1
|
Jung SC, Kang D, Ko EA. Roles of PDGF/PDGFR signaling in various organs. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2025; 29:139-155. [PMID: 39482238 PMCID: PMC11842291 DOI: 10.4196/kjpp.24.309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 10/03/2024] [Accepted: 10/07/2024] [Indexed: 11/03/2024]
Abstract
Platelet-derived growth factors (PDGFs) ligands and their corresponding receptors, PDGF receptor (PDGFR)α and PDGFRβ, play a crucial role in controlling diverse biological functions, including cell growth, viability and migration. These growth factors bind to PDGFRs, which are receptor tyrosine kinases present on the surface of target cells. The interaction between PDGFs and PDGFRs induces receptor dimerization and subsequent activation through auto-phosphorylation, which in turn triggers a cascade of intracellular signaling pathways. PDGF/PDGFR signaling is essential for maintaining normal physiological functions, including tissue regeneration and growth. However, dysregulation of this signaling pathway leads to pathological conditions, including fibrosis, atherosclerosis, and cancer development in various organs. The pathological impact of PDGF/PDGFR signaling primarily stems from its capacity to promote excessive cell proliferation, enhanced migration, and increased extracellular matrix deposition, resulting in tissue overgrowth, scarring, and abnormal vessel formation. These processes are integral to the pathogenesis of fibrotic, neoplastic, and vascular disorders. Therefore, understanding these pathways is crucial for developing targeted treatments designed to inhibit PDGF/PDGFR signaling in these diseases. This review delves into the dual role of PDGF/PDGFR signaling in both physiological and pathophysiological contexts across different organs and provides insights into current pharmacological therapies designed to target the PDGF signaling pathway.
Collapse
Affiliation(s)
- Sung-Cherl Jung
- Department of Physiology, College of Medicine, Jeju National University, Jeju 63243, Korea
| | - Dawon Kang
- Department of Physiology, College of Medicine and Institute of Medical Sciences, Gyeongsang National University, Jinju 52727, Korea
| | - Eun-A Ko
- Department of Physiology, College of Medicine, Jeju National University, Jeju 63243, Korea
| |
Collapse
|
2
|
Luo S, Wang Y, Hisatsune T. P2Y1 receptor in Alzheimer's disease. Neural Regen Res 2025; 20:440-453. [PMID: 38819047 PMCID: PMC11317937 DOI: 10.4103/nrr.nrr-d-23-02103] [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: 12/29/2023] [Revised: 03/15/2024] [Accepted: 03/28/2024] [Indexed: 06/01/2024] Open
Abstract
Alzheimer's disease is the most frequent form of dementia characterized by the deposition of amyloid-beta plaques and neurofibrillary tangles consisting of hyperphosphorylated tau. Targeting amyloid-beta plaques has been a primary direction for developing Alzheimer's disease treatments in the last decades. However, existing drugs targeting amyloid-beta plaques have not fully yielded the expected results in the clinic, necessitating the exploration of alternative therapeutic strategies. Increasing evidence unravels that astrocyte morphology and function alter in the brain of Alzheimer's disease patients, with dysregulated astrocytic purinergic receptors, particularly the P2Y1 receptor, all of which constitute the pathophysiology of Alzheimer's disease. These receptors are not only crucial for maintaining normal astrocyte function but are also highly implicated in neuroinflammation in Alzheimer's disease. This review delves into recent insights into the association between P2Y1 receptor and Alzheimer's disease to underscore the potential neuroprotective role of P2Y1 receptor in Alzheimer's disease by mitigating neuroinflammation, thus offering promising avenues for developing drugs for Alzheimer's disease and potentially contributing to the development of more effective treatments.
Collapse
Affiliation(s)
- Shan Luo
- Department of Integrated Biosciences, The University of Tokyo, Kashiwa, Japan
| | - Yifei Wang
- Department of Integrated Biosciences, The University of Tokyo, Kashiwa, Japan
| | - Tatsuhiro Hisatsune
- Department of Integrated Biosciences, The University of Tokyo, Kashiwa, Japan
| |
Collapse
|
3
|
Balzano T, Pineda-Pardo JA, Esteban-García N, López-Aguirre M, Reinares-Sebastián A, Trigo-Damas I, Takada M, Obeso JA, Blesa J. Temporal dynamics of neurovascular unit changes following blood-brain barrier opening in the putamen of non-human primates. J Control Release 2025; 377:116-126. [PMID: 39547418 DOI: 10.1016/j.jconrel.2024.11.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 10/18/2024] [Accepted: 11/09/2024] [Indexed: 11/17/2024]
Abstract
Low-intensity focused ultrasound (LIFU) combined with intravenously circulating microbubbles has recently emerged as a novel approach for increasing delivery through the blood-brain barrier (BBB). This technique safely and transiently enables therapeutic agents to overcome the BBB, which typically poses a significant obstacle for treatment of brain disorders. However, the full impact of LIFU on the entire neurovascular unit (NVU), as well as the mechanisms and factors involved in restoring BBB integrity still require further elucidation. We conducted immunohistochemical analyses of the putamen in non-human primates to monitor changes over time [immediately post-treatment (3 h) and at 7- and 30-days post-BBB opening] in vascular, glial, and immune cells. Additionally, we examined the dynamic interactions among these elements and their role in the restorative process at the BBB level. A mild inflammatory response primarily involving microglia, astrocytes, and T- and B-lymphocytes was observed in the treated putamen acutely after BBB opening. These cells, recruited in response to the vascular changes, stimulate upregulation of PDGFRβ, a pericyte-specific marker, and VEGF-A, a pro-angiogenic factor. This was associated with vascular sprouting by 7 days post-BBB opening. Importantly, no notable long-term alterations were observed in the NVU 30 days post-BBB opening. These results offer further evidence regarding the efficacy and safety of LIFU in achieving BBB opening in the primate brain, indicating that nearly all changes in the NVU revert to baseline within 30 days post-treatment. This also suggests that angiogenesis may play an important role in restoring vascular integrity after BBB opening.
Collapse
Affiliation(s)
- Tiziano Balzano
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain; Instituto de Investigación Sanitaria HM Hospitales.
| | - José A Pineda-Pardo
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain; Instituto de Investigación Sanitaria HM Hospitales; CIBERNED (Center for Networked Biomedical Research on Neurodegenerative Diseases), Instituto Carlos III, Madrid, Spain
| | - Noelia Esteban-García
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain; Instituto de Investigación Sanitaria HM Hospitales; Neuroscience Autonoma de Madrid University-Cajal Institute, Madrid, Spain
| | - Miguel López-Aguirre
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain; Instituto de Investigación Sanitaria HM Hospitales; CIBERNED (Center for Networked Biomedical Research on Neurodegenerative Diseases), Instituto Carlos III, Madrid, Spain; Physics, Complutense University of Madrid, Madrid, Spain
| | - Alejandro Reinares-Sebastián
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain; Instituto de Investigación Sanitaria HM Hospitales; CIBERNED (Center for Networked Biomedical Research on Neurodegenerative Diseases), Instituto Carlos III, Madrid, Spain
| | - Inés Trigo-Damas
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain; Instituto de Investigación Sanitaria HM Hospitales; CIBERNED (Center for Networked Biomedical Research on Neurodegenerative Diseases), Instituto Carlos III, Madrid, Spain; Facultad HM de Ciencias de la Salud de la Universidad Camilo José Cela, Madrid, Spain
| | - Masahiko Takada
- Systems Neuroscience Section, Department of Neuroscience, Primate Research Institute, and Center for the Evolutionary Origins of Human Behavior, Kyoto University, Inuyama, Aichi 484-8506, Japan; Department of Neurology, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - José A Obeso
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain; Instituto de Investigación Sanitaria HM Hospitales; CIBERNED (Center for Networked Biomedical Research on Neurodegenerative Diseases), Instituto Carlos III, Madrid, Spain
| | - Javier Blesa
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain; Instituto de Investigación Sanitaria HM Hospitales; CIBERNED (Center for Networked Biomedical Research on Neurodegenerative Diseases), Instituto Carlos III, Madrid, Spain; Facultad HM de Ciencias de la Salud de la Universidad Camilo José Cela, Madrid, Spain.
| |
Collapse
|
4
|
Yin Q, Yang G, Su R, Bu J, Li Y, Zhang H, Zhang Y, Zhuang P. Zi Shen Wan Fang repaired blood-brain barrier integrity in diabetic cognitive impairment mice via preventing cerebrovascular cells senescence. Chin Med 2024; 19:169. [PMID: 39696612 DOI: 10.1186/s13020-024-01041-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 11/25/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Blood-brain barrier (BBB) integrity disruption is a key pathological link of diabetes-induced cognitive impairment (DCI), but the detailed mechanism of how the diabetic environment induces BBB integrity disruption is not fully understood. Our previous study found that Zi Shen Wan Fang (ZSWF), an optimized prescription consisting of Anemarrhenae Rhizoma (Anemarrhena asphodeloides Bge.), Phellodendri Chinensis Cortex (Phellodendron chinense Schneid.) and Cistanches Herba (Cistanche deserticola Y.C.Ma) has excellent efficacy in alleviating DCI, however, whether its mechanism is related to repairing BBB integrity remains unclear. This study aims to reveal the mechanism of BBB integrity destruction in DCI mice, and to elucidate the mechanism by which ZSWF repairs BBB integrity and improves cognitive function in DCI mice. METHODS Diabetic mouse model was established by feeding a 60% high-fat diet combined with a single intraperitoneal injection of 120 mg/kg streptozotocin (STZ). DCI mice were screened with morris water maze (MWM) after 8 weeks of sustained hyperglycemic stimulation. ZSWF was administered daily at doses of 9.36 and 18.72 g/kg for 8 weeks. Cognitive function was evaluated using MWM, blood-brain-barrier (BBB) integrity was tested using immunostaining and western blot, the underlying mechanisms were explored using single-cell RNA sequencing (scRNA-seq), validation experiments were performed with immunofluorescence analysis, and the potential active ingredients of ZSWF against cerebrovascular senescence were predicted using molecular docking. Moreover, cerebral microvascular endothelial cells were cultured, and the effects of mangiferin on the expression of p21 and Vcam1 were investigated by immunofluorescence staining and RT-qPCR. RESULTS ZSWF treatment significantly ameliorated cognitive function and repaired BBB integrity in DCI mice. Using scRNA-seq, we identified 14 brain cell types. In BBB constituent cells (endothelial cells and pericytes), we found that Cdkn1a and senescence-associated secretory phenotype (SASP) genes were significantly overexpressed in DCI mice, while ZSWF intervention significantly inhibited the expression of Cdkn1a and SASP genes in cerebrovascular cells of DCI mice. Moreover, we also found that the communication between brain endothelial cells and pericytes was decreased in DCI mice, while ZSWF significantly increased the communication between them, especially the expression of PDGFRβ in pericytes. Molecular docking results showed that mangiferin, the blood component of ZSWF, had a stronger affinity with the upstream proteins of p21. In vitro experiments showed that high glucose significantly increased the expression of p21 and Vcam1 in bEnd.3 cells, while mangiferin significantly inhibited the expression of p21 and Vcam1 induced by high glucose. CONCLUSION Our study reveals that ZSWF can ameliorate cognitive function in DCI mice by repairing BBB integrity, and the specific mechanism of which may be related to preventing cerebrovascular cells senescence, and mangiferin is its key active ingredient.
Collapse
Affiliation(s)
- Qingsheng Yin
- State Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Jinghai District, Tianjin, 301617, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Genhui Yang
- State Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Jinghai District, Tianjin, 301617, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Runtao Su
- State Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Jinghai District, Tianjin, 301617, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Jie Bu
- State Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Jinghai District, Tianjin, 301617, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Ying Li
- State Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Jinghai District, Tianjin, 301617, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Han Zhang
- State Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Jinghai District, Tianjin, 301617, China.
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| | - Yanjun Zhang
- State Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Jinghai District, Tianjin, 301617, China.
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
- Department of Integrated Rehabilitation, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China.
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China.
| | - Pengwei Zhuang
- State Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Jinghai District, Tianjin, 301617, China.
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
- Department of Integrated Rehabilitation, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China.
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China.
| |
Collapse
|
5
|
Jiang H, Zhang C, Lin M, Yin Y, Deng S, Liu W, Zhuo B, Tian G, Du Y, Meng Z. Deciphering the mechanistic impact of acupuncture on the neurovascular unit in acute ischemic stroke: Insights from basic research in a narrative review. Ageing Res Rev 2024; 101:102536. [PMID: 39384155 DOI: 10.1016/j.arr.2024.102536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 09/27/2024] [Accepted: 10/01/2024] [Indexed: 10/11/2024]
Abstract
Ischemic stroke(IS), a severe acute cerebrovascular disease, not only imposes a heavy economic burden on society but also presents numerous challenges in treatment. During the acute phase, while thrombolysis and thrombectomy serve as primary treatments, these approaches are restricted by a narrow therapeutic window. During rehabilitation, commonly used neuroprotective agents struggle with their low drug delivery efficiency and inadequate preclinical testing, and the long-term pharmacological and toxicity effects of nanomedicines remain undefined. Meanwhile, acupuncture as a therapeutic approach is widely acknowledged for its effectiveness in treating IS and has been recommended by the World Health Organization (WHO) as an alternative and complementary therapy, even though its exact mechanisms remain unclear. This review aims to summarize the known mechanisms of acupuncture and electroacupuncture (EA) in the treatment of IS. Research shows that acupuncture treatment mainly protects the neurovascular unit through five mechanisms: 1) reducing neuronal apoptosis and promoting neuronal repair and proliferation; 2) maintaining the integrity of the blood-brain barrier (BBB); 3) inhibiting the overactivation and polarization imbalance of microglia; 4) regulating the movement of vascular smooth muscle (VSM) cells; 5) promoting the proliferation of oligodendrocyte precursors. Through an in-depth analysis, this review reveals the multi-level, multi-dimensional impact of acupuncture treatment on the neurovascular unit (NVU) following IS, providing stronger evidence and a theoretical basis for its clinical application.
Collapse
Affiliation(s)
- Hailun Jiang
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Chao Zhang
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Mengxuan Lin
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Yu Yin
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Shizhe Deng
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Wei Liu
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Bifang Zhuo
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Guang Tian
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Yuzheng Du
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China.
| | - Zhihong Meng
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China.
| |
Collapse
|
6
|
Tseng PH, Huang LC, Huang XL, Huang BR, Lin SZ, Tsai ST, Huang HY. Blood-brain barrier-associated biomarker correlated with cerebral small vessel disease and shunt outcome in normal pressure hydrocephalus: a prospective cohort study. Int J Surg 2024; 110:6962-6971. [PMID: 39166950 PMCID: PMC11573114 DOI: 10.1097/js9.0000000000002038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 07/30/2024] [Indexed: 08/23/2024]
Abstract
BACKGROUND Blood-brain barrier (BBB) breakdown is associated with neurodegeneration and cognitive impairment. Cerebral small vessel disease (CSVD) is also common in idiopathic normal pressure hydrocephalus (iNPH). Biomarkers in the cerebrospinal fluid (CSF) may reflect the severity of neuropathological damage and indicate a relationship between BBB integrity and iNPH and its surgical outcome. The authors investigated the association of CSVD and comorbidity-related CSF biomarkers with shunt outcomes in iNPH. MATERIALS AND METHODS This prospective cohort study recruited 53 patients with iNPH, who were subgrouped by CSVD severity. CSF proteins were analyzed, including soluble platelet-derived growth factor receptor-β (sPDGFR-β), Alzheimer's disease biomarkers, neurofilament light chain (NfL), and triggering receptor expressed on myeloid cells 2 (Trem2). We assessed symptom improvement, investigated its association with biomarkers levels, calculated protein cutoffs for surgical outcomes using receiver operating characteristic (ROC) curves, and compared model predictions using different proteins through hierarchical regression analysis. RESULTS Among patients with iNPH, 74% had comorbid CSVD. Patients with severe CSVD exhibited significantly higher sPDGFR-β levels ( P =0.019) and better postoperative performance (β=0.332, t=2.174, P =0.039; r =0.573, P =0.001). Analysis of the predictive potential of the biomarkers showed that sPDGFR-β was predictive of surgical outcomes (area under curve=0.82, sensitivity=66.8%, specificity=94.7%). A Comparison of the models revealed a greater effect of sPDGFR-β (Adjusted R 2 =0.247, ∆R 2 =0.160, ∆F(1, 37)=8.238, P =0.007) on cognitive improvement. CONCLUSION This study highlighted the relevance of CSF biomarkers in assessing CSVD severity and predicting iNPH surgical outcomes. CSF shunt surgery may provide an alternative treatment for neurodegenerative diseases with BBB breakdown and dysfunctional CSF clearance.
