1
|
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 2025; 45:617-629. [PMID: 39053491 PMCID: PMC11571979 DOI: 10.1177/0271678x241261946] [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: 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
|
2
|
Geng Z, Zhang Z, Wang M, Yu Z, Wang S, Lu J, Wang S, Guan S, Li J, Liu T, Zhu C. Targeting stromal cells in tumor microenvironment as a novel treatment strategy for glioma. Cancer Cell Int 2025; 25:58. [PMID: 39985022 PMCID: PMC11846374 DOI: 10.1186/s12935-025-03692-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Accepted: 02/12/2025] [Indexed: 02/23/2025] Open
Abstract
Glioma is the most common primary malignant tumor of the central nervous system in adults, characterized by high mortality, low cure rate and high recurrence rate. Among gliomas, glioblastoma multiforme (GBM) is the most malignant subtype. Currently, the standard treatment for patients with GBM is maximum surgical excision combined with radiotherapy and chemotherapy. But only a small percentage of patients benefit from this standard treatment. The tumor microenvironment plays an important role in the occurrence and development of most tumors. It is primarily composed of tumor cells, peripheral blood vessels, extracellular matrix, signaling molecules, stromal cells, and immune cells. The role of stromal cells in GBM has emerged as the focus of current research. The interaction among tumor, stromal, and immune cells within the tumor microenvironment can influence tumor development. Traditional research and drug therapy in glioma mainly focus on the tumor cells themselves, but recent studies have found that targeting stromal cells in the tumor microenvironment can also modulate tumor progression in GBM. Here, we review the influence of stromal cells in the tumor microenvironment of GBM on tumor cells and its related mechanism, as well as related molecular targets and signaling pathways, providing new ideas for the treatment and prognosis of GBM.
Collapse
Affiliation(s)
- Ziang Geng
- Department of Neurosurgery, The First Hospital of China Medical University, Nanjing Street 155, Heping District, Shenyang, 110001, Liaoning, China
| | - Zheyuan Zhang
- Department of Surgical Oncology and Breast Surgery, The First Hospital of China Medical University, Nanjing Street 155, Heping District, Shenyang, 110001, Liaoning, China
| | - Miaohan Wang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100021, China
| | - Zhongxue Yu
- Department of Cardiovascular Ultrasound, The First Hospital of China Medical University, Nanjing Street 155, Heping District, Shenyang, 110001, Liaoning, China
| | - Siqi Wang
- Department of Radiation Oncology, The First Hospital of China Medical University, Nanjing Street 155, Heping District, Shenyang, 110001, Liaoning, China
| | - Jun Lu
- Department of Neurosurgery, The First Hospital of China Medical University, Nanjing Street 155, Heping District, Shenyang, 110001, Liaoning, China
| | - Shisong Wang
- Department of Neurosurgery, The First Hospital of China Medical University, Nanjing Street 155, Heping District, Shenyang, 110001, Liaoning, China
| | - Shu Guan
- Department of Surgical Oncology and Breast Surgery, The First Hospital of China Medical University, Nanjing Street 155, Heping District, Shenyang, 110001, Liaoning, China
| | - Jinna Li
- Department of Oncology, Shengjing Hospital of China Medical University, Sanhao Street 36, Heping District, Shenyang, 110001, Liaoning, China.
| | - Tiancong Liu
- Department of Otolaryngology, Shengjing Hospital of China Medical University, Sanhao Street 36, Heping District, Shenyang, 110001, Liaoning, China.
| | - Chen Zhu
- Department of Neurosurgery, The First Hospital of China Medical University, Nanjing Street 155, Heping District, Shenyang, 110001, Liaoning, China.
