1
|
Pisani S, Evangelista A, Chesi L, Croce S, Avanzini MA, Dorati R, Genta I, Benazzo M, Comoli P, Conti B. Nanofibrous Scaffolds' Ability to Induce Mesenchymal Stem Cell Differentiation for Soft Tissue Regenerative Applications. Pharmaceuticals (Basel) 2025; 18:239. [PMID: 40006052 PMCID: PMC11859969 DOI: 10.3390/ph18020239] [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: 01/06/2025] [Revised: 01/27/2025] [Accepted: 02/07/2025] [Indexed: 02/27/2025] Open
Abstract
Mesenchymal stem cells (MSCs) have gained recognition as a highly versatile and promising cell source for repopulating bioengineered scaffolds due to their inherent capacity to differentiate into multiple cell types. However, MSC implantation techniques have often yielded inconsistent clinical results, underscoring the need for advanced approaches to enhance their therapeutic efficacy. Recent developments in three-dimensional (3D) bioengineered scaffolds have provided a significant breakthrough by closely mimicking the in vivo environment, addressing the limitations of traditional two-dimensional (2D) cell cultures. Among these, nanofibrous scaffolds have proven particularly effective, offering an optimal 3D framework, growth-permissive substrates, and the delivery of trophic factors crucial for MSC survival and regeneration. Furthermore, the selection of appropriate biomaterials can amplify the paracrine effects of MSCs, promoting both proliferation and targeted differentiation. The synergistic combination of MSCs with nanofibrous scaffolds has demonstrated remarkable potential in achieving repair, regeneration, and tissue-specific differentiation with enhanced safety and efficacy, paving the way for routine clinical applications. In this review, we examine the most recent studies (2013-2023) that explore the combined use of MSCs and nanofibrous scaffolds for differentiation into cardiogenic, epithelial, myogenic, tendon, and vascular cell lineages. Using PubMed, we identified and analyzed 275 relevant articles based on the search terms "Nanofibers", "Electrospinning", "Mesenchymal stem cells", and "Differentiation". This review highlights the critical advancements in the use of nanofibrous scaffolds as a platform for MSC differentiation and tissue regeneration. By summarizing key findings from the last decade, it provides valuable insights for researchers and clinicians aiming to optimize scaffold design, MSC integration, and translational applications. These insights could significantly influence future research directions and the development of more effective regenerative therapies.
Collapse
Affiliation(s)
- Silvia Pisani
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy; (L.C.); (R.D.); (I.G.); (B.C.)
| | - Aleksandra Evangelista
- Otorhinolaryngology Unit, Department of Surgical Sciences, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy; (A.E.); (M.B.)
| | - Luca Chesi
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy; (L.C.); (R.D.); (I.G.); (B.C.)
| | - Stefania Croce
- Department of Clinical, Surgical, Diagnostic & Pediatric Sciences, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy; (S.C.); (M.A.A.); (P.C.)
| | - Maria Antonietta Avanzini
- Department of Clinical, Surgical, Diagnostic & Pediatric Sciences, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy; (S.C.); (M.A.A.); (P.C.)
| | - Rossella Dorati
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy; (L.C.); (R.D.); (I.G.); (B.C.)
| | - Ida Genta
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy; (L.C.); (R.D.); (I.G.); (B.C.)
| | - Marco Benazzo
- Otorhinolaryngology Unit, Department of Surgical Sciences, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy; (A.E.); (M.B.)
| | - Patrizia Comoli
- Department of Clinical, Surgical, Diagnostic & Pediatric Sciences, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy; (S.C.); (M.A.A.); (P.C.)
| | - Bice Conti
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy; (L.C.); (R.D.); (I.G.); (B.C.)
| |
Collapse
|
2
|
Younesi FS, Hinz B. The Myofibroblast Fate of Therapeutic Mesenchymal Stromal Cells: Regeneration, Repair, or Despair? Int J Mol Sci 2024; 25:8712. [PMID: 39201399 PMCID: PMC11354465 DOI: 10.3390/ijms25168712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 07/31/2024] [Accepted: 08/06/2024] [Indexed: 09/02/2024] Open
Abstract
Mesenchymal stromal cells (MSCs) can be isolated from various tissues of healthy or patient donors to be retransplanted in cell therapies. Because the number of MSCs obtained from biopsies is typically too low for direct clinical application, MSC expansion in cell culture is required. However, ex vivo amplification often reduces the desired MSC regenerative potential and enhances undesired traits, such as activation into fibrogenic myofibroblasts. Transiently activated myofibroblasts restore tissue integrity after organ injury by producing and contracting extracellular matrix into scar tissue. In contrast, persistent myofibroblasts cause excessive scarring-called fibrosis-that destroys organ function. In this review, we focus on the relevance and molecular mechanisms of myofibroblast activation upon contact with stiff cell culture plastic or recipient scar tissue, such as hypertrophic scars of large skin burns. We discuss cell mechanoperception mechanisms such as integrins and stretch-activated channels, mechanotransduction through the contractile actin cytoskeleton, and conversion of mechanical signals into transcriptional programs via mechanosensitive co-transcription factors, such as YAP, TAZ, and MRTF. We further elaborate how prolonged mechanical stress can create persistent myofibroblast memory by direct mechanotransduction to the nucleus that can evoke lasting epigenetic modifications at the DNA level, such as histone methylation and acetylation. We conclude by projecting how cell culture mechanics can be modulated to generate MSCs, which epigenetically protected against myofibroblast activation and transport desired regeneration potential to the recipient tissue environment in clinical therapies.
Collapse
Affiliation(s)
- Fereshteh Sadat Younesi
- Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1G6, Canada;
- Keenan Research Institute for Biomedical Science, St. Michael’s Hospital, Toronto, ON M5B 1T8, Canada
| | - Boris Hinz
- Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1G6, Canada;
- Keenan Research Institute for Biomedical Science, St. Michael’s Hospital, Toronto, ON M5B 1T8, Canada
| |
Collapse
|
3
|
Jenkner S, Clark JM, Gronthos S, O’Hare Doig RL. Molars to Medicine: A Focused Review on the Pre-Clinical Investigation and Treatment of Secondary Degeneration following Spinal Cord Injury Using Dental Stem Cells. Cells 2024; 13:817. [PMID: 38786039 PMCID: PMC11119219 DOI: 10.3390/cells13100817] [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: 04/01/2024] [Revised: 05/01/2024] [Accepted: 05/07/2024] [Indexed: 05/25/2024] Open
Abstract
Spinal cord injury (SCI) can result in the permanent loss of mobility, sensation, and autonomic function. Secondary degeneration after SCI both initiates and propagates a hostile microenvironment that is resistant to natural repair mechanisms. Consequently, exogenous stem cells have been investigated as a potential therapy for repairing and recovering damaged cells after SCI and other CNS disorders. This focused review highlights the contributions of mesenchymal (MSCs) and dental stem cells (DSCs) in attenuating various secondary injury sequelae through paracrine and cell-to-cell communication mechanisms following SCI and other types of neurotrauma. These mechanistic events include vascular dysfunction, oxidative stress, excitotoxicity, apoptosis and cell loss, neuroinflammation, and structural deficits. The review of studies that directly compare MSC and DSC capabilities also reveals the superior capabilities of DSC in reducing the effects of secondary injury and promoting a favorable microenvironment conducive to repair and regeneration. This review concludes with a discussion of the current limitations and proposes improvements in the future assessment of stem cell therapy through the reporting of the effects of DSC viability and DSC efficacy in attenuating secondary damage after SCI.
Collapse
Affiliation(s)
- Sandra Jenkner
- School of Biomedicine, Faculty of Health and Medical Sciences, University of Adelaide, North Terrace, Adelaide 5000, Australia; (S.J.); (S.G.)
- Neil Sachse Centre for Spinal Cord Research, Lifelong Health Theme, South Australian Health and Medical Research Institute, North Terrace, Adelaide 5000, Australia;
| | - Jillian Mary Clark
- Neil Sachse Centre for Spinal Cord Research, Lifelong Health Theme, South Australian Health and Medical Research Institute, North Terrace, Adelaide 5000, Australia;
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, North Terrace, Adelaide 5000, Australia
| | - Stan Gronthos
- School of Biomedicine, Faculty of Health and Medical Sciences, University of Adelaide, North Terrace, Adelaide 5000, Australia; (S.J.); (S.G.)
- Mesenchymal Stem Cell Laboratory, Precision Medicine Theme, South Australian Health and Medical Research Institute, North Terrace, Adelaide 5000, Australia
| | - Ryan Louis O’Hare Doig
- Neil Sachse Centre for Spinal Cord Research, Lifelong Health Theme, South Australian Health and Medical Research Institute, North Terrace, Adelaide 5000, Australia;
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, North Terrace, Adelaide 5000, Australia
| |
Collapse
|
4
|
Jevtovic F, Zheng D, Claiborne A, Biagioni EM, Wisseman BL, Krassovskaia PM, Collier DN, Isler C, DeVente JE, Neufer PD, Houmard JA, May LE. Effects of maternal exercise on infant mesenchymal stem cell mitochondrial function, insulin action, and body composition in infancy. Physiol Rep 2024; 12:e16028. [PMID: 38684442 PMCID: PMC11058002 DOI: 10.14814/phy2.16028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 03/30/2024] [Accepted: 04/12/2024] [Indexed: 05/02/2024] Open
Abstract
Maternal exercise (ME) has been established as a useful non-pharmacological intervention to improve infant metabolic health; however, mechanistic insight behind these adaptations remains mostly confined to animal models. Infant mesenchymal stem cells (MSCs) give rise to infant tissues (e.g., skeletal muscle), and remain involved in mature tissue maintenance. Importantly, these cells maintain metabolic characteristics of an offspring donor and provide a model for the investigation of mechanisms behind infant metabolic health improvements. We used undifferentiated MSC to investigate if ME affects infant MSC mitochondrial function and insulin action, and if these adaptations are associated with lower infant adiposity. We found that infants from exercising mothers have improvements in MSC insulin signaling related to higher MSC respiration and fat oxidation, and expression and activation of energy-sensing and redox-sensitive proteins. Further, we found that infants exposed to exercise in utero were leaner at 1 month of age, with a significant inverse correlation between infant MSC respiration and infant adiposity at 6 months of age. These data suggest that infants from exercising mothers are relatively leaner, and this is associated with higher infant MSC mitochondrial respiration, fat use, and insulin action.
Collapse
Affiliation(s)
- Filip Jevtovic
- Department of KinesiologyEast Carolina UniversityGreenvilleNorth CarolinaUSA
- Human Performance LaboratoryEast Carolina UniversityGreenvilleNorth CarolinaUSA
- East Carolina Diabetes and Obesity InstituteEast Carolina UniversityGreenvilleNorth CarolinaUSA
| | - Donghai Zheng
- Department of KinesiologyEast Carolina UniversityGreenvilleNorth CarolinaUSA
- Human Performance LaboratoryEast Carolina UniversityGreenvilleNorth CarolinaUSA
- East Carolina Diabetes and Obesity InstituteEast Carolina UniversityGreenvilleNorth CarolinaUSA
| | - Alex Claiborne
- Department of KinesiologyEast Carolina UniversityGreenvilleNorth CarolinaUSA
- Human Performance LaboratoryEast Carolina UniversityGreenvilleNorth CarolinaUSA
- East Carolina Diabetes and Obesity InstituteEast Carolina UniversityGreenvilleNorth CarolinaUSA
| | - Ericka M. Biagioni
- Department of KinesiologyEast Carolina UniversityGreenvilleNorth CarolinaUSA
- Human Performance LaboratoryEast Carolina UniversityGreenvilleNorth CarolinaUSA
- East Carolina Diabetes and Obesity InstituteEast Carolina UniversityGreenvilleNorth CarolinaUSA
| | - Breanna L. Wisseman
- Department of KinesiologyEast Carolina UniversityGreenvilleNorth CarolinaUSA
- Human Performance LaboratoryEast Carolina UniversityGreenvilleNorth CarolinaUSA
- East Carolina Diabetes and Obesity InstituteEast Carolina UniversityGreenvilleNorth CarolinaUSA
| | - Polina M. Krassovskaia
- Department of KinesiologyEast Carolina UniversityGreenvilleNorth CarolinaUSA
- Human Performance LaboratoryEast Carolina UniversityGreenvilleNorth CarolinaUSA
- East Carolina Diabetes and Obesity InstituteEast Carolina UniversityGreenvilleNorth CarolinaUSA
| | - David N. Collier
- East Carolina Diabetes and Obesity InstituteEast Carolina UniversityGreenvilleNorth CarolinaUSA
- Department of Pediatrics, Brody School of MedicineEast Carolina UniversityGreenvilleNorth CarolinaUSA
| | - Christy Isler
- Department of Obstetrics and Gynecology, Brody School of MedicineEast Carolina UniversityGreenvilleNorth CarolinaUSA
| | - James E. DeVente
- Department of Obstetrics and Gynecology, Brody School of MedicineEast Carolina UniversityGreenvilleNorth CarolinaUSA
| | - P. Darrell Neufer
- East Carolina Diabetes and Obesity InstituteEast Carolina UniversityGreenvilleNorth CarolinaUSA
- Department of Physiology, Brody School of MedicineEast Carolina UniversityGreenvilleNorth CarolinaUSA
- Department of Biochemistry & Molecular Biology, Brody School of MedicineEast Carolina UniversityGreenvilleNorth CarolinaUSA
| | - Joseph A. Houmard
- Department of KinesiologyEast Carolina UniversityGreenvilleNorth CarolinaUSA
- Human Performance LaboratoryEast Carolina UniversityGreenvilleNorth CarolinaUSA
- East Carolina Diabetes and Obesity InstituteEast Carolina UniversityGreenvilleNorth CarolinaUSA
| | - Linda E. May
- Department of KinesiologyEast Carolina UniversityGreenvilleNorth CarolinaUSA
- Human Performance LaboratoryEast Carolina UniversityGreenvilleNorth CarolinaUSA
- East Carolina Diabetes and Obesity InstituteEast Carolina UniversityGreenvilleNorth CarolinaUSA
- Department of Obstetrics and Gynecology, Brody School of MedicineEast Carolina UniversityGreenvilleNorth CarolinaUSA
| |
Collapse
|
5
|
Pires F, Silva JC, Ferreira FC, Portugal CAM. Heparinized Acellular Hydrogels for Magnetically Induced Wound Healing Applications. ACS APPLIED MATERIALS & INTERFACES 2024; 16:9908-9924. [PMID: 38381140 DOI: 10.1021/acsami.3c18877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2024]
Abstract
The control of angiogenesis has the potential to be used for regulation of several pathological and physiological processes, which can be instrumental on the development of anticancer and wound healing therapeutical approaches. In this study, mesenchymal stem/stromal cells (MSCs) were seeded on magnetic-responsive gelatin, with or without heparin functionalization, and exposed to a static 0.08 T magnetic field (MF), for controlling their anti-inflammatory and angiogenic activity, with the aim of accelerating tissue healing. For the first time, it was examined how the amount of heparin and magnetic nanoparticles (MNPs) distributed on gelatin scaffolds affected the mechanical properties of the hydrogels and the morphology, proliferation, and secretome profiling of MSCs. The findings demonstrated that the addition of MNPs and heparin affects the hydrogel swelling capacity and renders distinct MSC proliferation rates. Additionally, MF acts as a topographical cue to guide MSCs alignment and increases the level of expression of specific genes and proteins that promote angiogenesis. The results also suggested that the presence of higher amounts of heparin (10 μg/cm3) interferes with the secretion and limits the capacity of angiogenic factors to diffuse through the hydrogel and into the culture medium. Ultimately, this study shows that acellular heparinized hydrogels efficiently retain the angiogenic growth factors released by magnetically stimulated MSCs thus rendering superior wound contraction (55.8% ± 0.4%) and cell migration rate (49.4% ± 0.4%), in comparison to nonheparinized hydrogels (35.2% ± 0.7% and 37.8% ± 0.7%, respectively). Therefore, these heparinized magnetic hydrogels can be used to facilitate angiogenesis in various forms of tissue damage including bone defects, skin wounds, and cardiovascular diseases, leading to enhanced tissue regeneration.
