1
|
Chen K, Wang F, Sun X, Ge W, Zhang M, Wang L, Zheng H, Zheng S, Tang H, Zhou Z, Wu G. 3D-printed zinc oxide nanoparticles modified barium titanate/hydroxyapatite ultrasound-responsive piezoelectric ceramic composite scaffold for treating infected bone defects. Bioact Mater 2025; 45:479-495. [PMID: 39717367 PMCID: PMC11664295 DOI: 10.1016/j.bioactmat.2024.11.015] [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: 07/25/2024] [Revised: 10/22/2024] [Accepted: 11/10/2024] [Indexed: 12/25/2024] Open
Abstract
Clinically, infectious bone defects represent a significant threat, leading to osteonecrosis, severely compromising patient prognosis, and prolonging hospital stays. Thus, there is an urgent need to develop a bone graft substitute that combines broad-spectrum antibacterial efficacy and bone-inductive properties, providing an effective treatment option for infectious bone defects. In this study, the precision of digital light processing (DLP) 3D printing technology was utilized to construct a scaffold, incorporating zinc oxide nanoparticles (ZnO-NPs) modified barium titanate (BT) with hydroxyapatite (HA), resulting in a piezoelectric ceramic scaffold designed for the repair of infected bone defects. The results indicated that the addition of ZnO-NPs significantly improved the piezoelectric properties of BT, facilitating a higher HA content within the ceramic scaffold system, which is essential for bone regeneration. In vitro antibacterial assessments highlighted the scaffold's potent antibacterial capabilities. Moreover, combining the synergistic effects of low-intensity pulsed ultrasound (LIPUS) and piezoelectricity, results demonstrated that the scaffold promoted notable osteogenic and angiogenic potential, enhancing bone growth and repair. Furthermore, transcriptomics analysis results suggested that the early growth response-1 (EGR1) gene might be crucial in this process. This study introduces a novel method for constructing piezoelectric ceramic scaffolds exhibiting outstanding osteogenic, angiogenic, and antibacterial properties under the combined influence of LIPUS, offering a promising treatment strategy for infectious bone defects.
Collapse
Affiliation(s)
- Kai Chen
- Department of Oral, Plastic and Aesthetic Surgery, Hospital of Stomatology, Jilin University, Changchun, 130021, China
| | - Fang Wang
- Department of Pathogeny Biology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| | - Xiumei Sun
- Department of Orthodontics, Hospital of Stomatology, Jilin University, Changchun, 130021, China
| | - Wenwei Ge
- Key Laboratory of Automobile Materials of Ministry of Education, School of Materials Science and Engineering, Jilin University, Changchun, 130021, China
| | - Mingjun Zhang
- Department of Oral, Plastic and Aesthetic Surgery, Hospital of Stomatology, Jilin University, Changchun, 130021, China
| | - Lin Wang
- Department of Oral, Plastic and Aesthetic Surgery, Hospital of Stomatology, Jilin University, Changchun, 130021, China
| | - Haoyu Zheng
- Department of Pathogeny Biology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| | - Shikang Zheng
- Department of Oral, Plastic and Aesthetic Surgery, Hospital of Stomatology, Jilin University, Changchun, 130021, China
| | - Haoyu Tang
- Key Laboratory of Automobile Materials of Ministry of Education, School of Materials Science and Engineering, Jilin University, Changchun, 130021, China
| | - Zhengjie Zhou
- Department of Pathogeny Biology, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| | - Guomin Wu
- Department of Oral, Plastic and Aesthetic Surgery, Hospital of Stomatology, Jilin University, Changchun, 130021, China
| |
Collapse
|
2
|
Li XH, Guo D, Chen LQ, Chang ZH, Shi JX, Hu N, Chen C, Zhang XW, Bao SQ, Chen MM, Ming D. Low-intensity ultrasound ameliorates brain organoid integration and rescues microcephaly deficits. Brain 2024; 147:3817-3833. [PMID: 38739753 DOI: 10.1093/brain/awae150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 03/12/2024] [Accepted: 03/26/2024] [Indexed: 05/16/2024] Open
Abstract
Human brain organoids represent a remarkable platform for modelling neurological disorders and a promising brain repair approach. However, the effects of physical stimulation on their development and integration remain unclear. Here, we report that low-intensity ultrasound significantly increases neural progenitor cell proliferation and neuronal maturation in cortical organoids. Histological assays and single-cell gene expression analyses revealed that low-intensity ultrasound improves the neural development in cortical organoids. Following organoid grafts transplantation into the injured somatosensory cortices of adult mice, longitudinal electrophysiological recordings and histological assays revealed that ultrasound-treated organoid grafts undergo advanced maturation. They also exhibit enhanced pain-related gamma-band activity and more disseminated projections into the host brain than the untreated groups. Finally, low-intensity ultrasound ameliorates neuropathological deficits in a microcephaly brain organoid model. Hence, low-intensity ultrasound stimulation advances the development and integration of brain organoids, providing a strategy for treating neurodevelopmental disorders and repairing cortical damage.
