1
|
Kunrath MF, Garaicoa‐Pazmino C, Giraldo‐Osorno PM, Haj Mustafa A, Dahlin C, Larsson L, Asa'ad F. Implant surface modifications and their impact on osseointegration and peri-implant diseases through epigenetic changes: A scoping review. J Periodontal Res 2024; 59:1095-1114. [PMID: 38747072 PMCID: PMC11626700 DOI: 10.1111/jre.13273] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 04/09/2024] [Accepted: 04/11/2024] [Indexed: 12/10/2024]
Abstract
Dental implant surfaces and their unique properties can interact with the surrounding oral tissues through epigenetic cues. The present scoping review provides current perspectives on surface modifications of dental implants, their impact on the osseointegration process, and the interaction between implant surface properties and epigenetics, also in peri-implant diseases. Findings of this review demonstrate the impact of innovative surface treatments on the epigenetic mechanisms of cells, showing promising results in the early stages of osseointegration. Dental implant surfaces with properties of hydrophilicity, nanotexturization, multifunctional coatings, and incorporated drug-release systems have demonstrated favorable outcomes for early bone adhesion, increased antibacterial features, and improved osseointegration. The interaction between modified surface morphologies, different chemical surface energies, and/or release of molecules within the oral tissues has been shown to influence epigenetic mechanisms of the surrounding tissues caused by a physical-chemical interaction. Epigenetic changes around dental implants in the state of health and disease are different. In conclusion, emerging approaches in surface modifications for dental implants functionalized with epigenetics have great potential with a significant impact on modulating bone healing during osseointegration.
Collapse
Affiliation(s)
- Marcel F. Kunrath
- Department of Biomaterials, Institute of Clinical Sciences, Sahlgrenska AcademyUniversity of GothenburgGöteborgSweden
- Department of Dentistry, School of Health and Life SciencesPontifical Catholic University of Rio Grande do Sul (PUCRS)Porto AlegreBrazil
| | - Carlos Garaicoa‐Pazmino
- Department of PeriodonticsUniversity of Iowa College of DentistryIowa CityIowaUSA
- Research Center, School of DentistryEspiritu Santo UniversitySamborondónEcuador
| | - Paula Milena Giraldo‐Osorno
- Department of Biomaterials, Institute of Clinical Sciences, Sahlgrenska AcademyUniversity of GothenburgGöteborgSweden
| | - Aya Haj Mustafa
- Institute of Odontology, Sahlgrenska AcademyUniversity of GothenburgGöteborgSweden
| | - Christer Dahlin
- Department of Biomaterials, Institute of Clinical Sciences, Sahlgrenska AcademyUniversity of GothenburgGöteborgSweden
| | - Lena Larsson
- Department of Oral Biochemistry, Institute of Odontology, Sahlgrenska AcademyUniversity of GothenburgGöteborgSweden
| | - Farah Asa'ad
- Department of Biomaterials, Institute of Clinical Sciences, Sahlgrenska AcademyUniversity of GothenburgGöteborgSweden
- Department of Oral Biochemistry, Institute of Odontology, Sahlgrenska AcademyUniversity of GothenburgGöteborgSweden
| |
Collapse
|
2
|
Cotrut CM, Blidisel A, Vranceanu DM, Vladescu (Dragomir) A, Ungureanu E, Pana I, Dinu M, Vitelaru C, Parau AC, Pruna V, Magurean MS, Titorencu I. Evaluation of the In Vitro Behavior of Electrochemically Deposited Plate-like Crystal Hydroxyapatite Coatings. Biomimetics (Basel) 2024; 9:704. [PMID: 39590276 PMCID: PMC11592108 DOI: 10.3390/biomimetics9110704] [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: 10/24/2024] [Revised: 11/10/2024] [Accepted: 11/13/2024] [Indexed: 11/28/2024] Open
Abstract
The purpose of coatings is to protect or enhance the functionality of the substrate material, irrespective of the field in which the material was designed. The use of coatings in medicine is rapidly expanding with the objective of enhancing the osseointegration ability of metallic materials such as titanium. The aim of this study was to obtain biomimetic hydroxyapatite (HAp)-based coatings on titanium by using the pulsed galvanostatic method. The morphology of the HAp-based coatings revealed the presence of very thin and wide plate-like crystals, grown perpendicular to the Ti substrate, while the chemical composition highlighted a Ca/P ratio of 1.66, which is close to that of stoichiometric HAp (1.67). The main phases and chemical bonds identified confirmed the presence of the HAp phase in the developed coatings. A roughness of 228 nm and a contact angle of approx. 17° were obtained for the HAp coatings, highlighting a hydrophilic character. In terms of biomineralization and electrochemical behavior, it was shown that the HAp coatings have significantly enhanced the titanium properties. Finally, the in vitro cell tests carried out with human mesenchymal stem cells showed that the Ti samples coated with HAp have increased cell viability, extracellular matrix, and Ca intracellular deposition when compared with the uncoated Ti, indicating the beneficial effect.
Collapse
Affiliation(s)
- Cosmin M. Cotrut
- Faculty of Materials Science and Engineering, National University of Science and Technology Politehnica Bucharest, 313 Independentei Street, 060042 Bucharest, Romania; (C.M.C.); (E.U.)
| | - Alexandru Blidisel
- Hepato-Bilio-Pancreatic Surgery Center, University Clinic Surgical Semiology and Thoracic Surgery, “Victor Babes” University of Medicine and Pharmacy, Sq. Eftimie Murgu No. 2, 300041 Timisoara, Romania
| | - Diana M. Vranceanu
- Faculty of Materials Science and Engineering, National University of Science and Technology Politehnica Bucharest, 313 Independentei Street, 060042 Bucharest, Romania; (C.M.C.); (E.U.)
| | - Alina Vladescu (Dragomir)
- Department for Advanced Surface Processing and Analysis by Vacuum Technologies, National Institute of Research and Development for Optoelectronics—INOE 2000, 077125 Magurele, Romania
| | - Elena Ungureanu
- Faculty of Materials Science and Engineering, National University of Science and Technology Politehnica Bucharest, 313 Independentei Street, 060042 Bucharest, Romania; (C.M.C.); (E.U.)
| | - Iulian Pana
- Department for Advanced Surface Processing and Analysis by Vacuum Technologies, National Institute of Research and Development for Optoelectronics—INOE 2000, 077125 Magurele, Romania
| | - Mihaela Dinu
- Department for Advanced Surface Processing and Analysis by Vacuum Technologies, National Institute of Research and Development for Optoelectronics—INOE 2000, 077125 Magurele, Romania
| | - Catalin Vitelaru
- Department for Advanced Surface Processing and Analysis by Vacuum Technologies, National Institute of Research and Development for Optoelectronics—INOE 2000, 077125 Magurele, Romania
| | - Anca C. Parau
- Department for Advanced Surface Processing and Analysis by Vacuum Technologies, National Institute of Research and Development for Optoelectronics—INOE 2000, 077125 Magurele, Romania
| | - Vasile Pruna
- Romanian Academy Institute of Cellular Biology and Pathology “Nicolae Simionescu”, 8 B.P. Hasdeu, 050568 Bucharest, Romania
| | | | - Irina Titorencu
- Romanian Academy Institute of Cellular Biology and Pathology “Nicolae Simionescu”, 8 B.P. Hasdeu, 050568 Bucharest, Romania
| |
Collapse
|
3
|
Jiang Y, Harberts J, Assadi A, Chen Y, Spatz JP, Duan W, Nisbet DR, Voelcker NH, Elnathan R. The Roles of Micro- and Nanoscale Materials in Cell-Engineering Systems. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2410908. [PMID: 39401098 DOI: 10.1002/adma.202410908] [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: 07/26/2024] [Revised: 09/13/2024] [Indexed: 11/29/2024]
Abstract
Customizable manufacturing of ex vivo cell engineering is driven by the need for innovations in the biomedical field and holds substantial potential for addressing current therapeutic challenges; but it is still only in its infancy. Micro- and nanoscale-engineered materials are increasingly used to control core cell-level functions in cellular engineering. By reprogramming or redirecting targeted cells for extremely precise functions, these advanced materials offer new possibilities. This influences the modularity of cell reprogramming and reengineering, making these materials part of versatile and emerging technologies. Here, the roles of micro- and nanoscale materials in cell engineering are highlighted, demonstrating how they can be adaptively controlled to regulate cellular reprogramming and core cell-level functions, including differentiation, proliferation, adhesion, user-defined gene expression, and epigenetic changes. The current reprogramming routes used to achieve pluripotency from somatic cells and the significant potential of induced pluripotent stem cell technology for translational biomedical research are covered. Recent advances in nonviral intracellular delivery modalities for cell reprogramming and their constraints are evaluated. This paper focuses on emerging physical and combinatorial approaches of intracellular delivery for cell engineering, revealing the capabilities and limitations of these routes. It is showcased how these programmable materials are continually being explored as customizable tools for inducing biophysical stimulation. Harnessing the power of micro- and nanoscale-engineered materials will be a step change in the design of cell engineering, producing a suite of powerful tools for addressing potential future challenges in therapeutic cell engineering.
Collapse
Affiliation(s)
- Yuan Jiang
- Faculty of Health, School of Medicine, Deakin University, Waurn Ponds, Victoria, 3216, Australia
- The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Waurn Ponds, Victoria, 3216, Australia
| | - Jann Harberts
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, Victoria, Clayton, 3168, Australia
| | - Artin Assadi
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, Victoria, Clayton, 3168, Australia
| | - Yaping Chen
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052, Australia
- Oujiang Laboratory, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, Wenzhou Medical University, Zhejiang, 325000, China
| | - Joachim P Spatz
- Department of Cellular Biophysics, Max Planck Institute for Medical Research, 69120, Heidelberg, Germany
- Institute for Molecular Systems Engineering (IMSE), Heidelberg University, 69120, Heidelberg, Germany
- Max Planck School Matter to Life, Max Planck Schools, 69120, Heidelberg, Germany
| | - Wei Duan
- Faculty of Health, School of Medicine, Deakin University, Waurn Ponds, Victoria, 3216, Australia
| | - David R Nisbet
- The Graeme Clark Institute, University of Melbourne, Parkville, Victoria, 3010, Australia
- Department of Biomedical Engineering, Faculty of Engineering and Information Technology, University of Melbourne, Parkville, Victoria, 3010, Australia
- Medical School, Faculty of Medicine, Dentistry and Health Science, The University of Melbourne, Melbourne, Parkville, VIC, 3010, Australia
| | - Nicolas H Voelcker
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, Victoria, Clayton, 3168, Australia
| | - Roey Elnathan
- Faculty of Health, School of Medicine, Deakin University, Waurn Ponds, Victoria, 3216, Australia
- The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Waurn Ponds, Victoria, 3216, Australia
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, Victoria, Clayton, 3168, Australia
- Institute for Frontier Materials, Deakin University, Waurn Ponds, Victoria, 3216, Australia
| |
Collapse
|
4
|
Castilla-Casadiego DA, Loh DH, Pineda-Hernandez A, Rosales AM. Stimuli-Responsive Substrates to Control the Immunomodulatory Potential of Stromal Cells. Biomacromolecules 2024; 25:6319-6337. [PMID: 39283807 PMCID: PMC11506505 DOI: 10.1021/acs.biomac.4c00835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/06/2024]
Abstract
Mesenchymal stromal cells (MSCs) have broad immunomodulatory properties that range from regulation, proliferation, differentiation, and immune cell activation to secreting bioactive molecules that inhibit inflammation and regulate immune response. These properties provide MSCs with high therapeutic potency that has been shown to be relevant to tissue engineering and regenerative medicine. Hence, researchers have explored diverse strategies to control the immunomodulatory potential of stromal cells using polymeric substrates or scaffolds. These substrates alter the immunomodulatory response of MSCs, especially through biophysical cues such as matrix mechanical properties. To leverage these cell-matrix interactions as a strategy for priming MSCs, emerging studies have explored the use of stimuli-responsive substrates to enhance the therapeutic value of stromal cells. This review highlights how stimuli-responsive materials, including chemo-responsive, microenvironment-responsive, magneto-responsive, mechano-responsive, and photo-responsive substrates, have specifically been used to promote the immunomodulatory potential of stromal cells by controlling their secretory activity.
Collapse
Affiliation(s)
- David A Castilla-Casadiego
- Mcketta Department of Chemical Engineering, University of Texas at Austin, Austin, Texas 78712, United States
| | - Darren H Loh
- Mcketta Department of Chemical Engineering, University of Texas at Austin, Austin, Texas 78712, United States
| | - Aldaly Pineda-Hernandez
- Mcketta Department of Chemical Engineering, University of Texas at Austin, Austin, Texas 78712, United States
| | - Adrianne M Rosales
- Mcketta Department of Chemical Engineering, University of Texas at Austin, Austin, Texas 78712, United States
| |
Collapse
|
5
|
He X, Yamada M, Watanabe J, Pengyu Q, Chen J, Egusa H. Titanium nanotopography enhances mechano-response of osteocyte three-dimensional network toward osteoblast activation. BIOMATERIALS ADVANCES 2024; 163:213939. [PMID: 38954876 DOI: 10.1016/j.bioadv.2024.213939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 06/19/2024] [Accepted: 06/27/2024] [Indexed: 07/04/2024]
Abstract
The bone turnover capability influences the acquisition and maintenance of osseointegration. The architectures of osteocyte three-dimensional (3D) networks determine the direction and activity of bone turnover through osteocyte intercellular crosstalk, which exchanges prostaglandins through gap junctions in response to mechanical loading. Titanium nanosurfaces with anisotropically patterned dense nanospikes promote the development of osteocyte lacunar-canalicular networks. We investigated the effects of titanium nanosurfaces on intercellular network development and regulatory capabilities of bone turnover in osteocytes under cyclic compressive loading. MLO-Y4 mouse osteocyte-like cell lines embedded in type I collagen 3D gels on titanium nanosurfaces promoted the formation of intercellular networks and gap junctions even under static culture conditions, in contrast to the poor intercellular connectivity in machined titanium surfaces. The osteocyte 3D network on the titanium nanosurfaces further enhanced gap junction formation after additional culturing under cyclic compressive loading simulating masticatory loading, beyond the degree observed on machined titanium surfaces. A prostaglandin synthesis inhibitor cancelled the dual effects of titanium nanosurfaces and cyclic compressive loading on the upregulation of gap junction-related genes in the osteocyte 3D culture. Supernatants from osteocyte monolayer culture on titanium nanosurfaces promoted osteocyte maturation and intercellular connections with gap junctions. With cyclic loading, titanium nanosurfaces induced expression of the regulatory factors of bone turnover in osteocyte 3D cultures, toward higher osteoblast activation than that observed on machined surfaces. Titanium nanosurfaces with anisotropically patterned dense nanospikes promoted intercellular 3D network development and regulatory function toward osteoblast activation in osteocytes activated by cyclic compressive loading, through intercellular crosstalk by prostaglandin.
Collapse
Affiliation(s)
- Xindie He
- Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, Sendai, Miyagi, Japan; Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, Fujian, PR China
| | - Masahiro Yamada
- Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, Sendai, Miyagi, Japan.
| | - Jun Watanabe
- Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, Sendai, Miyagi, Japan
| | - Qu Pengyu
- Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, Sendai, Miyagi, Japan
| | - Jiang Chen
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, Fujian, PR China
| | - Hiroshi Egusa
- Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, Sendai, Miyagi, Japan; Center for Advanced Stem Cell and Regenerative Research, Tohoku University Graduate School of Dentistry, Sendai, Miyagi, Japan.
| |
Collapse
|
6
|
Wang H, Weng X, Chen Y, Mao S, Gao Y, Wu Q, Huang Y, Guan X, Xu Z, Lai Y. Biomimetic concentric microgrooved titanium surfaces influence bone marrow-derived mesenchymal stem cell osteogenic differentiation via H3K4 trimethylation epigenetic regulation. Dent Mater J 2024; 43:683-692. [PMID: 39135261 DOI: 10.4012/dmj.2023-327] [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: 10/01/2024]
Abstract
Material surface micromorphology can modulate cellular behavior and promote osteogenic differentiation through cytoskeletal rearrangement. Bone reconstruction requires precise regulation of gene expression in cells, a process governed by epigenetic mechanisms such as histone modifications, DNA methylation, and chromatin remodeling. We constructed osteon-mimetic concentric microgrooved titanium surfaces with different groove sizes and cultured bone marrow-derived mesenchymal stem cells (BMSCs) on the material surfaces to study how they regulate cell biological behavior and osteogenic differentiation through epigenetics. We found that the cells arranged in concentric circles along the concentric structure in the experimental group, and the concentric microgrooved surface did not inhibit cell proliferation. The results of a series of osteogenic differentiation experiments showed that the concentric microgrooves facilitated calcium deposition and promoted osteogenic differentiation of the BMSCs. Concentric microgrooved titanium surfaces that were 30 μm wide and 10 μm deep promoted osteogenic differentiation of BMSC by increasing WDR5 expression via H3K4 trimethylation upregulation.
