1
|
Li D, Yong Y, Qiao C, Jiang H, Lin J, Wei J, Zhou Y, Li F. Low-Intensity Pulsed Ultrasound Dynamically Modulates the Migration of BV2 Microglia. ULTRASOUND IN MEDICINE & BIOLOGY 2025; 51:494-507. [PMID: 39632209 DOI: 10.1016/j.ultrasmedbio.2024.11.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 10/18/2024] [Accepted: 11/06/2024] [Indexed: 12/07/2024]
Abstract
OBJECTIVE Low-intensity pulsed ultrasound (LIPUS) is a promising modality for neuromodulation. Microglia are the resident immune cells in the brain and their mobility is critical for maintaining brain homeostasis and alleviating neuroimmune pathologies. However, it is unclear whether and how LIPUS modulates microglial migration in physiological conditions. METHODS Here we examined the in vitro effects of LIPUS on the mobility of BV2 microglia by live cell imaging. Single-cell tracing of BV2 microglia migration was analyzed using ImageJ and Chemotaxis and Migration Tool software. Pharmacological manipulation was performed to determine the key molecular players involved in regulating ultrasound-dependent microglia migration. RESULTS We found that the distance of microglial migration was enhanced by LIPUS with increasing acoustic pressure. Removing the extracellular Ca2+ influx or depletion of intracellular Ca2+ stores suppressed ultrasound-enhanced BV2 migration. Furthermore, we found that blocking the reorganization of actin, or suppressing purinergic signaling by application of apyrase or hemi-channel inhibitors, both diminished ultrasound-induced BV2 migration. LIPUS stimulation also enhanced microglial migration in a lipopolysaccharide (LPS)-induced inflammatory environment. CONCLUSION LIPUS promoted microglia migration in both physiological and inflammatory environments. Calcium, cytoskeleton, and purinergic signaling were involved in regulating ultrasound-dependent microglial mobility. Our study reveals the biomechanical impact of ultrasound on microglial migration and highlights the potential of using ultrasound-based tools for modulation of microglial function.
Collapse
Affiliation(s)
- Dandan Li
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, Shenzhen, China
| | - Yu Yong
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, Shenzhen, China
| | - Chaofeng Qiao
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, Shenzhen, China; School of Basic Medical Sciences, Beihua University, Jilin City, China
| | - Hao Jiang
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, Shenzhen, China
| | - Jiawei Lin
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, Shenzhen, China
| | - Jianpeng Wei
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, Shenzhen, China
| | - Yufeng Zhou
- Chongqing Medical University, State Key Laboratory of Ultrasound in Medicine and Engineering, Chongqing City, China
| | - Fenfang Li
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, Shenzhen, China.
| |
Collapse
|
2
|
Tabuchi Y, Kuroda K, Furusawa Y, Hirano T, Nagaoka R, Omura M, Hasegawa H, Hirayama J, Suzuki N. Genes involved in osteogenic differentiation induced by low‑intensity pulsed ultrasound in goldfish scales. Biomed Rep 2025; 22:18. [PMID: 39651404 PMCID: PMC11621920 DOI: 10.3892/br.2024.1896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 07/09/2024] [Indexed: 12/11/2024] Open
Abstract
The teleost scale is a unique calcified tissue that contains osteoclasts, osteoblasts, osteocytes and the bone matrix, similar to mammalian bone. Here, the effects of low-intensity pulsed ultrasound (LIPUS) on osteoblasts and osteoclasts in goldfish scales were investigated. Scales were treated with LIPUS, which is equivalent to use under clinical conditions (30 mW/cm2 for 20 min), then cultured at 15˚C. Alkaline phosphatase activity, a marker of osteoblasts, or tartrate-resistant acid phosphatase (TRAP) activity, a marker of osteoclasts was measured. The gene expression profile was examined using RNA-sequencing. Gene network and biological function analyses were performed using the Ingenuity® Pathways Knowledge Base. A single exposure of LIPUS significantly increased ALP activity but did not affect TRAP activity. These data indicated that LIPUS induced osteoblastic activation in goldfish scales. Using RNA-sequencing, numerous genes that were significantly and differentially expressed 3, 6, and 24 h after LIPUS exposure were observed. Ingenuity® pathway analysis demonstrated that three gene networks, GN-3h, GN-6h, and GN-24h, were obtained from upregulated genes at 3, 6 and 24 h culture, respectively, and included several genes associated with osteoblast differentiation, such as protein kinase D1, prostaglandin-endoperoxide synthase 2, TNFRSF11B (tumor necrosis factor receptor superfamily, member 11b) and WNT3A (Wnt family member 3A). A significant upregulation of expression levels of these genes in scales treated with LIPUS was confirmed by reverse transcription-quantitative polymerase chain reaction. These results contribute to elucidating the molecular mechanisms of osteoblast activation induced by LIPUS.
Collapse
Affiliation(s)
- Yoshiaki Tabuchi
- Division of Molecular Genetics Research, Life Science Research Center, University of Toyama, Toyama 930-0194, Japan
| | - Kouhei Kuroda
- Noto Marine Laboratory, Institute of Nature and Environmental Technology, Kanazawa University, Ishikawa 927-0553, Japan
| | - Yukihiro Furusawa
- Department of Pharmaceutical Engineering, Faculty of Engineering, Toyama Prefectural University, Toyama 939-0398, Japan
| | - Tetsushi Hirano
- Division of Molecular Genetics Research, Life Science Research Center, University of Toyama, Toyama 930-0194, Japan
| | - Ryo Nagaoka
- Laboratory of Medical Information Sensing, Faculty of Engineering, University of Toyama, Toyama 930-8555, Japan
| | - Masaaki Omura
- Laboratory of Medical Information Sensing, Faculty of Engineering, University of Toyama, Toyama 930-8555, Japan
| | - Hideyuki Hasegawa
- Laboratory of Medical Information Sensing, Faculty of Engineering, University of Toyama, Toyama 930-8555, Japan
| | - Jun Hirayama
- Department of Clinical Engineering, Faculty of Health Sciences, Komatsu University, Ishikawa 923-0961, Japan
| | - Nobuo Suzuki
- Noto Marine Laboratory, Institute of Nature and Environmental Technology, Kanazawa University, Ishikawa 927-0553, Japan
| |
Collapse
|
3
|
Han Y, Gao H, Gao J, Yang Y, He C. Low-intensity pulsed ultrasound regulates bone marrow mesenchymal stromal cells differentiation and inhibits bone loss by activating the IL-11-Wnt/β-catenin signaling pathway. Int Immunopharmacol 2024; 143:113380. [PMID: 39405933 DOI: 10.1016/j.intimp.2024.113380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 09/28/2024] [Accepted: 10/07/2024] [Indexed: 10/30/2024]
Abstract
BACKGROUND Osteoporosis (OP) is a common metabolic bone disease. Low-intensity pulsed ultrasound (LIPUS) can effectively promote bone formation and fracture healing. The Wnt/β-catenin signaling pathway is crucial for regulating bone homeostasis and bone diseases, and its downregulation is one of the main mechanisms of osteoporosis pathogenesis. Interleukin-11 (IL-11), which is regulated by mechanical stress, is a key factor in bone remodeling. Here, we investigated the optimal intervention parameters for LIPUS, the relationships among LIPUS, IL-11, and the Wnt/β-catenin signaling pathway, and the effects of LIPUS on bone loss and potential molecular mechanisms in ovariectomized (OVX) mice. METHODS Bone marrow mesenchymal stromal cells (BMSCs) were subjected to LIPUS intervention for 0, 10, or 20 min to determine the optimal intervention time. The mediating role of IL-11 in LIPUS intervention was explored through IL-11 knockdown and overexpression. Finally, animal experiments were conducted to investigate the in vivo therapeutic effects of LIPUS. RESULTS The optimal intervention time for LIPUS was 20 min. LIPUS promoted IL-11 expression and upregulated the Wnt/β-catenin signaling pathway, thereby promoting osteogenic differentiation and inhibiting adipogenic differentiation of BMSCs. IL-11 mediates the regulation of the Wnt/β-catenin signaling pathway by LIPUS. Additionally, LIPUS effectively improved the bone microstructure in ovariectomized mice, inhibited bone loss, promoted IL-11 expression in bone tissue, and activated the Wnt/β-catenin signaling pathway in the femur. CONCLUSION Low-intensity pulsed ultrasound can regulate BMSCs differentiation and inhibit bone loss by promoting IL-11 expression and activating the Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Yijing Han
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China, 610000; Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, Sichuan, China, 610000
| | - Hui Gao
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China, 610000; Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, Sichuan, China, 610000
| | - Jing Gao
- LIFU Medical Research Center, Sichuan Taiyou Technology Co., Ltd., Chengdu, Sichuan, China, 610000
| | - Yonghong Yang
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China, 610000; Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, Sichuan, China, 610000.
| | - Chengqi He
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China, 610000; Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, Sichuan, China, 610000.
| |
Collapse
|
4
|
Zamfirov L, Nguyen NM, Fernández-Sánchez ME, Cambronera Ghiglione P, Teston E, Dizeux A, Tiennot T, Farge E, Demené C, Tanter M. Acoustic-pressure-driven ultrasonic activation of the mechanosensitive receptor RET and of cell proliferation in colonic tissue. Nat Biomed Eng 2024:10.1038/s41551-024-01300-9. [PMID: 39706982 DOI: 10.1038/s41551-024-01300-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 10/31/2024] [Indexed: 12/23/2024]
Abstract
Ultrasound generates both compressive and shear mechanical forces in soft tissues. However, the specific mechanisms by which these forces activate cellular processes remain unclear. Here we show that low-intensity focused ultrasound can activate the mechanosensitive RET signalling pathway. Specifically, in mouse colon tissues ex vivo and in vivo, focused ultrasound induced RET phosphorylation in colonic crypts cells, which correlated with markers of proliferation and stemness when using hours-long insonication. The activation of the RET pathway is non-thermal, is linearly related to acoustic pressure and is independent of radiation-force-induced shear strain in tissue. Our findings suggest that ultrasound could be used to regulate cell proliferation, particularly in the context of regenerative medicine, and highlight the importance of adhering to current ultrasound-safety regulations for medical imaging.
Collapse
Affiliation(s)
- Laura Zamfirov
- Institute Physics for Medicine Paris, ESPCI PSL Paris, INSERM U1273, CNRS UMR 8361, Paris Sciences et Lettres University, Paris, France
| | - Ngoc-Minh Nguyen
- Mechanics and Genetics of Embryonic and Tumoral Development team, Physics of Cells and Cancer UMR168, INSERM, Université PSL, Sorbonne Université, Institut Curie, Paris, France
| | - Maria Elena Fernández-Sánchez
- Mechanics and Genetics of Embryonic and Tumoral Development team, Physics of Cells and Cancer UMR168, INSERM, Université PSL, Sorbonne Université, Institut Curie, Paris, France
| | - Paula Cambronera Ghiglione
- Mechanics and Genetics of Embryonic and Tumoral Development team, Physics of Cells and Cancer UMR168, INSERM, Université PSL, Sorbonne Université, Institut Curie, Paris, France
| | - Eliott Teston
- Institute Physics for Medicine Paris, ESPCI PSL Paris, INSERM U1273, CNRS UMR 8361, Paris Sciences et Lettres University, Paris, France
| | - Alexandre Dizeux
- Institute Physics for Medicine Paris, ESPCI PSL Paris, INSERM U1273, CNRS UMR 8361, Paris Sciences et Lettres University, Paris, France
| | - Thomas Tiennot
- Institute Physics for Medicine Paris, ESPCI PSL Paris, INSERM U1273, CNRS UMR 8361, Paris Sciences et Lettres University, Paris, France
| | - Emmanuel Farge
- Mechanics and Genetics of Embryonic and Tumoral Development team, Physics of Cells and Cancer UMR168, INSERM, Université PSL, Sorbonne Université, Institut Curie, Paris, France.
| | - Charlie Demené
- Institute Physics for Medicine Paris, ESPCI PSL Paris, INSERM U1273, CNRS UMR 8361, Paris Sciences et Lettres University, Paris, France.
| | - Mickael Tanter
- Institute Physics for Medicine Paris, ESPCI PSL Paris, INSERM U1273, CNRS UMR 8361, Paris Sciences et Lettres University, Paris, France.
| |
Collapse
|
5
|
Sahni J, McCue IS, Johnson AR, Schake MA, Sotelo LD, Turner JA, Pedrigi RM. Ultrasound Induces Similar Temporal Endothelial Expression Patterns of eNOS and KLF2 as Normal Flow. ULTRASOUND IN MEDICINE & BIOLOGY 2024; 50:1893-1902. [PMID: 39306482 PMCID: PMC11490374 DOI: 10.1016/j.ultrasmedbio.2024.08.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 07/22/2024] [Accepted: 08/23/2024] [Indexed: 10/21/2024]
Abstract
OBJECTIVE To determine the sensitivity of vascular endothelial cells to long durations of low-intensity pulsed ultrasound (LIPUS) compared to normal flow and identify the duration that maximizes expression of two mechanosensitive genes related to healthy endothelial function, endothelial nitric oxide synthase (eNOS) and Krüppel-like factor 2 (KLF2). METHODS Custom ultrasound exposure tanks were developed and the acoustic field was characterized. Human umbilical vein endothelial cells were seeded into culture plates and exposed to LIPUS at a frequency of 1 MHz and acoustic pressure of 217 kPa for 20 min, 1 h, 6 h, 9 h, or 24 h. As a comparator, other cells were exposed to normal flow. RT-qPCR was used to assess mRNA expression of eNOS and KLF2. RESULTS Maximum eNOS and KLF2 expression occurred at 6 h and was localized to the beam path. Both genes exhibited qualitatively similar patterns of expression under LIPUS compared to normal flow. LIPUS induced a more rapid beneficial response compared to normal flow, but flow induced higher expression of both genes. eNOS expression after 6 h of LIPUS was dependent on RNA yield and culture duration prior to experiments. CONCLUSION Endothelial cells exposed to longer durations of LIPUS than typically employed exhibited greater expression of beneficial genes. The temporal gene expression patterns resulting from LIPUS and normal flow suggest activation of similar signaling pathways. However, LIPUS also caused increased RNA yield that may be linked to proliferation, which would suggest more of a wound healing than atheroprotective phenotype.
Collapse
Affiliation(s)
- Jaideep Sahni
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Ian S McCue
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Adam R Johnson
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Morgan A Schake
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Luz D Sotelo
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Joseph A Turner
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Ryan M Pedrigi
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, USA; Nebraska Center for Integrated Biomolecular Communication, University of Nebraska-Lincoln, Lincoln, NE, USA; Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
6
|
Sarkar AK, Sarkar AR, Sahoo R, Jana NR, Jana NR. Designed Nanodrugs for Ultrasonic Removal of Toxic Polyglutamine Aggregates from Neuron Cells. NANO LETTERS 2024; 24:13473-13480. [PMID: 39413815 DOI: 10.1021/acs.nanolett.4c02243] [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: 10/18/2024]
Abstract
Clearing of toxic polyglutamine aggregates from neuronal cells is crucial for ameliorating Huntington's disease. However, such clearance is challenging, requiring the targeting of affected neuron cells in the brain, followed by the removal of polyglutamine from cells. Here we report a designed nanodrug that can be used for the ultrasound-based removal of toxic polyglutamine aggregates from neuron cells. The nanodrug is composed of a sonosensitizer molecule, chlorin e6- or protoporphyrin IX-loaded polymer micelle of 20-30 nm in size that rapidly delivers the sonosensitizer into the cell nucleus. Ultrasound exposure of these cells generates singlet oxygen in the nucleus/perinuclear region that induces strong autophagic flux and clears toxic polyglutamine aggregates from cells. It has been demonstrated that the nanodrug and ultrasound treatment can enhance the cell survival against polyglutamine aggregates by 4 times. This result suggests that the nanodrug can be designed for focused ultrasound-based wireless treatment of various neurodegenerative diseases.