Collapse
Affiliation(s)
- Filipa Pires
- Instituto de Telecomunicações, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
- LAQV-REQUIMTE, Department of Chemistry, NOVA School of Science and Technology, Universidade Nova de Lisboa, 2829-516 Caparica, Portugal
| | - João Carlos Silva
- Department of Bioengineering and iBB - Institute of Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Frederico Castelo Ferreira
- Department of Bioengineering and iBB - Institute of Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Carla A M Portugal
- LAQV-REQUIMTE, Department of Chemistry, NOVA School of Science and Technology, Universidade Nova de Lisboa, 2829-516 Caparica, Portugal
| |
Collapse
|
6
|
Koung Ngeun S, Shimizu M, Kaneda M. Myogenic Differentiation and Immunomodulatory Properties of Rat Adipose-Derived Mesenchymal Stem/Stromal Cells. BIOLOGY 2024; 13:72. [PMID: 38392291 PMCID: PMC10886144 DOI: 10.3390/biology13020072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 01/19/2024] [Accepted: 01/24/2024] [Indexed: 02/24/2024]
Abstract
The myogenic differentiation potential of MSCs is a key factor in their potential use as a cell source for muscle tissue repair and regeneration. Additionally, evaluating the immunomodulatory properties of MSCs is important to highlight their potential for regulating inflammation and supporting tissue regeneration. Given the limited literature on muscle differentiation potential and immunomodulatory properties, this study aims to characterize rat ADP MSCs for treating muscle disease. We isolated MSCs from adipose tissues around the periscapular region of the rats. We used a monoculture method for the myogenic differentiation and modified the myogenic induction medium by supplementing it with the growth factors FGF, HGF, and IGF. In rat ADP MSCs, expression of the MSC-specific marker, CD90, was 87.7%, while CD44 was 42.8%. For genes involved in immunomodulation, IGF1 and TGFB1 were highly expressed, while IL6 was poorly expressed. In addition to their trilineage differentiation potential, ADP MSCs exhibited the capacity to differentiate into myogenic cell lines, as evidenced by changes in cell morphology, leading to elongated and aligned structures and the expression of the MyoD and MYOG antibodies. The study found that ADP MSCs show great clinical promise for muscle regeneration.
Collapse
Affiliation(s)
- Sai Koung Ngeun
- Laboratory of Veterinary Diagnostic Imaging, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo 183-8509, Japan
| | - Miki Shimizu
- Laboratory of Veterinary Diagnostic Imaging, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo 183-8509, Japan
| | - Masahiro Kaneda
- Laboratory of Veterinary Anatomy, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, Tokyo 183-8509, Japan
| |
Collapse
|
7
|
O’Brien JG, Willis AB, Long AM, Kwon J, Lee G, Li FW, Page PG, Vo AH, Hadhazy M, Spencer MJ, Crosbie RH, Demonbreun AR, McNally EM. The super-healing MRL strain promotes muscle growth in muscular dystrophy through a regenerative extracellular matrix. JCI Insight 2024; 9:e173246. [PMID: 38175727 PMCID: PMC11143963 DOI: 10.1172/jci.insight.173246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 12/21/2023] [Indexed: 01/05/2024] Open
Abstract
The Murphy Roths Large (MRL) mouse strain has "super-healing" properties that enhance recovery from injury. In mice, the DBA/2J strain intensifies many aspects of muscular dystrophy, so we evaluated the ability of the MRL strain to suppress muscular dystrophy in the Sgcg-null mouse model of limb girdle muscular dystrophy. A comparative analysis of Sgcg-null mice in the DBA/2J versus MRL strains showed greater myofiber regeneration, with reduced structural degradation of muscle in the MRL strain. Transcriptomic profiling of dystrophic muscle indicated strain-dependent expression of extracellular matrix (ECM) and TGF-β signaling genes. To investigate the MRL ECM, cellular components were removed from dystrophic muscle sections to generate decellularized myoscaffolds. Decellularized myoscaffolds from dystrophic mice in the protective MRL strain had significantly less deposition of collagen and matrix-bound TGF-β1 and TGF-β3 throughout the matrix. Dystrophic myoscaffolds from the MRL background, but not the DBA/2J background, were enriched in myokines like IGF-1 and IL-6. C2C12 myoblasts seeded onto decellularized matrices from Sgcg-/- MRL and Sgcg-/- DBA/2J muscles showed the MRL background induced greater myoblast differentiation compared with dystrophic DBA/2J myoscaffolds. Thus, the MRL background imparts its effect through a highly regenerative ECM, which is active even in muscular dystrophy.
Collapse
Affiliation(s)
- Joseph G. O’Brien
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Alexander B. Willis
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Ashlee M. Long
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Jason Kwon
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - GaHyun Lee
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Frank W. Li
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Patrick G.T. Page
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Andy H. Vo
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Michele Hadhazy
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Melissa J. Spencer
- Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, California, USA
| | - Rachelle H. Crosbie
- Department of Integrative Biology and Physiology, Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, California, USA
| | - Alexis R. Demonbreun
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Elizabeth M. McNally
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
8
|
Grobbelaar S, Mercier AE, van den Bout I, Durandt C, Pepper MS. Considerations for enhanced mesenchymal stromal/stem cell myogenic commitment in vitro. Clin Transl Sci 2024; 17:e13703. [PMID: 38098144 PMCID: PMC10787211 DOI: 10.1111/cts.13703] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 11/16/2023] [Accepted: 12/09/2023] [Indexed: 01/15/2024] Open
Abstract
The generation of tissue from stem cells is an alluring concept as it holds a number of potential applications in clinical therapeutics and regenerative medicine. Mesenchymal stromal/stem cells (MSCs) can be isolated from a number of different somatic sources, and have the capacity to differentiate into adipogenic, osteogenic, chondrogenic, and myogenic lineages. Although the first three have been extensively investigated, there remains a paucity of literature on the latter. This review looks at the various strategies available in vitro to enhance harvested MSC commitment and differentiation into the myogenic pathway. These include chemical inducers, myogenic-enhancing cell culture substrates, and mechanical and dynamic culturing conditions. Drawing on information from embryonic and postnatal myogenesis from somites, satellite, and myogenic progenitor cells, the mechanisms behind the chemical and mechanical induction strategies can be studied, and the sequential gene and signaling cascades can be used to monitor the progression of myogenic differentiation in the laboratory. Increased understanding of the stimuli and signaling mechanisms in the initial stages of MSC myogenic commitment will provide tools with which we can enhance their differentiation efficacy and advance the process to clinical translation.
Collapse
Affiliation(s)
- Simone Grobbelaar
- Department of Physiology, School of Medicine, Faculty of Health SciencesUniversity of PretoriaPretoriaSouth Africa
- Institute for Cellular and Molecular Medicine, Department of Immunology, and South African Medical Research Council Extramural Unit for Stem Cell Research and Therapy, School of Medicine, Faculty of Health SciencesUniversity of PretoriaPretoriaSouth Africa
| | - Anne E. Mercier
- Department of Physiology, School of Medicine, Faculty of Health SciencesUniversity of PretoriaPretoriaSouth Africa
| | - Iman van den Bout
- Department of Physiology, School of Medicine, Faculty of Health SciencesUniversity of PretoriaPretoriaSouth Africa
- Centre for Neuroendocrinology, Department of Immunology, School of Medicine, Faculty of Health SciencesUniversity of PretoriaPretoriaSouth Africa
| | - Chrisna Durandt
- Institute for Cellular and Molecular Medicine, Department of Immunology, and South African Medical Research Council Extramural Unit for Stem Cell Research and Therapy, School of Medicine, Faculty of Health SciencesUniversity of PretoriaPretoriaSouth Africa
| | - Michael S. Pepper
- Institute for Cellular and Molecular Medicine, Department of Immunology, and South African Medical Research Council Extramural Unit for Stem Cell Research and Therapy, School of Medicine, Faculty of Health SciencesUniversity of PretoriaPretoriaSouth Africa
| |
Collapse
|
9
|
Chepeleva EV. Cell Therapy in the Treatment of Coronary Heart Disease. Int J Mol Sci 2023; 24:16844. [PMID: 38069167 PMCID: PMC10706847 DOI: 10.3390/ijms242316844] [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: 11/07/2023] [Revised: 11/24/2023] [Accepted: 11/27/2023] [Indexed: 12/18/2023] Open
Abstract
Heart failure is a leading cause of death in patients who have suffered a myocardial infarction. Despite the timely use of modern reperfusion therapies such as thrombolysis, surgical revascularization and balloon angioplasty, they are sometimes unable to prevent the development of significant areas of myocardial damage and subsequent heart failure. Research efforts have focused on developing strategies to improve the functional status of myocardial injury areas. Consequently, the restoration of cardiac function using cell therapy is an exciting prospect. This review describes the characteristics of various cell types relevant to cellular cardiomyoplasty and presents findings from experimental and clinical studies investigating cell therapy for coronary heart disease. Cell delivery methods, optimal dosage and potential treatment mechanisms are discussed.
Collapse
Affiliation(s)
- Elena V. Chepeleva
- Federal State Budgetary Institution National Medical Research Center Named after Academician E.N. Meshalkin of the Ministry of Health of the Russian Federation, 15, Rechkunovskaya Str., 630055 Novosibirsk, Russia;
- Research Institute of Clinical and Experimental Lymphology—Branch of the Institute of Cytology and Genetics Siberian Branch of Russian Academy of Sciences, 2, Timakova Str., 630060 Novosibirsk, Russia
| |
Collapse
|
10
|
Franchi-Mendes T, Silva M, Cartaxo AL, Fernandes-Platzgummer A, Cabral JMS, da Silva CL. Bioprocessing Considerations towards the Manufacturing of Therapeutic Skeletal and Smooth Muscle Cells. Bioengineering (Basel) 2023; 10:1067. [PMID: 37760170 PMCID: PMC10525286 DOI: 10.3390/bioengineering10091067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/31/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
Tissue engineering approaches within the muscle context represent a promising emerging field to address the current therapeutic challenges related with multiple pathological conditions affecting the muscle compartments, either skeletal muscle or smooth muscle, responsible for involuntary and voluntary contraction, respectively. In this review, several features and parameters involved in the bioprocessing of muscle cells are addressed. The cell isolation process is depicted, depending on the type of tissue (smooth or skeletal muscle), followed by the description of the challenges involving the use of adult donor tissue and the strategies to overcome the hurdles of reaching relevant cell numbers towards a clinical application. Specifically, the use of stem/progenitor cells is highlighted as a source for smooth and skeletal muscle cells towards the development of a cellular product able to maintain the target cell's identity and functionality. Moreover, taking into account the need for a robust and cost-effective bioprocess for cell manufacturing, the combination of muscle cells with biomaterials and the need for scale-up envisioning clinical applications are also approached.
Collapse
Affiliation(s)
- Teresa Franchi-Mendes
- Department of Bioengineering, iBB—Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal; (T.F.-M.); (M.S.); (A.L.C.); (A.F.-P.); (J.M.S.C.)
- Associate Laboratory, i4HB—Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Marília Silva
- Department of Bioengineering, iBB—Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal; (T.F.-M.); (M.S.); (A.L.C.); (A.F.-P.); (J.M.S.C.)
- Associate Laboratory, i4HB—Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Ana Luísa Cartaxo
- Department of Bioengineering, iBB—Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal; (T.F.-M.); (M.S.); (A.L.C.); (A.F.-P.); (J.M.S.C.)
- Associate Laboratory, i4HB—Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Ana Fernandes-Platzgummer
- Department of Bioengineering, iBB—Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal; (T.F.-M.); (M.S.); (A.L.C.); (A.F.-P.); (J.M.S.C.)
- Associate Laboratory, i4HB—Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Joaquim M. S. Cabral
- Department of Bioengineering, iBB—Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal; (T.F.-M.); (M.S.); (A.L.C.); (A.F.-P.); (J.M.S.C.)
- Associate Laboratory, i4HB—Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Cláudia L. da Silva
- Department of Bioengineering, iBB—Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal; (T.F.-M.); (M.S.); (A.L.C.); (A.F.-P.); (J.M.S.C.)
- Associate Laboratory, i4HB—Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| |
Collapse
|
11
|
González Díaz EC, Tai M, Monette CEF, Wu JY, Yang F. Spatially patterned 3D model mimics key features of cancer metastasis to bone. Biomaterials 2023; 299:122163. [PMID: 37236137 PMCID: PMC10621670 DOI: 10.1016/j.biomaterials.2023.122163] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 05/01/2023] [Accepted: 05/14/2023] [Indexed: 05/28/2023]
Abstract
Bone is the most common target of metastasis in breast cancer and prostate cancer, leading to significant mortality due to lack of effective treatments. The discovery of novel therapies has been hampered by a lack of physiologically relevant in vitro models that can mimic key clinical features of bone metastases. To fill this critical gap, here we report spatially patterned, tissue engineered 3D models of breast cancer and prostate cancer bone metastasis which mimic bone-specific invasion, cancer aggressiveness, cancer-induced dysregulation of bone remodeling, and in vivo drug response. We demonstrate the potential of integrating such 3D models with single-cell RNA sequencing to identify key signaling drivers of cancer metastasis to bone. Together, these results validate that spatially patterned 3D bone metastasis models mimic key clinical features of bone metastasis and can serve as a novel research tool to elucidate bone metastasis biology and expedite drug discovery.
Collapse
Affiliation(s)
- Eva C González Díaz
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA.
| | - Michelle Tai
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA
| | - Callan E F Monette
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA
| | - Joy Y Wu
- Division of Endocrinology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Fan Yang
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA; Department of Orthopaedic Surgery, Stanford University, Stanford, CA, 94305, USA.
| |
Collapse
|
12
|
Ishkaeva RA, Khaertdinov NN, Yakovlev AV, Esmeteva MV, Salakhieva DV, Nizamov IS, Sitdikova GF, Abdullin TI. Characterization of Glutathione Dithiophosphates as Long-Acting H 2S Donors. Int J Mol Sci 2023; 24:11063. [PMID: 37446245 DOI: 10.3390/ijms241311063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 06/28/2023] [Accepted: 06/30/2023] [Indexed: 07/15/2023] Open
Abstract
Considering the important cytoprotective and signaling roles but relatively narrow therapeutic index of hydrogen sulfide (H2S), advanced H2S donors are required to achieve a therapeutic effect. In this study, we proposed glutathione dithiophosphates as new combination donors of H2S and glutathione. The kinetics of H2S formation in dithiophosphate solutions suggested a continuous H2S release by the donors, which was higher for the dithiophosphate of reduced glutathione than oxidized glutathione. The compounds, unlike NaHS, inhibited the proliferation of C2C12 myoblasts at submillimolar concentrations due to an efficient increase in intracellular H2S. The H2S donors more profoundly affected reactive oxygen species and reduced glutathione levels in C2C12 myocytes, in which these parameters were elevated compared to myoblasts. Oxidized glutathione dithiophosphate as well as control donors exerted antioxidant action toward myocytes, whereas the effect of reduced glutathione dithiophosphate at (sub-)micromolar concentrations was rather modulating. This dithiophosphate showed an enhanced negative inotropic effect mediated by H2S upon contraction of the atrial myocardium, furthermore, its activity was prolonged and reluctant for washing. These findings identify glutathione dithiophosphates as redox-modulating H2S donors with long-acting profile, which are of interest for further pharmacological investigation.
