1
|
Su X, Wang M, Yuan R, Guo L, Han Y, Huang C, Li A, Kaplan DL, Wang X. Organoids in Dynamic Culture: Microfluidics and 3D Printing Technologies. ACS Biomater Sci Eng 2025. [PMID: 40248908 DOI: 10.1021/acsbiomaterials.4c02245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2025]
Abstract
With the rapid advancement of biomaterials and tissue engineering technologies, organoid research and its applications have made significant strides. Organoids are increasingly utilized in pharmacology, regenerative medicine, and precision clinical medicine. Current trends in organoid research are moving toward multifunctional composite three-dimensional cultivation and dynamic cultivation strategies. Key technologies driving this evolution, including 3D printing and microfluidics, continue to impact new areas of discovery and clinical relevance. This review provides a systematic overview of these emerging trends, discussing the strengths and limitations of these critical technologies and offering insight and research directions for professionals working in the organoid field.
Collapse
Affiliation(s)
- Xin Su
- College of Basic Medical Sciences, Dalian Medical University, Dalian, China 116044
| | - Mingqi Wang
- College of Basic Medical Sciences, Dalian Medical University, Dalian, China 116044
| | - Ruqiang Yuan
- College of Basic Medical Sciences, Dalian Medical University, Dalian, China 116044
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China 116044
| | - Lina Guo
- College of Basic Medical Sciences, Dalian Medical University, Dalian, China 116044
| | - Yinhe Han
- College of Basic Medical Sciences, Dalian Medical University, Dalian, China 116044
| | - Chun Huang
- College of Basic Medical Sciences, Dalian Medical University, Dalian, China 116044
| | - Ang Li
- College of Basic Medical Sciences, Dalian Medical University, Dalian, China 116044
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, United States
| | - Xiuli Wang
- College of Basic Medical Sciences, Dalian Medical University, Dalian, China 116044
| |
Collapse
|
2
|
Limyati Y, Lucretia T, Gunadi JW, Vitriana V, Jasaputra DK, De Mello Wahyudi K, Lesmana R. Chronic moderate‑intensity exercise can induce physiological hypertrophy in aged cardiomyocytes through autophagy, with minimal Yap/Taz involvement. Biomed Rep 2025; 22:44. [PMID: 39882338 PMCID: PMC11775639 DOI: 10.3892/br.2025.1922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 12/13/2024] [Indexed: 01/31/2025] Open
Abstract
Aging is known to cause increased comorbidities associated with cardiovascular decline. Physical exercises were known to be an effective intervention for the age-associated decline in cardiac function. Exercise caused physiological hypertrophy influenced by Yap/Taz, autophagy and myosin heavy chain (MHC) dynamics. However, whether exercise-induced changes are associated with aging has yet to be determined. The present study explored the effects of moderate-intensity exercises on autophagy, MHC dynamics, and Yap/Taz activity to understand their complex interactions at the molecular effects on the cardiac function of aging cardiac tissue. The present study used male Wistar (Rattus norvegicus) rats (80 weeks-old) randomly divided into two groups (n=12): control and intervention. The intervention group was given an intervention using an animal treadmill. After 8 weeks, the animal was sacrificed, and data were collected. Statistical analysis was conducted using an independent t-test or Mann-Whitney U test when appropriate. Exercise in aged rats can induce physiological hypertrophy, as shown by gross measurement and histological features. Yap/Taz did not mediate the effects of exercise on hypertrophy. Autophagy function was shown to increase, which may cause the low expression of Yap/Taz. In conclusion, exercise is a viable intervention in increasing heart mass and potentially delaying the decline in function associated with aging.
Collapse
Affiliation(s)
- Yenni Limyati
- Pasca Sarjana Faculty of Medicine Universitas Padjadjaran, Bandung, West Java 40164, Indonesia
- Department of Clinical Skills, Faculty of Medicine, Maranatha Christian University, Bandung, West Java 40164, Indonesia
- Department of Physical Medicine and Rehabilitation, Unggul Karsa Medika Hospital, Bandung, West Java 40164, Indonesia
| | - Teresa Lucretia
- Department of Histology, Faculty of Medicine, Maranatha Christian University, Bandung, West Java 40164, Indonesia
| | - Julia Windi Gunadi
- Department of Physiology, Faculty of Medicine, Maranatha Christian University, Bandung, West Java 40164, Indonesia
| | - Vitriana Vitriana
- Department of Physical Medicine and Rehabilitation, Faculty of Medicine Universitas Padjadjaran/Dr. Hasan Sadikin General Hospital Bandung, West Java 40164, Indonesia
| | - Diana Krisanti Jasaputra
- Department of Pharmacology, Faculty of Medicine, Maranatha Christian University, Bandung, West Java 40164, Indonesia
| | - Kevin De Mello Wahyudi
- Undergraduate Program in Medicine, Faculty of Medicine, Maranatha Christian University, Bandung, West Java 40164, Indonesia
| | - Ronny Lesmana
- Physiology Molecular, Biological Activity Division, Central Laboratory, Sumedang, West Java 45363, Indonesia
- Department of Biomedical Science, Faculty of Medicine, Universitas Padjadjaran, Bandung, West Java 40164, Indonesia
| |
Collapse
|
3
|
Nguyen MT, Ly QK, Ngo THP, Lee W. Calponin 3 Regulates Myoblast Proliferation and Differentiation Through Actin Cytoskeleton Remodeling and YAP1-Mediated Signaling in Myoblasts. Cells 2025; 14:142. [PMID: 39851570 PMCID: PMC11764405 DOI: 10.3390/cells14020142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 01/09/2025] [Accepted: 01/17/2025] [Indexed: 01/26/2025] Open
Abstract
An actin-binding protein, known as Calponin 3 (CNN3), modulates the remodeling of the actin cytoskeleton, a fundamental process for the maintenance of skeletal muscle homeostasis. Although the roles of CNN3 in actin remodeling have been established, its biological significance in myoblast differentiation remains largely unknown. This study investigated the functional significance of CNN3 in myogenic differentiation, along with its effects on actin remodeling and mechanosensitive signaling in C2C12 myoblasts. CNN3 knockdown led to a marked increase in filamentous actin, which promoted the nuclear localization of Yes-associated protein 1 (YAP1), a mechanosensitive transcriptional coactivator required for response to the mechanical cues that drive cell proliferation. Subsequently, CNN3 depletion enhanced myoblast proliferation by upregulating the expression of the YAP1 target genes related to cell cycle progression, such as cyclin B1, cyclin D1, and PCNA. According to a flow cytometry analysis, CNN3-deficient cells displayed higher S and G2/M phase fractions, which concurred with elevated proliferation rates. Furthermore, CNN3 knockdown impaired myogenic differentiation, as evidenced by reduced levels of MyoD, MyoG, and MyHC, key markers of myogenic commitment and maturation, and immunocytochemistry showed that myotube formation was diminished in CNN3-suppressed cells, which was supported by lower differentiation and fusion indices. These findings reveal that CNN3 is essential for myogenic differentiation, playing a key role in regulating actin remodeling and cellular localization of YAP1 to orchestrate the proliferation and differentiation in myogenic progenitor cells. This study highlights CNN3 as a critical regulator of skeletal myogenesis and suggests its therapeutic potential as a target for muscle atrophy and related disorders.
Collapse
Affiliation(s)
- Mai Thi Nguyen
- Department of Biochemistry, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju 38066, Republic of Korea; (M.T.N.); (Q.K.L.); (T.H.P.N.)
| | - Quoc Kiet Ly
- Department of Biochemistry, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju 38066, Republic of Korea; (M.T.N.); (Q.K.L.); (T.H.P.N.)
| | - Thanh Huu Phan Ngo
- Department of Biochemistry, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju 38066, Republic of Korea; (M.T.N.); (Q.K.L.); (T.H.P.N.)
| | - Wan Lee
- Department of Biochemistry, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju 38066, Republic of Korea; (M.T.N.); (Q.K.L.); (T.H.P.N.)
- Section of Molecular and Cellular Medicine, Medical Institute of Dongguk University, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju 38066, Republic of Korea
- Channelopathy Research Center (CRC), Dongguk University College of Medicine, 32 Dongguk-ro, Ilsan Dong-gu, Goyang 10326, Republic of Korea
| |
Collapse
|
4
|
Qin X, Niu W, Zhao K, Luo Y, Wang W, He Y, Yang F, Cao B, Du M, Su H. Resveratrol enhances post-injury muscle regeneration by regulating antioxidant and mitochondrial biogenesis. Curr Res Food Sci 2025; 10:100972. [PMID: 39896273 PMCID: PMC11787617 DOI: 10.1016/j.crfs.2025.100972] [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: 10/18/2024] [Revised: 01/03/2025] [Accepted: 01/06/2025] [Indexed: 02/04/2025] Open
Abstract
Resveratrol (RES), a natural polyphenolic compound, has shown promise in enhancing skeletal muscle regeneration and metabolic function. This study aims to explore the impact of RES on muscle regeneration after injury through the regulation of antioxidant capacity and mitochondrial biogenesis. RES treatment significantly increased the ratio of tibialis anterior muscle mass to body weight, alongside reduced fasting glucose levels. Following cardiotoxin-induced injury, RES treatment improved muscle regeneration, characterized by greater formation of new fibers and better structural repair compared to the control. Notably, gene expression analyses demonstrated that RES-treated mice exhibited elevated levels of myogenic markers, such as paired box 7 (Pax7), myogenic factor 5 (Myf5), myoblast determination protein (MyoD), and Myogenin (MyoG). Concurrently, yes-associated protein (YAP) increased, underscoring its role in regulating satellite cell activity. Transcriptomic analysis identified enriched pathways related to muscle regeneration and mitochondrial biogenesis, with increased expression of mitochondrial transcription factors and higher mitochondrial DNA content in RES-treated mice. Furthermore, RES enhanced antioxidant capacity via the Kelch-like ECH-associated protein 1 (KEAP-1)/nuclear factor erythroid 2-related factor 2 (NRF2)/heme oxygenase-1 (HO-1) signaling pathway, as indicated by elevated activities of total antioxidant capacity, Glutathione peroxidase (GSH-PX), and superoxidase dismutase (SOD). Collectively, these findings suggest that RES not only promotes muscle regeneration but also supports mitochondrial function and antioxidant defenses, positioning it as a food-derived pharmaceutical for targeting muscle repair after injury.
Collapse
Affiliation(s)
- Xiaoli Qin
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Wenjing Niu
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, 100193, China
| | - Kai Zhao
- Faculty of Engineering and Applied Science, University of Regina, Regina, Saskatchewan, S4S0A2, Canada
| | - Yawen Luo
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Wenfang Wang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Yang He
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Fuyu Yang
- College of Grassland Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Binghai Cao
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Min Du
- Department of Animal Sciences, Washington State University, Pullman, WA, USA
| | - Huawei Su
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| |
Collapse
|
5
|
Mihaly E, Chellu N, Iyer SR, Su EY, Altamirano DE, Dias ST, Grayson WL. Neuromuscular Regeneration of Volumetric Muscle Loss Injury in Response to Agrin-Functionalized Tissue Engineered Muscle Grafts and Rehabilitative Exercise. Adv Healthc Mater 2025; 14:e2403028. [PMID: 39523723 PMCID: PMC11803514 DOI: 10.1002/adhm.202403028] [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: 08/13/2024] [Revised: 10/29/2024] [Indexed: 11/16/2024]
Abstract
Neuromuscular deficits compound the loss of contractile tissue in volumetric muscle loss (VML). Two avenues for promoting recovery are neuromuscular junction (NMJ)-promoting substrates (e.g., agrin) and endurance exercise. Although mechanical stimulation enhances agrin-induced NMJ formation, the two modalities have yet to be evaluated combinatorially. It is hypothesized that the implantation of human myogenic progenitor-seeded tissue-engineered muscle grafts (hTEMGs) in combination with agrin treatment and/or exercise will enhance neuromuscular recovery after VML. The hTEMGs alone transplant into VML defects promote significant regeneration with minimal scarring. A sex-appropriate, low-intensity continuous running exercise paradigm increases acetylcholine receptor (AChR) cluster density in male mice twofold relative to hTEMG alone after 7 weeks of treadmill training (p < 0.05). To further promote neuromuscular recovery, agrin is incorporated into the scaffolds via covalent tethering. In vitro, agrin increases the proliferation of hMPs, and trends toward greater myogenic maturity and AChR clustering. Upon transplantation, both hTEMGs + agrin and hTEMGs + exercise induce near 100% recovery of muscle mass and increase twitch and tetanic force output (p > 0.05). However, agrin treatment in combination with exercise produces no additional benefit. These data highlight the unprecedented regenerative potential of using hTEMGs together with either agrin or exercise supplementation to treat VML injuries.
Collapse
Affiliation(s)
- Eszter Mihaly
- Translational Tissue Engineering Center, School of Medicine, Johns Hopkins University, Baltimore, MD 21231, USA
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Neha Chellu
- Translational Tissue Engineering Center, School of Medicine, Johns Hopkins University, Baltimore, MD 21231, USA
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Shama R. Iyer
- School of Science, Mathematics & Engineering, Marymount University, Arlington VA 22207, USA
| | - Eileen Y. Su
- Translational Tissue Engineering Center, School of Medicine, Johns Hopkins University, Baltimore, MD 21231, USA
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Dallas E. Altamirano
- Translational Tissue Engineering Center, School of Medicine, Johns Hopkins University, Baltimore, MD 21231, USA
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Shaquielle T. Dias
- Translational Tissue Engineering Center, School of Medicine, Johns Hopkins University, Baltimore, MD 21231, USA
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Warren L. Grayson
- Translational Tissue Engineering Center, School of Medicine, Johns Hopkins University, Baltimore, MD 21231, USA
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
- Department of Materials Science & Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Chemical & Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Institute for NanoBioTechnology (INBT), Johns Hopkins University School of Engineering, Baltimore, MD 21218, USA
| |
Collapse
|
6
|
Wang Y, Li X, Li N, Du J, Qin X, Sun X, Wang Y, Li C. Integrated Proteomic and Metabolomic Analysis of Muscle Atrophy Induced by Hindlimb Unloading. Biomolecules 2024; 15:14. [PMID: 39858409 PMCID: PMC11764416 DOI: 10.3390/biom15010014] [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: 11/27/2024] [Revised: 12/17/2024] [Accepted: 12/19/2024] [Indexed: 01/27/2025] Open
Abstract
Skeletal muscle atrophy, which is induced by factors such as disuse, spaceflight, certain medications, neurological disorders, and malnutrition, is a global health issue that lacks effective treatment. Hindlimb unloading is a commonly used model of muscle atrophy. However, the underlying mechanism of muscle atrophy induced by hindlimb unloading remains unclear, particularly from the perspective of the myocyte proteome and metabolism. We first used mass spectrometry for proteomic sequencing and untargeted metabolomics to analyze soleus muscle changes in rats with hindlimb unloading. The study found 1052 proteins and 377 metabolites (with the MS2 name) that were differentially expressed between the hindlimb unloading group and the control group. Proteins like ACTN3, MYH4, MYBPC2, and MYOZ1, typically found in fast-twitch muscles, were upregulated, along with metabolism-related proteins GLUL, GSTM4, and NDUFS4. Metabolites arachidylcarnitine and 7,8-dihydrobiopterin, as well as pathways like histidine, taurine, and hypotaurine metabolism, were linked to muscle atrophy. Protein and metabolism joint analyses revealed that some pathways, such as glutathione metabolism, ferroptosis, and lysosome pathways, were likely to be involved in soleus atrophy. In this study, we have applied integrated deep proteomic and metabolomic analyses. The upregulation of proteins that are expressed in fast-twitch fibers indicates the conversion of slow-twitch fibers to fast-twitch fibers under hindlimb unloading. In addition, some differentially abundant metabolites and pathways revealed the important role of metabolism in muscle atrophy of the soleus. As shown in the graphical abstract, our study provides insights into the pathogenesis and treatment of muscle atrophy that results from unloading by integrating proteomics and metabolomics of the soleus muscles.
