhesion dynamics in cellular function and disease. Cell Signal 2021; 85:110046. [PMID: 34004332 DOI: 10.1016/j.cellsig.2021.110046] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 05/13/2021] [Indexed: 02/06/2023]
Abstract
Acting as a bridge between the cytoskeleton of the cell and the extra cellular matrix (ECM), the cell-ECM adhesions with integrins at their core, play a major role in cell signalling to direct mechanotransduction, cell migration, cell cycle progression, proliferation, differentiation, growth and repair. Biochemically, these adhesions are composed of diverse, yet an organised group of structural proteins, receptors, adaptors, various enzymes including protein kinases, phosphatases, GTPases, proteases, etc. as well as scaffolding molecules. The major integrin adhesion complexes (IACs) characterised are focal adhesions (FAs), invadosomes (podosomes and invadopodia), hemidesmosomes (HDs) and reticular adhesions (RAs). The varied composition and regulation of the IACs and their signalling, apart from being an integral part of normal cell survival, has been shown to be of paramount importance in various developmental and pathological processes. This review per-illustrates the recent advancements in the research of IACs, their crucial roles in normal as well as diseased states. We have also touched on few of the various methods that have been developed over the years to visualise IACs, measure the forces they exert and study their signalling and molecular composition. Having such pertinent roles in the context of various pathologies, these IACs need to be understood and studied to develop therapeutical targets. We have given an update to the studies done in recent years and described various techniques which have been applied to study these structures, thereby, providing context in furthering research with respect to IAC targeted therapeutics.
Collapse
Affiliation(s)
- Yasaswi Gayatri Mishra
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad 500046, India
| | - Bramanandam Manavathi
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad 500046, India.
| |
Collapse
|
34
|
Yu ZH, Ji YC, Li K, Liang T, Liu B, Chen HL, Ni L, Luo ZP, Yang HL. Stiffness of the extracellular matrix affects apoptosis of nucleus pulposus cells by regulating the cytoskeleton and activating the TRPV2 channel protein. Cell Signal 2021; 84:110005. [PMID: 33862152 DOI: 10.1016/j.cellsig.2021.110005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/26/2021] [Accepted: 04/05/2021] [Indexed: 11/26/2022]
Abstract
It is known that nucleus pulposus cells (NPs) play an important role in intervertebral disc degeneration (IVDD), and a previous study indicated that the stiffness of NP tissue changes during the degeneration process. However, the mechanism underlying the cellular response to ECM stiffness is still unclear. To analyze the effects of extracellular matrix (ECM) with different degrees of stiffness on NPs, we prepared polyacrylamide (PA) gels with different elastic moduli, and cells grown under different stiffness conditions were obtained and analyzed. The results showed that the spreading morphology of NPs changed significantly under increased ECM elastic modulus conditions and that TRPV2 and the PI3K / AKT signaling pathway were activated by stiffer ECM. At the same time, mitochondria released cytochrome c (Cyt c) and activated caspase proteins to promote the apoptosis of NPs. After TRPV2 was specifically knocked out, the activation of the PI3K / AKT signaling pathway decreased, and the release of Cyt c and NP apoptosis were reduced. These results indicate that TRPV2 is closely linked to the detection of extracellular mechanical signals, and that conversion of mechanical and biological signals plays an important role in regulating the biological behavior of cells. This study offers a new perspective on the cellular and biochemical events underlying IVDD which could result in novel treatments.
Collapse
Affiliation(s)
- Zhao-Hui Yu
- Department of Orthopaedics, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China; Orthopaedic Institute, Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Yi-Chao Ji
- Department of Orthopaedics, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China; Orthopaedic Institute, Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Kun Li
- Department of Orthopaedics, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Ting Liang
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Bo Liu
- Department of Orthopaedics, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China; Orthopaedic Institute, Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Hai-Lei Chen
- Department of Neurosurgery, Jiangsu Rudong County People's Hospital, Nantong City, Jiangsu Province, China
| | - Li Ni
- Department of Orthopaedics, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China; Orthopaedic Institute, Medical College, Soochow University, Suzhou, Jiangsu, China.
| | - Zong-Ping Luo
- Department of Orthopaedics, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China; Orthopaedic Institute, Medical College, Soochow University, Suzhou, Jiangsu, China.
| | - Hui-Lin Yang
- Department of Orthopaedics, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China; Orthopaedic Institute, Medical College, Soochow University, Suzhou, Jiangsu, China.
| |
Collapse
|
35
|
Budharaju H, Subramanian A, Sethuraman S. Recent advancements in cardiovascular bioprinting and bioprinted cardiac constructs. Biomater Sci 2021; 9:1974-1994. [DOI: 10.1039/d0bm01428a] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Three-dimensionally bioprinted cardiac constructs with biomimetic bioink helps to create native-equivalent cardiac tissues to treat patients with myocardial infarction.