Collapse
Affiliation(s)
- Rezeda A Ishkaeva
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, 18 Kremlyovskaya St., 420008 Kazan, Russia
- Scientific and Educational Center of Pharmaceutics, Kazan (Volga Region) Federal University, 18 Kremlyovskaya St., 420008 Kazan, Russia
| | - Nail N Khaertdinov
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, 18 Kremlyovskaya St., 420008 Kazan, Russia
| | - Aleksey V Yakovlev
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, 18 Kremlyovskaya St., 420008 Kazan, Russia
| | - Marina V Esmeteva
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, 18 Kremlyovskaya St., 420008 Kazan, Russia
- Scientific and Educational Center of Pharmaceutics, Kazan (Volga Region) Federal University, 18 Kremlyovskaya St., 420008 Kazan, Russia
| | - Diana V Salakhieva
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, 18 Kremlyovskaya St., 420008 Kazan, Russia
- Scientific and Educational Center of Pharmaceutics, Kazan (Volga Region) Federal University, 18 Kremlyovskaya St., 420008 Kazan, Russia
| | - Ilyas S Nizamov
- Alexander Butlerov Institute of Chemistry, Kazan (Volga Region) Federal University, 420008 Kazan, Russia
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS, 8 Arbuzov St., 420088 Kazan, Russia
| | - Guzel F Sitdikova
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, 18 Kremlyovskaya St., 420008 Kazan, Russia
| | - Timur I Abdullin
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, 18 Kremlyovskaya St., 420008 Kazan, Russia
- Scientific and Educational Center of Pharmaceutics, Kazan (Volga Region) Federal University, 18 Kremlyovskaya St., 420008 Kazan, Russia
| |
Collapse
|
13
|
Ichiseki T, Shimasaki M, Ueda S, Hirata H, Souma D, Kawahara N, Ueda Y. Efficacy of Rectal Systemic Administration of Mesenchymal Stem Cells to Injury Sites via the CXCL12/CXCR4 Axis to Promote Regeneration in a Rabbit Skeletal Muscle Injury Model. Cells 2023; 12:1729. [PMID: 37443763 PMCID: PMC10340610 DOI: 10.3390/cells12131729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 06/22/2023] [Accepted: 06/22/2023] [Indexed: 07/15/2023] Open
Abstract
Mesenchymal stem cells (MSCs) have been transplanted directly into lesions or injected intravenously. The administration of MSCs using these delivery methods requires specialized knowledge, techniques, and facilities. Here, we describe intrarectal systemic administration of MSCs, a simple, non-invasive route for homing to the injury sites to promote the regeneration of skeletal muscle injuries. Using a cardiotoxin (CTX)-induced rabbit skeletal muscle injury model, homing to the site of muscle injury was confirmed by intrarectal administration of MSCs; the time required for homing after intrarectal administration was approximately 5 days. In addition, the C-X-C chemokine ligand 12 (CXCL12)/C-X-C chemokine receptor-4 (CXCR4) axis was found to be involved in the homing process. Histopathological examinations showed that skeletal muscle regeneration was promoted in the MSCs-administered group compared to the CTX-only group. Myosin heavy polypeptide 3 (Myh3) expression, an indicator of early muscle regeneration, was detected earlier in the intrarectal MSCs group compared to the CTX-only group. These findings indicate that intrarectal administration of MSCs is effective in homing to the injured area, where they promote injury repair. Since intrarectal administration is a simple and non-invasive delivery route, these findings may be valuable in future research on stem cell therapy.
Collapse
Affiliation(s)
- Toru Ichiseki
- Department of Orthopaedic Surgery, Kanazawa Medical University, Daigaku 1-1, Uchinada-machi, Kahoku 920-0293, Japan; (S.U.)
| | - Miyako Shimasaki
- Department of Pathology 2, Kanazawa Medical University, Daigaku 1-1, Uchinada-machi, Kahoku 920-0293, Japan;
| | - Shusuke Ueda
- Department of Orthopaedic Surgery, Kanazawa Medical University, Daigaku 1-1, Uchinada-machi, Kahoku 920-0293, Japan; (S.U.)
| | - Hiroaki Hirata
- Department of Orthopaedic Surgery, Kanazawa Medical University, Daigaku 1-1, Uchinada-machi, Kahoku 920-0293, Japan; (S.U.)
| | - Daisuke Souma
- Department of Orthopaedic Surgery, Kanazawa Medical University, Daigaku 1-1, Uchinada-machi, Kahoku 920-0293, Japan; (S.U.)
| | - Norio Kawahara
- Department of Orthopaedic Surgery, Kanazawa Medical University, Daigaku 1-1, Uchinada-machi, Kahoku 920-0293, Japan; (S.U.)
| | - Yoshimichi Ueda
- Department of Pathology, Keiju Medical Center, 94, Tomioka-machi, Nanao 926-0816, Japan
| |
Collapse
|
14
|
Cai A, Schneider P, Zheng ZM, Beier JP, Himmler M, Schubert DW, Weisbach V, Horch RE, Arkudas A. Myogenic differentiation of human myoblasts and Mesenchymal stromal cells under GDF11 on NPoly-ɛ-caprolactone-collagen I-Polyethylene-nanofibers. BMC Mol Cell Biol 2023; 24:18. [PMID: 37189080 PMCID: PMC10184409 DOI: 10.1186/s12860-023-00478-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 04/27/2023] [Indexed: 05/17/2023] Open
Abstract
BACKGROUND For the purpose of skeletal muscle engineering, primary myoblasts (Mb) and adipogenic mesenchymal stem cells (ADSC) can be co-cultured and myogenically differentiated. Electrospun composite nanofiber scaffolds represent suitable matrices for tissue engineering of skeletal muscle, combining both biocompatibility and stability Although growth differentiation factor 11 (GDF11) has been proposed as a rejuvenating circulating factor, restoring skeletal muscle function in aging mice, some studies have also described a harming effect of GDF11. Therefore, the aim of the study was to analyze the effect of GDF11 on co-cultures of Mb and ADSC on poly-ε-caprolactone (PCL)-collagen I-polyethylene oxide (PEO)-nanofibers. RESULTS Human Mb were co-cultured with ADSC two-dimensionally (2D) as monolayers or three-dimensionally (3D) on aligned PCL-collagen I-PEO-nanofibers. Differentiation media were either serum-free with or without GDF11, or serum containing as in a conventional differentiation medium. Cell viability was higher after conventional myogenic differentiation compared to serum-free and serum-free + GDF11 differentiation as was creatine kinase activity. Immunofluorescence staining showed myosine heavy chain expression in all groups after 28 days of differentiation without any clear evidence of more or less pronounced expression in either group. Gene expression of myosine heavy chain (MYH2) increased after serum-free + GDF11 stimulation compared to serum-free stimulation alone. CONCLUSIONS This is the first study analyzing the effect of GDF11 on myogenic differentiation of Mb and ADSC co-cultures under serum-free conditions. The results of this study show that PCL-collagen I-PEO-nanofibers represent a suitable matrix for 3D myogenic differentiation of Mb and ADSC. In this context, GDF11 seems to promote myogenic differentiation of Mb and ADSC co-cultures compared to serum-free differentiation without any evidence of a harming effect.
Collapse
Affiliation(s)
- Aijia Cai
- Department of Plastic and Hand Surgery, Laboratory for Tissue Engineering and Regenerative Medicine, University Hospital of Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), 91054, Erlangen, Germany.
| | - Paul Schneider
- Department of Plastic and Hand Surgery, Laboratory for Tissue Engineering and Regenerative Medicine, University Hospital of Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), 91054, Erlangen, Germany
| | - Zeng-Ming Zheng
- Department of Plastic and Hand Surgery, Laboratory for Tissue Engineering and Regenerative Medicine, University Hospital of Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), 91054, Erlangen, Germany
| | - Justus P Beier
- Department of Plastic Surgery, Hand Surgery - Burn Center, University Hospital RWTH Aachen, 52074, Aachen, Germany
| | - Marcus Himmler
- Institute of Polymer Materials, Department of Materials Science and Engineering, University of Erlangen-Nürnberg (FAU), 91058, Erlangen, Germany
| | - Dirk W Schubert
- Institute of Polymer Materials, Department of Materials Science and Engineering, University of Erlangen-Nürnberg (FAU), 91058, Erlangen, Germany
| | - Volker Weisbach
- Department of Transfusion Medicine, University Hospital of Erlangen, Friedrich-Alexander- University Erlangen-Nürnberg (FAU), 91054, Erlangen, Germany
| | - Raymund E Horch
- Department of Plastic and Hand Surgery, Laboratory for Tissue Engineering and Regenerative Medicine, University Hospital of Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), 91054, Erlangen, Germany
| | - Andreas Arkudas
- Department of Plastic and Hand Surgery, Laboratory for Tissue Engineering and Regenerative Medicine, University Hospital of Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), 91054, Erlangen, Germany
| |
Collapse
|
15
|
Lee K, Jackson A, John N, Zhang R, Ozhava D, Bhatia M, Mao Y. Bovine Fibroblast-Derived Extracellular Matrix Promotes the Growth and Preserves the Stemness of Bovine Stromal Cells during In Vitro Expansion. J Funct Biomater 2023; 14:jfb14040218. [PMID: 37103308 PMCID: PMC10144935 DOI: 10.3390/jfb14040218] [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: 03/24/2023] [Revised: 04/06/2023] [Accepted: 04/11/2023] [Indexed: 04/28/2023] Open
Abstract
Cultivated meat is a fast-growing research field and an industry with great potential to overcome the limitations of traditional meat production. Cultivated meat utilizes cell culture and tissue engineering technologies to culture a vast number of cells in vitro and grow/assemble them into structures mimicking the muscle tissues of livestock animals. Stem cells with self-renewal and lineage-specific differentiation abilities have been considered one of the key cell sources for cultivated meats. However, the extensive in vitro culturing/expansion of stem cells results in a reduction in their abilities to proliferate and differentiate. Extracellular matrix (ECM) has been used as a culturing substrate to support cell expansion for cell-based therapies in regenerative medicine due to its resemblance to the native microenvironment of cells. In this study, the effect of the ECM on the expansion of bovine umbilical cord stromal cells (BUSC) in vitro was evaluated and characterized. BUSCs with multi-lineage differentiation potentials were isolated from bovine placental tissue. Decellularized ECM prepared from a confluent monolayer of bovine fibroblasts (BF) is free of cellular components but contains major ECM proteins such as fibronectin and type I collagen and ECM-associated growth factors. Expansion of BUSC on ECM for three passages (around three weeks) resulted in about 500-fold amplification, while cells were amplified less than 10-fold when cultured on standard tissue culture plates (TCP). Moreover, the presence of ECM reduced the requirement for serum in the culture medium. Importantly, the cells amplified on ECM retained their differentiation abilities better than cells cultured on TCP. The results of our study support the notion that monolayer cell-derived ECM may be a strategy to expand bovine cells in vitro effectively and efficiently.
Collapse
Affiliation(s)
- Kathleen Lee
- Laboratory for Biomaterials Research, Department of Chemistry and Chemical Biology, Rutgers University, 145 Bevier Rd., Piscataway, NJ 08854, USA
| | - Anisha Jackson
- Laboratory for Biomaterials Research, Department of Chemistry and Chemical Biology, Rutgers University, 145 Bevier Rd., Piscataway, NJ 08854, USA
| | - Nikita John
- Laboratory for Biomaterials Research, Department of Chemistry and Chemical Biology, Rutgers University, 145 Bevier Rd., Piscataway, NJ 08854, USA
| | - Ryan Zhang
- Laboratory for Biomaterials Research, Department of Chemistry and Chemical Biology, Rutgers University, 145 Bevier Rd., Piscataway, NJ 08854, USA
| | - Derya Ozhava
- Laboratory for Biomaterials Research, Department of Chemistry and Chemical Biology, Rutgers University, 145 Bevier Rd., Piscataway, NJ 08854, USA
| | - Mohit Bhatia
- Atelier Meats, 666 Burrard Street, Suite 500, Vancouver, BC V6C 3P6, Canada
| | - Yong Mao
- Laboratory for Biomaterials Research, Department of Chemistry and Chemical Biology, Rutgers University, 145 Bevier Rd., Piscataway, NJ 08854, USA
| |
Collapse
|
16
|
Bomkamp C, Musgrove L, Marques DMC, Fernando GF, Ferreira FC, Specht EA. Differentiation and Maturation of Muscle and Fat Cells in Cultivated Seafood: Lessons from Developmental Biology. MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2023; 25:1-29. [PMID: 36374393 PMCID: PMC9931865 DOI: 10.1007/s10126-022-10174-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 10/10/2022] [Indexed: 06/16/2023]
Abstract
Cultivated meat, also known as cultured or cell-based meat, is meat produced directly from cultured animal cells rather than from a whole animal. Cultivated meat and seafood have been proposed as a means of mitigating the substantial harms associated with current production methods, including damage to the environment, antibiotic resistance, food security challenges, poor animal welfare, and-in the case of seafood-overfishing and ecological damage associated with fishing and aquaculture. Because biomedical tissue engineering research, from which cultivated meat draws a great deal of inspiration, has thus far been conducted almost exclusively in mammals, cultivated seafood suffers from a lack of established protocols for producing complex tissues in vitro. At the same time, fish such as the zebrafish Danio rerio have been widely used as model organisms in developmental biology. Therefore, many of the mechanisms and signaling pathways involved in the formation of muscle, fat, and other relevant tissue are relatively well understood for this species. The same processes are understood to a lesser degree in aquatic invertebrates. This review discusses the differentiation and maturation of meat-relevant cell types in aquatic species and makes recommendations for future research aimed at recapitulating these processes to produce cultivated fish and shellfish.
Collapse
Affiliation(s)
- Claire Bomkamp
- Department of Science & Technology, The Good Food Institute, Washington, DC USA
| | - Lisa Musgrove
- University of the Sunshine Coast, Sippy Downs, Queensland Australia
| | - Diana M. C. Marques
- Department of Bioengineering and Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Gonçalo F. Fernando
- Department of Science & Technology, The Good Food Institute, Washington, DC USA
| | - Frederico C. Ferreira
- Department of Bioengineering and Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Elizabeth A. Specht
- Department of Science & Technology, The Good Food Institute, Washington, DC USA
| |
Collapse
|
17
|
Scala P, Manzo P, Lamparelli EP, Lovecchio J, Ciardulli MC, Giudice V, Selleri C, Giordano E, Rehak L, Maffulli N, Della Porta G. Peripheral blood mononuclear cells contribute to myogenesis in a 3D bioengineered system of bone marrow mesenchymal stem cells and myoblasts. Front Bioeng Biotechnol 2023; 10:1075715. [PMID: 36704300 PMCID: PMC9871311 DOI: 10.3389/fbioe.2022.1075715] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 12/28/2022] [Indexed: 01/12/2023] Open
Abstract
In this work, a 3D environment obtained using fibrin scaffold and two cell populations, such as bone marrow-derived mesenchymal stem cells (BM-MSCs), and primary skeletal muscle cells (SkMs), was assembled. Peripheral blood mononuclear cells (PBMCs) fraction obtained after blood filtration with HemaTrate® filter was then added to the 3D culture system to explore their influence on myogenesis. The best cell ratio into a 3D fibrin hydrogel was 1:1 (BM-MSCs plus SkMs:PBMCs) when cultured in a perfusion bioreactor; indeed, excellent viability and myogenic event induction were observed. Myogenic genes were significantly overexpressed when cultured with PBMCs, such as MyoD1 of 118-fold at day 14 and Desmin 6-fold at day 21. Desmin and Myosin Heavy Chain were also detected at protein level by immunostaining along the culture. Moreover, the presence of PBMCs in 3D culture induced a significant downregulation of pro-inflammatory cytokine gene expression, such as IL6. This smart biomimetic environment can be an excellent tool for investigation of cellular crosstalk and PBMC influence on myogenic processes.
