1
|
Hubertus V, Meyer L, Waldmann L, Roolfs L, Taheri N, Kersting K, von Bronewski E, Nieminen-Kelhä M, Kremenetskaia I, Uhl C, Fiedler KC, Ode JE, Rex A, Prüß H, Rakhymzhan A, Hauser AE, Niesner R, Heppner FL, Fehlings MG, Vajkoczy P. Identification of a Therapeutic Window for Neurovascular Unit Repair after Experimental Spinal Cord Injury. J Neurotrauma 2024. [PMID: 39585767 DOI: 10.1089/neu.2024.0233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2024] Open
Abstract
Traumatic spinal cord injury (SCI) is a devastating condition for which effective neuroregenerative and neuroreparative strategies are lacking. The post-traumatic disruption of the blood-spinal cord barrier (BSCB) as part of the neurovascular unit (NVU) is one major factor in the complex pathophysiology of SCI, which is associated with edema, inflammation, and cell death in the penumbra regions of the spinal cord adjacent to the lesion epicenter. Thus, the preservation of an intact NVU and vascular integrity to facilitate the regenerative capacity following SCI is a desirable therapeutic target. This study aims to identify a therapeutic window of opportunity for NVU repair after SCI by characterizing the timeframe of its post-traumatic disintegration and reintegration with implications for functional spinal cord recovery. Following thoracic clip-compression SCI or sham injury, adult C57BL/6J mice were followed up from one to 28 days. At one, three, seven, 14, and 28 days after SCI/sham, seven-Tesla magnetic resonance imaging (MRI), neurobehavioral analysis (Basso mouse scale, Tally subscore, CatWalk® gait analysis), and following sacrifice immunohistochemistry were performed, assessing vessel permeability via Evans blue (EVB) extravasation, (functional) vessel density, and NVU integrity. Thy1-yellow fluorescent protein+ mice were additionally implanted with a customized spinal window chamber and received longitudinal in vivo two-photon excitation imaging (2PM) with the injection of rhodamine-B-isothiocyanate-dextran for the combined imaging of axons and vasculature up to 14 days after SCI/sham injury. Post-traumatic edema formation as assessed by MRI volumetry peaked at one to three days after injury, while EVB permeability quantification revealed a thoroughly injured BSCB up to 14 days after SCI. Partial regeneration of functional vasculature via endogenous revascularization was detected after one to four weeks, however, with only 50-54% of existing vessels regaining functional perfusion. Longitudinal in vivo 2PM visualized the progressive degeneration of initially preserved spinal cord axons in the peri-traumatic zone after SCI while displaying a rarefication of functionally perfused vessels up to two weeks after injury. Neurobehavioral recovery started after one week but remained impaired over the whole observation period of four weeks after SCI. With this study, a therapeutic window to address the impaired NVU starting from the first days to two weeks after SCI is identified. A number of lines of evidence including in vivo 2PM, assessment of NVU integrity, and neurobehavioral assessments point to the critical nature of targeting the NVU to enhance axonal preservation and regeneration after SCI. Continuous multifactorial therapy applications targeting the integrity of the NVU over the identified therapeutic window of opportunity appears promising to ameliorate functional vessel perseverance and the spinal cord's regenerative capacity.
Collapse
Affiliation(s)
- Vanessa Hubertus
- Department of Neurosurgery, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Lea Meyer
- Department of Neurosurgery, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Lilly Waldmann
- Department of Neurosurgery, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Laurens Roolfs
- Department of Neurosurgery, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Nima Taheri
- Department of Neurosurgery, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Katharina Kersting
- Department of Neurosurgery, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Emily von Bronewski
- Department of Neurosurgery, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Melina Nieminen-Kelhä
- Department of Neurosurgery, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Irina Kremenetskaia
- Department of Neurosurgery, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Christian Uhl
- Department of Neurosurgery, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Kim C Fiedler
- Department of Neurosurgery, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Jan-Erik Ode
- Scientific Workshops, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Andre Rex
- Department of Experimental Neurology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Harald Prüß
- Department of Experimental Neurology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
| | - Asylkhan Rakhymzhan
- Deutsches Rheuma-Forschungszentrum, A Leibniz Institute, Berlin, Germany and Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Anja E Hauser
- Deutsches Rheuma-Forschungszentrum, A Leibniz Institute, Berlin, Germany and Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Raluca Niesner
- Deutsches Rheuma-Forschungszentrum, A Leibniz Institute, Berlin, Germany and Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Frank L Heppner
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Department of Neuropathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Cluster of Excellence, NeuroCure, Berlin, Germany
| | - Michael G Fehlings
- Division of Neurosurgery and Krembil Neuroscience Centre, Toronto Western Hospital, University Health Network and University of Toronto, Toronto, Canada
| | - Peter Vajkoczy
- Department of Neurosurgery, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
2
|
Zhang N, Hu J, Liu W, Cai W, Xu Y, Wang X, Li S, Ru B. Advances in Novel Biomaterial-Based Strategies for Spinal Cord Injury Treatment. Mol Pharm 2024; 21:4764-4785. [PMID: 39235393 DOI: 10.1021/acs.molpharmaceut.3c01104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/06/2024]
Abstract
Spinal cord injury (SCI) is a highly disabling neurological disorder. Its pathological process comprises an initial acute injury phase (primary injury) and a secondary injury phase (subsequent chronic injury). Although surgical, drug, and cell therapies have made some progress in treating SCI, there is no exact therapeutic strategy for treating SCI and promoting nerve regeneration due to the complexity of the pathological SCI process. The development of novel drug delivery systems to treat SCI is expected to significantly impact the individualized treatment of SCI due to its unique and excellent properties, such as active targeting and controlled release. In this review, we first describe the pathological progression of the SCI response, including primary and secondary injuries. Next, we provide a concise overview of newly developed nanoplatforms and their potential application in regulating and treating different pathological processes of SCI. Then, we introduce the existing potential problems and future clinical application perspectives of biomedical engineering-based therapies for SCI.
Collapse
Affiliation(s)
- Nannan Zhang
- Center for Rehabilitation Medicine, Department of Pain Management, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 330004, China
| | - Jiaqi Hu
- Center for Rehabilitation Medicine, Department of Pain Management, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 330004, China
| | - Wenlong Liu
- Center for Rehabilitation Medicine, Department of Pain Management, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 330004, China
| | - Wenjun Cai
- Center for Rehabilitation Medicine, Department of Pain Management, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 330004, China
| | - Yun Xu
- Center for Rehabilitation Medicine, Department of Pain Management, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 330004, China
| | - Xiaojuan Wang
- Department of Clinical Pharmacy, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Shun Li
- Center for Rehabilitation Medicine, Department of Pain Management, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 330004, China
| | - Bin Ru
- Center for Rehabilitation Medicine, Department of Pain Management, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 330004, China
| |
Collapse
|
3
|
Bennett HC, Zhang Q, Wu YT, Manjila SB, Chon U, Shin D, Vanselow DJ, Pi HJ, Drew PJ, Kim Y. Aging drives cerebrovascular network remodeling and functional changes in the mouse brain. Nat Commun 2024; 15:6398. [PMID: 39080289 PMCID: PMC11289283 DOI: 10.1038/s41467-024-50559-8] [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: 06/09/2023] [Accepted: 07/15/2024] [Indexed: 08/02/2024] Open
Abstract
Aging is frequently associated with compromised cerebrovasculature and pericytes. However, we do not know how normal aging differentially impacts vascular structure and function in different brain areas. Here we utilize mesoscale microscopy methods and in vivo imaging to determine detailed changes in aged murine cerebrovascular networks. Whole-brain vascular tracing shows an overall ~10% decrease in vascular length and branching density with ~7% increase in vascular radii in aged brains. Light sheet imaging with 3D immunolabeling reveals increased arteriole tortuosity of aged brains. Notably, vasculature and pericyte densities show selective and significant reductions in the deep cortical layers, hippocampal network, and basal forebrain areas. We find increased blood extravasation, implying compromised blood-brain barrier function in aged brains. Moreover, in vivo imaging in awake mice demonstrates reduced baseline and on-demand blood oxygenation despite relatively intact neurovascular coupling. Collectively, we uncover regional vulnerabilities of cerebrovascular network and physiological changes that can mediate cognitive decline in normal aging.
Collapse
Affiliation(s)
- Hannah C Bennett
- Department of Neural and Behavioral Sciences, The Pennsylvania State University, Hershey, PA, 17033, USA
| | - Qingguang Zhang
- Center for Neural Engineering, Department of Engineering Science and Mechanics, The Pennsylvania State University, University Park, PA, 16802, USA
- Department of Physiology, Michigan State University, East Lansing, MI, 48824, USA
| | - Yuan-Ting Wu
- Department of Neural and Behavioral Sciences, The Pennsylvania State University, Hershey, PA, 17033, USA
- Department of Neurosurgery, Department of Computational Biomedicine, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Steffy B Manjila
- Department of Neural and Behavioral Sciences, The Pennsylvania State University, Hershey, PA, 17033, USA
| | - Uree Chon
- Department of Neural and Behavioral Sciences, The Pennsylvania State University, Hershey, PA, 17033, USA
- Neurosciences Graduate Program, Stanford University, Stanford, CA, 94305, USA
| | - Donghui Shin
- Department of Neural and Behavioral Sciences, The Pennsylvania State University, Hershey, PA, 17033, USA
| | - Daniel J Vanselow
- Department of Neural and Behavioral Sciences, The Pennsylvania State University, Hershey, PA, 17033, USA
| | - Hyun-Jae Pi
- Department of Neural and Behavioral Sciences, The Pennsylvania State University, Hershey, PA, 17033, USA
| | - Patrick J Drew
- Center for Neural Engineering, Department of Engineering Science and Mechanics, The Pennsylvania State University, University Park, PA, 16802, USA
- Department of Biomedical Engineering, Biology, and Neurosurgery, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Yongsoo Kim
- Department of Neural and Behavioral Sciences, The Pennsylvania State University, Hershey, PA, 17033, USA.
- Center for Neural Engineering, Department of Engineering Science and Mechanics, The Pennsylvania State University, University Park, PA, 16802, USA.
| |
Collapse
|
4
|
Kim HW, Wu KLK, Tam KW, Chan YS, Shum DKY. Pericyte derivation and transplantation for blood-CNS barrier reconstitution in CNS disorders. IBRO Neurosci Rep 2024; 16:147-154. [PMID: 39007089 PMCID: PMC11240299 DOI: 10.1016/j.ibneur.2023.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 12/09/2023] [Accepted: 12/29/2023] [Indexed: 07/16/2024] Open
Abstract
Disruption of the blood-central nervous system barrier (BCB) is increasingly recognized as a pathological factor in diseases and trauma of the central nervous system. Despite the neuropathological impact, current treatment modalities do not target the BCB; strategies to reconstitute the impaired BCB have been restricted to nutritional and dietary remedies. As an integral cell type in the neurovascular unit, pericytes are crucial to the development, maintenance, and repair of the BCB. As such, pericytes are well poised as cellular agents for reconstitution of the impaired BCB. Here, we summarize recent revelations regarding the role of BCB disruption in diseases and trauma of the central nervous system and highlight how pericytes are harnessed to provide targeted therapeutic effect in each case. This review will also address how recent advances in pericyte derivation strategies can serve to overcome practical hurdles in the clinical use of pericytes.
