1
|
Wen ZH, Wu ZS, Cheng HJ, Huang SY, Tang SH, Teng WN, Su FW, Chen NF, Sung CS. Intrathecal Fumagillin Alleviates Chronic Neuropathy-Induced Nociceptive Sensitization and Modulates Spinal Astrocyte-Neuronal Glycolytic and Angiogenic Proteins. Mol Neurobiol 2025; 62:246-263. [PMID: 38837104 DOI: 10.1007/s12035-024-04254-w] [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: 03/13/2024] [Accepted: 05/21/2024] [Indexed: 06/06/2024]
Abstract
Nociceptive sensitization is accompanied by the upregulation of glycolysis in the central nervous system in neuropathic pain. Growing evidence has demonstrated glycolysis and angiogenesis to be related to the inflammatory processes. This study investigated whether fumagillin inhibits neuropathic pain by regulating glycolysis and angiogenesis. Fumagillin was administered through an intrathecal catheter implanted in rats with chronic constriction injury (CCI) of the sciatic nerve. Nociceptive, behavioral, and immunohistochemical analyses were performed to evaluate the effects of the inhibition of spinal glycolysis-related enzymes and angiogenic factors on CCI-induced neuropathic pain. Fumagillin reduced CCI-induced thermal hyperalgesia and mechanical allodynia from postoperative days (POD) 7 to 14. The expression of angiogenic factors, vascular endothelial growth factor (VEGF) and angiopoietin 2 (ANG2), increased in the ipsilateral lumbar spinal cord dorsal horn (SCDH) following CCI. The glycolysis-related enzymes, pyruvate kinase M2 (PKM2) and lactate dehydrogenase A (LDHA) significantly increased in the ipsilateral lumbar SCDH following CCI on POD 7 and 14 compared to those in the control rats. Double immunofluorescence staining indicated that VEGF and PKM2 were predominantly expressed in the astrocytes, whereas ANG2 and LDHA were predominantly expressed in the neurons. Intrathecal infusion of fumagillin significantly reduced the expression of angiogenic factors and glycolytic enzymes upregulated by CCI. The expression of hypoxia-inducible factor-1α (HIF-1α), a crucial transcription factor that regulates angiogenesis and glycolysis, was also upregulated after CCI and inhibited by fumagillin. We concluded that intrathecal fumagillin may reduce the expression of ANG2 and LDHA in neurons and VEGF and PKM2 in the astrocytes of the SCDH, further attenuating spinal angiogenesis in neuropathy-induced nociceptive sensitization. Hence, fumagillin may play a role in the inhibition of peripheral neuropathy-induced neuropathic pain by modulating glycolysis and angiogenesis.
Collapse
Affiliation(s)
- Zhi-Hong Wen
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung, 804201, Taiwan
- Institute of Biopharmaceutical Sciences, National Sun Yat-sen University, Kaohsiung, 80424, Taiwan
| | - Zong-Sheng Wu
- Division of Pain Management, Department of Anesthesiology, Taipei Veterans General Hospital, Taipei, 112201, Taiwan
| | - Hao-Jung Cheng
- Institute of Biopharmaceutical Sciences, National Sun Yat-sen University, Kaohsiung, 80424, Taiwan
| | - Shi-Ying Huang
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen, 361021, China
| | - Shih-Hsuan Tang
- Division of Pain Management, Department of Anesthesiology, Taipei Veterans General Hospital, Taipei, 112201, Taiwan
| | - Wei-Nung Teng
- Division of Pain Management, Department of Anesthesiology, Taipei Veterans General Hospital, Taipei, 112201, Taiwan
- School of Medicine, National Yang-Ming Chiao Tung University, Taipei, 112304, Taiwan
| | - Fu-Wei Su
- Division of Pain Management, Department of Anesthesiology, Taipei Veterans General Hospital, Taipei, 112201, Taiwan
- School of Medicine, National Yang-Ming Chiao Tung University, Taipei, 112304, Taiwan
| | - Nan-Fu Chen
- Division of Neurosurgery, Department of Surgery, Kaohsiung Armed Forces General Hospital, Kaohsiung, 80284, Taiwan
- Institute of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung, 804201, Taiwan
| | - Chun-Sung Sung
- Division of Pain Management, Department of Anesthesiology, Taipei Veterans General Hospital, Taipei, 112201, Taiwan.
- School of Medicine, National Yang-Ming Chiao Tung University, Taipei, 112304, Taiwan.
| |
Collapse
|
2
|
Su W, Liao C, Liu X. Angiogenic and neurogenic potential of dental-derived stem cells for functional pulp regeneration: A narrative review. Int Endod J 2024. [PMID: 39660369 DOI: 10.1111/iej.14180] [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: 04/29/2024] [Revised: 10/26/2024] [Accepted: 11/22/2024] [Indexed: 12/12/2024]
Abstract
BACKGROUND Dental pulp tissue engineering is expected to become an ideal treatment for irreversible pulpitis and apical periodontitis. However, angiogenesis and neurogenesis for functional pulp regeneration have not yet met the standard for large-scale clinical application, and need further research. OBJECTIVE This review focused on the potential mechanisms of angiogenesis and neurogenesis in pulp regeneration, including stem cell types, upstream and downstream regulatory molecules and cascade signalling pathways, thereby providing a theoretical basis and inspiring new ideas to improve the effectiveness of dental pulp tissue engineering. METHODS An electronic literature search was carried out using the keywords of 'pulp regeneration', 'stem cell transplantation', 'dental pulp stem cells', 'angiogenesis' and 'neurogenesis'. The resulting literature was screened and reviewed. RESULTS Stem cells used in dental pulp tissue engineering can be classified as dental-derived and non-dental-derived stem cells, amongst which dental pulp stem cells (DPSC) have achieved promising results in animal experiments and clinical trials. Multiple molecules and signalling pathways are involved in the process of DPSC-mediated angiogenic and neurogenetic regeneration. In order to promote angiogenesis and neurogenesis in pulp regeneration, feasible measures include the addition of growth factors, the modulation of transcription factors and signalling pathways, the use of extracellular vesicles and the modification of bioscaffold materials. CONCLUSION Dental pulp tissue engineering has had breakthroughs in preclinical and clinical studies in vivo. Overcoming difficulties in pulpal angiogenesis and neurogenesis, and achieving functional pulp regeneration will lead to a significant impact in endodontics.
Collapse
Affiliation(s)
- Wanting Su
- School of Stomatology, Jinan University, Guangzhou, China
| | - Chufang Liao
- School of Stomatology, Jinan University, Guangzhou, China
- Clinical Research Platform for Interdiscipline of Stomatology, Jinan University, Guangzhou, China
- Hospital of stomatology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Xiangning Liu
- School of Stomatology, Jinan University, Guangzhou, China
- Clinical Research Platform for Interdiscipline of Stomatology, Jinan University, Guangzhou, China
- Hospital of stomatology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| |
Collapse
|
3
|
Cheng C, Deng M, Cheng C, Wu H, Wang Y, Lu M, Yao Z, Li K, Zhang X, Yu B. FOXO1-mTOR pathway in vascular pericyte regulates the formation of type H vessels to control bone metabolism. J Orthop Translat 2024; 49:246-263. [PMID: 39524152 PMCID: PMC11546805 DOI: 10.1016/j.jot.2024.08.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 07/31/2024] [Accepted: 08/13/2024] [Indexed: 11/16/2024] Open
Abstract
Background As the population aging progresses, age-related osteoporosis has become one of the most common and severe chronic degenerative diseases. Due to insufficient understanding of its complex pathomechanisms, current clinical treatments often suffer from many negative effects. Type H vessels play critical role in bone remodeling owing to their specialized function in coupling angiogenesis and osteogenesis. Increasing evidences have shown a close association between the age-related decline of type H vessels and bone loss. However, the underlying mechanisms whereby the regression of type H vessels with aging remain largely unknown. Methods Col2-Cre ERT /Foxo1 flox/flox mice and FOXO1 inhibitor (AS1842856) treated adult (6 months) and middle aged (10 months) mice were utilized for evaluating the variations in bone volume, bone microarchitecture and type H vessels through micro-CT scanning analysis, histological staining and immunofluorescence staining. In vitro tube-forming and scratch assays were applied to evaluate the angiogenic capacity of human umbilical vein endothelial cells (HUVECs) exposed to AS1842856 or conditioned culture milieu of Human Brain Vascular Pericytes (HBVPs). The expression of pericyte marker proteins, myofibroblast-related proteins and genes in inhibitors-stimulated HBVPs were detected via western blot analysis and Reverse transcription-quantitative PCR (RT-qPCR). Furthermore, perivascular myofibroblastic-like transformation was confirmed in AS1842856-treated animal models through immunofluorescence staining. We also constructed Adipoq-Cre/Foxo1 flox/flox conditional knockout mice and measured their bone mass and type H vessels by micro-CT and immunofluorescence staining. Mechanistic experiments in vitro were conducted via detection of mTOR signalling expression in HBVPs with pharmacological intervention (AS1842856 and rapamycin), genetic knockdown of Foxo1, or FOXO1-overexpression plasmid treatment, verified by RT-qPCR, western blot analysis and cellular immunofluorescence staining. In vivo validation was conducted on Adipoq-Cre/Foxo1 flox/flox mice using immunofluorescence staining. Finally, alterations in osteo-morphology and type H vessels were verified in AS1842856-treated and rapamycin-treated aged mouse models. Results This study identified FOXO1 in pericytes as key components for the formation of type H vessels. We found that FOXO1 expression in pericytes decreases with aging, and pharmacological blocking with AS1842856 promoted type H vessels degeneration and increased bone loss in adult and middle-aged mice, while rapamycin prevented the above pathology in middle-aged mice. We further showed that the loss of FOXO1 in Adipoq + pericytes led to degeneration of type H vessels and bone loss in mice. Mechanistically, the inhibition of FOXO1 by AS1842856 or knockdown of Foxo1 by siRNAs activated mTOR signaling, thereby resulting in the myofibroblastic transformation of pericytes. Furthermore, blocking mTOR signaling by rapamycin rescued the above effects in vitro and in vivo. Conclusion Our findings uncover a hitherto unknown role of FOXO1 in maintaining the phenotype and function of pericytes, thereby promoting formation of type H vessels. This suggests that targeting the FOXO1-mTOR pathway in pericytes could be a potential therapeutic approach to overcome the regression of type H vessels and bone degeneration with aging. The translational potential of this article Our research uncovers a previously unidentified role of FOXO1 in preserving pericyte characteristics and promoting the development of type H vessels. Future translational research targeting the FOXO1-mTOR pathway in pericytes may provide new strategies for the prevention and treatment of age-related osteoporosis in the clinic.
