1
|
Nikitina N, Wadsworth J, Goelzer M, Goldfeldt M, Bursa N, Howard S, Crandall C, Semodji A, Zavala AG, Judex S, Rubin J, Lujan TJ, Fitzpatrick CK, Rubin CT, Satici A, Uzer G. Small Accelerations of the cell generate sufficient nuclear motion to modulate transcriptional activity, driving cellular response independent of matrix strain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.07.647583. [PMID: 40291652 PMCID: PMC12026902 DOI: 10.1101/2025.04.07.647583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
The cell's mechanical environment is a fundamental determinant of its activity. Ostensibly, the cellular response is dependent on interactions between extracellular matrix deformations and the cell adhesome. Low-intensity vibration (LIV) induces sinusoidal mechanical accelerations that stimulate mesenchymal stem cell (MSC) anabolism despite generating minimal matrix strain. In this study, we tested the hypothesis that accelerations of less than 1g cause nuclear motions relative to the cell membrane in adherent cells, resulting in elevated stresses in the cytoskeleton that promote transcriptional activity. Coupling a piezoelectric vibration platform with real-time microscopy, we applied a 0.7g, 90Hz LIV signal that oscillates the cell with displacements of up to ±11 µm. Live-cell tracking revealed that the sinusoidal vibrations caused the nucleus to move ±1.27 µm (17% of total displacement) out of phase with the cell membrane. Disruption of the LINC complex, which mechanically couples the nucleoskeleton to the cytoskeleton, doubled the magnitude of this relative motion, indicating that the nucleo-cytoskeletal configuration plays a major role in regulating nuclear motion. Consistent with a previously reported increase in nuclear stiffness caused by LIV, machine-learning-based image segmentation of confocal micrographs showed that LIV increased both apical and basal F-actin fiber numbers, generating a denser, more branched actin network near the nucleus. Following six 20 min bouts of LIV applied to MSC, RNA sequencing identified 372 differentially expressed genes. Upregulated gene sets were linked to F-actin assembly and focal adhesion pathways. Finite element simulations showed that nuclear stresses increased by LIV up to 18% were associated with nuclei flattening and a 30-50% increase in actin-generated forces. These findings demonstrate that low-intensity accelerations, independent of matrix strain, can directly activate a response of the nucleus, leading to cytoskeletal reorganization and heightened nuclear stresses. Thus, even very small oscillatory mechanical signals can markedly influence cell outcomes, establishing a mechanosensing pathway independent of extracellular strains.
Collapse
|
2
|
Regner AM, DeLeon M, Gibbons KD, Howard S, Nesbitt DQ, Darghiasi SF, Zavala AG, Lujan TJ, Fitzpatrick CK, Farach-Carson MC, Wu D, Uzer G. Increased deformations are dispensable for encapsulated cell mechanoresponse in engineered bone analogs mimicking aging bone marrow. MECHANOBIOLOGY IN MEDICINE 2025; 3:100097. [PMID: 40134991 PMCID: PMC11936507 DOI: 10.1016/j.mbm.2024.100097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/27/2025]
Abstract
Aged individuals and astronauts experience bone loss despite rigorous physical activity. Bone mechanoresponse is in-part regulated by mesenchymal stem cells (MSCs) that respond to mechanical stimuli. Direct delivery of low intensity vibration (LIV) recovers MSC proliferation in senescence and simulated microgravity models, indicating that age-related reductions in mechanical signal delivery within bone marrow may contribute to declining bone mechanoresponse. To answer this question, we developed a 3D bone marrow analog that controls trabecular geometry, marrow mechanics and external stimuli. Validated finite element (FE) models were developed to quantify strain environment within hydrogels during LIV. Bone marrow analogs with gyroid-based trabeculae of scaffold volume fractions (SV/TV) corresponding to adult (25 %) and aged (13 %) mice were printed using polylactic acid (PLA). MSCs encapsulated in migration-permissive hydrogels within printed trabeculae showed robust cell populations on both PLA surface and hydrogel within a week. Following 14 days of LIV treatment (1 g, 100 Hz, 1 h/day), cell proliferation, type-I collagen (Collagen-I) and filamentous actin (F-actin) were quantified for the cells in the hydrogel fraction. While LIV increased all measured outcomes, FE models predicted higher von Mises strains for the 13 % SV/TV groups (0.2 %) when compared to the 25 % SV/TV group (0.1 %). While LIV increased collagen-I volume 34 % more in 13 % SV/TV groups when compared to 25 % SV/TV groups, collagen-I and F-actin measures remained lower in the 13 % SV/TV groups when compared to 25 % SV/TV counterparts, indicating that both LIV-induced strains and scaffold volume fraction (i.e. available scaffold surface) affect cell behavior in the hydrogel phase. Overall, bone marrow analogs offer a robust and repeatable platform to study bone mechanobiology.
Collapse
Affiliation(s)
- Alexander M. Regner
- Mechanical and Biomedical Engineering Department, Boise State University, USA
| | - Maximilien DeLeon
- Department of Diagnostic and Biomedical Sciences, UTHealth Houston School of Dentistry, USA
- Department of Bioengineering, Rice University, USA
| | - Kalin D. Gibbons
- Mechanical and Biomedical Engineering Department, Boise State University, USA
| | - Sean Howard
- Mechanical and Biomedical Engineering Department, Boise State University, USA
| | | | | | - Anamaria G. Zavala
- Mechanical and Biomedical Engineering Department, Boise State University, USA
| | - Trevor J. Lujan
- Mechanical and Biomedical Engineering Department, Boise State University, USA
| | | | - Mary C. Farach-Carson
- Department of Diagnostic and Biomedical Sciences, UTHealth Houston School of Dentistry, USA
- Department of Bioengineering, Rice University, USA
- Department of Biosciences, Rice University, USA
| | - Danielle Wu
- Department of Diagnostic and Biomedical Sciences, UTHealth Houston School of Dentistry, USA
- Department of Bioengineering, Rice University, USA
| | - Gunes Uzer
- Mechanical and Biomedical Engineering Department, Boise State University, USA
| |
Collapse
|
3
|
Xu J, Song Z. The role of different physical exercises as an anti-aging factor in different stem cells. Biogerontology 2025; 26:63. [PMID: 40009244 DOI: 10.1007/s10522-025-10205-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 02/14/2025] [Indexed: 02/27/2025]
Abstract
The senescence process is connected to the characteristics of cellular aging. Understanding their causal network helps develop a framework for creating new treatments to slow down the senescence process. A growing body of research indicates that aging may adversely affect stem cells (SCs). SCs change their capability to differentiate into different cell types and decrease their potential for renewal as they age. Research has indicated that consistent physical exercise offers several health advantages, including a reduced risk of age-associated ailments like tumors, heart disease, diabetes, and neurological disorders. Exercise is a potent physiological stressor linked to higher red blood cell counts and an enhanced immune system, promoting disease resistance. Sports impact mesenchymal SCs (MSCs), hematopoietic SCs (HSCs), neuronal SCs (NuSCs), and muscular SCs (MuSCs), among other aged SCs types. These changes to the niche will probably affect the amount and capability of adult SCs after exercise. In this work, we looked into how different types of SCs age. The impact of physical activity on the aging process has been studied. Additionally, there has been discussion and study on the impact of different sports and physical activities on SCs as an anti-aging component.