Collapse
Affiliation(s)
- Hong Wang
- Department of Stomatology, Engineering Research Center of Fujian University for Stomatological Biomaterials, Xiamen Medical College
- Stomatological Hospital of Xiamen Medical college
| | - Xinze Weng
- Department of Stomatology, Engineering Research Center of Fujian University for Stomatological Biomaterials, Xiamen Medical College
| | - Yan Chen
- Department of Stomatology, Engineering Research Center of Fujian University for Stomatological Biomaterials, Xiamen Medical College
| | - Shunjie Mao
- Department of Stomatology, Engineering Research Center of Fujian University for Stomatological Biomaterials, Xiamen Medical College
| | - Yuerong Gao
- Department of Stomatology of The Third Affiliated Hospital of Xi'an Medical University
| | - Qinglin Wu
- Department of Stomatology, Engineering Research Center of Fujian University for Stomatological Biomaterials, Xiamen Medical College
| | - Yanling Huang
- Department of Stomatology, Engineering Research Center of Fujian University for Stomatological Biomaterials, Xiamen Medical College
| | - Xin Guan
- Department of Stomatology, Engineering Research Center of Fujian University for Stomatological Biomaterials, Xiamen Medical College
| | - Zhiqiang Xu
- Department of Stomatology, Affiliated Hospital of Putian University
| | - Yingzhen Lai
- Department of Stomatology, Engineering Research Center of Fujian University for Stomatological Biomaterials, Xiamen Medical College
| |
Collapse
|
7
|
Davis ZG, Koch DW, Watson SL, Scull GM, Brown AC, Schnabel LV, Fisher MB. Controlled Stiffness of Direct-Write, Near-Field Electrospun Gelatin Fibers Generates Differences in Tenocyte Morphology and Gene Expression. J Biomech Eng 2024; 146:091008. [PMID: 38529730 PMCID: PMC11080953 DOI: 10.1115/1.4065163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 03/18/2024] [Accepted: 03/18/2024] [Indexed: 03/27/2024]
Abstract
Tendinopathy is a leading cause of mobility issues. Currently, the cell-matrix interactions involved in the development of tendinopathy are not fully understood. In vitro tendon models provide a unique tool for addressing this knowledge gap as they permit fine control over biochemical, micromechanical, and structural aspects of the local environment to explore cell-matrix interactions. In this study, direct-write, near-field electrospinning of gelatin solution was implemented to fabricate micron-scale fibrous scaffolds that mimic native collagen fiber size and orientation. The stiffness of these fibrous scaffolds was found to be controllable between 1 MPa and 8 MPa using different crosslinking methods (EDC, DHT, DHT+EDC) or through altering the duration of crosslinking with EDC (1 h to 24 h). EDC crosslinking provided the greatest fiber stability, surviving up to 3 weeks in vitro. Differences in stiffness resulted in phenotypic changes for equine tenocytes with low stiffness fibers (∼1 MPa) promoting an elongated nuclear aspect ratio while those on high stiffness fibers (∼8 MPa) were rounded. High stiffness fibers resulted in the upregulation of matrix metalloproteinase (MMPs) and proteoglycans (possible indicators for tendinopathy) relative to low stiffness fibers. These results demonstrate the feasibility of direct-written gelatin scaffolds as tendon in vitro models and provide evidence that matrix mechanical properties may be crucial factors in cell-matrix interactions during tendinopathy formation.
Collapse
Affiliation(s)
- Zachary G. Davis
- Joint Department of Biomedical Engineering, North Carolina State University, University of North Carolina at Chapel Hill, Raleigh, NC 27695; Comparative Medicine Institute, North Carolina State University, Raleigh, NC 27695
| | - Drew W. Koch
- College of Veterinary Medicine, North Carolina State University, Raleigh, NC 27695; Comparative Medicine Institute, North Carolina State University, Raleigh, NC 27695
- North Carolina State University
| | - Samantha L. Watson
- Joint Department of Biomedical Engineering, North Carolina State University, University of North Carolina at Chapel Hill, Raleigh, NC 27695
| | - Grant M. Scull
- Joint Department of Biomedical Engineering, North Carolina State University, University of North Carolina at Chapel Hill, Raleigh, NC 27695; Comparative Medicine Institute, North Carolina State University, Raleigh, NC 27695
| | - Ashley C. Brown
- Joint Department of Biomedical Engineering, North Carolina State University, University of North Carolina at Chapel Hill, Raleigh, NC 27695; Comparative Medicine Institute, North Carolina State University, Raleigh, NC 27695
| | - Lauren V. Schnabel
- College of Veterinary Medicine, North Carolina State University, Raleigh, NC 27695; Comparative Medicine Institute, North Carolina State University, Raleigh, NC 27695
- North Carolina State University
| | - Matthew B. Fisher
- Joint Department of Biomedical Engineering, North Carolina State University, University of North Carolina at Chapel Hill, Raleigh, NC 27695; Comparative Medicine Institute, North Carolina State University, Raleigh, NC 27695; Department of Orthopaedics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| |
Collapse
|
8
|
Ferrai C, Schulte C. Mechanotransduction in stem cells. Eur J Cell Biol 2024; 103:151417. [PMID: 38729084 DOI: 10.1016/j.ejcb.2024.151417] [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: 12/27/2023] [Revised: 04/28/2024] [Accepted: 04/29/2024] [Indexed: 05/12/2024] Open
Abstract
Nowadays, it is an established concept that the capability to reach a specialised cell identity via differentiation, as in the case of multi- and pluripotent stem cells, is not only determined by biochemical factors, but that also physical aspects of the microenvironment play a key role; interpreted by the cell through a force-based signalling pathway called mechanotransduction. However, the intricate ties between the elements involved in mechanotransduction, such as the extracellular matrix, the glycocalyx, the cell membrane, Integrin adhesion complexes, Cadherin-mediated cell/cell adhesion, the cytoskeleton, and the nucleus, are still far from being understood in detail. Here we report what is currently known about these elements in general and their specific interplay in the context of multi- and pluripotent stem cells. We furthermore merge this overview to a more comprehensive picture, that aims to cover the whole mechanotransductive pathway from the cell/microenvironment interface to the regulation of the chromatin structure in the nucleus. Ultimately, with this review we outline the current picture of the interplay between mechanotransductive cues and epigenetic regulation and how these processes might contribute to stem cell dynamics and fate.
Collapse
Affiliation(s)
- Carmelo Ferrai
- Institute of Pathology, University Medical Centre Göttingen, Germany.
| | - Carsten Schulte
- Department of Biomedical and Clinical Sciences and Department of Physics "Aldo Pontremoli", University of Milan, Italy.
| |
Collapse
|
9
|
Chen Y, Li Y, Zhu W, Liu Q. Biomimetic gradient scaffolds for the tissue engineering and regeneration of rotator cuff enthesis. Biofabrication 2024; 16:032005. [PMID: 38697099 DOI: 10.1088/1758-5090/ad467d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 05/02/2024] [Indexed: 05/04/2024]
Abstract
Rotator cuff tear is one of the most common musculoskeletal disorders, which often results in recurrent shoulder pain and limited movement. Enthesis is a structurally complex and functionally critical interface connecting tendon and bone that plays an essential role in maintaining integrity of the shoulder joint. Despite the availability of advanced surgical procedures for rotator cuff repair, there is a high rate of failure following surgery due to suboptimal enthesis healing and regeneration. Novel strategies based on tissue engineering are gaining popularity in improving tendon-bone interface (TBI) regeneration. Through incorporating physical and biochemical cues into scaffold design which mimics the structure and composition of native enthesis is advantageous to guide specific differentiation of seeding cells and facilitate the formation of functional tissues. In this review, we summarize the current state of research in enthesis tissue engineering highlighting the development and application of biomimetic scaffolds that replicate the gradient TBI. We also discuss the latest techniques for fabricating potential translatable scaffolds such as 3D bioprinting and microfluidic device. While preclinical studies have demonstrated encouraging results of biomimetic gradient scaffolds, the translation of these findings into clinical applications necessitates a comprehensive understanding of their safety and long-term efficacy.
Collapse
Affiliation(s)
- Yang Chen
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Yexin Li
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Weihong Zhu
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Qian Liu
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, Changsha, People's Republic of China
| |
Collapse
|
10
|
Wu Y, Song Y, Soto J, Hoffman T, Zhang A, Han X, Fang Z, Eoh J, Gu L, Gu Z, Li S. Viscoelastic Extracellular Matrix Enhances Epigenetic Remodeling and Cellular Plasticity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.14.589442. [PMID: 38659850 PMCID: PMC11042188 DOI: 10.1101/2024.04.14.589442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Living tissue and extracellular matrices possess viscoelastic properties, but understanding how viscoelastic matrix regulates chromatin and the epigenome is limited. Here, we find that the regulation of the epigenetic state by the viscoelastic matrix is more pronounced on softer matrices. Cells on viscoelastic matrices exhibit larger nuclei, increased nuclear lamina ruffling, loosely organized chromatin, and faster chromatin dynamics, compared to those on elastic matrices. These changes are accompanied by a global increase in euchromatic marks and a local increase in chromatin accessibility at the cis -regulatory elements associated with neuronal and pluripotent genes. Consequently, viscoelastic matrices enhanced the efficiency of reprogramming fibroblasts into neurons and induced pluripotent stem cells, respectively. Together, our findings demonstrate the key roles of matrix viscoelasticity in the regulation of epigenetic state, and uncover a new mechanism of biophysical regulation of chromatin and cell reprogramming, with implications for the design of smart materials to engineer cell fate.
Collapse
|
11
|
Kim NY, Choi YY, Kim TH, Ha JH, Kim TH, Kang T, Chung BG. Synergistic Effect of Electrical and Biochemical Stimulation on Human iPSC-Derived Neural Differentiation in a Microfluidic Electrode Array Chip. ACS APPLIED MATERIALS & INTERFACES 2024; 16:15730-15740. [PMID: 38527279 DOI: 10.1021/acsami.3c17108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/27/2024]
Abstract
Neural differentiation is crucial for advancing our understanding of the nervous system and developing treatments for neurological disorders. The advanced methods and the ability to manipulate the alignment, proliferation, and differentiation of stem cells are essential for studying neuronal development and synaptic interactions. However, the utilization of human induced pluripotent stem cells (iPSCs) for disease modeling of neurodegenerative conditions may be constrained by the prolonged duration and uncontrolled cell differentiation required for functional neural cell differentiation. Here, we developed a microfluidic chip to enhance the differentiation and maturation of specific neural lineages by placing aligned microelectrodes on the glass surface to regulate the neural differentiation of human iPSCs. The utilization of electrical stimulation (ES) in conjunction with neurotrophic factors (NF) significantly enhanced the efficiency in generating functional neurons from human iPSCs. We also observed that the simultaneous application of NF and ES to human iPSCs promoted their differentiation and maturation into functional neurons while increasing synaptic interactions. Our research demonstrated the effect of combining NF and ES on human iPSC-derived neural differentiation.
Collapse
Affiliation(s)
- Na Yeon Kim
- Department of Biomedical Engineering, Sogang University, Seoul 04107, Korea
| | - Yoon Young Choi
- Institute of Integrated Biotechnology, Sogang University, Seoul 04107, Korea
| | - Tae Hyeon Kim
- Department of Mechanical Engineering, Sogang University, Seoul 04107, Korea
| | - Jang Ho Ha
- Department of Mechanical Engineering, Sogang University, Seoul 04107, Korea
| | - Tae-Hyung Kim
- School of Integrative Engineering, Chung-Ang University, Seoul 06974, Korea
| | - Taewook Kang
- Institute of Integrated Biotechnology, Sogang University, Seoul 04107, Korea
- Department of Chemical and Biomolecular Engineering, Sogang University, Seoul 04107, Korea
| | - Bong Geun Chung
- Department of Biomedical Engineering, Sogang University, Seoul 04107, Korea
- Institute of Integrated Biotechnology, Sogang University, Seoul 04107, Korea
- Department of Mechanical Engineering, Sogang University, Seoul 04107, Korea
- Institute of Smart Biosensor, Sogang University, Seoul 04107, Korea
| |
Collapse
|
12
|
Xuan Y, Guo Y, Li L, Yuzhang, Zhang C, RuiJin, Yin X, Zhang Z. 3D-printed bredigite scaffolds with ordered arrangement structures promote bone regeneration by inducing macrophage polarization in onlay grafts. J Nanobiotechnology 2024; 22:102. [PMID: 38468312 PMCID: PMC10926610 DOI: 10.1186/s12951-024-02362-2] [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: 11/07/2023] [Accepted: 02/21/2024] [Indexed: 03/13/2024] Open
Abstract
Bone tissue engineering scaffolds may provide a potential strategy for onlay bone grafts for oral implants. For determining the fate of scaffold biomaterials and osteogenesis effects, the host immune response is crucial. In the present study, bredigite (BRT) bioceramic scaffolds with an ordered arrangement structure (BRT-O) and a random morphology (BRT-R) were fabricated. The physicochemical properties of scaffolds were first characterized by scanning electron microscopy, mechanical test and micro-Fourier transform infrared spectroscopy. In addition, their osteogenic and immunomodulatory properties in an onlay grafting model were investigated. In vitro, the BRT-O scaffolds facilitated the macrophage polarization towards a pro-regenerative M2 phenotype, which subsequently facilitated the migration and osteogenic differentiation of bone marrow-derived mesenchymal stem cells. In vivo, an onlay grafting model was successfully established in the cranium of rabbits. In addition, the BRT-O scaffolds grafted on rabbit cranium promoted bone regeneration and CD68 + CD206 + M2 macrophage polarization. In conclusion, the 3D-printed BRT-O scaffold presents as a promising scaffold biomaterial for onlay grafts by regulating the local immune microenvironment.
Collapse
Affiliation(s)
- Yaowei Xuan
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Department of Periodontology, School of Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, The Fourth Military Medical University, Xi'an, 710032, China
| | - Yibo Guo
- Department of Oral & Maxillofacial-Head & Neck Oncology, Shanghai Ninth People's Hospital, College of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai, 200011, China
| | - Lin Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Department of Periodontology, School of Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, The Fourth Military Medical University, Xi'an, 710032, China
| | - Yuzhang
- Department of Oral and Maxillofacial Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Chenping Zhang
- Department of Oral & Maxillofacial-Head & Neck Oncology, Shanghai Ninth People's Hospital, College of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai, 200011, China
| | - RuiJin
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Department of Periodontology, School of Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, The Fourth Military Medical University, Xi'an, 710032, China
| | - Xuelai Yin
- Department of Oral & Maxillofacial-Head & Neck Oncology, Shanghai Ninth People's Hospital, College of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai, 200011, China.
| | - Zhen Zhang
- Department of Oral and Maxillofacial Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
- Department of Oral & Maxillofacial-Head & Neck Oncology, Shanghai Ninth People's Hospital, College of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai, 200011, China.
| |
Collapse
|
13
|
Mishra J, Chakraborty S, Niharika, Roy A, Manna S, Baral T, Nandi P, Patra SK. Mechanotransduction and epigenetic modulations of chromatin: Role of mechanical signals in gene regulation. J Cell Biochem 2024; 125:e30531. [PMID: 38345428 DOI: 10.1002/jcb.30531] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 01/08/2024] [Accepted: 01/26/2024] [Indexed: 03/12/2024]
Abstract
Mechanical forces may be generated within a cell due to tissue stiffness, cytoskeletal reorganization, and the changes (even subtle) in the cell's physical surroundings. These changes of forces impose a mechanical tension within the intracellular protein network (both cytosolic and nuclear). Mechanical tension could be released by a series of protein-protein interactions often facilitated by membrane lipids, lectins and sugar molecules and thus generate a type of signal to drive cellular processes, including cell differentiation, polarity, growth, adhesion, movement, and survival. Recent experimental data have accentuated the molecular mechanism of this mechanical signal transduction pathway, dubbed mechanotransduction. Mechanosensitive proteins in the cell's plasma membrane discern the physical forces and channel the information to the cell interior. Cells respond to the message by altering their cytoskeletal arrangement and directly transmitting the signal to the nucleus through the connection of the cytoskeleton and nucleoskeleton before the information despatched to the nucleus by biochemical signaling pathways. Nuclear transmission of the force leads to the activation of chromatin modifiers and modulation of the epigenetic landscape, inducing chromatin reorganization and gene expression regulation; by the time chemical messengers (transcription factors) arrive into the nucleus. While significant research has been done on the role of mechanotransduction in tumor development and cancer progression/metastasis, the mechanistic basis of force-activated carcinogenesis is still enigmatic. Here, in this review, we have discussed the various cues and molecular connections to better comprehend the cellular mechanotransduction pathway, and we also explored the detailed role of some of the multiple players (proteins and macromolecular complexes) involved in mechanotransduction. Thus, we have described an avenue: how mechanical stress directs the epigenetic modifiers to modulate the epigenome of the cells and how aberrant stress leads to the cancer phenotype.
Collapse
Affiliation(s)
- Jagdish Mishra
- Epigenetics and Cancer Research Laboratory, Department of Life Science, Biochemistry and Molecular Biology Group, National Institute of Technology, Rourkela, Odisha, India
| | - Subhajit Chakraborty
- Epigenetics and Cancer Research Laboratory, Department of Life Science, Biochemistry and Molecular Biology Group, National Institute of Technology, Rourkela, Odisha, India
| | - Niharika
- Epigenetics and Cancer Research Laboratory, Department of Life Science, Biochemistry and Molecular Biology Group, National Institute of Technology, Rourkela, Odisha, India
| | - Ankan Roy
- Epigenetics and Cancer Research Laboratory, Department of Life Science, Biochemistry and Molecular Biology Group, National Institute of Technology, Rourkela, Odisha, India
| | - Soumen Manna
- Epigenetics and Cancer Research Laboratory, Department of Life Science, Biochemistry and Molecular Biology Group, National Institute of Technology, Rourkela, Odisha, India
| | - Tirthankar Baral
- Epigenetics and Cancer Research Laboratory, Department of Life Science, Biochemistry and Molecular Biology Group, National Institute of Technology, Rourkela, Odisha, India
| | - Piyasa Nandi
- Epigenetics and Cancer Research Laboratory, Department of Life Science, Biochemistry and Molecular Biology Group, National Institute of Technology, Rourkela, Odisha, India
| | - Samir K Patra
- Epigenetics and Cancer Research Laboratory, Department of Life Science, Biochemistry and Molecular Biology Group, National Institute of Technology, Rourkela, Odisha, India
| |
Collapse
|
14
|
Thomasy SM, Leonard BC, Greiner MA, Skeie JM, Raghunathan VK. Squishy matters - Corneal mechanobiology in health and disease. Prog Retin Eye Res 2024; 99:101234. [PMID: 38176611 PMCID: PMC11193890 DOI: 10.1016/j.preteyeres.2023.101234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 12/22/2023] [Accepted: 12/27/2023] [Indexed: 01/06/2024]
Abstract
The cornea, as a dynamic and responsive tissue, constantly interacts with mechanical forces in order to maintain its structural integrity, barrier function, transparency and refractive power. Cells within the cornea sense and respond to various mechanical forces that fundamentally regulate their morphology and fate in development, homeostasis and pathophysiology. Corneal cells also dynamically regulate their extracellular matrix (ECM) with ensuing cell-ECM crosstalk as the matrix serves as a dynamic signaling reservoir providing biophysical and biochemical cues to corneal cells. Here we provide an overview of mechanotransduction signaling pathways then delve into the recent advances in corneal mechanobiology, focusing on the interplay between mechanical forces and responses of the corneal epithelial, stromal, and endothelial cells. We also identify species-specific differences in corneal biomechanics and mechanotransduction to facilitate identification of optimal animal models to study corneal wound healing, disease, and novel therapeutic interventions. Finally, we identify key knowledge gaps and therapeutic opportunities in corneal mechanobiology that are pressing for the research community to address especially pertinent within the domains of limbal stem cell deficiency, keratoconus and Fuchs' endothelial corneal dystrophy. By furthering our understanding corneal mechanobiology, we can contextualize discoveries regarding corneal diseases as well as innovative treatments for them.