Collapse
Affiliation(s)
- Hyun Woo Kim
- Department of Orthopedics and Traumatology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Kenneth Lap Kei Wu
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Kin-Wai Tam
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Ying-Shing Chan
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- State Key Laboratory for Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong SAR, China
| | - Daisy Kwok-Yan Shum
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- State Key Laboratory for Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
5
|
Xu F, Tian Z, Wang Z. Cilostazol protects against degenerative cervical myelopathy injury and cell pyroptosis via TXNIP-NLRP3 pathway. Cell Div 2024; 19:2. [PMID: 38233884 DOI: 10.1186/s13008-024-00108-y] [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: 11/19/2023] [Accepted: 01/11/2024] [Indexed: 01/19/2024] Open
Abstract
Degenerative cervical myelopathy (DCM) is one of the most common and serious neurological diseases. Cilostazol has protective effects of anterior horn motor neurons and prevented the cell apoptosis. However, there was no literatures of Cilostazol on DCM. In this study, we established the DCM rat model to detect the effects of Cilostazol. Meanwhile, the neurobehavioral assessments, histopathology changes, inflammatory cytokines, Thioredoxin-interacting protein (TXNIP), NOD‑like receptor pyrin domain containing 3 (NLRP3) and pro-caspase-1 expressions were detected by Basso, Beattie, and Bresnahan score assessment, Hematoxylin and Eosin Staining, Enzyme-linked immunosorbent assay, immunofluorescence and Western blotting, respectively. After treated with Cilostazol, the Basso, Beattie, and Bresnahan (BBB) score, inclined plane test and forelimb grip strength in DCM rats were significantly increased meanwhile the histopathology injury and inflammatory cytokines were decreased. Additionally, TXNIP, NLRP3 and pro-caspase-1 expressions levels were decreased in Cilostazol treated DCM rats. Interestingly, the using of siTXNIP significantly changed inflammatory cytokines, TXNIP, NLRP3 and pro-caspase-1 expressions, however there was no significance between siTXNIP and Cilostazol + siTXNIP group. These observations showed that Cilostazol rescues DCM injury and ameliorates neuronal destruction mediated by TXNIP/NLRP3/caspase-1 and pro-inflammatory cytokines. As a result of our study, these findings provide further evidence that Cilostazol may represent promising therapeutic candidates for DCM.
Collapse
Affiliation(s)
- Fei Xu
- Department of Neck-Shoulder and Lumbocrural Pain, Yantai hospital of traditional Chinese medicine, 39 Xingfu Road, Zhifu District, Yantai, 264000, Shandong, P.R. China
| | - Zhuo Tian
- Department of General Surgery, Yantai hospital of traditional Chinese medicine, Yantai, Shandong, China
| | - Zhengguang Wang
- Department of Neck-Shoulder and Lumbocrural Pain, Yantai hospital of traditional Chinese medicine, 39 Xingfu Road, Zhifu District, Yantai, 264000, Shandong, P.R. China.
| |
Collapse
|
6
|
Yan Q, Xun Y, Lei D, Zhai H. Tanshinone IIA protects motor neuron-like NSC-34 cells against lipopolysaccharide-induced cell injury by the regulation of the lncRNA TCTN2/miR-125a-5p/DUSP1 axis. Regen Ther 2023; 24:417-425. [PMID: 37727797 PMCID: PMC10506057 DOI: 10.1016/j.reth.2023.03.007] [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: 06/20/2022] [Revised: 03/10/2023] [Accepted: 03/31/2023] [Indexed: 09/21/2023] Open
Abstract
Background Tanshinone IIA (TSIIA) exerts a protective role in spinal cord injury (SCI). However, the mechanism of TSIIA activity in SCI remains to be elucidated. Methods Cell viability and apoptosis were gauged by CCK-8 assay and flow cytometry, respectively. The expression levels of lncRNA TCTN2, miR-125a-5p and DUSP1 were detected by qRT-PCR and western blot. Direct relationship between miR-125a-5p and TCTN2 or DUSP1 was verified by dual-luciferase reporter assay. Results In mouse NSC-34 cells, LPS reduced the expression of TCTN2. TSIIA alleviated cell injury induced by LPS and increased TCTN2 expression in LPS-exposed NSC-34 cells. TCTN2 was a downstream mediator of TSIIA activity. TCTN2 targeted miR-125a-5p, and TCTN2 over-expression attenuated LPS-induced cell damage in NSC-34 cells by down-regulating miR-125a-5p. TCTN2 functioned as a post-transcriptional regulator of DUSP1 expression through miR-125a-5p. DUSP1 was a functional target of miR-125a-5p in controlling NSC-34 cell injury induced by LPS. TSIIA inhibited miR-125a-5p expression and increased the level of DUSP1 protein in LPS-exposed NSC-34 cells. Conclusion Our study establishes a novel mechanism, the TCTN2/miR-125a-5p/DUSP1 axis, at least in part, for the protective activity of TSIIA in cell injury induced by LPS.
Collapse
Affiliation(s)
| | | | - Debao Lei
- Department of Rehabilitation Medicine, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang City, 441000, Hubei, China
| | - Hongyu Zhai
- Department of Rehabilitation Medicine, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang City, 441000, Hubei, China
| |
Collapse
|
7
|
Fu J, Liang H, Yuan P, Wei Z, Zhong P. Brain pericyte biology: from physiopathological mechanisms to potential therapeutic applications in ischemic stroke. Front Cell Neurosci 2023; 17:1267785. [PMID: 37780206 PMCID: PMC10536258 DOI: 10.3389/fncel.2023.1267785] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 08/30/2023] [Indexed: 10/03/2023] Open
Abstract
Pericytes play an indispensable role in various organs and biological processes, such as promoting angiogenesis, regulating microvascular blood flow, and participating in immune responses. Therefore, in this review, we will first introduce the discovery and development of pericytes, identification methods and functional characteristics, then focus on brain pericytes, on the one hand, to summarize the functions of brain pericytes under physiological conditions, mainly discussing from the aspects of stem cell characteristics, contractile characteristics and paracrine characteristics; on the other hand, to summarize the role of brain pericytes under pathological conditions, mainly taking ischemic stroke as an example. Finally, we will discuss and analyze the application and development of pericytes as therapeutic targets, providing the research basis and direction for future microvascular diseases, especially ischemic stroke treatment.
Collapse
Affiliation(s)
- Jiaqi Fu
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
- Department of Neurology, Shidong Hospital, Yangpu District, Shanghai, China
| | - Huazheng Liang
- Monash Suzhou Research Institute, Suzhou, Jiangsu, China
| | - Ping Yuan
- Department of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhenyu Wei
- Department of Neurology, Shidong Hospital, Yangpu District, Shanghai, China
| | - Ping Zhong
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
- Department of Neurology, Shidong Hospital, Yangpu District, Shanghai, China
| |
Collapse
|
8
|
Bennett HC, Zhang Q, Wu YT, Chon U, Pi HJ, Drew PJ, Kim Y. Aging drives cerebrovascular network remodeling and functional changes in the mouse brain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.23.541998. [PMID: 37305850 PMCID: PMC10257218 DOI: 10.1101/2023.05.23.541998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Aging is the largest risk factor for neurodegenerative disorders, and commonly associated with compromised cerebrovasculature and pericytes. However, we do not know how normal aging differentially impacts the vascular structure and function in different brain areas. Here we utilize mesoscale microscopy methods (serial two-photon tomography and light sheet microscopy) and in vivo imaging (wide field optical spectroscopy and two-photon imaging) to determine detailed changes in aged cerebrovascular networks. Whole-brain vascular tracing showed an overall ~10% decrease in vascular length and branching density, and light sheet imaging with 3D immunolabeling revealed increased arteriole tortuosity in aged brains. Vasculature and pericyte densities showed significant reductions in the deep cortical layers, hippocampal network, and basal forebrain areas. Moreover, in vivo imaging in awake mice identified delays in neurovascular coupling and disrupted blood oxygenation. Collectively, we uncover regional vulnerabilities of cerebrovascular network and physiological changes that can mediate cognitive decline in normal aging.
Collapse
Affiliation(s)
- Hannah C Bennett
- Department of Neural and Behavioral Sciences, The Pennsylvania State University, Hershey, PA, 17033, USA
- Equal contribution
| | - Qingguang Zhang
- Center for Neural Engineering, Department of Engineering Science and Mechanics, The Pennsylvania State University, University Park, PA, 16802, USA
- Equal contribution
| | - Yuan-Ting Wu
- Department of Neural and Behavioral Sciences, The Pennsylvania State University, Hershey, PA, 17033, USA
| | - Uree Chon
- Department of Neural and Behavioral Sciences, The Pennsylvania State University, Hershey, PA, 17033, USA
| | - Hyun-Jae Pi
- Department of Neural and Behavioral Sciences, The Pennsylvania State University, Hershey, PA, 17033, USA
| | - Patrick J Drew
- Center for Neural Engineering, Department of Engineering Science and Mechanics, The Pennsylvania State University, University Park, PA, 16802, USA
- Biomedical Engineering, Biology, and Neurosurgery, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Yongsoo Kim
- Department of Neural and Behavioral Sciences, The Pennsylvania State University, Hershey, PA, 17033, USA
- Center for Neural Engineering, Department of Engineering Science and Mechanics, The Pennsylvania State University, University Park, PA, 16802, USA
- Lead contact
| |
Collapse
|
9
|
Zhang X, Jiang W, Lu Y, Mao T, Gu Y, Ju D, Dong C. Exosomes combined with biomaterials in the treatment of spinal cord injury. Front Bioeng Biotechnol 2023; 11:1077825. [PMID: 36994357 PMCID: PMC10040754 DOI: 10.3389/fbioe.2023.1077825] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Accepted: 02/27/2023] [Indexed: 03/16/2023] Open
Abstract
Spinal cord injury (SCI) is a serious and disabling disease with a high mortality rate. It often leads to complete or partial sensory and motor dysfunction and is accompanied by a series of secondary outcomes, such as pressure sores, pulmonary infections, deep vein thrombosis in the lower extremities, urinary tract infections, and autonomic dysfunction. Currently, the main treatments for SCI include surgical decompression, drug therapy, and postoperative rehabilitation. Studies have shown that cell therapy plays a beneficial role in the treatment of SCI. Nonetheless, there is controversy regarding the therapeutic effect of cell transplantation in SCI models. Meanwhile exosomes, as a new therapeutic medium for regenerative medicine, possess the advantages of small size, low immunogenicity, and the ability to cross the blood-spinal cord barrier. Certain studies have shown that stem cell-derived exosomes have anti-inflammatory effects and can play an irreplaceable role in the treatment of SCI. In this case, it is difficult for a single treatment method to play an effective role in the repair of neural tissue after SCI. The combination of biomaterial scaffolds and exosomes can better transfer and fix exosomes to the injury site and improve their survival rate. This paper first reviews the current research status of stem cell-derived exosomes and biomaterial scaffolds in the treatment of SCI respectively, and then describes the application of exosomes combined with biomaterial scaffolds in the treatment of SCI, as well as the challenges and prospects.
Collapse
|
10
|
Rocha BGS, Picoli CC, Gonçalves BOP, Silva WN, Costa AC, Moraes MM, Costa PAC, Santos GSP, Almeida MR, Silva LM, Singh Y, Falchetti M, Guardia GDA, Guimarães PPG, Russo RC, Resende RR, Pinto MCX, Amorim JH, Azevedo VAC, Kanashiro A, Nakaya HI, Rocha EL, Galante PAF, Mintz A, Frenette PS, Birbrair A. Tissue-resident glial cells associate with tumoral vasculature and promote cancer progression. Angiogenesis 2023; 26:129-166. [PMID: 36183032 DOI: 10.1007/s10456-022-09858-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 09/08/2022] [Indexed: 11/01/2022]
Abstract
Cancer cells are embedded within the tissue and interact dynamically with its components during cancer progression. Understanding the contribution of cellular components within the tumor microenvironment is crucial for the success of therapeutic applications. Here, we reveal the presence of perivascular GFAP+/Plp1+ cells within the tumor microenvironment. Using in vivo inducible Cre/loxP mediated systems, we demonstrated that these cells derive from tissue-resident Schwann cells. Genetic ablation of endogenous Schwann cells slowed down tumor growth and angiogenesis. Schwann cell-specific depletion also induced a boost in the immune surveillance by increasing tumor-infiltrating anti-tumor lymphocytes, while reducing immune-suppressor cells. In humans, a retrospective in silico analysis of tumor biopsies revealed that increased expression of Schwann cell-related genes within melanoma was associated with improved survival. Collectively, our study suggests that Schwann cells regulate tumor progression, indicating that manipulation of Schwann cells may provide a valuable tool to improve cancer patients' outcomes.
