1
|
Zhao X, Yao M, Wang Y, Feng C, Yang Y, Tian L, Bao C, Li X, Zhu X, Zhang X. Neuroregulation during Bone Formation and Regeneration: Mechanisms and Strategies. ACS APPLIED MATERIALS & INTERFACES 2025; 17:7223-7250. [PMID: 39869030 DOI: 10.1021/acsami.4c16786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
The skeleton is highly innervated by numerous nerve fibers. These nerve fibers, in addition to transmitting information within the bone and mediating bone sensations, play a crucial role in regulating bone tissue formation and regeneration. Traditional bone tissue engineering (BTE) often fails to achieve satisfactory outcomes when dealing with large-scale bone defects, which is frequently related to the lack of effective reconstruction of the neurovascular network. In recent years, increasing research has revealed the critical role of nerves in bone metabolism. Nerve fibers regulate bone cells through neurotransmitters, neuropeptides, and peripheral glial cells. Furthermore, nerves also coordinate with the vascular and immune systems to jointly construct a microenvironment favorable for bone regeneration. As a signaling driver of bone formation, neuroregulation spans the entire process of bone physiological activities from the embryonic formation to postmaturity remodeling and repair. However, there is currently a lack of comprehensive summaries of these regulatory mechanisms. Therefore, this review sketches out the function of nerves during bone formation and regeneration. Then, we elaborate on the mechanisms of neurovascular coupling and neuromodulation of bone immunity. Finally, we discuss several novel strategies for neuro-bone tissue engineering (NBTE) based on neuroregulation of bone, focusing on the coordinated regeneration of nerve and bone tissue.
Collapse
Affiliation(s)
- Xiangrong Zhao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Med-X Center for Materials, Sichuan University, Chengdu 610041, Sichuan, China
| | - Meilin Yao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Med-X Center for Materials, Sichuan University, Chengdu 610041, Sichuan, China
| | - Yuyi Wang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Cong Feng
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Yuhan Yang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Med-X Center for Materials, Sichuan University, Chengdu 610041, Sichuan, China
| | - Luoqiang Tian
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Chongyun Bao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Med-X Center for Materials, Sichuan University, Chengdu 610041, Sichuan, China
| | - Xiangfeng Li
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Xiangdong Zhu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| |
Collapse
|
2
|
Wei X, Li M, You J, Luo J, Zhai J, Zhang J, Feng J, Wang H, Zhou Y. A Procedural Overview of the Involvement of Small Molecules in the Nervous System in the Regulation of Bone Healing. Int J Nanomedicine 2025; 20:1263-1284. [PMID: 39906525 PMCID: PMC11792627 DOI: 10.2147/ijn.s505677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 01/14/2025] [Indexed: 02/06/2025] Open
Abstract
Clinically, a multitude of factors can contribute to the development of bone defects. In the process of bone healing, the nervous system plays a vital role in bone regeneration. Small molecules from the nervous system, such as neurotrophic factors and neuropeptides, have been found to stimulate osteoblast proliferation and differentiation by activating signaling pathways associated with bone calcification and angiogenesis. These small molecules play a crucial regulatory role at various stages of bone healing. The systematic release mechanism of small molecules within the nervous system through diverse bone tissue engineering materials holds significant clinical implications for the controlled regulation of the bone healing process. This review provides an overview of the involvement of various nervous system small molecules at different stages of bone healing and discusses their regulatory mechanisms, aiming to establish a theoretical foundation for programmed regulation in bone regeneration and design of replacement materials in bone tissue engineering.
Collapse
Affiliation(s)
- Xuyan Wei
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
| | - Mucong Li
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
| | - Jiaqian You
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
| | - Jiaxin Luo
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
| | - Jingjie Zhai
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
| | - Jiameng Zhang
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
| | - Jian Feng
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
| | - Hanchi Wang
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
| | - Yanmin Zhou
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
| |
Collapse
|
3
|
Bai L, Li J, Li G, Zhou D, Su J, Liu C. Skeletal interoception and prospective application in biomaterials for bone regeneration. Bone Res 2025; 13:1. [PMID: 39743568 DOI: 10.1038/s41413-024-00378-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 10/08/2024] [Accepted: 10/21/2024] [Indexed: 01/04/2025] Open
Abstract
Accumulating research has shed light on the significance of skeletal interoception, in maintaining physiological and metabolic homeostasis related to bone health. This review provides a comprehensive analysis of how skeletal interoception influences bone homeostasis, delving into the complex interplay between the nervous system and skeletal system. One key focus of the review is the role of various factors such as prostaglandin E2 (PGE2) in skeletal health via skeletal interoception. It explores how nerves innervating the bone tissue communicate with the central nervous system to regulate bone remodeling, a process critical for maintaining bone strength and integrity. Additionally, the review highlights the advancements in biomaterials designed to utilize skeletal interoception for enhancing bone regeneration and treatment of bone disorders. These biomaterials, tailored to interact with the body's interoceptive pathways, are positioned at the forefront of innovative treatments for conditions like osteoporosis and fractures. They represent a convergence of bioengineering, neuroscience, and orthopedics, aiming to create more efficient and targeted therapies for bone-related disorders. In conclusion, the review underscores the importance of skeletal interoception in physiological regulation and its potential in developing more effective therapies for bone regeneration. It emphasizes the need for further research to fully understand the mechanisms of skeletal interoception and to harness its therapeutic potential fully.
Collapse
Affiliation(s)
- Long Bai
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, China
- Wenzhou Institute of Shanghai University, Wenzhou, Zhejiang, China
| | - Jilong Li
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, China
| | - Guangfeng Li
- Department of Orthopedics, Shanghai Zhongye Hospital, Shanghai, China
| | - Dongyang Zhou
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, China.
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, China.
| | - Jiacan Su
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, China.
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, China.
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Changsheng Liu
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, China.
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, China.
- Key Laboratory for Ultrafine Materials of Ministry of Education, Engineering Research Center for Biomedical Materials of the Ministry of Education, East China University of Science and Technology, Shanghai, China.
| |
Collapse
|
4
|
Kim S, Hong HS. Substance P alleviates liver fibrosis by modulating inflammation and mobilizing reparative stem cells. Int Immunopharmacol 2024; 142:113211. [PMID: 39321699 DOI: 10.1016/j.intimp.2024.113211] [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: 05/12/2024] [Revised: 09/11/2024] [Accepted: 09/17/2024] [Indexed: 09/27/2024]
Abstract
Repetitive hepatic damage resulting from viral hepatitis, toxins, and alcohol abuse induces chronic inflammation and excessive accumulation of the extracellular matrix, leading to the development of liver cirrhosis. Substance P (SP) promotes endogenous wound healing by mobilizing bone marrow stem cells and stimulating anti-inflammatory responses. This study aimed to investigate whether SP exerts a therapeutic effect on liver fibrosis by recruiting endogenous stem cells and modulating immune responses. A non-clinical model of liver cirrhosis was established through repeated injections of thioacetamide and recombinant leptin. After confirming liver fibrosis, SP was administered intravenously for 6 weeks. SP treatment decreased the formation of hepatic micronodules on the external surface of the liver and the infiltration of immune cells. Furthermore, SP treatment notably reduced the deposition of collagen and the activation of hepatic stellate cells, concomitant with decreased levels of transforming growth factor-β1 and matrix metalloproteinase activity. In the context of severe hepatic damage, SP increased the number of circulating stem cells, leading to the restoration of the reparative stem cell pool in the bone marrow. The findings of this study suggest that SP alleviates liver fibrosis by modulating the mobilization of functional stem cells and the immune response.
Collapse
Affiliation(s)
- Suna Kim
- Department of Genetic Engineering, Graduate School of Biotechnology, Kyung Hee University, Deokyoung dae-ro, 1732, Yong In 17104, Republic of Korea; Kyung Hee Institute of Regenerative Medicine (KIRM), Medical Science Research Institute, Kyung Hee University Medical Center, Kyung Hee dae-ro 23, Hoegi-dong, Seoul 02447, Republic of Korea
| | - Hyun Sook Hong
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, Kyung Hee dae-ro, 24, Seoul 02461, Republic of Korea; East-West Medical Research Institute, Kyung Hee University, Kyung Hee dae-ro, 24, Hoegi-dong, Seoul 02461, Republic of Korea; Kyung Hee Institute of Regenerative Medicine (KIRM), Medical Science Research Institute, Kyung Hee University Medical Center, Kyung Hee dae-ro 23, Hoegi-dong, Seoul 02447, Republic of Korea.
| |
Collapse
|
5
|
Toan NK, Kim S, Ahn S. Neuropeptides regulate embryonic salivary gland branching through the FGF/FGFR pathway in aging klotho-deficient mice. Aging Cell 2024; 23:e14329. [PMID: 39239870 PMCID: PMC11634708 DOI: 10.1111/acel.14329] [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: 03/14/2024] [Revised: 06/19/2024] [Accepted: 07/18/2024] [Indexed: 09/07/2024] Open
Abstract
Salivary gland branching morphogenesis is regulated by the functional integration of neuronal signaling, but the underlying mechanisms are not fully understood in aging accelerated klotho-deficient (Kl-/-) mice. Here, we investigated whether the neuropeptides substance P (SP) and neuropeptide Y (NPY) affect the branching morphogenesis of embryonic salivary glands in aging Kl-/- mice. In the salivary glands of embryonic Kl-/- mice, morphological analysis and immunostaining revealed that epithelial bud formation, neuronal cell proliferation/differentiation, and the expression of the salivary gland functional marker ZO-1 were decreased in embryonic ductal cells. Incubation with SP/NPY at E12-E13d promoted branching morphogenesis, parasympathetic innervation, and epithelial proliferation in salivary glands of embryonic Kl-/- mice. The ERK inhibitor U0126 specifically inhibited neuronal substance-induced epithelial bud formation in the embryonic salivary gland. RNA-seq profiling analysis revealed that the expression of fibroblast growth factors/fibroblast growth factors (FGFs/FGFRs) and their receptors was significantly regulated by SP/NPY treatment in the embryonic salivary gland (E15). The FGFR inhibitor BGJ389 inhibited new branching formation induced by SP and NPY treatment and ERK1/2 expression. These results showed that aging may affect virtually the development of salivary gland by neuronal dysfunction. The neuropeptides SP/NPY induced embryonic salivary gland development through FGF/FGFR/ERK1/2-mediated signaling.
Collapse
Affiliation(s)
- Nguyen Khanh Toan
- Department of Pathology, School of DentistryChosun UniversityGwangjuRepublic of Korea
| | - Soo‐A Kim
- Department of Biochemistry, School of Oriental MedicineDongguk UniversityGyeongjuRepublic of Korea
| | - Sang‐Gun Ahn
- Department of Pathology, School of DentistryChosun UniversityGwangjuRepublic of Korea
| |
Collapse
|
6
|
Borda M, Sierra R, Cantero MJ, Gómez Bustillo S, Fiore EJ, Giardelli G, Martino Garcet M, Rebottaro ML, Bayo Fina JM, Schiavone M, Rubione J, García MG, Montaner A, Mazzolini GD, Aquino JB. The antifibrotic potential of IMT504: modulation of GLAST + Wnt1 + bone marrow stromal progenitors and hepatic microenvironment. Stem Cell Res Ther 2024; 15:278. [PMID: 39227908 PMCID: PMC11373403 DOI: 10.1186/s13287-024-03896-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: 05/08/2024] [Accepted: 08/23/2024] [Indexed: 09/05/2024] Open
Abstract
BACKGROUND The immunomodulatory oligodeoxynucleotide (ODN) IMT504 might harbor antifibrotic properties within the liver. METHODS Fibrosis models were induced in mice through thioacetamide (TAA) administration and bile-duct ligation. Cre-loxP mice were utilized to identify GLAST + Wnt1 + bone marrow stromal progenitors (BMSPs) and to examine their contribution with cells in the liver. In vivo and in vitro assays; flow-cytometry, immunohistochemistry, and qPCR were conducted. RESULTS IMT504 demonstrated significant inhibition of liver fibrogenesis progression and reversal of established fibrosis. Early responses to IMT504 involved the suppression of profibrogenic and proinflammatory markers, coupled with an augmentation of hepatocyte proliferation. Additionally, this ODN stimulated the proliferation and mobilization of GLAST + Wnt1 + BMSPs, likely amplifying their contribution with endothelial- and hepatocytes-like cells. Moreover, IMT504 significantly modulated the expression levels of Wnt ligands and signaling pathway/target genes specifically within GLAST + Wnt1 + BMSPs, with minimal impact on other BMSPs. Intriguingly, both IMT504 and conditioned media from IMT504-pre-treated GLAST + Wnt1 + BMSPs shifted the phenotype of fibrotic macrophages, hepatic stellate cells, and hepatocytes, consistent with the potent antifibrotic effects observed. CONCLUSION In summary, our findings identify IMT504 as a promising candidate molecule with potent antifibrotic properties, operating through both direct and indirect mechanisms, including the activation of GLAST + Wnt1 + BMSPs.
Collapse
Affiliation(s)
- Maximiliano Borda
- Developmental Biology & Regenerative Medicine Laboratory, Instituto de Investigaciones en Medicina Traslacional, CONICET-Universidad Austral, Derqui, Pilar, Buenos Aires, Argentina
- Facultad de Ciencias Biomédicas, Universidad Austral, Pilar, Argentina
| | - Romina Sierra
- Developmental Biology & Regenerative Medicine Laboratory, Instituto de Investigaciones en Medicina Traslacional, CONICET-Universidad Austral, Derqui, Pilar, Buenos Aires, Argentina
- Facultad de Ciencias Biomédicas, Universidad Austral, Pilar, Argentina
| | - María José Cantero
- Facultad de Ciencias Biomédicas, Universidad Austral, Pilar, Argentina
- Gene Therapy Laboratory, Instituto de Investigaciones en Medicina Traslacional, CONICET- Universidad Austral, Buenos Aires, Argentina
| | - Sofía Gómez Bustillo
- Instituto de Ciencia y Tecnología Dr. César Milstein. Fundación Pablo Cassará, Buenos Aires City, Argentina
| | - Esteban Juan Fiore
- Facultad de Ciencias Biomédicas, Universidad Austral, Pilar, Argentina
- Gene Therapy Laboratory, Instituto de Investigaciones en Medicina Traslacional, CONICET- Universidad Austral, Buenos Aires, Argentina
| | - Gianlucca Giardelli
- Developmental Biology & Regenerative Medicine Laboratory, Instituto de Investigaciones en Medicina Traslacional, CONICET-Universidad Austral, Derqui, Pilar, Buenos Aires, Argentina
- Facultad de Ciencias Biomédicas, Universidad Austral, Pilar, Argentina
| | - Matías Martino Garcet
- Developmental Biology & Regenerative Medicine Laboratory, Instituto de Investigaciones en Medicina Traslacional, CONICET-Universidad Austral, Derqui, Pilar, Buenos Aires, Argentina
- Facultad de Ciencias Biomédicas, Universidad Austral, Pilar, Argentina
| | - María Luz Rebottaro
- Developmental Biology & Regenerative Medicine Laboratory, Instituto de Investigaciones en Medicina Traslacional, CONICET-Universidad Austral, Derqui, Pilar, Buenos Aires, Argentina
| | - Juan Miguel Bayo Fina
- Facultad de Ciencias Biomédicas, Universidad Austral, Pilar, Argentina
- Gene Therapy Laboratory, Instituto de Investigaciones en Medicina Traslacional, CONICET- Universidad Austral, Buenos Aires, Argentina
| | - Máximo Schiavone
- Developmental Biology & Regenerative Medicine Laboratory, Instituto de Investigaciones en Medicina Traslacional, CONICET-Universidad Austral, Derqui, Pilar, Buenos Aires, Argentina
- Facultad de Ciencias Biomédicas, Universidad Austral, Pilar, Argentina
| | - Julia Rubione
- Mechanisms and Therapeutic Innovation in Pain Laboratory, Instituto de Investigaciones en Medicina Traslacional, CONICET-Universidad Austral, Buenos Aires, Argentina
| | - Mariana Gabriela García
- Facultad de Ciencias Biomédicas, Universidad Austral, Pilar, Argentina
- Gene Therapy Laboratory, Instituto de Investigaciones en Medicina Traslacional, CONICET- Universidad Austral, Buenos Aires, Argentina
| | - Alejandro Montaner
- Instituto de Ciencia y Tecnología Dr. César Milstein. Fundación Pablo Cassará, Buenos Aires City, Argentina
| | - Guillermo Daniel Mazzolini
- Facultad de Ciencias Biomédicas, Universidad Austral, Pilar, Argentina
- Gene Therapy Laboratory, Instituto de Investigaciones en Medicina Traslacional, CONICET- Universidad Austral, Buenos Aires, Argentina
| | - Jorge Benjamín Aquino
- Developmental Biology & Regenerative Medicine Laboratory, Instituto de Investigaciones en Medicina Traslacional, CONICET-Universidad Austral, Derqui, Pilar, Buenos Aires, Argentina.