Collapse
Affiliation(s)
- Yuan Wang
- Department of Aerospace Medical Training, School of Aerospace Medicine, Fourth Military Medical University, Xi’an 710032, China; (Y.W.); (X.L.); (N.L.); (X.S.)
| | - Xi Li
- Department of Aerospace Medical Training, School of Aerospace Medicine, Fourth Military Medical University, Xi’an 710032, China; (Y.W.); (X.L.); (N.L.); (X.S.)
| | - Na Li
- Department of Aerospace Medical Training, School of Aerospace Medicine, Fourth Military Medical University, Xi’an 710032, China; (Y.W.); (X.L.); (N.L.); (X.S.)
| | - Jiawei Du
- Key Laboratory of Sports and Physical Fitness of the Ministry of Education, Beijing Sport University, Beijing 100084, China;
| | - Xiaodong Qin
- Department of Aerospace Medical Training, School of Aerospace Medicine, Fourth Military Medical University, Xi’an 710032, China; (Y.W.); (X.L.); (N.L.); (X.S.)
| | - Xiqing Sun
- Department of Aerospace Medical Training, School of Aerospace Medicine, Fourth Military Medical University, Xi’an 710032, China; (Y.W.); (X.L.); (N.L.); (X.S.)
| | - Yongchun Wang
- Department of Aerospace Medical Training, School of Aerospace Medicine, Fourth Military Medical University, Xi’an 710032, China; (Y.W.); (X.L.); (N.L.); (X.S.)
| | - Chengfei Li
- Department of Aerospace Medical Training, School of Aerospace Medicine, Fourth Military Medical University, Xi’an 710032, China; (Y.W.); (X.L.); (N.L.); (X.S.)
| |
Collapse
|
7
|
Ly QK, Nguyen MT, Ngo THP, Lee W. Essential Role of Cortactin in Myogenic Differentiation: Regulating Actin Dynamics and Myocardin-Related Transcription Factor A-Serum Response Factor (MRTFA-SRF) Signaling. Int J Mol Sci 2024; 25:13564. [PMID: 39769327 PMCID: PMC11677934 DOI: 10.3390/ijms252413564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 12/17/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025] Open
Abstract
Cortactin (CTTN) is an actin-binding protein regulating actin polymerization and stabilization, which are vital processes for maintaining skeletal muscle homeostasis. Despite the established function of CTTN in actin cytoskeletal dynamics, its role in the myogenic differentiation of progenitor cells remains largely unexplored. In this study, we investigated the role of CTTN in the myogenic differentiation of C2C12 myoblasts by analyzing its effects on actin cytoskeletal remodeling, myocardin-related transcription factor A (MRTFA) nuclear translocation, serum response factor (SRF) activation, expression of myogenic transcription factors, and myotube formation. CTTN expression declined during myogenic differentiation, paralleling the reduction in MyoD, suggesting a potential role in the early stages of myogenesis. We also found that CTTN knockdown in C2C12 myoblasts reduced filamentous actin, enhanced globular actin levels, and inhibited the nuclear translocation of MRTFA, resulting in suppressed SRF activity. This led to the subsequent downregulation of myogenic regulatory factors, such as MyoD and MyoG. Furthermore, CTTN knockdown reduced the nuclear localization of YAP1, a mechanosensitive transcription factor, further supporting its regulatory roles in cell cycle and proliferation. Consequently, CTTN depletion impeded proliferation, differentiation, and myotube formation in C2C12 myoblasts, highlighting its dual role in the coordination of cell cycle regulation and myogenic differentiation of progenitor cells during myogenesis. This study identifies CTTN as an essential regulator of myogenic differentiation via affecting the actin remodeling-MRTFA-SRF signaling axis and cell proliferation.
Collapse
Affiliation(s)
- Quoc Kiet Ly
- Department of Biochemistry, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju 38066, Republic of Korea; (Q.K.L.); (M.T.N.); (T.H.P.N.)
| | - Mai Thi Nguyen
- Department of Biochemistry, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju 38066, Republic of Korea; (Q.K.L.); (M.T.N.); (T.H.P.N.)
| | - Thanh Huu Phan Ngo
- Department of Biochemistry, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju 38066, Republic of Korea; (Q.K.L.); (M.T.N.); (T.H.P.N.)
| | - Wan Lee
- Department of Biochemistry, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju 38066, Republic of Korea; (Q.K.L.); (M.T.N.); (T.H.P.N.)
- Section of Molecular and Cellular Medicine, Medical Institute of Dongguk University, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju 38066, Republic of Korea
- Channelopathy Research Center (CRC), Dongguk University College of Medicine, 32 Dongguk-ro, Ilsan Dong-gu, Goyang 10326, Republic of Korea
| |
Collapse
|
8
|
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
|
9
|
Donati L, Valicenti ML, Giannoni S, Morena F, Martino S. Biomaterials Mimicking Mechanobiology: A Specific Design for a Specific Biological Application. Int J Mol Sci 2024; 25:10386. [PMID: 39408716 PMCID: PMC11476540 DOI: 10.3390/ijms251910386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 10/20/2024] Open
Abstract
Mechanosensing and mechanotransduction pathways between the Extracellular Matrix (ECM) and cells form the essential crosstalk that regulates cell homeostasis, tissue development, morphology, maintenance, and function. Understanding these mechanisms involves creating an appropriate cell support that elicits signals to guide cellular functions. In this context, polymers can serve as ideal molecules for producing biomaterials designed to mimic the characteristics of the ECM, thereby triggering responsive mechanisms that closely resemble those induced by a natural physiological system. The generated specific stimuli depend on the different natural or synthetic origins of the polymers, the chemical composition, the assembly structure, and the physical and surface properties of biomaterials. This review discusses the most widely used polymers and their customization to develop biomaterials with tailored properties. It examines how the characteristics of biomaterials-based polymers can be harnessed to replicate the functions of biological cells, making them suitable for biomedical and biotechnological applications.
Collapse
Affiliation(s)
- Leonardo Donati
- Department of Chemistry, Biology and Biotechnology, Biochemical and Biotechnological Sciences, University of Perugia, 06122 Perugia, Italy
| | - Maria Luisa Valicenti
- Department of Chemistry, Biology and Biotechnology, Biochemical and Biotechnological Sciences, University of Perugia, 06122 Perugia, Italy
| | - Samuele Giannoni
- Department of Chemistry, Biology and Biotechnology, Biochemical and Biotechnological Sciences, University of Perugia, 06122 Perugia, Italy
| | - Francesco Morena
- Department of Chemistry, Biology and Biotechnology, Biochemical and Biotechnological Sciences, University of Perugia, 06122 Perugia, Italy
| | - Sabata Martino
- Department of Chemistry, Biology and Biotechnology, Biochemical and Biotechnological Sciences, University of Perugia, 06122 Perugia, Italy
- Centro di Eccellenza Materiali Innovativi Nanostrutturati per Applicazioni Chimiche Fisiche e Biomediche (CEMIN), University of Perugia, 06123 Perugia, Italy
| |
Collapse
|
10
|
Fowler A, Knaus KR, Khuu S, Khalilimeybodi A, Schenk S, Ward SR, Fry AC, Rangamani P, McCulloch AD. Network model of skeletal muscle cell signalling predicts differential responses to endurance and resistance exercise training. Exp Physiol 2024; 109:939-955. [PMID: 38643471 PMCID: PMC11140181 DOI: 10.1113/ep091712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 03/20/2024] [Indexed: 04/22/2024]
Abstract
Exercise-induced muscle adaptations vary based on exercise modality and intensity. We constructed a signalling network model from 87 published studies of human or rodent skeletal muscle cell responses to endurance or resistance exercise in vivo or simulated exercise in vitro. The network comprises 259 signalling interactions between 120 nodes, representing eight membrane receptors and eight canonical signalling pathways regulating 14 transcriptional regulators, 28 target genes and 12 exercise-induced phenotypes. Using this network, we formulated a logic-based ordinary differential equation model predicting time-dependent molecular and phenotypic alterations following acute endurance and resistance exercises. Compared with nine independent studies, the model accurately predicted 18/21 (85%) acute responses to resistance exercise and 12/16 (75%) acute responses to endurance exercise. Detailed sensitivity analysis of differential phenotypic responses to resistance and endurance training showed that, in the model, exercise regulates cell growth and protein synthesis primarily by signalling via mechanistic target of rapamycin, which is activated by Akt and inhibited in endurance exercise by AMP-activated protein kinase. Endurance exercise preferentially activates inflammation via reactive oxygen species and nuclear factor κB signalling. Furthermore, the expected preferential activation of mitochondrial biogenesis by endurance exercise was counterbalanced in the model by protein kinase C in response to resistance training. This model provides a new tool for investigating cross-talk between skeletal muscle signalling pathways activated by endurance and resistance exercise, and the mechanisms of interactions such as the interference effects of endurance training on resistance exercise outcomes.
Collapse
Affiliation(s)
- Annabelle Fowler
- Department of BioengineeringUniversity of California SanDiegoLa JollaCaliforniaUSA
| | - Katherine R. Knaus
- Department of BioengineeringUniversity of California SanDiegoLa JollaCaliforniaUSA
| | - Stephanie Khuu
- Department of BioengineeringUniversity of California SanDiegoLa JollaCaliforniaUSA
| | - Ali Khalilimeybodi
- Department of Mechanical and Aerospace EngineeringUniversity of California San DiegoLa JollaCaliforniaUSA
| | - Simon Schenk
- Department of Orthopaedic SurgeryUniversity of California San DiegoLa JollaCaliforniaUSA
| | - Samuel R. Ward
- Department of Orthopaedic SurgeryUniversity of California San DiegoLa JollaCaliforniaUSA
| | - Andrew C. Fry
- Department of Health, Sport and Exercise SciencesUniversity of KansasLawrenceKansasUSA
| | - Padmini Rangamani
- Department of Mechanical and Aerospace EngineeringUniversity of California San DiegoLa JollaCaliforniaUSA
| | - Andrew D. McCulloch
- Department of BioengineeringUniversity of California SanDiegoLa JollaCaliforniaUSA
- Department of MedicineUniversity of California San DiegoLa JollaCaliforniaUSA
| |
Collapse
|
11
|
Kim J, Lee H, Lee G, Ryu D, Kim G. Fabrication of fully aligned self-assembled cell-laden collagen filaments for tissue engineering via a hybrid bioprinting process. Bioact Mater 2024; 36:14-29. [PMID: 38425743 PMCID: PMC10900255 DOI: 10.1016/j.bioactmat.2024.02.020] [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: 12/19/2023] [Revised: 02/02/2024] [Accepted: 02/16/2024] [Indexed: 03/02/2024] Open
Abstract
Cell-laden structures play a pivotal role in various tissue engineering applications, particularly in tissue restoration. Interactions between cells within bioprinted structures are crucial for successful tissue development and regulation of stem cell fate through intricate cell-to-cell signaling pathways. In this study, we developed a new technique that combines polyethylene glycol (PEG)-infused submerged bioprinting with a stretching procedure. This approach facilitated the generation of fully aligned collagen structures consisting of myoblasts and a low concentration (2 wt%) of collagen to efficiently encourage muscle tissue regeneration. By adjusting several processing parameters, we obtained biologically safe and mechanically stable cell-laden collagen filaments with uniaxial alignment. Notably, the cell filaments exhibited markedly elevated cellular activities compared to those exhibited by conventional bioprinted filaments, even at similar cell densities. Moreover, when we implanted structures containing adipose stem cells into mice, we observed a significantly increased level of myogenesis compared to that in normally bioprinted struts. Thus, this promising approach has the potential to revolutionize tissue engineering by fostering enhanced cellular interactions and promoting improved outcomes in regenerative medicine.
Collapse
Affiliation(s)
- JuYeon Kim
- Department of Precision Medicine, Sungkyunkwan University School of Medicine (SKKU-SOM), Suwon, 16419, Republic of Korea
| | - Hyeongjin Lee
- Department of Biotechnology and Bioinformatics, Korea University, Sejong, Republic of Korea
| | - Gyudo Lee
- Department of Biotechnology and Bioinformatics, Korea University, Sejong, Republic of Korea
| | - Dongryeol Ryu
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - GeunHyung Kim
- Department of Precision Medicine, Sungkyunkwan University School of Medicine (SKKU-SOM), Suwon, 16419, Republic of Korea
| |
Collapse
|
12
|
Noom A, Sawitzki B, Knaus P, Duda GN. A two-way street - cellular metabolism and myofibroblast contraction. NPJ Regen Med 2024; 9:15. [PMID: 38570493 PMCID: PMC10991391 DOI: 10.1038/s41536-024-00359-x] [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: 07/24/2023] [Accepted: 03/20/2024] [Indexed: 04/05/2024] Open
Abstract
Tissue fibrosis is characterised by the high-energy consumption associated with myofibroblast contraction. Although myofibroblast contraction relies on ATP production, the role of cellular metabolism in myofibroblast contraction has not yet been elucidated. Studies have so far only focused on myofibroblast contraction regulators, such as integrin receptors, TGF-β and their shared transcription factor YAP/TAZ, in a fibroblast-myofibroblast transition setting. Additionally, the influence of the regulators on metabolism and vice versa have been described in this context. However, this has so far not yet been connected to myofibroblast contraction. This review focuses on the known and unknown of how cellular metabolism influences the processes leading to myofibroblast contraction and vice versa. We elucidate the signalling cascades responsible for myofibroblast contraction by looking at FMT regulators, mechanical cues, biochemical signalling, ECM properties and how they can influence and be influenced by cellular metabolism. By reviewing the existing knowledge on the link between cellular metabolism and the regulation of myofibroblast contraction, we aim to pinpoint gaps of knowledge and eventually help identify potential research targets to identify strategies that would allow switching tissue fibrosis towards tissue regeneration.