Collapse
Affiliation(s)
- Caiyu Cheng
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, PR China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, PR China
| | - Mingye Deng
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, PR China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, PR China
| | - Chubin Cheng
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, PR China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, PR China
| | - Hangtian Wu
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, PR China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, PR China
| | - Yutian Wang
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, PR China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, PR China
| | - Mincheng Lu
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, PR China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, PR China
| | - Zilong Yao
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, PR China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, PR China
| | - Kaiqun Li
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, PR China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, PR China
| | - Xianrong Zhang
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, PR China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, PR China
| | - Bin Yu
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, PR China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, PR China
| |
Collapse
|
4
|
Yang S, Leung AYP, Wang Z, Yiu CKY, Dissanayaka WL. Proanthocyanidin surface preconditioning of dental pulp stem cell spheroids enhances dimensional stability and biomineralization in vitro. Int Endod J 2024; 57:1639-1654. [PMID: 39046812 DOI: 10.1111/iej.14126] [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: 11/27/2023] [Revised: 04/15/2024] [Accepted: 07/12/2024] [Indexed: 07/27/2024]
Abstract
AIM Lack of adequate mechanical strength and progressive shrinkage over time remain challenges in scaffold-free microtissue-based dental pulp regeneration. Surface collagen cross-linking holds the promise to enhance the mechanical stability of microtissue constructs and trigger biological regulations. In this study, we proposed a novel strategy for surface preconditioning microtissues using a natural collagen cross-linker, proanthocyanidin (PA). We evaluated its effects on cell viability, tissue integrity, and biomineralization of dental pulp stem cell (DPSCs)-derived 3D cell spheroids. METHODOLOGY Microtissue and macrotissue spheroids were fabricated from DPSCs and incubated with PA solution for surface collagen cross-linking. Microtissue viability was examined by live/dead staining and 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, with transverse dimension change monitored. Microtissue surface stiffness was measured by an atomic force microscope (AFM). PA-preconditioned microtissues and macrotissues were cultured under basal or osteogenic conditions. Immunofluorescence staining of PA-preconditioned microtissues was performed to detect dentin sialophosphoprotein (DSPP) and F-actin expressions. PA-preconditioned macrotissues were subjected to histological analysis, including haematoxylin-eosin (HE), alizarin red, and Masson trichrome staining. Immunohistochemistry staining was used to detect alkaline phosphatase (ALP) and dentin matrix acidic phosphoprotein 1 (DMP-1) expressions. RESULTS PA preconditioning had no adverse effects on microtissue spheroid viability and increased surface stiffness. It reduced dimensional shrinkage for over 7 days in microtissues and induced a larger transverse-section area in the macrotissue. PA preconditioning enhanced collagen formation, mineralized nodule formation, and elevated ALP and DMP-1 expressions in macrotissues. Additionally, PA preconditioning induced higher F-actin and DSPP expression in microtissues, while inhibition of F-actin activity by cytochalasin B attenuated PA-induced dimensional change and DSPP upregulation. CONCLUSION PA surface preconditioning of DPSCs spheroids demonstrates excellent biocompatibility while effectively enhancing tissue structure stability and promoting biomineralization. This strategy strengthens tissue integrity in DPSC-derived spheroids and amplifies osteogenic differentiation potential, advancing scaffold-free tissue engineering applications in regenerative dentistry.
Collapse
Affiliation(s)
- Shengyan Yang
- Applied Oral Sciences & Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China
| | - Andy Yu Pan Leung
- Applied Oral Sciences & Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China
| | - Zheng Wang
- Department of Mechanical Engineering, The University of Hong Kong, Hong Kong SAR, China
| | - Cynthia Kar Yung Yiu
- Paediatric Dentistry & Orthodontics, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China
| | - Waruna Lakmal Dissanayaka
- Applied Oral Sciences & Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
5
|
Li Y, Zhang L, Yang W, Lin L, Pan J, Lu M, Zhang Z, Li Y, Li C. Notoginsenoside R 1 decreases intraplaque neovascularization by governing pericyte-endothelial cell communication via Ang1/Tie2 axis in atherosclerosis. Phytother Res 2024; 38:4036-4052. [PMID: 38886264 DOI: 10.1002/ptr.8257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 04/30/2024] [Accepted: 05/16/2024] [Indexed: 06/20/2024]
Abstract
Atherosclerosis represents the major cause of mortality worldwide and triggers higher risk of acute cardiovascular events. Pericytes-endothelial cells (ECs) communication is orchestrated by ligand-receptor interaction generating a microenvironment which results in intraplaque neovascularization, that is closely associated with atherosclerotic plaque instability. Notoginsenoside R1 (R1) exhibits anti-atherosclerotic bioactivity, but its effect on angiogenesis in atherosclerotic plaque remains elusive. The aim of our study is to explore the therapeutic effect of R1 on vulnerable plaque and investigate its potential mechanism against intraplaque neovascularization. The impacts of R1 on plaque stability and intraplaque neovascularization were assessed in ApoE-/- mice induced by high-fat diet. Pericytes-ECs direct or non-direct contact co-cultured with VEGF-A stimulation were used as the in vitro angiogenesis models. Overexpressing Ang1 in pericytes was performed to investigate the underlying mechanism. In vivo experiments, R1 treatment reversed atherosclerotic plaque vulnerability and decreased the presence of neovessels in ApoE-/- mice. Additionally, R1 reduced the expression of Ang1 in pericytes. In vitro experiments demonstrated that R1 suppressed pro-angiogenic behavior of ECs induced by pericytes cultured with VEGF-A. Mechanistic studies revealed that the anti-angiogenic effect of R1 was dependent on the inhibition of Ang1 and Tie2 expression, as the effects were partially reversed after Ang1 overexpressing in pericytes. Our study demonstrated that R1 treatment inhibited intraplaque neovascularization by governing pericyte-EC association via suppressing Ang1-Tie2/PI3K-AKT paracrine signaling pathway. R1 represents a novel therapeutic strategy for atherosclerotic vulnerable plaques in clinical application.
Collapse
Affiliation(s)
- Yuan Li
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Lei Zhang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
- Department of Cardiovascular, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wenqing Yang
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Lin Lin
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jinyuan Pan
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Mengkai Lu
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zhiyuan Zhang
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yunlun Li
- Department of Cardiovascular, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Chao Li
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
6
|
Koutrouli A, Machla F, Arapostathis K, Kokoti M, Bakopoulou A. "Biological responses of two calcium-silicate-based cements on a tissue-engineered 3D organotypic deciduous pulp analogue". Dent Mater 2024; 40:e14-e25. [PMID: 38431482 DOI: 10.1016/j.dental.2024.02.024] [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: 09/25/2023] [Revised: 02/14/2024] [Accepted: 02/18/2024] [Indexed: 03/05/2024]
Abstract
OBJECTIVES The biological responses of MTA and Biodentine™ has been assessed on a three-dimensional, tissue-engineered organotypic deciduous pulp analogue. METHODS Human endothelial (HUVEC) and dental mesenchymal stem cells (SHED) at a ratio of 3:1, were incorporated into a collagen I/fibrin hydrogel; succeeding Biodentine™ and MTA cylindrical specimens were placed in direct contact with the pulp analogue 48 h later. Cell viability/proliferation and morphology were evaluated through live/dead staining, MTT assay and Scanning Electron Microscopy (SEM), and expression of angiogenic, odontogenic markers through real time PCR. RESULTS Viable cells dominated at day 3 after treatment presenting typical morphology, firmly attached within the hydrogel structures, as shown by live/dead staining and SEM images. MTT assay at day 1 presented a significant increase of cell proliferation in Biodentine™ group. Real-time PCR showed significant upregulation of odontogenic markers DSPP, BMP-2 (day 3,6), RUNX2, ALP (day 3) in contact with Biodentine™ compared to MTA and the control, whereas MTA promoted significant upregulation of DSPP, BMP-2, RUNX2, Osterix (day 3) and ALP (day 6) compared to the control. MSX1 presented downregulation in both experimental groups. Expression of angiogenic markers VEGFa and ANGPT-1 at day 3 was significantly upregulated in contact with Biodentine™ and MTA respectively, while the receptors VEGFR1, VEGFR2 and Tie-2, as well as PECAM-1 were downregulated. SIGNIFICANCE Both calcium silicate-based materials are biocompatible and exert positive angiogenic and odontogenic effects, although Biodentine™ during the first days of culture, seems to induce higher cell proliferation and provoke a more profound odontogenic and angiogenic response from SHED.
Collapse
Affiliation(s)
- A Koutrouli
- Department of Paediatric Dentistry, School of Dentistry, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki GR-54124, Greece
| | - F Machla
- Department of Prosthodontics, School of Dentistry, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki GR-54124, Greece
| | - K Arapostathis
- Department of Paediatric Dentistry, School of Dentistry, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki GR-54124, Greece
| | - M Kokoti
- Department of Prosthodontics, School of Dentistry, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki GR-54124, Greece
| | - A Bakopoulou
- Department of Prosthodontics, School of Dentistry, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki GR-54124, Greece.
| |
Collapse
|
7
|
Di T, Feng C, Wang L, Xu J, Du Y, Cheng B, Chen Y, Wu L. Enhancing Vasculogenesis in Dental Pulp Development: DPSCs-ECs Communication via FN1-ITGA5 Signaling. Stem Cell Rev Rep 2024; 20:1060-1077. [PMID: 38418738 PMCID: PMC11087358 DOI: 10.1007/s12015-024-10695-6] [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] [Accepted: 02/12/2024] [Indexed: 03/02/2024]
Abstract
BACKGROUND Dental pulp regeneration therapy is a challenge to achieve early vascularization during treatment. Studying the regulatory mechanisms of vascular formation during human dental pulp development may provide insights for related therapies. In this study, we utilized single-cell sequencing analysis to compare the gene expression of dental pulp stem cells (DPSCs) and vascular endothelial cells (ECs) from developing and mature dental pulps. METHOD Immunohistochemistry, Western blot, and real-time polymerase chain reaction (RT-PCR) were used to detect fibronectin 1 (FN1) expression and molecules, such as PI3K/AKT. Cell proliferation assay, scratch assay, tube formation assay and were used to investigate the effects of DPSCs on the vasculogenetic capability of ECs. Additionally, animal experiments involving mice were conducted. RESULT The results revealed that DPSCs exist around dental pulp vasculature. FN1 expression was significantly higher in DPSCs from young permanent pulps than mature pulps, promoting HUVEC proliferation, migration, and tube formation via ITGA5 and the downstream PI3K/AKT signaling pathway. CONCLUSION Our data indicate that intercellular communication between DPSCs and ECs mediated by FN1-ITGA5 signaling is crucial for vascularizationduring dental pulp development, laying an experimental foundation for future clinical studies.