- Facultad de Ciencias Biomédicas, Universidad Austral, Pilar, Argentina.
| |
Collapse
|
7
|
Wang X, Hong CG, Duan R, Pang ZL, Zhang MN, Xie H, Liu ZZ. Transplantation of olfactory mucosa mesenchymal stromal cells repairs spinal cord injury by inducing microglial polarization. Spinal Cord 2024; 62:429-439. [PMID: 38849489 DOI: 10.1038/s41393-024-01004-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 05/27/2024] [Accepted: 06/03/2024] [Indexed: 06/09/2024]
Abstract
STUDY DESIGN Animal studies OBJECTIVES: To evaluate the therapeutic effect of olfactory mucosa mesenchymal stem cell (OM-MSCs) transplantation in mice with spinal cord injury (SCI) and to explore the mechanism by which OM-MSCs inhibit neuroinflammation and improve SCI. SETTING Xiangya Hospital, Central South University; Affiliated Hospital of Guangdong Medical University. METHODS Mice (C57BL/6, female, 6-week-old) were randomly divided into sham, SCI, and SCI + OM-MSC groups. The SCI mouse model was generated using Allen's method. OM-MSCs were immediately delivered to the lateral ventricle after SCI using stereotaxic brain injections. One day prior to injury and on days 1, 5, 7, 14, 21, and 28 post-injury, the Basso Mouse Scale and Rivlin inclined plate tests were performed. Inflammation and microglial polarization were evaluated using histological staining, immunofluorescence, and qRT-PCR. RESULTS OM-MSCs originating from the neuroectoderm have great potential in the management of SCI owing to their immunomodulatory effects. OM-MSCs administration improved motor function, alleviated inflammation, promoted the transformation of the M1 phenotype of microglia into the M2 phenotype, facilitated axonal regeneration, and relieved spinal cord injury in SCI mice. CONCLUSIONS OM-MSCs reduced the level of inflammation in the spinal cord tissue, protected neurons, and repaired spinal cord injury by regulating the M1/M2 polarization of microglia.
Collapse
Affiliation(s)
- Xin Wang
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Chun-Gu Hong
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Ran Duan
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Zhi-Lin Pang
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Min-Na Zhang
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Hui Xie
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| | - Zheng-Zhao Liu
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524001, China.
| |
Collapse
|
8
|
Pi HJ, Huang B, Yuan Q, Jing JJ. Neural regulation of mesenchymal stem cells in craniofacial bone: development, homeostasis and repair. Front Physiol 2024; 15:1423539. [PMID: 39135707 PMCID: PMC11318092 DOI: 10.3389/fphys.2024.1423539] [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: 04/26/2024] [Accepted: 07/15/2024] [Indexed: 08/15/2024] Open
Abstract
Mesenchymal stem cells endow various functions, including proliferation, multipotency, migration, etc. Craniofacial bones originate from the cranial neural crest and are developed mainly through intramembranous ossification, which are different from long bones. There are varied mesenchymal stem cells existing in the craniofacial bone, including Gli1 + cells, Axin2 + cells, Prx1 + cells, etc. Nerves distributed in craniofacial area are also derived from the neural crest, and the trigeminal nerve is the major sensory nerve in craniofacial area. The nerves and the skeleton are tightly linked spatially, and the skeleton is broadly innervated by sensory and sympathetic nerves, which also participate in bone development, homeostasis and healing process. In this review, we summarize mesenchymal stem cells located in craniofacial bone or, to be more specific, in jaws, temporomandibular joint and cranial sutures. Then we discuss the research advance concerning neural regulation of mesenchymal stem cells in craniofacial bone, mainly focused on development, homeostasis and repair. Discovery of neural regulation of mesenchymal stem cells may assist in treatment in the craniofacial bone diseases or injuries.
Collapse
Affiliation(s)
| | | | - Quan Yuan
- *Correspondence: Quan Yuan, ; Jun-Jun Jing,
| | | |
Collapse
|
9
|
Kim HE, Ju HJ, Kim S, Kim YH, Lee S, Choi S, Yoon HC, Choi HS, Kim MS. Amplifying endogenous stem cell migration for in situ bone tissue formation: Substance P analog and BMP mimetic peptide-loaded click-crosslinked hyaluronic acid hydrogel. Mater Today Bio 2024; 26:101070. [PMID: 38711939 PMCID: PMC11070699 DOI: 10.1016/j.mtbio.2024.101070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 04/20/2024] [Accepted: 04/25/2024] [Indexed: 05/08/2024] Open
Abstract
Endogenous stem cell-driven in situ bone tissue formation has recently garnered increasing attention. Therefore, our study sought to refine methods to enhance the migration and subsequent osteogenic differentiation of these cells. Our innovative approach involves using an injectable hydrogel that combines click cross-linking sites and a BMP-2 mimetic peptide (BP) with hyaluronic acid (HA). This injectable formulation, hereinafter referred to as SPa + Cx-HA-BP, incorporates a substance P analog peptide (SPa) with Cx-HA-BP, proving versatile for in vitro and in vivo applications without cytotoxicity. The controlled release of SPa creates a gradient that guides endogenous stem cells towards the Cx-HA scaffold from specific tissue niches. Both Cx-HA and SPa+Cx-HA induced minimal changes in the expression of genes associated with osteogenic differentiation. In contrast, these genes were robustly induced by both SPa + Cx-HA+BP and SPa + Cx-HA-BP, in which BP was respectively integrated via physical and chemical methods. Remarkably, chemically incorporating BP (Cx-HA-BP) resulted in 4-9 times higher osteogenic gene expression than physically mixed BP in Cx-HA+BP. This study validates the role of SPa role in guiding endogenous stem cells toward the hydrogel and underscores the substantial impact of sustained BP presence within the hydrogel. Collectively, our findings offer valuable insights for the development of innovative strategies to promote endogenous stem cell-based tissue regeneration. The developed hydrogel effectively guides stem cells from their natural locations and facilitates sustained osteogenic differentiation, thus holding great promise for applications in regenerative medicine.
Collapse
Affiliation(s)
- Hee Eun Kim
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, South Korea
| | - Hyeon Jin Ju
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, South Korea
| | - Shina Kim
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, South Korea
| | - Young Hun Kim
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, South Korea
| | - Soyeon Lee
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, South Korea
| | - Sangdun Choi
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, South Korea
| | - Hyun C. Yoon
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, South Korea
| | - Hak Soo Choi
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Moon Suk Kim
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, South Korea
- Research Institute, Medipolymer, Woncheon Dong 274, Suwon, 16522, South Korea
| |
Collapse
|
10
|
Chin JY, Liu C, Lee IXY, Lin MTY, Cheng CY, Wong JHF, Teo CL, Mehta JS, Liu YC. Impact of Age on the Characteristics of Corneal Nerves and Corneal Epithelial Cells in Healthy Adults. Cornea 2024; 43:409-418. [PMID: 37643477 PMCID: PMC10906190 DOI: 10.1097/ico.0000000000003363] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 05/26/2023] [Accepted: 07/02/2023] [Indexed: 08/31/2023]
Abstract
PURPOSE The aim of this study was to investigate age-related changes in corneal nerves and corneal epithelial cell parameters and to establish age-adjusted reference values. METHODS A total of 7025 corneal nerve images and 4215 corneal epithelial images obtained using in vivo confocal microscopy from 281 eyes of 143 healthy participants were included. Seven corneal nerve parameters and 3 corneal epithelial cell parameters were quantified using 2 automatic analytic software and analyzed across 6 age groups ranging from 21 to 80 years. RESULTS There was a declining trend in all 7 nerve parameters with advancing age. In particular, corneal nerve fiber length and corneal nerve fiber density demonstrated a significant decrease in subjects aged 65 years or older compared with subjects younger than 65 years (10.8 ± 2.6 mm/mm 2 vs. 9.9 ± 2.0 mm/mm 2 , P = 0.011 in corneal nerve fiber length; 15.8 ± 5.2 fibers/mm 2 vs. 14.4 ± 4.3 fibers/mm 2 , P = 0.046 in corneal nerve fiber density), whereas corneal nerve fractal dimension demonstrated a borderline significant decrease ( P = 0.057). Similarly, there was a general declining trend in all epithelial cell parameters with advancing age. Corneal epithelial cell circularity was significantly lower in subjects aged 65 years and older as compared to subjects younger than 65 years (0.722 ± 0.021 μm 2 vs. 0.714 ± 0.021 μm 2 ; P = 0.011). CONCLUSIONS Advancing age results in reduced corneal nerve metrics and alteration of corneal cell morphology. Aging effects should be considered when evaluating patients with corneal neuropathy.
Collapse
Affiliation(s)
- Jia Ying Chin
- Tissue Engineering and Stem Cell Group, Singapore Eye Research Institute, Singapore
| | - Chang Liu
- Tissue Engineering and Stem Cell Group, Singapore Eye Research Institute, Singapore
| | - Isabelle Xin Yu Lee
- Tissue Engineering and Stem Cell Group, Singapore Eye Research Institute, Singapore
| | - Molly Tzu Yu Lin
- Tissue Engineering and Stem Cell Group, Singapore Eye Research Institute, Singapore
| | - Ching-Yu Cheng
- Ophthalmology and Visual Sciences Academic Clinical Program, Duke-NUS Medical School, Singapore
- Epidemiology Group, Singapore Eye Research Institute, Singapore
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore and National University Health System, Singapore, Singapore
| | - Jipson Hon Fai Wong
- Tissue Engineering and Stem Cell Group, Singapore Eye Research Institute, Singapore
| | - Cong Ling Teo
- Epidemiology Group, Singapore Eye Research Institute, Singapore
| | - Jodhbir S. Mehta
- Tissue Engineering and Stem Cell Group, Singapore Eye Research Institute, Singapore
- Ophthalmology and Visual Sciences Academic Clinical Program, Duke-NUS Medical School, Singapore
- Department of Cornea and External Eye Disease, Singapore National Eye Centre, Singapore; and
| | - Yu-Chi Liu
- Tissue Engineering and Stem Cell Group, Singapore Eye Research Institute, Singapore
- Ophthalmology and Visual Sciences Academic Clinical Program, Duke-NUS Medical School, Singapore
- Department of Cornea and External Eye Disease, Singapore National Eye Centre, Singapore; and
- Department of Ophthalmology, National Taiwan University Hospital, Taipei, Taiwan
| |
Collapse
|
11
|
Seal A, Hughes M, Wei F, Pugazhendhi AS, Ngo C, Ruiz J, Schwartzman JD, Coathup MJ. Sphingolipid-Induced Bone Regulation and Its Emerging Role in Dysfunction Due to Disease and Infection. Int J Mol Sci 2024; 25:3024. [PMID: 38474268 PMCID: PMC10932382 DOI: 10.3390/ijms25053024] [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: 02/09/2024] [Revised: 03/02/2024] [Accepted: 03/04/2024] [Indexed: 03/14/2024] Open
Abstract
The human skeleton is a metabolically active system that is constantly regenerating via the tightly regulated and highly coordinated processes of bone resorption and formation. Emerging evidence reveals fascinating new insights into the role of sphingolipids, including sphingomyelin, sphingosine, ceramide, and sphingosine-1-phosphate, in bone homeostasis. Sphingolipids are a major class of highly bioactive lipids able to activate distinct protein targets including, lipases, phosphatases, and kinases, thereby conferring distinct cellular functions beyond energy metabolism. Lipids are known to contribute to the progression of chronic inflammation, and notably, an increase in bone marrow adiposity parallel to elevated bone loss is observed in most pathological bone conditions, including aging, rheumatoid arthritis, osteoarthritis, and osteomyelitis. Of the numerous classes of lipids that form, sphingolipids are considered among the most deleterious. This review highlights the important primary role of sphingolipids in bone homeostasis and how dysregulation of these bioactive metabolites appears central to many chronic bone-related diseases. Further, their contribution to the invasion, virulence, and colonization of both viral and bacterial host cell infections is also discussed. Many unmet clinical needs remain, and data to date suggest the future use of sphingolipid-targeted therapy to regulate bone dysfunction due to a variety of diseases or infection are highly promising. However, deciphering the biochemical and molecular mechanisms of this diverse and extremely complex sphingolipidome, both in terms of bone health and disease, is considered the next frontier in the field.
Collapse
Affiliation(s)
- Anouska Seal
- Biionix Cluster, University of Central Florida, Orlando, FL 32827, USA; (A.S.); (F.W.); (A.S.P.); (C.N.)
| | - Megan Hughes
- School of Biosciences, Cardiff University, Cardiff CF10 3AT, UK;
| | - Fei Wei
- Biionix Cluster, University of Central Florida, Orlando, FL 32827, USA; (A.S.); (F.W.); (A.S.P.); (C.N.)
- College of Medicine, University of Central Florida, Orlando, FL 32827, USA (J.D.S.)
| | - Abinaya S. Pugazhendhi
- Biionix Cluster, University of Central Florida, Orlando, FL 32827, USA; (A.S.); (F.W.); (A.S.P.); (C.N.)
- College of Medicine, University of Central Florida, Orlando, FL 32827, USA (J.D.S.)
| | - Christopher Ngo
- Biionix Cluster, University of Central Florida, Orlando, FL 32827, USA; (A.S.); (F.W.); (A.S.P.); (C.N.)
- College of Medicine, University of Central Florida, Orlando, FL 32827, USA (J.D.S.)
| | - Jonathan Ruiz
- College of Medicine, University of Central Florida, Orlando, FL 32827, USA (J.D.S.)
| | | | - Melanie J. Coathup
- Biionix Cluster, University of Central Florida, Orlando, FL 32827, USA; (A.S.); (F.W.); (A.S.P.); (C.N.)
- College of Medicine, University of Central Florida, Orlando, FL 32827, USA (J.D.S.)
| |
Collapse
|
12
|
Morris AJ, Parker RS, Nazzal MK, Natoli RM, Fehrenbacher JC, Kacena MA, White FA. Cracking the Code: The Role of Peripheral Nervous System Signaling in Fracture Repair. Curr Osteoporos Rep 2024; 22:193-204. [PMID: 38236511 PMCID: PMC10912155 DOI: 10.1007/s11914-023-00846-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/19/2023] [Indexed: 01/19/2024]
Abstract
PURPOSE OF REVIEW The traditionally understated role of neural regulation in fracture healing is gaining prominence, as recent findings underscore the peripheral nervous system's critical contribution to bone repair. Indeed, it is becoming more evident that the nervous system modulates every stage of fracture healing, from the onset of inflammation to repair and eventual remodeling. RECENT FINDINGS Essential to this process are neurotrophins and neuropeptides, such as substance P, calcitonin gene-related peptide, and neuropeptide Y. These molecules fulfill key roles in promoting osteogenesis, influencing inflammation, and mediating pain. The sympathetic nervous system also plays an important role in the healing process: while local sympathectomies may improve fracture healing, systemic sympathetic denervation impairs fracture healing. Furthermore, chronic activation of the sympathetic nervous system, often triggered by stress, is a potential impediment to effective fracture healing, marking an important area for further investigation. The potential to manipulate aspects of the nervous system offers promising therapeutic possibilities for improving outcomes in fracture healing. This review article is part of a series of multiple manuscripts designed to determine the utility of using artificial intelligence for writing scientific reviews.