Collapse
Affiliation(s)
- Sara M Thomasy
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California - Davis, Davis, CA, United States; Department of Ophthalmology & Vision Science, School of Medicine, University of California - Davis, Davis, CA, United States; California National Primate Research Center, Davis, CA, United States.
| | - Brian C Leonard
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California - Davis, Davis, CA, United States; Department of Ophthalmology & Vision Science, School of Medicine, University of California - Davis, Davis, CA, United States
| | - Mark A Greiner
- Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, IA, United States; Iowa Lions Eye Bank, Coralville, IA, United States
| | - Jessica M Skeie
- Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, IA, United States; Iowa Lions Eye Bank, Coralville, IA, United States
| | | |
Collapse
|
15
|
Lopes V, Moreira G, Bramini M, Capasso A. The potential of graphene coatings as neural interfaces. NANOSCALE HORIZONS 2024; 9:384-406. [PMID: 38231692 DOI: 10.1039/d3nh00461a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
Recent advances in nanotechnology design and fabrication have shaped the landscape for the development of ideal cell interfaces based on biomaterials. A holistic evaluation of the requirements for a cell interface is a highly complex task. Biocompatibility is a crucial requirement which is affected by the interface's properties, including elemental composition, morphology, and surface chemistry. This review explores the current state-of-the-art on graphene coatings produced by chemical vapor deposition (CVD) and applied as neural interfaces, detailing the key properties required to design an interface capable of physiologically interacting with neural cells. The interfaces are classified into substrates and scaffolds to differentiate the planar and three-dimensional environments where the cells can adhere and proliferate. The role of specific features such as mechanical properties, porosity and wettability are investigated. We further report on the specific brain-interface applications where CVD graphene paved the way to revolutionary advances in biomedicine. Future studies on the long-term effects of graphene-based materials in vivo will unlock even more potentially disruptive neuro-applications.
Collapse
Affiliation(s)
- Vicente Lopes
- International Iberian Nanotechnology Laboratory, 4715-330 Braga, Portugal.
| | - Gabriel Moreira
- International Iberian Nanotechnology Laboratory, 4715-330 Braga, Portugal.
| | - Mattia Bramini
- Department of Cell Biology, Facultad de Ciencias, Universidad de Granada, 18071 Granada, Spain.
| | - Andrea Capasso
- International Iberian Nanotechnology Laboratory, 4715-330 Braga, Portugal.
| |
Collapse
|
16
|
Song Y, Soto J, Wong SY, Wu Y, Hoffman T, Akhtar N, Norris S, Chu J, Park H, Kelkhoff DO, Ang CE, Wernig M, Kasko A, Downing TL, Poo MM, Li S. Biphasic regulation of epigenetic state by matrix stiffness during cell reprogramming. SCIENCE ADVANCES 2024; 10:eadk0639. [PMID: 38354231 PMCID: PMC10866547 DOI: 10.1126/sciadv.adk0639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 01/12/2024] [Indexed: 02/16/2024]
Abstract
We investigate how matrix stiffness regulates chromatin reorganization and cell reprogramming and find that matrix stiffness acts as a biphasic regulator of epigenetic state and fibroblast-to-neuron conversion efficiency, maximized at an intermediate stiffness of 20 kPa. ATAC sequencing analysis shows the same trend of chromatin accessibility to neuronal genes at these stiffness levels. Concurrently, we observe peak levels of histone acetylation and histone acetyltransferase (HAT) activity in the nucleus on 20 kPa matrices, and inhibiting HAT activity abolishes matrix stiffness effects. G-actin and cofilin, the cotransporters shuttling HAT into the nucleus, rises with decreasing matrix stiffness; however, reduced importin-9 on soft matrices limits nuclear transport. These two factors result in a biphasic regulation of HAT transport into nucleus, which is directly demonstrated on matrices with dynamically tunable stiffness. Our findings unravel a mechanism of the mechano-epigenetic regulation that is valuable for cell engineering in disease modeling and regenerative medicine applications.
Collapse
Affiliation(s)
- Yang Song
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Jennifer Soto
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Sze Yue Wong
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Yifan Wu
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Tyler Hoffman
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Navied Akhtar
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92617, USA
| | - Sam Norris
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Julia Chu
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Hyungju Park
- Division of Neurobiology, Department of Molecular and Cell Biology, Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA
- Department of Structure and Function of Neural Network, Korea Brain Research Institute (KBRI), Daegu 41068, South Korea
| | - Douglas O. Kelkhoff
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Cheen Euong Ang
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
- Department of Pathology and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA 94305, USA
| | - Marius Wernig
- Department of Pathology and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA 94305, USA
| | - Andrea Kasko
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Timothy L. Downing
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA 92617, USA
| | - Mu-ming Poo
- Division of Neurobiology, Department of Molecular and Cell Biology, Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA
- Institute of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Song Li
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
17
|
Chu Q, Han W, He Z, Hao L, Fu X. Suppression of LPS-activated inflammatory responses and chromosomal histone modifications in macrophages by micropattern-induced nuclear deformation. J Biomed Mater Res A 2024; 112:250-259. [PMID: 37740539 DOI: 10.1002/jbm.a.37617] [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: 05/11/2023] [Revised: 08/24/2023] [Accepted: 09/07/2023] [Indexed: 09/24/2023]
Abstract
Macrophages are important immune effector cells which participate various physiological and pathological conditions. Numerous studies have demonstrated the regulation of macrophage phenotype by micropatterns. It is well accepted that micropatterns affect cellular behaviors through changing cell shape and modulating the associated mechanical sensors on the plasma membrane and cytoskeleton. However, the role of nucleus, which serves as a critical physical sensing device, is often ignored. Herein, we found the nuclear deformation and the subsequently increased chromosomal histone methylation (H3K36me2) may contribute to the micropattern-induced suppression of macrophage inflammatory responses. Specifically, macrophages on micropatterned surfaces expressed lower levels of key inflammatory genes, compared with those on flat surfaces. Further investigation on macrophage nuclei showed that micropatterned surfaces cause shrinkage of nucleus volume and compaction of chromatin. Moreover, micropatterned surfaces elevated the methylation level of H3K36me2 in macrophages, while decreased the methylation level of H3K4me3. Our study provides new mechanistic insight into how micropatterns affect macrophage phenotype and highlights the importance of nuclear shape and chromatin histone modification in mediating micropattern-induced change in cell behaviors.
Collapse
Affiliation(s)
- Qi Chu
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, People's Republic of China
- National Engineering Research Center for Tissue Restoration and Reconstruction and Innovation Center for Tissue Restoration and Reconstruction, Guangzhou, People's Republic of China
- Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou, People's Republic of China
| | - Weiju Han
- National Engineering Research Center for Tissue Restoration and Reconstruction and Innovation Center for Tissue Restoration and Reconstruction, Guangzhou, People's Republic of China
- Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou, People's Republic of China
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, People's Republic of China
| | - Zhichun He
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, People's Republic of China
- National Engineering Research Center for Tissue Restoration and Reconstruction and Innovation Center for Tissue Restoration and Reconstruction, Guangzhou, People's Republic of China
- Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou, People's Republic of China
| | - Lijing Hao
- National Engineering Research Center for Tissue Restoration and Reconstruction and Innovation Center for Tissue Restoration and Reconstruction, Guangzhou, People's Republic of China
- Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou, People's Republic of China
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, People's Republic of China
| | - Xiaoling Fu
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, People's Republic of China
- National Engineering Research Center for Tissue Restoration and Reconstruction and Innovation Center for Tissue Restoration and Reconstruction, Guangzhou, People's Republic of China
- Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou, People's Republic of China
| |
Collapse
|
18
|
Song J, Zeng X, Li C, Yin H, Mao S, Ren D. Alteration in cartilage matrix stiffness as an indicator and modulator of osteoarthritis. Biosci Rep 2024; 44:BSR20231730. [PMID: 38014522 PMCID: PMC10794814 DOI: 10.1042/bsr20231730] [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: 10/11/2023] [Revised: 11/22/2023] [Accepted: 11/24/2023] [Indexed: 11/29/2023] Open
Abstract
Osteoarthritis (OA) is characterized by cartilage degeneration and destruction, leading to joint ankylosis and disability. The major challenge in diagnosing OA at early stage is not only lack of clinical symptoms but also the insufficient histological and immunohistochemical signs. Alteration in cartilage stiffness during OA progression, especially at OA initiation, has been confirmed by growing evidences. Moreover, the stiffness of cartilage extracellular matrix (ECM), pericellular matrix (PCM) and chondrocytes during OA development are dynamically changed in unique and distinct fashions, revealing possibly inconsistent conclusions when detecting cartilage matrix stiffness at different locations and scales. In addition, it will be discussed regarding the mechanisms through which OA-related cartilage degenerations exhibit stiffened or softened matrix, highlighting some critical events that generally incurred to cartilage stiffness alteration, as well as some typical molecules that participated in constituting the mechanical properties of cartilage. Finally, in vitro culturing chondrocytes in various stiffness-tunable scaffolds provided a reliable method to explore the matrix stiffness-dependent modulation of chondrocyte metabolism, which offers valuable information on optimizing implant scaffolds to maximally promote cartilage repair and regeneration during OA. Overall, this review systematically and comprehensively elucidated the current progresses in the relationship between cartilage stiffness alteration and OA progression. We hope that deeper attention and understanding in this researching field will not only develop more innovative methods in OA early detection and diagnose but also provide promising ideas in OA therapy and prognosis.
Collapse
Affiliation(s)
- Jing Song
- Qingdao University Affiliated Qingdao Women and Children’s Hospital, Department of Stomatology Medical Center, Qingdao University, Qingdao, Shandong, CN, China
| | - Xuemin Zeng
- The Affiliated Hospital of Qingdao University, Department of Stomatology Medical Center, Qingdao University, Qingdao, Shandong, CN, China
| | - Chenzhi Li
- The Affiliated Hospital of Qingdao University, Department of Stomatology Medical Center, Qingdao University, Qingdao, Shandong, CN, China
| | - Hongyan Yin
- Institute of Hybrid Materials, College of Materials Science and Engineering, Qingdao University, Qingdao, Shandong, CN, China
| | - Sui Mao
- Institute of Hybrid Materials, College of Materials Science and Engineering, Qingdao University, Qingdao, Shandong, CN, China
| | - Dapeng Ren
- The Affiliated Hospital of Qingdao University, Department of Stomatology Medical Center, Qingdao University, Qingdao, Shandong, CN, China
| |
Collapse
|
19
|
Suarato G, Pressi S, Menna E, Ruben M, Petrini EM, Barberis A, Miele D, Sandri G, Salerno M, Schirato A, Alabastri A, Athanassiou A, Proietti Zaccaria R, Papadopoulou EL. Modified Carbon Nanotubes Favor Fibroblast Growth by Tuning the Cell Membrane Potential. ACS APPLIED MATERIALS & INTERFACES 2024; 16:3093-3105. [PMID: 38206310 PMCID: PMC10811621 DOI: 10.1021/acsami.3c14527] [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: 09/28/2023] [Revised: 12/20/2023] [Accepted: 12/25/2023] [Indexed: 01/12/2024]
Abstract
As is known, carbon nanotubes favor cell growth in vitro, although the underlying mechanisms are not yet fully elucidated. In this study, we explore the hypothesis that electrostatic fields generated at the interface between nonexcitable cells and appropriate scaffold might favor cell growth by tuning their membrane potential. We focused on primary human fibroblasts grown on electrospun polymer fibers (poly(lactic acid)─PLA) with embedded multiwall carbon nanotubes (MWCNTs). The MWCNTs were functionalized with either the p-methoxyphenyl (PhOME) or the p-acetylphenyl (PhCOMe) moiety, both of which allowed uniform dispersion in a solvent, good mixing with PLA and the consequent smooth and homogeneous electrospinning process. The inclusion of the electrically conductive MWCNTs in the insulating PLA matrix resulted in differences in the surface potential of the fibers. Both PLA and PLA/MWCNT fiber samples were found to be biocompatible. The main features of fibroblasts cultured on different substrates were characterized by scanning electron microscopy, immunocytochemistry, Rt-qPCR, and electrophysiology revealing that fibroblasts grown on PLA/MWCNT reached a healthier state as compared to pure PLA. In particular, we observed physiological spreading, attachment, and Vmem of fibroblasts on PLA/MWCNT. Interestingly, the electrical functionalization of the scaffold resulted in a more suitable extracellular environment for the correct biofunctionality of these nonexcitable cells. Finally, numerical simulations were also performed in order to understand the mechanism behind the different cell behavior when grown either on PLA or PLA/MWCNT samples. The results show a clear effect on the cell membrane potential, depending on the underlying substrate.
Collapse
Affiliation(s)
- Giulia Suarato
- Istituto
Italiano di Tecnologia, via Morego 30, 16163 Genova, Italy
| | - Samuel Pressi
- Department
of Chemical Sciences, University of Padua, via Marzolo 1, 35131 Padova, Italy
- Interdepartmental
Centre Giorgio Levi Cases for Energy Economics and Technology, University of Padua, via Marzolo 9, 35131 Padova, Italy
| | - Enzo Menna
- Department
of Chemical Sciences, University of Padua, via Marzolo 1, 35131 Padova, Italy
- Interdepartmental
Centre Giorgio Levi Cases for Energy Economics and Technology, University of Padua, via Marzolo 9, 35131 Padova, Italy
| | - Massimo Ruben
- Istituto
Italiano di Tecnologia, via Morego 30, 16163 Genova, Italy
| | | | - Andrea Barberis
- Istituto
Italiano di Tecnologia, via Morego 30, 16163 Genova, Italy
| | - Dalila Miele
- Department
of Drug Sciences, University of Pavia, via Taramelli 12, 27100 Pavia, Italy
| | - Giuseppina Sandri
- Department
of Drug Sciences, University of Pavia, via Taramelli 12, 27100 Pavia, Italy
| | - Marco Salerno
- Istituto
Italiano di Tecnologia, via Morego 30, 16163 Genova, Italy
| | - Andrea Schirato
- Istituto
Italiano di Tecnologia, via Morego 30, 16163 Genova, Italy
- Dipartimento
di Fisica, Politecnico di Milano, Pizza Leonardo da Vinci 32, Milan 20133, Italy
- Department
of Electrical and Computer Engineering, Rice University, 6100 Main Street, Houston, Texas 77005, United States
| | - Alessandro Alabastri
- Department
of Electrical and Computer Engineering, Rice University, 6100 Main Street, Houston, Texas 77005, United States
| | | | | | | |
Collapse
|
20
|
Yang X, Xiong M, Fu X, Sun X. Bioactive materials for in vivo sweat gland regeneration. Bioact Mater 2024; 31:247-271. [PMID: 37637080 PMCID: PMC10457517 DOI: 10.1016/j.bioactmat.2023.07.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 07/30/2023] [Accepted: 07/30/2023] [Indexed: 08/29/2023] Open
Abstract
Loss of sweat glands (SwGs) commonly associated with extensive skin defects is a leading cause of hyperthermia and heat stroke. In vivo tissue engineering possesses the potential to take use of the body natural ability to regenerate SwGs, making it more conducive to clinical translation. Despite recent advances in regenerative medicine, reconstructing SwG tissue with the same structure and function as native tissue remains challenging. Elucidating the SwG generation mechanism and developing biomaterials for in vivo tissue engineering is essential for understanding and developing in vivo SwG regenerative strategies. Here, we outline the cell biology associated with functional wound healing and the characteristics of bioactive materials. We critically summarize the recent progress in bioactive material-based cell modulation approaches for in vivo SwG regeneration, including the recruitment of endogenous cells to the skin lesion for SwG regeneration and in vivo cellular reprogramming for SwG regeneration. We discussed the re-establishment of microenvironment via bioactive material-mediated regulators. Besides, we offer promising perspectives for directing in situ SwG regeneration via bioactive material-based cell-free strategy, which is a simple and effective approach to regenerate SwG tissue with both fidelity of structure and function. Finally, we discuss the opportunities and challenges of in vivo SwG regeneration in detail. The molecular mechanisms and cell fate modulation of in vivo SwG regeneration will provide further insights into the regeneration of patient-specific SwGs and the development of potential intervention strategies for gland-derived diseases.