Collapse
Affiliation(s)
- Jia Xu
- College of Physical Education, North-West Normal University, Lanzhou, 730070, China
| | - Zhe Song
- Cangzhou Medical College, Cangzhou, 061001, China.
| |
Collapse
|
4
|
Chan ME, Ashdown C, Strait L, Pasumarthy S, Hassan A, Crimarco S, Singh C, Patel VS, Pagnotti G, Khan O, Uzer G, Rubin CT. Low intensity mechanical signals promote proliferation in a cell-specific manner: Tailoring a non-drug strategy to enhance biomanufacturing yields. MECHANOBIOLOGY IN MEDICINE 2024; 2:100080. [PMID: 39717386 PMCID: PMC11666124 DOI: 10.1016/j.mbm.2024.100080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/25/2024]
Abstract
Biomanufacturing relies on living cells to produce biotechnology-based therapeutics, tissue engineering constructs, vaccines, and a vast range of agricultural and industrial products. With the escalating demand for these bio-based products, any process that could improve yields and shorten outcome timelines by accelerating cell proliferation would have a significant impact across the discipline. While these goals are primarily achieved using biological or chemical strategies, harnessing cell mechanosensitivity represents a promising - albeit less studied - physical pathway to promote bioprocessing endpoints, yet identifying which mechanical parameters influence cell activities has remained elusive. We tested the hypothesis that mechanical signals, delivered non-invasively using low-intensity vibration (LIV; <1 g, 10-500 Hz), will enhance cell expansion, and determined that any unique signal configuration was not equally influential across a range of cell types. Varying frequency, intensity, duration, refractory period, and daily doses of LIV increased proliferation in Chinese Hamster Ovary (CHO)-adherent cells (+79% in 96 hr) using a particular set of LIV parameters (0.2 g, 500 Hz, 3 × 30 min/d, 2 hr refractory period), yet this same mechanical input suppressed proliferation in CHO-suspension cells (-13%). Another set of LIV parameters (30 Hz, 0.7 g, 2 × 60 min/d, 2 hr refractory period) however, were able to increase the proliferation of CHO-suspension cells by 210% and T-cells by 20.3%. Importantly, we also reported that T-cell response to LIV was in-part dependent upon AKT phosphorylation, as inhibiting AKT phosphorylation reduced the proliferative effect of LIV by over 60%, suggesting that suspension cells utilize mechanism(s) similar to adherent cells to sense specific LIV signals. Particle image velocimetry combined with finite element modeling showed high transmissibility of these signals across fluids (>90%), and LIV effectively scaled up to T75 flasks. Ultimately, when LIV is tailored to the target cell population, it's highly efficient transmission across media represents a means to non-invasively augment biomanufacturing endpoints for both adherent and suspended cells, and holds immediate applications, ranging from small-scale, patient-specific personalized medicine to large-scale commercial biocentric production challenges.
Collapse
Affiliation(s)
- M. Ete Chan
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, 11794-5280, USA
| | - Christopher Ashdown
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, 11794-5280, USA
- Medical Scientist Training Program, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Lia Strait
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, 11794-5280, USA
| | - Sishir Pasumarthy
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, 11794-5280, USA
| | - Abdullah Hassan
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, 11794-5280, USA
| | - Steven Crimarco
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, 11794-5280, USA
| | - Chanpreet Singh
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, 11794-5280, USA
| | - Vihitaben S. Patel
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, 11794-5280, USA
| | - Gabriel Pagnotti
- Department of Endocrine Neoplasia and Hormonal Disorders, MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Omor Khan
- Department of Mechanical and Biomedical Engineering, College of Engineering, Boise State University, Boise, ID, 83725-205, USA
| | - Gunes Uzer
- Department of Mechanical and Biomedical Engineering, College of Engineering, Boise State University, Boise, ID, 83725-205, USA
| | - Clinton T. Rubin
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, 11794-5280, USA
- Center for Biotechnology, New York State Center for Advanced Technology in Medical Biotechnology, Stony Brook University, Stony Brook, NY, 11794-5281, USA
| |
Collapse
|
5
|
Bedoux A, Lallemant-Dudek P, Bensidhoum M, Potier E, Larochette N, Mary P, Vialle R, Hoc T, Bachy M. Effects of shear stress on mesenchymal stem cells of patients with osteogenesis imperfecta. Orthop Traumatol Surg Res 2024:104067. [PMID: 39581491 DOI: 10.1016/j.otsr.2024.104067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 03/15/2024] [Accepted: 04/04/2024] [Indexed: 11/26/2024]
Abstract
INTRODUCTION Osteogenesis imperfecta (OI) is a rare genetic bone disorder, mainly caused by autosomal dominant mutations of the COL1A1 or COL1A2 genes that encode the alpha chains of type 1 collagen. In severe forms and in nonambulatory patients, for whom physical exercise is difficult, exposing the bone to mechanical stimuli by promoting movement, especially with physiotherapy and mobility aids, is an essential part of clinical practice. However, the effects of mechanical stimulation at the cellular level remain unknown for this disease. HYPOTHESIS The study hypothesis was that human mesenchymal stem cells (hMSCs) from patients with OI were as sensitive to mechanical stimulation as those from healthy patients, validating the current clinical practice. MATERIALS AND METHODS hMSCs were harvested from 3 healthy control subjects and 3 patients with OI during an elective osteotomy of a long bone of the lower limb. The healthy and OI hMSCs were then exposed to mechanical stimuli, such as intermittent shear stress of 0, 0.7, 1.5, and 3 Pascal (Pa) at a frequency of 2.8 Hertz (Hz) for 30 minutes using a commercial ibidi system. The immediate early gene expression of themechanosensitive prostaglandin-endoperoxide synthase 2 (PTGS2) was examined 1 hour after stimulation to determine the best level of mechanical stimulation. The expression of 7 other mechanosensitive genes was also examined for this level of mechanical stimulation after applying intermittent shear stress at 1.5 Pa. RESULTS In all hMSCs, mechanical stimulation induced PTGS2 gene overexpression with a maximum after exposure to intermittent shear stress of 1.5 Pa and without significant differences between OI and healthy donors. Except for fibroblast growth factor 2, gene expression in OI donors was found to be significantly different from that in hMSCs not exposed to shear stress. Moreover, the relative expression associated with mechanical stimulation was not significantly different between healthy and OI donors for most other genes. DISCUSSION This is the first study to demonstrate that hMSCs from patients with OI are as sensitive to mechanical shear stress as those from healthy donors. The mechanical stress that resulted in the greatest change in the expression of PTGS2 in patients with OI was similar to that previously reported in the literature for healthy subjects. These findings are an important step toward further fundamental research aimed at confirming the effects of mechanical stress at the cellular level over the long term and, more importantly, toward developing clinical protocols for delivering mechanical stimuli to these patients. LEVEL OF EVIDENCE III; comparative case-control study.
