1
|
Bril M, Boesveld JN, Coelho-Rato LS, Sahlgren CM, Bouten CVC, Kurniawan NA. Dynamic substrate topographies drive actin- and vimentin-mediated nuclear mechanoprotection events in human fibroblasts. BMC Biol 2025; 23:94. [PMID: 40189524 PMCID: PMC11974106 DOI: 10.1186/s12915-025-02199-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Accepted: 03/27/2025] [Indexed: 04/09/2025] Open
Abstract
BACKGROUND Dynamic physical changes in the extracellular environment of living tissues present a mechanical challenge for resident cells that can lead to damage to the nucleus, genome, and DNA. Recent studies have started to uncover nuclear mechanoprotection mechanisms that prevent excessive mechanical deformations of the nucleus. Here, we hypothesized that dynamic topographical changes in the cellular environment can be mechanically transmitted to the nucleus and trigger nuclear mechanoprotection events. We tested this using a photoresponsive hydrogel whose surface topography can be reversibly changed on demand upon light illumination, allowing us to subject cells to recurring microscale topographical changes. RESULTS With each recurring topographical change, fibroblasts were found to increasingly compact and relocate their nuclei away from the dynamic regions of the hydrogel. These cell-scale reorganization events were accompanied by an increase of global histone acetylation and decreased methylation in cells on the dynamic topographies, resulting in a minimization of DNA strand breakage. We further found that these nuclear mechanoprotection events were mediated by both vimentin intermediate filaments and the actin cytoskeleton. CONCLUSIONS Together, these data reveal that fibroblasts actively protect their nuclei in the presence of dynamic topographical changes through cytoskeleton-mediated mechanisms. Broadly, these results stress the importance of gaining a deeper fundamental understanding of the cellular mechanoresponse under dynamically changing conditions.
Collapse
Affiliation(s)
- Maaike Bril
- Department of Biomedical Engineering, Eindhoven University of Technology, PO Box 513, 5600 MB, Eindhoven, The Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, PO Box 513, 5600 MB, Eindhoven, The Netherlands
| | - Jules N Boesveld
- Department of Biomedical Engineering, Eindhoven University of Technology, PO Box 513, 5600 MB, Eindhoven, The Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, PO Box 513, 5600 MB, Eindhoven, The Netherlands
| | - Leila S Coelho-Rato
- Faculty of Science and Engineering, Åbo Akademi University, 20520, Turku, Finland
| | - Cecilia M Sahlgren
- Department of Biomedical Engineering, Eindhoven University of Technology, PO Box 513, 5600 MB, Eindhoven, The Netherlands
- Faculty of Science and Engineering, Åbo Akademi University, 20520, Turku, Finland
| | - Carlijn V C Bouten
- Department of Biomedical Engineering, Eindhoven University of Technology, PO Box 513, 5600 MB, Eindhoven, The Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, PO Box 513, 5600 MB, Eindhoven, The Netherlands
| | - Nicholas A Kurniawan
- Department of Biomedical Engineering, Eindhoven University of Technology, PO Box 513, 5600 MB, Eindhoven, The Netherlands.
- Institute for Complex Molecular Systems, Eindhoven University of Technology, PO Box 513, 5600 MB, Eindhoven, The Netherlands.
| |
Collapse
|
2
|
Akter MZ, Tufail F, Ahmad A, Oh YW, Kim JM, Kim S, Hasan MM, Li L, Lee DW, Kim YS, Lee SJ, Kim HS, Ahn Y, Choi YJ, Yi HG. Harnessing native blueprints for designing bioinks to bioprint functional cardiac tissue. iScience 2025; 28:111882. [PMID: 40177403 PMCID: PMC11964760 DOI: 10.1016/j.isci.2025.111882] [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] [Indexed: 04/05/2025] Open
Abstract
Cardiac tissue lacks regenerative capacity, making heart transplantation the primary treatment for end-stage heart failure. Engineered cardiac tissues developed through three-dimensional bioprinting (3DBP) offer a promising alternative. However, reproducing the native structure, cellular diversity, and functionality of cardiac tissue requires advanced cardiac bioinks. Major obstacles in CTE (cardiac tissue engineering) include accurately characterizing bioink properties, replicating the cardiac microenvironment, and achieving precise spatial organization. Optimizing bioink properties to closely mimic the extracellular matrix (ECM) is essential, as deviations may result in pathological effects. This review encompasses the rheological and electromechanical properties of bioinks and the function of the cardiac microenvironment in the design of functional cardiac constructs. Furthermore, it focuses on improving the rheological characteristics, printability, and functionality of bioinks, offering valuable perspectives for developing new bioinks especially designed for CTE.
Collapse
Affiliation(s)
- Mst Zobaida Akter
- Department of Convergence Biosystems Engineering, Chonnam National University, Gwangju 61186, Republic of Korea
- Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Gwangju 61186, Republic of Korea
- Advanced Medical Device Research Center for Cardiovascular Disease, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Fatima Tufail
- Department of Convergence Biosystems Engineering, Chonnam National University, Gwangju 61186, Republic of Korea
- Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Gwangju 61186, Republic of Korea
- Advanced Medical Device Research Center for Cardiovascular Disease, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Ashfaq Ahmad
- Department of Convergence Biosystems Engineering, Chonnam National University, Gwangju 61186, Republic of Korea
- Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Gwangju 61186, Republic of Korea
- Advanced Medical Device Research Center for Cardiovascular Disease, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Yoon Wha Oh
- Department of Convergence Biosystems Engineering, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Jung Min Kim
- Department of Convergence Biosystems Engineering, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Seoyeon Kim
- Department of Convergence Biosystems Engineering, Chonnam National University, Gwangju 61186, Republic of Korea
- Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Md Mehedee Hasan
- Department of Convergence Biosystems Engineering, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Longlong Li
- Department of Mechanical Engineering, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Dong-Weon Lee
- Advanced Medical Device Research Center for Cardiovascular Disease, Chonnam National University, Gwangju 61186, Republic of Korea
- Department of Mechanical Engineering, Chonnam National University, Gwangju 61186, Republic of Korea
- Center for Next-Generation Sensor Research and Development, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Yong Sook Kim
- Biomedical Research Institute, Chonnam National University Hospital, Gwangju 61469, Republic of Korea
| | - Su-jin Lee
- Biomedical Research Institute, Chonnam National University Hospital, Gwangju 61469, Republic of Korea
- Department of Forensic Medicine, Chonnam National University Medical School, Gwangju 61469, Republic of Korea
| | - Hyung-Seok Kim
- Department of Forensic Medicine, Chonnam National University Medical School, Gwangju 61469, Republic of Korea
| | - Youngkeun Ahn
- Division of Cardiology, Department of Internal Medicine, Chonnam National University Hospital, Chonnam National University Medical School, Gwangju, 61469, Republic of Korea
| | - Yeong-Jin Choi
- Advanced Bio and Healthcare Materials Research Division, Korea Institute of Materials Science (KIMS), Changwon 51508, Republic of Korea
- Advanced Materials Engineering, Korea National University of Science and Technology (UST), Changwon, Republic of Korea
| | - Hee-Gyeong Yi
- Department of Convergence Biosystems Engineering, Chonnam National University, Gwangju 61186, Republic of Korea
- Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Gwangju 61186, Republic of Korea
- Advanced Medical Device Research Center for Cardiovascular Disease, Chonnam National University, Gwangju 61186, Republic of Korea
| |
Collapse
|
3
|
Rodriguez Ayala A, Christ G, Griffin D. Cell-scale porosity minimizes foreign body reaction and promotes innervated myofiber formation after volumetric muscle loss. NPJ Regen Med 2025; 10:12. [PMID: 40025057 PMCID: PMC11873130 DOI: 10.1038/s41536-025-00395-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 01/29/2025] [Indexed: 03/04/2025] Open
Abstract
Volumetric muscle loss (VML) from severe traumatic injuries results in irreversible loss of contractile tissue and permanent functional deficits. These injuries resist endogenous healing and clinical treatment due to excessive inflammation, leading to fibrosis, muscle fiber denervation, and impaired regeneration. Using a rodent tibialis anterior VML model, this study demonstrates microporous annealed particle (MAP) hydrogel scaffolds as a biomaterial platform for improved muscle regeneration. Unlike bulk (nanoporous) hydrogel scaffolds, MAP scaffolds enhance integration by preventing a foreign body reaction, slowing implant degradation, and promoting regenerative macrophage polarization. Cell migration and angiogenesis occur throughout the implant before MAP scaffold degradation, with muscle fibers and neuromuscular junctions forming within the scaffolds. These structures continue developing as the implant degrades, suggesting MAP hydrogel scaffolds offer a promising therapeutic approach for VML injuries.
Collapse
Affiliation(s)
- Areli Rodriguez Ayala
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - George Christ
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA.
- Department of Orthopaedic Surgery, University of Virginia, Charlottesville, VA, USA.
| | - Donald Griffin
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA.
- Department of Chemical Engineering, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
4
|
Mayer MN, Köhler LM, Paulus M, Iberl S, Heinrich M, Wagner S, Maier LS, Dietl A. MATLAB-based Methods Allow Precise, High-Throughput Quantification of Nuclear Morphology and Texture in Tachycardiomyopathy. J Histochem Cytochem 2025; 73:115-128. [PMID: 40219615 PMCID: PMC11993534 DOI: 10.1369/00221554251332331] [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: 10/10/2024] [Accepted: 03/19/2025] [Indexed: 04/14/2025] Open
Abstract
The understanding of cardiomyopathies is hindered by a lack of quantitative histologic data. To address this methodical gap, we wrote a MATLAB-based image analysis platform to quantify nuclear and cellular disarray. We validated its utility in an animal model of tachycardiomyopathy (T-CM), whose ultrastructural remodeling processes have only partially been characterized and differ substantially from more prevalent cardiomyopathies. Six rabbits received right ventricular pacemaker implants. Three animals were paced incrementally up to 380 bpm for 30 days to induce T-CM. In three control rabbits, the pacemaker remained inactive (SHAM). Left ventricular tissue was collected, fixed in formalin, embedded in paraffin, stained, and digitized for nuclear morphometry, texture analysis, orientation analysis, and vascular architecture evaluation. Nuclear segmentation performed by the software was highly accurate, closely matching manual counts (mean manual nuclear count per slide = 81.3 ± 3.8, mean automated nuclear count per slide = 81.9 ± 4.3, r = 0.981, p<0.001). In T-CM, nuclei were enlarged [SHAM (a.u.) = 2362, T-CM (a.u.) = 2660, p=0.0042]. Texture patterns differed between the groups with higher nuclear contrast in T-CM [SHAM (a.u.) = 0.0169, T-CM (a.u.) = 0.0247, p=0.0149], highlighting structural remodeling at the nuclear level. Median vessel size increased in T-CM [SHAM (a.u.) = 1532, T-CM (a.u.) = 2421, p<0.0001]. In conclusion, our MATLAB-based image analysis platform allows high-throughput quantification of nuclear and extracellular disarray. It identified enlargement of nuclei and increased nuclear contrast as part of ultrastructural remodeling in tachycardiomyopathy.
Collapse
Affiliation(s)
- Moritz N.S. Mayer
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Lisa M. Köhler
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Michael Paulus
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Sabine Iberl
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Maria Heinrich
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Stefan Wagner
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Lars S. Maier
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Alexander Dietl
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
5
|
House A, Santillan A, Correa E, Youssef V, Guvendiren M. Cellular Alignment and Matrix Stiffening Induced Changes in Human Induced Pluripotent Stem Cell Derived Cardiomyocytes. Adv Healthc Mater 2025; 14:e2402228. [PMID: 39468891 DOI: 10.1002/adhm.202402228] [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: 07/03/2024] [Revised: 10/15/2024] [Indexed: 10/30/2024]
Abstract
Biological processes are inherently dynamic, necessitating biomaterial platforms capable of spatiotemporal control over cellular organization and matrix stiffness for accurate study of tissue development, wound healing, and disease. However, most in vitro platforms remain static. In this study, a dynamic biomaterial platform comprising a stiffening hydrogel is introduced and achieved through a stepwise approach of addition followed by light-mediated crosslinking, integrated with an elastomeric substrate featuring strain-responsive lamellar surface patterns. Employing this platform, the response of human induced pluripotent stem cell-derived cardiomyocytes (hIPSC-CMs) is investigated to dynamic stiffening from healthy to fibrotic tissue stiffness. The results demonstrate that culturing hIPSC-CMs on physiologically relevant healthy stiffness significantly enhances their function, as evidenced by increased sarcomere fraction, wider sarcomere width, significantly higher connexin-43 content, and elevated cell beating frequency compared to cells cultured on fibrotic matrix. Conversely, dynamic matrix stiffening negatively impacts hIPSC-CM function, with earlier stiffening events exerting a more pronounced hindering effect. These findings provide valuable insights into material-based approaches for addressing existing challenges in hIPSC-CM maturation and have broader implications across various tissue models, including muscle, tendon, nerve, and cornea, where both cellular alignment and matrix stiffening play pivotal roles in tissue development and regeneration.
Collapse
Affiliation(s)
- Andrew House
- Department of Chemical and Materials Engineering, New Jersey Institute of Technology, University Heights, Newark, NJ, 07102, USA
| | - Anjeli Santillan
- Department of Biomedical Engineering, New Jersey Institute of Technology, University Heights, Newark, NJ, 07102, USA
| | - Evan Correa
- Federated Department of Biological Sciences, New Jersey Institute of Technology, University Heights, Newark, NJ, 07102, USA
| | - Victoria Youssef
- Federated Department of Biological Sciences, New Jersey Institute of Technology, University Heights, Newark, NJ, 07102, USA
| | - Murat Guvendiren
- Department of Chemical and Materials Engineering, New Jersey Institute of Technology, University Heights, Newark, NJ, 07102, USA
- Department of Biomedical Engineering, New Jersey Institute of Technology, University Heights, Newark, NJ, 07102, USA
| |
Collapse
|
6
|
Zimmerman JF, Drennan DJ, Ikeda J, Jin Q, Ardoña HAM, Kim SL, Ishii R, Parker KK. Bioinspired design of a tissue-engineered ray with machine learning. Sci Robot 2025; 10:eadr6472. [PMID: 39937888 DOI: 10.1126/scirobotics.adr6472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 01/14/2025] [Indexed: 02/14/2025]
Abstract
In biomimetic design, researchers recreate existing biological structures to form functional devices. For biohybrid robotic swimmers assembled with tissue engineering, this is problematic because most devices operate at different length scales than their naturally occurring counterparts, resulting in reduced performance. To overcome these challenges, here, we demonstrate how machine learning-directed optimization (ML-DO) can be used to inform the design of a biohybrid robot, outperforming other nonlinear optimization techniques, such as Bayesian optimization, in the selection of high-performance geometries. We show how this approach can be used to maximize the thrust generated by a tissue-engineered mobuliform miniray. This results in devices that can swim at the millimeter scale while more closely preserving natural locomotive scaling laws. Overall, this work provides a quantitatively rigorous approach for the engineering design of muscular structure-function relationships in an automated fashion.