Collapse
Affiliation(s)
- Pasqualina Scala
- Department of Medicine, Surgery and Dentistry, University of Salerno, Baronissi, Italy
| | - Paola Manzo
- Department of Medicine, Surgery and Dentistry, University of Salerno, Baronissi, Italy,Hematology and Transplant Center, University Hospital “San Giovanni di Dio e Ruggi D’Aragona”, Salerno, Italy
| | | | - Joseph Lovecchio
- Department of Electrical, Electronic and Information Engineering “Guglielmo Marconi” (DEI), University of Bologna, Bologna, Italy
| | | | - Valentina Giudice
- Department of Medicine, Surgery and Dentistry, University of Salerno, Baronissi, Italy,Hematology and Transplant Center, University Hospital “San Giovanni di Dio e Ruggi D’Aragona”, Salerno, Italy
| | - Carmine Selleri
- Department of Medicine, Surgery and Dentistry, University of Salerno, Baronissi, Italy,Hematology and Transplant Center, University Hospital “San Giovanni di Dio e Ruggi D’Aragona”, Salerno, Italy
| | - Emanuele Giordano
- Department of Electrical, Electronic and Information Engineering “Guglielmo Marconi” (DEI), University of Bologna, Bologna, Italy
| | - Laura Rehak
- Athena Biomedical innovations, Florence, Italy
| | - Nicola Maffulli
- Department of Medicine, Surgery and Dentistry, University of Salerno, Baronissi, Italy,Centre for Sports and Exercise Medicine, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, England
| | - Giovanna Della Porta
- Department of Medicine, Surgery and Dentistry, University of Salerno, Baronissi, Italy,Interdepartment Centre BIONAM, University of Salerno, Fisciano, Italy,*Correspondence: Giovanna Della Porta,
| |
Collapse
|
18
|
Scala P, Lovecchio J, Lamparelli EP, Vitolo R, Giudice V, Giordano E, Selleri C, Rehak L, Maffulli N, Della Porta G. Myogenic commitment of human stem cells by myoblasts Co-culture: a static vs. a dynamic approach. ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY 2022; 50:49-58. [PMID: 35188030 DOI: 10.1080/21691401.2022.2039684] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
An in-vitro model of human bone marrow mesenchymal stem cells (hBM-MSCs) myogenic commitment by synergic effect of a differentiation media coupled with human primary skeletal myoblasts (hSkMs) co-culture was developed adopting both conventional static co-seeding and perfused culture systems. Static co-seeding provided a notable outcome in terms of gene expression with a significant increase of Desmin (141-fold) and Myosin heavy chain II (MYH2, 32-fold) at day 21, clearly detected also by semi-quantitative immunofluorescence. Under perfusion conditions, myogenic induction ability of hSkMs on hBM-MSCs was exerted by paracrine effect with an excellent gene overexpression and immunofluorescence detection of MYH2 protein; furthermore, due to the dynamic cell culture in separate wells, western blot data were acquired confirming a successful cell commitment at day 14. A significant increase of anti-inflammatory cytokine gene expression, including IL-10 and IL-4 (15-fold and 11-fold, respectively) at day 14, with respect to the pro-inflammatory cytokines IL-12A (7-fold at day 21) and IL-1β (1.4-fold at day 7) was also detected during dynamic culture, confirming the immunomodulatory activity of hBM-MSCs along with commitment events. The present study opens interesting perspectives on the use of dynamic culture based on perfusion as a versatile tool to study myogenic events and paracrine cross-talk compared to the simple co-seeding static culture.
Collapse
Affiliation(s)
- Pasqualina Scala
- Translational Medicine Laboratory, Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 84081 Baronissi, Salerno (SA), Italy
| | - J Lovecchio
- Mol Cel Eng. Lab "S. Cavalcanti", Department of Electrical, Electronic and Information Engineering "Guglielmo Marconi" (DEI), University of Bologna, Via dell'Universitá 50, 47522 Cesena, Forlí-Cesena (FC), Italy.,Health Sciences and Technologies - Interdepartmental Center for Industrial Research (HST-ICIR), University of Bologna, Via Tolara di Sopra 41/E, 40064 Ozzano dell'Emilia, Bologna (BO), Italy
| | - E P Lamparelli
- Translational Medicine Laboratory, Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 84081 Baronissi, Salerno (SA), Italy
| | - R Vitolo
- Translational Medicine Laboratory, Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 84081 Baronissi, Salerno (SA), Italy
| | - V Giudice
- Translational Medicine Laboratory, Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 84081 Baronissi, Salerno (SA), Italy
| | - E Giordano
- Mol Cel Eng. Lab "S. Cavalcanti", Department of Electrical, Electronic and Information Engineering "Guglielmo Marconi" (DEI), University of Bologna, Via dell'Universitá 50, 47522 Cesena, Forlí-Cesena (FC), Italy.,Health Sciences and Technologies - Interdepartmental Center for Industrial Research (HST-ICIR), University of Bologna, Via Tolara di Sopra 41/E, 40064 Ozzano dell'Emilia, Bologna (BO), Italy.,Advanced Research Center on Electronic Systems (ARCES), University of Bologna, Via Vincenzo Toffano 2/2, 40125 Bologna (BO), Italy
| | - C Selleri
- Translational Medicine Laboratory, Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 84081 Baronissi, Salerno (SA), Italy
| | - L Rehak
- Athena Biomedical innovations, Viale Europa 139, Florence (FI), 50126, Italy
| | - N Maffulli
- Translational Medicine Laboratory, Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 84081 Baronissi, Salerno (SA), Italy
| | - G Della Porta
- Translational Medicine Laboratory, Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 84081 Baronissi, Salerno (SA), Italy.,Interdepartment Centre BIONAM, Università di Salerno, via Giovanni Paolo I, 84084 Fisciano, Salerno (SA), Italy
| |
Collapse
|
19
|
Tissue Engineering Applied to Skeletal Muscle: Strategies and Perspectives. BIOENGINEERING (BASEL, SWITZERLAND) 2022; 9:bioengineering9120744. [PMID: 36550950 PMCID: PMC9774646 DOI: 10.3390/bioengineering9120744] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 11/21/2022] [Accepted: 11/28/2022] [Indexed: 12/04/2022]
Abstract
Muscle tissue is formed by elongated and contractile cells with specific morphofunctional characteristics. Thus, it is divided into three basic types: smooth muscle tissue, cardiac striated muscle tissue and skeletal striated muscle tissue. The striated skeletal muscle tissue presents high plasticity, regeneration and growth capacity due to the presence of satellite cells, quiescent myoblasts that are activated in case of injury to the tissue and originate new muscle fibers when they differentiate. In more severe deficiencies or injuries there is a loss of their regenerative capacity, thus compromising the body's functionality at different levels. Tissue engineering studies the development of biomaterials capable of stimulating the recovery of cellular activity in injured body tissues, as well as the activity of cells with muscle differentiation potential in injury repair. However, the need for three-dimensional re-assembly in a complex organization makes it difficult to mimic this tissue and fully regenerate it for the sake of precise and effective movements. Thus, this article aims to provide a narrative review of tissue engineering strategies applied to the regeneration of skeletal muscle, in a critical evaluation of research, whether aimed at injury or atrophies such as spinal muscular atrophy.
Collapse
|
20
|
Yazdanian M, Alam M, Abbasi K, Rahbar M, Farjood A, Tahmasebi E, Tebyaniyan H, Ranjbar R, Hesam Arefi A. Synthetic materials in craniofacial regenerative medicine: A comprehensive overview. Front Bioeng Biotechnol 2022; 10:987195. [PMID: 36440445 PMCID: PMC9681815 DOI: 10.3389/fbioe.2022.987195] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 10/26/2022] [Indexed: 07/25/2023] Open
Abstract
The state-of-the-art approach to regenerating different tissues and organs is tissue engineering which includes the three parts of stem cells (SCs), scaffolds, and growth factors. Cellular behaviors such as propagation, differentiation, and assembling the extracellular matrix (ECM) are influenced by the cell's microenvironment. Imitating the cell's natural environment, such as scaffolds, is vital to create appropriate tissue. Craniofacial tissue engineering refers to regenerating tissues found in the brain and the face parts such as bone, muscle, and artery. More biocompatible and biodegradable scaffolds are more commensurate with tissue remodeling and more appropriate for cell culture, signaling, and adhesion. Synthetic materials play significant roles and have become more prevalent in medical applications. They have also been used in different forms for producing a microenvironment as ECM for cells. Synthetic scaffolds may be comprised of polymers, bioceramics, or hybrids of natural/synthetic materials. Synthetic scaffolds have produced ECM-like materials that can properly mimic and regulate the tissue microenvironment's physical, mechanical, chemical, and biological properties, manage adherence of biomolecules and adjust the material's degradability. The present review article is focused on synthetic materials used in craniofacial tissue engineering in recent decades.
Collapse
Affiliation(s)
- Mohsen Yazdanian
- Research Center for Prevention of Oral and Dental Diseases, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mostafa Alam
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kamyar Abbasi
- Department of Prosthodontics, School of Dentistry, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahdi Rahbar
- Department of Restorative Dentistry, School of Dentistry, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Amin Farjood
- Orthodontic Department, Dental School, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Elahe Tahmasebi
- Research Center for Prevention of Oral and Dental Diseases, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Hamid Tebyaniyan
- Department of Science and Research, Islimic Azade University, Tehran, Iran
| | - Reza Ranjbar
- Research Center for Prevention of Oral and Dental Diseases, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Arian Hesam Arefi
- Dental Research Center, Zahedan University of Medical Sciences, Zahedan, Iran
| |
Collapse
|
21
|
Khodabukus A, Guyer T, Moore AC, Stevens MM, Guldberg RE, Bursac N. Translating musculoskeletal bioengineering into tissue regeneration therapies. Sci Transl Med 2022; 14:eabn9074. [PMID: 36223445 PMCID: PMC7614064 DOI: 10.1126/scitranslmed.abn9074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Musculoskeletal injuries and disorders are the leading cause of physical disability worldwide and a considerable socioeconomic burden. The lack of effective therapies has driven the development of novel bioengineering approaches that have recently started to gain clinical approvals. In this review, we first discuss the self-repair capacity of the musculoskeletal tissues and describe causes of musculoskeletal dysfunction. We then review the development of novel biomaterial, immunomodulatory, cellular, and gene therapies to treat musculoskeletal disorders. Last, we consider the recent regulatory changes and future areas of technological progress that can accelerate translation of these therapies to clinical practice.
Collapse
Affiliation(s)
- Alastair Khodabukus
- Department of Biomedical Engineering, Duke University; Durham, NC, 27708 USA
| | - Tyler Guyer
- Knight Campus for Accelerating Scientific Impact, University of Oregon, Eugene, OR, 97403 USA
| | - Axel C. Moore
- Department of Materials, Department of Bioengineering and Institute of Biomedical Engineering, Imperial College London; London, SW7 2AZ UK
- Department of Biomedical Engineering, University of Delaware; Newark, DE, 19716 USA
| | - Molly M. Stevens
- Department of Materials, Department of Bioengineering and Institute of Biomedical Engineering, Imperial College London; London, SW7 2AZ UK
- Department of Medical Biochemistry and Biophysics, Karolinska Institute; Stockholm, 17177 SE
| | - Robert E. Guldberg
- Knight Campus for Accelerating Scientific Impact, University of Oregon, Eugene, OR, 97403 USA
| | - Nenad Bursac
- Department of Biomedical Engineering, Duke University; Durham, NC, 27708 USA
| |
Collapse
|
22
|
Li Q, Yang Q, Liu X, Liang W, Zhang X, Wang Y. Effect and mechanism of a novel Mg-Nd-Gd-Sr alloy on osteogenic differentiation of bone marrow mesenchymal stem cells. J Biomater Appl 2022; 37:829-837. [PMID: 35977627 DOI: 10.1177/08853282221121880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We investigated the effect and mechanism of a novel Mg-3Nd-1Gd-0.3Sr-0.2Zn-0.4Zr (abbreviated to Mg-Nd-Gd-Sr) alloy on the osteogenic differentiation of bone marrow mesenchymal stem cells extracted from Sprague-Dawley rats. Cultured cells were divided into five groups: a control group cultured in osteogenic induction medium alone without Mg-Nd-Gd-Sr alloy extract, and four experimental groups cultured in the same medium with 25%, 50%, 75%, and 100% Mg-Nd-Gd-Sr alloy extracts, respectively. After 14 days of culture, ALP activity was determined and expressions of osteogenesis-related factors Runx2, OCN, and OPN at the mRNA level and Runx2, OCN, and OPN at the protein level were detected by RT-PCR and western blot, respectively. After 21 days of culture, mineralized nodules were detected by alizarin red staining. The results showed that bone marrow mesenchymal stem cells from Sprague-Dawley rats were successfully isolated in vitro using the whole bone marrow adherence method. Flow cytometry revealed that the cells expressed high levels of CD44 and CD90, but low levels of CD31 and CD45. Alizarin red staining indicated the formation of mineralized nodules in all five groups. Compared with the control group, the number of mineralized nodules was increased significantly in the four experimental groups (p < 0.05). The ALP activity in each group was significantly higher on day 14 than on day 7, and was significantly higher in the four experimental groups compared with the control group (p < 0.05). Moreover, the ALP activity was highest when the concentration of Mg-Nd-Gd-Sr alloy extract was 75% (p < 0.05). RT-PCR results showed that, compared with the control group, the mRNA expression of Runx2, OPN, and OCN was significantly higher in the four experimental groups (p < 0.05), and the highest mRNA expression of Runx2, OPN, and OCN was observed in the 75% experimental group (p < 0.05). Western blotting showed that Mg-Nd-Gd-Sr alloy extract significantly increased the protein expression of Runx2, OCN, and OPN compared with the control group (p < 0.05). Our data indicate that the novel Mg-Nd-Gd-Sr alloy can promotes the osteogenic differentiation of bone marrow mesenchymal stem cells isolated from Sprague-Dawley rats. During this process, there is an increase in the expressions of Runx2, OPN, and OCN mRNAs and Runx2, OCN, and OPN proteins.
Collapse
Affiliation(s)
- Qiangqiang Li
- Department of Orthopedics, 117741the First Hospital of Lanzhou University, Lanzhou, China
| | - Qinglin Yang
- Department of Orthopedics, 117741the First Hospital of Lanzhou University, Lanzhou, China
| | - Xiaorong Liu
- College of Clinical Medicine, 12426Northwest University for Nationalities, Lanzhou, China.,Department of Laboratory, the Second People's Hospital of Gansu Province, Lanzhou, China
| | - Wenqiang Liang
- Department of Orthopedics, 117741the First Hospital of Lanzhou University, Lanzhou, China
| | - Xiaobo Zhang
- School of Materials Science and Engineering, Nanjing Institute of Technology, Nanjing, China
| | - Yongping Wang
- Department of Orthopedics, 117741the First Hospital of Lanzhou University, Lanzhou, China
| |
Collapse
|
23
|
The Microenvironment That Regulates Vascular Wall Stem/Progenitor Cells in Vascular Injury and Repair. BIOMED RESEARCH INTERNATIONAL 2022; 2022:9377965. [PMID: 35958825 PMCID: PMC9357805 DOI: 10.1155/2022/9377965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 07/15/2022] [Accepted: 07/19/2022] [Indexed: 11/17/2022]
Abstract
Vascular repair upon injury is a frequently encountered pathology in cardiovascular diseases, which is crucial for the maintenance of arterial homeostasis and function. Stem/progenitor cells located on vascular walls have multidirectional differentiation potential and regenerative ability. It has been demonstrated that stem/progenitor cells play an essential role in the basic medical research and disease treatment. The dynamic microenvironment around the vascular wall stem/progenitor cells (VW-S/PCs) possesses many stem cell niche-like characteristics to support and regulate cells' activities, maintaining the properties of stem cells. Under physiological conditions, vascular homeostasis is a cautiously balanced and efficient interaction between stem cells and the microenvironment. These interactions contribute to the vascular repair and remodeling upon vessel injury. However, the signaling mechanisms involved in the regulation of microenvironment on stem cells remain to be further elucidated. Understanding the functional characteristics and potential mechanisms of VW-S/PCs is of great significance for both basic and translational research. This review underscores the microenvironment-derived signals that regulate VW-S/PCs and aims at providing new targets for the treatment of related cardiovascular diseases.