Collapse
Affiliation(s)
- Xiao-Hong Li
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Di Guo
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Li-Qun Chen
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Zhe-Han Chang
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Jian-Xin Shi
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Nan Hu
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Chong Chen
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Xiao-Wang Zhang
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Shuang-Qing Bao
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Meng-Meng Chen
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Dong Ming
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| |
Collapse
|
3
|
Wang Y, Tian J, Liu D, Li T, Mao Y, Zhu C. Microglia in radiation-induced brain injury: Cellular and molecular mechanisms and therapeutic potential. CNS Neurosci Ther 2024; 30:e14794. [PMID: 38867379 PMCID: PMC11168970 DOI: 10.1111/cns.14794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 05/15/2024] [Accepted: 05/23/2024] [Indexed: 06/14/2024] Open
Abstract
BACKGROUND Radiation-induced brain injury is a neurological condition resulting from radiotherapy for malignant tumors, with its underlying pathogenesis still not fully understood. Current hypotheses suggest that immune cells, particularly the excessive activation of microglia in the central nervous system and the migration of peripheral immune cells into the brain, play a critical role in initiating and progressing the injury. This review aimed to summarize the latest advances in the cellular and molecular mechanisms and the therapeutic potential of microglia in radiation-induced brain injury. METHODS This article critically examines recent developments in understanding the role of microglia activation in radiation-induced brain injury. It elucidates associated mechanisms and explores novel research pathways and therapeutic options for managing this condition. RESULTS Post-irradiation, activated microglia release numerous inflammatory factors, exacerbating neuroinflammation and facilitating the onset and progression of radiation-induced damage. Therefore, controlling microglial activation and suppressing the secretion of related inflammatory factors is crucial for preventing radiation-induced brain injury. While microglial activation is a primary factor in neuroinflammation, the precise mechanisms by which radiation prompts this activation remain elusive. Multiple signaling pathways likely contribute to microglial activation and the progression of radiation-induced brain injury. CONCLUSIONS The intricate microenvironment and molecular mechanisms associated with radiation-induced brain injury underscore the crucial roles of immune cells in its onset and progression. By investigating the interplay among microglia, neurons, astrocytes, and peripheral immune cells, potential strategies emerge to mitigate microglial activation, reduce the release of inflammatory agents, and impede the entry of peripheral immune cells into the brain.
Collapse
Affiliation(s)
- Yafeng Wang
- Henan Neurodevelopment Engineering Research Center for Children, Children's Hospital Affiliated to Zhengzhou University, Department of PediatricsHenan Children's Hospital Zhengzhou Children's HospitalZhengzhouChina
- Department of Hematology and Oncology, Children's Hospital Affiliated to Zhengzhou UniversityHenan Children's Hospital Zhengzhou Children's HospitalZhengzhouChina
| | - Jiayu Tian
- Henan Neurodevelopment Engineering Research Center for Children, Children's Hospital Affiliated to Zhengzhou University, Department of PediatricsHenan Children's Hospital Zhengzhou Children's HospitalZhengzhouChina
| | - Dandan Liu
- Department of Electrocardiogram, Children's Hospital Affiliated to Zhengzhou UniversityHenan Children's Hospital Zhengzhou Children's HospitalZhengzhouChina
| | - Tao Li
- Henan Neurodevelopment Engineering Research Center for Children, Children's Hospital Affiliated to Zhengzhou University, Department of PediatricsHenan Children's Hospital Zhengzhou Children's HospitalZhengzhouChina
| | - Yanna Mao
- Department of Hematology and Oncology, Children's Hospital Affiliated to Zhengzhou UniversityHenan Children's Hospital Zhengzhou Children's HospitalZhengzhouChina
| | - Changlian Zhu
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Department of PediatricsInstitute of Neuroscience and Third Affiliated Hospital of Zhengzhou UniversityKangfuqian Street 7Zhengzhou450052None SelectedChina
- Center for Brain Repair and Rehabilitation, Department of Clinical NeuroscienceInstitute of Neuroscience and Physiology, Sahlgrenska Academy, University of GothenburgMedicinaregtan 11Göteborg40530Sweden
| |
Collapse
|
4
|
Duan H, Chen S, Mai X, Fu L, Huang L, Xiao L, Liao M, Chen H, Liu G, Xie L. Low-intensity pulsed ultrasound (LIPUS) promotes skeletal muscle regeneration by regulating PGC-1α/AMPK/GLUT4 pathways in satellite cells/myoblasts. Cell Signal 2024; 117:111097. [PMID: 38355078 DOI: 10.1016/j.cellsig.2024.111097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 01/19/2024] [Accepted: 02/11/2024] [Indexed: 02/16/2024]
Abstract
Low-Intensity Pulsed Ultrasound (LIPUS) holds therapeutic potential in promoting skeletal muscle regeneration, a biological process mediated by satellite cells and myoblasts. Despite their central roles in regeneration, the detailed mechanistic of LIPUS influence on satellite cells and myoblasts are not fully underexplored. In the current investigation, we administrated LIPUS treatment to injured skeletal muscles and C2C12 myoblasts over five consecutive days. Muscle samples were collected on days 6 and 30 post-injury for an in-depth histological and molecular assessment, both in vivo and in vitro with immunofluorescence analysis. During the acute injury phase, LIPUS treatment significantly augmented the satellite cell population, concurrently enhancing the number and size of newly formed myofibers whilst reducing fibrosis levels. At 30 days post-injury, the LIPUS-treated group demonstrated a more robust satellite cell pool and a higher myofiber count, suggesting that early LIPUS intervention facilitates satellite cell proliferation and differentiation, thereby promoting long-term recovery. Additionally, LIPUS markedly accelerated C2C12 myoblast differentiation, with observed increases in AMPK phosphorylation in myoblasts, leading to elevated expression of Glut4 and PGC-1α, and subsequent glucose uptake and mitochondrial biogenesis. These findings imply that LIPUS-induced modulation of myoblasts may culminate in enhanced cellular energy availability, laying a theoretical groundwork for employing LIPUS in ameliorating skeletal muscle regeneration post-injury. NEW & NOTEWORTHY: Utilizing the cardiotoxin (CTX) muscle injury model, we investigated the influence of LIPUS on satellite cell homeostasis and skeletal muscle regeneration. Our findings indicate that LIPUS promotes satellite cell proliferation and differentiation, thereby facilitating skeletal muscle repair. Additionally, in vitro investigations lend credence to the hypothesis that the regulatory effect of LIPUS on satellite cells may be attributed to its capability to enhance cellular energy metabolism.