Collapse
Affiliation(s)
- Pao-Hui Tseng
- Department of Neurosurgery, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation
- Department of Nursing, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation
- Institute of Medical Sciences, Tzu Chi University
| | - Li-Chuan Huang
- Department of Medical Imaging, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien
- Department of Medical Imaging and Radiological Sciences, Tzu Chi University, Hualien, Taiwan
| | - Xiang-Ling Huang
- Department of Neurosurgery, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation
- Department of Nursing, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation
- Institute of Medical Sciences, Tzu Chi University
| | - Bor-Ren Huang
- Department of Neurosurgery, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung
- School of Medicine, Tzu Chi University
| | - Shinn-Zong Lin
- Department of Neurosurgery, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation
- Buddhist Tzu Chi Bioinnovation Center, Buddhist Tzu Chi Medical Foundation
| | - Sheng-Tzung Tsai
- Department of Neurosurgery, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation
- Institute of Medical Sciences, Tzu Chi University
- School of Medicine, Tzu Chi University
| | - Hsin-Yi Huang
- Department of Medical Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation
| |
Collapse
|
7
|
Rust R, Nih LR, Liberale L, Yin H, El Amki M, Ong LK, Zlokovic BV. Brain repair mechanisms after cell therapy for stroke. Brain 2024; 147:3286-3305. [PMID: 38916992 PMCID: PMC11449145 DOI: 10.1093/brain/awae204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 06/04/2024] [Accepted: 06/08/2024] [Indexed: 06/27/2024] Open
Abstract
Cell-based therapies hold great promise for brain repair after stroke. While accumulating evidence confirms the preclinical and clinical benefits of cell therapies, the underlying mechanisms by which they promote brain repair remain unclear. Here, we briefly review endogenous mechanisms of brain repair after ischaemic stroke and then focus on how different stem and progenitor cell sources can promote brain repair. Specifically, we examine how transplanted cell grafts contribute to improved functional recovery either through direct cell replacement or by stimulating endogenous repair pathways. Additionally, we discuss recently implemented preclinical refinement methods, such as preconditioning, microcarriers, genetic safety switches and universal (immune evasive) cell transplants, as well as the therapeutic potential of these pharmacologic and genetic manipulations to further enhance the efficacy and safety of cell therapies. By gaining a deeper understanding of post-ischaemic repair mechanisms, prospective clinical trials may be further refined to advance post-stroke cell therapy to the clinic.
Collapse
Affiliation(s)
- Ruslan Rust
- Department of Physiology and Neuroscience, University of Southern California, Los Angeles, CA 90033, USA
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Institute for Regenerative Medicine, University of Zurich, 8952 Schlieren, Switzerland
| | - Lina R Nih
- Department of Brain Health, University of Nevada, Las Vegas, NV 89154, USA
| | - Luca Liberale
- Department of Internal Medicine, University of Genoa, 16132 Genova, Italy
- IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
| | - Hao Yin
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 3K7, Canada
| | - Mohamad El Amki
- Department of Neurology, University Hospital and University of Zurich, 8091 Zurich, Switzerland
| | - Lin Kooi Ong
- School of Health and Medical Sciences & Centre for Health Research, University of Southern Queensland, Toowoomba, QLD 4350, Australia
| | - Berislav V Zlokovic
- Department of Physiology and Neuroscience, University of Southern California, Los Angeles, CA 90033, USA
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
8
|
Ding X, Chen Y, Zhang X, Duan Y, Yuan G, Liu C. Research progress on the protection and mechanism of active peptides in Alzheimer's disease and Parkinson's disease. Neuropeptides 2024; 107:102457. [PMID: 39068763 DOI: 10.1016/j.npep.2024.102457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 07/21/2024] [Accepted: 07/23/2024] [Indexed: 07/30/2024]
Abstract
Neurodegenerative diseases are the main causes of death and morbidity among elderly people worldwide. From the pathological point of view, oxidative stress, neuroinflammation, mitochondrial damage and apoptosis are the causes of neuronal diseases, and play a harmful role in the process of neuronal cell death and neurodegeneration. The most common neurodegenerative diseases are Alzheimer's disease(AD) and Parkinson's disease(PD), and there is no effective treatment. The physiological role of active peptides in the human body is significant. Modern medical research has found that animal and plant peptides, natural peptides in human body, can act on the central nervous system, and their active components can improve learning and memory ability, and play the roles of antioxidation, anti-inflammation, anti-apoptosis and maintaining the structure and function of mitochondria. This review reviews the reports on neurodegenerative diseases such as AD and PD by active peptides from animals and plants and natural peptides from the human body, and summarizes the neuroprotective mechanism of peptides. A theoretical basis for further research and development of active peptides was provided by examining the research and application of peptides, which provided a theoretical basis for further research and development.
Collapse
Affiliation(s)
- Xuying Ding
- College of Pharmacy, Beihua University, Jilin, Jilin 132013, PR China
| | - Yutong Chen
- College of Pharmacy, Beihua University, Jilin, Jilin 132013, PR China
| | - Xiaojun Zhang
- State key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Changchun, jilin 130022, PR China
| | - Yanming Duan
- College of Pharmacy, Beihua University, Jilin, Jilin 132013, PR China
| | - Guojing Yuan
- College of Pharmacy, Beihua University, Jilin, Jilin 132013, PR China
| | - Chang Liu
- College of Pharmacy, Beihua University, Jilin, Jilin 132013, PR China.
| |
Collapse
|
9
|
Li ZR, Wang YY, Wang ZH, Qin QL, Huang C, Shi GS, He HY, Deng YH, He XY, Zhao XM. The positive role of transforming growth factor-β1 in ischemic stroke. Cell Signal 2024; 121:111301. [PMID: 39019338 DOI: 10.1016/j.cellsig.2024.111301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 07/05/2024] [Accepted: 07/12/2024] [Indexed: 07/19/2024]
Abstract
Ischemic stroke is one of the most disabling and fatal diseases around the world. The damaged brain tissues will undergo excessive autophagy, vascular endothelial cells injury, blood-brain barrier (BBB) impairment and neuroinflammation after ischemic stroke. However, there is no unified viewpoint on the underlying mechanism of brain damage. Transforming growth factor-β1 (TGF-β1), as a multi-functional cytokine, plays a crucial role in the intricate pathological processes and helps maintain the physiological homeostasis of brain tissues through various signaling pathways after ischemic stroke. In this review, we summarize the protective role of TGF-β1 in autophagic flux, BBB, vascular remodeling, neuroinflammation and other aspects after ischemic stroke. Based on the review, we believe that TGF-β1 could serve as a key target for treating ischemic stroke.
Collapse
Affiliation(s)
- Zi-Rong Li
- Faculty of Medicine, Kunming University of Science and Technology, Kunming, China.
| | - Yong-Yan Wang
- Faculty of Medicine, Kunming University of Science and Technology, Kunming, China.
| | - Zi-Han Wang
- Faculty of Medicine, Kunming University of Science and Technology, Kunming, China.
| | - Qi-Lin Qin
- Faculty of Medicine, Kunming University of Science and Technology, Kunming, China.
| | - Cheng Huang
- Faculty of Medicine, Kunming University of Science and Technology, Kunming, China.
| | - Guang-Sen Shi
- Faculty of Medicine, Kunming University of Science and Technology, Kunming, China.
| | - Hong-Yun He
- Faculty of Medicine, Kunming University of Science and Technology, Kunming, China; Anning First People's Hospital Affiliated to Kunming University of Science and Technology, Kunming, China.
| | - Yi-Hao Deng
- Faculty of Medicine, Kunming University of Science and Technology, Kunming, China.
| | - Xiu-Ying He
- Department of Anesthesiology, Institute of Neurological Disease, West China Hospital, Sichuan University, Chengdu, China.
| | - Xiao-Ming Zhao
- Faculty of Medicine, Kunming University of Science and Technology, Kunming, China; Anning First People's Hospital Affiliated to Kunming University of Science and Technology, Kunming, China.
| |
Collapse
|
10
|
Meijer WC, Gorter JA. Role of blood-brain barrier dysfunction in the development of poststroke epilepsy. Epilepsia 2024; 65:2519-2536. [PMID: 39101543 DOI: 10.1111/epi.18072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 07/12/2024] [Accepted: 07/17/2024] [Indexed: 08/06/2024]
Abstract
Stroke is a major contributor to mortality and morbidity worldwide and the most common cause of epilepsy in the elderly in high income nations. In recent years, it has become increasingly evident that both ischemic and hemorrhagic strokes induce dysfunction of the blood-brain barrier (BBB), and that this impairment can contribute to epileptogenesis. Nevertheless, studies directly comparing BBB dysfunction and poststroke epilepsy (PSE) are largely absent. Therefore, this review summarizes the role of BBB dysfunction in the development of PSE in animal models and clinical studies. There are multiple mechanisms whereby stroke induces BBB dysfunction, including increased transcytosis, tight junction dysfunction, spreading depolarizations, astrocyte and pericyte loss, reactive astrocytosis, angiogenesis, matrix metalloproteinase activation, neuroinflammation, adenosine triphosphate depletion, oxidative stress, and finally cell death. The degree to which these effects occur is dependent on the severity of the ischemia, whereby cell death is a more prominent mechanism of BBB disruption in regions of critical ischemia. BBB dysfunction can contribute to epileptogenesis by increasing the risk of hemorrhagic transformation, increasing stroke size and the amount of cerebral vasogenic edema, extravasation of excitatory compounds, and increasing neuroinflammation. Furthermore, albumin extravasation after BBB dysfunction contributes to epileptogenesis primarily via increased transforming growth factor β signaling. Finally, seizures themselves induce BBB dysfunction, thereby contributing to epileptogenesis in a cyclical manner. In repairing this BBB dysfunction, pericyte migration via platelet-derived growth factor β signaling is indispensable and required for reconstruction of the BBB, whereby astrocytes also play a role. Although animal stroke models have their limitations, they provide valuable insights into the development of potential therapeutics designed to restore the BBB after stroke, with the ultimate goal of improving outcomes and minimizing the occurrence of PSE. In pursuit of this goal, rapamycin, statins, losartan, semaglutide, and metformin show promise, whereby modulation of pericyte migration could also be beneficial.
Collapse
Affiliation(s)
- Wouter C Meijer
- Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Amsterdam, the Netherlands
| | - Jan A Gorter
- Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
11
|
Roth M, Carlsson R, Buizza C, Enström A, Paul G. Pericyte response to ischemic stroke precedes endothelial cell death and blood-brain barrier breakdown. J Cereb Blood Flow Metab 2024:271678X241261946. [PMID: 39053491 PMCID: PMC11571979 DOI: 10.1177/0271678x241261946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 05/14/2024] [Accepted: 05/18/2024] [Indexed: 07/27/2024]
Abstract
Stroke is one of the leading causes of death and disability, yet the cellular response to the ischemic insult is poorly understood limiting therapeutic options. Brain pericytes are crucial for maintaining blood-brain barrier (BBB) integrity and are known to be one of the first responders to ischemic stroke. The exact timeline of cellular events after stroke, however, remains elusive. Using the permanent middle cerebral artery occlusion stroke model, we established a detailed timeline of microvascular events after experimental stroke. Our results show that pericytes respond already within 1 hour after the ischemic insult. We find that approximately 30% of the pericyte population dies as early as 1 hour after stroke, while ca 50% express markers that indicate activation. A decrease of endothelial tight junctions, signs of endothelial cell death and reduction in blood vessel length are only detected at time points after the initial pericyte response. Consistently, markers of BBB leakage are observed several hours after pericyte cell death and/or vascular detachment. Our results suggest that the pericyte response to stroke occurs early and precedes both the endothelial response and the BBB breakdown. This highlights pericytes as an important target cell type to develop new diagnostic and therapeutic tools.
Collapse
Affiliation(s)
- Michaela Roth
- Translational Neurology Group, Department of Clinical Science, Wallenberg Neuroscience Center, Lund University, Lund, Sweden
| | - Robert Carlsson
- Translational Neurology Group, Department of Clinical Science, Wallenberg Neuroscience Center, Lund University, Lund, Sweden
| | - Carolina Buizza
- Translational Neurology Group, Department of Clinical Science, Wallenberg Neuroscience Center, Lund University, Lund, Sweden
| | - Andreas Enström
- Translational Neurology Group, Department of Clinical Science, Wallenberg Neuroscience Center, Lund University, Lund, Sweden
| | - Gesine Paul
- Translational Neurology Group, Department of Clinical Science, Wallenberg Neuroscience Center, Lund University, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
- Department of Neurology, Scania University Hospital, Lund, Sweden
| |
Collapse
|
12
|
Shuvalova M, Dmitrieva A, Belousov V, Nosov G. The role of reactive oxygen species in the regulation of the blood-brain barrier. Tissue Barriers 2024:2361202. [PMID: 38808582 DOI: 10.1080/21688370.2024.2361202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 05/23/2024] [Indexed: 05/30/2024] Open
Abstract
The blood-brain barrier (BBB) regulates the exchange of metabolites and cells between the blood and brain, and maintains central nervous system homeostasis. Various factors affect BBB barrier functions, including reactive oxygen species (ROS). ROS can act as stressors, damaging biological molecules, but they also serve as secondary messengers in intracellular signaling cascades during redox signaling. The impact of ROS on the BBB has been observed in multiple sclerosis, stroke, trauma, and other neurological disorders, making blocking ROS generation a promising therapeutic strategy for BBB dysfunction. However, it is important to consider ROS generation during normal BBB functioning for signaling purposes. This review summarizes data on proteins expressed by BBB cells that can be targets of redox signaling or oxidative stress. It also provides examples of signaling molecules whose impact may cause ROS generation in the BBB, as well as discusses the most common diseases associated with BBB dysfunction and excessive ROS generation, open questions that arise in the study of this problem, and possible ways to overcome them.
Collapse
Affiliation(s)
- Margarita Shuvalova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia
- Department of metabolism and redox biology, Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Anastasiia Dmitrieva
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Vsevolod Belousov
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia
- Department of metabolism and redox biology, Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, Moscow, Russia
- Life Improvement by Future Technologies (LIFT) Center, Skolkovo, Moscow, Russia
| | - Georgii Nosov
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia
- Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, Moscow, Russia
- Life Improvement by Future Technologies (LIFT) Center, Skolkovo, Moscow, Russia
| |
Collapse
|
13
|
Bernard M, Menet R, Lecordier S, ElAli A. Endothelial PDGF-D contributes to neurovascular protection after ischemic stroke by rescuing pericyte functions. Cell Mol Life Sci 2024; 81:225. [PMID: 38769116 PMCID: PMC11106055 DOI: 10.1007/s00018-024-05244-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 03/29/2024] [Accepted: 04/19/2024] [Indexed: 05/22/2024]
Abstract
Ischemic stroke induces neovascularization of the injured tissue as an attempt to promote structural repair and neurological recovery. Angiogenesis is regulated by pericytes that potently react to ischemic stroke stressors, ranging from death to dysfunction. Platelet-derived growth factor (PDGF) receptor (PDGFR)β controls pericyte survival, migration, and interaction with brain endothelial cells. PDGF-D a specific ligand of PDGFRβ is expressed in the brain, yet its regulation and role in ischemic stroke pathobiology remains unexplored. Using experimental ischemic stroke mouse model, we found that PDGF-D is transiently induced in brain endothelial cells at the injury site in the subacute phase. To investigate the biological significance of PDGF-D post-ischemic stroke regulation, its subacute expression was either downregulated using siRNA or upregulated using an active recombinant form. Attenuation of PDGF-D subacute induction exacerbates neuronal loss, impairs microvascular density, alters vascular permeability, and increases microvascular stalling. Increasing PDGF-D subacute bioavailability rescues neuronal survival and improves neurological recovery. PDGF-D subacute enhanced bioavailability promotes stable neovascularization of the injured tissue and improves brain perfusion. Notably, PDGF-D enhanced bioavailability improves pericyte association with brain endothelial cells. Cell-based assays using human brain pericyte and brain endothelial cells exposed to ischemia-like conditions were applied to investigate the underlying mechanisms. PDGF-D stimulation attenuates pericyte loss and fibrotic transition, while increasing the secretion of pro-angiogenic and vascular protective factors. Moreover, PDGF-D stimulates pericyte migration required for optimal endothelial coverage and promotes angiogenesis. Our study unravels new insights into PDGF-D contribution to neurovascular protection after ischemic stroke by rescuing the functions of pericytes.