Collapse
Affiliation(s)
- Xinling Yang
- Research Center for Tissue Repair and Regeneration Affiliated to Medical Innovation Research Department and 4th Medical Center, PLA General Hospital and PLA Medical College, China
- PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, China
- Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, PR China
| | - Mingchen Xiong
- Research Center for Tissue Repair and Regeneration Affiliated to Medical Innovation Research Department and 4th Medical Center, PLA General Hospital and PLA Medical College, China
- PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, China
- Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, PR China
| | - Xiaobing Fu
- Research Center for Tissue Repair and Regeneration Affiliated to Medical Innovation Research Department and 4th Medical Center, PLA General Hospital and PLA Medical College, China
- PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, China
- Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, PR China
| | - Xiaoyan Sun
- Research Center for Tissue Repair and Regeneration Affiliated to Medical Innovation Research Department and 4th Medical Center, PLA General Hospital and PLA Medical College, China
- PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, China
- Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, PR China
| |
Collapse
|
21
|
Zhang Z, Zhu H, Zhao G, Miao Y, Zhao L, Feng J, Zhang H, Miao R, Sun L, Gao B, Zhang W, Wang Z, Zhang J, Zhang Y, Guo H, Xu F, Lu TJ, Genin GM, Lin M. Programmable and Reversible Integrin-Mediated Cell Adhesion Reveals Hysteresis in Actin Kinetics that Alters Subsequent Mechanotransduction. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2302421. [PMID: 37849221 PMCID: PMC10724447 DOI: 10.1002/advs.202302421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 08/16/2023] [Indexed: 10/19/2023]
Abstract
Dynamically evolving adhesions between cells and extracellular matrix (ECM) transmit time-varying signals that control cytoskeletal dynamics and cell fate. Dynamic cell adhesion and ECM stiffness regulate cellular mechanosensing cooperatively, but it has not previously been possible to characterize their individual effects because of challenges with controlling these factors independently. Therefore, a DNA-driven molecular system is developed wherein the integrin-binding ligand RGD can be reversibly presented and removed to achieve cyclic cell attachment/detachment on substrates of defined stiffness. Using this culture system, it is discovered that cyclic adhesion accelerates F-actin kinetics and nuclear mechanosensing in human mesenchymal stem cells (hMSCs), with the result that hysteresis can completely change how hMSCs transduce ECM stiffness. Results are dramatically different from well-known results for mechanotransduction on static substrates, but are consistent with a mathematical model of F-actin fragments retaining structure following loss of integrin ligation and participating in subsequent repolymerization. These findings suggest that cyclic integrin-mediated adhesion alters the mechanosensing of ECM stiffness by hMSCs through transient, hysteretic memory that is stored in F-actin.
Collapse
Affiliation(s)
- Zheng Zhang
- The Key Laboratory of Biomedical Information Engineering of Ministry of EducationSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'an710049P. R. China
- Bioinspired Engineering and Biomechanics Center (BEBC)Xi'an Jiaotong UniversityXi'an710049P. R. China
| | - Hongyuan Zhu
- The Key Laboratory of Biomedical Information Engineering of Ministry of EducationSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'an710049P. R. China
- Bioinspired Engineering and Biomechanics Center (BEBC)Xi'an Jiaotong UniversityXi'an710049P. R. China
| | - Guoqing Zhao
- The Key Laboratory of Biomedical Information Engineering of Ministry of EducationSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'an710049P. R. China
- Bioinspired Engineering and Biomechanics Center (BEBC)Xi'an Jiaotong UniversityXi'an710049P. R. China
| | - Yunyi Miao
- The Key Laboratory of Biomedical Information Engineering of Ministry of EducationSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'an710049P. R. China
- Bioinspired Engineering and Biomechanics Center (BEBC)Xi'an Jiaotong UniversityXi'an710049P. R. China
| | - Lingzhu Zhao
- The Key Laboratory of Biomedical Information Engineering of Ministry of EducationSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'an710049P. R. China
- Bioinspired Engineering and Biomechanics Center (BEBC)Xi'an Jiaotong UniversityXi'an710049P. R. China
| | - Jinteng Feng
- Department of Medical OncologyFirst Affiliated Hospital of Xi'an Jiaotong UniversityXi'an710061P. R. China
| | - Huan Zhang
- The Key Laboratory of Biomedical Information Engineering of Ministry of EducationSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'an710049P. R. China
- Bioinspired Engineering and Biomechanics Center (BEBC)Xi'an Jiaotong UniversityXi'an710049P. R. China
| | - Run Miao
- The Key Laboratory of Biomedical Information Engineering of Ministry of EducationSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'an710049P. R. China
- Bioinspired Engineering and Biomechanics Center (BEBC)Xi'an Jiaotong UniversityXi'an710049P. R. China
| | - Lin Sun
- The Key Laboratory of Biomedical Information Engineering of Ministry of EducationSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'an710049P. R. China
- Bioinspired Engineering and Biomechanics Center (BEBC)Xi'an Jiaotong UniversityXi'an710049P. R. China
| | - Bin Gao
- Department of EndocrinologySecond Affiliated Hospital of Air Force Military Medical UniversityXi'an710038P. R. China
| | - Wencheng Zhang
- Department of EndocrinologySecond Affiliated Hospital of Air Force Military Medical UniversityXi'an710038P. R. China
| | - Zheng Wang
- Department of Hepatobiliary SurgeryFirst Affiliated Hospital of Xi'an Jiaotong UniversityXi'an710061P. R. China
| | - Jianfang Zhang
- Department of Gynaecology and Obstetrics of Xijing Hospital, Fourth Military Medical University710054Xi'anP. R. China
| | - Ying Zhang
- Xijing 986 Hospital DepartmentFourth Military Medical UniversityXi'an710054P. R. China
| | - Hui Guo
- Department of Medical OncologyFirst Affiliated Hospital of Xi'an Jiaotong UniversityXi'an710061P. R. China
| | - Feng Xu
- The Key Laboratory of Biomedical Information Engineering of Ministry of EducationSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'an710049P. R. China
- Bioinspired Engineering and Biomechanics Center (BEBC)Xi'an Jiaotong UniversityXi'an710049P. R. China
| | - Tian Jian Lu
- State Key Laboratory of Mechanics and Control of Mechanical StructuresNanjing University of Aeronautics and AstronauticsNanjing210016P. R. China
| | - Guy M. Genin
- The Key Laboratory of Biomedical Information Engineering of Ministry of EducationSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'an710049P. R. China
- Bioinspired Engineering and Biomechanics Center (BEBC)Xi'an Jiaotong UniversityXi'an710049P. R. China
- Department of Mechanical Engineering & Materials ScienceWashington University in St. LouisSt. LouisMO63130USA
- NSF Science and Technology Center for Engineering MechanobiologyWashington University in St. LouisSt. LouisMO63130USA
| | - Min Lin
- The Key Laboratory of Biomedical Information Engineering of Ministry of EducationSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'an710049P. R. China
- Bioinspired Engineering and Biomechanics Center (BEBC)Xi'an Jiaotong UniversityXi'an710049P. R. China
| |
Collapse
|
22
|
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
|
23
|
Qu J, Wang X, Zhang Y, Hu R, Hao Y, Zhao X, Dong C, Yang C, Zhang W, Sui J, Huang Y, Liu P, Yu J, Chen X, Fan Y. Cell reprogramming in a predictable manner on the superhydrophobic microwell array chip. Biomaterials 2023; 301:122215. [PMID: 37406601 DOI: 10.1016/j.biomaterials.2023.122215] [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: 05/11/2022] [Revised: 05/03/2023] [Accepted: 06/23/2023] [Indexed: 07/07/2023]
Abstract
Reprogramming of somatic cells into the pluripotent state is stochastic and inefficient using the conventional culture plates. Novel micro-culture systems employing precisely controlled biophysical cues can improve the reprogramming efficiencies dramatically. Here we perform iPSC induction on our previously developed superhydrophobic microwell array chip (SMAR-chip) where cells undergo distinctive morphology change, switching from 2D monolayers to 3D clumps, and develop into bona fide colonies in more than 90% of the microwells. The PDMS substrate, together with the microwell structure and the superhydrophobic layer constitute a well-controlled microenvironment favorable for the morphogenesis and pluripotency induction. Investigation of the molecular roadmap demonstrates that the SMAR-chip promotes the transition from the initiation phase to the maturation phase and overcomes the roadblocks for reprogramming. In addition, the SMAR-chip also promotes the reprogramming of human cells, opening our method for translational applications. In summary, our study provides a novel platform for efficient cell reprogramming and emphasizes the advantages of employing the insoluble microenvironmental cues for the precise control of cell fate conversion.
Collapse
Affiliation(s)
- Jianan Qu
- Beijing Advanced Innovation Centre for Biomedical Engineering, Key Laboratory for Biomechanics and Mechanobiology of Chinese Education Ministry, School of Biological Science and Medical Engineering, Beihang University, No.37, Xueyuan Road, Haidian District, Beijing, China
| | - Xiaoqing Wang
- Beijing Advanced Innovation Centre for Biomedical Engineering, Key Laboratory for Biomechanics and Mechanobiology of Chinese Education Ministry, School of Biological Science and Medical Engineering, Beihang University, No.37, Xueyuan Road, Haidian District, Beijing, China
| | - Yang Zhang
- Beijing Advanced Innovation Centre for Biomedical Engineering, Key Laboratory for Biomechanics and Mechanobiology of Chinese Education Ministry, School of Biological Science and Medical Engineering, Beihang University, No.37, Xueyuan Road, Haidian District, Beijing, China
| | - Ruowen Hu
- Beijing Advanced Innovation Centre for Biomedical Engineering, Key Laboratory for Biomechanics and Mechanobiology of Chinese Education Ministry, School of Biological Science and Medical Engineering, Beihang University, No.37, Xueyuan Road, Haidian District, Beijing, China
| | - Yunqi Hao
- Beijing Advanced Innovation Centre for Biomedical Engineering, Key Laboratory for Biomechanics and Mechanobiology of Chinese Education Ministry, School of Biological Science and Medical Engineering, Beihang University, No.37, Xueyuan Road, Haidian District, Beijing, China
| | - Xuechen Zhao
- Beijing Advanced Innovation Centre for Biomedical Engineering, Key Laboratory for Biomechanics and Mechanobiology of Chinese Education Ministry, School of Biological Science and Medical Engineering, Beihang University, No.37, Xueyuan Road, Haidian District, Beijing, China
| | - Chunhui Dong
- Beijing Advanced Innovation Centre for Biomedical Engineering, Key Laboratory for Biomechanics and Mechanobiology of Chinese Education Ministry, School of Biological Science and Medical Engineering, Beihang University, No.37, Xueyuan Road, Haidian District, Beijing, China
| | - Chengxi Yang
- Beijing Advanced Innovation Centre for Biomedical Engineering, Key Laboratory for Biomechanics and Mechanobiology of Chinese Education Ministry, School of Biological Science and Medical Engineering, Beihang University, No.37, Xueyuan Road, Haidian District, Beijing, China
| | - Weirong Zhang
- Beijing Advanced Innovation Centre for Biomedical Engineering, Key Laboratory for Biomechanics and Mechanobiology of Chinese Education Ministry, School of Biological Science and Medical Engineering, Beihang University, No.37, Xueyuan Road, Haidian District, Beijing, China
| | - Jingchao Sui
- Beijing Advanced Innovation Centre for Biomedical Engineering, Key Laboratory for Biomechanics and Mechanobiology of Chinese Education Ministry, School of Biological Science and Medical Engineering, Beihang University, No.37, Xueyuan Road, Haidian District, Beijing, China
| | - Yan Huang
- Beijing Advanced Innovation Centre for Biomedical Engineering, Key Laboratory for Biomechanics and Mechanobiology of Chinese Education Ministry, School of Biological Science and Medical Engineering, Beihang University, No.37, Xueyuan Road, Haidian District, Beijing, China
| | - Peng Liu
- Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing, China
| | - Jian Yu
- School of Engineering Medicine, Beihang University, No.37, Xueyuan Road, Haidian District, Beijing, China
| | - Xiaofang Chen
- Beijing Advanced Innovation Centre for Biomedical Engineering, Key Laboratory for Biomechanics and Mechanobiology of Chinese Education Ministry, School of Biological Science and Medical Engineering, Beihang University, No.37, Xueyuan Road, Haidian District, Beijing, China.
| | - Yubo Fan
- Beijing Advanced Innovation Centre for Biomedical Engineering, Key Laboratory for Biomechanics and Mechanobiology of Chinese Education Ministry, School of Biological Science and Medical Engineering, Beihang University, No.37, Xueyuan Road, Haidian District, Beijing, China; School of Engineering Medicine, Beihang University, No.37, Xueyuan Road, Haidian District, Beijing, China.
| |
Collapse
|
24
|
Sharlow ER, Llaneza DC, Grever WE, Mingledorff GA, Mendelson AJ, Bloom GS, Lazo JS. High content screening miniaturization and single cell imaging of mature human feeder layer-free iPSC-derived neurons. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2023; 28:275-283. [PMID: 36273809 PMCID: PMC10119332 DOI: 10.1016/j.slasd.2022.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 09/27/2022] [Accepted: 10/11/2022] [Indexed: 11/05/2022]
Abstract
Human induced pluripotent stem cell (iPSC)-derived neurons are being increasingly used for high content imaging and screening. However, iPSC-derived neuronal differentiation and maturation is time-intensive, often requiring >8 weeks. Unfortunately, the differentiating and maturing iPSC-derived neuronal cultures also tend to migrate and coalesce into ganglion-like clusters making single-cell analysis challenging, especially in miniaturized formats. Using our defined extracellular matrix and low oxygen culturing conditions for the differentiation and maturation of human cortical neurons, we further modified neuronal progenitor cell seeding densities and feeder layer-free culturing conditions in miniaturized formats (i.e., 96 well) to decrease neuronal clustering, enhance single-cell identification and reduce edge effects usually observed after extended neuronal cell culture. Subsequent algorithm development refined capabilities to distinguish and identify single mature neurons, as identified by NeuN expression, from large cellular aggregates, which were excluded from image analysis. Incorporation of astrocyte conditioned medium during differentiation and maturation periods significantly increased the percentage (i.e., ∼10% to ∼30%) of mature neurons (i.e., NeuN+) detected at 4-weeks post-differentiation. Pilot, proof of concept studies using this optimized assay system yielded negligible edge effects and robust Z-factors in population-based as well as image-based neurotoxicity assay formats. Moreover, moxidectin, an FDA-approved drug with documented neurotoxic adverse effects, was identified as a hit using both screening formats. This miniaturized, feeder layer-free format and image analysis algorithm provides a foundational imaging and screening platform, which enables quantitative single-cell analysis of differentiated human neurons.
Collapse
Affiliation(s)
- Elizabeth R Sharlow
- Department of Pharmacology, University of Virginia, 340 Jefferson Park Avenue, Pinn Hall, 5th Floor, P.O. Box 800735, Charlottesville, VA 22908-0735, USA.
| | - Danielle C Llaneza
- Department of Pharmacology, University of Virginia, 340 Jefferson Park Avenue, Pinn Hall, 5th Floor, P.O. Box 800735, Charlottesville, VA 22908-0735, USA
| | | | - Garnett A Mingledorff
- Department of Pharmacology, University of Virginia, 340 Jefferson Park Avenue, Pinn Hall, 5th Floor, P.O. Box 800735, Charlottesville, VA 22908-0735, USA
| | - Anna J Mendelson
- Department of Pharmacology, University of Virginia, 340 Jefferson Park Avenue, Pinn Hall, 5th Floor, P.O. Box 800735, Charlottesville, VA 22908-0735, USA
| | - George S Bloom
- Department of Biology, University of Virginia, 420 Gilmer Hall, 485 McCormick Road, P.O. Box 400328, Charlottesville VA 22904, USA; Department of Cell Biology, University of Virginia, 420 Gilmer Hall, 485 McCormick Road, P.O. Box 400328, Charlottesville VA 22904, USA; Department of Neuroscience, University of Virginia, 420 Gilmer Hall, 485 McCormick Road, P.O. Box 400328, Charlottesville VA 22904, USA
| | - John S Lazo
- Department of Pharmacology, University of Virginia, 340 Jefferson Park Avenue, Pinn Hall, 5th Floor, P.O. Box 800735, Charlottesville, VA 22908-0735, USA
| |
Collapse
|
25
|
Han X, Lu T, Zhang Z, Wang H, Lu S. Tremella polysaccharide-based conductive hydrogel with anti-freezing and self-healing ability for motion monitoring and intelligent interaction. Int J Biol Macromol 2023; 248:125987. [PMID: 37516220 DOI: 10.1016/j.ijbiomac.2023.125987] [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: 06/14/2023] [Revised: 07/20/2023] [Accepted: 07/23/2023] [Indexed: 07/31/2023]
Abstract
The application of conductive hydrogels in flexible wearable devices has garnered significant attention. In this study, a self-healing, anti-freezing, and fire-resistant hydrogel strain sensor is successfully synthesized by incorporating sustainable natural biological materials, viz. Tremella polysaccharide and silk fiber, into a polyvinyl alcohol matrix with borax cross-linking. The resulting hydrogel exhibits excellent transparency, thermoplasticity, and remarkable mechanical properties, including a notable elongation (1107.3 %) and high self-healing rate (91.11 %) within 5 min, attributed to the dynamic cross-linking effect of hydrogen bonds and borax. A strain sensor based on the prepared hydrogel sensor can be used to accurately monitor diverse human movements, while maintaining exceptional sensing stability and durability under repeated strain cycles. Additionally, a hydrogel touch component is designed that can successfully interact with intelligent electronic devices, encompassing functions like clicking, writing, and drawing. These inherent advantages make the prepared hydrogel a promising candidate for applications in human health monitoring and intelligent electronic device interaction.