Collapse
Affiliation(s)
- John F Zimmerman
- Disease Biophysics Group, John A. Paulson School of Engineering and Applied Science, Harvard University, Boston, MA 02134, USA
| | - Daniel J Drennan
- Disease Biophysics Group, John A. Paulson School of Engineering and Applied Science, Harvard University, Boston, MA 02134, USA
| | - James Ikeda
- Disease Biophysics Group, John A. Paulson School of Engineering and Applied Science, Harvard University, Boston, MA 02134, USA
| | - Qianru Jin
- Disease Biophysics Group, John A. Paulson School of Engineering and Applied Science, Harvard University, Boston, MA 02134, USA
| | - Herdeline Ann M Ardoña
- Disease Biophysics Group, John A. Paulson School of Engineering and Applied Science, Harvard University, Boston, MA 02134, USA
| | - Sean L Kim
- Disease Biophysics Group, John A. Paulson School of Engineering and Applied Science, Harvard University, Boston, MA 02134, USA
| | - Ryoma Ishii
- Disease Biophysics Group, John A. Paulson School of Engineering and Applied Science, Harvard University, Boston, MA 02134, USA
- Medical and Health Informatics Laboratory, NTT Research Inc., Sunnyvale, CA 94085, USA
| | - Kevin Kit Parker
- Disease Biophysics Group, John A. Paulson School of Engineering and Applied Science, Harvard University, Boston, MA 02134, USA
| |
Collapse
|
7
|
Hatano R, Smith AM, Raman R, Zamora JE, Bashir R, McCloskey KE. Comparing fabrication techniques for engineered cardiac tissue. J Biomed Mater Res A 2024; 112:1921-1929. [PMID: 38752415 DOI: 10.1002/jbm.a.37737] [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: 07/11/2023] [Revised: 04/25/2024] [Accepted: 05/02/2024] [Indexed: 09/03/2024]
Abstract
Tissue engineering can provide in vitro models for drug testing, disease modeling, and perhaps someday, tissue/organ replacements. For building 3D heart tissue, the alignment of cardiac cells or cardiomyocytes (CMs) is important in generating a synchronously contracting tissue. To that end, researchers have generated several fabrication methods for building heart tissue, but direct comparisons of pros and cons using the same cell source is lacking. Here, we derived cardiomyocytes (CMs) from human induced pluripotent stem cells (hiPSCs) and compare the assembly of these cells using three fabrication methods: cardiospheres, muscle rings, and muscle strips. All three protocols successfully generated compacted tissue comprised of hiPSC-derived CMs stable for at least 2 weeks. The percentage of aligned cells was greatest in the muscle strip (55%) and the muscle ring (50%) compared with the relatively unaligned cardiospheres (35%). The iPSC-derived CMs within the muscle strip also exhibited the greatest elongation, with elongation factor at 2.0 compared with 1.5 for the muscle ring and 1.2 for the cardiospheres. This is the first direct comparison of various fabrication techniques using the same cell source.
Collapse
Affiliation(s)
- Rachel Hatano
- Graduate Program in Bioengineering and Small-scale Technologies, University of California, Merced, USA
| | - Ariell M Smith
- Bioengineering Department, University of California, Merced, USA
| | - Ritu Raman
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, USA
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, USA
| | - Jose E Zamora
- Graduate Program in Materials and Biomaterials Science and Engineering, University of California, Merced, USA
| | - Rashid Bashir
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, USA
| | - Kara E McCloskey
- Graduate Program in Bioengineering and Small-scale Technologies, University of California, Merced, USA
- Materials Science and Engineering Department, University of California, Merced, USA
| |
Collapse
|
8
|
Kahvecioğlu B, Mutlu Avinç G, Arslan Selçuk S. Biomimetic Adaptive Building Façade Modeling for Sustainable Urban Freshwater Ecosystems: Integration of Nature's Water-Harvesting Strategy into Sun-Breakers. Biomimetics (Basel) 2024; 9:569. [PMID: 39329591 PMCID: PMC11429842 DOI: 10.3390/biomimetics9090569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 09/06/2024] [Accepted: 09/12/2024] [Indexed: 09/28/2024] Open
Abstract
Urban freshwater ecosystems have many critical functions, such as providing water to all living things and supporting biodiversity. Factors such as water pollution, increased water consumption, habitat loss, climate change, and drought threaten the health of urban freshwater ecosystems. Looking for solutions to these challenges, this article aims to recycle water and return it to its life cycle using a climate-sensitive water collection strategy. The model focuses on the biomimetic method as a basic strategy. In this regard, the concept of water-harvesting has been examined in detail by conducting a deep literature review, including architecture and engineering disciplines. With all these data obtained, a synthesis/integration study was carried out by developing a model proposal based on adaptive building façade elements to solve the water problems experienced in cities. The model proposal, which is directly related to the titles of "Clean Water and Sanitation (SDG 6)" and "Sustainable Cities and Communities (SDG 11)", which are among the Sustainable Development Goals (SDGs), aims to provide different perspectives on the disciplines with its superficial and functional features. In this context, it is anticipated that the article will become an indispensable resource for other researchers working on the subject.
Collapse
Affiliation(s)
- Berkan Kahvecioğlu
- Department of Architecture, Faculty of Architecture, Gazi University, 06560 Ankara, Turkey
- Department of Construction and Technical, Erciyes University, 38280 Kayseri, Turkey
| | - Güneş Mutlu Avinç
- Department of Architecture, Faculty of Engineering and Architecture, Mus Alparslan University, 49250 Mus, Turkey
| | - Semra Arslan Selçuk
- Department of Architecture, Faculty of Architecture, Gazi University, 06560 Ankara, Turkey
| |
Collapse
|
9
|
Mastrodimos M, Jain S, Badv M, Shen J, Montazerian H, Meyer CE, Annabi N, Weiss PS. Human Skeletal Muscle Myoblast Culture in Aligned Bacterial Nanocellulose and Commercial Matrices. ACS APPLIED MATERIALS & INTERFACES 2024; 16:47150-47162. [PMID: 39206938 PMCID: PMC11403597 DOI: 10.1021/acsami.4c07612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 08/06/2024] [Accepted: 08/06/2024] [Indexed: 09/04/2024]
Abstract
Bacterial nanocellulose (BNC) is a durable, flexible, and dynamic biomaterial capable of serving a wide variety of fields, sectors, and applications within biotechnology, healthcare, electronics, agriculture, fashion, and others. BNC is produced spontaneously in carbohydrate-rich bacterial culture media, forming a cellulosic pellicle via a nanonetwork of fibrils extruded from certain genera. Herein, we demonstrate engineering BNC-based scaffolds with tunable physical and mechanical properties through postprocessing. Human skeletal muscle myoblasts (HSMMs) were cultured on these scaffolds, and in vitro electrical stimulation was applied to promote cellular function for tissue engineering applications. We compared physiologic maturation markers of human skeletal muscle myoblast development using a 2.5-dimensional culture paradigm in fabricated BNC scaffolds, compared to two-dimensional (2D) controls. We demonstrate that the culture of human skeletal muscle myoblasts on BNC scaffolds developed under electrical stimulation produced highly aligned, physiologic morphology of human skeletal muscle myofibers compared to unstimulated BNC and standard 2D culture. Furthermore, we compared an array of metrics to assess the BNC scaffold in a rigorous head-to-head study with commercially available, clinically approved matrices, Kerecis Omega3 Wound Matrix (Marigen) and Phoenix as well as a gelatin methacryloyl (GelMA) hydrogel. The BNC scaffold outcompeted industry standard matrices as well as a 20% GelMA hydrogel in durability and sustained the support of human skeletal muscle myoblasts in vitro. This work offers a robust demonstration of BNC scaffold cytocompatibility with human skeletal muscle cells and sets the basis for future work in healthcare, bioengineering, and medical implant technological development.
Collapse
Affiliation(s)
- Melina Mastrodimos
- California
NanoSystems Institute, University of California,
Los Angeles, Los Angeles, California 90095, United States
- Department
of Bioengineering, University of California,
Los Angeles, 410 Westwood Plaza, Los Angeles, California 90095, United States
| | - Saumya Jain
- Department
of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, California 90095, United States
| | - Maryam Badv
- Department
of Biomedical Engineering, Schulich School of Engineering, University of Calgary, Calgary, Alberta T2N 1N4, Canada
| | - Jun Shen
- California
NanoSystems Institute, University of California,
Los Angeles, Los Angeles, California 90095, United States
- Department
of Chemistry & Biochemistry, University
of California, Los Angeles, Los
Angeles, California 90095, United States
| | - Hossein Montazerian
- California
NanoSystems Institute, University of California,
Los Angeles, Los Angeles, California 90095, United States
- Department
of Bioengineering, University of California,
Los Angeles, 410 Westwood Plaza, Los Angeles, California 90095, United States
- Terasaki
Institute for Biomedical Innovation, Los Angeles, California 90024, United States
| | - Claire E. Meyer
- Department
of Chemistry & Biochemistry, University
of California, Los Angeles, Los
Angeles, California 90095, United States
| | - Nasim Annabi
- Department
of Bioengineering, University of California,
Los Angeles, 410 Westwood Plaza, Los Angeles, California 90095, United States
- Department
of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, California 90095, United States
| | - Paul S. Weiss
- California
NanoSystems Institute, University of California,
Los Angeles, Los Angeles, California 90095, United States
- Department
of Bioengineering, University of California,
Los Angeles, 410 Westwood Plaza, Los Angeles, California 90095, United States
- Department
of Materials Science and Engineering, University
of California, Los Angeles, Los
Angeles, California 90095, United States
- Department
of Chemistry & Biochemistry, University
of California, Los Angeles, Los
Angeles, California 90095, United States
| |
Collapse
|
10
|
Chen TA, Zhao BB, Balbin RA, Sharma S, Ha D, Kamp TJ, Zhou Y, Zhao F. Engineering a robust and anisotropic cardiac-specific extracellular matrix scaffold for cardiac patch tissue engineering. Matrix Biol Plus 2024; 23:100151. [PMID: 38882397 PMCID: PMC11176808 DOI: 10.1016/j.mbplus.2024.100151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 04/22/2024] [Accepted: 05/18/2024] [Indexed: 06/18/2024] Open
Abstract
Extracellular matrix (ECM) fabricated using human induced pluripotent stem cells (hiPSCs)-derived cardiac fibroblasts (hiPSC-CFs) could serve as a completely biological scaffold for an engineered cardiac patch, leveraging the unlimited source and outstanding reproducibility of hiPSC-CFs. Additionally, hiPSC-CF-derived ECM (hiPSC-CF-ECM) holds the potential to enhance maturation of exogenous cardiomyocytes, such as hiPSC-derived cardiomyocytes (hiPSC-CMs), by providing a microenvironment rich in cardiac-specific biochemical and signaling cues. However, achieving sufficient robustness of hiPSC-CF-ECM is challenging. This study aims to achieve appropriate ECM deposition, scaffold thickness, and mechanical strength of an aligned hiPSC-CF-ECM by optimizing the culture period, ranging from 2 to 10 weeks, of hiPSC-CFs grown on micro-grated substrates, which can direct the alignment of both hiPSC-CFs and their secreted ECM. The hiPSC-CFs demonstrated a production rate of 13.5 µg ECM per day per 20,000 cells seeded. An anisotropic nanofibrous hiPSC-CF-ECM scaffold with a thickness of 20.0 ± 2.1 µm was achieved after 6 weeks of culture, followed by decellularization. Compositional analysis through liquid chromatography-mass spectrometry (LC-MS) revealed the presence of cardiac-specific fibrillar collagens, non-fibrillar collagens, and matricellular proteins. Uniaxial tensile stretching of the hiPSC-CF-ECM scaffold indicated robust tensile resilience. Finally, hiPSCs-CMs cultured on the hiPSC-CF-ECM exhibited alignment following the guidance of ECM nanofibers and demonstrated mature organization of key structural proteins. The culture duration of the anisotropic hiPSC-CF-ECM was successfully refined to achieve a robust scaffold containing structural proteins that resembles cardiac microenvironment. This completely biological, anisotropic, and cardiac-specific ECM holds great potential for cardiac patch engineering.
Collapse
Affiliation(s)
- Te-An Chen
- Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Brandon B. Zhao
- Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Richard A. Balbin
- Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Sameeksha Sharma
- Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Donggi Ha
- Department of Mechanical Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Timothy J. Kamp
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
- Stem Cell and Regenerative Medicine Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Yuxiao Zhou
- Department of Mechanical Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Feng Zhao
- Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
11
|
Cyr JA, Burdett C, Pürstl JT, Thompson RP, Troughton SC, Sinha S, Best SM, Cameron RE. Characterizing collagen scaffold compliance with native myocardial strains using an ex-vivo cardiac model: The physio-mechanical influence of scaffold architecture and attachment method. Acta Biomater 2024; 184:239-253. [PMID: 38942187 DOI: 10.1016/j.actbio.2024.06.031] [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: 03/09/2024] [Revised: 05/30/2024] [Accepted: 06/21/2024] [Indexed: 06/30/2024]
Abstract
Applied to the epicardium in-vivo, regenerative cardiac patches support the ventricular wall, reduce wall stresses, encourage ventricular wall thickening, and improve ventricular function. Scaffold engraftment, however, remains a challenge. After implantation, scaffolds are subject to the complex, time-varying, biomechanical environment of the myocardium. The mechanical capacity of engineered tissue to biomimetically deform and simultaneously support the damaged native tissue is crucial for its efficacy. To date, however, the biomechanical response of engineered tissue applied directly to live myocardium has not been characterized. In this paper, we utilize optical imaging of a Langendorff ex-vivo cardiac model to characterize the native deformation of the epicardium as well as that of attached engineered scaffolds. We utilize digital image correlation, linear strain, and 2D principal strain analysis to assess the mechanical compliance of acellular ice templated collagen scaffolds. Scaffolds had either aligned or isotropic porous architecture and were adhered directly to the live epicardial surface with either sutures or cyanoacrylate glue. We demonstrate that the biomechanical characteristics of native myocardial deformation on the epicardial surface can be reproduced by an ex-vivo cardiac model. Furthermore, we identified that scaffolds with unidirectionally aligned pores adhered with suture fixation most accurately recapitulated the deformation of the native epicardium. Our study contributes a translational characterization methodology to assess the physio-mechanical performance of engineered cardiac tissue and adds to the growing body of evidence showing that anisotropic scaffold architecture improves the functional biomimetic capacity of engineered cardiac tissue. STATEMENT OF SIGNIFICANCE: Engineered cardiac tissue offers potential for myocardial repair, but engraftment remains a challenge. In-vivo, engineered scaffolds are subject to complex biomechanical stresses and the mechanical capacity of scaffolds to biomimetically deform is critical. To date, the biomechanical response of engineered scaffolds applied to live myocardium has not been characterized. In this paper, we utilize optical imaging of an ex-vivo cardiac model to characterize the deformation of the native epicardium and scaffolds attached directly to the heart. Comparing scaffold architecture and fixation method, we demonstrate that sutured scaffolds with anisotropic pores aligned with the native alignment of the superficial myocardium best recapitulate native deformation. Our study contributes a physio-mechanical characterization methodology for cardiac tissue engineering scaffolds.