Collapse
Affiliation(s)
- Ankan Kumar Sarkar
- School of Materials Sciences, Indian Association for the Cultivation of Science, Kolkata-700032, India
| | - Abu Raihan Sarkar
- School of Materials Sciences, Indian Association for the Cultivation of Science, Kolkata-700032, India
| | - Rajkumar Sahoo
- School of Materials Sciences, Indian Association for the Cultivation of Science, Kolkata-700032, India
| | - Nihar R Jana
- Department of Bioscience and Biotechnology, Indian Institute of Technology, Kharagpur-721302, India
| | - Nikhil R Jana
- School of Materials Sciences, Indian Association for the Cultivation of Science, Kolkata-700032, India
| |
Collapse
|
7
|
Xiong H, Cao M, Yu Y, Duan X, Sun L, Tang L, Fan X. Study on the Effects of Low-Intensity Pulsed Ultrasound and Iron Ions for Proliferation and Differentiation of Osteoblasts. ULTRASOUND IN MEDICINE & BIOLOGY 2024:S0301-5629(24)00265-5. [PMID: 39209558 DOI: 10.1016/j.ultrasmedbio.2024.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Revised: 06/25/2024] [Accepted: 07/08/2024] [Indexed: 09/04/2024]
Abstract
OBJECTIVE This study involved the proliferation and differentiation of osteoblasts treated with low-intensity pulsed ultrasound (LIPUS) and iron (Fe3+) ions, respectively. The biological effects of LIPUS and Fe3+ ions on the proliferation and differentiation of osteoblasts were also evaluated. METHODS MC3T3-E1 cells were seeded in six-well plates with the medium, which contained different concentrations of Fe3+ (0, 100, 200, 300, 400, 500, 600 and 700 μg L-1, respectively). LIPUS treatment was directed at the bottom of the plate for 20 min at an intensity of 80 mW cm-2 every day. RESULTS Viability results showed that a dose of 400 μg L-1 Fe3+ ions had the best effect at promoting osteogenic proliferation in cell culture. The results of alkaline phosphatase staining and mineralization indicated that the differentiation of osteoblasts was promoted by LIPUS and Fe3+ ions. Fluorescence staining results showed that the number of cell nuclei in the LIPUS, Fe3+ and LIPUS-Fe groups increased by 37.20%, 55.81% and 89.76%, respectively. Migration data indicated that migration and proliferation rates were increased by LIPUS and Fe3+, and the results of protein expression indicated that LIPUS and Fe3+ may increase the expression of Wnt, β-catenin, and Runx2, hence promoting normal bone regeneration and development. CONCLUSION The combination of LIPUS (1.5 MHz, 80 mW cm-2) and Fe3+ accelerates the proliferation and differentiation of osteoblasts significantly compared with single-factor treatment (stimulated by LIPUS and Fe3+ ions, respectively). This study could establish a foundation for LIPUS-responsive biomaterials in the repair and regeneration of bone tissues.
Collapse
Affiliation(s)
- Huanbin Xiong
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, China
| | - Mengshu Cao
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, China
| | - Yanan Yu
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, China
| | - Xueyou Duan
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, China
| | - Lijun Sun
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, China
| | - Liang Tang
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, China
| | - Xiushan Fan
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, China.
| |
Collapse
|
8
|
Williams JA, Campsie P, Gibson R, Johnson-Love O, Werner A, Sprott M, Meechan R, Huesa C, Windmill JFC, Purcell M, Coupaud S, Dalby MJ, Childs P, Riddell JS, Reid S. Developing and Investigating a Nanovibration Intervention for the Prevention/Reversal of Bone Loss Following Spinal Cord Injury. ACS NANO 2024; 18:17630-17641. [PMID: 38924391 PMCID: PMC11238619 DOI: 10.1021/acsnano.4c02104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 06/13/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024]
Abstract
Osteoporosis disrupts the fine-tuned balance between bone formation and resorption, leading to reductions in bone quantity and quality and ultimately increasing fracture risk. Prevention and treatment of osteoporotic fractures is essential for reductions in mortality, morbidity, and the economic burden, particularly considering the aging global population. Extreme bone loss that mimics time-accelerated osteoporosis develops in the paralyzed limbs following complete spinal cord injury (SCI). In vitro nanoscale vibration (1 kHz, 30 or 90 nm amplitude) has been shown to drive differentiation of mesenchymal stem cells toward osteoblast-like phenotypes, enhancing osteogenesis and inhibiting osteoclastogenesis simultaneously. Here, we develop and characterize a wearable device designed to deliver and monitor continuous nanoamplitude vibration to the hindlimb long bones of rats with complete SCI. We investigate whether a clinically feasible dose of nanovibration (two 2 h/day, 5 days/week for 6 weeks) is effective at reversing the established SCI-induced osteoporosis. Laser interferometry and finite element analysis confirmed transmission of nanovibration into the bone, and microcomputed tomography and serum bone formation and resorption markers assessed effectiveness. The intervention did not reverse SCI-induced osteoporosis. However, serum analysis indicated an elevated concentration of the bone formation marker procollagen type 1 N-terminal propeptide (P1NP) in rats receiving 40 nm amplitude nanovibration, suggesting increased synthesis of type 1 collagen, the major organic component of bone. Therefore, enhanced doses of nanovibrational stimulus may yet prove beneficial in attenuating/reversing osteoporosis, particularly in less severe forms of osteoporosis.
Collapse
Affiliation(s)
- Jonathan A. Williams
- Centre
for the Cellular Microenvironment, Department of Biomedical Engineering,
Wolfson Centre, University of Strathclyde, Glasgow G4 0NW, U.K.
- School
of Psychology and Neuroscience, College of Medical, Veterinary and
Life Sciences, University of Glasgow, Glasgow G12 8QQ, U.K.
- Scottish
Centre for Innovation in Spinal Cord Injury, Queen Elizabeth National
Spinal Injuries Unit, Queen Elizabeth University
Hospital, Glasgow G51 4TF, U.K.
| | - Paul Campsie
- Centre
for the Cellular Microenvironment, Department of Biomedical Engineering,
Wolfson Centre, University of Strathclyde, Glasgow G4 0NW, U.K.
| | - Richard Gibson
- Centre
for the Cellular Microenvironment, Department of Biomedical Engineering,
Wolfson Centre, University of Strathclyde, Glasgow G4 0NW, U.K.
| | - Olivia Johnson-Love
- Centre
for the Cellular Microenvironment, Department of Biomedical Engineering,
Wolfson Centre, University of Strathclyde, Glasgow G4 0NW, U.K.
| | - Anna Werner
- Centre
for the Cellular Microenvironment, Department of Biomedical Engineering,
Wolfson Centre, University of Strathclyde, Glasgow G4 0NW, U.K.
| | - Mark Sprott
- Centre
for the Cellular Microenvironment, Institute of Molecular, Cell and
Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, U.K.
| | - Ryan Meechan
- Centre
for the Cellular Microenvironment, Department of Biomedical Engineering,
Wolfson Centre, University of Strathclyde, Glasgow G4 0NW, U.K.
| | - Carmen Huesa
- Institute
of Infection, Immunity and Inflammation, College of Medical, Veterinary
and Life Sciences, University of Glasgow, Glasgow G12 8QQ, U.K.
| | - James F. C. Windmill
- Department
of Electronic and Electrical Engineering, Royal College Building, University of Strathclyde, Glasgow G1 1XW, U.K.
| | - Mariel Purcell
- Scottish
Centre for Innovation in Spinal Cord Injury, Queen Elizabeth National
Spinal Injuries Unit, Queen Elizabeth University
Hospital, Glasgow G51 4TF, U.K.
| | - Sylvie Coupaud
- Centre
for the Cellular Microenvironment, Department of Biomedical Engineering,
Wolfson Centre, University of Strathclyde, Glasgow G4 0NW, U.K.
- Scottish
Centre for Innovation in Spinal Cord Injury, Queen Elizabeth National
Spinal Injuries Unit, Queen Elizabeth University
Hospital, Glasgow G51 4TF, U.K.
| | - Matthew J. Dalby
- Centre
for the Cellular Microenvironment, Institute of Molecular, Cell and
Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, U.K.
| | - Peter Childs
- Centre
for the Cellular Microenvironment, Department of Biomedical Engineering,
Wolfson Centre, University of Strathclyde, Glasgow G4 0NW, U.K.
| | - John S. Riddell
- School
of Psychology and Neuroscience, College of Medical, Veterinary and
Life Sciences, University of Glasgow, Glasgow G12 8QQ, U.K.
| | - Stuart Reid
- Centre
for the Cellular Microenvironment, Department of Biomedical Engineering,
Wolfson Centre, University of Strathclyde, Glasgow G4 0NW, U.K.
| |
Collapse
|
9
|
Guo X, Lv M, Lin J, Guo J, Lin J, Li S, Sun Y, Zhang X. Latest Progress of LIPUS in Fracture Healing: A Mini-Review. JOURNAL OF ULTRASOUND IN MEDICINE : OFFICIAL JOURNAL OF THE AMERICAN INSTITUTE OF ULTRASOUND IN MEDICINE 2024; 43:643-655. [PMID: 38224522 DOI: 10.1002/jum.16403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 12/09/2023] [Accepted: 12/17/2023] [Indexed: 01/17/2024]
Abstract
The use of low-intensity pulsed ultrasound (LIPUS) for promoting fracture healing has been Food and Drug Administration (FDA)-approved since 1994 due to largely its non-thermal effects of sound flow sound radiation force and so on. Numerous clinical and animal studies have shown that LIPUS can accelerate the healing of fresh fractures, nonunions, and delayed unions in pulse mode regardless of LIPUS devices or circumstantial factors. Rare clinical studies show limitations of LIPUS for treating fractures with intramedullary nail fixation or low patient compliance. The biological effect is achieved by regulating various cellular behaviors involving mesenchymal stem/stromal cells (MSCs), osteoblasts, chondrocytes, and osteoclasts and with dose dependency on LIPUS intensity and time. Specifically, LIPUS promotes the osteogenic differentiation of MSCs through the ROCK-Cot/Tpl2-MEK-ERK signaling. Osteoblasts, in turn, respond to the mechanical signal of LIPUS through integrin, angiotensin type 1 (AT1), and PIEZO1 mechano-receptors, leading to the production of inflammatory factors such as COX-2, MCP-1, and MIP-1β fracture repair. LIPUS also induces CCN2 expression in chondrocytes thereby coordinating bone regeneration. Finally, LIPUS suppresses osteoclast differentiation and gene expression by interfering with the ERK/c-Fos/NFATc1 cascade. This mini-review revisits the known effects and mechanisms of LIPUS on bone fracture healing and strengthens the need for further investigation into the underlying mechanisms.
Collapse
Affiliation(s)
- Xin Guo
- School of Rehabilitation Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
- Department of Sports Medicine and Rehabilitation, Peking University Shenzhen Hospital, Shenzhen, China
| | - Maojiang Lv
- Department of Sports Medicine and Rehabilitation, Peking University Shenzhen Hospital, Shenzhen, China
- Zun Yi Medical University, Zhuhai, China
| | - Jie Lin
- Department of Joint Laboratory for Translational Medicine Research, Liaocheng People's Hospital, Liaocheng, China
| | - Jiang Guo
- Department of Sports Medicine and Rehabilitation, Peking University Shenzhen Hospital, Shenzhen, China
| | - Jianjing Lin
- Department of Sports Medicine and Rehabilitation, Peking University Shenzhen Hospital, Shenzhen, China
| | - Shun Li
- Department of Sports Medicine and Rehabilitation, Peking University Shenzhen Hospital, Shenzhen, China
| | - Yi Sun
- Department of Sports Medicine and Rehabilitation, Peking University Shenzhen Hospital, Shenzhen, China
- Department of Orthopaedics and Traumatology, The University of Hong Kong, Hong Kong SAR, China
| | - Xintao Zhang
- School of Rehabilitation Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
- Department of Sports Medicine and Rehabilitation, Peking University Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
10
|
Volarić D, Žauhar G, Chen J, Jerbić Radetić AT, Omrčen H, Raič A, Pirović R, Cvijanović Peloza O. The Effect of Low-Intensity Pulsed Ultrasound on Bone Regeneration and the Expression of Osterix and Cyclooxygenase-2 during Critical-Size Bone Defect Repair. Int J Mol Sci 2024; 25:3882. [PMID: 38612693 PMCID: PMC11012169 DOI: 10.3390/ijms25073882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 03/23/2024] [Accepted: 03/29/2024] [Indexed: 04/14/2024] Open
Abstract
Low-intensity pulsed ultrasound (LIPUS) is a form of ultrasound that utilizes low-intensity pulsed waves. Its effect on bones that heal by intramembranous ossification has not been sufficiently investigated. In this study, we examined LIPUS and the autologous bone, to determine their effect on the healing of the critical-size bone defect (CSBD) of the rat calvaria. The bone samples underwent histological, histomorphometric and immunohistochemical analyses. Both LIPUS and autologous bone promoted osteogenesis, leading to almost complete closure of the bone defect. On day 30, the bone volume was the highest in the autologous bone group (20.35%), followed by the LIPUS group (19.12%), and the lowest value was in the control group (5.11%). The autologous bone group exhibited the highest intensities of COX-2 (167.7 ± 1.1) and Osx (177.1 ± 0.9) expression on day 30. In the LIPUS group, the highest intensity of COX-2 expression was found on day 7 (169.7 ±1.6) and day 15 (92.7 ± 2.2), while the highest Osx expression was on day 7 (131.9 ± 0.9). In conclusion, this study suggests that LIPUS could represent a viable alternative to autologous bone grafts in repairing bone defects that are ossified by intramembranous ossification.
Collapse
Affiliation(s)
- Darian Volarić
- Department of Physical Medicine and Rehabilitation, Thalassotherapia Crikvenica—Special Hospital for Medical Rehabilitation, Gajevo Šetalište 21, 51260 Crikvenica, Croatia;
- Doctoral School of Biomedicine and Health, Faculty of Medicine, University of Rijeka, Braće Branchetta 20/1, 51000 Rijeka, Croatia
| | - Gordana Žauhar
- Department of Medical Physics and Biophysics, Faculty of Medicine, University of Rijeka, Braće Branchetta 20/1, 51000 Rijeka, Croatia
- Faculty of Physics, University of Rijeka, Radmile Matejčić 2, 51000 Rijeka, Croatia
| | - Jie Chen
- Department of Electrical and Computer Engineering, University of Alberta, Edmonton, AB T6G 2V4, Canada;
- Academy for Engineering and Technology, Fudan University, Shanghai 200433, China
| | - Ana Terezija Jerbić Radetić
- Department of Anatomy, Faculty of Medicine, University of Rijeka, Braće Branchetta 20/1, 51000 Rijeka, Croatia; (A.T.J.R.); (O.C.P.)
| | - Hrvoje Omrčen
- Department of Clinical Microbiology, Teaching Institute of Public Health of Primorsko-Goranska County, Krešimirova 52a, 51000 Rijeka, Croatia;
| | - Antonio Raič
- University Integrated Undergraduate and Graduate Study Programme of Medicine, Faculty of Medicine, University of Rijeka, Braće Branchetta 20/1, 51000 Rijeka, Croatia; (A.R.); (R.P.)
| | - Roko Pirović
- University Integrated Undergraduate and Graduate Study Programme of Medicine, Faculty of Medicine, University of Rijeka, Braće Branchetta 20/1, 51000 Rijeka, Croatia; (A.R.); (R.P.)
| | - Olga Cvijanović Peloza
- Department of Anatomy, Faculty of Medicine, University of Rijeka, Braće Branchetta 20/1, 51000 Rijeka, Croatia; (A.T.J.R.); (O.C.P.)
| |
Collapse
|
11
|
Kong Q, Wang Y, Jiang N, Wang Y, Wang R, Hu X, Mao J, Shi X. Exosomes as Promising Therapeutic Tools for Regenerative Endodontic Therapy. Biomolecules 2024; 14:330. [PMID: 38540750 PMCID: PMC10967740 DOI: 10.3390/biom14030330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 03/03/2024] [Accepted: 03/08/2024] [Indexed: 11/06/2024] Open
Abstract
Pulpitis is a common and frequent disease in dental clinics. Although vital pulp therapy and root canal treatment can stop the progression of inflammation, they do not allow for genuine structural regeneration and functional reconstruction of the pulp-dentin complex. In recent years, with the development of tissue engineering and regenerative medicine, research on stem cell-based regenerative endodontic therapy (RET) has achieved satisfactory preliminary results, significantly enhancing its clinical translational prospects. As one of the crucial paracrine effectors, the roles and functions of exosomes in pulp-dentin complex regeneration have gained considerable attention. Due to their advantages of cost-effectiveness, extensive sources, favorable biocompatibility, and high safety, exosomes are considered promising therapeutic tools to promote dental pulp regeneration. Accordingly, in this article, we first focus on the biological properties of exosomes, including their biogenesis, uptake, isolation, and characterization. Then, from the perspectives of cell proliferation, migration, odontogenesis, angiogenesis, and neurogenesis, we aim to reveal the roles and mechanisms of exosomes involved in regenerative endodontics. Lastly, immense efforts are made to illustrate the clinical strategies and influencing factors of exosomes applied in dental pulp regeneration, such as types of parental cells, culture conditions of parent cells, exosome concentrations, and scaffold materials, in an attempt to lay a solid foundation for exploring and facilitating the therapeutic strategy of exosome-based regenerative endodontic procedures.