Collapse
Affiliation(s)
- Tiankai Di
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Pediatric Dentistry, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, People's Republic of China
- Department of Stomatology, No.969 Hospital, Joint Logistics Support Force of the Chinese People's Liberation Army, Hohhot, Inner Mongolia, 010000, People's Republic of China
| | - Chao Feng
- Center for Computational Biology, Institute of Military Cognition and Brain Sciences, Academy of Military Medical Sciences, Beijing, 100850, People's Republic of China
- Department of Clinical Laboratory, No.969 Hospital, Joint Logistics Support Force of the Chinese People's Liberation Army, Hohhot, Inner Mongolia, 010000, People's Republic of China
| | - Lulu Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Pediatric Dentistry, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, People's Republic of China
| | - Jinlong Xu
- Department of Stomatology, No.969 Hospital, Joint Logistics Support Force of the Chinese People's Liberation Army, Hohhot, Inner Mongolia, 010000, People's Republic of China
| | - Yang Du
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Pediatric Dentistry, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, People's Republic of China
| | - Baixiang Cheng
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, Clinical Research Center of Shaanxi Province for Dental and Maxillofacial Diseases, Department of General Dentistry, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710032, People's Republic of China
| | - Yujiang Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Pediatric Dentistry, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, People's Republic of China.
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, Xi'an, Shaanxi, 710032, People's Republic of China.
| | - Lian Wu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Pediatric Dentistry, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, People's Republic of China.
| |
Collapse
|
8
|
Yang X, Wu M, Kong X, Wang Y, Hu C, Zhu D, Kong L, Qiu F, Jiang W. Exosomal miR-3174 induced by hypoxia promotes angiogenesis and metastasis of hepatocellular carcinoma by inhibiting HIPK3. iScience 2024; 27:108955. [PMID: 38322996 PMCID: PMC10845063 DOI: 10.1016/j.isci.2024.108955] [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: 03/05/2023] [Revised: 11/11/2023] [Accepted: 01/15/2024] [Indexed: 02/08/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is a highly malignant tumor with rich blood supply. HCC-derived exosomes containing hereditary substances including microRNAs (miRNAs) were involved in regulating tumor angiogenesis and metastasis. Subsequently, series experiments were performed to evaluate the effect of exosomal miR-3174 on HCC angiogenesis and metastasis. HCC-derived exosomal miR-3174 was ingested by human umbilical vein endothelial cells (HUVECs) in which HIPK3 was targeted and silenced, causing subsequent inhibition of Fas and p53 signaling pathways. Furthermore, exosomal miR-3174 induced permeability and angiogenesis of HUVECs to enhance HCC progression and metastasis. Under hypoxia, upregulated HIF-1α further promoted the transcription of miR-3174. Moreover, HNRNPA1 augmented the package of miR-3174 into exosomes. Clinical data analysis confirmed that HCC patients with high-level miR-3174 were correlated with worse prognosis. Thus, exosomal miR-3174 induced by hypoxia promotes angiogenesis and metastasis of HCC by inhibiting HIPK3/p53 and HIPK3/Fas signaling pathways. Our findings might provide potential targets for anti-tumor therapy.
Collapse
Affiliation(s)
- Xiao Yang
- Department of Hepatobiliary Surgery, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi People’s Hospital, Wuxi Medical Center, Nanjing Medical University, 299 Qingyang Road, Wuxi, Jiangsu 214023, China
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, Jiangsu 210000, China
| | - Mingyu Wu
- Department of Hepatobiliary Surgery, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi People’s Hospital, Wuxi Medical Center, Nanjing Medical University, 299 Qingyang Road, Wuxi, Jiangsu 214023, China
| | - Xiangxu Kong
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, Jiangsu 210000, China
| | - Yun Wang
- Department of Hepatobiliary Surgery, Xuzhou City Central Hospital, The Affiliated Hospital of the Southeast University Medical School (Xu zhou), The Tumor Research Institute of the Southeast University (Xu zhou), Xuzhou clinical college of Xuzhou Medical University, 199 Jiefang South Road, Xuzhou, Jiangsu 221009, China
| | - Chunyang Hu
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, Jiangsu 210000, China
| | - Deming Zhu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Lianbao Kong
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, Jiangsu 210000, China
| | - Fei Qiu
- Department of Anesthesiology, The Second Hospital of Nanjing, Nanjing, Jiangsu 210000, China
| | - Wangjie Jiang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, Jiangsu 210000, China
| |
Collapse
|
9
|
Zhang Y, Lin S, Liu J, Chen Q, Kang J, Zhong J, Hu M, Basabrain MS, Liang Y, Yuan C, Zhang C. Ang1/Tie2/VE-Cadherin Signaling Regulates DPSCs in Vascular Maturation. J Dent Res 2024; 103:101-110. [PMID: 38058134 DOI: 10.1177/00220345231210227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/08/2023] Open
Abstract
Adding dental pulp stem cells (DPSCs) to vascular endothelial cell-formed vessel-like structures can increase the longevity of these vessel networks. DPSCs display pericyte-like cell functions and closely assemble endothelial cells (ECs). However, the mechanisms of DPSC-derived pericyte-like cells in stabilizing the vessel networks are not fully understood. In this study, we investigated the functions of E-DPSCs, which were DPSCs isolated from the direct coculture of human umbilical vein endothelial cells (HUVECs) and DPSCs, and T-DPSCs, which were DPSCs treated by transforming growth factor beta 1 (TGF-β1), in stabilizing blood vessels in vitro and in vivo. A 3-dimensional coculture spheroid sprouting assay was conducted to compare the functions of E-DPSCs and T-DPSCs in vitro. Dental pulp angiogenesis in the severe combined immunodeficiency (SCID) mouse model was used to explore the roles of E-DPSCs and T-DPSCs in vascularization in vivo. The results demonstrated that both E-DPSCs and T-DPSCs possess smooth muscle cell-like cell properties, exhibiting higher expression of the mural cell-specific markers and the suppression of HUVEC sprouting. E-DPSCs and T-DPSCs inhibited HUVEC sprouting by activating TEK tyrosine kinase (Tie2) signaling, upregulating vascular endothelial (VE)-cadherin, and downregulating vascular endothelial growth factor receptor 2 (VEGFR2). In vivo study revealed more perfused and total blood vessels in the HUVEC + E-DPSC group, HUVEC + T-DPSC group, angiopoietin 1 (Ang1) pretreated group, and vascular endothelial protein tyrosine phosphatase (VE-PTP) inhibitor pretreated group, compared to HUVEC + DPSC group. In conclusion, these data indicated that E-DPSCs and T-DPSCs could stabilize the newly formed blood vessels and accelerate their perfusion. The critical regulating pathways are Ang1/Tie2/VE-cadherin and VEGF/VEGFR2 signaling.
Collapse
Affiliation(s)
- Y Zhang
- Restorative Dental Sciences, Endodontics, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - S Lin
- Restorative Dental Sciences, Endodontics, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - J Liu
- Restorative Dental Sciences, Endodontics, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - Q Chen
- Applied Oral Sciences & Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - J Kang
- Restorative Dental Sciences, Endodontics, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - J Zhong
- Restorative Dental Sciences, Endodontics, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - M Hu
- Restorative Dental Sciences, Endodontics, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - M S Basabrain
- Restorative Dental Sciences, Endodontics, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - Y Liang
- Restorative Dental Sciences, Endodontics, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - C Yuan
- School of Stomatology, Xuzhou Medical University, Department of Dental Implant, The Affiliated Stomatological Hospital of Xuzhou Medical University, Xuzhou, China
| | - C Zhang
- Restorative Dental Sciences, Endodontics, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
10
|
Bassett C, Triplett H, Lott K, Howard KM, Kingsley K. Differential Expression of MicroRNA (MiR-27, MiR-145) among Dental Pulp Stem Cells (DPSCs) Following Neurogenic Differentiation Stimuli. Biomedicines 2023; 11:3003. [PMID: 38002003 PMCID: PMC10669296 DOI: 10.3390/biomedicines11113003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 11/03/2023] [Accepted: 11/07/2023] [Indexed: 11/26/2023] Open
Abstract
This study sought to evaluate the expression of previously identified microRNAs known to regulate neuronal differentiation in mesenchymal stem cells (MSCs), including miR-27, miR-125, miR-128, miR-135, miR-140, miR-145, miR-218 and miR-410, among dental pulp stem cells (DPSCs) under conditions demonstrated to induce neuronal differentiation. Using an approved protocol, n = 12 DPSCs were identified from an existing biorepository and treated with basic fibroblast growth factor (bFGF) and epidermal growth factor (EGF), which were previously demonstrated to induce neural differentiation markers including Sox1, Pax6 and NFM among these DPSCs. This study revealed that some microRNAs involved in the neuronal differentiation of MSCs were also differentially expressed among the DPSCs, including miR-27 and miR-145. In addition, this study also revealed that administration of bFGF and EGF was sufficient to modulate miR-27 and miR-145 expression in all of the stimulus-responsive DPSCs but not among all of the non-responsive DPSCs-suggesting that further investigation of the downstream targets of these microRNAs may be needed to fully evaluate and understand these observations.