Collapse
Affiliation(s)
- Ashlyn J Morris
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Reginald S Parker
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Murad K Nazzal
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Roman M Natoli
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jill C Fehrenbacher
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, USA
- Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Melissa A Kacena
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, USA.
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA.
- Richard L. Roudebush VA Medical Center, Indianapolis, IN, USA.
| | - Fletcher A White
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA.
- Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA.
- Richard L. Roudebush VA Medical Center, Indianapolis, IN, USA.
- Department of Anesthesia, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
13
|
Park DS, Oh S, Jin YJ, Na MH, Kim M, Kim JH, Hyun DY, Cho KH, Hong YJ, Kim JH, Ahn Y, Hermida-Prieto M, Vázquez-Rodríguez JM, Gutiérrez-Chico JL, Mariñas-Pardo L, Lim KS, Park JK, Byeon DH, Cho YN, Kee SJ, Sim DS, Jeong MH. Preliminary Investigation on Efficacy and Safety of Substance P-Coated Stent for Promoting Re-Endothelialization: A Porcine Coronary Artery Restenosis Model. Tissue Eng Regen Med 2024; 21:53-64. [PMID: 37973692 PMCID: PMC10764706 DOI: 10.1007/s13770-023-00608-y] [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: 07/26/2023] [Revised: 09/10/2023] [Accepted: 10/11/2023] [Indexed: 11/19/2023] Open
Abstract
BACKGROUND Current polymer-based drug-eluting stents (DESs) have fundamental issues about inflammation and delayed re-endothelializaton of the vessel wall. Substance-P (SP), which plays an important role in inflammation and endothelial cells, has not yet been applied to coronary stents. Therefore, this study compares poly lactic-co-glycolic acid (PLGA)-based everolimus-eluting stents (PLGA-EESs) versus 2-methacryloyloxyethyl phosphorylcholine (MPC)-based SP-eluting stents (MPC-SPs) in in-vitro and in-vivo models. METHODS The morphology of the stent surface and peptide/drug release kinetics from stents were evaluated. The in-vitro proliferative effect of SP released from MPC-SP is evaluated using human umbilical vein endothelial cell. Finally, the safety and efficacy of the stent are evaluated after inserting it into a pig's coronary artery. RESULTS Similar to PLGA-EES, MPC-SP had a uniform surface morphology with very thin coating layer thickness (2.074 μm). MPC-SP showed sustained drug release of SP for over 2 weeks. Endothelial cell proliferation was significantly increased in groups treated with SP (n = 3) compared with the control (n = 3) and those with everolimus (n = 3) (SP: 118.9 ± 7.61% vs. everolimus: 64.3 ± 12.37% vs. the control: 100 ± 6.64%, p < 0.05). In the animal study, the percent stenosis was higher in MPC-SP group (n = 7) compared to PLGA-EES group (n = 7) (MPC-SP: 28.6 ± 10.7% vs. PLGA-EES: 16.7 ± 6.3%, p < 0.05). MPC-SP group showed, however, lower inflammation (MPC-SP: 0.3 ± 0.26 vs. PLGA-EES: 1.2 ± 0.48, p < 0.05) and fibrin deposition (MPC-SP: 1.0 ± 0.73 vs. PLGA-EES: 1.5 ± 0.59, p < 0.05) around the stent strut. MPC-SP showed more increased expression of cluster of differentiation 31, suggesting enhanced re-endothelialization. CONCLUSION Compared to PLGA-EES, MPC-SP demonstrated more decreased inflammation of the vascular wall and enhanced re-endothelialization and stent coverage. Hence, MPC-SP has the potential therapeutic benefits for the treatment of coronary artery disease by solving limitations of currently available DESs.
Collapse
Affiliation(s)
- Dae Sung Park
- The Korea Cardiovascular Stent Research Institute, Chonnam National University, Gwangju, Korea
- The Cardiovascular Convergence Research Center of Chonnam National University Hospital Designated by Korea Ministry of Health and Welfare, Gwangju, Korea
- The Research Institute of Medical Sciences, Chonnam National University, Gwangju, Korea
| | - Seok Oh
- The Korea Cardiovascular Stent Research Institute, Chonnam National University, Gwangju, Korea
- The Cardiovascular Convergence Research Center of Chonnam National University Hospital Designated by Korea Ministry of Health and Welfare, Gwangju, Korea
- Department of Cardiovascular Medicine, Chonnam National University Hospital, Gwangju, Korea
| | - Yu Jeong Jin
- The Korea Cardiovascular Stent Research Institute, Chonnam National University, Gwangju, Korea
| | - Mi Hyang Na
- The Korea Cardiovascular Stent Research Institute, Chonnam National University, Gwangju, Korea
| | - Munki Kim
- The Korea Cardiovascular Stent Research Institute, Chonnam National University, Gwangju, Korea
- The Cardiovascular Convergence Research Center of Chonnam National University Hospital Designated by Korea Ministry of Health and Welfare, Gwangju, Korea
| | - Jeong Ha Kim
- The Korea Cardiovascular Stent Research Institute, Chonnam National University, Gwangju, Korea
- The Cardiovascular Convergence Research Center of Chonnam National University Hospital Designated by Korea Ministry of Health and Welfare, Gwangju, Korea
| | - Dae Young Hyun
- The Korea Cardiovascular Stent Research Institute, Chonnam National University, Gwangju, Korea
- The Cardiovascular Convergence Research Center of Chonnam National University Hospital Designated by Korea Ministry of Health and Welfare, Gwangju, Korea
- Department of Cardiovascular Medicine, Chonnam National University Hospital, Gwangju, Korea
| | - Kyung Hoon Cho
- The Korea Cardiovascular Stent Research Institute, Chonnam National University, Gwangju, Korea
- The Cardiovascular Convergence Research Center of Chonnam National University Hospital Designated by Korea Ministry of Health and Welfare, Gwangju, Korea
- Department of Cardiovascular Medicine, Chonnam National University Hospital, Gwangju, Korea
- Department of Cardiovascular Medicine, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Young Joon Hong
- The Korea Cardiovascular Stent Research Institute, Chonnam National University, Gwangju, Korea
- The Cardiovascular Convergence Research Center of Chonnam National University Hospital Designated by Korea Ministry of Health and Welfare, Gwangju, Korea
- Department of Cardiovascular Medicine, Chonnam National University Hospital, Gwangju, Korea
- Department of Cardiovascular Medicine, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Ju Han Kim
- The Korea Cardiovascular Stent Research Institute, Chonnam National University, Gwangju, Korea
- The Cardiovascular Convergence Research Center of Chonnam National University Hospital Designated by Korea Ministry of Health and Welfare, Gwangju, Korea
- Department of Cardiovascular Medicine, Chonnam National University Hospital, Gwangju, Korea
- Department of Cardiovascular Medicine, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Youngkeun Ahn
- The Korea Cardiovascular Stent Research Institute, Chonnam National University, Gwangju, Korea
- The Cardiovascular Convergence Research Center of Chonnam National University Hospital Designated by Korea Ministry of Health and Welfare, Gwangju, Korea
- Department of Cardiovascular Medicine, Chonnam National University Hospital, Gwangju, Korea
- Department of Cardiovascular Medicine, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Manuel Hermida-Prieto
- Instituto de Investigación Biomédica de A Coruña (INIBIC), Universidade da Coruña (UDC), A Coruña, Spain
| | - José Manuel Vázquez-Rodríguez
- Instituto de Investigación Biomédica de A Coruña (INIBIC), Universidade da Coruña (UDC), A Coruña, Spain
- Servicio de Cardiología, Complexo Hospitalario Universitario de A Coruña, A Coruña, Spain
| | - Juan Luis Gutiérrez-Chico
- Bundeswehrzentralkrankenhaus (Federal Army Central Military Hospital), Koblenz, Germany
- Universidad Alfonso X el Sabio, Madrid, Spain
| | - Luis Mariñas-Pardo
- Facultad de Ciencias de La Salud, Universidad Internacional de Valencia (VIU), Valencia, Spain
| | - Kyung Seob Lim
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Ochang, Korea
| | | | | | - Young-Nan Cho
- Department of Clinical Laboratory Medicine, Chonnam National University Hospital, Gwangju, Korea
| | - Seung-Jung Kee
- Department of Clinical Laboratory Medicine, Chonnam National University Hospital, Gwangju, Korea
| | - Doo Sun Sim
- The Korea Cardiovascular Stent Research Institute, Chonnam National University, Gwangju, Korea.
- The Cardiovascular Convergence Research Center of Chonnam National University Hospital Designated by Korea Ministry of Health and Welfare, Gwangju, Korea.
- Department of Cardiovascular Medicine, Chonnam National University Hospital, Gwangju, Korea.
- Department of Cardiovascular Medicine, Chonnam National University Medical School, Gwangju, Republic of Korea.
| | - Myung Ho Jeong
- The Korea Cardiovascular Stent Research Institute, Chonnam National University, Gwangju, Korea.
- The Cardiovascular Convergence Research Center of Chonnam National University Hospital Designated by Korea Ministry of Health and Welfare, Gwangju, Korea.
- Department of Cardiovascular Medicine, Chonnam National University Hospital, Gwangju, Korea.
- Department of Cardiovascular Medicine, Chonnam National University Medical School, Gwangju, Republic of Korea.
| |
Collapse
|
14
|
Yang X, Xiong M, Fu X, Sun X. Bioactive materials for in vivo sweat gland regeneration. Bioact Mater 2024; 31:247-271. [PMID: 37637080 PMCID: PMC10457517 DOI: 10.1016/j.bioactmat.2023.07.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 07/30/2023] [Accepted: 07/30/2023] [Indexed: 08/29/2023] Open
Abstract
Loss of sweat glands (SwGs) commonly associated with extensive skin defects is a leading cause of hyperthermia and heat stroke. In vivo tissue engineering possesses the potential to take use of the body natural ability to regenerate SwGs, making it more conducive to clinical translation. Despite recent advances in regenerative medicine, reconstructing SwG tissue with the same structure and function as native tissue remains challenging. Elucidating the SwG generation mechanism and developing biomaterials for in vivo tissue engineering is essential for understanding and developing in vivo SwG regenerative strategies. Here, we outline the cell biology associated with functional wound healing and the characteristics of bioactive materials. We critically summarize the recent progress in bioactive material-based cell modulation approaches for in vivo SwG regeneration, including the recruitment of endogenous cells to the skin lesion for SwG regeneration and in vivo cellular reprogramming for SwG regeneration. We discussed the re-establishment of microenvironment via bioactive material-mediated regulators. Besides, we offer promising perspectives for directing in situ SwG regeneration via bioactive material-based cell-free strategy, which is a simple and effective approach to regenerate SwG tissue with both fidelity of structure and function. Finally, we discuss the opportunities and challenges of in vivo SwG regeneration in detail. The molecular mechanisms and cell fate modulation of in vivo SwG regeneration will provide further insights into the regeneration of patient-specific SwGs and the development of potential intervention strategies for gland-derived diseases.
Collapse
Affiliation(s)
- Xinling Yang
- Research Center for Tissue Repair and Regeneration Affiliated to Medical Innovation Research Department and 4th Medical Center, PLA General Hospital and PLA Medical College, China
- PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, China
- Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, PR China
| | - Mingchen Xiong
- Research Center for Tissue Repair and Regeneration Affiliated to Medical Innovation Research Department and 4th Medical Center, PLA General Hospital and PLA Medical College, China
- PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, China
- Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, PR China
| | - Xiaobing Fu
- Research Center for Tissue Repair and Regeneration Affiliated to Medical Innovation Research Department and 4th Medical Center, PLA General Hospital and PLA Medical College, China
- PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, China
- Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, PR China
| | - Xiaoyan Sun
- Research Center for Tissue Repair and Regeneration Affiliated to Medical Innovation Research Department and 4th Medical Center, PLA General Hospital and PLA Medical College, China
- PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, China
- Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, PR China
| |
Collapse
|
15
|
Rodriguez FD, Covenas R. Association of Neurokinin-1 Receptor Signaling Pathways with Cancer. Curr Med Chem 2024; 31:6460-6486. [PMID: 37594106 DOI: 10.2174/0929867331666230818110812] [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: 02/03/2023] [Revised: 06/14/2023] [Accepted: 07/01/2023] [Indexed: 08/19/2023]
Abstract
BACKGROUND Numerous biochemical reactions leading to altered cell proliferation cause tumorigenesis and cancer treatment resistance. The mechanisms implicated include genetic and epigenetic changes, modified intracellular signaling, and failure of control mechanisms caused by intrinsic and extrinsic factors alone or combined. No unique biochemical events are responsible; entangled molecular reactions conduct the resident cells in a tissue to display uncontrolled growth and abnormal migration. Copious experimental research supports the etiological responsibility of NK-1R (neurokinin-1 receptor) activation, alone or cooperating with other mechanisms, in cancer appearance in different tissues. Consequently, a profound study of this receptor system in the context of malignant processes is essential to design new treatments targeting NK-1R-deviated activity. METHODS This study reviews and discusses recent literature that analyzes the main signaling pathways influenced by the activation of neurokinin 1 full and truncated receptor variants. Also, the involvement of NK-1R in cancer development is discussed. CONCLUSION NK-1R can signal through numerous pathways and cross-talk with other receptor systems. The participation of override or malfunctioning NK-1R in malignant processes needs a more precise definition in different types of cancers to apply satisfactory and effective treatments. A long way has already been traveled: the current disposal of selective and effective NK-1R antagonists and the capacity to develop new drugs with biased agonistic properties based on the receptor's structural states with functional significance opens immediate research action and clinical application.
Collapse
Affiliation(s)
- Francisco David Rodriguez
- Department of Biochemistry and Molecular Biology, Faculty of Chemical Sciences, University of Salamanca, 37007 Salamanca, Spain
- Group GIR USAL: BMD (Bases Moleculares del Desarrollo), University of Salamanca, Salamanca, Spain
| | - Rafael Covenas
- Group GIR USAL: BMD (Bases Moleculares del Desarrollo), University of Salamanca, Salamanca, Spain
- Laboratory of Neuroanatomy of the Peptidergic Systems, Institute of Neurosciences of Castilla y León (INCYL), University of Salamanca, 37007 Salamanca, Spain
| |
Collapse
|
16
|
Piao J, Cho H, Park JH, Yoo KH, Jeong I, Hong HS. Preconditioning with Substance P Restores Therapeutic Efficacy of Aged ADSC by Elevating TNFR2 and Paracrine Potential. BIOLOGY 2023; 12:1458. [PMID: 38132284 PMCID: PMC10740808 DOI: 10.3390/biology12121458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 11/16/2023] [Accepted: 11/21/2023] [Indexed: 12/23/2023]
Abstract
Aging leads to a decline in stem cell activity by reducing the repopulation rate and paracrine potential, ultimately diminishing efficacy in vivo. TNF-α can exert inflammatory and cell death actions via Erk by binding to TNFR-1, and survival and tissue repair actions via Akt by binding to TNFR-2. Aged cells are reported to have insufficient expression of TNFR-2, indicating that aged adipose-derived stem cells (ADSCs-E) lack the ability for cell survival and immune control compared to young ADSCs (ADSCs-Y). This study aims to assess the preconditioning effect of SP on the response of ADSCs-E to inflammation. ADSCs-E were treated with SP and then exposed to a high dose of TNF-α for 24 h. Consequently, ADSC-E exhibited weaker viability and lower TNFR2 levels compared to ADSC-Y. In response to TNF-α, the difference in TNFR2 expression became more pronounced in ADSC-E and ADSC-Y. Moreover, ADSC-E showed a severe deficiency in proliferation and paracrine activity. However, preconditioning with SP significantly enhanced the viability of ADSCs-E and also restored TNFR2 expression and paracrine potential, similar to ADSC-Y under inflammatory conditions. Our findings support the idea that preconditioning with SP has the potential to restore the cellular function of senescent stem cells before transplantation.