Collapse
|
24
|
Chaves A, Weyrauch LA, Zheng D, Biagioni EM, Krassovskaia PM, Davidson BL, Broskey NT, Boyle KE, May LE, Houmard JA. Influence of Maternal Exercise on Glucose and Lipid Metabolism in Offspring Stem Cells: ENHANCED by Mom. J Clin Endocrinol Metab 2022; 107:e3353-e3365. [PMID: 35511592 DOI: 10.1210/clinem/dgac270] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Indexed: 02/06/2023]
Abstract
CONTEXT Recent preclinical data suggest exercise during pregnancy can improve the metabolic phenotype not only of the mother, but of the developing offspring as well. However, investigations in human offspring are lacking. OBJECTIVE To characterize the effect of maternal aerobic exercise on the metabolic phenotype of the offspring's mesenchymal stem cells (MSCs). DESIGN Randomized controlled trial. SETTING Clinical research facility. PATIENTS Healthy female adults between 18 and 35 years of age and ≤ 16 weeks' gestation. INTERVENTION Mothers were randomized into 1 of 2 groups: aerobic exercise (AE, n = 10) or nonexercise control (CTRL, n = 10). The AE group completed 150 minutes of weekly moderate-intensity exercise, according to American College of Sports Medicine guidelines, during pregnancy, whereas controls attended stretching sessions. MAIN OUTCOME MEASURES Following delivery, MSCs were isolated from the umbilical cord of the offspring and metabolic tracer and immunoblotting experiments were completed in the undifferentiated (D0) or myogenically differentiated (D21) state. RESULTS AE-MSCs at D0 had an elevated fold-change over basal in insulin-stimulated glycogen synthesis and reduced nonoxidized glucose metabolite (NOGM) production (P ≤ 0.05). At D21, AE-MSCs had a significant elevation in glucose partitioning toward oxidation (oxidation/NOGM ratio) compared with CTRL (P ≤ 0.05). Immunoblot analysis revealed elevated complex I expression in the AE-MSCs at D21 (P ≤ 0.05). Basal and palmitate-stimulated lipid metabolism was similar between groups at D0 and D21. CONCLUSIONS These data provide evidence of a programmed metabolic phenotype in human offspring with maternal AE during pregnancy.
Collapse
Affiliation(s)
- Alec Chaves
- Department of Kinesiology, East Carolina University, Greenville, NC 27834, USA
- Human Performance Laboratory, East Carolina University, Greenville, NC 27834, USA
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC 27834, USA
| | - Luke A Weyrauch
- Department of Kinesiology, East Carolina University, Greenville, NC 27834, USA
- Human Performance Laboratory, East Carolina University, Greenville, NC 27834, USA
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC 27834, USA
| | - Donghai Zheng
- Department of Kinesiology, East Carolina University, Greenville, NC 27834, USA
- Human Performance Laboratory, East Carolina University, Greenville, NC 27834, USA
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC 27834, USA
| | - Ericka M Biagioni
- Department of Kinesiology, East Carolina University, Greenville, NC 27834, USA
- Human Performance Laboratory, East Carolina University, Greenville, NC 27834, USA
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC 27834, USA
| | - Polina M Krassovskaia
- Department of Kinesiology, East Carolina University, Greenville, NC 27834, USA
- Human Performance Laboratory, East Carolina University, Greenville, NC 27834, USA
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC 27834, USA
| | - Breanna L Davidson
- Department of Kinesiology, East Carolina University, Greenville, NC 27834, USA
- Human Performance Laboratory, East Carolina University, Greenville, NC 27834, USA
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC 27834, USA
| | - Nicholas T Broskey
- Department of Kinesiology, East Carolina University, Greenville, NC 27834, USA
- Human Performance Laboratory, East Carolina University, Greenville, NC 27834, USA
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC 27834, USA
| | - Kristen E Boyle
- The Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, Aurora, CO 80045, USA
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Linda E May
- Department of Kinesiology, East Carolina University, Greenville, NC 27834, USA
- Human Performance Laboratory, East Carolina University, Greenville, NC 27834, USA
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC 27834, USA
| | - Joseph A Houmard
- Department of Kinesiology, East Carolina University, Greenville, NC 27834, USA
- Human Performance Laboratory, East Carolina University, Greenville, NC 27834, USA
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC 27834, USA
| |
Collapse
|
25
|
Therapeutic Strategy of Mesenchymal-Stem-Cell-Derived Extracellular Vesicles as Regenerative Medicine. Int J Mol Sci 2022; 23:ijms23126480. [PMID: 35742923 PMCID: PMC9224400 DOI: 10.3390/ijms23126480] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 06/07/2022] [Accepted: 06/08/2022] [Indexed: 12/11/2022] Open
Abstract
Extracellular vesicles (EVs) are lipid bilayer membrane particles that play critical roles in intracellular communication through EV-encapsulated informative content, including proteins, lipids, and nucleic acids. Mesenchymal stem cells (MSCs) are pluripotent stem cells with self-renewal ability derived from bone marrow, fat, umbilical cord, menstruation blood, pulp, etc., which they use to induce tissue regeneration by their direct recruitment into injured tissues, including the heart, liver, lung, kidney, etc., or secreting factors, such as vascular endothelial growth factor or insulin-like growth factor. Recently, MSC-derived EVs have been shown to have regenerative effects against various diseases, partially due to the post-transcriptional regulation of target genes by miRNAs. Furthermore, EVs have garnered attention as novel drug delivery systems, because they can specially encapsulate various target molecules. In this review, we summarize the regenerative effects and molecular mechanisms of MSC-derived EVs.
Collapse
|
26
|
Fibrous Protein Composite Scaffolds (3D) for Tissue Regeneration: An in vitro Study on Skeletal Muscle Regeneration. Colloids Surf B Biointerfaces 2022; 217:112656. [DOI: 10.1016/j.colsurfb.2022.112656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 06/04/2022] [Accepted: 06/21/2022] [Indexed: 11/15/2022]
|
27
|
Logan A, Nagy Z, Barnes NM, Belli A, Di Pietro V, Tavazzi B, Lazzarino G, Lazzarino G, Bruce L, Persson LI. A phase II open label clinical study of the safety, tolerability and efficacy of ILB® for Amyotrophic Lateral Sclerosis. PLoS One 2022; 17:e0267183. [PMID: 35613082 PMCID: PMC9132272 DOI: 10.1371/journal.pone.0267183] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 03/04/2022] [Indexed: 12/23/2022] Open
Abstract
Introduction Amyotrophic lateral sclerosis (ALS) is an invariably lethal progressive disease, causing degeneration of neurons and muscle. No current treatment halts or reverses disease advance. This single arm, open label, clinical trial in patients with ALS investigated the safety and tolerability of a novel modified low molecular weight dextran sulphate (LMW-DS, named ILB®) previously proven safe for use in healthy volunteers and shown to exert potent neurotrophic effects in pre-clinical studies. Secondary endpoints relate to efficacy and exploratory biomarkers. Methods Thirteen patients with ALS were treated with 5 weekly subcutaneous injections of ILB®. Safety and efficacy outcome measures were recorded weekly during treatment and at regular intervals for a further 70 days. Functional and laboratory biomarkers were assessed before, during and after treatment. Results No deaths, serious adverse events or participant withdrawals occurred during or after ILB® treatment and no significant drug-related changes in blood safety markers were evident, demonstrating safety and tolerability of the drug in this cohort of patients with ALS. The PK of ILB® in patients with ALS was similar to that seen in healthy controls. The ILB® injection elicited a transient elevation of plasma Hepatocyte Growth Factor, a neurotrophic and myogenic growth factor. Following the ILB® injections patients reported increased vitality, decreased spasticity and increased mobility. The ALSFRS-R rating improved from 36.31 ± 6.66 to 38.77 ± 6.44 and the Norris rating also improved from 70.61 ± 13.91 to 77.85 ± 14.24 by Day 36. The improvement of functions was associated with a decrease in muscle atrophy biomarkers. These therapeutic benefits decreased 3–4 weeks after the last dosage. Conclusions This pilot clinical study demonstrates safety and tolerability of ILB® in patients with ALS. The exploratory biomarker and functional measures must be cautiously interpreted but suggest clinical benefit and have a bearing on the mechanism of action of ILB®. The results support the drug’s potential as the first disease modifying treatment for patients with ALS. Trial registration EudraCT 2017-005065-47.
Collapse
Affiliation(s)
- Ann Logan
- Axolotl Consulting Ltd, Droitwich, United Kingdom
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, United Kingdom
- * E-mail:
| | - Zsuzsanna Nagy
- College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Nicholas M. Barnes
- College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Antonio Belli
- College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Valentina Di Pietro
- College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Barbara Tavazzi
- Department of Basic Biotechnological Sciences, Intensive and Perioperative Clinics, Catholic University of Rome, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Giuseppe Lazzarino
- Department of Biomedical and Biotechnological Sciences, Division of Medical Biochemistry, University of Catania, Catania, Italy
| | - Giacomo Lazzarino
- UniCamillus, Saint Camillus International University of Health Sciences, Rome, Italy
| | | | - Lennart I. Persson
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
28
|
Fan J, Abedi-Dorcheh K, Sadat Vaziri A, Kazemi-Aghdam F, Rafieyan S, Sohrabinejad M, Ghorbani M, Rastegar Adib F, Ghasemi Z, Klavins K, Jahed V. A Review of Recent Advances in Natural Polymer-Based Scaffolds for Musculoskeletal Tissue Engineering. Polymers (Basel) 2022; 14:polym14102097. [PMID: 35631979 PMCID: PMC9145843 DOI: 10.3390/polym14102097] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 05/09/2022] [Accepted: 05/17/2022] [Indexed: 02/01/2023] Open
Abstract
The musculoskeletal (MS) system consists of bone, cartilage, tendon, ligament, and skeletal muscle, which forms the basic framework of the human body. This system plays a vital role in appropriate body functions, including movement, the protection of internal organs, support, hematopoiesis, and postural stability. Therefore, it is understandable that the damage or loss of MS tissues significantly reduces the quality of life and limits mobility. Tissue engineering and its applications in the healthcare industry have been rapidly growing over the past few decades. Tissue engineering has made significant contributions toward developing new therapeutic strategies for the treatment of MS defects and relevant disease. Among various biomaterials used for tissue engineering, natural polymers offer superior properties that promote optimal cell interaction and desired biological function. Natural polymers have similarity with the native ECM, including enzymatic degradation, bio-resorb and non-toxic degradation products, ability to conjugate with various agents, and high chemical versatility, biocompatibility, and bioactivity that promote optimal cell interaction and desired biological functions. This review summarizes recent advances in applying natural-based scaffolds for musculoskeletal tissue engineering.
Collapse
Affiliation(s)
- Jingzhi Fan
- Rudolfs Cimdins Riga Biomaterials Innovations and Development Centre of RTU, Institute of General Chemical Engineering, Faculty of Materials Science and Applied Chemistry, Riga Technical University, Pulka St 3, LV-1007 Riga, Latvia;
- Baltic Biomaterials Centre of Excellence, Headquarters at Riga Technical University, Pulka St 3, LV-1007 Riga, Latvia
| | - Keyvan Abedi-Dorcheh
- Biomedical Engineering Division, Faculty of Chemical Engineering, Tarbiat Modares University, Tehran 14117-13116, Iran; (K.A.-D.); (A.S.V.); (F.K.-A.); (S.R.); (M.S.); (M.G.); (F.R.A.); (Z.G.)
| | - Asma Sadat Vaziri
- Biomedical Engineering Division, Faculty of Chemical Engineering, Tarbiat Modares University, Tehran 14117-13116, Iran; (K.A.-D.); (A.S.V.); (F.K.-A.); (S.R.); (M.S.); (M.G.); (F.R.A.); (Z.G.)
| | - Fereshteh Kazemi-Aghdam
- Biomedical Engineering Division, Faculty of Chemical Engineering, Tarbiat Modares University, Tehran 14117-13116, Iran; (K.A.-D.); (A.S.V.); (F.K.-A.); (S.R.); (M.S.); (M.G.); (F.R.A.); (Z.G.)
| | - Saeed Rafieyan
- Biomedical Engineering Division, Faculty of Chemical Engineering, Tarbiat Modares University, Tehran 14117-13116, Iran; (K.A.-D.); (A.S.V.); (F.K.-A.); (S.R.); (M.S.); (M.G.); (F.R.A.); (Z.G.)
| | - Masoume Sohrabinejad
- Biomedical Engineering Division, Faculty of Chemical Engineering, Tarbiat Modares University, Tehran 14117-13116, Iran; (K.A.-D.); (A.S.V.); (F.K.-A.); (S.R.); (M.S.); (M.G.); (F.R.A.); (Z.G.)
| | - Mina Ghorbani
- Biomedical Engineering Division, Faculty of Chemical Engineering, Tarbiat Modares University, Tehran 14117-13116, Iran; (K.A.-D.); (A.S.V.); (F.K.-A.); (S.R.); (M.S.); (M.G.); (F.R.A.); (Z.G.)
| | - Fatemeh Rastegar Adib
- Biomedical Engineering Division, Faculty of Chemical Engineering, Tarbiat Modares University, Tehran 14117-13116, Iran; (K.A.-D.); (A.S.V.); (F.K.-A.); (S.R.); (M.S.); (M.G.); (F.R.A.); (Z.G.)
| | - Zahra Ghasemi
- Biomedical Engineering Division, Faculty of Chemical Engineering, Tarbiat Modares University, Tehran 14117-13116, Iran; (K.A.-D.); (A.S.V.); (F.K.-A.); (S.R.); (M.S.); (M.G.); (F.R.A.); (Z.G.)
| | - Kristaps Klavins
- Rudolfs Cimdins Riga Biomaterials Innovations and Development Centre of RTU, Institute of General Chemical Engineering, Faculty of Materials Science and Applied Chemistry, Riga Technical University, Pulka St 3, LV-1007 Riga, Latvia;
- Baltic Biomaterials Centre of Excellence, Headquarters at Riga Technical University, Pulka St 3, LV-1007 Riga, Latvia
- Correspondence: (K.K.); (V.J.)
| | - Vahid Jahed
- Rudolfs Cimdins Riga Biomaterials Innovations and Development Centre of RTU, Institute of General Chemical Engineering, Faculty of Materials Science and Applied Chemistry, Riga Technical University, Pulka St 3, LV-1007 Riga, Latvia;
- Baltic Biomaterials Centre of Excellence, Headquarters at Riga Technical University, Pulka St 3, LV-1007 Riga, Latvia
- Correspondence: (K.K.); (V.J.)
| |
Collapse
|
29
|
Carotenuto F, Politi S, Ul Haq A, De Matteis F, Tamburri E, Terranova ML, Teodori L, Pasquo A, Di Nardo P. From Soft to Hard Biomimetic Materials: Tuning Micro/Nano-Architecture of Scaffolds for Tissue Regeneration. MICROMACHINES 2022; 13:mi13050780. [PMID: 35630247 PMCID: PMC9144100 DOI: 10.3390/mi13050780] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 03/30/2022] [Accepted: 05/13/2022] [Indexed: 11/23/2022]
Abstract
Failure of tissues and organs resulting from degenerative diseases or trauma has caused huge economic and health concerns around the world. Tissue engineering represents the only possibility to revert this scenario owing to its potential to regenerate or replace damaged tissues and organs. In a regeneration strategy, biomaterials play a key role promoting new tissue formation by providing adequate space for cell accommodation and appropriate biochemical and biophysical cues to support cell proliferation and differentiation. Among other physical cues, the architectural features of the biomaterial as a kind of instructive stimuli can influence cellular behaviors and guide cells towards a specific tissue organization. Thus, the optimization of biomaterial micro/nano architecture, through different manufacturing techniques, is a crucial strategy for a successful regenerative therapy. Over the last decades, many micro/nanostructured biomaterials have been developed to mimic the defined structure of ECM of various soft and hard tissues. This review intends to provide an overview of the relevant studies on micro/nanostructured scaffolds created for soft and hard tissue regeneration and highlights their biological effects, with a particular focus on striated muscle, cartilage, and bone tissue engineering applications.
Collapse
Affiliation(s)
- Felicia Carotenuto
- Dipartimento di Scienze Cliniche e Medicina Traslazionale, Università Degli Studi di Roma “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy;
- Department of Fusion and Technologies for Nuclear Safety and Security, Diagnostic and Metrology (FSN-TECFIS-DIM), ENEA, CR Frascati, 00044 Rome, Italy; (S.P.); (L.T.); (A.P.)
- Centro di Ricerca Interdipartimentale di Medicina Rigenerativa (CIMER), Università Degli Studi di Roma “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy; (F.D.M.); (E.T.); (M.L.T.)
- Correspondence: (F.C.); (P.D.N.)
| | - Sara Politi
- Department of Fusion and Technologies for Nuclear Safety and Security, Diagnostic and Metrology (FSN-TECFIS-DIM), ENEA, CR Frascati, 00044 Rome, Italy; (S.P.); (L.T.); (A.P.)