Collapse
Affiliation(s)
- Agathe Bedoux
- Université de Paris Cité, B3OA, UMR CNRS 7052, INSERM U1271, 10 avenue de Verdun, 75010 Paris, France
| | - Pauline Lallemant-Dudek
- Service de Médecine Physique et Réadaptation fonctionnelle pédiatrique, APHP-Sorbonne Université, Hôpital Armand Trousseau, 26 avenue du Docteur Arnold-Netter, 75012 Paris, France
| | - Morad Bensidhoum
- Université de Paris Cité, B3OA, UMR CNRS 7052, INSERM U1271, 10 avenue de Verdun, 75010 Paris, France
| | - Esther Potier
- Université de Paris Cité, B3OA, UMR CNRS 7052, INSERM U1271, 10 avenue de Verdun, 75010 Paris, France
| | - Nathanael Larochette
- Université de Paris Cité, B3OA, UMR CNRS 7052, INSERM U1271, 10 avenue de Verdun, 75010 Paris, France
| | - Pierre Mary
- Service de Chirurgie Orthopédique et Réparatrice de l'Enfant, APHP-Sorbonne Université, Hôpital Armand Trousseau, 26 avenue du Docteur Arnold-Netter, 75012 Paris, France
| | - Raphaël Vialle
- Service de Chirurgie Orthopédique et Réparatrice de l'Enfant, APHP-Sorbonne Université, Hôpital Armand Trousseau, 26 avenue du Docteur Arnold-Netter, 75012 Paris, France
| | - Thierry Hoc
- Université de Paris Cité, B3OA, UMR CNRS 7052, INSERM U1271, 10 avenue de Verdun, 75010 Paris, France; Ecole Centrale de Lyon, Département de mécanique, MSGMGC, 36 Avenue Guy de Collongue, 69134 Ecully Cedex, France
| | - Manon Bachy
- Université de Paris Cité, B3OA, UMR CNRS 7052, INSERM U1271, 10 avenue de Verdun, 75010 Paris, France; Service de Chirurgie Orthopédique et Réparatrice de l'Enfant, APHP-Sorbonne Université, Hôpital Armand Trousseau, 26 avenue du Docteur Arnold-Netter, 75012 Paris, France.
| |
Collapse
|
6
|
Camacho-Cardenosa M, Pulido-Escribano V, Torrecillas-Baena B, Quesada-Gómez JM, Herrera-Martínez AD, Sola-Guirado RR, Dorado G, Gálvez-Moreno MÁ, Casado-Díaz A. Combined Effects of Cyclic Hypoxic and Mechanical Stimuli on Human Bone Marrow Mesenchymal Stem Cell Differentiation: A New Approach to the Treatment of Bone Loss. J Clin Med 2024; 13:5805. [PMID: 39407866 PMCID: PMC11476683 DOI: 10.3390/jcm13195805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 09/24/2024] [Accepted: 09/26/2024] [Indexed: 10/20/2024] Open
Abstract
Background: The prevention and treatment of bone loss and osteoporotic fractures is a public health challenge. Combined with normobaric hypoxia, whole-body vibration has a high clinic potential in bone health and body composition. The effect of this therapy may be mediated by its action on bone marrow mesenchymal stem cells (MSCs). Objectives: Evaluate the effects of cyclic low-vibration stimuli and/or hypoxia on bone marrow-derived human MSC differentiation. Methods: MSCs were exposed four days per week, two hours/day, to hypoxia (3% O2) and/or vibration before they were induced to differentiate or during differentiation into osteoblasts or adipocytes. Gene and protein expression of osteoblastic, adipogenic, and cytoskeletal markers were studied, as well as extracellular matrix mineralization and lipid accumulation. Results: early osteoblastic markers increased in undifferentiated MSCs, pretreated in hypoxia and vibration. This pretreatment also increased mRNA levels of osteoblastic genes and beta-catenin protein in the early stages of differentiation into osteoblasts without increasing mineralization. When MSCs were exposed to vibration under hypoxia or normoxia during osteoblastic differentiation, mineralization increased with respect to cultures without vibrational stimuli. In MSCs differentiated into adipocytes, both in those pretreated as well as exposed to different conditions during differentiation, lipid formation decreased. Changes in adipogenic gene expression and increased beta-catenin protein were observed in cultures treated during differentiation. Conclusions: Exposure to cyclic hypoxia in combination with low-intensity vibratory stimuli had positive effects on osteoblastic differentiation and negative ones on adipogenesis of bone marrow-derived MSCs. These results suggest that in elderly or frail people with difficulty performing physical activity, exposure to normobaric cyclic hypoxia and low-density vibratory stimuli could improve bone metabolism and health.
Collapse
Affiliation(s)
- Marta Camacho-Cardenosa
- Unidad de Gestión Clínica de Endocrinología y Nutrición, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Hospital Universitario Reina Sofía, 14004 Córdoba, Spain; (V.P.-E.); (B.T.-B.); (J.M.Q.-G.); (A.D.H.-M.); (M.Á.G.-M.)
| | - Victoria Pulido-Escribano
- Unidad de Gestión Clínica de Endocrinología y Nutrición, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Hospital Universitario Reina Sofía, 14004 Córdoba, Spain; (V.P.-E.); (B.T.-B.); (J.M.Q.-G.); (A.D.H.-M.); (M.Á.G.-M.)
| | - Bárbara Torrecillas-Baena
- Unidad de Gestión Clínica de Endocrinología y Nutrición, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Hospital Universitario Reina Sofía, 14004 Córdoba, Spain; (V.P.-E.); (B.T.-B.); (J.M.Q.-G.); (A.D.H.-M.); (M.Á.G.-M.)
| | - Jose Manuel Quesada-Gómez
- Unidad de Gestión Clínica de Endocrinología y Nutrición, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Hospital Universitario Reina Sofía, 14004 Córdoba, Spain; (V.P.-E.); (B.T.-B.); (J.M.Q.-G.); (A.D.H.-M.); (M.Á.G.-M.)
| | - Aura D. Herrera-Martínez
- Unidad de Gestión Clínica de Endocrinología y Nutrición, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Hospital Universitario Reina Sofía, 14004 Córdoba, Spain; (V.P.-E.); (B.T.-B.); (J.M.Q.-G.); (A.D.H.-M.); (M.Á.G.-M.)
| | - Rafael R. Sola-Guirado
- Department Mecánica, Escuela Politécnica Superior, Universidad de Córdoba, 14071 Córdoba, Spain;
| | - Gabriel Dorado
- Department Bioquímica y Biología Molecular, Campus Rabanales C6-1-E17, Campus de Excelencia Internacional Agroalimentario (ceiA3), Universidad de Córdoba, 14071 Córdoba, Spain;
- CIBER Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, 08003 Madrid, Spain
| | - María Ángeles Gálvez-Moreno
- Unidad de Gestión Clínica de Endocrinología y Nutrición, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Hospital Universitario Reina Sofía, 14004 Córdoba, Spain; (V.P.-E.); (B.T.-B.); (J.M.Q.-G.); (A.D.H.-M.); (M.Á.G.-M.)
| | - Antonio Casado-Díaz
- Unidad de Gestión Clínica de Endocrinología y Nutrición, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Hospital Universitario Reina Sofía, 14004 Córdoba, Spain; (V.P.-E.); (B.T.-B.); (J.M.Q.-G.); (A.D.H.-M.); (M.Á.G.-M.)
