1
|
Zhu S, Liao X, Xu Y, Zhou N, Pan Y, Song J, Zheng T, Zhang L, Bai L, Wang Y, Zhou X, Gou M, Tao J, Liu R. 3D bioprinting of high-performance hydrogel with in-situ birth of stem cell spheroids. Bioact Mater 2025; 43:392-405. [PMID: 39399841 PMCID: PMC11470575 DOI: 10.1016/j.bioactmat.2024.09.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 09/11/2024] [Accepted: 09/23/2024] [Indexed: 10/15/2024] Open
Abstract
Digital light processing (DLP)-based bioprinting technology holds immense promise for the advancement of hydrogel constructs in biomedical applications. However, creating high-performance hydrogel constructs with this method is still a challenge, as it requires balancing the physicochemical properties of the matrix while also retaining the cellular activity of the encapsulated cells. Herein, we propose a facile and practical strategy for the 3D bioprinting of high-performance hydrogel constructs through the in-situ birth of stem cell spheroids. The strategy is achieved by loading the cell/dextran microdroplets within gelatin methacryloyl (GelMA) emulsion, where dextran functions as a decoy to capture and aggregate the cells for bioprinting while GelMA enables the mechanical support without losing the structural complexity and fidelity. Post-bioprinting, the leaching of dextran results in a smooth curved surface that promotes in-situ birth of spheroids within hydrogel constructs. This process significant enhances differentiation potential of encapsulated stem cells. As a proof-of-concept, we encapsulate dental pulp stem cells (DPSCs) within hydrogel constructs, showcasing their regenerative capabilities in dentin and neovascular-like structures in vivo. The strategy in our study enables high-performance hydrogel tissue construct fabrication with DLP-based bioprinting, which is anticipated to pave a promising way for diverse biomedical applications.
Collapse
Affiliation(s)
- Shunyao Zhu
- Department of Stomatology, Daping Hospital, Army Medical University (The Third Military Medical University), Chongqing, 400042, China
| | - Xueyuan Liao
- Department of Stomatology, Daping Hospital, Army Medical University (The Third Military Medical University), Chongqing, 400042, China
| | - Yue Xu
- Department of Stomatology, Daping Hospital, Army Medical University (The Third Military Medical University), Chongqing, 400042, China
| | - Nazi Zhou
- Department of Stomatology, Daping Hospital, Army Medical University (The Third Military Medical University), Chongqing, 400042, China
| | - Yingzi Pan
- Department of Stomatology, Daping Hospital, Army Medical University (The Third Military Medical University), Chongqing, 400042, China
| | - Jinlin Song
- College of Stomatology, Chongqing Medical University, Chongqing, China
| | - Taijing Zheng
- Department of Stomatology, Daping Hospital, Army Medical University (The Third Military Medical University), Chongqing, 400042, China
| | - Lin Zhang
- Department of Stomatology, Daping Hospital, Army Medical University (The Third Military Medical University), Chongqing, 400042, China
| | - Liyun Bai
- Department of Stomatology, Daping Hospital, Army Medical University (The Third Military Medical University), Chongqing, 400042, China
| | - Yu Wang
- Department of Stomatology, Daping Hospital, Army Medical University (The Third Military Medical University), Chongqing, 400042, China
| | - Xia Zhou
- Department of Stomatology, Daping Hospital, Army Medical University (The Third Military Medical University), Chongqing, 400042, China
- State Key Laboratory of Trauma, Burn and Combined Injury, Third Military Medical University, Chongqing, 400042, China
| | - Maling Gou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, 610065, China
| | - Jie Tao
- Department of Stomatology, Daping Hospital, Army Medical University (The Third Military Medical University), Chongqing, 400042, China
| | - Rui Liu
- Department of Stomatology, Daping Hospital, Army Medical University (The Third Military Medical University), Chongqing, 400042, China
| |
Collapse
|
2
|
Baig MA, Du Y, Zan Z, Fan Z. Influence of cell shape on sonoporation efficiency in microbubble-facilitated delivery using micropatterned cell arrays. Sci Rep 2024; 14:30845. [PMID: 39730459 PMCID: PMC11680583 DOI: 10.1038/s41598-024-81410-1] [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: 09/04/2024] [Accepted: 11/26/2024] [Indexed: 12/29/2024] Open
Abstract
Microbubble-facilitated sonoporation is a rapid, versatile, and non-viral intracellular delivery technique with potential for clinical and ex vivo cell engineering applications. We developed a micropatterning-based approach to investigate the impact of cell shape on sonoporation efficacy. Cationic microbubbles were employed to enhance sonoporation by binding to the cell membrane electrostatically. NIH/3T3 fibroblasts were micropatterned into circle, square, triangle, and rectangle. A two-plate system ensured high-throughput and efficient sonoporation by controlling cationic microbubble-cell attachment. High-speed video microscopy captured the acoustic dynamics of microbubbles under short ultrasound pulses. Our findings reveal that for NIH/3T3 fibroblasts, rectangular cells achieved the highest sonoporation and survival rate, while square-shaped cells demonstrated the greatest propidium iodide uptake. Triangle-shaped NIH/3T3 fibroblasts displayed an initial rise then a plateau in the sonoporation and survival rate as the ultrasound pulse duration increased from 10 cycles to 100 cycles, and then to 200 cycles. Conversely, rectangle-shaped cells showed a decrease followed by a stabilization. Circle-shaped and rectangle-shaped HeLa cells exhibited similar sonoporation outcomes, which were not as effective as NIH/3T3 fibroblasts. This study underscores the significance of cell shape in optimizing sonoporation efficiency and highlights the potential of combining micropatterning with controlled targeting sonoporation to advance intracellular delivery technologies.
Collapse
Affiliation(s)
- Mirza Albash Baig
- State Key Laboratory of Precision Measurement Technology and Instruments, Tianjin University, Tianjin, 300072, China
| | - Yanyao Du
- State Key Laboratory of Precision Measurement Technology and Instruments, Tianjin University, Tianjin, 300072, China
| | - Zhaoguang Zan
- State Key Laboratory of Precision Measurement Technology and Instruments, Tianjin University, Tianjin, 300072, China
| | - Zhenzhen Fan
- State Key Laboratory of Precision Measurement Technology and Instruments, Tianjin University, Tianjin, 300072, China.
- State Key Laboratory of Acoustics, Institute of Acoustics, Chinese Academy of Sciences, Beijing, 100190, China.
| |
Collapse
|
3
|
Kao CY, Ho CT, Huang TH, Kao CT. The Effects of Photobiomodulation Therapy on Tensile-Cultured Cementoblasts Cells. Photobiomodul Photomed Laser Surg 2024; 42:754-759. [PMID: 39330936 DOI: 10.1089/photob.2024.0076] [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] [Indexed: 09/28/2024] Open
Abstract
Background: Studies show that photobiomodulation therapy (PBMT) boosts cellular ATP production and cell growth and reduces inflammation. Additionally, mechanical tension affects gene expression, impacting cellular functions like proliferation and migration. Objective: We investigated the impact of PBMT on OCCM-30 cementoblast cells under tensile stress, focusing on cell survival, differentiation, and inflammatory responses, particularly relating to orthodontic tooth movement and root resorption. Methods: Cultured OCCM-30 cells under negative pressure received PBMT with a 10.6 μm wavelength in continuous mode at 1.0 W power for 3, 5, or 10 sec, corresponding to energy densities of 3, 5, or 10 J/cm2. We assessed cell viability with the Presto Blue assay and inflammatory markers Interleukin 6 (IL-6), inducible nitric oxide synthase (iNOS), and cyclooxygenase-2 (COX-2) through western blots at 1, 12, 24 h, and 7 days post-irradiation. Results: PBMT improved cell viability over time while maintaining levels of inflammatory markers. alkaline phosphatase levels dropped initially but increased after 7 days, suggesting enhanced cementoblast differentiation. IL-6 levels rose gradually, with 3J and 5J treatments showing significantly higher levels than the control. iNOS levels spiked within the first 24 h, then declined by day 7. COX-2 levels consistently rose, with the 5J treatment showing greater increases. Conclusions: PBMT appears to support cementoblast survival and differentiation while managing inflammation, potentially aiding root repair during orthodontic treatments and reducing inflammatory root resorption.
Collapse
Affiliation(s)
- Chuan-Yi Kao
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Psychiatry, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Chun-Te Ho
- School of Dentistry, Chung Shan Medical University, Taichung, Taiwan
- Department of Dentistry, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Tsui-Hsein Huang
- School of Dentistry, Chung Shan Medical University, Taichung, Taiwan
- Department of Dentistry, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Chia-Tze Kao
- School of Dentistry, Chung Shan Medical University, Taichung, Taiwan
- Department of Dentistry, Chung Shan Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
4
|
Sun P, Kraus CN, Zhao W, Xu J, Suh S, Nguyen Q, Jia Y, Nair A, Oakes M, Tinoco R, Shiu J, Sun B, Elsensohn A, Atwood SX, Nie Q, Dai X. Single-cell and spatial transcriptomics of vulvar lichen sclerosus reveal multi-compartmental alterations in gene expression and signaling cross-talk. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.14.607986. [PMID: 39211101 PMCID: PMC11361165 DOI: 10.1101/2024.08.14.607986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Vulvar diseases are a critical yet often neglected area of women's health, profoundly affecting patients' quality of life and frequently resulting in long-term physical and psychological challenges. Lichen sclerosus (LS) is a chronic inflammatory skin disorder that predominantly affects the vulva, leading to severe itching, pain, scarring, and an increased risk of malignancy. Despite its profound impact on affected individuals, the molecular pathogenesis of vulvar LS (VLS) is not well understood, hindering the development of FDA-approved therapies. Here, we utilize single-cell and spatial transcriptomics to analyze lesional and non-lesional skin from VLS patients, as well as healthy control vulvar skin. Our findings demonstrate histologic, cellular, and molecular heterogeneities within VLS, yet highlight unifying molecular changes across keratinocytes, fibroblasts, immune cells, and melanocytes in lesional skin. They reveal cellular stress and damage in fibroblasts and keratinocytes, enhanced T cell activation and cytotoxicity, aberrant cell-cell signaling, and increased activation of the IFN, JAK/STAT, and p53 pathways in specific cell types. Using both monolayer and organotypic culture models, we also demonstrate that knockdown of select genes, which are downregulated in VLS lesional keratinocytes, partially recapitulates VLS-like stress-associated changes. Collectively, these data provide novel insights into the pathogenesis of VLS, identifying potential biomarkers and therapeutic targets for future research.
Collapse
|
5
|
Bhattacharya S, Ettela A, Haydak J, Hobson CM, Stern A, Yoo M, Chew TL, Gusella GL, Gallagher EJ, Hone JC, Azeloglu EU. A high-throughput microfabricated platform for rapid quantification of metastatic potential. SCIENCE ADVANCES 2024; 10:eadk0015. [PMID: 39151003 PMCID: PMC11328906 DOI: 10.1126/sciadv.adk0015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 07/12/2024] [Indexed: 08/18/2024]
Abstract
Assays that measure morphology, proliferation, motility, deformability, and migration are used to study the invasiveness of cancer cells. However, native invasive potential of cells may be hidden from these contextual metrics because they depend on culture conditions. We created a micropatterned chip that mimics the native environmental conditions, quantifies the invasive potential of tumor cells, and improves our understanding of the malignancy signatures. Unlike conventional assays, which rely on indirect measurements of metastatic potential, our method uses three-dimensional microchannels to measure the basal native invasiveness without chemoattractants or microfluidics. No change in cell death or proliferation is observed on our chips. Using six cancer cell lines, we show that our system is more sensitive than other motility-based assays, measures of nuclear deformability, or cell morphometrics. In addition to quantifying metastatic potential, our platform can distinguish between motility and invasiveness, help study molecular mechanisms of invasion, and screen for targeted therapeutics.