Collapse
Affiliation(s)
- Jiyuan Piao
- Department of Genetic Engineering, Graduate School of Biotechnology, Kyung Hee University, Yongin-si 17104, Republic of Korea; (J.P.)
| | - Hyunchan Cho
- Department of Genetic Engineering, Graduate School of Biotechnology, Kyung Hee University, Yongin-si 17104, Republic of Korea; (J.P.)
| | - Jong Hyun Park
- Department of Dance, College of Performing Arts & Sport, Han Yang University, Seoul 04763, Republic of Korea
| | - Ki Hyun Yoo
- SIMPLE Planet Inc., Seoul 04790, Republic of Korea
| | - Ildoo Jeong
- SIMPLE Planet Inc., Seoul 04790, Republic of Korea
| | - Hyun Sook Hong
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
- East-West Medical Research Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Kyung Hee Institute of Regenerative Medicine (KIRM), Medical Science Research Institute, Kyung Hee University Medical Center, Seoul 02447, Republic of Korea
| |
Collapse
|
17
|
Chen J, Ye P, Gu R, Zhu H, He W, Mu X, Wu X, Pang H, Han F, Nie X. Neuropeptide substance P: A promising regulator of wound healing in diabetic foot ulcers. Biochem Pharmacol 2023; 215:115736. [PMID: 37549795 DOI: 10.1016/j.bcp.2023.115736] [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: 06/12/2023] [Revised: 08/01/2023] [Accepted: 08/02/2023] [Indexed: 08/09/2023]
Abstract
In the past, neuropeptide substance P (SP) was predominantly recognized as a neuroinflammatory factor, while its potent healing activity was overlooked. This paper aims to review the regulatory characteristics of neuropeptide SP in both normal and diabetic wound healing. SP actively in the regulation of wound healing-related cells directly and indirectly, exhibiting robust inflammatory properties, promoting cell proliferation and migration and restoring the activity and paracrine ability of skin cells under diabetic conditions. Furthermore, SP not only regulates healing-related cells but also orchestrates the immune environment, thereby presenting unique and promising application prospects in wound intervention. As new SP-based preparations are being explored, SP-related drugs are poised to become an effective therapeutic intervention for diabetic foot ulcers (DFU).
Collapse
Affiliation(s)
- Jitao Chen
- College of Pharmacy, Zunyi Medical University, Zunyi 563000, China; Key Lab of the Basic Pharmacology of the Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Chinese Ministry of Education, Zunyi Medical University, Zunyi 563000, China
| | - Penghui Ye
- College of Pharmacy, Zunyi Medical University, Zunyi 563000, China; Key Lab of the Basic Pharmacology of the Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Chinese Ministry of Education, Zunyi Medical University, Zunyi 563000, China
| | - Rifang Gu
- University Medical Office, Zunyi Medical University, Zunyi 563000, China
| | - Huan Zhu
- College of Pharmacy, Zunyi Medical University, Zunyi 563000, China; Key Lab of the Basic Pharmacology of the Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Chinese Ministry of Education, Zunyi Medical University, Zunyi 563000, China
| | - Wenjie He
- College of Pharmacy, Zunyi Medical University, Zunyi 563000, China; Key Lab of the Basic Pharmacology of the Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Chinese Ministry of Education, Zunyi Medical University, Zunyi 563000, China
| | - Xingrui Mu
- College of Pharmacy, Zunyi Medical University, Zunyi 563000, China; Key Lab of the Basic Pharmacology of the Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Chinese Ministry of Education, Zunyi Medical University, Zunyi 563000, China
| | - Xingqian Wu
- College of Pharmacy, Zunyi Medical University, Zunyi 563000, China; Key Lab of the Basic Pharmacology of the Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Chinese Ministry of Education, Zunyi Medical University, Zunyi 563000, China
| | - Huiwen Pang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Felicity Han
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Xuqiang Nie
- College of Pharmacy, Zunyi Medical University, Zunyi 563000, China; Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia; Key Lab of the Basic Pharmacology of the Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Chinese Ministry of Education, Zunyi Medical University, Zunyi 563000, China.
| |
Collapse
|
18
|
Liu N, Liu D, Li Y, Zhang X, He J, Jiang Y, Wang Y, Ma Y, Jin H, Shen L. Effects and mechanisms of substance P on the proliferation and angiogenic differentiation of bone marrow mesenchymal stem cells: Bioinformatics and in vitro experiments. Genomics 2023; 115:110679. [PMID: 37423397 DOI: 10.1016/j.ygeno.2023.110679] [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: 04/16/2023] [Revised: 06/25/2023] [Accepted: 07/05/2023] [Indexed: 07/11/2023]
Abstract
The slight release of substance P (SP) from the end of peripheral nerve fibers causes a neurogenic inflammatory reaction, promotes vascular dilation and increases vascular permeability. However, whether SP can promote the angiogenesis of bone marrow mesenchymal stem cells (BMSCs) under high glucose conditions has not been reported. This study analyzed the targets, biological processes and molecular mechanisms underlying the effects of SP on BMSCs. BMSCs cultured in vitro were divided into a normal control group, high glucose control group, high glucose SP group and high glucose Akt inhibitor group to verify the effects of SP on BMSCs proliferation, migration and angiogenic differentiation. SP was found to act on 28 targets of BMSCs and participate in angiogenesis. Thirty-six core proteins, including AKT1, APP, BRCA1, CREBBP and EGFR, were identified. In a high glucose environment, SP increased the BMSCs proliferation optical density value and cell migration number and reduced the BMSCs apoptosis rate. In addition, SP induced BMSCs to highly express the CD31 protein, maintain the wall structure integrity of the matrix glue mesh and promote increases in the number of matrix glue meshes. These experiments showed that in a high glucose environment, SP acts on 28 targets of BMSCs that encode core proteins, such as AKT1, APP and BRCA1, and improves BMSCs proliferation, migration and angiogenic differentiation through the Akt signaling pathway.
Collapse
Affiliation(s)
- Na Liu
- Department of Anatomy, Qiqihar Medical University, No. 333, Bukui North Street, Jianhua District, Qiqihar 161006, China
| | - Danyang Liu
- Department of Histology & Embryology, Qiqihar Medical University, No. 333, Bukui North Street, Jianhua District, Qiqihar 161006, China
| | - Yongtao Li
- Department of Anatomy, Qiqihar Medical University, No. 333, Bukui North Street, Jianhua District, Qiqihar 161006, China
| | - Xiaodong Zhang
- Department of Anatomy, Qiqihar Medical University, No. 333, Bukui North Street, Jianhua District, Qiqihar 161006, China
| | - Jun He
- Department of Anatomy, Qiqihar Medical University, No. 333, Bukui North Street, Jianhua District, Qiqihar 161006, China
| | - Yang Jiang
- Department of Anatomy, Qiqihar Medical University, No. 333, Bukui North Street, Jianhua District, Qiqihar 161006, China
| | - Yang Wang
- Department of physiology, Qiqihar Medical University, No. 333, Basic Medical Research Center, Bukui North Street, Jianhua District, Qiqihar 161006, China
| | - Yong Ma
- Department of Anatomy, Qiqihar Medical University, No. 333, Bukui North Street, Jianhua District, Qiqihar 161006, China
| | - Haifeng Jin
- Department of Anatomy, Qiqihar Medical University, No. 333, Bukui North Street, Jianhua District, Qiqihar 161006, China; Basic Medical Research Center, Qiqihar Medical University, No. 333, Bukui North Street, Jianhua District, Qiqihar 161006, China.
| | - Lei Shen
- Department of Anatomy, Qiqihar Medical University, No. 333, Bukui North Street, Jianhua District, Qiqihar 161006, China; Basic Medical Research Center, Qiqihar Medical University, No. 333, Bukui North Street, Jianhua District, Qiqihar 161006, China.
| |
Collapse
|
19
|
Al-Baadani MA, Xu L, Cai K, Yie KHR, Shen Y, Al-Bishari AM, Al-Shaaobi BA, Ma P, Shen X, Liu J. Preparation of co-electrospinning membrane loaded with simvastatin and substance P to accelerate bone regeneration by promoting cell homing, angiogenesis and osteogenesis. Mater Today Bio 2023; 21:100692. [PMID: 37455818 PMCID: PMC10338360 DOI: 10.1016/j.mtbio.2023.100692] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 05/23/2023] [Accepted: 06/05/2023] [Indexed: 07/18/2023] Open
Abstract
Bone regeneration is a complex process that requires the coordination of various biological events. Developing a tissue regeneration membrane that can regulate this cascade of events is challenging. In this study, we aimed to fabricate a membrane that can enrich the damaged area with mesenchymal stem cells, improve angiogenesis, and continuously induce osteogenesis. Our approach involved creating a hierarchical polycaprolactone/gelatin (PCL/GEL) co-electrospinning membrane that incorporated substance P (SP)-loaded GEL fibers and simvastatin (SIM)-loaded PCL fibers. The membrane could initiate a burst release of SP and a slow/sustained release of SIM for over a month. In vitro experiments, including those related to angiogenesis and osteogenesis (e.g., migration, endothelial network formation, alkaline phosphatase activity, mineralization, and gene expression), clearly demonstrated the membrane's superior ability to improve cell homing, revascularization, and osteogenic differentiation. Furthermore, a series of in vivo studies, including immunofluorescence of CD29+/CD90+ double-positive cells and immunohistochemical staining for CD34 and vWF, confirmed the co-electrospinning membrane's ability to enhance MSC migration and revascularization response after five days of implantation. After one month, the Micro-CT and histological (Masson and H&E) results showed accelerated bone regeneration. Our findings suggest that a co-electrospinning membrane with time-tunable drug delivery could advance the development of tissue engineering therapeutic strategies and potentially improve patient outcomes.
Collapse
Affiliation(s)
- Mohammed A. Al-Baadani
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, 325027, People's Republic of China
| | - Lihua Xu
- Department of Stomatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325016, People's Republic of China
| | - Kexin Cai
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, 325027, People's Republic of China
| | - Kendrick Hii Ru Yie
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, 325027, People's Republic of China
| | - Yiding Shen
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, 325027, People's Republic of China
| | - Abdullrahman M. Al-Bishari
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, 325027, People's Republic of China
| | - Bilal A. Al-Shaaobi
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, 325027, People's Republic of China
| | - Pingping Ma
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, 325027, People's Republic of China
| | - Xinkun Shen
- Science and Education Division, The Third Affiliated Hospital of Wenzhou Medical University (Ruian People's Hospital), Wenzhou, 325016, People's Republic of China
| | - Jinsong Liu
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, 325027, People's Republic of China
| |
Collapse
|
20
|
Whitlock AE, Moskowitzova K, Kycia I, Zurakowski D, Fauza DO. Morphometric, Developmental, and Anti-Inflammatory Effects of Transamniotic Stem Cell Therapy (TRASCET) on the Fetal Heart and Lungs in a Model of Intrauterine Growth Restriction. Stem Cells Dev 2023; 32:484-490. [PMID: 37358376 DOI: 10.1089/scd.2023.0040] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/27/2023] Open
Abstract
Transamniotic stem cell therapy (TRASCET) with mesenchymal stem cells (MSCs) can attenuate placental inflammation and minimize intrauterine growth restriction (IUGR). We sought to determine whether MSC-based TRASCET could mitigate fetal cardiopulmonary effects of IUGR. Pregnant Sprague-Dawley dams were exposed to alternating 12-h hypoxia (10.5% O2) cycles in the last fourth of gestation. Their fetuses (n = 155) were divided into 4 groups. One group remained untreated (n = 42), while three groups received volume-matched intra-amniotic injections of either saline (sham; n = 34), or of syngeneic amniotic fluid-derived MSCs, either in their native state (TRASCET; n = 36) or "primed" by exposure to interferon-gamma and interleukin-1beta before administration in vivo (TRASCET-primed; n = 43). Normal fetuses served as additional controls (n = 30). Multiple morphometric and biochemical analyses were performed at term for select markers of cardiopulmonary development and inflammation previously shown to be affected by IUGR. Among survivors (75%; 117/155), fetal heart-to-body weight ratio was increased in both the sham and untreated groups (P < 0.001 for both) but normalized in the TRASCET and TRASCET-primed groups (P = 0.275, 0.069, respectively). Cardiac b-type natriuretic peptide levels were increased in all hypoxia groups compared with normal (P < 0.001), but significantly decreased from sham and untreated in both TRASCET groups (P < 0.0001-0.005). Heart tumor necrosis factor-alpha levels were significantly elevated in sham and TRASCET groups (P = 0.009, 0.002), but normalized in the untreated and TRASCET-primed groups (P = 0.256, 0.456). Lung transforming growth factor-beta levels were significantly increased in both sham and untreated groups (P < 0.001, 0.003), but normalized in both TRASCET groups (P = 0.567, 0.303). Similarly, lung endothelin-1 levels were elevated in sham and untreated groups (P < 0.001 for both), but normalized in both TRASCET groups (P = 0.367, 0.928). We conclude that TRASCET with MSCs decreases markers of fetal cardiac strain, insufficiency, and inflammation, as well as of pulmonary fibrosis and hypertension in the rodent model of IUGR.
Collapse
Affiliation(s)
- Ashlyn E Whitlock
- Department of Surgery, Boston Children's Hospital/Harvard Medical School, Boston, Massachusetts, USA
| | - Kamila Moskowitzova
- Department of Surgery, Boston Children's Hospital/Harvard Medical School, Boston, Massachusetts, USA
| | - Ina Kycia
- Department of Surgery, Boston Children's Hospital/Harvard Medical School, Boston, Massachusetts, USA
| | - David Zurakowski
- Department of Surgery, Boston Children's Hospital/Harvard Medical School, Boston, Massachusetts, USA
| | - Dario O Fauza
- Department of Surgery, Boston Children's Hospital/Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
21
|
Wei J, Xia X, Xiao S, Jin S, Zou Q, Zuo Y, Li Y, Li J. Sequential Dual-Biofactor Release from the Scaffold of Mesoporous HA Microspheres and PLGA Matrix for Boosting Endogenous Bone Regeneration. Adv Healthc Mater 2023; 12:e2300624. [PMID: 36938866 DOI: 10.1002/adhm.202300624] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Indexed: 03/21/2023]
Abstract
The combined design of scaffold structure and multi-biological factors is a prominent strategy to promote bone regeneration. Herein, a composite scaffold of mesoporous hydroxyapatite (HA) microspheres loaded with the bone morphogenetic protein-2 (BMP-2) and a poly(DL-lactic-co-glycolic acid) (PLGA) matrix is constructed by 3D printing. Furthermore, the chemokine stromal cell-derived factor-1α (SDF-1α) is adsorbed on a scaffold surface to achieve the sequential release of the dual-biofactors. The results indicate that the rapid release of SDF-1α chemokine on the scaffold surface effectively recruits bone marrow-derived mesenchymal stem cells (BMSCs) to the target defect area, whereas the long-term sustained release of BMP-2 from the HA microspheres in the degradable PLGA matrix successfully triggers the osteogenic differentiation in the recruited BMSCs, significantly promoting bone regeneration and reconstruction. In addition, these structures/biofactors specially combining scaffold exhibit significantly better biological performance than that of other combined scaffolds, including the bare HA/PLGA scaffold, the scaffold loaded with SDF-1α or BMP-2 biofactor alone, and the scaffold with surface SDF-1α and BMP-2 dual-biofactors. The utilization of mesoporous HA, the assembly method, and sequential release of the two biofactors in the 3D printed composite scaffold present a new method for future design of high-performance bone repairing scaffolds.