- CIBER Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, 08003 Madrid, Spain
| |
Collapse
|
7
|
Birks S, Howard S, O’Rourke C, Thompson WR, Lau A, Uzer G. Osterix-driven LINC complex disruption in vivo diminishes osteogenesis at 8 weeks but not at 15 weeks. J Orthop Res 2024; 42:2007-2016. [PMID: 38602438 PMCID: PMC11293982 DOI: 10.1002/jor.25849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 03/15/2024] [Accepted: 03/24/2024] [Indexed: 04/12/2024]
Abstract
The Linker of Nucleoskeleton and Cytoskeleton (LINC) complex is a crucial connective component between the nuclear envelope and the cytoskeleton involving various cellular processes including nuclear positioning, nuclear architecture, and mechanotransduction. How LINC complexes regulate bone formation in vivo, however, is not well understood. To start bridging this gap, here we created a LINC disruption murine model using transgenic mice expressing Cre recombinase enzyme under the control of the Osterix (Osx-Cre) which is primarily active in pre-osteoblasts and floxed Tg(CAG-LacZ/EGFP-KASH2) mice. Tg(CAG-LacZ/EGFP-KASH2) mice contain a lox-STOP-lox flanked LacZ gene which is deleted upon cre recombination allowing for the overexpression of an EGFP-KASH2 fusion protein. This overexpressed protein disrupts endogenous Nesprin-Sun binding leading to disruption of LINC complexes. Thus, crossing these two lines results in an Osx- driven LINC disruption (ODLD) specific to pre-osteoblasts. In this study, we investigated how this LINC disruption affects exercise-induced bone accrual. ODLD cells had decreased osteogenic and adipogenic potential in vitro compared to non-disrupted controls and sedentary ODLD mice showed decreased bone quality at 8 weeks. Upon access to a voluntary running wheel, ODLD animals showed increased running time and distance; however, our 6-week exercise intervention did not significantly affect bone microarchitecture and bone mechanical properties.
Collapse
Affiliation(s)
- Scott Birks
- Boise State University, Micron School of Materials Science and Engineering
| | - Sean Howard
- Boise State University, Mechanical and Biomedical Engineering
| | | | | | - Anthony Lau
- The College of New Jersey, Biomedical Engineering
| | - Gunes Uzer
- Boise State University, Mechanical and Biomedical Engineering
| |
Collapse
|
8
|
Regner AM, DeLeon M, Gibbons KD, Howard S, Nesbitt DQ, Lujan TJ, Fitzpatrick CK, Farach-Carson MC, Wu D, Uzer G. Increased deformations are dispensable for cell mechanoresponse in engineered bone analogs mimicking aging bone marrow. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.24.559187. [PMID: 37905032 PMCID: PMC10614733 DOI: 10.1101/2023.09.24.559187] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Aged individuals and astronauts experience bone loss despite rigorous physical activity. Bone mechanoresponse is in-part regulated by mesenchymal stem cells (MSCs) that respond to mechanical stimuli. Direct delivery of low intensity vibration (LIV) recovers MSC proliferation in senescence and simulated microgravity models, indicating that age-related reductions in mechanical signal delivery within bone marrow may contribute to declining bone mechanoresponse. To answer this question, we developed a 3D bone marrow analog that controls trabecular geometry, marrow mechanics and external stimuli. Validated finite element (FE) models were developed to quantify strain environment within hydrogels during LIV. Bone marrow analogs with gyroid-based trabeculae of bone volume fractions (BV/TV) corresponding to adult (25%) and aged (13%) mice were printed using polylactic acid (PLA). MSCs encapsulated in migration-permissive hydrogels within printed trabeculae showed robust cell populations on both PLA surface and hydrogel within a week. Following 14 days of LIV treatment (1g, 100 Hz, 1 hour/day), type-I collagen and F-actin were quantified for the cells in the hydrogel fraction. While LIV increased all measured outcomes, FE models predicted higher von Mises strains for the 13% BV/TV groups (0.2%) when compared to the 25% BV/TV group (0.1%). Despite increased strains, collagen-I and F-actin measures remained lower in the 13% BV/TV groups when compared to 25% BV/TV counterparts, indicating that cell response to LIV does not depend on hydrogel strains and that bone volume fraction (i.e. available bone surface) directly affects cell behavior in the hydrogel phase independent of the external stimuli. Overall, bone marrow analogs offer a robust and repeatable platform to study bone mechanobiology.
Collapse
Affiliation(s)
- Alexander M Regner
- Mechanical and Biomedical Engineering Department, Boise State University
| | - Maximilien DeLeon
- Department of Diagnostic and Biomedical Sciences, UTHealth Houston School of Dentistry
- Department of Bioengineering, Rice University
- Department of Biosciences, Rice University
| | - Kalin D. Gibbons
- Mechanical and Biomedical Engineering Department, Boise State University
| | - Sean Howard
- Mechanical and Biomedical Engineering Department, Boise State University
| | | | - Trevor J. Lujan
- Mechanical and Biomedical Engineering Department, Boise State University
| | | | - Mary C Farach-Carson
- Department of Diagnostic and Biomedical Sciences, UTHealth Houston School of Dentistry
- Department of Bioengineering, Rice University
- Department of Biosciences, Rice University
| | - Danielle Wu
- Department of Diagnostic and Biomedical Sciences, UTHealth Houston School of Dentistry
- Department of Bioengineering, Rice University
- Department of Biosciences, Rice University
| | - Gunes Uzer
- Mechanical and Biomedical Engineering Department, Boise State University
| |
Collapse
|
9
|
Chan ME, Strait L, Ashdown C, Pasumarthy S, Hassan A, Crimarco S, Singh C, Patel VS, Pagnotti G, Khan O, Uzer G, Rubin CT. Low intensity mechanical signals promote proliferation in a cell-specific manner: Tailoring a non-drug strategy to enhance biomanufacturing yields. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.05.547864. [PMID: 37461507 PMCID: PMC10350023 DOI: 10.1101/2023.07.05.547864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2024]
Abstract
Biomanufacturing relies on living cells to produce biotechnology-based therapeutics, tissue engineering constructs, vaccines, and a vast range of agricultural and industrial products. With the escalating demand for these bio-based products, any process that could improve yields and shorten outcome timelines by accelerating cell proliferation would have a significant impact across the discipline. While these goals are primarily achieved using biological or chemical strategies, harnessing cell mechanosensitivity represents a promising - albeit less studied - physical pathway to promote bioprocessing endpoints, yet identifying which mechanical parameters influence cell activities has remained elusive. We tested the hypothesis that mechanical signals, delivered non-invasively using low-intensity vibration (LIV; <1g, 10-500Hz), will enhance cell expansion, and determined that any unique signal configuration was not equally influential across a range of cell types. Varying frequency, intensity, duration, refractory period, and daily doses of LIV increased proliferation in CHO-adherent cells (+79% in 96h) using a particular set of LIV parameters (0.2g, 500Hz, 3x30 min/d, 2h refractory period), yet this same mechanical input suppressed proliferation in CHO-suspension cells (-13%). Exposing these same CHO-suspension cells to distinct LIV parameters (30Hz, 0.7g, 2x60 min/d, 2h refractory period) increased proliferation by 210%. Particle image velocimetry combined with finite element modeling showed high transmissibility of these signals across fluids (>90%), and LIV effectively scaled up to T75 flasks. Ultimately, when LIV is tailored to the target cell population, its highly efficient transmission across media represents a means to non-invasively augment biomanufacturing endpoints for both adherent and suspended cells, and holds immediate applications, ranging from small-scale, patient-specific personalized medicine to large-scale commercial bio-centric production challenges.