Collapse
Affiliation(s)
- Smiti Bhattacharya
- Barbara T. Murphy Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Mechanical Engineering, Columbia University, New York, NY, USA
| | - Abora Ettela
- Division of Endocrinology, Diabetes and Bone Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jonathan Haydak
- Barbara T. Murphy Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Chad M. Hobson
- Advanced Imaging Center, Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Alan Stern
- Barbara T. Murphy Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Miran Yoo
- Barbara T. Murphy Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Teng-Leong Chew
- Advanced Imaging Center, Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - G. Luca Gusella
- Barbara T. Murphy Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Emily J. Gallagher
- Division of Endocrinology, Diabetes and Bone Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - James C. Hone
- Department of Mechanical Engineering, Columbia University, New York, NY, USA
| | - Evren U. Azeloglu
- Barbara T. Murphy Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
6
|
Han Y, Shao Z, Zhang Y, Zhao H, Sun Z, Yang C, Tang H, Han Y, Gao C. 3D matrix stiffness modulation unveils cardiac fibroblast phenotypic switching. Sci Rep 2024; 14:17015. [PMID: 39043765 PMCID: PMC11266583 DOI: 10.1038/s41598-024-67646-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 07/15/2024] [Indexed: 07/25/2024] Open
Abstract
This study investigates how dynamic fluctuations in matrix stiffness affect the behavior of cardiac fibroblasts (CFs) within a three-dimensional (3D) hydrogel environment. Using hybrid hydrogels with tunable stiffness, we created an in vitro model to mimic the varying stiffness of the cardiac microenvironment. By manipulating hydrogel stiffness, we examined CF responses, particularly the expression of α-smooth muscle actin (α-SMA), a marker of myofibroblast differentiation. Our findings reveal that increased matrix stiffness promotes the differentiation of CFs into myofibroblasts, while matrix softening reverses this process. Additionally, we identified the role of focal adhesions and integrin β1 in mediating stiffness-induced phenotypic switching. This study provides significant insights into the mechanobiology of cardiac fibrosis and suggests that modulating matrix stiffness could be a potential therapeutic strategy for treating cardiovascular diseases.
Collapse
Affiliation(s)
- Yan Han
- Department of Structural Heart Disease, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, 451464, Henan, China
| | - Zehua Shao
- Department of Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100871, China
| | - Yuanhao Zhang
- Department of Structural Heart Disease, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, 451464, Henan, China
| | - Huan Zhao
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Zirui Sun
- Department of Structural Heart Disease, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, 451464, Henan, China
| | - Chaokuan Yang
- Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, 451464, Henan, China
| | - Hao Tang
- Zhengzhou Key Laboratory of Cardiovascular Aging, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, 451464, Henan, China.
- National Health Commission Key Laboratory of Cardiovascular Regenerative Medicine, Central China Branch of National Center for Cardiovascular Diseases, Zhengzhou, 451464, Henan, China.
| | - Yu Han
- Department of Structural Heart Disease, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, 451464, Henan, China.
| | - Chuanyu Gao
- Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, 451464, Henan, China.
| |
Collapse
|
7
|
Haydak J, Azeloglu EU. Role of biophysics and mechanobiology in podocyte physiology. Nat Rev Nephrol 2024; 20:371-385. [PMID: 38443711 PMCID: PMC12103212 DOI: 10.1038/s41581-024-00815-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/30/2024] [Indexed: 03/07/2024]
Abstract
Podocytes form the backbone of the glomerular filtration barrier and are exposed to various mechanical forces throughout the lifetime of an individual. The highly dynamic biomechanical environment of the glomerular capillaries greatly influences the cell biology of podocytes and their pathophysiology. Throughout the past two decades, a holistic picture of podocyte cell biology has emerged, highlighting mechanobiological signalling pathways, cytoskeletal dynamics and cellular adhesion as key determinants of biomechanical resilience in podocytes. This biomechanical resilience is essential for the physiological function of podocytes, including the formation and maintenance of the glomerular filtration barrier. Podocytes integrate diverse biomechanical stimuli from their environment and adapt their biophysical properties accordingly. However, perturbations in biomechanical cues or the underlying podocyte mechanobiology can lead to glomerular dysfunction with severe clinical consequences, including proteinuria and glomerulosclerosis. As our mechanistic understanding of podocyte mechanobiology and its role in the pathogenesis of glomerular disease increases, new targets for podocyte-specific therapeutics will emerge. Treating glomerular diseases by targeting podocyte mechanobiology might improve therapeutic precision and efficacy, with potential to reduce the burden of chronic kidney disease on individuals and health-care systems alike.
Collapse
Affiliation(s)
- Jonathan Haydak
- Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Evren U Azeloglu
- Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
8
|
Zhang Y, Remy M, Leste-Lasserre T, Durrieu MC. Manipulating Stem Cell Fate with Disordered Bioactive Cues on Surfaces: The Role of Bioactive Ligand Selection. ACS APPLIED MATERIALS & INTERFACES 2024; 16:18474-18489. [PMID: 38581548 DOI: 10.1021/acsami.4c00262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/08/2024]
Abstract
The development of 2D or 3D bioactive platforms for rapidly isolating pure populations of cells from adult stem cells holds promise for advancing the understanding of cellular mechanisms, drug testing, and tissue engineering. Over the years, methods have emerged to synthesize bioactive micro- and nanostructured 2D materials capable of directing stem cell fate. We introduce a novel method for randomly micro- or nanopatterning any protein/peptide onto both 2D and 3D scaffolds via spray technology. Our goal is to investigate the impact of arranging bioactive micropatterns (ordered vs disordered) on surfaces to guide human mesenchymal stem cell (hMSC) differentiation. The spray technology efficiently coats materials with controlled, cost-effective bioactive micropatterns in various sizes and shapes. BMP-2 mimetic peptides were covalently grafted, individually or in combination with RGD peptides, onto activated polyethylene terephthalate (PET) surfaces through a spraying process, incorporating nano/microscale parameters like size, shape, and composition. The study explores different peptide distributions on surfaces and various peptide combinations. Four surfaces were homogeneously functionalized with these peptides (M1 to M4 with various densities of peptides), and six surfaces with disordered micro- and nanopatterns of peptides (S0 to S5 with different sizes of peptide patterns) were synthesized. Fluorescence microscopy assessed peptide distribution, followed by hMSC culture for 2 weeks, and evaluated osteogenic differentiation via immunocytochemistry and RT-qPCR for osteoblast and osteocyte markers. Cells on uniformly peptide-functionalized surfaces exhibited cuboidal forms, while those on surfaces with disordered patterns tended toward columnar or cuboidal shapes. Surfaces S4 and S5 showed dendrite-like formations resembling an osteocyte morphology. S5 showed significant overexpression of osteoblast (OPN) and osteocyte markers (E11, DMP1, and SOST) compared to control surfaces and other micropatterned surfaces. Notably, despite sharing an equivalent quantity of peptides with a homogeneous functionalized surface, S5 displayed a distinct distribution of peptides, resulting in enhanced osteogenic differentiation of hMSCs.
Collapse
Affiliation(s)
- Yujie Zhang
- CNRS, Bordeaux INP, CBMN, Univ. Bordeaux, UMR 5248, Pessac33600,France
| | - Murielle Remy
- CNRS, Bordeaux INP, CBMN, Univ. Bordeaux, UMR 5248, Pessac33600,France
| | | | | |
Collapse
|
9
|
Sun F, Li H, Hu Y, Zhang M, Wang W, Chen W, Liu Z. Exploring Mechanical Responses of Cells to Geometric Information Using Micropatterned DNA-Based Molecular Tension Probes. ACS NANO 2023; 17:18584-18595. [PMID: 37713214 DOI: 10.1021/acsnano.3c07088] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/16/2023]
Abstract
The geometric shape of a cell is strongly influenced by the cytoskeleton, which, in turn, is regulated by integrin-mediated cell-extracellular matrix (ECM) interactions. To investigate the mechanical role of integrin in the geometrical interplay between cells and the ECM, we proposed a single-cell micropatterning technique combined with molecular tension fluorescence microscopy (MTFM), which allows us to characterize the mechanical properties of cells with prescribed geometries. Our results show that the curvature is a key geometric cue for cells to differentiate shapes in a membrane-tension- and actomyosin-dependent manner. Specifically, curvatures affect the size of focal adhesions (FAs) and induce a curvature-dependent density and spatial distribution of strong integrins. In addition, we found that the integrin subunit β1 plays a critical role in the detection of geometric information. Overall, the integration of MTFM and single-cell micropatterning offers a robust approach for investigating the nexus between mechanical cues and cellular responses, holding potential for advancing our understanding of mechanobiology.
Collapse
Affiliation(s)
- Feng Sun
- TaiKang Center for Life and Medical Sciences, the Institute for Advanced Studies, Wuhan University, Wuhan 430072, China
| | - Hongyun Li
- TaiKang Center for Life and Medical Sciences, the Institute for Advanced Studies, Wuhan University, Wuhan 430072, China
| | - Yuru Hu
- TaiKang Center for Life and Medical Sciences, the Institute for Advanced Studies, Wuhan University, Wuhan 430072, China
| | - Mengsheng Zhang
- TaiKang Center for Life and Medical Sciences, the Institute for Advanced Studies, Wuhan University, Wuhan 430072, China
| | - Wenxu Wang
- TaiKang Center for Life and Medical Sciences, the Institute for Advanced Studies, Wuhan University, Wuhan 430072, China
| | - Wei Chen
- TaiKang Center for Life and Medical Sciences, the Institute for Advanced Studies, Wuhan University, Wuhan 430072, China
| | - Zheng Liu
- TaiKang Center for Life and Medical Sciences, the Institute for Advanced Studies, Wuhan University, Wuhan 430072, China
| |
Collapse
|
10
|
Sucre JM, Bock F, Negretti NM, Benjamin JT, Gulleman PM, Dong X, Ferguson KT, Jetter CS, Han W, Liu Y, Kook S, Gokey JJ, Guttentag SH, Kropski JA, Blackwell TS, Zent R, Plosa EJ. Alveolar repair following LPS-induced injury requires cell-ECM interactions. JCI Insight 2023; 8:e167211. [PMID: 37279065 PMCID: PMC10443799 DOI: 10.1172/jci.insight.167211] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 05/31/2023] [Indexed: 06/07/2023] Open
Abstract
During alveolar repair, alveolar type 2 (AT2) epithelial cell progenitors rapidly proliferate and differentiate into flat AT1 epithelial cells. Failure of normal alveolar repair mechanisms can lead to loss of alveolar structure (emphysema) or development of fibrosis, depending on the type and severity of injury. To test if β1-containing integrins are required during repair following acute injury, we administered E. coli lipopolysaccharide (LPS) by intratracheal injection to mice with a postdevelopmental deletion of β1 integrin in AT2 cells. While control mice recovered from LPS injury without structural abnormalities, β1-deficient mice had more severe inflammation and developed emphysema. In addition, recovering alveoli were repopulated with an abundance of rounded epithelial cells coexpressing AT2 epithelial, AT1 epithelial, and mixed intermediate cell state markers, with few mature type 1 cells. AT2 cells deficient in β1 showed persistently increased proliferation after injury, which was blocked by inhibiting NF-κB activation in these cells. Lineage tracing experiments revealed that β1-deficient AT2 cells failed to differentiate into mature AT1 epithelial cells. Together, these findings demonstrate that functional alveolar repair after injury with terminal alveolar epithelial differentiation requires β1-containing integrins.