Collapse
Affiliation(s)
- Charlton Bassett
- School of Medicine, University of Nevada, Las Vegas 1700 West Charleston Boulevard, Las Vegas, NV 89106, USA; (C.B.); (H.T.); (K.L.)
| | - Hunter Triplett
- School of Medicine, University of Nevada, Las Vegas 1700 West Charleston Boulevard, Las Vegas, NV 89106, USA; (C.B.); (H.T.); (K.L.)
| | - Keegan Lott
- School of Medicine, University of Nevada, Las Vegas 1700 West Charleston Boulevard, Las Vegas, NV 89106, USA; (C.B.); (H.T.); (K.L.)
| | - Katherine M. Howard
- School of Dental Medicine, University of Nevada, Las Vegas 1001 Shadow Lane, Las Vegas, NV 89106, USA;
| | - Karl Kingsley
- School of Dental Medicine, University of Nevada, Las Vegas 1001 Shadow Lane, Las Vegas, NV 89106, USA;
| |
Collapse
|
11
|
Qin X, Ruan H, Yuan L, Lin L. Colorectal cancer tumor stem cells mediate bevacizumab resistance through the signal IL-22-STAT3 signaling pathway. 3 Biotech 2023; 13:327. [PMID: 37663749 PMCID: PMC10473997 DOI: 10.1007/s13205-023-03742-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Accepted: 08/09/2023] [Indexed: 09/05/2023] Open
Abstract
Bevacizumab is the standard treatment for colorectal cancer (CRC) in the advanced stage. However, poor diagnosis identified due to the bevacizumab resistance in many CRC patients. Previous studies have found that CRC stem cells (CCSCs) and interleukin 22 (IL-22) are involved in the resistance of bevacizumab, however, the mechanism of remains unclear. In this study, we established the bevacizumab drug-resistant cell line HCT-116-R by concentration gradient method, and the cell viability was detected by CCK-8 assay. The resistance of bevacizumab in CRC cell lines HCT-116-R was identified by characterizing epithelial-mesenchymal transition (EMT). Additionally, HCT-116-R cell lines were isolated from CCSCs and their tumorigenicity was validated in nude mice. We observed that that compared with the matched group, the expression of IL-22, IL-22R, STAT3, and GP130 in drug-resistant cells increased distinctly, with blocked IL-22 cells were successfully constructed by lentiviral interference. The level of proteins in stem cell landmarks (EpCAM, CD133), and stem cell landmarks (Oct4, Sox2) was identified by western blotting. Furthermore, the IL-22 role was evaluated by xenograft model. We found that short hairpin RNA (shRNA) suppression of IL-22 expression can restore the sensitivity of drug-resistant CCSCs to bevacizumab, Moreover, xenograft tumor models show that suppression of IL-22 can increase the anti-tumor influence of bevacizumab. In summary, we demonstrated that CCSCs play a major part in bevacizumab-resistant CRC. Inhibiting the signaling pathway of IL-22/STAT3 can improve the anti-tumor influence on bevacizumab in vitro and in vivo. Thus, IL-22 may represent a new anti-bevacizumab target in CRC.
Collapse
Affiliation(s)
- Xiaoning Qin
- The Third Department of General Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000 Hebei China
| | - Hongxun Ruan
- The Third Department of General Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000 Hebei China
| | - Liqing Yuan
- The Second Department of Gynaecology, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000 Hebei China
| | - Lin Lin
- The Third Department of General Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000 Hebei China
| |
Collapse
|
12
|
Liu B, Zhang C, Zhao H, Gao J, Hu J. Chitosan Hydrogel-Delivered ABE8e Corrects PAX9 Mutant in Dental Pulp Stem Cells. Gels 2023; 9:436. [PMID: 37367107 DOI: 10.3390/gels9060436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 05/19/2023] [Accepted: 05/23/2023] [Indexed: 06/28/2023] Open
Abstract
Hypodontia (dental agenesis) is a genetic disorder, and it has been identified that the mutation C175T in PAX9 could lead to hypodontia. Cas9 nickase (nCas9)-mediated homology-directed repair (HDR) and base editing were used for the correction of this mutated point. This study aimed to investigate the effect of HDR and the base editor ABE8e in editing PAX9 mutant. It was found that the chitosan hydrogel was efficient in delivering naked DNA into dental pulp stem cells (DPSCs). To explore the influence of the C175T mutation in PAX9 on the proliferation of DPSCs, hydrogel was employed to deliver PAX9 mutant vector into DPSCs, finding that the PAX9-containing C175T mutation failed to promote the proliferation of DPSCs. Firstly, DPSCs stably carrying PAX9 mutant were constructed. Either an HDR or ABE8e system was delivered into the above-mentioned stable DPSCs, and then the correction efficiency using Sanger sequencing and Western blotting was determined. Meanwhile, the ABE8e presented significantly higher efficiency in correcting C175T compared with HDR. Furthermore, the corrected PAX9 presented enhanced viability and differentiation capacity for osteogenic and neurogenic lineages; the corrected PAX9 even possessed extremely enhanced transcriptional activation ability. In summary, this study has powerful implications for studies into base editors, chitosan hydrogel, and DPSCs in treating hypodontia.
Collapse
Affiliation(s)
- Bowen Liu
- Outpatient Department of Oral and Maxillofacial Surgery, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Tian Tan Xi Li No. 4, Beijing 100050, China
| | - Chenjiao Zhang
- Department of General, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Tian Tan Xi Li No. 4, Beijing 100050, China
| | - Han Zhao
- Multi-Disciplinary Treatment Center, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Tian Tan Xi Li No. 4, Beijing 100050, China
| | - Jian Gao
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Jingchao Hu
- Department of Periodontics, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Tian Tan Xi Li No. 4, Beijing 100050, China
| |
Collapse
|
13
|
Ruan Q, Tan S, Guo L, Ma D, Wen J. Prevascularization techniques for dental pulp regeneration: potential cell sources, intercellular communication and construction strategies. Front Bioeng Biotechnol 2023; 11:1186030. [PMID: 37274160 PMCID: PMC10232868 DOI: 10.3389/fbioe.2023.1186030] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 05/10/2023] [Indexed: 06/06/2023] Open
Abstract
One of the difficulties of pulp regeneration is the rapid vascularization of transplanted engineered tissue, which is crucial for the initial survival of the graft and subsequent pulp regeneration. At present, prevascularization techniques, as emerging techniques in the field of pulp regeneration, has been proposed to solve this challenge and have broad application prospects. In these techniques, endothelial cells and pericytes are cocultured to induce intercellular communication, and the cell coculture is then introduced into the customized artificial vascular bed or induced to self-assembly to simulate the interaction between cells and extracellular matrix, which would result in construction of a prevascularization system, preformation of a functional capillary network, and rapid reconstruction of a sufficient blood supply in engineered tissue after transplantation. However, prevascularization techniques for pulp regeneration remain in their infancy, and there remain unresolved problems regarding cell sources, intercellular communication and the construction of prevascularization systems. This review focuses on the recent advances in the application of prevascularization techniques for pulp regeneration, considers dental stem cells as a potential cell source of endothelial cells and pericytes, discusses strategies for their directional differentiation, sketches the mechanism of intercellular communication and the potential application of communication mediators, and summarizes construction strategies for prevascularized systems. We also provide novel ideas for the extensive application and follow-up development of prevascularization techniques for dental pulp regeneration.
Collapse
Affiliation(s)
| | | | | | - Dandan Ma
- *Correspondence: Dandan Ma, ; Jun Wen,
| | - Jun Wen
- *Correspondence: Dandan Ma, ; Jun Wen,
| |
Collapse
|
14
|
Differential Effects of Extracellular Matrix Glycoproteins Fibronectin and Laminin-5 on Dental Pulp Stem Cell Phenotypes and Responsiveness. J Funct Biomater 2023; 14:jfb14020091. [PMID: 36826890 PMCID: PMC9963712 DOI: 10.3390/jfb14020091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 01/31/2023] [Accepted: 02/03/2023] [Indexed: 02/11/2023] Open
Abstract
Dental pulp stem cells (DPSCs) are mesenchymal stem cells (MSCs) with the potential to differentiate in a limited number of other tissue types. Some evidence has suggested the modulation of DPSC growth may be mediated, in part, by exogenous extracellular matrix (ECM) glycoproteins, including fibronectin (FN) and laminin-5 (LN5). Although preliminary research suggests that some ECM glycoproteins may work as functional biomaterials to modulate DPSC growth responses, the primary goal of this project is to determine the specific effects of FN and LN5 on DPSC growth and viability. Using an existing DPSC repository, n = 16 DPSC isolates were cultured and 96-well growth assays were performed, which revealed FN, LN5 and the combination of these were sufficient to induce statistically significant changes in growth among five (n = 5) DPSC isolates. In addition, the administration of FN (either alone or in combination) was sufficient to induce the expression of alkaline phosphatase (ALP) and dentin sialophosphoprotein (DSPP), while LN5 induced the expression of ALP only, suggesting differential responsiveness among DPSCs. Moreover, these responses appeared to correlate with the expression of MSC biomarkers NANOG, Oct4 and Sox2. These results add to the growing body of evidence suggesting that functional biomaterials, such as ECM glycoproteins FN and LN5, are sufficient to induce phenotypic and differentiation-specific effects in a specific subset of DPSC isolates. More research will be needed to determine which biomarkers or additional factors are necessary and sufficient to induce the differentiation and development of DPSCs ex vivo and in vitro for biomedical applications.
Collapse
|
15
|
Hu N, Li W, Jiang W, Wen J, Gu S. Creating a Microenvironment to Give Wings to Dental Pulp Regeneration-Bioactive Scaffolds. Pharmaceutics 2023; 15:158. [PMID: 36678787 PMCID: PMC9861529 DOI: 10.3390/pharmaceutics15010158] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/13/2022] [Accepted: 12/23/2022] [Indexed: 01/05/2023] Open
Abstract
Dental pulp and periapical diseases make patients suffer from acute pain and economic loss. Although root canal therapies, as demonstrated through evidence-based medicine, can relieve symptoms and are commonly employed by dentists, it is still difficult to fully restore a dental pulp's nutrition, sensory, and immune-regulation functions. In recent years, researchers have made significant progress in tissue engineering to regenerate dental pulp in a desired microenvironment. With breakthroughs in regenerative medicine and material science, bioactive scaffolds play a pivotal role in creating a suitable microenvironment for cell survival, proliferation, and differentiation, following dental restoration and regeneration. This article focuses on current challenges and novel perspectives about bioactive scaffolds in creating a microenvironment to promote dental pulp regeneration. We hope our readers will gain a deeper understanding and new inspiration of dental pulp regeneration through our summary.