Collapse
Affiliation(s)
- Jiawei Wei
- Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu, 610064, P. R. China
| | - Xue Xia
- Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu, 610064, P. R. China
| | - Shiqi Xiao
- Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu, 610064, P. R. China
| | - Shue Jin
- Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu, 610064, P. R. China
| | - Qin Zou
- Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu, 610064, P. R. China
| | - Yi Zuo
- Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu, 610064, P. R. China
| | - Yubao Li
- Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu, 610064, P. R. China
| | - Jidong Li
- Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu, 610064, P. R. China
| |
Collapse
|
22
|
Ahn W, Chi G, Kim S, Son Y, Zhang M. Substance P Reduces Infarct Size and Mortality After Ischemic Stroke, Possibly Through the M2 Polarization of Microglia/Macrophages and Neuroprotection in the Ischemic Rat Brain. Cell Mol Neurobiol 2023; 43:2035-2052. [PMID: 36112332 PMCID: PMC11412183 DOI: 10.1007/s10571-022-01284-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 09/08/2022] [Indexed: 12/12/2022]
Abstract
Substance-P (SP) is an 11 amino acid neuropeptide that is known to stimulate the peripheral mobilization of bone marrow mesenchymal stem cells and M2 polarization in monocytes/macrophages in a variety of acute and chronic tissue injuries. To examine the role of SP in protection and recovery from acute ischemic brain injury, experimental ischemic stroke was induced by transient middle cerebral artery occlusion (tMCAo) in rats for 1 h with subsequent reperfusion. Two injections of SP, immediately and one day post-tMCAo, resulted in approximately threefold lower mortality and 40% less infarct volume than those of saline-treated rats at seven days post-tMCAo. At 4.5 h, SP markedly increased CD11b/c+CD163+/CD 206+ cells in the blood, which were concomitantly decreased in the bone marrow, suggesting that SP preferentially mobilized M2-polarized monocytes. After two days, SP increased the expression of neuroprotective and anti-inflammatory genes in the ischemic brain and induced neuronal survival in the brain penumbra. Additionally, SP markedly increased CD68+CD163+ and CD68+CD206+ M2 microglia/macrophages in the ischemic brain during seven days post-tMCAo. Furthermore, SP preserved the blood‒brain barrier in the ischemic brain, which was confirmed by the abundant levels of SMI71+ brain endothelial cells that colocalized with α-SMA+ pericytes. The beneficial effects of SP on functional recovery and tissue preservation were maintained for six weeks. Collectively, SP treatment in the early phase of ischemic stroke markedly suppressed the destructive inflammatory response and improved the microenvironment for tissue protection and repair.
Collapse
Affiliation(s)
- Woosung Ahn
- Department of Genetics and Biotechnology, College of Life Science and Graduate School of Biotechnology, Kyung Hee University, Seocheon-dong, Kiheung-gu 446-701, Yongin-Si, Republic of Korea
| | - Guangfan Chi
- Department of Genetics and Biotechnology, College of Life Science and Graduate School of Biotechnology, Kyung Hee University, Seocheon-dong, Kiheung-gu 446-701, Yongin-Si, Republic of Korea
| | - Sumin Kim
- Department of Genetics and Biotechnology, College of Life Science and Graduate School of Biotechnology, Kyung Hee University, Seocheon-dong, Kiheung-gu 446-701, Yongin-Si, Republic of Korea
| | - Youngsook Son
- Department of Genetics and Biotechnology, College of Life Science and Graduate School of Biotechnology, Kyung Hee University, Seocheon-dong, Kiheung-gu 446-701, Yongin-Si, Republic of Korea.
| | - Mingzi Zhang
- Department of Genetics and Biotechnology, College of Life Science and Graduate School of Biotechnology, Kyung Hee University, Seocheon-dong, Kiheung-gu 446-701, Yongin-Si, Republic of Korea
| |
Collapse
|
23
|
Garcia Garcia JM, Vannuzzi V, Donati C, Bernacchioni C, Bruni P, Petraglia F. Endometriosis: Cellular and Molecular Mechanisms Leading to Fibrosis. Reprod Sci 2023; 30:1453-1461. [PMID: 36289173 PMCID: PMC10160154 DOI: 10.1007/s43032-022-01083-x] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 09/09/2022] [Indexed: 10/31/2022]
Abstract
Endometriosis is a chronic inflammatory condition affecting women of reproductive age. A relevant feature of endometriosis is the presence of fibrotic tissue inside and around the lesions, thus contributing to the classic endometriosis-related symptoms, pain, and infertility. The molecular mechanisms responsible for the development of fibrosis in endometriosis are not yet defined. The present review aimed to examine the biological mechanisms and signalling pathways involved in fibrogenesis of endometriotic lesions, highlighting the difference between deep infiltrating and ovarian endometriosis. The main cell types involved in the development of fibrosis are platelets, myofibroblasts, macrophages, and sensory nerve fibers. Members of the transforming growth factor (TGF) -β family, as well as the receptor Notch, or the bioactive sphingolipid sphingosine 1-phosphate (S1P), play a role in the development of tissue fibrosis, resulting in their metabolism and/or their signalling pathways altered in endometriotic lesions. It is relevant the knowledge of the molecular mechanisms that guide and support fibrosis in endometriosis, to identify new drug targets and provide new therapeutic approaches to patients.
Collapse
Affiliation(s)
- Jose Manuel Garcia Garcia
- Obstetrics and Gynecology and Molecular Biology, Department of Experimental and Clinical Biomedical Sciences "M. Serio, " University of Florence, Florence, Italy
| | - Valentina Vannuzzi
- Obstetrics and Gynecology and Molecular Biology, Department of Experimental and Clinical Biomedical Sciences "M. Serio, " University of Florence, Florence, Italy
| | - Chiara Donati
- Obstetrics and Gynecology and Molecular Biology, Department of Experimental and Clinical Biomedical Sciences "M. Serio, " University of Florence, Florence, Italy
| | - Caterina Bernacchioni
- Obstetrics and Gynecology and Molecular Biology, Department of Experimental and Clinical Biomedical Sciences "M. Serio, " University of Florence, Florence, Italy
| | - Paola Bruni
- Obstetrics and Gynecology and Molecular Biology, Department of Experimental and Clinical Biomedical Sciences "M. Serio, " University of Florence, Florence, Italy
| | - Felice Petraglia
- Obstetrics and Gynecology and Molecular Biology, Department of Experimental and Clinical Biomedical Sciences "M. Serio, " University of Florence, Florence, Italy.
| |
Collapse
|
24
|
Lee D, Hong HS. Substance P Alleviates Retinal Pigment Epithelium Dysfunction Caused by High Glucose-Induced Stress. Life (Basel) 2023; 13:life13051070. [PMID: 37240715 DOI: 10.3390/life13051070] [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: 04/10/2023] [Accepted: 04/21/2023] [Indexed: 05/28/2023] Open
Abstract
When the retina is constantly affected by high glucose (HG) due to diabetes, the barrier function of the retinal pigment epithelium (RPE) is impaired, accompanied by unnecessary vascularization. This eventually leads to the development of diabetic retinopathy (DR). This study investigated the recovery effect of substance P (SP) on RPE injured by HG. RPE was treated with HG for 24 h, and HG-induced cellular injuries were confirmed. SP was added to the dysfunctional RPE. Compared to RPE in low glucose (LG) conditions, HG-damaged RPE had large, fibrotic cell shapes, and its cellular viability decreased. HG treatment reduced tight junction protein expression levels and caused oxidative stress by interrupting the antioxidant system; this was followed by inflammatory factor intracellular adhesion molecule-1 (ICAM-1), Monocyte chemotactic protein-1 (MCP-1), and angiogenesis factor vascular endothelial growth factor (VEGF) expression. SP treatment contributed to RPE recovery by enhancing cell viability, tight junction protein expression, and RPE function under HG conditions, possibly by activating the Akt signaling pathway. Importantly, SP treatment reduced ICAM-1, MCP-1, and VEGF expression. Collectively, SP activated survival signals to suppress oxidative stress and improve retinal barrier function in RPE, accompanied by immune suppression. This suggests the possible application of SP to diabetic retinal injuries.
Collapse
Affiliation(s)
- Dahyeon Lee
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Hyun Sook Hong
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
- East-West Medical Research Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Kyung Hee Institute of Regenerative Medicine (KIRM), Medical Science Research Institute, Kyung Hee University Medical Center, Seoul 02447, Republic of Korea
| |
Collapse
|
25
|
Kim J, Kim J, Park HJ, Jeon EJ, Cho SW. A microfluidic platform for simulating stem cell migration using in vivo-like gradients of stem cell mobilizer. KOREAN J CHEM ENG 2023. [DOI: 10.1007/s11814-023-1390-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/09/2023]
|
26
|
Moskowitzova K, Fauza DO. Transamniotic stem cell therapy (TRASCET): An emerging minimally invasive strategy for intrauterine stem cell delivery. Semin Perinatol 2023; 47:151728. [PMID: 36990923 DOI: 10.1016/j.semperi.2023.151728] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
Abstract
Transamniotic stem cell therapy (TRASCET) is an emerging strategy for prenatal stem cell therapy involving the least invasive method described to date of delivering select stem cells to virtually any anatomical site in the fetus, including the blood and bone marrow, as well as to fetal annexes, including the placenta. Such broad therapeutic potential derives, to a large extent, from unique routing patterns following stem cell delivery into the amniotic fluid, which have commonalities with naturally occurring fetal cell kinetics. First reported experimentally only less than a decade ago, TRASCET has yet to be attempted clinically, though a first clinical trial appears imminent. Despite significant experimental advances, much promise and perhaps excessive publicity, most cell-based therapies have yet to deliver meaningful large-scale impact to patient care. The few exceptions typically consist of therapies based on the amplification of the normal biological role played by the given cells in their natural environment. Therein lays much of the appeal of TRASCET, in that it, too, is in essence a magnification of naturally occurring processes in the distinctive environment of the maternal-fetal unit. As much as fetal stem cells possess unique characteristics compared with other stem cells, so does the fetus when compared with any other age group, converging into a scenario that enables therapeutic paradigms exclusive to prenatal life. This review summarizes the diversity of applications and biological responses associated with the TRASCET principle.
Collapse
Affiliation(s)
- Kamila Moskowitzova
- Department of Surgery, Boston Children's Hospital and Harvard Medical School, 300 Longwood Avenue - Fegan 3, Boston, MA 02115, USA
| | - Dario O Fauza
- Department of Surgery, Boston Children's Hospital and Harvard Medical School, 300 Longwood Avenue - Fegan 3, Boston, MA 02115, USA.
| |
Collapse
|
27
|
Kokabi F, Ebrahimi S, Mirzavi F, Ghiasi Nooghabi N, Hashemi SF, Hashemy SI. The neuropeptide substance P/neurokinin-1 receptor system and diabetes: From mechanism to therapy. Biofactors 2023. [PMID: 36651605 DOI: 10.1002/biof.1935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 12/22/2022] [Indexed: 01/19/2023]
Abstract
Diabetes is a significant public health issue known as the world's fastest-growing disease condition. It is characterized by persistent hyperglycemia and subsequent chronic complications leading to organ dysfunction and, ultimately, the failure of target organs. Substance P (SP) is an undecapeptide that belongs to the family of tachykinin (TK) peptides. The SP-mediated activation of the neurokinin 1 receptor (NK1R) regulates many pathophysiological processes in the body. There is also a relation between the SP/NK1R system and diabetic processes. Importantly, deregulated expression of SP has been reported in diabetes and diabetes-associated chronic complications. SP can induce both diabetogenic and antidiabetogenic effects and thus affect the pathology of diabetes destructively or protectively. Here, we review the current knowledge of the functional relevance of the SP/NK1R system in diabetes pathogenesis and its exploitation for diabetes therapy. A comprehensive understanding of the role of the SP/NK1R system in diabetes is expected to shed further light on developing new therapeutic possibilities for diabetes and its associated chronic conditions.
Collapse
Affiliation(s)
- Fariba Kokabi
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Safieh Ebrahimi
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Farshad Mirzavi
- Cardiovascular Diseases Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | | | | | - Seyed Isaac Hashemy
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Surgical Oncology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
28
|
Tokumoto M, Nakasa T, Shirakawa Y, Nekomoto A, Ikuta Y, Ishikawa M, Miyaki S, Adachi N. The role of substance P on maintaining ligament homeostasis by inhibiting endochondral ossification during osteoarthritis progression. Connect Tissue Res 2023; 64:82-92. [PMID: 35856812 DOI: 10.1080/03008207.2022.2099847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
PURPOSE Osteoarthritis (OA) is characterized by the degeneration of various tissues, including ligaments. However, pathological changes such as chondrogenesis and ossification in ligaments during OA are still unclear. Substance P (SP), a neuropeptide, has various functions including bone metabolism. This study aimed to analyze the expression and function of SP in OA ligaments, and the therapeutic potential of SP agonists in OA mice. MATERIALS AND METHODS Expressions of SP, SOX9, and MMP13 were histologically analyzed in the posterior cruciate ligament (PCL) in humans with OA and Senescence-accelerated mouse-prone 8 (SAMP8) mice as a spontaneous OA model. The effect of SP agonists on chondrogenesis was evaluated using human ligament cells. Finally, SP agonists were administered intraperitoneally to destabilized medial meniscus (DMM) mice, and the PCL was histologically evaluated. RESULTS In PCL of humans and mice, the expression of SP, SOX9, and MMP13 was upregulated as OA progressed, but their expression was downregulated in severe degeneration. SP and SOX9 were co-expressed in chondrocyte-like cells. In ligament cells, SP agonists downregulated SOX9, RUNX2, and COL10A1. On evaluating chondrogenesis in ligament cells, pellet diameter was reduced in those treated with the SP agonists compared to those untreated. Administration of SP agonists ameliorated PCL degeneration in DMM mice. The Osteoarthritis Research Society and ligament scores in mice with SP agonists were significantly lower than those without SP agonists. CONCLUSIONS SP plays an important role in maintaining ligament homeostasis by inhibiting endochondral ossification during OA progression. Targeting SP has therapeutic potential for preventing ligament degeneration.
Collapse
Affiliation(s)
- Maya Tokumoto
- Department of Orthopaedic Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Tomoyuki Nakasa
- Department of Orthopaedic Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan.,Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima, Japan
| | - Yoshiko Shirakawa
- Department of Orthopaedic Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Akinori Nekomoto
- Department of Orthopaedic Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yasunari Ikuta
- Department of Orthopaedic Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Masakazu Ishikawa
- Department of Orthopaedic Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan.,Department of Artificial Joints and Biomaterials, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Shigeru Miyaki
- Department of Orthopaedic Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan.,Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima, Japan
| | - Nobuo Adachi
- Department of Orthopaedic Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
29
|
Chauhan A, Alam MA, Kaur A, Malviya R. Advancements and Utilizations of Scaffolds in Tissue Engineering and Drug Delivery. Curr Drug Targets 2023; 24:13-40. [PMID: 36221880 DOI: 10.2174/1389450123666221011100235] [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: 01/05/2022] [Revised: 03/02/2022] [Accepted: 03/09/2022] [Indexed: 11/22/2022]
Abstract
The drug development process requires a thorough understanding of the scaffold and its three-dimensional structure. Scaffolding is a technique for tissue engineering and the formation of contemporary functioning tissues. Tissue engineering is sometimes referred to as regenerative medicine. They also ensure that drugs are delivered with precision. Information regarding scaffolding techniques, scaffolding kinds, and other relevant facts, such as 3D nanostructuring, are discussed in depth in this literature. They are specific and demonstrate localized action for a specific reason. Scaffold's acquisition nature and flexibility make it a new drug delivery technology with good availability and structural parameter management.