Collapse
Affiliation(s)
- Jennifer M.S. Sucre
- Department of Pediatrics, Division of Neonatology
- Department of Cell and Developmental Biology
| | - Fabian Bock
- Department of Medicine, Division of Nephrology and Hypertension; and
| | | | | | | | - Xinyu Dong
- Department of Medicine, Division of Nephrology and Hypertension; and
| | | | | | - Wei Han
- Department of Medicine, Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Yang Liu
- Department of Medicine, Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | | | - Jason J. Gokey
- Department of Medicine, Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | | | - Jonathan A. Kropski
- Department of Cell and Developmental Biology
- Department of Medicine, Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Nashville Veterans Affairs Medical Center, Nashville, Tennessee, USA
| | - Timothy S. Blackwell
- Department of Cell and Developmental Biology
- Department of Medicine, Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Nashville Veterans Affairs Medical Center, Nashville, Tennessee, USA
| | - Roy Zent
- Department of Cell and Developmental Biology
- Department of Medicine, Division of Nephrology and Hypertension; and
- Nashville Veterans Affairs Medical Center, Nashville, Tennessee, USA
| | | |
Collapse
|
11
|
Li J, Li X, Song S, Sun Z, Li Y, Yang L, Xie Z, Cai Y, Zhao Y. Mitochondria spatially and temporally modulate VSMC phenotypes via interacting with cytoskeleton in cardiovascular diseases. Redox Biol 2023; 64:102778. [PMID: 37321061 DOI: 10.1016/j.redox.2023.102778] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 05/31/2023] [Accepted: 06/06/2023] [Indexed: 06/17/2023] Open
Abstract
Cardiovascular diseases caused by atherosclerosis (AS) seriously endanger human health, which is closely related to vascular smooth muscle cell (VSMC) phenotypes. VSMC phenotypic transformation is marked by the alteration of phenotypic marker expression and cellular behaviour. Intriguingly, the mitochondrial metabolism and dynamics altered during VSMC phenotypic transformation. Firstly, this review combs VSMC mitochondrial metabolism in three aspects: mitochondrial ROS generation, mutated mitochondrial DNA (mtDNA) and calcium metabolism respectively. Secondly, we summarized the role of mitochondrial dynamics in regulating VSMC phenotypes. We further emphasized the association between mitochondria and cytoskelton via presenting cytoskeletal support during mitochondrial dynamics process, and discussed its impact on their respective dynamics. Finally, considering that both mitochondria and cytoskeleton are mechano-sensitive organelles, we demonstrated their direct and indirect interaction under extracellular mechanical stimuli through several mechano-sensitive signaling pathways. We additionally discussed related researches in other cell types in order to inspire deeper thinking and reasonable speculation of potential regulatory mechanism in VSMC phenotypic transformation.
Collapse
Affiliation(s)
- Jingwen Li
- Laboratory of Tissue and Cell Biology, Lab Teaching & Management Center, Chongqing Medical University, NO.1 Medical College Road, Yuzhong District, Chongqing, 400016, China
| | - Xinyue Li
- Laboratory of Tissue and Cell Biology, Lab Teaching & Management Center, Chongqing Medical University, NO.1 Medical College Road, Yuzhong District, Chongqing, 400016, China
| | - Sijie Song
- Laboratory of Tissue and Cell Biology, Lab Teaching & Management Center, Chongqing Medical University, NO.1 Medical College Road, Yuzhong District, Chongqing, 400016, China
| | - Zhengwen Sun
- Laboratory of Tissue and Cell Biology, Lab Teaching & Management Center, Chongqing Medical University, NO.1 Medical College Road, Yuzhong District, Chongqing, 400016, China
| | - Yuanzhu Li
- Laboratory of Tissue and Cell Biology, Lab Teaching & Management Center, Chongqing Medical University, NO.1 Medical College Road, Yuzhong District, Chongqing, 400016, China
| | - Long Yang
- Laboratory of Tissue and Cell Biology, Lab Teaching & Management Center, Chongqing Medical University, NO.1 Medical College Road, Yuzhong District, Chongqing, 400016, China
| | - Zhenhong Xie
- Laboratory of Tissue and Cell Biology, Lab Teaching & Management Center, Chongqing Medical University, NO.1 Medical College Road, Yuzhong District, Chongqing, 400016, China
| | - Yikui Cai
- Laboratory of Tissue and Cell Biology, Lab Teaching & Management Center, Chongqing Medical University, NO.1 Medical College Road, Yuzhong District, Chongqing, 400016, China
| | - Yinping Zhao
- Laboratory of Tissue and Cell Biology, Lab Teaching & Management Center, Chongqing Medical University, NO.1 Medical College Road, Yuzhong District, Chongqing, 400016, China.
| |
Collapse
|
12
|
Tang RZ, Liu XQ. Biophysical cues of in vitro biomaterials-based artificial extracellular matrix guide cancer cell plasticity. Mater Today Bio 2023; 19:100607. [PMID: 36960095 PMCID: PMC10027567 DOI: 10.1016/j.mtbio.2023.100607] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 02/10/2023] [Accepted: 03/07/2023] [Indexed: 03/13/2023] Open
Abstract
Clinical evidence supports a role for the extracellular matrix (ECM) in cancer plasticity across multiple tumor types. The lack of in vitro models that represent the native ECMs is a significant challenge for cancer research and drug discovery. Therefore, a major motivation for developing new tumor models is to create the artificial ECM in vitro. Engineered biomaterials can closely mimic the architectural and mechanical properties of ECM to investigate their specific effects on cancer progression, offering an alternative to animal models for the testing of cancer cell behaviors. In this review, we focused on the biomaterials from different sources applied in the fabrication of the artificial ECM and their biophysical cues to recapitulate key features of tumor niche. Furthermore, we summarized how the distinct biophysical cues guided cell behaviors of cancer plasticity, including morphology, epithelial-to-mesenchymal transition (EMT), enrichment of cancer stem cells (CSCs), proliferation, migration/invasion and drug resistance. We also discuss the future opportunities in using the artificial ECM for applications of tumorigenesis research and precision medicine, as well as provide useful messages of principles for designing suitable biomaterial scaffolds.
Collapse
Affiliation(s)
- Rui-Zhi Tang
- Key Laboratory for Molecular Diagnosis of Hubei Province, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430014, PR China
| | - Xi-Qiu Liu
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
| |
Collapse
|
13
|
Martins de Sousa K, Linklater DP, Murdoch BJ, Al Kobaisi M, Crawford RJ, Judge R, Dashper S, Sloan AJ, Losic D, Ivanova EP. Modulation of MG-63 Osteogenic Response on Mechano-Bactericidal Micronanostructured Titanium Surfaces. ACS APPLIED BIO MATERIALS 2023; 6:1054-1070. [PMID: 36880728 DOI: 10.1021/acsabm.2c00952] [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] [Indexed: 03/08/2023]
Abstract
Despite recent advances in the development of orthopedic devices, implant-related failures that occur as a result of poor osseointegration and nosocomial infection are frequent. In this study, we developed a multiscale titanium (Ti) surface topography that promotes both osteogenic and mechano-bactericidal activity using a simple two-step fabrication approach. The response of MG-63 osteoblast-like cells and antibacterial activity toward Pseudomonas aeruginosa and Staphylococcus aureus bacteria was compared for two distinct micronanoarchitectures of differing surface roughness created by acid etching, using either hydrochloric acid (HCl) or sulfuric acid (H2SO4), followed by hydrothermal treatment, henceforth referred to as either MN-HCl or MN-H2SO4. The MN-HCl surfaces were characterized by an average surface microroughness (Sa) of 0.8 ± 0.1 μm covered by blade-like nanosheets of 10 ± 2.1 nm thickness, whereas the MN-H2SO4 surfaces exhibited a greater Sa value of 5.8 ± 0.6 μm, with a network of nanosheets of 20 ± 2.6 nm thickness. Both micronanostructured surfaces promoted enhanced MG-63 attachment and differentiation; however, cell proliferation was only significantly increased on MN-HCl surfaces. In addition, the MN-HCl surface exhibited increased levels of bactericidal activity, with only 0.6% of the P. aeruginosa cells and approximately 5% S. aureus cells remaining viable after 24 h when compared to control surfaces. Thus, we propose the modulation of surface roughness and architecture on the micro- and nanoscale to achieve efficient manipulation of osteogenic cell response combined with mechanical antibacterial activity. The outcomes of this study provide significant insight into the further development of advanced multifunctional orthopedic implant surfaces.
Collapse
Affiliation(s)
| | - Denver P Linklater
- School of Science, STEM College, RMIT University, Melbourne, Victoria 3000, Australia
- Department of Biomedical Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Billy J Murdoch
- RMIT Microscopy and Microanalysis Facility, STEM College, RMIT University, Melbourne, Victoria 3000, Australia
| | - Mohammad Al Kobaisi
- School of Engineering, STEM College, RMIT University, Melbourne, Victoria 3000, Australia
| | - Russell J Crawford
- School of Science, STEM College, RMIT University, Melbourne, Victoria 3000, Australia
| | - Roy Judge
- Melbourne Dental School, Faculty of Medicine, Dentistry & Health Sciences, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Stuart Dashper
- Melbourne Dental School, Faculty of Medicine, Dentistry & Health Sciences, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Alastair J Sloan
- Melbourne Dental School, Faculty of Medicine, Dentistry & Health Sciences, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Dusan Losic
- School of Chemical Engineering and Advanced Materials, The University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Elena P Ivanova
- School of Science, STEM College, RMIT University, Melbourne, Victoria 3000, Australia
| |
Collapse
|
14
|
Li JH, Trivedi V, Diz-Muñoz A. Understanding the interplay of membrane trafficking, cell surface mechanics, and stem cell differentiation. Semin Cell Dev Biol 2023; 133:123-134. [PMID: 35641408 PMCID: PMC9703995 DOI: 10.1016/j.semcdb.2022.05.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 04/08/2022] [Accepted: 05/14/2022] [Indexed: 01/17/2023]
Abstract
Stem cells can generate a diversity of cell types during development, regeneration and adult tissue homeostasis. Differentiation changes not only the cell fate in terms of gene expression but also the physical properties and functions of cells, e.g. the secretory activity, cell shape, or mechanics. Conversely, these activities and properties can also regulate differentiation itself. Membrane trafficking is known to modulate signal transduction and thus has the potential to control stem cell differentiation. On the other hand, membrane trafficking, particularly from and to the plasma membrane, depends on the mechanical properties of the cell surface such as tension within the plasma membrane or the cortex. Indeed, recent findings demonstrate that cell surface mechanics can also control cell fate. Here, we review the bidirectional relationships between these three fundamental cellular functions, i.e. membrane trafficking, cell surface mechanics, and stem cell differentiation. Furthermore, we discuss commonly used methods in each field and how combining them with new tools will enhance our understanding of their interplay. Understanding how membrane trafficking and cell surface mechanics can guide stem cell fate holds great potential as these concepts could be exploited for directed differentiation of stem cells for the fields of tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Jia Hui Li
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory (EMBL), Meyerhofstraße 1, Heidelberg 69117, Germany
| | - Vikas Trivedi
- EMBL, PRBB, Dr. Aiguader, 88, Barcelona 08003, Spain,Developmental Biology Unit, EMBL, Meyerhofstraße 1, Heidelberg 69117, Germany
| | - Alba Diz-Muñoz
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory (EMBL), Meyerhofstraße 1, Heidelberg 69117, Germany.