Collapse
Affiliation(s)
- Anne Noom
- Julius Wolff Institute (JWI), Berlin Institute of Health and Center for Musculoskeletal Surgery at Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany
| | - Birgit Sawitzki
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt University of Berlin, 13353, Berlin, Germany
- Center of Immunomics, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany
| | - Petra Knaus
- Institute of Chemistry and Biochemistry - Biochemistry, Freie Universität Berlin, 14195, Berlin, Germany
| | - Georg N Duda
- Julius Wolff Institute (JWI), Berlin Institute of Health and Center for Musculoskeletal Surgery at Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany.
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany.
| |
Collapse
|
13
|
Sun C, Serra C, Kalicharan BH, Harding J, Rao M. Challenges and Considerations of Preclinical Development for iPSC-Based Myogenic Cell Therapy. Cells 2024; 13:596. [PMID: 38607035 PMCID: PMC11011706 DOI: 10.3390/cells13070596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 03/21/2024] [Accepted: 03/22/2024] [Indexed: 04/13/2024] Open
Abstract
Cell therapies derived from induced pluripotent stem cells (iPSCs) offer a promising avenue in the field of regenerative medicine due to iPSCs' expandability, immune compatibility, and pluripotent potential. An increasing number of preclinical and clinical trials have been carried out, exploring the application of iPSC-based therapies for challenging diseases, such as muscular dystrophies. The unique syncytial nature of skeletal muscle allows stem/progenitor cells to integrate, forming new myonuclei and restoring the expression of genes affected by myopathies. This characteristic makes genome-editing techniques especially attractive in these therapies. With genetic modification and iPSC lineage specification methodologies, immune-compatible healthy iPSC-derived muscle cells can be manufactured to reverse the progression of muscle diseases or facilitate tissue regeneration. Despite this exciting advancement, much of the development of iPSC-based therapies for muscle diseases and tissue regeneration is limited to academic settings, with no successful clinical translation reported. The unknown differentiation process in vivo, potential tumorigenicity, and epigenetic abnormality of transplanted cells are preventing their clinical application. In this review, we give an overview on preclinical development of iPSC-derived myogenic cell transplantation therapies including processes related to iPSC-derived myogenic cells such as differentiation, scaling-up, delivery, and cGMP compliance. And we discuss the potential challenges of each step of clinical translation. Additionally, preclinical model systems for testing myogenic cells intended for clinical applications are described.
Collapse
Affiliation(s)
- Congshan Sun
- Vita Therapeutics, Baltimore, MD 21043, USA (M.R.)
| | - Carlo Serra
- Department of Neurology, Baylor College of Medicine, Houston, TX 77030, USA
| | | | | | - Mahendra Rao
- Vita Therapeutics, Baltimore, MD 21043, USA (M.R.)
| |
Collapse
|
14
|
Helzer D, Kannan P, Reynolds JC, Gibbs DE, Crosbie RH. Role of microenvironment on muscle stem cell function in health, adaptation, and disease. Curr Top Dev Biol 2024; 158:179-201. [PMID: 38670705 DOI: 10.1016/bs.ctdb.2024.02.002] [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: 04/28/2024]
Abstract
The role of the cellular microenvironment has recently gained attention in the context of muscle health, adaption, and disease. Emerging evidence supports major roles for the extracellular matrix (ECM) in regeneration and the dynamic regulation of the satellite cell niche. Satellite cells normally reside in a quiescent state in healthy muscle, but upon muscle injury, they activate, proliferate, and fuse to the damaged fibers to restore muscle function and architecture. This chapter reviews the composition and mechanical properties of skeletal muscle ECM and the role of these factors in contributing to the satellite cell niche that impact muscle regeneration. In addition, the chapter details the effects of satellite cell-matrix interactions and provides evidence that there is bidirectional regulation affecting both the cellular and extracellular microenvironment within skeletal muscle. Lastly, emerging methods to investigate satellite cell-matrix interactions will be presented.
Collapse
Affiliation(s)
- Daniel Helzer
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, United States
| | - Pranav Kannan
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, United States
| | - Joseph C Reynolds
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, United States
| | - Devin E Gibbs
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, United States; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, United States
| | - Rachelle H Crosbie
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, United States; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, United States; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, United States; Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States.
| |
Collapse
|
15
|
dos Santos AEA, Guadalupe JL, Albergaria JDS, Almeida IA, Moreira AMS, Copola AGL, de Araújo IP, de Paula AM, Neves BRA, Santos JPF, da Silva AB, Jorge EC, Andrade LDO. Random cellulose acetate nanofibers: a breakthrough for cultivated meat production. Front Nutr 2024; 10:1297926. [PMID: 38249608 PMCID: PMC10796801 DOI: 10.3389/fnut.2023.1297926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 11/27/2023] [Indexed: 01/23/2024] Open
Abstract
Overcoming the challenge of creating thick, tissue-resembling muscle constructs is paramount in the field of cultivated meat production. This study investigates the remarkable potential of random cellulose acetate nanofibers (CAN) as a transformative scaffold for muscle tissue engineering (MTE), specifically in the context of cultivated meat applications. Through a comparative analysis between random and aligned CAN, utilizing C2C12 and H9c2 myoblasts, we unveil the unparalleled capabilities of random CAN in facilitating muscle differentiation, independent of differentiation media, by exploiting the YAP/TAZ-related mechanotransduction pathway. In addition, we have successfully developed a novel process for stacking cell-loaded CAN sheets, enabling the production of a three-dimensional meat product. C2C12 and H9c2 loaded CAN sheets were stacked (up to four layers) to form a ~300-400 μm thick tissue 2 cm in length, organized in a mesh of uniaxial aligned cells. To further demonstrate the effectiveness of this methodology for cultivated meat purposes, we have generated thick and viable constructs using chicken muscle satellite cells (cSCs) and random CAN. This groundbreaking discovery offers a cost-effective and biomimetic solution for cultivating and differentiating muscle cells, forging a crucial link between tissue engineering and the pursuit of sustainable and affordable cultivated meat production.
Collapse
Affiliation(s)
- Ana Elisa Antunes dos Santos
- Department of Morphology, Institute of Biological Science, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Jorge Luís Guadalupe
- Department of Morphology, Institute of Biological Science, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Juliano Douglas Silva Albergaria
- Laboratory of Biomaterials, Department of Materials Engineering, Federal Center for Technological Education of Minas Gerais (CEFET-MG), Belo Horizonte, Brazil
| | - Itallo Augusto Almeida
- Department of Morphology, Institute of Biological Science, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Amanda Maria Siqueira Moreira
- Department of Morphology, Institute of Biological Science, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Aline Gonçalves Lio Copola
- Laboratory of Biomaterials, Department of Materials Engineering, Federal Center for Technological Education of Minas Gerais (CEFET-MG), Belo Horizonte, Brazil
| | - Isabella Paula de Araújo
- Laboratory of Biomaterials, Department of Materials Engineering, Federal Center for Technological Education of Minas Gerais (CEFET-MG), Belo Horizonte, Brazil
| | - Ana Maria de Paula
- Department of Physics, Institute of Exact Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Bernardo Ruegger Almeida Neves
- Department of Physics, Institute of Exact Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - João Paulo Ferreira Santos
- Laboratory of Biomaterials, Department of Materials Engineering, Federal Center for Technological Education of Minas Gerais (CEFET-MG), Belo Horizonte, Brazil
| | - Aline Bruna da Silva
- Laboratory of Biomaterials, Department of Materials Engineering, Federal Center for Technological Education of Minas Gerais (CEFET-MG), Belo Horizonte, Brazil
| | - Erika Cristina Jorge
- Department of Morphology, Institute of Biological Science, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Luciana de Oliveira Andrade
- Department of Morphology, Institute of Biological Science, Federal University of Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
16
|
Nguyen MT, Ly QK, Kim HJ, Lee W. WAVE2 Is a Vital Regulator in Myogenic Differentiation of Progenitor Cells through the Mechanosensitive MRTFA-SRF Axis. Cells 2023; 13:9. [PMID: 38201213 PMCID: PMC10778525 DOI: 10.3390/cells13010009] [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: 11/17/2023] [Revised: 12/15/2023] [Accepted: 12/19/2023] [Indexed: 01/12/2024] Open
Abstract
Skeletal myogenesis is an intricate process involving the differentiation of progenitor cells into myofibers, which is regulated by actin cytoskeletal dynamics and myogenic transcription factors. Although recent studies have demonstrated the pivotal roles of actin-binding proteins (ABPs) as mechanosensors and signal transducers, the biological significance of WAVE2 (Wiskott-Aldrich syndrome protein family member 2), an ABP essential for actin polymerization, in myogenic differentiation of progenitor cells has not been investigated. Our study provides important insights into the regulatory roles played by WAVE2 in the myocardin-related transcription factor A (MRTFA)-serum response factor (SRF) signaling axis and differentiation of myoblasts. We demonstrate that WAVE2 expression is induced during myogenic differentiation and plays a pivotal role in actin cytoskeletal remodeling in C2C12 myoblasts. Knockdown of WAVE2 in C2C12 cells reduced filamentous actin levels, increased globular actin accumulation, and impaired the nuclear translocation of MRTFA. Furthermore, WAVE2 depletion in myoblasts inhibited the expression and transcriptional activity of SRF and suppressed cell proliferation in myoblasts. Consequently, WAVE2 knockdown suppressed myogenic regulatory factors (i.e., MyoD, MyoG, and SMYD1) expressions, thereby hindering the differentiation of myoblasts. Thus, this study suggests that WAVE2 is essential for myogenic differentiation of progenitor cells by modulating the mechanosensitive MRTFA-SRF axis.
Collapse
Affiliation(s)
- Mai Thi Nguyen
- Department of Biochemistry, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju 38066, Republic of Korea; (M.T.N.); (Q.K.L.); (H.-J.K.)
| | - Quoc Kiet Ly
- Department of Biochemistry, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju 38066, Republic of Korea; (M.T.N.); (Q.K.L.); (H.-J.K.)
| | - Hyun-Jung Kim
- Department of Biochemistry, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju 38066, Republic of Korea; (M.T.N.); (Q.K.L.); (H.-J.K.)
| | - Wan Lee
- Department of Biochemistry, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju 38066, Republic of Korea; (M.T.N.); (Q.K.L.); (H.-J.K.)
- Channelopathy Research Center, Dongguk University College of Medicine, 32 Dongguk-ro, Goyang 10326, Republic of Korea
| |
Collapse
|
17
|
Nguyen MT, Dash R, Jeong K, Lee W. Role of Actin-Binding Proteins in Skeletal Myogenesis. Cells 2023; 12:2523. [PMID: 37947600 PMCID: PMC10650911 DOI: 10.3390/cells12212523] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 10/22/2023] [Accepted: 10/23/2023] [Indexed: 11/12/2023] Open
Abstract
Maintenance of skeletal muscle quantity and quality is essential to ensure various vital functions of the body. Muscle homeostasis is regulated by multiple cytoskeletal proteins and myogenic transcriptional programs responding to endogenous and exogenous signals influencing cell structure and function. Since actin is an essential component in cytoskeleton dynamics, actin-binding proteins (ABPs) have been recognized as crucial players in skeletal muscle health and diseases. Hence, dysregulation of ABPs leads to muscle atrophy characterized by loss of mass, strength, quality, and capacity for regeneration. This comprehensive review summarizes the recent studies that have unveiled the role of ABPs in actin cytoskeletal dynamics, with a particular focus on skeletal myogenesis and diseases. This provides insight into the molecular mechanisms that regulate skeletal myogenesis via ABPs as well as research avenues to identify potential therapeutic targets. Moreover, this review explores the implications of non-coding RNAs (ncRNAs) targeting ABPs in skeletal myogenesis and disorders based on recent achievements in ncRNA research. The studies presented here will enhance our understanding of the functional significance of ABPs and mechanotransduction-derived myogenic regulatory mechanisms. Furthermore, revealing how ncRNAs regulate ABPs will allow diverse therapeutic approaches for skeletal muscle disorders to be developed.
Collapse
Affiliation(s)
- Mai Thi Nguyen
- Department of Biochemistry, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju 38066, Republic of Korea; (M.T.N.); (K.J.)
| | - Raju Dash
- Department of Anatomy, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju 38066, Republic of Korea;
- Department of New Biology, Daegu Gyeongbuk Institute of Science & Technology (DGIST), Daegu 42988, Republic of Korea
| | - Kyuho Jeong
- Department of Biochemistry, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju 38066, Republic of Korea; (M.T.N.); (K.J.)
| | - Wan Lee
- Department of Biochemistry, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju 38066, Republic of Korea; (M.T.N.); (K.J.)
- Channelopathy Research Center, Dongguk University College of Medicine, 32 Dongguk-ro, Ilsan Dong-gu, Goyang 10326, Republic of Korea
| |
Collapse
|
18
|
Nguyen MT, Ly QK, Kim HJ, Lee W. FLII Modulates the Myogenic Differentiation of Progenitor Cells via Actin Remodeling-Mediated YAP1 Regulation. Int J Mol Sci 2023; 24:14335. [PMID: 37762638 PMCID: PMC10531566 DOI: 10.3390/ijms241814335] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 09/16/2023] [Accepted: 09/19/2023] [Indexed: 09/29/2023] Open
Abstract
The dynamic rearrangement of the actin cytoskeleton plays an essential role in myogenesis, which is regulated by diverse mechanisms, such as mechanotransduction, modulation of the Hippo signaling pathway, control of cell proliferation, and the influence of morphological changes. Despite the recognized importance of actin-binding protein Flightless-1 (FLII) during actin remodeling, the role played by FLII in the differentiation of myogenic progenitor cells has not been explored. Here, we investigated the roles of FLII in the proliferation and differentiation of myoblasts. FLII was found to be enriched in C2C12 myoblasts, and its expression was stable during the early stages of differentiation but down-regulated in fully differentiated myotubes. Knockdown of FLII in C2C12 myoblasts resulted in filamentous actin (F-actin) accumulation and inhibited Yes-associated protein 1 (YAP1) phosphorylation, which triggers its nuclear translocation from the cytoplasm. Consequently, the expressions of YAP1 target genes, including PCNA, CCNB1, and CCND1, were induced, and the cell cycle and proliferation of myoblasts were promoted. Moreover, FLII knockdown significantly inhibited the expression of myogenic regulatory factors, i.e., MyoD and MyoG, thereby impairing myoblast differentiation, fusion, and myotube formation. Thus, our findings demonstrate that FLII is crucial for the differentiation of myoblasts via modulation of the F-actin/YAP1 axis and suggest that FLII is a putative novel therapeutic target for muscle wasting.
Collapse
Affiliation(s)
- Mai Thi Nguyen
- Department of Biochemistry, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju 38066, Republic of Korea; (M.T.N.); (Q.K.L.); (H.-J.K.)
| | - Quoc Kiet Ly
- Department of Biochemistry, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju 38066, Republic of Korea; (M.T.N.); (Q.K.L.); (H.-J.K.)
| | - Hyun-Jung Kim
- Department of Biochemistry, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju 38066, Republic of Korea; (M.T.N.); (Q.K.L.); (H.-J.K.)
| | - Wan Lee
- Department of Biochemistry, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju 38066, Republic of Korea; (M.T.N.); (Q.K.L.); (H.-J.K.)