Collapse
Affiliation(s)
- Maxime Bernard
- Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Quebec City, QC, Canada
- Neuroscience Axis, Research Center of CHU de Québec (CHUQ)-Université Laval, 2705 Laurier Boulevard, Quebec City, QC, G1V 4G2, Canada
| | - Romain Menet
- Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Quebec City, QC, Canada
- Neuroscience Axis, Research Center of CHU de Québec (CHUQ)-Université Laval, 2705 Laurier Boulevard, Quebec City, QC, G1V 4G2, Canada
| | - Sarah Lecordier
- Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Quebec City, QC, Canada
- Neuroscience Axis, Research Center of CHU de Québec (CHUQ)-Université Laval, 2705 Laurier Boulevard, Quebec City, QC, G1V 4G2, Canada
| | - Ayman ElAli
- Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Quebec City, QC, Canada.
- Neuroscience Axis, Research Center of CHU de Québec (CHUQ)-Université Laval, 2705 Laurier Boulevard, Quebec City, QC, G1V 4G2, Canada.
| |
Collapse
|
14
|
Chen X, Luo J, Song M, Pan L, Qu Z, Huang B, Yu S, Shu H. Challenges and prospects in geriatric epilepsy treatment: the role of the blood-brain barrier in pharmacotherapy and drug delivery. Front Aging Neurosci 2024; 16:1342366. [PMID: 38389560 PMCID: PMC10882099 DOI: 10.3389/fnagi.2024.1342366] [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: 11/21/2023] [Accepted: 01/29/2024] [Indexed: 02/24/2024] Open
Abstract
The blood-brain barrier (BBB) is pivotal in maintaining neuronal physiology within the brain. This review delves into the alterations of the BBB specifically in the context of geriatric epilepsy. We examine how age-related changes in the BBB contribute to the pathogenesis of epilepsy in the elderly and present significant challenges in pharmacotherapy. Subsequently, we evaluate recent advancements in drug delivery methods targeting the BBB, as well as alternative approaches that could bypass the BBB's restrictive nature. We particularly highlight the use of neurotropic viruses and various synthetic nanoparticles that have been investigated for delivering a range of antiepileptic drugs. Additionally, the advantage and limitation of these diverse delivery methods are discussed. Finally, we analyze the potential efficacy of different drug delivery approaches in the treatment of geriatric epilepsy, aiming to provide insights into more effective management of this condition in the elderly population.
Collapse
Affiliation(s)
- Xin Chen
- Department of Neurosurgery, Western Theater General Hospital, Chengdu, Sichuan, China
| | - Juan Luo
- Department of Neurosurgery, Western Theater General Hospital, Chengdu, Sichuan, China
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Min Song
- Department of Neurosurgery, Western Theater General Hospital, Chengdu, Sichuan, China
| | - Liang Pan
- Department of Pediatrics, Western Theater General Hospital, Chengdu, Sichuan, China
| | - Zhichuang Qu
- Department of Neurosurgery, Meishan City People's Hospital, Meishan, Sichuan, China
| | - Bo Huang
- Department of Burn and Plastic, Western Theater General Hospital, Chengdu, Sichuan, China
| | - Sixun Yu
- Department of Neurosurgery, Western Theater General Hospital, Chengdu, Sichuan, China
| | - Haifeng Shu
- Department of Neurosurgery, Western Theater General Hospital, Chengdu, Sichuan, China
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
15
|
Okekawa A, Wada T, Onogi Y, Takeda Y, Miyazawa Y, Sasahara M, Tsuneki H, Sasaoka T. Platelet-derived growth factor signaling in pericytes promotes hypothalamic inflammation and obesity. Mol Med 2024; 30:21. [PMID: 38317079 PMCID: PMC10845801 DOI: 10.1186/s10020-024-00793-z] [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: 10/05/2023] [Accepted: 01/25/2024] [Indexed: 02/07/2024] Open
Abstract
BACKGROUND Pericytes are a vital component of the blood-brain barrier, and their involvement in acute inflammation was recently suggested. However, it remains unclear whether pericytes contribute to hypothalamic chronic inflammation and energy metabolism in obesity. The present study investigated the impact of pericytes on the pathophysiology of obesity by focusing on platelet-derived growth factor (PDGF) signaling, which regulates pericyte functions. METHODS Tamoxifen-inducible systemic conditional PDGF receptor β knockout mice (Pdgfrb∆SYS-KO) and Calcium/calmodulin-dependent protein kinase type IIa (CaMKIIa)-positive neuron-specific PDGF receptor β knockout mice (Pdgfrb∆CaMKII-KO) were fed a high-fat diet, and metabolic phenotypes before and 3 to 4 weeks after dietary loading were examined. Intracellular energy metabolism and relevant signal transduction in lipopolysaccharide- and/or platelet-derived growth factor-BB (PDGF-BB)-stimulated human brain pericytes (HBPCs) were assessed by the Seahorse XFe24 Analyzer and Western blotting. The pericyte secretome in conditioned medium from HBPCs was studied using cytokine array kit, and its impact on polarization was examined in bone marrow-derived macrophages (BMDMs), which are microglia-like cells. RESULTS Energy consumption increased and body weight gain decreased after high-fat diet loading in Pdgfrb∆SYS-KO mice. Cellular oncogene fos (cFos) expression increased in proopiomelanocortin (POMC) neurons, whereas microglial numbers and inflammatory gene expression decreased in the hypothalamus of Pdgfrb∆SYS-KO mice. No significant changes were observed in Pdgfrb∆CaMKII-KO mice. In HBPCs, a co-stimulation with lipopolysaccharide and PDGF-BB shifted intracellular metabolism towards glycolysis, activated mitogen-activated protein kinase (MAPK), and modulated the secretome to the inflammatory phenotype. Consequently, the secretome showed an increase in various proinflammatory chemokines and growth factors including Epithelial-derived neutrophil-activating peptide 78 (C-X-C motif chemokine ligand (CXCL)5), Thymus and activation-regulated chemokine (C-C motif chemokine (CCL)17), Monocyte chemoattractant protein 1 (CCL2), and Growth-regulated oncogene α (CXCL1). Furthermore, conditioned medium from HBPCs stimulated the inflammatory priming of BMDMs, and this change was abolished by the C-X-C motif chemokine receptor (CXCR) inhibitor. Consistently, mRNA expression of CXCL5 was elevated by lipopolysaccharide and PDGF-BB treatment in HBPCs, and the expression was significantly lower in the hypothalamus of Pdgfrb∆SYS-KO mice than in control Pdgfrbflox/flox mice (FL) following 4 weeks of HFD feeding. CONCLUSIONS PDGF receptor β signaling in hypothalamic pericytes promotes polarization of macrophages by changing their secretome and contributes to the progression of obesity.
Collapse
Affiliation(s)
- Akira Okekawa
- Department of Clinical Pharmacology, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Tsutomu Wada
- Department of Clinical Pharmacology, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan.
| | - Yasuhiro Onogi
- Department of Clinical Pharmacology, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
- Research Center for Pre-Disease Science, University of Toyama, 2630 Sugitani, Toyama, Japan
| | - Yuki Takeda
- Department of Clinical Pharmacology, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Yuichiro Miyazawa
- Department of Clinical Pharmacology, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Masakiyo Sasahara
- Department of Pathology, University of Toyama, 2630 Sugitani, Toyama, Japan
| | - Hiroshi Tsuneki
- Department of Clinical Pharmacology, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
- Department of Integrative Pharmacology, University of Toyama, 2630 Sugitani, Toyama, Japan
| | - Toshiyasu Sasaoka
- Department of Clinical Pharmacology, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan.
| |
Collapse
|
16
|
Rademeyer KM, R Nass S, Jones AM, Ohene-Nyako M, Hauser KF, McRae M. Fentanyl dysregulates neuroinflammation and disrupts blood-brain barrier integrity in HIV-1 Tat transgenic mice. J Neurovirol 2024; 30:1-21. [PMID: 38280928 PMCID: PMC11232468 DOI: 10.1007/s13365-023-01186-4] [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: 06/17/2023] [Revised: 10/29/2023] [Accepted: 11/16/2023] [Indexed: 01/29/2024]
Abstract
Opioid overdose deaths have dramatically increased by 781% from 1999 to 2021. In the setting of HIV, opioid drug abuse exacerbates neurotoxic effects of HIV in the brain, as opioids enhance viral replication, promote neuronal dysfunction and injury, and dysregulate an already compromised inflammatory response. Despite the rise in fentanyl abuse and the close association between opioid abuse and HIV infection, the interactive comorbidity between fentanyl abuse and HIV has yet to be examined in vivo. The HIV-1 Tat-transgenic mouse model was used to understand the interactive effects between fentanyl and HIV. Tat is an essential protein produced during HIV that drives the transcription of new virions and exerts neurotoxic effects within the brain. The Tat-transgenic mouse model uses a glial fibrillary acidic protein (GFAP)-driven tetracycline promoter which limits Tat production to the brain and this model is well used for examining mechanisms related to neuroHIV. After 7 days of fentanyl exposure, brains were harvested. Tight junction proteins, the vascular cell adhesion molecule, and platelet-derived growth factor receptor-β were measured to examine the integrity of the blood brain barrier. The immune response was assessed using a mouse-specific multiplex chemokine assay. For the first time in vivo, we demonstrate that fentanyl by itself can severely disrupt the blood-brain barrier and dysregulate the immune response. In addition, we reveal associations between inflammatory markers and tight junction proteins at the blood-brain barrier.
Collapse
Affiliation(s)
- Kara M Rademeyer
- Department of Pharmacotherapy and Outcomes Science, Virginia Commonwealth University, Richmond, VA, 23298, U.S.A
| | - Sara R Nass
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, U.S.A
| | - Austin M Jones
- Department of Pharmacotherapy and Outcomes Science, Virginia Commonwealth University, Richmond, VA, 23298, U.S.A
| | - Michael Ohene-Nyako
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, U.S.A
| | - Kurt F Hauser
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, U.S.A
| | - MaryPeace McRae
- Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA, 22908, U.S.A..
| |
Collapse
|
17
|
Shen J, Zhang T, Guan H, Li X, Zhang S, Xu G. PDGFR-beta signaling mediates endogenous neurogenesis after postischemic neural stem/progenitor cell transplantation in mice. Brain Inj 2023; 37:1345-1354. [PMID: 37975626 DOI: 10.1080/02699052.2023.2280894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 10/30/2023] [Indexed: 11/19/2023]
Abstract
OBJECTIVE Although platelet-derived growth factor receptor (PDGFR)-β mediates the self-renewal and multipotency of neural stem/progenitor cells (NSPCs) in vitro and in vivo, its mechanisms of activating endogenous NSPCs following ischemic stroke still remain unproven. METHODS The exogenous NSPCs were transplanted into the ischemic striatum of PDGFR-β conditionally neuroepithelial knockout (KO) mice at 24 h after transient middle cerebral artery occlusion (tMCAO). 5-Bromo-2'-deoxyuridine (BrdU) was intraperitoneally injected to label the newly formed endogenous NSPCs. Infarction volume was measured, and behavioral tests were performed. In the subventricular zone (SVZ), proliferation of endogenous NSPCs was tested, and synapse formation and expression of nutritional factors were measured. RESULTS Compared with control mice, KO mice showed larger infarction volume, delayed neurological recovery, reduced numbers of BrdU positive cells, decreased expression of neurogenic factors (including neurofilament, synaptophysin, and brain-derived neurotrophic factor), and decreased synaptic regeneration in SVZ after tMCAO. Moreover, exogenous NSPC transplantation significantly alleviated neurologic dysfunction, promoted neurogenesis, increased expression of neurologic factors, and diminished synaptic deformation in SVZ of FL mice after tMCAO but had no beneficial effect in KO mice. CONCLUSION PDGFR-β signaling may promote activation of endogenous NSPCs after postischemic NSPC transplantation, and thus represents a novel potential regeneration-based therapeutic target.
Collapse
Affiliation(s)
- Jie Shen
- Department of Neurology, Dongguan Binhaiwan Central Hospital, Dongguan, Guang Dong, China
| | - Tong Zhang
- School of Medicine, Shanxi Datong University, Datong, Shanxi, China
- Institute of Brain Science, Shanxi Datong University, Datong, Shanxi, China
| | - Hong Guan
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China
| | - Xin Li
- Department of Pulmonary and Critical Care Medicine, Inner Mongolia Autonomous Region People's Hospital, Hohhot, China
| | - Sainan Zhang
- Department of Pulmonary and Critical Care Medicine, Inner Mongolia Autonomous Region People's Hospital, Hohhot, China
| | - Guihua Xu
- Department of Science and Education, Dongguan Binhaiwan Central Hospital, Dongguan, Guang Dong, China
- Dongguan Key Laboratory of Precision Medicine
| |
Collapse
|
18
|
Kim HW, Yong H, Shea GKH. Blood-spinal cord barrier disruption in degenerative cervical myelopathy. Fluids Barriers CNS 2023; 20:68. [PMID: 37743487 PMCID: PMC10519090 DOI: 10.1186/s12987-023-00463-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 08/12/2023] [Indexed: 09/26/2023] Open
Abstract
Degenerative cervical myelopathy (DCM) is the most prevalent cause of spinal cord dysfunction in the aging population. Significant neurological deficits may result from a delayed diagnosis as well as inadequate neurological recovery following surgical decompression. Here, we review the pathophysiology of DCM with an emphasis on how blood-spinal cord barrier (BSCB) disruption is a critical yet neglected pathological feature affecting prognosis. In patients suffering from DCM, compromise of the BSCB is evidenced by elevated cerebrospinal fluid (CSF) to serum protein ratios and abnormal contrast-enhancement upon magnetic resonance imaging (MRI). In animal model correlates, there is histological evidence of increased extravasation of tissue dyes and serum contents, and pathological changes to the neurovascular unit. BSCB dysfunction is the likely culprit for ischemia-reperfusion injury following surgical decompression, which can result in devastating neurological sequelae. As there are currently no therapeutic approaches specifically targeting BSCB reconstitution, we conclude the review by discussing potential interventions harnessed for this purpose.