Collapse
Affiliation(s)
- Xiaokun Han
- Key Laboratory of Rubber-Plastics of Ministry of Education/Shandong Provincial Key Laboratory of Rubber, Plastics, School of Polymer Science and Engineering, Qingdao University of Science & Technology, Qingdao 266061, PR China; Key Laboratory of New Processing Technology for Nonferrous Metals and Materials, Ministry of Education, School of Material Science and Engineering, Guilin University of Technology, Guilin 541004, PR China
| | - Tianyun Lu
- Key Laboratory of Rubber-Plastics of Ministry of Education/Shandong Provincial Key Laboratory of Rubber, Plastics, School of Polymer Science and Engineering, Qingdao University of Science & Technology, Qingdao 266061, PR China
| | - Zuocai Zhang
- Shenzhen Key Laboratory of Polymer Science and Technology, Guangdong Research Center for Interfacial Engineering of Functional Materials, College of Materials Science and Engineering, Shenzhen University, Shenzhen 518060, PR China
| | - He Wang
- Key Laboratory of Rubber-Plastics of Ministry of Education/Shandong Provincial Key Laboratory of Rubber, Plastics, School of Polymer Science and Engineering, Qingdao University of Science & Technology, Qingdao 266061, PR China
| | - Shaorong Lu
- Key Laboratory of New Processing Technology for Nonferrous Metals and Materials, Ministry of Education, School of Material Science and Engineering, Guilin University of Technology, Guilin 541004, PR China.
| |
Collapse
|
26
|
Goldshmid R, Simaan-Yameen H, Ifergan L, Loebel C, Burdick JA, Seliktar D. Modulus-dependent effects on neurogenic, myogenic, and chondrogenic differentiation of human mesenchymal stem cells in three-dimensional hydrogel cultures. J Biomed Mater Res A 2023; 111:1441-1458. [PMID: 37066837 DOI: 10.1002/jbm.a.37545] [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: 09/21/2022] [Revised: 03/23/2023] [Accepted: 03/25/2023] [Indexed: 04/18/2023]
Abstract
Human mesenchymal stromal cells (hMSCs) are of significant interest as a renewable source of therapeutically useful cells. In tissue engineering, hMSCs are implanted within a scaffold to provide enhanced capacity for tissue repair. The present study evaluates how mechanical properties of that scaffold can alter the phenotype and genotype of the cells, with the aim of augmenting hMSC differentiation along the myogenic, neurogenic or chondrogenic linages. The hMSCs were grown three-dimensionally (3D) in a hydrogel comprised of poly(ethylene glycol) (PEG)-conjugated to fibrinogen. The hydrogel's shear storage modulus (G'), which was controlled by increasing the amount of PEG-diacrylate cross-linker in the matrix, was varied in the range of 100-2000 Pascal (Pa). The differentiation into each lineage was initiated by a defined culture medium, and the hMSCs grown in the different modulus hydrogels were characterized using gene and protein expression. Materials having lower storage moduli (G' = 100 Pa) exhibited more hMSCs differentiating to neurogenic lineages. Myogenesis was favored in materials having intermediate modulus values (G' = 500 Pa), whereas chondrogenesis was favored in materials with a higher modulus (G' = 1000 Pa). Enhancing the differentiation pathway of hMSCs in 3D hydrogel scaffolds using simple modifications to mechanical properties represents an important achievement toward the effective application of these cells in tissue engineering.
Collapse
Affiliation(s)
- Revital Goldshmid
- The Faculty of Biomedical Engineering, Technion-Israel Institute of Technology, Haifa, Israel
- The Interdisciplinary Program for Biotechnology, Technion-Israel Institute of Technology, Haifa, Israel
| | - Haneen Simaan-Yameen
- The Faculty of Biomedical Engineering, Technion-Israel Institute of Technology, Haifa, Israel
- The Interdisciplinary Program for Biotechnology, Technion-Israel Institute of Technology, Haifa, Israel
| | - Liaura Ifergan
- The Faculty of Biomedical Engineering, Technion-Israel Institute of Technology, Haifa, Israel
| | - Claudia Loebel
- Materials Science & Engineering Department, University of Michigan, Ann Arbor, Michigan, USA
| | - Jason A Burdick
- BioFrontiers Institute and Department of Chemical and Biological Engineering, University of Colorado, Boulder, Colorado, USA
| | - Dror Seliktar
- The Faculty of Biomedical Engineering, Technion-Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
27
|
Guinard I, Nguyen T, Brassard-Jollive N, Weber J, Ruch L, Reininger L, Brouard N, Eckly A, Collin D, Lanza F, Léon C. Matrix stiffness controls megakaryocyte adhesion, fibronectin fibrillogenesis, and proplatelet formation through Itgβ3. Blood Adv 2023; 7:4003-4018. [PMID: 37171626 PMCID: PMC10410137 DOI: 10.1182/bloodadvances.2022008680] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 04/13/2023] [Accepted: 04/17/2023] [Indexed: 05/13/2023] Open
Abstract
Megakaryocytes (MKs) are the precursor cells of platelets, located in the bone marrow (BM). Once mature, they extend elongated projections named proplatelets through sinusoid vessels, emerging from the marrow stroma into the circulating blood. Not all signals from the microenvironment that regulate proplatelet formation are understood, particularly those from the BM biomechanics. We sought to investigate how MKs perceive and adapt to modifications of the stiffness of their environment. Although the BM is one of the softest tissue of the body, its rigidification results from excess fibronectin (FN), and other matrix protein deposition occur upon myelofibrosis. Here, we have shown that mouse MKs are able to detect the stiffness of a FN-coated substrate and adapt their morphology accordingly. Using a polydimethylsiloxane substrate with stiffness varying from physiological to pathological marrow, we found that a stiff matrix favors spreading, intracellular contractility, and FN fibrils assembly at the expense of proplatelet formation. Itgb3, but not Itgb1, is required for stiffness sensing, whereas both integrins are involved in fibrils assembly. In contrast, soft substrates promote proplatelet formation in an Itgb3-dependent manner, consistent with the ex vivo decrease in proplatelet formation and the in vivo decrease in platelet number in Itgb3-deficient mice. Our findings demonstrate the importance of environmental stiffness for MK functions with potential pathophysiological implications during pathologies that deregulate FN deposition and modulate stiffness in the marrow.
Collapse
Affiliation(s)
- Ines Guinard
- UMR_S1255, INSERM, Etablissement Français du Sang-Grand Est, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Strasbourg, France
| | - Thao Nguyen
- UMR_S1255, INSERM, Etablissement Français du Sang-Grand Est, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Strasbourg, France
| | - Noémie Brassard-Jollive
- UMR_S1255, INSERM, Etablissement Français du Sang-Grand Est, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Strasbourg, France
| | - Josiane Weber
- UMR_S1255, INSERM, Etablissement Français du Sang-Grand Est, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Strasbourg, France
| | - Laurie Ruch
- UMR_S1255, INSERM, Etablissement Français du Sang-Grand Est, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Strasbourg, France
| | - Laura Reininger
- UMR_S1255, INSERM, Etablissement Français du Sang-Grand Est, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Strasbourg, France
| | - Nathalie Brouard
- UMR_S1255, INSERM, Etablissement Français du Sang-Grand Est, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Strasbourg, France
| | - Anita Eckly
- UMR_S1255, INSERM, Etablissement Français du Sang-Grand Est, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Strasbourg, France
| | | | - François Lanza
- UMR_S1255, INSERM, Etablissement Français du Sang-Grand Est, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Strasbourg, France
| | - Catherine Léon
- UMR_S1255, INSERM, Etablissement Français du Sang-Grand Est, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Strasbourg, France
| |
Collapse
|
28
|
Lou Y, Sun M, Zhang J, Wang Y, Ma H, Sun Z, Li S, Weng X, Ying B, Liu C, Yu M, Wang H. Ultraviolet Light-Based Micropattern Printing on Titanium Surfaces to Promote Early Osseointegration. Adv Healthc Mater 2023; 12:e2203300. [PMID: 37119120 DOI: 10.1002/adhm.202203300] [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: 12/19/2022] [Revised: 04/24/2023] [Indexed: 04/30/2023]
Abstract
Patterned interfaces are widely used for surface modification of biomaterials because of a morphological unit similar to that of native tissue. However, engineering fast and cost-effective high-resolution micropatterns directly onto titanium surfaces remains a grand challenge. Herein, a simply designed ultraviolet (UV) light-based micropattern printing to obtain geometrical patterns on implant interfaces is fabricated by utilizing customized photomasks and titanium dioxide (TiO2 ) nanorods as a photo-responsive platform. The technique manipulates the cytoskeleton of micropatterning cells on the surface of TiO2 nanorods. The linear pattern surface shows the elongated morphology and parallel linear arrangements of human mesenchymal stem cells (hMSCs), significantly enhancing their osteogenic differentiation. In addition to the upregulated expression of key osteo-specific function genes in vitro, the accelerated osseointegration between the implant and the host bone is obtained in vivo. Further investigation indicates that the developed linear pattern surface has an outstanding effect on the cytoskeletal system, and finally activates Yes-Associated Protein (YAP)-mediated mechanotransduction pathways, initiating hMSCs osteogenic differentiation. This study not only offers a microfabrication method that can be extended to fabricate various shape- and size-controlled micropatterns on titanium surfaces, but also provides insight into the surface structure design for enhanced bone regeneration.
Collapse
Affiliation(s)
- Yiting Lou
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, 395 Yan'an road, Hangzhou, Zhejiang, 310000, China
| | - Mouyuan Sun
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, 395 Yan'an road, Hangzhou, Zhejiang, 310000, China
| | - Jingyu Zhang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, 395 Yan'an road, Hangzhou, Zhejiang, 310000, China
| | - Yu Wang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, 395 Yan'an road, Hangzhou, Zhejiang, 310000, China
| | - Haiying Ma
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, 395 Yan'an road, Hangzhou, Zhejiang, 310000, China
| | - Zheyuan Sun
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, 395 Yan'an road, Hangzhou, Zhejiang, 310000, China
| | - Shengjie Li
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, 395 Yan'an road, Hangzhou, Zhejiang, 310000, China
- Department of Stomatology, The First Affiliated Hospital of Ningbo University, 59 Liuting street, Ningbo, Zhejiang, 315000, China
| | - Xiaoyan Weng
- The Third Affiliated Hospital of Wenzhou Medical University (Ruian People's Hospital), 168 Ruifeng Avenue, Wenzhou, Zhejiang, 325016, China
| | - Binbin Ying
- Department of Stomatology, The First Affiliated Hospital of Ningbo University, 59 Liuting street, Ningbo, Zhejiang, 315000, China
| | - Chao Liu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, 395 Yan'an road, Hangzhou, Zhejiang, 310000, China
| | - Mengfei Yu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, 395 Yan'an road, Hangzhou, Zhejiang, 310000, China
| | - Huiming Wang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, 395 Yan'an road, Hangzhou, Zhejiang, 310000, China
| |
Collapse
|
29
|
Babaliari E, Ranella A, Stratakis E. Microfluidic Systems for Neural Cell Studies. Bioengineering (Basel) 2023; 10:902. [PMID: 37627787 PMCID: PMC10451731 DOI: 10.3390/bioengineering10080902] [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: 06/02/2023] [Revised: 07/05/2023] [Accepted: 07/25/2023] [Indexed: 08/27/2023] Open
Abstract
Whereas the axons of the peripheral nervous system (PNS) spontaneously regenerate after an injury, the occurring regeneration is rarely successful because axons are usually directed by inappropriate cues. Therefore, finding successful ways to guide neurite outgrowth, in vitro, is essential for neurogenesis. Microfluidic systems reflect more appropriately the in vivo environment of cells in tissues such as the normal fluid flow within the body, consistent nutrient delivery, effective waste removal, and mechanical stimulation due to fluid shear forces. At the same time, it has been well reported that topography affects neuronal outgrowth, orientation, and differentiation. In this review, we demonstrate how topography and microfluidic flow affect neuronal behavior, either separately or in synergy, and highlight the efficacy of microfluidic systems in promoting neuronal outgrowth.
Collapse
Affiliation(s)
- Eleftheria Babaliari
- Foundation for Research and Technology—Hellas (F.O.R.T.H.), Institute of Electronic Structure and Laser (I.E.S.L.), Vasilika Vouton, 70013 Heraklion, Greece;
| | - Anthi Ranella
- Foundation for Research and Technology—Hellas (F.O.R.T.H.), Institute of Electronic Structure and Laser (I.E.S.L.), Vasilika Vouton, 70013 Heraklion, Greece;
| | - Emmanuel Stratakis
- Foundation for Research and Technology—Hellas (F.O.R.T.H.), Institute of Electronic Structure and Laser (I.E.S.L.), Vasilika Vouton, 70013 Heraklion, Greece;
- Department of Physics, University of Crete, 70013 Heraklion, Greece
| |
Collapse
|
30
|
Kohler TN, De Jonghe J, Ellermann AL, Yanagida A, Herger M, Slatery EM, Weberling A, Munger C, Fischer K, Mulas C, Winkel A, Ross C, Bergmann S, Franze K, Chalut K, Nichols J, Boroviak TE, Hollfelder F. Plakoglobin is a mechanoresponsive regulator of naive pluripotency. Nat Commun 2023; 14:4022. [PMID: 37419903 PMCID: PMC10329048 DOI: 10.1038/s41467-023-39515-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 06/09/2023] [Indexed: 07/09/2023] Open
Abstract
Biomechanical cues are instrumental in guiding embryonic development and cell differentiation. Understanding how these physical stimuli translate into transcriptional programs will provide insight into mechanisms underlying mammalian pre-implantation development. Here, we explore this type of regulation by exerting microenvironmental control over mouse embryonic stem cells. Microfluidic encapsulation of mouse embryonic stem cells in agarose microgels stabilizes the naive pluripotency network and specifically induces expression of Plakoglobin (Jup), a vertebrate homolog of β-catenin. Overexpression of Plakoglobin is sufficient to fully re-establish the naive pluripotency gene regulatory network under metastable pluripotency conditions, as confirmed by single-cell transcriptome profiling. Finally, we find that, in the epiblast, Plakoglobin was exclusively expressed at the blastocyst stage in human and mouse embryos - further strengthening the link between Plakoglobin and naive pluripotency in vivo. Our work reveals Plakoglobin as a mechanosensitive regulator of naive pluripotency and provides a paradigm to interrogate the effects of volumetric confinement on cell-fate transitions.
Collapse
Affiliation(s)
- Timo N Kohler
- Department of Biochemistry, University of Cambridge, Hopkins Building, Tennis Court Road, Cambridge, CB2 1QW, UK
- Wellcome Trust - Medical Research Council Stem Cell Institute, University of Cambridge, Jeffrey Cheah Biomedical Centre, Puddicombe Way, Cambridge, CB2 0AW, UK
| | - Joachim De Jonghe
- Department of Biochemistry, University of Cambridge, Hopkins Building, Tennis Court Road, Cambridge, CB2 1QW, UK
| | - Anna L Ellermann
- Department of Biochemistry, University of Cambridge, Hopkins Building, Tennis Court Road, Cambridge, CB2 1QW, UK
| | - Ayaka Yanagida
- Wellcome Trust - Medical Research Council Stem Cell Institute, University of Cambridge, Jeffrey Cheah Biomedical Centre, Puddicombe Way, Cambridge, CB2 0AW, UK
- Department of Veterinary Anatomy, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, 113-8657, Japan
- Stem Cell Therapy Laboratory, Advanced Research Institute, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Michael Herger
- Department of Biochemistry, University of Cambridge, Hopkins Building, Tennis Court Road, Cambridge, CB2 1QW, UK
| | - Erin M Slatery
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, CB2 3DY, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, CB2 3EG, UK
| | - Antonia Weberling
- Department of Biochemistry, University of Cambridge, Hopkins Building, Tennis Court Road, Cambridge, CB2 1QW, UK
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, CB2 3DY, UK
| | - Clara Munger
- Department of Biochemistry, University of Cambridge, Hopkins Building, Tennis Court Road, Cambridge, CB2 1QW, UK
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, CB2 3DY, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, CB2 3EG, UK
| | - Katrin Fischer
- Department of Biochemistry, University of Cambridge, Hopkins Building, Tennis Court Road, Cambridge, CB2 1QW, UK
| | - Carla Mulas
- Wellcome Trust - Medical Research Council Stem Cell Institute, University of Cambridge, Jeffrey Cheah Biomedical Centre, Puddicombe Way, Cambridge, CB2 0AW, UK
- Randall Centre for Cell and Molecular Biophysics, King's College London, London, SE1 1UL, UK
- Altos Labs, Cambridge Institute of Science, Cambridge, UK
| | - Alex Winkel
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, CB2 3DY, UK
| | - Connor Ross
- Wellcome Trust - Medical Research Council Stem Cell Institute, University of Cambridge, Jeffrey Cheah Biomedical Centre, Puddicombe Way, Cambridge, CB2 0AW, UK
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, CB2 3DY, UK
- MRC Human Genetics Unit, Institute of Genetics and Cancer, The University of Edinburgh, Crewe Road, Edinburgh, EH4 2XU, UK
| | - Sophie Bergmann
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, CB2 3DY, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, CB2 3EG, UK
| | - Kristian Franze
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, CB2 3DY, UK
- Institute of Medical Physics, Friedrich-Alexander-Universität Erlangen-Nürnberg, Henkestr. 91, 91052, Erlangen, Germany
- Max-Planck-Zentrum für Physik und Medizin, 91054, Erlangen, Germany
| | - Kevin Chalut
- Wellcome Trust - Medical Research Council Stem Cell Institute, University of Cambridge, Jeffrey Cheah Biomedical Centre, Puddicombe Way, Cambridge, CB2 0AW, UK
- Altos Labs, Cambridge Institute of Science, Cambridge, UK
| | - Jennifer Nichols
- Wellcome Trust - Medical Research Council Stem Cell Institute, University of Cambridge, Jeffrey Cheah Biomedical Centre, Puddicombe Way, Cambridge, CB2 0AW, UK
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, CB2 3DY, UK
- MRC Human Genetics Unit, Institute of Genetics and Cancer, The University of Edinburgh, Crewe Road, Edinburgh, EH4 2XU, UK
| | - Thorsten E Boroviak
- Wellcome Trust - Medical Research Council Stem Cell Institute, University of Cambridge, Jeffrey Cheah Biomedical Centre, Puddicombe Way, Cambridge, CB2 0AW, UK.