- Channelopathy Research Center, Dongguk University College of Medicine, 32 Dongguk-ro, Goyang 10326, Republic of Korea
| |
Collapse
|
19
|
Koushki N, Ghagre A, Srivastava LK, Molter C, Ehrlicher AJ. Nuclear compression regulates YAP spatiotemporal fluctuations in living cells. Proc Natl Acad Sci U S A 2023; 120:e2301285120. [PMID: 37399392 PMCID: PMC10334804 DOI: 10.1073/pnas.2301285120] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 06/04/2023] [Indexed: 07/05/2023] Open
Abstract
Yes-associated protein (YAP) is a key mechanotransduction protein in diverse physiological and pathological processes; however, a ubiquitous YAP activity regulatory mechanism in living cells has remained elusive. Here, we show that YAP nuclear translocation is highly dynamic during cell movement and is driven by nuclear compression arising from cell contractile work. We resolve the mechanistic role of cytoskeletal contractility in nuclear compression by manipulation of nuclear mechanics. Disrupting the linker of nucleoskeleton and cytoskeleton complex reduces nuclear compression for a given contractility and correspondingly decreases YAP localization. Conversely, decreasing nuclear stiffness via silencing of lamin A/C increases nuclear compression and YAP nuclear localization. Finally, using osmotic pressure, we demonstrated that nuclear compression even without active myosin or filamentous actin regulates YAP localization. The relationship between nuclear compression and YAP localization captures a universal mechanism for YAP regulation with broad implications in health and biology.
Collapse
Affiliation(s)
- Newsha Koushki
- Department of Bioengineering, McGill University, Montreal, QCH3A 0E9, Canada
| | - Ajinkya Ghagre
- Department of Bioengineering, McGill University, Montreal, QCH3A 0E9, Canada
| | | | - Clayton Molter
- Department of Bioengineering, McGill University, Montreal, QCH3A 0E9, Canada
| | - Allen J. Ehrlicher
- Department of Bioengineering, McGill University, Montreal, QCH3A 0E9, Canada
- Department of Anatomy and Cell Biology, McGill University, Montreal, QCH3A 0C7, Canada
- Department of Biomedical Engineering, McGill University, Montreal, QCH3A 2B4, Canada
- Department of Mechanical Engineering, McGill University, Montreal, QCH3A 0C3, Canada
- Centre for Structural Biology, McGill University, Montreal, QCH3G 0B1, Canada
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QCH3A 1A3, Canada
| |
Collapse
|
20
|
Furrer R, Hawley JA, Handschin C. The molecular athlete: exercise physiology from mechanisms to medals. Physiol Rev 2023; 103:1693-1787. [PMID: 36603158 PMCID: PMC10110736 DOI: 10.1152/physrev.00017.2022] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 12/12/2022] [Accepted: 12/19/2022] [Indexed: 01/07/2023] Open
Abstract
Human skeletal muscle demonstrates remarkable plasticity, adapting to numerous external stimuli including the habitual level of contractile loading. Accordingly, muscle function and exercise capacity encompass a broad spectrum, from inactive individuals with low levels of endurance and strength to elite athletes who produce prodigious performances underpinned by pleiotropic training-induced muscular adaptations. Our current understanding of the signal integration, interpretation, and output coordination of the cellular and molecular mechanisms that govern muscle plasticity across this continuum is incomplete. As such, training methods and their application to elite athletes largely rely on a "trial-and-error" approach, with the experience and practices of successful coaches and athletes often providing the bases for "post hoc" scientific enquiry and research. This review provides a synopsis of the morphological and functional changes along with the molecular mechanisms underlying exercise adaptation to endurance- and resistance-based training. These traits are placed in the context of innate genetic and interindividual differences in exercise capacity and performance, with special consideration given to aging athletes. Collectively, we provide a comprehensive overview of skeletal muscle plasticity in response to different modes of exercise and how such adaptations translate from "molecules to medals."
Collapse
Affiliation(s)
| | - John A Hawley
- Exercise and Nutrition Research Program, Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne, Victoria, Australia
| | | |
Collapse
|
21
|
Nguyen MT, Lee W. Induction of miR-665-3p Impairs the Differentiation of Myogenic Progenitor Cells by Regulating the TWF1-YAP1 Axis. Cells 2023; 12:cells12081114. [PMID: 37190023 DOI: 10.3390/cells12081114] [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: 03/10/2023] [Revised: 04/03/2023] [Accepted: 04/06/2023] [Indexed: 05/17/2023] Open
Abstract
Actin dynamics are known to orchestrate various myogenic processes in progenitor cells. Twinfilin-1 (TWF1) is an actin-depolymerizing factor that plays a crucial role in the differentiation of myogenic progenitor cells. However, little is known about the mechanisms underlying the epigenetic regulation of TWF1 expression and impaired myogenic differentiation in the background of muscle wasting. This study investigated how miR-665-3p affects TWF1 expression, actin filaments' modulation, proliferation, and myogenic differentiation in progenitor cells. Palmitic acid, the most prevalent saturated fatty acid (SFA) in food, suppressed TWF1 expression and inhibited the myogenic differentiation of C2C12 cells while increasing the level of miR-665-3p expression. Interestingly, miR-665-3p inhibited TWF1 expression by targeting TWF1 3'UTR directly. In addition, miR-665-3p accumulated filamentous actin (F-actin) and enhanced the nuclear translocation of Yes-associated protein 1 (YAP1), consequently promoting cell cycle progression and proliferation. Furthermore, miR-665-3p suppressed the expressions of myogenic factors, i.e., MyoD, MyoG, and MyHC, and consequently impaired myoblast differentiation. In conclusion, this study suggests that SFA-inducible miR-665-3p suppresses TWF1 expression epigenetically and inhibits myogenic differentiation by facilitating myoblast proliferation via the F-actin/YAP1 axis.
Collapse
Affiliation(s)
- Mai Thi Nguyen
- Department of Biochemistry, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju 38066, Republic of Korea
| | - Wan Lee
- Department of Biochemistry, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju 38066, Republic of Korea
- Channelopathy Research Center, Dongguk University College of Medicine, 32 Dongguk-ro, Ilsan Dong-gu, Goyang 10326, Republic of Korea
| |
Collapse
|
22
|
Nguyen MT, Lee W. Mir-302a/TWF1 Axis Impairs the Myogenic Differentiation of Progenitor Cells through F-Actin-Mediated YAP1 Activation. Int J Mol Sci 2023; 24:ijms24076341. [PMID: 37047312 PMCID: PMC10094299 DOI: 10.3390/ijms24076341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 03/17/2023] [Accepted: 03/21/2023] [Indexed: 03/30/2023] Open
Abstract
Actin cytoskeleton dynamics have been found to regulate myogenesis in various progenitor cells, and twinfilin-1 (TWF1), an actin-depolymerizing factor, plays a vital role in actin dynamics and myoblast differentiation. Nevertheless, the molecular mechanisms underlying the epigenetic regulation and biological significance of TWF1 in obesity and muscle wasting have not been explored. Here, we investigated the roles of miR-302a in TWF1 expression, actin filament modulation, proliferation, and myogenic differentiation in C2C12 progenitor cells. Palmitic acid, the most prevalent saturated fatty acid (SFA) in the diet, decreased the expression of TWF1 and impeded myogenic differentiation while increasing the miR-302a levels in C2C12 myoblasts. Interestingly, miR-302a inhibited TWF1 expression directly by targeting its 3′UTR. Furthermore, ectopic expression of miR-302a promoted cell cycle progression and proliferation by increasing the filamentous actin (F-actin) accumulation, which facilitated the nuclear translocation of Yes-associated protein 1 (YAP1). Consequently, by suppressing the expressions of myogenic factors, i.e., MyoD, MyoG, and MyHC, miR-302a impaired myoblast differentiation. Hence, this study demonstrated that SFA-inducible miR-302a suppresses TWF1 expression epigenetically and impairs myogenic differentiation by facilitating myoblast proliferation via F-actin-mediated YAP1 activation.
Collapse
Affiliation(s)
- Mai Thi Nguyen
- Department of Biochemistry, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju 38066, Republic of Korea
| | - Wan Lee
- Department of Biochemistry, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju 38066, Republic of Korea
- Channelopathy Research Center, Dongguk University College of Medicine, 32 Dongguk-ro, Ilsan Dong-gu, Goyang 10326, Republic of Korea
- Correspondence: ; Tel.: +82-54-770-2409
| |
Collapse
|
23
|
Gallardo FS, Córdova-Casanova A, Bock-Pereda A, Rebolledo DL, Ravasio A, Casar JC, Brandan E. Denervation Drives YAP/TAZ Activation in Muscular Fibro/Adipogenic Progenitors. Int J Mol Sci 2023; 24:ijms24065585. [PMID: 36982659 PMCID: PMC10059792 DOI: 10.3390/ijms24065585] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 02/24/2023] [Accepted: 02/25/2023] [Indexed: 03/17/2023] Open
Abstract
Loss of motoneuron innervation (denervation) is a hallmark of neurodegeneration and aging of the skeletal muscle. Denervation induces fibrosis, a response attributed to the activation and expansion of resident fibro/adipogenic progenitors (FAPs), i.e., multipotent stromal cells with myofibroblast potential. Using in vivo and in silico approaches, we revealed FAPs as a novel cell population that activates the transcriptional coregulators YAP/TAZ in response to skeletal muscle denervation. Here, we found that denervation induces the expression and transcriptional activity of YAP/TAZ in whole muscle lysates. Using the PdgfraH2B:EGFP/+ transgenic reporter mice to trace FAPs, we demonstrated that denervation leads to increased YAP expression that accumulates within FAPs nuclei. Consistently, re-analysis of published single-nucleus RNA sequencing (snRNA-seq) data indicates that FAPs from denervated muscles have a higher YAP/TAZ signature level than control FAPs. Thus, our work provides the foundations to address the functional role of YAP/TAZ in FAPs in a neurogenic pathological context, which could be applied to develop novel therapeutic approaches for the treatment of muscle disorders triggered by motoneuron degeneration.
Collapse
Affiliation(s)
- Felipe S. Gallardo
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile
- Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Santiago 7750000, Chile
| | - Adriana Córdova-Casanova
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile
- Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Santiago 7750000, Chile
| | - Alexia Bock-Pereda
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile
- Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Santiago 7750000, Chile
| | - Daniela L. Rebolledo
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas 6213515, Chile
| | - Andrea Ravasio
- Institute for Biological and Medical Engineering, School of Engineering, Medicine and Biological Sciences, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile
| | - Juan Carlos Casar
- Departamento de Neurología, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile
| | - Enrique Brandan
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile
- Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Santiago 7750000, Chile
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago 7510602, Chile
- Correspondence:
| |
Collapse
|
24
|
Battey E, Ross JA, Hoang A, Wilson DGS, Han Y, Levy Y, Pollock RD, Kalakoutis M, Pugh JN, Close GL, Ellison-Hughes GM, Lazarus NR, Iskratsch T, Harridge SDR, Ochala J, Stroud MJ. Myonuclear alterations associated with exercise are independent of age in humans. J Physiol 2023. [PMID: 36597809 DOI: 10.1113/jp284128] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 12/19/2022] [Indexed: 01/05/2023] Open
Abstract
Age-related decline in skeletal muscle structure and function can be mitigated by regular exercise. However, the precise mechanisms that govern this are not fully understood. The nucleus plays an active role in translating forces into biochemical signals (mechanotransduction), with the nuclear lamina protein lamin A regulating nuclear shape, nuclear mechanics and ultimately gene expression. Defective lamin A expression causes muscle pathologies and premature ageing syndromes, but the roles of nuclear structure and function in physiological ageing and in exercise adaptations remain obscure. Here, we isolated single muscle fibres and carried out detailed morphological and functional analyses on myonuclei from young and older exercise-trained individuals. Strikingly, myonuclei from trained individuals were more spherical, less deformable, and contained a thicker nuclear lamina than those from untrained individuals. Complementary to this, exercise resulted in increased levels of lamin A and increased myonuclear stiffness in mice. We conclude that exercise is associated with myonuclear remodelling, independently of age, which may contribute to the preservative effects of exercise on muscle function throughout the lifespan. KEY POINTS: The nucleus plays an active role in translating forces into biochemical signals. Myonuclear aberrations in a group of muscular dystrophies called laminopathies suggest that the shape and mechanical properties of myonuclei are important for maintaining muscle function. Here, striking differences are presented in myonuclear shape and mechanics associated with exercise, in both young and old humans. Myonuclei from trained individuals were more spherical, less deformable and contained a thicker nuclear lamina than untrained individuals. It is concluded that exercise is associated with age-independent myonuclear remodelling, which may help to maintain muscle function throughout the lifespan.
Collapse
Affiliation(s)
- E Battey
- Centre for Human & Applied Physiological Sciences, Faculty of Life Sciences & Medicine, King's College London, London, UK
- British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, King's College London, London, UK
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - J A Ross
- British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, King's College London, London, UK
| | - A Hoang
- British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, King's College London, London, UK
| | - D G S Wilson
- School of Engineering and Materials Science, Queen Mary University of London, London, UK
| | - Y Han
- Centre for Human & Applied Physiological Sciences, Faculty of Life Sciences & Medicine, King's College London, London, UK
| | - Y Levy
- Centre for Human & Applied Physiological Sciences, Faculty of Life Sciences & Medicine, King's College London, London, UK
| | - R D Pollock
- Centre for Human & Applied Physiological Sciences, Faculty of Life Sciences & Medicine, King's College London, London, UK
| | - M Kalakoutis
- Centre for Human & Applied Physiological Sciences, Faculty of Life Sciences & Medicine, King's College London, London, UK
- Randall Centre for Cell and Molecular Biophysics, Faculty of Life Sciences & Medicine, King's College London, London, UK
| | - J N Pugh
- School of Sport and Exercise Sciences, Liverpool John Moores University, Liverpool, UK
| | - G L Close
- School of Sport and Exercise Sciences, Liverpool John Moores University, Liverpool, UK
| | - G M Ellison-Hughes
- Centre for Human & Applied Physiological Sciences, Faculty of Life Sciences & Medicine, King's College London, London, UK
| | - N R Lazarus
- Centre for Human & Applied Physiological Sciences, Faculty of Life Sciences & Medicine, King's College London, London, UK
| | - T Iskratsch
- School of Engineering and Materials Science, Queen Mary University of London, London, UK
- Randall Centre for Cell and Molecular Biophysics, Faculty of Life Sciences & Medicine, King's College London, London, UK
| | - S D R Harridge
- Centre for Human & Applied Physiological Sciences, Faculty of Life Sciences & Medicine, King's College London, London, UK
| | - J Ochala
- Centre for Human & Applied Physiological Sciences, Faculty of Life Sciences & Medicine, King's College London, London, UK
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - M J Stroud
- British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, King's College London, London, UK
| |
Collapse
|
25
|
Campbell MD, Martín-Pérez M, Egertson JD, Gaffrey MJ, Wang L, Bammler T, Rabinovitch PS, MacCoss M, Qian WJ, Villen J, Marcinek D. Elamipretide effects on the skeletal muscle phosphoproteome in aged female mice. GeroScience 2022; 44:2913-2924. [PMID: 36322234 PMCID: PMC9768078 DOI: 10.1007/s11357-022-00679-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 10/20/2022] [Indexed: 12/24/2022] Open
Abstract
The age-related decline in skeletal muscle mass and function is known as sarcopenia. Sarcopenia progresses based on complex processes involving protein dynamics, cell signaling, oxidative stress, and repair. We have previously found that 8-week treatment with elamipretide improves skeletal muscle function, reverses redox stress, and restores protein S-glutathionylation changes in aged female mice. This study tested whether 8-week treatment with elamipretide also affects global phosphorylation in skeletal muscle consistent with functional improvements and S-glutathionylation. Using female 6-7-month-old mice and 28-29-month-old mice, we found that phosphorylation changes did not relate to S-glutathionylation modifications, but that treatment with elamipretide did partially reverse age-related changes in protein phosphorylation in mouse skeletal muscle.