Collapse
Affiliation(s)
- Jamie A Cyr
- Department of Materials Science & Metallurgy Cambridge University 27 Charles Babbage Road, Cambridge CB3 0FS, UK.
| | - Clare Burdett
- Department of Materials Science & Metallurgy Cambridge University 27 Charles Babbage Road, Cambridge CB3 0FS, UK
| | - Julia T Pürstl
- Department of Materials Science & Metallurgy Cambridge University 27 Charles Babbage Road, Cambridge CB3 0FS, UK
| | - Robert P Thompson
- Department of Materials Science & Metallurgy Cambridge University 27 Charles Babbage Road, Cambridge CB3 0FS, UK
| | - Samuel C Troughton
- Department of Materials Science & Metallurgy Cambridge University 27 Charles Babbage Road, Cambridge CB3 0FS, UK
| | - Sanjay Sinha
- Wellcome-MRC Cambridge Stem Cell Institute, Cambridge University, Jeffrey Cheah Biomedical Centre, Puddicombe Way, Cambridge CB2 0AW, UK
| | - Serena M Best
- Department of Materials Science & Metallurgy Cambridge University 27 Charles Babbage Road, Cambridge CB3 0FS, UK.
| | - Ruth E Cameron
- Department of Materials Science & Metallurgy Cambridge University 27 Charles Babbage Road, Cambridge CB3 0FS, UK.
| |
Collapse
|
12
|
Garibyan M, Hoffman T, Makaske T, Do SK, Wu Y, Williams BA, March AR, Cho N, Pedroncelli N, Lima RE, Soto J, Jackson B, Santoso JW, Khademhosseini A, Thomson M, Li S, McCain ML, Morsut L. Engineering programmable material-to-cell pathways via synthetic notch receptors to spatially control differentiation in multicellular constructs. Nat Commun 2024; 15:5891. [PMID: 39003263 PMCID: PMC11246427 DOI: 10.1038/s41467-024-50126-1] [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: 06/16/2023] [Accepted: 07/02/2024] [Indexed: 07/15/2024] Open
Abstract
Synthetic Notch (synNotch) receptors are genetically encoded, modular synthetic receptors that enable mammalian cells to detect environmental signals and respond by activating user-prescribed transcriptional programs. Although some materials have been modified to present synNotch ligands with coarse spatial control, applications in tissue engineering generally require extracellular matrix (ECM)-derived scaffolds and/or finer spatial positioning of multiple ligands. Thus, we develop here a suite of materials that activate synNotch receptors for generalizable engineering of material-to-cell signaling. We genetically and chemically fuse functional synNotch ligands to ECM proteins and ECM-derived materials. We also generate tissues with microscale precision over four distinct reporter phenotypes by culturing cells with two orthogonal synNotch programs on surfaces microcontact-printed with two synNotch ligands. Finally, we showcase applications in tissue engineering by co-transdifferentiating fibroblasts into skeletal muscle or endothelial cell precursors in user-defined micropatterns. These technologies provide avenues for spatially controlling cellular phenotypes in mammalian tissues.
Collapse
Affiliation(s)
- Mher Garibyan
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA
- Eli and Edythe Broad Center, University of Southern California, Los Angeles, CA, 90033, USA
- Alfred E. Mann Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA, USA
| | - Tyler Hoffman
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, USA
| | - Thijs Makaske
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA
- Eli and Edythe Broad Center, University of Southern California, Los Angeles, CA, 90033, USA
- Utrecht University in the lab of Prof. Dr. Lukas Kapitein, Los Angeles, CA, 90024, USA
| | - Stephanie K Do
- Alfred E. Mann Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA, USA
| | - Yifan Wu
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, USA
| | - Brian A Williams
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Alexander R March
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA
- Eli and Edythe Broad Center, University of Southern California, Los Angeles, CA, 90033, USA
| | - Nathan Cho
- Alfred E. Mann Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA, USA
| | - Nicolas Pedroncelli
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, USA
| | - Ricardo Espinosa Lima
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, USA
| | - Jennifer Soto
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, USA
| | - Brooke Jackson
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, USA
| | - Jeffrey W Santoso
- Alfred E. Mann Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA, USA
| | - Ali Khademhosseini
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, USA
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA, 90024, USA
| | - Matt Thomson
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Song Li
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, USA
- Broad Stem Cell Center, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Megan L McCain
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA.
- Alfred E. Mann Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA, USA.
| | - Leonardo Morsut
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA.
- Eli and Edythe Broad Center, University of Southern California, Los Angeles, CA, 90033, USA.
- Alfred E. Mann Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
13
|
Morita T, Nie M, Takeuchi S. Human induced pluripotent stem cell-derived cardiac muscle rings for biohybrid self-beating actuator. LAB ON A CHIP 2024; 24:3377-3387. [PMID: 38916038 DOI: 10.1039/d4lc00276h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Cardiac muscle, a subtype of striated muscle composing our heart, has garnered attention as a source of autonomously driven actuators due to its inherent capability for spontaneous contraction. However, conventional cardiac biohybrid robots have utilized planar (2D) cardiac tissue consisting of a thin monolayer of cardiac myotubes with a thickness of 3-5 μm, which can generate a limited contractile force per unit footprint. In this study, 3D cardiac muscle rings were proposed as robotic actuator units. These units not only exhibit higher contractile force per unit footprint compared to their 2D counterparts due to their increased height, but they can also be integrated into desired 3D configurations. We fabricated cardiac muscle rings from human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs), evaluated their driving characteristics, and verified the actuation effects by integrating them with artificial components. After the 10th day from culture, the cardiac muscle rings exhibited rhythmic spontaneous contraction and increased contractile force in response to stretching stimuli. Furthermore, after constructing a centimeter-sized biohybrid self-beating actuator with an antagonistic pair structure of cardiac muscle rings, the periodic antagonistic beating motion at its tail portion was confirmed. We believe that 3D cardiac muscle rings, possessing high contractile force and capable of being positioned within limited 3D space, can be used as potent biohybrid robotic actuators.
Collapse
Affiliation(s)
- Tomohiro Morita
- Department of Mechano-Informatics, Graduate School of Information Science and Technology, The University of Tokyo, 113-8656, Japan.
| | - Minghao Nie
- Department of Mechano-Informatics, Graduate School of Information Science and Technology, The University of Tokyo, 113-8656, Japan.
| | - Shoji Takeuchi
- Department of Mechano-Informatics, Graduate School of Information Science and Technology, The University of Tokyo, 113-8656, Japan.
- Institude of Industrial Science (IIS), The University of Tokyo, 153-8505, Japan
- International Research Center for Neurointelligence (WPI-IRCN), the University of Tokyo Institutes for Advanced Study (UTIAS), 113-0033, Japan
| |
Collapse
|
14
|
Taylor A, Xu J, Rogozinski N, Fu H, Molina Cortez L, McMahan S, Perez K, Chang Y, Pan Z, Yang H, Liao J, Hong Y. Reduced Graphene-Oxide-Doped Elastic Biodegradable Polyurethane Fibers for Cardiomyocyte Maturation. ACS Biomater Sci Eng 2024; 10:3759-3774. [PMID: 38800901 DOI: 10.1021/acsbiomaterials.3c01908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Conductive biomaterials offer promising solutions to enhance the maturity of cultured cardiomyocytes. While the conventional culture of cardiomyocytes on nonconductive materials leads to more immature characteristics, conductive microenvironments have the potential to support sarcomere development, gap junction formation, and beating of cardiomyocytes in vitro. In this study, we systematically investigated the behaviors of cardiomyocytes on aligned electrospun fibrous membranes composed of elastic and biodegradable polyurethane (PU) doped with varying concentrations of reduced graphene oxide (rGO). Compared to PU and PU-4%rGO membranes, the PU-10%rGO membrane exhibited the highest conductivity, approaching levels close to those of native heart tissue. The PU-rGO membranes retained anisotropic viscoelastic behavior similar to that of the porcine left ventricle and a superior tensile strength. Neonatal rat cardiomyocytes (NRCMs) and human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) on the PU-rGO membranes displayed enhanced maturation with cell alignment and enhanced sarcomere structure and gap junction formation with PU-10%rGO having the most improved sarcomere structure and CX-43 presence. hiPSC-CMs on the PU-rGO membranes exhibited a uniform and synchronous beating pattern compared with that on PU membranes. Overall, PU-10%rGO exhibited the best performance for cardiomyocyte maturation. The conductive PU-rGO membranes provide a promising matrix for in vitro cardiomyocyte culture with promoted cell maturation/functionality and the potential for cardiac disease treatment.
Collapse
Affiliation(s)
- Alan Taylor
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas 76019, United States
| | - Jiazhu Xu
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas 76019, United States
| | - Nicholas Rogozinski
- Department of Biomedical Engineering, University of North Texas, Denton, Texas 76207, United States
| | - Huikang Fu
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas 76019, United States
| | - Lia Molina Cortez
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas 76019, United States
| | - Sara McMahan
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas 76019, United States
| | - Karla Perez
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas 76019, United States
| | - Yan Chang
- Department of Graduate Nursing, University of Texas at Arlington, Arlington, Texas 76010, United States
| | - Zui Pan
- Department of Graduate Nursing, University of Texas at Arlington, Arlington, Texas 76010, United States
| | - Huaxiao Yang
- Department of Biomedical Engineering, University of North Texas, Denton, Texas 76207, United States
| | - Jun Liao
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas 76019, United States
| | - Yi Hong
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas 76019, United States
| |
Collapse
|
15
|
Yao ZF, Kuang Y, Wu HT, Lundqvist E, Fu X, Celt N, Pei J, Yee A, Ardoña HAM. Selective Induction of Molecular Assembly to Tissue-Level Anisotropy on Peptide-Based Optoelectronic Cardiac Biointerfaces. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2312231. [PMID: 38335948 PMCID: PMC11126358 DOI: 10.1002/adma.202312231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 01/19/2024] [Indexed: 02/12/2024]
Abstract
The conduction efficiency of ions in excitable tissues and of charged species in organic conjugated materials both benefit from having ordered domains and anisotropic pathways. In this study, a photocurrent-generating cardiac biointerface is presented, particularly for investigating the sensitivity of cardiomyocytes to geometrically comply to biomacromolecular cues differentially assembled on a conductive nanogrooved substrate. Through a polymeric surface-templated approach, photoconductive substrates with symmetric peptide-quaterthiophene (4T)-peptide units assembled as 1D nanostructures on nanoimprinted polyalkylthiophene (P3HT) surface are developed. The 4T-based peptides studied here can form 1D nanostructures on prepatterned polyalkylthiophene substrates, as directed by hydrogen bonding, aromatic interactions between 4T and P3HT, and physical confinement on the nanogrooves. It is observed that smaller 4T-peptide units that can achieve a higher degree of assembly order within the polymeric templates serve as a more efficient driver of cardiac cytoskeletal anisotropy than merely presenting aligned -RGD bioadhesive epitopes on a nanotopographic surface. These results unravel some insights on how cardiomyocytes perceive submicrometer dimensionality, local molecular order, and characteristics of surface cues in their immediate environment. Overall, the work offers a cardiac patterning platform that presents the possibility of a gene modification-free cardiac photostimulation approach while controlling the conduction directionality of the biotic and abiotic components.
Collapse
Affiliation(s)
- Ze-Fan Yao
- Department of Chemical and Biomolecular Engineering, Samueli School of Engineering, University of California, Irvine, CA 92697, USA
- Department of Chemistry, School of Physical Sciences, University of California, Irvine, CA 92697, USA
| | - Yuyao Kuang
- Department of Chemical and Biomolecular Engineering, Samueli School of Engineering, University of California, Irvine, CA 92697, USA
| | - Hao-Tian Wu
- Beijing National Laboratory for Molecular Sciences (BNLMS), Key Laboratory of Polymer Chemistry and Physics of Ministry of Education, Center of Soft Matter Science and Engineering, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Emil Lundqvist
- Department of Biomedical Engineering, Samueli School of Engineering, University of California, Irvine, CA 92697, USA
| | - Xin Fu
- Department of Materials Science and Engineering, Samueli School of Engineering, University of California, Irvine, CA 92697, USA
| | - Natalie Celt
- Department of Biomedical Engineering, Samueli School of Engineering, University of California, Irvine, CA 92697, USA
| | - Jian Pei
- Beijing National Laboratory for Molecular Sciences (BNLMS), Key Laboratory of Polymer Chemistry and Physics of Ministry of Education, Center of Soft Matter Science and Engineering, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Albert Yee
- Department of Chemical and Biomolecular Engineering, Samueli School of Engineering, University of California, Irvine, CA 92697, USA
| | - Herdeline Ann M. Ardoña
- Department of Chemical and Biomolecular Engineering, Samueli School of Engineering, University of California, Irvine, CA 92697, USA
- Department of Chemistry, School of Physical Sciences, University of California, Irvine, CA 92697, USA
- Department of Biomedical Engineering, Samueli School of Engineering, University of California, Irvine, CA 92697, USA
- Sue & Bill Gross Stem Cell Research Center, University of California, Irvine, CA 92697, USA
| |
Collapse
|
16
|
Jilberto J, DePalma SJ, Lo J, Kobeissi H, Quach L, Lejeune E, Baker BM, Nordsletten D. A data-driven computational model for engineered cardiac microtissues. Acta Biomater 2023; 172:123-134. [PMID: 37879587 PMCID: PMC10938557 DOI: 10.1016/j.actbio.2023.10.025] [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: 06/01/2023] [Revised: 10/17/2023] [Accepted: 10/19/2023] [Indexed: 10/27/2023]
Abstract
Engineered heart tissues (EHTs) present a potential solution to some of the current challenges in the treatment of heart disease; however, the development of mature, adult-like cardiac tissues remains elusive. Mechanical stimuli have been observed to improve whole-tissue function and cardiomyocyte (CM) maturation, although our ability to fully utilize these mechanisms is hampered, in part, by our incomplete understanding of the mechanobiology of EHTs. In this work, we leverage experimental data, produced by a mechanically tunable experimental setup, to introduce a tissue-specific computational modeling pipeline of EHTs. Our new modeling pipeline generates simulated, image-based EHTs, capturing ECM and myofibrillar structure as well as functional parameters estimated directly from experimental data. This approach enables the unique estimation of EHT function by data-based estimation of CM active stresses. We use this experimental and modeling pipeline to study different mechanical environments, where we contrast the force output of the tissue with the computed active stress of CMs. We show that the significant differences in measured experimental forces can largely be explained by the levels of myofibril formation achieved by the CMs in the distinct mechanical environments, with active stress showing more muted variations across conditions. The presented model also enables us to dissect the relative contributions of myofibrils and extracellular matrix to tissue force output, a task difficult to address experimentally. These results highlight the importance of tissue-specific modeling to augment EHT experiments, providing deeper insights into the mechanobiology driving EHT function. STATEMENT OF SIGNIFICANCE: Engineered heart tissues (EHTs) have the potential to revolutionize the way heart disease is treated. However, developing mature cardiomyocytes (CM) in these tissues remains a challenge due, in part, to our incomplete understanding of the fundamental biomechanical mechanisms that drive EHT development. This work integrates the experimental data of an EHT platform developed to study the influence of mechanics in CM maturation with computational biomechanical models. This approach is used to augment conclusions obtained in-vitro - by measuring quantities such as cell stress and strain - and to dissect the relevance of each component in the whole tissue performance. Our results show how a combination of specialized in-silico and in-vitro approaches can help us better understand the mechanobiology of EHTs.
Collapse
Affiliation(s)
- Javiera Jilberto
- Department of Biomedical Engineering, University of Michigan, MI, USA.
| | - Samuel J DePalma
- Department of Biomedical Engineering, University of Michigan, MI, USA
| | - Jason Lo
- Department of Biomedical Engineering, University of Michigan, MI, USA
| | - Hiba Kobeissi
- Department of Mechanical Engineering, Boston University, MA, USA
| | - Lani Quach
- Department of Biomedical Engineering, University of Michigan, MI, USA
| | - Emma Lejeune
- Department of Mechanical Engineering, Boston University, MA, USA
| | - Brendon M Baker
- Department of Biomedical Engineering, University of Michigan, MI, USA
| | - David Nordsletten
- Department of Biomedical Engineering, University of Michigan, MI, USA; Department of Cardiac Surgery, University of Michigan, MI, USA; Department of Biomedical Engineering, School of Imaging Sciences and Biomedical Engineering, King's College London, London, UK
| |
Collapse
|
17
|
Cyr JA, Colzani M, Bayraktar S, Köhne M, Bax DV, Graup V, Farndale R, Sinha S, Best SM, Cameron RE. Extracellular macrostructure anisotropy improves cardiac tissue-like construct function and phenotypic cellular maturation. BIOMATERIALS ADVANCES 2023; 155:213680. [PMID: 37944449 DOI: 10.1016/j.bioadv.2023.213680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 10/02/2023] [Accepted: 10/30/2023] [Indexed: 11/12/2023]
Abstract
Regenerative cardiac tissue is a promising field of study with translational potential as a therapeutic option for myocardial repair after injury, however, poor electrical and contractile function has limited translational utility. Emerging research suggests scaffolds that recapitulate the structure of the native myocardium improve physiological function. Engineered cardiac constructs with anisotropic extracellular architecture demonstrate improved tissue contractility, signaling synchronicity, and cellular organization when compared to constructs with reduced architectural order. The complexity of scaffold fabrication, however, limits isolated variation of individual structural and mechanical characteristics. Thus, the isolated impact of scaffold macroarchitecture on tissue function is poorly understood. Here, we produce isotropic and aligned collagen scaffolds seeded with embryonic stem cell derived cardiomyocytes (hESC-CM) while conserving all confounding physio-mechanical features to independently assess the effects of macroarchitecture on tissue function. We quantified spatiotemporal tissue function through calcium signaling and contractile strain. We further examined intercellular organization and intracellular development. Aligned tissue constructs facilitated improved signaling synchronicity and directional contractility as well as dictated uniform cellular alignment. Cells on aligned constructs also displayed phenotypic and genetic markers of increased maturity. Our results isolate the influence of scaffold macrostructure on tissue function and inform the design of optimized cardiac tissue for regenerative and model medical systems.