Collapse
Affiliation(s)
- Qingyue Kong
- Center of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Q.K.); (Y.W.); (Y.W.); (R.W.)
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Yujie Wang
- Center of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Q.K.); (Y.W.); (Y.W.); (R.W.)
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Nan Jiang
- Central Laboratory, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing 100081, China;
| | - Yifan Wang
- Center of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Q.K.); (Y.W.); (Y.W.); (R.W.)
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Rui Wang
- Center of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Q.K.); (Y.W.); (Y.W.); (R.W.)
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Xiaohan Hu
- Outpatient Department Office, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China;
| | - Jing Mao
- Center of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Q.K.); (Y.W.); (Y.W.); (R.W.)
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Xin Shi
- Center of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Q.K.); (Y.W.); (Y.W.); (R.W.)
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| |
Collapse
|
12
|
Doveri E, Majnooni M, Guivier-Curien C, Baron C, Lasaygues P. Computational model to address lens-based acoustic field aperture in the in vitro ultrasonic cell stimulation. ULTRASONICS 2024; 138:107226. [PMID: 38103352 DOI: 10.1016/j.ultras.2023.107226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 11/10/2023] [Accepted: 12/13/2023] [Indexed: 12/19/2023]
Abstract
Low-Intensity Pulsed Ultrasound Stimulation (LIPUS) is a therapeutic modality used for bone tissue regeneration and healing. Its clinical efficacy is still debated, as the underlying physical phenomena remain poorly understood. The interaction between ultrasonic waves and cells, likely to trigger mechanotransduction inducing bone regeneration, is at the center of scientific concerns on the subject. In order to get new insights into these phenomena, the development of in vitro experiments is a key step but special attentions should be paid concerning to the actual acoustic area covered that has to be sufficiently large and homogeneous. To address this issue, an acoustic lens can be placed on the transducer to improve the homogeneity of the acoustic field over the entire cell culture area. A computational model is developed to test several shapes and heights of acoustic lenses and compare their effectiveness in order to find a compromise between the surface covered, the homogeneity of the intensity distribution and the acoustic pressure loss. All the lenses studied improve the enlargement of the field and its homogeneity but they all generate pressure acoustic loss. The best performing lens in terms of field homogeneity is the one that minimizes pressure acoustic loss but covers only 22% of the target surface. The best enlargement (68% of the surface covered) is obtained for a lens that produces a field that is 4 times less homogeneous and 3 times less efficient in terms of pressure acoustic loss. As no one lens is ideal, the choice of the lens should be the result of a compromise taking into account the prioritization of criteria.
Collapse
Affiliation(s)
- Elise Doveri
- Aix Marseille Univ, CNRS, Centrale Marseille, LMA UMR 7031, 4 impasse Nikola Tesla, 13453, Marseille, France.
| | - Meysam Majnooni
- Aix Marseille Univ, CNRS, Centrale Marseille, IRPHE UMR 7342, 49 rue Frédéric Joliot-Curie, 13384, Marseille, France.
| | - Carine Guivier-Curien
- Aix Marseille Univ, CNRS, Centrale Marseille, IRPHE UMR 7342, 49 rue Frédéric Joliot-Curie, 13384, Marseille, France.
| | - Cécile Baron
- Aix Marseille Univ, CNRS, Centrale Marseille, IRPHE UMR 7342, 49 rue Frédéric Joliot-Curie, 13384, Marseille, France.
| | - Philippe Lasaygues
- Aix Marseille Univ, CNRS, Centrale Marseille, LMA UMR 7031, 4 impasse Nikola Tesla, 13453, Marseille, France.
| |
Collapse
|
13
|
Fu S, Guo Z, Xu X, Li Y, Choi S, Zhao P, Shen W, Gao F, Wang C, Chen S, Li Y, Tian J, Sun P. Protective effect of low-intensity pulsed ultrasound on immune checkpoint inhibitor-related myocarditis via fine-tuning CD4 + T-cell differentiation. Cancer Immunol Immunother 2024; 73:15. [PMID: 38236243 PMCID: PMC10796578 DOI: 10.1007/s00262-023-03590-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 12/06/2023] [Indexed: 01/19/2024]
Abstract
PURPOSE Immune checkpoint inhibitors (ICIs) have transformed traditional cancer treatments. Specifically, ICI-related myocarditis is an immune-related adverse event (irAE) with high mortality. ICIs activate CD4+ T-lymphocyte reprogramming, causing an imbalance between Th17 and Treg cell differentiation, ultimately leading to myocardial inflammatory damage. Low-intensity pulsed ultrasound (LIPUS) can limit inflammatory responses, with positive therapeutic effects across various cardiovascular inflammatory diseases; however, its role in the pathogenesis of ICI-related myocarditis and CD4+ T-cell dysfunction remains unclear. Accordingly, this study investigated whether LIPUS can alleviate ICI-related myocarditis inflammatory damage and, if so, aimed to elucidate the beneficial effects of LIPUS and its underlying molecular mechanisms. METHODS An in vivo model of ICI-related myocarditis was obtained by intraperitonially injecting male A/J mice with an InVivoPlus anti-mouse PD-1 inhibitor. LIPUS treatment was performed via an ultrasound-guided application to the heart via the chest wall. The echocardiographic parameters were observed and cardiac function was assessed using an in vivo imaging system. The expression of core components of the HIPPO pathway was analyzed via western blotting. RESULTS LIPUS treatment reduced cardiac immune responses and inflammatory cardiac injury. Further, LIPUS treatment alleviated the inflammatory response in mice with ICI-related myocarditis. Mechanistically, in the HIPPO pathway, the activation of Mst1-TAZ axis improved autoimmune inflammation by altering the interaction between the transcription factors FOXP3 and RORγt and regulating the differentiation of Treg and Th17 cells. CONCLUSION LIPUS therapy was shown to reduce ICI-related myocarditis inflammatory damage and improve cardiac function, representing an exciting finding for irAEs treatment.
Collapse
Affiliation(s)
- Shuai Fu
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
- Ultrasound Molecular Imaging Joint Laboratory of Heilongjiang Province, Harbin, Heilongjiang Province, China
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang Province, China
| | - Zihong Guo
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
- Ultrasound Molecular Imaging Joint Laboratory of Heilongjiang Province, Harbin, Heilongjiang Province, China
| | - Xiangli Xu
- Department of Ultrasound, The Second Hospital of Harbin, Harbin, Heilongjiang Province, China
| | - Yifei Li
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
- Ultrasound Molecular Imaging Joint Laboratory of Heilongjiang Province, Harbin, Heilongjiang Province, China
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang Province, China
| | - Stephen Choi
- SXULTRASONIC Ltd. Kerry Rehabilitation Medicine Research Institute, Shenzhen, Guangdong Province, China
| | - Peng Zhao
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
- Ultrasound Molecular Imaging Joint Laboratory of Heilongjiang Province, Harbin, Heilongjiang Province, China
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang Province, China
| | - Wenqian Shen
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Fei Gao
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
- Ultrasound Molecular Imaging Joint Laboratory of Heilongjiang Province, Harbin, Heilongjiang Province, China
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang Province, China
| | - Chao Wang
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
- Ultrasound Molecular Imaging Joint Laboratory of Heilongjiang Province, Harbin, Heilongjiang Province, China
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang Province, China
| | - Shuang Chen
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
- Ultrasound Molecular Imaging Joint Laboratory of Heilongjiang Province, Harbin, Heilongjiang Province, China
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang Province, China
| | - You Li
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
- Ultrasound Molecular Imaging Joint Laboratory of Heilongjiang Province, Harbin, Heilongjiang Province, China
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang Province, China
| | - Jiawei Tian
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China.
| | - Ping Sun
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China.
- Ultrasound Molecular Imaging Joint Laboratory of Heilongjiang Province, Harbin, Heilongjiang Province, China.
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang Province, China.
| |
Collapse
|
14
|
Zhang B, Zeng J, Zhang J, Song K, Kuang L, Wu X, Zhao G, Shang H, Ni Z, Chen L. Research trends and perspective of low-intensity pulsed ultrasound in orthopedic rehabilitation treatment based on Web of Science: A bibliometric analysis. J Back Musculoskelet Rehabil 2024; 37:1189-1203. [PMID: 38758991 DOI: 10.3233/bmr-230273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/19/2024]
Abstract
BACKGROUND Ultrasound has a long history as a diagnostic and therapeutic tool. Low-intensity pulsed ultrasound (LIPUS), whose intensity is below 300 mW/cm2, has been widely used in orthopedic rehabilitation treatment. However, the detailed bioeffects and underlying mechanisms of LIPUS treatment need to be explored. OBJECTIVE To make a comprehensive view of the field, bibliometric and visualization analysis was used to reveal the global research trends of LIPUS in orthopedics and rehabilitation treatment between 1994 and 2023. METHODS All literature data on LIPUS were retrieved from the Web of Science Core Collection database. VOSviewer and CiteSpace were applied for the bibliometric and visualization analysis. RESULTS A total of 760 publications were included. The distribution of publications generally showed an unstable rising trend. China had the highest number of publications (28.0%), and Chong Qing Medical University was the organization with the highest number of publications (5.8%). Ultrasound in Medicine and Biology had the highest number of publications (8.8%), while BMJ-British Medical Journal had the highest impact factor among the retrieved journals. Ling Qin from the Chinese University of Hong Kong was the most active researcher. Our overlay visualization map showed that the keywords such as pain, knee osteoarthritis, apoptosis, chondrocytes, cartilage, and autophagy, which link to osteoarthritis, have becoming the new research trends and hotspots. CONCLUSION LIPUS is a popular and increasingly important area of orthopedic rehabilitation, and collaboration of authors from different countries should be further strengthened. Predictably, clinical application of LIPUS on chronic inflammation-related diseases and regenerative medicine, and in-depth biological mechanisms are the orientations of LIPUS in orthopedic rehabilitation treatment.
Collapse
Affiliation(s)
- Bin Zhang
- Department of Wound Repair and Rehabilitation Medicine, Center of Bone Metabolism and Repair, Laboratory for Prevention and Rehabilitation of Training Injuries, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
- Rehabilitation Department, Key Specialty of Neck and Low Back Pain Rehabilitation, Xingcheng Special Duty Sanatorium, Xingcheng, Liaoning, China
- Department of Wound Repair and Rehabilitation Medicine, Center of Bone Metabolism and Repair, Laboratory for Prevention and Rehabilitation of Training Injuries, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Jiahao Zeng
- Rehabilitation Department, Key Specialty of Neck and Low Back Pain Rehabilitation, Xingcheng Special Duty Sanatorium, Xingcheng, Liaoning, China
- Department of Wound Repair and Rehabilitation Medicine, Center of Bone Metabolism and Repair, Laboratory for Prevention and Rehabilitation of Training Injuries, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Jiayi Zhang
- Rehabilitation Department, Key Specialty of Neck and Low Back Pain Rehabilitation, Xingcheng Special Duty Sanatorium, Xingcheng, Liaoning, China
- Department of Wound Repair and Rehabilitation Medicine, Center of Bone Metabolism and Repair, Laboratory for Prevention and Rehabilitation of Training Injuries, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Keyan Song
- Rehabilitation Department, Key Specialty of Neck and Low Back Pain Rehabilitation, Xingcheng Special Duty Sanatorium, Xingcheng, Liaoning, China
| | - Liang Kuang
- Department of Wound Repair and Rehabilitation Medicine, Center of Bone Metabolism and Repair, Laboratory for Prevention and Rehabilitation of Training Injuries, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Xiangbo Wu
- Department of Rehabilitation Medicine, Xi-Jing Hospital, Air Force Medical University (Fourth Military Medical University), Xi'an, Shaanxi, China
| | - Guang Zhao
- Rehabilitation Department, Key Specialty of Neck and Low Back Pain Rehabilitation, Xingcheng Special Duty Sanatorium, Xingcheng, Liaoning, China
| | - Huijuan Shang
- Rehabilitation Department, Key Specialty of Neck and Low Back Pain Rehabilitation, Xingcheng Special Duty Sanatorium, Xingcheng, Liaoning, China
| | - Zhenhong Ni
- Department of Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, Army Medical University, Chongqing, China
| | - Lin Chen
- Department of Wound Repair and Rehabilitation Medicine, Center of Bone Metabolism and Repair, Laboratory for Prevention and Rehabilitation of Training Injuries, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
15
|
Zheng Q, Liu M, He M, Sun S, Liu C, Li Y, Jiang L, Ta D. Low-Intensity Pulsed Ultrasound Promotes the Repair of Achilles Tendinopathy by Downregulating the JAK/STAT Signaling Pathway in Rabbits. IEEE TRANSACTIONS ON ULTRASONICS, FERROELECTRICS, AND FREQUENCY CONTROL 2024; 71:141-152. [PMID: 38060355 DOI: 10.1109/tuffc.2023.3340721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Tendinopathy is a complex tendon injury or pathology outcome, potentially leading to permanent impairment. Low-intensity pulsed ultrasound (LIPUS) is emerging as a treatment modality for tendon disorders. However, the optimal treatment duration and its effect on tendons remain unclear. This study aims to investigate the efficacy of LIPUS in treating injured tendons, delineate the appropriate treatment duration, and elucidate the underlying treatment mechanisms through animal experiments. Ninety-six three-month-old New Zealand white rabbits were divided into normal control (NC) and model groups. The model group received Prostaglandin E2 (PGE2) injections to induce Achilles tendinopathy. They were then divided into model control (MC) and LIPUS treatment (LT) groups. LT received LIPUS intervention with a 1-MHz frequency, a pulse repetition frequency (PRF) of 1 kHz, and spatial average temporal average sound intensity ( [Formula: see text]) of 100 mW/cm2. MC underwent a sham ultrasound, and NC received no treatment. Assessments on 1, 4, 7, 14, and 28 days after LT included shear wave elastography (SWE), mechanical testing, histologic evaluation, ribonucleic acid sequencing (RNA-seq), polymerase chain reaction (PCR), and western blot (WB) analysis. SWE results showed that the shear modulus in the LT group was significantly higher than that in the MC group after LT for seven days. Histological results demonstrated improved tendon tissue alignment and fibroblast distribution after LT. Molecular analyses suggested that LIPUS may downregulate the Janus kinase/signal transducer and activator of transcription (JAK/STAT) signaling pathway and regulate inflammatory and matrix-related factors. We concluded that LT enhanced injured tendon elasticity and accelerated Achilles tendon healing. The study highlighted the JAK/STAT signaling pathway as a potential therapeutic target for LT of Achilles tendinopathy, guiding future research.
Collapse
|
16
|
Casal D, Casimiro MH, Ferreira LM, Leal JP, Rodrigues G, Lopes R, Moura DL, Gonçalves L, Lago JB, Pais D, Santos PMP. Review of Piezoelectrical Materials Potentially Useful for Peripheral Nerve Repair. Biomedicines 2023; 11:3195. [PMID: 38137416 PMCID: PMC10740581 DOI: 10.3390/biomedicines11123195] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 11/21/2023] [Accepted: 11/29/2023] [Indexed: 12/24/2023] Open
Abstract
It has increasingly been recognized that electrical currents play a pivotal role in cell migration and tissue repair, in a process named "galvanotaxis". In this review, we summarize the current evidence supporting the potential benefits of electric stimulation (ES) in the physiology of peripheral nerve repair (PNR). Moreover, we discuss the potential of piezoelectric materials in this context. The use of these materials has deserved great attention, as the movement of the body or of the external environment can be used to power internally the electrical properties of devices used for providing ES or acting as sensory receptors in artificial skin (e-skin). The fact that organic materials sustain spontaneous degradation inside the body means their piezoelectric effect is limited in duration. In the case of PNR, this is not necessarily problematic, as ES is only required during the regeneration period. Arguably, piezoelectric materials have the potential to revolutionize PNR with new biomedical devices that range from scaffolds and nerve-guiding conduits to sensory or efferent components of e-skin. However, much remains to be learned regarding piezoelectric materials, their use in manufacturing of biomedical devices, and their sterilization process, to fine-tune their safe, effective, and predictable in vivo application.
Collapse
Affiliation(s)
- Diogo Casal
- Departamento de Anatomia, NOVA Medical School|Faculdade de Ciências Médicas (NMS|FCM), Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal; (L.G.); (D.P.)