Collapse
Affiliation(s)
- Hyun Woo Kim
- Department of Orthopaedics and Traumatology, LKS Faulty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Hu Yong
- Department of Orthopaedics and Traumatology, LKS Faulty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Graham Ka Hon Shea
- Department of Orthopaedics and Traumatology, LKS Faulty of Medicine, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
19
|
Nakisli S, Lagares A, Nielsen CM, Cuervo H. Pericytes and vascular smooth muscle cells in central nervous system arteriovenous malformations. Front Physiol 2023; 14:1210563. [PMID: 37601628 PMCID: PMC10437819 DOI: 10.3389/fphys.2023.1210563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Accepted: 06/29/2023] [Indexed: 08/22/2023] Open
Abstract
Previously considered passive support cells, mural cells-pericytes and vascular smooth muscle cells-have started to garner more attention in disease research, as more subclassifications, based on morphology, gene expression, and function, have been discovered. Central nervous system (CNS) arteriovenous malformations (AVMs) represent a neurovascular disorder in which mural cells have been shown to be affected, both in animal models and in human patients. To study consequences to mural cells in the context of AVMs, various animal models have been developed to mimic and predict human AVM pathologies. A key takeaway from recently published work is that AVMs and mural cells are heterogeneous in their molecular, cellular, and functional characteristics. In this review, we summarize the observed perturbations to mural cells in human CNS AVM samples and CNS AVM animal models, and we discuss various potential mechanisms relating mural cell pathologies to AVMs.
Collapse
Affiliation(s)
- Sera Nakisli
- Department of Biological Sciences, Ohio University, Athens, OH, United States
- Neuroscience Program, Ohio University, Athens, OH, United States
| | - Alfonso Lagares
- Department of Neurosurgery, University Hospital 12 de Octubre, Madrid, Spain
- Department of Surgery, Universidad Complutense de Madrid, Madrid, Spain
- Instituto de Investigación Imas12, Madrid, Spain
| | - Corinne M. Nielsen
- Department of Biological Sciences, Ohio University, Athens, OH, United States
- Neuroscience Program, Ohio University, Athens, OH, United States
- Molecular and Cellular Biology Program, Ohio University, Athens, OH, United States
| | - Henar Cuervo
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (F.S.P), Madrid, Spain
| |
Collapse
|
20
|
Payne LB, Abdelazim H, Hoque M, Barnes A, Mironovova Z, Willi CE, Darden J, Houk C, Sedovy MW, Johnstone SR, Chappell JC. A Soluble Platelet-Derived Growth Factor Receptor-β Originates via Pre-mRNA Splicing in the Healthy Brain and Is Upregulated during Hypoxia and Aging. Biomolecules 2023; 13:711. [PMID: 37189457 PMCID: PMC10136073 DOI: 10.3390/biom13040711] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 04/17/2023] [Accepted: 04/19/2023] [Indexed: 05/17/2023] Open
Abstract
The platelet-derived growth factor-BB (PDGF-BB) pathway provides critical regulation of cerebrovascular pericytes, orchestrating their investment and retention within the brain microcirculation. Dysregulated PDGF Receptor-beta (PDGFRβ) signaling can lead to pericyte defects that compromise blood-brain barrier (BBB) integrity and cerebral perfusion, impairing neuronal activity and viability, which fuels cognitive and memory deficits. Receptor tyrosine kinases such as PDGF-BB and vascular endothelial growth factor-A (VEGF-A) are often modulated by soluble isoforms of cognate receptors that establish signaling activity within a physiological range. Soluble PDGFRβ (sPDGFRβ) isoforms have been reported to form by enzymatic cleavage from cerebrovascular mural cells, and pericytes in particular, largely under pathological conditions. However, pre-mRNA alternative splicing has not been widely explored as a possible mechanism for generating sPDGFRβ variants, and specifically during tissue homeostasis. Here, we found sPDGFRβ protein in the murine brain and other tissues under normal, physiological conditions. Utilizing brain samples for follow-on analysis, we identified mRNA sequences corresponding to sPDGFRβ isoforms, which facilitated construction of predicted protein structures and related amino acid sequences. Human cell lines yielded comparable sequences and protein model predictions. Retention of ligand binding capacity was confirmed for sPDGFRβ by co-immunoprecipitation. Visualizing fluorescently labeled sPDGFRβ transcripts revealed a spatial distribution corresponding to murine brain pericytes alongside cerebrovascular endothelium. Soluble PDGFRβ protein was detected throughout the brain parenchyma in distinct regions, such as along the lateral ventricles, with signals also found more broadly adjacent to cerebral microvessels consistent with pericyte labeling. To better understand how sPDGFRβ variants might be regulated, we found elevated transcript and protein levels in the murine brain with age, and acute hypoxia increased sPDGFRβ variant transcripts in a cell-based model of intact vessels. Our findings indicate that soluble isoforms of PDGFRβ likely arise from pre-mRNA alternative splicing, in addition to enzymatic cleavage mechanisms, and these variants exist under normal physiological conditions. Follow-on studies will be needed to establish potential roles for sPDGFRβ in regulating PDGF-BB signaling to maintain pericyte quiescence, BBB integrity, and cerebral perfusion-critical processes underlying neuronal health and function, and in turn, memory and cognition.
Collapse
Affiliation(s)
- Laura Beth Payne
- Fralin Biomedical Research Institute (FBRI) at Virginia Tech-Carilion (VTC), Roanoke, VA 24016, USA
- FBRI Center for Vascular and Heart Research, Roanoke, VA 24016, USA
| | - Hanaa Abdelazim
- Fralin Biomedical Research Institute (FBRI) at Virginia Tech-Carilion (VTC), Roanoke, VA 24016, USA
- FBRI Center for Vascular and Heart Research, Roanoke, VA 24016, USA
| | - Maruf Hoque
- Fralin Biomedical Research Institute (FBRI) at Virginia Tech-Carilion (VTC), Roanoke, VA 24016, USA
- FBRI Center for Vascular and Heart Research, Roanoke, VA 24016, USA
| | - Audra Barnes
- Fralin Biomedical Research Institute (FBRI) at Virginia Tech-Carilion (VTC), Roanoke, VA 24016, USA
- FBRI Center for Vascular and Heart Research, Roanoke, VA 24016, USA
- Department of Biomedical Engineering and Mechanics, School of Biomedical Engineering and Sciences, Virginia Tech, Blacksburg, VA 24061, USA
| | - Zuzana Mironovova
- Fralin Biomedical Research Institute (FBRI) at Virginia Tech-Carilion (VTC), Roanoke, VA 24016, USA
- FBRI Center for Vascular and Heart Research, Roanoke, VA 24016, USA
| | - Caroline E. Willi
- Fralin Biomedical Research Institute (FBRI) at Virginia Tech-Carilion (VTC), Roanoke, VA 24016, USA
- FBRI Center for Vascular and Heart Research, Roanoke, VA 24016, USA
| | - Jordan Darden
- Fralin Biomedical Research Institute (FBRI) at Virginia Tech-Carilion (VTC), Roanoke, VA 24016, USA
- FBRI Center for Vascular and Heart Research, Roanoke, VA 24016, USA
| | - Clifton Houk
- Fralin Biomedical Research Institute (FBRI) at Virginia Tech-Carilion (VTC), Roanoke, VA 24016, USA
- Virginia Tech Carilion School of Medicine, Roanoke, VA 24016, USA
| | - Meghan W. Sedovy
- Fralin Biomedical Research Institute (FBRI) at Virginia Tech-Carilion (VTC), Roanoke, VA 24016, USA
- FBRI Center for Vascular and Heart Research, Roanoke, VA 24016, USA
| | - Scott R. Johnstone
- Fralin Biomedical Research Institute (FBRI) at Virginia Tech-Carilion (VTC), Roanoke, VA 24016, USA
- FBRI Center for Vascular and Heart Research, Roanoke, VA 24016, USA
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA 24061, USA
| | - John C. Chappell
- Fralin Biomedical Research Institute (FBRI) at Virginia Tech-Carilion (VTC), Roanoke, VA 24016, USA
- FBRI Center for Vascular and Heart Research, Roanoke, VA 24016, USA
- Department of Biomedical Engineering and Mechanics, School of Biomedical Engineering and Sciences, Virginia Tech, Blacksburg, VA 24061, USA
- Virginia Tech Carilion School of Medicine, Roanoke, VA 24016, USA
| |
Collapse
|
21
|
Cheng J, Jiang J, He B, Lin WJ, Li Y, Duan J, Li H, Huang X, Cai J, Xie J, Zhang Z, Yang Y, Xu Y, Hu X, Wu M, Zhuo X, Liu Q, Shi Z, Yu P, Rong X, Ye X, Saw PE, Wu LJ, Simone CB, Chua MLK, Mai HQ, Tang Y. A phase 2 study of thalidomide for the treatment of radiation-induced blood-brain barrier injury. Sci Transl Med 2023; 15:eabm6543. [PMID: 36812346 DOI: 10.1126/scitranslmed.abm6543] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
Abstract
Radiation-induced brain injury (RIBI) is a debilitating sequela after radiotherapy to treat head and neck cancer, and 20 to 30% of patients with RIBI fail to respond to or have contraindications to the first-line treatments of bevacizumab and corticosteroids. Here, we reported a Simon's minmax two-stage, single-arm, phase 2 clinical trial (NCT03208413) to assess the efficacy of thalidomide in patients with RIBI who were unresponsive to or had contraindications to bevacizumab and corticosteroid therapies. The trial met its primary endpoint, with 27 of 58 patients enrolled showing ≥25% reduction in the volume of cerebral edema on fluid-attenuated inversion recovery-magnetic resonance imaging (FLAIR-MRI) after treatment (overall response rate, 46.6%; 95% CI, 33.3 to 60.1%). Twenty-five (43.1%) patients demonstrated a clinical improvement based on the Late Effects Normal Tissues-Subjective, Objective, Management, Analytic (LENT/SOMA) scale, and 36 (62.1%) experienced cognitive improvement based on the Montreal Cognitive Assessment (MoCA) scores. In a mouse model of RIBI, thalidomide restored the blood-brain barrier and cerebral perfusion, which were attributed to the functional rescue of pericytes secondary to elevation of platelet-derived growth factor receptor β (PDGFRβ) expression by thalidomide. Our data thus demonstrate the therapeutic potential of thalidomide for the treatment of radiation-induced cerebral vasculature impairment.
Collapse
Affiliation(s)
- Jinping Cheng
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.,Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Jingru Jiang
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.,Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Baixuan He
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.,Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Wei-Jye Lin
- Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.,Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China.,Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-sen Memorial Hospital, Foshan 528200, China
| | - Yi Li
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.,Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Jingjing Duan
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Honghong Li
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.,Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Xiaolong Huang
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.,Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Jinhua Cai
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.,Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Jiatian Xie
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.,Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Zhan Zhang
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.,Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Yuhua Yang
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.,Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Yongteng Xu
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.,Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Xia Hu
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.,Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Minyi Wu
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.,Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Xiaohuang Zhuo
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.,Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Qiang Liu
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.,Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Zhongshan Shi
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.,Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Pei Yu
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.,Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Xiaoming Rong
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.,Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Xiaojing Ye
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China.,Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China.,Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Sun Yat-sen University, Guangzhou 510080, China
| | - Phei Er Saw
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA.,Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA.,Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA
| | - Charles B Simone
- Department of Radiation Oncology, New York Proton Center, New York, NY 10035, USA
| | - Melvin L K Chua
- Department of Head and Neck and Thoracic Cancers, Division of Radiation Oncology, National Cancer Centre Singapore, Singapore 169610, Singapore.,Division of Medical Sciences, National Cancer Centre Singapore, Singapore 169610, Singapore.,Oncology Academic Programme, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Hai-Qiang Mai
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou 510060, China
| | - Yamei Tang
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.,Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.,Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China.,Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-sen Memorial Hospital, Foshan 528200, China
| |
Collapse
|
22
|
The Role of Pericytes in Regulation of Innate and Adaptive Immunity. Biomedicines 2023; 11:biomedicines11020600. [PMID: 36831136 PMCID: PMC9953719 DOI: 10.3390/biomedicines11020600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 02/03/2023] [Accepted: 02/16/2023] [Indexed: 02/22/2023] Open
Abstract
Pericytes are perivascular multipotent cells wrapping microvascular capillaries, where they support vasculature functioning, participate in tissue regeneration, and regulate blood flow. However, recent evidence suggests that in addition to traditionally credited structural function, pericytes also manifest immune properties. In this review, we summarise recent data regarding pericytes' response to different pro-inflammatory stimuli and their involvement in innate immune responses through expression of pattern-recognition receptors. Moreover, pericytes express various adhesion molecules, thus regulating trafficking of immune cells across vessel walls. Additionally, the role of pericytes in modulation of adaptive immunity is discussed. Finally, recent reports have suggested that the interaction with cancer cells evokes immunosuppression function in pericytes, thus facilitating immune evasion and facilitating cancer proliferation and metastasis. However, such complex and multi-faceted cross-talks of pericytes with immune cells also suggest a number of potential pericyte-based therapeutic methods and techniques for cancer immunotherapy and treatment of autoimmune and auto-inflammatory disorders.
Collapse
|
23
|
Payne LB, Abdelazim H, Hoque M, Barnes A, Mironovova Z, Willi CE, Darden J, Jenkins-Houk C, Sedovy MW, Johnstone SR, Chappell JC. A Soluble Platelet-Derived Growth Factor Receptor-β Originates via Pre-mRNA Splicing in the Healthy Brain and is Differentially Regulated during Hypoxia and Aging. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.03.527005. [PMID: 36778261 PMCID: PMC9915746 DOI: 10.1101/2023.02.03.527005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The platelet-derived growth factor-BB (PDGF-BB) pathway provides critical regulation of cerebrovascular pericytes, orchestrating their investment and retention within the brain microcirculation. Dysregulated PDGF Receptor-beta (PDGFRβ) signaling can lead to pericyte defects that compromise blood-brain barrier (BBB) integrity and cerebral perfusion, impairing neuronal activity and viability, which fuels cognitive and memory deficits. Receptor tyrosine kinases (RTKs) like PDGF-BB and vascular endothelial growth factor-A (VEGF-A) are often modulated by soluble isoforms of cognate receptors that establish signaling activity within a physiological range. Soluble PDGFRβ (sPDGFRβ) isoforms have been reported to form by enzymatic cleavage from cerebrovascular mural cells, and pericytes in particular, largely under pathological conditions. However, pre-mRNA alternative splicing has not been widely explored as a possible mechanism for generating sPDGFRβ variants, and specifically during tissue homeostasis. Here, we found sPDGFRβ protein in the murine brain and other tissues under normal, physiological conditions. Utilizing brain samples for follow-on analysis, we identified mRNA sequences corresponding to sPDGFRβ isoforms, which facilitated construction of predicted protein structures and related amino acid sequences. Human cell lines yielded comparable sequences and protein model predictions. Retention of ligand binding capacity was confirmed for sPDGFRβ by co-immunoprecipitation. Visualizing fluorescently labeled sPDGFRβ transcripts revealed a spatial distribution corresponding to murine brain pericytes alongside cerebrovascular endothelium. Soluble PDGFRβ protein was detected throughout the brain parenchyma in distinct regions such as along the lateral ventricles, with signals also found more broadly adjacent to cerebral microvessels consistent with pericyte labeling. To better understand how sPDGFRβ variants might be regulated, we found elevated transcript and protein levels in the murine brain with age, and acute hypoxia increased sPDGFRβ variant transcripts in a cell-based model of intact vessels. Our findings indicate that soluble isoforms of PDGFRβ likely arise from pre-mRNA alternative splicing, in addition to enzymatic cleavage mechanisms, and these variants exist under normal physiological conditions. Follow-on studies will be needed to establish potential roles for sPDGFRβ in regulating PDGF-BB signaling to maintain pericyte quiescence, BBB integrity, and cerebral perfusion - critical processes underlying neuronal health and function, and in turn memory and cognition.