Collapse
Affiliation(s)
- Beatriz G S Rocha
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Caroline C Picoli
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Bryan O P Gonçalves
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Walison N Silva
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Alinne C Costa
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Michele M Moraes
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Pedro A C Costa
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Gabryella S P Santos
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Milla R Almeida
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Luciana M Silva
- Department of Cell Biology, Ezequiel Dias Foundation, Belo Horizonte, MG, Brazil
| | - Youvika Singh
- Hospital Israelita Albert Einstein, São Paulo, SP, Brazil
| | - Marcelo Falchetti
- Department of Microbiology and Immunology, Federal University of Santa Catarina, Florianópolis, Brazil
| | | | - Pedro P G Guimarães
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Remo C Russo
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Rodrigo R Resende
- Department of Biochemistry and Immunology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Mauro C X Pinto
- Institute of Biological Sciences, Federal University of Goiás, Goiânia, GO, Brazil
| | - Jaime H Amorim
- Center of Biological Sciences and Health, Federal University of Western Bahia, Barreiras, BA, Brazil
| | - Vasco A C Azevedo
- Department of Genetics, Ecology and Evolution, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Alexandre Kanashiro
- Department of Dermatology, University of Wisconsin-Madison, Medical Sciences Center, Rm 4385, 1300 University Avenue, Madison, WI, 53706, USA
| | | | - Edroaldo L Rocha
- Department of Microbiology and Immunology, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Pedro A F Galante
- Centro de Oncologia Molecular, Hospital Sirio-Libanes, Sao Paulo, SP, Brazil
| | - Akiva Mintz
- Department of Radiology, Columbia University Medical Center, New York, NY, USA
| | - Paul S Frenette
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, New York, NY, USA
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Alexander Birbrair
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil.
- Department of Dermatology, University of Wisconsin-Madison, Medical Sciences Center, Rm 4385, 1300 University Avenue, Madison, WI, 53706, USA.
- Department of Radiology, Columbia University Medical Center, New York, NY, USA.
| |
Collapse
|
11
|
Hu R, Shi M, Xu H, Wu X, He K, Chen Y, Wu L, Ma R. Integrated bioinformatics analysis identifies the effects of Sema3A/NRP1 signaling in oligodendrocytes after spinal cord injury in rats. PeerJ 2022; 10:e13856. [PMID: 35990904 PMCID: PMC9390322 DOI: 10.7717/peerj.13856] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 07/16/2022] [Indexed: 01/18/2023] Open
Abstract
Objective To investigate the effect of Sema3A/NRP1 signaling in oligodendrocytes (OLs) after spinal cord injury. Methods Three analysis strategies, namely differential expression gene analysis, Gene Ontology (GO) enrichment, and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis, were applied. The protein-protein interaction (PPI) network was constructed using the STRING website to explore the correlation between Sema3A/NRP1 and oligodendrocytes. Then, the T10 spinal cord segment of rats was injured by the Allen method to establish a spinal cord injury (SCI) model. Real-time quantitative PCR, Western blotting, Nissl staining and immunofluorescence staining were used to detect the effect of Sema3A/NRP1 signaling on oligodendrocytes in vivo. Results After the SCI model was established, significantly fewer oligodendrocytes were observed. At the same time, R software was used to analyze the expression of related genes, and NRP1 expression was increased. PCR also demonstrated similar results, and NRP1 ligand Sema3A was also upregulated. KEGG and GO functional enrichment analysis indicated that the SCI model was mainly related to cytokine interaction, cell proliferation, differentiation and maturation. Interestingly, we found that NRP1 was involved in semaphorin-plexin signaling pathway neuronal projection guidance and axon guidance, mediating cell growth and migration. Moreover, Sema3A/NRP1 signaling was closely associated with platelet-derived growth factor receptor α (PDGFRα) in the PPI network. When Sema3A/NRP1 signaling was specifically blocked at early stages, PDGFRα expression was effectively inhibited, and the expression of OLs was promoted. Furthermore, inhibition of Sema3A/NRP1 signaling increased the Basso-Beattie-Bresnahan (BBB) score of lower limb motor function in SCI rats and promoted the survival of motor neurons in the ventral horn of the injured spinal cord. Conclusion Our data suggest that Sema3A/NRP1 signaling may regulate the development of OPCs and OLs after SCI, thereby affecting functional recovery.
Collapse
Affiliation(s)
- Rong Hu
- Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Third School of Clinical Medicine (School of Rehabilitation Medicine), HangZhou, China
| | - Mengting Shi
- Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Third School of Clinical Medicine (School of Rehabilitation Medicine), HangZhou, China
| | - Haipeng Xu
- Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Third School of Clinical Medicine (School of Rehabilitation Medicine), HangZhou, China
| | - Xingying Wu
- Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Third School of Clinical Medicine (School of Rehabilitation Medicine), HangZhou, China
| | - Kelin He
- Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Third School of Clinical Medicine (School of Rehabilitation Medicine), HangZhou, China,Department of Acupuncture and Moxibustion, The Third Affiliated Hospital of Zhejiang Chinese Medical University (Zhongshan Hospital of Zhejiang Province), HangZhou, China
| | - Yi Chen
- Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Third School of Clinical Medicine (School of Rehabilitation Medicine), HangZhou, China
| | - Lei Wu
- Department of Acupuncture and Moxibustion, The Third Affiliated Hospital of Zhejiang Chinese Medical University (Zhongshan Hospital of Zhejiang Province), HangZhou, China
| | - Ruijie Ma
- Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Third School of Clinical Medicine (School of Rehabilitation Medicine), HangZhou, China,Department of Acupuncture and Moxibustion, The Third Affiliated Hospital of Zhejiang Chinese Medical University (Zhongshan Hospital of Zhejiang Province), HangZhou, China
| |
Collapse
|
12
|
The Provenance, Providence, and Position of Endothelial Cells in Injured Spinal Cord Vascular Pathology. Cell Mol Neurobiol 2022; 43:1519-1535. [PMID: 35945301 DOI: 10.1007/s10571-022-01266-9] [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: 01/06/2022] [Accepted: 07/21/2022] [Indexed: 11/03/2022]
Abstract
Endothelial cells (ECs) and pericytes are present in all blood vessels. Their position confers an important role in controlling oxygen and nutrient transportation to the different organs. ECs can adopt different morphologies based on their need and functions. Both ECs and pericytes express different surface markers that help in their identification, but heterogeneity and overlapping between markers among different cells pose a challenge for their precise identification. Spatiotemporal association of ECs and pericytes have great importance in sprout formation and vessel stabilization. Any traumatic injury in CNS may lead to vascular damage along with neuronal damage. Hence, ECs-pericyte interaction by physical contact and paracrine molecules is crucial in recovering the epicenter region by promoting angiogenesis. ECs can transform into other types of cells through endothelial-mesenchymal transition (EndMT), promoting wound healing in the epicenter region. Various signaling pathways mediate the interaction of ECs with pericytes that have an extensive role in angiogenesis. In this review, we discussed ECs and pericytes surface markers, the spatiotemporal association and interaction of ECs-pericytes, and signaling associated with the pathology of traumatic SCI. Linking the brain or spinal cord-specific pathologies and human vascular pathology will pave the way toward identifying new therapeutic targets and developing innovative preventive strategies. Endothelial-pericyte interaction strategic for formation of functional neo-vessels that are crucial for neurological recovery.
Collapse
|
13
|
Bernardes SS, Pinto MCX, Amorim JH, Azevedo VADC, Resende RR, Mintz A, Birbrair A. Glioma Pericytes Promote Angiogenesis by Producing Periostin. Cell Mol Neurobiol 2022; 42:557-564. [PMID: 33010018 PMCID: PMC8018985 DOI: 10.1007/s10571-020-00975-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 09/27/2020] [Indexed: 12/14/2022]
Abstract
Glioma is the prevalent aggressive primary brain tumor, with a very poor prognosis. The absence of advanced understanding of the roles played by the cells within the glioma microenvironment limits the development of effective drugs. A recent study indicates that periostin expressed by pericytes is crucial for glioma angiogenesis. Here, we describe succinctly the results and implications of this discovery in what we know about pericytes within the glioma microenvironment. The emerging knowledge from this work will benefit the development of therapies for gliomas.
Collapse
Affiliation(s)
- Sara Santos Bernardes
- Tissue Microenvironment Laboratory, Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Mauro Cunha Xavier Pinto
- Laboratory of Neuropharmacology and Neurochemistry, Institute of Biological Sciences, Federal University of Goiás, Goiânia, GO, Brazil
| | - Jaime Henrique Amorim
- Center of Biological Sciences and Health, Federal University of West Bahia, Barreiras, BA, Brazil
| | - Vasco Ariston de Carvalho Azevedo
- Cellular and Molecular Genetics Laboratory, Department of Genetics, Ecology and Evolution, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Rodrigo Ribeiro Resende
- Department of Biochemistry and Immunology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Akiva Mintz
- Department of Radiology, Columbia University Medical Center, New York, NY, USA
| | - Alexander Birbrair
- Tissue Microenvironment Laboratory, Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil.
- Department of Radiology, Columbia University Medical Center, New York, NY, USA.
| |
Collapse
|
14
|
Adhikari K, Dolma S, Mamidi T, Roy A, Pathak Z, Kumar H. Tomographic Imaging and Correlation to Quantify Vascular and Inflammatory Changes in an Experimental Spinal Cord Injury. ACS Chem Neurosci 2021; 12:3864-3872. [PMID: 34628864 DOI: 10.1021/acschemneuro.1c00390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Spinal cord injury (SCI) is a devastating condition causing the loss of sensory and motor functions. SCI pathology is multifaceted, encompassing inflammation, scarring, neuronal damage, and vascular and tissue remodeling. The dynamics of SCI rapidly transform from acute, sub-acute, and chronic phases. The rapidly changing environment necessitates the real-time monitoring of disease severity. Therefore, in this study, we used the IVIS spectrum, a noninvasive fluorescence imaging modality, to monitor the disease pathology in live animals. We used near-infrared fluorescence imaging agents including Angiosense 750 EX, a probe that detects vascular changes, and Cat B 680 FAST, a probe that detects inflammation at various day points post injury (DPI), that is, DPI-1, DPI-14, and DPI-28. We quantified the pathophysiological changes after SCI using IVIS in live animals. As a result, we observed distinct differences in the disease progression between injured and sham mice. Moreover, live imaging showed a good correlation with behavioral studies, protein expression, and immunohistological analysis. Hence, the goal of this study was to introduce a new optical imaging modality that offers a determination of disease severity and the advantage of accelerated imaging of the correlated biomarkers in a real-time and dynamic manner. This study concluded that Cat B 680 Fast and Angiosense 750 EX could be used to assess the disease severity after SCI. Furthermore, our study suggests that the noninvasive fluorescence optical imaging modality offers a unique approach in monitoring neuroinflammatory diseases in live animals.
Collapse
Affiliation(s)
- Kirti Adhikari
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Sonam Dolma
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Teena Mamidi
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Abhishek Roy
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Zarna Pathak
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Hemant Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat 382355, India
| |
Collapse
|
15
|
Wu H, Li Y, Wang X, Zhang Z, Huang Y. Long non-coding RNA TUG1 knockdown prevents neurons from death to alleviate acute spinal cord injury via the microRNA-338/BIK axis. Bioengineered 2021; 12:5566-5582. [PMID: 34517787 PMCID: PMC8806874 DOI: 10.1080/21655979.2021.1966258] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Taurine up-regulated gene 1 (TUG1) is a cancer-associated long noncoding RNA (lncRNA) and engages in the development of spinal cord injury (SCI), a suffering neuropathological disorder. However, the regulatory role of TUG1 in acute SCI (ASCI) is still underdetermined. RT-qPCR and western blot analysis were applied to measure the expression of TUG1, microRNA-338 (miR-338), Bcl2-interacting killer (BIK), cleaved caspase 3 (c-caspase 3) and hypoxia-inducible factor-1 alpha (HIF-1α) in ASCI rats and hypoxic cells. Cell death was evaluated using flow cytometric analysis. The relationships among miR-338, TUG1 or BIK were confirmed by luciferase reporter assay, RNA immunoprecipitation and RNA pull-down. Accordingly, we monitored higher expression of TUG1 and BIK, but lower expression of miR-338 in ASCI rats and hypoxic cells. In vitro, hypoxia expedited cell death and c-caspase 3 levels. In vivo, ASCI rats were successfully developed as evidenced by diminished Basso-Beattie-Bresnahan (BBB) locomotor score and enhanced c-caspase 3 and HIF-1α expression. Nevertheless, TUG1 knockdown mitigated the cell death in ASCI rats and hypoxic cells. Mechanically, TUG1 interacted with miR-338 to regulate the BIK expression. Together, TUG1 silencing could alleviate the death in neurons and ASCI models via modulating the miR-338/BIK axis.