| |
Collapse
|
3
|
Salazar-Saura I, Pinilla-Sala M, Megías J, Navarro L, Roselló-Sastre E, San-Miguel T. Pericytes in Glioblastoma: Hidden Regulators of Tumor Vasculature and Therapy Resistance. Cancers (Basel) 2024; 17:15. [PMID: 39796646 PMCID: PMC11718950 DOI: 10.3390/cancers17010015] [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: 11/27/2024] [Revised: 12/19/2024] [Accepted: 12/22/2024] [Indexed: 01/13/2025] Open
Abstract
Glioblastoma IDH wild type (GB), the most common malignant primary brain tumor, is characterized by rapid proliferation, extensive infiltration into surrounding brain tissue, and significant resistance to current therapies. Median survival is only 15 months despite extensive clinical efforts. The tumor microenvironment (TME) in GB is highly specialized, supporting the tumor's aggressive behavior and its ability to evade conventional treatments. One critical component is the aberrant vascular network that complicates the delivery of chemotherapy across the blood-brain barrier. Antiangiogenic therapies emerged as a promising option but have shown limited efficacy in extending the survival of these patients. Comprehension of the complex vascular network of GB may be a key to overcoming the limitations of current therapies. Pericytes are gaining recognition within the context of the TME. These mural cells are essential for vascular integrity and may contribute to tumor progression and therapeutic resistance. Although their role has been evidenced in other tumors, they remain underexplored in GB. Pericytes are known to respond to tumor hypoxia and interact with vascular endothelia, influencing responses to DNA damage and antiangiogenic treatments. They actively regulate not only angiogenesis but also the different vasculogenic strategies for tumor neovascularization. Additionally, they affect leukocyte trafficking and tumor-associated macrophages. This review aims to integrate the various functions controlled by pericytes to favor deeper investigation into their actionable potential. Pericytes may represent a promising target for novel therapeutic strategies in order to improve patient outcomes.
Collapse
Affiliation(s)
- Irene Salazar-Saura
- Pathology Service, Consorcio Hospital General Universitario de Valencia, 46014 Valencia, Spain; (I.S.-S.); (L.N.); (E.R.-S.)
| | - María Pinilla-Sala
- Research Group on Tumors of the Central Nervous System, Pathology Department, University of Valencia, 46010 Valencia, Spain;
- INCLIVA Foundation, 46010 Valencia, Spain
| | - Javier Megías
- Research Group on Tumors of the Central Nervous System, Pathology Department, University of Valencia, 46010 Valencia, Spain;
- INCLIVA Foundation, 46010 Valencia, Spain
| | - Lara Navarro
- Pathology Service, Consorcio Hospital General Universitario de Valencia, 46014 Valencia, Spain; (I.S.-S.); (L.N.); (E.R.-S.)
| | - Esther Roselló-Sastre
- Pathology Service, Consorcio Hospital General Universitario de Valencia, 46014 Valencia, Spain; (I.S.-S.); (L.N.); (E.R.-S.)
| | - Teresa San-Miguel
- Research Group on Tumors of the Central Nervous System, Pathology Department, University of Valencia, 46010 Valencia, Spain;
- INCLIVA Foundation, 46010 Valencia, Spain
| |
Collapse
|
4
|
Luo N, Zhu W, Li X, Fu M, Zhang Y, Yang F, Zhang Y, Chen Z, Zhang Q, Peng B, Li Q, Chen X, Liu Y, Hu G, Peng X. Defective autophagy of pericytes enhances radiation-induced senescence promoting radiation brain injury. Neuro Oncol 2024; 26:2288-2304. [PMID: 39110121 PMCID: PMC11630511 DOI: 10.1093/neuonc/noae153] [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] [Indexed: 12/12/2024] Open
Abstract