| |
Collapse
|
15
|
Stern AD, Smith GR, Santos LC, Sarmah D, Zhang X, Lu X, Iuricich F, Pandey G, Iyengar R, Birtwistle MR. Relating individual cell division events to single-cell ERK and Akt activity time courses. Sci Rep 2022; 12:18077. [PMID: 36302844 PMCID: PMC9613772 DOI: 10.1038/s41598-022-23071-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 10/25/2022] [Indexed: 02/01/2023] Open
Abstract
Biochemical correlates of stochastic single-cell fates have been elusive, even for the well-studied mammalian cell cycle. We monitored single-cell dynamics of the ERK and Akt pathways, critical cell cycle progression hubs and anti-cancer drug targets, and paired them to division events in the same single cells using the non-transformed MCF10A epithelial line. Following growth factor treatment, in cells that divide both ERK and Akt activities are significantly higher within the S-G2 time window (~ 8.5-40 h). Such differences were much smaller in the pre-S-phase, restriction point window which is traditionally associated with ERK and Akt activity dependence, suggesting unappreciated roles for ERK and Akt in S through G2. Simple metrics of central tendency in this time window are associated with subsequent cell division fates. ERK activity was more strongly associated with division fates than Akt activity, suggesting Akt activity dynamics may contribute less to the decision driving cell division in this context. We also find that ERK and Akt activities are less correlated with each other in cells that divide. Network reconstruction experiments demonstrated that this correlation behavior was likely not due to crosstalk, as ERK and Akt do not interact in this context, in contrast to other transformed cell types. Overall, our findings support roles for ERK and Akt activity throughout the cell cycle as opposed to just before the restriction point, and suggest ERK activity dynamics may be more important than Akt activity dynamics for driving cell division in this non-transformed context.
Collapse
Affiliation(s)
- Alan D Stern
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Gregory R Smith
- Department of Neurology, Center for Advanced Research on Diagnostic Assays, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Luis C Santos
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Deepraj Sarmah
- Department of Chemical and Biomolecular Engineering, Clemson University, Clemson, SC, USA
| | - Xiang Zhang
- School of Computing, Clemson University, Clemson, SC, USA
| | - Xiaoming Lu
- Department of Chemical and Biomolecular Engineering, Clemson University, Clemson, SC, USA
| | | | - Gaurav Pandey
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ravi Iyengar
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Marc R Birtwistle
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Chemical and Biomolecular Engineering, Clemson University, Clemson, SC, USA.
| |
Collapse
|
16
|
Ganjian M, Modaresifar K, Rompolas D, Fratila-Apachitei LE, Zadpoor AA. Nanoimprinting for high-throughput replication of geometrically precise pillars in fused silica to regulate cell behavior. Acta Biomater 2022; 140:717-729. [PMID: 34875357 DOI: 10.1016/j.actbio.2021.12.001] [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: 08/30/2021] [Revised: 11/04/2021] [Accepted: 12/01/2021] [Indexed: 12/31/2022]
Abstract
Developing high-throughput nanopatterning techniques that also allow for precise control over the dimensions of the fabricated features is essential for the study of cell-nanopattern interactions. Here, we developed a process that fulfills both of these criteria. Firstly, we used electron-beam lithography (EBL) to fabricate precisely controlled arrays of submicron pillars with varying values of interspacing on a large area of fused silica. Two types of etching procedures with two different systems were developed to etch the fused silica and create the final desired height. We then studied the interactions of preosteoblasts (MC3T3-E1) with these pillars. Varying interspacing was observed to significantly affect the morphological characteristics of the cell, the organization of actin fibers, and the formation of focal adhesions. The expression of osteopontin (OPN) significantly increased on the patterns, indicating the potential of the pillars for inducing osteogenic differentiation. The EBL pillars were thereafter used as master molds in two subsequent processing steps, namely soft lithography and thermal nanoimprint lithography for high-fidelity replication of the pillars on the substrates of interest. The molding parameters were optimized to maximize the fidelity of the generated patterns and minimize the wear and tear of the master mold. Comparing the replicated feature with those present on the original mold confirmed that the geometry and dimensions of the replicated pillars closely resemble those of the original ones. The method proposed in this study, therefore, enables the precise fabrication of submicron- and nanopatterns on a wide variety of materials that are relevant for systematic cell studies. STATEMENT OF SIGNIFICANCE: Submicron pillars with specific dimensions on the bone implants have been proven to be effective in controlling cell behaviors. Nowadays, numerous methods have been proposed to produce bio-instructive submicron-topographies. However, most of these techniques are suffering from being low-throughput, low-precision, and expensive. Here, we developed a high-throughput nanopatterning technique that allows for control over the dimensions of the features for the study of cell-nanotopography interactions. Assessing the adaptation of preosteoblast cells showed the potential of the pillars for inducing osteogenic differentiation. Afterward, the pillars were used for high-fidelity replication of the bio-instructive features on the substrates of interest. The results show the advantages of nanoimprint lithography as a unique technique for the patterning of large areas of bio-instructive surfaces.
Collapse
|
17
|
Vermeulen S, Birgani ZT, Habibovic P. Biomaterial-induced pathway modulation for bone regeneration. Biomaterials 2022; 283:121431. [DOI: 10.1016/j.biomaterials.2022.121431] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 01/28/2022] [Accepted: 02/17/2022] [Indexed: 12/18/2022]
|
18
|
Haroon M, Boers HE, Bakker AD, Bloks NGC, Hoogaars WMH, Giordani L, Musters RJP, Deldicque L, Koppo K, Le Grand F, Klein-Nulend J, Jaspers RT. Reduced growth rate of aged muscle stem cells is associated with impaired mechanosensitivity. Aging (Albany NY) 2022; 14:28-53. [PMID: 35023852 PMCID: PMC8791224 DOI: 10.18632/aging.203830] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 01/03/2022] [Indexed: 11/25/2022]
Abstract
Aging-associated muscle wasting and impaired regeneration are caused by deficiencies in muscle stem cell (MuSC) number and function. We postulated that aged MuSCs are intrinsically impaired in their responsiveness to omnipresent mechanical cues through alterations in MuSC morphology, mechanical properties, and number of integrins, culminating in impaired proliferative capacity. Here we show that aged MuSCs exhibited significantly lower growth rate and reduced integrin-α7 expression as well as lower number of phospho-paxillin clusters than young MuSCs. Moreover, aged MuSCs were less firmly attached to matrigel-coated glass substrates compared to young MuSCs, as 43% of the cells detached in response to pulsating fluid shear stress (1 Pa). YAP nuclear localization was 59% higher than in young MuSCs, yet YAP target genes Cyr61 and Ctgf were substantially downregulated. When subjected to pulsating fluid shear stress, aged MuSCs exhibited reduced upregulation of proliferation-related genes. Together these results indicate that aged MuSCs exhibit impaired mechanosensitivity and growth potential, accompanied by altered morphology and mechanical properties as well as reduced integrin-α7 expression. Aging-associated impaired muscle regenerative capacity and muscle wasting is likely due to aging-induced intrinsic MuSC alterations and dysfunctional mechanosensitivity.
Collapse
Affiliation(s)
- Mohammad Haroon
- Laboratory for Myology, Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam 1081 HZ, The Netherlands
| | - Heleen E Boers
- Laboratory for Myology, Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam 1081 HZ, The Netherlands
| | - Astrid D Bakker
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam, University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam 1081 LA, The Netherlands
| | - Niek G C Bloks
- Laboratory for Myology, Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam 1081 HZ, The Netherlands
| | - Willem M H Hoogaars
- Laboratory for Myology, Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam 1081 HZ, The Netherlands
| | - Lorenzo Giordani
- Sorbonne Université, INSERM UMRS974, Center for Research in Myology, Paris 75013, France
| | - René J P Musters
- Department of Physiology, Amsterdam University Medical Center VUmc, Amsterdam Cardiovascular Sciences, Amsterdam 1081 HZ, The Netherlands
| | - Louise Deldicque
- Institute of Neuroscience, Université Catholique de Louvain, Louvain-la-Neuve 1348, Belgium
| | - Katrien Koppo
- Exercise Physiology Research Group, Department of Movement Sciences, KU Leuven, Leuven 3001, Belgium
| | - Fabien Le Grand
- Faculty of Medicine and Pharmacy, NeuroMyoGène UCBL-CNRS UMR 5310, INSERM U1217, Lyon 69008, France
| | - Jenneke Klein-Nulend
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam, University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam 1081 LA, The Netherlands
| | - Richard T Jaspers
- Laboratory for Myology, Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam 1081 HZ, The Netherlands
| |
Collapse
|
19
|
Dieffenbach PB, Mallarino Haeger C, Rehman R, Corcoran AM, Coronata AMF, Vellarikkal SK, Chrobak I, Waxman AB, Vitali SH, Sholl LM, Padera RF, Lagares D, Polverino F, Owen CA, Fredenburgh LE. A Novel Protective Role for Matrix Metalloproteinase-8 in the Pulmonary Vasculature. Am J Respir Crit Care Med 2021; 204:1433-1451. [PMID: 34550870 PMCID: PMC8865706 DOI: 10.1164/rccm.202108-1863oc] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 09/22/2021] [Indexed: 11/16/2022] Open
Abstract
Rationale: Mechanical signaling through cell-matrix interactions plays a major role in progressive vascular remodeling in pulmonary arterial hypertension (PAH). MMP-8 (matrix metalloproteinase-8) is an interstitial collagenase involved in regulating inflammation and fibrosis of the lung and systemic vasculature, but its role in PAH pathogenesis remains unexplored. Objectives: To evaluate MMP-8 as a modulator of pathogenic mechanical signaling in PAH. Methods: MMP-8 levels were measured in plasma from patients with pulmonary hypertension (PH) and controls by ELISA. MMP-8 vascular expression was examined in lung tissue from patients with PAH and rodent models of PH. MMP-8-/- and MMP-8+/+ mice were exposed to normobaric hypoxia or normoxia for 4-8 weeks. PH severity was evaluated by right ventricular systolic pressure, echocardiography, pulmonary artery morphometry, and immunostaining. Proliferation, migration, matrix component expression, and mechanical signaling were assessed in MMP-8-/- and MMP-8+/+ pulmonary artery smooth muscle cells (PASMCs). Measurements and Main Results: MMP-8 expression was significantly increased in plasma and pulmonary arteries of patients with PH compared with controls and induced in the pulmonary vasculature in rodent PH models. Hypoxia-exposed MMP-8-/- mice had significant mortality, increased right ventricular systolic pressure, severe right ventricular dysfunction, and exaggerated vascular remodeling compared with MMP-8+/+ mice. MMP-8-/- PASMCs demonstrated exaggerated proliferation and migration mediated by altered matrix protein expression, elevated integrin-β3 levels, and induction of FAK (focal adhesion kinase) and downstream YAP (Yes-associated protein)/TAZ (transcriptional coactivator with PDZ-binding motif) activity. Conclusions: MMP-8 is a novel protective factor upregulated in the pulmonary vasculature during PAH pathogenesis. MMP-8 opposes pathologic mechanobiological feedback by altering matrix composition and disrupting integrin-β3/FAK and YAP/TAZ-dependent mechanical signaling in PASMCs.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Izabela Chrobak
- Lovelace Respiratory Research Institute, Albuquerque, New Mexico
| | | | - Sally H. Vitali
- Department of Anesthesiology, Critical Care, and Pain Medicine, Boston Children’s Hospital, Boston, Massachusetts; and
| | - Lynette M. Sholl
- Department of Pathology, Brigham and Women’s Hospital, Boston, Massachusetts
| | - Robert F. Padera
- Department of Pathology, Brigham and Women’s Hospital, Boston, Massachusetts
| | - David Lagares
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | | | | | | |
Collapse
|
20
|
Williams S, Charest J, Pollak M, Subramanian BK. Bioengineering Strategies To Develop Podocyte Culture Systems. TISSUE ENGINEERING PART B-REVIEWS 2021; 28:938-948. [PMID: 34541902 PMCID: PMC9419930 DOI: 10.1089/ten.teb.2021.0154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Unraveling the complex behavior of healthy and disease podocytes by analyzing the changes in their unique arrangement of foot processes, slit diaphragm and the 3D morphology is a long-standing goal in kidney-glomerular research. The complexities surrounding the podocytes' accessibility in animal models and growing evidence of differences between humans and animal systems have compelled researchers to look for alternate approaches to study podocyte behaviors. With the advent of bioengineered models, an increasingly powerful and diverse set of tools is available to develop novel podocyte culture systems. This review discusses the pertinence of various culture models of podocytes to study podocyte mechanisms in both normal physiology and disease conditions. While no one in vitro system comprehensively recapitulates podocytes' in vivo architecture, we emphasize how the existing systems can be exploited to answer targeted questions on podocyte structure and function. We highlight the distinct advantages and limitations of using these models to study podocyte behaviors and screen therapeutics. Finally, we discuss various considerations and potential engineering strategies for developing next-generation complex 3D culture models for studying podocyte behaviors in vitro.