- Plastic and Reconstructive Surgery Department and Burn Unit, Centro Hospitalar Universitário de Lisboa Central, Rua José António Serrano, 1169-045 Lisbon, Portugal
| | - Maria Helena Casimiro
- Centro de Ciências e Tecnologias Nucleares (C2TN), Instituto Superior Técnico (IST), Universidade de Lisboa, 2695-066 Bobadela, Portugal; (M.H.C.); (P.M.P.S.)
| | - Luís M. Ferreira
- Departamento de Engenharia e Ciências Nucleares (DECN), Instituto Superior Técnico (IST), Universidade de Lisboa, 2695-066 Bobadela, Portugal;
| | - João Paulo Leal
- Centro de Química Estrutural (CQE), Institute of Molecular Sciences (IMS), Instituto Superior Técnico (IST), Universidade de Lisboa, 2695-066 Bobadela, Portugal;
| | - Gabriela Rodrigues
- Centro de Ecologia, Evolução e Alterações Ambientais (cE3c) & CHANGE—Global Change and Sustainability Institute, Departamento de Biologia Animal, Faculdade de Ciências, Universidade de Lisboa (FCUL), 1749-016 Lisboa, Portugal;
| | - Raquel Lopes
- Gynaecology and Obstetrics Department, Maternidade Alfredo da Costa, Centro Hospitalar Universitário de Lisboa Central, R. Viriato 1, 2890-495 Lisboa, Portugal;
| | - Diogo Lino Moura
- Anatomy Institute and Orthopedics Department, Faculty of Medicine, University of Coimbra, 3004-504 Coimbra, Portugal;
- Spine Unit, Orthopedics Department, Coimbra University Hospital, 3000-602 Coimbra, Portugal
| | - Luís Gonçalves
- Departamento de Anatomia, NOVA Medical School|Faculdade de Ciências Médicas (NMS|FCM), Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal; (L.G.); (D.P.)
| | - João B. Lago
- Departamento de Biologia Animal, Faculdade de Ciências, Universidade de Lisboa (FCUL), 1749-016 Lisboa, Portugal;
| | - Diogo Pais
- Departamento de Anatomia, NOVA Medical School|Faculdade de Ciências Médicas (NMS|FCM), Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal; (L.G.); (D.P.)
| | - Pedro M. P. Santos
- Centro de Ciências e Tecnologias Nucleares (C2TN), Instituto Superior Técnico (IST), Universidade de Lisboa, 2695-066 Bobadela, Portugal; (M.H.C.); (P.M.P.S.)
| |
Collapse
|
17
|
Chen R, Du W, Zhang X, Xu R, Jiang W, Zhang C, Yang Y, Zhang H, Xie X, Song D, Yuan Y, Zhang X. Protective effects of low-intensity pulsed ultrasound (LIPUS) against cerebral ischemic stroke in mice by promoting brain vascular remodeling via the inhibition of ROCK1/p-MLC2 signaling pathway. Cereb Cortex 2023; 33:10984-10996. [PMID: 37771006 DOI: 10.1093/cercor/bhad330] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 08/21/2023] [Accepted: 08/22/2023] [Indexed: 09/30/2023] Open
Abstract
Vascular remodeling is essential for patients with cerebral ischemic stroke (CIS). Our previous study proved that low-intensity pulsed ultrasound (LIPUS) could increase cortical hemodynamics. However, the effects and mechanisms of LIPUS on cerebral vascular remodeling after CIS are still unknown. In this study, we applied LIPUS to the mouse brain at 0.5 h after distal middle cerebral artery occlusion (dMCAO) and subsequently daily for a stimulation time of 30 min. Results showed that compared with the dMCAO group, LIPUS markedly increased cerebral blood flow (CBF), reduced brain swelling, and improved functional recovery at day 3 after CIS. LIPUS promoted leptomeningeal vasculature remodeling, enlarged vascular diameter, and increased the average vessel length and density at day 3 after CIS. Proteomic analysis highlighted that LIPUS mainly participated in the regulation of actin cytoskeleton pathway. Rho kinase 1 (ROCK1) was downregulated by LIPUS and participated in regulation of actin cytoskeleton. Subsequently, we verified that ROCK1 was mainly expressed in pericytes. Furthermore, we demonstrated that LIPUS inhibited ROCK1/p-MLC2 signaling pathway after CIS, which had positive effects on vascular remodeling and cerebral blood circulation. In conclusion, our preliminary study revealed the vascular remodeling effects and mechanism of LIPUS in CIS, provided evidence for potential clinical application of LIPUS.
Collapse
Affiliation(s)
- Rong Chen
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
- Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-Cerebrovascular Disease, Hebei Medical University, Shijiazhuang, Hebei 050011, China
- The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang, Hebei 050000, China
| | - Wei Du
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
- Department of Orthopedics, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei 050051, China
| | - Xiao Zhang
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
- Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-Cerebrovascular Disease, Hebei Medical University, Shijiazhuang, Hebei 050011, China
- The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang, Hebei 050000, China
| | - Renhao Xu
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
- Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-Cerebrovascular Disease, Hebei Medical University, Shijiazhuang, Hebei 050011, China
- The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang, Hebei 050000, China
| | - Wei Jiang
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
- Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-Cerebrovascular Disease, Hebei Medical University, Shijiazhuang, Hebei 050011, China
- The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang, Hebei 050000, China
| | - Cong Zhang
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
- Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-Cerebrovascular Disease, Hebei Medical University, Shijiazhuang, Hebei 050011, China
- The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang, Hebei 050000, China
| | - Yi Yang
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
- Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-Cerebrovascular Disease, Hebei Medical University, Shijiazhuang, Hebei 050011, China
- The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang, Hebei 050000, China
| | - Huiran Zhang
- Department of Biological Pharmacy, Hebei Medical University, Shijiazhuang, Hebei 050011, China
| | - Xiaoli Xie
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
| | - Degang Song
- Department of Neurology, First Hospital of Qinhuangdao, Hebei Medical University, No. 258, Wenhua Road, Qinhuangdao, Hebei 066000, China
| | - Yi Yuan
- School of Electrical Engineering, Key Laboratory of Intelligent Rehabilitation and Neuromodulation of Hebei Province, Yanshan University, Qinhuangdao, Hebei 066004, China
| | - Xiangjian Zhang
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
- Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-Cerebrovascular Disease, Hebei Medical University, Shijiazhuang, Hebei 050011, China
- The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang, Hebei 050000, China
| |
Collapse
|
18
|
Zhu H, He M, Wang Y, Zhang Y, Dong J, Chen B, Li Y, Zhou L, Du L, Liu Y, Zhang W, Ta D, Duan S. Low-intensity pulsed ultrasound alleviates doxorubicin-induced cardiotoxicity via inhibition of S100a8/a9-mediated cardiac recruitment of neutrophils. Bioeng Transl Med 2023; 8:e10570. [PMID: 38023700 PMCID: PMC10658545 DOI: 10.1002/btm2.10570] [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: 04/06/2023] [Revised: 06/05/2023] [Accepted: 06/25/2023] [Indexed: 12/01/2023] Open
Abstract
Doxorubicin (DOX)-induced cardiotoxicity limits its broad use as a chemotherapy agent. The development of effective and non-invasive strategies to prevent DOX-associated adverse cardiac events is urgently needed. We aimed to examine whether and how low-intensity pulsed ultrasound (LIPUS) plays a protective role in DOX-induced cardiotoxicity. Male C57BL/6J mice were used to establish models of both acute and chronic DOX-induced cardiomyopathy. Non-invasive LIPUS therapy was conducted for four consecutive days after DOX administration. Cardiac contractile function was evaluated by echocardiography. Myocardial apoptosis, oxidative stress, and fibrosis were analyzed using terminal deoxynucleotidyl transferase-mediated dUTP nick end labelling (TUNEL) staining, dihydroethidium (DHE) staining, and picrosirius red staining assays. RNA-seq analysis was performed to unbiasedly explore the possible downstream regulatory mechanisms. Neutrophil recruitment and infiltration in the heart were analyzed by flow cytometry. The S100a8/a9 inhibitor ABR-238901 was utilized to identify the effect of S100a8/a9 signaling. We found that LIPUS therapy elicited a great benefit on DOX-induced heart contractile dysfunction in both acute and chronic DOX models. Chronic DOX administration increased serum creatine kinase and lactate dehydrogenase levels, as well as myocardial apoptosis, all of which were significantly mitigated by LIPUS. In addition, LIPUS treatment prevented chronic DOX-induced cardiac oxidative stress and fibrosis. RNA-seq analysis revealed that LIPUS treatment partially reversed alterations of gene expression induced by DOX. Gene ontology (GO) analysis of the downregulated genes between DOX-LIPUS and DOX-Sham groups indicated that inhibition of neutrophil chemotaxis might be involved in the protective effects of LIPUS therapy. Flow cytometry analysis illustrated the inhibitory effects of LIPUS on DOX-induced neutrophil recruitment and infiltration in the heart. Moreover, S100 calcium binding protein A8/A9 (S100a8/a9) was identified as a potential key target of LIPUS therapy. S100a8/a9 inhibition by ABR-238901 showed a similar heart protective effect against DOX-induced cardiomyopathy to LIPUS treatment. LIPUS therapy prevents DOX-induced cardiotoxicity through inhibition of S100a8/a9-mediated neutrophil recruitment to the heart, suggesting its potential application in cancer patients undergoing chemotherapy with DOX.
Collapse
Affiliation(s)
- Hong Zhu
- Laboratory of Oral Microbiota and Systemic DiseasesShanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of MedicineShanghaiChina
- Translational Medical Center for Stem Cell Therapy & Institutes for Regenerative Medicine, Shanghai East Hospital, Tongji University School of MedicineShanghaiChina
| | - Min He
- Center for Biomedical Engineering, School of Information Science and Technology, Fudan UniversityShanghaiChina
| | - Yong‐Li Wang
- Laboratory of Oral Microbiota and Systemic DiseasesShanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of MedicineShanghaiChina
- National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of StomatologyShanghaiChina
| | - Yuanxin Zhang
- Department of CardiologyNinth People's Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Jingsong Dong
- Center for Biomedical Engineering, School of Information Science and Technology, Fudan UniversityShanghaiChina
| | - Bo‐Yan Chen
- Laboratory of Oral Microbiota and Systemic DiseasesShanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of MedicineShanghaiChina
- National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of StomatologyShanghaiChina
| | - Yu‐Lin Li
- Laboratory of Oral Microbiota and Systemic DiseasesShanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of MedicineShanghaiChina
- National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of StomatologyShanghaiChina
| | - Lu‐Jun Zhou
- Laboratory of Oral Microbiota and Systemic DiseasesShanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of MedicineShanghaiChina
- National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of StomatologyShanghaiChina
| | - Lin‐Juan Du
- Laboratory of Oral Microbiota and Systemic DiseasesShanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of MedicineShanghaiChina
- National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of StomatologyShanghaiChina
| | - Yuan Liu
- Laboratory of Oral Microbiota and Systemic DiseasesShanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of MedicineShanghaiChina
- National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of StomatologyShanghaiChina
| | - Wu‐Chang Zhang
- Laboratory of Oral Microbiota and Systemic DiseasesShanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of MedicineShanghaiChina
- National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of StomatologyShanghaiChina
| | - Dean Ta
- Center for Biomedical Engineering, School of Information Science and Technology, Fudan UniversityShanghaiChina
- Department of Rehabilitation MedicineHuashan Hospital, Fudan UniversityShanghaiChina
| | - Sheng‐Zhong Duan
- Laboratory of Oral Microbiota and Systemic DiseasesShanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of MedicineShanghaiChina
- National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of StomatologyShanghaiChina
| |
Collapse
|
19
|
Jia W, Zhou Z, Zhan W. Musculoskeletal Biomaterials: Stimulated and Synergized with Low Intensity Pulsed Ultrasound. J Funct Biomater 2023; 14:504. [PMID: 37888169 PMCID: PMC10607075 DOI: 10.3390/jfb14100504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 09/10/2023] [Accepted: 09/21/2023] [Indexed: 10/28/2023] Open
Abstract
Clinical biophysical stimulating strategies, which have significant effects on improving the function of organs or treating diseases by causing the salutary response of body, have shown many advantages, such as non-invasiveness, few side effects, and controllable treatment process. As a critical technique for stimulation, the low intensity pulsed ultrasound (LIPUS) has been explored in regulating osteogenesis, which has presented great promise in bone repair by delivering a combined effect with biomaterials. This review summarizes the musculoskeletal biomaterials that can be synergized with LIPUS for enhanced biomedical application, including bone regeneration, spinal fusion, osteonecrosis/osteolysis, cartilage repair, and nerve regeneration. Different types of biomaterials are categorized for summary and evaluation. In each subtype, the verified biological mechanisms are listed in a table or graphs to prove how LIPUS was effective in improving musculoskeletal tissue regeneration. Meanwhile, the acoustic excitation parameters of LIPUS that were promising to be effective for further musculoskeletal tissue engineering are discussed, as well as their limitations and some perspectives for future research. Overall, coupled with biomimetic scaffolds and platforms, LIPUS may be a powerful therapeutic approach to accelerate musculoskeletal tissue repair and even in other regenerative medicine applications.
Collapse
Affiliation(s)
- Wanru Jia
- Department of Ultrasound, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China;
| | - Zifei Zhou
- Department of Orthopedics, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Weiwei Zhan
- Department of Ultrasound, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China;
| |
Collapse
|
20
|
Hao S, Wang M, Yin Z, Jing Y, Bai L, Su J. Microenvironment-targeted strategy steers advanced bone regeneration. Mater Today Bio 2023; 22:100741. [PMID: 37576867 PMCID: PMC10413201 DOI: 10.1016/j.mtbio.2023.100741] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 06/26/2023] [Accepted: 07/19/2023] [Indexed: 08/15/2023] Open
Abstract
Treatment of large bone defects represents a great challenge in orthopedic and craniomaxillofacial surgery. Traditional strategies in bone tissue engineering have focused primarily on mimicking the extracellular matrix (ECM) of bone in terms of structure and composition. However, the synergistic effects of other cues from the microenvironment during bone regeneration are often neglected. The bone microenvironment is a sophisticated system that includes physiological (e.g., neighboring cells such as macrophages), chemical (e.g., oxygen, pH), and physical factors (e.g., mechanics, acoustics) that dynamically interact with each other. Microenvironment-targeted strategies are increasingly recognized as crucial for successful bone regeneration and offer promising solutions for advancing bone tissue engineering. This review provides a comprehensive overview of current microenvironment-targeted strategies and challenges for bone regeneration and further outlines prospective directions of the approaches in construction of bone organoids.
Collapse
Affiliation(s)
- Shuyue Hao
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
| | - Mingkai Wang
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
| | - Zhifeng Yin
- Department of Orthopedics, Shanghai Zhongye Hospital, Shanghai, 201941, China
| | - Yingying Jing
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
| | - Long Bai
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
| | - Jiacan Su
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Department of Orthopedic Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200444, China
| |
Collapse
|
21
|
Ali M, Bathaei MJ, Istif E, Karimi SNH, Beker L. Biodegradable Piezoelectric Polymers: Recent Advancements in Materials and Applications. Adv Healthc Mater 2023; 12:e2300318. [PMID: 37235849 PMCID: PMC11469082 DOI: 10.1002/adhm.202300318] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 05/21/2023] [Indexed: 05/28/2023]
Abstract
Recent materials, microfabrication, and biotechnology improvements have introduced numerous exciting bioelectronic devices based on piezoelectric materials. There is an intriguing evolution from conventional unrecyclable materials to biodegradable, green, and biocompatible functional materials. As a fundamental electromechanical coupling material in numerous applications, novel piezoelectric materials with a feature of degradability and desired electrical and mechanical properties are being developed for future wearable and implantable bioelectronics. These bioelectronics can be easily integrated with biological systems for applications, including sensing physiological signals, diagnosing medical problems, opening the blood-brain barrier, and stimulating healing or tissue growth. Therefore, the generation of piezoelectricity from natural and synthetic bioresorbable polymers has drawn great attention in the research field. Herein, the significant and recent advancements in biodegradable piezoelectric materials, including natural and synthetic polymers, their principles, advanced applications, and challenges for medical uses, are reviewed thoroughly. The degradation methods of these piezoelectric materials through in vitro and in vivo studies are also investigated. These improvements in biodegradable piezoelectric materials and microsystems could enable new applications in the biomedical field. In the end, potential research opportunities regarding the practical applications are pointed out that might be significant for new materials research.