Collapse
|
24
|
Protective Effects of Human Pericyte-like Adipose-Derived Mesenchymal Stem Cells on Human Retinal Endothelial Cells in an In Vitro Model of Diabetic Retinopathy: Evidence for Autologous Cell Therapy. Int J Mol Sci 2023; 24:ijms24020913. [PMID: 36674425 PMCID: PMC9860961 DOI: 10.3390/ijms24020913] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 12/23/2022] [Accepted: 12/31/2022] [Indexed: 01/06/2023] Open
Abstract
Diabetic retinopathy (DR) is characterized by morphologic and metabolic alterations in endothelial cells (ECs) and pericytes (PCs) of the blood-retinal barrier (BRB). The loss of interendothelial junctions, increased vascular permeability, microaneurysms, and finally, EC detachment are the main features of DR. In this scenario, a pivotal role is played by the extensive loss of PCs. Based on previous results, the aim of this study was to assess possible beneficial effects exerted by adipose mesenchymal stem cells (ASCs) and their pericyte-like differentiated phenotype (P-ASCs) on human retinal endothelial cells (HRECs) in high glucose conditions (25 mM glucose, HG). P-ASCs were more able to preserve BRB integrity than ASCs in terms of (a) increased transendothelial electrical resistance (TEER); (b) increased expression of adherens junction and tight junction proteins (VE-cadherin and ZO-1); (c) reduction in mRNA levels of inflammatory cytokines TNF-α, IL-1β, and MMP-9; (d) reduction in the angiogenic factor VEGF and in fibrotic TGF-β1. Moreover, P-ASCs counteracted the HG-induced activation of the pro-inflammatory phospho-ERK1/2/phospho-cPLA2/COX-2 pathway. Finally, crosstalk between HRECs and ASCs or P-ASCs based on the PDGF-B/PDGFR-β axis at the mRNA level is described herein. Thus, P-ASCs might be considered valuable candidates for therapeutic approaches aimed at countering BRB disruption in DR.
Collapse
|
25
|
Xu S, Li X, Li Y, Li X, Lv E, Zhang X, Shi Y, Wang Y. Neuroprotective effect of Dl-3-n-butylphthalide against ischemia-reperfusion injury is mediated by ferroptosis regulation via the SLC7A11/GSH/GPX4 pathway and the attenuation of blood-brain barrier disruption. Front Aging Neurosci 2023; 15:1028178. [PMID: 36909944 PMCID: PMC9995665 DOI: 10.3389/fnagi.2023.1028178] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 02/07/2023] [Indexed: 02/25/2023] Open
Abstract
Background Stroke is one of the most severe diseases worldwide, resulting in physical and mental problems. Dl-3-n-butylphthalide, a compound derived from celery seed, has been approved for treating ischemic stroke in China. No study has evaluated how Dl-3-n-butylphthalide affects the ferroptosis SLC7A11/GSH/GPX4 signal pathway and blood-brain barrier (BBB) PDGFRβ/PI3K/Akt signal pathways in the rat middle cerebral artery occlusion/reperfusion (MCAO/R) model of ischemic stroke. Methods Sprague-Dawley rats were used to develop the MCAO/R model. Our study used three incremental doses (10, 20, and 30) of Dl-3-n-butylphthalide injected intraperitoneally 24 h after MCAO/R surgery. The neuroprotective effect and success of the model were evaluated using the neurofunction score, brain water content determination, and triphenyl-tetrazolium chloride-determined infarction area changes. Pathological changes in the brain tissue and the degree of apoptosis were examined by hematoxylin and eosin, Nissl, and terminal deoxynucleotidyl transferase dUTP nick end labeling staining. In addition, pathway proteins and RNA expression levels were studied to verify the effects of Dl-3-n-butyphthalide on both pathways. At the same time, commercial kits were used to detect glutathione, reactive oxygen species, and malondialdehyde, to detect oxidative stress in brain tissues. Results The middle dose of Dl-3-n-butylphthalide not only improved MCAO-induced brain dysfunction and alleviated pathological damage, brain inflammatory response, oxidative stress, and apoptosis but also protected against ferroptosis and reduced BBB damage. These changes resulted in improved neurological function in the cerebral cortex. Conclusion We speculate that Dl-3-n-butylphthalide has a neuroprotective effect on focal cerebral ischemia/reperfusion, which may be mediated through ferroptosis-dependent SLC7A11/GSH/GPX4 signal pathway and PDGFRβ/PI3/Akt signal pathway.
Collapse
Affiliation(s)
- Shuangli Xu
- Emergency Department, The Affiliated Hospital of Weifang Medical University, Weifang, Shandong, China
| | - Xuewei Li
- Department of Rheumatology, The Affiliated Hospital of Weifang Medical University, Weifang, Shandong, China
| | - Yutian Li
- School of Pharmacy, Weifang Medical University, Weifang, Shandong, China
| | - Xiangling Li
- Department of Internal Medicine, The Affiliated Hospital of Weifang Medical University, Weifang, Shandong, China
| | - E Lv
- Department of Histology and Embryology, Weifang Medical University, Weifang, Shandong, China
| | - Xiaojun Zhang
- Department II of Neurology, The Affiliated Hospital of Weifang Medical University, Weifang, Shandong, China
| | - Youkui Shi
- Emergency Department, The Affiliated Hospital of Weifang Medical University, Weifang, Shandong, China
| | - Yanqiang Wang
- Department of Rheumatology, The Affiliated Hospital of Weifang Medical University, Weifang, Shandong, China
| |
Collapse
|
26
|
Gürler G, Soylu KO, Yemisci M. Importance of Pericytes in the Pathophysiology of Cerebral Ischemia. Noro Psikiyatr Ars 2022; 59:S29-S35. [PMID: 36578988 PMCID: PMC9767130 DOI: 10.29399/npa.28171] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 04/02/2022] [Indexed: 12/31/2022] Open
Abstract
Various cell types contribute to pathological changes observed in the brain following cerebral ischemia. Pericytes, as a component of neurovascular unit (NVU) and blood brain barrier (BBB), play a key role for cerebral blood flow control and regulation of vessel permeability. It was shown that pericytes can control cerebral blood flow at the level of capillaries, by their contractile property. Their role in BBB development and maintenance are crucial for guidance of brain vessel development, new vessel formation and stabilization of the newly formed vessels. Additionally, they can contribute to inflammation in response to inflammatory stimuli and can differentiate to various cell types by their multipotent differentiation properties. This cell type which is intimately associated with cerebral circulation also plays important roles during cerebral ischemia. Here, we review the properties and physiological functions of pericytes, how these functions change during ischemia to affect the pathophysiology of ischemic stroke and post stroke cognitive impairment. Pericytes are a neglected cell type and they are not unambiguously characterized which in turn led to contradictory findings in the literature. Clear characterization of pericytes by current methods will help better understanding of their role in the pathophysiology of stroke. With the information gained from these efforts it will be possible to develop pericyte specific therapeutic targets and achieve important breakthroughs in clinical recovery in ischemic stroke treatment.
Collapse
Affiliation(s)
- Gökçe Gürler
- Hacettepe University, Institute of Neurological Sciences and Psychiatry, Ankara, Turkey
| | - Kadir Oğuzhan Soylu
- Hacettepe University, Institute of Neurological Sciences and Psychiatry, Ankara, Turkey
| | - Müge Yemisci
- Hacettepe University, Institute of Neurological Sciences and Psychiatry, Ankara, Turkey,Hacettepe University, Faculty of Medicine, Department of Neurology, Ankara, Turkey,Neuroscience and Neurotechnology Center of Excellence (NÖROM), Ankara, Turkey,Correspondence Address: Müge Yemişci, Hacettepe Üniversitesi Nörolojik Bilimler ve Psikiyatri Enstitüsü, 06230 Sıhhiye Ankara, Turkey • E-mail:
| |
Collapse
|
27
|
Components of Salvia miltiorrhiza and Panax notoginseng Protect Pericytes Against OGD/R-Induced Injury via Regulating the PI3K/AKT/mTOR and JNK/ERK/P38 Signaling Pathways. J Mol Neurosci 2022; 72:2377-2388. [PMID: 36394713 DOI: 10.1007/s12031-022-02082-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: 08/22/2022] [Accepted: 11/10/2022] [Indexed: 11/18/2022]
Abstract
Salvia miltiorrhiza (SAL) and Panax notoginseng (PNS) are widely used in treating of ischemic stroke. However, it is unknown which components of SAL and PNS protect brain microvascular pericytes after an ischemic stroke. We evaluated the protective effects and mechanisms of SAL and PNS components in pericytes subjected to oxygen-glucose deprivation/reoxygenation (OGD/R). Pericytes were subjected to OGD/R. Cell Counting Kit-8 (CCK-8) was used to evaluate cell viability. ROS and SOD kits were used to detect oxidative stress. Flow cytometry was performed to analyze cell apoptosis. To evaluate cell migration, a scratch assay was performed. Expression of cleaved caspase-3, Bcl-2, Bax, VEGF, Ang-1, PDGFR-β, PI3K/AKT/mTOR, and JNK/ERK/P38 signaling pathways were identified using western blot. The results revealed that salvianolic acid B (Sal B), salvianolic acid D (Sal D), notoginsenoside R1 (R1), ginsenoside Rb1 (Rb1), and ginsenoside Rg1 (Rg1) increased the cell viability of pericytes subjected to OGD/R, reduced the level of ROS, and increased the expression of SOD. The components reduced cell apoptosis, increased the protein level of Bcl-2/Bax, reduced the level of cleaved caspase-3/caspase-3, increased cell migration, and enhanced the levels of Ang-1, PDGFR-β, and VEGF. The components could activate PI3K/AKT/mTOR pathway while inhibiting the JNK/ERK/P38 pathway. Studies found that Sal B, Sal D, R1, Rb1, and Rg1 inhibited oxidative stress and apoptosis while increasing the release of pro-angiogenic regulators of pericytes related to the PI3K/AKT/mTOR and JNK/ERK/P38 signaling pathways. This provides a potential foundation for developing monomeric drugs for treating ischemic stroke.
Collapse
|
28
|
Cognitive Impairments and blood-brain Barrier Damage in a Mouse Model of Chronic Cerebral Hypoperfusion. Neurochem Res 2022; 47:3817-3828. [DOI: 10.1007/s11064-022-03799-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 10/07/2022] [Accepted: 10/16/2022] [Indexed: 10/31/2022]
Abstract
AbstractChronic cerebral hypoperfusion (CCH) is commonly involved in various brain diseases. Tight junction proteins (TJs) are key components constituting the anatomical substrate of the blood-brain barrier (BBB). Changes in cognitive function and BBB after CCH and their relationship need further exploration. To investigate the effect of CCH on cognition and BBB, we developed a bilateral common carotid artery stenosis (BCAS) model in Tie2-GFP mice. Mice manifested cognitive impairments accompanied with increased microglia after the BCAS operation. BCAS mice also exhibited increased BBB permeability at all time points set from D1 to D42. Furthermore, BCAS mice showed reduced expression of TJs 42 d after the operation. In addition, correct entrances of mice in radial arm maze test had a moderate negative correlation with EB extravasation. Our data suggested that BCAS could lead to cognitive deficits, microglia increase and BBB dysfunction characterized by increased BBB permeability and reduced TJs expression level. BBB permeability may be involved in the cognitive impairments induced by CCH.
Collapse
|
29
|
Janssen Daalen JM, Meinders MJ, Giardina F, Roes KCB, Stunnenberg BC, Mathur S, Ainslie PN, Thijssen DHJ, Bloem BR. Multiple N-of-1 trials to investigate hypoxia therapy in Parkinson's disease: study rationale and protocol. BMC Neurol 2022; 22:262. [PMID: 35836147 PMCID: PMC9281145 DOI: 10.1186/s12883-022-02770-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 06/24/2022] [Indexed: 11/30/2022] Open
Abstract
Background Parkinson’s disease (PD) is a neurodegenerative disease, for which no disease-modifying therapies exist. Preclinical and clinical evidence suggest that hypoxia-based therapy might have short- and long-term benefits in PD. We present the contours of the first study to assess the safety, feasibility and physiological and symptomatic impact of hypoxia-based therapy in individuals with PD. Methods/Design In 20 individuals with PD, we will investigate the safety, tolerability and short-term symptomatic efficacy of continuous and intermittent hypoxia using individual, double-blind, randomized placebo-controlled N-of-1 trials. This design allows for dose finding and for including more individualized outcomes, as each individual serves as its own control. A wide range of exploratory outcomes is deployed, including the Movement Disorders Society Unified Parkinson’s Disease Rating scale (MDS-UPDRS) part III, Timed Up & Go Test, Mini Balance Evaluation Systems (MiniBES) test and wrist accelerometry. Also, self-reported impression of overall symptoms, motor and non-motor symptoms and urge to take dopaminergic medication will be assessed on a 10-point Likert scale. As part of a hypothesis-generating part of the study, we also deploy several exploratory outcomes to probe possible underlying mechanisms of action, including cortisol, erythropoietin and platelet-derived growth factor β. Efficacy will be assessed primarily by a Bayesian analysis. Discussion This evaluation of hypoxia therapy could provide insight in novel pathways that may be pursued for PD treatment. This trial also serves as a proof of concept for deploying an N-of-1 design and for including individualized outcomes in PD research, as a basis for personalized treatment approaches. Trial registration ClinicalTrials.gov Identifier: NCT05214287 (registered January 28, 2022).
Supplementary Information The online version contains supplementary material available at 10.1186/s12883-022-02770-7.
Collapse
Affiliation(s)
- Jules M Janssen Daalen
- Center of Expertise for Parkinson & Movement Disorders; Nijmegen, the Netherlands, Department of Neurology, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Marjan J Meinders
- Center of Expertise for Parkinson & Movement Disorders; Nijmegen, the Netherlands, Department of Neurology, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, The Netherlands.,IQ Healthcare, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Federica Giardina
- Department of Health Evidence, Radboud Institute for Health Sciences, Radboud University Medical Center, Section Biostatistics, Nijmegen, The Netherlands
| | - Kit C B Roes
- Department of Health Evidence, Radboud Institute for Health Sciences, Radboud University Medical Center, Section Biostatistics, Nijmegen, The Netherlands
| | - Bas C Stunnenberg
- Department of Neurology, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, The Netherlands.,Department of Neurology, Rijnstate Hospital, Arnhem, Netherlands
| | | | - Philip N Ainslie
- Center for Heart, Lung and Vascular Health, School of Health and Exercise Sciences, University of British Columbia, Kelowna, Canada
| | - Dick H J Thijssen
- Department of Physiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Bastiaan R Bloem
- Center of Expertise for Parkinson & Movement Disorders; Nijmegen, the Netherlands, Department of Neurology, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, The Netherlands.
| |
Collapse
|
30
|
Morihara R, Yamashita T, Osakada Y, Feng T, Hu X, Fukui Y, Tadokoro K, Takemoto M, Abe K. Efficacy and safety of spot heating and ultrasound irradiation on in vitro and in vivo thrombolysis models. J Cereb Blood Flow Metab 2022; 42:1322-1334. [PMID: 35130767 PMCID: PMC9207486 DOI: 10.1177/0271678x221079127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The feasibility of transcranial sonothrombolysis has been demonstrated, although little is known about the relationships between thermal or mechanical mechanisms and thrombolytic outcomes. Therefore, the present study aims to reveal the effect and safety of temperature and ultrasound through in vitro and in vivo thrombolysis models. Artificial clots in microtubes were heated in a water bath or sonicated by ultrasound irradiation, and then clots weight decrease with rising temperature and sonication time was confirmed. In the in vitro thrombotic occlusion model, based on spot heating, clot volume was reduced and clots moved to the distal side, followed by recanalization of the occlusion. In the in vivo study, the common carotid artery of rats was exposed to a spot heater or to sonication. No brain infarct or brain blood barrier disruption was shown, but endothelial junctional dysintegrity and an inflammatory response in the carotid artery were detected. The present spot heating and ultrasound irradiation models seem to be effective for disintegrating clots in vitro, but the safety of the in vivo model was not fully supported by the data. However, the data indicates that a shorter time exposure could be less invasive than a longer exposure.