Collapse
|
10
|
Goh D, Yang Y, Lee EH, Hui JHP, Yang Z. Managing the Heterogeneity of Mesenchymal Stem Cells for Cartilage Regenerative Therapy: A Review. Bioengineering (Basel) 2023; 10:bioengineering10030355. [PMID: 36978745 PMCID: PMC10045936 DOI: 10.3390/bioengineering10030355] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 03/12/2023] [Accepted: 03/12/2023] [Indexed: 03/18/2023] Open
Abstract
Articular cartilage defects commonly result from trauma and are associated with significant morbidity. Since cartilage is an avascular, aneural, and alymphatic tissue with a poor intrinsic healing ability, the regeneration of functional hyaline cartilage remains a difficult clinical problem. Mesenchymal stem cells (MSCs) are multipotent cells with multilineage differentiation potential, including the ability to differentiate into chondrocytes. Due to their availability and ease of ex vivo expansion, clinicians are increasingly applying MSCs in the treatment of cartilage lesions. However, despite encouraging pre-clinical and clinical data, inconsistencies in MSC proliferative and chondrogenic potential depending on donor, tissue source, cell subset, culture conditions, and handling techniques remain a key barrier to widespread clinical application of MSC therapy in cartilage regeneration. In this review, we highlight the strategies to manage the heterogeneity of MSCs ex vivo for more effective cartilage repair, including reducing the MSC culture expansion period, and selecting MSCs with higher chondrogenic potential through specific genetic markers, surface markers, and biophysical attributes. The accomplishment of a less heterogeneous population of culture-expanded MSCs may improve the scalability, reproducibility, and standardisation of MSC therapy for clinical application in cartilage regeneration.
Collapse
Affiliation(s)
- Doreen Goh
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, NUHS Tower block Level 11, Singapore 119288, Singapore
- NUS Tissue Engineering Program, Life Sciences Institute, National University of Singapore, 27 Medical Drive, DSO (Kent Ridge) Building, Level 4, Singapore 11751, Singapore
| | - Yanmeng Yang
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, NUHS Tower block Level 11, Singapore 119288, Singapore
- NUS Tissue Engineering Program, Life Sciences Institute, National University of Singapore, 27 Medical Drive, DSO (Kent Ridge) Building, Level 4, Singapore 11751, Singapore
- Critical Analytics for Manufacturing Personalised-Medicine, Singapore-MIT Alliance for Research and Technology, Singapore 138602, Singapore
| | - Eng Hin Lee
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, NUHS Tower block Level 11, Singapore 119288, Singapore
- NUS Tissue Engineering Program, Life Sciences Institute, National University of Singapore, 27 Medical Drive, DSO (Kent Ridge) Building, Level 4, Singapore 11751, Singapore
- Critical Analytics for Manufacturing Personalised-Medicine, Singapore-MIT Alliance for Research and Technology, Singapore 138602, Singapore
| | - James Hoi Po Hui
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, NUHS Tower block Level 11, Singapore 119288, Singapore
- NUS Tissue Engineering Program, Life Sciences Institute, National University of Singapore, 27 Medical Drive, DSO (Kent Ridge) Building, Level 4, Singapore 11751, Singapore
| | - Zheng Yang
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, NUHS Tower block Level 11, Singapore 119288, Singapore
- NUS Tissue Engineering Program, Life Sciences Institute, National University of Singapore, 27 Medical Drive, DSO (Kent Ridge) Building, Level 4, Singapore 11751, Singapore
- Critical Analytics for Manufacturing Personalised-Medicine, Singapore-MIT Alliance for Research and Technology, Singapore 138602, Singapore
- Correspondence: ; Tel.: +65-6516-5398
| |
Collapse
|
11
|
Wang W, Li N, Zhao Y, Wu H, Wang M, Chen X. Effect of stretch frequency on osteogenesis of periodontium during periodontal ligament distraction. Orthod Craniofac Res 2023; 26:53-61. [PMID: 35384280 DOI: 10.1111/ocr.12577] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 03/20/2022] [Accepted: 03/31/2022] [Indexed: 01/07/2023]
Abstract
OBJECTIVES Periodontal ligament distraction (PDLD) can accelerate orthodontic tooth movement (OTM). However, the effect of stretch frequency on osseous formation during PDLD remains unclear. Here, we sought to identify the effect of PDLD frequency on the osteogenic remodelling of the periodontium. MATERIALS AND METHODS (i) In vitro, five human periodontal ligament stem cell (PDLSC) cultures were randomized to either static conditions or exposure to a cyclic stretch force involving 12% deformation at frequencies of 0.3, 0.5, 0.7 or 1.0 Hz for 12 h, and the osteogenic differentiation of PDLSCs was assessed using Western blotting. (ii) In vivo, 18 beagle dogs underwent orthodontic distalization of bilateral maxillary first premolars. In the test groups, PDLD was performed at a frequency of two or six times/day, while Ni-Ti coil springs were applied to mimic traditional OTM in the control group. The amount of OTM and histological staining was estimated after force loading for 5, 10 and 15 days. RESULTS (i) In vitro, the expression of osteogenic-specific markers (runt-related transcription factor 2 [Runx2], type I collagen [COL-I] and osteocalcin [OCN]) increased with the frequency of tensile force, to a peak at 0.7 Hz. (ii) In vivo, both PDLD groups displayed a greater rate of OTM and a higher bone metabolism than the control group. The expression of COL-I and OCN was significantly reinforced in the six times/day-PDLD group in comparison to the two times/day-PDLD group. CONCLUSIONS The cyclic stretch force enhances osteogenesis of the periodontium in a frequency-dependent manner.
Collapse
Affiliation(s)
- W Wang
- Department of Stomatology, First Affiliated Hospital, College of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - N Li
- Department of Orthodontics, Yantai Hospital of Stomatology, Yantai, China
| | - Y Zhao
- Department of Stomatology, First Affiliated Hospital, College of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - H Wu
- Department of Stomatology, First Affiliated Hospital, College of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - M Wang
- Anesthesiology Department, Second Affiliated Hospital, College of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - X Chen
- Department of Stomatology, First Affiliated Hospital, College of Medicine, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
12
|
Rubin J, van Wijnen AJ, Uzer G. Architectural control of mesenchymal stem cell phenotype through nuclear actin. Nucleus 2022; 13:35-48. [PMID: 35133922 PMCID: PMC8837231 DOI: 10.1080/19491034.2022.2029297] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 01/10/2022] [Accepted: 01/11/2022] [Indexed: 11/18/2022] Open
Abstract
There is growing appreciation that architectural components of the nucleus regulate gene accessibility by altering chromatin organization. While nuclear membrane connector proteins link the mechanosensitive actin cytoskeleton to the nucleoskeleton, actin's contribution to the inner architecture of the nucleus remains enigmatic. Control of actin transport into the nucleus, plus the presence of proteins that control actin structure (the actin tool-box) within the nucleus, suggests that nuclear actin may support biomechanical regulation of gene expression. Cellular actin structure is mechanoresponsive: actin cables generated through forces experienced at the plasma membrane transmit force into the nucleus. We posit that dynamic actin remodeling in response to such biomechanical cues provides a novel level of structural control over the epigenetic landscape. We here propose to bring awareness to the fact that mechanical forces can promote actin transfer into the nucleus and control structural arrangements as illustrated in mesenchymal stem cells, thereby modulating lineage commitment.