Collapse
Affiliation(s)
- Mohsin Ali
- Department of Biomedical Sciences and EngineeringKoç UniversityRumelifeneri YoluSarıyerIstanbul34450Turkey
| | - Mohammad Javad Bathaei
- Department of Biomedical Sciences and EngineeringKoç UniversityRumelifeneri YoluSarıyerIstanbul34450Turkey
| | - Emin Istif
- Department of Mechanical EngineeringKoç UniversityRumelifeneri YoluSarıyerIstanbul34450Turkey
- Faculty of Engineering and Natural SciencesKadir Has UniversityCibaliIstanbul34083Turkey
| | - Seyed Nasir Hosseini Karimi
- Koç University Research Center for Translational Research (KUTTAM)Rumelifeneri YoluSarıyerIstanbul34450Turkey
| | - Levent Beker
- Department of Biomedical Sciences and EngineeringKoç UniversityRumelifeneri YoluSarıyerIstanbul34450Turkey
- Department of Mechanical EngineeringKoç UniversityRumelifeneri YoluSarıyerIstanbul34450Turkey
- Koç University Research Center for Translational Research (KUTTAM)Rumelifeneri YoluSarıyerIstanbul34450Turkey
| |
Collapse
|
22
|
Pereira LF, Fontes-Pereira AJ, de Albuquerque Pereira WC. Influence of Low-Intensity Pulsed Ultrasound Parameters on the Bone Mineral Density in Rat Model: A Systematic Review. ULTRASOUND IN MEDICINE & BIOLOGY 2023; 49:1687-1698. [PMID: 37121881 DOI: 10.1016/j.ultrasmedbio.2023.03.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 02/23/2023] [Accepted: 03/03/2023] [Indexed: 06/19/2023]
Abstract
OBJECTIVE Bone recovery typically depends on the age of organisms or the prevalence of metabolic disorders such as osteoporosis, which is a metabolic condition characterized by decreased bone strength and bone mineral density (BMD). Therefore, low-intensity pulsed ultrasound (LIPUS), a non-invasive method for osteogenic stimulation, presents promising results. However, heterogeneity in animal study designs is a typical characteristic. Hence, we conducted a systematic review to evaluate the effectiveness of LIPUS in the recovery of experimental bone defects using rat models. We examined the areal and volumetric BMD to identify LIPUS doses to be applied and evaluated the accuracy reported by previous studies. METHODS The Virtual Health Library regional portal, PubMed, Embase, EBSCOhost, Scopus and CAPES were reviewed for animal studies that compared fracture treatments based on LIPUS with sham or no treatments using rat models and reported BMD as an outcome. The tool provided by the Systematic Review Centre for Laboratory Animal Experimentation (SYRCLE) and the Collaborative Approach to Meta-Analysis and Review of Animal Data from Experimental Studies (CAMARADES) checklist were used to assess the bias and quality of such studies. RESULTS Of the six studies reviewed, the most frequently used LIPUS dose had an ultrasonic frequency of 1.0 MHz, repetition rate of 0.1 kHz and pulse duration of 2000 μs. An intensity (ISATA) of 30 mW/cm2 was the most preferred for bone recovery. However, the BMD could not solely irrefutably evaluate the effectiveness of LIPUS in bone recovery as the results were discordant with each other. The discrepancies in experimental methodologies, low-quality classifications and high risk of bias in the selected studies, however, did not validate the undertaking of a meta-analysis. CONCLUSION On the basis of the BMD results, no sufficient evidence was found to recommend the use of LIPUS for bone recovery in rat models. Thus, this systematic review indicates that the accuracy of such reports must be improved to improve their scientific quality to facilitate a transition of LIPUS applications from pre-clinical research to clinic use.
Collapse
Affiliation(s)
- Luiz Fernando Pereira
- Biomedical Engineering Program/COPPE, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Aldo José Fontes-Pereira
- Biomedical Engineering Program/COPPE, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil; Centro Universitário Serra dos Órgãos-Unifeso, Teresópolis, Rio de Janeiro, Brazil
| | | |
Collapse
|
23
|
Chen Y, Sun S, Zhou X, He M, Li Y, Liu C, Ta D. Low-intensity pulsed ultrasound and parathyroid hormone improve muscle atrophy in estrogen deficiency mice. ULTRASONICS 2023; 132:106984. [PMID: 36944299 DOI: 10.1016/j.ultras.2023.106984] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 02/12/2023] [Accepted: 03/09/2023] [Indexed: 05/29/2023]
Abstract
Due to aging and long-term estrogen deficiency, postmenopausal women suffer muscle atrophy (MA), which is characterized by decreased muscle mass and muscle quality. Low-intensity pulsed ultrasound (LIPUS) is an acoustic wave inducing biological effects mainly by the mechanical stimulation and used as a non-invasive physical therapy for muscle repair. Parathyroid hormone (PTH) is an 84-amino-acid polypeptide, and its bioactive fragment [PTH (1-34)] has potential application in the treatment of MA. We speculate that the combination of physical therapy (i.e., the LIPUS) and regulatory hormone (i.e., the PTH) would be more effective in the treatment of MA. The objective of this study was to evaluate the individual and combined effects of LIPUS and PTH therapy on MA in estrogen deficiency mice. Seventy 8-week-old female C57BL/6J mice were used in this study and the MA model was induced by an intraperitoneal injection of 4-vinylcyclohexene diepoxide (VCD) for 20 consecutive days. The VCD-induced MA mice were randomly divided into MA, LIPUS, PTH and LIPUS + PTH (Combined) groups (n = 10/group). In the LIPUS group, the mice were treated by LIPUS in bilateral quadriceps muscles for 20 min, five times a week for 6 weeks. In the PTH group, the mice received subcutaneous injection of PTH (1-34) (80 ug/kg/d) five times a week, for 6 weeks. In the Combined group, the PTH was administrated 30 min before each LIPUS session. Hematoxylin-eosin (H&E) staining, serum biochemical analysis and quantitative real-time polymerase chain reaction (qRT-PCR) were applied to evaluate the therapeutic effects of related treatments. The results showed that the MA mice had a disordered estrus cycle, significantly decreased muscle mass and myofibers cross-sectional area (CSA). After treatments, LIPUS, PTH and Combined groups had a significantly increased CSA, compared with the MA mice without treatment. In addition, Combined group had a significantly increased mRNA expression of Pax7, MyoD and MyoG, compared with LIPUS and PTH monotherapy groups. Our findings indicated that the combination of LIPUS and PTH treatment improves muscle regeneration ability, which might have potential for treating MA in postmenopausal women.
Collapse
Affiliation(s)
- Yuefu Chen
- Academy for Engineering and Technology, Fudan University, Shanghai 200433, China
| | - Shuxin Sun
- Academy for Engineering and Technology, Fudan University, Shanghai 200433, China
| | - Xinyan Zhou
- Center for Biomedical Engineering, School of Information Science and Technology, Fudan University, Shanghai 200438, China
| | - Min He
- Center for Biomedical Engineering, School of Information Science and Technology, Fudan University, Shanghai 200438, China
| | - Ying Li
- Center for Biomedical Engineering, School of Information Science and Technology, Fudan University, Shanghai 200438, China.
| | - Chengcheng Liu
- Academy for Engineering and Technology, Fudan University, Shanghai 200433, China; State Key Laboratory of Integrated Chips and Systems, Fudan University, Shanghai 201203, China.
| | - Dean Ta
- Academy for Engineering and Technology, Fudan University, Shanghai 200433, China; Center for Biomedical Engineering, School of Information Science and Technology, Fudan University, Shanghai 200438, China; State Key Laboratory of Integrated Chips and Systems, Fudan University, Shanghai 201203, China
| |
Collapse
|
24
|
Shin EJ, Park S, Kang S, Kim J, Chang JH. Improving the quality of ultrasound images acquired using a therapeutic transducer. ULTRASONICS 2023; 134:107063. [PMID: 37300907 DOI: 10.1016/j.ultras.2023.107063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 05/01/2023] [Accepted: 05/30/2023] [Indexed: 06/12/2023]
Abstract
To enhance the effectiveness and safety of focused ultrasound (FUS) therapy, ultrasound image-based guidance and treatment monitoring are crucial. However, the use of FUS transducers for both therapy and imaging is impractical due to their low spatial resolution, signal-to-noise ratio (SNR), and contrast-to-noise ratio (CNR). To address this issue, we propose a new method that significantly improve the quality of images obtained by a FUS transducer. The proposed method employs coded excitation to enhance SNR and Wiener deconvolution to solve the problem of low axial resolution resulting from the narrow spectral bandwidth of FUS transducers. Specifically, the method eliminates the impulse response of a FUS transducer from received ultrasound signals using Wiener deconvolution, and pulse compression is performed using a mismatched filter. Simulation and commercial phantom experiments confirmed that the proposed method significantly improves the quality of images acquired by the FUS transducer. The -6 dB axial resolution was improved 1.27 mm to 0.37 mm that was similar to the resolution achieved by the imaging transducer, i.e., 0.33 mm. SNR and CNR also increased from 16.5 dB and 0.69 to 29.1 dB and 3.03, respectively, that were also similar to those by the imaging transducer (27.8 dB and 3.16). Based on the results, we believe that the proposed method has great potential to enhance the clinical utility of FUS transducers in ultrasound image-guided therapy.
Collapse
Affiliation(s)
- Eui-Ji Shin
- Department of Electronic Engineering, Sogang University, Seoul, Korea
| | - Sunghun Park
- Department of Electronic Engineering, Sogang University, Seoul, Korea
| | - Sungwoo Kang
- Department of Electrical Engineering and Computer Science, DGIST (Daegu Gyeongbuk Institute of Science and Technology), Daegu, Korea
| | - Jinwoo Kim
- Department of Electrical Engineering and Computer Science, DGIST (Daegu Gyeongbuk Institute of Science and Technology), Daegu, Korea
| | - Jin Ho Chang
- Department of Electrical Engineering and Computer Science, DGIST (Daegu Gyeongbuk Institute of Science and Technology), Daegu, Korea.
| |
Collapse
|
25
|
Phan TN, Fan CH, Yeh CK. Application of Ultrasound to Enhancing Stem Cells Associated Therapies. Stem Cell Rev Rep 2023:10.1007/s12015-023-10546-w. [PMID: 37119453 DOI: 10.1007/s12015-023-10546-w] [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] [Accepted: 04/12/2023] [Indexed: 05/01/2023]
Abstract
Pluripotent stem cell therapy exhibits self-renewal capacity and multi-directional differentiation potential and is considered an important regenerative approach for the treatment of several diseases. However, insufficient cell transplantation efficiency, uncontrollable differentiation, low cell viability, and difficult tracing limit its clinical applications and treatment outcome. Ultrasound (US) has mechanical, cavitation, and thermal effects that can produce different biological effects on organs, tissues, and cells. US can be combined with different US-responsive particles for enhanced physical-chemical stimulation and drug delivery. In the meantime, US also can provide a noninvasive and harmless imaging modality for deep tissue in vivo. An in-depth evaluation of the role and mechanism of action of US in stem cell therapy would enhance understanding of US and encourage research in this field. In this article, we comprehensively review progress in the application of US alone and combined with US-responsive particles for the promotion of proliferation, differentiation, migration, and in vivo detection of stem cells and the potential clinical applications.
Collapse
Affiliation(s)
- Thi-Nhan Phan
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Ching-Hsiang Fan
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan
- Medical Device Innovation Center, National Cheng Kung University, Tainan, Taiwan
| | - Chih-Kuang Yeh
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan.
| |
Collapse
|
26
|
Wang J, Yuan B, Yin R, Zhang H. Inflammation Responses to Bone Scaffolds under Mechanical Stimuli in Bone Regeneration. J Funct Biomater 2023; 14:jfb14030169. [PMID: 36976093 PMCID: PMC10059255 DOI: 10.3390/jfb14030169] [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: 02/05/2023] [Revised: 03/05/2023] [Accepted: 03/18/2023] [Indexed: 03/29/2023] Open
Abstract
Physical stimuli play an important role in one tissue engineering. Mechanical stimuli, such as ultrasound with cyclic loading, are widely used to promote bone osteogenesis; however, the inflammatory response under physical stimuli has not been well studied. In this paper, the signaling pathways related to inflammatory responses in bone tissue engineering are evaluated, and the application of physical stimulation to promote osteogenesis and its related mechanisms are reviewed in detail; in particular, how physical stimulation alleviates inflammatory responses during transplantation when employing a bone scaffolding strategy is discussed. It is concluded that physical stimulation (e.g., ultrasound and cyclic stress) helps to promote osteogenesis while reducing the inflammatory response. In addition, apart from 2D cell culture, more consideration should be given to the mechanical stimuli applied to 3D scaffolds and the effects of different force moduli while evaluating inflammatory responses. This will facilitate the application of physiotherapy in bone tissue engineering.
Collapse
Affiliation(s)
- Junjie Wang
- School of Mechanical and Power Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Bo Yuan
- Spine Center, Department of Orthopaedics, Shanghai Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Ruixue Yin
- School of Mechanical and Power Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Hongbo Zhang
- School of Mechanical and Power Engineering, East China University of Science and Technology, Shanghai 200237, China
| |
Collapse
|
27
|
Puts R, Khaffaf A, Shaka M, Zhang H, Raum K. Focused Low-Intensity Pulsed Ultrasound (FLIPUS) Mitigates Apoptosis of MLO-Y4 Osteocyte-like Cells. Bioengineering (Basel) 2023; 10:bioengineering10030387. [PMID: 36978778 PMCID: PMC10045139 DOI: 10.3390/bioengineering10030387] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/01/2023] [Accepted: 03/15/2023] [Indexed: 03/30/2023] Open
Abstract
Long cytoplasmic processes of osteocytes orchestrate bone activity by integration of biochemical and mechanical signals and regulate load-induced bone adaptation. Low-Intensity Pulsed Ultrasound (LIPUS) is a clinically used technique for fracture healing that delivers mechanical impulses to the damaged bone tissue in a non-invasive and non-ionizing manner. The mechanism of action of LIPUS is still controversially discussed in the scientific community. In this study, the effect of focused LIPUS (FLIPUS) on the survival of starved MLO-Y4 osteocytes was investigated in vitro. Osteocytes stimulated for 10 min with FLIPUS exhibited extended dendrites, which formed frequent connections to neighboring cells and spanned longer distances. The sonicated cells displayed thick actin bundles and experienced increase in expression of connexin 43 (Cx43) proteins, especially on their dendrites, and E11 glycoprotein, which is responsible for the elongation of cellular cytoplasmic processes. After stimulation, expression of cell growth and survival genes as well as genes related to cell-cell communication was augmented. In addition, cell viability was improved after the sonication, and a decrease in ATP release in the medium was observed. In summary, FLIPUS mitigated apoptosis of starved osteocytes, which is likely related to the formation of the extensive dendritic network that ensured cell survival.
Collapse
Affiliation(s)
- Regina Puts
- Center for Biomedicine, Charité-Universitätsmedizin, 12203 Berlin, Germany
- Berlin Institute of Health (BIH) Center for Regenerative Therapies, Charité-Universitätsmedizin, 13353 Berlin, Germany
| | - Aseel Khaffaf
- Center for Biomedicine, Charité-Universitätsmedizin, 12203 Berlin, Germany
| | - Maria Shaka
- Center for Biomedicine, Charité-Universitätsmedizin, 12203 Berlin, Germany
| | - Hui Zhang
- Center for Biomedicine, Charité-Universitätsmedizin, 12203 Berlin, Germany
| | - Kay Raum
- Center for Biomedicine, Charité-Universitätsmedizin, 12203 Berlin, Germany
| |
Collapse
|
28
|
Effects and mechanotransduction pathways of therapeutic ultrasound on healthy and osteoarthritic chondrocytes: a systematic review of in vitro studies. Osteoarthritis Cartilage 2023; 31:317-339. [PMID: 36481451 DOI: 10.1016/j.joca.2022.07.014] [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: 02/24/2022] [Revised: 07/11/2022] [Accepted: 07/12/2022] [Indexed: 12/12/2022]
Abstract
OBJECTIVE To investigate the effects and mechanotransduction pathways of therapeutic ultrasound on chondrocytes. METHOD PubMed, EMBASE and Web of Science databases were searched up to 19th September 2021 to identify in vitro studies exploring ultrasound to stimulate chondrocytes for osteoarthritis (OA) treatment. Study characteristics, ultrasound parameters, in vitro setup, and mechanotransduction pathways were collected. Risk of bias was judged using the Risk of Bias Assessment for Non-randomized Studies (RoBANS) tool. RESULTS Thirty-one studies were included comprising healthy and OA chondrocytes and explants. Most studies had high risk of performance, detection and pseudoreplication bias due to lack of temperature control, setup calibration, inadequate semi-quantitatively analyzes and independent experiments. Ultrasound was applied to the culture plate via acoustic gel, water bath or culture media. Regardless of the setup used, ultrasound stimulated the cartilage production and suppressed its degradation, although the effect size was nonsignificant. Ultrasound inhibited p38, c-Jun N-terminal kinases (JNK) and factor nuclear kappa B (NFκB) pathways in OA chondrocytes to reduce apoptosis, inflammation and matrix degradation, while triggered phosphoinositide-3-kinase/akt (PI3K/Akt), extracellular signal-regulated kinase (ERK), p38 and JNK pathways in healthy chondrocytes to promote matrix synthesis. CONCLUSION The included studies suggest that ultrasound application induces therapeutic effects on chondrocytes. However, these results should be interpreted with caution because high risk of performance, detection and pseudoreplication bias were identified. Future studies should explore the application of ultrasound on human OA chondrocytes cultures to potentiate the applicability of ultrasound towards cartilage regeneration of knee with OA.