Collapse
Affiliation(s)
- Matthew D Campbell
- Department of Radiology, University of Washington, South Lake Union Campus, 850 Republican St., Brotman D142, Box 358050, Seattle, WA, 98109, USA
| | | | - Jarrett D Egertson
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Matthew J Gaffrey
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Lu Wang
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, USA
| | - Theo Bammler
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, USA
| | - Peter S Rabinovitch
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Michael MacCoss
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Wei-Jun Qian
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Judit Villen
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - David Marcinek
- Department of Radiology, University of Washington, South Lake Union Campus, 850 Republican St., Brotman D142, Box 358050, Seattle, WA, 98109, USA.
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA.
| |
Collapse
|
26
|
Peyton MP, Yang TY, Higgins L, Markowski TW, Vue C, Parker LL, Lowe DA. Global phosphoproteomic profiling of skeletal muscle in ovarian hormone-deficient mice. Physiol Genomics 2022; 54:417-432. [PMID: 36062884 PMCID: PMC9639773 DOI: 10.1152/physiolgenomics.00104.2022] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/05/2022] [Accepted: 08/30/2022] [Indexed: 11/22/2022] Open
Abstract
Protein phosphorylation is important in skeletal muscle development, growth, regeneration, and contractile function. Alterations in the skeletal muscle phosphoproteome due to aging have been reported in males; however, studies in females are lacking. We have demonstrated that estrogen deficiency decreases muscle force, which correlates with decreased myosin regulatory light chain phosphorylation. Thus, we questioned whether the decline of estrogen in females that occurs with aging might alter the skeletal muscle phosphoproteome. C57BL/6J female mice (6 mo) were randomly assigned to a sham-operated (Sham) or ovariectomy (Ovx) group to investigate the effects of estrogen deficiency on skeletal muscle protein phosphorylation in a resting, noncontracting condition. After 16 wk of estrogen deficiency, the tibialis anterior muscle was dissected and prepped for label-free nano-liquid chromatography-tandem mass spectrometry phosphoproteomic analysis. We identified 4,780 phosphopeptides in tibialis anterior muscles of ovariectomized (Ovx) and Sham-operated (Sham) control mice. Further analysis revealed 647 differentially regulated phosphopeptides (Benjamini-Hochberg adjusted P value < 0.05 and 1.5-fold change ratio) that corresponded to 130 proteins with 22 proteins differentially phosphorylated (3 unique to Ovx, 2 unique to Sham, 6 upregulated, and 11 downregulated). Differentially phosphorylated proteins associated with the sarcomere, cytoplasm, and metabolic and calcium signaling pathways were identified. Our work provides the first global phosphoproteomic analysis in females and how estrogen deficiency impacts the skeletal muscle phosphoproteome.
Collapse
Affiliation(s)
- Mina P Peyton
- Division of Rehabilitation Science, Department of Rehabilitation Medicine, University of Minnesota, Twin Cities, Minneapolis, Minnesota
- Department of Computer Science, Bioinformatics and Computational Biology Program, University of Minnesota, Minneapolis, Minnesota
| | - Tzu-Yi Yang
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Twin Cities, Minneapolis, Minnesota
| | - LeeAnn Higgins
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Twin Cities, Minneapolis, Minnesota
| | - Todd W Markowski
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Twin Cities, Minneapolis, Minnesota
| | - Cha Vue
- Division of Rehabilitation Science, Department of Rehabilitation Medicine, University of Minnesota, Twin Cities, Minneapolis, Minnesota
| | - Laurie L Parker
- Department of Computer Science, Bioinformatics and Computational Biology Program, University of Minnesota, Minneapolis, Minnesota
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Twin Cities, Minneapolis, Minnesota
| | - Dawn A Lowe
- Division of Rehabilitation Science, Department of Rehabilitation Medicine, University of Minnesota, Twin Cities, Minneapolis, Minnesota
- Division of Physical Therapy, Department of Rehabilitation Medicine, University of Minnesota, Twin Cities, Minneapolis, Minnesota
| |
Collapse
|
27
|
Kalukula Y, Stephens AD, Lammerding J, Gabriele S. Mechanics and functional consequences of nuclear deformations. Nat Rev Mol Cell Biol 2022; 23:583-602. [PMID: 35513718 PMCID: PMC9902167 DOI: 10.1038/s41580-022-00480-z] [Citation(s) in RCA: 183] [Impact Index Per Article: 61.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/29/2022] [Indexed: 02/08/2023]
Abstract
As the home of cellular genetic information, the nucleus has a critical role in determining cell fate and function in response to various signals and stimuli. In addition to biochemical inputs, the nucleus is constantly exposed to intrinsic and extrinsic mechanical forces that trigger dynamic changes in nuclear structure and morphology. Emerging data suggest that the physical deformation of the nucleus modulates many cellular and nuclear functions. These functions have long been considered to be downstream of cytoplasmic signalling pathways and dictated by gene expression. In this Review, we discuss an emerging perspective on the mechanoregulation of the nucleus that considers the physical connections from chromatin to nuclear lamina and cytoskeletal filaments as a single mechanical unit. We describe key mechanisms of nuclear deformations in time and space and provide a critical review of the structural and functional adaptive responses of the nucleus to deformations. We then consider the contribution of nuclear deformations to the regulation of important cellular functions, including muscle contraction, cell migration and human disease pathogenesis. Collectively, these emerging insights shed new light on the dynamics of nuclear deformations and their roles in cellular mechanobiology.
Collapse
Affiliation(s)
- Yohalie Kalukula
- University of Mons, Soft Matter and Biomaterials group, Interfaces and Complex Fluids Laboratory, Research Institute for Biosciences, CIRMAP, Place du Parc, 20 B-7000 Mons, Belgium
| | - Andrew D. Stephens
- Biology Department, University of Massachusetts Amherst, Amherst, MA, USA
| | - Jan Lammerding
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853, USA,Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Sylvain Gabriele
- University of Mons, Soft Matter and Biomaterials group, Interfaces and Complex Fluids Laboratory, Research Institute for Biosciences, CIRMAP, Place du Parc, 20 B-7000 Mons, Belgium
| |
Collapse
|
28
|
Shen Y, Jing D, Zhao Z. The effect of AKT in extracellular matrix stiffness induced osteogenic differentiation of hBMSCs. Cell Signal 2022; 99:110404. [PMID: 35835331 DOI: 10.1016/j.cellsig.2022.110404] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 07/02/2022] [Accepted: 07/08/2022] [Indexed: 02/05/2023]
Abstract
Extracellular matrix (ECM) stiffness is an important biophysical factor in human bone marrow mesenchymal stem cells (hBMSCs) differentiation. Although there is evidence that Yes-associated protein (YAP) plays an important role in ECM elasticity induced osteogenesis, but the regulatory mechanism and signaling pathways have not been distinctly uncovered. In this study, hBMSCs were cultured on collagen-coated polydimethylsiloxane hydrogels with stiffness corresponding to Young's moduli of 0.5 kPa and 32 kPa, and gene chip analyses revealed the phosphoinositide 3-kinase (PI3K)-AKT pathway was highly correlated with ECM stiffness. Following western blots indicated that AKT phosphorylation was evidently affected in 5th-7th days after ECM stiffness stimulation, while PI3K showed little difference. The AKT activator SC79 and inhibitor MK2206 were utilized to modulate AKT phosphorylation. SC79 and MK2206 caused alteration in the mRNA expression and protein level of alkaline phosphatase (ALP), collagen type I alpha 1 (COL1A1) and runt related transcription factor 2 (RUNX2). On 32 kPa substrates, YAP enrichment in nucleus were significantly promoted by SC79 and remarkably decreased by MK2206. Besides, the ratio of YAP/p-YAP is upregulated by SC79 on both 32 kPa and 0.5 kPa substrates. In conclusion, these findings suggest that AKT is involved in the modulation of ECM stiffness induced osteogenesis, and AKT phosphorylation also influences the subcellular localization and activation of YAP.
Collapse
Affiliation(s)
- Yu Shen
- School of Basic Medical Sciences, Chengdu University, Chengdu 610106, China
| | - Dian Jing
- Department of Orthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, China.
| | - Zhihe Zhao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
29
|
Nguyen MT, Lee W. Kank1 Is Essential for Myogenic Differentiation by Regulating Actin Remodeling and Cell Proliferation in C2C12 Progenitor Cells. Cells 2022; 11:cells11132030. [PMID: 35805114 PMCID: PMC9265739 DOI: 10.3390/cells11132030] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 06/24/2022] [Accepted: 06/25/2022] [Indexed: 02/05/2023] Open
Abstract
Actin cytoskeleton dynamics are essential regulatory processes in muscle development, growth, and regeneration due to their modulation of mechanotransduction, cell proliferation, differentiation, and morphological changes. Although the KN motif and ankyrin repeat domain-containing protein 1 (Kank1) plays a significant role in cell adhesion dynamics, actin polymerization, and cell proliferation in various cells, the functional significance of Kank1 during the myogenic differentiation of progenitor cells has not been explored. Here, we report that Kank1 acts as a critical regulator of the proliferation and differentiation of muscle progenitor cells. Kank1 was found to be expressed at a relatively high level in C2C12 myoblasts, and its expression was modulated during the differentiation. Depletion of Kank1 by siRNA (siKank1) increased the accumulation of filamentous actin (F-actin). Furthermore, it facilitated the nuclear localization of Yes-associated protein 1 (YAP1) by diminishing YAP1 phosphorylation in the cytoplasm, which activated the transcriptions of YAP1 target genes and promoted proliferation and cell cycle progression in myoblasts. Notably, depletion of Kank1 suppressed the protein expression of myogenic regulatory factors (i.e., MyoD and MyoG) and dramatically inhibited myoblast differentiation and myotube formation. Our results show that Kank1 is an essential regulator of actin dynamics, YAP1 activation, and cell proliferation and that its depletion impairs the myogenic differentiation of progenitor cells by promoting myoblast proliferation triggered by the F-actin-induced nuclear translocation of YAP1.
Collapse
Affiliation(s)
- Mai Thi Nguyen
- Department of Biochemistry, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju 38066, Korea;
| | - Wan Lee
- Department of Biochemistry, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju 38066, Korea;
- Channelopathy Research Center, Dongguk University College of Medicine, 32 Dongguk-ro, Ilsan Dong-gu, Gyeonggi-do, Goyang 10326, Korea
- Correspondence: ; Tel.: +82-54-770-2409
| |
Collapse
|
30
|
Abstract
Skeletal muscle is a mechanical organ that not only produces force but also uses mechanical stimuli as a signal to regulate cellular responses. Duchenne and Becker muscular dystrophy are lethal muscle wasting diseases that affect 1 in 3,500 boys and is caused by the absence or malfunction of dystrophin protein, respectively. There is a lack of understanding on how the integration of these mechanical signals is dysregulated in muscular dystrophy and how they may contribute to disease progression. In this study, we show that patient-relevant dystrophin mutations alter the mechanical signaling axis in muscle cells, leading to impaired migration. This work proposes dystrophin as a component of the cellular force-sensing machinery, furthering our knowledge in the pathomechanism of muscular dystrophy. Dystrophin is an essential muscle protein that contributes to cell membrane stability by mechanically linking the actin cytoskeleton to the extracellular matrix via an adhesion complex called the dystrophin–glycoprotein complex. The absence or impaired function of dystrophin causes muscular dystrophy. Focal adhesions (FAs) are also mechanosensitive adhesion complexes that connect the cytoskeleton to the extracellular matrix. However, the interplay between dystrophin and FA force transmission has not been investigated. Using a vinculin-based bioluminescent tension sensor, we measured FA tension in transgenic C2C12 myoblasts expressing wild-type (WT) dystrophin, a nonpathogenic single nucleotide polymorphism (SNP) (I232M), or two missense mutations associated with Duchenne (L54R), or Becker muscular dystrophy (L172H). Our data revealed cross talk between dystrophin and FAs, as the expression of WT or I232M dystrophin increased FA tension compared to dystrophin-less nontransgenic myoblasts. In contrast, the expression of L54R or L172H did not increase FA tension, indicating that these disease-causing mutations compromise the mechanical function of dystrophin as an FA allosteric regulator. Decreased FA tension caused by these mutations manifests as defective migration, as well as decreased Yes-associated protein 1 (YAP) activation, possibly by the disruption of the ability of FAs to transmit forces between the extracellular matrix and cytoskeleton. Our results indicate that dystrophin influences FA tension and suggest that dystrophin disease-causing missense mutations may disrupt a cellular tension-sensing pathway in dystrophic skeletal muscle.
Collapse
|
31
|
Vanmunster M, Rojo Garcia AV, Pacolet A, Dalle S, Koppo K, Jonkers I, Lories R, Suhr F. Mechanosensors control skeletal muscle mass, molecular clocks, and metabolism. Cell Mol Life Sci 2022; 79:321. [PMID: 35622133 PMCID: PMC11072145 DOI: 10.1007/s00018-022-04346-7] [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: 02/11/2022] [Revised: 04/12/2022] [Accepted: 05/03/2022] [Indexed: 11/03/2022]
Abstract
BACKGROUND Skeletal muscles (SkM) are mechanosensitive, with mechanical unloading resulting in muscle-devastating conditions and altered metabolic properties. However, it remains unexplored whether these atrophic conditions affect SkM mechanosensors and molecular clocks, both crucial for their homeostasis and consequent physiological metabolism. METHODS We induced SkM atrophy through 14 days of hindlimb suspension (HS) in 10 male C57BL/6J mice and 10 controls (CTR). SkM histology, gene expressions and protein levels of mechanosensors, molecular clocks and metabolism-related players were examined in the m. Gastrocnemius and m. Soleus. Furthermore, we genetically reduced the expression of mechanosensors integrin-linked kinase (Ilk1) and kindlin-2 (Fermt2) in myogenic C2C12 cells and analyzed the gene expression of mechanosensors, clock components and metabolism-controlling genes. RESULTS Upon hindlimb suspension, gene expression levels of both core molecular clocks and mechanosensors were moderately upregulated in m. Gastrocnemius but strongly downregulated in m. Soleus. Upon unloading, metabolism- and protein biosynthesis-related genes were moderately upregulated in m. Gastrocnemius but downregulated in m. Soleus. Furthermore, we identified very strong correlations between mechanosensors, metabolism- and circadian clock-regulating genes. Finally, genetically induced downregulations of mechanosensors Ilk1 and Fermt2 caused a downregulated mechanosensor, molecular clock and metabolism-related gene expression in the C2C12 model. CONCLUSIONS Collectively, these data shed new lights on mechanisms that control muscle loss. Mechanosensors are identified to crucially control these processes, specifically through commanding molecular clock components and metabolism.