Collapse
Affiliation(s)
- Hongbo Wu
- Department of Orthopaedics, Huizhou City Center People's Hospital, Huizhou Guangdong, P.R. China
| | - Yi Li
- Department of Orthopaedics, Huizhou City Center People's Hospital, Huizhou Guangdong, P.R. China
| | - Xiaofeng Wang
- Department of Orthopaedics, Huizhou City Center People's Hospital, Huizhou Guangdong, P.R. China
| | - Zhiwen Zhang
- Department of Orthopaedics, Huizhou City Center People's Hospital, Huizhou Guangdong, P.R. China
| | - Yuliang Huang
- Department of Orthopaedics, Huizhou City Center People's Hospital, Huizhou Guangdong, P.R. China
| |
Collapse
|
16
|
Picoli CC, Gonçalves BÔP, Santos GSP, Rocha BGS, Costa AC, Resende RR, Birbrair A. Pericytes cross-talks within the tumor microenvironment. Biochim Biophys Acta Rev Cancer 2021; 1876:188608. [PMID: 34384850 DOI: 10.1016/j.bbcan.2021.188608] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 07/14/2021] [Accepted: 08/05/2021] [Indexed: 02/07/2023]
Abstract
Cancer cells are embedded within the tumor microenvironment and interact dynamically with its components during tumor progression. Understanding the molecular mechanisms by which the tumor microenvironment components communicate is crucial for the success of therapeutic applications. Recent studies show, by using state-of-the-art technologies, including sophisticated in vivo inducible Cre/loxP mediated systems and CRISPR-Cas9 gene editing, that pericytes communicate with cancer cells. The arising knowledge on cross-talks within the tumor microenvironment will be essential for the development of new therapies against cancer. Here, we review recent progress in our understanding of pericytes roles within tumors.
Collapse
Affiliation(s)
- Caroline C Picoli
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Bryan Ô P Gonçalves
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Gabryella S P Santos
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Beatriz G S Rocha
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Alinne C Costa
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Rodrigo R Resende
- Department of Biochemistry and Immunology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Alexander Birbrair
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil; Department of Radiology, Columbia University Medical Center, New York, NY, USA.
| |
Collapse
|
17
|
Man W, Yang S, Cao Z, Lu J, Kong X, Sun X, Zhao L, Guo Y, Yao S, Wang G, Wang X. A multi-modal delivery strategy for spinal cord regeneration using a composite hydrogel presenting biophysical and biochemical cues synergistically. Biomaterials 2021; 276:120971. [PMID: 34242812 DOI: 10.1016/j.biomaterials.2021.120971] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 06/04/2021] [Accepted: 06/10/2021] [Indexed: 01/11/2023]
Abstract
Extensive tissue engineering studies have supported the enhanced spinal cord regeneration by implantable scaffolds loaded with bioactive cues. However, scaffolds with single-cue delivery showed unsatisfactory effects, most likely due to the complex nature of hostile niches in the lesion area. In this regard, strategies of multi-modal delivery of multiple heterogeneous cell-regulatory cues are unmet needs for enhancing spinal cord repair, which requires a thorough understanding of the regenerative niche associated with spinal cord injury. Here, by combining hierarchically aligned fibrin hydrogel (AFG) and functionalized self-assembling peptides (fSAP), a novel multifunctional nanofiber composite hydrogel AFG/fSAP characterized with interpenetrating network is designed. Serving as a source of both biophysical and biochemical cues, AFG/fSAP can facilitate spinal cord regeneration via guiding regenerated tissues, accelerating axonal regrowth and remyelination, and promoting angiogenesis. Giving the synergistic effect of multiple cues, AFG/fSAP implantation contributes to anatomical, electrophysiological, and motor functional restorations in rats with spinal cord hemisection. This study provides a novel multi-modal approach for regeneration in central nervous system, which has potentials for clinical practice of spinal cord injury.
Collapse
Affiliation(s)
- Weitao Man
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, China; Department of Neurosurgery, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 102218, China
| | - Shuhui Yang
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, China
| | - Zheng Cao
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, China
| | - Jiaju Lu
- School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, China; Zhejiang-Mauritius Joint Research Center for Biomaterials and Tissue Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Xiangdong Kong
- School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, China; Zhejiang-Mauritius Joint Research Center for Biomaterials and Tissue Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Xiaodan Sun
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, China
| | - Lingyun Zhao
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, China
| | - Yi Guo
- School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Shenglian Yao
- School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing, 100083, China
| | - Guihuai Wang
- Department of Neurosurgery, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 102218, China.
| | - Xiumei Wang
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
18
|
Shu J, Cheng F, Gong Z, Ying L, Wang C, Yu C, Zhou X, Xiao M, Wang J, Xia K, Huang X, Tao Y, Shi K, Liu Y, Liang C, Chen Q, Feng X, Li F. Transplantation Strategies for Spinal Cord Injury Based on Microenvironment Modulation. Curr Stem Cell Res Ther 2021; 15:522-530. [PMID: 32316901 DOI: 10.2174/1574888x15666200421112622] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 02/27/2020] [Accepted: 03/02/2020] [Indexed: 12/20/2022]
Abstract
Spinal cord injury (SCI) is different from peripheral nerve injury; it results in devastating and permanent damage to the spine, leading to severe motor, sensory and autonomic dysfunction. SCI produces a complex microenvironment that can result in hemorrhage, inflammation and scar formation. Not only does it significantly limit regeneration, but it also challenges a multitude of transplantation strategies. In order to promote regeneration, researchers have recently begun to focus their attention on strategies that manipulate the complicated microenvironment produced by SCI. And some have achieved great therapeutic effects. Hence, reconstructing an appropriate microenvironment after transplantation could be a potential therapeutic solution for SCI. In this review, first, we aim to summarize the influential compositions of the microenvironment and their different effects on regeneration. Second, we highlight recent research that used various transplantation strategies to modulate different microenvironments produced by SCI in order to improve regeneration. Finally, we discuss future transplantation strategies regarding SCI.
Collapse
Affiliation(s)
- Jiawei Shu
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, Zhejiang, China
| | - Feng Cheng
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, Zhejiang, China
| | - Zhe Gong
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, Zhejiang, China
| | - Liwei Ying
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, Zhejiang, China
| | - Chenggui Wang
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, Zhejiang, China
| | - Chao Yu
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, Zhejiang, China
| | - Xiaopeng Zhou
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, Zhejiang, China
| | - Mu Xiao
- The MOE Key Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jingkai Wang
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, Zhejiang, China
| | - Kaishun Xia
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, Zhejiang, China
| | - Xianpeng Huang
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, Zhejiang, China
| | - Yiqing Tao
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, Zhejiang, China
| | - Kesi Shi
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, Zhejiang, China
| | - Yuemei Liu
- The MOE Key Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang, China
| | - Chengzhen Liang
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, Zhejiang, China
| | - Qixin Chen
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, Zhejiang, China
| | - Xinhua Feng
- The MOE Key Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang, China
| | - Fangcai Li
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, Zhejiang, China
| |
Collapse
|
19
|
Coimbra-Campos LMC, Silva WN, Baltazar LM, Costa PAC, Prazeres PHDM, Picoli CC, Costa AC, Rocha BGS, Santos GSP, Oliveira FMS, Pinto MCX, Amorim JH, Azevedo VAC, Souza DG, Russo RC, Resende RR, Mintz A, Birbrair A. Circulating Nestin-GFP + Cells Participate in the Pathogenesis of Paracoccidioides brasiliensis in the Lungs. Stem Cell Rev Rep 2021; 17:1874-1888. [PMID: 34003465 DOI: 10.1007/s12015-021-10181-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/06/2021] [Indexed: 02/06/2023]
Abstract
Multiple infectious diseases lead to impaired lung function. Revealing the cellular mechanisms involved in this impairment is crucial for the understanding of how the lungs shift from a physiologic to a pathologic state in each specific condition. In this context, we explored the pathogenesis of Paracoccidioidomycosis, which affects pulmonary functioning. The presence of cells expressing Nestin-GFP has been reported in different tissues, and their roles as tissue-specific progenitors have been stablished in particular organs. Here, we explored how Nestin-GFP+ cells are affected after lung infection by Paracoccidioides brasiliensis, a model of lung granulomatous inflammation with fibrotic outcome. We used Nestin-GFP transgenic mice, parabiosis surgery, confocal microscopy and flow cytometry to investigate the participation of Nestin-GFP+ cells in Paracoccidioides brasiliensis pathogenesis. We revealed that these cells increase in the lungs post-Paracoccidioides brasiliensis infection, accumulating around granulomas. This increase was due mainly to Nestin-GPF+ cells derived from the blood circulation, not associated to blood vessels, that co-express markers suggestive of hematopoietic cells (Sca-1, CD45 and CXCR4). Therefore, our findings suggest that circulating Nestin-GFP+ cells participate in the Paracoccidioides brasiliensis pathogenesis in the lungs.
Collapse
Affiliation(s)
| | - Walison N Silva
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Ludmila M Baltazar
- Department of Microbiology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Pedro A C Costa
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Pedro H D M Prazeres
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Caroline C Picoli
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Alinne C Costa
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Beatriz G S Rocha
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Gabryella S P Santos
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Fabrício M S Oliveira
- Laboratory of Pulmonary Immunology and Mechanics, Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Mauro C X Pinto
- Laboratory of Neuropharmacology and Neurochemistry, Institute of Biological Sciences, Federal University of Goiás, Goiânia, GO, Brazil
| | - Jaime H Amorim
- Center of Biological Sciences and Health, Federal University of West Bahia, Barreiras, BA, Brazil
| | - Vasco A C Azevedo
- Cellular and Molecular Genetics Laboratory, Department of Genetics, Ecology and Evolution, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Danielle G Souza
- Department of Microbiology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Remo C Russo
- Laboratory of Pulmonary Immunology and Mechanics, Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Rodrigo R Resende
- Department of Biochemistry and Immunology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Akiva Mintz
- Department of Radiology, Columbia University Medical Center, New York, NY, USA
| | - Alexander Birbrair
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil.
| |
Collapse
|
20
|
Li J, Wei W, Xu F, Wang Y, Liu Y, Fu C. Clinical Therapy of Metastatic Spinal Tumors. Front Surg 2021; 8:626873. [PMID: 33937314 PMCID: PMC8084350 DOI: 10.3389/fsurg.2021.626873] [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: 11/07/2020] [Accepted: 03/23/2021] [Indexed: 11/13/2022] Open
Abstract
Metastatic spinal tumors (MST) have high rates of morbidity and mortality. MST can destroy the vertebral body or compress the nerve roots, resulting in an increased risk of pathological fractures and intractable pain. Here, we elaborately reviewed the currently available therapeutic options for MST according to the following four aspects: surgical management, minimally invasive therapy (MIT), radiation therapy, and systemic therapy. In particular, these aspects were classified and introduced to show their developmental process, clinical effects, advantages, and current limitations. Furthermore, with the improvement of treatment concepts and techniques, we discovered the prevalent trend toward the use of radiation therapy and MIT in clinic therapies. Finally, the future directions of these treatment options were discussed. We hoped that along with future advances and study will lead to the improvement of living standard and present status of treatment in patients with MST.