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, CB2 3DY, UK.
- Centre for Trophoblast Research, University of Cambridge, Cambridge, CB2 3EG, UK.
| | - Florian Hollfelder
- Department of Biochemistry, University of Cambridge, Hopkins Building, Tennis Court Road, Cambridge, CB2 1QW, UK.
| |
Collapse
|
31
|
Xu KL, Mauck RL, Burdick JA. Modeling development using hydrogels. Development 2023; 150:dev201527. [PMID: 37387575 PMCID: PMC10323241 DOI: 10.1242/dev.201527] [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: 07/01/2023]
Abstract
The development of multicellular complex organisms relies on coordinated signaling from the microenvironment, including both biochemical and mechanical interactions. To better understand developmental biology, increasingly sophisticated in vitro systems are needed to mimic these complex extracellular features. In this Primer, we explore how engineered hydrogels can serve as in vitro culture platforms to present such signals in a controlled manner and include examples of how they have been used to advance our understanding of developmental biology.
Collapse
Affiliation(s)
- Karen L. Xu
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Robert L. Mauck
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA 19104, USA
| | - Jason A. Burdick
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80303, USA
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO 80303, USA
| |
Collapse
|
32
|
Chi C, Song K. Cellular reprogramming of fibroblasts in heart regeneration. J Mol Cell Cardiol 2023; 180:84-93. [PMID: 36965699 PMCID: PMC10347886 DOI: 10.1016/j.yjmcc.2023.03.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 03/10/2023] [Accepted: 03/21/2023] [Indexed: 03/27/2023]
Abstract
Myocardial infarction causes the loss of cardiomyocytes and the formation of cardiac fibrosis due to the activation of cardiac fibroblasts, leading to cardiac dysfunction and heart failure. Unfortunately, current therapeutic interventions can only slow the disease progression. Furthermore, they cannot fully restore cardiac function, likely because the adult human heart lacks sufficient capacity to regenerate cardiomyocytes. Therefore, intensive efforts have focused on developing therapeutics to regenerate the damaged heart. Several strategies have been intensively investigated, including stimulation of cardiomyocyte proliferation, transplantation of stem cell-derived cardiomyocytes, and conversion of fibroblasts into cardiac cells. Resident cardiac fibroblasts are critical in the maintenance of the structure and contractility of the heart. Fibroblast plasticity makes this type of cells be reprogrammed into many cell types, including but not limited to induced pluripotent stem cells, induced cardiac progenitor cells, and induced cardiomyocytes. Fibroblasts have become a therapeutic target due to their critical roles in cardiac pathogenesis. This review summarizes the reprogramming of fibroblasts into induced pluripotent stem cell-derived cardiomyocytes, induced cardiac progenitor cells, and induced cardiomyocytes to repair a damaged heart, outlines recent findings in utilizing fibroblast-derived cells for heart regeneration, and discusses the limitations and challenges.
Collapse
Affiliation(s)
- Congwu Chi
- Division of Cardiology, Department of Medicine, The University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Kunhua Song
- Division of Cardiology, Department of Medicine, The University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Gates Center for Regenerative Medicine and Stem Cell Biology, The University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.
| |
Collapse
|
33
|
Papalamprou A, Yu V, Chen A, Stefanovic T, Kaneda G, Salehi K, Castaneda CM, Gertych A, Glaeser JD, Sheyn D. Directing iPSC differentiation into iTenocytes using combined scleraxis overexpression and cyclic loading. J Orthop Res 2023; 41:1148-1161. [PMID: 36203346 PMCID: PMC10076443 DOI: 10.1002/jor.25459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 09/08/2022] [Accepted: 09/22/2022] [Indexed: 02/04/2023]
Abstract
Regenerative therapies for tendon are falling behind other tissues due to the lack of an appropriate and potent cell therapeutic candidate. This study aimed to induce tenogenesis using stable Scleraxis (Scx) overexpression in combination with uniaxial mechanical stretch of iPSC-derived mesenchymal stromal-like cells (iMSCs). Scx is the single direct molecular regulator of tendon differentiation known to date. Bone marrow-derived (BM-)MSCs were used as reference. Scx overexpression alone resulted in significantly higher upregulation of tenogenic markers in iMSCs compared to BM-MSCs. Mechanoregulation is known to be a central element guiding tendon development and healing. Mechanical stimulation combined with Scx overexpression resulted in morphometric and cytoskeleton-related changes, upregulation of early and late tendon markers, and increased extracellular matrix deposition and alignment, and tenomodulin perinuclear localization in iMSCs. Our findings suggest that these cells can be differentiated into tenocytes and might be a better candidate for tendon cell therapy applications than BM-MSCs.
Collapse
Affiliation(s)
- Angela Papalamprou
- Orthopaedic Stem Cell Research Laboratory, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Victoria Yu
- Orthopaedic Stem Cell Research Laboratory, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Angel Chen
- Orthopaedic Stem Cell Research Laboratory, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Tina Stefanovic
- Orthopaedic Stem Cell Research Laboratory, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Giselle Kaneda
- Orthopaedic Stem Cell Research Laboratory, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Khosrowdad Salehi
- Orthopaedic Stem Cell Research Laboratory, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Chloe M. Castaneda
- Orthopaedic Stem Cell Research Laboratory, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Arkadiusz Gertych
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Juliane D. Glaeser
- Orthopaedic Stem Cell Research Laboratory, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Orthopedics, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Dmitriy Sheyn
- Orthopaedic Stem Cell Research Laboratory, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Orthopedics, Cedars-Sinai Medical Center, Los Angeles, California, USA
| |
Collapse
|
34
|
Kang Z, Wu B, Zhang L, Liang X, Guo D, Yuan S, Xie D. Metabolic regulation by biomaterials in osteoblast. Front Bioeng Biotechnol 2023; 11:1184463. [PMID: 37324445 PMCID: PMC10265685 DOI: 10.3389/fbioe.2023.1184463] [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: 03/11/2023] [Accepted: 05/19/2023] [Indexed: 06/17/2023] Open
Abstract
The repair of bone defects resulting from high-energy trauma, infection, or pathological fracture remains a challenge in the field of medicine. The development of biomaterials involved in the metabolic regulation provides a promising solution to this problem and has emerged as a prominent research area in regenerative engineering. While recent research on cell metabolism has advanced our knowledge of metabolic regulation in bone regeneration, the extent to which materials affect intracellular metabolic remains unclear. This review provides a detailed discussion of the mechanisms of bone regeneration, an overview of metabolic regulation in bone regeneration in osteoblasts and biomaterials involved in the metabolic regulation for bone regeneration. Furthermore, it introduces how materials, such as promoting favorable physicochemical characteristics (e.g., bioactivity, appropriate porosity, and superior mechanical properties), incorporating external stimuli (e.g., photothermal, electrical, and magnetic stimulation), and delivering metabolic regulators (e.g., metal ions, bioactive molecules like drugs and peptides, and regulatory metabolites such as alpha ketoglutarate), can affect cell metabolism and lead to changes of cell state. Considering the growing interests in cell metabolic regulation, advanced materials have the potential to help a larger population in overcoming bone defects.
Collapse
Affiliation(s)
- Zhengyang Kang
- Department of Orthopedics, The Second People’s Hospital of Panyu Guangzhou, Guangzhou, China
- Department of Joint Surgery and Sports Medicine, Center for Orthopedic Surgery, Orthopedic Hospital of Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Bin Wu
- Department of Orthopedics, The Second People’s Hospital of Panyu Guangzhou, Guangzhou, China
| | - Luhui Zhang
- Department of Joint Surgery and Sports Medicine, Center for Orthopedic Surgery, Orthopedic Hospital of Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Xinzhi Liang
- Department of Joint Surgery and Sports Medicine, Center for Orthopedic Surgery, Orthopedic Hospital of Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Dong Guo
- Department of Joint Surgery and Sports Medicine, Center for Orthopedic Surgery, Orthopedic Hospital of Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Shuai Yuan
- Department of Joint Surgery and Sports Medicine, Center for Orthopedic Surgery, Orthopedic Hospital of Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Denghui Xie
- Department of Joint Surgery and Sports Medicine, Center for Orthopedic Surgery, Orthopedic Hospital of Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Guangxi Key Laboratory of Bone and Joint Degeneration Diseases, Youjiang Medical University For Nationalities, Baise, China
| |
Collapse
|
35
|
Yue M, Liu Y, Zhang P, Li Z, Zhou Y. Integrative Analysis Reveals the Diverse Effects of 3D Stiffness upon Stem Cell Fate. Int J Mol Sci 2023; 24:9311. [PMID: 37298263 PMCID: PMC10253631 DOI: 10.3390/ijms24119311] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 05/09/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023] Open
Abstract
The origin of life and native tissue development are dependent on the heterogeneity of pluripotent stem cells. Bone marrow mesenchymal stem cells (BMMSCs) are located in a complicated niche with variable matrix stiffnesses, resulting in divergent stem cell fates. However, how stiffness drives stem cell fate remains unknown. For this study, we performed whole-gene transcriptomics and precise untargeted metabolomics sequencing to elucidate the complex interaction network of stem cell transcriptional and metabolic signals in extracellular matrices (ECMs) with different stiffnesses, and we propose a potential mechanism involved in stem cell fate decision. In a stiff (39~45 kPa) ECM, biosynthesis of aminoacyl-tRNA was up-regulated, and increased osteogenesis was also observed. In a soft (7~10 kPa) ECM, biosynthesis of unsaturated fatty acids and deposition of glycosaminoglycans were increased, accompanied by enhanced adipogenic/chondrogenic differentiation of BMMSCs. In addition, a panel of genes responding to the stiffness of the ECM were validated in vitro, mapping out the key signaling network that regulates stem cells' fate decisions. This finding of "stiffness-dependent manipulation of stem cell fate" provides a novel molecular biological basis for development of potential therapeutic targets within tissue engineering, from both a cellular metabolic and a biomechanical perspective.
Collapse
Affiliation(s)
- Muxin Yue
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing 100081, China; (M.Y.); (Y.L.); (P.Z.)
- National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China
| | - Yunsong Liu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing 100081, China; (M.Y.); (Y.L.); (P.Z.)
- National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China
| | - Ping Zhang
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing 100081, China; (M.Y.); (Y.L.); (P.Z.)
- National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China
| | - Zheng Li
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing 100081, China; (M.Y.); (Y.L.); (P.Z.)
- National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China
| | - Yongsheng Zhou
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing 100081, China; (M.Y.); (Y.L.); (P.Z.)
- National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China
| |
Collapse
|
36
|
Zhang X, Cao D, Xu L, Xu Y, Gao Z, Pan Y, Jiang M, Wei Y, Wang L, Liao Y, Wang Q, Yang L, Xu X, Gao Y, Gao S, Wang J, Yue R. Harnessing matrix stiffness to engineer a bone marrow niche for hematopoietic stem cell rejuvenation. Cell Stem Cell 2023; 30:378-395.e8. [PMID: 37028404 DOI: 10.1016/j.stem.2023.03.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 11/23/2022] [Accepted: 03/08/2023] [Indexed: 04/09/2023]
Abstract
Hematopoietic stem cell (HSC) self-renewal and aging are tightly regulated by paracrine factors from the bone marrow niche. However, whether HSC rejuvenation could be achieved by engineering a bone marrow niche ex vivo remains unknown. Here, we show that matrix stiffness fine-tunes HSC niche factor expression by bone marrow stromal cells (BMSCs). Increased stiffness activates Yap/Taz signaling to promote BMSC expansion upon 2D culture, which is largely reversed by 3D culture in soft gelatin methacrylate hydrogels. Notably, 3D co-culture with BMSCs promotes HSC maintenance and lymphopoiesis, reverses aging hallmarks of HSCs, and restores their long-term multilineage reconstitution capacity. In situ atomic force microscopy analysis reveals that mouse bone marrow stiffens with age, which correlates with a compromised HSC niche. Taken together, this study highlights the biomechanical regulation of the HSC niche by BMSCs, which could be harnessed to engineer a soft bone marrow niche for HSC rejuvenation.
Collapse
Affiliation(s)
- Xiaoying Zhang
- Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Dandan Cao
- Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Liting Xu
- Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Yanhua Xu
- Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China; Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Zehua Gao
- The State Key Laboratory of Bioreactor Engineering, Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai 200237, China
| | - Yuanzhong Pan
- The State Key Laboratory of Bioreactor Engineering, Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai 200237, China
| | - Ming Jiang
- The State Key Laboratory of Bioreactor Engineering, Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai 200237, China
| | - Yuhui Wei
- The Interdisciplinary Research Center, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China
| | - Lihua Wang
- The Interdisciplinary Research Center, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China
| | - Yue Liao
- School of Chemical Science and Engineering, Tongji University, Shanghai 200092, China
| | - Qigang Wang
- School of Chemical Science and Engineering, Tongji University, Shanghai 200092, China
| | - Lei Yang
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Xiaocui Xu
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Yawei Gao
- Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China; Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai 200120, China
| | - Shaorong Gao
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Jing Wang
- The State Key Laboratory of Bioreactor Engineering, Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai 200237, China
| | - Rui Yue
- Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China; Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai 200120, China.
| |
Collapse
|
37
|
Hu X, Xue Y, Liu D, Zhang J, Wang T, Wu Z, Lei W. Effects of material nano-topography on the angiogenesis of type H vessels: Size dependence, cell heterogeneity and intercellular communication. BIOMATERIALS ADVANCES 2023; 147:213307. [PMID: 36746099 DOI: 10.1016/j.bioadv.2023.213307] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 12/31/2022] [Accepted: 01/17/2023] [Indexed: 01/22/2023]
Abstract
Type H vessel, a vascular subtype in bone, is a critical regulator of osteogenesis, but how material properties affect this organ-specific vessel remains unknown. Here, titania nanotubes were fabricated on bone implant surface to investigate the effects of nano-topography on type H vessels. In vivo, surface nanotubes with 20-100 nm diameters promoted the angiogenesis of type H vessels and bone regeneration in mouse femurs to different extents, with the best effects induced by 70 nm diameter. In vitro, bone-specific endothelial cells (BECs) and artery endothelial cells (AECs) presented significantly different behaviors on the same material. Nanotubes with 20 nm small diameters significantly improved the adhesion, proliferation, type H differentiation of BECs and their paracrine function to regulate pre-osteoblasts (POBs), possibly via binding integrin β1 on the cell membrane, but these effects weakened when tube diameter increased, which conflicted with the results in vivo. Further study suggested that the better in vivo effects by larger diameters of 70-100 nm might be exerted indirectly through remodeling the regulation from POBs to BECs, highlighting the underappreciated indirect bio-effects of materials via intercellular communication. These suggest that nanoscale material topography makes significant impact on the angiogenesis of type H vessels, directly via binding integrins on the cell membrane of BECs and indirectly via modulating the regulation from osteoblastic cells to BECs, both in a size-dependent manner. Cells of the same type but from different tissues may show different responses to the same material, thus material properties should be tailored to the specific cell population. In research on material-tissue interactions, conclusions from in vitro experiments exposing a single type of cell to material might deviate from the truth in vivo, because materials may indirectly influence the targeted cells through modulating intercellular communication. These provide new insights into material-tissue interactions.
Collapse
Affiliation(s)
- Xiaofan Hu
- Department of Orthopedics, Xijing Hospital, The Fourth Military Medical University, Xi'an 710032, China
| | - Yumeng Xue
- State Key Laboratory of Solidification Processing, Center for Nano Energy Materials, School of Materials Science and Engineering, Northwestern Polytechnical University and Shaanxi Joint Laboratory of Graphene (NPU), Xi'an 710072, China
| | - Daming Liu
- Department of Orthopedics, Xijing Hospital, The Fourth Military Medical University, Xi'an 710032, China
| | - Jianming Zhang
- Department of Orthopedics, Xijing Hospital, The Fourth Military Medical University, Xi'an 710032, China
| | - Tianji Wang
- Department of Orthopedics, Xijing Hospital, The Fourth Military Medical University, Xi'an 710032, China
| | - Zixiang Wu
- Department of Orthopedics, Xijing Hospital, The Fourth Military Medical University, Xi'an 710032, China.
| | - Wei Lei
- Department of Orthopedics, Xijing Hospital, The Fourth Military Medical University, Xi'an 710032, China.
| |
Collapse
|
38
|
Hussien AA, Niederoest B, Bollhalder M, Goedecke N, Snedeker JG. The Stiffness-Sensitive Transcriptome of Human Tendon Stromal Cells. Adv Healthc Mater 2023; 12:e2101216. [PMID: 36509005 PMCID: PMC11468939 DOI: 10.1002/adhm.202101216] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 11/20/2022] [Indexed: 12/14/2022]
Abstract
Extracellular matrix stiffness is a major regulator of cellular states. Stiffness-sensing investigations are typically performed using cells that have acquired "mechanical memory" through prolonged conditioning in rigid environments, e.g., tissue culture plastic (TCP). This potentially masks the full extent of the matrix stiffness-driven mechanosensing programs. Here, a biomaterial composed of 2D mechanovariant silicone substrates with simplified and scalable surface biofunctionalization chemistry is developed to facilitate large-scale cell culture expansion processes. Using RNA sequencing, stiffness-mediated mechano-responses of human tendon-derived stromal cells are broadly mapped. Matrix elasticity (E.) approximating tendon microscale stiffness range (E. ≈ 35 kPa) distinctly favors transcriptional programs related to chromatin remodeling and Hippo signaling; whereas compliant stiffnesses (E. ≈ 2 kPa) are enriched in processes related to cell stemness, synapse assembly, and angiogenesis. While tendon stromal cells undergo dramatic phenotypic drift on conventional TCP, mechanovariant substrates abrogate this activation with tenogenic stiffnesses inducing a transcriptional program that strongly correlates with established tendon tissue-specific expression signature. Computational inference predicts that AKT1 and ERK1/2 are major stiffness-sensing signaling hubs. Together, these findings highlight how matrix biophysical cues may dictate the transcriptional identity of tendon cells, and how matrix mechano-reciprocity regulates diverse sets of previously underappreciated mechanosensitive processes in tendon fibroblasts.