Collapse
Affiliation(s)
- Harshavardhan Budharaju
- Tissue Engineering & Additive Manufacturing (TEAM) Lab
- Centre for Nanotechnology & Advanced Biomaterials
- ACBDE Innovation Centre
- School of Chemical & Biotechnology
- SASTRA Deemed to be University
| | - Anuradha Subramanian
- Tissue Engineering & Additive Manufacturing (TEAM) Lab
- Centre for Nanotechnology & Advanced Biomaterials
- ACBDE Innovation Centre
- School of Chemical & Biotechnology
- SASTRA Deemed to be University
| | - Swaminathan Sethuraman
- Tissue Engineering & Additive Manufacturing (TEAM) Lab
- Centre for Nanotechnology & Advanced Biomaterials
- ACBDE Innovation Centre
- School of Chemical & Biotechnology
- SASTRA Deemed to be University
| |
Collapse
|
36
|
Missirlis D, Haraszti T, Heckmann L, Spatz JP. Substrate Resistance to Traction Forces Controls Fibroblast Polarization. Biophys J 2020; 119:2558-2572. [PMID: 33217384 DOI: 10.1016/j.bpj.2020.10.043] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 10/21/2020] [Accepted: 10/23/2020] [Indexed: 12/25/2022] Open
Abstract
The mechanics of fibronectin-rich extracellular matrix regulate cell physiology in a number of diseases, prompting efforts to elucidate cell mechanosensing mechanisms at the molecular and cellular scale. Here, the use of fibronectin-functionalized silicone elastomers that exhibit considerable frequency dependence in viscoelastic properties unveiled the presence of two cellular processes that respond discreetly to substrate mechanical properties. Weakly cross-linked elastomers supported efficient focal adhesion maturation and fibroblast spreading because of an apparent stiff surface layer. However, they did not enable cytoskeletal and fibroblast polarization; elastomers with high cross-linking and low deformability were required for polarization. Our results suggest as an underlying reason for this behavior the inability of soft elastomer substrates to resist traction forces rather than a lack of sufficient traction force generation. Accordingly, mild inhibition of actomyosin contractility rescued fibroblast polarization even on the softer elastomers. Our findings demonstrate differential dependence of substrate physical properties on distinct mechanosensitive processes and provide a premise to reconcile previously proposed local and global models of cell mechanosensing.
Collapse
Affiliation(s)
- Dimitris Missirlis
- Max-Planck-Institute for Medical Research, Department of Cellular Biophysics, Heidelberg, Germany.
| | - Tamás Haraszti
- DWI-Leibniz Institute for Interactive Materials, Aachen, Germany; RWTH Aachen University, Institute for Technical and Macromolecular Chemistry, Aachen, Germany
| | - Lara Heckmann
- Max-Planck-Institute for Medical Research, Department of Cellular Biophysics, Heidelberg, Germany
| | - Joachim P Spatz
- Max-Planck-Institute for Medical Research, Department of Cellular Biophysics, Heidelberg, Germany; Heidelberg University, Department of Biophysical Chemistry, Physical Chemistry Institute, Heidelberg, Germany
| |
Collapse
|
37
|
Review of PIP2 in Cellular Signaling, Functions and Diseases. Int J Mol Sci 2020; 21:ijms21218342. [PMID: 33172190 PMCID: PMC7664428 DOI: 10.3390/ijms21218342] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 10/30/2020] [Accepted: 11/03/2020] [Indexed: 12/27/2022] Open
Abstract
Phosphoinositides play a crucial role in regulating many cellular functions, such as actin dynamics, signaling, intracellular trafficking, membrane dynamics, and cell-matrix adhesion. Central to this process is phosphatidylinositol bisphosphate (PIP2). The levels of PIP2 in the membrane are rapidly altered by the activity of phosphoinositide-directed kinases and phosphatases, and it binds to dozens of different intracellular proteins. Despite the vast literature dedicated to understanding the regulation of PIP2 in cells over past 30 years, much remains to be learned about its cellular functions. In this review, we focus on past and recent exciting results on different molecular mechanisms that regulate cellular functions by binding of specific proteins to PIP2 or by stabilizing phosphoinositide pools in different cellular compartments. Moreover, this review summarizes recent findings that implicate dysregulation of PIP2 in many diseases.
Collapse
|
38
|
Sitarska E, Diz-Muñoz A. Pay attention to membrane tension: Mechanobiology of the cell surface. Curr Opin Cell Biol 2020; 66:11-18. [PMID: 32416466 PMCID: PMC7594640 DOI: 10.1016/j.ceb.2020.04.001] [Citation(s) in RCA: 97] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 04/01/2020] [Accepted: 04/02/2020] [Indexed: 02/09/2023]
Abstract
The cell surface is a mechanobiological unit that encompasses the plasma membrane, its interacting proteins, and the complex underlying cytoskeleton. Recently, attention has been directed to the mechanics of the plasma membrane, and in particular membrane tension, which has been linked to diverse cellular processes such as cell migration and membrane trafficking. However, how tension across the plasma membrane is regulated and propagated is still not completely understood. Here, we review recent efforts to study the interplay between membrane tension and the cytoskeletal machinery and how they control cell form and function. We focus on factors that have been proposed to affect the propagation of membrane tension and as such could determine whether it can act as a global or local regulator of cell behavior. Finally, we discuss the limitations of the available tool kit as new approaches that reveal its dynamics in cells are needed to decipher how membrane tension regulates diverse cellular processes.
Collapse
Affiliation(s)
- Ewa Sitarska
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Meyerhofstrasse 1, 69117, Heidelberg, Germany
| | - Alba Diz-Muñoz
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Meyerhofstrasse 1, 69117, Heidelberg, Germany.