Collapse
Affiliation(s)
- Jamie A Cyr
- Department of Materials Science & Metallurgy, Cambridge University, 27 Charles Babbage Road, Cambridge CB3 0FS, UK
| | - Maria Colzani
- Wellcome-MRC Cambridge Stem Cell Institute, Cambridge University, Jeffrey Cheah Biomedical Centre, Puddicombe Way, Cambridge CB2 0AW, UK
| | - Semih Bayraktar
- Wellcome-MRC Cambridge Stem Cell Institute, Cambridge University, Jeffrey Cheah Biomedical Centre, Puddicombe Way, Cambridge CB2 0AW, UK
| | - Maria Köhne
- Wellcome-MRC Cambridge Stem Cell Institute, Cambridge University, Jeffrey Cheah Biomedical Centre, Puddicombe Way, Cambridge CB2 0AW, UK
| | - Daniel V Bax
- Department of Materials Science & Metallurgy, Cambridge University, 27 Charles Babbage Road, Cambridge CB3 0FS, UK
| | - Vera Graup
- Department of Materials Science & Metallurgy, Cambridge University, 27 Charles Babbage Road, Cambridge CB3 0FS, UK
| | - Richard Farndale
- Department of Biochemistry, Cambridge University, Hopkins Building Tennis Court Road, Cambridge CB2 1QW, UK
| | - Sanjay Sinha
- Wellcome-MRC Cambridge Stem Cell Institute, Cambridge University, Jeffrey Cheah Biomedical Centre, Puddicombe Way, Cambridge CB2 0AW, UK.
| | - Serena M Best
- Department of Materials Science & Metallurgy, Cambridge University, 27 Charles Babbage Road, Cambridge CB3 0FS, UK.
| | - Ruth E Cameron
- Department of Materials Science & Metallurgy, Cambridge University, 27 Charles Babbage Road, Cambridge CB3 0FS, UK.
| |
Collapse
|
18
|
Venturino I, Vurro V, Bonfadini S, Moschetta M, Perotto S, Sesti V, Criante L, Bertarelli C, Lanzani G. Skeletal muscle cells opto-stimulation by intramembrane molecular transducers. Commun Biol 2023; 6:1148. [PMID: 37952040 PMCID: PMC10640616 DOI: 10.1038/s42003-023-05538-y] [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: 04/21/2023] [Accepted: 11/02/2023] [Indexed: 11/14/2023] Open
Abstract
Optical stimulation and control of muscle cell contraction opens up a number of interesting applications in hybrid robotic and medicine. Here we show that recently designed molecular phototransducer can be used to stimulate C2C12 skeletal muscle cells, properly grown to exhibit collective behaviour. C2C12 is a skeletal muscle cell line that does not require animal sacrifice Furthermore, it is an ideal cell model for evaluating the phototransducer pacing ability due to its negligible spontaneous activity. We study the stimulation process and analyse the distribution of responses in multinuclear cells, in particular looking at the consistency between stimulus and contraction. Contractions are detected by using an imaging software for object recognition. We find a deterministic response to light stimuli, yet with a certain distribution of erratic behaviour that is quantified and correlated to light intensity or stimulation frequency. Finally, we compare our optical stimulation with electrical stimulation showing advantages of the optical approach, like the reduced cell stress.
Collapse
Affiliation(s)
- Ilaria Venturino
- Dipartimento di Fisica, Politecnico di Milano, Milano, Italy
- Center for Nano Science and Technology, Istituto Italiano di Tecnologia, Milano, Italy
| | - Vito Vurro
- Center for Nano Science and Technology, Istituto Italiano di Tecnologia, Milano, Italy
| | - Silvio Bonfadini
- Center for Nano Science and Technology, Istituto Italiano di Tecnologia, Milano, Italy
| | - Matteo Moschetta
- Center for Nano Science and Technology, Istituto Italiano di Tecnologia, Milano, Italy
| | - Sara Perotto
- Center for Nano Science and Technology, Istituto Italiano di Tecnologia, Milano, Italy
| | - Valentina Sesti
- Center for Nano Science and Technology, Istituto Italiano di Tecnologia, Milano, Italy
- Dipartimento di Chimica, Materiali e Ingegneria Chimica "Giulio Natta" Politecnico di Milano, Milano, Italy
| | - Luigino Criante
- Center for Nano Science and Technology, Istituto Italiano di Tecnologia, Milano, Italy
| | - Chiara Bertarelli
- Center for Nano Science and Technology, Istituto Italiano di Tecnologia, Milano, Italy
- Dipartimento di Chimica, Materiali e Ingegneria Chimica "Giulio Natta" Politecnico di Milano, Milano, Italy
| | - Guglielmo Lanzani
- Dipartimento di Fisica, Politecnico di Milano, Milano, Italy.
- Center for Nano Science and Technology, Istituto Italiano di Tecnologia, Milano, Italy.
| |
Collapse
|
19
|
Bernava G, Iop L. Advances in the design, generation, and application of tissue-engineered myocardial equivalents. Front Bioeng Biotechnol 2023; 11:1247572. [PMID: 37811368 PMCID: PMC10559975 DOI: 10.3389/fbioe.2023.1247572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 08/29/2023] [Indexed: 10/10/2023] Open
Abstract
Due to the limited regenerative ability of cardiomyocytes, the disabling irreversible condition of myocardial failure can only be treated with conservative and temporary therapeutic approaches, not able to repair the damage directly, or with organ transplantation. Among the regenerative strategies, intramyocardial cell injection or intravascular cell infusion should attenuate damage to the myocardium and reduce the risk of heart failure. However, these cell delivery-based therapies suffer from significant drawbacks and have a low success rate. Indeed, cardiac tissue engineering efforts are directed to repair, replace, and regenerate native myocardial tissue function. In a regenerative strategy, biomaterials and biomimetic stimuli play a key role in promoting cell adhesion, proliferation, differentiation, and neo-tissue formation. Thus, appropriate biochemical and biophysical cues should be combined with scaffolds emulating extracellular matrix in order to support cell growth and prompt favorable cardiac microenvironment and tissue regeneration. In this review, we provide an overview of recent developments that occurred in the biomimetic design and fabrication of cardiac scaffolds and patches. Furthermore, we sift in vitro and in situ strategies in several preclinical and clinical applications. Finally, we evaluate the possible use of bioengineered cardiac tissue equivalents as in vitro models for disease studies and drug tests.
Collapse
Affiliation(s)
| | - Laura Iop
- Department of Cardiac Thoracic Vascular Sciences and Public Health, Padua Medical School, University of Padua, Padua, Italy
| |
Collapse
|
20
|
Tan Y, Coyle RC, Barrs RW, Silver SE, Li M, Richards DJ, Lin Y, Jiang Y, Wang H, Menick DR, Deleon-Pennell K, Tian B, Mei Y. Nanowired human cardiac organoid transplantation enables highly efficient and effective recovery of infarcted hearts. SCIENCE ADVANCES 2023; 9:eadf2898. [PMID: 37540743 PMCID: PMC10403216 DOI: 10.1126/sciadv.adf2898] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 07/06/2023] [Indexed: 08/06/2023]
Abstract
Human cardiac organoids hold remarkable potential for cardiovascular disease modeling and human pluripotent stem cell-derived cardiomyocyte (hPSC-CM) transplantation. Here, we show cardiac organoids engineered with electrically conductive silicon nanowires (e-SiNWs) significantly enhance the therapeutic efficacy of hPSC-CMs to treat infarcted hearts. We first demonstrated the biocompatibility of e-SiNWs and their capacity to improve cardiac microtissue engraftment in healthy rat myocardium. Nanowired human cardiac organoids were then engineered with hPSC-CMs, nonmyocyte supporting cells, and e-SiNWs. Nonmyocyte supporting cells promoted greater ischemia tolerance of cardiac organoids, and e-SiNWs significantly improved electrical pacing capacity. After transplantation into ischemia/reperfusion-injured rat hearts, nanowired cardiac organoids significantly improved contractile development of engrafted hPSC-CMs, induced potent cardiac functional recovery, and reduced maladaptive left ventricular remodeling. Compared to contemporary studies with an identical injury model, greater functional recovery was achieved with a 20-fold lower dose of hPSC-CMs, revealing therapeutic synergy between conductive nanomaterials and human cardiac organoids for efficient heart repair.
Collapse
Affiliation(s)
- Yu Tan
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
| | - Robert C. Coyle
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
| | - Ryan W. Barrs
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
| | - Sophia E. Silver
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
| | - Mei Li
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
| | - Dylan J. Richards
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
| | - Yiliang Lin
- Department of Chemistry, The James Franck Institute and the Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL 60637, USA
| | - Yuanwen Jiang
- Department of Chemistry, The James Franck Institute and the Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL 60637, USA
| | - Hongjun Wang
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Donald R. Menick
- Division of Cardiology, Department of Medicine, Gazes Cardiac Research Institute, Ralph H. Johnson Veterans Affairs Medical Center, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Kristine Deleon-Pennell
- Division of Cardiology, Department of Medicine, Gazes Cardiac Research Institute, Ralph H. Johnson Veterans Affairs Medical Center, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Bozhi Tian
- Department of Chemistry, The James Franck Institute and the Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL 60637, USA
| | - Ying Mei
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
21
|
House A, Cornick J, Butt Q, Guvendiren M. Elastomeric platform with surface wrinkling patterns to control cardiac cell alignment. J Biomed Mater Res A 2023; 111:1228-1242. [PMID: 36762538 DOI: 10.1002/jbm.a.37511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 12/07/2022] [Accepted: 01/29/2023] [Indexed: 02/11/2023]
Abstract
There is a growing interest in creating 2D cardiac tissue models that display native extracellular matrix (ECM) cues of the heart tissue. Cellular alignment alone is known to be a crucial cue for cardiac tissue development by regulating cell-cell and cell-ECM interactions. In this study, we report a simple and robust approach to create lamellar surface wrinkling patterns enabling spatial control of pattern dimensions with a wide range of pattern amplitude (A ≈ 2-55 μm) and wavelength (λ ≈ 35-100 μm). For human cardiomyocytes (hCMs) and human cardiac fibroblasts (hCFs), our results indicate that the degree of cellular alignment and pattern recognition are correlated with pattern A and λ. We also demonstrate fabrication of devices composed of micro-well arrays with user-defined lamellar patterns on the bottom surface of each well for high-throughput screening studies. Results from a screening study indicate that cellular alignment is strongly diminished with increasing seeding density. In another study, we show our ability to vary hCM/hCF seeding ratio for each well to create co-culture systems where seeding ratio is independent of cellular alignment.
Collapse
Affiliation(s)
- Andrew House
- Otto H. York Department of Chemical and Materials Engineering, New Jersey Institute of Technology, Newark, New Jersey, USA
| | - Jason Cornick
- Otto H. York Department of Chemical and Materials Engineering, New Jersey Institute of Technology, Newark, New Jersey, USA
| | - Quratulain Butt
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, New Jersey, USA
| | - Murat Guvendiren
- Otto H. York Department of Chemical and Materials Engineering, New Jersey Institute of Technology, Newark, New Jersey, USA
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, New Jersey, USA
| |
Collapse
|
22
|
Shang Y, Liu R, Gan J, Yang Y, Sun L. Construction of cardiac fibrosis for biomedical research. SMART MEDICINE 2023; 2:e20230020. [PMID: 39188350 PMCID: PMC11235890 DOI: 10.1002/smmd.20230020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 07/22/2023] [Indexed: 08/28/2024]
Abstract
Cardiac remodeling is critical for effective tissue recuperation, nevertheless, excessive formation and deposition of extracellular matrix components can result in the onset of cardiac fibrosis. Despite the emergence of novel therapies, there are still no lifelong therapeutic solutions for this issue. Understanding the detrimental cardiac remodeling may aid in the development of innovative treatment strategies to prevent or reverse fibrotic alterations in the heart. Further combining the latest understanding of disease pathogenesis with cardiac tissue engineering has provided the conversion of basic laboratory studies into the therapy of cardiac fibrosis patients as an increasingly viable prospect. This review presents the current main mechanisms and the potential tissue engineering of cardiac fibrosis. Approaches using biomedical materials-based cardiac constructions are reviewed to consider key issues for simulating in vitro cardiac fibrosis, outlining a future perspective for preclinical applications.
Collapse
Affiliation(s)
- Yixuan Shang
- Department of Medical Supplies SupportNanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
| | - Rui Liu
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
| | - Jingjing Gan
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
| | - Yuzhi Yang
- Department of Medical Supplies SupportNanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
| | - Lingyun Sun
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
| |
Collapse
|
23
|
Liu L, Xu F, Jin H, Qiu B, Yang J, Zhang W, Gao Q, Lin B, Chen S, Sun D. Integrated Manufacturing of Suspended and Aligned Nanofibrous Scaffold for Structural Maturation and Synchronous Contraction of HiPSC-Derived Cardiomyocytes. Bioengineering (Basel) 2023; 10:702. [PMID: 37370633 DOI: 10.3390/bioengineering10060702] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 05/30/2023] [Accepted: 06/02/2023] [Indexed: 06/29/2023] Open
Abstract
Electrospun nanofiber constructs represent a promising alternative for mimicking the natural extracellular matrix in vitro and have significant potential for cardiac patch applications. While the effect of fiber orientation on the morphological structure of cardiomyocytes has been investigated, fibers only provide contact guidance without accounting for substrate stiffness due to their deposition on rigid substrates (e.g., glass or polystyrene). This paper introduces an in situ fabrication method for suspended and well aligned nanofibrous scaffolds via roller electrospinning, providing an anisotropic microenvironment with reduced stiffness for cardiac tissue engineering. A fiber surface modification strategy, utilizing oxygen plasma treatment combined with sodium dodecyl sulfate solution, was proposed to maintain the hydrophilicity of polycaprolactone (PCL) fibers, promoting cellular adhesion. Human-induced pluripotent stem cell (hiPSC)-derived cardiomyocytes (CMs), cultured on aligned fibers, exhibited an elongated morphology with extension along the fiber axis. In comparison to Petri dishes and suspended random fiber scaffolds, hiPSC-CMs on suspended aligned fiber scaffolds demonstrated enhanced sarcomere organization, spontaneous synchronous contraction, and gene expression indicative of maturation. This work demonstrates the suspended and aligned nano-fibrous scaffold provides a more realistic biomimetic environment for hiPSC-CMs, which promoted further research on the inducing effect of fiber scaffolds on hiPSC-CMs microstructure and gene-level expression.