Collapse
Affiliation(s)
- Huimin Duan
- Department of Rehabilitation Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510000, China
| | - Shujie Chen
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China; Department of Anesthesiology, The Seventh Affiliated Hospital, Southern Medical University, Foshan 528244, Guangdong, China
| | - Xudong Mai
- Department of Endocrinology and Metabolism, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Liping Fu
- Department of Rehabilitation Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510000, China
| | - Liujing Huang
- Medical Affairs Department, Guangzhou Betrue Technology Co., Ltd, Guangzhou 510700, China
| | - Lanling Xiao
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China; Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan 410125, China
| | - Miaomiao Liao
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China
| | - Hong Chen
- Department of Endocrinology and Metabolism, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Gang Liu
- Department of Rehabilitation Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510000, China.
| | - Liwei Xie
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China; Department of Anesthesiology, The Seventh Affiliated Hospital, Southern Medical University, Foshan 528244, Guangdong, China; Department of Internal Medicine, Shunde Women and Children's Hospital (Maternity and Child Healthcare Hospital of Shunde Foshan), Guangdong Medical University, Foshan, Guangdong, China; Department of Endocrinology and Metabolism, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China; College of Life and Health Sciences, Guangdong Industry Polytechnic, Guangzhou, Guangdong 510300, China.
| |
Collapse
|
5
|
Wang W, Li Z, Yan Y, Wu S, Yao X, Gao C, Liu L, Yu Y. LIPUS-induced neurogenesis:A potential therapeutic strategy for cognitive dysfunction in traumatic brain injury. Exp Neurol 2024; 371:114588. [PMID: 37907126 DOI: 10.1016/j.expneurol.2023.114588] [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: 07/13/2023] [Revised: 10/02/2023] [Accepted: 10/25/2023] [Indexed: 11/02/2023]
Abstract
Traumatic brain injury (TBI) precipitates cellular membrane degeneration, phospholipid degradation, neuronal demise, impaired brain electrical activity, and compromised neuroplasticity, ultimately leading to acute and chronic brain dysfunction. Low-intensity pulsed ultrasound (LIPUS) is an emerging brain therapy with the characteristics of non-invasive, high spatial resolution, and high stimulation depth. Herein, we established a controlled cortical impact model to investigate the potential reparative mechanisms of LIPUS in TBI, employing a multi-faceted research methodology encompassing behavioral assessments, immunofluorescence, neuroelectrophysiology, scratch detection of primary cortical neurons, metabolomics and transcriptomics. Our findings demonstrate that LIPUS promotes hippocampal neurogenesis following brain injury, accomplished through the elevation of phosphatidylcholine levels in the hippocampus of TBI mice. Consequently, LIPUS enhances neural electrical activity and augments neural plasticity within the CA1 subregion of the hippocampus, effectively restoring neuronal function and cognitive capabilities in TBI mice. These findings shed light on the promising role of LIPUS in TBI brain rehabilitation, offering new perspectives and theoretical foundations for future studies in this domain.
Collapse
Affiliation(s)
- Wenzhu Wang
- China Rehabilitation Science Institute, China Rehabilitation Research Center, School of Rehabilitation, Capital Medical University, Beijing, PR China; Wenzhou Medical University, Zhejiang, PR China; Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, PR China; Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, PR China
| | - Zihan Li
- China Rehabilitation Science Institute, China Rehabilitation Research Center, School of Rehabilitation, Capital Medical University, Beijing, PR China; Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, PR China; Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, PR China
| | - Yitong Yan
- China Rehabilitation Science Institute, China Rehabilitation Research Center, School of Rehabilitation, Capital Medical University, Beijing, PR China; Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, PR China; Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, PR China
| | - Shuo Wu
- First Hospital of Qinhuangdao, Qinhuangdao, Hebei, PR China
| | - Xinyu Yao
- First Hospital of Qinhuangdao, Qinhuangdao, Hebei, PR China
| | - Chen Gao
- China Rehabilitation Science Institute, China Rehabilitation Research Center, School of Rehabilitation, Capital Medical University, Beijing, PR China; Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, PR China; Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, PR China
| | - Lanxiang Liu
- First Hospital of Qinhuangdao, Qinhuangdao, Hebei, PR China.