Collapse
Affiliation(s)
- Akash Chauhan
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, Uttar Pradesh, India
| | - Md Aftab Alam
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, Uttar Pradesh, India
| | - Awaneet Kaur
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, Uttar Pradesh, India
| | - Rishabha Malviya
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, Uttar Pradesh, India
| |
Collapse
|
30
|
Changes in TRPV1 Expression as Well as Substance P and Vasoactive Intestinal Peptide Levels Are Associated with Recurrence of Pterygium. Int J Mol Sci 2022; 23:ijms232415692. [PMID: 36555331 PMCID: PMC9779225 DOI: 10.3390/ijms232415692] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 12/06/2022] [Accepted: 12/07/2022] [Indexed: 12/14/2022] Open
Abstract
Pterygium, a disease of the ocular surface, is characterized by the proliferation and invasion of fibrovascular tissue. Chronic inflammation contributes to pterygium occurrence. Sensory neuropeptides of TRPV1-positive nerve fibers are involved in inflammation and corneal wound healing. The possible association between TRPV1 in nerve fibers and neuropeptides such as Substance P (SP) and Vasoactive Intestinal Peptide (VIP) in the recurrence of pterygium has not been examined before. The pterygia from 64 patients were used to determine changes in SP and VIP levels using 10 min acetic-acid extraction that yielded mainly neuronal peptides. There was a sufficient amount of pterygium tissues from the 35 patients for further immunohistochemical analysis of TRPV1 and S100, which is a glial marker to visualize nerve fibers. SP and VIP levels increased markedly in cases with primary and secondary recurrences, and there was a close correlation between SP and VIP levels. TRPV1 expression increased in the epithelium, while stromal expression decreased in recurrences. Nerve fibers were demonstrated mainly in the stroma, and serial sections confirmed the localization of TRPV1 with the nerve fibers. These results together with previous findings demonstrated that the increased epithelial expression of TRPV1 in recurrent pterygia might be involved in the pathogenesis, and the inhibition of epithelial TRPV1 activity may prevent recurrence.
Collapse
|
31
|
Qin Q, Lee S, Patel N, Walden K, Gomez-Salazar M, Levi B, James AW. Neurovascular coupling in bone regeneration. Exp Mol Med 2022; 54:1844-1849. [PMID: 36446849 PMCID: PMC9722927 DOI: 10.1038/s12276-022-00899-6] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 10/11/2022] [Accepted: 10/11/2022] [Indexed: 11/30/2022] Open
Abstract
The mammalian skeletal system is densely innervated by both neural and vascular networks. Peripheral nerves in the skeleton include sensory and sympathetic nerves. The crosstalk between skeletal and neural tissues is critical for skeletal development and regeneration. The cellular processes of osteogenesis and angiogenesis are coupled in both physiological and pathophysiological contexts. The cellular and molecular regulation of osteogenesis and angiogenesis have yet to be fully defined. This review will provide a detailed characterization of the regulatory role of nerves and blood vessels during bone regeneration. Furthermore, given the importance of the spatial relationship between nerves and blood vessels in bone, we discuss neurovascular coupling during physiological and pathological bone formation. A better understanding of the interactions between nerves and blood vessels will inform future novel therapeutic neural and vascular targeting for clinical bone repair and regeneration.
Collapse
Affiliation(s)
- Qizhi Qin
- grid.21107.350000 0001 2171 9311Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
| | - Seungyong Lee
- grid.260024.20000 0004 0627 4571Department of Physiology, College of Graduate Studies, Midwestern University, Glendale, AZ 85308 USA ,grid.412977.e0000 0004 0532 7395Department of Physical Education, Incheon National University, Incheon, 22012 South Korea
| | - Nirali Patel
- grid.260024.20000 0004 0627 4571Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ 85308 USA
| | - Kalah Walden
- grid.260024.20000 0004 0627 4571Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ 85308 USA
| | - Mario Gomez-Salazar
- grid.21107.350000 0001 2171 9311Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
| | - Benjamin Levi
- grid.267313.20000 0000 9482 7121Departments of Surgery, UT Southwestern Medical Center, Dallas, TX 75390 USA
| | - Aaron W. James
- grid.21107.350000 0001 2171 9311Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
| |
Collapse
|
32
|
Micropattern-based nerve guidance conduit with hundreds of microchannels and stem cell recruitment for nerve regeneration. NPJ Regen Med 2022; 7:62. [PMID: 36261427 PMCID: PMC9582221 DOI: 10.1038/s41536-022-00257-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 10/05/2022] [Indexed: 11/12/2022] Open
Abstract
Guiding the regrowth of thousands of nerve fibers within a regeneration-friendly environment enhances the regeneration capacity in the case of peripheral nerve injury (PNI) and spinal cord injury (SCI). Although clinical treatments are available and several studies have been conducted, the development of nerve guidance conduits (NGCs) with desirable properties, including controllable size, hundreds of nerve bundle-sized microchannels, and host stem-cell recruitment, remains challenging. In this study, the micropattern-based fabrication method was combined with stem-cell recruitment factor (substance P, SP) immobilization onto the main material to produce a size-tunable NGC with hundreds of microchannels with stem-cell recruitment capability. The SP-immobilized multiple microchannels aligned the regrowth of nerve fibers and recruited the host stem cells, which enhanced the functional regeneration capacity. This method has wide applicability in the modification and augmentation of NGCs, such as bifurcated morphology or directional topographies on microchannels. Additional improvements in fabrication will advance the regeneration technology and improve the treatment of PNI/SCI.
Collapse
|
33
|
Gao X, Frakich N, Filippini P, Edwards LJ, Vinkemeier U, Gran B, Tanasescu R, Bayraktutan U, Colombo S, Constantinescu CS. Effects of substance P on human cerebral microvascular endothelial cell line hCMEC/D3 are mediated exclusively through a truncated NK-1 receptor and depend on cell confluence. Neuropeptides 2022; 95:102265. [PMID: 35696961 DOI: 10.1016/j.npep.2022.102265] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 05/01/2022] [Accepted: 06/02/2022] [Indexed: 01/15/2023]
Abstract
The neuropeptide substance P (SP) mediates pain transmission, immune modulation, vasodilation and neurogenic inflammation. Its role in the peripheral nervous system has been well characterised. However, its actions on the blood-brain barrier (BBB) are less clear and warrant further study. The aim of this study was to characterise the effect of SP on the brain microvascular endothelial cells using the immortalized human brain microvascular endothelial cell line hCMEC/D3. As part of our studies, we have evaluated changes in expression, at mRNA and protein levels, of genes involved in the function of the blood-brain barrier such as occludin, induced by exposure to SP. We show that the effect of SP is dependent on cell confluence status. Thus, at low confluence but not at full confluence, SP treatment reduced occludin expression. The expression of the SP receptor, neurokinin-1 receptor (NK-1R) (the truncated form of the receptor expressed exclusively in this cell line) was also modulated in a similar pattern. SP treatment stimulated extracellular signal-regulated kinase (Erk2) phosphorylation which was not associated to changes in Interleukin-6 (IL-6), Interleukin-8 (IL-8), or Intercellular Adhesion Molecule 1 (ICAM-1) protein expression. In addition, SP treatment effectively recovered nitric oxide production on cells exposed to tumour necrosis factor alpha (TNF-α). SP did not trigger intracellular calcium release in hCMEC/D3 cells. We conclude that hCMEC/D3 cells are partially responsive to SP, that the effects are mediated through the truncated form of the receptor and are dependent on the confluence status of these cells.
Collapse
Affiliation(s)
- Xin Gao
- Academic Unit of Mental Health and Clinical Neuroscience, University of Nottingham, Queen's Medical Centre, Nottingham, UK.
| | - Nanci Frakich
- Academic Unit of Mental Health and Clinical Neuroscience, University of Nottingham, Queen's Medical Centre, Nottingham, UK
| | - Perla Filippini
- Academic Unit of Mental Health and Clinical Neuroscience, University of Nottingham, Queen's Medical Centre, Nottingham, UK
| | - Laura J Edwards
- Division of Medical Sciences and Graduate Entry Medicine, Medical School, Royal Derby Hospital, Uttoxeter Road, Derby DE22 3DT, University of Nottingham, UK
| | - Uwe Vinkemeier
- School of Life Science, Action Medical Research Professor of Cell Biology, University of Nottingham, Nottingham, UK
| | - Bruno Gran
- Academic Unit of Mental Health and Clinical Neuroscience, University of Nottingham, Queen's Medical Centre, Nottingham, UK
| | - Radu Tanasescu
- Academic Unit of Mental Health and Clinical Neuroscience, University of Nottingham, Queen's Medical Centre, Nottingham, UK; Department of Neurology, Nottingham University Hospitals NHS Trust, Nottingham, UK
| | - Ulvi Bayraktutan
- Academic Unit of Mental Health and Clinical Neuroscience, University of Nottingham, Queen's Medical Centre, Nottingham, UK
| | - Sergio Colombo
- School of Science & Technology, Nottingham Trent University, Clifton Lane, Nottingham NG11 8NS, UK
| | - Cris S Constantinescu
- Academic Unit of Mental Health and Clinical Neuroscience, University of Nottingham, Queen's Medical Centre, Nottingham, UK; Department of Neurology, Cooper University Hospital, Cooper Neurological Institute, Camden, NJ 08103, USA.
| |
Collapse
|
34
|
V. K. AD, Ray S, Arora U, Mitra S, Sionkowska A, Jaiswal AK. Dual drug delivery platforms for bone tissue engineering. Front Bioeng Biotechnol 2022; 10:969843. [PMID: 36172012 PMCID: PMC9511792 DOI: 10.3389/fbioe.2022.969843] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 08/23/2022] [Indexed: 11/22/2022] Open
Abstract
The dual delivery platforms used in bone tissue engineering provide supplementary bioactive compounds that include distinct medicines and growth factors thereby aiding enhanced bone regeneration. The delivery of these compounds can be adjusted for a short or prolonged time based on the requirement by altering various parameters of the carrier platform. The platforms thus used are fabricated to mimic the niche of the bone microenvironment, either in the form of porous 3D structures, microspheres, or films. Thus, this review article focuses on the concept of dual drug delivery platform and its importance, classification of various platforms for dual drug delivery specific to bone tissue engineering, and finally highlights the foresight into the future direction of these techniques for better clinical applications.
Collapse
Affiliation(s)
- Anupama Devi V. K.
- Tissue Engineering Group, Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology (VIT), Vellore, India
- School of Bio Sciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore, India
| | - Sarbajit Ray
- School of Bio Sciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore, India
| | - Udita Arora
- School of Bio Sciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore, India
| | - Sunrito Mitra
- School of Bio Sciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore, India
| | | | - Amit Kumar Jaiswal
- Tissue Engineering Group, Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology (VIT), Vellore, India
- *Correspondence: Amit Kumar Jaiswal,
| |
Collapse
|
35
|
Xu J, Zhang Z, Zhao J, Meyers CA, Lee S, Qin Q, James AW. Interaction between the nervous and skeletal systems. Front Cell Dev Biol 2022; 10:976736. [PMID: 36111341 PMCID: PMC9468661 DOI: 10.3389/fcell.2022.976736] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 08/08/2022] [Indexed: 11/14/2022] Open
Abstract
The skeleton is one of the largest organ systems in the body and is richly innervated by the network of nerves. Peripheral nerves in the skeleton include sensory and sympathetic nerves. Crosstalk between bones and nerves is a hot topic of current research, yet it is not well understood. In this review, we will explore the role of nerves in bone repair and remodeling, as well as summarize the molecular mechanisms by which neurotransmitters regulate osteogenic differentiation. Furthermore, we discuss the skeleton’s role as an endocrine organ that regulates the innervation and function of nerves by secreting bone-derived factors. An understanding of the interactions between nerves and bone can help to prevent and treat bone diseases caused by abnormal innervation or nerve function, develop new strategies for clinical bone regeneration, and improve patient outcomes.
Collapse
Affiliation(s)
- Jiajia Xu
- Department of Pathology, Johns Hopkins University, Baltimore, MD, United States
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Academy of Orthopedics, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Zhongmin Zhang
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Junjie Zhao
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Carolyn A. Meyers
- Department of Pathology, Johns Hopkins University, Baltimore, MD, United States
| | - Seungyong Lee
- Department of Pathology, Johns Hopkins University, Baltimore, MD, United States
- Department of Physical Education, Incheon National University, Incheon, South Korea
| | - Qizhi Qin
- Department of Pathology, Johns Hopkins University, Baltimore, MD, United States
| | - Aaron W. James
- Department of Pathology, Johns Hopkins University, Baltimore, MD, United States
- *Correspondence: Aaron W. James,
| |
Collapse
|
36
|
Puri S, Kenyon BM, Hamrah P. Immunomodulatory Role of Neuropeptides in the Cornea. Biomedicines 2022; 10:1985. [PMID: 36009532 PMCID: PMC9406019 DOI: 10.3390/biomedicines10081985] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/11/2022] [Accepted: 08/12/2022] [Indexed: 12/21/2022] Open
Abstract
The transparency of the cornea along with its dense sensory innervation and resident leukocyte populations make it an ideal tissue to study interactions between the nervous and immune systems. The cornea is the most densely innervated tissue of the body and possesses both immune and vascular privilege, in part due to its unique repertoire of resident immune cells. Corneal nerves produce various neuropeptides that have a wide range of functions on immune cells. As research in this area expands, further insights are made into the role of neuropeptides and their immunomodulatory functions in the healthy and diseased cornea. Much remains to be known regarding the details of neuropeptide signaling and how it contributes to pathophysiology, which is likely due to complex interactions among neuropeptides, receptor isoform-specific signaling events, and the inflammatory microenvironment in disease. However, progress in this area has led to an increase in studies that have begun modulating neuropeptide activity for the treatment of corneal diseases with promising results, necessitating the need for a comprehensive review of the literature. This review focuses on the role of neuropeptides in maintaining the homeostasis of the ocular surface, alterations in disease settings, and the possible therapeutic potential of targeting these systems.
Collapse
Affiliation(s)
- Sudan Puri
- Center for Translational Ocular Immunology, Tufts Medical Center, Tufts University School of Medicine, Boston, MA 02111, USA
- Department of Ophthalmology, Tufts Medical Center, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Brendan M. Kenyon
- Center for Translational Ocular Immunology, Tufts Medical Center, Tufts University School of Medicine, Boston, MA 02111, USA
- Program in Neuroscience, Graduate School of Biomedical Sciences, Tufts University, Boston, MA 02111, USA
| | - Pedram Hamrah
- Center for Translational Ocular Immunology, Tufts Medical Center, Tufts University School of Medicine, Boston, MA 02111, USA
- Department of Ophthalmology, Tufts Medical Center, Tufts University School of Medicine, Boston, MA 02111, USA
- Program in Neuroscience, Graduate School of Biomedical Sciences, Tufts University, Boston, MA 02111, USA
- Departments of Immunology and Neuroscience, Tufts University School of Medicine, Boston, MA 02111, USA
- Cornea Service, Tufts New England Eye Center, Boston, MA 02111, USA
| |
Collapse
|
37
|
Circulating Osteogenic Progenitor Cells Enhanced with Teriparatide or Denosumab Treatment. J Clin Med 2022; 11:jcm11164749. [PMID: 36012987 PMCID: PMC9409740 DOI: 10.3390/jcm11164749] [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: 06/30/2022] [Revised: 08/10/2022] [Accepted: 08/11/2022] [Indexed: 11/17/2022] Open
Abstract
Circulating osteogenic precursor (COP) cells are peripheral blood cells with a capacity for osteogenesis. The objective of our study was to ascertain the percentage of COPs as an early biomarker of osteoporosis and the effect of these cells in response to Denosumab (DmAb) (anti-resorptive) or to Teriparatide (TPDP) (anabolic) as very effective drugs in the treatment of the illness. A first study was conducted on healthy volunteers, with three age ranges, to determine the percentage of COPs and relate it to their anthropometric and biochemical characteristics, followed by a second longitudinal study on patients with osteoporosis, whereby one group of patients was treated with TPTD and another with DmAb. All were analyzed by cytometry for COP percentage in blood, bone turnover markers, and bone mass. Our findings show that COPs are influenced by age and become more prolific in the stages of growth and skeletal maturation. A higher percentage of COPs is found in osteoporotic disease, which could constitute a predictive marker thereof. We also show how treatment with TPTD or DmAb mobilizes circulating osteogenic precursors in the blood. Significant increases in % COPs were observed after 12 months of treatment with Dmb (21.9%) and TPTD (17%). These results can be related to an increase in osteogenesis and, consequently, a better and more efficient repair of bone tissue.