BACKGROUND Radiation-induced brain injury (RBI) represents a major challenge for cancer patients undergoing cranial radiotherapy. However, the molecular mechanisms and therapeutic strategies of RBI remain inconclusive. With the continuous exploration of the mechanisms of RBI, an increasing number of studies have implicated cerebrovascular dysfunction as a key factor in RBI-related cognitive impairment. As pericytes are a component of the neurovascular unit, there is still a lack of understanding in current research about the specific role and function of pericytes in RBI. METHODS We constructed a mouse model of RBI-associated cognitive dysfunction in vivo and an in vitro radiation-induced pericyte model to explore the effects of senescent pericytes on the blood-brain barrier (BBB) and normal central nervous system cells, even glioma cells. To further clarify the effects of pericyte autophagy on senescence, molecular mechanisms were explored at the animal and cellular levels. Finally, we validated the clearance of pericyte senescence by using a senolytic drug and all-trans retinoic acid to investigate the role of radiation-induced pericyte senescence. RESULTS Our findings indicated that radiation-induced pericyte senescence plays a key role in BBB dysfunction, leading to RBI and subsequent cognitive decline. Strikingly, pericyte senescence also contributed to the growth and invasion of glioma cells. We further demonstrated that defective autophagy in pericytes is a vital regulatory mechanism for pericyte senescence. Moreover, autophagy activated by rapamycin could reverse pericyte senescence. Notably, the elimination of senescent cells by senolytic drugs significantly mitigated radiation-induced cognitive dysfunction. CONCLUSIONS Our results demonstrated that pericyte senescence may be a promising therapeutic target for RBI and glioma progression.
Collapse
Affiliation(s)
- Na Luo
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenjun Zhu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoyu Li
- Department of Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Min Fu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | | | - Feng Yang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yiling Zhang
- Department of Radiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ziqi Chen
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qiang Zhang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bi Peng
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qianxia Li
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xin Chen
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuanhui Liu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guangyuan Hu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaohong Peng
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
5
|
Dai J, Jiang Y, Hu H, Zhang S, Chen Y. Extracellular vesicles as modulators of glioblastoma progression and tumor microenvironment. Pathol Oncol Res 2024; 30:1611549. [PMID: 38379858 PMCID: PMC10876843 DOI: 10.3389/pore.2024.1611549] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 01/16/2024] [Indexed: 02/22/2024]
Abstract
Glioblastoma is the most aggressive brain tumor with extremely poor prognosis in adults. Routine treatments include surgery, chemotherapy, and radiotherapy; however, these may lead to rapid relapse and development of therapy-resistant tumor. Glioblastoma cells are known to communicate with macrophages, microglia, endothelial cells, astrocytes, and immune cells in the tumor microenvironment (TME) to promote tumor preservation. It was recently demonstrated that Glioblastoma-derived extracellular vesicles (EVs) participate in bidirectional intercellular communication in the TME. Apart from promoting glioblastoma cell proliferation, migration, and angiogenesis, EVs and their cargos (primarily proteins and miRNAs) can act as biomarkers for tumor diagnosis and prognosis. Furthermore, they can be used as therapeutic tools. In this review, the mechanisms of Glioblastoma-EVs biogenesis and intercellular communication with TME have been summarized. Moreover, there is discussion surrounding EVs as novel diagnostic structures and therapeutic tools for glioblastoma. Finally, unclear questions that require future investigation have been reviewed.