- Dipartimento di Scienze e Tecnologie Chimiche, Università Degli Studi di Roma “Tor Vergata”, Via della Ricerca Scientifica, 00133 Rome, Italy
| | - Arsalan Ul Haq
- Dipartimento di Scienze Cliniche e Medicina Traslazionale, Università Degli Studi di Roma “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy;
- Centro di Ricerca Interdipartimentale di Medicina Rigenerativa (CIMER), Università Degli Studi di Roma “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy; (F.D.M.); (E.T.); (M.L.T.)
| | - Fabio De Matteis
- Centro di Ricerca Interdipartimentale di Medicina Rigenerativa (CIMER), Università Degli Studi di Roma “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy; (F.D.M.); (E.T.); (M.L.T.)
- Dipartimento Ingegneria Industriale, Università Degli Studi di Roma “Tor Vergata”, Via del Politecnico, 00133 Roma, Italy
| | - Emanuela Tamburri
- Centro di Ricerca Interdipartimentale di Medicina Rigenerativa (CIMER), Università Degli Studi di Roma “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy; (F.D.M.); (E.T.); (M.L.T.)
- Dipartimento di Scienze e Tecnologie Chimiche, Università Degli Studi di Roma “Tor Vergata”, Via della Ricerca Scientifica, 00133 Rome, Italy
| | - Maria Letizia Terranova
- Centro di Ricerca Interdipartimentale di Medicina Rigenerativa (CIMER), Università Degli Studi di Roma “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy; (F.D.M.); (E.T.); (M.L.T.)
- Dipartimento di Scienze e Tecnologie Chimiche, Università Degli Studi di Roma “Tor Vergata”, Via della Ricerca Scientifica, 00133 Rome, Italy
| | - Laura Teodori
- Department of Fusion and Technologies for Nuclear Safety and Security, Diagnostic and Metrology (FSN-TECFIS-DIM), ENEA, CR Frascati, 00044 Rome, Italy; (S.P.); (L.T.); (A.P.)
- Centro di Ricerca Interdipartimentale di Medicina Rigenerativa (CIMER), Università Degli Studi di Roma “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy; (F.D.M.); (E.T.); (M.L.T.)
| | - Alessandra Pasquo
- Department of Fusion and Technologies for Nuclear Safety and Security, Diagnostic and Metrology (FSN-TECFIS-DIM), ENEA, CR Frascati, 00044 Rome, Italy; (S.P.); (L.T.); (A.P.)
| | - Paolo Di Nardo
- Dipartimento di Scienze Cliniche e Medicina Traslazionale, Università Degli Studi di Roma “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy;
- Centro di Ricerca Interdipartimentale di Medicina Rigenerativa (CIMER), Università Degli Studi di Roma “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy; (F.D.M.); (E.T.); (M.L.T.)
- Correspondence: (F.C.); (P.D.N.)
| |
Collapse
|
30
|
Schwann Cells Promote Myogenic Differentiation of Myoblasts and Adipogenic Mesenchymal Stromal Cells on Poly-ɛ-Caprolactone-Collagen I-Nanofibers. Cells 2022; 11:cells11091436. [PMID: 35563742 PMCID: PMC9100029 DOI: 10.3390/cells11091436] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 04/16/2022] [Accepted: 04/20/2022] [Indexed: 02/07/2023] Open
Abstract
For the purpose of skeletal muscle tissue engineering, different cell types have been investigated regarding their myogenic differentiation potential, including co-cultured myoblasts and adipogenic mesenchymal stromal cells (Mb/ADSC). As neural cells enhance synaptic junction formation, the aim of this study was to co-culture Schwann cells (SCs) with Mb/ADSC on biocompatible electrospun aligned poly-ε-polycaprolacton (PCL)-collagen I-nanofibers. It was hypothesized that SCs, as part of the peripheral nervous system, promote the myogenic differentiation of Mb/ADSC co-cultures. Mb/ADSC were compared to Mb/ADSC/SC regarding their capacity for myogenic differentiation via immunofluorescent staining and gene expression of myogenic markers. Mb/ADSC/SC showed more myotubes after 28 days of differentiation (p ≤ 0.05). After 28 days of differentiation on electrospun aligned PCL-collagen I-nanofibers, gene expression of myosin heavy chains (MYH2) and myogenin (MYOG) was upregulated in Mb/ADSC/SC compared to Mb/ADSC (p ≤ 0.01 and p ≤ 0.05, respectively). Immunofluorescent staining for MHC showed highly aligned multinucleated cells as possible myotube formation in Mb/ADSC/SC. In conclusion, SCs promote myogenic differentiation of Mb/ADSC. The co-culture of primary Mb/ADSC/SC on PCL-collagen I-nanofibers serves as a physiological model for skeletal muscle tissue engineering, applicable to future clinical applications.
Collapse
|
31
|
Cai A, Zheng Z, Müller-Seubert W, Biggemann J, Fey T, Beier JP, Horch RE, Frieß B, Arkudas A. Microsurgical Transplantation of Pedicled Muscles in an Isolation Chamber—A Novel Approach to Engineering Muscle Constructs via Perfusion-Decellularization. J Pers Med 2022; 12:jpm12030442. [PMID: 35330443 PMCID: PMC8951001 DOI: 10.3390/jpm12030442] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 03/07/2022] [Accepted: 03/09/2022] [Indexed: 02/06/2023] Open
Abstract
Decellularized whole muscle constructs represent an ideal scaffold for muscle tissue engineering means as they retain the network and proteins of the extracellular matrix of skeletal muscle tissue. The presence of a vascular pedicle enables a more efficient perfusion-based decellularization protocol and allows for subsequent recellularization and transplantation of the muscle construct in vivo. The goal of this study was to create a baseline for transplantation of decellularized whole muscle constructs by establishing an animal model for investigating a complete native muscle isolated on its pedicle in terms of vascularization and functionality. The left medial gastrocnemius muscles of 5 male Lewis rats were prepared and raised from their beds for in situ muscle stimulation. The stimulation protocol included twitches, tetanic stimulation, fatigue testing, and stretching of the muscles. Peak force, maximum rate of contraction and relaxation, time to maximum contraction and relaxation, and maximum contraction and relaxation rate were determined. Afterwards, muscles were explanted and transplanted heterotopically in syngeneic rats in an isolation chamber by microvascular anastomosis. After 2 weeks, transplanted gastrocnemius muscles were exposed and stimulated again followed by intravascular perfusion with a contrast agent for µCT analysis. Muscle constructs were then paraffin embedded for immunohistological staining. Peak twitch and tetanic force values all decreased significantly after muscle transplantation while fatigue index and passive stretch properties did not differ between the two groups. Vascular analysis revealed retained perfused vessels most of which were in a smaller radius range of up to 20 µm and 45 µm. In this study, a novel rat model of heterotopic microvascular muscle transplantation in an isolation chamber was established. With the assessment of in situ muscle contraction properties as well as vessel distribution after 2 weeks of transplantation, this model serves as a base for future studies including the transplantation of perfusion-decellularized muscle constructs.
Collapse
Affiliation(s)
- Aijia Cai
- Department of Plastic and Hand Surgery and Laboratory for Tissue Engineering and Regenerative Medicine, University Hospital of Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Krankenhausstraße 12, 91054 Erlangen, Germany; (Z.Z.); (W.M.-S.); (R.E.H.); (B.F.); (A.A.)
- Correspondence: ; Tel.: +49-9131-85-33296; Fax: +49-9131-85-39327
| | - Zengming Zheng
- Department of Plastic and Hand Surgery and Laboratory for Tissue Engineering and Regenerative Medicine, University Hospital of Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Krankenhausstraße 12, 91054 Erlangen, Germany; (Z.Z.); (W.M.-S.); (R.E.H.); (B.F.); (A.A.)
| | - Wibke Müller-Seubert
- Department of Plastic and Hand Surgery and Laboratory for Tissue Engineering and Regenerative Medicine, University Hospital of Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Krankenhausstraße 12, 91054 Erlangen, Germany; (Z.Z.); (W.M.-S.); (R.E.H.); (B.F.); (A.A.)
| | - Jonas Biggemann
- Department of Materials Science and Engineering, Institute of Glass and Ceramics, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Martensstr. 5, 91058 Erlangen, Germany; (J.B.); (T.F.)
| | - Tobias Fey
- Department of Materials Science and Engineering, Institute of Glass and Ceramics, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Martensstr. 5, 91058 Erlangen, Germany; (J.B.); (T.F.)
- Frontier Research Institute for Materials Science, Nagoya Institute of Technology, Gokiso-cho, Showa-ku, Nagoya 466-8555, Japan
| | - Justus P. Beier
- Department of Plastic Surgery, Hand Surgery, Burn Center, University Hospital RWTH Aachen, Pauwelsstr. 30, 52074 Aachen, Germany;
| | - Raymund E. Horch
- Department of Plastic and Hand Surgery and Laboratory for Tissue Engineering and Regenerative Medicine, University Hospital of Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Krankenhausstraße 12, 91054 Erlangen, Germany; (Z.Z.); (W.M.-S.); (R.E.H.); (B.F.); (A.A.)
| | - Benjamin Frieß
- Department of Plastic and Hand Surgery and Laboratory for Tissue Engineering and Regenerative Medicine, University Hospital of Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Krankenhausstraße 12, 91054 Erlangen, Germany; (Z.Z.); (W.M.-S.); (R.E.H.); (B.F.); (A.A.)
| | - Andreas Arkudas
- Department of Plastic and Hand Surgery and Laboratory for Tissue Engineering and Regenerative Medicine, University Hospital of Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Krankenhausstraße 12, 91054 Erlangen, Germany; (Z.Z.); (W.M.-S.); (R.E.H.); (B.F.); (A.A.)
| |
Collapse
|
32
|
Ding DC, Li PC. Stem-cell therapy in stress urinary incontinence: A review. Tzu Chi Med J 2022. [DOI: 10.4103/tcmj.tcmj_145_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
33
|
Zhang SY, Ren JY, Yang B. Priming strategies for controlling stem cell fate: Applications and challenges in dental tissue regeneration. World J Stem Cells 2021; 13:1625-1646. [PMID: 34909115 PMCID: PMC8641023 DOI: 10.4252/wjsc.v13.i11.1625] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 05/14/2021] [Accepted: 08/27/2021] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stromal cells (MSCs) have attracted intense interest in the field of dental tissue regeneration. Dental tissue is a popular source of MSCs because MSCs can be obtained with minimally invasive procedures. MSCs possess distinct inherent properties of self-renewal, immunomodulation, proangiogenic potential, and multilineage potency, as well as being readily available and easy to culture. However, major issues, including poor engraftment and low survival rates in vivo, remain to be resolved before large-scale application is feasible in clinical treatments. Thus, some recent investigations have sought ways to optimize MSC functions in vitro and in vivo. Currently, priming culture conditions, pretreatment with mechanical and physical stimuli, preconditioning with cytokines and growth factors, and genetic modification of MSCs are considered to be the main strategies; all of which could contribute to improving MSC efficacy in dental regenerative medicine. Research in this field has made tremendous progress and continues to gather interest and stimulate innovation. In this review, we summarize the priming approaches for enhancing the intrinsic biological properties of MSCs such as migration, antiapoptotic effect, proangiogenic potential, and regenerative properties. Challenges in current approaches associated with MSC modification and possible future solutions are also indicated. We aim to outline the present understanding of priming approaches to improve the therapeutic effects of MSCs on dental tissue regeneration.
Collapse
Affiliation(s)
- Si-Yuan Zhang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Jia-Yin Ren
- Department of Oral Radiology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Bo Yang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan Province, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan Province, China
| |
Collapse
|
34
|
Aghali A. Craniofacial Bone Tissue Engineering: Current Approaches and Potential Therapy. Cells 2021; 10:cells10112993. [PMID: 34831216 PMCID: PMC8616509 DOI: 10.3390/cells10112993] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/16/2021] [Accepted: 10/22/2021] [Indexed: 01/10/2023] Open
Abstract
Craniofacial bone defects can result from various disorders, including congenital malformations, tumor resection, infection, severe trauma, and accidents. Successfully regenerating cranial defects is an integral step to restore craniofacial function. However, challenges managing and controlling new bone tissue formation remain. Current advances in tissue engineering and regenerative medicine use innovative techniques to address these challenges. The use of biomaterials, stromal cells, and growth factors have demonstrated promising outcomes in vitro and in vivo. Natural and synthetic bone grafts combined with Mesenchymal Stromal Cells (MSCs) and growth factors have shown encouraging results in regenerating critical-size cranial defects. One of prevalent growth factors is Bone Morphogenetic Protein-2 (BMP-2). BMP-2 is defined as a gold standard growth factor that enhances new bone formation in vitro and in vivo. Recently, emerging evidence suggested that Megakaryocytes (MKs), induced by Thrombopoietin (TPO), show an increase in osteoblast proliferation in vitro and bone mass in vivo. Furthermore, a co-culture study shows mature MKs enhance MSC survival rate while maintaining their phenotype. Therefore, MKs can provide an insight as a potential therapy offering a safe and effective approach to regenerating critical-size cranial defects.
Collapse
Affiliation(s)
- Arbi Aghali
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA;
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47908, USA
| |
Collapse
|
35
|
Takegaki J, Sase K, Kono Y, Nakano D, Fujita T, Konishi S, Fujita S. Intramuscular injection of mesenchymal stem cells activates anabolic and catabolic systems in mouse skeletal muscle. Sci Rep 2021; 11:21224. [PMID: 34707171 PMCID: PMC8551189 DOI: 10.1038/s41598-021-00627-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 10/01/2021] [Indexed: 01/06/2023] Open
Abstract
Skeletal muscle mass is critical for good quality of life. Mesenchymal stem cells (MSCs) are multipotent stem cells distributed across various tissues. They are characterized by the capacity to secrete growth factors and differentiate into skeletal muscle cells. These capabilities suggest that MSCs might be beneficial for muscle growth. Nevertheless, little is known regarding the effects on muscle protein anabolic and catabolic systems of intramuscular injection of MSCs into skeletal muscle. Therefore, in the present study, we measured changes in mechanistic target of rapamycin complex 1 (mTORC1) signaling, the ubiquitin–proteasome system, and autophagy-lysosome system-related factors after a single intramuscular injection of MSCs with green fluorescence protein (GFP) into mouse muscles. The intramuscularly-injected MSCs were retained in the gastrocnemius muscle for 7 days after the injection, indicated by detection of GFP and expression of platelet-derived growth factor receptor-alpha. The injection of MSCs increased the expression of satellite cell-related genes, activated mTORC1 signaling and muscle protein synthesis, and increased protein ubiquitination and autophagosome formation (indicated by the expression of microtubule-associated protein 1 light chain 3-II). These results suggest that the intramuscular injection of MSCs activated muscle anabolic and catabolic systems and accelerated muscle protein turnover.