Collapse
Affiliation(s)
- Nan Hu
- Department of Endodontics, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, Shanghai 200011, China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai 200011, China
| | - Weiping Li
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai 200011, China
- Department of Oral and Maxillofacial Head & Neck Oncology, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University, Shanghai 200011, China
| | - Wentao Jiang
- Department of Endodontics, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, Shanghai 200011, China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai 200011, China
| | - Jin Wen
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai 200011, China
- Department of Prosthodontics, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, Shanghai 200011, China
- Shanghai Key Laboratory of Stomatology, Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai 200125, China
| | - Shensheng Gu
- Department of Endodontics, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, Shanghai 200011, China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai 200011, China
| |
Collapse
|
16
|
Ni Y, Chen Y, Jiang X, Pu T, Zhang L, Li S, Hu L, Bai B, Hu T, Yu L, Yang Y. Transplantation of Human Amniotic Mesenchymal Stem Cells Up-Regulates Angiogenic Factor Expression to Attenuate Diabetic Kidney Disease in Rats. Diabetes Metab Syndr Obes 2023; 16:331-343. [PMID: 36785675 PMCID: PMC9921454 DOI: 10.2147/dmso.s371752] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 01/10/2023] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND AND AIMS Diabetic kidney disease (DKD) is a prevalent and intractable microvascular complication of diabetes mellitus (DM), the process of which is closely related to abnormal expression of angiogenesis-regulating factors (ARFs). Stem cell transplantation might be a novel strategy for treating DKD. This study aims to explore the effect of transplantation of human amniotic mesenchymal stem cells (hAMSCs) on renal microangiopathy in a type 1 DKD rat model (T1DRM). METHODS Seventy-two rats were randomly divided into three groups, including normal control group, DKD group, and hAMSCs transplantation group. T1DRM was established using a rat tail vein injection of streptozotocin (STZ) (55 mg/kg). hAMSCs were obtained from placental amniotic membranes during cesarean delivery and transplanted at 3 and 4 weeks through penile veins. At 6, 8, and 12 weeks following transplantation, blood glucose levels, renal function, pathological kidney alterations, and the expressions of ARFs' mRNA and protein were analyzed. RESULTS In T1DRM, transplanted hAMSCs that were homed at the injured site of kidneys increased ARFs' expression and decreased blood glucose levels. Compared to the DKD group, the levels of 24-h urinary protein, serum creatinine, urea, and kidney injury molecule-1 (KIM-1) were reduced in hAMSCs transplantation group. In terms of renal pathology such as the degree of basement membrane thickening, hAMSCs transplantation was also less severe than the DKD group, thereby alleviating kidney injury. CONCLUSION hAMSCs transplantation might ameliorate STZ-induced chronic kidney injury through increasing ARFs' expression in kidneys and lowering blood glucose levels.
Collapse
Affiliation(s)
- Yu Ni
- Department of Nephrology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, People’s Republic of China
| | - Yuqin Chen
- Key Laboratory of Cell Engineering of Guizhou Province, Zunyi City, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, People’s Republic of China
| | - Xuheng Jiang
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, People’s Republic of China
| | - Tao Pu
- Department of Nephrology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, People’s Republic of China
| | - Ling Zhang
- Zhuhai Campus of Zunyi Medical University, Zhuhai, 519041, People’s Republic of China
| | - Shaobin Li
- Key Laboratory of Cell Engineering of Guizhou Province, Zunyi City, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, People’s Republic of China
| | - Linhong Hu
- Department of Nephrology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, People’s Republic of China
| | - Bing Bai
- Department of Nephrology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, People’s Republic of China
| | - Tingting Hu
- Department of Nephrology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, People’s Republic of China
| | - Limei Yu
- Key Laboratory of Cell Engineering of Guizhou Province, Zunyi City, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, People’s Republic of China
- Correspondence: Limei Yu, Department of Nephrology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, People’s Republic of China, Email
| | - Yibin Yang
- Department of Nephrology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, People’s Republic of China
- Key Laboratory of Cell Engineering of Guizhou Province, Zunyi City, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, People’s Republic of China
- Yibin Yang, Key Laboratory of Cell Engineering of Guizhou Province, Zunyi City, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, People’s Republic of China, Email
| |
Collapse
|
17
|
Kuang G, Shu Z, Zhu C, Li H, Zhang C. The promoting effect of modified Dioscorea pills on vascular remodeling in chronic cerebral hypoperfusion via the Ang/Tie signaling pathway. Transl Neurosci 2023; 14:20220302. [PMID: 37635842 PMCID: PMC10448306 DOI: 10.1515/tnsci-2022-0302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 07/14/2023] [Accepted: 07/26/2023] [Indexed: 08/29/2023] Open
Abstract
Objective The objective of this study was to investigate the effect of modified Dioscorea pills (MDP) on microcirculatory remodeling in the hippocampus of rats with chronic cerebral hypoperfusion (CCH) through the angiopoietin (Ang)/tyrosine kinase receptor tyrosine kinase with immunoglobulin-like and EGF-like domains (Ang receptor) 2 (Tie-2) signaling pathways, which may underlie the cognitive improvement observed in CCH rats. Methods Forty male Sprague-Dawley rats raised under specific pathogen-free conditions were randomly divided into three groups: control group (10 rats), model group (15 rats), and MDP group (15 rats). The rats in the model group and MDP group underwent bilateral common carotid artery occlusion using the 2-vessel occlusion (2-VO) method to induce CCH. Rats in the control group underwent the same surgical procedures as those in the model group, except for ligation and occlusion of the carotid arteries. After 1 week of 2-VO, rats in the MDP group were administered MDP condensed decoction intragastrically at a dose of 1 ml/100 g body weight (prepared by the Preparation Room of Hubei Provincial Hospital of Traditional Chinese Medicine) for 45 days, while rats in the other two groups received normal saline intragastrically with the same dose and duration as the MDP group. After the intervention, all rats were euthanized, and brain perfusion was performed to obtain the hippocampal tissue for analysis. Immunohistochemical staining for CD43 was performed to assess microvessel density (MVD); western blot and the reverse transcription-polymerase chain reaction (RT-PCR) were used to analyze the expression of proteins and genes in angiopoietin-1 (Ang-1), angiopoietin-2 (Ang-2), Tie-2, and vascular endothelial growth factor (VEGF) proteins and genes in the hippocampal tissue and compute the Ang-1/Ang-2 ratio. Results MDP treatment reduced neuronal loss and promoted restoration of the damaged hippocampal structure in CCH rats. The model group showed significantly higher MVD (14.93 ± 1.92) compared to the control group (5.78 ± 1.65) (P < 0.01), whereas MDP treatment further increased MVD (21.19 ± 2.62). Western blot and RT-PCR analysis revealed that CCH significantly increased the expression of Ang-1, Ang-2, Tie-2, and VEGF proteins and genes, while MDP treatment further significantly upregulated the expression of these proteins and genes. In addition, MDP significantly elevated the gene and protein expression of the Ang-1/Ang-2 ratio compared to the control group (P = 0.041, P = 0.029). Conclusion CCH induces microvascular neogenesis in the hippocampus, and MDP promotes angiogenesis and microcirculation remodeling in CCH rats via the Ang/Tie signaling pathway, which may be an important mechanism for its restorative effects on hippocampal perfusion and improvement of cognitive function in CCH rats.
Collapse
Affiliation(s)
- Guiying Kuang
- Neurological Department, Wuhan Red Cross Hospital, Wuhan, Hubei Province, 436015, China
| | - Zhigang Shu
- Neurological Department, Ezhou Central Hospital, Ezhou, Hubei Province, 436000, China
| | - Chunli Zhu
- Neurological Department, Wuhan Red Cross Hospital, Wuhan, Hubei Province, 436015, China
| | - Hongbing Li
- Emergency Department, The First People’s Hospital of Guiyang, Guiyang, Guizhou Province, 550002, China
| | - Cheng Zhang
- Emergency Department, The First People’s Hospital of Guiyang, Guiyang, Guizhou Province, 550002, China
| |
Collapse
|
18
|
Khanna A, Oropeza BP, Huang NF. Engineering Spatiotemporal Control in Vascularized Tissues. BIOENGINEERING (BASEL, SWITZERLAND) 2022; 9:bioengineering9100555. [PMID: 36290523 PMCID: PMC9598830 DOI: 10.3390/bioengineering9100555] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 10/10/2022] [Accepted: 10/11/2022] [Indexed: 11/06/2022]
Abstract
A major challenge in engineering scalable three-dimensional tissues is the generation of a functional and developed microvascular network for adequate perfusion of oxygen and growth factors. Current biological approaches to creating vascularized tissues include the use of vascular cells, soluble factors, and instructive biomaterials. Angiogenesis and the subsequent generation of a functional vascular bed within engineered tissues has gained attention and is actively being studied through combinations of physical and chemical signals, specifically through the presentation of topographical growth factor signals. The spatiotemporal control of angiogenic signals can generate vascular networks in large and dense engineered tissues. This review highlights the developments and studies in the spatiotemporal control of these biological approaches through the coordinated orchestration of angiogenic factors, differentiation of vascular cells, and microfabrication of complex vascular networks. Fabrication strategies to achieve spatiotemporal control of vascularization involves the incorporation or encapsulation of growth factors, topographical engineering approaches, and 3D bioprinting techniques. In this article, we highlight the vascularization of engineered tissues, with a focus on vascularized cardiac patches that are clinically scalable for myocardial repair. Finally, we discuss the present challenges for successful clinical translation of engineered tissues and biomaterials.