Collapse
Affiliation(s)
- Amro A. Hussien
- Institute for BiomechanicsETH ZurichZurich8092Switzerland
- Balgrist University HospitalUniversity of ZurichZurich8008Switzerland
| | - Barbara Niederoest
- Institute for BiomechanicsETH ZurichZurich8092Switzerland
- Balgrist University HospitalUniversity of ZurichZurich8008Switzerland
| | - Maja Bollhalder
- Institute for BiomechanicsETH ZurichZurich8092Switzerland
- Balgrist University HospitalUniversity of ZurichZurich8008Switzerland
| | - Nils Goedecke
- Institute for BiomechanicsETH ZurichZurich8092Switzerland
- Balgrist University HospitalUniversity of ZurichZurich8008Switzerland
| | - Jess G. Snedeker
- Institute for BiomechanicsETH ZurichZurich8092Switzerland
- Balgrist University HospitalUniversity of ZurichZurich8008Switzerland
| |
Collapse
|
39
|
Zhang B, Leng J, Ouyang Z, Yang Z, Zhang Q, Li Q, Li D, Zhao H. Superhydrophilic and topography-regulatable surface grafting on PEEK to improve cellular affinity. BIOMATERIALS ADVANCES 2023; 146:213310. [PMID: 36716597 DOI: 10.1016/j.bioadv.2023.213310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 12/29/2022] [Accepted: 01/18/2023] [Indexed: 01/26/2023]
Abstract
Polyetheretherketone (PEEK) has been widely used in the preparation of orthopedic implants due to its biological inertness and similar mechanical modulus to natural bone. However, the affinity between biological tissue (bone and soft tissue) and PEEK surface is weak, leading to low osseointegration and an increased risk of inflammation. The situation could be improved by modifying PEEK surface. Surfaces with good hydrophilicity and proper microtopography would promote cellular adhesion and proliferation. This work presented a two-step surface modification method to achieve the effect. Polyacrylic acid (PAA) chains were grafted on PEEK surface by UV irradiation. Then, ethylenediamine (EDA) was added to introduce amino groups and promote the cross-linking of PAA chains. Furthermore, a mathematical model was built to describe and regulate the surface topography growth process semi-quantitatively. The model fits experimental data quite well (adjusted R2 = 0.779). Results showed that the modified PEEK surface obtained superhydrophilicity. It significantly improved the adhesion and proliferation of BMSCs and MFBs by activating the FAK pathway and Rho family GTPase. The cellular affinity performed better when the surface topography was in network structure with holes in about 25 μm depth and 20-50 μm diameter. Good hydrophilicity seems necessary for the FAK pathway activation, but simply improving surface hydrophilicity might not be enough for cellular affinity improvement. Surface topography at micron scale should be a more important cue. This simple surface modification method could be contributed to further study of cell-microtopography interaction and have potential applications in clinical PEEK orthopedic implants.
Collapse
Affiliation(s)
- Bowen Zhang
- State Key Laboratory for Manufacturing System Engineering, School of Mechanical Engineering, Xi'an Jiaotong University, 710054 Xi'an, Shaanxi, China; National Medical Products Administration (NMPA), Key Laboratory for Research and Evaluation of Additive Manufacturing Medical Devices, Xi'an Jiaotong University, 710054 Xi'an, Shaanxi, China
| | - Junqing Leng
- State Key Laboratory for Manufacturing System Engineering, School of Mechanical Engineering, Xi'an Jiaotong University, 710054 Xi'an, Shaanxi, China; National Medical Products Administration (NMPA), Key Laboratory for Research and Evaluation of Additive Manufacturing Medical Devices, Xi'an Jiaotong University, 710054 Xi'an, Shaanxi, China
| | - Zhicong Ouyang
- Academy of Orthopedics·Guangdong Province, Orthopedic Hospital of Guangdong Province, Department of Spine Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, 510665 Guangzhou, China
| | - Zijian Yang
- Academy of Orthopedics·Guangdong Province, Orthopedic Hospital of Guangdong Province, Department of Spine Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, 510665 Guangzhou, China
| | - Qing Zhang
- State Key Laboratory for Manufacturing System Engineering, School of Mechanical Engineering, Xi'an Jiaotong University, 710054 Xi'an, Shaanxi, China; Center for Medical Device Evaluation, National Medical Products Administration (NMPA), 100081 Beijing, China
| | - Qingchu Li
- Academy of Orthopedics·Guangdong Province, Orthopedic Hospital of Guangdong Province, Department of Spine Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, 510665 Guangzhou, China
| | - Dichen Li
- State Key Laboratory for Manufacturing System Engineering, School of Mechanical Engineering, Xi'an Jiaotong University, 710054 Xi'an, Shaanxi, China; National Medical Products Administration (NMPA), Key Laboratory for Research and Evaluation of Additive Manufacturing Medical Devices, Xi'an Jiaotong University, 710054 Xi'an, Shaanxi, China.
| | - Huiyu Zhao
- Academy of Orthopedics·Guangdong Province, Orthopedic Hospital of Guangdong Province, Department of Spine Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, 510665 Guangzhou, China.
| |
Collapse
|
40
|
Iijima H, Gilmer G, Wang K, Bean AC, He Y, Lin H, Tang WY, Lamont D, Tai C, Ito A, Jones JJ, Evans C, Ambrosio F. Age-related matrix stiffening epigenetically regulates α-Klotho expression and compromises chondrocyte integrity. Nat Commun 2023; 14:18. [PMID: 36627269 PMCID: PMC9832042 DOI: 10.1038/s41467-022-35359-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 11/29/2022] [Indexed: 01/12/2023] Open
Abstract
Extracellular matrix stiffening is a quintessential feature of cartilage aging, a leading cause of knee osteoarthritis. Yet, the downstream molecular and cellular consequences of age-related biophysical alterations are poorly understood. Here, we show that epigenetic regulation of α-Klotho represents a novel mechanosensitive mechanism by which the aged extracellular matrix influences chondrocyte physiology. Using mass spectrometry proteomics followed by a series of genetic and pharmacological manipulations, we discovered that increased matrix stiffness drove Klotho promoter methylation, downregulated Klotho gene expression, and accelerated chondrocyte senescence in vitro. In contrast, exposing aged chondrocytes to a soft matrix restored a more youthful phenotype in vitro and enhanced cartilage integrity in vivo. Our findings demonstrate that age-related alterations in extracellular matrix biophysical properties initiate pathogenic mechanotransductive signaling that promotes Klotho promoter methylation and compromises cellular health. These findings are likely to have broad implications even beyond cartilage for the field of aging research.
Collapse
Affiliation(s)
- Hirotaka Iijima
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, PA, USA.
- Japan Society for the Promotion of Science, Tokyo, Japan.
- Institute for Advanced Research, Nagoya University, Nagoya, Japan.
| | - Gabrielle Gilmer
- Medical Scientist Training Program, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
- Cellular and Molecular Pathology Graduate Program, University of Pittsburgh, Pittsburgh, PA, USA
- Discovery Center for Musculoskeletal Recovery, Schoen Adams Research Institute at Spaulding, Boston, MA, USA
- Department of Physical Medicine & Rehabilitation, Harvard Medical School, Boston, MA, USA
| | - Kai Wang
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, PA, USA
- Discovery Center for Musculoskeletal Recovery, Schoen Adams Research Institute at Spaulding, Boston, MA, USA
- Department of Physical Medicine & Rehabilitation, Harvard Medical School, Boston, MA, USA
| | - Allison C Bean
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, PA, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yuchen He
- Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Hang Lin
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Wan-Yee Tang
- Department of Environmental and Occupational Health, University of Pittsburgh School of Public Health, Pittsburgh, PA, USA
| | - Daniel Lamont
- Petersen Institute of Nanoscience and Engineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Chia Tai
- Department of Motor Function Analysis, Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Akira Ito
- Department of Motor Function Analysis, Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Jeffrey J Jones
- Proteome Exploration Laboratory, Beckman Institute, California Institute of Technology, Pasadena, CA, USA
| | - Christopher Evans
- Department of Physical Medicine & Rehabilitation, Mayo Clinic, Rochester, MN, USA
| | - Fabrisia Ambrosio
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA.
- Discovery Center for Musculoskeletal Recovery, Schoen Adams Research Institute at Spaulding, Boston, MA, USA.
- Department of Physical Medicine & Rehabilitation, Harvard Medical School, Boston, MA, USA.
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Environmental and Occupational Health, University of Pittsburgh School of Public Health, Pittsburgh, PA, USA.
| |
Collapse
|
41
|
Mongera A, Pochitaloff M, Gustafson HJ, Stooke-Vaughan GA, Rowghanian P, Kim S, Campàs O. Mechanics of the cellular microenvironment as probed by cells in vivo during zebrafish presomitic mesoderm differentiation. NATURE MATERIALS 2023; 22:135-143. [PMID: 36577855 PMCID: PMC9812792 DOI: 10.1038/s41563-022-01433-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 11/03/2022] [Indexed: 05/19/2023]
Abstract
Tissue morphogenesis, homoeostasis and repair require cells to constantly monitor their three-dimensional microenvironment and adapt their behaviours in response to local biochemical and mechanical cues. Yet the mechanical parameters of the cellular microenvironment probed by cells in vivo remain unclear. Here, we report the mechanics of the cellular microenvironment that cells probe in vivo and in situ during zebrafish presomitic mesoderm differentiation. By quantifying both endogenous cell-generated strains and tissue mechanics, we show that individual cells probe the stiffness associated with deformations of the supracellular, foam-like tissue architecture. Stress relaxation leads to a perceived microenvironment stiffness that decreases over time, with cells probing the softest regime. We find that most mechanical parameters, including those probed by cells, vary along the anteroposterior axis as mesodermal progenitors differentiate. These findings expand our understanding of in vivo mechanosensation and might aid the design of advanced scaffolds for tissue engineering applications.
Collapse
Affiliation(s)
- Alessandro Mongera
- Department of Mechanical Engineering, University of California, Santa Barbara, CA, USA
- Department of Pathology, Brigham and Women's Hospital and Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Marie Pochitaloff
- Department of Mechanical Engineering, University of California, Santa Barbara, CA, USA
| | - Hannah J Gustafson
- Department of Mechanical Engineering, University of California, Santa Barbara, CA, USA
- Biomolecular Science and Engineering Program, University of California, Santa Barbara, CA, USA
| | | | - Payam Rowghanian
- Department of Mechanical Engineering, University of California, Santa Barbara, CA, USA
| | - Sangwoo Kim
- Department of Mechanical Engineering, University of California, Santa Barbara, CA, USA
| | - Otger Campàs
- Department of Mechanical Engineering, University of California, Santa Barbara, CA, USA.
- Center for Systems Biology Dresden, Dresden, Germany.
- Cluster of Excellence Physics of Life, TU Dresden, Dresden, Germany.
| |
Collapse
|
42
|
Fernandez Cabada T, Ruben M, El Merhie A, Proietti Zaccaria R, Alabastri A, Petrini EM, Barberis A, Salerno M, Crepaldi M, Davis A, Ceseracciu L, Catelani T, Athanassiou A, Pellegrino T, Cingolani R, Papadopoulou EL. Electrostatic polarization fields trigger glioblastoma stem cell differentiation. NANOSCALE HORIZONS 2022; 8:95-107. [PMID: 36426604 PMCID: PMC9765404 DOI: 10.1039/d2nh00453d] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 11/17/2022] [Indexed: 06/16/2023]
Abstract
Over the last few years it has been understood that the interface between living cells and the underlying materials can be a powerful tool to manipulate cell functions. In this study, we explore the hypothesis that the electrical cell/material interface can regulate the differentiation of cancer stem-like cells (CSCs). Electrospun polymer fibres, either polyamide 66 or poly(lactic acid), with embedded graphene nanoplatelets (GnPs), have been fabricated as CSC scaffolds, providing both the 3D microenvironment and a suitable electrical environment favorable for CSCs adhesion, growth and differentiation. We have investigated the impact of these scaffolds on the morphological, immunostaining and electrophysiological properties of CSCs extracted from human glioblastoma multiform (GBM) tumor cell line. Our data provide evidence in favor of the ability of GnP-incorporating scaffolds to promote CSC differentiation to the glial phenotype. Numerical simulations support the hypothesis that the electrical interface promotes the hyperpolarization of the cell membrane potential, thus triggering the CSC differentiation. We propose that the electrical cell/material interface can regulate endogenous bioelectrical cues, through the membrane potential manipulation, resulting in the differentiation of CSCs. Material-induced differentiation of stem cells and particularly of CSCs, can open new horizons in tissue engineering and new approaches to cancer treatment, especially GBM.
Collapse
Affiliation(s)
| | - Massimo Ruben
- Istituto Italiano di Tecnologia, via Morego 30, 16163 Genova, Italy.
| | - Amira El Merhie
- Istituto Italiano di Tecnologia, via Morego 30, 16163 Genova, Italy.
| | | | - Alessandro Alabastri
- Department of Electrical and Computer Engineering, Rice University, 6100 Main Street, Houston, TX, 77005, USA
| | | | - Andrea Barberis
- Istituto Italiano di Tecnologia, via Morego 30, 16163 Genova, Italy.
| | - Marco Salerno
- Istituto Italiano di Tecnologia, via Morego 30, 16163 Genova, Italy.
| | - Marco Crepaldi
- Istituto Italiano di Tecnologia, via Melen 83, 16152 Genova, Italy
| | - Alexander Davis
- Istituto Italiano di Tecnologia, via Morego 30, 16163 Genova, Italy.
| | - Luca Ceseracciu
- Istituto Italiano di Tecnologia, via Morego 30, 16163 Genova, Italy.
| | - Tiziano Catelani
- Istituto Italiano di Tecnologia, via Morego 30, 16163 Genova, Italy.
| | | | - Teresa Pellegrino
- Istituto Italiano di Tecnologia, via Morego 30, 16163 Genova, Italy.
| | - Roberto Cingolani
- Istituto Italiano di Tecnologia, via Morego 30, 16163 Genova, Italy.
| | | |
Collapse
|
43
|
Zhong JX, Raghavan P, Desai TA. Harnessing Biomaterials for Immunomodulatory-Driven Tissue Engineering. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2022; 9:224-239. [PMID: 37333620 PMCID: PMC10272262 DOI: 10.1007/s40883-022-00279-6] [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: 05/03/2022] [Revised: 08/08/2022] [Accepted: 09/07/2022] [Indexed: 11/29/2022]
Abstract
Abstract The immune system plays a crucial role during tissue repair and wound healing processes. Biomaterials have been leveraged to assist in this in situ tissue regeneration process to dampen the foreign body response by evading or suppressing the immune system. An emerging paradigm within regenerative medicine is to use biomaterials to influence the immune system and create a pro-reparative microenvironment to instigate endogenously driven tissue repair. In this review, we discuss recent studies that focus on immunomodulation of innate and adaptive immune cells for tissue engineering applications through four biomaterial-based mechanisms of action: biophysical cues, chemical modifications, drug delivery, and sequestration. These materials enable augmented regeneration in various contexts, including vascularization, bone repair, wound healing, and autoimmune regulation. While further understanding of immune-material interactions is needed to design the next generation of immunomodulatory biomaterials, these materials have already demonstrated great promise for regenerative medicine. Lay Summary The immune system plays an important role in tissue repair. Many biomaterial strategies have been used to promote tissue repair, and recent work in this area has looked into the possibility of doing repair by tuning. Thus, we examined the literature for recent works showcasing the efficacy of these approaches in animal models of injuries. In these studies, we found that biomaterials successfully tuned the immune response and improved the repair of various tissues. This highlights the promise of immune-modulating material strategies to improve tissue repair.