Collapse
Affiliation(s)
- Mathias Vanmunster
- Department of Movement Sciences, Exercise Physiology Research Group, KU Leuven, 3001, Leuven, Belgium
| | - Ana Victoria Rojo Garcia
- Department of Movement Sciences, Exercise Physiology Research Group, KU Leuven, 3001, Leuven, Belgium
| | - Alexander Pacolet
- Department of Movement Sciences, Exercise Physiology Research Group, KU Leuven, 3001, Leuven, Belgium
| | - Sebastiaan Dalle
- Department of Movement Sciences, Exercise Physiology Research Group, KU Leuven, 3001, Leuven, Belgium
| | - Katrien Koppo
- Department of Movement Sciences, Exercise Physiology Research Group, KU Leuven, 3001, Leuven, Belgium
| | - Ilse Jonkers
- Department of Movement Sciences, Human Movement Biomechanics Research Group, KU Leuven, 3001, Leuven, Belgium
| | - Rik Lories
- Department of Development and Regeneration, Skeletal Biology and Engineering Research Center, KU Leuven, 3000, Leuven, Belgium
| | - Frank Suhr
- Department of Movement Sciences, Exercise Physiology Research Group, KU Leuven, 3001, Leuven, Belgium.
| |
Collapse
|
32
|
Nguyen MT, Won YH, Kwon TW, Lee W. Twinfilin-1 is an essential regulator of myogenic differentiation through the modulation of YAP in C2C12 myoblasts. Biochem Biophys Res Commun 2022; 599:17-23. [DOI: 10.1016/j.bbrc.2022.02.021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 02/07/2022] [Indexed: 12/28/2022]
|
33
|
Cao H, Duan L, Zhang Y, Cao J, Zhang K. Current hydrogel advances in physicochemical and biological response-driven biomedical application diversity. Signal Transduct Target Ther 2021; 6:426. [PMID: 34916490 PMCID: PMC8674418 DOI: 10.1038/s41392-021-00830-x] [Citation(s) in RCA: 410] [Impact Index Per Article: 102.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 11/10/2021] [Accepted: 11/11/2021] [Indexed: 02/05/2023] Open
Abstract
Hydrogel is a type of versatile platform with various biomedical applications after rational structure and functional design that leverages on material engineering to modulate its physicochemical properties (e.g., stiffness, pore size, viscoelasticity, microarchitecture, degradability, ligand presentation, stimulus-responsive properties, etc.) and influence cell signaling cascades and fate. In the past few decades, a plethora of pioneering studies have been implemented to explore the cell-hydrogel matrix interactions and figure out the underlying mechanisms, paving the way to the lab-to-clinic translation of hydrogel-based therapies. In this review, we first introduced the physicochemical properties of hydrogels and their fabrication approaches concisely. Subsequently, the comprehensive description and deep discussion were elucidated, wherein the influences of different hydrogels properties on cell behaviors and cellular signaling events were highlighted. These behaviors or events included integrin clustering, focal adhesion (FA) complex accumulation and activation, cytoskeleton rearrangement, protein cyto-nuclei shuttling and activation (e.g., Yes-associated protein (YAP), catenin, etc.), cellular compartment reorganization, gene expression, and further cell biology modulation (e.g., spreading, migration, proliferation, lineage commitment, etc.). Based on them, current in vitro and in vivo hydrogel applications that mainly covered diseases models, various cell delivery protocols for tissue regeneration and disease therapy, smart drug carrier, bioimaging, biosensor, and conductive wearable/implantable biodevices, etc. were further summarized and discussed. More significantly, the clinical translation potential and trials of hydrogels were presented, accompanied with which the remaining challenges and future perspectives in this field were emphasized. Collectively, the comprehensive and deep insights in this review will shed light on the design principles of new biomedical hydrogels to understand and modulate cellular processes, which are available for providing significant indications for future hydrogel design and serving for a broad range of biomedical applications.
Collapse
Affiliation(s)
- Huan Cao
- Department of Nuclear Medicine, West China Hospital, and National Engineering Research Center for Biomaterials, Sichuan University, 610064, Chengdu, P. R. China
- Department of Medical Ultrasound and Central Laboratory, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, 200072, Shanghai, People's Republic of China
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore
| | - Lixia Duan
- Department of Medical Ultrasound and Central Laboratory, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, 200072, Shanghai, People's Republic of China
| | - Yan Zhang
- Department of Medical Ultrasound and Central Laboratory, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, 200072, Shanghai, People's Republic of China
| | - Jun Cao
- Department of Nuclear Medicine, West China Hospital, and National Engineering Research Center for Biomaterials, Sichuan University, 610064, Chengdu, P. R. China.
| | - Kun Zhang
- Department of Medical Ultrasound and Central Laboratory, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, 200072, Shanghai, People's Republic of China.
| |
Collapse
|
34
|
Leser JM, Harriot A, Buck HV, Ward CW, Stains JP. Aging, Osteo-Sarcopenia, and Musculoskeletal Mechano-Transduction. FRONTIERS IN REHABILITATION SCIENCES 2021; 2:782848. [PMID: 36004321 PMCID: PMC9396756 DOI: 10.3389/fresc.2021.782848] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 11/10/2021] [Indexed: 11/13/2022]
Abstract
The decline in the mass and function of bone and muscle is an inevitable consequence of healthy aging with early onset and accelerated decline in those with chronic disease. Termed osteo-sarcopenia, this condition predisposes the decreased activity, falls, low-energy fractures, and increased risk of co-morbid disease that leads to musculoskeletal frailty. The biology of osteo-sarcopenia is most understood in the context of systemic neuro-endocrine and immune/inflammatory alterations that drive inflammation, oxidative stress, reduced autophagy, and cellular senescence in the bone and muscle. Here we integrate these concepts to our growing understanding of how bone and muscle senses, responds and adapts to mechanical load. We propose that age-related alterations in cytoskeletal mechanics alter load-sensing and mechano-transduction in bone osteocytes and muscle fibers which underscores osteo-sarcopenia. Lastly, we examine the evidence for exercise as an effective countermeasure to osteo-sarcopenia.
Collapse
Affiliation(s)
| | | | | | | | - Joseph P. Stains
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
35
|
Liu Z, Lin L, Zhu H, Wu Z, Ding X, Hu R, Jiang Y, Tang C, Ding S, Guo R. YAP Promotes Cell Proliferation and Stemness Maintenance of Porcine Muscle Stem Cells under High-Density Condition. Cells 2021; 10:cells10113069. [PMID: 34831292 PMCID: PMC8621012 DOI: 10.3390/cells10113069] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/29/2021] [Accepted: 11/03/2021] [Indexed: 12/16/2022] Open
Abstract
Muscle stem cells (MuSCs) isolated ex vivo are essential original cells to produce cultured meat. Currently, one of the main obstacles for cultured meat production derives from the limited capacity of large-scale amplification of MuSCs, especially under high-density culture condition. Here, we show that at higher cell densities, proliferation and differentiation capacities of porcine MuSCs are impaired. We investigate the roles of Hippo-YAP signaling, which is important regulators in response to cell contact inhibition. Interestingly, abundant but not functional YAP proteins are accumulated in MuSCs seeded at high density. When treated with lysophosphatidic acid (LPA), the activator of YAP, porcine MuSCs exhibit increased proliferation and elevated differentiation potential compared with control cells. Moreover, constitutively active YAP with deactivated phosphorylation sites, but not intact YAP, promotes cell proliferation and stemness maintenance of MuSCs. Together, we reveal a potential molecular target that enables massive MuSCs expansion for large-scale cultured meat production under high-density condition.
Collapse
Affiliation(s)
- Zheng Liu
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (Z.L.); (L.L.); (H.Z.); (Z.W.); (X.D.); (R.H.); (Y.J.); (C.T.)
- National Center of Meat Quality and Safety Control, Key Laboratory of Meat Processing and Quality Control, Key Laboratory of Meat Processing, Nanjing Agricultural University, Nanjing 210095, China
| | - Ling Lin
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (Z.L.); (L.L.); (H.Z.); (Z.W.); (X.D.); (R.H.); (Y.J.); (C.T.)
- National Center of Meat Quality and Safety Control, Key Laboratory of Meat Processing and Quality Control, Key Laboratory of Meat Processing, Nanjing Agricultural University, Nanjing 210095, China
| | - Haozhe Zhu
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (Z.L.); (L.L.); (H.Z.); (Z.W.); (X.D.); (R.H.); (Y.J.); (C.T.)
- National Center of Meat Quality and Safety Control, Key Laboratory of Meat Processing and Quality Control, Key Laboratory of Meat Processing, Nanjing Agricultural University, Nanjing 210095, China
| | - Zhongyuan Wu
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (Z.L.); (L.L.); (H.Z.); (Z.W.); (X.D.); (R.H.); (Y.J.); (C.T.)
- National Center of Meat Quality and Safety Control, Key Laboratory of Meat Processing and Quality Control, Key Laboratory of Meat Processing, Nanjing Agricultural University, Nanjing 210095, China
| | - Xi Ding
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (Z.L.); (L.L.); (H.Z.); (Z.W.); (X.D.); (R.H.); (Y.J.); (C.T.)
- National Center of Meat Quality and Safety Control, Key Laboratory of Meat Processing and Quality Control, Key Laboratory of Meat Processing, Nanjing Agricultural University, Nanjing 210095, China
| | - Rongrong Hu
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (Z.L.); (L.L.); (H.Z.); (Z.W.); (X.D.); (R.H.); (Y.J.); (C.T.)
- National Center of Meat Quality and Safety Control, Key Laboratory of Meat Processing and Quality Control, Key Laboratory of Meat Processing, Nanjing Agricultural University, Nanjing 210095, China
| | - Yichen Jiang
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (Z.L.); (L.L.); (H.Z.); (Z.W.); (X.D.); (R.H.); (Y.J.); (C.T.)
- National Center of Meat Quality and Safety Control, Key Laboratory of Meat Processing and Quality Control, Key Laboratory of Meat Processing, Nanjing Agricultural University, Nanjing 210095, China
| | - Changbo Tang
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (Z.L.); (L.L.); (H.Z.); (Z.W.); (X.D.); (R.H.); (Y.J.); (C.T.)
- National Center of Meat Quality and Safety Control, Key Laboratory of Meat Processing and Quality Control, Key Laboratory of Meat Processing, Nanjing Agricultural University, Nanjing 210095, China
| | - Shijie Ding
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (Z.L.); (L.L.); (H.Z.); (Z.W.); (X.D.); (R.H.); (Y.J.); (C.T.)
- National Center of Meat Quality and Safety Control, Key Laboratory of Meat Processing and Quality Control, Key Laboratory of Meat Processing, Nanjing Agricultural University, Nanjing 210095, China
- Correspondence: (S.D.); (R.G.)
| | - Renpeng Guo
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (Z.L.); (L.L.); (H.Z.); (Z.W.); (X.D.); (R.H.); (Y.J.); (C.T.)
- Correspondence: (S.D.); (R.G.)
| |
Collapse
|
36
|
Günay KA, Silver JS, Chang TL, Bednarski OJ, Bannister KL, Rogowski CJ, Olwin BB, Anseth KS. Myoblast mechanotransduction and myotube morphology is dependent on BAG3 regulation of YAP and TAZ. Biomaterials 2021; 277:121097. [PMID: 34481290 DOI: 10.1016/j.biomaterials.2021.121097] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 07/26/2021] [Accepted: 08/24/2021] [Indexed: 11/30/2022]
Abstract
Skeletal muscle tissue is mechanically dynamic with changes in stiffness influencing function, maintenance, and regeneration. We modeled skeletal muscle mechanical changes in culture with dynamically stiffening hydrogels demonstrating that the chaperone protein BAG3 transduces matrix stiffness by redistributing YAP and TAZ subcellular localization in muscle progenitor cells. BAG3 depletion increases cytoplasmic retention of YAP and TAZ, desensitizing myoblasts to changes in hydrogel elastic moduli. Upon differentiation, muscle progenitors depleted of BAG3 formed enlarged, round myotubes lacking the typical cylindrical morphology. The aberrant morphology is dependent on YAP/TAZ signaling, which was sequestered in the cytoplasm in BAG3-depleted myotubes but predominately nuclear in cylindrical myotubes of control cells. Control progenitor cells induced to differentiate on soft (E' = 4 and 12 kPa) hydrogels formed circular myotubes similar to those observed in BAG3-depleted cells. Inhibition of the Hippo pathway partially restored myotube morphologies, permitting nuclear translocation of YAP and TAZ in BAG3-depleted myogenic progenitors. Thus, BAG3 is a critical mediator of dynamic stiffness changes in muscle tissue, coupling mechanical alterations to intracellular signals and inducing changes in gene expression that influence muscle progenitor cell morphology and differentiation.
Collapse
Affiliation(s)
- K Arda Günay
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, CO, USA; BioFrontiers Institute, University of Colorado, Boulder, CO, USA
| | - Jason S Silver
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, CO, USA; BioFrontiers Institute, University of Colorado, Boulder, CO, USA; Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO, USA; Medical Scientist Training Program, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Tze-Ling Chang
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, CO, USA; BioFrontiers Institute, University of Colorado, Boulder, CO, USA
| | - Olivia J Bednarski
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, CO, USA; BioFrontiers Institute, University of Colorado, Boulder, CO, USA
| | - Kendra L Bannister
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, CO, USA; BioFrontiers Institute, University of Colorado, Boulder, CO, USA
| | - Cameron J Rogowski
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, CO, USA; BioFrontiers Institute, University of Colorado, Boulder, CO, USA
| | - Bradley B Olwin
- BioFrontiers Institute, University of Colorado, Boulder, CO, USA; Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO, USA.
| | - Kristi S Anseth
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, CO, USA; BioFrontiers Institute, University of Colorado, Boulder, CO, USA.
| |
Collapse
|
37
|
Zhang X, Wang D, Mak KLK, Tuan RS, Ker DFE. Engineering Musculoskeletal Grafts for Multi-Tissue Unit Repair: Lessons From Developmental Biology and Wound Healing. Front Physiol 2021; 12:691954. [PMID: 34504435 PMCID: PMC8421786 DOI: 10.3389/fphys.2021.691954] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 07/22/2021] [Indexed: 12/13/2022] Open
Abstract
In the musculoskeletal system, bone, tendon, and skeletal muscle integrate and act coordinately as a single multi-tissue unit to facilitate body movement. The development, integration, and maturation of these essential components and their response to injury are vital for conferring efficient locomotion. The highly integrated nature of these components is evident under disease conditions, where rotator cuff tears at the bone-tendon interface have been reported to be associated with distal pathological alterations such as skeletal muscle degeneration and bone loss. To successfully treat musculoskeletal injuries and diseases, it is important to gain deep understanding of the development, integration and maturation of these musculoskeletal tissues along with their interfaces as well as the impact of inflammation on musculoskeletal healing and graft integration. This review highlights the current knowledge of developmental biology and wound healing in the bone-tendon-muscle multi-tissue unit and perspectives of what can be learnt from these biological and pathological processes within the context of musculoskeletal tissue engineering and regenerative medicine. Integrating these knowledge and perspectives can serve as guiding principles to inform the development and engineering of musculoskeletal grafts and other tissue engineering strategies to address challenging musculoskeletal injuries and diseases.