Collapse
|
29
|
Quarato CMI, Lacedonia D, Salvemini M, Tuccari G, Mastrodonato G, Villani R, Fiore LA, Scioscia G, Mirijello A, Saponara A, Sperandeo M. A Review on Biological Effects of Ultrasounds: Key Messages for Clinicians. Diagnostics (Basel) 2023; 13:855. [PMID: 36899998 PMCID: PMC10001275 DOI: 10.3390/diagnostics13050855] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 02/18/2023] [Accepted: 02/21/2023] [Indexed: 02/25/2023] Open
Abstract
Ultrasound (US) is acoustic energy that interacts with human tissues, thus, producing bioeffects that may be hazardous, especially in sensitive organs (i.e., brain, eye, heart, lung, and digestive tract) and embryos/fetuses. Two basic mechanisms of US interaction with biological systems have been identified: thermal and non-thermal. As a result, thermal and mechanical indexes have been developed to provide a means of assessing the potential for biological effects from exposure to diagnostic US. The main aims of this paper were to describe the models and assumptions used to estimate the "safety" of acoustic outputs and indices and to summarize the current state of knowledge about US-induced effects on living systems deriving from in vitro models and in vivo experiments on animals. This review work has made it possible to highlight the limits associated with the use of the estimated safety values of thermal and mechanical indices relating above all to the use of new US technologies, such as contrast-enhanced ultrasound (CEUS) and acoustic radiation force impulse (ARFI) shear wave elastography (SWE). US for diagnostic and research purposes has been officially declared safe, and no harmful biological effects in humans have yet been demonstrated with new imaging modalities; however, physicians should be adequately informed on the potential risks of biological effects. US exposure, according to the ALARA (As Low As Reasonably Achievable) principle, should be as low as reasonably possible.
Collapse
Affiliation(s)
- Carla Maria Irene Quarato
- Department of Medical and Surgical Sciences, Institute of Respiratory Diseases, Policlinico Universitario “Riuniti” di Foggia, University of Foggia, 71122 Foggia, Italy
| | - Donato Lacedonia
- Department of Medical and Surgical Sciences, Institute of Respiratory Diseases, Policlinico Universitario “Riuniti” di Foggia, University of Foggia, 71122 Foggia, Italy
| | - Michela Salvemini
- Department of Medical and Surgical Sciences, Institute of Respiratory Diseases, Policlinico Universitario “Riuniti” di Foggia, University of Foggia, 71122 Foggia, Italy
| | - Giulia Tuccari
- Department of Medical and Surgical Sciences, Institute of Geriatric, Policlinico Universitario “Riuniti” di Foggia, University of Foggia, 71122 Foggia, Italy
| | - Grazia Mastrodonato
- Department of Basic Medical Science, Neuroscience and Sensory Organs, Institute of Sports Medicine, University “Aldo Moro” of Bari, 70122 Bari, Italy
| | - Rosanna Villani
- Department of Medical and Surgical Sciences, Institute of Internal Medicine, Liver Unit, Policlinico Universitario “Riuniti” di Foggia, University of Foggia, 71122 Foggia, Italy
| | - Lucia Angela Fiore
- Department of Medical and Surgical Sciences, Institute of Geriatric, Policlinico Universitario “Riuniti” di Foggia, University of Foggia, 71122 Foggia, Italy
| | - Giulia Scioscia
- Department of Medical and Surgical Sciences, Institute of Respiratory Diseases, Policlinico Universitario “Riuniti” di Foggia, University of Foggia, 71122 Foggia, Italy
| | - Antonio Mirijello
- Department of Internal of Medicine, IRCCS Fondazione Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy
| | | | - Marco Sperandeo
- Unit of Interventional and Diagnostic Ultrasound of Internal Medicine, IRCCS Fondazione Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy
| |
Collapse
|
30
|
Zhao Z, Liu J, Weir MD, Schneider A, Ma T, Oates TW, Xu HHK, Zhang K, Bai Y. Periodontal ligament stem cell-based bioactive constructs for bone tissue engineering. Front Bioeng Biotechnol 2022; 10:1071472. [PMID: 36532583 PMCID: PMC9755356 DOI: 10.3389/fbioe.2022.1071472] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 11/17/2022] [Indexed: 09/29/2023] Open
Abstract
Objectives: Stem cell-based tissue engineering approaches are promising for bone repair and regeneration. Periodontal ligament stem cells (PDLSCs) are a promising cell source for tissue engineering, especially for maxillofacial bone and periodontal regeneration. Many studies have shown potent results via PDLSCs in bone regeneration. In this review, we describe recent cutting-edge researches on PDLSC-based bone regeneration and periodontal tissue regeneration. Data and sources: An extensive search of the literature for papers related to PDLSCs-based bioactive constructs for bone tissue engineering was made on the databases of PubMed, Medline and Google Scholar. The papers were selected by three independent calibrated reviewers. Results: Multiple types of materials and scaffolds have been combined with PDLSCs, involving xeno genic bone graft, calcium phosphate materials and polymers. These PDLSC-based constructs exhibit the potential for bone and periodontal tissue regeneration. In addition, various osteo inductive agents and strategies have been applied with PDLSCs, including drugs, biologics, gene therapy, physical stimulation, scaffold modification, cell sheets and co-culture. Conclusoin: This review article demonstrates the great potential of PDLSCs-based bioactive constructs as a promising approach for bone and periodontal tissue regeneration.
Collapse
Affiliation(s)
- Zeqing Zhao
- Department of Orthodontics, School of Stomatology, Capital Medical University, Beijing, China
| | - Jin Liu
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi’an Jiaotong University, Xi’an, China
| | - Michael D. Weir
- Biomaterials and Tissue Engineering Division, Department of Advanced Oral Sciences and Therapeutics, University of Maryland Dental School, Baltimore, MD, United States
| | - Abraham Schneider
- Department of Oncology and Diagnostic Sciences, University of Maryland School of Dentistry, Baltimore, MD, United States
| | - Tao Ma
- Department of Oncology and Diagnostic Sciences, University of Maryland School of Dentistry, Baltimore, MD, United States
| | - Thomas W. Oates
- Biomaterials and Tissue Engineering Division, Department of Advanced Oral Sciences and Therapeutics, University of Maryland Dental School, Baltimore, MD, United States
| | - Hockin H. K. Xu
- Biomaterials and Tissue Engineering Division, Department of Advanced Oral Sciences and Therapeutics, University of Maryland Dental School, Baltimore, MD, United States
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD, United States
- Center for Stem Cell Biology and Regenerative Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Ke Zhang
- Department of Orthodontics, School of Stomatology, Capital Medical University, Beijing, China
| | - Yuxing Bai
- Department of Orthodontics, School of Stomatology, Capital Medical University, Beijing, China
| |
Collapse
|
31
|
Ling L, Hou J, Wang Y, Shu H, Huang Y. Effects of Low-Intensity Pulsed Ultrasound on the Migration and Homing of Human Amnion-Derived Mesenchymal Stem Cells to Ovaries in Rats With Premature Ovarian Insufficiency. Cell Transplant 2022; 31:9636897221129171. [PMID: 36282038 PMCID: PMC9608022 DOI: 10.1177/09636897221129171] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Premature ovarian insufficiency (POI) can cause multiple sequelae and is currently incurable. Mesenchymal stem cell (MSC) transplantation might provide an effective treatment method for POI. However, the clinical application of systemic MSC transplantation is limited by the low efficiency of cell homing to target tissue in vivo, including systemic MSC transplantation for POI treatment. Thus, exploration of methods to promote MSC homing is necessary. This study was to investigate the effects of low-intensity pulsed ultrasound (LIPUS) on the migration and homing of transplanted human amnion–derived MSCs (hAD-MSCs) to ovaries in rats with chemotherapy-induced POI. For LIPUS treatment, hAD-MSCs were exposed to LIPUS or sham irradiation. Chemokine receptor expressions in hAD-MSCs were detected by polymerase chain reaction (PCR), Western blot, and immunofluorescence assays. hAD-MSC migration was detected by wound healing and transwell migration assays. Cyclophosphamide-induced POI rat models were established to evaluate the effects of LIPUS on the homing of systemically transplanted hAD-MSCs to chemotherapy-induced POI ovaries in vivo. We found that hAD-MSCs expressed chemokine receptors. The LIPUS promoted the expression of chemokine receptors, especially CXCR4, in hAD-MSCs. SDF-1 induced hAD-MSC migration. The LIPUS promoted hAD-MSC migration induced by SDF-1 through SDF-1/CXCR4 axis. SDF-1 levels significantly increased in ovaries induced by chemotherapy in POI rats. Pretreating hAD-MSCs with LIPUS increased the number of hAD-MSCs homing to ovaries in rats with chemotherapy-induced POI to some extent. However, the difference was not significant. Both hAD-MSC and LIPUS-pretreated hAD-MSC transplantation reduced ovarian injuries and improved ovarian function in rats with chemotherapy-induced POI. CXCR4 antagonist significantly reduced the number of hAD-MSCs- and LIPUS-pretreated hAD-MSCs homing to POI ovaries, and further reduced their efficacy in POI treatment. According to these findings, pretreating MSCs with LIPUS before transplantation might provide a novel, convenient, and safe technique to explore for improving the homing of systemically transplanted MSCs to target tissue.
Collapse
Affiliation(s)
- Li Ling
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China,Li Ling, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, No. 74, Linjiang Road, Chongqing 400010, China.
| | - Jiying Hou
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yan Wang
- State Key Laboratory of Ultrasound Engineering in Medicine Co-founded by Chongqing and the Ministry of Science and Technology, Chongqing Key Laboratory of Biomedical Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, China
| | - Han Shu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yubin Huang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
32
|
A Tissue Engineering Acoustophoretic (TEA) Set-up for the Enhanced Osteogenic Differentiation of Murine Mesenchymal Stromal Cells (mMSCs). Int J Mol Sci 2022; 23:ijms231911473. [PMID: 36232775 PMCID: PMC9570200 DOI: 10.3390/ijms231911473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/20/2022] [Accepted: 09/23/2022] [Indexed: 11/30/2022] Open
Abstract
Quickly developing precision medicine and patient-oriented treatment strategies urgently require novel technological solutions. The randomly cell-populated scaffolds usually used for tissue engineering often fail to mimic the highly anisotropic characteristics of native tissue. In this work, an ultrasound standing-wave-based tissue engineering acoustophoretic (TEA) set-up was developed to organize murine mesenchymal stromal cells (mMSCs) in an in situ polymerizing 3-D fibrin hydrogel. The resultant constructs, consisting of 17 cell layers spaced at 300 µm, were obtained by continuous wave ultrasound applied at a 2.5 MHz frequency. The patterned mMSCs preserved the structured behavior within 10 days of culturing in osteogenic conditions. Cell viability was moderately increased 1 day after the patterning; it subdued and evened out, with the cells randomly encapsulated in hydrogels, within 21 days of culturing. Cells in the structured hydrogels exhibited enhanced expression of certain osteogenic markers, i.e., Runt-related transcription factor 2 (RUNX2), osterix (Osx) transcription factor, collagen-1 alpha1 (COL1A1), osteopontin (OPN), osteocalcin (OCN), and osteonectin (ON), as well as of certain cell-cycle-progression-associated genes, i.e., Cyclin D1, cysteine-rich angiogenic inducer 61 (CYR61), and anillin (ANLN), when cultured with osteogenic supplements and, for ANLN, also in the expansion media. Additionally, OPN expression was also augmented on day 5 in the patterned gels cultured without the osteoinductive media, suggesting the pro-osteogenic influence of the patterned cell organization. The TEA set-up proposes a novel method for non-invasively organizing cells in a 3-D environment, potentially enhancing the regenerative properties of the designed anisotropic constructs for bone healing.
Collapse
|
33
|
Liang C, Liu X, Yan Y, Sun R, Li J, Geng W. Effectiveness and Mechanisms of Low-Intensity Pulsed Ultrasound on Osseointegration of Dental Implants and Biological Functions of Bone Marrow Mesenchymal Stem Cells. Stem Cells Int 2022; 2022:7397335. [PMID: 36199628 PMCID: PMC9529500 DOI: 10.1155/2022/7397335] [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: 05/31/2022] [Accepted: 09/09/2022] [Indexed: 11/27/2022] Open
Abstract
Dental implant restoration is the preferred choice for patients with dentition defects or edentulous patients, and obtaining stable osseointegration is the determining factor for successful implant healing. The risk of implant failure during the healing stage is still an urgent problem in clinical practice due to differences in bone quality at different implant sites and the impact of some systemic diseases on bone tissue metabolism. Low-intensity pulsed ultrasound (LIPUS) is a noninvasive physical intervention method widely recognized in the treatment of bone fracture and joint damage repair. Moreover, many studies indicated that LIPUS could effectively promote the osseointegration of dental implants and improve the osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs). This review is aimed at investigating the research progress on the use of LIPUS in dental implant medicine from three aspects: (1) discuss the promoting effects of LIPUS on osseointegration and peri-implant bone regeneration, (2) summarize the effects and associated mechanisms of LIPUS on the biological functions of BMSCs, and (3) introduce the application and prospects of LIPUS in the clinical work of dental implantation. Although many challenges need to be overcome in the future, LIPUS is bound to be an efficient and convenient therapeutic method to improve the dental implantation success rate and expand clinical implant indications.
Collapse
Affiliation(s)
- Chao Liang
- Department of Dental Implant Center, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing 100050, China
- Beijing Institute of Dental Research, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing 100050, China
| | - Xiu Liu
- Beijing Institute of Dental Research, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing 100050, China
| | - Yuwei Yan
- Department of Dental Implant Center, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing 100050, China
| | - Rongxin Sun
- Department of Dental Implant Center, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing 100050, China
| | - Jun Li
- Department of Dental Implant Center, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing 100050, China
- Beijing Institute of Dental Research, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing 100050, China
| | - Wei Geng
- Department of Dental Implant Center, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing 100050, China
| |
Collapse
|
34
|
Gupta D, Savva J, Li X, Chandler JH, Shelton RM, Scheven BA, Mulvana H, Valdastri P, Lucas M, Walmsley AD. Traditional Multiwell Plates and Petri Dishes Limit the Evaluation of the Effects of Ultrasound on Cells In Vitro. ULTRASOUND IN MEDICINE & BIOLOGY 2022; 48:1745-1761. [PMID: 35760602 DOI: 10.1016/j.ultrasmedbio.2022.05.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 04/29/2022] [Accepted: 05/02/2022] [Indexed: 06/15/2023]
Abstract
Ultrasound accelerates healing in fractured bone; however, the mechanisms responsible are poorly understood. Experimental setups and ultrasound exposures vary or are not adequately characterized across studies, resulting in inter-study variation and difficulty in concluding biological effects. This study investigated experimental variability introduced through the cell culture platform used. Continuous wave ultrasound (45 kHz; 10, 25 or 75 mW/cm2, 5 min/d) was applied, using a Duoson device, to Saos-2 cells seeded in multiwell plates or Petri dishes. Pressure field and vibration quantification and finite-element modelling suggested formation of complex interference patterns, resulting in localized displacement and velocity gradients, more pronounced in multiwell plates. Cell experiments revealed lower metabolic activities in both culture platforms at higher ultrasound intensities and absence of mineralization in certain regions of multiwell plates but not in Petri dishes. Thus, the same transducer produced variable results in different cell culture platforms. Analysis on Petri dishes further revealed that higher intensities reduced vinculin expression and distorted cell morphology, while causing mitochondrial and endoplasmic reticulum damage and accumulation of cells in sub-G1 phase, leading to cell death. More defined experimental setups and reproducible ultrasound exposure systems are required to study the real effect of ultrasound on cells for development of effective ultrasound-based therapies not just limited to bone repair and regeneration.