Collapse
Affiliation(s)
- Lingling Liu
- Sabondong Micron Nano Science and Technology Research Institute, Xiamen University, Xiamen 361102, China
| | - Feng Xu
- Sabondong Micron Nano Science and Technology Research Institute, Xiamen University, Xiamen 361102, China
| | - Hang Jin
- Sabondong Micron Nano Science and Technology Research Institute, Xiamen University, Xiamen 361102, China
| | - Bin Qiu
- Sabondong Micron Nano Science and Technology Research Institute, Xiamen University, Xiamen 361102, China
| | - Jianhui Yang
- Sabondong Micron Nano Science and Technology Research Institute, Xiamen University, Xiamen 361102, China
| | - Wangzihan Zhang
- Sabondong Micron Nano Science and Technology Research Institute, Xiamen University, Xiamen 361102, China
| | - Qiang Gao
- Department of Cardiovascular Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangzhou 510080, China
- Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou 510080, China
| | - Bin Lin
- Guangdong Beating Origin Regenerative Medicine Co., Ltd., Foshan 528231, China
| | - Songyue Chen
- Sabondong Micron Nano Science and Technology Research Institute, Xiamen University, Xiamen 361102, China
| | - Daoheng Sun
- Sabondong Micron Nano Science and Technology Research Institute, Xiamen University, Xiamen 361102, China
| |
Collapse
|
24
|
Vurro V, Shani K, Ardoña HAM, Zimmerman JF, Sesti V, Lee KY, Jin Q, Bertarelli C, Parker KK, Lanzani G. Light-triggered cardiac microphysiological model. APL Bioeng 2023; 7:026108. [PMID: 37234844 PMCID: PMC10208677 DOI: 10.1063/5.0143409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 04/24/2023] [Indexed: 05/28/2023] Open
Abstract
Light is recognized as an accurate and noninvasive tool for stimulating excitable cells. Here, we report on a non-genetic approach based on organic molecular phototransducers that allows wiring- and electrode-free tissue modulation. As a proof of concept, we show photostimulation of an in vitro cardiac microphysiological model mediated by an amphiphilic azobenzene compound that preferentially dwells in the cell membrane. Exploiting this optical based stimulation technology could be a disruptive approach for highly resolved cardiac tissue stimulation.
Collapse
Affiliation(s)
- V. Vurro
- Center for Nanoscience and Technology, Istituto Italiano di Teconologia, Milano, 20133 Italy
| | - K. Shani
- Disease Biophysics Group, John A. Paulson School of Engineering and Applied Science, Harvard University, Boston, Massachusetts 02134, USA
| | | | - J. F. Zimmerman
- Disease Biophysics Group, John A. Paulson School of Engineering and Applied Science, Harvard University, Boston, Massachusetts 02134, USA
| | | | | | - Q. Jin
- Disease Biophysics Group, John A. Paulson School of Engineering and Applied Science, Harvard University, Boston, Massachusetts 02134, USA
| | | | - K. K. Parker
- Disease Biophysics Group, John A. Paulson School of Engineering and Applied Science, Harvard University, Boston, Massachusetts 02134, USA
| | - G. Lanzani
- Author to whom correspondence should be addressed:
| |
Collapse
|
25
|
Mostert D, Groenen B, Klouda L, Passier R, Goumans MJ, Kurniawan NA, Bouten CVC. Human pluripotent stem cell-derived cardiomyocytes align under cyclic strain when guided by cardiac fibroblasts. APL Bioeng 2022; 6:046108. [PMID: 36567768 PMCID: PMC9771596 DOI: 10.1063/5.0108914] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 11/23/2022] [Indexed: 12/24/2022] Open
Abstract
The myocardium is a mechanically active tissue typified by anisotropy of the resident cells [cardiomyocytes (CMs) and cardiac fibroblasts (cFBs)] and the extracellular matrix (ECM). Upon ischemic injury, the anisotropic tissue is replaced by disorganized scar tissue, resulting in loss of coordinated contraction. Efforts to re-establish tissue anisotropy in the injured myocardium are hampered by a lack of understanding of how CM and/or cFB structural organization is affected by the two major physical cues inherent in the myocardium: ECM organization and cyclic mechanical strain. Herein, we investigate the singular and combined effect of ECM (dis)organization and cyclic strain in a two-dimensional human in vitro co-culture model of the myocardial microenvironment. We show that (an)isotropic ECM protein patterning can guide the orientation of CMs and cFBs, both in mono- and co-culture. Subsequent application of uniaxial cyclic strain-mimicking the local anisotropic deformation of beating myocardium-causes no effect when applied parallel to the anisotropic ECM. However, when cultured on isotropic substrates, cFBs, but not CMs, orient away from the direction of cyclic uniaxial strain (strain avoidance). In contrast, CMs show strain avoidance via active remodeling of their sarcomeres only when co-cultured with at least 30% cFBs. Paracrine signaling or N-cadherin-mediated communication between CMs and cFBs was no contributing factor. Our findings suggest that the mechanoresponsive cFBs provide structural guidance for CM orientation and elongation. Our study, therefore, highlights a synergistic mechanobiological interplay between CMs and cFBs in shaping tissue organization, which is of relevance for regenerating functionally organized myocardium.
Collapse
Affiliation(s)
| | - Bart Groenen
- Department of Biomedical Engineering, Eindhoven University of Technology, PO Box 513, 5600 MB Eindhoven, The Netherlands
| | - Leda Klouda
- Department of Biomedical Engineering, Eindhoven University of Technology, PO Box 513, 5600 MB Eindhoven, The Netherlands
| | | | - Marie-Jose Goumans
- Department of Cell and Chemical Biology and Center for Biomedical Genetics, Leiden University Medical Centre, Leiden, The Netherlands
| | | | | |
Collapse
|
26
|
Morris TA, Eldeen S, Tran RDH, Grosberg A. A comprehensive review of computational and image analysis techniques for quantitative evaluation of striated muscle tissue architecture. BIOPHYSICS REVIEWS 2022; 3:041302. [PMID: 36407035 PMCID: PMC9667907 DOI: 10.1063/5.0057434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 10/03/2022] [Indexed: 06/16/2023]
Abstract
Unbiased evaluation of morphology is crucial to understanding development, mechanics, and pathology of striated muscle tissues. Indeed, the ability of striated muscles to contract and the strength of their contraction is dependent on their tissue-, cellular-, and cytoskeletal-level organization. Accordingly, the study of striated muscles often requires imaging and assessing aspects of their architecture at multiple different spatial scales. While an expert may be able to qualitatively appraise tissues, it is imperative to have robust, repeatable tools to quantify striated myocyte morphology and behavior that can be used to compare across different labs and experiments. There has been a recent effort to define the criteria used by experts to evaluate striated myocyte architecture. In this review, we will describe metrics that have been developed to summarize distinct aspects of striated muscle architecture in multiple different tissues, imaged with various modalities. Additionally, we will provide an overview of metrics and image processing software that needs to be developed. Importantly to any lab working on striated muscle platforms, characterization of striated myocyte morphology using the image processing pipelines discussed in this review can be used to quantitatively evaluate striated muscle tissues and contribute to a robust understanding of the development and mechanics of striated muscles.
Collapse
Affiliation(s)
| | - Sarah Eldeen
- Center for Complex Biological Systems, University of California, Irvine, California 92697-2700, USA
| | | | | |
Collapse
|
27
|
Huebsch N. Collective organization from cellular disorder. Biophys J 2022; 121:4239-4241. [PMID: 36272406 PMCID: PMC9703033 DOI: 10.1016/j.bpj.2022.10.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 10/03/2022] [Accepted: 10/06/2022] [Indexed: 12/14/2022] Open
Affiliation(s)
- Nathaniel Huebsch
- Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, Missouri; NSF Science and Technology Center for Engineering Mechanobiology, Washington University in Saint Louis, Saint Louis, Missouri; Center for Cardiovascular Research, Center for Regenerative Medicine, Center for Investigation of Membrane Excitability Diseases, Washington University in Saint Louis, Saint Louis, Missouri.
| |
Collapse
|
28
|
Kałużna E, Nadel A, Zimna A, Rozwadowska N, Kolanowski T. Modeling the human heart ex vivo-current possibilities and strive for future applications. J Tissue Eng Regen Med 2022; 16:853-874. [PMID: 35748158 PMCID: PMC9796015 DOI: 10.1002/term.3335] [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: 11/16/2021] [Revised: 04/20/2022] [Accepted: 06/03/2022] [Indexed: 12/30/2022]
Abstract
The high organ specification of the human heart is inversely proportional to its functional recovery after damage. The discovery of induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) has accelerated research in human heart regeneration and physiology. Nevertheless, due to the immaturity of iPSC-CMs, they are far from being an representative model of the adult heart physiology. Therefore, number of laboratories strive to obtain a heart tissues by engineering methods by structuring iPSC-CMs into complex and advanced platforms. By using the iPSC-CMs and arranging them in 3D cultures it is possible to obtain a human heart muscle with physiological capabilities potentially similar to the adult heart, while remaining in vitro. Here, we attempt to describe existing examples of heart muscle either in vitro or ex vivo models and discuss potential options for the further development of such structures. This will be a crucial step for ultimate derivation of complete heart tissue-mimicking organs and their future use in drug development, therapeutic approaches testing, pre-clinical studies, and clinical applications. This review particularly aims to compile available models of advanced human heart tissue for scientists considering which model would best fit their research needs.
Collapse
Affiliation(s)
- Ewelina Kałużna
- Institute of Human GeneticsPolish Academy of SciencesPoznanPoland
| | - Agnieszka Nadel
- Institute of Human GeneticsPolish Academy of SciencesPoznanPoland
| | - Agnieszka Zimna
- Institute of Human GeneticsPolish Academy of SciencesPoznanPoland
| | | | | |
Collapse
|
29
|
Wang Y, Yu M, Hao K, Lei W, Tang M, Hu S. Cardiomyocyte Maturation-the Road is not Obstructed. Stem Cell Rev Rep 2022; 18:2966-2981. [PMID: 35788883 DOI: 10.1007/s12015-022-10407-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/30/2022] [Indexed: 12/29/2022]
Abstract
Human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) represent one of the most promising ways to treat cardiovascular diseases. High-purity cardiomyocytes (CM) from different cell sources could be obtained at present. However, the immature nature of these cardiomyocytes hinders its further clinical application. From immature to mature state, it involves structural, functional, and metabolic changes in cardiomyocytes. Generally, two types of culturing (2D and 3D) systems have been reported to induce cardiomyocyte maturation. 2D culture mainly achieves the maturation of cardiomyocytes through long-term culture, co-culture, supplementation of small molecule compounds, and the application of biophysical cues. The combined use of biomaterial's surface topography and biophysical cues also facilitates the maturation of cardiomyocytes. Cardiomyocyte maturation is a complex process involving many signaling pathways, and current methods fail to fully reproduce this process. Therefore, analyzing the signaling pathway network related to the maturation and producing hPSC-CMs with adult-like phenotype is a challenge. In this review, we summarized the structural and functional differences between hPSC-CMs and mature cardiomyocytes, and introduced various methods to induce cardiomyocyte maturation.
Collapse
Affiliation(s)
- Yaning Wang
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, 215000, China
| | - Miao Yu
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, 215000, China
| | - Kaili Hao
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, 215000, China
| | - Wei Lei
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, 215000, China
| | - Mingliang Tang
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, 215000, China.
| | - Shijun Hu
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, 215000, China.
| |
Collapse
|
30
|
Ghovvati M, Kharaziha M, Ardehali R, Annabi N. Recent Advances in Designing Electroconductive Biomaterials for Cardiac Tissue Engineering. Adv Healthc Mater 2022; 11:e2200055. [PMID: 35368150 PMCID: PMC9262872 DOI: 10.1002/adhm.202200055] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 03/12/2022] [Indexed: 12/19/2022]
Abstract
Implantable cardiac patches and injectable hydrogels are among the most promising therapies for cardiac tissue regeneration following myocardial infarction. Incorporating electrical conductivity into these patches and hydrogels is found to be an efficient method to improve cardiac tissue function. Conductive nanomaterials such as carbon nanotube, graphene oxide, gold nanorod, as well as conductive polymers such as polyaniline, polypyrrole, and poly(3,4-ethylenedioxythiophene):polystyrene sulfonate are appealing because they possess the electroconductive properties of semiconductors with ease of processing and have potential to restore electrical signaling propagation through the infarct area. Numerous studies have utilized these materials for regeneration of biological tissues that possess electrical activities, such as cardiac tissue. In this review, recent studies on the use of electroconductive materials for cardiac tissue engineering and their fabrication methods are summarized. Moreover, recent advances in developing electroconductive materials for delivering therapeutic agents as one of emerging approaches for treating heart diseases and regenerating damaged cardiac tissues are highlighted.
Collapse
Affiliation(s)
- Mahsa Ghovvati
- Department of Chemical and Biomolecular Engineering, University of California - Los Angeles, Los Angeles, CA 90095, USA
| | - Mahshid Kharaziha
- Biomaterials Research Group, Department of Materials Engineering, Isfahan University of Technology, Isfahan 84156-83111, Iran
| | - Reza Ardehali
- Division of Cardiology, Department of Internal Medicine, David Geffen School of Medicine, University of California – Los Angeles, Los Angeles, CA 90095, USA
| | - Nasim Annabi
- Department of Chemical and Biomolecular Engineering, University of California - Los Angeles, Los Angeles, CA 90095, USA
- Department of Bioengineering, University of California - Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
31
|
Tang X, He Y, Liu J. Soft bioelectronics for cardiac interfaces. BIOPHYSICS REVIEWS 2022; 3:011301. [PMID: 38505226 PMCID: PMC10903430 DOI: 10.1063/5.0069516] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 12/10/2021] [Indexed: 03/21/2024]
Abstract
Bioelectronics for interrogation and intervention of cardiac systems is important for the study of cardiac health and disease. Interfacing cardiac systems by using conventional rigid bioelectronics is limited by the structural and mechanical disparities between rigid electronics and soft tissues as well as their limited performance. Recently, advances in soft electronics have led to the development of high-performance soft bioelectronics, which is flexible and stretchable, capable of interfacing with cardiac systems in ways not possible with conventional rigid bioelectronics. In this review, we first review the latest developments in building flexible and stretchable bioelectronics for the epicardial interface with the heart. Next, we introduce how stretchable bioelectronics can be integrated with cardiac catheters for a minimally invasive in vivo heart interface. Then, we highlight the recent progress in the design of soft bioelectronics as a new class of biomaterials for integration with different in vitro cardiac models. In particular, we highlight how these devices unlock opportunities to interrogate the cardiac activities in the cardiac patch and cardiac organoid models. Finally, we discuss future directions and opportunities using soft bioelectronics for the study of cardiac systems.