Collapse
Affiliation(s)
- Sarah Williams
- Beth Israel Deaconess Medical Center, 1859, Boston, Massachusetts, United States;
| | - Joseph Charest
- Draper Laboratory, Biomedical Engineering, 555 Technology Square, Cambridge, Massachusetts, United States, 02139;
| | - Martin Pollak
- Beth Israel Deaconess Medical Center, 1859, Boston, Massachusetts, United States;
| | | |
Collapse
|
21
|
Wang L, Zheng F, Song R, Zhuang L, Yang M, Suo J, Li L. Integrins in the Regulation of Mesenchymal Stem Cell Differentiation by Mechanical Signals. Stem Cell Rev Rep 2021; 18:126-141. [PMID: 34536203 DOI: 10.1007/s12015-021-10260-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/02/2021] [Indexed: 10/20/2022]
Abstract
Mesenchymal stem cells (MSCs) can sense and convert mechanical stimuli signals into a chemical response. Integrins are involved in the mechanotransduction from inside to outside and from outside to inside, and ultimately affect the fate of MSCs responding to different mechanical signals. Different integrins participate in different signaling pathways to regulate MSCs multi-differentiation. In this review, we summarize the latest advances in the effects of mechanical signals on the differentiation of MSCs, the importance of integrins in mechanotransduction, the relationship between integrin heterodimers and different mechanical signals, and the interaction among mechanical signals. We put forward our views on the prospect and challenges of developing mechanical biology in tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Lei Wang
- Department of Gastrointestinal Surgery, Jilin University First Hospital, Jilin University, 130021, Changchun, People's Republic of China
| | - Fuwen Zheng
- Norman Bethune College of Medicine, Jilin University, 130021, Changchun, People's Republic of China
| | - Ruixue Song
- Norman Bethune College of Medicine, Jilin University, 130021, Changchun, People's Republic of China
| | - Lequan Zhuang
- Norman Bethune College of Medicine, Jilin University, 130021, Changchun, People's Republic of China
| | - Ming Yang
- Department of Molecular Biology, College of Basic Medical Sciences, Jilin University, 130021, Changchun, People's Republic of China.
| | - Jian Suo
- Department of Gastrointestinal Surgery, Jilin University First Hospital, Jilin University, 130021, Changchun, People's Republic of China.
| | - Lisha Li
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 130021, Changchun, People's Republic of China.
| |
Collapse
|
22
|
Castillo-Badillo JA, Gautam N. An optogenetic model reveals cell shape regulation through FAK and fascin. J Cell Sci 2021; 134:269115. [PMID: 34114634 DOI: 10.1242/jcs.258321] [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] [Received: 12/15/2020] [Accepted: 06/01/2021] [Indexed: 12/17/2022] Open
Abstract
Cell shape regulation is important, but the mechanisms that govern shape are not fully understood, in part due to limited experimental models in which cell shape changes and underlying molecular processes can be rapidly and non-invasively monitored in real time. Here, we used an optogenetic tool to activate RhoA in the middle of mononucleated macrophages to induce contraction, resulting in a side with the nucleus that retained its shape and a non-nucleated side that was unable to maintain its shape and collapsed. In cells overexpressing focal adhesion kinase (FAK; also known as PTK2), the non-nucleated side exhibited a wide flat morphology and was similar in adhesion area to the nucleated side. In cells overexpressing fascin, an actin-bundling protein, the non-nucleated side assumed a spherical shape and was similar in height to the nucleated side. This effect of fascin was also observed in fibroblasts even without inducing furrow formation. Based on these results, we conclude that FAK and fascin work together to maintain cell shape by regulating adhesion area and height, respectively, in different cell types. This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Jean A Castillo-Badillo
- Department of Anesthesiology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - N Gautam
- Department of Anesthesiology, Washington University School of Medicine, St Louis, MO 63110, USA.,Department of Genetics, Washington University School of Medicine, St Louis, MO 63110, USA
| |
Collapse
|
23
|
Ebefors K, Lassén E, Anandakrishnan N, Azeloglu EU, Daehn IS. Modeling the Glomerular Filtration Barrier and Intercellular Crosstalk. Front Physiol 2021; 12:689083. [PMID: 34149462 PMCID: PMC8206562 DOI: 10.3389/fphys.2021.689083] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 05/05/2021] [Indexed: 12/11/2022] Open
Abstract
The glomerulus is a compact cluster of capillaries responsible for blood filtration and initiating urine production in the renal nephrons. A trilaminar structure in the capillary wall forms the glomerular filtration barrier (GFB), composed of glycocalyx-enriched and fenestrated endothelial cells adhering to the glomerular basement membrane and specialized visceral epithelial cells, podocytes, forming the outermost layer with a molecular slit diaphragm between their interdigitating foot processes. The unique dynamic and selective nature of blood filtration to produce urine requires the functionality of each of the GFB components, and hence, mimicking the glomerular filter in vitro has been challenging, though critical for various research applications and drug screening. Research efforts in the past few years have transformed our understanding of the structure and multifaceted roles of the cells and their intricate crosstalk in development and disease pathogenesis. In this review, we present a new wave of technologies that include glomerulus-on-a-chip, three-dimensional microfluidic models, and organoids all promising to improve our understanding of glomerular biology and to enable the development of GFB-targeted therapies. Here, we also outline the challenges and the opportunities of these emerging biomimetic systems that aim to recapitulate the complex glomerular filter, and the evolving perspectives on the sophisticated repertoire of cellular signaling that comprise the glomerular milieu.
Collapse
Affiliation(s)
- Kerstin Ebefors
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Emelie Lassén
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Nanditha Anandakrishnan
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Evren U Azeloglu
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Ilse S Daehn
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
24
|
|
25
|
Páez-Pérez M, López-Duarte I, Vyšniauskas A, Brooks NJ, Kuimova MK. Imaging non-classical mechanical responses of lipid membranes using molecular rotors. Chem Sci 2020; 12:2604-2613. [PMID: 34164028 PMCID: PMC8179291 DOI: 10.1039/d0sc05874b] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 12/22/2020] [Indexed: 12/15/2022] Open
Abstract
Lipid packing in cellular membranes has a direct effect on membrane tension and microviscosity, and plays a central role in cellular adaptation, homeostasis and disease. According to conventional mechanical descriptions, viscosity and tension are directly interconnected, with increased tension leading to decreased membrane microviscosity. However, the intricate molecular interactions that combine to build the structure and function of a cell membrane suggest a more complex relationship between these parameters. In this work, a viscosity-sensitive fluorophore ('molecular rotor') is used to map changes in microviscosity in model membranes under conditions of osmotic stress. Our results suggest that the relationship between membrane tension and microviscosity is strongly influenced by the bilayer's lipid composition. In particular, we show that the effects of increasing tension are minimised for membranes that exhibit liquid disordered (Ld) - liquid ordered (Lo) phase coexistence; while, surprisingly, membranes in pure gel and Lo phases exhibit a negative compressibility behaviour, i.e. they soften upon compression.
Collapse
Affiliation(s)
- Miguel Páez-Pérez
- MSRH, Department of Chemistry, Imperial College London Wood Lane London W12 0BZ UK
| | - Ismael López-Duarte
- MSRH, Department of Chemistry, Imperial College London Wood Lane London W12 0BZ UK
- Departamento de Química Orgánica, Universidad Autónoma de Madrid Cantoblanco 28049 Madrid Spain
| | - Aurimas Vyšniauskas
- MSRH, Department of Chemistry, Imperial College London Wood Lane London W12 0BZ UK
- Center of Physical Sciences and Technology Saulėtekio av. 3 Vilnius Lithuania
| | - Nicholas J Brooks
- MSRH, Department of Chemistry, Imperial College London Wood Lane London W12 0BZ UK
| | - Marina K Kuimova
- MSRH, Department of Chemistry, Imperial College London Wood Lane London W12 0BZ UK
| |
Collapse
|
26
|
Vasilevich AS, Vermeulen S, Kamphuis M, Roumans N, Eroumé S, Hebels DGAJ, van de Peppel J, Reihs R, Beijer NRM, Carlier A, Carpenter AE, Singh S, de Boer J. On the correlation between material-induced cell shape and phenotypical response of human mesenchymal stem cells. Sci Rep 2020; 10:18988. [PMID: 33149200 PMCID: PMC7642380 DOI: 10.1038/s41598-020-76019-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 10/13/2020] [Indexed: 12/21/2022] Open
Abstract
Learning rules by which cell shape impacts cell function would enable control of cell physiology and fate in medical applications, particularly, on the interface of cells and material of the implants. We defined the phenotypic response of human bone marrow-derived mesenchymal stem cells (hMSCs) to 2176 randomly generated surface topographies by probing basic functions such as migration, proliferation, protein synthesis, apoptosis, and differentiation using quantitative image analysis. Clustering the surfaces into 28 archetypical cell shapes, we found a very strict correlation between cell shape and physiological response and selected seven cell shapes to describe the molecular mechanism leading to phenotypic diversity. Transcriptomics analysis revealed a tight link between cell shape, molecular signatures, and phenotype. For instance, proliferation is strongly reduced in cells with limited spreading, resulting in down-regulation of genes involved in the G2/M cycle and subsequent quiescence, whereas cells with large filopodia are related to activation of early response genes and inhibition of the osteogenic process. In this paper we were aiming to identify a universal set of genes that regulate the material-induced phenotypical response of human mesenchymal stem cells. This will allow designing implants that can actively regulate cellular, molecular signalling through cell shape. Here we are proposing an approach to tackle this question.