| | - Yan Yu
- China Rehabilitation Science Institute, China Rehabilitation Research Center, School of Rehabilitation, Capital Medical University, Beijing, PR China; Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, PR China; Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, PR China.
| |
Collapse
|
6
|
He YF, Wang XL, Deng SP, Wang YL, Huang QQ, Lin S, Lyu GR. Latest progress in low-intensity pulsed ultrasound for studying exosomes derived from stem/progenitor cells. Front Endocrinol (Lausanne) 2023; 14:1286900. [PMID: 38089611 PMCID: PMC10715436 DOI: 10.3389/fendo.2023.1286900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 11/06/2023] [Indexed: 12/18/2023] Open
Abstract
Stem cells have self-renewal, replication, and multidirectional differentiation potential, while progenitor cells are undifferentiated, pluripotent or specialized stem cells. Stem/progenitor cells secrete various factors, such as cytokines, exosomes, non-coding RNAs, and proteins, and have a wide range of applications in regenerative medicine. However, therapies based on stem cells and their secreted exosomes present limitations, such as insufficient source materials, mature differentiation, and low transplantation success rates, and methods addressing these problems are urgently required. Ultrasound is gaining increasing attention as an emerging technology. Low-intensity pulsed ultrasound (LIPUS) has mechanical, thermal, and cavitation effects and produces vibrational stimuli that can lead to a series of biochemical changes in organs, tissues, and cells, such as the release of extracellular bodies, cytokines, and other signals. These changes can alter the cellular microenvironment and affect biological behaviors, such as cell differentiation and proliferation. Here, we discuss the effects of LIPUS on the biological functions of stem/progenitor cells, exosomes, and non-coding RNAs, alterations involved in related pathways, various emerging applications, and future perspectives. We review the roles and mechanisms of LIPUS in stem/progenitor cells and exosomes with the aim of providing a deeper understanding of LIPUS and promoting research and development in this field.
Collapse
Affiliation(s)
- Yi-fang He
- Department of Ultrasound, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Xia-li Wang
- Department of Ultrasound, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
- Departments of Medical Imaging, Quanzhou Medical College, Quanzhou, China
| | - Shuang-ping Deng
- Department of Ultrasound, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Yan-li Wang
- Department of Ultrasound, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Qing-qing Huang
- Department of Ultrasound, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Shu Lin
- Centre of Neurological and Metabolic Research, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, Darlinghurst, Sydney, NSW, Australia
| | - Guo-rong Lyu
- Department of Ultrasound, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
- Departments of Medical Imaging, Quanzhou Medical College, Quanzhou, China
| |
Collapse
|
7
|
Hu Y, Hu X, Luo J, Huang J, Sun Y, Li H, Qiao Y, Wu H, Li J, Zhou L, Zheng S. Liver organoid culture methods. Cell Biosci 2023; 13:197. [PMID: 37915043 PMCID: PMC10619312 DOI: 10.1186/s13578-023-01136-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 09/20/2023] [Indexed: 11/03/2023] Open
Abstract
Organoids, three-dimensional structures cultured in vitro, can recapitulate the microenvironment, complex architecture, and cellular functions of in vivo organs or tissues. In recent decades, liver organoids have been developed rapidly, and their applications in biomedicine, such as drug screening, disease modeling, and regenerative medicine, have been widely recognized. However, the lack of repeatability and consistency, including the lack of standardized culture conditions, has been a major obstacle to the development and clinical application of liver organoids. It is time-consuming for researchers to identify an appropriate medium component scheme, and the usage of some ingredients remains controversial. In this review, we summarized and compared different methods for liver organoid cultivation that have been published in recent years, focusing on controversial medium components and discussing their advantages and drawbacks. We aimed to provide an effective reference for the development and standardization of liver organoid cultivation.
Collapse
Affiliation(s)
- Yiqing Hu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
| | - Xiaoyi Hu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
| | - Jia Luo
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
| | - Jiacheng Huang
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
| | - Yaohan Sun
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
| | - Haoyu Li
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
| | - Yinbiao Qiao
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
| | - Hao Wu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
| | - Jianhui Li
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
- Department of Hepatobiliary and Pancreatic Surgery, Shulan (Hangzhou) Hospital, Zhejiang Shuren University School of Medicine, Hangzhou, 310015, China
- The Organ Repair and Regeneration Medicine Institute of Hangzhou, Hangzhou, 310003, China
| | - Lin Zhou
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China.
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, 250117, China.
| | - Shusen Zheng
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China.
- Department of Hepatobiliary and Pancreatic Surgery, Shulan (Hangzhou) Hospital, Zhejiang Shuren University School of Medicine, Hangzhou, 310015, China.