Collapse
Affiliation(s)
- Xin Tang
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, Massachusetts 02134, USA
| | - Yichun He
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, Massachusetts 02134, USA
| | - Jia Liu
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, Massachusetts 02134, USA
| |
Collapse
|
32
|
Rothermel TM, Cook BL, Alford PW. Cellular Microbiaxial Stretching Assay for Measurement and Characterization of the Anisotropic Mechanical Properties of Micropatterned Cells. Curr Protoc 2022; 2:e370. [PMID: 35195953 DOI: 10.1002/cpz1.370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Characterizing the mechanical properties of single cells is important for developing descriptive models of tissue mechanics and improving the understanding of mechanically driven cell processes. Standard methods for measuring single-cell mechanical properties typically provide isotropic mechanical descriptions. However, many cells exhibit specialized geometries in vivo, with anisotropic cytoskeletal architectures reflective of their function, and are exposed to dynamic multiaxial loads, raising the need for more complete descriptions of their anisotropic mechanical properties under complex deformations. Here, we describe the cellular microbiaxial stretching (CμBS) assay in which controlled deformations are applied to micropatterned cells while simultaneously measuring cell stress. CμBS utilizes a set of linear actuators to apply tensile or compressive, short- or long-term deformations to cells micropatterned on a fluorescent bead-doped polyacrylamide gel. Using traction force microscopy principles and the known geometry of the cell and the mechanical properties of the underlying gel, we calculate the stress within the cell to formulate stress-strain curves that can be further used to create mechanical descriptions of the cells, such as strain energy density functions. © 2022 Wiley Periodicals LLC. Basic Protocol 1: Assembly of CμBS stretching constructs Basic Protocol 2: Polymerization of micropatterned, bead-doped polyacrylamide gel on an elastomer membrane Support Protocol: Cell culture and seeding onto CμBS constructs Basic Protocol 3: Implementing CμBS stretching protocols and traction force microscopy Basic Protocol 4: Data analysis and cell stress measurements.
Collapse
Affiliation(s)
- Taylor M Rothermel
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota
| | - Bernard L Cook
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota
| | - Patrick W Alford
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
33
|
Béland J, Duverger JE, Comtois P. Novel Analysis Method for Beating Cells Videomicroscopy Data: Functional Characterization of Culture Samples. Front Physiol 2022; 13:733706. [PMID: 35242049 PMCID: PMC8886216 DOI: 10.3389/fphys.2022.733706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 01/06/2022] [Indexed: 11/13/2022] Open
Abstract
Cell culture of cardiac tissue analog is becoming increasingly interesting for regenerative medicine (cell therapy and tissue engineering) and is widely used for high throughput cardiotoxicity. As a cost-effective approach to rapidly discard new compounds with high toxicity risks, cardiotoxicity evaluation is firstly done in vitro requiring cells/tissue with physiological/pathological characteristics (close to in vivo properties). Studying multicellular electrophysiological and contractile properties is needed to assess drug effects. Techniques favoring process automation which could help in simplifying screening drug candidates are thus of central importance. A lot of effort has been made to ameliorate in vitro models including several in vitro platforms for engineering neonatal rat cardiac tissues. However, most of the initial evaluation is done by studying the rate of activity. In this study, we present new approaches that use the videomicroscopy video of monolayer activity to study contractile properties of beating cells in culture. Two new variables are proposed which are linked to the contraction dynamics and are dependent on the rhythm of activity. Methods for evaluation of regional synchronicity within the image field of view are also presented that can rapidly determine regions with abnormal activity or heterogeneity in contraction dynamics.
Collapse
Affiliation(s)
- Jonathan Béland
- Research Centre, Montreal Heart Institute, Montreal, QC, Canada
- Department of Pharmacology and Physiology, Université de Montréal, Montreal, QC, Canada
| | - James Elber Duverger
- Research Centre, Montreal Heart Institute, Montreal, QC, Canada
- Institute of Biomedical Engineering, Université de Montréal, Montreal, QC, Canada
| | - Philippe Comtois
- Research Centre, Montreal Heart Institute, Montreal, QC, Canada
- Department of Pharmacology and Physiology, Université de Montréal, Montreal, QC, Canada
- Institute of Biomedical Engineering, Université de Montréal, Montreal, QC, Canada
- *Correspondence: Philippe Comtois,
| |
Collapse
|
34
|
He C, Liu M, Jiang D, Wu J, Qin C, Liang T, Wu P, Han C, Huang L, Hsia KJ, Wang P. Fabricating Tissues In Situ with the Controlled Cellular Alignments. Adv Healthc Mater 2022; 11:e2100934. [PMID: 34648692 DOI: 10.1002/adhm.202100934] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 09/15/2021] [Indexed: 11/07/2022]
Abstract
Tissue engineering techniques have enabled to replicate the geometrical architecture of native tissues but usually fail to reproduce their exact cellular arrangements during the fabricating process, while it is critical for manufacturing physiologically relevant tissues. To address this problem, a "sewing-like" method of controlling cellular alignment during the fabricating process is reported here. By integrating the stretching step into the fabricating process, a static mechanical environment is created which, in turn, regulates the subsequent cellular alignment, elongation, and differentiation in the generated tissues. With this method, patterned cellular constructs can be fabricated with controlled cellular alignment. Moreover, this method shows a potent capability to fabricate physiologically relevant skeletal muscle constructs in vitro by mechanically inducing myoblast fusion and maturation. As a potential clinical application, aligned myofibers are directly fabricated onto injured muscles in vivo, which repair the damaged tissues effectively. This study shows that the "sewing-like" method can produce engineered tissues with precise control of cellular arrangements and more clinically viable functionalities.
Collapse
Affiliation(s)
- Chuanjiang He
- Biosensor National Special Laboratory Key Laboratory for Biomedical Engineering Ministry of Education Department of Biomedical Engineering Zhejiang University Hangzhou 310027 China
- State Key Laboratory of Transducer Technology Chinese Academy of Sciences Shanghai 200050 China
| | - Mengxue Liu
- Biosensor National Special Laboratory Key Laboratory for Biomedical Engineering Ministry of Education Department of Biomedical Engineering Zhejiang University Hangzhou 310027 China
| | - Deming Jiang
- Biosensor National Special Laboratory Key Laboratory for Biomedical Engineering Ministry of Education Department of Biomedical Engineering Zhejiang University Hangzhou 310027 China
| | - Jianguo Wu
- Biosensor National Special Laboratory Key Laboratory for Biomedical Engineering Ministry of Education Department of Biomedical Engineering Zhejiang University Hangzhou 310027 China
| | - Chunlian Qin
- Biosensor National Special Laboratory Key Laboratory for Biomedical Engineering Ministry of Education Department of Biomedical Engineering Zhejiang University Hangzhou 310027 China
| | - Tao Liang
- Biosensor National Special Laboratory Key Laboratory for Biomedical Engineering Ministry of Education Department of Biomedical Engineering Zhejiang University Hangzhou 310027 China
| | - Pan Wu
- Department of Burns and Wound Care Center The Second Affiliated Hospital of Zhejiang University School of Medicine Zhejiang University Hangzhou 310009 China
| | - Chunmao Han
- Department of Burns and Wound Care Center The Second Affiliated Hospital of Zhejiang University School of Medicine Zhejiang University Hangzhou 310009 China
| | - Liquan Huang
- College of Life Sciences Zhejiang University Hangzhou 310058 China
- Monell Chemical Senses Center Philadelphia PA 19104 USA
| | - K. Jimmy Hsia
- Schools of Chemical and Biomedical Engineering and Mechanical and Aerospace Engineering Nanyang Technological University Singapore 639798 Singapore
| | - Ping Wang
- Biosensor National Special Laboratory Key Laboratory for Biomedical Engineering Ministry of Education Department of Biomedical Engineering Zhejiang University Hangzhou 310027 China
- State Key Laboratory of Transducer Technology Chinese Academy of Sciences Shanghai 200050 China
| |
Collapse
|
35
|
Basara G, Saeidi-Javash M, Ren X, Bahcecioglu G, Wyatt BC, Anasori B, Zhang Y, Zorlutuna P. Electrically conductive 3D printed Ti 3C 2T x MXene-PEG composite constructs for cardiac tissue engineering. Acta Biomater 2022; 139:179-189. [PMID: 33352299 PMCID: PMC8213874 DOI: 10.1016/j.actbio.2020.12.033] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 11/30/2020] [Accepted: 12/15/2020] [Indexed: 02/03/2023]
Abstract
Tissue engineered cardiac patches have great potential as a therapeutic treatment for myocardial infarction (MI). However, for successful integration with the native tissue and proper function of the cells comprising the patch, it is crucial for these patches to mimic the ordered structure of the native extracellular matrix and the electroconductivity of the human heart. In this study, a new composite construct that can provide both conductive and topographical cues for human induced pluripotent stem cell derived cardiomyocytes (iCMs) is developed for cardiac tissue engineering applications. The constructs are fabricated by 3D printing conductive titanium carbide (Ti3C2Tx) MXene in pre-designed patterns on polyethylene glycol (PEG) hydrogels, using aerosol jet printing, at a cell-level resolution and then seeded with iCMs and cultured for one week with no signs of cytotoxicity. The results presented in this work illustrate the vital role of 3D-printed Ti3C2Tx MXene on aligning iCMs with a significant increase in MYH7, SERCA2, and TNNT2 expressions, and with an improved synchronous beating as well as conduction velocity. This study demonstrates that 3D printed Ti3C2Tx MXene can potentially be used to create physiologically relevant cardiac patches for the treatment of MI. STATEMENT OF SIGNIFICANCE: As cardiovascular diseases and specifically myocardial infarction (MI) continue to be the leading cause of death worldwide, it is critical that new clinical interventions be developed. Tissue engineered cardiac patches have shown significant potential as clinical therapeutics to promote recovery following MI. Unfortunately, current constructs lack the ordered structure and electroconductivity of native human heart. In this study, we engineered a composite construct that can provide both conductive and topographical cues for human induced pluripotent stem cell derived cardiomyocytes. By 3D printing conductive Ti3C2Tx MXene in pre-designed patterns on polyethylene glycol hydrogels, using aerosol jet printing, at a cell-level resolution, we developed tissue engineered patches that have the potential for providing a new clinical therapeutic to combat cardiovascular disease.
Collapse
Affiliation(s)
- Gozde Basara
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, Indiana 46556, USA
| | - Mortaza Saeidi-Javash
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, Indiana 46556, USA
| | - Xiang Ren
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, Indiana 46556, USA
| | - Gokhan Bahcecioglu
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, Indiana 46556, USA
| | - Brian C. Wyatt
- Integrated Nanosystems Development Institute and Department of Mechanical and Energy Engineering, Purdue School of Engineering and Technology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202, USA
| | - Babak Anasori
- Integrated Nanosystems Development Institute and Department of Mechanical and Energy Engineering, Purdue School of Engineering and Technology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202, USA
| | - Yanliang Zhang
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, Indiana 46556, USA
| | - Pinar Zorlutuna
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, Indiana 46556, USA,Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana 46556, USA,Corresponding author: Pinar Zorlutuna, , Address: 143 Multidisciplinary Research Building, University of Notre Dame, Notre Dame, IN 46556, Phone no: +1 574 631 8543, Fax no: +1 574 631 8341
| |
Collapse
|
36
|
Petersen AP, McCain ML. Quantifying Propagation Velocity from Engineered Cardiac Tissues with High-Speed Fluorescence Microscopy and Automated Analysis Software. Methods Mol Biol 2022; 2485:133-145. [PMID: 35618903 DOI: 10.1007/978-1-0716-2261-2_9] [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: 06/15/2023]
Abstract
Many acquired or inherited forms of heart disease as well as drugs are known to increase the susceptibility of patients to arrhythmias. To predict arrhythmogenic events and discover new therapeutic strategies to mitigate them, approaches to efficiently quantify the velocity of propagation in engineered cardiac tissues are important research tools. In this chapter, we describe how to collect videos of propagating calcium waves in engineered cardiac tissues with a high-speed camera mounted on an inverted fluorescence microscope. We also provide instructions for downloading and using our software package to analyze these videos and calculate propagation velocity. These techniques should be compatible with a variety of voltage- or calcium-sensitive fluorescent dyes or genetically encoded sensors. Although these approaches were originally developed for engineered neonatal rat cardiac tissues, the same procedures can likely be used with human-induced pluripotent stem cell-derived cardiac myocytes, paving the way for patient-specific analysis of propagation due to features such as tissue architecture, substrate rigidity, genetic mutations, or drug treatments.
Collapse
Affiliation(s)
- Andrew P Petersen
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA, USA
| | - Megan L McCain
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA, USA.
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
37
|
Giardini F, Lazzeri E, Vitale G, Ferrantini C, Costantini I, Pavone FS, Poggesi C, Bocchi L, Sacconi L. Quantification of Myocyte Disarray in Human Cardiac Tissue. Front Physiol 2021; 12:750364. [PMID: 34867455 PMCID: PMC8635020 DOI: 10.3389/fphys.2021.750364] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 09/24/2021] [Indexed: 11/13/2022] Open
Abstract
Proper three-dimensional (3D)-cardiomyocyte orientation is important for an effective tension production in cardiac muscle. Cardiac diseases can cause severe remodeling processes in the heart, such as cellular misalignment, that can affect both the electrical and mechanical functions of the organ. To date, a proven methodology to map and quantify myocytes disarray in massive samples is missing. In this study, we present an experimental pipeline to reconstruct and analyze the 3D cardiomyocyte architecture in massive samples. We employed tissue clearing, staining, and advanced microscopy techniques to detect sarcomeres in relatively large human myocardial strips with micrometric resolution. Z-bands periodicity was exploited in a frequency analysis approach to extract the 3D myofilament orientation, providing an orientation map used to characterize the tissue organization at different spatial scales. As a proof-of-principle, we applied the proposed method to healthy and pathologically remodeled human cardiac tissue strips. Preliminary results suggest the reliability of the method: strips from a healthy donor are characterized by a well-organized tissue, where the local disarray is log-normally distributed and slightly depends on the spatial scale of analysis; on the contrary, pathological strips show pronounced tissue disorganization, characterized by local disarray significantly dependent on the spatial scale of analysis. A virtual sample generator is developed to link this multi-scale disarray analysis with the underlying cellular architecture. This approach allowed us to quantitatively assess tissue organization in terms of 3D myocyte angular dispersion and may pave the way for developing novel predictive models based on structural data at cellular resolution.
Collapse
Affiliation(s)
- Francesco Giardini
- Laboratory of Non-Linear Spectroscopy (LENS), University of Florence, Sesto Fiorentino, Italy
| | - Erica Lazzeri
- Laboratory of Non-Linear Spectroscopy (LENS), University of Florence, Sesto Fiorentino, Italy
| | - Giulia Vitale
- Division of Physiology, Department of Clinical and Experimental Medicine, University of Florence, Florence, Italy
| | - Cecilia Ferrantini
- Laboratory of Non-Linear Spectroscopy (LENS), University of Florence, Sesto Fiorentino, Italy.,Division of Physiology, Department of Clinical and Experimental Medicine, University of Florence, Florence, Italy
| | - Irene Costantini
- Laboratory of Non-Linear Spectroscopy (LENS), University of Florence, Sesto Fiorentino, Italy.,National Institute of Optics, National Research Council, University of Florence, Florence, Italy.,Department of Biology, University of Florence, Florence, Italy
| | - Francesco S Pavone
- Laboratory of Non-Linear Spectroscopy (LENS), University of Florence, Sesto Fiorentino, Italy.,National Institute of Optics, National Research Council, University of Florence, Florence, Italy.,Department of Physics, University of Florence, Florence, Italy
| | - Corrado Poggesi
- Division of Physiology, Department of Clinical and Experimental Medicine, University of Florence, Florence, Italy
| | - Leonardo Bocchi
- Laboratory of Non-Linear Spectroscopy (LENS), University of Florence, Sesto Fiorentino, Italy.,Department of Information Engineering, University of Florence, Florence, Italy
| | - Leonardo Sacconi
- Laboratory of Non-Linear Spectroscopy (LENS), University of Florence, Sesto Fiorentino, Italy.,National Institute of Optics, National Research Council, University of Florence, Florence, Italy.,Faculty of Medicine, Institute for Experimental Cardiovascular Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| |
Collapse
|
38
|
Tran RDH, Morris TA, Gonzalez D, Hetta AHSHA, Grosberg A. Quantitative Evaluation of Cardiac Cell Interactions and Responses to Cyclic Strain. Cells 2021; 10:3199. [PMID: 34831422 PMCID: PMC8625419 DOI: 10.3390/cells10113199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 10/14/2021] [Accepted: 10/27/2021] [Indexed: 11/17/2022] Open
Abstract
The heart has a dynamic mechanical environment contributed by its unique cellular composition and the resultant complex tissue structure. In pathological heart tissue, both the mechanics and cell composition can change and influence each other. As a result, the interplay between the cell phenotype and mechanical stimulation needs to be considered to understand the biophysical cell interactions and organization in healthy and diseased myocardium. In this work, we hypothesized that the overall tissue organization is controlled by varying densities of cardiomyocytes and fibroblasts in the heart. In order to test this hypothesis, we utilized a combination of mechanical strain, co-cultures of different cell types, and inhibitory drugs that block intercellular junction formation. To accomplish this, an image analysis pipeline was developed to automatically measure cell type-specific organization relative to the stretch direction. The results indicated that cardiac cell type-specific densities influence the overall organization of heart tissue such that it is possible to model healthy and fibrotic heart tissue in vitro. This study provides insight into how to mimic the dynamic mechanical environment of the heart in engineered tissue as well as providing valuable information about the process of cardiac remodeling and repair in diseased hearts.