| |
Collapse
|
39
|
Ouyang M, Qian Z, Bu B, Jin Y, Wang J, Zhu Y, Liu L, Pan Y, Deng L. Sensing Traction Force on the Matrix Induces Cell-Cell Distant Mechanical Communications for Self-Assembly. ACS Biomater Sci Eng 2020; 6:5833-5848. [PMID: 33320570 DOI: 10.1021/acsbiomaterials.0c01035] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The long-range biomechanical force propagating across a large scale may reserve the capability to trigger coordinative responses within cell population such as during angiogenesis, epithelial tubulogenesis, and cancer metastasis. How cells communicate in a distant manner within the group for self-assembly remains largely unknown. Here, we found that airway smooth muscle cells (ASMCs) rapidly self-assembled into a well-constructed network on 3D Matrigel containing type I collagen (COL), which relied on long-range biomechanical force across the matrix to direct cell-cell distant interactions. Similar results happened by HUVEC cells to mimic angiogenesis. Interestingly, single ASMCs initiated multiple extended protrusions precisely pointing to neighboring cells in distance (100-300 μm away or 5-10 folds of the diameter of a round single cell), depending on traction force sensing. Individual ASMCs mechanosensed each other to move directionally on both nonfibrous Matrigel only and Matrigel containing fibrous COL but lost mutual sensing on the cross-linked gel or coated glass due to no long-range force transmission. The bead tracking assay demonstrated distant transmission of traction force (up to 400 μm) during the matrix deformation, and finite element method modeling confirmed the consistency between maximum strain distribution on the matrix and cell directional movements in experiments. Furthermore, ASMCs recruited COL from the hydrogel to build a fibrous network to mechanically stabilize the cell network. Our results revealed principally that cells can sense traction force transmitted through the matrix to initiate cell-cell distant mechanical communications, resulting in cell directional migration and coordinated cell and COL self-assembly with active matrix remodeling. As an interesting phenomenon, cells seem to be able to "make a phone call" via long-range biomechanics, which implicates physiological importance such as for tissue pattern formation.
Collapse
Affiliation(s)
- Mingxing Ouyang
- Institute of Biomedical Engineering and Health Sciences, School of Medicine, Changzhou University, 1 Gehu Road, Wujin District, Changzhou City, Jiangsu Province 213164, China
| | - Zhili Qian
- Institute of Biomedical Engineering and Health Sciences, School of Medicine, Changzhou University, 1 Gehu Road, Wujin District, Changzhou City, Jiangsu Province 213164, China
| | - Bing Bu
- Institute of Biomedical Engineering and Health Sciences, School of Medicine, Changzhou University, 1 Gehu Road, Wujin District, Changzhou City, Jiangsu Province 213164, China
| | - Yang Jin
- Institute of Biomedical Engineering and Health Sciences, School of Medicine, Changzhou University, 1 Gehu Road, Wujin District, Changzhou City, Jiangsu Province 213164, China
| | - Jiajia Wang
- Institute of Biomedical Engineering and Health Sciences, School of Medicine, Changzhou University, 1 Gehu Road, Wujin District, Changzhou City, Jiangsu Province 213164, China
| | - Yiming Zhu
- Institute of Biomedical Engineering and Health Sciences, School of Medicine, Changzhou University, 1 Gehu Road, Wujin District, Changzhou City, Jiangsu Province 213164, China
| | - Lei Liu
- Institute of Biomedical Engineering and Health Sciences, School of Medicine, Changzhou University, 1 Gehu Road, Wujin District, Changzhou City, Jiangsu Province 213164, China
| | - Yan Pan
- Institute of Biomedical Engineering and Health Sciences, School of Medicine, Changzhou University, 1 Gehu Road, Wujin District, Changzhou City, Jiangsu Province 213164, China
| | - Linhong Deng
- Institute of Biomedical Engineering and Health Sciences, School of Medicine, Changzhou University, 1 Gehu Road, Wujin District, Changzhou City, Jiangsu Province 213164, China
| |
Collapse
|
40
|
Schwager SC, Reinhart-King CA. Mechanobiology of microvesicle release, uptake, and microvesicle-mediated activation. CURRENT TOPICS IN MEMBRANES 2020; 86:255-278. [PMID: 33837695 DOI: 10.1016/bs.ctm.2020.08.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Microvesicles are small, membrane-bound vesicles that are shed from the plasma membrane of cells into the extracellular space. Microvesicles contain a variety of cargo not typically thought to be released from cells, including receptor tyrosine kinases, cytosolic signaling proteins, and microRNAs, which are transferred from donor cells to recipient cells. The transfer of microvesicle cargo can result in the transformation of recipient cells thereby supporting disease progression, including modified fibroblast metabolism, epithelial cell contractility, vascular remodeling, and immune cell inflammatory signaling. Additionally, microvesicles are believed to play prominent roles in cell-cell communication and disease progression as they are detected at elevated concentrations in diseased tissues. As microvesicle uptake by recipient cells can modulate cell function to promote disease progression, understanding the mechanisms and mechanosensitivity of microvesicle release, internalization, and the resulting signaling is crucial to fully comprehend their functions in disease. Here, we review recent advances in the understanding of actomyosin-regulated microvesicle biogenesis, microvesicle uptake via pinocytosis, and the resulting cellular transformation. We discuss the effects of altered cell contractility, mode of cell migration, and extracellular matrix compliance on microvesicle signaling, with direct implications in disease progression and identifying future therapeutic targets.
Collapse
Affiliation(s)
- Samantha C Schwager
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, United States
| | | |
Collapse
|
41
|
Schnauß J, Schmidt BUS, Brazel CB, Dogan S, Losert W, Anderegg U, Käs JA. Influence of hyaluronic acid binding on the actin cortex measured by optical forces. JOURNAL OF BIOPHOTONICS 2020; 13:e201960215. [PMID: 32246559 DOI: 10.1002/jbio.201960215] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 03/24/2020] [Accepted: 03/25/2020] [Indexed: 06/11/2023]
Abstract
Melanoma cells are often surrounded by hyaluronic acid (HA) rich environments, which are considered to promote tumor progression and metastasis. Induced effects in compound materials consisting of cells embedded in an extracellular matrix have been studied, however, alterations of the single cells have never been addressed. Here, we explicitly addressed single cell properties and measured HA-induced biomechanical changes via deformations induced solely by optical forces. With the optical stretcher setup, cells were deformed after culturing them in either the presence or absence of HA revealing the crucial interplay of HA with the CD44 receptor. To assess the role of CD44 in transducing effects of HA, we compared a CD44 expressing variant of the melanoma cell line RPM-MC to its natural CD44-negative counterpart. Our measurements revealed a significant stiffness change, which we attribute to changes of the actin cytoskeleton.