Collapse
Affiliation(s)
- Junya Takegaki
- Research Organization of Science and Technology, Ritsumeikan University, 1-1-1 Noji-Higashi, Kusatsu, Shiga, 525-8577, Japan.,Ritsumeikan Global Innovation Research Organization, Ritsumeikan University, 1-1-1 Noji-Higashi, Kusatsu, Shiga, 525-8577, Japan
| | - Kohei Sase
- Faculty of Sport and Health Science, Ritsumeikan University, 1-1-1 Noji-Higashi, Kusatsu, Shiga, 525-8577, Japan
| | - Yusuke Kono
- Ritsumeikan Global Innovation Research Organization, Ritsumeikan University, 1-1-1 Noji-Higashi, Kusatsu, Shiga, 525-8577, Japan
| | - Daiki Nakano
- Ritsumeikan Global Innovation Research Organization, Ritsumeikan University, 1-1-1 Noji-Higashi, Kusatsu, Shiga, 525-8577, Japan
| | - Takuya Fujita
- College of Pharmaceutical Sciences, Ritsumeikan University, 1-1-1 Noji-Higashi, Kusatsu, Shiga, 525-8577, Japan
| | - Satoshi Konishi
- Faculty of Science and Engineering, Ritsumeikan University, 1-1-1 Noji-Higashi, Kusatsu, Shiga, 525-8577, Japan
| | - Satoshi Fujita
- Faculty of Sport and Health Science, Ritsumeikan University, 1-1-1 Noji-Higashi, Kusatsu, Shiga, 525-8577, Japan.
| |
Collapse
|
36
|
MacDonald A, Gross A, Jones B, Dhar M. Muscle Regeneration of the Tongue: A review of current clinical and regenerative research strategies. TISSUE ENGINEERING PART B-REVIEWS 2021; 28:1022-1034. [PMID: 34693743 DOI: 10.1089/ten.teb.2021.0133] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Various abnormalities of the tongue, including cancers, commonly require surgical removal to sequester growth and metastasis. However, even minor resections can affect functional outcomes such as speech and swallowing, thereby reducing quality of life. Surgical resections alone create volumetric muscle loss whereby muscle tissue cannot self-regenerate within the tongue. In these cases, the tongue is reconstructed typically in the form of autologous skin flaps. However, flap reconstruction has many limitations and unfortunately is the primary option for oral and reconstructive surgeons to treat tongue defects. The alternative, but yet undeveloped strategy for tongue reconstruction is regenerative medicine, which widely focuses on building new organs with stem cells. Regenerative medicine has successfully treated many tissues, but research has inadequately addressed the tongue as a vital organ in need of tissue engineering. In this review, we address the current standard for tongue reconstruction, the cellular mechanisms of muscle cell development, and the stem cell studies that have attempted muscle engineering within the tongue. Until now, no review has focused on engineering the tongue with regenerative medicine, which could guide innovative strategies for tongue reconstruction.
Collapse
Affiliation(s)
- Amber MacDonald
- The University of Tennessee Knoxville College of Veterinary Medicine, 70737, Large Animal Clinical Sciences, 2407 River Drive, Knoxville, Tennessee, United States, 37996-4539;
| | - Andrew Gross
- The University of Tennessee Medical Center, 21823, Knoxville, Tennessee, United States;
| | - Brady Jones
- The University of Tennessee Medical Center, 21823, Knoxville, Tennessee, United States;
| | - Madhu Dhar
- University of Tennessee Knoxville College of Veterinary Medicine, 70737, Large Animal Clinical Sciences, College of Veterinary Medicine, 2407 River Drive, Knoxville, Tennessee, United States, 37996.,University of Tennessee;
| |
Collapse
|
37
|
Advancing Regenerative Medicine Through the Development of Scaffold, Cell Biology, Biomaterials and Strategies of Smart Material. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2021. [DOI: 10.1007/s40883-021-00227-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
38
|
Peper S, Vo T, Ahuja N, Awad K, Mikos AG, Varanasi V. Bioprinted nanocomposite hydrogels: A proposed approach to functional restoration of skeletal muscle and vascular tissue following volumetric muscle loss. Curr Opin Pharmacol 2021; 58:35-43. [PMID: 33853025 PMCID: PMC8718378 DOI: 10.1016/j.coph.2021.03.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 02/27/2021] [Accepted: 03/11/2021] [Indexed: 01/03/2023]
Abstract
Musculoskeletal conditions are the highest contributor to global disability, accounting for 16% of all ages lived with disability. Volumetric muscle loss (VML) is classified as significant damage to skeletal muscle compartments and motor units, leading to significant tissue loss, functional deficits, and long-term disability. In this review, the current tissue engineering approaches in terms of fabrication techniques, materials, cell sources, and growth factors for enhanced angiogenesis and neuromuscular junction (NMJ) in VML repair, are discussed. Review of results recently published in the literature suggested that bioprinted nanocomposite hydrogels (NC gels) seeded with adult muscle progenitor cells that promote secretion of endogenous vascular growth factors have potential applications in promoting skeletal muscle regeneration, revascularization, and NMJ repair (Figure 1). Despite recent advancements, future research is needed on NC gels and the complex processes underlying vascular infiltration and NMJ repair in VML injuries.
Collapse
Affiliation(s)
- Sara Peper
- Bone Muscle Research Center, College of Nursing & Health Innovation, The University of Texas at Arlington, 701 South Nedderman Drive, Arlington, TX, 76019, USA; Department of Bioengineering, College of Engineering, The University of Texas at Arlington, 701 South Nedderman Drive, Box 19138, Arlington, TX, 76019, USA
| | - Thy Vo
- Bone Muscle Research Center, College of Nursing & Health Innovation, The University of Texas at Arlington, 701 South Nedderman Drive, Arlington, TX, 76019, USA; Department of Kinesiology, College of Nursing & Health Innovation, The University of Texas at Arlington, 411 South Nedderman Drive, Box 19407, Arlington, TX, 76019, USA
| | - Neelam Ahuja
- Bone Muscle Research Center, College of Nursing & Health Innovation, The University of Texas at Arlington, 701 South Nedderman Drive, Arlington, TX, 76019, USA; Department of Kinesiology, College of Nursing & Health Innovation, The University of Texas at Arlington, 411 South Nedderman Drive, Box 19407, Arlington, TX, 76019, USA
| | - Kamal Awad
- Bone Muscle Research Center, College of Nursing & Health Innovation, The University of Texas at Arlington, 701 South Nedderman Drive, Arlington, TX, 76019, USA; Department of Materials Science & Engineering, College of Engineering, The University of Texas at Arlington, 701 South Nedderman Drive, Box 19138, Arlington, TX, 76019 & National Research Center, 12622, Egypt
| | - Antonios G Mikos
- Center for Engineering Complex Tissues, Center for Excellence in Tissue Engineering, J.W. Cox Laboratory for Biomedical Engineering, Rice University, P.O. Box 1892, Houston, TX, 77251, USA
| | - Venu Varanasi
- Bone Muscle Research Center, College of Nursing & Health Innovation, The University of Texas at Arlington, 701 South Nedderman Drive, Arlington, TX, 76019, USA; Department of Nursing, College of Nursing & Health Innovation, The University of Texas at Arlington, 411 South Nedderman Drive Box 19407, Arlington, TX, 76019, USA.
| |
Collapse
|
39
|
Li M, Zhao Q, Belloli R, Duffy CR, Cai HN. Insulator foci distance correlates with cellular and nuclear morphology in early Drosophila embryos. Dev Biol 2021; 476:189-199. [PMID: 33844976 DOI: 10.1016/j.ydbio.2021.03.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 02/16/2021] [Accepted: 03/26/2021] [Indexed: 11/25/2022]
Abstract
The three-dimensional (3D) organization of the genome is highly dynamic, changing during development and varying across different tissues and cell types. Recent studies indicate that these changes alter regulatory interactions, leading to changes in gene expression. Despite its importance, the mechanisms that influence genomic organization remain poorly understood. We have previously identified a network of chromatin boundary elements, or insulators, in the Drosophila Antennapedia homeotic complex (ANT-C). These genomic elements interact with one another to tether chromatin loops that could block or promote enhancer-promoter interactions. To understand the function of these insulators, we assessed their interactions by measuring their 3D nuclear distance in developing animal tissues. Our data suggest that the ANT-C Hox complex might be in a folded or looped configuration rather than in a random or extended form. The architecture of the ANT-C complex, as read out by the pair-wise distance between insulators, undergoes a strong compression during late embryogenesis, coinciding with the reduction of cell and nuclear diameters due to continued cell divisions in post-cleavage cells. Our results suggest that genomic architecture and gene regulation may be influenced by cellular morphology and movement during development.
Collapse
Affiliation(s)
- Mo Li
- Department of Cellular Biology, University of Georgia, Athens GA, 30602, USA
| | - Qing Zhao
- Department of Cellular Biology, University of Georgia, Athens GA, 30602, USA
| | - Ryan Belloli
- Department of Cellular Biology, University of Georgia, Athens GA, 30602, USA
| | - Carly R Duffy
- Department of Cellular Biology, University of Georgia, Athens GA, 30602, USA
| | - Haini N Cai
- Department of Cellular Biology, University of Georgia, Athens GA, 30602, USA.
| |
Collapse
|
40
|
Choi JS, Lee MS, Kim J, Eom MR, Jeong EJ, Lee M, Park SA, Jeong JH, Kwon SK. Hyaluronic Acid Coating on Hydrophobic Tracheal Scaffold Enhances Mesenchymal Stem Cell Adhesion and Tracheal Regeneration. Tissue Eng Regen Med 2021; 18:225-233. [PMID: 33765289 PMCID: PMC8012419 DOI: 10.1007/s13770-021-00335-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Revised: 02/05/2021] [Accepted: 02/19/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Long segmental tracheal repair is challenging in regenerative medicine due to low adhesion of stem cells to tracheal scaffolds. Optimal transplantation of stem cells for tracheal defects has not been established. We evaluated the role of hyaluronic acid (HA) coating of tracheal scaffolds in mesenchymal stem cell (MSC) adhesion and tracheal regeneration in a rabbit model. METHODS A three-dimensionally printed tubular tracheal prosthesis was incubated with dopa-HA-fluorescein isothiocyanate in phosphate-buffered saline for 2 days. MSCs were incubated with an HA-coated scaffold, and their adhesion was evaluated by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. HA coated scaffolds with or without MSC seeding were transplanted at the circumferential tracheal defect in rabbits, and survival, rigid bronchoscopy, radiologic findings, and histologic findings were compared between the two groups. RESULTS HA-coated scaffolds showed better MSC adhesion than non-coated scaffolds. The HA-coated scaffolds with MSC group showed a wider airway and greater mucosal regeneration compared to the HA-coated scaffolds without MSC group. CONCLUSION HA coating of scaffolds can promote MSC adhesion and tracheal regeneration.
Collapse
Affiliation(s)
- Ji Suk Choi
- Department of Otorhinolaryngology-Head and Neck Surgery, Biomedical Research Institute, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu Seoul, 03080, Republic of Korea
| | - Min Sang Lee
- School of Pharmacy, Theranostic Macromolecules Research Center, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, 16419, Republic of Korea
| | - Jooyoung Kim
- Department of Otorhinolaryngology-Head and Neck Surgery, Biomedical Research Institute, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu Seoul, 03080, Republic of Korea
| | - Min Rye Eom
- Department of Otorhinolaryngology-Head and Neck Surgery, Biomedical Research Institute, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu Seoul, 03080, Republic of Korea
| | - Eun Ji Jeong
- Department of Otorhinolaryngology-Head and Neck Surgery, Biomedical Research Institute, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu Seoul, 03080, Republic of Korea
| | - Minhyung Lee
- Department of Otorhinolaryngology-Head and Neck Surgery, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu Seoul, 03080, Republic of Korea
- Department of Otorhinolaryngology-Head and Neck Surgery, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu Seoul, 03080, Republic of Korea
| | - Su A Park
- Department of Nature-Inspired Nanoconvergence Systems, Korea Institute of Machinery and Materials, 156 Gajeongbuk-ro, Yuseong-gu, Daejeon, 34103, Republic of Korea
| | - Ji Hoon Jeong
- School of Pharmacy, Theranostic Macromolecules Research Center, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, 16419, Republic of Korea.
| | - Seong Keun Kwon
- Department of Otorhinolaryngology-Head and Neck Surgery, Biomedical Research Institute, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu Seoul, 03080, Republic of Korea.
- Department of Otorhinolaryngology-Head and Neck Surgery, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu Seoul, 03080, Republic of Korea.
- Department of Otorhinolaryngology-Head and Neck Surgery, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu Seoul, 03080, Republic of Korea.
- Cancer Research Institute, Seoul National University, 101 Daehak-ro, Jongno-gu Seoul, 03080, Republic of Korea.
| |
Collapse
|
41
|
Wang D, Zhang X, Huang S, Liu Y, Fu BSC, Mak KKL, Blocki AM, Yung PSH, Tuan RS, Ker DFE. Engineering multi-tissue units for regenerative Medicine: Bone-tendon-muscle units of the rotator cuff. Biomaterials 2021; 272:120789. [PMID: 33845368 DOI: 10.1016/j.biomaterials.2021.120789] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 03/25/2021] [Accepted: 03/26/2021] [Indexed: 12/13/2022]
Abstract
Our body systems are comprised of numerous multi-tissue units. For the musculoskeletal system, one of the predominant functional units is comprised of bone, tendon/ligament, and muscle tissues working in tandem to facilitate locomotion. To successfully treat musculoskeletal injuries and diseases, critical consideration and thoughtful integration of clinical, biological, and engineering aspects are necessary to achieve translational bench-to-bedside research. In particular, identifying ideal biomaterial design specifications, understanding prior and recent tissue engineering advances, and judicious application of biomaterial and fabrication technologies will be crucial for addressing current clinical challenges in engineering multi-tissue units. Using rotator cuff tears as an example, insights relevant for engineering a bone-tendon-muscle multi-tissue unit are presented. This review highlights the tissue engineering strategies for musculoskeletal repair and regeneration with implications for other bone-tendon-muscle units, their derivatives, and analogous non-musculoskeletal tissue structures.
Collapse
Affiliation(s)
- Dan Wang
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR; School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR; Ministry of Education Key Laboratory for Regenerative Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR; Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR
| | - Xu Zhang
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR; School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR
| | - Shuting Huang
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR; School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR
| | - Yang Liu
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR
| | - Bruma Sai-Chuen Fu
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR; Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR
| | | | - Anna Maria Blocki
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR; School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR; Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR
| | - Patrick Shu-Hang Yung
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR; Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR
| | - Rocky S Tuan
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR; School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR
| | - Dai Fei Elmer Ker
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR; School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR; Ministry of Education Key Laboratory for Regenerative Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR; Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR.
| |
Collapse
|
42
|
Vaghela R, Arkudas A, Horch RE, Hessenauer M. Actually Seeing What Is Going on - Intravital Microscopy in Tissue Engineering. Front Bioeng Biotechnol 2021; 9:627462. [PMID: 33681162 PMCID: PMC7925911 DOI: 10.3389/fbioe.2021.627462] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 01/26/2021] [Indexed: 12/21/2022] Open
Abstract
Intravital microscopy (IVM) study approach offers several advantages over in vitro, ex vivo, and 3D models. IVM provides real-time imaging of cellular events, which provides us a comprehensive picture of dynamic processes. Rapid improvement in microscopy techniques has permitted deep tissue imaging at a higher resolution. Advances in fluorescence tagging methods enable tracking of specific cell types. Moreover, IVM can serve as an important tool to study different stages of tissue regeneration processes. Furthermore, the compatibility of different tissue engineered constructs can be analyzed. IVM is also a promising approach to investigate host reactions on implanted biomaterials. IVM can provide instant feedback for improvising tissue engineering strategies. In this review, we aim to provide an overview of the requirements and applications of different IVM approaches. First, we will discuss the history of IVM development, and then we will provide an overview of available optical modalities including the pros and cons. Later, we will summarize different fluorescence labeling methods. In the final section, we will discuss well-established chronic and acute IVM models for different organs.
Collapse
Affiliation(s)
- Ravikumar Vaghela
- Department of Plastic and Hand Surgery, University Hospital of Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Andreas Arkudas
- Department of Plastic and Hand Surgery, University Hospital of Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Raymund E Horch
- Department of Plastic and Hand Surgery, University Hospital of Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Maximilian Hessenauer
- Department of Plastic and Hand Surgery, University Hospital of Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| |
Collapse
|
43
|
Yu D, Cai Z, Li D, Zhang Y, He M, Yang Y, Liu D, Xie W, Li Y, Xiao W. Myogenic Differentiation of Stem Cells for Skeletal Muscle Regeneration. Stem Cells Int 2021; 2021:8884283. [PMID: 33628275 PMCID: PMC7884123 DOI: 10.1155/2021/8884283] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 12/22/2020] [Accepted: 01/08/2021] [Indexed: 12/11/2022] Open
Abstract
Stem cells have become a hot research topic in the field of regenerative medicine due to their self-renewal and differentiation capabilities. Skeletal muscle tissue is one of the most important tissues in the human body, and it is difficult to recover when severely damaged. However, conventional treatment methods can cause great pain to patients. Stem cell-based tissue engineering can repair skeletal muscle to the greatest extent with little damage. Therefore, the application of stem cells to skeletal muscle regeneration is very promising. In this review, we discuss scaffolds and stem cells for skeletal muscle regeneration and put forward our ideas for future development.