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, 250117, China.
| |
Collapse
|
8
|
Su WS, Wu CH, Song WS, Chen SF, Yang FY. Low-intensity pulsed ultrasound ameliorates glia-mediated inflammation and neuronal damage in experimental intracerebral hemorrhage conditions. J Transl Med 2023; 21:565. [PMID: 37620888 PMCID: PMC10464049 DOI: 10.1186/s12967-023-04377-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 07/21/2023] [Indexed: 08/26/2023] Open
Abstract
BACKGROUND Intracerebral hemorrhage (ICH) is a condition associated with high morbidity and mortality, and glia-mediated inflammation is a major contributor to neurological deficits. However, there is currently no proven effective treatment for clinical ICH. Recently, low-intensity pulsed ultrasound (LIPUS), a non-invasive method, has shown potential for neuroprotection in neurodegenerative diseases. This study aimed to investigate the neuroprotective effects and potential mechanisms of LIPUS on glia-mediated inflammation in ICH. METHODS This study used 289 mice to investigate the effects of LIPUS on ICH. ICH was induced by injecting bacterial collagenase (type VII-S; 0.0375 U) into the striatum of the mice. LIPUS was applied noninvasively for 3 days, including a 2-h-delayed intervention to mimic clinical usage. The study evaluated neurological function, histology, brain water content, hemoglobin content, MRI, and protein expression of neurotrophic factors, inflammatory molecules, and apoptosis. In vitro studies investigated glia-mediated inflammation by adding thrombin (10 U/mL) or conditioned media to primary and cell line cultures. The PI3K inhibitor LY294002 was used to confirm the effects of PI3K/Akt signaling after LIPUS treatment. RESULTS LIPUS treatment improved neurological deficits and reduced tissue loss, edema, and neurodegeneration after ICH. The protective effects of LIPUS resulted from decreased glia-mediated inflammation by inhibiting PI3K/Akt-NF-κB signaling, which reduced cytokine expression and attenuated microglial activation-induced neuronal damage in vitro. CONCLUSIONS LIPUS treatment improved neurological outcomes and reduced glia-mediated inflammation by inhibiting PI3K/Akt-NF-κB signaling after ICH. LIPUS may provide a non-invasive potential management strategy for ICH.
Collapse
Affiliation(s)
- Wei-Shen Su
- Department of Biomedical Imaging and Radiological Sciences, School of Biomedical Science and Engineering, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Li-Nong Street, Taipei, 11221, Taiwan
| | - Chun-Hu Wu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Wen-Shin Song
- Division of Neurosurgery, Cheng Hsin General Hospital, Taipei, Taiwan
- Department of Neurological Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Szu-Fu Chen
- Department of Physical Medicine and Rehabilitation, Cheng Hsin General Hospital, No. 45, Cheng Hsin Street, Taipei, 11221, Taiwan.
- Department of Physiology and Biophysics, National Defense Medical Center, Taipei, Taiwan.
| | - Feng-Yi Yang
- Department of Biomedical Imaging and Radiological Sciences, School of Biomedical Science and Engineering, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Li-Nong Street, Taipei, 11221, Taiwan.
| |
Collapse
|
9
|
Ambattu LA, Yeo LY. Sonomechanobiology: Vibrational stimulation of cells and its therapeutic implications. BIOPHYSICS REVIEWS 2023; 4:021301. [PMID: 38504927 PMCID: PMC10903386 DOI: 10.1063/5.0127122] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 02/27/2023] [Indexed: 03/21/2024]
Abstract
All cells possess an innate ability to respond to a range of mechanical stimuli through their complex internal machinery. This comprises various mechanosensory elements that detect these mechanical cues and diverse cytoskeletal structures that transmit the force to different parts of the cell, where they are transcribed into complex transcriptomic and signaling events that determine their response and fate. In contrast to static (or steady) mechanostimuli primarily involving constant-force loading such as compression, tension, and shear (or forces applied at very low oscillatory frequencies (≤ 1 Hz) that essentially render their effects quasi-static), dynamic mechanostimuli comprising more complex vibrational forms (e.g., time-dependent, i.e., periodic, forcing) at higher frequencies are less well understood in comparison. We review the mechanotransductive processes associated with such acoustic forcing, typically at ultrasonic frequencies (> 20 kHz), and discuss the various applications that arise from the cellular responses that are generated, particularly for regenerative therapeutics, such as exosome biogenesis, stem cell differentiation, and endothelial barrier modulation. Finally, we offer perspectives on the possible existence of a universal mechanism that is common across all forms of acoustically driven mechanostimuli that underscores the central role of the cell membrane as the key effector, and calcium as the dominant second messenger, in the mechanotransduction process.
Collapse
Affiliation(s)
- Lizebona August Ambattu
- Micro/Nanophysics Research Laboratory, School of Engineering, RMIT University, Melbourne VIC 3000, Australia
| | - Leslie Y. Yeo
- Micro/Nanophysics Research Laboratory, School of Engineering, RMIT University, Melbourne VIC 3000, Australia
| |
Collapse
|
10
|
Blackmore DG, Razansky D, Götz J. Ultrasound as a versatile tool for short- and long-term improvement and monitoring of brain function. Neuron 2023; 111:1174-1190. [PMID: 36917978 DOI: 10.1016/j.neuron.2023.02.018] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 01/19/2023] [Accepted: 02/13/2023] [Indexed: 03/15/2023]
Abstract
Treating the brain with focused ultrasound (FUS) at low intensities elicits diverse responses in neurons, astroglia, and the extracellular matrix. In combination with intravenously injected microbubbles, FUS also opens the blood-brain barrier (BBB) and facilitates focal drug delivery. However, an incompletely understood cellular specificity and a wide parameter space currently limit the optimal application of FUS in preclinical and human studies. In this perspective, we discuss how different FUS modalities can be utilized to achieve short- and long-term improvements, thereby potentially treating brain disorders. We review the ongoing efforts to determine which parameters induce neuronal inhibition versus activation and how mechanoreceptors and signaling cascades are activated to induce long-term changes, including memory improvements. We suggest that optimal FUS treatments may require different FUS modalities and devices, depending on the targeted brain area or local pathology, and will be greatly enhanced by new techniques for monitoring FUS efficacy.