Collapse
Affiliation(s)
- Jie Li
- Department of Spine Surgery, The First Hospital of Jilin University, Changchun, China.,Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China
| | - Wenjie Wei
- Key Laboratory of Pathobiology, Ministry of Education, School of Basic Medical Sciences, Jilin University, Changchun, China
| | - Feng Xu
- Department of Spine Surgery, The First Hospital of Jilin University, Changchun, China
| | - Yuanyi Wang
- Department of Spine Surgery, The First Hospital of Jilin University, Changchun, China
| | - Yadong Liu
- Department of Spine Surgery, The First Hospital of Jilin University, Changchun, China
| | - Changfeng Fu
- Department of Spine Surgery, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
21
|
Bennett HC, Kim Y. Pericytes Across the Lifetime in the Central Nervous System. Front Cell Neurosci 2021; 15:627291. [PMID: 33776651 PMCID: PMC7994897 DOI: 10.3389/fncel.2021.627291] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 02/23/2021] [Indexed: 12/25/2022] Open
Abstract
The pericyte is a perivascular cell type that encapsulates the microvasculature of the brain and spinal cord. Pericytes play a crucial role in the development and maintenance of the blood-brain barrier (BBB) and have a multitude of important functions in the brain. Recent evidence indicates that pericyte impairment has been implicated in neurovascular pathology associated with various human diseases such as diabetes mellitus, Alzheimer's disease (AD), and stroke. Although the pericyte is essential for normal brain function, knowledge about its developmental trajectory and anatomical distribution is limited. This review article summarizes the scientific community's current understanding of pericytes' regional heterogeneity in the brain and their changes during major life stages. More specifically, this review article focuses on pericyte differentiation and migration during brain development, regional population differences in the adult brain, and changes during normal and pathological aging. Most of what is known about pericytes come from studies of the cerebral cortex and hippocampus. Therefore, we highlight the need to expand our understanding of pericyte distribution and function in the whole brain to better delineate this cell type's role in the normal brain and pathological conditions.
Collapse
Affiliation(s)
- Hannah C Bennett
- Department of Neural and Behavioral Sciences, Penn State University, Hershey, PA, United States
| | - Yongsoo Kim
- Department of Neural and Behavioral Sciences, Penn State University, Hershey, PA, United States
| |
Collapse
|
22
|
Zhu S, Ying Y, He Y, Zhong X, Ye J, Huang Z, Chen M, Wu Q, Zhang Y, Xiang Z, Tu Y, Ying W, Xiao J, Li X, Ye Q, Wang Z. Hypoxia response element-directed expression of bFGF in dental pulp stem cells improve the hypoxic environment by targeting pericytes in SCI rats. Bioact Mater 2021; 6:2452-2466. [PMID: 33553827 PMCID: PMC7850944 DOI: 10.1016/j.bioactmat.2021.01.024] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 01/07/2021] [Accepted: 01/19/2021] [Indexed: 02/08/2023] Open
Abstract
Cell-based transplantation strategies possess great potential for spinal cord injury (SCI) repair. Basic fibroblast growth factor (bFGF) has been reported to have multiple neuro-promoting effects on developing and adult nervous system of mammals and considered a promising therapy for nerve injury following SCI. Human dental pulp stem cells (DPSCs) are abundant stem cells with low immune rejection, which can be considered for cell replacement therapy. The purpose of this study was to investigate the roles of DPSCs which express bFGF under the regulation of five hypoxia-responsive elements (5HRE) using an adeno-associated virus (AAV-5HRE-bFGF-DPSCs) in SCI repairing model. In this study, DPSCs were revealed to differentiate into CD13+ pericytes and up-regulate N-cadherin expression to promote the re-attachment of CD13+ pericytes to vascular endothelial cells. The re-attachment of CD13+ pericytes to vascular endothelial cells subsequently increased the flow rate of blood in microvessels via the contraction of protuberance. As a result, increased numbers of red blood cells carried more oxygen to the damaged area and the local hypoxia microenvironment in SCI was improved. Thus, this study represents a step forward towards the potential use of AAV-5HRE-bFGF-DPSCs in SCI treatment in clinic. ) 5HRE-bFGF-DPSCs secrete bFGF in a hypoxia dependent manner, making the administration more precise. CD13+ pericyte regulate vascular diameter and promote the recovery of hypoxia microenvironment via DDC-5HT-5HT-1B in SCI. 5HRE-bFGF-DPSCs can differentiate into CD13+ pericyte to compensate for the mass death of CD13+ pericyte after SCI. 5HRE-bFGF-DPSCs promote CD13+ pericyte adhesion to vascular endothelial cell by secreting bFGF through N-cadherin. 5HRE-bFGF-DPSCs promote the recovery of SCI by restoring hypoxic microenvironment and inhibit autophagy pathway.
Collapse
Affiliation(s)
- Sipin Zhu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Yibo Ying
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Yan He
- Laboratory of Regenerative Medicine, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, 430064, China
| | - Xingxing Zhong
- The Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, China
| | - Jiahui Ye
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Zhiyang Huang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Min Chen
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Qiuji Wu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Yifan Zhang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Ziyue Xiang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Yurong Tu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Weiyang Ying
- Department of Pain Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Jian Xiao
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China.,Research Units of Clinical Translation of Cell Growth Factors and Diseases Research, Chinese Academy of Medical Science, China
| | - Xiaokun Li
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China.,Research Units of Clinical Translation of Cell Growth Factors and Diseases Research, Chinese Academy of Medical Science, China
| | - Qingsong Ye
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, 325035, China.,Massachusetts General Hospital, Harvard University, Boston, 02114, USA.,Center of Regenerative Medicine, Renmin Hospital of Wuhan University, Wuhan, 630060, China
| | - Zhouguang Wang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China.,Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
23
|
Alterations of Dopamine-Related Transcripts in A11 Diencephalospinal Pathways after Spinal Cord Injury. Neural Plast 2021; 2021:8838932. [PMID: 33510781 PMCID: PMC7822663 DOI: 10.1155/2021/8838932] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 12/04/2020] [Accepted: 01/05/2021] [Indexed: 12/15/2022] Open
Abstract
The diencephalic A11 nuclei are the primary source of spinal dopamine (DA). Neurons in this region project to all levels of the spinal cord. Traumatic spinal cord injury (SCI) often interrupts descending and ascending neuronal pathways and further elicits injury-induced neuronal plasticity. However, it is unknown how A11 neurons and projections respond to SCI-induced axotomy. Based on preliminary observation, we hypothesized that A11 DA-ergic neurons rostral to the lesion site might change their capacity to synthesize DA after SCI. Adult rats received a complete spinal cord transection at the 10th thoracic (T10) level. After 3 or 8 weeks, rostral (T5) and caudal (L1) spinal cord tissue was collected to measure mRNA levels of DA-related genes. Meanwhile, A11 neurons in the brain were explicitly isolated by laser capture microdissection, and single-cell qPCR was employed to evaluate mRNA levels in the soma. Histological analysis was conducted to assess the number of A11 DA-ergic neurons. The results showed that, compared to naïve rats, mRNA levels of tyrosine hydroxylase (TH), dopamine decarboxylase (DDC), and D2 receptors in the T5 spinal segment had a transient decrease and subsequent recovery. However, dopamine-β-hydroxylase (DBH), D1 receptors, and DA-associated transcription factors did not change following SCI. Furthermore, axon degeneration below the lesion substantially reduced mRNA levels of TH and D2 in the L1 spinal segment. However, DDC transcript underwent only a temporary decrease. Similar mRNA levels of DA-related enzymes were detected in the A11 neuronal soma between naïve and SCI rats. In addition, immunostaining revealed that the number of A11 DA neurons did not change after SCI, indicating a sustention of capacity to synthesize DA in the neuroplasm. Thus, impaired A11 diencephalospinal pathways following SCI may transiently reduce DA production in the spinal cord rostral to the lesion but not in the brain.
Collapse
|
24
|
Picoli CC, Costa AC, Rocha BGS, Silva WN, Santos GSP, Prazeres PHDM, Costa PAC, Oropeza A, da Silva RA, Azevedo VAC, Resende RR, Cunha TM, Mintz A, Birbrair A. Sensory nerves in the spotlight of the stem cell niche. Stem Cells Transl Med 2020; 10:346-356. [PMID: 33112056 PMCID: PMC7900586 DOI: 10.1002/sctm.20-0284] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 08/27/2020] [Accepted: 09/26/2020] [Indexed: 12/16/2022] Open
Abstract
Niches are specialized tissue microenvironments that control stem cells functioning. The bone marrow mesenchymal stem cell niche defines a location within the marrow in which mesenchymal stem cells are retained and produce new cells throughout life. Deciphering the signaling mechanisms by which the niche regulates stem cell fate will facilitate the use of these cells for therapy. Recent studies, by using state-of-the-art methodologies, including sophisticated in vivo inducible genetic techniques, such as lineage-tracing Cre/loxP mediated systems, in combination with pharmacological inhibition, provide evidence that sensory neuron is an important component of the bone marrow mesenchymal stem cell niche. Strikingly, knockout of a specific receptor in sensory neurons blocked stem cell function in the bone marrow. The knowledge arising from these discoveries will be crucial for stem cell manipulation in the future. Here, we review recent progress in our understanding of sensory nerves biology in the stem cell niche.
Collapse
Affiliation(s)
- Caroline C Picoli
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Alinne C Costa
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Beatriz G S Rocha
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Walison N Silva
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Gabryella S P Santos
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Pedro H D M Prazeres
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Pedro A C Costa
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Anderson Oropeza
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Rodrigo A da Silva
- Department of Dentistry, University of Taubaté, Taubaté, São Paulo, Brazil
| | - Vasco A C Azevedo
- Cellular and Molecular Genetics Laboratory, Department of Genetics, Ecology and Evolution, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Rodrigo R Resende
- Department of Biochemistry and Immunology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Thiago M Cunha
- Department of Pharmacology, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Akiva Mintz
- Department of Radiology, Columbia University Medical Center, New York, New York, USA
| | - Alexander Birbrair
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.,Department of Radiology, Columbia University Medical Center, New York, New York, USA
| |
Collapse
|
25
|
Lim DY, Hwang DM, Cho KH, Moon CW, Ahn SY. A Fully Immersive Virtual Reality Method for Upper Limb Rehabilitation in Spinal Cord Injury. Ann Rehabil Med 2020; 44:311-319. [PMID: 32721988 PMCID: PMC7463118 DOI: 10.5535/arm.19181] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 12/06/2019] [Indexed: 11/25/2022] Open
Abstract
Objective To determine whether a fully immersive virtual reality (VR) intervention combined with conventional rehabilitation (CR) can improve upper limb function more than CR alone in patients with spinal cord injury (SCI), we conducted a prospective, randomized, controlled clinical trial. Methods Participants were randomly assigned to either the control group (CG; n=10) or experimental group (EG; n=10). The participants in the CG received 60 minutes of conventional therapy per day, 4 days per week for 4 weeks, whereas those in the EG received 30 minutes of VR training and 30 minutes of conventional therapy per day, 4 days per week for 4 weeks. The clinical outcome measures included Medical Research Council grade, the American Spinal Injury Association upper extremity motor score (ASIA-UEMS), and scores in the Hand Strength Test, Box and Block Test, Nine-Hole Peg Test, Action Research Arm Test, and Korean version of the Spinal Cord Independence Measure (K-SCIM). The assessments were performed at the beginning (T0) and end of the intervention (T1). Results Grip power and K-SCIM score significantly improved in the EG after the intervention. When comparing differences between the groups, elbow extensor, wrist extensor, ASIA-UEMS, grip power, lateral pinch power, and palmar pinch power were all significantly improved. Conclusion VR training of upper limb function after SCI can provide an acceptable adjunctive rehabilitation method without significant adverse effects.