Collapse
Affiliation(s)
- Aliaksei S Vasilevich
- BIS-Biointerface Science in Regenerative Medicine, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Steven Vermeulen
- BIS-Biointerface Science in Regenerative Medicine, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands.,Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, The Netherlands
| | - Marloes Kamphuis
- Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, The Netherlands
| | - Nadia Roumans
- Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, The Netherlands
| | - Said Eroumé
- Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, The Netherlands
| | - Dennie G A J Hebels
- Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, The Netherlands
| | - Jeroen van de Peppel
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Rika Reihs
- Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, The Netherlands
| | - Nick R M Beijer
- Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, The Netherlands
| | - Aurélie Carlier
- Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, The Netherlands
| | - Anne E Carpenter
- Imaging Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Shantanu Singh
- Imaging Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jan de Boer
- BIS-Biointerface Science in Regenerative Medicine, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands.
| |
Collapse
|
27
|
Calizo RC, Bell MK, Ron A, Hu M, Bhattacharya S, Wong NJ, Janssen WGM, Perumal G, Pederson P, Scarlata S, Hone J, Azeloglu EU, Rangamani P, Iyengar R. Cell shape regulates subcellular organelle location to control early Ca 2+ signal dynamics in vascular smooth muscle cells. Sci Rep 2020; 10:17866. [PMID: 33082406 PMCID: PMC7576209 DOI: 10.1038/s41598-020-74700-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 10/05/2020] [Indexed: 12/23/2022] Open
Abstract
The shape of the cell is connected to its function; however, we do not fully understand underlying mechanisms by which global shape regulates a cell's functional capabilities. Using theory, experiments and simulation, we investigated how physiologically relevant cell shape changes affect subcellular organization, and consequently intracellular signaling, to control information flow needed for phenotypic function. Vascular smooth muscle cells going from a proliferative and motile circular shape to a contractile fusiform shape show changes in the location of the sarcoplasmic reticulum, inter-organelle distances, and differential distribution of receptors in the plasma membrane. These factors together lead to the modulation of signals transduced by the M3 muscarinic receptor/Gq/PLCβ pathway at the plasma membrane, amplifying Ca2+ dynamics in the cytoplasm, and the nucleus resulting in phenotypic changes, as determined by increased activity of myosin light chain kinase in the cytoplasm and enhanced nuclear localization of the transcription factor NFAT. Taken together, our observations show a systems level phenomenon whereby global cell shape affects subcellular organization to modulate signaling that enables phenotypic changes.
Collapse
Affiliation(s)
- R C Calizo
- Department of Pharmacological Sciences, Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1215, New York, NY, 10029, USA
| | - M K Bell
- Department of Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - A Ron
- Department of Mechanical Engineering, Columbia University, New York, NY, 10027, USA
| | - M Hu
- Department of Mechanical Engineering, Columbia University, New York, NY, 10027, USA
| | - S Bhattacharya
- Department of Mechanical Engineering, Columbia University, New York, NY, 10027, USA
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - N J Wong
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - W G M Janssen
- Department of Pharmacological Sciences, Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1215, New York, NY, 10029, USA
| | - G Perumal
- Carl Zeiss Microscopy LLC, White Plains, NY, 10601, USA
| | - P Pederson
- Carl Zeiss Microscopy LLC, White Plains, NY, 10601, USA
| | - S Scarlata
- Department of Chemistry and Biochemistry, Worcester Polytechnic Institute, Worcester, MA, 01609, USA
| | - J Hone
- Department of Mechanical Engineering, Columbia University, New York, NY, 10027, USA
| | - E U Azeloglu
- Department of Pharmacological Sciences, Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1215, New York, NY, 10029, USA
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - P Rangamani
- Department of Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, CA, 92093, USA.
| | - R Iyengar
- Department of Pharmacological Sciences, Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1215, New York, NY, 10029, USA.
| |
Collapse
|
28
|
Song D, Dong L, Gupta M, Li L, Klaas O, Loghin A, Beall M, Chen CS, Oberai AA. Recovery of Tractions Exerted by Single Cells in Three-Dimensional Nonlinear Matrices. J Biomech Eng 2020; 142:081012. [PMID: 32320015 PMCID: PMC7477711 DOI: 10.1115/1.4046974] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 04/07/2020] [Indexed: 01/30/2023]
Abstract
Cell-generated tractions play an important role in various physiological and pathological processes such as stem-cell differentiation, cell migration, wound healing, and cancer metastasis. Traction force microscopy (TFM) is a technique for quantifying cellular tractions during cell-matrix interactions. Most applications of this technique have heretofore assumed that the matrix surrounding the cells is linear elastic and undergoes infinitesimal strains, but recent experiments have shown that the traction-induced strains can be large (e.g., more than 50%). In this paper, we propose a novel three-dimensional (3D) TFM approach that consistently accounts for both the geometric nonlinearity introduced by large strains in the matrix, and the material nonlinearity due to strain-stiffening of the matrix. In particular, we pose the TFM problem as a nonlinear inverse hyperelasticity problem in the stressed configuration of the matrix, with the objective of determining the cellular tractions that are consistent with the measured displacement field in the matrix. We formulate the inverse problem as a constrained minimization problem and develop an efficient adjoint-based minimization procedure to solve it. We first validate our approach using simulated data, and quantify its sensitivity to noise. We then employ the new approach to recover tractions exerted by NIH 3T3 cells fully encapsulated in hydrogel matrices of varying stiffness. We find that neglecting nonlinear effects can induce significant errors in traction reconstructions. We also find that cellular tractions roughly increase with gel stiffness, while the strain energy appears to saturate.
Collapse
Affiliation(s)
- Dawei Song
- Department of Aerospace and Mechanical Engineering, University of Southern California, Los Angeles, CA 90089
| | - Li Dong
- Walker Department of Mechanical Engineering, Oden Institute for Computational Engineering and Sciences, University of Texas at Austin, Austin, TX 78712
| | - Mukund Gupta
- Department of Biomedical Engineering, Biological Design Center, Boston University, Boston, MA 02215
| | - Linqing Li
- Department of Biomedical Engineering, Biological Design Center, Boston University, Boston, MA 02215
| | | | | | | | - Christopher S. Chen
- Department of Biomedical Engineering, Biological Design Center, Boston University, Boston, MA 02215
| | - Assad A. Oberai
- Department of Aerospace and Mechanical Engineering, University of Southern California, Los Angeles, CA 90089
| |
Collapse
|
29
|
Ge X, Zhang T, Yu X, Muwonge AN, Anandakrishnan N, Wong NJ, Haydak JC, Reid JM, Fu J, Wong JS, Bhattacharya S, Cuttitta CM, Zhong F, Gordon RE, Salem F, Janssen W, Hone JC, Zhang A, Li H, He JC, Gusella GL, Campbell KN, Azeloglu EU. LIM-Nebulette Reinforces Podocyte Structural Integrity by Linking Actin and Vimentin Filaments. J Am Soc Nephrol 2020; 31:2372-2391. [PMID: 32737144 DOI: 10.1681/asn.2019121261] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 06/06/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Maintenance of the intricate interdigitating morphology of podocytes is crucial for glomerular filtration. One of the key aspects of specialized podocyte morphology is the segregation and organization of distinct cytoskeletal filaments into different subcellular components, for which the exact mechanisms remain poorly understood. METHODS Cells from rats, mice, and humans were used to describe the cytoskeletal configuration underlying podocyte structure. Screening the time-dependent proteomic changes in the rat puromycin aminonucleoside-induced nephropathy model correlated the actin-binding protein LIM-nebulette strongly with glomerular function. Single-cell RNA sequencing and immunogold labeling were used to determine Nebl expression specificity in podocytes. Automated high-content imaging, super-resolution microscopy, atomic force microscopy (AFM), live-cell imaging of calcium, and measurement of motility and adhesion dynamics characterized the physiologic role of LIM-nebulette in podocytes. RESULTS Nebl knockout mice have increased susceptibility to adriamycin-induced nephropathy and display morphologic, cytoskeletal, and focal adhesion abnormalities with altered calcium dynamics, motility, and Rho GTPase activity. LIM-nebulette expression is decreased in diabetic nephropathy and FSGS patients at both the transcript and protein level. In mice, rats, and humans, LIM-nebulette expression is localized to primary, secondary, and tertiary processes of podocytes, where it colocalizes with focal adhesions as well as with vimentin fibers. LIM-nebulette shRNA knockdown in immortalized human podocytes leads to dysregulation of vimentin filament organization and reduced cellular elasticity as measured by AFM indentation. CONCLUSIONS LIM-nebulette is a multifunctional cytoskeletal protein that is critical in the maintenance of podocyte structural integrity through active reorganization of focal adhesions, the actin cytoskeleton, and intermediate filaments.
Collapse
Affiliation(s)
- Xuhua Ge
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Tao Zhang
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Xiaoxia Yu
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Alecia N Muwonge
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Nanditha Anandakrishnan
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Nicholas J Wong
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Jonathan C Haydak
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Jordan M Reid
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Jia Fu
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Jenny S Wong
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Smiti Bhattacharya
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York.,Department of Mechanical Engineering, Columbia University, New York, New York
| | - Christina M Cuttitta
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Fang Zhong
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Ronald E Gordon
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Fadi Salem
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - William Janssen
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York
| | - James C Hone
- Department of Mechanical Engineering, Columbia University, New York, New York
| | - Aihua Zhang
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Hong Li
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers University-New Jersey Medical School, Newark, New Jersey
| | - John C He
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York.,Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - G Luca Gusella
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Kirk N Campbell
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Evren U Azeloglu
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York .,Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
30
|
Controlling osteoblast morphology and proliferation via surface micro-topographies of implant biomaterials. Sci Rep 2020; 10:12810. [PMID: 32732908 PMCID: PMC7393177 DOI: 10.1038/s41598-020-69685-6] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 07/15/2020] [Indexed: 12/20/2022] Open
Abstract
Current research on surface modifications has yielded advanced implant biomaterials. Various implant surface modifications have been shown to be promising in improving bone target cell response, but more comprehensive studies whether certain implant surface modifications can directly target cell behavioural features such as morphogenesis and proliferation are needed. Here, we studied the response of primary alveolar bone cells on various implant surface modifications in terms of osteoblast morphology and proliferation in vitro. Analyses of surface modifications led to surface-related test parameters including the topographical parameters micro-roughness, texture aspect and surface enlargement as well as the physicochemical parameter surface wettability. We compared osteoblast morphology and proliferation towards the above-mentioned parameters and found that texture aspect and surface enlargement but not surface roughness or wettability exhibited significant impact on osteoblast morphology and proliferation. Detailed analysis revealed osteoblast proliferation as a function of cell morphology, substantiated by an osteoblast size- and morphology-dependent increase in mitotic activity. These findings show that implant surface topography controls cell behavioural morphology and subsequently cell proliferation, thereby opening the road for cell instructive biomaterials.