Collapse
Affiliation(s)
- Jörg Schnauß
- Leipzig University, Faculty of Physics and Earth Sciences, Peter Debye Institute for Soft Matter Physics, Leipzig, Germany
- Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| | - B U Sebastian Schmidt
- University of Maryland, Institute for Physical Science and Technology, Physical Sciences Complex, College Park, MD, USA
| | - Christina B Brazel
- Department of Dermatology, Leipzig University, Medical Faculty, Leipzig, Germany
| | - Senol Dogan
- Leipzig University, Faculty of Physics and Earth Sciences, Peter Debye Institute for Soft Matter Physics, Leipzig, Germany
| | - Wolfgang Losert
- University of Maryland, Institute for Physical Science and Technology, Physical Sciences Complex, College Park, MD, USA
| | - Ulf Anderegg
- Department of Dermatology, Leipzig University, Medical Faculty, Leipzig, Germany
| | - Josef A Käs
- Leipzig University, Faculty of Physics and Earth Sciences, Peter Debye Institute for Soft Matter Physics, Leipzig, Germany
| |
Collapse
|
42
|
Yu L, Hou Y, Xie W, Camacho JLC, Cheng C, Holle A, Young J, Trappmann B, Zhao W, Melzig MF, Cavalcanti-Adam EA, Zhao C, Spatz JP, Wei Q, Haag R. Ligand Diffusion Enables Force-Independent Cell Adhesion via Activating α5β1 Integrin and Initiating Rac and RhoA Signaling. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e2002566. [PMID: 32537880 DOI: 10.1002/adma.202002566] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 05/12/2020] [Indexed: 05/12/2023]
Abstract
Cells reside in a dynamic microenvironment in which adhesive ligand availability, density, and diffusivity are key factors regulating cellular behavior. Here, the cellular response to integrin-binding ligand dynamics by directly controlling ligand diffusivity via tunable ligand-surface interactions is investigated. Interestingly, cell spread on the surfaces with fast ligand diffusion is independent of myosin-based force generation. Fast ligand diffusion enhances α5β1 but not αvβ3 integrin activation and initiates Rac and RhoA but not ROCK signaling, resulting in lamellipodium-based fast cell spreading. Meanwhile, on surfaces with immobile ligands, αvβ3 and α5β1 integrins synergistically initiate intracellular-force-based canonical mechanotransduction pathways to enhance cell adhesion and osteogenic differentiation of stem cells. These results indicate the presence of heretofore-unrecognized pathways, distinct from canonical actomyosin-driven mechanisms, that are capable of promoting cell adhesion.
Collapse
Affiliation(s)
- Leixiao Yu
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, Berlin, 14195, Germany
| | - Yong Hou
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, Berlin, 14195, Germany
| | - Wenyan Xie
- Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Str. 2+4, Berlin, 14195, Germany
| | - Jose Luis Cuellar Camacho
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, Berlin, 14195, Germany
| | - Chong Cheng
- State Key Laboratory of Polymer Materials and Engineering, College of Polymer Science and Engineering, Sichuan University, Chengdu, 610065, China
| | - Andrew Holle
- Department of Cellular Biophysics, Max Planck Institute for Medical Research, Jahnstraße 29, Heidelberg, 69120, Germany
- Department of Biophysical Chemistry, Institute of Physical Chemistry, University of Heidelberg, Im Neuenheimer Feld 253, Heidelberg, 69120, Germany
| | - Jennifer Young
- Department of Cellular Biophysics, Max Planck Institute for Medical Research, Jahnstraße 29, Heidelberg, 69120, Germany
- Department of Biophysical Chemistry, Institute of Physical Chemistry, University of Heidelberg, Im Neuenheimer Feld 253, Heidelberg, 69120, Germany
| | - Britta Trappmann
- Max Planck Institute for Molecular Biomedicine, Röntgenstr. 20, Münster, 48149, Germany
| | - Weifeng Zhao
- State Key Laboratory of Polymer Materials and Engineering, College of Polymer Science and Engineering, Sichuan University, Chengdu, 610065, China
| | - Matthias F Melzig
- Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Str. 2+4, Berlin, 14195, Germany
| | - Elisabetta A Cavalcanti-Adam
- Department of Cellular Biophysics, Max Planck Institute for Medical Research, Jahnstraße 29, Heidelberg, 69120, Germany
- Department of Biophysical Chemistry, Institute of Physical Chemistry, University of Heidelberg, Im Neuenheimer Feld 253, Heidelberg, 69120, Germany
- Central Scientific Facility "Cellular Biotechnology", Jahnstr. 29, Heidelberg, 69120, Germany
| | - Changsheng Zhao
- State Key Laboratory of Polymer Materials and Engineering, College of Polymer Science and Engineering, Sichuan University, Chengdu, 610065, China
| | - Joachim P Spatz
- Department of Cellular Biophysics, Max Planck Institute for Medical Research, Jahnstraße 29, Heidelberg, 69120, Germany
- Department of Biophysical Chemistry, Institute of Physical Chemistry, University of Heidelberg, Im Neuenheimer Feld 253, Heidelberg, 69120, Germany
| | - Qiang Wei
- State Key Laboratory of Polymer Materials and Engineering, College of Polymer Science and Engineering, Sichuan University, Chengdu, 610065, China
| | - Rainer Haag