Collapse
Affiliation(s)
- Ryuta Morihara
- Department of Neurology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Toru Yamashita
- Department of Neurology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Yosuke Osakada
- Department of Neurology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Tian Feng
- Department of Neurology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Xinran Hu
- Department of Neurology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Yusuke Fukui
- Department of Neurology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Koh Tadokoro
- Department of Neurology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Mami Takemoto
- Department of Neurology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Koji Abe
- Department of Neurology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| |
Collapse
|
31
|
Vázquez-Villaseñor I, Smith CI, Thang YJR, Heath PR, Wharton SB, Blackburn DJ, Ridger VC, Simpson JE. RNA-Seq Profiling of Neutrophil-Derived Microvesicles in Alzheimer's Disease Patients Identifies a miRNA Signature That May Impact Blood-Brain Barrier Integrity. Int J Mol Sci 2022; 23:5913. [PMID: 35682592 PMCID: PMC9180128 DOI: 10.3390/ijms23115913] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 05/21/2022] [Accepted: 05/21/2022] [Indexed: 02/01/2023] Open
Abstract
(1) Background: Systemic infection is associated with increased neuroinflammation and accelerated cognitive decline in AD patients. Activated neutrophils produce neutrophil-derived microvesicles (NMV), which are internalised by human brain microvascular endothelial cells and increase their permeability in vitro, suggesting that NMV play a role in blood-brain barrier (BBB) integrity during infection. The current study investigated whether microRNA content of NMV from AD patients is significantly different compared to healthy controls and could impact cerebrovascular integrity. (2) Methods: Neutrophils isolated from peripheral blood samples of five AD and five healthy control donors without systemic infection were stimulated to produce NMV. MicroRNAs isolated from NMV were analysed by RNA-Seq, and online bioinformatic tools were used to identify significantly differentially expressed microRNAs in the NMV. Target and pathway analyses were performed to predict the impact of the candidate microRNAs on vascular integrity. (3) Results: There was no significant difference in either the number of neutrophils (p = 0.309) or the number of NMV (p = 0.3434) isolated from AD donors compared to control. However, 158 microRNAs were significantly dysregulated in AD NMV compared to controls, some of which were associated with BBB dysfunction, including miR-210, miR-20b-5p and miR-126-5p. Pathway analysis revealed numerous significantly affected pathways involved in regulating vascular integrity, including the TGFβ and PDGFB pathways, as well as Hippo, IL-2 and DNA damage signalling. (4) Conclusions: NMV from AD patients contain miRNAs that may alter the integrity of the BBB and represent a novel neutrophil-mediated mechanism for BBB dysfunction in AD and the accelerated cognitive decline seen as a result of a systemic infection.
Collapse
Affiliation(s)
- Irina Vázquez-Villaseñor
- Sheffield Institute for Translational Neuroscience, The University of Sheffield, Sheffield S10 2HQ, UK; (C.I.S.); (Y.J.R.T.); (P.R.H.); (S.B.W.); (D.J.B.)
| | - Cynthia I. Smith
- Sheffield Institute for Translational Neuroscience, The University of Sheffield, Sheffield S10 2HQ, UK; (C.I.S.); (Y.J.R.T.); (P.R.H.); (S.B.W.); (D.J.B.)
| | - Yung J. R. Thang
- Sheffield Institute for Translational Neuroscience, The University of Sheffield, Sheffield S10 2HQ, UK; (C.I.S.); (Y.J.R.T.); (P.R.H.); (S.B.W.); (D.J.B.)
| | - Paul R. Heath
- Sheffield Institute for Translational Neuroscience, The University of Sheffield, Sheffield S10 2HQ, UK; (C.I.S.); (Y.J.R.T.); (P.R.H.); (S.B.W.); (D.J.B.)
| | - Stephen B. Wharton
- Sheffield Institute for Translational Neuroscience, The University of Sheffield, Sheffield S10 2HQ, UK; (C.I.S.); (Y.J.R.T.); (P.R.H.); (S.B.W.); (D.J.B.)
| | - Daniel J. Blackburn
- Sheffield Institute for Translational Neuroscience, The University of Sheffield, Sheffield S10 2HQ, UK; (C.I.S.); (Y.J.R.T.); (P.R.H.); (S.B.W.); (D.J.B.)
| | - Victoria C. Ridger
- Department of Infection, Immunity and Cardiovascular Disease, Medical School, The University of Sheffield, Beech Hill Road, Sheffield S10 2RX, UK;
| | - Julie E. Simpson
- Sheffield Institute for Translational Neuroscience, The University of Sheffield, Sheffield S10 2HQ, UK; (C.I.S.); (Y.J.R.T.); (P.R.H.); (S.B.W.); (D.J.B.)
| |
Collapse
|
32
|
Zhao Y, Gan L, Ren L, Lin Y, Ma C, Lin X. Factors influencing the blood-brain barrier permeability. Brain Res 2022; 1788:147937. [PMID: 35568085 DOI: 10.1016/j.brainres.2022.147937] [Citation(s) in RCA: 79] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 04/28/2022] [Accepted: 05/03/2022] [Indexed: 12/14/2022]
Abstract
The blood-brain barrier (BBB) is a dynamic structure that protects the brain from harmful blood-borne, endogenous and exogenous substances and maintains the homeostatic microenvironment. All constituent cell types play indispensable roles in the BBB's integrity, and other structural BBB components, such as tight junction proteins, adherens junctions, and junctional proteins, can control the barrier permeability. Regarding the need to exchange nutrients and toxic materials, solute carriers, ATP-binding case families, and ion transporter, as well as transcytosis regulate the influx and efflux transport, while the difference in localisation and expression can contribute to functional differences in transport properties. Numerous chemical mediators and other factors such as non-physicochemical factors have been identified to alter BBB permeability by mediating the structural components and barrier function, because of the close relationship with inflammation. In this review, we highlight recently gained mechanistic insights into the maintenance and disruption of the BBB. A better understanding of the factors influencing BBB permeability could contribute to supporting promising potential therapeutic targets for protecting the BBB and the delivery of central nervous system drugs via BBB permeability interventions under pathological conditions.
Collapse
Affiliation(s)
- Yibin Zhao
- The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China; Department of Neurobiology and Acupuncture Research, Zhejiang Chinese Medical University, Hangzhou, China
| | - Lin Gan
- The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China; Department of Neurobiology and Acupuncture Research, Zhejiang Chinese Medical University, Hangzhou, China
| | - Li Ren
- The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China; Department of Neurobiology and Acupuncture Research, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yubo Lin
- The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China; Department of Neurobiology and Acupuncture Research, Zhejiang Chinese Medical University, Hangzhou, China
| | - Congcong Ma
- The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China; Department of Neurobiology and Acupuncture Research, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xianming Lin
- The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China; Department of Neurobiology and Acupuncture Research, Zhejiang Chinese Medical University, Hangzhou, China.
| |
Collapse
|
33
|
Li SY, Johnson R, Smyth LC, Dragunow M. Platelet-derived growth factor signalling in neurovascular function and disease. Int J Biochem Cell Biol 2022; 145:106187. [PMID: 35217189 DOI: 10.1016/j.biocel.2022.106187] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 02/08/2022] [Accepted: 02/21/2022] [Indexed: 11/25/2022]
Abstract
Platelet-derived growth factors are critical for cerebrovascular development and homeostasis. Abnormalities in this signalling pathway are implicated in neurological diseases, especially those where neurovascular dysfunction and neuroinflammation plays a prominent role in disease pathologies, such as stroke and Alzheimer's disease; the angiogenic nature of this pathway also draws its significance in brain malignancies such as glioblastoma where tumour angiogenesis is profuse. In this review, we provide an updated overview of the actions of the platelet-derived growth factors on neurovascular function, their role in the regulation of perivascular cell types expressing the cognate receptors, neurological diseases associated with aberrance in signalling, and highlight the clinical relevance and therapeutic potentials of this pathway for central nervous system diseases.
Collapse
Affiliation(s)
- Susan Ys Li
- Department of Pharmacology and Centre for Brain Research, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand.
| | - Rebecca Johnson
- Department of Pharmacology and Centre for Brain Research, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand.
| | - Leon Cd Smyth
- Center for Brain Immunology and Glia, Department of Pathology and Immunology, Washington University in St Louis, MO, USA.
| | - Mike Dragunow
- Department of Pharmacology and Centre for Brain Research, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand.
| |
Collapse
|
34
|
Gallego I, Villate-Beitia I, Saenz-Del-Burgo L, Puras G, Pedraz JL. Therapeutic Opportunities and Delivery Strategies for Brain Revascularization in Stroke, Neurodegeneration, and Aging. Pharmacol Rev 2022; 74:439-461. [PMID: 35302047 DOI: 10.1124/pharmrev.121.000418] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 01/18/2022] [Accepted: 01/22/2022] [Indexed: 12/25/2022] Open
Abstract
Central nervous system (CNS) diseases, especially acute ischemic events and neurodegenerative disorders, constitute a public health problem with no effective treatments to allow a persistent solution. Failed therapies targeting neuronal recovery have revealed the multifactorial and intricate pathophysiology underlying such CNS disorders as ischemic stroke, Alzheimeŕs disease, amyotrophic lateral sclerosis, vascular Parkisonism, vascular dementia, and aging, in which cerebral microvasculature impairment seems to play a key role. In fact, a reduction in vessel density and cerebral blood flow occurs in these scenarios, contributing to neuronal dysfunction and leading to loss of cognitive function. In this review, we provide an overview of healthy brain microvasculature structure and function in health and the effect of the aforementioned cerebral CNS diseases. We discuss the emerging new therapeutic opportunities, and their delivery approaches, aimed at recovering brain vascularization in this context. SIGNIFICANCE STATEMENT: The lack of effective treatments, mainly focused on neuron recovery, has prompted the search of other therapies to treat cerebral central nervous system diseases. The disruption and degeneration of cerebral microvasculature has been evidenced in neurodegenerative diseases, stroke, and aging, constituting a potential target for restoring vascularization, neuronal functioning, and cognitive capacities by the development of therapeutic pro-angiogenic strategies.
Collapse
Affiliation(s)
- Idoia Gallego
- NanoBioCel Research Group, Laboratory of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain (I.G., I.V.-B., L.S.-B., G.P., J.L.P); Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine, Institute of Health Carlos III, Madrid, Spain (I.G., I.V.-B., L.S.-B., G.P., J.L.P.); and Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain (I.G., I.V.-B., L.S.-B., G.P., J.L.P.)
| | - Ilia Villate-Beitia
- NanoBioCel Research Group, Laboratory of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain (I.G., I.V.-B., L.S.-B., G.P., J.L.P); Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine, Institute of Health Carlos III, Madrid, Spain (I.G., I.V.-B., L.S.-B., G.P., J.L.P.); and Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain (I.G., I.V.-B., L.S.-B., G.P., J.L.P.)
| | - Laura Saenz-Del-Burgo
- NanoBioCel Research Group, Laboratory of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain (I.G., I.V.-B., L.S.-B., G.P., J.L.P); Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine, Institute of Health Carlos III, Madrid, Spain (I.G., I.V.-B., L.S.-B., G.P., J.L.P.); and Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain (I.G., I.V.-B., L.S.-B., G.P., J.L.P.)
| | - Gustavo Puras
- NanoBioCel Research Group, Laboratory of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain (I.G., I.V.-B., L.S.-B., G.P., J.L.P); Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine, Institute of Health Carlos III, Madrid, Spain (I.G., I.V.-B., L.S.-B., G.P., J.L.P.); and Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain (I.G., I.V.-B., L.S.-B., G.P., J.L.P.)
| | - José Luis Pedraz
- NanoBioCel Research Group, Laboratory of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain (I.G., I.V.-B., L.S.-B., G.P., J.L.P); Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine, Institute of Health Carlos III, Madrid, Spain (I.G., I.V.-B., L.S.-B., G.P., J.L.P.); and Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain (I.G., I.V.-B., L.S.-B., G.P., J.L.P.)
| |
Collapse
|
35
|
Characterisation of PDGF-BB:PDGFRβ signalling pathways in human brain pericytes: evidence of disruption in Alzheimer's disease. Commun Biol 2022; 5:235. [PMID: 35301433 PMCID: PMC8931009 DOI: 10.1038/s42003-022-03180-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 02/17/2022] [Indexed: 01/03/2023] Open
Abstract
Platelet-derived growth factor-BB (PDGF-BB):PDGF receptor-β (PDGFRβ) signalling in brain pericytes is critical to the development, maintenance and function of a healthy blood-brain barrier (BBB). Furthermore, BBB impairment and pericyte loss in Alzheimer’s disease (AD) is well documented. We found that PDGF-BB:PDGFRβ signalling components were altered in human AD brains, with a marked reduction in vascular PDGFB. We hypothesised that reduced PDGF-BB:PDGFRβ signalling in pericytes may impact on the BBB. We therefore tested the effects of PDGF-BB on primary human brain pericytes in vitro to define pathways related to BBB function. Using pharmacological inhibitors, we dissected distinct aspects of the PDGF-BB response that are controlled by extracellular signal-regulated kinase (ERK) and Akt pathways. PDGF-BB promotes the proliferation of pericytes and protection from apoptosis through ERK signalling. In contrast, PDGF-BB:PDGFRβ signalling through Akt augments pericyte-derived inflammatory secretions. It may therefore be possible to supplement PDGF-BB signalling to stabilise the cerebrovasculature in AD. Smyth et al. use tissue microarrays from Alzheimer’s disease (AD) patient brains to show that PDGF-BB:PDGFRβ signalling components are reduced in AD. They then use primary human brain pericytes to elucidate a pathway by which PDGF-BB:PDGFRβ signalling in brain pericytes is disrupted in AD, thus impairing the blood brain barrier.
Collapse
|
36
|
van Vliet EA, Marchi N. Neurovascular unit dysfunction as a mechanism of seizures and epilepsy during aging. Epilepsia 2022; 63:1297-1313. [PMID: 35218208 PMCID: PMC9321014 DOI: 10.1111/epi.17210] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 02/23/2022] [Accepted: 02/23/2022] [Indexed: 11/30/2022]
Abstract
The term neurovascular unit (NVU) describes the structural and functional liaison between specialized brain endothelium, glial and mural cells, and neurons. Within the NVU, the blood‐brain barrier (BBB) is the microvascular structure regulating neuronal physiology and immune cross‐talk, and its properties adapt to brain aging. Here, we analyze a research framework where NVU dysfunction, caused by acute insults or disease progression in the aging brain, represents a converging mechanism underlying late‐onset seizures or epilepsy and neurological or neurodegenerative sequelae. Furthermore, seizure activity may accelerate brain aging by sustaining regional NVU dysfunction, and a cerebrovascular pathology may link seizures to comorbidities. Next, we focus on NVU diagnostic approaches that could be tailored to seizure conditions in the elderly. We also examine the impending disease‐modifying strategies based on the restoration of the NVU and, more in general, the homeostatic control of anti‐ and pro‐inflammatory players. We conclude with an outlook on current pre‐clinical knowledge gaps and clinical challenges pertinent to seizure onset and conditions in an aging population.
Collapse
Affiliation(s)
- Erwin A van Vliet
- Amsterdam UMC, University of Amsterdam, dept. of (Neuro)pathology, Amsterdam, the Netherlands.,University of Amsterdam, Swammerdam Institute for Life Sciences, Center for Neuroscience, Amsterdam, the Netherlands
| | - Nicola Marchi
- Cerebrovascular and Glia Research, Department of Neuroscience, Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France
| |
Collapse
|
37
|
Fujisawa M, Takeshita Y, Fujikawa S, Matsuo K, Okamoto M, Tamada M, Shimizu F, Sano Y, Koga M, Kanda T. Exploring lipophilic compounds that induce BDNF secretion in astrocytes beyond the BBB using a new multi-cultured human in vitro BBB model. J Neuroimmunol 2022; 362:577783. [PMID: 34902709 DOI: 10.1016/j.jneuroim.2021.577783] [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: 07/27/2021] [Revised: 11/04/2021] [Accepted: 12/02/2021] [Indexed: 10/19/2022]
Abstract
Brain-derived neurotrophic factor (BDNF) cannot cross the blood-brain barrier (BBB) when administered peripherally, which hinders its therapeutic potential. We utilized an in vitro BBB model-a tri-culture of a human endothelial cell line, a pericyte cell line, and an astrocyte cell line-to study the effect of twenty candidate lipophilic compounds on stimulating BDNF secretion in pericytes and astrocytes. The prostaglandin E2 receptor 4 agonist and sphingosine-1-phosphate receptor 5 agonist facilitated secretion of BDNF in the astrocyte, but did not decrease the transendothelial electrical resistance. These compounds may be promising agents for neurodegenerative and neuroinflammatory diseases.