Collapse
Affiliation(s)
- Janet Rubin
- Department of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Andre J. van Wijnen
- Department of Biochemistry, University of Vermont Medical School, Burlington, Vt, USA
| | - Gunes Uzer
- Department of Mechanical & Biomedical Engineering, Boise State University, Boise, ID, USA
| |
Collapse
|
13
|
Su T, Xu M, Lu F, Chang Q. Adipogenesis or osteogenesis: destiny decision made by mechanical properties of biomaterials. RSC Adv 2022; 12:24501-24510. [PMID: 36128379 PMCID: PMC9425444 DOI: 10.1039/d2ra02841g] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 07/24/2022] [Indexed: 11/21/2022] Open
Abstract
Regenerative medicine affords an effective approach for restoring defect-associated diseases, and biomaterials play a pivotal role as cell niches to support the cell behavior and decide the destiny of cell differentiation. Except for chemical inducers, mechanical properties such as stiffness, pore size and topography of biomaterials play a crucial role in the regulation of cell behaviors and functions. Stiffness may determine the adipogenesis or osteogenesis of mesenchymal stem cells (MSCs) via the translocation of yes-associated protein (YAP) and the transcriptional coactivator with a PDZ-binding motif (TAZ). External forces transmit through cytoskeleton reorientation to assist nuclear deformation and molecule transport, meanwhile, signal pathways including the Hippo, FAK/RhoA/ROCK, and Wnt/β-catenin have been evidenced to participate in the mechanotransduction. Different pore sizes not only tailor the scaffold stiffness but also conform to the requirements of cell migration and vessels in-growth. Topography guides cell geometry along with mobility and determines the cell fate ascribed to micro/nano-scale contact. Herein, we highlight the recent progress in exploring the regulation mechanism by the physical properties of biomaterials, which might lead to more innovative regenerative strategies for adipose or bone tissue repair.
Collapse
Affiliation(s)
- Ting Su
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University 510515 China
| | - Mimi Xu
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University 510515 China
| | - Feng Lu
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University 510515 China
| | - Qiang Chang
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University 510515 China
| |
Collapse
|
14
|
Amuk M, Gül Amuk N, Hacıoğlu Z. Effects of orofacial applications of low-magnitude, high-frequency mechanical vibration on cranial sutures and calvarial bones: A micro-computed tomography study in rats. Am J Orthod Dentofacial Orthop 2022; 162:459-468. [PMID: 35777991 DOI: 10.1016/j.ajodo.2021.04.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 04/04/2021] [Accepted: 04/04/2021] [Indexed: 11/20/2022]
Abstract
INTRODUCTION The purpose of this study was to assess the effects of orthodontically aimed low-magnitude, high-frequency mechanical vibration (OLMHFMV) on intact calvarial bone, specifically the parietal and temporal, and cranial sutures, including the sagittal and parietotemporal, of rats in differing stages of growth and development. METHODS Forty Wistar rats were divided into 4 groups: 2 control groups and 2 OLMHFMV groups. Subsequently, 0.3 cN of force with a frequency of 30 Hz was applied as OLMHFMV on the temporomandibular joint region in the rats in the OLMHFMV-1 group, with the protocol of 20 min/d for 5 d/wk for 1 month, whereas the rats in the OLMHFMV-2 group received mechanical stimuli for 2 months with the same protocol. Morphometric and structural analyses, including suture width, cranial width and height, bone mineral density, bone volume/tissue volume, trabecular number, trabecular separation, and trabecular thickness analyses, were carried out using micro-computed tomography. RESULTS The width of the parietotemporal and sagittal sutures and the cranial height and width increased significantly by OLMHFMV (P <0.021). The structural analysis revealed that trabecular number and trabecular separation increased, whereas trabecular thickness decreased in the OLMHFMV groups compared with the control groups (P <0.048). Bone volume/tissue volume remained unchanged despite reducing the bone mineral density of the OLMHFMV groups. CONCLUSIONS OLMHFMV had a potential for modulating sutural and cranial growth in adolescent rats. OLMHFMV increased the structural quality of the temporal and parietal bones. These effects may have clinical implications as a treatment option for patients suffering from craniofacial anomalies such as craniosynostosis or a supportive approach for dentofacial orthodontic treatments.
Collapse
Affiliation(s)
- Mehmet Amuk
- Department of Oral and Maxillofacial Radiology, Faculty of Dentistry, Erciyes University, Kayseri, Turkey
| | - Nisa Gül Amuk
- Department of Orthodontics, Faculty of Dentistry, Erciyes University, Kayseri, Turkey.
| | - Zeynep Hacıoğlu
- Department of Orthodontics, Faculty of Dentistry, Erciyes University, Kayseri, Turkey
| |
Collapse
|
15
|
Little-Letsinger SE, Rubin J, Diekman B, Rubin CT, McGrath C, Pagnotti GM, Klett EL, Styner M. Exercise to Mend Aged-tissue Crosstalk in Bone Targeting Osteoporosis & Osteoarthritis. Semin Cell Dev Biol 2022; 123:22-35. [PMID: 34489173 PMCID: PMC8840966 DOI: 10.1016/j.semcdb.2021.08.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 08/16/2021] [Accepted: 08/19/2021] [Indexed: 12/16/2022]
Abstract
Aging induces alterations in bone structure and strength through a multitude of processes, exacerbating common aging- related diseases like osteoporosis and osteoarthritis. Cellular hallmarks of aging are examined, as related to bone and the marrow microenvironment, and ways in which these might contribute to a variety of age-related perturbations in osteoblasts, osteocytes, marrow adipocytes, chondrocytes, osteoclasts, and their respective progenitors. Cellular senescence, stem cell exhaustion, mitochondrial dysfunction, epigenetic and intracellular communication changes are central pathways and recognized as associated and potentially causal in aging. We focus on these in musculoskeletal system and highlight knowledge gaps in the literature regarding cellular and tissue crosstalk in bone, cartilage, and the bone marrow niche. While senolytics have been utilized to target aging pathways, here we propose non-pharmacologic, exercise-based interventions as prospective "senolytics" against aging effects on the skeleton. Increased bone mass and delayed onset or progression of osteoporosis and osteoarthritis are some of the recognized benefits of regular exercise across the lifespan. Further investigation is needed to delineate how cellular indicators of aging manifest in bone and the marrow niche and how altered cellular and tissue crosstalk impact disease progression, as well as consideration of exercise as a therapeutic modality, as a means to enhance discovery of bone-targeted therapies.