Collapse
Affiliation(s)
- Da Young Lim
- Department of Rehabilitation Medicine, Chungnam National University College of Medicine, Daejeon, Korea
| | - Dong Min Hwang
- Department of Rehabilitation Medicine, Chungnam National University College of Medicine, Daejeon, Korea
| | - Kang Hee Cho
- Department of Rehabilitation Medicine, Chungnam National University College of Medicine, Daejeon, Korea.,Biomedical Institute, Chungnam National University, Daejeon, Korea
| | - Chang Won Moon
- Department of Rehabilitation Medicine, Chungnam National University College of Medicine, Daejeon, Korea
| | - So Young Ahn
- Department of Rehabilitation Medicine, Chungnam National University College of Medicine, Daejeon, Korea
| |
Collapse
|
26
|
Téglási V, Csűry DT, Dezső K, Bugyik E, Szabó V, Szállási Z, Paku S, Reiniger L. Origin and Distribution of Connective Tissue and Pericytes Impacting Vascularization in Brain Metastases With Different Growth Patterns. J Neuropathol Exp Neurol 2020; 78:326-339. [PMID: 30816955 DOI: 10.1093/jnen/nlz007] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The impact of growth pattern on the distribution of connective tissue and on the vascularization of brain metastases (40 colon, lung and breast carcinoma samples) was analyzed. Most of the cases showed either a "pushing-type" (18/40, mostly colon and lung carcinomas) or a "papillary-type" (19/40, mostly breast carcinomas) growth pattern. There was a striking difference in the growth pattern and vascularization of colon/lung versus breast carcinoma metastases. Pushing-type brain metastases incorporated fewer vessels and accumulated more collagen in the adjacent brain parenchyma, whereas papillary-type brain metastases incorporated more vessels and accumulated collagen in the center of the tumor. We observed duplication of the PDGFRβ-positive pericyte layer accompanied by an increase in the amount of collagen within the vessel walls. The outer layer of pericytes and the collagen was removed from the vessel by invasive activity of the tumors, which occurred either peri- or intratumorally, depending on the growth pattern of the metastasis. Our findings suggest that pericytes are the main source of the connective tissue in brain metastases. Vascularization and connective tissue accumulation of the brain metastases largely depend on the growth pattern of the tumors.
Collapse
Affiliation(s)
- Vanda Téglási
- 1st Department of Pathology and Experimental Cancer Research
| | - Dániel T Csűry
- 1st Department of Pathology and Experimental Cancer Research
| | - Katalin Dezső
- 1st Department of Pathology and Experimental Cancer Research
| | - Edina Bugyik
- 1st Department of Pathology and Experimental Cancer Research
| | - Vanessza Szabó
- 1st Department of Pathology and Experimental Cancer Research
| | - Zoltán Szállási
- Brain Metastasis Research Group, Hungarian Academy of Sciences, 2nd Department of Pathology, Semmelweis University, Budapest, Hungary.,Computational Health Informatics Program, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts.,Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Sándor Paku
- 1st Department of Pathology and Experimental Cancer Research
| | - Lilla Reiniger
- 1st Department of Pathology and Experimental Cancer Research.,Brain Metastasis Research Group, Hungarian Academy of Sciences, 2nd Department of Pathology, Semmelweis University, Budapest, Hungary
| |
Collapse
|
27
|
Andreotti JP, Silva WN, Costa AC, Picoli CC, Bitencourt FCO, Coimbra-Campos LMC, Resende RR, Magno LAV, Romano-Silva MA, Mintz A, Birbrair A. Neural stem cell niche heterogeneity. Semin Cell Dev Biol 2019; 95:42-53. [PMID: 30639325 PMCID: PMC6710163 DOI: 10.1016/j.semcdb.2019.01.005] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 12/02/2018] [Accepted: 01/08/2019] [Indexed: 12/15/2022]
Abstract
In mammals, new neurons can be generated from neural stem cells in specific regions of the adult brain. Neural stem cells are characterized by their abilities to differentiate into all neural lineages and to self-renew. The specific microenvironments regulating neural stem cells, commonly referred to as neurogenic niches, comprise multiple cell populations whose precise contributions are under active current exploration. Understanding the cross-talk between neural stem cells and their niche components is essential for the development of therapies against neurological disorders in which neural stem cells function is altered. In this review, we describe and discuss recent studies that identified novel components in the neural stem cell niche. These discoveries bring new concepts to the field. Here, we evaluate these recent advances that change our understanding of the neural stem cell niche heterogeneity and its influence on neural stem cell function.
Collapse
Affiliation(s)
- Julia P Andreotti
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Walison N Silva
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Alinne C Costa
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Caroline C Picoli
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Flávia C O Bitencourt
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | | | - Rodrigo R Resende
- Department of Biochemistry and Immunology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Luiz A V Magno
- Department of Mental Health, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Marco A Romano-Silva
- Department of Mental Health, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Akiva Mintz
- Department of Radiology, Columbia University Medical Center, New York, NY, USA
| | - Alexander Birbrair
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil; Department of Radiology, Columbia University Medical Center, New York, NY, USA.
| |
Collapse
|
28
|
Leonel C, Sena IFG, Silva WN, Prazeres PHDM, Fernandes GR, Mancha Agresti P, Martins Drumond M, Mintz A, Azevedo VAC, Birbrair A. Staphylococcus epidermidis role in the skin microenvironment. J Cell Mol Med 2019; 23:5949-5955. [PMID: 31278859 PMCID: PMC6714221 DOI: 10.1111/jcmm.14415] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 04/12/2019] [Accepted: 04/30/2019] [Indexed: 02/06/2023] Open
Abstract
Wound healing is a complex dynamic physiological process in response to cutaneous destructive stimuli that aims to restore the cutaneous' barrier role. Deciphering the underlying mechanistic details that contribute to wound healing will create novel therapeutic strategies for skin repair. Recently, by using state-of-the-art technologies, it was revealed that the cutaneous microbiota interact with skin immune cells. Strikingly, commensal Staphylococcus epidermidis-induced CD8+ T cells induce re-epithelization of the skin after injury, accelerating wound closure. From a drug development perspective, the microbiota may provide new therapeutic candidate molecules to accelerate skin healing. Here, we summarize and evaluate recent advances in the understanding of the microbiota in the skin microenvironment.
Collapse
Affiliation(s)
- Caroline Leonel
- Departamento de PatologiaUniversidade Federal de Minas GeraisBelo HorizonteBrasil
| | - Isadora F. G. Sena
- Departamento de PatologiaUniversidade Federal de Minas GeraisBelo HorizonteBrasil
| | - Walison N. Silva
- Departamento de PatologiaUniversidade Federal de Minas GeraisBelo HorizonteBrasil
| | | | | | - Pamela Mancha Agresti
- Departamento de Biologia GeralUniversidade Federal de Minas GeraisBelo HorizonteBrasil
| | | | - Akiva Mintz
- Department of RadiologyColumbia University Medical CenterNew YorkNew York
| | - Vasco A. C. Azevedo
- Departamento de Biologia GeralUniversidade Federal de Minas GeraisBelo HorizonteBrasil
| | - Alexander Birbrair
- Departamento de PatologiaUniversidade Federal de Minas GeraisBelo HorizonteBrasil
- Department of RadiologyColumbia University Medical CenterNew YorkNew York
| |
Collapse
|
29
|
Santos GSP, Magno LAV, Romano-Silva MA, Mintz A, Birbrair A. Pericyte Plasticity in the Brain. Neurosci Bull 2019; 35:551-560. [PMID: 30367336 PMCID: PMC6527663 DOI: 10.1007/s12264-018-0296-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 08/06/2018] [Indexed: 12/16/2022] Open
Abstract
Cerebral pericytes are perivascular cells that stabilize blood vessels. Little is known about the plasticity of pericytes in the adult brain in vivo. Recently, using state-of-the-art technologies, including two-photon microscopy in combination with sophisticated Cre/loxP in vivo tracing techniques, a novel role of pericytes was revealed in vascular remodeling in the adult brain. Strikingly, after pericyte ablation, neighboring pericytes expand their processes and prevent vascular dilatation. This new knowledge provides insights into pericyte plasticity in the adult brain.
Collapse
Affiliation(s)
- Gabryella S P Santos
- Departamento de Patologia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, 31270-901, Brazil
| | - Luiz A V Magno
- Department of Mental Health, Federal University of Minas Gerais, Belo Horizonte, MG, 30130-100, Brazil
| | - Marco A Romano-Silva
- Department of Mental Health, Federal University of Minas Gerais, Belo Horizonte, MG, 30130-100, Brazil
| | - Akiva Mintz
- Department of Radiology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Alexander Birbrair
- Departamento de Patologia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, 31270-901, Brazil.
- Department of Radiology, Columbia University Medical Center, New York, NY, 10032, USA.
| |
Collapse
|
30
|
Azevedo PO, Paiva AE, Santos GSP, Lousado L, Andreotti JP, Sena IFG, Tagliati CA, Mintz A, Birbrair A. Cross-talk between lung cancer and bones results in neutrophils that promote tumor progression. Cancer Metastasis Rev 2019; 37:779-790. [PMID: 30203108 DOI: 10.1007/s10555-018-9759-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Lung cancer is the leading cause of cancer mortality around the world. The lack of detailed understanding of the cellular and molecular mechanisms participating in the lung tumor progression restrains the development of efficient treatments. Recently, by using state-of-the-art technologies, including in vivo sophisticated Cre/loxP technologies in combination with lung tumor models, it was revealed that osteoblasts activate neutrophils that promote tumor growth in the lung. Strikingly, genetic ablation of osteoblasts abolished lung tumor progression via interruption of SiglecFhigh-expressing neutrophils supply to the tumor microenvironment. Interestingly, SiglecFhigh neutrophil signature was associated with worse lung adenocarcinoma patients outcome. This study identifies novel cellular targets for lung cancer treatment. Here, we summarize and evaluate recent advances in our understanding of lung tumor microenvironment.
Collapse
Affiliation(s)
- Patrick O Azevedo
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Ana E Paiva
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Gabryella S P Santos
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Luiza Lousado
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Julia P Andreotti
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Isadora F G Sena
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Carlos A Tagliati
- Department of Clinical and Toxicological Analysis, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Akiva Mintz
- Department of Radiology, Columbia University Medical Center, New York, NY, USA
| | - Alexander Birbrair
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil. .,Department of Radiology, Columbia University Medical Center, New York, NY, USA.
| |
Collapse
|
31
|
Picoli CC, Coimbra-Campos LMC, Guerra DAP, Silva WN, Prazeres PHDM, Costa AC, Magno LAV, Romano-Silva MA, Mintz A, Birbrair A. Pericytes Act as Key Players in Spinal Cord Injury. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 189:1327-1337. [PMID: 31014955 DOI: 10.1016/j.ajpath.2019.03.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Revised: 03/08/2019] [Accepted: 03/28/2019] [Indexed: 02/06/2023]
Abstract
Spinal cord injury results in locomotor impairment attributable to the formation of an inhibitory fibrous scar, which prevents axonal regeneration after trauma. The scarcity of knowledge about the molecular and cellular mechanisms involved in scar formation after spinal cord lesion impede the design of effective therapies. Recent studies, by using state-of-the-art technologies, including genetic tracking and blockage of pericytes in combination with optogenetics, reveal that pericyte blockage facilitates axonal regeneration and neuronal integration into the local neural circuitry. Strikingly, a pericyte subset is essential during scarring after spinal cord injury, and its arrest results in motor performance improvement. The arising knowledge from current research will contribute to novel approaches to develop therapies for spinal cord injury. We review novel advances in our understanding of pericyte biology in the spinal cord.
Collapse
Affiliation(s)
- Caroline C Picoli
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | | | - Daniel A P Guerra
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Walison N Silva
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Pedro H D M Prazeres
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Alinne C Costa
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Luiz A V Magno
- Department of Mental Health, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Marco A Romano-Silva
- Department of Mental Health, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Akiva Mintz
- Department of Radiology, Columbia University Medical Center, New York, New York
| | - Alexander Birbrair
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, Brazil; Department of Radiology, Columbia University Medical Center, New York, New York.
| |
Collapse
|
32
|
Guerra DAP, Paiva AE, Sena IFG, Azevedo PO, Silva WN, Mintz A, Birbrair A. Targeting glioblastoma-derived pericytes improves chemotherapeutic outcome. Angiogenesis 2018; 21:667-675. [PMID: 29761249 PMCID: PMC6238207 DOI: 10.1007/s10456-018-9621-x] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 05/08/2018] [Indexed: 12/12/2022]
Abstract
Glioblastoma is the most common malignant brain cancer in adults, with poor prognosis. The blood-brain barrier limits the arrival of several promising anti-glioblastoma drugs, and restricts the design of efficient therapies. Recently, by using state-of-the-art technologies, including thymidine kinase targeting system in combination with glioblastoma xenograft mouse models, it was revealed that targeting glioblastoma-derived pericytes improves chemotherapy efficiency. Strikingly, ibrutinib treatment enhances chemotherapeutic effectiveness, by targeting pericytes, improving blood-brain barrier permeability, and prolonging survival. This study identifies glioblastoma-derived pericyte as a novel target in the brain tumor microenvironment during carcinogenesis. Here, we summarize and evaluate recent advances in the understanding of pericyte's role in the glioblastoma microenvironment.