Collapse
Affiliation(s)
- Miwako Fujisawa
- Department of Neurology and Clinical Neuroscience, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan.
| | - Yukio Takeshita
- Department of Neurology and Clinical Neuroscience, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan.
| | - Susumu Fujikawa
- Department of Neurology and Clinical Neuroscience, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan.
| | - Kinya Matsuo
- Department of Neurology and Clinical Neuroscience, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan.
| | - Masashi Okamoto
- Department of Neurology and Clinical Neuroscience, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan.
| | - Masaya Tamada
- Department of Neurology and Clinical Neuroscience, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan.
| | - Fumitaka Shimizu
- Department of Neurology and Clinical Neuroscience, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan.
| | - Yasuteru Sano
- Department of Neurology and Clinical Neuroscience, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan.
| | - Michiaki Koga
- Department of Neurology and Clinical Neuroscience, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan.
| | - Takashi Kanda
- Department of Neurology and Clinical Neuroscience, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan.
| |
Collapse
|
38
|
Platelet-derived growth factor-BB and white matter hyperintensity burden in APOE4 carriers. CEREBRAL CIRCULATION - COGNITION AND BEHAVIOR 2022; 3. [PMID: 35844252 PMCID: PMC9286493 DOI: 10.1016/j.cccb.2022.100131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Background The apolipoprotein-e4 (APOE4) gene increases risk for developing late-onset Alzheimer's disease (AD) and has been linked to increased microvascular dysfunction, including pericyte degeneration and blood-brain barrier breakdown. Platelet-derived growth factor-BB (PDGF-BB) is a glycoprotein involved in blood-brain barrier and pericyte maintenance. Increased PDGF-BB levels have been reported in white matter in AD brain tissue. However, the association between circulating levels of PDGF-BB and cerebral white matter damage in older adults remains unknown. Methods Participants included community-dwelling older adults (age range 55–90 years, M = 73.1 years; SD = 7.5; 61.0% male) from the Alzheimer's Disease Neuroimaging Initiative who underwent venipuncture and blood plasma immunoassay for PDGF-BB, brain MRI scanning with T2-FLAIR for volumetric quantification of white matter hyperintensities (WMH) and APOE4 genotyping (N = 64). Linear regression analyses examined the relationship between plasma PDGF-BB levels and WMH volume, adjusting for age, sex, intracranial volume (ICV) and stratifying by APOE4 status. Results Greater levels of circulating PDGF-BB were related to greater WMH volume, even after accounting for age, sex, ICV and APOE4 carrier status (p = 0.040). Nineteen (29.2%) were APOE4 carriers. When stratified by APOE4 status, the relationship between PDGF-BB and WMH volume was only significant for APOE4 carriers (p = 0.007), but not non-carriers (p = 0.448), after adjusting for age, sex and ICV. Discussion These findings reveal a differential relationship between PDGF-BB and WMH volume for APOE4 carriers versus non-carriers. The APOE4 variant leads to accelerated cerebrovascular injury and cognitive decline. Elevated levels of PDGF-BB in carriers may suggest a role for pericytes and blood-brain barrier dysfunction in white matter damage, vascular cognitive impairment and AD. Additional studies will elucidate the role of PDGF ligands and receptors in these conditions.
Collapse
|
39
|
Peters EC, Gee MT, Pawlowski LN, Kath AM, Polk FD, Vance CJ, Sacoman JL, Pires PW. Amyloid- β disrupts unitary calcium entry through endothelial NMDA receptors in mouse cerebral arteries. J Cereb Blood Flow Metab 2022; 42:145-161. [PMID: 34465229 PMCID: PMC8721780 DOI: 10.1177/0271678x211039592] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 07/14/2021] [Accepted: 07/15/2021] [Indexed: 01/07/2023]
Abstract
Transient increases in intracellular Ca2+ activate endothelium-dependent vasodilatory pathways. This process is impaired in cerebral amyloid angiopathy, where amyloid-β(1-40) accumulates around blood vessels. In neurons, amyloid-β impairs the Ca2+-permeable N-methyl-D-aspartate receptor (NMDAR), a mediator of endothelium-dependent dilation in arteries. We hypothesized that amyloid-β(1-40) reduces NMDAR-elicited Ca2+ signals in mouse cerebral artery endothelial cells, blunting dilation. Cerebral arteries isolated from 4-5 months-old, male and female cdh5:Gcamp8 mice were used for imaging of unitary Ca2+ influx through NMDAR (NMDAR sparklets) and intracellular Ca2+ transients. The NMDAR agonist NMDA (10 µmol/L) increased frequency of NMDAR sparklets and intracellular Ca2+ transients in endothelial cells; these effects were prevented by NMDAR antagonists D-AP5 and MK-801. Next, we tested if amyloid-β(1-40) impairs NMDAR-elicited Ca2+ transients. Cerebral arteries incubated with amyloid-β(1-40) (5 µmol/L) exhibited reduced NMDAR sparklets and intracellular Ca2+ transients. Lastly, we observed that NMDA-induced dilation of pial arteries is reduced by acute intraluminal amyloid-β(1-40), as well as in a mouse model of Alzheimer's disease, the 5x-FAD, linked to downregulation of Grin1 mRNA compared to wild-type littermates. These data suggest that endothelial NMDAR mediate dilation via Ca2+-dependent pathways, a process disrupted by amyloid-β(1-40) and impaired in 5x-FAD mice.
Collapse
Affiliation(s)
- Emily C Peters
- Department of Physiology, University of Arizona College of Medicine Tucson, Tucson, AZ, USA
| | - Michael T Gee
- Department of Physiology, University of Arizona College of Medicine Tucson, Tucson, AZ, USA
| | - Lukas N Pawlowski
- Department of Physiology, University of Arizona College of Medicine Tucson, Tucson, AZ, USA
| | - Allison M Kath
- Department of Physiology, University of Arizona College of Medicine Tucson, Tucson, AZ, USA
| | - Felipe D Polk
- Department of Physiology, University of Arizona College of Medicine Tucson, Tucson, AZ, USA
| | - Christopher J Vance
- Department of Physiology, University of Arizona College of Medicine Tucson, Tucson, AZ, USA
| | - Juliana L Sacoman
- Department of Physiology, University of Arizona College of Medicine Tucson, Tucson, AZ, USA
| | - Paulo W Pires
- Department of Physiology, University of Arizona College of Medicine Tucson, Tucson, AZ, USA
- Sarver Heart Center, University of Arizona College of Medicine Tucson, Tucson, AZ, USA
| |
Collapse
|
40
|
Chen S, Nazeri A, Baek H, Ye D, Yang Y, Yuan J, Rubin JB, Chen H. A review of bioeffects induced by focused ultrasound combined with microbubbles on the neurovascular unit. J Cereb Blood Flow Metab 2022; 42:3-26. [PMID: 34551608 PMCID: PMC8721781 DOI: 10.1177/0271678x211046129] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 08/16/2021] [Accepted: 08/22/2021] [Indexed: 01/29/2023]
Abstract
Focused ultrasound combined with circulating microbubbles (FUS+MB) can transiently enhance blood-brain barrier (BBB) permeability at targeted brain locations. Its great promise in improving drug delivery to the brain is reflected by a rapidly growing number of clinical trials using FUS+MB to treat various brain diseases. As the clinical applications of FUS+MB continue to expand, it is critical to have a better understanding of the molecular and cellular effects induced by FUS+MB to enhance the efficacy of current treatment and enable the discovery of new therapeutic strategies. Existing studies primarily focus on FUS+MB-induced effects on brain endothelial cells, the major cellular component of BBB. However, bioeffects induced by FUS+MB expand beyond the BBB to cells surrounding blood vessels, including astrocytes, microglia, and neurons. Together these cell types comprise the neurovascular unit (NVU). In this review, we examine cell-type-specific bioeffects of FUS+MB on different NVU components, including enhanced permeability in endothelial cells, activation of astrocytes and microglia, as well as increased intraneuron protein metabolism and neuronal activity. Finally, we discuss knowledge gaps that must be addressed to further advance clinical applications of FUS+MB.
Collapse
Affiliation(s)
- Si Chen
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO, USA
| | - Arash Nazeri
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Hongchae Baek
- Imaging Institute and Neurological Institute, Cleveland Clinic, Cleveland Clinic, Cleveland, OH, USA
| | - Dezhuang Ye
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO, USA
| | - Yaoheng Yang
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO, USA
| | - Jinyun Yuan
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO, USA
| | - Joshua B Rubin
- Department of Pediatrics, Washington University School of Medicine, Saint Louis, MO, USA
- Department of Neuroscience, Washington University School of Medicine, Saint Louis, MO, USA
| | - Hong Chen
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO, USA
- Department of Radiation Oncology, Washington University School of Medicine, Saint Louis, MO, USA
| |
Collapse
|
41
|
Tian Y, Zhan Y, Jiang Q, Lu W, Li X. Expression and function of PDGF-C in development and stem cells. Open Biol 2021; 11:210268. [PMID: 34847773 PMCID: PMC8633783 DOI: 10.1098/rsob.210268] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Platelet-derived growth factor C (PDGF-C) is a relatively new member of the PDGF family, discovered nearly 20 years after the finding of platelet-derived growth factor A (PDGF-A) and platelet-derived growth factor B (PDGF-B). PDGF-C is generally expressed in most organs and cell types. Studies from the past 20 years have demonstrated critical roles of PDGF-C in numerous biological, physiological and pathological processes, such as development, angiogenesis, tumour growth, tissue remodelling, wound healing, atherosclerosis, fibrosis, stem/progenitor cell regulation and metabolism. Understanding PDGF-C expression and activities thus will be of great importance to various research disciplines. In this review, however, we mainly discuss the expression and functions of PDGF-C and its receptors in development and stem cells.
Collapse
Affiliation(s)
- Yi Tian
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, People’s Republic of China
| | - Ying Zhan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, People’s Republic of China
| | - Qin Jiang
- Ophthalmic Department, Affiliated Eye Hospital of Nanjing Medical University, Nanjing, People's Republic of China
| | - Weisi Lu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, People’s Republic of China
| | - Xuri Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, People’s Republic of China
| |
Collapse
|
42
|
Guo ZN, Liu J, Chang J, Zhang P, Jin H, Sun X, Yang Y. GAS6/Axl Signaling Modulates Blood-Brain Barrier Function Following Intravenous Thrombolysis in Acute Ischemic Stroke. Front Immunol 2021; 12:742359. [PMID: 34733281 PMCID: PMC8558492 DOI: 10.3389/fimmu.2021.742359] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 09/30/2021] [Indexed: 01/27/2023] Open
Abstract
Background and Purpose Recent studies have shown that several proteins, including Axl, are related to hemorrhagic transformation (HT) following intravenous thrombolysis by affecting blood-brain barrier (BBB) function. However, the effects of these proteins on BBB function have been studied primarily in animal models. In this study, we aimed to identify serum protein markers that predict HT following intravenous thrombolysis in patients with acute ischemic stroke (AIS) and verify whether these serum proteins regulate BBB function and HT in animal stroke models. Methods First, 118 AIS patients were enrolled in this study, including 52 HT patients and 66 non-HT patients. In Step 1, baseline serum levels of Axl, angiopoietin-like 4, C-reactive protein, ferritin, hypoxia-inducible factor-1 alpha, HTRA2, Lipocalin2, matrix metallopeptidase 9, platelet-derived growth factor-BB, and tumor necrosis factor alpha were measured using a quantitative cytokine chip. Next, sequence mutations and variations in genes encoding the differentially expressed proteins identified in Step 1 and subsequent function-related proteins were detected. Finally, we verified whether manipulation of differentially expressed proteins affected BBB function and HT in a hyperglycemia-induced rat stroke model. Results Serum Axl levels were significantly lower in the HT group than in the non-HT group; none of the other protein markers differed significantly between the two groups. Genetic testing revealed that sequence variations of GAS6 (the gene encoding the Axl ligand)-derived long non-coding RNA, GAS6-AS1, were significantly correlated with an increased risk of HT after intravenous thrombolysis. In animal studies, administration of recombinant GAS6 significantly reduced brain infarction and neurological deficits and attenuated BBB disruption and HT. Conclusions Lower serum Axl levels, which may result from sequence variations in GAS6-AS1, are correlated with an increased risk of HT after intravenous thrombolysis in stroke patients. Activation of the Axl signaling pathway by the GAS6 protein may serve as a therapeutic strategy to reduce HT in AIS patients.
Collapse
Affiliation(s)
- Zhen-Ni Guo
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Changchun, China.,Clinical Trial and Research Center for Stroke, Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Jie Liu
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Junlei Chang
- Center for Protein and Cell-based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Peng Zhang
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Changchun, China.,Clinical Trial and Research Center for Stroke, Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Hang Jin
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Xin Sun
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Yi Yang
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Changchun, China.,Clinical Trial and Research Center for Stroke, Department of Neurology, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
43
|
Gastfriend BD, Foreman KL, Katt ME, Palecek SP, Shusta EV. Integrative analysis of the human brain mural cell transcriptome. J Cereb Blood Flow Metab 2021; 41:3052-3068. [PMID: 34027687 PMCID: PMC8756477 DOI: 10.1177/0271678x211013700] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Brain mural cells, including pericytes and vascular smooth muscle cells, are important for vascular development, blood-brain barrier function, and neurovascular coupling, but the molecular characteristics of human brain mural cells are incompletely characterized. Single cell RNA-sequencing (scRNA-seq) is increasingly being applied to assess cellular diversity in the human brain, but the scarcity of mural cells in whole brain samples has limited their molecular profiling. Here, we leverage the combined power of multiple independent human brain scRNA-seq datasets to build a transcriptomic database of human brain mural cells. We use this combined dataset to determine human-mouse species differences in mural cell transcriptomes, culture-induced dedifferentiation of human brain pericytes, and human mural cell organotypicity, with several key findings validated by RNA fluorescence in situ hybridization. Together, this work improves knowledge regarding the molecular constituents of human brain mural cells, serves as a resource for hypothesis generation in understanding brain mural cell function, and will facilitate comparative evaluation of animal and in vitro models.
Collapse
Affiliation(s)
- Benjamin D Gastfriend
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Koji L Foreman
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Moriah E Katt
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Sean P Palecek
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Eric V Shusta
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI, USA.,Department of Neurological Surgery, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
44
|
Ji Y, Wang T, Gao Q, Huang X, Chang J. Normalization of non-canonical Wnt signalings does not compromise blood-brain barrier protection conferred by upregulating endothelial Wnt/β-catenin signaling following ischemic stroke. CNS Neurosci Ther 2021; 27:1085-1096. [PMID: 34056869 PMCID: PMC8339534 DOI: 10.1111/cns.13661] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 05/05/2021] [Accepted: 05/06/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Endothelial canonical (Wnt/β-catenin) and non-canonical Wnt signalings (Wnt/PCP and Wnt/Ca2+ ) promote blood-brain barrier (BBB) development and antagonize each other. However, the effects of ischemic stroke on endothelial canonical and non-canonical Wnt signalings are unclear. Further, how non-canonical Wnt signalings are influenced by upregulation of endothelial Wnt/β-catenin signaling and subsequently affect BBB function following ischemic stroke have not been studied. METHODS First, we determined the levels of Wnt signaling markers including TCF/LEF1 transcription activity, Axin2 mRNA, phospho-JNKThr183/Tyr185 , and NFAT in brain endothelial cells (ECs) with the deletion of Wnt receptor Frizzled (Fzd)4 or Fzd6, the two most abundant Fzds in brain ECs. Next, we observed the effect of ischemia/reperfusion injury on Wnt signalings in brain ECs and adult mice. Last, we assessed the changes of non-canonical Wnt signalings and BBB injury in the early stage of ischemic stroke in mice with endothelial β-catenin activation (β-cat mice). RESULTS Fzd4 or Fzd6 deletion dampened both Wnt/β-catenin and Wnt/PCP signalings but enhanced Wnt/Ca2+ signaling in brain ECs. Both canonical and non-canonical Wnt signalings in brain ECs were downregulated after ischemia/reperfusion injury in vitro and in vivo. Upregulating endothelial Wnt/β-catenin signaling in β-cat mice normalized the downregulated non-canonical Wnt signalings, which did not compromise its protective effects on BBB integrity and endothelial tight junction following ischemic stroke. CONCLUSIONS The BBB protection induced by upregulation of endothelial Wnt/β-catenin signaling may be not interfered by the normalization of non-canonical Wnt signalings in the early stage of ischemic stroke.