Collapse
Affiliation(s)
| | - Beu P. Oropeza
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA 94305, USA
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA 94305, USA
- Center for Tissue Regeneration, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304, USA
| | - Ngan F. Huang
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA 94305, USA
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA 94305, USA
- Center for Tissue Regeneration, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304, USA
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
- Correspondence:
| |
Collapse
|
19
|
Dieterle MP, Gross T, Steinberg T, Tomakidi P, Becker K, Vach K, Kremer K, Proksch S. Characterization of a Stemness-Optimized Purification Method for Human Dental-Pulp Stem Cells: An Approach to Standardization. Cells 2022; 11:cells11203204. [PMID: 36291072 PMCID: PMC9600643 DOI: 10.3390/cells11203204] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/06/2022] [Accepted: 10/09/2022] [Indexed: 11/16/2022] Open
Abstract
Human dental pulp stem cells (hDPSCs) are promising for oral/craniofacial regeneration, but their purification and characterization is not yet standardized. hDPSCs from three donors were purified by magnetic activated cell sorting (MACS)-assisted STRO-1-positive cell enrichment (+), colony derivation (c), or a combination of both (c/+). Immunophenotype, clonogenicity, stemness marker expression, senescence, and proliferation were analyzed. Multilineage differentiation was assessed by qPCR, immunohistochemistry, and extracellular matrix mineralization. To confirm the credibility of the results, repeated measures analysis and post hoc p-value adjustment were applied. All hDPSC fractions expressed STRO-1 and were similar for several surface markers, while their clonogenicity and expression of CD10/44/105/146, and 166 varied with the purification method. (+) cells proliferated significantly faster than (c/+), while (c) showed the highest increase in metabolic activity. Colony formation was most efficient in (+) cells, which also exhibited the lowest cellular senescence. All hDPSCs produced mineralized extracellular matrix. Regarding osteogenic induction, (c/+) revealed a significant increase in mRNA expression of COL5A1 and COL6A1, while osteogenic marker genes were detected at varying levels. (c/+) were the only population missing BDNF gene transcription increase during neurogenic induction. All hDPSCs were able to differentiate into chondrocytes. In summary, the three hDPSCs populations showed differences in phenotype, stemness, proliferation, and differentiation capacity. The data suggest that STRO-1-positive cell enrichment is the optimal choice for hDPSCs purification to maintain hDPSCs stemness. Furthermore, an (immuno) phenotypic characterization is the minimum requirement for quality control in hDPSCs studies.
Collapse
Affiliation(s)
- Martin Philipp Dieterle
- Division of Oral Biotechnology, Center for Dental Medicine, Medical Center—University of Freiburg, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Hugstetter Str. 55, 79106 Freiburg, Germany
| | - Tara Gross
- Department of Operative Dentistry and Periodontology, Centre for Dental Medicine Medical Center—University of Freiburg, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, 79106 Freiburg, Germany
- G.E.R.N. Center for Tissue Replacement, Regeneration & Neogenesis, Medical Center—University of Freiburg, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, 79108 Freiburg, Germany
| | - Thorsten Steinberg
- Division of Oral Biotechnology, Center for Dental Medicine, Medical Center—University of Freiburg, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Hugstetter Str. 55, 79106 Freiburg, Germany
- Correspondence: ; Tel.: +49-761-27047460
| | - Pascal Tomakidi
- Division of Oral Biotechnology, Center for Dental Medicine, Medical Center—University of Freiburg, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Hugstetter Str. 55, 79106 Freiburg, Germany
| | - Kathrin Becker
- Department of Operative Dentistry and Periodontology, Centre for Dental Medicine Medical Center—University of Freiburg, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, 79106 Freiburg, Germany
| | - Kirstin Vach
- Institute of Medical Biometry and Statistics, Medical Center—University of Freiburg, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, 79104 Freiburg, Germany
| | - Katrin Kremer
- Department of Oral and Maxillofacial Surgery, Center for Dental Medicine, Medical Center—University of Freiburg, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, 79106 Freiburg, Germany
| | - Susanne Proksch
- Department of Operative Dentistry and Periodontology, Centre for Dental Medicine Medical Center—University of Freiburg, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, 79106 Freiburg, Germany
- G.E.R.N. Center for Tissue Replacement, Regeneration & Neogenesis, Medical Center—University of Freiburg, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, 79108 Freiburg, Germany
| |
Collapse
|
20
|
Kim CK, Hwang JY, Hong TH, Lee DM, Lee K, Nam H, Joo KM. Combination stem cell therapy using dental pulp stem cells and human umbilical vein endothelial cells for critical hindlimb ischemia. BMB Rep 2022. [PMID: 35168701 PMCID: PMC9340082 DOI: 10.5483/bmbrep.2022.55.7.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Narrowing of arteries supplying blood to the limbs provokes critical hindlimb ischemia (CLI). Although CLI results in irreversible sequelae, such as amputation, few therapeutic options induce the formation of new functional blood vessels. Based on the proangiogenic potentials of stem cells, in this study, it was examined whether a combination of dental pulp stem cells (DPSCs) and human umbilical vein endothelial cells (HUVECs) could result in enhanced therapeutic effects of stem cells for CLI compared with those of DPSCs or HUVECs alone. The DPSCs+ HUVECs combination therapy resulted in significantly higher blood flow and lower ischemia damage than DPSCs or HUVECs alone. The improved therapeutic effects in the DPSCs+ HUVECs group were accompanied by a significantly higher number of microvessels in the ischemic tissue than in the other groups. In vitro proliferation and tube formation assay showed that VEGF in the conditioned media of DPSCs induced proliferation and vessel-like tube formation of HUVECs. Altogether, our results demonstrated that the combination of DPSCs and HUVECs had significantly better therapeutic effects on CLI via VEGF-mediated crosstalk. This combinational strategy could be used to develop novel clinical protocols for CLI proangiogenic regenerative treatments.
Collapse
Affiliation(s)
- Chung Kwon Kim
- Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Korea
- Medical Innovation Technology Inc. (MEDINNO Inc.), Seoul 08517, Korea
| | - Ji-Yoon Hwang
- Medical Innovation Technology Inc. (MEDINNO Inc.), Seoul 08517, Korea
- Department of Anatomy and Cell Biology, Sungkyunkwan University School of Medicine, Suwon 16149, Korea
| | - Tae Hee Hong
- Medical Innovation Technology Inc. (MEDINNO Inc.), Seoul 08517, Korea
| | - Du Man Lee
- Medical Innovation Technology Inc. (MEDINNO Inc.), Seoul 08517, Korea
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul 06351, Korea
| | - Kyunghoon Lee
- Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Korea
- Department of Anatomy and Cell Biology, Sungkyunkwan University School of Medicine, Suwon 16149, Korea
| | - Hyun Nam
- Department of Anatomy and Cell Biology, Sungkyunkwan University School of Medicine, Suwon 16149, Korea
- Stem Cell and Regenerative Medicine Institute, Research Institute for Future Medicine, Samsung Medical Center, Seoul 06351, Korea
- Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon 16149, Korea
| | - Kyeung Min Joo
- Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Korea
- Medical Innovation Technology Inc. (MEDINNO Inc.), Seoul 08517, Korea
- Department of Anatomy and Cell Biology, Sungkyunkwan University School of Medicine, Suwon 16149, Korea
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul 06351, Korea
- Stem Cell and Regenerative Medicine Institute, Research Institute for Future Medicine, Samsung Medical Center, Seoul 06351, Korea
| |
Collapse
|
21
|
Luo L, Xing Z, Liao X, Li Y, Luo Y, Ai Y, He Y, Ye Q. Dental pulp stem cells-based therapy for the oviduct injury via immunomodulation and angiogenesis in vivo. Cell Prolif 2022; 55:e13293. [PMID: 35822247 PMCID: PMC9528759 DOI: 10.1111/cpr.13293] [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: 05/17/2022] [Revised: 06/03/2022] [Accepted: 06/07/2022] [Indexed: 01/07/2023] Open
Abstract
OBJECTIVES As a result of the current limitation of therapeutic strategies, the repair and regeneration of oviduct injuries required an alternative treatment. We present a novel approach to treat oviduct injuries through a dental pulp stem cells (DPSCs)-based therapy. MATERIALS AND METHODS In vitro and in vivo models have been established. Immunofluorescence staining, flow cytometry and enzyme-linked immunosorbent assay (ELISA) analysis were used to investigate the features and angiogenic properties of DPSCs, as well as their impact on macrophages, in vitro. For the in vivo experiment with female SD rat model, immunohistochemical staining and ELISA analysis were used to assess the effects of DPSCs on the repair and regeneration of damaged oviducts. RESULTS The present data showed that intraperitoneal injection of DPSCs reduced the expression of IL-6 and TNF-α to inhibit the immunoreaction in injured sites, as well as increased the expression of VEGF to promote the in situ formation of vessel-like structures, thus the repair and recovery process could be initiated. CONCLUSIONS We concluded that DPSCs-based therapy could be a novel potential technique for restoring the structure and function of damaged oviduct by enhancing immuno-regulated effect and promoting angiogenic property.
Collapse
Affiliation(s)
- Lihua Luo
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Zhenjie Xing
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Xiangyan Liao
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Yejian Li
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Yu Luo
- Center of Regenerative Medicine, Renmin Hospital of Wuhan University, Wuhan, China.,Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
| | - Yilong Ai
- Foshan Stomatological Hospital, School of Medicine, Foshan University, Foshan, China
| | - Yan He
- Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
| | - Qingsong Ye
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China.,Center of Regenerative Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
22
|
Kim CK, Hwang JY, Hong TH, Lee DM, Lee K, Nam H, Joo KM. Combination stem cell therapy using dental pulp stem cells and human umbilical vein endothelial cells for critical hindlimb ischemia. BMB Rep 2022; 55:336-341. [PMID: 35168701 PMCID: PMC9340082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 01/25/2022] [Accepted: 02/11/2022] [Indexed: 03/08/2024] Open
Abstract
Narrowing of arteries supplying blood to the limbs provokes critical hindlimb ischemia (CLI). Although CLI results in irreversible sequelae, such as amputation, few therapeutic options induce the formation of new functional blood vessels. Based on the proangiogenic potentials of stem cells, in this study, it was examined whether a combination of dental pulp stem cells (DPSCs) and human umbilical vein endothelial cells (HUVECs) could result in enhanced therapeutic effects of stem cells for CLI compared with those of DPSCs or HUVECs alone. The DPSCs+ HUVECs combination therapy resulted in significantly higher blood flow and lower ischemia damage than DPSCs or HUVECs alone. The improved therapeutic effects in the DPSCs+ HUVECs group were accompanied by a significantly higher number of microvessels in the ischemic tissue than in the other groups. In vitro proliferation and tube formation assay showed that VEGF in the conditioned media of DPSCs induced proliferation and vessel-like tube formation of HUVECs. Altogether, our results demonstrated that the combination of DPSCs and HUVECs had significantly better therapeutic effects on CLI via VEGF-mediated crosstalk. This combinational strategy could be used to develop novel clinical protocols for CLI proangiogenic regenerative treatments. [BMB Reports 2022; 55(7): 336-341].