Collapse
|
31
|
Honig F, Vermeulen S, Zadpoor AA, de Boer J, Fratila-Apachitei LE. Natural Architectures for Tissue Engineering and Regenerative Medicine. J Funct Biomater 2020; 11:E47. [PMID: 32645945 PMCID: PMC7565607 DOI: 10.3390/jfb11030047] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 06/27/2020] [Accepted: 07/03/2020] [Indexed: 02/07/2023] Open
Abstract
The ability to control the interactions between functional biomaterials and biological systems is of great importance for tissue engineering and regenerative medicine. However, the underlying mechanisms defining the interplay between biomaterial properties and the human body are complex. Therefore, a key challenge is to design biomaterials that mimic the in vivo microenvironment. Over millions of years, nature has produced a wide variety of biological materials optimised for distinct functions, ranging from the extracellular matrix (ECM) for structural and biochemical support of cells to the holy lotus with special wettability for self-cleaning effects. Many of these systems found in biology possess unique surface properties recognised to regulate cell behaviour. Integration of such natural surface properties in biomaterials can bring about novel cell responses in vitro and provide greater insights into the processes occurring at the cell-biomaterial interface. Using natural surfaces as templates for bioinspired design can stimulate progress in the field of regenerative medicine, tissue engineering and biomaterials science. This literature review aims to combine the state-of-the-art knowledge in natural and nature-inspired surfaces, with an emphasis on material properties known to affect cell behaviour.
Collapse
Affiliation(s)
- Floris Honig
- Laboratory for Cell Biology-Inspired Tissue Engineering, MERLN Institute, University of Maastricht, 6229 ET Maastricht, The Netherlands; (F.H.); (S.V.)
| | - Steven Vermeulen
- Laboratory for Cell Biology-Inspired Tissue Engineering, MERLN Institute, University of Maastricht, 6229 ET Maastricht, The Netherlands; (F.H.); (S.V.)
- BioInterface Science Group, Department of Biomedical Engineering, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands;
| | - Amir A. Zadpoor
- Biomaterials and Tissue Biomechanics Section, Department of Biomechanical Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology, 2628 CD Delft, The Netherlands;
| | - Jan de Boer
- BioInterface Science Group, Department of Biomedical Engineering, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands;
| | - Lidy E. Fratila-Apachitei
- Biomaterials and Tissue Biomechanics Section, Department of Biomechanical Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology, 2628 CD Delft, The Netherlands;
| |
Collapse
|
32
|
Mohan AS, Dean KM, Isogai T, Kasitinon SY, Murali VS, Roudot P, Groisman A, Reed DK, Welf ES, Han SJ, Noh J, Danuser G. Enhanced Dendritic Actin Network Formation in Extended Lamellipodia Drives Proliferation in Growth-Challenged Rac1 P29S Melanoma Cells. Dev Cell 2020; 49:444-460.e9. [PMID: 31063759 DOI: 10.1016/j.devcel.2019.04.007] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 01/21/2019] [Accepted: 04/05/2019] [Indexed: 12/19/2022]
Abstract
Actin assembly supplies the structural framework for cell morphology and migration. Beyond structure, this actin framework can also be engaged to drive biochemical signaling programs. Here, we describe how the hyperactivation of Rac1 via the P29S mutation (Rac1P29S) in melanoma hijacks branched actin network assembly to coordinate proliferative cues that facilitate metastasis and drug resistance. Upon growth challenge, Rac1P29S-harboring melanoma cells massively upregulate lamellipodia formation by dendritic actin polymerization. These extended lamellipodia form a signaling microdomain that sequesters and phospho-inactivates the tumor suppressor NF2/Merlin, driving Rac1P29S cell proliferation in growth suppressive conditions. These biochemically active lamellipodia require cell-substrate attachment but not focal adhesion assembly and drive proliferation independently of the ERK/MAPK pathway. These data suggest a critical link between cell morphology and cell signaling and reconcile the dichotomy of Rac1's regulation of both proliferation and actin assembly by revealing a mutual signaling axis wherein actin assembly drives proliferation in melanoma.
Collapse
Affiliation(s)
- Ashwathi S Mohan
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Kevin M Dean
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Tadamoto Isogai
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Stacy Y Kasitinon
- Children's Research Institute and the Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Vasanth S Murali
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Philippe Roudot
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Alex Groisman
- Department of Physics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Dana K Reed
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Erik S Welf
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Sangyoon J Han
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Biomedical Engineering, Michigan Technological University, Houghton, MI 49931, USA
| | - Jungsik Noh
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Gaudenz Danuser
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
33
|
Müller S, Ueda M, Isoshima T, Ushida T, Ito Y. Stretching of fibroblast cells on micropatterned gelatin on silicone elastomer. J Mater Chem B 2020; 8:416-425. [DOI: 10.1039/c9tb02203a] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Micropatterned gelatin was formed on the silicone elastomer surface. The micropattern enabled cell alignment, regulation of the cell shape, and endowed the cells with resistance against mechanical stress.
Collapse
Affiliation(s)
- Stefan Müller
- Emergent Bioengineering Materials Research Team
- RIKEN Center for Emergent Matter Science
- Saitama
- Japan
- Graduate School of Medicine
| | - Motoki Ueda
- Emergent Bioengineering Materials Research Team
- RIKEN Center for Emergent Matter Science
- Saitama
- Japan
- Nano Medical Engineering Laboratory
| | - Takashi Isoshima
- Nano Medical Engineering Laboratory
- RIKEN Cluster for Pioneering Research
- Saitama
- Japan
| | - Takashi Ushida
- Graduate School of Medicine
- The University of Tokyo
- Tokyo
- Japan
| | - Yoshihiro Ito
- Emergent Bioengineering Materials Research Team
- RIKEN Center for Emergent Matter Science
- Saitama
- Japan
- Nano Medical Engineering Laboratory
| |
Collapse
|
34
|
Vasan R, Maleckar MM, Williams CD, Rangamani P. DLITE Uses Cell-Cell Interface Movement to Better Infer Cell-Cell Tensions. Biophys J 2019; 117:1714-1727. [PMID: 31648791 PMCID: PMC6838938 DOI: 10.1016/j.bpj.2019.09.034] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 09/17/2019] [Accepted: 09/23/2019] [Indexed: 11/30/2022] Open
Abstract
Cell shapes and connectivities evolve over time as the colony changes shape or embryos develop. Shapes of intercellular interfaces are closely coupled with the forces resulting from actomyosin interactions, membrane tension, or cell-cell adhesions. Although it is possible to computationally infer cell-cell forces from a mechanical model of collective cell behavior, doing so for temporally evolving forces in a manner robust to digitization difficulties is challenging. Here, we introduce a method for dynamic local intercellular tension estimation (DLITE) that infers such evolution in temporal force with less sensitivity to digitization ambiguities or errors. This method builds upon previous work on single time points (cellular force-inference toolkit). We validate our method using synthetic geometries. DLITE's inferred cell colony tension evolutions correlate better with ground truth for these synthetic geometries as compared to tension values inferred from methods that consider each time point in isolation. We introduce cell connectivity errors, angle estimate errors, connection mislocalization, and connection topological changes to synthetic data and show that DLITE has reduced sensitivity to these conditions. Finally, we apply DLITE to time series of human-induced pluripotent stem cell colonies with endogenously expressed GFP-tagged zonulae occludentes-1. We show that DLITE offers improved stability in the inference of cell-cell tensions and supports a correlation between the dynamics of cell-cell forces and colony rearrangement.
Collapse
Affiliation(s)
- Ritvik Vasan
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, San Diego, California
| | | | | | - Padmini Rangamani
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, San Diego, California.
| |
Collapse
|
35
|
Jang YH, Jin X, Shankar P, Lee JH, Jo K, Lim KI. Molecular-Level Interactions between Engineered Materials and Cells. Int J Mol Sci 2019; 20:E4142. [PMID: 31450647 PMCID: PMC6747072 DOI: 10.3390/ijms20174142] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 08/19/2019] [Accepted: 08/21/2019] [Indexed: 12/13/2022] Open
Abstract
Various recent experimental observations indicate that growing cells on engineered materials can alter their physiology, function, and fate. This finding suggests that better molecular-level understanding of the interactions between cells and materials may guide the design and construction of sophisticated artificial substrates, potentially enabling control of cells for use in various biomedical applications. In this review, we introduce recent research results that shed light on molecular events and mechanisms involved in the interactions between cells and materials. We discuss the development of materials with distinct physical, chemical, and biological features, cellular sensing of the engineered materials, transfer of the sensing information to the cell nucleus, subsequent changes in physical and chemical states of genomic DNA, and finally the resulting cellular behavior changes. Ongoing efforts to advance materials engineering and the cell-material interface will eventually expand the cell-based applications in therapies and tissue regenerations.
Collapse
Affiliation(s)
- Yoon-Ha Jang
- Department of Chemical and Biological Engineering, Sookmyung Women's University, Seoul 04310, Korea
| | - Xuelin Jin
- Department of Chemistry and Integrated Biotechnology, Sogang University, Seoul 04107, Korea
| | - Prabakaran Shankar
- School of Advanced Materials Science and Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Korea
| | - Jung Heon Lee
- School of Advanced Materials Science and Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Korea.
| | - Kyubong Jo
- Department of Chemistry and Integrated Biotechnology, Sogang University, Seoul 04107, Korea.
| | - Kwang-Il Lim
- Department of Chemical and Biological Engineering, Sookmyung Women's University, Seoul 04310, Korea.
| |
Collapse
|
36
|
Calizo RC, Bhattacharya S, van Hasselt JGC, Wei C, Wong JS, Wiener RJ, Ge X, Wong NJ, Lee JJ, Cuttitta CM, Jayaraman G, Au VH, Janssen W, Liu T, Li H, Salem F, Jaimes EA, Murphy B, Campbell KN, Azeloglu EU. Disruption of podocyte cytoskeletal biomechanics by dasatinib leads to nephrotoxicity. Nat Commun 2019; 10:2061. [PMID: 31053734 PMCID: PMC6499885 DOI: 10.1038/s41467-019-09936-x] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 04/05/2019] [Indexed: 12/22/2022] Open
Abstract
Nephrotoxicity is a critical adverse event that leads to discontinuation of kinase inhibitor (KI) treatment. Here we show, through meta-analyses of FDA Adverse Event Reporting System, that dasatinib is associated with high risk for glomerular toxicity that is uncoupled from hypertension, suggesting a direct link between dasatinib and podocytes. We further investigate the cellular effects of dasatinib and other comparable KIs with varying risks of nephrotoxicity. Dasatinib treated podocytes show significant changes in focal adhesions, actin cytoskeleton, and morphology that are not observed with other KIs. We use phosphoproteomics and kinome profiling to identify the molecular mechanisms of dasatinib-induced injury to the actin cytoskeleton, and atomic force microscopy to quantify impairment to cellular biomechanics. Furthermore, chronic administration of dasatinib in mice causes reversible glomerular dysfunction, loss of stress fibers, and foot process effacement. We conclude that dasatinib induces nephrotoxicity through altered podocyte actin cytoskeleton, leading to injurious cellular biomechanics.