Collapse
Affiliation(s)
- Jie Dai
- Department of Pathology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Yong Jiang
- Department of Neurosurgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Haoyue Hu
- Department of Medical Oncology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Shuang Zhang
- Department of Pathology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Yue Chen
- Department of Pathology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
6
|
Merk L, Regel K, Eckhardt H, Evers M, El-Ayoubi A, Mittelbronn M, Krüger M, Gérardy JJ, Mack AF, Naumann U. Blocking TGF-β- and Epithelial-to-Mesenchymal Transition (EMT)-mediated activation of vessel-associated mural cells in glioblastoma impacts tumor angiogenesis. FREE NEUROPATHOLOGY 2024; 5:4. [PMID: 38455669 PMCID: PMC10919159 DOI: 10.17879/freeneuropathology-2024-5188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 02/07/2024] [Indexed: 03/09/2024]
Abstract
Glioblastoma (GBM) is the most common malignant primary brain tumor in adults. GBM displays excessive and unfunctional vascularization which may, among others, be a reason for its devastating prognosis. Pericytes have been identified as the major component of the irregular vessel structure in GBM. In vitro data suggest an epithelial-to-mesenchymal transition (EMT)-like activation of glioma-associated pericytes, stimulated by GBM-secreted TGF-β, to be involved in the formation of a chaotic and dysfunctional tumor vasculature. This study investigated whether TGF-β impacts the function of vessel associated mural cells (VAMCs) in vivo via the induction of the EMT transcription factor SLUG and whether this is associated with the development of GBM-associated vascular abnormalities. Upon preventing the TGF-β-/SLUG-mediated EMT induction in VAMCs, the number of PDGFRβ and αSMA positive cells was significantly reduced, regardless of whether TGF-β secretion by GBM cells was blocked or whether SLUG was specifically knocked out in VAMCs. The reduced amount of PDGFRβ+ or αSMA+ cells observed under those conditions correlated with a lower vessel density and fewer vascular abnormalities. Our data provide evidence that the SLUG-mediated modulation of VAMC activity is induced by GBM-secreted TGF-β¬ and that activated VAMCs are key contributors in neo-angiogenic processes. We suggest that a pathologically altered activation of GA-Peris in the tumor microenvironment is responsible for the unstructured tumor vasculature. There is emerging evidence that vessel normalization alleviates tumor hypoxia, reduces tumor-associated edema and improves drug delivery. Therefore, avoiding the generation of an unstructured and non-functional tumor vasculature during tumor recurrence might be a promising treatment approach for GBM and identifies pericytes as a potential novel therapeutic target.
Collapse
Affiliation(s)
- Luisa Merk
- Molecular Neuro-Oncology, Department of Vascular Neurology, Hertie Institute for Clinical Brain Research and Center Neurology, University Hospital of Tübingen, Germany
| | - Katja Regel
- Molecular Neuro-Oncology, Department of Vascular Neurology, Hertie Institute for Clinical Brain Research and Center Neurology, University Hospital of Tübingen, Germany
| | - Hermann Eckhardt
- Molecular Neuro-Oncology, Department of Vascular Neurology, Hertie Institute for Clinical Brain Research and Center Neurology, University Hospital of Tübingen, Germany
| | - Marietheres Evers
- Molecular Neuro-Oncology, Department of Vascular Neurology, Hertie Institute for Clinical Brain Research and Center Neurology, University Hospital of Tübingen, Germany
| | - Ali El-Ayoubi
- Molecular Neuro-Oncology, Department of Vascular Neurology, Hertie Institute for Clinical Brain Research and Center Neurology, University Hospital of Tübingen, Germany
| | - Michel Mittelbronn
- Department of Cancer Research (DOCR), Luxembourg Institute of Health (LIH), Luxembourg
- Luxembourg Centre of Neuropathology (LCNP), Luxembourg
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Luxembourg
- Department of Life Sciences and Medicine (DLSM), University of Luxembourg, Esch-sur-Alzette, Luxembourg
- Faculty of Science, Technology and Medicine (FSTM), University of Luxembourg, Esch-sur-Alzette, Luxembourg
- National Center of Pathology (NCP), Laboratoire Nationale de Santé (LNS), Luxembourg
| | - Marcel Krüger
- Department of Preclinical Imaging and Radiopharmacy, University of Tübingen, Tübingen, Germany
| | - Jean-Jacques Gérardy
- Luxembourg Centre of Neuropathology (LCNP), Luxembourg
- National Center of Pathology (NCP), Laboratoire Nationale de Santé (LNS), Luxembourg
| | - Andreas F. Mack
- Institute for Clinical Anatomy and Cell Analytics, University of Tübingen, Germany
| | - Ulrike Naumann
- Molecular Neuro-Oncology, Department of Vascular Neurology, Hertie Institute for Clinical Brain Research and Center Neurology, University Hospital of Tübingen, Germany