Collapse
|
38
|
Modulating the tachykinin: Role of substance P and neurokinin receptor expression in ocular surface disorders. Ocul Surf 2022; 25:142-153. [PMID: 35779793 DOI: 10.1016/j.jtos.2022.06.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/21/2022] [Accepted: 06/24/2022] [Indexed: 01/19/2023]
Abstract
Substance P (SP) is a tachykinin expressed by various cells in the nervous and immune systems. SP is predominantly released by neurons and exerts its biological and immunological effects through the neurokinin receptors, primarily the neurokinin-1 receptor (NK1R). SP is essential for maintaining ocular surface homeostasis, and its reduced levels in disorders like diabetic neuropathy disrupt the corneal tissue. It also plays an essential role in promoting corneal wound healing by promoting the migration of keratocytes. In this review, we briefly discuss the structure, expression, and function of SP and its principal receptor NK1R. In addition, SP induces pro-inflammatory effects through autocrine or paracrine action on the immune cells in various ocular surface pathologies, including dry eye disease, herpes simplex virus keratitis, and Pseudomonas keratitis. We provide an in-depth review of the pathogenic role of SP in various ocular surface diseases and several new approaches developed to counter the immune-mediated effects of SP either through modulating its production or blocking its target receptor.
Collapse
|
39
|
Labuz DF, Whitlock AE, Kycia I, Zurakowski D, Fauza DO. Intrauterine Growth Restriction (IUGR) as a potential target for transamniotic stem cell therapy. J Pediatr Surg 2022; 57:999-1003. [PMID: 35277250 DOI: 10.1016/j.jpedsurg.2022.01.062] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 01/31/2022] [Indexed: 12/28/2022]
Abstract
BACKGROUND We sought to determine whether intrauterine growth restriction (IUGR) could be a target for mesenchymal stem cell (MSC)-based transamniotic stem cell therapy (TRASCET). METHODS Pregnant dams subjected to hypoxia (10.5% O2) cycles had their fetuses divided into four groups: untreated (n = 24) and three groups receiving volume-matched intra-amniotic injections of either saline (sham; n = 16), or suspensions of luciferase-labeled, syngeneic amniotic fluid-derived MSCs that were either native (TRASCET-unprimed; n = 29), or primed by exposure to IFNγ and IL-1β (TRASCET-primed; n = 31). Normal fetuses served as additional controls (n = 22). Multiple analyses were performed at term. RESULTS Compared to normal, fetal weights were significantly decreased in all hypoxia groups (p = 0.002 to <0.001), except for TRASCET-primed. Placental efficiency (fetal/placental weight) was significantly decreased in all hypoxia groups (p = 0.002 to <0.001), but normalized in both TRASCET groups. A significant increase in metrial expression of IFNγ in both the untreated and sham groups (p = 0.04 to 0.02) was reversed only in the TRASCET-primed group. Luciferase DNA was present in both TRASCET groups' placentas. CONCLUSIONS Transamniotic stem cell therapy with primed mesenchymal stem cells reverses some of the effects of intrauterine growth restriction in a rat model. Further study into this novel approach for the treatment of this disease is warranted. LEVEL OF EVIDENCE N/A (Animal and Laboratory Study).
Collapse
Affiliation(s)
- Daniel F Labuz
- Department of Surgery, Boston Children's Hospital/Harvard Medical School, Boston, MA, United States of America
| | - Ashlyn E Whitlock
- Department of Surgery, Boston Children's Hospital/Harvard Medical School, Boston, MA, United States of America
| | - Ina Kycia
- Department of Surgery, Boston Children's Hospital/Harvard Medical School, Boston, MA, United States of America
| | - David Zurakowski
- Department of Surgery, Boston Children's Hospital/Harvard Medical School, Boston, MA, United States of America
| | - Dario O Fauza
- Department of Surgery, Boston Children's Hospital/Harvard Medical School, Boston, MA, United States of America.
| |
Collapse
|
40
|
Mahmoudi Z, Farahpour MR. Accelerated wound healing and its promoting effects of topical codeine on the healing of full-thickness cutaneous wound, evidences for modulating cytokines involved in pain, inflammation and collagen biosynthesis. Eur J Trauma Emerg Surg 2022; 48:4735-4744. [PMID: 35612602 DOI: 10.1007/s00068-022-01999-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 05/06/2022] [Indexed: 11/28/2022]
Abstract
INTRODUCTION The inflammation and pain occur in all the wounds. Opioids drugs decrease pain and may act as an anti-inflammation. The current study was conducted to investigate the efficiency of the topical uses of Codeine on full-thickness excision wound models by focusing on relationship between pain mediators, inflammation and wound healing rate. METHODS Following the induction of anesthesia, a skin wound with a size of 7-mm punch was induced on the dorsal surfaces of each mouse. The mice were divided into five categories: groups I-III were daily administered 2.5%, 5%, and 10% Codeine gel; those in group IV were administered phenytoin cream, and group V (controls) received base ointment. To assess the effects of Codeine gel on the wound healing process, the wound area, histological parameters, and the relative protein expression of CXCR1, CXCR2, IL-6, IL-6R, PDGF, PDGFR, and COL1A along with the plasma concentrations of IL-1β, IL-10, and TNF-α were investigated on days 3, 7, and 14. RESULTS On days 7 and 14, the wound area was significantly lower in the treated mice compared to the controls (P < 0.05). Angiogenesis, collagen deposition, and epithelium thickness were significantly higher in the treatment groups compared to the control group (P < 0.05). The relative protein expressions of CXCR1, CXCR2, IL-6, and IL-6R and the plasma concentrations of IL-1β and TNF-α were significantly lower in the treated groups. Meanwhile, the relative protein expressions of PDGF, PDGFR, and COL1A and the plasma concentration of IL-10 were significantly higher in the treated mice (P < 0.05). CONCLUSION Administration of Codeine gel accelerated wound healing through decreasing the pain mediators, inflammation and promoting proliferative phase.
Collapse
Affiliation(s)
- Zhila Mahmoudi
- Department of Basic Sciences, Faculty of Veterinary Medicine, Urmia Branch, Islamic Azad University, Urmia, Iran
| | - Mohammad Reza Farahpour
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Urmia Branch, Islamic Azad University, Urmia, Iran.
| |
Collapse
|
41
|
Erin N, Shurin GV, Baraldi JH, Shurin MR. Regulation of Carcinogenesis by Sensory Neurons and Neuromediators. Cancers (Basel) 2022; 14:2333. [PMID: 35565462 PMCID: PMC9102554 DOI: 10.3390/cancers14092333] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 04/26/2022] [Accepted: 05/05/2022] [Indexed: 12/12/2022] Open
Abstract
Interactions between the immune system and the nervous system are crucial in maintaining homeostasis, and disturbances of these neuro-immune interactions may participate in carcinogenesis and metastasis. Nerve endings have been identified within solid tumors in humans and experimental animals. Although the involvement of the efferent sympathetic and parasympathetic innervation in carcinogenesis has been extensively investigated, the role of the afferent sensory neurons and the neuropeptides in tumor development, growth, and progression is recently appreciated. Similarly, current findings point to the significant role of Schwann cells as part of neuro-immune interactions. Hence, in this review, we mainly focus on local and systemic effects of sensory nerve activity as well as Schwann cells in carcinogenesis and metastasis. Specific denervation of vagal sensory nerve fibers, or vagotomy, in animal models, has been reported to markedly increase lung metastases of breast carcinoma as well as pancreatic and gastric tumor growth, with the formation of liver metastases demonstrating the protective role of vagal sensory fibers against cancer. Clinical studies have revealed that patients with gastric ulcers who have undergone a vagotomy have a greater risk of stomach, colorectal, biliary tract, and lung cancers. Protective effects of vagal activity have also been documented by epidemiological studies demonstrating that high vagal activity predicts longer survival rates in patients with colon, non-small cell lung, prostate, and breast cancers. However, several studies have reported that inhibition of sensory neuronal activity reduces the development of solid tumors, including prostate, gastric, pancreatic, head and neck, cervical, ovarian, and skin cancers. These contradictory findings are likely to be due to the post-nerve injury-induced activation of systemic sensory fibers, the level of aggressiveness of the tumor model used, and the local heterogeneity of sensory fibers. As the aggressiveness of the tumor model and the level of the inflammatory response increase, the protective role of sensory nerve fibers is apparent and might be mostly due to systemic alterations in the neuro-immune response. Hence, more insights into inductive and permissive mechanisms, such as systemic, cellular neuro-immunological mechanisms of carcinogenesis and metastasis formation, are needed to understand the role of sensory neurons in tumor growth and spread.
Collapse
Affiliation(s)
- Nuray Erin
- Department of Medical Pharmacology, Immunopharmacology, and Immuno-Oncology Unit, School of Medicine, Akdeniz University, 07070 Antalya, Turkey
| | - Galina V. Shurin
- Department of Pathology, University of Pittsburgh Medical Center and University of Pittsburgh Cancer Institute, Pittsburgh, 15213 PA, USA; (G.V.S.); (M.R.S.)
| | - James H. Baraldi
- Department of Neuroscience, University of Pittsburgh Medical Center and University of Pittsburgh Cancer Institute, Pittsburgh, 15213 PA, USA;
| | - Michael R. Shurin
- Department of Pathology, University of Pittsburgh Medical Center and University of Pittsburgh Cancer Institute, Pittsburgh, 15213 PA, USA; (G.V.S.); (M.R.S.)
- Department of Immunology, University of Pittsburgh Medical Center and University of Pittsburgh Cancer Institute, Pittsburgh, 15213 PA, USA
| |
Collapse
|
42
|
Williams MAC, Mair DB, Lee W, Lee E, Kim DH. Engineering Three-Dimensional Vascularized Cardiac Tissues. TISSUE ENGINEERING. PART B, REVIEWS 2022; 28:336-350. [PMID: 33559514 PMCID: PMC9063162 DOI: 10.1089/ten.teb.2020.0343] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 02/08/2021] [Indexed: 12/23/2022]
Abstract
Heart disease is one of the largest burdens to human health worldwide and has very limited therapeutic options. Engineered three-dimensional (3D) vascularized cardiac tissues have shown promise in rescuing cardiac function in diseased hearts and may serve as a whole organ replacement in the future. One of the major obstacles in reconstructing these thick myocardial tissues to a clinically applicable scale is the integration of functional vascular networks capable of providing oxygen and nutrients throughout whole engineered constructs. Without perfusion of oxygen and nutrient flow throughout the entire engineered tissue not only is tissue viability compromised, but also overall tissue functionality is lost. There are many supporting technologies and approaches that have been developed to create vascular networks such as 3D bioprinting, co-culturing hydrogels, and incorporation of soluble angiogenic factors. In this state-of-the-art review, we discuss some of the most current engineered vascular cardiac tissues reported in the literature and future directions in the field. Impact statement The field of cardiac tissue engineering is rapidly evolving and is now closer than ever to having engineered tissue models capable of predicting preclinical responses to therapeutics, modeling diseases, and being used as a means of rescuing cardiac function following injuries to the native myocardium. However, a major obstacle of engineering thick cardiac tissue remains to be the integration of functional vasculature. In this review, we highlight seminal and recently published works that have influenced and pushed the field of cardiac tissue engineering toward achieving vascularized functional tissues.
Collapse
Affiliation(s)
| | - Devin B. Mair
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Wonjae Lee
- Department of Neurosurgery, Stanford School of Medicine, Stanford, California, USA
| | - Esak Lee
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, USA
| | - Deok-Ho Kim
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
43
|
Hao Z, Li H, Wang Y, Hu Y, Chen T, Zhang S, Guo X, Cai L, Li J. Supramolecular Peptide Nanofiber Hydrogels for Bone Tissue Engineering: From Multihierarchical Fabrications to Comprehensive Applications. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2103820. [PMID: 35128831 PMCID: PMC9008438 DOI: 10.1002/advs.202103820] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 01/02/2022] [Indexed: 05/03/2023]
Abstract
Bone tissue engineering is becoming an ideal strategy to replace autologous bone grafts for surgical bone repair, but the multihierarchical complexity of natural bone is still difficult to emulate due to the lack of suitable biomaterials. Supramolecular peptide nanofiber hydrogels (SPNHs) are emerging biomaterials because of their inherent biocompatibility, satisfied biodegradability, high purity, facile functionalization, and tunable mechanical properties. This review initially focuses on the multihierarchical fabrications by SPNHs to emulate natural bony extracellular matrix. Structurally, supramolecular peptides based on distinctive building blocks can assemble into nanofiber hydrogels, which can be used as nanomorphology-mimetic scaffolds for tissue engineering. Biochemically, bioactive motifs and bioactive factors can be covalently tethered or physically absorbed to SPNHs to endow various functions depending on physiological and pharmacological requirements. Mechanically, four strategies are summarized to optimize the biophysical microenvironment of SPNHs for bone regeneration. Furthermore, comprehensive applications about SPNHs for bone tissue engineering are reviewed. The biomaterials can be directly used in the form of injectable hydrogels or composite nanoscaffolds, or they can be used to construct engineered bone grafts by bioprinting or bioreactors. Finally, continuing challenges and outlook are discussed.
Collapse
Affiliation(s)
- Zhuowen Hao
- Department of OrthopedicsZhongnan Hospital of Wuhan UniversityDonghu Road 169Wuhan430071China
| | - Hanke Li
- Department of OrthopedicsZhongnan Hospital of Wuhan UniversityDonghu Road 169Wuhan430071China
| | - Yi Wang
- Department of OrthopedicsZhongnan Hospital of Wuhan UniversityDonghu Road 169Wuhan430071China
| | - Yingkun Hu
- Department of OrthopedicsZhongnan Hospital of Wuhan UniversityDonghu Road 169Wuhan430071China
| | - Tianhong Chen
- Department of OrthopedicsZhongnan Hospital of Wuhan UniversityDonghu Road 169Wuhan430071China
| | - Shuwei Zhang
- Department of OrthopedicsZhongnan Hospital of Wuhan UniversityDonghu Road 169Wuhan430071China
| | - Xiaodong Guo
- Department of OrthopedicsUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyJiefang Road 1277Wuhan430022China
| | - Lin Cai
- Department of OrthopedicsZhongnan Hospital of Wuhan UniversityDonghu Road 169Wuhan430071China
| | - Jingfeng Li
- Department of OrthopedicsZhongnan Hospital of Wuhan UniversityDonghu Road 169Wuhan430071China
| |
Collapse
|
44
|
Lasagni Vitar R, Triani F, Barbariga M, Fonteyne P, Rama P, Ferrari G. Substance P/neurokinin-1 receptor pathway blockade ameliorates limbal stem cell deficiency by modulating mTOR pathway and preventing cell senescence. Stem Cell Reports 2022; 17:849-863. [PMID: 35334220 PMCID: PMC9023781 DOI: 10.1016/j.stemcr.2022.02.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 02/18/2022] [Accepted: 02/21/2022] [Indexed: 11/01/2022] Open
Abstract
Severe ocular surface diseases can lead to limbal stem cell deficiency (LSCD), which is accompanied by defective healing. We aimed to evaluate the role of the substance P (SP)/neurokinin-1 receptor (NK1R) pathway in corneal epithelium wound healing in a pre-clinical model of LSCD. SP ablation or NK1R blockade significantly increased epithelial wound healing (p < 0.001) and corneal transparency (p < 0.001), compared with wild type (WT). In addition, a reduced number of infiltrating goblet and conjunctival cells (p < 0.05) and increased number of epithelial stem cells (p < 0.01), which also expressed NK1R, was observed. The mammalian target of rapamycin (mTOR) pathway was significantly inhibited (p < 0.05) and expression of γH2AX was significantly reduced (p < 0.05) after SP ablation. These results suggest that excessive expression of SP is associated with LSCD and results in accelerated senescence and exhaustion of residual stem cells. Topical treatment with NK1R antagonist ameliorates clinical signs associated with LSCD and could be used as an adjuvant treatment in LSCD.