Collapse
Affiliation(s)
- Daniel A P Guerra
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Ana E Paiva
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Isadora F G Sena
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Patrick O Azevedo
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Walison N Silva
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Akiva Mintz
- Department of Radiology, Columbia University Medical Center, New York, NY, USA
| | - Alexander Birbrair
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil.
- Department of Radiology, Columbia University Medical Center, New York, NY, USA.
| |
Collapse
|
33
|
de Alvarenga EC, Silva WN, Vasconcellos R, Paredes-Gamero EJ, Mintz A, Birbrair A. Promyelocytic leukemia protein in mesenchymal stem cells is essential for leukemia progression. Ann Hematol 2018; 97:1749-1755. [PMID: 30069705 DOI: 10.1007/s00277-018-3463-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Accepted: 07/25/2018] [Indexed: 12/28/2022]
Abstract
The dynamic interactions between leukemic cells and cells resident within the bone marrow microenvironment are vital for leukemia progression. The lack of detailed knowledge about the cellular and molecular mechanisms involved in this cross-talk restricts the design of effective treatments. Guarnerio et al. (2018) by using state-of-the-art techniques, including sophisticated Cre/loxP technologies in combination with leukemia mouse models, reveal that mesenchymal stem cells via promyelocytic leukemia protein (Pml) maintain leukemic cells in the bone marrow niche. Strikingly, genetic deletion of Pml in mesenchymal stem cells raised survival of leukemic mice under chemotherapeutic treatment. The emerging knowledge from this research provides a novel target in the bone marrow niche for therapeutic benefit in leukemia.
Collapse
Affiliation(s)
- Erika Costa de Alvarenga
- Department of Natural Sciences, Federal University of São João del Rei, São João Del Rey, MG, Brazil
| | - Walison N Silva
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Rebecca Vasconcellos
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Edgar J Paredes-Gamero
- Department of Biochemistry, Federal University of São Paulo, São Paulo, SP, Brazil.,Faculty of Pharmaceutical Sciences, Food and Nutrition, Federal University of Mato Grosso do Sul, Campo Grande, MS, Brazil
| | - Akiva Mintz
- Department of Radiology, Columbia University Medical Center, New York, NY, USA
| | - Alexander Birbrair
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil. .,Department of Radiology, Columbia University Medical Center, New York, NY, USA.
| |
Collapse
|
34
|
Andreotti JP, Prazeres PHDM, Magno LAV, Romano-Silva MA, Mintz A, Birbrair A. Neurogenesis in the postnatal cerebellum after injury. Int J Dev Neurosci 2018; 67:33-36. [PMID: 29555564 PMCID: PMC6069997 DOI: 10.1016/j.ijdevneu.2018.03.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Revised: 03/02/2018] [Accepted: 03/08/2018] [Indexed: 12/21/2022] Open
Abstract
The cerebellum plays major role in motor coordination and learning. It contains half of the neurons in the brain. Thus, deciphering the mechanisms by which cerebellar neurons are generated is essential to understand the cerebellar functions and the pathologies associated with it. In a recent study, Wojcinski et al. (2017) by using in vivo Cre/loxP technologies reveal that Nestin-expressing progenitors repopulated the external granular cell layer after injury. Depletion of postnatal external granular cell layer is not sufficient to induce motor behavior defects in adults, as the cerebellum recovers these neurons. Strikingly, Nestin-expressing progenitors differentiate into granule cell precursors and mature granule neurons after ablation of perinatal external granular layer, either by irradiation or by genetic ablation. This work identified a novel role of Nestin-expressing progenitors in the cerebellar microenvironment during development, and revealed that extracellular signals can convert specified progenitors into multipotent stem cells. Here, we discuss the findings from this study, and evaluate recent advances in our understanding of the cerebellar neurogenesis.
Collapse
Affiliation(s)
- Julia P Andreotti
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Pedro H D M Prazeres
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Luiz A V Magno
- Department of Mental Health, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Marco A Romano-Silva
- Department of Mental Health, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Akiva Mintz
- Department of Radiology, Columbia University Medical Center, New York, NY, USA
| | - Alexander Birbrair
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil; Department of Radiology, Columbia University Medical Center, New York, NY, USA.
| |
Collapse
|
35
|
Prazeres PHDM, Turquetti AOM, Azevedo PO, Barreto RSN, Miglino MA, Mintz A, Delbono O, Birbrair A. Perivascular cell αv integrins as a target to treat skeletal muscle fibrosis. Int J Biochem Cell Biol 2018; 99:109-113. [PMID: 29627438 PMCID: PMC6159891 DOI: 10.1016/j.biocel.2018.04.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 04/02/2018] [Accepted: 04/03/2018] [Indexed: 02/06/2023]
Abstract
Fibrosis following injury leads to aberrant regeneration and incomplete functional recovery of skeletal muscle, but the lack of detailed knowledge about the cellular and molecular mechanisms involved hampers the design of effective treatments. Using state-of-the-art technologies, Murray et al. (2017) found that perivascular PDGFRβ-expressing cells generate fibrotic cells in the skeletal muscle. Strikingly, genetic deletion of αv integrins from perivascular PDGFRβ-expressing cells significantly inhibited skeletal muscle fibrosis without affecting muscle vascularization or regeneration. In addition, the authors showed that a small molecule inhibitor of αv integrins, CWHM 12, attenuates skeletal muscle fibrosis. From a drug-development perspective, this study identifies a new cellular and molecular target to treat skeletal muscle fibrosis.
Collapse
Affiliation(s)
- Pedro H D M Prazeres
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Anaelise O M Turquetti
- Anatomy of Domestic and Wild Animals Program, Department of Surgery, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, SP, Brazil
| | - Patrick O Azevedo
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Rodrigo S N Barreto
- Anatomy of Domestic and Wild Animals Program, Department of Surgery, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, SP, Brazil
| | - Maria A Miglino
- Anatomy of Domestic and Wild Animals Program, Department of Surgery, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, SP, Brazil
| | - Akiva Mintz
- Department of Radiology, Columbia University Medical Center, New York, NY, USA
| | - Osvaldo Delbono
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Alexander Birbrair
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil; Anatomy of Domestic and Wild Animals Program, Department of Surgery, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, SP, Brazil; Department of Radiology, Columbia University Medical Center, New York, NY, USA.
| |
Collapse
|
36
|
Silva WN, Prazeres PHDM, Paiva AE, Lousado L, Turquetti AOM, Barreto RSN, de Alvarenga EC, Miglino MA, Gonçalves R, Mintz A, Birbrair A. Macrophage-derived GPNMB accelerates skin healing. Exp Dermatol 2018; 27:630-635. [PMID: 29505115 PMCID: PMC6013359 DOI: 10.1111/exd.13524] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/23/2018] [Indexed: 12/22/2022]
Abstract
Healing is a vital response important for the re-establishment of the skin integrity following injury. Delayed or aberrant dermal wound healing leads to morbidity in patients. The development of therapies to improve dermal healing would be useful. Currently, the design of efficient treatments is stalled by the lack of detailed knowledge about the cellular and molecular mechanisms involved in wound healing. Recently, using state-of-the-art technologies, it was revealed that macrophages signal via GPNMB to mesenchymal stem cells, accelerating skin healing. Strikingly, transplantation of macrophages expressing GPNMB improves skin healing in GPNMB-mutant mice. Additionally, topical treatment with recombinant GPNMB restored mesenchymal stem cells recruitment and accelerated wound closure in the diabetic skin. From a drug development perspective, this GPNMB is a new candidate for skin healing.
Collapse
Affiliation(s)
- Walison N. Silva
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | | | - Ana E. Paiva
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Luiza Lousado
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Anaelise O. M. Turquetti
- Anatomy of Domestic and Wild Animals Program, Department of Surgery, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, SP, Brazil
| | - Rodrigo S. N. Barreto
- Anatomy of Domestic and Wild Animals Program, Department of Surgery, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, SP, Brazil
| | - Erika Costa de Alvarenga
- Department of Natural Sciences, Federal University of São João del Rei, São João Del Rey, MG, Brazil
| | - Maria A. Miglino
- Anatomy of Domestic and Wild Animals Program, Department of Surgery, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, SP, Brazil
| | - Ricardo Gonçalves
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Akiva Mintz
- Department of Radiology, Columbia University Medical Center, New York, NY, USA
| | - Alexander Birbrair
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
- Anatomy of Domestic and Wild Animals Program, Department of Surgery, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, SP, Brazil
| |
Collapse
|
37
|
Paiva AE, Lousado L, Guerra DAP, Azevedo PO, Sena IFG, Andreotti JP, Santos GSP, Gonçalves R, Mintz A, Birbrair A. Pericytes in the Premetastatic Niche. Cancer Res 2018; 78:2779-2786. [PMID: 29789421 PMCID: PMC6044472 DOI: 10.1158/0008-5472.can-17-3883] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 01/29/2018] [Accepted: 03/09/2018] [Indexed: 12/20/2022]
Abstract
The premetastatic niche formed by primary tumor-derived molecules contributes to fixation of cancer metastasis. The design of efficient therapies is limited by the current lack of knowledge about the details of cellular and molecular mechanisms involved in the premetastatic niche formation. Recently, the role of pericytes in the premetastatic niche formation and lung metastatic tropism was explored by using state-of-the-art techniques, including in vivo lineage-tracing and mice with pericyte-specific KLF4 deletion. Strikingly, genetic inactivation of KLF4 in pericytes inhibits pulmonary pericyte expansion and decreases metastasis in the lung. Here, we summarize and evaluate recent advances in the understanding of pericyte contribution to premetastatic niche formation. Cancer Res; 78(11); 2779-86. ©2018 AACR.
Collapse
Affiliation(s)
- Ana E Paiva
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Luiza Lousado
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Daniel A P Guerra
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Patrick O Azevedo
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Isadora F G Sena
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Julia P Andreotti
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Gabryella S P Santos
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Ricardo Gonçalves
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Akiva Mintz
- Department of Radiology, Columbia University Medical Center, New York, New York
| | - Alexander Birbrair
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.
- Department of Radiology, Columbia University Medical Center, New York, New York
| |
Collapse
|
38
|
Sena IFG, Paiva AE, Prazeres PHDM, Azevedo PO, Lousado L, Bhutia SK, Salmina AB, Mintz A, Birbrair A. Glioblastoma-activated pericytes support tumor growth via immunosuppression. Cancer Med 2018; 7:1232-1239. [PMID: 29479841 PMCID: PMC5911609 DOI: 10.1002/cam4.1375] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 12/26/2017] [Accepted: 01/10/2018] [Indexed: 12/13/2022] Open
Abstract
Glioblastoma multiforme is the most common and aggressive primary brain tumor, with an extremely poor prognosis. The lack of detailed knowledge about the cellular and molecular mechanisms involved in glioblastoma development restricts the design of efficient therapies. A recent study using state-of-art technologies explores the role of pericytes in the glioblastoma microenvironment. Glioblastoma-activated pericytes develop an immunosuppressive phenotype, reducing T-cell activation through the induction of an anti-inflammatory response. Strikingly, pericytes support glioblastoma growth in vitro and in vivo. Here, we describe succinctly the results and implications of the findings reported in pericytes' and glioblastomas' biology. The emerging knowledge from this study will be essential for the treatment of brain tumors.