Collapse
Affiliation(s)
- Daniel G Blackmore
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Daniel Razansky
- Institute for Biomedical Engineering, Institute of Pharmacology and Toxicology, Faculty of Medicine, University of Zurich, 8057 Zurich, Switzerland; Institute for Biomedical Engineering, Department of Information Technology and Electrical Engineering, ETH Zurich, 8093 Zurich, Switzerland
| | - Jürgen Götz
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia.
| |
Collapse
|
11
|
Lin Z, Gao L, Hou N, Zhi X, Zhang Y, Che Z, Deng A. Application of low-intensity pulsed ultrasound on tissue resident stem cells: Potential for ophthalmic diseases. Front Endocrinol (Lausanne) 2023; 14:1153793. [PMID: 37008913 PMCID: PMC10063999 DOI: 10.3389/fendo.2023.1153793] [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] [Received: 01/30/2023] [Accepted: 03/07/2023] [Indexed: 03/19/2023] Open
Abstract
INTRODUCTION Tissue-resident stem cells (TRSCs) have the ability to self-renew and differentiate throughout an individual's lifespan, and they utilize both mechanisms to maintain homeostasis and regenerate damaged tissues. Several studies suggest that these stem cells can serve as a potential source for cell-replacement-based therapy by promoting differentiation or expansion. In recent years, low-intensity pulsed ultrasound (LIPUS) has been demonstrated to effectively stimulate stem cell proliferation and differentiation, promote tissue regeneration, and inhibit inflammatory responses. AIMS To present a comprehensive overview of current application and mechanism of LIPUS on tissue resident stem cells. METHODS We searched PubMed, Web of Science for articles on the effects of LIPUS on tissue resident stem cells and its application. RESULTS The LIPUS could modulate cellular activities such as cell viability, proliferation and differentiation of tissue resident stem cells and related cells through various cellular signaling pathways. Currently, LIPUS, as the main therapeutic ultrasound, is being widely used in the treatment of preclinical and clinical diseases. CONCLUSION The stem cell research is the hot topic in the biological science, while in recent years, increasing evidence has shown that TRSCs are good targets for LIPUS-regulated regenerative medicine. LIPUS may be a novel and valuable therapeutic approach for the treatment of ophthalmic diseases. How to further improve its efficiency and accuracy, as well as the biological mechanism therein, will be the focus of future research.
Collapse
|
12
|
Esmaeili J, Barati A, Charelli LE. Discussing the final size and shape of the reconstructed tissues in tissue engineering. J Artif Organs 2022:10.1007/s10047-022-01360-1. [PMID: 36125581 DOI: 10.1007/s10047-022-01360-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 08/15/2022] [Indexed: 11/30/2022]
Abstract
Tissue engineering (TE) has made a revolution in repairing, replacing, or regenerating tissues or organs, but it has still a long way ahead. The mechanical properties along with suitable physicochemical and biological characteristics are the initial criteria for scaffolds in TE that should be fulfilled. This research will provide another point of view toward TE challenges concerning the morphological and geometrical aspects of the reconstructed tissue and which parameters may affect it. Based on our survey, there is a high possibility that the final reconstructed tissue may be different in size and shape compared to the original design scaffold. Thereby, the 3D-printed scaffold might not guarantee an accurate tissue reconstruction. The main justification for this is the unpredicted behavior of cells, specifically in the outer layer of the scaffold. It can also be a concern when the scaffold is implanted while cell migration cannot be controlled through the in vivo signaling pathways, which might cause cancer challenges. To sum up, it is concluded that more studies are necessary to focus on the size and geometry of the final reconstructed tissue.
Collapse
Affiliation(s)
- Javad Esmaeili
- Department of Chemical Engineering, Faculty of Engineering, Arak University, Arak, 38156-88349, Iran.,Tissue Engineering Department, TISSUEHUB Co., Tehran, Iran
| | - Aboulfazl Barati
- Department of Chemical Engineering, Faculty of Engineering, Arak University, Arak, 38156-88349, Iran.