Collapse
Affiliation(s)
- Xu Zhang
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, China
- Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, China
| | - Dan Wang
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, China
- Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, China
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, China
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, China
| | - King-Lun Kingston Mak
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health-Guangdong Laboratory), Guangzhou, China
| | - Rocky S. Tuan
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, China
- Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, China
| | - Dai Fei Elmer Ker
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, China
- Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, China
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, China
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, China
| |
Collapse
|
38
|
Whitely ME, Collins PB, Iwamoto M, Wenke JC. Administration of a selective retinoic acid receptor-γ agonist improves neuromuscular strength in a rodent model of volumetric muscle loss. J Exp Orthop 2021; 8:58. [PMID: 34383202 PMCID: PMC8360252 DOI: 10.1186/s40634-021-00378-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 07/30/2021] [Indexed: 12/24/2022] Open
Abstract
PURPOSE Volumetric muscle loss is a uniquely challenging pathology that results in irrecoverable functional deficits. Furthermore, a breakthrough drug or bioactive factor has yet to be established that adequately improves repair of these severe skeletal muscle injuries. This study sought to assess the ability of an orally administered selective retinoic acid receptor-γ agonist, palovarotene, to improve recovery of neuromuscular strength in a rat model of volumetric muscle loss. METHODS An irrecoverable, full thickness defect was created in the tibialis anterior muscle of Lewis rats and animals were survived for 4 weeks. Functional recovery of the tibialis anterior muscle was assessed in vivo via neural stimulation and determination of peak isometric torque. Histological staining was performed to qualitatively assess fibrous scarring of the defect site. RESULTS Treatment with the selective retinoic acid receptor-γ agonist, palovarotene, resulted in a 38% improvement of peak isometric torque in volumetric muscle loss affected limbs after 4 weeks of healing compared to untreated controls. Additionally, preliminary histological assessment suggests that oral administration of palovarotene reduced fibrous scarring at the defect site. CONCLUSIONS These results highlight the potential role of selective retinoic acid receptor-γ agonists in the design of regenerative medicine platforms to maximize skeletal muscle healing. Additional studies are needed to further elucidate cellular responses, optimize therapeutic delivery, and characterize synergistic potential with adjunct therapies.
Collapse
Affiliation(s)
- Michael E. Whitely
- Orthopaedic Trauma Department, United States Army Institute of Surgical Research, 3698 Chambers Pass, Building 3611, JBSA Fort Sam Houston, San Antonio, TX 78234 USA
| | - Patrick B. Collins
- Orthopaedic Trauma Department, United States Army Institute of Surgical Research, 3698 Chambers Pass, Building 3611, JBSA Fort Sam Houston, San Antonio, TX 78234 USA
| | - Masahiro Iwamoto
- Department of Orthopaedics, University of Maryland School of Medicine, 655 W Baltimore St, Baltimore, MD 21201 USA
| | - Joseph C. Wenke
- Orthopaedic Trauma Department, United States Army Institute of Surgical Research, 3698 Chambers Pass, Building 3611, JBSA Fort Sam Houston, San Antonio, TX 78234 USA
| |
Collapse
|
39
|
Chiang MY, Lo YC, Lai YH, Yong YYA, Chang SJ, Chen WL, Chen SY. Protein-based soft actuator with high photo-response and easy modulation for anisotropic cell alignment and proliferation in a liquid environment. J Mater Chem B 2021; 9:6634-6645. [PMID: 34365493 DOI: 10.1039/d1tb01198g] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Cell alignment and elongation, which are critical factors correlated with differentiation and maturation in cell biology and tissue engineering, have been widely studied in organisms. Several strategies such as external mechanical strain, geometric topography, micropatterning approaches, and microfabricated substrates have been developed to guide cell alignment, but these methodologies cannot be used for easily denatured natural proteins to modulate the cell behaviour. Herein, for the first time, a novel biocompatible light-controlled protein-based bilayer soft actuator composed of elastin-like polypeptides (ELPs), silk fibroin (SF), graphene oxide (GO), and reduced graphene oxide (rGO), named ESGRG, is developed for efficiently driving cellular orientation and elongation with anisotropic features on soft actuator via remote NIR laser exposure. The actuation of ESGRG could be manipulated by modulating the intensity of NIR and the relative ratio of GO to rGO for promoting myoblasts alignment and nucleus elongation to generate different motions. The results indicate that the YAP and MHC protein expression of C2C12 skeletal muscle cells on ESGRG can be rapidly induced and enhanced by controlling the relative ratio of rGO/GO = 1/4 at a multiple-cycle stimulation with a very low power intensity of 1.2 W cm-2 in friendly liquid environments. This study demonstrates that the ESGRG hydrogel actuator system can modulate the cell-level behaviors via light-driven cyclic bending-motions and can be utilized in applications of soft robotic and tissue engineering such as artificial muscle and maturation of cardiomyocytes.
Collapse
Affiliation(s)
- Min-Yu Chiang
- Department of Materials Science and Engineering, National Yang Ming Chiao Tung University, No. 1001 Ta-Hsueh Rd, Hsinchu, Taiwan 300, Republic of China.
| | | | | | | | | | | | | |
Collapse
|
40
|
YAP1 nuclear efflux and transcriptional reprograming follow membrane diminution upon VSV-G-induced cell fusion. Nat Commun 2021; 12:4502. [PMID: 34301937 PMCID: PMC8302681 DOI: 10.1038/s41467-021-24708-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 06/24/2021] [Indexed: 02/07/2023] Open
Abstract
Cells in many tissues, such as bone, muscle, and placenta, fuse into syncytia to acquire new functions and transcriptional programs. While it is known that fused cells are specialized, it is unclear whether cell-fusion itself contributes to programmatic-changes that generate the new cellular state. Here, we address this by employing a fusogen-mediated, cell-fusion system to create syncytia from undifferentiated cells. RNA-Seq analysis reveals VSV-G-induced cell fusion precedes transcriptional changes. To gain mechanistic insights, we measure the plasma membrane surface area after cell-fusion and observe it diminishes through increases in endocytosis. Consequently, glucose transporters internalize, and cytoplasmic glucose and ATP transiently decrease. This reduced energetic state activates AMPK, which inhibits YAP1, causing transcriptional-reprogramming and cell-cycle arrest. Impairing either endocytosis or AMPK activity prevents YAP1 inhibition and cell-cycle arrest after fusion. Together, these data demonstrate plasma membrane diminishment upon cell-fusion causes transient nutrient stress that may promote transcriptional-reprogramming independent from extrinsic cues.
Collapse
|
41
|
Kwon H, Kim J, Jho EH. Role of the Hippo pathway and mechanisms for controlling cellular localization of YAP/TAZ. FEBS J 2021; 289:5798-5818. [PMID: 34173335 DOI: 10.1111/febs.16091] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 06/05/2021] [Accepted: 06/24/2021] [Indexed: 12/26/2022]
Abstract
The Hippo pathway is a crucial signaling mechanism that inhibits the growth of cells and organs during development and in disease. When the Hippo pathway is activated, YAP/TAZ transcriptional coactivators are phosphorylated by upstream kinases, preventing nuclear localization of YAP/TAZ. However, when the Hippo pathway is inhibited, YAP/TAZ localize mainly in the nucleus and induce the expression of target genes related to cell proliferation. Abnormal proliferation of cells is one of the hallmarks of cancer initiation, and activation of Hippo pathway dampens such cell proliferation. Various types of diseases including cancer can occur due to the dysregulation of the Hippo pathway. Therefore, a better understanding of the Hippo pathway signaling mechanisms, and in particular how YAP/TAZ exist in the nucleus, may lead to the identification of new therapeutic targets for treating cancer and other diseases. In this review, we summarize the overall Hippo pathway and discuss mechanisms related to nuclear localization of YAP/TAZ.
Collapse
Affiliation(s)
- Hyeryun Kwon
- Department of Life Science, University of Seoul, Korea
| | - Jiyoung Kim
- Department of Life Science, University of Seoul, Korea
| | - Eek-Hoon Jho
- Department of Life Science, University of Seoul, Korea
| |
Collapse
|
42
|
Boso D, Carraro E, Maghin E, Todros S, Dedja A, Giomo M, Elvassore N, De Coppi P, Pavan PG, Piccoli M. Porcine Decellularized Diaphragm Hydrogel: A New Option for Skeletal Muscle Malformations. Biomedicines 2021; 9:biomedicines9070709. [PMID: 34206569 PMCID: PMC8301461 DOI: 10.3390/biomedicines9070709] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 06/17/2021] [Accepted: 06/18/2021] [Indexed: 12/14/2022] Open
Abstract
Hydrogels are biomaterials that, thanks to their unique hydrophilic and biomimetic characteristics, are used to support cell growth and attachment and promote tissue regeneration. The use of decellularized extracellular matrix (dECM) from different tissues or organs significantly demonstrated to be far superior to other types of hydrogel since it recapitulates the native tissue’s ECM composition and bioactivity. Different muscle injuries and malformations require the application of patches or fillers to replenish the defect and boost tissue regeneration. Herein, we develop, produce, and characterize a porcine diaphragmatic dECM-derived hydrogel for diaphragmatic applications. We obtain a tissue-specific biomaterial able to mimic the complex structure of skeletal muscle ECM; we characterize hydrogel properties in terms of biomechanical properties, biocompatibility, and adaptability for in vivo applications. Lastly, we demonstrate that dECM-derived hydrogel obtained from porcine diaphragms can represent a useful biological product for diaphragmatic muscle defect repair when used as relevant acellular stand-alone patch.
Collapse
Affiliation(s)
- Daniele Boso
- Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Corso Stati Uniti 4, 35127 Padova, Italy; (D.B.); (E.C.); (E.M.); (P.G.P.)
- Department of Industrial Engineering, University of Padova, Via Venezia 1, 35131 Padova, Italy;
| | - Eugenia Carraro
- Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Corso Stati Uniti 4, 35127 Padova, Italy; (D.B.); (E.C.); (E.M.); (P.G.P.)
- Department of Biomedical Sciences, University of Padova, Via Ugo Bassi 58/B, 35131 Padova, Italy
| | - Edoardo Maghin
- Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Corso Stati Uniti 4, 35127 Padova, Italy; (D.B.); (E.C.); (E.M.); (P.G.P.)
- Department of Industrial Engineering, University of Padova, Via Venezia 1, 35131 Padova, Italy;
| | - Silvia Todros
- Department of Industrial Engineering, University of Padova, Via Venezia 1, 35131 Padova, Italy;
| | - Arben Dedja
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Via Giustiniani 2, 35128 Padova, Italy;
| | - Monica Giomo
- Department of Industrial Engineering, University of Padova, Via Marzolo 9, 35131 Padova, Italy; (M.G.); (N.E.)
| | - Nicola Elvassore
- Department of Industrial Engineering, University of Padova, Via Marzolo 9, 35131 Padova, Italy; (M.G.); (N.E.)
- Veneto Institute of Molecular Medicine, Via G. Orus 2, 35127 Padova, Italy
- Shanghai Institute for Advanced Immunochemical Studies (SIAIS), ShanghaiTech University, Y Building, No. 393 Middle Huaxia Road, Pudong, Shanghai 201210, China
- NIHR Biomedical Research Center, Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK;
| | - Paolo De Coppi
- NIHR Biomedical Research Center, Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK;
- Specialist Neonatal and Pediatric Surgery, Great Ormond Street Hospital, London WC1N 3JH, UK
| | - Piero Giovanni Pavan
- Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Corso Stati Uniti 4, 35127 Padova, Italy; (D.B.); (E.C.); (E.M.); (P.G.P.)
- Department of Industrial Engineering, University of Padova, Via Venezia 1, 35131 Padova, Italy;
| | - Martina Piccoli
- Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Corso Stati Uniti 4, 35127 Padova, Italy; (D.B.); (E.C.); (E.M.); (P.G.P.)
- Correspondence:
| |
Collapse
|
43
|
Shi N, Li Y, Chang L, Zhao G, Jin G, Lyu Y, Genin GM, Ma Y, Xu F. A 3D, Magnetically Actuated, Aligned Collagen Fiber Hydrogel Platform Recapitulates Physical Microenvironment of Myoblasts for Enhancing Myogenesis. SMALL METHODS 2021; 5:e2100276. [PMID: 34927916 DOI: 10.1002/smtd.202100276] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 04/23/2021] [Indexed: 06/14/2023]
Abstract
Many cell responses that underlie the development, maturation, and function of tissues are guided by the architecture and mechanical loading of the extracellular matrix (ECM). Because mechanical stimulation must be transmitted through the ECM architecture, the synergy between these two factors is important. However, recapitulating the synergy of these physical microenvironmental cues in vitro remains challenging. To address this, a 3D magnetically actuated collagen hydrogel platform is developed that enables combined control of ECM architecture and mechanical stimulation. With this platform, it is demonstrated how these factors synergistically promote cell alignment of C2C12 myoblasts and enhance myogenesis. This promotion is driven in part by the dynamics of Yes-associated protein and structure of cellular microtubule networks. This facile platform holds great promises for regulating cell behavior and fate, generating a broad range of engineered physiologically representative microtissues in vitro, and quantifying the mechanobiology underlying their functions.