Collapse
Affiliation(s)
- Isadora F. G. Sena
- Department of PathologyFederal University of Minas Gerais (UFMG)Belo HorizonteMGBrazil
| | - Ana E. Paiva
- Department of PathologyFederal University of Minas Gerais (UFMG)Belo HorizonteMGBrazil
| | | | - Patrick O. Azevedo
- Department of PathologyFederal University of Minas Gerais (UFMG)Belo HorizonteMGBrazil
| | - Luiza Lousado
- Department of PathologyFederal University of Minas Gerais (UFMG)Belo HorizonteMGBrazil
| | - Sujit K. Bhutia
- Department of Life ScienceNational Institute of TechnologyRourkelaOdishaIndia
| | - Alla B. Salmina
- Department of BiochemistryKrasnoyarsk State Medical UniversityKrasnoyarskRussia
| | - Akiva Mintz
- Department of RadiologyColumbia University Medical CenterNew YorkNew York
| | - Alexander Birbrair
- Department of PathologyFederal University of Minas Gerais (UFMG)Belo HorizonteMGBrazil
| |
Collapse
|
39
|
Costa MA, Paiva AE, Andreotti JP, Cardoso MV, Cardoso CD, Mintz A, Birbrair A. Pericytes constrict blood vessels after myocardial ischemia. J Mol Cell Cardiol 2018; 116:1-4. [PMID: 29371134 PMCID: PMC6089363 DOI: 10.1016/j.yjmcc.2018.01.014] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 01/21/2018] [Indexed: 12/22/2022]
Abstract
No-reflow phenomenon is defined as the reduced blood flow after myocardial ischemia. If prolonged it leads to profound damages in the myocardium. The lack of a detailed knowledge about the cells mediating no-reflow restricts the design of effective therapies. Recently, O'Farrell et al. (2017) by using state-of-the-art technologies, including high-resolution confocal imaging in combination with myocardial ischemia/reperfusion mouse model, reveal that pericytes contribute to the no-reflow phenomenon post-ischemia in the heart. Strikingly, intravenous adenosine increased vascular diameter at pericyte site after cardiac ischemia. This study provides a novel therapeutic target to inhibit no-reflow phenomenon after myocardial ischemia.
Collapse
Affiliation(s)
- Matheus A Costa
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Ana E Paiva
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Julia P Andreotti
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Marcus V Cardoso
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Carlos D Cardoso
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Akiva Mintz
- Department of Radiology, Columbia University Medical Center, New York, NY, USA
| | - Alexander Birbrair
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil; Department of Radiology, Columbia University Medical Center, New York, NY, USA.
| |
Collapse
|
40
|
Guerra DAP, Paiva AE, Sena IFG, Azevedo PO, Batista ML, Mintz A, Birbrair A. Adipocytes role in the bone marrow niche. Cytometry A 2018; 93:167-171. [PMID: 29236351 PMCID: PMC6067923 DOI: 10.1002/cyto.a.23301] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 11/21/2017] [Accepted: 11/28/2017] [Indexed: 12/19/2022]
Abstract
Adipocyte infiltration in the bone marrow follows chemotherapy or irradiation. Previous studies indicate that bone marrow fat cells inhibit hematopoietic stem cell function. Recently, Zhou et al. (2017) using state-of-the-art techniques, including sophisticated Cre/loxP technologies, confocal microscopy, in vivo lineage-tracing, flow cytometry, and bone marrow transplantation, reveal that adipocytes promote hematopoietic recovery after irradiation. This study challenges the current view of adipocytes as negative regulators of the hematopoietic stem cells niche, and reopens the discussion about adipocytes' roles in the bone marrow. Strikingly, genetic deletion of stem cell factor specifically from adipocytes leads to deficiency in hematopoietic stem cells, and reduces animal survival after myeloablation, The emerging knowledge from this research will be important for the treatment of multiple hematologic disorders. © 2017 International Society for Advancement of Cytometry.
Collapse
Affiliation(s)
- Daniel A. P. Guerra
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Ana E. Paiva
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Isadora F. G. Sena
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Patrick O. Azevedo
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Miguel Luiz Batista
- Laboratory of Adipose Tissue Biology, University of Mogi das Cruzes, Mogi das Cruzes, SP, Brazil
| | - Akiva Mintz
- Department of Radiology, Columbia University Medical Center, New York, NY, USA
| | - Alexander Birbrair
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| |
Collapse
|
41
|
Azevedo PO, Lousado L, Paiva AE, Andreotti JP, Santos GSP, Sena IFG, Prazeres PHDM, Filev R, Mintz A, Birbrair A. Endothelial cells maintain neural stem cells quiescent in their niche. Neuroscience 2017; 363:62-65. [PMID: 28893649 PMCID: PMC6089873 DOI: 10.1016/j.neuroscience.2017.08.059] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 08/24/2017] [Accepted: 08/31/2017] [Indexed: 01/02/2023]
Abstract
Niches are specialized microenvironments that regulate stem cells' activity. The neural stem cell (NSC) niche defines a zone in which NSCs are retained and produce new cells of the nervous system throughout life. Understanding the signaling mechanisms by which the niche controls the NSC fate is crucial for the success of clinical applications. In a recent study, Sato and colleagues, by using state-of-the-art techniques, including sophisticated in vivo lineage-tracing technologies, provide evidence that endothelial amyloid precursor protein (APP) is an important component of the NSC niche. Strikingly, depletion of APP increased NSC proliferation in the subventricular zone, indicating that endothelial cells negatively regulate NSCs' growth. The emerging knowledge from this research will be important for the treatment of several neurological diseases.
Collapse
Affiliation(s)
- Patrick O Azevedo
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Luiza Lousado
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Ana E Paiva
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Julia P Andreotti
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Gabryella S P Santos
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Isadora F G Sena
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Pedro H D M Prazeres
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Renato Filev
- Laboratory of Neurobiology, Federal University of São Paulo, SP, Brazil
| | - Akiva Mintz
- Department of Radiology, Columbia University Medical Center, New York, NY, USA
| | - Alexander Birbrair
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil; Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, USA; Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
42
|
Paiva AE, Lousado L, Almeida VM, Andreotti JP, Santos GSP, Azevedo PO, Sena IFG, Prazeres PHDM, Borges IT, Azevedo V, Mintz A, Birbrair A. Endothelial Cells as Precursors for Osteoblasts in the Metastatic Prostate Cancer Bone. Neoplasia 2017; 19:928-931. [PMID: 28957694 PMCID: PMC5619995 DOI: 10.1016/j.neo.2017.08.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 08/20/2017] [Accepted: 08/22/2017] [Indexed: 01/27/2023]
Abstract
Prostate cancer cells metastasize to the bones, causing ectopic bone formation, which results in fractures and pain. The cellular mechanisms underlying new bone production are unknown. In a recent study, Lin and colleagues, by using state-of-the-art techniques, including prostate cancer mouse models in combination with sophisticated in vivo lineage-tracing technologies, revealed that endothelial cells form osteoblasts induced by prostate cancer metastasis in the bone. Strikingly, genetic deletion of osteorix protein from endothelial cells affected prostate cancer-induced osteogenesis in vivo. Deciphering the osteoblasts origin in the bone microenvironment may result in the development of promising new molecular targets for prostate cancer therapy.
Collapse
Affiliation(s)
- Ana E Paiva
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Luiza Lousado
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Viviani M Almeida
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Julia P Andreotti
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Gabryella S P Santos
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Patrick O Azevedo
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Isadora F G Sena
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Pedro H D M Prazeres
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Isabella T Borges
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Vasco Azevedo
- Department of General Biology of Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Akiva Mintz
- Department of Radiology, Columbia University Medical Center, New York, NY, USA
| | - Alexander Birbrair
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil.
| |
Collapse
|
43
|
Sena IFG, Borges IT, Lousado L, Azevedo PO, Andreotti JP, Almeida VM, Paiva AE, Santos GSP, Guerra DAP, Prazeres PHDM, Souto L, Mintz A, Birbrair A. LepR+ cells dispute hegemony with Gli1+ cells in bone marrow fibrosis. Cell Cycle 2017; 16:2018-2022. [PMID: 28976809 DOI: 10.1080/15384101.2017.1367072] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Bone marrow fibrosis is a reactive process, and a central pathological feature of primary myelofibrosis. Revealing the origin of fibroblastic cells in the bone marrow is crucial, as these cells are considered an ideal, and essential target for anti-fibrotic therapy. In 2 recent studies, Decker et al. (2017) and Schneider et al. (2017), by using state-of-the-art techniques including in vivo lineage-tracing, provide evidence that leptin receptor (LepR)-expressing and Gli1-expressing cells are responsible for fibrotic tissue deposition in the bone marrow. However, what is the relationship between these 2 bone marrow cell populations, and what are their relative contributions to bone marrow fibrosis remain unclear. From a drug development perspective, these works bring new cellular targets for bone marrow fibrosis.
Collapse
Affiliation(s)
- Isadora F G Sena
- a Department of Pathology , Federal University of Minas Gerais , Belo Horizonte , MG , Brazil
| | - Isabella T Borges
- a Department of Pathology , Federal University of Minas Gerais , Belo Horizonte , MG , Brazil
| | - Luiza Lousado
- a Department of Pathology , Federal University of Minas Gerais , Belo Horizonte , MG , Brazil
| | - Patrick O Azevedo
- a Department of Pathology , Federal University of Minas Gerais , Belo Horizonte , MG , Brazil
| | - Julia P Andreotti
- a Department of Pathology , Federal University of Minas Gerais , Belo Horizonte , MG , Brazil
| | - Viviani M Almeida
- a Department of Pathology , Federal University of Minas Gerais , Belo Horizonte , MG , Brazil
| | - Ana E Paiva
- a Department of Pathology , Federal University of Minas Gerais , Belo Horizonte , MG , Brazil
| | - Gabryella S P Santos
- a Department of Pathology , Federal University of Minas Gerais , Belo Horizonte , MG , Brazil
| | - Daniel A P Guerra
- a Department of Pathology , Federal University of Minas Gerais , Belo Horizonte , MG , Brazil
| | - Pedro H D M Prazeres
- a Department of Pathology , Federal University of Minas Gerais , Belo Horizonte , MG , Brazil
| | - Luanny Souto
- a Department of Pathology , Federal University of Minas Gerais , Belo Horizonte , MG , Brazil
| | - Akiva Mintz
- b Department of Radiology , Columbia University Medical Center , New York , NY , USA
| | - Alexander Birbrair
- a Department of Pathology , Federal University of Minas Gerais , Belo Horizonte , MG , Brazil.,c Department of Cell Biology , Albert Einstein College of Medicine , Bronx , NY , USA.,d Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine , Bronx , NY , USA
| |
Collapse
|
44
|
Prazeres PHDM, Almeida VM, Lousado L, Andreotti JP, Paiva AE, Santos GSP, Azevedo PO, Souto L, Almeida GG, Filev R, Mintz A, Gonçalves R, Birbrair A. Macrophages Generate Pericytes in the Developing Brain. Cell Mol Neurobiol 2017; 38:777-782. [PMID: 28894964 DOI: 10.1007/s10571-017-0549-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 09/06/2017] [Indexed: 01/01/2023]
Abstract
Pericytes are defined by their anatomical location encircling blood vessels' walls with their long projections. The exact embryonic sources of cerebral pericytes remain poorly understood, especially because of their recently revealed diversity. Yamamoto et al. (Sci Rep 7(1):3855, 2017) using state-of-the-art techniques, including several transgenic mice models, reveal that a subpopulation of brain pericytes are derived from phagocytic macrophages during vascular development. This work highlights a new possible ancestor of brain pericytes. The emerging knowledge from this research may provide new approaches for the treatment of several neurodevelopmental disorders in the future.
Collapse
Affiliation(s)
- Pedro H D M Prazeres
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Viviani M Almeida
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Luiza Lousado
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Julia P Andreotti
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Ana E Paiva
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Gabryella S P Santos
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Patrick O Azevedo
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Luanny Souto
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Gregório G Almeida
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Renato Filev
- Laboratory of Neurobiology, Federal University of São Paulo, São Paulo, SP, Brazil
| | - Akiva Mintz
- Department of Radiology, Columbia University Medical Center, New York, NY, USA
| | - Ricardo Gonçalves
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Alexander Birbrair
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil.
| |
Collapse
|