Collapse
Affiliation(s)
- Ya‐bin Ji
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular ImmunomodulationInstitute of Biomedicine and BiotechnologyShenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhenChina
- Department of NeurologyNanfang HospitalSouthern Medical UniversityGuangzhouChina
- Baiyun affiliated Cerebrovascular HospitalNanfang Hospital Baiyun BranchSouthern Medical UniversityGuangzhouChina
| | - Tian‐xi Wang
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular ImmunomodulationInstitute of Biomedicine and BiotechnologyShenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhenChina
| | - Qiang Gao
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular ImmunomodulationInstitute of Biomedicine and BiotechnologyShenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhenChina
- Department of NeurosurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou UniversityZhengzhouChina
| | - Xiao‐wen Huang
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular ImmunomodulationInstitute of Biomedicine and BiotechnologyShenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhenChina
| | - Junlei Chang
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular ImmunomodulationInstitute of Biomedicine and BiotechnologyShenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhenChina
| |
Collapse
|
45
|
Chico TJA, Kugler EC. Cerebrovascular development: mechanisms and experimental approaches. Cell Mol Life Sci 2021; 78:4377-4398. [PMID: 33688979 PMCID: PMC8164590 DOI: 10.1007/s00018-021-03790-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 02/04/2021] [Accepted: 02/12/2021] [Indexed: 12/13/2022]
Abstract
The cerebral vasculature plays a central role in human health and disease and possesses several unique anatomic, functional and molecular characteristics. Despite their importance, the mechanisms that determine cerebrovascular development are less well studied than other vascular territories. This is in part due to limitations of existing models and techniques for visualisation and manipulation of the cerebral vasculature. In this review we summarise the experimental approaches used to study the cerebral vessels and the mechanisms that contribute to their development.
Collapse
Affiliation(s)
- Timothy J A Chico
- Department of Infection, Immunity and Cardiovascular Disease, Medical School, University of Sheffield, Beech Hill Road, Sheffield, S10 2RX, UK.
- The Bateson Centre, Firth Court, University of Sheffield, Western Bank, Sheffield, S10 2TN, UK.
- Insigneo Institute for in Silico Medicine, The Pam Liversidge Building, Sheffield, S1 3JD, UK.
| | - Elisabeth C Kugler
- Department of Infection, Immunity and Cardiovascular Disease, Medical School, University of Sheffield, Beech Hill Road, Sheffield, S10 2RX, UK.
- The Bateson Centre, Firth Court, University of Sheffield, Western Bank, Sheffield, S10 2TN, UK.
- Insigneo Institute for in Silico Medicine, The Pam Liversidge Building, Sheffield, S1 3JD, UK.
| |
Collapse
|
46
|
Schofield CL, Rodrigo-Navarro A, Dalby MJ, Van Agtmael T, Salmeron-Sanchez M. Biochemical‐ and Biophysical‐Induced Barriergenesis in the Blood–Brain Barrier: A Review of Barriergenic Factors for Use in In Vitro Models. ADVANCED NANOBIOMED RESEARCH 2021. [DOI: 10.1002/anbr.202000068] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Affiliation(s)
| | | | - Matthew J. Dalby
- Centre for the Cellular Microenvironment University of Glasgow Glasgow UK
| | - Tom Van Agtmael
- Institute of Cardiovascular and Medical Sciences University of Glasgow Glasgow UK
| | | |
Collapse
|
47
|
Zhao M, Wang Z, Yang M, Ding Y, Zhao M, Wu H, Zhang Y, Lu Q. The Roles of Orphan G Protein-Coupled Receptors in Autoimmune Diseases. Clin Rev Allergy Immunol 2021; 60:220-243. [PMID: 33411320 DOI: 10.1007/s12016-020-08829-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/16/2020] [Indexed: 12/26/2022]
Abstract
G protein-coupled receptors (GPCRs) constitute the largest family of plasma membrane receptors in nature and mediate the effects of a variety of extracellular signals, such as hormone, neurotransmitter, odor, and light signals. Due to their involvement in a broad range of physiological and pathological processes and their accessibility, GPCRs are widely used as pharmacological targets of treatment. Orphan G protein-coupled receptors (oGPCRs) are GPCRs for which no natural ligands have been found, and they not only play important roles in various physiological functions, such as sensory perception, reproduction, development, growth, metabolism, and responsiveness, but are also closely related to many major diseases, such as central nervous system (CNS) diseases, metabolic diseases, and cancer. Recently, many studies have reported that oGPCRs play increasingly important roles as key factors in the occurrence and progression of autoimmune diseases. Therefore, oGPCRs are likely to become potential therapeutic targets and may provide a breakthrough in the study of autoimmune diseases. In this article, we focus on reviewing the recent research progress and clinical treatment effects of oGPCRs in three common autoimmune diseases: multiple sclerosis (MS), rheumatoid arthritis (RA), and systemic lupus erythematosus (SLE), shedding light on novel strategies for treatments.
Collapse
Affiliation(s)
- Mingming Zhao
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zheyu Wang
- University of South China, Hengyang, Hunan, China.,Maternal & Child Health Care Hospital Hainan Province, Haikou, Hainan, China
| | - Ming Yang
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yan Ding
- Maternal & Child Health Care Hospital Hainan Province, Haikou, Hainan, China.,Hainan Province Dermatol Disease Hospital, Haikou, Hainan, China
| | - Ming Zhao
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Haijing Wu
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, Second Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Yan Zhang
- Department of Biophysics, and Department of Pathology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China. .,Zhejiang Laboratory for Systems & Precison Medicine, Zhejiang University Medical Center, Hangzhou, 311121, China. .,Zhejiang Provincial Key Laboratory of Immunity and Inflammatory Diseases, Hangzhou, 310058, China. .,MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Medicine, Hangzhou, 310058, China.
| | - Qianjin Lu
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, Second Xiangya Hospital, Central South University, Changsha, Hunan, China. .,Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China.
| |
Collapse
|
48
|
Girolamo F, de Trizio I, Errede M, Longo G, d'Amati A, Virgintino D. Neural crest cell-derived pericytes act as pro-angiogenic cells in human neocortex development and gliomas. Fluids Barriers CNS 2021; 18:14. [PMID: 33743764 PMCID: PMC7980348 DOI: 10.1186/s12987-021-00242-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 02/13/2021] [Indexed: 02/07/2023] Open
Abstract
Central nervous system diseases involving the parenchymal microvessels are frequently associated with a ‘microvasculopathy’, which includes different levels of neurovascular unit (NVU) dysfunction, including blood–brain barrier alterations. To contribute to the understanding of NVU responses to pathological noxae, we have focused on one of its cellular components, the microvascular pericytes, highlighting unique features of brain pericytes with the aid of the analyses carried out during vascularization of human developing neocortex and in human gliomas. Thanks to their position, centred within the endothelial/glial partition of the vessel basal lamina and therefore inserted between endothelial cells and the perivascular and vessel-associated components (astrocytes, oligodendrocyte precursor cells (OPCs)/NG2-glia, microglia, macrophages, nerve terminals), pericytes fulfil a central role within the microvessel NVU. Indeed, at this critical site, pericytes have a number of direct and extracellular matrix molecule- and soluble factor-mediated functions, displaying marked phenotypical and functional heterogeneity and carrying out multitasking services. This pericytes heterogeneity is primarily linked to their position in specific tissue and organ microenvironments and, most importantly, to their ontogeny. During ontogenesis, pericyte subtypes belong to two main embryonic germ layers, mesoderm and (neuro)ectoderm, and are therefore expected to be found in organs ontogenetically different, nonetheless, pericytes of different origin may converge and colonize neighbouring areas of the same organ/apparatus. Here, we provide a brief overview of the unusual roles played by forebrain pericytes in the processes of angiogenesis and barriergenesis by virtue of their origin from midbrain neural crest stem cells. A better knowledge of the ontogenetic subpopulations may support the understanding of specific interactions and mechanisms involved in pericyte function/dysfunction, including normal and pathological angiogenesis, thereby offering an alternative perspective on cell subtype-specific therapeutic approaches. ![]()
Collapse
Affiliation(s)
- Francesco Girolamo
- Department of Basic Medical Sciences, Neuroscience and Sensory Organs, Human Anatomy and Histology Unit, University of Bari School of Medicine, Bari, Italy.
| | - Ignazio de Trizio
- Department of Basic Medical Sciences, Neuroscience and Sensory Organs, Human Anatomy and Histology Unit, University of Bari School of Medicine, Bari, Italy.,Intensive Care Unit, Department of Intensive Care, Regional Hospital of Lugano, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Mariella Errede
- Department of Basic Medical Sciences, Neuroscience and Sensory Organs, Human Anatomy and Histology Unit, University of Bari School of Medicine, Bari, Italy
| | - Giovanna Longo
- Department of Basic Medical Sciences, Neuroscience and Sensory Organs, Molecular Biology Unit, University of Bari School of Medicine, Bari, Italy
| | - Antonio d'Amati
- Department of Basic Medical Sciences, Neuroscience and Sensory Organs, Human Anatomy and Histology Unit, University of Bari School of Medicine, Bari, Italy.,Department of Emergency and Organ Transplantation, Pathology Section, University of Bari School of Medicine, Bari, Italy
| | - Daniela Virgintino
- Department of Basic Medical Sciences, Neuroscience and Sensory Organs, Human Anatomy and Histology Unit, University of Bari School of Medicine, Bari, Italy
| |
Collapse
|
49
|
Girolamo F, de Trizio I, Errede M, Longo G, d’Amati A, Virgintino D. Neural crest cell-derived pericytes act as pro-angiogenic cells in human neocortex development and gliomas. Fluids Barriers CNS 2021. [DOI: 10.1186/s12987-021-00242-7 union select null--] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
AbstractCentral nervous system diseases involving the parenchymal microvessels are frequently associated with a ‘microvasculopathy’, which includes different levels of neurovascular unit (NVU) dysfunction, including blood–brain barrier alterations. To contribute to the understanding of NVU responses to pathological noxae, we have focused on one of its cellular components, the microvascular pericytes, highlighting unique features of brain pericytes with the aid of the analyses carried out during vascularization of human developing neocortex and in human gliomas. Thanks to their position, centred within the endothelial/glial partition of the vessel basal lamina and therefore inserted between endothelial cells and the perivascular and vessel-associated components (astrocytes, oligodendrocyte precursor cells (OPCs)/NG2-glia, microglia, macrophages, nerve terminals), pericytes fulfil a central role within the microvessel NVU. Indeed, at this critical site, pericytes have a number of direct and extracellular matrix molecule- and soluble factor-mediated functions, displaying marked phenotypical and functional heterogeneity and carrying out multitasking services. This pericytes heterogeneity is primarily linked to their position in specific tissue and organ microenvironments and, most importantly, to their ontogeny. During ontogenesis, pericyte subtypes belong to two main embryonic germ layers, mesoderm and (neuro)ectoderm, and are therefore expected to be found in organs ontogenetically different, nonetheless, pericytes of different origin may converge and colonize neighbouring areas of the same organ/apparatus. Here, we provide a brief overview of the unusual roles played by forebrain pericytes in the processes of angiogenesis and barriergenesis by virtue of their origin from midbrain neural crest stem cells. A better knowledge of the ontogenetic subpopulations may support the understanding of specific interactions and mechanisms involved in pericyte function/dysfunction, including normal and pathological angiogenesis, thereby offering an alternative perspective on cell subtype-specific therapeutic approaches.
Collapse
|
50
|
Nguyen QL, Okuno N, Hamashima T, Dang ST, Fujikawa M, Ishii Y, Enomoto A, Maki T, Nguyen HN, Nguyen VT, Fujimori T, Mori H, Andrae J, Betsholtz C, Takao K, Yamamoto S, Sasahara M. Vascular PDGFR-alpha protects against BBB dysfunction after stroke in mice. Angiogenesis 2021; 24:35-46. [PMID: 32918673 DOI: 10.1007/s10456-020-09742-w] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 09/01/2020] [Indexed: 02/06/2023]
Abstract
Blood-brain barrier (BBB) dysfunction underlies the pathogenesis of many neurological diseases. Platelet-derived growth factor receptor-alpha (PDGFRα) induces hemorrhagic transformation (HT) downstream of tissue plasminogen activator in thrombolytic therapy of acute stroke. Thus, PDGFs are attractive therapeutic targets for BBB dysfunction. In the present study, we examined the role of PDGF signaling in the process of tissue remodeling after middle cerebral arterial occlusion (MCAO) in mice. Firstly, we found that imatinib increased lesion size after permanent MCAO in wild-type mice. Moreover, imatinib-induced HT only when administrated in the subacute phase of MCAO, but not in the acute phase. Secondly, we generated genetically mutated mice (C-KO mice) that showed decreased expression of perivascular PDGFRα. Additionally, transient MCAO experiments were performed in these mice. We found that the ischemic lesion size was not affected; however, the recruitment of PDGFRα/type I collagen-expressing perivascular cells was significantly downregulated, and HT and IgG leakage was augmented only in the subacute phase of stroke in C-KO mice. In both experiments, we found that the expression of tight junction proteins and PDGFRβ-expressing pericyte coverage was not significantly affected in imatinib-treated mice and in C-KO mice. The specific implication of PDGFRα signaling was suggestive of protective effects against BBB dysfunction during the subacute phase of stroke. Vascular TGF-β1 expression was downregulated in both imatinib-treated and C-KO mice, along with sustained levels of MMP9. Therefore, PDGFRα effects may be mediated by TGF-β1 which exerts potent protective effects in the BBB.
Collapse
Affiliation(s)
- Quang Linh Nguyen
- Department of Pathology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, 930-0194, Japan
- Stroke Center, The 108 Military Central Hospital, Ha Noi, Vietnam
| | - Noriko Okuno
- Department of Pathology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, 930-0194, Japan
| | - Takeru Hamashima
- Department of Pathology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, 930-0194, Japan
| | - Son Tung Dang
- Department of Pathology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, 930-0194, Japan
| | - Miwa Fujikawa
- Department of Pathology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, 930-0194, Japan
| | - Yoko Ishii
- Department of Health Science, Faculty of Health and Human Development, The University of Nagano, Nagano, Japan
| | - Atsushi Enomoto
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takakuni Maki
- Department of Neurology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | | | - Van Tuyen Nguyen
- Stroke Center, The 108 Military Central Hospital, Ha Noi, Vietnam
| | - Toshihiko Fujimori
- Division of Embryology, National Institute for Basic Biology, Okazaki, Japan
| | - Hisashi Mori
- Department of Molecular Neuroscience, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Johanna Andrae
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Christer Betsholtz
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
- Integrated Cardio Metabolic Center, Karolinska Institute, Huddinge, Sweden
| | - Keizo Takao
- Division of Animal Resources and Development, Life Science Research Center, University of Toyama, Toyama, Japan
| | - Seiji Yamamoto
- Department of Pathology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, 930-0194, Japan.
| | - Masakiyo Sasahara
- Department of Pathology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, 930-0194, Japan.
| |
Collapse
|