Collapse
Affiliation(s)
- Chung Kwon Kim
- Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Korea
- Medical Innovation Technology Inc. (MEDINNO Inc.), Seoul 08517, Korea
| | - Ji-Yoon Hwang
- Medical Innovation Technology Inc. (MEDINNO Inc.), Seoul 08517, Korea
- Department of Anatomy and Cell Biology, Sungkyunkwan University School of Medicine, Suwon 16149, Korea
| | - Tae Hee Hong
- Medical Innovation Technology Inc. (MEDINNO Inc.), Seoul 08517, Korea
| | - Du Man Lee
- Medical Innovation Technology Inc. (MEDINNO Inc.), Seoul 08517, Korea
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul 06351, Korea
| | - Kyunghoon Lee
- Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Korea
- Department of Anatomy and Cell Biology, Sungkyunkwan University School of Medicine, Suwon 16149, Korea
| | - Hyun Nam
- Department of Anatomy and Cell Biology, Sungkyunkwan University School of Medicine, Suwon 16149, Korea
- Stem Cell and Regenerative Medicine Institute, Research Institute for Future Medicine, Samsung Medical Center, Seoul 06351, Korea
- Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon 16149, Korea
| | - Kyeung Min Joo
- Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Korea
- Medical Innovation Technology Inc. (MEDINNO Inc.), Seoul 08517, Korea
- Department of Anatomy and Cell Biology, Sungkyunkwan University School of Medicine, Suwon 16149, Korea
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul 06351, Korea
- Stem Cell and Regenerative Medicine Institute, Research Institute for Future Medicine, Samsung Medical Center, Seoul 06351, Korea
- Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon 16149, Korea
| |
Collapse
|
23
|
Wintruba KL, Hill JC, Richards TD, Lee YC, Kaczorowski DJ, Sultan I, Badylak SF, Billaud M, Gleason TG, Phillippi JA. Adventitia-derived extracellular matrix hydrogel enhances contractility of human vasa vasorum-derived pericytes via α 2 β 1 integrin and TGFβ receptor. J Biomed Mater Res A 2022; 110:1912-1920. [PMID: 35770946 DOI: 10.1002/jbm.a.37422] [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: 04/25/2022] [Revised: 06/07/2022] [Accepted: 06/15/2022] [Indexed: 11/11/2022]
Abstract
Pericytes are essential components of small blood vessels and are found in human aortic vasa vasorum. Prior work uncovered lower vasa vasorum density and decreased levels of pro-angiogenic growth factors in adventitial specimens of human ascending thoracic aortic aneurysm. We hypothesized that adventitial extracellular matrix (ECM) from normal aorta promotes pericyte function by increasing pericyte contractile function through mechanisms deficient in ECM derived from aneurysmal aortic adventitia. ECM biomaterials were prepared as lyophilized particulates from decellularized adventitial specimens of human and porcine aorta. Immortalized human aortic adventitia-derived pericytes were cultured within Type I collagen gels in the presence or absence of human or porcine adventitial ECMs. Cell contractility index was quantified by measuring the gel area immediately following gelation and after 48 h of culture. Normal human and porcine adventitial ECM increased contractility of pericytes when compared with pericytes cultured in absence of adventitial ECM. In contrast, aneurysm-derived human adventitial ECM failed to promote pericyte contractility. Pharmacological inhibition of TGFβR1 and antibody blockade of α2 β1 integrin independently decreased porcine adventitial ECM-induced pericyte contractility. By increasing pericyte contractility, adventitial ECM may improve microvascular function and thus represents a candidate biomaterial for less invasive and preventative treatment of human ascending aortic disease.
Collapse
Affiliation(s)
- Kaitlyn L Wintruba
- Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Jennifer C Hill
- Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Tara D Richards
- Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Yoojin C Lee
- Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - David J Kaczorowski
- Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Heart and Vascular Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA.,McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Ibrahim Sultan
- Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Heart and Vascular Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Stephen F Badylak
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Marie Billaud
- Department of Surgery, Division of Thoracic and Cardiac Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Thomas G Gleason
- Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Julie A Phillippi
- Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Heart and Vascular Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA.,Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
24
|
Mu R, Chen B, Bi B, Yu H, Liu J, Li J, He M, Rong L, Liu B, Liu K, Zhu L, Shi X, Shuai Y, Jin L. LIM Mineralization Protein-1 Enhances the Committed Differentiation of Dental Pulp Stem Cells through the ERK1/2 and p38 MAPK Pathways and BMP Signaling. Int J Med Sci 2022; 19:1307-1319. [PMID: 35928717 PMCID: PMC9346378 DOI: 10.7150/ijms.70411] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 06/24/2022] [Indexed: 11/23/2022] Open
Abstract
Tissue regeneration is the preferred treatment for dentin and bone tissue defects. Dental pulp stem cells (DPSCs) have been extensively studied for their use in tissue regeneration, including the regeneration of dentin and bone tissue. LIM mineralization protein-1 (LMP-1) is an intracellular non-secretory protein that plays a positive regulatory role in the mineralization process. In this study, an LMP-1-induced DPSCs model was used to explore the effect of LMP-1 on the proliferation and odonto/osteogenic differentiation of DPSCs, as well as the underlying mechanisms. As indicated by the cell counting kit-8 assay, the results showed that LMP-1 did not affect the proliferation of DPSCs. Overexpression of LMP-1 significantly promoted the committed differentiation of DPSCs and vice versa, as shown by alkaline phosphatase activity assay, alizarin red staining, western blot assay, quantitative real-time polymerase chain reaction assay, and in vivo mineralized tissue formation assay. Furthermore, inhibiting the activation of the extracellular signal-regulated kinase 1/2 (ERK1/2), p38 mitogen-activated protein kinase (MAPK), and c-Jun N-terminal kinase (JNK) pathways using specific pathway inhibitors showed that the ERK1/2 and p38 MAPK pathways attenuated the differentiation of DPSCs. Besides, the expression of BMP signaling pathway components were also determined, which suggested that LMP-1 could activate BMP-2/Smad1/5 signaling pathway. Our results not only indicated the underlying mechanism of LMP-1 treated DPSCs but also provided valuable insight into therapeutic strategies in regenerative medicine.
Collapse
Affiliation(s)
- Rui Mu
- Department of Stomatology, Jinling Hospital, Medical School of Nanjing University, School of Stomatology of Southern Medical University, Clinical Medical School of Nanjing Medical University, Nanjing 210002, China.,Stomatology Center, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Guangdong province, Shenzhen 518036, China
| | - Bo Chen
- Department of Stomatology, Jinling Hospital, Medical School of Nanjing University, School of Stomatology of Southern Medical University, Clinical Medical School of Nanjing Medical University, Nanjing 210002, China
| | - Bo Bi
- Department of Stomatology, Jinling Hospital, Medical School of Nanjing University, School of Stomatology of Southern Medical University, Clinical Medical School of Nanjing Medical University, Nanjing 210002, China
| | - Hongchuan Yu
- Department of Stomatology, Jinling Hospital, Medical School of Nanjing University, School of Stomatology of Southern Medical University, Clinical Medical School of Nanjing Medical University, Nanjing 210002, China
| | - Juan Liu
- Department of Stomatology, Jinling Hospital, Medical School of Nanjing University, School of Stomatology of Southern Medical University, Clinical Medical School of Nanjing Medical University, Nanjing 210002, China
| | - Junxia Li
- Department of Stomatology, Jinling Hospital, Medical School of Nanjing University, School of Stomatology of Southern Medical University, Clinical Medical School of Nanjing Medical University, Nanjing 210002, China
| | - Maodian He
- Department of Stomatology, Jinling Hospital, Medical School of Nanjing University, School of Stomatology of Southern Medical University, Clinical Medical School of Nanjing Medical University, Nanjing 210002, China
| | - Liang Rong
- Department of Stomatology, Jinling Hospital, Medical School of Nanjing University, School of Stomatology of Southern Medical University, Clinical Medical School of Nanjing Medical University, Nanjing 210002, China
| | - Bingyao Liu
- Department of Stomatology, Jinling Hospital, Medical School of Nanjing University, School of Stomatology of Southern Medical University, Clinical Medical School of Nanjing Medical University, Nanjing 210002, China
| | - Ke Liu
- Department of Stomatology, Jinling Hospital, Medical School of Nanjing University, School of Stomatology of Southern Medical University, Clinical Medical School of Nanjing Medical University, Nanjing 210002, China
| | - Lei Zhu
- Department of Stomatology, Jinling Hospital, Medical School of Nanjing University, School of Stomatology of Southern Medical University, Clinical Medical School of Nanjing Medical University, Nanjing 210002, China
| | - Xiaolei Shi
- Department of Stomatology, Jinling Hospital, Medical School of Nanjing University, School of Stomatology of Southern Medical University, Clinical Medical School of Nanjing Medical University, Nanjing 210002, China
| | - Yi Shuai
- Department of Stomatology, Jinling Hospital, Medical School of Nanjing University, School of Stomatology of Southern Medical University, Clinical Medical School of Nanjing Medical University, Nanjing 210002, China
| | - Lei Jin
- Department of Stomatology, Jinling Hospital, Medical School of Nanjing University, School of Stomatology of Southern Medical University, Clinical Medical School of Nanjing Medical University, Nanjing 210002, China
| |
Collapse
|
25
|