| | - Letícia Emiliano Charelli
- Nanotechnology Engineering Program, Alberto Luiz Coimbra Institute for Graduate Studies and Research in Engineering, COPPE, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| |
Collapse
|
13
|
Hu Y, Jia Y, Wang H, Cao Q, Yang Y, Zhou Y, Tan T, Huang X, Zhou Q. Low-intensity pulsed ultrasound promotes cell viability and inhibits apoptosis of H9C2 cardiomyocytes in 3D bioprinting scaffolds via PI3K-Akt and ERK1/2 pathways. J Biomater Appl 2022; 37:402-414. [PMID: 35574901 DOI: 10.1177/08853282221102669] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The aim of this study was to investigate whether low-intensity pulsed ultrasound (LIPUS) promotes myocardial cell viability in three-dimensional (3D) cell-laden gelatin methacryloyl (GelMA) scaffolds. Cardiomyoblasts (H9C2s) were mixed in 6% (w/v) GelMA bio-inks and printed using an extrusion-based 3D bioprinter. These scaffolds were exposed to LIPUS with different parameters or sham-irradiated to optimize the LIPUS treatment. The viability of H9C2s was measured using Cell Counting Kit-8 (CCK8), cell cycle, and live and dead cell double-staining assays. Western blot analysis was performed to determine the protein expression levels. We successfully fabricated 3D bio-printed cell-laden GelMA scaffolds. CCK8 and live and dead cell double-staining assays indicated that the optimal conditions for LIPUS were a frequency of 0.5 MHz and an exposure time of 10 min. Cell cycle analysis showed that LIPUS promoted the entry of cells into the S and G2/M phases from the G0/G1 phase. Western blot analysis revealed that LIPUS promoted the phosphorylation and activation of ERK1/2 and PI3K-Akt. The ERK1/2 inhibitor (U0126) and PI3K inhibitor (LY294002) significantly reduced LIPUS-induced phosphorylation of ERK1/2 and PI3K-Akt, respectively, which in turn reduced the LIPUS-induced viability of H9C2s in 3D bio-printed cell-laden GelMA scaffolds. A frequency of 0.5 MHz and exposure time of 10 min for LIPUS exposure can be adapted to achieve optimized culture effects on myocardial cells in 3D bio-printed cell-laden GelMA scaffolds via the ERK1/2 and PI3K-Akt signaling pathways.
Collapse
Affiliation(s)
- Yugang Hu
- Department of Ultrasound Imaging, 117921Renmin Hospital of Wuhan University, Wuhan, China
| | - Yan Jia
- Department of Ultrasound Imaging, 117921Renmin Hospital of Wuhan University, Wuhan, China
| | - Hao Wang
- Department of Ultrasound Imaging, 117921Renmin Hospital of Wuhan University, Wuhan, China
| | - Quan Cao
- Department of Ultrasound Imaging, 117921Renmin Hospital of Wuhan University, Wuhan, China
| | - Yuanting Yang
- Department of Ultrasound Imaging, 117921Renmin Hospital of Wuhan University, Wuhan, China
| | - Yanxiang Zhou
- Department of Ultrasound Imaging, 117921Renmin Hospital of Wuhan University, Wuhan, China
| | - Tuantuan Tan
- Department of Ultrasound Imaging, 117921Renmin Hospital of Wuhan University, Wuhan, China
| | - Xin Huang
- Department of Ultrasound Imaging, 117921Renmin Hospital of Wuhan University, Wuhan, China
| | - Qing Zhou
- Department of Ultrasound Imaging, 117921Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
14
|
Qin HC, Luo ZW, Zhu YL. Physical energy-based ultrasound shifts M1 macrophage differentiation towards M2 state. World J Stem Cells 2022; 14:214-218. [PMID: 35432733 PMCID: PMC8963378 DOI: 10.4252/wjsc.v14.i2.214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 12/12/2021] [Accepted: 02/16/2022] [Indexed: 02/06/2023] Open
Abstract
Recently, we read with interest the article entitled “Unveiling the Morphogenetic Code: A New Path at the Intersection of Physical Energies and Chemical Signaling”. In this paper, the investigation into the systematic and comprehensive bio-effects of physical energies prompted us to reflect on our research. We believe that ultrasound, which possesses a special physical energy, also has a certain positive regulatory effect on macrophages, and we have already obtained some preliminary research results that support our hypothesis.
Collapse
Affiliation(s)
- Hao-Cheng Qin
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Zhi-Wen Luo
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Yu-Lian Zhu
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| |
Collapse
|
15
|
ZHOU YUAN, Liu G, Guo S. Advances in Ultrasound-Responsive Hydrogels for Biomedical Applications. J Mater Chem B 2022; 10:3947-3958. [DOI: 10.1039/d2tb00541g] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Various intelligent hydrogels have been developed for biomedical applications because they can achieve multiple, variable, controllable and reversible changes in their shape and properties in a spatial and temporal manner,...
Collapse
|
16
|
Ultrasound-Enabled Therapeutic Delivery and Regenerative Medicine: Physical and Biological Perspectives. ACS Biomater Sci Eng 2021; 7:4371-4387. [PMID: 34460238 DOI: 10.1021/acsbiomaterials.1c00276] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The role of ultrasound in medicine and biological sciences is expanding rapidly beyond its use in conventional diagnostic imaging. Numerous studies have reported the effects of ultrasound on cellular and tissue physiology. Advances in instrumentation and electronics have enabled successful in vivo applications of therapeutic ultrasound. Despite path breaking advances in understanding the biophysical and biological mechanisms at both microscopic and macroscopic scales, there remain substantial gaps. With the progression of research in this area, it is important to take stock of the current understanding of the field and to highlight important areas for future work. We present herein key developments in the biological applications of ultrasound especially in the context of nanoparticle delivery, drug delivery, and regenerative medicine. We conclude with a brief perspective on the current promise, limitations, and future directions for interfacing ultrasound technology with biological systems, which could provide guidance for future investigations in this interdisciplinary area.