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, Berlin, 14195, Germany
| |
Collapse
|
43
|
Li Y, Randriantsilefisoa R, Chen J, Cuellar-Camacho JL, Liang W, Li W. Matrix Stiffness Regulates Chemosensitivity, Stemness Characteristics, and Autophagy in Breast Cancer Cells. ACS APPLIED BIO MATERIALS 2020; 3:4474-4485. [DOI: 10.1021/acsabm.0c00448] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Yan Li
- Institute of Chemistry and Biochemistry, Free University of Berlin, Takustr. 3, 14195 Berlin, Germany
| | | | - Jie Chen
- Institute of Chemistry and Biochemistry, Free University of Berlin, Takustr. 3, 14195 Berlin, Germany
| | - Jose Luis Cuellar-Camacho
- Institute of Chemistry and Biochemistry, Free University of Berlin, Takustr. 3, 14195 Berlin, Germany
| | - Wanjun Liang
- Institute of Chemistry and Biochemistry, Free University of Berlin, Takustr. 3, 14195 Berlin, Germany
| | - Wenzhong Li
- Institute of Chemistry and Biochemistry, Free University of Berlin, Takustr. 3, 14195 Berlin, Germany
| |
Collapse
|
44
|
Serna-Márquez N, Rodríguez-Hernández A, Ayala-Reyes M, Martínez-Hernández LO, Peña-Rico MÁ, Carretero-Ortega J, Hautefeuille M, Vázquez-Victorio G. Fibrillar Collagen Type I Participates in the Survival and Aggregation of Primary Hepatocytes Cultured on Soft Hydrogels. Biomimetics (Basel) 2020; 5:E30. [PMID: 32630500 PMCID: PMC7345357 DOI: 10.3390/biomimetics5020030] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 06/15/2020] [Accepted: 06/23/2020] [Indexed: 12/11/2022] Open
Abstract
Liver is an essential organ that carries out multiple functions such as glycogen storage, the synthesis of plasma proteins, and the detoxification of xenobiotics. Hepatocytes are the parenchyma that sustain almost all the functions supported by this organ. Hepatocytes and non-parenchymal cells respond to the mechanical alterations that occur in the extracellular matrix (ECM) caused by organogenesis and regenerating processes. Rearrangements of the ECM modify the composition and mechanical properties that result in specific dedifferentiation programs inside the hepatic cells. Quiescent hepatocytes are embedded in the soft ECM, which contains an important concentration of fibrillar collagens in combination with a basement membrane-associated matrix (BM). This work aims to evaluate the role of fibrillar collagens and BM on actin cytoskeleton organization and the function of rat primary hepatocytes cultured on soft elastic polyacrylamide hydrogels (PAA HGs). We used rat tail collagen type I and Matrigel® as references of fibrillar collagens and BM respectively and mixed different percentages of collagen type I in combination with BM. We also used peptides obtained from decellularized liver matrices (dECM). Remarkably, hepatocytes showed a poor adhesion in the absence of collagen on soft PAA HGs. We demonstrated that collagen type I inhibited apoptosis and activated extracellular signal-regulated kinases 1/2 (ERK1/2) in primary hepatocytes cultured on soft hydrogels. Epidermal growth factor (EGF) was not able to rescue cell viability in conjugated BM but affected cell aggregation in soft PAA HGs conjugated with combinations of different proportions of collagen and BM. Interestingly, actin cytoskeleton was localized and preserved close to plasma membrane (cortical actin) and proximal to intercellular ducts (canaliculi-like structures) in soft conditions; however, albumin protein expression was not preserved, even though primary hepatocytes did not remodel their actin cytoskeleton significantly in soft conditions. This investigation highlights the important role of fibrillar collagens on soft hydrogels for the maintenance of survival and aggregation of the hepatocytes. Data suggest evaluating the conditions that allow the establishment of optimal biomimetic environments for physiology and cell biology studies, where the phenotype of primary cells may be preserved for longer periods of time.
Collapse
Affiliation(s)
- Nathalia Serna-Márquez
- Laboratorio Nacional de Soluciones Biomiméticas para Diagnóstico y Terapia (LaNSBioDyT), Facultad de Ciencias, Universidad Nacional Autónoma de Mexico, Ciudad de México CP 04510, Mexico; (N.S.-M.); (A.R.-H.); (M.A.-R.); (L.O.M.-H.); (J.C.-O.); (M.H.)
| | - Adriana Rodríguez-Hernández
- Laboratorio Nacional de Soluciones Biomiméticas para Diagnóstico y Terapia (LaNSBioDyT), Facultad de Ciencias, Universidad Nacional Autónoma de Mexico, Ciudad de México CP 04510, Mexico; (N.S.-M.); (A.R.-H.); (M.A.-R.); (L.O.M.-H.); (J.C.-O.); (M.H.)
| | - Marisol Ayala-Reyes
- Laboratorio Nacional de Soluciones Biomiméticas para Diagnóstico y Terapia (LaNSBioDyT), Facultad de Ciencias, Universidad Nacional Autónoma de Mexico, Ciudad de México CP 04510, Mexico; (N.S.-M.); (A.R.-H.); (M.A.-R.); (L.O.M.-H.); (J.C.-O.); (M.H.)
| | - Lorena Omega Martínez-Hernández
- Laboratorio Nacional de Soluciones Biomiméticas para Diagnóstico y Terapia (LaNSBioDyT), Facultad de Ciencias, Universidad Nacional Autónoma de Mexico, Ciudad de México CP 04510, Mexico; (N.S.-M.); (A.R.-H.); (M.A.-R.); (L.O.M.-H.); (J.C.-O.); (M.H.)