Collapse
Affiliation(s)
- SE Little-Letsinger
- Department of Medicine, Division of Endocrinology & Metabolism, University of North Carolina at Chapel Hill
| | - J Rubin
- Department of Medicine, Division of Endocrinology & Metabolism, University of North Carolina at Chapel Hill,North Carolina Diabetes Research Center (NCDRC), University of North Carolina at Chapel Hill,Department of Medicine, Thurston Arthritis Research Center (TARC), University of North Carolina at Chapel Hill
| | - B Diekman
- Department of Medicine, Thurston Arthritis Research Center (TARC), University of North Carolina at Chapel Hill,Joint Departments of Biomedical Engineering NC State & University of North Carolina at Chapel Hill
| | - CT Rubin
- Department of Biomedical Engineering, State University of New York at Stony Brook
| | - C McGrath
- Department of Medicine, Division of Endocrinology & Metabolism, University of North Carolina at Chapel Hill
| | - GM Pagnotti
- Dept of Endocrine, Neoplasia, and Hormonal Disorders, University Texas MD Anderson Cancer Center, Houston
| | - EL Klett
- Department of Medicine, Division of Endocrinology & Metabolism, University of North Carolina at Chapel Hill,Department of Nutrition, School of Public Health, University of North Carolina at Chapel Hill
| | - M Styner
- Department of Medicine, Division of Endocrinology & Metabolism, University of North Carolina at Chapel Hill,North Carolina Diabetes Research Center (NCDRC), University of North Carolina at Chapel Hill,Department of Medicine, Thurston Arthritis Research Center (TARC), University of North Carolina at Chapel Hill
| |
Collapse
|
16
|
Hamid HA, Sarmadi VH, Prasad V, Ramasamy R, Miskon A. Electromagnetic field exposure as a plausible approach to enhance the proliferation and differentiation of mesenchymal stem cells in clinically relevant scenarios. J Zhejiang Univ Sci B 2022; 23:42-57. [PMID: 35029087 PMCID: PMC8758935 DOI: 10.1631/jzus.b2100443] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Mesenchymal stem/stromal cell (MSC)-based therapy has been regarded as one of the most revolutionary breakthroughs in the history of modern medicine owing to its myriad of immunoregulatory and regenerative properties. With the rapid progress in the fields of osteo- and musculoskeletal therapies, the demand for MSC-based treatment modalities is becoming increasingly prominent. In this endeavor, researchers around the world have devised new and innovative techniques to support the proliferation of MSCs while minimizing the loss of hallmark features of stem cells. One such example is electromagnetic field (EMF) exposure, which is an alternative approach with promising potential. In this review, we present a critical discourse on the efficiency, practicability, and limitations of some of the relevant methods, with insurmountable evidence backing the implementation of EMF as a feasible strategy for the clinically relevant expansion of MSCs.
Collapse
Affiliation(s)
- Haslinda Abdul Hamid
- Bio-artificial Organ and Regenerative Medicine Unit, National Defense University of Malaysia, Kuala Lumpur 57000, Malaysia
| | - Vahid Hosseinpour Sarmadi
- Cellular and Molecular Research Centre, Iran University of Medical Sciences, Tehran 144961 4535, Iran.,Institutes of Regenerative Medicine, Iran University of Medical Sciences, Tehran 199671 4353, Iran
| | - Vivek Prasad
- Stem Cell and Immunity Research Group, Immunology Laboratory, Department of Pathology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia (UPM), Selangor 43400, Malaysia
| | - Rajesh Ramasamy
- Stem Cell and Immunity Research Group, Immunology Laboratory, Department of Pathology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia (UPM), Selangor 43400, Malaysia
| | - Azizi Miskon
- Bio-artificial Organ and Regenerative Medicine Unit, National Defense University of Malaysia, Kuala Lumpur 57000, Malaysia.
| |
Collapse
|
17
|
Ebid A, El-Boshy M, El-Shamy S, Thabet A, Abedalla M, Ali T. Long-term effect of full-body pulsed electromagnetic field and exercise protocol in the treatment of men with osteopenia or osteoporosis: A randomized placebo-controlled trial. F1000Res 2021; 10:649. [PMID: 34900231 PMCID: PMC8637238 DOI: 10.12688/f1000research.54519.3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/26/2021] [Indexed: 11/25/2022] Open
Abstract
Background: Osteoporosis is the most prevalent metabolic disease affecting bones.
Objective: To investigate the long-term effect of pulsed electromagnetic field (PEMF) combined with exercise protocol on bone mineral density (BMD) and bone markers in men with osteopenia or osteoporosis.
Methods: Ninety-five males with osteopenia or osteoporosis (mean age, 51.26 ± 2.41 years; mean height, 176 ± 2.02 cm; mean weight, 83.08 ± 2.60 kg; mean body–mass index (BMI), 26.08 ± 1.09 kg/m
2) participated in the study, and they were randomly assigned to one of three groups: Group 1 received a full-body PEMF and exercise protocol (PEMF +EX), Group 2 received a placebo full-body PEMF and exercise protocol (PPEMF +EX), and Group 3 received a full-body PEMF alone (PEMF). PEMF was applied for the whole body using a full-body mat three times per week for 12 weeks, with an exercise protocol that includes flexibility, aerobic exercise, strengthening, weight-bearing, and balance exercises followed by whole-body vibration (WBV) training. Outcome measures include BMD of total hip and lumbar spine and bone markers [serum osteocalcin (s-OC), Serum amino-terminal cross-linking telopeptide of type I collagen (s-NTX), Serum carboxy-terminal cross-linking telopeptide of type I collagen (s-CTX), Parathyroid hormones (PTH), Bone-specific Alkaline Phosphatase (BSAP), and 25-hydroxy vitamin D (Vit D)].
Results: The
BMD of total hip and lumbar spine was significantly increased post-treatment in all groups, and more so in Group 1 and Group 2 than Group 3. There was a significant difference in bone markers in all groups, more so in Group 1 and Group 2 than in Group 3.
Conclusion: PEMF combined with exercise protocol exerts a potent role for treating OP, is more effective than exercise and PEMF alone for increasing BMD and enhancing bone formation, and suppresses bone-resorption markers after 12-weeks of treatment with the impact lasting up to 6 months.
Collapse
Affiliation(s)
- Anwar Ebid
- Physical Therapy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Mohamed El-Boshy
- Laboratory Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | | | - Ali Thabet
- Physical Therapy, Umm Al-Qura University, Makkah, Saudi Arabia
| | | | - Tariq Ali
- Umm Al-Qura University Medical Center, Umm Al-Qura University, Makkah, Saudi Arabia
| |
Collapse
|
18
|
Kennedy Z, Newberg J, Goelzer M, Judex S, Fitzpatrick CK, Uzer G. Modeling stem cell nucleus mechanics using confocal microscopy. Biomech Model Mechanobiol 2021; 20:2361-2372. [PMID: 34424419 PMCID: PMC8599651 DOI: 10.1007/s10237-021-01513-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 08/14/2021] [Indexed: 11/24/2022]
Abstract
Nuclear mechanics is emerging as a key component of stem cell function and differentiation. While changes in nuclear structure can be visually imaged with confocal microscopy, mechanical characterization of the nucleus and its sub-cellular components require specialized testing equipment. A computational model permitting cell-specific mechanical information directly from confocal and atomic force microscopy of cell nuclei would be of great value. Here, we developed a computational framework for generating finite element models of isolated cell nuclei from multiple confocal microscopy scans and simple atomic force microscopy (AFM) tests. Confocal imaging stacks of isolated mesenchymal stem cells were converted into finite element models and siRNA-mediated Lamin A/C depletion isolated chromatin and Lamin A/C structures. Using AFM-measured experimental stiffness values, a set of conversion factors were determined for both chromatin and Lamin A/C to map the voxel intensity of the original images to the element stiffness, allowing the prediction of nuclear stiffness in an additional set of other nuclei. The developed computational framework will identify the contribution of a multitude of sub-nuclear structures and predict global nuclear stiffness of multiple nuclei based on simple nuclear isolation protocols, confocal images and AFM tests.