Collapse
Affiliation(s)
- Dhanak Gupta
- School of Dentistry, University of Birmingham, Birmingham, UK.
| | - Jill Savva
- Centre for Medical & Industrial Ultrasonics, James Watt School of Engineering, University of Glasgow, Glasgow, UK
| | - Xuan Li
- Centre for Medical & Industrial Ultrasonics, James Watt School of Engineering, University of Glasgow, Glasgow, UK
| | - James H Chandler
- Science and Technology of Robotics in Medicine (STORM) Laboratory UK, School of Electronic and Electrical Engineering, University of Leeds, Leeds, UK
| | | | - Ben A Scheven
- School of Dentistry, University of Birmingham, Birmingham, UK
| | - Helen Mulvana
- Department of Biomedical Engineering, University of Strathclyde, Glasgow, UK
| | - Pietro Valdastri
- Science and Technology of Robotics in Medicine (STORM) Laboratory UK, School of Electronic and Electrical Engineering, University of Leeds, Leeds, UK
| | - Margaret Lucas
- Centre for Medical & Industrial Ultrasonics, James Watt School of Engineering, University of Glasgow, Glasgow, UK
| | | |
Collapse
|
35
|
Voxtalisib and Low Intensity Pulsed Ultrasound Combinatorial Effect On Glioblastoma Multiforme Cancer Stem Cells Via PI3K/AKT/mTOR. Pathol Res Pract 2022; 239:154145. [DOI: 10.1016/j.prp.2022.154145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/15/2022] [Accepted: 09/26/2022] [Indexed: 11/22/2022]
|
36
|
Majnooni M, Lasaygues P, Long V, Scimeca JC, Momier D, Rico F, Buzhinsky N, Guivier-Curien C, Baron C. Monitoring of in-vitro ultrasonic stimulation of cells by numerical modeling. ULTRASONICS 2022; 124:106714. [PMID: 35344779 DOI: 10.1016/j.ultras.2022.106714] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 12/16/2021] [Accepted: 02/15/2022] [Indexed: 06/14/2023]
Abstract
Ultrasound stimulation of living tissues is a promising technique that can be safely applied for regenerative treatments. However, the ultrasound-induced mechanotransduction is still not well understood because of the large number of parameters involved at different scales and their difficult experimental accessibility. In this context, in-vitro studies may help to gain insight into the interaction between ultrasound and cells. Nevertheless, to conduct a reliable analysis of ultrasound effects on cell culture, the monitoring of the acoustic intensity delivered to the cells is of prime interest. Thanks to the development of an innovative custom experimental set-up inspired from ultrasound stimulation of bone regeneration conditions, major disturbing phenomena such as multiple reflections and standing wave formation inside the Petri dish are eliminated. Thus, the level of ultrasound stimulation, especially, in terms of spatial average temporal average intensity (ISATA), delivered to the cells can be monitored. Then, to properly estimate the level of ultrasound stimulation, a finite element model representing the experimental in-vitro configuration is developed. The numerical model manages on capturing the characteristics of the experimentally measured acoustic intensity distribution as illustrated by the experimental and numerical ISATA values of 42.3 and 45.8 mW/cm2 respectively, i.e. a relative difference of 8%. The numerical model would therefore allow exploring data inaccessible to experimental measurement and parametric studies to be carried out and facilitates the investigation of different virtual experimental configurations.
Collapse
Affiliation(s)
- M Majnooni
- Aix-Marseille Université, CNRS, ISM UMR 7287, Marseille, France; Aix-Marseille Université, CNRS, Centrale Marseille, IRPHE UMR 7342, Marseille, France.
| | - P Lasaygues
- Aix-Marseille Université, CNRS, Centrale Marseille, LMA UMR 7031, Marseille, France
| | - V Long
- Aix-Marseille Université, CNRS, Centrale Marseille, LMA UMR 7031, Marseille, France
| | - J-C Scimeca
- Université Côte d'Azur, CNRS, iBV UMR 7277, INSERM U1091, Nice, France
| | - D Momier
- Université Côte d'Azur, CNRS, iBV UMR 7277, INSERM U1091, Nice, France
| | - F Rico
- Aix-Marseille Université, CNRS, LAI UMR 7333, INSERM UMR 1067, Marseille, France
| | - N Buzhinsky
- Aix-Marseille Université, CNRS, LAI UMR 7333, INSERM UMR 1067, Marseille, France
| | - C Guivier-Curien
- Aix-Marseille Université, CNRS, Centrale Marseille, IRPHE UMR 7342, Marseille, France
| | - C Baron
- Aix-Marseille Université, CNRS, ISM UMR 7287, Marseille, France
| |
Collapse
|
37
|
Xia P, Shi Y, Wang X, Li X. Advances in the application of low-intensity pulsed ultrasound to mesenchymal stem cells. Stem Cell Res Ther 2022; 13:214. [PMID: 35619156 PMCID: PMC9137131 DOI: 10.1186/s13287-022-02887-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 05/03/2022] [Indexed: 11/10/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are stem cells that exhibit self-renewal capacity and multi-directional differentiation potential. They can be extracted from the bone marrow and umbilical cord, as well as adipose, amnion, and other tissues. They are widely used in tissue engineering and are currently considered an important source of cells in the field of regenerative medicine. Since certain limitations, such as an insufficient cell source, mature differentiation, and low transplantation efficiency, are still associated with MSCs, researchers have currently focused on improving the efficacy of MSCs. Low-intensity pulsed ultrasound (LIPUS) has mechanical, cavitation, and thermal effects that can produce different biological effects on organs, tissues, and cells. It can be used for fracture treatment, cartilage repair, and stem cell applications. An in-depth study of the role and mechanism of action of LIPUS in MSC treatment would promote our understanding of LIPUS and promote research in this field. In this article, we have reviewed the progress in research on the use of LIPUS with various MSCs and comprehensively discussed the progress in the use of LIPUS for promoting the proliferation, differentiation, and migration of MSCs, as well as its future prospects.
Collapse
Affiliation(s)
- Peng Xia
- Department of Rehabilitation Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China.
| | - Yi Shi
- Department of Rehabilitation Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Xiaoju Wang
- Department of Rehabilitation Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Xueping Li
- Department of Rehabilitation Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China.
| |
Collapse
|
38
|
Hua Z, Li S, Liu Q, Yu M, Liao M, Zhang H, Xiang X, Wu Q. Low-Intensity Pulsed Ultrasound Promotes Osteogenic Potential of iPSC-Derived MSCs but Fails to Simplify the iPSC-EB-MSC Differentiation Process. Front Bioeng Biotechnol 2022; 10:841778. [PMID: 35656194 PMCID: PMC9152674 DOI: 10.3389/fbioe.2022.841778] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 04/07/2022] [Indexed: 11/29/2022] Open
Abstract
Induced pluripotent stem cell (iPSC)-derived mesenchymal stem cells (iMSCs) are a promising cell source for bone tissue engineering. However, iMSCs have less osteogenic potential than BMSCs, and the classical iPSC-EB-iMSC process to derive iMSCs from iPSCs is too laborious as it involves multiple in vitro steps. Low-intensity pulsed ultrasound (LIPUS) is a safe therapeutic modality used to promote osteogenic differentiation of stem cells. Whether LIPUS can facilitate osteogenic differentiation of iMSCs and simplify the iPSC-EB-iMSC process is unknown. We stimulated iMSCs with LIPUS at different output intensities (20, 40, and 60 mW/cm2) and duty cycles (20, 50, and 80%). Results of ALP activity assay, osteogenic gene expression, and mineralization quantification demonstrated that LIPUS was able to promote osteogenic differentiation of iMSCs, and it worked best at the intensity of 40 mW/cm2 and the duty cycle of 50% (LIPUS40/50). The Wnt/β-catenin signaling pathway was involved in LIPUS40/50-mediated osteogenesis. When cranial bone defects were implanted with iMSCs, LIPUS40/50 stimulation resulted in a significant higher new bone filling rate (72.63 ± 17.04)% than the non-stimulated ones (34.85 ± 4.53)%. Daily exposure to LIPUS40/50 may accelerate embryoid body (EB)-MSC transition, but it failed to drive iPSCs or EB cells to an osteogenic lineage directly. This study is the first to demonstrate the pro-osteogenic effect of LIPUS on iMSCs. Although LIPUS40/50 failed to simplify the classical iPSC-EB-MSC differentiation process, our preliminary results suggest that LIPUS with a more suitable parameter set may achieve the goal. LIPUS is a promising method to establish an efficient model for iPSC application.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Qingqing Wu
- *Correspondence: Qingqing Wu, ; Xuerong Xiang,
| |
Collapse
|
39
|
Cha JM, Hwang YS, Kang DK, Lee J, Cooper ES, Mantalaris A. Development of a Novel Perfusion Rotating Wall Vessel Bioreactor with Ultrasound Stimulation for Mass-Production of Mineralized Tissue Constructs. Tissue Eng Regen Med 2022; 19:739-754. [PMID: 35532736 PMCID: PMC9294093 DOI: 10.1007/s13770-022-00447-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 02/06/2022] [Accepted: 02/20/2022] [Indexed: 10/18/2022] Open
Abstract
BACKGROUND As stem cells are considered a promising cell source for tissue engineering, many culture strategies have been extensively studied to generate in vitro stem cell-based tissue constructs. However, most approaches using conventional tissue culture plates are limited by the lack of biological relevance in stem cell microenvironments required for neotissue formation. In this study, a novel perfusion rotating wall vessel (RWV) bioreactor was developed for mass-production of stem cell-based 3D tissue constructs. METHODS An automated RWV bioreactor was fabricated, which is capable of controlling continuous medium perfusion, highly efficient gas exchange with surrounding air, as well as low-intensity pulsed ultrasound (LIPUS) stimulation. Embryonic stem cells encapsulated in alginate/gelatin hydrogel were cultured in the osteogenic medium by using our bioreactor system. Cellular viability, growth kinetics, and osteogenesis/mineralization were thoroughly evaluated, and culture media were profiled at real time. The in vivo efficacy was examined by a rabbit cranial defect model. RESULTS Our bioreactor successfully maintained the optimal culture environments for stem cell proliferation, osteogenic differentiation, and mineralized tissue formation during the culture period. The mineralized tissue constructs produced by our bioreactor demonstrated higher void filling efficacy in the large bone defects compared to the group implanted with hydrogel beads only. In addition, the LIPUS modules mounted on our bioreactor successfully reached higher mineralization of the tissue constructs compared to the groups without LIPUS stimulation. CONCLUSION This study suggests an effective biomanufacturing strategy for mass-production of implantable mineralized tissue constructs from stem cells that could be applicable to future clinical practice.
Collapse
|
40
|
Díaz-Alejo JF, González Gómez I, Earl J. Ultrasounds in cancer therapy: A summary of their use and unexplored potential. Oncol Rev 2022; 16:531. [PMID: 35340884 PMCID: PMC8941342 DOI: 10.4081/oncol.2022.531] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 09/17/2021] [Indexed: 11/26/2022] Open
Abstract
Ultrasounds (US) are a non-ionizing mechanical wave, with less adverse effects than conventional pharmacological or surgical treatments. Different biological effects are induced in tissues and cells by ultrasound actuation depending on acoustic parameters, such as the wave intensity, frequency and treatment dose. This non-ionizing radiation has considerable applications in biomedicine including surgery, medical imaging, physical therapy and cancer therapy. Depending on the wave intensity, US are applied as high-intensity ultrasounds (HIUS) and low-intensity pulsed ultrasounds (LIPUS), with different effects on cells and tissues. HIUS produce thermal and mechanical effects, resulting in a large localized temperature increase, leading to tissue ablation and even tumor necrosis. This can be achieved by focusing low intensity waves emitted from different electrically shifted transducers, known as high-intensity focused ultrasounds (HIFU). LIPUS have been used extensively as a therapeutic, surgical and diagnostic tool, with diverse biological effects observed in tissues and cultured cells. US represent a non-invasive treatment strategy that can be applied to selected areas of the body, with limited adverse effects. In fact, tumor ablation using HIFU has been used as a curative treatment in patients with an early-stage pancreatic tumor and is an effective palliative treatment in patients with advanced stage disease. However, the biological effects, dose standardization, benefit-risk ratio and safety are not fully understood. Thus, it is an emerging field that requires further research in order to reach its full potential.
Collapse
Affiliation(s)
- Jesús Frutos Díaz-Alejo
- Molecular Epidemiology and Predictive Tumor Markers Group, Ramón y Cajal Health Research Institute (IRYCIS), Madrid
- Faculty of Medicine and Health Sciences, University of Alcalá de Henares (UAH), Madrid
| | | | - Julie Earl
- Molecular Epidemiology and Predictive Tumor Markers Group, Ramón y Cajal Health Research Institute (IRYCIS), Madrid
- Biomedical Research Network in Cancer (CIBERONC), Madrid, Spain
| |
Collapse
|
41
|
Fielder M, Nair AK. Effects of scattering on ultrasound wave transmission through bioinspired scaffolds. J Mech Behav Biomed Mater 2022; 126:105065. [PMID: 34974324 DOI: 10.1016/j.jmbbm.2021.105065] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/23/2021] [Accepted: 12/24/2021] [Indexed: 01/24/2023]
Abstract
Enhancing tissue growth in scaffolds using ultrasound waves while maintaining the structural integrity of the scaffolds is a challenging problem. Previous studies have primarily focused on the effect of ultrasound waves directly on the tissue, but how the ultrasound wave interacts with the scaffold needs to be further understood, which will have a significant effect on the response of tissue to mechanical stimulation. In this study we investigate how ultrasound wave transmission differs between scaffolds with uniform pore shapes (triangle, square, rectangle, hexagon) and a bioinspired scaffold with higher structural integrity that is inspired from the atomic structure of hydroxyapatite which is a primary component of bone. We use finite element method and ultrasound experiments on 3D-printed scaffolds composed of Acrylonitrile butadiene styrene (ABS) with constant porosity to predict the effect of pore shape and wave signal frequency in the range of 1-20 MHz on acoustic wave scattering and transmission. We find that the pore shape of the scaffold affects the magnitude of ultrasound transmission even when porosity is constant, and that the bioinspired scaffolds can allow as much as 67% more wave transmission compared to scaffolds with rectangular or square pore shapes at 1 MHz frequency. Triangular and hexagonal pores are also found to produce more nonuniform transmitted wavefronts compared to the square and rectangular pores. Peak density is defined as the number of local extrema of the transmitted wave frequency power spectrum and measures the uniformity of the transmitted wave. We find that a higher peak density value for the bioinspired scaffold due to its nonsymmetric structure further produces more nonuniform wave scattering. The results of this study are important for designing bioinspired tissue scaffold geometries to control ultrasound wave penetration and to enhance mechanical stimulation for tissue growth and will also aid in the ultrasonic characterization of porous structures based on changes in pore geometry.
Collapse
Affiliation(s)
- Marco Fielder
- Multiscale Materials Modeling Lab, Department of Mechanical Engineering, University of Arkansas, Fayetteville, AR, USA
| | - Arun K Nair
- Multiscale Materials Modeling Lab, Department of Mechanical Engineering, University of Arkansas, Fayetteville, AR, USA; Institute for Nanoscience and Engineering, 731 W. Dickson Street, University of Arkansas, Fayetteville, AR, USA.
| |
Collapse
|
42
|
Sawauchi K, Fukui T, Oe K, Kumabe Y, Oda T, Yoshikawa R, Takase K, Matsushita T, Matsumoto T, Hayashi S, Kuroda R, Niikura T. Low-Intensity Pulsed Ultrasound Promotes Osteogenic Differentiation of Reamer-Irrigator-Aspirator Graft-Derived Cells in Vitro. ULTRASOUND IN MEDICINE & BIOLOGY 2022; 48:313-322. [PMID: 34785092 DOI: 10.1016/j.ultrasmedbio.2021.10.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 09/29/2021] [Accepted: 10/04/2021] [Indexed: 06/13/2023]
Abstract
Recently, reamer-irrigator-aspirator (RIA) systems have been increasingly used to harvest autologous bone grafts. RIA graft materials contain bone marrow, which provides a viable source to derive large numbers of mesenchymal stem cells. Low-intensity pulsed ultrasound (LIPUS) significantly accelerates the differentiation of stem cells derived from bone marrow. This in vitro study investigated the effect of LIPUS on the osteogenic activity and differentiation of RIA graft-derived cells. A small amount of RIA graft was obtained from seven patients. After the cells derived from RIA grafts were cultured, they were divided into two groups: the LIPUS and control groups. LIPUS was applied once daily for 20 min (1.5 MHz, pulse duration: 200 µs, pulse repetition rate: 1 kHz, spatial average-temporal average intensity: 30 mW/cm2). Alkaline phosphatase activity (113.4% and 130.1% on days 7 and 14), expression of osteoblast-related genes (ALP, Runx2) and mineralization (135.2% on day 21) of the RIA graft-derived cells were significantly higher in the LIPUS group than in the control group. However, LIPUS did not affect the cell proliferation of RIA graft-derived cells. This study indicates that LIPUS may enhance the healing of non-union and critical bone defects treated by autologous bone grafting using the RIA system.