Collapse
Affiliation(s)
- Romina Lasagni Vitar
- Cornea and Ocular Surface Disease Unit, Eye Repair Lab, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy
| | - Francesca Triani
- Cornea and Ocular Surface Disease Unit, Eye Repair Lab, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy
| | - Marco Barbariga
- Cornea and Ocular Surface Disease Unit, Eye Repair Lab, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy
| | - Philippe Fonteyne
- Cornea and Ocular Surface Disease Unit, Eye Repair Lab, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy
| | - Paolo Rama
- Cornea and Ocular Surface Disease Unit, Eye Repair Lab, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy
| | - Giulio Ferrari
- Cornea and Ocular Surface Disease Unit, Eye Repair Lab, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy.
| |
Collapse
|
45
|
Mehta M, Zhao C, Liu A, Innocent C, Kohane DS. Prolonged Retrobulbar Local Anesthesia of the Cornea Does Not Cause Keratopathy in Mice. Transl Vis Sci Technol 2022; 11:33. [PMID: 35072700 PMCID: PMC8802029 DOI: 10.1167/tvst.11.1.33] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose Prolonged local anesthesia (PLA) of the cornea is currently assumed to cause neurotrophic keratitis and is strongly discouraged. We investigate whether PLA of the cornea per se causes neurotrophic keratitis. Methods PLA of the cornea was induced in 12 female albino BALB/c mice by retrobulbar injection of a polymeric prodrug (PGS-TTX) where the site 1 sodium channel blocker tetrodotoxin (TTX) was slowly released from the polymer polyglycerol sebacate. The duration and depth of corneal anesthesia was monitored by the Cochet-Bonnet esthesiometer. Corneal injury from PLA was assessed by slit lamp examination with 2% sodium fluorescein dye, histology, corneal nerve density by immunohistochemistry with anti-β III tubulin antibody and confocal microscopy, and corneal neurotrophin levels (substance P and neurokinin A) by an enzyme-linked immunosorbent assay. PLA was also induced by topical amitriptyline (80 mM), used as a positive control for local anesthetic-induced corneal injury. Frequent ocular lubrication was provided. Results Retrobulbar PGS-TTX resulted in complete corneal anesthesia lasting 50.1 ± 3.6 hours and mean time to complete resolution of block of 55.1 ± 3.6 hours with no keratopathy provided lubrication was provided. Topical 80 mM amitriptyline induced complete corneal anesthesia for 24 hours and developed keratopathy. There was no difference in the histology, levels of corneal neurotrophins, and corneal nerve density between the retrobulbar PGS-TTX group and normal cornea. Conclusions In the absence of topical toxicity or corneal exposure, PLA of the cornea per se does not cause keratitis. Translational Relevance PLA of the cornea could be highly beneficial in acute and chronic painful corneal conditions.
Collapse
Affiliation(s)
- Manisha Mehta
- Laboratory for Biomaterials and Drug Delivery, Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Harvard Institutes of Medicine, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA.,Boston Children's Hospital, Boston, MA, USA
| | - Chao Zhao
- Laboratory for Biomaterials and Drug Delivery, Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Harvard Institutes of Medicine, Boston, MA, USA.,Boston Children's Hospital, Boston, MA, USA
| | - Andong Liu
- Laboratory for Biomaterials and Drug Delivery, Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Harvard Institutes of Medicine, Boston, MA, USA.,Boston Children's Hospital, Boston, MA, USA
| | | | - Daniel S Kohane
- Laboratory for Biomaterials and Drug Delivery, Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Harvard Institutes of Medicine, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA.,Boston Children's Hospital, Boston, MA, USA
| |
Collapse
|
46
|
Merkel MFR, Hellsten Y, Magnusson SP, Kjaer M. Tendon blood flow, angiogenesis, and tendinopathy pathogenesis. TRANSLATIONAL SPORTS MEDICINE 2021. [DOI: 10.1002/tsm2.280] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Max Flemming Ravn Merkel
- Institute of Sports Medicine Department of Orthopedic Surgery Copenhagen University Hospital ‐ Bispebjerg‐Frederiksberg University of Copenhagen Copenhagen Denmark
- Department of Nutrition, Exercise and Sports University of Copenhagen Copenhagen Denmark
| | - Ylva Hellsten
- Department of Nutrition, Exercise and Sports University of Copenhagen Copenhagen Denmark
| | - Stig Peter Magnusson
- Institute of Sports Medicine Department of Orthopedic Surgery Copenhagen University Hospital ‐ Bispebjerg‐Frederiksberg University of Copenhagen Copenhagen Denmark
- Center for Healthy Aging Department of Clinical Medicine University of Copenhagen Copenhagen Denmark
| | - Michael Kjaer
- Institute of Sports Medicine Department of Orthopedic Surgery Copenhagen University Hospital ‐ Bispebjerg‐Frederiksberg University of Copenhagen Copenhagen Denmark
- Center for Healthy Aging Department of Clinical Medicine University of Copenhagen Copenhagen Denmark
| |
Collapse
|
47
|
Ji H, Wang Y, Liu H, Liu Y, Zhang X, Xu J, Li Z, Luo E. Programmed core-shell electrospun nanofibers to sequentially regulate osteogenesis-osteoclastogenesis balance for promoting immediate implant osseointegration. Acta Biomater 2021; 135:274-288. [PMID: 34492371 DOI: 10.1016/j.actbio.2021.08.050] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 08/06/2021] [Accepted: 08/29/2021] [Indexed: 02/05/2023]
Abstract
The biology of immediate post-extraction implant osseointegration is mediated by a coordinated cascade of osteoblast-osteoclast interactions. The aim of this study was to develop a dual-delivery system that allowed sequential release of substance P (SP) to promote bone regeneration and alendronate (ALN) to reduce bone resorption, which will improve the implant osseointegration. We used coaxial electrospinning to fabricate the core-shell poly lactic-co-glycolic acid (PLGA)/gelatin nanofibers, which consists of SP in the shell and ALN in the core. This programmed delivery system was shown to release SP and ALN sequentially to match the spatio-temporal specificity of bone healing. The migration assay demonstrated that the SP-ALN dual-delivery system increased bone marrow mesenchymal stem cells (BMSCs) transmigration. Besides, the expression of osteogenic/osteoclastic markers, Alizarin Red staining, tartrate-resistant acid phosphatase (TRAP) staining, F-actin staining and bone resorption experiment showed that the dual-delivery system can render a microenvironment favorable for osteogenic differentiation and adverse to osteoclastogenesis. Using a rat immediate implant model, we validated the promoted osteogenic property and osseointegration around the implants of SP-ALN dual-delivery system by micro-computed tomography (micro-CT) and histological analysis. These findings suggest that the dual-delivery system with time-controlled release of SP and ALN by core-shell nanofibers provides a promising strategy to facilitate immediate implant osseointegration through favorable osteogenesis. STATEMENT OF SIGNIFICANCE: Immediate implant placement is potentially challenged by the difficulties in achieving primary implant stability and early osteogenesis. Initial period of osteointegration is regulated by osteoblastic/osteoclastic cells resulting in a coordinated healing process. To have an efficient bone regeneration, the coaxial electrospinning was used to fabricate a programmed dual-delivery system. The SP released rapidly and favored for BMSCs migration and osteogenic differentiation, while the sustained release of ALN can reduce the bone resorption. The rat immediate implant model indicated that the SP-ALN dual-delivery system could present the promoted peri‑implant osteogenic property and osseointegration through modulating the osteogenesis-osteoclastogenesis balance. This work highlights the sequential dual delivery of SP and ALN has a promising potential of achieving enhanced osseointegration for immediate implant placement.
Collapse
Affiliation(s)
- Huanzhong Ji
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, No. 14 Section 3, Renmin South Road, Chengdu, Sichuan 610041, PR China
| | - Yiyao Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, No. 14 Section 3, Renmin South Road, Chengdu, Sichuan 610041, PR China; Department of Oral and Maxillofacial Surgery, Sichuan Hospital of Stomatology, Chengdu 610031, PR China
| | - Hanghang Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, No. 14 Section 3, Renmin South Road, Chengdu, Sichuan 610041, PR China
| | - Yao Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, No. 14 Section 3, Renmin South Road, Chengdu, Sichuan 610041, PR China
| | - Xiaohui Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, No. 14 Section 3, Renmin South Road, Chengdu, Sichuan 610041, PR China
| | - Jiazhuang Xu
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, PR China
| | - Zhongming Li
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, PR China
| | - En Luo
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, No. 14 Section 3, Renmin South Road, Chengdu, Sichuan 610041, PR China.
| |
Collapse
|
48
|
Torrecillas-Baena B, Gálvez-Moreno MÁ, Quesada-Gómez JM, Dorado G, Casado-Díaz A. Influence of Dipeptidyl Peptidase-4 (DPP4) on Mesenchymal Stem-Cell (MSC) Biology: Implications for Regenerative Medicine - Review. Stem Cell Rev Rep 2021; 18:56-76. [PMID: 34677817 DOI: 10.1007/s12015-021-10285-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/12/2021] [Indexed: 12/16/2022]
Abstract
Dipeptidyl peptidase IV (DPP4) is a ubiquitous protease that can be found in membrane-anchored or soluble form. Incretins are one of the main DPP4 substrates. These hormones regulate glucose levels, by stimulating insulin secretion and decreasing glucagon production. Because DPP4 levels are high in diabetes, DPP4 inhibitor (DPP4i) drugs derived from gliptin are widespread used as hypoglycemic agents for its treatment. However, as DPP4 recognizes other substrates such as chemokines, growth factors and neuropeptides, pleiotropic effects have been observed in patients treated with DPP4i. Several of these substrates are part of the stem-cell niche. Thus, they may affect different physiological aspects of mesenchymal stem-cells (MSC). They include viability, differentiation, mobilization and immune response. MSC are involved in tissue homeostasis and regeneration under both physiological and pathological conditions. Therefore, such cells and their secretomes have a high clinical potential in regenerative medicine. In this context, DPP4 activity may modulate different aspects of MSC regenerative capacity. Therefore, the aim of this review is to analyze the effect of different DPP4 substrates on MSC. Likewise, how the regulation of DPP4 activity by DPP4i can be applied in regenerative medicine. That includes treatment of cardiovascular and bone pathologies, cutaneous ulcers, organ transplantation and pancreatic beta-cell regeneration, among others. Thus, DPP4i has an important clinical potential as a complement to therapeutic strategies in regenerative medicine. They involve enhancing the differentiation, immunomodulation and mobilization capacity of MSC for regenerative purposes.
Collapse
Affiliation(s)
- Bárbara Torrecillas-Baena
- Unidad de Gestión Clínica de Endocrinología y Nutrición - GC17, Instituto Maimónides de Investigación Biomédica de Córdoba, Hospital Universitario Reina Sofía, CIBERFES, Avda. Menéndez Pidal s/n, 14004, Córdoba, Spain
| | - María Ángeles Gálvez-Moreno
- Unidad de Gestión Clínica de Endocrinología y Nutrición - GC17, Instituto Maimónides de Investigación Biomédica de Córdoba, Hospital Universitario Reina Sofía, CIBERFES, Avda. Menéndez Pidal s/n, 14004, Córdoba, Spain
| | - José Manuel Quesada-Gómez
- Unidad de Gestión Clínica de Endocrinología y Nutrición - GC17, Instituto Maimónides de Investigación Biomédica de Córdoba, Hospital Universitario Reina Sofía, CIBERFES, Avda. Menéndez Pidal s/n, 14004, Córdoba, Spain
| | - Gabriel Dorado
- Dep. Bioquímica y Biología Molecular, Campus Rabanales C6-1-E17, Campus de Excelencia Internacional Agroalimentario (ceiA3), Universidad de Córdoba, CIBERFES, 14071, Córdoba, Spain
| | - Antonio Casado-Díaz
- Unidad de Gestión Clínica de Endocrinología y Nutrición - GC17, Instituto Maimónides de Investigación Biomédica de Córdoba, Hospital Universitario Reina Sofía, CIBERFES, Avda. Menéndez Pidal s/n, 14004, Córdoba, Spain.
| |
Collapse
|
49
|
Shirakawa Y, Nakasa T, Kanemitsu M, Nekomoto A, Ishikawa M, Yimiti D, Miyaki S, Adachi N. Therapeutic effect of targeting Substance P on the progression of osteoarthritis. Mod Rheumatol 2021; 32:1175-1185. [PMID: 34915568 DOI: 10.1093/mr/roab089] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 06/23/2021] [Accepted: 09/29/2021] [Indexed: 12/13/2022]
Abstract
OBJECTIVES Substance P (SP) modulates NK1 and has various functions such as regulation of pain response, bone metabolism, and angiogenesis, which are recognized as important factors in osteoarthritis (OA). We aimed to evaluate the therapeutic effect of targeting SP on OA progression. METHODS SP expression patterns were analysed histologically in articular cartilage and subchondral bone of human knees from OA patients and autopsy donors as non-OA samples and in mouse articular cartilage. Moreover, to examine the effect of SP on the progression of OA, we administered drugs to mice following the surgical destabilization of the medial meniscus: Phosphate-buffered saline (PBS), septide (NK1 receptor agonist), or aprepitant (NK1 receptor antagonist). Histological analysis and bone morphologic analysis using micro-computed tomography were performed. RESULTS In human analysis, the expression of SP in mild OA samples was significantly higher than that in severe OA, and that in healthy cartilage was significantly higher than that in OA. In mouse analysis, Osteoarthritis Research Society International scores in the septide group were significantly lower than those in the control group. Computed tomography analysis showed that the subchondral bone's epiphysis in the control group had sclerotic change, not observed in the septide group. CONCLUSIONS The administration of septide ameliorates OA progression through preventing subchondral bone sclerosis.
Collapse
Affiliation(s)
- Yoshiko Shirakawa
- Department of Orthopaedic Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Tomoyuki Nakasa
- Department of Orthopaedic Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
- Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima, Japan
| | - Munekazu Kanemitsu
- Department of Orthopaedic Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Akinori Nekomoto
- Department of Orthopaedic Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Masakazu Ishikawa
- Department of Orthopaedic Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Dilimulati Yimiti
- Department of Orthopaedic Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Shigeru Miyaki
- Department of Orthopaedic Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
- Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima, Japan
| | - Nobuo Adachi
- Department of Orthopaedic Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
50
|
Park SH, Ju HJ, Ji YB, Shah M, Min BH, Choi HS, Choi S, Kim MS. Endogenous Stem Cell-Based In Situ Tissue Regeneration Using Electrostatically Interactive Hydrogel with a Newly Discovered Substance P Analog and VEGF-Mimicking Peptide. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2103244. [PMID: 34480409 DOI: 10.1002/smll.202103244] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 07/22/2021] [Indexed: 06/13/2023]
Abstract
The use of chemoattractants to promote endogenous stem cell-based in situ tissue regeneration has recently garnered much attention. This study is the first to assess the endogenous stem cell migration using a newly discovered substance P (SP) analog (SP1) by molecular dynamics simulations as an efficient chemoattractant. Further, a novel strategy based on electrostatic interaction using cationic chitosan (Ch) and anionic hyaluronic acid (HA) to prepare an SP1-loaded injectable C/H formulation without SP1 loss is developed. The formulation quickly forms an SP1-loaded C/H hydrogel in situ through in vivo injection. The newly discovered SP1 is found to possess human mesenchymal stromal cells (hMSCs) migration-inducing ability that is approximately two to three times higher than that of the existing SP. The designed VEGF-mimicking peptide (VP) chemically reacts with the hydrogel (C/H-VP) to sustain the release of VP, thus inducing vasculogenic differentiation of the hMSCs that migrate toward the C/H-VP hydrogel. Similarly, in animal experiments, SP1 attracts a large number of hMSCs toward the C/H-VP hydrogel, after which VP induces vasculogenic differentiation. Collectively, these findings indicate that SP1-loaded C/H-VP hydrogels are a promising strategy to facilitate endogenous stem cell-based in situ tissue regeneration.
Collapse
Affiliation(s)
- Seung Hun Park
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, Korea
| | - Hyeon Jin Ju
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, Korea
| | - Yun Bae Ji
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, Korea
| | - Masaud Shah
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, Korea
| | - Byoung Hyun Min
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, Korea
| | - Hak Soo Choi
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Sangdun Choi
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, Korea
| | - Moon Suk Kim
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, Korea
- Medipolymers, Research Institute, Woncheon Dong 332-2, Suwon, 16522, Korea
| |
Collapse
|