- Instituto de Biotecnología, Universidad del Papaloapan, Tuxtepec CP 68301, Oaxaca, Mexico;
| | - Miguel Ángel Peña-Rico
- Instituto de Biotecnología, Universidad del Papaloapan, Tuxtepec CP 68301, Oaxaca, Mexico;
| | - Jorge Carretero-Ortega
- Laboratorio Nacional de Soluciones Biomiméticas para Diagnóstico y Terapia (LaNSBioDyT), Facultad de Ciencias, Universidad Nacional Autónoma de Mexico, Ciudad de México CP 04510, Mexico; (N.S.-M.); (A.R.-H.); (M.A.-R.); (L.O.M.-H.); (J.C.-O.); (M.H.)
| | - Mathieu Hautefeuille
- Laboratorio Nacional de Soluciones Biomiméticas para Diagnóstico y Terapia (LaNSBioDyT), Facultad de Ciencias, Universidad Nacional Autónoma de Mexico, Ciudad de México CP 04510, Mexico; (N.S.-M.); (A.R.-H.); (M.A.-R.); (L.O.M.-H.); (J.C.-O.); (M.H.)
- Departamento de Física, Facultad de Ciencias, Universidad Nacional Autónoma de Mexico, Ciudad de México CP 04510, Mexico
| | - Genaro Vázquez-Victorio
- Laboratorio Nacional de Soluciones Biomiméticas para Diagnóstico y Terapia (LaNSBioDyT), Facultad de Ciencias, Universidad Nacional Autónoma de Mexico, Ciudad de México CP 04510, Mexico; (N.S.-M.); (A.R.-H.); (M.A.-R.); (L.O.M.-H.); (J.C.-O.); (M.H.)
- Departamento de Física, Facultad de Ciencias, Universidad Nacional Autónoma de Mexico, Ciudad de México CP 04510, Mexico
| |
Collapse
|
45
|
Hong F, Gao Y, Li Y, Zheng L, Xu F, Li X. Inhibition of HIF1A-AS1 promoted starvation-induced hepatocellular carcinoma cell apoptosis by reducing HIF-1α/mTOR-mediated autophagy. World J Surg Oncol 2020; 18:113. [PMID: 32473641 PMCID: PMC7261383 DOI: 10.1186/s12957-020-01884-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Accepted: 05/15/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is still a major health burden in China considering its high incidence and mortality. Long non-coding RNAs (lncRNAs) were found playing vital roles in tumor progression, suggesting a new way of diagnosis and prognosis prediction, or treatment of HCC. This study was designed to investigate the role of HIF1A-AS1 during the progression of HCC and to explore its related mechanisms. METHODS The expression of HIF1A-AS1 was detected in 50 paired carcinoma tissues and adjacent normal tissues by quantitative real-time PCR assay. HCC cell apoptosis was induced by nutrient-deficient culture medium and detected by Cell Counting Kit-8 and flow cytometer assays. HIF1A-AS1 inhibition in HCC cells was accomplished by small interfering RNA transfection. RESULTS HIF1A-AS1 was overexpressed in HCC tissues and was associated with tumor size, TNM stage, and lymph node metastasis. Compared with the low HIF1A-AS1 group, the high HIF1A-AS1 group had a shorter overall survival and a worse disease-free survival. HIF1A-AS1 expression was significantly higher in HCC cell lines (7721 and Huh7) than that in normal hepatocyte cell line L02 under normal culture condition. However, under nutrient-deficient condition, HIF1A-AS1 expression was significantly increased in both HCC and normal hepatocyte cell lines and was increased with the prolongation of nutrient-free culture. Inhibition of HIF1A-AS1 promoted starvation-induced HCC cell apoptosis. Furthermore, inhibition of HIF1A-AS1 could also reduce starvation-induced HCC cell autophagy. The expression of HIF-1α and phosphorylated mTOR was significantly decreased in HCC cells after HIF1A-AS1 inhibition. CONCLUSIONS HIF1A-AS1, overexpressed in HCC and associated with HCC prognosis, could regulate starvation-induced HCC cell apoptosis by reducing HIF-1α/mTOR-mediated autophagy, promoting HCC cell progression.
Collapse
Affiliation(s)
- Fenfen Hong
- Division of Gastroenterology and Hepatology, Yinzhou Hospital Affiliated to Medical School of Ningbo University, Ningbo, Zhejiang, 315000, China
| | - Yu Gao
- Hongkou Branch of Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Yang Li
- Department of Cardiovascular Surgery, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Linfeng Zheng
- Department of Radiology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, 200080, China
| | - Feng Xu
- Division of Gastroenterology and Hepatology, Yinzhou Hospital Affiliated to Medical School of Ningbo University, Ningbo, Zhejiang, 315000, China. .,, Ningbo, China.
| | - Xianpeng Li
- Division of Gastroenterology and Hepatology, Yinzhou Hospital Affiliated to Medical School of Ningbo University, Ningbo, Zhejiang, 315000, China. .,, Ningbo, China.
| |
Collapse
|
46
|
Tian Z, Wang Z, Chen Y, Qu S, Liu C, Chen F, Ma L, Zhu J. Bioinformatics Analysis of Prognostic Tumor Microenvironment-Related Genes in the Tumor Microenvironment of Hepatocellular Carcinoma. Med Sci Monit 2020; 26:e922159. [PMID: 32231177 PMCID: PMC7146066 DOI: 10.12659/msm.922159] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Background Growing evidence shows that the tumor microenvironment plays a crucial role in the pathogenesis of hepatocellular carcinoma (HCC). The present work aimed to screen tumor microenvironment-related genes strongly related to prognosis and to construct a prognostic gene expression model for HCC. Material/Methods We downloaded gene expression data of 371 HCC patients in The Cancer Genome Atlas (TCGA). A novel ESTIMATE algorithm was applied to calculate immune scores and stromal scores for each patient. Then, the differentially-expressed genes (DEGs) were detected according to the immune and stromal scores, and tumor microenvironment-related genes were further explored. Univariate, Lasso, and multivariate Cox analyses were performed to build the tumor microenvironment-related prediction model. Results Stromal and immune scores were calculated and were found to be correlated with the 3-year prognosis of HCC patients. DEGs were detected according to the stromal and immune scores. There were 49 genes with prognostic value in both TCGA and ICGC (International Cancer Genome Consortium) considered as prognostic tumor microenvironment-related genes. Univariate, Lasso, and multivariate Cox analyses were conducted. A novel 2-gene signature (IL18RAP and GPR182) was built for HCC 3-year prognosis prediction. The 2-gene signature was regarded as an independent prognostic predictor that was correlated with 3-year survival rate, as shown by Cox regression analysis. Conclusions This study offers a novel 2-gene signature to predict overall survival of patients with HCC, which has the potential to be used as an independent prognostic predictor. Overall, this study reveals more details about the tumor microenvironment in HCC and offers novel candidate biomarkers.