- Gene and RNA Therapy Center (GRTC), Faculty of Medicine University Tübingen, Germany
| |
Collapse
|
7
|
Waseem A, Rashid S, Rashid K, Khan MA, Khan R, Haque R, Seth P, Raza SS. Insight into the transcription factors regulating Ischemic Stroke and Glioma in Response to Shared Stimuli. Semin Cancer Biol 2023; 92:102-127. [PMID: 37054904 DOI: 10.1016/j.semcancer.2023.04.006] [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: 11/23/2022] [Revised: 03/28/2023] [Accepted: 04/09/2023] [Indexed: 04/15/2023]
Abstract
Cerebral ischemic stroke and glioma are the two leading causes of patient mortality globally. Despite physiological variations, 1 in 10 people who have an ischemic stroke go on to develop brain cancer, most notably gliomas. In addition, glioma treatments have also been shown to increase the risk of ischemic strokes. Stroke occurs more frequently in cancer patients than in the general population, according to traditional literature. Unbelievably, these events share multiple pathways, but the precise mechanism underlying their co-occurrence remains unknown. Transcription factors (TFs), the main components of gene expression programmes, finally determine the fate of cells and homeostasis. Both ischemic stroke and glioma exhibit aberrant expression of a large number of TFs, which are strongly linked to the pathophysiology and progression of both diseases. The precise genomic binding locations of TFs and how TF binding ultimately relates to transcriptional regulation remain elusive despite a strong interest in understanding how TFs regulate gene expression in both stroke and glioma. As a result, the importance of continuing efforts to understand TF-mediated gene regulation is highlighted in this review, along with some of the primary shared events in stroke and glioma.
Collapse
Affiliation(s)
- Arshi Waseem
- Laboratory for Stem Cell & Restorative Neurology, Department of Biotechnology, Era's Lucknow Medical College and Hospital, Era University, Sarfarazganj, Lucknow-226003, India
| | - Sumaiya Rashid
- Department of Pharmacology & Toxicology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, P.O. Box 173, Al-Kharj 11942, Saudi Arabia
| | - Khalid Rashid
- Department of Cancer Biology, Vontz Center for Molecular Studies, Cincinnati, OH 45267-0521
| | | | - Rehan Khan
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City,Mohali, Punjab 140306, India
| | - Rizwanul Haque
- Department of Biotechnology, Central University of South Bihar, Gaya -824236, India
| | - Pankaj Seth
- Molecular and Cellular Neuroscience, Neurovirology Section, National Brain Research Centre, Manesar, Haryana-122052, India
| | - Syed Shadab Raza
- Laboratory for Stem Cell & Restorative Neurology, Department of Biotechnology, Era's Lucknow Medical College and Hospital, Era University, Sarfarazganj, Lucknow-226003, India; Department of Stem Cell Biology and Regenerative Medicine, Era's Lucknow Medical College Hospital, Era University, Sarfarazganj, Lucknow-226003, India
| |
Collapse
|
8
|
Schumacher L, Slimani R, Zizmare L, Ehlers J, Kleine Borgmann F, Fitzgerald JC, Fallier-Becker P, Beckmann A, Grißmer A, Meier C, El-Ayoubi A, Devraj K, Mittelbronn M, Trautwein C, Naumann U. TGF-Beta Modulates the Integrity of the Blood Brain Barrier In Vitro, and Is Associated with Metabolic Alterations in Pericytes. Biomedicines 2023; 11:214. [PMID: 36672722 PMCID: PMC9855966 DOI: 10.3390/biomedicines11010214] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 01/11/2023] [Indexed: 01/18/2023] Open
Abstract
The blood-brain barrier (BBB) is a selectively permeable boundary that separates the circulating blood from the extracellular fluid of the brain and is an essential component for brain homeostasis. In glioblastoma (GBM), the BBB of peritumoral vessels is often disrupted. Pericytes, being important to maintaining BBB integrity, can be functionally modified by GBM cells which induce proliferation and cell motility via the TGF-β-mediated induction of central epithelial to mesenchymal transition (EMT) factors. We demonstrate that pericytes strengthen the integrity of the BBB in primary endothelial cell/pericyte co-cultures as an in vitro BBB model, using TEER measurement of the barrier integrity. In contrast, this effect was abrogated by TGF-β or conditioned medium from TGF-β secreting GBM cells, leading to the disruption of a so far intact and tight BBB. TGF-β notably changed the metabolic behavior of pericytes, by shutting down the TCA cycle, driving energy generation from oxidative phosphorylation towards glycolysis, and by modulating pathways that are necessary for the biosynthesis of molecules used for proliferation and cell division. Combined metabolomic and transcriptomic analyses further underscored that the observed functional and metabolic changes of TGF-β-treated pericytes are closely connected with their role as important supporting cells during angiogenic processes.