Collapse
Affiliation(s)
- Richard Duc Hien Tran
- Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, CA 92617-2700, USA; (R.D.H.T.); (T.A.M.); (D.G.); (A.H.S.H.A.H.)
- Department of Biomedical Engineering, University of California, Irvine, CA 92617, USA
| | - Tessa Altair Morris
- Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, CA 92617-2700, USA; (R.D.H.T.); (T.A.M.); (D.G.); (A.H.S.H.A.H.)
- Center for Complex Biological Systems, University of California, Irvine, CA 92697, USA
- NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, CA 92697, USA
| | - Daniela Gonzalez
- Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, CA 92617-2700, USA; (R.D.H.T.); (T.A.M.); (D.G.); (A.H.S.H.A.H.)
- Department of Biomedical Engineering, University of California, Irvine, CA 92617, USA
| | - Ali Hatem Salaheldin Hassan Ahmed Hetta
- Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, CA 92617-2700, USA; (R.D.H.T.); (T.A.M.); (D.G.); (A.H.S.H.A.H.)
- Department of Biomedical Engineering, University of California, Irvine, CA 92617, USA
| | - Anna Grosberg
- Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, CA 92617-2700, USA; (R.D.H.T.); (T.A.M.); (D.G.); (A.H.S.H.A.H.)
- Department of Biomedical Engineering, University of California, Irvine, CA 92617, USA
- Center for Complex Biological Systems, University of California, Irvine, CA 92697, USA
- NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, CA 92697, USA
- Department of Chemical and Biomolecular Engineering, University of California, Irvine, CA 92617, USA
| |
Collapse
|
39
|
Nicin L, Wagner JUG, Luxán G, Dimmeler S. Fibroblast-mediated intercellular crosstalk in the healthy and diseased heart. FEBS Lett 2021; 596:638-654. [PMID: 34787896 DOI: 10.1002/1873-3468.14234] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 10/28/2021] [Accepted: 11/04/2021] [Indexed: 01/07/2023]
Abstract
Cardiac fibroblasts constitute a major cell population in the heart. They secrete extracellular matrix components and various other factors shaping the microenvironment of the heart. In silico analysis of intercellular communication based on single-cell RNA sequencing revealed that fibroblasts are the source of the majority of outgoing signals to other cell types. This observation suggests that fibroblasts play key roles in orchestrating cellular interactions that maintain organ homeostasis but that can also contribute to disease states. Here, we will review the current knowledge of fibroblast interactions in the healthy, diseased, and aging heart. We focus on the interactions that fibroblasts establish with other cells of the heart, specifically cardiomyocytes, endothelial cells and immune cells, and particularly those relying on paracrine, electrical, and exosomal communication modes.
Collapse
Affiliation(s)
- Luka Nicin
- Institute for Cardiovascular Regeneration, Goethe University, Frankfurt am Main, Germany.,German Center for Cardiovascular Research (DZHK), Frankfurt am Main, Germany.,Cardio-Pulmonary Institute (CPI), Frankfurt am Main, Germany
| | - Julian U G Wagner
- Institute for Cardiovascular Regeneration, Goethe University, Frankfurt am Main, Germany.,German Center for Cardiovascular Research (DZHK), Frankfurt am Main, Germany.,Cardio-Pulmonary Institute (CPI), Frankfurt am Main, Germany
| | - Guillermo Luxán
- Institute for Cardiovascular Regeneration, Goethe University, Frankfurt am Main, Germany.,German Center for Cardiovascular Research (DZHK), Frankfurt am Main, Germany.,Cardio-Pulmonary Institute (CPI), Frankfurt am Main, Germany
| | - Stefanie Dimmeler
- Institute for Cardiovascular Regeneration, Goethe University, Frankfurt am Main, Germany.,German Center for Cardiovascular Research (DZHK), Frankfurt am Main, Germany.,Cardio-Pulmonary Institute (CPI), Frankfurt am Main, Germany
| |
Collapse
|
40
|
Shih ED, Provenzano PP, Witzenburg CM, Barocas VH, Grande AW, Alford PW. Characterizing Tissue Remodeling and Mechanical Heterogeneity in Cerebral Aneurysms. J Vasc Res 2021; 59:34-42. [PMID: 34758464 DOI: 10.1159/000519694] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 09/14/2021] [Indexed: 11/19/2022] Open
Abstract
Accurately assessing the complex tissue mechanics of cerebral aneurysms (CAs) is critical for elucidating how CAs grow and whether that growth will lead to rupture. The factors that have been implicated in CA progression - blood flow dynamics, immune infiltration, and extracellular matrix remodeling - all occur heterogeneously throughout the CA. Thus, it stands to reason that the mechanical properties of CAs are also spatially heterogeneous. Here, we present a new method for characterizing the mechanical heterogeneity of human CAs using generalized anisotropic inverse mechanics, which uses biaxial stretching experiments and inverse analyses to determine the local Kelvin moduli and principal alignments within the tissue. Using this approach, we find that there is significant mechanical heterogeneity within a single acquired human CA. These results were confirmed using second harmonic generation imaging of the CA's fiber architecture and a correlation was observed. This approach provides a single-step method for determining the complex heterogeneous mechanics of CAs, which has important implications for future identification of metrics that can improve accuracy in prediction risk of rupture.
Collapse
Affiliation(s)
- Elizabeth D Shih
- Department of Biomedical Engineering, University of Minnesota Twin Cities, Minneapolis, Minnesota, USA
| | - Paolo P Provenzano
- Department of Biomedical Engineering, University of Minnesota Twin Cities, Minneapolis, Minnesota, USA
| | - Colleen M Witzenburg
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Victor H Barocas
- Department of Biomedical Engineering, University of Minnesota Twin Cities, Minneapolis, Minnesota, USA
| | - Andrew W Grande
- Department of Neurosurgery, University of Minnesota Twin Cities, Minneapolis, Minnesota, USA
| | - Patrick W Alford
- Department of Biomedical Engineering, University of Minnesota Twin Cities, Minneapolis, Minnesota, USA
| |
Collapse
|
41
|
An YH, Kim SH. Facile Fabrication of Three-Dimensional Hydrogel Film with Complex Tissue Morphology. Bioengineering (Basel) 2021; 8:bioengineering8110164. [PMID: 34821730 PMCID: PMC8614799 DOI: 10.3390/bioengineering8110164] [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: 09/03/2021] [Revised: 10/22/2021] [Accepted: 10/24/2021] [Indexed: 11/16/2022] Open
Abstract
In this study, we proposed a simple and easy method for fabricating a three-dimensional (3D) structure that can recapitulate the morphology of a tissue surface and deliver biological molecules into complex-shaped target tissues. To fabricate the 3D hydrogel film structure, we utilized a direct tissue casting method that can recapitulate tissue structure in micro-/macroscale using polydimethylsiloxane (PDMS). A replica 3D negative mold was manufactured by a polyurethane acrylate (PUA)-based master mold. Then, we poured the catechol-conjugated alginate (ALG-C) solution into the mold and evaporated it to form a dried film, followed by crosslinking the film using calcium chloride. The ALG-C hydrogel film had a tensile modulus of 725.2 ± 123.4 kPa and maintained over 95% of initial weight after 1 week without significant degradation. The ALG-C film captured over 4.5 times as much macromolecule (FITC-dextran) compared to alginate film (ALG). The cardiomyoblast cells exhibited high cell viability over 95% on ALG-C film. Moreover, the ALG-C film had about 70% of surface-bound lentivirus (1% in ALG film), which finally exhibited much higher viral transfection efficiency of GFP protein to C2C12 cells on the film than ALG film. In conclusion, we demonstrated a 3D film structure of biofunctionalized hydrogel for substrate-mediated drug delivery, and this approach could be utilized to recapitulate the complex-shaped tissues.
Collapse
Affiliation(s)
- Young-Hyeon An
- BioMax/N-Bio Institute, Seoul National University, Seoul 08826, Korea;
| | - Su-Hwan Kim
- Department of Chemical Engineering (BK 21 FOUR), Dong-A University, Busan 49315, Korea
- Correspondence:
| |
Collapse
|
42
|
Lock R, Al Asafen H, Fleischer S, Tamargo M, Zhao Y, Radisic M, Vunjak-Novakovic G. A framework for developing sex-specific engineered heart models. NATURE REVIEWS. MATERIALS 2021; 7:295-313. [PMID: 34691764 PMCID: PMC8527305 DOI: 10.1038/s41578-021-00381-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 08/20/2021] [Indexed: 05/02/2023]
Abstract
The convergence of tissue engineering and patient-specific stem cell biology has enabled the engineering of in vitro tissue models that allow the study of patient-tailored treatment modalities. However, sex-related disparities in health and disease, from systemic hormonal influences to cellular-level differences, are often overlooked in stem cell biology, tissue engineering and preclinical screening. The cardiovascular system, in particular, shows considerable sex-related differences, which need to be considered in cardiac tissue engineering. In this Review, we analyse sex-related properties of the heart muscle in the context of health and disease, and discuss a framework for including sex-based differences in human cardiac tissue engineering. We highlight how sex-based features can be implemented at the cellular and tissue levels, and how sex-specific cardiac models could advance the study of cardiovascular diseases. Finally, we define design criteria for sex-specific cardiac tissue engineering and provide an outlook to future research possibilities beyond the cardiovascular system.
Collapse
Affiliation(s)
- Roberta Lock
- Department of Biomedical Engineering, Columbia University, New York, NY USA
| | - Hadel Al Asafen
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario Canada
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario Canada
| | - Sharon Fleischer
- Department of Biomedical Engineering, Columbia University, New York, NY USA
| | - Manuel Tamargo
- Department of Biomedical Engineering, Columbia University, New York, NY USA
| | - Yimu Zhao
- Department of Biomedical Engineering, Columbia University, New York, NY USA
| | - Milica Radisic
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario Canada
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario Canada
| | - Gordana Vunjak-Novakovic
- Department of Biomedical Engineering, Columbia University, New York, NY USA
- Department of Medicine, Columbia University, New York, NY USA
| |
Collapse
|
43
|
Sokolova OV. [Forensic medical characteristics of metabolic myocardial lesions affecting cardiac contractile ability in cases of death from alcoholic cardiomyopathy]. Sud Med Ekspert 2021; 64:27-31. [PMID: 34644030 DOI: 10.17116/sudmed20216405127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Objective - a morphological study of myocardial tissue was carried out in order to characterize the metabolic lesions that influence the heart contractility in cases of sudden cardiac death from alcoholic cardiomyopathy. The occurrence of metabolic damages in myocardial tissue in cases of alcoholic cardiomyopathy is a vivid reflection of the toxic effects on the cardiac muscle of ethanol and its metabolites. The toxic damage of the main structural components of the microcirculatory vessels contributes to the disruption of the transport of electrolytes and nutrients with the development of trophic disorders and the increasing phenomena of hypoxia that is the cause dystrophic and necrobiotic changes in myocardial tissue. The contracture damages of cardiomyocytes, intracellular myocytolysis, and cationic decomposition of myofibrils were revealed in polarized light and they were mosaic in nature and played a direct role in the occurrence of cardiac rhythm disturbances. An immunohistochemical study of desmin expression can be recommended as a marker of ischemic and necrobiotic changes in cardiomyocytes, that development is possible on the background of subtotal concentrations of ethanol in the blood. The results of a morphometric study of the parenchymal component of myocardial tissue have established that the relative area of the parenchyma is not directly related to the age and gender of those who died from alcoholic cardiomyopathy. It is recommended to use a combination of morphological methods including light microscopy, polarization microscopy methods and immunohistochemical method for the diagnosis of alcoholic cardiomyopathy in forensic practice.
Collapse
Affiliation(s)
- O V Sokolova
- Academician I.P. Pavlov First Saint Petersburg State Medical University, St. Petersburg, Russia
| |
Collapse
|
44
|
Cook BL, Chao CJ, Alford PW. Architecture-Dependent Mechano-Adaptation in Single Vascular Smooth Muscle Cells. J Biomech Eng 2021; 143:101002. [PMID: 33972987 DOI: 10.1115/1.4051117] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Indexed: 01/03/2023]
Abstract
Arteries grow and remodel following mechanical perturbation. Vascular smooth muscle cells (VSMCs) within the artery undergo hyperplasia, hypertrophy, or change their contractility following sustained changes in loading. Experimental evidence in vivo and in vitro suggests that VSMCs grow and remodel to maintain a constant transmural stress, or "target" stress. This behavior is often described using a stress-dependent finite growth framework. Typically, computational models of arterial growth and remodeling account for VSMC behavior in a constrained mixture formulation that incorporates behavior of each component of the artery. However, these models do not account for differential VSMC architecture observed in situ, which may significantly influence growth and remodeling behavior. Here, we used cellular microbiaxial stretching (CμBS) to characterize how VSMCs with different cytoskeletal architectures respond to a sustained step change in strain. We find that VSMC F-actin architecture becomes more aligned following stretch and retains this alignment after 24 h. Further, we find that VSMC stress magnitude depends on cellular architecture. Qualitatively, however, stress behavior following stretch is consistent across cell architectures-stress increases following stretch and returns to prestretch magnitudes after 24 h. Finally, we formulated an architecture-dependent targeted growth law that accounts for experimentally measured cytoskeletal alignment and attributes stress evolution to individual fiber growth and find that this model robustly captures long-term stress evolution in single VSMCs. These results suggest that VSMC mechano-adaptation depends on cellular architecture, which has implications for growth and remodeling in regions of arteries with differential architecture, such as at bifurcations.