Collapse
Affiliation(s)
- Dengjie Yu
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410008 Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008 Hunan, China
| | - Zijun Cai
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410008 Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008 Hunan, China
| | - Daishi Li
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yi Zhang
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410008 Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008 Hunan, China
| | - Miao He
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410008 Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008 Hunan, China
| | - Yuntao Yang
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410008 Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008 Hunan, China
| | - Di Liu
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410008 Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008 Hunan, China
| | - Wenqing Xie
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410008 Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008 Hunan, China
| | - Yusheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410008 Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008 Hunan, China
| | - Wenfeng Xiao
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410008 Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008 Hunan, China
| |
Collapse
|
44
|
Guan F, Wu J, Li J, Liu S, Weng Y, Chen T, Yang T, Fang K. RETRACTED ARTICLE: Inducible nitric oxide synthase promotes differentiation of satellite cells and prevents stress urinary incontinence via HGF-mediated p38/MAPK signaling. World J Urol 2021; 39:633. [PMID: 32556674 DOI: 10.1007/s00345-020-03289-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Accepted: 05/28/2020] [Indexed: 01/06/2023] Open
Affiliation(s)
- Fei Guan
- Department of Urology, The 2nd Hospital of Kunming Medical University, No. 374, Dianmian AvenueYunnan Province, Kunming, 650101, People's Republic of China
- The 4th Hospital of Changde, Changde, 415000, People's Republic of China
| | - Jing Wu
- Department of Biochemistry and Molecular Biology, The Primary Medicine School of Kunming Medical University, Kunming, 650500, People's Republic of China
| | - Jiang Li
- Department of Urology, The 1st Hospital of Qujing, Qujing, 650500, People's Republic of China
| | - Shang Liu
- Department of Urology, The 2nd Hospital of Kunming Medical University, No. 374, Dianmian AvenueYunnan Province, Kunming, 650101, People's Republic of China
| | - Yuting Weng
- Department of Urology, The 2nd Hospital of Kunming Medical University, No. 374, Dianmian AvenueYunnan Province, Kunming, 650101, People's Republic of China
| | - Tao Chen
- Department of Urology, The 2nd Hospital of Kunming Medical University, No. 374, Dianmian AvenueYunnan Province, Kunming, 650101, People's Republic of China
| | - Tongxin Yang
- Department of Urology, The 2nd Hospital of Kunming Medical University, No. 374, Dianmian AvenueYunnan Province, Kunming, 650101, People's Republic of China
| | - Kewei Fang
- Department of Urology, The 2nd Hospital of Kunming Medical University, No. 374, Dianmian AvenueYunnan Province, Kunming, 650101, People's Republic of China.
| |
Collapse
|
45
|
Wang S, Hashemi S, Stratton S, Arinzeh TL. The Effect of Physical Cues of Biomaterial Scaffolds on Stem Cell Behavior. Adv Healthc Mater 2021; 10:e2001244. [PMID: 33274860 DOI: 10.1002/adhm.202001244] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 10/09/2020] [Indexed: 02/06/2023]
Abstract
Stem cells have been sought as a promising cell source in the tissue engineering field due to their proliferative capacity as well as differentiation potential. Biomaterials have been utilized to facilitate the delivery of stem cells in order to improve their engraftment and long-term viability upon implantation. Biomaterials also have been developed as scaffolds to promote stem cell induced tissue regeneration. This review focuses on the latter where the biomaterial scaffold is designed to provide physical cues to stem cells in order to promote their behavior for tissue formation. Recent work that explores the effect of scaffold physical properties, topography, mechanical properties and electrical properties, is discussed. Although still being elucidated, the biological mechanisms, including cell shape, focal adhesion distribution, and nuclear shape, are presented. This review also discusses emerging areas and challenges in clinical translation.
Collapse
Affiliation(s)
- Shuo Wang
- Department of Biomedical Engineering New Jersey Institute of Technology Newark NJ 07102 USA
| | - Sharareh Hashemi
- Department of Biomedical Engineering New Jersey Institute of Technology Newark NJ 07102 USA
| | - Scott Stratton
- Department of Biomedical Engineering New Jersey Institute of Technology Newark NJ 07102 USA
| | | |
Collapse
|
46
|
Nuge T, Liu Z, Liu X, Ang BC, Andriyana A, Metselaar HSC, Hoque ME. Recent Advances in Scaffolding from Natural-Based Polymers for Volumetric Muscle Injury. Molecules 2021; 26:699. [PMID: 33572728 PMCID: PMC7865392 DOI: 10.3390/molecules26030699] [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: 09/25/2020] [Revised: 01/03/2021] [Accepted: 01/06/2021] [Indexed: 02/07/2023] Open
Abstract
Volumetric Muscle Loss (VML) is associated with muscle loss function and often untreated and considered part of the natural sequelae of trauma. Various types of biomaterials with different physical and properties have been developed to treat VML. However, much work remains yet to be done before the scaffolds can pass from the bench to the bedside. The present review aims to provide a comprehensive summary of the latest developments in the construction and application of natural polymers-based tissue scaffolding for volumetric muscle injury. Here, the tissue engineering approaches for treating volumetric muscle loss injury are highlighted and recent advances in cell-based therapies using various sources of stem cells are elaborated in detail. An overview of different strategies of tissue scaffolding and their efficacy on skeletal muscle cells regeneration and migration are presented. Furthermore, the present paper discusses a wide range of natural polymers with a special focus on proteins and polysaccharides that are major components of the extracellular matrices. The natural polymers are biologically active and excellently promote cell adhesion and growth. These bio-characteristics justify natural polymers as one of the most attractive options for developing scaffolds for muscle cell regeneration.
Collapse
Affiliation(s)
- Tamrin Nuge
- Department of Mechanical, Materials and Manufacturing Engineering, Faculty of Science and Engineering, University of Nottingham Ningbo China, 199 Taikang East Road, Ningbo 315100, China; (T.N.); (Z.L.)
| | - Ziqian Liu
- Department of Mechanical, Materials and Manufacturing Engineering, Faculty of Science and Engineering, University of Nottingham Ningbo China, 199 Taikang East Road, Ningbo 315100, China; (T.N.); (Z.L.)
| | - Xiaoling Liu
- Department of Mechanical, Materials and Manufacturing Engineering, Faculty of Science and Engineering, University of Nottingham Ningbo China, 199 Taikang East Road, Ningbo 315100, China; (T.N.); (Z.L.)
| | - Bee Chin Ang
- Centre of Advanced Materials, Faculty of Engineering, University of Malaya, Kuala Lumpur 50603, Malaysia; (A.A.); (H.S.C.M.)
- Department of Chemical Engineering, Faculty of Engineering, University of Malaya, Kuala Lumpur 50603, Malaysia
| | - Andri Andriyana
- Centre of Advanced Materials, Faculty of Engineering, University of Malaya, Kuala Lumpur 50603, Malaysia; (A.A.); (H.S.C.M.)
- Department of Mechanical Engineering, Faculty of Engineering, University of Malaya, Kuala Lumpur 50603, Malaysia
| | - Hendrik Simon Cornelis Metselaar
- Centre of Advanced Materials, Faculty of Engineering, University of Malaya, Kuala Lumpur 50603, Malaysia; (A.A.); (H.S.C.M.)
- Department of Mechanical Engineering, Faculty of Engineering, University of Malaya, Kuala Lumpur 50603, Malaysia
| | - Md Enamul Hoque
- Department of Biomedical Engineering, Military Institute of Science and Technology (MIST), Dhaka 1216, Bangladesh;
| |
Collapse
|
47
|
Zhu J, Fu Q, Shao J, Peng J, Qian Q, Zhou Y, Chen Y. Over-expression of MEG3 promotes differentiation of bone marrow mesenchymal stem cells into chondrocytes by regulating miR-129-5p/RUNX1 axis. Cell Cycle 2021; 20:96-111. [PMID: 33410373 DOI: 10.1080/15384101.2020.1863043] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
This study explored the role of MEG3 in the cartilage differentiation of bone marrow mesenchymal stem cells (BMSCs). We investigated the effects of over-expression and knockdown of MEG3 on cell viability, cell differentiation, and the expressions of MEG3, miR-129-5p, COL2, chondrocyte differentiation-related genes (sry-type high-mobility-group box 9 (SOX9), SOX5, Aggrecan, silent information regulator 1 (SIRT1), and Cartilage oligomeric matrix protein (COMP)). The targeting relationship between MEG3 and miR-129-5p and the target gene of miR-129-5p was confirmed through Starbase, TargetScan and luciferase experiments. Finally, a series of rescue experiments were conducted to study the regulatory effects of MEG3 and miR-129-5p. BMSCs were identified as CD29+ and CD44+ positive, and their differentiation was time-dependent. As BMSCs differentiated, MEG3 expression was up-regulated, but miR-129-5p was down-regulated. Over-expressed MEG3 promoted the viability and differentiation of BMSCs, up-regulated the expressions of COL2 and chondrocyte differentiation-related genes, and inhibited miR-129-5p. Runt-related transcription factor 1 (RUNX1) was negatively regulated as a target gene of miR-129-5p. Results of rescue experiments showed that the inhibitory effect of miR-129-5p mimic on BMSCs could be partially reversed by MEG3. Over-expression of MEG3 regulated the miR-129-5p/RUNX1 axis to promote the differentiation of BMSCs into chondrocytes. This study provides a reliable basis for the application of lncRNA in articular cartilage injury.
Collapse
Affiliation(s)
- Jun Zhu
- Department of Joint Surgery and Orthopedic Medicine, Shanghai Changzheng Hospital, Second Military Medical University , Shanghai, China
| | - Qiwei Fu
- Department of Joint Surgery and Orthopedic Medicine, Shanghai Changzheng Hospital, Second Military Medical University , Shanghai, China
| | - Jiahua Shao
- Department of Joint Surgery and Orthopedic Medicine, Shanghai Changzheng Hospital, Second Military Medical University , Shanghai, China
| | - Jinhui Peng
- Department of Joint Surgery and Orthopedic Medicine, Shanghai Changzheng Hospital, Second Military Medical University , Shanghai, China
| | - Qirong Qian
- Department of Joint Surgery and Orthopedic Medicine, Shanghai Changzheng Hospital, Second Military Medical University , Shanghai, China
| | - Yiqin Zhou
- Department of Joint Surgery and Orthopedic Medicine, Shanghai Changzheng Hospital, Second Military Medical University , Shanghai, China
| | - Yi Chen
- Department of Joint Surgery and Orthopedic Medicine, Shanghai Changzheng Hospital, Second Military Medical University , Shanghai, China
| |
Collapse
|
48
|
Priester C, MacDonald A, Dhar M, Bow A. Examining the Characteristics and Applications of Mesenchymal, Induced Pluripotent, and Embryonic Stem Cells for Tissue Engineering Approaches across the Germ Layers. Pharmaceuticals (Basel) 2020; 13:E344. [PMID: 33114710 PMCID: PMC7692540 DOI: 10.3390/ph13110344] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 10/15/2020] [Accepted: 10/20/2020] [Indexed: 12/13/2022] Open
Abstract
The field of regenerative medicine utilizes a wide array of technologies and techniques for repairing and restoring function to damaged tissues. Among these, stem cells offer one of the most potent and promising biological tools to facilitate such goals. Implementation of mesenchymal stem cells (MSCs), induced pluripotent stem cells (iPSCs), and embryonic stem cells (ESCs) offer varying advantages based on availability and efficacy in the target tissue. The focus of this review is to discuss characteristics of these three subset stem cell populations and examine their utility in tissue engineering. In particular, the development of therapeutics that utilize cell-based approaches, divided by germinal layer to further assess research targeting specific tissues of the mesoderm, ectoderm, and endoderm. The combinatorial application of MSCs, iPSCs, and ESCs with natural and synthetic scaffold technologies can enhance the reparative capacity and survival of implanted cells. Continued efforts to generate more standardized approaches for these cells may provide improved study-to-study variations on implementation, thereby increasing the clinical translatability of cell-based therapeutics. Coupling clinically translatable research with commercially oriented methods offers the potential to drastically advance medical treatments for multiple diseases and injuries, improving the quality of life for many individuals.
Collapse
Affiliation(s)
- Caitlin Priester
- Department of Animal Science, University of Tennessee, Knoxville, TN 37998, USA;
| | - Amber MacDonald
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, 2407 River Drive, Knoxville, TN 37996, USA; (A.M.); (M.D.)
| | - Madhu Dhar
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, 2407 River Drive, Knoxville, TN 37996, USA; (A.M.); (M.D.)
| | - Austin Bow
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, 2407 River Drive, Knoxville, TN 37996, USA; (A.M.); (M.D.)
| |
Collapse
|
49
|
Zidarič T, Milojević M, Vajda J, Vihar B, Maver U. Cultured Meat: Meat Industry Hand in Hand with Biomedical Production Methods. FOOD ENGINEERING REVIEWS 2020. [DOI: 10.1007/s12393-020-09253-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
50
|
Rukavina P, Koch F, Wehrle M, Tröndle K, Björn Stark G, Koltay P, Zimmermann S, Zengerle R, Lampert F, Strassburg S, Finkenzeller G, Simunovic F. In vivo evaluation of bioprinted prevascularized bone tissue. Biotechnol Bioeng 2020; 117:3902-3911. [PMID: 32749669 DOI: 10.1002/bit.27527] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 07/16/2020] [Accepted: 08/01/2020] [Indexed: 12/21/2022]
Abstract
Bioprinting can be considered as a progression of the classical tissue engineering approach, in which cells are randomly seeded into scaffolds. Bioprinting offers the advantage that cells can be placed with high spatial fidelity within three-dimensional tissue constructs. A decisive factor to be addressed for bioprinting approaches of artificial tissues is that almost all tissues of the human body depend on a functioning vascular system for the supply of oxygen and nutrients. In this study, we have generated cuboid prevascularized bone tissue constructs by bioprinting human adipose-derived mesenchymal stem cells (ASCs) and human umbilical vein endothelial cells (HUVECs) by extrusion-based bioprinting and drop-on-demand (DoD) bioprinting, respectively. The computer-generated print design could be verified in vitro after printing. After subcutaneous implantation of bioprinted constructs in immunodeficient mice, blood vessel formation with human microvessels of different calibers could be detected arising from bioprinted HUVECs and stabilization of human blood vessels by mouse pericytes was observed. In addition, bioprinted ASCs were able to synthesize a calcified bone matrix as an indicator of ectopic bone formation. These results indicate that the combined bioprinting of ASCs and HUVECs represents a promising strategy to produce prevascularized artificial bone tissue for prospective applications in the treatment of critical-sized bone defects.
Collapse
Affiliation(s)
- Patrick Rukavina
- Department of Plastic and Hand Surgery, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Fritz Koch
- Laboratory for MEMS Applications, IMTEK-Department of Microsystems Engineering, University of Freiburg, Freiburg, Germany
| | - Maximilian Wehrle
- Department of Plastic and Hand Surgery, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Kevin Tröndle
- Laboratory for MEMS Applications, IMTEK-Department of Microsystems Engineering, University of Freiburg, Freiburg, Germany
| | - G Björn Stark
- Department of Plastic and Hand Surgery, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Peter Koltay
- Laboratory for MEMS Applications, IMTEK-Department of Microsystems Engineering, University of Freiburg, Freiburg, Germany.,Hahn-Schickard, Freiburg, Germany.,Freiburg Center for Interactive Materials and Bioinspired Technologies, University of Freiburg, Freiburg, Germany
| | - Stefan Zimmermann
- Laboratory for MEMS Applications, IMTEK-Department of Microsystems Engineering, University of Freiburg, Freiburg, Germany
| | - Roland Zengerle
- Laboratory for MEMS Applications, IMTEK-Department of Microsystems Engineering, University of Freiburg, Freiburg, Germany.,Hahn-Schickard, Freiburg, Germany
| | - Florian Lampert
- Department of Plastic and Hand Surgery, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Sandra Strassburg
- Department of Plastic and Hand Surgery, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Günter Finkenzeller
- Department of Plastic and Hand Surgery, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Filip Simunovic
- Department of Plastic and Hand Surgery, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|