Vakhrushev IV, Nezhurina EK, Karalkin PA, Tsvetkova AV, Sergeeva NS, Majouga AG, Yarygin KN. Heterotypic Multicellular Spheroids as Experimental and Preclinical Models of Sprouting Angiogenesis. BIOLOGY 2021; 11:18. [PMID: 35053016 PMCID: PMC8772844 DOI: 10.3390/biology11010018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 12/18/2021] [Accepted: 12/20/2021] [Indexed: 12/12/2022]
Abstract
Sprouting angiogenesis is the common response of live tissues to physiological and pathological angiogenic stimuli. Its accurate evaluation is of utmost importance for basic research and practical medicine and pharmacology and requires adequate experimental models. A variety of assays for angiogenesis were developed, none of them perfect. In vitro approaches are generally less physiologically relevant due to the omission of essential components regulating the process. However, only in vitro models can be entirely non-xenogeneic. The limitations of the in vitro angiogenesis assays can be partially overcome using 3D models mimicking tissue O2 and nutrient gradients, the influence of the extracellular matrix (ECM), and enabling cell-cell interactions. Here we present a review of the existing models of sprouting angiogenesis that are based on the use of endothelial cells (ECs) co-cultured with perivascular or other stromal cells. This approach provides an excellent in vitro platform for further decoding of the cellular and molecular mechanisms of sprouting angiogenesis under conditions close to the in vivo conditions, as well as for preclinical drug testing and preclinical research in tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Igor V. Vakhrushev
- Laboratory of Cell Biology, Institute of Biomedical Chemistry, 119121 Moscow, Russia;
| | - Elizaveta K. Nezhurina
- P.A. Hertsen Moscow Oncology Research Center, National Medical Research Radiological Center, 125284 Moscow, Russia;
| | - Pavel A. Karalkin
- Institute for Cluster Oncology, Sechenov University, 119435 Moscow, Russia;
| | | | - Nataliya S. Sergeeva
- Department of Biology, Pirogov Russian National Research Medical University, 117997 Moscow, Russia;
| | - Alexander G. Majouga
- Faculty of Chemical and Pharmaceutical Technologies and Biomedical Products, D. Mendeleev University of Chemical Technology of Russia, 125047 Moscow, Russia;
| | - Konstantin N. Yarygin
- Laboratory of Cell Biology, Institute of Biomedical Chemistry, 119121 Moscow, Russia;
| |
Collapse
|
26
|
Lia A, Annese T, Fornaro M, Giannini M, D'Abbicco D, Errede M, Lorusso L, Amati A, Tampoia M, Trojano M, Virgintino D, Ribatti D, Serlenga L, Iannone F, Girolamo F. Perivascular and endomysial macrophages expressing VEGF and CXCL12 promote angiogenesis in anti-HMGCR immune-mediated necrotizing myopathy. Rheumatology (Oxford) 2021; 61:3448-3460. [PMID: 34864921 DOI: 10.1093/rheumatology/keab900] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 11/29/2021] [Indexed: 11/12/2022] Open
Abstract
OBJECTIVES To study the phenotype of macrophage infiltrates and their role in angiogenesis in different Idiopathic Inflammatory Myopathies (IIMs). METHODS The density and distribution of the subpopulations of macrophages subsets (M1, inducible nitric oxide+, CD11c+; M2, arginase-1+), endomysial capillaries (CD31+, FLK1+), degenerating (C5b-9+), and regenerating (NCAM+) myofibers, were investigated by immunohistochemistry in human muscle samples of diagnostic biopsies from a large cohort of untreated patients (n: 81) suffering from anti-3-hydroxy-3-methylglutaryl coenzyme A reductase (anti-HMGCR)+ Immune Mediated Necrotizing Myopathy (IMNM), anti-signal recognition particle (anti-SRP)+ IMNM, seronegative IMNM, Dermatomyositis, Polymyositis, Polymyositis with mitochondrial pathology, sporadic Inclusion Body Myositis, Scleromyositis, and anti-Synthetase Syndrome. The samples were compared with mitochondrial myopathy and control muscle samples. RESULTS Compared with the other IIMs and controls, endomysial capillary density (CD) was higher in anti-HMGCR+ IMNM, where M1 and M2 macrophages, detected by confocal microscopy, infiltrated perivascular endomysium and expressed angiogenic molecules such as VEGF-A and CXCL12. These angiogenic macrophages were preferentially associated with CD31+ FLK1+ microvessels in anti-HMGCR+ IMNM. The VEGF-A+ M2 macrophage density was significantly correlated with CD (rS: 0.98; p: 0.0004). Western blot analyses revealed increased expression levels of VEGF-A, FLK1, HIF-1α, and CXCL12 in anti-HMGCR+ IMNM. CD and expression levels of these angiogenic molecules were not increased in anti-SRP+ and seronegative IMNM, offering additional, useful information for differential diagnosis among these IIM subtypes. CONCLUSION Our findings suggest that in IIMs, infiltrating macrophages and microvascular cells interactions play a pivotal role in coordinating myogenesis and angiogenesis. This reciprocal crosstalk seems to distinguish anti-HMGCR associated IMNM.
Collapse
Affiliation(s)
- Anna Lia
- Unit of Neurophysiopathology, Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari, Italy
| | - Tiziana Annese
- Unit of Human Anatomy and Histology, Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari, Italy
| | - Marco Fornaro
- Unit of Rheumatology, Department of Emergency and Organ Transplantation, University of Bari, Italy
| | - Margherita Giannini
- Unit of Rheumatology, Department of Emergency and Organ Transplantation, University of Bari, Italy.,Service de Physiologie, Unité d'Explorations Fonctionnelles Musculaires, Hôpitaux Universitaires de Strasbourg, France
| | - Dario D'Abbicco
- Institute of General Surgery "G. Marinaccio", Department of Emergency and Organ Transplantation, University of Bari
| | - Mariella Errede
- Unit of Human Anatomy and Histology, Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari, Italy
| | - Loredana Lorusso
- Unit of Human Anatomy and Histology, Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari, Italy
| | - Angela Amati
- Unit of Neurophysiopathology, Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari, Italy
| | - Marilina Tampoia
- Unit of Clinical Pathology, Ospedale SS., Annunziata, Taranto, Italy
| | - Maria Trojano
- Unit of Neurophysiopathology, Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari, Italy
| | - Daniela Virgintino
- Unit of Human Anatomy and Histology, Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari, Italy
| | - Domenico Ribatti
- Unit of Human Anatomy and Histology, Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari, Italy
| | - Luigi Serlenga
- Unit of Neurophysiopathology, Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari, Italy
| | - Florenzo Iannone
- Unit of Rheumatology, Department of Emergency and Organ Transplantation, University of Bari, Italy
| | - Francesco Girolamo
- Unit of Human Anatomy and Histology, Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari, Italy
| |
Collapse
|
27
|
Mantesso A, Zhang Z, Warner KA, Herzog AE, Pulianmackal AJ, Nör JE. Pulpbow: A Method to Study the Vasculogenic Potential of Mesenchymal Stem Cells from the Dental Pulp. Cells 2021; 10:2804. [PMID: 34831027 PMCID: PMC8616523 DOI: 10.3390/cells10112804] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 10/14/2021] [Accepted: 10/15/2021] [Indexed: 01/06/2023] Open
Abstract
Understanding how Mesenchymal Stem Cells (MSCs) form blood vessels is critical for creating mechanism-based approaches for the therapeutic use of these cells. In addition, understanding the determinants and factors involved in lineage hierarchy is fundamental to creating accurate and reliable techniques for the study of stem cells in tissue engineering and repair. Dental Pulp Stem Cells (DPSC) from permanent teeth and Stem cells from Human Exfoliated Deciduous teeth (SHED) are particularly interesting sources for tissue engineering as they are easily accessible and expandable. Previously, we have shown that DPSCs and SHEDs can differentiate into endothelial cells and form functional blood vessels through vasculogenesis. Here, we described how we created the "pulpbow" (pulp + rainbow), a multicolor tag experimental model that is stable, permanent, unique to each cell and passed through generations. We used the pulpbow to understand how dental pulp stem cells contributed to blood vessel formation in 3D models in in vitro and ex vivo live cell tracking, and in vivo transplantation assays. Simultaneous tracking of cells during sprout formation revealed that no single multicolor-tagged cell was more prone to vasculogenesis. During this process, there was intense cell motility with minimal proliferation in early time points. In later stages, when the availability of undifferentiated cells around the forming sprout decreased, there was local clonal proliferation mediated by proximity. These results unveiled that the vasculogenesis process mediated by dental pulp stem cells is dynamic and proximity to the sprouting area is critical for cell fate decisions.
Collapse
Affiliation(s)
- Andrea Mantesso
- Angiogenesis Research Laboratory, Department of Cariology, Restorative Sciences and Endodontics, University of Michigan School of Dentistry, Ann Arbor, MI 48109, USA; (A.M.); (Z.Z.); (K.A.W.); (A.E.H.)
| | - Zhaocheng Zhang
- Angiogenesis Research Laboratory, Department of Cariology, Restorative Sciences and Endodontics, University of Michigan School of Dentistry, Ann Arbor, MI 48109, USA; (A.M.); (Z.Z.); (K.A.W.); (A.E.H.)
| | - Kristy A. Warner
- Angiogenesis Research Laboratory, Department of Cariology, Restorative Sciences and Endodontics, University of Michigan School of Dentistry, Ann Arbor, MI 48109, USA; (A.M.); (Z.Z.); (K.A.W.); (A.E.H.)
| | - Alexandra E. Herzog
- Angiogenesis Research Laboratory, Department of Cariology, Restorative Sciences and Endodontics, University of Michigan School of Dentistry, Ann Arbor, MI 48109, USA; (A.M.); (Z.Z.); (K.A.W.); (A.E.H.)
| | - Ajai J. Pulianmackal
- Department of Molecular, Cellular and Developmental Biology, University of Michigan College of Literature, Science and the Arts, Ann Arbor, MI 48109, USA;
| | - Jacques E. Nör
- Angiogenesis Research Laboratory, Department of Cariology, Restorative Sciences and Endodontics, University of Michigan School of Dentistry, Ann Arbor, MI 48109, USA; (A.M.); (Z.Z.); (K.A.W.); (A.E.H.)
- Department of Biomedical Engineering, University of Michigan College of Engineering, Ann Arbor, MI 48109, USA
- Department of Otolaryngology, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| |
Collapse
|