Collapse
Affiliation(s)
- Bernard L Cook
- Department of Biomedical Engineering, University of Minnesota, Nils Hasselmo Hall, Room 7-105 312 Church Street SE, Minneapolis, MN 55455
| | - Christina J Chao
- Department of Biomedical Engineering, University of Minnesota, Nils Hasselmo Hall, Room 7-105 312 Church Street SE, Minneapolis, MN 55455
| | - Patrick W Alford
- Department of Biomedical Engineering, University of Minnesota, Nils Hasselmo Hall, Room 7-105 312 Church Street SE, Minneapolis, MN 55455
| |
Collapse
|
45
|
Mehrabi M, Morris TA, Cang Z, Nguyen CHH, Sha Y, Asad MN, Khachikyan N, Greene TL, Becker DM, Nie Q, Zaragoza MV, Grosberg A. A Study of Gene Expression, Structure, and Contractility of iPSC-Derived Cardiac Myocytes from a Family with Heart Disease due to LMNA Mutation. Ann Biomed Eng 2021; 49:3524-3539. [PMID: 34585335 PMCID: PMC8671287 DOI: 10.1007/s10439-021-02850-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 08/06/2021] [Indexed: 12/18/2022]
Abstract
Genetic mutations to the Lamin A/C gene (LMNA) can cause heart disease, but the mechanisms making cardiac tissues uniquely vulnerable to the mutations remain largely unknown. Further, patients with LMNA mutations have highly variable presentation of heart disease progression and type. In vitro patient-specific experiments could provide a powerful platform for studying this phenomenon, but the use of induced pluripotent stem cell-derived cardiomyocytes (iPSC-CM) introduces heterogeneity in maturity and function thus complicating the interpretation of the results of any single experiment. We hypothesized that integrating single cell RNA sequencing (scRNA-seq) with analysis of the tissue architecture and contractile function would elucidate some of the probable mechanisms. To test this, we investigated five iPSC-CM lines, three controls and two patients with a (c.357-2A>G) mutation. The patient iPSC-CM tissues had significantly weaker stress generation potential than control iPSC-CM tissues demonstrating the viability of our in vitro approach. Through scRNA-seq, differentially expressed genes between control and patient lines were identified. Some of these genes, linked to quantitative structural and functional changes, were cardiac specific, explaining the targeted nature of the disease progression seen in patients. The results of this work demonstrate the utility of combining in vitro tools in exploring heart disease mechanics.
Collapse
Affiliation(s)
- Mehrsa Mehrabi
- Department of Biomedical Engineering, University of California, Irvine, CA, 92697, USA.,UCI Edwards Lifesciences Foundation Cardiovascular Innovation and Research Center (CIRC), University of California, Irvine, CA, 92697, USA
| | - Tessa A Morris
- UCI Edwards Lifesciences Foundation Cardiovascular Innovation and Research Center (CIRC), University of California, Irvine, CA, 92697, USA.,Center for Complex Biological Systems, University of California, Irvine, CA, 92697, USA
| | - Zixuan Cang
- Department of Mathematics and Developmental & Cell Biology, University of California, Irvine, CA, 92697, USA.,The NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, CA, 92697, USA
| | - Cecilia H H Nguyen
- Genetics & Genomics Division, Department of Pediatrics, School of Medicine, University of California, Irvine, CA, 92697, USA
| | - Yutong Sha
- Department of Mathematics and Developmental & Cell Biology, University of California, Irvine, CA, 92697, USA
| | - Mira N Asad
- Department of Biomedical Engineering, University of California, Irvine, CA, 92697, USA.,UCI Edwards Lifesciences Foundation Cardiovascular Innovation and Research Center (CIRC), University of California, Irvine, CA, 92697, USA
| | - Nyree Khachikyan
- Department of Biomedical Engineering, University of California, Irvine, CA, 92697, USA.,UCI Edwards Lifesciences Foundation Cardiovascular Innovation and Research Center (CIRC), University of California, Irvine, CA, 92697, USA
| | - Taylor L Greene
- Department of Biomedical Engineering, University of California, Irvine, CA, 92697, USA.,UCI Edwards Lifesciences Foundation Cardiovascular Innovation and Research Center (CIRC), University of California, Irvine, CA, 92697, USA
| | - Danielle M Becker
- Department of Biomedical Engineering, University of California, Irvine, CA, 92697, USA.,UCI Edwards Lifesciences Foundation Cardiovascular Innovation and Research Center (CIRC), University of California, Irvine, CA, 92697, USA
| | - Qing Nie
- Department of Biomedical Engineering, University of California, Irvine, CA, 92697, USA.,Department of Mathematics and Developmental & Cell Biology, University of California, Irvine, CA, 92697, USA.,The NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, CA, 92697, USA
| | - Michael V Zaragoza
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, CA, 92697, USA.,Genetics & Genomics Division, Department of Pediatrics, School of Medicine, University of California, Irvine, CA, 92697, USA
| | - Anna Grosberg
- Department of Biomedical Engineering, University of California, Irvine, CA, 92697, USA. .,UCI Edwards Lifesciences Foundation Cardiovascular Innovation and Research Center (CIRC), University of California, Irvine, CA, 92697, USA. .,Center for Complex Biological Systems, University of California, Irvine, CA, 92697, USA. .,The NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, CA, 92697, USA. .,Department of Chemical and Biomolecular Engineering, University of California, Irvine, CA, 92697, USA. .,The Henry Samueli School of Engineering, University of California, Irvine, 2418 Engineering Hall, Irvine, CA, 92697, USA.
| |
Collapse
|
46
|
Bliley JM, Vermeer MCSC, Duffy RM, Batalov I, Kramer D, Tashman JW, Shiwarski DJ, Lee A, Teplenin AS, Volkers L, Coffin B, Hoes MF, Kalmykov A, Palchesko RN, Sun Y, Jongbloed JDH, Bomer N, de Boer RA, Suurmeijer AJH, Pijnappels DA, Bolling MC, van der Meer P, Feinberg AW. Dynamic loading of human engineered heart tissue enhances contractile function and drives a desmosome-linked disease phenotype. Sci Transl Med 2021; 13:13/603/eabd1817. [PMID: 34290054 DOI: 10.1126/scitranslmed.abd1817] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 07/02/2021] [Indexed: 12/23/2022]
Abstract
The role that mechanical forces play in shaping the structure and function of the heart is critical to understanding heart formation and the etiology of disease but is challenging to study in patients. Engineered heart tissues (EHTs) incorporating human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes have the potential to provide insight into these adaptive and maladaptive changes. However, most EHT systems cannot model both preload (stretch during chamber filling) and afterload (pressure the heart must work against to eject blood). Here, we have developed a new dynamic EHT (dyn-EHT) model that enables us to tune preload and have unconstrained contractile shortening of >10%. To do this, three-dimensional (3D) EHTs were integrated with an elastic polydimethylsiloxane strip providing mechanical preload and afterload in addition to enabling contractile force measurements based on strip bending. Our results demonstrated that dynamic loading improves the function of wild-type EHTs on the basis of the magnitude of the applied force, leading to improved alignment, conduction velocity, and contractility. For disease modeling, we used hiPSC-derived cardiomyocytes from a patient with arrhythmogenic cardiomyopathy due to mutations in the desmoplakin gene. We demonstrated that manifestation of this desmosome-linked disease state required dyn-EHT conditioning and that it could not be induced using 2D or standard 3D EHT approaches. Thus, a dynamic loading strategy is necessary to provoke the disease phenotype of diastolic lengthening, reduction of desmosome counts, and reduced contractility, which are related to primary end points of clinical disease, such as chamber thinning and reduced cardiac output.
Collapse
Affiliation(s)
- Jacqueline M Bliley
- Regenerative Biomaterials and Therapeutics Group, Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Mathilde C S C Vermeer
- Department of Cardiology, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, Netherlands
| | - Rebecca M Duffy
- Regenerative Biomaterials and Therapeutics Group, Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Ivan Batalov
- Regenerative Biomaterials and Therapeutics Group, Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Duco Kramer
- Department of Dermatology, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, Netherlands
| | - Joshua W Tashman
- Regenerative Biomaterials and Therapeutics Group, Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Daniel J Shiwarski
- Regenerative Biomaterials and Therapeutics Group, Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Andrew Lee
- Regenerative Biomaterials and Therapeutics Group, Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Alexander S Teplenin
- Department of Cardiology, Heart Lung Center Leiden, Leiden University Medical Center, 2333 ZA Leiden, Netherlands
| | - Linda Volkers
- Department of Cardiology, Heart Lung Center Leiden, Leiden University Medical Center, 2333 ZA Leiden, Netherlands
| | - Brian Coffin
- Department of Materials Science and Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Martijn F Hoes
- Department of Cardiology, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, Netherlands
| | - Anna Kalmykov
- Regenerative Biomaterials and Therapeutics Group, Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Rachelle N Palchesko
- Regenerative Biomaterials and Therapeutics Group, Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Yan Sun
- Regenerative Biomaterials and Therapeutics Group, Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Jan D H Jongbloed
- Department of Genetics, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, Netherlands
| | - Nils Bomer
- Department of Cardiology, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, Netherlands
| | - Rudolf A de Boer
- Department of Cardiology, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, Netherlands
| | - Albert J H Suurmeijer
- Department of Pathology, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, Netherlands
| | - Daniel A Pijnappels
- Department of Cardiology, Heart Lung Center Leiden, Leiden University Medical Center, 2333 ZA Leiden, Netherlands
| | - Maria C Bolling
- Department of Dermatology, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, Netherlands
| | - Peter van der Meer
- Department of Cardiology, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, Netherlands.
| | - Adam W Feinberg
- Regenerative Biomaterials and Therapeutics Group, Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA. .,Department of Materials Science and Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| |
Collapse
|
47
|
Rieger M, Duran P, Cook M, Schenk S, Shah M, Jacobs M, Christman K, Kado DM, Alperin M. Quantifying the Effects of Aging on Morphological and Cellular Properties of Human Female Pelvic Floor Muscles. Ann Biomed Eng 2021; 49:1836-1847. [PMID: 33683527 PMCID: PMC8376748 DOI: 10.1007/s10439-021-02748-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 02/04/2021] [Indexed: 12/11/2022]
Abstract
Age-related pelvic floor muscle (PFM) dysfunction is a critical defect in the progression to pelvic floor disorders (PFDs). Despite dramatic prevalence of PFDs in older women, the underlying pathophysiology of age-related PFM dysfunction remains poorly understood. Using cadaveric specimens, we quantified aging effects on functionally relevant PFM properties and compared PFMs with the appendicular muscles from the same donors. PFMs, obturator internus, and vastus lateralis were procured from younger (N = 4) and older (N = 11) donors with known obstetrical and medical history. Our findings demonstrate that PFMs undergo degenerative, rather than atrophic, alterations. Importantly, age-related fibrotic degeneration disproportionally impacts PFMs compared to the appendicular muscles. We identified intramuscular lipid accumulation as another contributing factor to the pathological alterations of PFMs with aging. We observed a fourfold decrease in muscle stem cell (MuSC) pool of aged relative to younger PFMs, but the MuSC pool of appendicular muscles from the same older donors was only twofold lower than in younger group, although these differences were not statistically significant. Age-related degeneration appears to disproportionally impact PFMs relative to the appendicular muscles from the same donors. Knowledge of tissue- and cell-level changes in aged PFMs is essential to promote our understanding of the mechanisms governing PFM dysfunction in older women.
Collapse
Affiliation(s)
- Mary Rieger
- Division of Female Pelvic Medicine and Reconstructive Surgery, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0863, USA
| | - Pamela Duran
- Department of Bioengineering and Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, USA
| | - Mark Cook
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, USA
| | - Simon Schenk
- Department of Orthopedic Surgery, University of California San Diego, La Jolla, USA
| | - Manali Shah
- Department of Bioengineering, University of California San Diego, La Jolla, USA
| | - Marni Jacobs
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego, La Jolla, USA
| | - Karen Christman
- Department of Bioengineering and Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, USA
| | - Deborah M Kado
- Herbert Wertheim School of Public Health and Human Longevity Science, University of California San Diego, La Jolla, USA
- Department of Medicine, University of California San Diego, La Jolla, USA
| | - Marianna Alperin
- Division of Female Pelvic Medicine and Reconstructive Surgery, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0863, USA.
| |
Collapse
|
48
|
Khalil NN, McCain ML. Engineering the Cellular Microenvironment of Post-infarct Myocardium on a Chip. Front Cardiovasc Med 2021; 8:709871. [PMID: 34336962 PMCID: PMC8316619 DOI: 10.3389/fcvm.2021.709871] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 06/14/2021] [Indexed: 01/02/2023] Open
Abstract
Myocardial infarctions are one of the most common forms of cardiac injury and death worldwide. Infarctions cause immediate necrosis in a localized region of the myocardium, which is followed by a repair process with inflammatory, proliferative, and maturation phases. This repair process culminates in the formation of scar tissue, which often leads to heart failure in the months or years after the initial injury. In each reparative phase, the infarct microenvironment is characterized by distinct biochemical, physical, and mechanical features, such as inflammatory cytokine production, localized hypoxia, and tissue stiffening, which likely each contribute to physiological and pathological tissue remodeling by mechanisms that are incompletely understood. Traditionally, simplified two-dimensional cell culture systems or animal models have been implemented to elucidate basic pathophysiological mechanisms or predict drug responses following myocardial infarction. However, these conventional approaches offer limited spatiotemporal control over relevant features of the post-infarct cellular microenvironment. To address these gaps, Organ on a Chip models of post-infarct myocardium have recently emerged as new paradigms for dissecting the highly complex, heterogeneous, and dynamic post-infarct microenvironment. In this review, we describe recent Organ on a Chip models of post-infarct myocardium, including their limitations and future opportunities in disease modeling and drug screening.
Collapse
Affiliation(s)
- Natalie N Khalil
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA, United States
| | - Megan L McCain
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA, United States.,Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
49
|
Wang Z, Wang L, Li T, Liu S, Guo B, Huang W, Wu Y. 3D bioprinting in cardiac tissue engineering. Am J Cancer Res 2021; 11:7948-7969. [PMID: 34335973 PMCID: PMC8315053 DOI: 10.7150/thno.61621] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 06/06/2021] [Indexed: 12/22/2022] Open
Abstract
Heart disease is the main cause of death worldwide. Because death of the myocardium is irreversible, it remains a significant clinical challenge to rescue myocardial deficiency. Cardiac tissue engineering (CTE) is a promising strategy for repairing heart defects and offers platforms for studying cardiac tissue. Numerous achievements have been made in CTE in the past decades based on various advanced engineering approaches. 3D bioprinting has attracted much attention due to its ability to integrate multiple cells within printed scaffolds with complex 3D structures, and many advancements in bioprinted CTE have been reported recently. Herein, we review the recent progress in 3D bioprinting for CTE. After a brief overview of CTE with conventional methods, the current 3D printing strategies are discussed. Bioink formulations based on various biomaterials are introduced, and strategies utilizing composite bioinks are further discussed. Moreover, several applications including heart patches, tissue-engineered cardiac muscle, and other bionic structures created via 3D bioprinting are summarized. Finally, we discuss several crucial challenges and present our perspective on 3D bioprinting techniques in the field of CTE.
Collapse
|
50
|
Dwyer KD, Coulombe KL. Cardiac mechanostructure: Using mechanics and anisotropy as inspiration for developing epicardial therapies in treating myocardial infarction. Bioact Mater 2021; 6:2198-2220. [PMID: 33553810 PMCID: PMC7822956 DOI: 10.1016/j.bioactmat.2020.12.015] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 12/18/2020] [Accepted: 12/18/2020] [Indexed: 12/14/2022] Open
Abstract
The mechanical environment and anisotropic structure of the heart modulate cardiac function at the cellular, tissue and organ levels. During myocardial infarction (MI) and subsequent healing, however, this landscape changes significantly. In order to engineer cardiac biomaterials with the appropriate properties to enhance function after MI, the changes in the myocardium induced by MI must be clearly identified. In this review, we focus on the mechanical and structural properties of the healthy and infarcted myocardium in order to gain insight about the environment in which biomaterial-based cardiac therapies are expected to perform and the functional deficiencies caused by MI that the therapy must address. From this understanding, we discuss epicardial therapies for MI inspired by the mechanics and anisotropy of the heart focusing on passive devices, which feature a biomaterials approach, and active devices, which feature robotic and cellular components. Through this review, a detailed analysis is provided in order to inspire further development and translation of epicardial therapies for MI.
Collapse
Affiliation(s)
- Kiera D. Dwyer
- Center for Biomedical Engineering, School of Engineering, Brown University, Providence, RI, USA
| | - Kareen L.K. Coulombe
- Center for Biomedical Engineering, School of Engineering, Brown University, Providence, RI, USA
| |
Collapse
|