Collapse
Affiliation(s)
- Leonie Schumacher
- Molecular Neurooncology, Department of Vascular Neurology, Hertie Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, 72076 Tübingen, Germany
| | - Rédouane Slimani
- Department of Cancer Research (DOCR), Luxembourg Institute of Health (LIH), 1445 Strassen, Luxembourg
- Luxembourg Centre of Neuropathology (LCNP), 3555 Dudelange, Luxembourg
| | - Laimdota Zizmare
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, University of Tübingen, 72076 Tübingen, Germany
| | - Jakob Ehlers
- Molecular Neurooncology, Department of Vascular Neurology, Hertie Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, 72076 Tübingen, Germany
| | - Felix Kleine Borgmann
- Department of Cancer Research (DOCR), Luxembourg Institute of Health (LIH), 1445 Strassen, Luxembourg
- Luxembourg Centre of Neuropathology (LCNP), 3555 Dudelange, Luxembourg
| | - Julia C. Fitzgerald
- Mitochondrial Biology of Parkinson’s Disease, Department of Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, 72076 Tübingen, Germany
| | - Petra Fallier-Becker
- Institute for Pathology and Neuropathology, University of Tübingen, 72076 Tübingen, Germany
| | - Anja Beckmann
- Department of Anatomy and Cell Biology, Saarland University, 66421 Homburg, Germany
| | - Alexander Grißmer
- Department of Anatomy and Cell Biology, Saarland University, 66421 Homburg, Germany
| | - Carola Meier
- Department of Anatomy and Cell Biology, Saarland University, 66421 Homburg, Germany
| | - Ali El-Ayoubi
- Molecular Neurooncology, Department of Vascular Neurology, Hertie Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, 72076 Tübingen, Germany
| | - Kavi Devraj
- German Cancer Consortium (DKTK), 69120 Heidelberg, Germany
- Edinger Institute (Neurological Institute), Goethe University Hospital, 60528 Frankfurt am Main, Germany
| | - Michel Mittelbronn
- Department of Cancer Research (DOCR), Luxembourg Institute of Health (LIH), 1445 Strassen, Luxembourg
- Luxembourg Centre of Neuropathology (LCNP), 3555 Dudelange, Luxembourg
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 4365 Esch-sur-Alzette, Luxembourg
- Department of Life Sciences and Medicine (DLSM), University of Luxembourg, 4365 Esch-sur-Alzette, Luxembourg
- Faculty of Science, Technology and Medicine (FSTM), University of Luxembourg, 4365 Esch-sur-Alzette, Luxembourg
- National Center of Pathology (NCP), Laboratoire Nationale de Santé (LNS), 3555 Dudelange, Luxembourg
| | - Christoph Trautwein
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, University of Tübingen, 72076 Tübingen, Germany
| | - Ulrike Naumann
- Molecular Neurooncology, Department of Vascular Neurology, Hertie Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, 72076 Tübingen, Germany
| |
Collapse
|
9
|
Lin W, Wang Q, Chen Y, Wang N, Ni Q, Qi C, Wang Q, Zhu Y. Identification of a 6-RBP gene signature for a comprehensive analysis of glioma and ischemic stroke: Cognitive impairment and aging-related hypoxic stress. Front Aging Neurosci 2022; 14:951197. [PMID: 36118697 PMCID: PMC9476601 DOI: 10.3389/fnagi.2022.951197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 07/25/2022] [Indexed: 11/13/2022] Open
Abstract