Collapse
Affiliation(s)
- Kenichi Sawauchi
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, 650-0017, Japan
| | - Tomoaki Fukui
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, 650-0017, Japan
| | - Keisuke Oe
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, 650-0017, Japan
| | - Yohei Kumabe
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, 650-0017, Japan
| | - Takahiro Oda
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, 650-0017, Japan
| | - Ryo Yoshikawa
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, 650-0017, Japan
| | - Kyohei Takase
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, 650-0017, Japan
| | - Takehiko Matsushita
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, 650-0017, Japan
| | - Tomoyuki Matsumoto
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, 650-0017, Japan
| | - Shinya Hayashi
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, 650-0017, Japan
| | - Ryosuke Kuroda
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, 650-0017, Japan
| | - Takahiro Niikura
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, 650-0017, Japan.
| |
Collapse
|
43
|
Tang L, Wu T, Zhou Y, Zhong Y, Sun L, Guo J, Fan X, Ta D. Study on synergistic effects of carboxymethyl cellulose and LIPUS for bone tissue engineering. Carbohydr Polym 2022; 286:119278. [DOI: 10.1016/j.carbpol.2022.119278] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 01/28/2022] [Accepted: 02/18/2022] [Indexed: 02/07/2023]
|
44
|
Mouratidis PXE, ter Haar G. Latest Advances in the Use of Therapeutic Focused Ultrasound in the Treatment of Pancreatic Cancer. Cancers (Basel) 2022; 14:638. [PMID: 35158903 PMCID: PMC8833696 DOI: 10.3390/cancers14030638] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/17/2022] [Accepted: 01/19/2022] [Indexed: 02/07/2023] Open
Abstract
Traditional oncological interventions have failed to improve survival for pancreatic cancer patients significantly. Novel treatment modalities able to release cancer-specific antigens, render immunologically "cold" pancreatic tumours "hot" and disrupt or reprogram the pancreatic tumour microenvironment are thus urgently needed. Therapeutic focused ultrasound exerts thermal and mechanical effects on tissue, killing cancer cells and inducing an anti-cancer immune response. The most important advances in therapeutic focused ultrasound use for initiation and augmentation of the cancer immunity cycle against pancreatic cancer are described. We provide a comprehensive review of the use of therapeutic focused ultrasound for the treatment of pancreatic cancer patients and describe recent studies that have shown an ultrasound-induced anti-cancer immune response in several tumour models. Published studies that have investigated the immunological effects of therapeutic focused ultrasound in pancreatic cancer are described. This article shows that therapeutic focused ultrasound has been deemed to be a safe technique for treating pancreatic cancer patients, providing pain relief and improving survival rates in pancreatic cancer patients. Promotion of an immune response in the clinic and sensitisation of tumours to the effects of immunotherapy in preclinical models of pancreatic cancer is shown, making it a promising candidate for use in the clinic.
Collapse
Affiliation(s)
- Petros X. E. Mouratidis
- Department of Physics, Division of Radiotherapy and Imaging, The Institute of Cancer Research: Royal Marsden Hospital, Sutton, London SM25NG, UK;
| | | |
Collapse
|
45
|
Tynan A, Brines M, Chavan SS. Control of inflammation using non-invasive neuromodulation: past, present and promise. Int Immunol 2022; 34:119-128. [PMID: 34558623 PMCID: PMC8783606 DOI: 10.1093/intimm/dxab073] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 09/20/2021] [Indexed: 12/14/2022] Open
Abstract
The nervous system has been increasingly recognized as a novel and accessible target in the regulation of inflammation. The use of implantable and invasive devices targeting neural circuits has yielded successful results in clinical settings but does have some risk or adverse effects. Recent advances in technology and understanding of mechanistic pathways have opened new avenues of non-invasive neuromodulation. Through this review we discuss the novel research and outcomes of major modalities of non-invasive neuromodulation in the context of inflammation including transcutaneous electrical, magnetic and ultrasound neuromodulation. In addition to highlighting the scientific observations and breakthroughs, we discuss the underlying mechanisms and pathways for neural regulation of inflammation.
Collapse
Affiliation(s)
- Aisling Tynan
- Laboratory of Biomedical Science, Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY, USA
| | - Michael Brines
- Laboratory of Biomedical Science, Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY, USA
| | - Sangeeta S Chavan
- Laboratory of Biomedical Science, Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY, USA
- Elmezzi Graduate School of Molecular Medicine, 350 Community Drive, Manhasset, NY, USA
- Donald and Barbara Zucker School of Medicine at Hofstra University, Hempstead, NY, USA
| |
Collapse
|
46
|
Piezoelectric and Opto-Acoustic Material Properties of Bone. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1364:319-346. [DOI: 10.1007/978-3-030-91979-5_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
47
|
Chen H, Zhu N, Osman M, Biskowitz R, Liu J, Khandare S, Butler P, Wong PK, Kothapalli SR. A transparent low intensity pulsed ultrasound (LIPUS) chip for high-throughput cell stimulation. LAB ON A CHIP 2021; 21:4734-4742. [PMID: 34739019 DOI: 10.1039/d1lc00667c] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
We report an on-chip platform for low-intensity pulsed ultrasound (LIPUS) stimulation of cells directly cultured on a biocompatible surface of a transparent ultrasound transducer (TUT) fabricated using lithium niobate. The high light transmittance (>80%) and compact size (3 mm × 3 mm × 2 mm) of TUTs allowed easy integration with powerful optical microscopy techniques with no additional acoustic coupling and risk for contamination. TUTs were excited with varying acoustic excitation parameters (voltage amplitude and duty cycle) and resulting live cell calcium signaling was simultaneously imaged using time-lapse confocal microscopy, while the temperature change was measured by a thermocouple. Quantitative single-cell fluorescence analysis revealed the dynamic calcium signaling responses and together with the temperature measurements elucidated the optimal stimulation parameters for non-thermal and thermal effects. The fluorescence change profile was distinct from the recorded temperature change (<1 degree Celsius) profile under LIPUS treatment conditions. Cell dead assay results confirmed cells remain viable after the LIPUS treatment. These results confirmed that the TUT platform enables controllable, safe, high-throughput, and uniform mechanical stimulation of all plated cells. The on-chip LIPUS stimulation using TUTs has the potential to attract several in vitro and in vivo biomedical applications such as controlling stem cell differentiation and proliferation, studying biomechanical properties of cancer cells, and gaining fundamental insights into mechanotransduction pathways when integrated with state-of-the-art high-speed and high-resolution microscopy techniques.
Collapse
Affiliation(s)
- Haoyang Chen
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA.
| | - Ninghao Zhu
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA.
| | - Mohamed Osman
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA.
| | - Ryan Biskowitz
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA.
| | - Jinyun Liu
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA.
| | - Shubham Khandare
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA.
| | - Peter Butler
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA.
| | - Pak Kin Wong
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA.
- Department of Mechanical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
| | - Sri-Rajasekhar Kothapalli
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA.
- Penn State Cancer Institute, The Pennsylvania State University, Hershey, PA 17033, USA
| |
Collapse
|
48
|
Hao Z, Xu Z, Wang X, Wang Y, Li H, Chen T, Hu Y, Chen R, Huang K, Chen C, Li J. Biophysical Stimuli as the Fourth Pillar of Bone Tissue Engineering. Front Cell Dev Biol 2021; 9:790050. [PMID: 34858997 PMCID: PMC8630705 DOI: 10.3389/fcell.2021.790050] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 10/26/2021] [Indexed: 01/12/2023] Open
Abstract
The repair of critical bone defects remains challenging worldwide. Three canonical pillars (biomaterial scaffolds, bioactive molecules, and stem cells) of bone tissue engineering have been widely used for bone regeneration in separate or combined strategies, but the delivery of bioactive molecules has several obvious drawbacks. Biophysical stimuli have great potential to become the fourth pillar of bone tissue engineering, which can be categorized into three groups depending on their physical properties: internal structural stimuli, external mechanical stimuli, and electromagnetic stimuli. In this review, distinctive biophysical stimuli coupled with their osteoinductive windows or parameters are initially presented to induce the osteogenesis of mesenchymal stem cells (MSCs). Then, osteoinductive mechanisms of biophysical transduction (a combination of mechanotransduction and electrocoupling) are reviewed to direct the osteogenic differentiation of MSCs. These mechanisms include biophysical sensing, transmission, and regulation. Furthermore, distinctive application strategies of biophysical stimuli are presented for bone tissue engineering, including predesigned biomaterials, tissue-engineered bone grafts, and postoperative biophysical stimuli loading strategies. Finally, ongoing challenges and future perspectives are discussed.
Collapse
Affiliation(s)
- Zhuowen Hao
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Zhenhua Xu
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xuan Wang
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yi Wang
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Hanke Li
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Tianhong Chen
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yingkun Hu
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Renxin Chen
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Kegang Huang
- Wuhan Institute of Proactive Health Management Science, Wuhan, China
| | - Chao Chen
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Orthopedics, Hefeng Central Hospital, Enshi, China
| | - Jingfeng Li
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
49
|
Suarez-Castellanos I, Singh T, Chatterjee Bhowmick D, Cohen J, Jeremic A, Zderic V. Effect of Therapeutic Ultrasound on the Release of Insulin, Glucagon, and Alpha-Amylase from Ex Vivo Pancreatic Models. JOURNAL OF ULTRASOUND IN MEDICINE : OFFICIAL JOURNAL OF THE AMERICAN INSTITUTE OF ULTRASOUND IN MEDICINE 2021; 40:2709-2719. [PMID: 33595146 DOI: 10.1002/jum.15661] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 01/22/2021] [Accepted: 01/29/2021] [Indexed: 06/12/2023]
Abstract
OBJECTIVES Our previously published studies showed the potential of therapeutic ultrasound (US) as a novel non-pharmacological alternative for the treatment of secretory deficiencies in type 2 diabetes. Despite showing enhanced insulin release from beta cells, these studies did not explore the potential effects of US treatment on other cells in the islets of Langerhans such as glucagon-secreting alpha cells or acinar cells of the exocrine pancreas. METHODS We applied US parameters found capable of safely stimulating insulin secretion from pancreatic beta cells (f = 800 kHz, ISPTA = 0.5-1 W/cm2 , 5 minutes) to a diced rabbit pancreas model in culture plates (n = 6 per group). Released quantities of insulin and glucagon in response to US treatment were measured by collecting aliquots of the extracellular medium prior to the start of the treatment (t = 0 minute), immediately after treatment (t = 5 minutes) and 30 minutes after the end of treatment (t = 35 minutes). Potential release of digestive enzyme alpha-amylase as a result of US treatment was evaluated in rabbit pancreas experiments. Preliminary studies were also performed in a small number of human pancreatic islets in culture plates (n = 3 per group). The general integrity of the US-treated rabbit pancreatic tissue and human pancreatic islets was evaluated through histological analysis. RESULTS While sham-treated rabbit pancreas samples showed decreased extracellular insulin content, there was an increase in insulin release at t = 5 minutes from samples treated with US at 800 kHz and 1 W/cm2 (P <.005). Furthermore, no further insulin release was detected at t = 35 minutes. No statistically significant difference in extracellular glucagon and alpha-amylase concentrations was observed between US-treated and sham rabbit pancreas groups. Preliminary studies in human islets appeared to follow trends observed in rabbit pancreas studies. Islet and other pancreatic tissue integrity did not appear to be affected by the US treatment. CONCLUSION A potential US-based strategy for enhanced insulin release would require optimization of insulin secretion from pancreatic beta cells while minimizing glucagon and pancreatic enzyme secretions.
Collapse
Affiliation(s)
- Ivan Suarez-Castellanos
- Department of Biomedical Engineering, The George Washington University, Washington, District of Columbia, USA
- LabTAU, INSERM, Centre Léon Bérard, Université Lyon 1, Université Lyon 1, Université Lyon, Lyon, France
| | - Tania Singh
- Department of Biomedical Engineering, The George Washington University, Washington, District of Columbia, USA
| | - Diti Chatterjee Bhowmick
- Department of Biological Sciences, The George Washington University, Washington, District of Columbia, USA
| | - Joshua Cohen
- Department of Endocrinology, Medical Faculty Associates, The George Washington University, Washington, District of Columbia, USA
| | - Aleksandar Jeremic
- Department of Biological Sciences, The George Washington University, Washington, District of Columbia, USA
| | - Vesna Zderic
- Department of Biomedical Engineering, The George Washington University, Washington, District of Columbia, USA
| |
Collapse
|
50
|
Sang F, Xu J, Chen Z, Liu Q, Jiang W. Low-Intensity Pulsed Ultrasound Alleviates Osteoarthritis Condition Through Focal Adhesion Kinase-Mediated Chondrocyte Proliferation and Differentiation. Cartilage 2021; 13:196S-203S. [PMID: 32281401 PMCID: PMC8804760 DOI: 10.1177/1947603520912322] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVE Osteoarthritis (OA) is a prevalent chronic multifactorial degenerative disease characterized by joint tissue inflammation, osteophyte formation, subchondral bone sclerosis, and articular cartilage degradation. Low-intensity pulsed ultrasound (LIPUS), a noninvasive ultrasound technique, is widely used to attenuate diseases. The aim of this study was to investigate whether LIPUS can ameliorate OA, and to explore its underlying molecular mechanism. DESIGN The OA model was established in a C57BL/6 mouse by the anterior cruciate ligament transaction method. OA was assessed using arthritis scoring and weightbearing parameters. Chondrocyte proliferation was detected by a CCK-8 assay. The levels of interleukin-6 (IL-6), IL-8 and tumor necrosis factor-α (TNF-α) in synovial fluid of the mice were measured by enzyme-linked immunosorbent assay. RESULTS In OA mice, the arthritis score and weightbearing abilities were dramatically improved by LIPUS treatment. LIPUS also remarkably declined the levels of inflammatory cytokines IL-6, IL-8, and TNF-α in synovial fluid of OA mice. Moreover, LIPUS promoted chondrocyte proliferation and differentiation by activating focal adhesion kinase (FAK) signaling. Inhibition of FAK significantly blocked LIPUS-mediated cell proliferation and differentiation in vitro, as well as inflammation condition in OA mice. CONCLUSION LIPUS alleviates OA through promoting chondrocytes proliferation and differentiation by activating FAK, which could act as an intervening target for OA treatment.
Collapse
Affiliation(s)
- Fei Sang
- Department of Orthopaedics,
Lianshui County People’s Hospital, The Affiliated Lianshui County People’s
Hospital of Kangda College of Nanjing Medical University, Huai’an, Jiangsu,
China
| | - Jin Xu
- Department of Orthopaedics, The
Affiliated Huai’an Hospital of Xuzhou Medical University and The Second
People’s Hospital of Huai’an, Huai’an, Jiangsu, China
| | - Zheng Chen
- Department of Emergency Surgery,
The Affiliated Huai’an No. 1 People’s Hospital of Nanjing Medical
University, Huai’an, Jiangsu, China
| | - Qingbai Liu
- Department of Orthopaedics,
Lianshui County People’s Hospital, The Affiliated Lianshui County People’s
Hospital of Kangda College of Nanjing Medical University, Huai’an, Jiangsu,
China
| | - Wenchao Jiang
- Department of Orthopaedics, Wujin
Hospital Affiliated with Jiangsu University, the Wujin Clinical College of
Xuzhou Medical University, Changzhou, Jiangsu, China,Wenchao Jiang, Department of
Orthopedics, Wujin People’s Hospital, No. 2 of Wujin North Road,
Changzhou, Jiangsu 213017, China.
| |
Collapse
|