Collapse
Affiliation(s)
- Nianyuan Shi
- Bioinspired Engineering and Biomechanics Center (BEBC), The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Yuhui Li
- Bioinspired Engineering and Biomechanics Center (BEBC), The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Le Chang
- Bioinspired Engineering and Biomechanics Center (BEBC), The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Guoxu Zhao
- School of Material Science and Chemical Engineering, Xi'an Technological University, Xi'an, 710021, China
| | - Guorui Jin
- Bioinspired Engineering and Biomechanics Center (BEBC), The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Yi Lyu
- Department of Hepatobiliary Surgery and Institute of Advanced Surgical Technology and Engineering, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Guy M Genin
- Bioinspired Engineering and Biomechanics Center (BEBC), The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Xi'an Jiaotong University, Xi'an, 710049, China
- NSF Science and Technology Center for Engineering MechanoBiology, Washington University in St. Louis, St. Louis, MO, 63130, USA
| | - Yufei Ma
- Bioinspired Engineering and Biomechanics Center (BEBC), The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Feng Xu
- Bioinspired Engineering and Biomechanics Center (BEBC), The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Xi'an Jiaotong University, Xi'an, 710049, China
| |
Collapse
|
44
|
Rebolledo DL, Acuña MJ, Brandan E. Role of Matricellular CCN Proteins in Skeletal Muscle: Focus on CCN2/CTGF and Its Regulation by Vasoactive Peptides. Int J Mol Sci 2021; 22:5234. [PMID: 34063397 PMCID: PMC8156781 DOI: 10.3390/ijms22105234] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 04/28/2021] [Accepted: 05/12/2021] [Indexed: 02/08/2023] Open
Abstract
The Cellular Communication Network (CCN) family of matricellular proteins comprises six proteins that share conserved structural features and play numerous biological roles. These proteins can interact with several receptors or soluble proteins, regulating cell signaling pathways in various tissues under physiological and pathological conditions. In the skeletal muscle of mammals, most of the six CCN family members are expressed during embryonic development or in adulthood. Their roles during the adult stage are related to the regulation of muscle mass and regeneration, maintaining vascularization, and the modulation of skeletal muscle fibrosis. This work reviews the CCNs proteins' role in skeletal muscle physiology and disease, focusing on skeletal muscle fibrosis and its regulation by Connective Tissue Growth factor (CCN2/CTGF). Furthermore, we review evidence on the modulation of fibrosis and CCN2/CTGF by the renin-angiotensin system and the kallikrein-kinin system of vasoactive peptides.
Collapse
Affiliation(s)
- Daniela L. Rebolledo
- Centro de Envejecimiento y Regeneración, CARE Chile UC, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile;
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas 6213515, Chile
| | - María José Acuña
- Centro de Envejecimiento y Regeneración, CARE Chile UC, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile;
- Centro Integrativo de Biología y Química Aplicada (CIBQA), Universidad Bernardo O Higgins, Santiago 8370854, Chile
| | - Enrique Brandan
- Centro de Envejecimiento y Regeneración, CARE Chile UC, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile;
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
- Fundación Ciencia & Vida, Santiago 7810000, Chile
| |
Collapse
|
45
|
Millward DJ. Interactions between Growth of Muscle and Stature: Mechanisms Involved and Their Nutritional Sensitivity to Dietary Protein: The Protein-Stat Revisited. Nutrients 2021; 13:729. [PMID: 33668846 PMCID: PMC7996181 DOI: 10.3390/nu13030729] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 02/15/2021] [Accepted: 02/22/2021] [Indexed: 02/07/2023] Open
Abstract
Childhood growth and its sensitivity to dietary protein is reviewed within a Protein-Stat model of growth regulation. The coordination of growth of muscle and stature is a combination of genetic programming, and of two-way mechanical interactions involving the mechanotransduction of muscle growth through stretching by bone length growth, the core Protein-Stat feature, and the strengthening of bone through muscle contraction via the mechanostat. Thus, growth in bone length is the initiating event and this is always observed. Endocrine and cellular mechanisms of growth in stature are reviewed in terms of the growth hormone-insulin like growth factor-1 (GH-IGF-1) and thyroid axes and the sex hormones, which together mediate endochondral ossification in the growth plate and bone lengthening. Cellular mechanisms of muscle growth during development are then reviewed identifying (a) the difficulties posed by the need to maintain its ultrastructure during myofibre hypertrophy within the extracellular matrix and the concept of muscle as concentric "bags" allowing growth to be conceived as bag enlargement and filling, (b) the cellular and molecular mechanisms involved in the mechanotransduction of satellite and mesenchymal stromal cells, to enable both connective tissue remodelling and provision of new myonuclei to aid myofibre hypertrophy and (c) the implications of myofibre hypertrophy for protein turnover within the myonuclear domain. Experimental data from rodent and avian animal models illustrate likely changes in DNA domain size and protein turnover during developmental and stretch-induced muscle growth and between different muscle fibre types. Growth of muscle in male rats during adulthood suggests that "bag enlargement" is achieved mainly through the action of mesenchymal stromal cells. Current understanding of the nutritional regulation of protein deposition in muscle, deriving from experimental studies in animals and human adults, is reviewed, identifying regulation by amino acids, insulin and myofibre volume changes acting to increase both ribosomal capacity and efficiency of muscle protein synthesis via the mechanistic target of rapamycin complex 1 (mTORC1) and the phenomenon of a "bag-full" inhibitory signal has been identified in human skeletal muscle. The final section deals with the nutritional sensitivity of growth of muscle and stature to dietary protein in children. Growth in length/height as a function of dietary protein intake is described in the context of the breastfed child as the normative growth model, and the "Early Protein Hypothesis" linking high protein intakes in infancy to later adiposity. The extensive paediatric studies on serum IGF-1 and child growth are reviewed but their clinical relevance is of limited value for understanding growth regulation; a role in energy metabolism and homeostasis, acting with insulin to mediate adiposity, is probably more important. Information on the influence of dietary protein on muscle mass per se as opposed to lean body mass is limited but suggests that increased protein intake in children is unable to promote muscle growth in excess of that linked to genotypic growth in length/height. One possible exception is milk protein intake, which cohort and cross-cultural studies suggest can increase height and associated muscle growth, although such effects have yet to be demonstrated by randomised controlled trials.
Collapse
Affiliation(s)
- D Joe Millward
- Department of Nutritional Sciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XH, UK
| |
Collapse
|
46
|
Connon CJ, Gouveia RM. Milliscale Substrate Curvature Promotes Myoblast Self-Organization and Differentiation. Adv Biol (Weinh) 2021; 5:e2000280. [PMID: 33852180 DOI: 10.1002/adbi.202000280] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 01/23/2021] [Indexed: 11/06/2022]
Abstract
Biological tissues comprise complex structural environments known to influence cell behavior via multiple interdependent sensing and transduction mechanisms. Yet, and despite the predominantly nonplanar geometry of these environments, the impact of tissue-size (milliscale) curvature on cell behavior is largely overlooked or underestimated. This study explores how concave, hemicylinder-shaped surfaces 3-50 mm in diameter affect the migration, proliferation, orientation, and differentiation of C2C12 myoblasts. Notably, these milliscale cues significantly affect cell responses compared with planar substrates, with myoblasts grown on surfaces 7.5-15 mm in diameter showing prevalent migration and alignment parallel to the curvature axis. Moreover, surfaces within this curvature range promote myoblast differentiation and the formation of denser, more compact tissues comprising highly oriented multinucleated myotubes. Based on the similarity of effects, it is further proposed that myoblast susceptibility to substrate curvature depends on mechanotransduction signaling. This model thus supports the notion that cellular responses to substrate curvature and compliance share the same molecular pathways and that control of cell behavior can be achieved via modulation of either individual parameter or in combination. This correlation is relevant for elucidating how muscle tissue forms and heals, as well as for designing better biomaterials and more appropriate cell-surface interfaces.
Collapse
Affiliation(s)
- Che J Connon
- Tissue Engineering Lab Biosciences Institute, Newcastle University, International Centre for Life, Newcastle upon Tyne, NE1 3BZ, UK
| | - Ricardo M Gouveia
- Biosciences Institute, Newcastle University, International Centre for Life, Newcastle upon Tyne, NE1 3BZ, UK
| |
Collapse
|
47
|
Jabre S, Hleihel W, Coirault C. Nuclear Mechanotransduction in Skeletal Muscle. Cells 2021; 10:cells10020318. [PMID: 33557157 PMCID: PMC7913907 DOI: 10.3390/cells10020318] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 01/29/2021] [Accepted: 02/01/2021] [Indexed: 12/11/2022] Open
Abstract
Skeletal muscle is composed of multinucleated, mature muscle cells (myofibers) responsible for contraction, and a resident pool of mononucleated muscle cell precursors (MCPs), that are maintained in a quiescent state in homeostatic conditions. Skeletal muscle is remarkable in its ability to adapt to mechanical constraints, a property referred as muscle plasticity and mediated by both MCPs and myofibers. An emerging body of literature supports the notion that muscle plasticity is critically dependent upon nuclear mechanotransduction, which is transduction of exterior physical forces into the nucleus to generate a biological response. Mechanical loading induces nuclear deformation, changes in the nuclear lamina organization, chromatin condensation state, and cell signaling, which ultimately impacts myogenic cell fate decisions. This review summarizes contemporary insights into the mechanisms underlying nuclear force transmission in MCPs and myofibers. We discuss how the cytoskeleton and nuclear reorganizations during myogenic differentiation may affect force transmission and nuclear mechanotransduction. We also discuss how to apply these findings in the context of muscular disorders. Finally, we highlight current gaps in knowledge and opportunities for further research in the field.
Collapse
Affiliation(s)
- Saline Jabre
- Sorbonne Université, INSERM UMRS-974 and Institut de Myologie, 75013 Paris, France;
- Department of Biology, Faculty of Arts and Sciences, Holy Spirit University of Kasik (USEK), Jounieh 446, Lebanon;
| | - Walid Hleihel
- Department of Biology, Faculty of Arts and Sciences, Holy Spirit University of Kasik (USEK), Jounieh 446, Lebanon;
- Department of Basic Health Sciences, Faculty of Medicine, Holy Spirit University of Kaslik (USEK), Jounieh 446, Lebanon
| | - Catherine Coirault
- Sorbonne Université, INSERM UMRS-974 and Institut de Myologie, 75013 Paris, France;
- Correspondence:
| |
Collapse
|
48
|
Reichenbach M, Mendez P, da Silva Madaleno C, Ugorets V, Rikeit P, Boerno S, Jatzlau J, Knaus P. Differential Impact of Fluid Shear Stress and YAP/TAZ on BMP/TGF‐β Induced Osteogenic Target Genes. Adv Biol (Weinh) 2021; 5:e2000051. [DOI: 10.1002/adbi.202000051] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 12/08/2020] [Indexed: 01/07/2023]
Affiliation(s)
- Maria Reichenbach
- Institute of Chemistry/Biochemistry Freie Universität Berlin Thielallee 63 Berlin 14195 Germany
| | - Paul‐Lennard Mendez
- International Max Planck Research School for Biology and Computation Max Planck Institute for Molecular Genetics Ihnestr. 63 Berlin 14195 Germany
| | - Carolina da Silva Madaleno
- Institute of Chemistry/Biochemistry Freie Universität Berlin Thielallee 63 Berlin 14195 Germany
- Berlin‐Brandenburg School for Regenerative Therapies (BSRT) Charité—Universitätsmedizin Berlin Föhrer Str. 15 Berlin 13353 Germany
| | - Vladimir Ugorets
- Institute of Chemistry/Biochemistry Freie Universität Berlin Thielallee 63 Berlin 14195 Germany
| | - Paul Rikeit
- Institute of Chemistry/Biochemistry Freie Universität Berlin Thielallee 63 Berlin 14195 Germany
- Berlin‐Brandenburg School for Regenerative Therapies (BSRT) Charité—Universitätsmedizin Berlin Föhrer Str. 15 Berlin 13353 Germany
| | - Stefan Boerno
- Sequencing Core Facility Max Planck Institute for Molecular Genetics Ihnestr. 63 Berlin 14195 Germany
| | - Jerome Jatzlau
- Institute of Chemistry/Biochemistry Freie Universität Berlin Thielallee 63 Berlin 14195 Germany
- Berlin‐Brandenburg School for Regenerative Therapies (BSRT) Charité—Universitätsmedizin Berlin Föhrer Str. 15 Berlin 13353 Germany
| | - Petra Knaus
- Institute of Chemistry/Biochemistry Freie Universität Berlin Thielallee 63 Berlin 14195 Germany
| |
Collapse
|
49
|
Peris-Moreno D, Cussonneau L, Combaret L, Polge C, Taillandier D. Ubiquitin Ligases at the Heart of Skeletal Muscle Atrophy Control. Molecules 2021; 26:molecules26020407. [PMID: 33466753 PMCID: PMC7829870 DOI: 10.3390/molecules26020407] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 01/08/2021] [Accepted: 01/10/2021] [Indexed: 02/07/2023] Open
Abstract
Skeletal muscle loss is a detrimental side-effect of numerous chronic diseases that dramatically increases mortality and morbidity. The alteration of protein homeostasis is generally due to increased protein breakdown while, protein synthesis may also be down-regulated. The ubiquitin proteasome system (UPS) is a master regulator of skeletal muscle that impacts muscle contractile properties and metabolism through multiple levers like signaling pathways, contractile apparatus degradation, etc. Among the different actors of the UPS, the E3 ubiquitin ligases specifically target key proteins for either degradation or activity modulation, thus controlling both pro-anabolic or pro-catabolic factors. The atrogenes MuRF1/TRIM63 and MAFbx/Atrogin-1 encode for key E3 ligases that target contractile proteins and key actors of protein synthesis respectively. However, several other E3 ligases are involved upstream in the atrophy program, from signal transduction control to modulation of energy balance. Controlling E3 ligases activity is thus a tempting approach for preserving muscle mass. While indirect modulation of E3 ligases may prove beneficial in some situations of muscle atrophy, some drugs directly inhibiting their activity have started to appear. This review summarizes the main signaling pathways involved in muscle atrophy and the E3 ligases implicated, but also the molecules potentially usable for future therapies.
Collapse
|
50
|
Cell preservation methods and its application to studying rare disease. Mol Cell Probes 2021; 56:101694. [PMID: 33429040 DOI: 10.1016/j.mcp.2021.101694] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/21/2020] [Accepted: 01/05/2021] [Indexed: 12/30/2022]
Abstract
The ability to preserve and transport human cells in a stable medium over long distances is critical to collaborative efforts and the advancement of knowledge in the study of human disease. This is particularly important in the study of rare diseases. Recently, advancements in the understanding of renal ciliopathies has been achieved via the use of patient urine-derived cells (UDCs). However, the traditional method of cryopreservation, although considered as the gold standard, can result in decreased sample viability of many cell types, including UDCs. Delays in transportation can have devastating effects upon the viability of samples, and may even result in complete destruction of cells following evaporation of dry ice or liquid nitrogen, leaving samples in cryoprotective agents, which are cytotoxic at room temperature. The loss of any patient sample in this manner is detrimental to research, however it is even more so when samples are from patients with a rare disease. In order to overcome the associated limitations of traditional practices, new methods of preservation and shipment, including cell encapsulation within hydrogels, and transport in specialised devices are continually being investigated. Here we summarise and compare traditional methods with emerging novel alternatives for the preservation and shipment of cells, and consider the effectiveness of such methods for use with UDCs to further enable the study and understanding of kidney diseases.
Collapse
|