Collapse
Affiliation(s)
- Zeke Kennedy
- Department of Mechanical and Biomedical Engineering, Boise State University, 1910 University Drive, MSd-2085, Boise, ID, 83725-2085, USA
| | - Joshua Newberg
- Department of Mechanical and Biomedical Engineering, Boise State University, 1910 University Drive, MSd-2085, Boise, ID, 83725-2085, USA
| | - Matthew Goelzer
- Department of Mechanical and Biomedical Engineering, Boise State University, 1910 University Drive, MSd-2085, Boise, ID, 83725-2085, USA
| | - Stefan Judex
- Biomedical Engineering, Stony Brook University, Stony Brook, USA
| | - Clare K Fitzpatrick
- Department of Mechanical and Biomedical Engineering, Boise State University, 1910 University Drive, MSd-2085, Boise, ID, 83725-2085, USA
| | - Gunes Uzer
- Department of Mechanical and Biomedical Engineering, Boise State University, 1910 University Drive, MSd-2085, Boise, ID, 83725-2085, USA.
| |
Collapse
|
19
|
Abstract
The nuclear envelope and nucleoskeleton are emerging as signaling centers that regulate how physical information from the extracellular matrix is biochemically transduced into the nucleus, affecting chromatin and controlling cell function. Bone is a mechanically driven tissue that relies on physical information to maintain its physiological function and structure. Disorder that present with musculoskeletal and cardiac symptoms, such as Emery-Dreifuss muscular dystrophies and progeria, correlate with mutations in nuclear envelope proteins including Linker of Nucleoskeleton and Cytoskeleton (LINC) complex, Lamin A/C, and emerin. However, the role of nuclear envelope mechanobiology on bone function remains underexplored. The mesenchymal stem cell (MSC) model is perhaps the most studied relationship between bone regulation and nuclear envelope function. MSCs maintain the musculoskeletal system by differentiating into multiple cell types including osteocytes and adipocytes, thus supporting the bone's ability to respond to mechanical challenge. In this review, we will focus on how MSC function is regulated by mechanical challenges both in vitro and in vivo within the context of bone function specifically focusing on integrin, β-catenin and YAP/TAZ signaling. The importance of the nuclear envelope will be explored within the context of musculoskeletal diseases related to nuclear envelope protein mutations and nuclear envelope regulation of signaling pathways relevant to bone mechanobiology in vitro and in vivo.
Collapse
Affiliation(s)
- Scott Birks
- Boise State University, Micron School of Materials Science and Engineering, United States of America
| | - Gunes Uzer
- Boise State University, Mechanical and Biomedical Engineering, United States of America.
| |
Collapse
|
20
|
Lamin A/C Is Dispensable to Mechanical Repression of Adipogenesis. Int J Mol Sci 2021; 22:ijms22126580. [PMID: 34205295 PMCID: PMC8234021 DOI: 10.3390/ijms22126580] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/10/2021] [Accepted: 06/16/2021] [Indexed: 12/15/2022] Open
Abstract
Mesenchymal stem cells (MSCs) maintain the musculoskeletal system by differentiating into multiple lineages, including osteoblasts and adipocytes. Mechanical signals, including strain and low-intensity vibration (LIV), are important regulators of MSC differentiation via control exerted through the cell structure. Lamin A/C is a protein vital to the nuclear architecture that supports chromatin organization and differentiation and contributes to the mechanical integrity of the nucleus. We investigated whether lamin A/C and mechanoresponsiveness are functionally coupled during adipogenesis in MSCs. siRNA depletion of lamin A/C increased the nuclear area, height, and volume and decreased the circularity and stiffness. Lamin A/C depletion significantly decreased markers of adipogenesis (adiponectin, cellular lipid content) as did LIV treatment despite depletion of lamin A/C. Phosphorylation of focal adhesions in response to mechanical challenge was also preserved during loss of lamin A/C. RNA-seq showed no major adipogenic transcriptome changes resulting from LIV treatment, suggesting that LIV regulation of adipogenesis may not occur at the transcriptional level. We observed that during both lamin A/C depletion and LIV, interferon signaling was downregulated, suggesting potentially shared regulatory mechanism elements that could regulate protein translation. We conclude that the mechanoregulation of adipogenesis and the mechanical activation of focal adhesions function independently from those of lamin A/C.
Collapse
|
21
|
Thompson M, Woods K, Newberg J, Oxford JT, Uzer G. Low-intensity vibration restores nuclear YAP levels and acute YAP nuclear shuttling in mesenchymal stem cells subjected to simulated microgravity. NPJ Microgravity 2020; 6:35. [PMID: 33298964 PMCID: PMC7708987 DOI: 10.1038/s41526-020-00125-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 10/08/2020] [Indexed: 12/18/2022] Open
Abstract
Reducing the musculoskeletal deterioration that astronauts experience in microgravity requires countermeasures that can improve the effectiveness of otherwise rigorous and time-expensive exercise regimens in space. The ability of low-intensity vibrations (LIV) to activate force-responsive signaling pathways in cells suggests LIV as a potential countermeasure to improve cell responsiveness to subsequent mechanical challenge. Mechanoresponse of mesenchymal stem cells (MSC), which maintain bone-making osteoblasts, is in part controlled by the "mechanotransducer" protein YAP (Yes-associated protein), which is shuttled into the nucleus in response to cyto-mechanical forces. Here, using YAP nuclear shuttling as a measurement outcome, we tested the effect of 72 h of clinostat-induced simulated microgravity (SMG) and daily LIV application (LIVDT) on the YAP nuclear entry driven by either acute LIV (LIVAT) or Lysophosphohaditic acid (LPA), applied after the 72 h period. We hypothesized that SMG-induced impairment of acute YAP nuclear entry would be alleviated by the daily application of LIVDT. Results showed that while both acute LIVAT and LPA treatments increased nuclear YAP entry by 50 and 87% over the basal levels in SMG-treated MSCs, nuclear YAP levels of all SMG groups were significantly lower than non-SMG controls. LIVDT, applied in parallel to SMG, restored the SMG-driven decrease in basal nuclear YAP to control levels as well as increased the LPA-induced but not LIVAT-induced YAP nuclear entry over SMG only, counterparts. These cell-level observations suggest that daily LIV treatments are a feasible countermeasure for restoring basal nuclear YAP levels and increasing the YAP nuclear shuttling in MSCs under SMG.
Collapse
Affiliation(s)
- Matthew Thompson
- Mechanical and Biomedical Engineering, Boise State University, Boise, ID, USA
| | - Kali Woods
- Biomolecular Sciences Graduate Program, Boise State University, Boise, ID, USA
| | - Joshua Newberg
- Mechanical and Biomedical Engineering, Boise State University, Boise, ID, USA
| | - Julia Thom Oxford
- Biomolecular Sciences Graduate Program, Boise State University, Boise, ID, USA
| | - Gunes Uzer
- Mechanical and Biomedical Engineering, Boise State University, Boise, ID, USA.
| |
Collapse
|