Collapse
Affiliation(s)
- Justin X. Zhong
- UC Berkeley – UCSF Graduate Program in Bioengineering, San Francisco, CA 94143 USA
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA 94143 USA
| | - Preethi Raghavan
- UC Berkeley – UCSF Graduate Program in Bioengineering, San Francisco, CA 94143 USA
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA 94143 USA
| | - Tejal A. Desai
- UC Berkeley – UCSF Graduate Program in Bioengineering, San Francisco, CA 94143 USA
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA 94143 USA
- Department of Bioengineering, University of California, Berkeley, CA 94720 USA
- School of Engineering, Brown University, Providence, RI 02912 USA
| |
Collapse
|
44
|
Walker CJ, Batan D, Bishop CT, Ramirez D, Aguado BA, Schroeder ME, Crocini C, Schwisow J, Moulton K, Macdougall L, Weiss RM, Allen MA, Dowell R, Leinwand LA, Anseth KS. Extracellular matrix stiffness controls cardiac valve myofibroblast activation through epigenetic remodeling. Bioeng Transl Med 2022; 7:e10394. [PMID: 36176599 PMCID: PMC9472021 DOI: 10.1002/btm2.10394] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 07/11/2022] [Accepted: 07/27/2022] [Indexed: 11/29/2022] Open
Abstract
Aortic valve stenosis (AVS) is a progressive fibrotic disease that is caused by thickening and stiffening of valve leaflets. At the cellular level, quiescent valve interstitial cells (qVICs) activate to myofibroblasts (aVICs) that persist within the valve tissue. Given the persistence of myofibroblasts in AVS, epigenetic mechanisms have been implicated. Here, we studied changes that occur in VICs during myofibroblast activation by using a hydrogel matrix to recapitulate different stiffnesses in the valve leaflet during fibrosis. We first compared the chromatin landscape of qVICs cultured on soft hydrogels and aVICs cultured on stiff hydrogels, representing the native and diseased phenotypes respectively. Using assay for transposase-accessible chromatin sequencing (ATAC-Seq), we found that open chromatin regions in aVICs were enriched for transcription factor binding motifs associated with mechanosensing pathways compared to qVICs. Next, we used RNA-Seq to show that the open chromatin regions in aVICs correlated with pro-fibrotic gene expression, as aVICs expressed higher levels of contractile fiber genes, including myofibroblast markers such as alpha smooth muscle actin (αSMA), compared to qVICs. In contrast, chromatin remodeling genes were downregulated in aVICs compared to qVICs, indicating qVICs may be protected from myofibroblast activation through epigenetic mechanisms. Small molecule inhibition of one of these remodelers, CREB Binding Protein (CREBBP), prevented qVICs from activating to aVICs. Notably, CREBBP is more abundant in valves from healthy patients compared to fibrotic valves. Our findings reveal the role of mechanical regulation in chromatin remodeling during VIC activation and quiescence and highlight one potential therapeutic target for treating AVS.
Collapse
Affiliation(s)
- Cierra J. Walker
- Materials Science and Engineering ProgramUniversity of Colorado BoulderBoulderColoradoUSA
- BioFrontiers Institute, University of Colorado BoulderBoulderColoradoUSA
| | - Dilara Batan
- BioFrontiers Institute, University of Colorado BoulderBoulderColoradoUSA
- Biochemistry DepartmentUniversity of Colorado BoulderBoulderColoradoUSA
| | - Carrie T. Bishop
- Chemical and Biological Engineering DepartmentUniversity of Colorado BoulderBoulderColoradoUSA
| | - Daniel Ramirez
- BioFrontiers Institute, University of Colorado BoulderBoulderColoradoUSA
- Molecular, Cellular, and Developmental Biology DepartmentUniversity of Colorado BoulderBoulderColoradoUSA
| | - Brian A. Aguado
- BioFrontiers Institute, University of Colorado BoulderBoulderColoradoUSA
- Chemical and Biological Engineering DepartmentUniversity of Colorado BoulderBoulderColoradoUSA
| | - Megan E. Schroeder
- BioFrontiers Institute, University of Colorado BoulderBoulderColoradoUSA
- Chemical and Biological Engineering DepartmentUniversity of Colorado BoulderBoulderColoradoUSA
| | - Claudia Crocini
- BioFrontiers Institute, University of Colorado BoulderBoulderColoradoUSA
- Molecular, Cellular, and Developmental Biology DepartmentUniversity of Colorado BoulderBoulderColoradoUSA
| | - Jessica Schwisow
- Division of CardiologyUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
| | - Karen Moulton
- Division of CardiologyUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
| | - Laura Macdougall
- BioFrontiers Institute, University of Colorado BoulderBoulderColoradoUSA
- Chemical and Biological Engineering DepartmentUniversity of Colorado BoulderBoulderColoradoUSA
| | - Robert M. Weiss
- Department of Internal MedicineUniversity of IowaIowa CityIowaUSA
| | - Mary A. Allen
- BioFrontiers Institute, University of Colorado BoulderBoulderColoradoUSA
- Molecular, Cellular, and Developmental Biology DepartmentUniversity of Colorado BoulderBoulderColoradoUSA
| | - Robin Dowell
- BioFrontiers Institute, University of Colorado BoulderBoulderColoradoUSA
- Molecular, Cellular, and Developmental Biology DepartmentUniversity of Colorado BoulderBoulderColoradoUSA
| | - Leslie A. Leinwand
- BioFrontiers Institute, University of Colorado BoulderBoulderColoradoUSA
- Molecular, Cellular, and Developmental Biology DepartmentUniversity of Colorado BoulderBoulderColoradoUSA
| | - Kristi S. Anseth
- BioFrontiers Institute, University of Colorado BoulderBoulderColoradoUSA
- Chemical and Biological Engineering DepartmentUniversity of Colorado BoulderBoulderColoradoUSA
| |
Collapse
|
45
|
Xu Z, Li Y, Li P, Sun Y, Lv S, Wang Y, He X, Xu J, Xu Z, Li L, Li Y. Soft substrates promote direct chemical reprogramming of fibroblasts into neurons. Acta Biomater 2022; 152:255-272. [PMID: 36041647 DOI: 10.1016/j.actbio.2022.08.049] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 08/13/2022] [Accepted: 08/23/2022] [Indexed: 11/01/2022]
Abstract
Fibroblasts can be directly reprogrammed via a combination of small molecules to generate induced neurons (iNs), bypassing intermediate stages. This method holds great promise for regenerative medicine; however, it remains inefficient. Recently, studies have suggested that physical cues may improve the direct reprogramming of fibroblasts into neurons, but the underlying mechanisms remain to be further explored, and the physical factors reported to date do not exhibit the full properties of the extracellular matrix (ECM). Previous in vitro studies mainly used rigid polystyrene dishes, while one of the characteristics of the native in-vivo environment of neurons is the soft nature of brain ECM. The reported stiffness of brain tissue is very soft ranging between 100 Pa and 3 kPa, and the effect of substrate stiffness on direct neuronal reprogramming has not been explored. Here, we show for the first time that soft substrates substantially improved the production efficiency and quality of iNs, without needing to co-culture with glial cells during reprogramming, producing more glutamatergic neurons with electrophysiological functions in a shorter time. Transcriptome sequencing indicated that soft substrates might promote glutamatergic neuron reprogramming through integrins, actin cytoskeleton, Hippo signalling pathway, and regulation of mesenchymal-to-epithelial transition, and competing endogenous RNA network analysis provided new targets for neuronal reprogramming. We demonstrated that soft substrates may promote neuronal reprogramming by inhibiting microRNA-615-3p-targeting integrin subunit beta 4. Our findings can aid the development of regenerative therapies and help improve our understanding of neuronal reprogramming. STATEMENT OF SIGNIFICANCE: : First, we have shown that low stiffness promotes direct reprogramming on the basis of small molecule combinations. To the best of our knowledge, this is the first report on this type of method, which may greatly promote the progress of neural reprogramming. Second, we found that miR-615-3p may interact with ITGB4, and the soft substrates may promote neural reprogramming by inhibiting microRNA (miR)-615-3p targeting integrin subunit beta 4 (ITGB4). We are the first to report on this mechanism. Our findings will provide more functional neurons for subsequent basic and clinical research in neurological regenerative medicine, and will help to improve the overall understanding of neural reprogramming. This work also provides new ideas for the design of medical biomaterials for nerve regeneration.
Collapse
Affiliation(s)
- Ziran Xu
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| | - Yan Li
- Division of Orthopedics and Biotechnology, Department for Clinical Intervention and Technology (CLINTEC), Karolinska Institute, Stockholm, Sweden.
| | - Pengdong Li
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan 511518, Guangdong, China.
| | - Yingying Sun
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; Department of Stomatology, The First Hospital of Jilin University, Changchun 130021, China.
| | - Shuang Lv
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| | - Yin Wang
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| | - Xia He
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; Department of Pathology, Shanxi Bethune Hospital, Taiyuan 030032, China.
| | - Jinying Xu
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; Department of Burns Surgery, The First Hospital of Jilin University, Changchun 130000, China.
| | - Zhixiang Xu
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| | - Lisha Li
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| | - Yulin Li
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| |
Collapse
|
46
|
Wang C, Yang J. Mechanical forces: The missing link between idiopathic pulmonary fibrosis and lung cancer. Eur J Cell Biol 2022; 101:151234. [DOI: 10.1016/j.ejcb.2022.151234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 05/05/2022] [Accepted: 05/05/2022] [Indexed: 11/25/2022] Open
|
47
|
Lecoutre S, Lambert M, Drygalski K, Dugail I, Maqdasy S, Hautefeuille M, Clément K. Importance of the Microenvironment and Mechanosensing in Adipose Tissue Biology. Cells 2022; 11:cells11152310. [PMID: 35954152 PMCID: PMC9367348 DOI: 10.3390/cells11152310] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 07/19/2022] [Accepted: 07/23/2022] [Indexed: 11/16/2022] Open
Abstract
The expansion of adipose tissue is an adaptive mechanism that increases nutrient buffering capacity in response to an overall positive energy balance. Over the course of expansion, the adipose microenvironment undergoes continual remodeling to maintain its structural and functional integrity. However, in the long run, adipose tissue remodeling, typically characterized by adipocyte hypertrophy, immune cells infiltration, fibrosis and changes in vascular architecture, generates mechanical stress on adipose cells. This mechanical stimulus is then transduced into a biochemical signal that alters adipose function through mechanotransduction. In this review, we describe the physical changes occurring during adipose tissue remodeling, and how they regulate adipose cell physiology and promote obesity-associated dysfunction in adipose tissue.
Collapse
Affiliation(s)
- Simon Lecoutre
- Nutrition and Obesities: Systemic Approaches Research Group (Nutri-Omics), Sorbonne Université, INSERM, F-75013 Paris, France; (S.L.); (K.D.); (I.D.)
| | - Mélanie Lambert
- Labex Inflamex, Université Sorbonne Paris Nord, INSERM, F-93000 Bobigny, France;
| | - Krzysztof Drygalski
- Nutrition and Obesities: Systemic Approaches Research Group (Nutri-Omics), Sorbonne Université, INSERM, F-75013 Paris, France; (S.L.); (K.D.); (I.D.)
| | - Isabelle Dugail
- Nutrition and Obesities: Systemic Approaches Research Group (Nutri-Omics), Sorbonne Université, INSERM, F-75013 Paris, France; (S.L.); (K.D.); (I.D.)
| | - Salwan Maqdasy
- Department of Medicine (H7), Karolinska Institutet Hospital, C2-94, 14186 Stockholm, Sweden;
| | - Mathieu Hautefeuille
- Laboratoire de Biologie du Développement (UMR 7622), IBPS, Sorbonne Université, F-75005 Paris, France;
| | - Karine Clément
- Nutrition and Obesities: Systemic Approaches Research Group (Nutri-Omics), Sorbonne Université, INSERM, F-75013 Paris, France; (S.L.); (K.D.); (I.D.)
- Assistance Publique Hôpitaux de Paris, Nutrition Department, CRNH Ile-de-France, Pitié-Salpêtrière Hospital, F-75013 Paris, France
- Correspondence: or
| |
Collapse
|
48
|
Rhodes ADY, Duran-Mota JA, Oliva N. Current progress in bionanomaterials to modulate the epigenome. Biomater Sci 2022; 10:5081-5091. [PMID: 35880652 DOI: 10.1039/d2bm01027e] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Recent advances in genomics during the 1990s have made it possible to study and identify genetic and epigenetic responses of cells and tissues to various drugs and environmental factors. This has accelerated the number of targets available to treat a range of diseases from cancer to wound healing disorders. Equally interesting is the understanding of how bio- and nanomaterials alter gene expression through epigenetic mechanisms, and whether they have the potential to elicit a positive therapeutic response without requiring additional biomolecule delivery. In fact, from a cell's perspective, a biomaterial is nothing more than an environmental factor, and so it has the power to epigenetically modulate gene expression of cells in contact with it. Understanding these epigenetic interactions between biomaterials and cells will open new avenues in the development of technologies that can not only provide biological signals (i.e. drugs, growth factors) necessary for therapy and regeneration, but also intimately interact with cells to promote the expression of genes of interest. This review article aims to summarise the current state-of-the-art and progress on the development of bio- and nanomaterials to modulate the epigenome.
Collapse
Affiliation(s)
- Anna D Y Rhodes
- Department of Bioengineering, Imperial College London, London W12 0BZ, UK.
| | - Jose Antonio Duran-Mota
- Department of Bioengineering, Imperial College London, London W12 0BZ, UK. .,Materials Engineering Group (GEMAT), IQS Barcelona, Barcelona 08017, Spain
| | - Nuria Oliva
- Department of Bioengineering, Imperial College London, London W12 0BZ, UK.
| |
Collapse
|
49
|
Hsu CC, Serio A, Gopal S, Gelmi A, Chiappini C, Desai RA, Stevens MM. Biophysical Regulations of Epigenetic State and Notch Signaling in Neural Development Using Microgroove Substrates. ACS APPLIED MATERIALS & INTERFACES 2022; 14:32773-32787. [PMID: 35830496 PMCID: PMC9335410 DOI: 10.1021/acsami.2c01996] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
A number of studies have recently shown how surface topography can alter the behavior and differentiation patterns of different types of stem cells. Although the exact mechanisms and molecular pathways involved remain unclear, a consistent portion of the literature points to epigenetic changes induced by nuclear remodeling. In this study, we investigate the behavior of clinically relevant neural populations derived from human pluripotent stem cells when cultured on polydimethylsiloxane microgrooves (3 and 10 μm depth grooves) to investigate what mechanisms are responsible for their differentiation capacity and functional behavior. Our results show that microgrooves enhance cell alignment, modify nuclear geometry, and significantly increase cellular stiffness, which we were able to measure at high resolution with a combination of light and electron microscopy, scanning ion conductance microscopy (SICM), and atomic force microscopy (AFM) coupled with quantitative image analysis. The microgrooves promoted significant changes in the epigenetic landscape, as revealed by the expression of key histone modification markers. The main behavioral change of neural stem cells on microgrooves was an increase of neuronal differentiation under basal conditions on the microgrooves. Through measurements of cleaved Notch1 levels, we found that microgrooves downregulate Notch signaling. We in fact propose that microgroove topography affects the differentiation potential of neural stem cells by indirectly altering Notch signaling through geometric segregation and that this mechanism in parallel with topography-dependent epigenetic modulations acts in concert to enhance stem cell neuronal differentiation.
Collapse
Affiliation(s)
- Chia-Chen Hsu
- Department
of Materials, Imperial College London, Exhibition Road, London SW7 2AZ, U.K.
- Department
of Bioengineering, Imperial College London, Exhibition Road, London SW7 2AZ, U.K.
- Institute
of Biomedical Engineering, Imperial College
London, Exhibition Road, London SW7 2AZ, U.K.
| | - Andrea Serio
- Department
of Materials, Imperial College London, Exhibition Road, London SW7 2AZ, U.K.
- Department
of Bioengineering, Imperial College London, Exhibition Road, London SW7 2AZ, U.K.
- Institute
of Biomedical Engineering, Imperial College
London, Exhibition Road, London SW7 2AZ, U.K.
| | - Sahana Gopal
- Department
of Materials, Imperial College London, Exhibition Road, London SW7 2AZ, U.K.
- Department
of Bioengineering, Imperial College London, Exhibition Road, London SW7 2AZ, U.K.
- Institute
of Biomedical Engineering, Imperial College
London, Exhibition Road, London SW7 2AZ, U.K.
| | - Amy Gelmi
- Department
of Materials, Imperial College London, Exhibition Road, London SW7 2AZ, U.K.
- Department
of Bioengineering, Imperial College London, Exhibition Road, London SW7 2AZ, U.K.
- Institute
of Biomedical Engineering, Imperial College
London, Exhibition Road, London SW7 2AZ, U.K.
| | - Ciro Chiappini
- Department
of Materials, Imperial College London, Exhibition Road, London SW7 2AZ, U.K.
- Department
of Bioengineering, Imperial College London, Exhibition Road, London SW7 2AZ, U.K.
- Institute
of Biomedical Engineering, Imperial College
London, Exhibition Road, London SW7 2AZ, U.K.
| | - Ravi A. Desai
- Department
of Materials, Imperial College London, Exhibition Road, London SW7 2AZ, U.K.
- Department
of Bioengineering, Imperial College London, Exhibition Road, London SW7 2AZ, U.K.
- Institute
of Biomedical Engineering, Imperial College
London, Exhibition Road, London SW7 2AZ, U.K.
| | - Molly M. Stevens
- Department
of Materials, Imperial College London, Exhibition Road, London SW7 2AZ, U.K.
- Department
of Bioengineering, Imperial College London, Exhibition Road, London SW7 2AZ, U.K.
- Institute
of Biomedical Engineering, Imperial College
London, Exhibition Road, London SW7 2AZ, U.K.
| |
Collapse
|
50
|
A Molecular View on Biomaterials and Dental Stem Cells Interactions: Literature Review. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12125815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Biomaterials and stem cells are essential components in the field of regenerative medicine. Various biomaterials have been designed that have appropriate biochemical and biophysical characteristics to mimic the microenvironment of an extracellular matrix. Dental stem cells (DT-MSCs) represent a novel source for the development of autologous therapies due to their easy availability. Although research on biomaterials and DT-MSCs has progressed, there are still challenges in the characteristics of biomaterials and the molecular mechanisms involved in regulating the behavior of DT-MSCs. In this review, the characteristics of biomaterials are summarized, and their classification according to their source, bioactivity, and different biological effects on the expansion and differentiation of DT-MSCs is summarized. Finally, advances in research on the interaction of biomaterials and the molecular components involved (mechanosensors and mechanotransduction) in DT-MSCs during their proliferation and differentiation are analyzed. Understanding the molecular dynamics of DT-MSCs and biomaterials can contribute to research in regenerative medicine and the development of autologous stem cell therapies.
Collapse
|