Collapse
Affiliation(s)
- Zongbiao Tian
- Department of Gastroenterology, TengZhou Central People's Hospital, Tengzhou, Shandong, China (mainland)
| | - Zheng Wang
- Department of Infectious Diseases, Qilu Hospital, Shandong University, Jinan, Shandong, China (mainland)
| | - Yanfeng Chen
- Department of Gastroenterology, TengZhou Central People's Hospital, Tengzhou, Shandong, China (mainland)
| | - Shuoying Qu
- Department of Clinical Laboratory, Qilu Hospital, Shandong University, Jinan, Shandong, China (mainland)
| | - Changhong Liu
- Department of Gastroenterology, Shandong Provincial Qianfoshan Hospital, The First Hospital Affiliated with Shandong First Medical University, Jinan, Shandong, China (mainland)
| | - Fengzhe Chen
- Department of Infectious Diseases, Qilu Hospital, Shandong University, Jinan, Shandong, China (mainland)
| | - Lixian Ma
- Department of Infectious Diseases, Qilu Hospital, Shandong University, Jinan, Shandong, China (mainland)
| | - Jie Zhu
- Department of Infectious Diseases, Qilu Hospital, Shandong University, Jinan, Shandong, China (mainland)
| |
Collapse
|
47
|
Mandal K, Gong Z, Rylander A, Shenoy VB, Janmey PA. Opposite responses of normal hepatocytes and hepatocellular carcinoma cells to substrate viscoelasticity. Biomater Sci 2020; 8:1316-1328. [PMID: 31903466 DOI: 10.1039/c9bm01339c] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The cellular microenvironment plays a critical role in cell differentiation, proliferation, migration, and homeostasis. Recent studies have shown the importance of substrate viscosity in determining cellular function. Here, we study the mechanoresponse of normal hepatocytes and hepatocellular carcinoma cells (HCC) to elastic and viscoelastic substrates using the Huh7 cell line derived from a human liver tumor and primary human hepatocytes (PHH). Unlike PHH and fibroblasts, which respond to viscoelastic substrates by reducing spreading area and actin bundle assembly compared to purely elastic substrates of the same stiffness, Huh7 cells spread faster on viscoelastic substrates than on purely elastic substrates. The steady state spreading areas of Huh7 cells are larger on viscoelastic substrates, whereas the opposite effect occurs with PHH cells. The viscoelasticity of the microenvironment also promotes motility and multiple long protrusions in Huh7 cells. Pharmacologic disruption of the actin assembly makes cells unable to spread on either elastic or viscoelastic substrates. In contrast, upon vimentin perturbation, cells still spread to a limited degree on elastic substrates but are unable to spread on viscoelastic substrates. The time evolution of cell traction force shows that the peak occurs at an earlier time point on viscoelastic substrates compared to elastic substrates. However, the total force generation at steady state is the same on both substrates after 4 hours. Our data suggest that stress relaxation time scales of the viscoelastic substrate regulate cell dynamics and traction force generation, indicating different binding-unbinding rates of the proteins that form cell attachment sites in HCC cells and normal hepatocytes. These results suggest that liver cancer cells may have different characteristic lifetimes of binding to the substrate in comparision to normal cells, which might cause differences in cell spreading and motility within the diseased tissue.
Collapse
Affiliation(s)
- Kalpana Mandal
- Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia 19104, USA.
| | | | | | | | | |
Collapse
|
48
|
Patteson AE, Pogoda K, Byfield FJ, Mandal K, Ostrowska-Podhorodecka Z, Charrier EE, Galie PA, Deptuła P, Bucki R, McCulloch CA, Janmey PA. Loss of Vimentin Enhances Cell Motility through Small Confining Spaces. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2019; 15:e1903180. [PMID: 31721440 PMCID: PMC6910987 DOI: 10.1002/smll.201903180] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 09/22/2019] [Indexed: 05/28/2023]
Abstract
The migration of cells through constricting spaces or along fibrous tracks in tissues is important for many biological processes and depends on the mechanical properties of a cytoskeleton made up of three different filaments: F-actin, microtubules, and intermediate filaments. The signaling pathways and cytoskeletal structures that control cell motility on 2D are often very different from those that control motility in 3D. Previous studies have shown that intermediate filaments can promote actin-driven protrusions at the cell edge, but have little effect on overall motility of cells on flat surfaces. They are however important for cells to maintain resistance to repeated compressive stresses that are expected to occur in vivo. Using mouse embryonic fibroblasts derived from wild-type and vimentin-null mice, it is found that loss of vimentin increases motility in 3D microchannels even though on flat surfaces it has the opposite effect. Atomic force microscopy and traction force microscopy experiments reveal that vimentin enhances perinuclear cell stiffness while maintaining the same level of acto-myosin contractility in cells. A minimal model in which a perinuclear vimentin cage constricts along with the nucleus during motility through confining spaces, providing mechanical resistance against large strains that could damage the structural integrity of cells, is proposed.