Collapse
Affiliation(s)
- Rhodora C Calizo
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.,Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Smiti Bhattacharya
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.,Department of Mechanical Engineering, Columbia University, New York, NY, 10027, USA
| | - J G Coen van Hasselt
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Chengguo Wei
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Jenny S Wong
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Robert J Wiener
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Xuhua Ge
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Nicholas J Wong
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Jia-Jye Lee
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Christina M Cuttitta
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Gomathi Jayaraman
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Vivienne H Au
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - William Janssen
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Tong Liu
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers University-New Jersey Medical School, Newark, NJ, 07103, USA
| | - Hong Li
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers University-New Jersey Medical School, Newark, NJ, 07103, USA
| | - Fadi Salem
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Edgar A Jaimes
- Renal Service, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Barbara Murphy
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Kirk N Campbell
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Evren U Azeloglu
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA. .,Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
37
|
Prasad A, Alizadeh E. Cell Form and Function: Interpreting and Controlling the Shape of Adherent Cells. Trends Biotechnol 2019; 37:347-357. [DOI: 10.1016/j.tibtech.2018.09.007] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 09/14/2018] [Accepted: 09/18/2018] [Indexed: 12/13/2022]
|
38
|
Kim JK, Shin YJ, Ha LJ, Kim DH, Kim DH. Unraveling the Mechanobiology of the Immune System. Adv Healthc Mater 2019; 8:e1801332. [PMID: 30614636 PMCID: PMC7700013 DOI: 10.1002/adhm.201801332] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 12/01/2018] [Indexed: 12/20/2022]
Abstract
Cells respond and actively adapt to environmental cues in the form of mechanical stimuli. This extends to immune cells and their critical role in the maintenance of tissue homeostasis. Multiple recent studies have begun illuminating underlying mechanisms of mechanosensation in modulating immune cell phenotypes. Since the extracellular microenvironment is critical to modify cellular physiology that ultimately determines the functionality of the cell, understanding the interactions between immune cells and their microenvironment is necessary. This review focuses on mechanoregulation of immune responses mediated by macrophages, dendritic cells, and T cells, in the context of modern mechanobiology.
Collapse
Affiliation(s)
- Jeong-Ki Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea
| | - Yu Jung Shin
- Department of Bioengineering, University of Washington, Seattle, WA 98109, USA
| | - Leslie Jaesun Ha
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea
| | - Deok-Ho Kim
- Department of Bioengineering, University of Washington, Seattle, WA 98109, USA
| | - Dong-Hwee Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea
| |
Collapse
|
39
|
Gates EM, LaCroix AS, Rothenberg KE, Hoffman BD. Improving Quality, Reproducibility, and Usability of FRET-Based Tension Sensors. Cytometry A 2018; 95:201-213. [PMID: 30523675 DOI: 10.1002/cyto.a.23688] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 10/30/2018] [Accepted: 11/05/2018] [Indexed: 02/07/2023]
Abstract
Mechanobiology, the study of how mechanical forces affect cellular behavior, is an emerging field of study that has garnered broad and significant interest. Researchers are currently seeking to better understand how mechanical signals are transmitted, detected, and integrated at a subcellular level. One tool for addressing these questions is a Förster resonance energy transfer (FRET)-based tension sensor, which enables the measurement of molecular-scale forces across proteins based on changes in emitted light. However, the reliability and reproducibility of measurements made with these sensors has not been thoroughly examined. To address these concerns, we developed numerical methods that improve the accuracy of measurements made using sensitized emission-based imaging. To establish that FRET-based tension sensors are versatile tools that provide consistent measurements, we used these methods, and demonstrated that a vinculin tension sensor is unperturbed by cell fixation, permeabilization, and immunolabeling. This suggests FRET-based tension sensors could be coupled with a variety of immuno-fluorescent labeling techniques. Additionally, as tension sensors are frequently employed in complex biological samples where large experimental repeats may be challenging, we examined how sample size affects the uncertainty of FRET measurements. In total, this work establishes guidelines to improve FRET-based tension sensor measurements, validate novel implementations of these sensors, and ensure that results are precise and reproducible. © 2018 International Society for Advancement of Cytometry.
Collapse
Affiliation(s)
- Evan M Gates
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, 27708
| | - Andrew S LaCroix
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, 27708
| | - Katheryn E Rothenberg
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, 27708
| | - Brenton D Hoffman
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, 27708
| |
Collapse
|
40
|
Korolj A, Laschinger C, James C, Hu E, Velikonja C, Smith N, Gu I, Ahadian S, Willette R, Radisic M, Zhang B. Curvature facilitates podocyte culture in a biomimetic platform. LAB ON A CHIP 2018; 18:3112-3128. [PMID: 30264844 DOI: 10.1039/c8lc00495a] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Most kidney diseases begin with abnormalities in glomerular podocytes, motivating the need for podocyte models to study pathophysiological mechanisms and new treatment options. However, podocytes cultured in vitro face a limited ability to maintain appreciable extents of differentiation hallmarks, raising concerns over the relevance of study results. Many key properties such as nephrin expression and morphology reach plateaus that are far from the in vivo levels. Here, we demonstrate that a biomimetic topography, consisting of microhemispheres arrayed over the cell culture substrate, promotes podocyte differentiation in vitro. We define new methods for fabricating microscale curvature on various substrates, including a thin porous membrane. By growing podocytes on our topographic substrates, we found that these biophysical cues augmented nephrin gene expression, supported full-size nephrin protein expression, encouraged structural arrangement of F-actin and nephrin within the cell, and promoted process formation and even interdigitation compared to the flat substrates. Furthermore, the topography facilitated nephrin localization on curved structures while nuclei lay in the valleys between them. The improved differentiation was also evidenced by tracking barrier function to albumin over time using our custom topomembranes. Overall, our work presents accessible methods for incorporating microcurvature on various common substrates, and demonstrates the importance of biophysical stimulation in supporting higher-fidelity podocyte cultivation in vitro.
Collapse
Affiliation(s)
- Anastasia Korolj
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Canada.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Bao M, Xie J, Huck WTS. Recent Advances in Engineering the Stem Cell Microniche in 3D. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2018; 5:1800448. [PMID: 30128252 PMCID: PMC6096985 DOI: 10.1002/advs.201800448] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 05/01/2018] [Indexed: 05/18/2023]
Abstract
Conventional 2D cell culture techniques have provided fundamental insights into key biochemical and biophysical mechanisms responsible for various cellular behaviors, such as cell adhesion, spreading, division, proliferation, and differentiation. However, 2D culture in vitro does not fully capture the physical and chemical properties of the native microenvironment. There is a growing body of research that suggests that cells cultured on 2D substrates differ greatly from those grown in vivo. This article focuses on recent progress in using bioinspired 3D matrices that recapitulate as many aspects of the natural extracellular matrix as possible. A range of techniques for the engineering of 3D microenvironment with precisely controlled biophysical and chemical properties, and the impact of these environments on cellular behavior, is reviewed. Finally, an outlook on future challenges for engineering the 3D microenvironment and how such approaches would further our understanding of the influence of the microenvironment on cell function is provided.
Collapse
Affiliation(s)
- Min Bao
- Institute for Molecules and MaterialsRadboud UniversityHeyendaalseweg 1356525 AJNijmegenThe Netherlands
| | - Jing Xie
- Institute for Molecules and MaterialsRadboud UniversityHeyendaalseweg 1356525 AJNijmegenThe Netherlands
| | - Wilhelm T. S. Huck
- Institute for Molecules and MaterialsRadboud UniversityHeyendaalseweg 1356525 AJNijmegenThe Netherlands
| |
Collapse
|
42
|
Spill F, Bakal C, Mak M. Mechanical and Systems Biology of Cancer. Comput Struct Biotechnol J 2018; 16:237-245. [PMID: 30105089 PMCID: PMC6077126 DOI: 10.1016/j.csbj.2018.07.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 07/03/2018] [Accepted: 07/11/2018] [Indexed: 12/18/2022] Open
Abstract
Mechanics and biochemical signaling are both often deregulated in cancer, leading toincreased cell invasiveness, proliferation, and survival. The dynamics and interactions of cytoskeletal components control basic mechanical properties, such as cell tension, stiffness, and engagement with the extracellular environment, which can lead to extracellular matrix remodeling. Intracellular mechanics can alter signaling and transcription factors, impacting cell decision making. Additionally, signaling from soluble and mechanical factors in the extracellular environment, such as substrate stiffness and ligand density, can modulate cytoskeletal dynamics. Computational models closely integrated with experimental support, incorporating cancer-specific parameters, can provide quantitative assessments and serve as predictive tools toward dissecting the feedback between signaling and mechanics and across multiple scales and domains in tumor progression.
Collapse
Affiliation(s)
- Fabian Spill
- School of Mathematics, University of Birmingham, Birmingham B15 2TT, UK
| | - Chris Bakal
- Division of Cancer Biology, Chester Beatty Laboratories, The Institute of Cancer Research, London SW3 6JB, UK
| | - Michael Mak
- Department of Biomedical Engineering, Yale University, New Haven, USA
| |
Collapse
|
43
|
Haupt A, Minc N. How cells sense their own shape - mechanisms to probe cell geometry and their implications in cellular organization and function. J Cell Sci 2018; 131:131/6/jcs214015. [PMID: 29581183 DOI: 10.1242/jcs.214015] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Cells come in a variety of shapes that most often underlie their functions. Regulation of cell morphogenesis implies that there are mechanisms for shape sensing that still remain poorly appreciated. Global and local cell geometry features, such as aspect ratio, size or membrane curvature, may be probed by intracellular modules, such as the cytoskeleton, reaction-diffusion systems or molecular complexes. In multicellular tissues, cell shape emerges as an important means to transduce tissue-inherent chemical and mechanical cues into intracellular organization. One emergent paradigm is that cell-shape sensing is most often based upon mechanisms of self-organization, rather than determinism. Here, we review relevant work that has elucidated some of the core principles of how cellular geometry may be conveyed into spatial information to guide processes, such as polarity, signaling, morphogenesis and division-plane positioning.
Collapse
Affiliation(s)
- Armin Haupt
- Institut Jacques Monod, CNRS UMR7592 and Université Paris Diderot, 15 rue Hélène Brion, 75205 Paris Cedex 13, France
| | - Nicolas Minc
- Institut Jacques Monod, CNRS UMR7592 and Université Paris Diderot, 15 rue Hélène Brion, 75205 Paris Cedex 13, France
| |
Collapse
|