Collapse
|
17
|
Li Q, Li B, Tao B, Zhao C, Fan B, Wang Q, Sun C, Duan H, Pang Y, Fu X, Feng S. Identification of four genes and biological characteristics associated with acute spinal cord injury in rats integrated bioinformatics analysis. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:570. [PMID: 33987268 PMCID: PMC8105796 DOI: 10.21037/atm-21-603] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 03/05/2021] [Indexed: 12/25/2022]
Abstract
BACKGROUND Spinal cord injury (SCI) is a serious condition that can cause physical disability and sensory dysfunction. Cytokines play an extremely important role in the acute phase of SCI. Clarifying the cytokine expression profile is of great importance. METHODS Cytokine array analysis was used to explore the changes in 67 different proteins at 0 hours, 2 hours, 1 day, 3 days, and 7 days after acute SCI in rats. The differentially expressed cytokines in the various periods were analyzed and compared. The biological processes related to the differentially expressed proteins were examined using Gene Ontology (GO) analysis. RESULTS Immediately after SCI (0 hours), only ciliary neurotrophic factor (CNTF) was slightly up-regulated, while 23 other proteins were down-regulated. At 2 hours after SCI, there were 3 upregulated and 21 downregulated proteins. At 1 day after SCI, there were 5 upregulated and 6 downregulated proteins. At 3 days after SCI, there were 6 upregulated and 4 downregulated proteins. At 7 days after SCI, there were 4 upregulated and 9 downregulated proteins. Erythropoietin (EPO) and Fms related tyrosine kinase 3 ligand (Flt-3L) were downregulated at all time points. CD48 was decreased at 2 hours to 7 days after SCI. Monocyte chemotactic protein-1 (MCP-1) was the only protein that was upregulated at 2 hours to 7 days. The GO and pathway analyses revealed that the cytokine-related pathways, cell death, and proliferation might play a key role during secondary SCI. CONCLUSIONS This study identified 3 downregulated proteins during SCI, that being EPO, Flt-3L, and CD48. MCP-1 was the only upregulated protein, and its expression was upregulated till day 7 following SCI. These 4 identified genes may be potential therapeutic targets for the treatment of SCI.
Collapse
Affiliation(s)
- Qiang Li
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
- International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin, China
- Department of Orthopedics, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, China
| | - Bo Li
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
- Department of Orthopedics, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
| | - Bo Tao
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
- International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin, China
| | - Chenxi Zhao
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
- International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin, China
| | - Baoyou Fan
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
- International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin, China
| | - Qi Wang
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
- International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin, China
- Department of Orthopedics, Tianjin Hospital of ITCWM Nankai Hospital, Tianjin, China
| | - Chao Sun
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
- International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin, China
| | - Huiquan Duan
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
- International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin, China
| | - Yilin Pang
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
- International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin, China
| | - Xuanhao Fu
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
- International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin, China
| | - Shiqing Feng
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
- International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin, China
| |
Collapse
|
18
|
Zheng K, Han X, Su Y, Wang Q, Ma Q, Zheng K. Effects of targeted Notch1 silencing on the biological processes of the T24 and 5637 cells in vitro. Oncol Lett 2021; 21:305. [PMID: 33732381 PMCID: PMC7905604 DOI: 10.3892/ol.2021.12566] [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: 01/20/2020] [Accepted: 11/26/2020] [Indexed: 11/30/2022] Open
Abstract
The present study aimed to investigate the roles of Notch1 in the biological processes of bladder cancer cells (BCCs) in vitro. Short hairpin (sh)RNA targeting Notch1 was designed and constructed, and the T24 and 5637 BCCs were selected for transfection. The cells were classified into two groups: shRNA negative control (NC) and Notch1 shRNA. MTT and Transwell assays, and flow cytometry were performed to examine the changes in cell proliferation, invasiveness, and apoptosis, respectively. In addition, reverse transcription-quantitative PCR and western blot analysis was used to detect the mRNA and protein expression levels of apoptosis-related proteins (Bax, Bid and Bcl2) and epithelial-mesenchymal transition factors (vimentin and E- and N-cadherin). Compared with that in the shRNA NC group, the Notch1 shRNA group showed significantly decreased cell proliferation rate and invasiveness; increased apoptotic rate; elevated mRNA expression levels of Bad, Bid and E-cadherin; and reduced mRNA expression levels of Bcl2, N-cadherin and vimentin. The trends for protein expression levels were the same as those for mRNA levels. Notch1 silencing inhibited invasion and promoted apoptosis of BCCs.
Collapse
Affiliation(s)
- Kewen Zheng
- Department of Urology, The First Affiliated Hospital of Wenzhou Medical University, The First Clinical College of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Xiaomin Han
- Blood Conservation Institute, School of Basic and Forensic Medicine, Baotou Medical College, Baotou, Inner Mongolia Autonomous Region 014040, P.R. China
| | - Yan Su
- Blood Conservation Institute, School of Basic and Forensic Medicine, Baotou Medical College, Baotou, Inner Mongolia Autonomous Region 014040, P.R. China
| | - Qinghai Wang
- Department of Kidney Transplantation, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Qiang Ma
- Blood Conservation Institute, School of Basic and Forensic Medicine, Baotou Medical College, Baotou, Inner Mongolia Autonomous Region 014040, P.R. China
| | - Kesi Zheng
- Department of Thyroid and Breast Surgery, Wenzhou People's Hospital, The Third Clinical College of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| |
Collapse
|