Collapse
Affiliation(s)
- Alison E. Patteson
- Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, PA 19104
- Physics Department, Syracuse University, Syracuse, NY 13244
| | - Katarzyna Pogoda
- Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, PA 19104
- Institute of Nuclear Physics, Polish Academy of Sciences, PL-31342 Krakow, Poland
| | - Fitzroy J. Byfield
- Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, PA 19104
| | - Kalpana Mandal
- Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, PA 19104
| | | | - Elisabeth E. Charrier
- Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, PA 19104
| | - Peter A. Galie
- Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, PA 19104
- Department of Biomedical Engineering, Rowan University, Glassboro, NJ 08028
| | - Piotr Deptuła
- Department of Microbiological and Nanobiomedical Engineering, Medical University of Białystok, Mickiewicza 2C, Białystok, Poland
| | - Robert Bucki
- Department of Microbiological and Nanobiomedical Engineering, Medical University of Białystok, Mickiewicza 2C, Białystok, Poland
| | | | - Paul A. Janmey
- Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, PA 19104
- Departments of Physiology and Physics & Astronomy, University of Pennsylvania, Philadelphia, PA 19104
| |
Collapse
|
49
|
Abstract
Physical stimuli are essential for the function of eukaryotic cells, and changes in physical signals are important elements in normal tissue development as well as in disease initiation and progression. The complexity of physical stimuli and the cellular signals they initiate are as complex as those triggered by chemical signals. One of the most important, and the focus of this review, is the effect of substrate mechanical properties on cell structure and function. The past decade has produced a nearly exponentially increasing number of mechanobiological studies to define how substrate stiffness alters cell biology using both purified systems and intact tissues. Here we attempt to identify common features of mechanosensing in different systems while also highlighting the numerous informative exceptions to what in early studies appeared to be simple rules by which cells respond to mechanical stresses.
Collapse
Affiliation(s)
- Paul A Janmey
- Department of Physiology, Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Bioengineering, University of California-Berkeley, Berkeley, California; and Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| | - Daniel A Fletcher
- Department of Physiology, Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Bioengineering, University of California-Berkeley, Berkeley, California; and Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| | - Cynthia A Reinhart-King
- Department of Physiology, Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Bioengineering, University of California-Berkeley, Berkeley, California; and Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| |
Collapse
|
50
|
Mandal K, Pogoda K, Nandi S, Mathieu S, Kasri A, Klein E, Radvanyi F, Goud B, Janmey PA, Manneville JB. Role of a Kinesin Motor in Cancer Cell Mechanics. NANO LETTERS 2019; 19:7691-7702. [PMID: 31565944 PMCID: PMC7737127 DOI: 10.1021/acs.nanolett.9b02592] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
Molecular motors play important roles in force generation, migration, and intracellular trafficking. Changes in specific motor activities are altered in numerous diseases. KIF20A, a motor protein of the kinesin-6 family, is overexpressed in bladder cancer, and KIF20A levels correlate negatively with clinical outcomes. We report here a new role for the KIF20A kinesin motor protein in intracellular mechanics. Using optical tweezers to probe intracellular mechanics and surface AFM to probe cortical mechanics, we first confirm that bladder urothelial cells soften with an increasing cancer grade. We then show that inhibiting KIF20A makes the intracellular environment softer for both high- and low-grade bladder cancer cells. Upon inhibition of KIF20A, cortical stiffness also decreases in lower grade cells, while it surprisingly increases in higher grade malignant cells. Changes in cortical stiffness correlate with the interaction of KIF20A with myosin IIA. Moreover, KIF20A inhibition negatively regulates bladder cancer cell motility irrespective of the underlying substrate stiffness. Our results reveal a central role for a microtubule motor in cell mechanics and migration in the context of bladder cancer.
Collapse
Affiliation(s)
- Kalpana Mandal
- Institute for Medicine and Engineering , University of Pennsylvania , Philadelphia , Pennsylvania 19104 , United States
| | - Katarzyna Pogoda
- Institute for Medicine and Engineering , University of Pennsylvania , Philadelphia , Pennsylvania 19104 , United States
- Institute of Nuclear Physics , Polish Academy of Sciences , PL-31342 Krakow 31-342 , Poland
| | - Satabdi Nandi
- School of Veterinary Medicine , University of Pennsylvania , Philadelphia , Pennsylvania 19104 , United States
- Laboratory of Molecular Biology and Immunology , National Institute on Aging , Baltimore , Maryland 21224 , United States
| | - Samuel Mathieu
- Institut Curie, PSL Research University, CNRS, UMR 144 , 26 rue d'Ulm , Paris Cedex 05 75248 , France
| | - Amal Kasri
- Institut Curie, PSL Research University, CNRS, UMR 144 , 26 rue d'Ulm , Paris Cedex 05 75248 , France
- ICM Brain and Spine Institute , Pitié Salpêtrière Hospital , 47-83 Boulevard de l'Hôpital , Paris 75013 , France
| | - Eric Klein
- Department of Biology , Rutgers University-Camden Waterfront Tech Center , Camden , New Jersey 08103 , United States
| | - François Radvanyi
- Institut Curie, PSL Research University, CNRS, UMR 144 , 26 rue d'Ulm , Paris Cedex 05 75248 , France
| | - Bruno Goud
- Institut Curie, PSL Research University, CNRS, UMR 144 , 26 rue d'Ulm , Paris Cedex 05 75248 , France
| | - Paul A Janmey
- Institute for Medicine and Engineering , University of Pennsylvania , Philadelphia , Pennsylvania 19104 , United States
- Departments of Physiology and Physics & Astronomy , University of Pennsylvania , Philadelphia , Pennsylvania 19104 , United States
| | - Jean-Baptiste Manneville
- Institut Curie, PSL Research University, CNRS, UMR 144 , 26 rue d'Ulm , Paris Cedex 05 75248 , France
| |
Collapse
|