There is mounting evidence that ischemic cerebral infarction contributes to vascular cognitive impairment and dementia in elderly. Ischemic stroke and glioma are two majorly fatal diseases worldwide, which promote each other's development based on some common underlying mechanisms. As a post-transcriptional regulatory protein, RNA-binding protein is important in the development of a tumor and ischemic stroke (IS). The purpose of this study was to search for a group of RNA-binding protein (RBP) gene markers related to the prognosis of glioma and the occurrence of IS, and elucidate their underlying mechanisms in glioma and IS. First, a 6-RBP (POLR2F, DYNC1H1, SMAD9, TRIM21, BRCA1, and ERI1) gene signature (RBPS) showing an independent overall survival prognostic prediction was identified using the transcriptome data from TCGA-glioma cohort (n = 677); following which, it was independently verified in the CGGA-glioma cohort (n = 970). A nomogram, including RBPS, 1p19q codeletion, radiotherapy, chemotherapy, grade, and age, was established to predict the overall survival of patients with glioma, convenient for further clinical transformation. In addition, an automatic machine learning classification model based on radiomics features from MRI was developed to stratify according to the RBPS risk. The RBPS was associated with immunosuppression, energy metabolism, and tumor growth of gliomas. Subsequently, the six RBP genes from blood samples showed good classification performance for IS diagnosis (AUC = 0.95, 95% CI: 0.902–0.997). The RBPS was associated with hypoxic responses, angiogenesis, and increased coagulation in IS. Upregulation of SMAD9 was associated with dementia, while downregulation of POLR2F was associated with aging-related hypoxic stress. Irf5/Trim21 in microglia and Taf7/Trim21 in pericytes from the mouse cerebral cortex were identified as RBPS-related molecules in each cell type under hypoxic conditions. The RBPS is expected to serve as a novel biomarker for studying the common mechanisms underlying glioma and IS.
Collapse
Affiliation(s)
- Weiwei Lin
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases of Zhejiang, Hangzhou, China
| | - Qiangwei Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases of Zhejiang, Hangzhou, China
| | - Yisheng Chen
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ning Wang
- Brain Center, Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qingbin Ni
- Postdoctoral Workstation, Department of Central Laboratory, The Affiliated Taian City Central Hospital of Qingdao University, Taian, China
| | - Chunhua Qi
- Postdoctoral Workstation, Department of Central Laboratory, The Affiliated Taian City Central Hospital of Qingdao University, Taian, China
| | - Qian Wang
- Postdoctoral Workstation, Department of Central Laboratory, The Affiliated Taian City Central Hospital of Qingdao University, Taian, China
- *Correspondence: Qian Wang
| | - Yongjian Zhu
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases of Zhejiang, Hangzhou, China
- College of Mathematical Medicine, Zhejiang Normal University, Jinhua, China
- Yongjian Zhu
| |
Collapse
|
10
|
Shuai W, Chen Q, Zhou X. Up-regulation of MiR-146b-5p Inhibits Fibrotic Lung Pericytes via Inactivation of the Notch1/PDGFRβ/ROCK1 Pathway. Folia Biol (Praha) 2022; 68:180-188. [PMID: 37256552 DOI: 10.14712/fb2022068050180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Lung fibrosis is a serious human pathology. MiR-146b-5p is down-regulated in idiopathic pulmonary fibrosis, and the Notch1/PDGFRβ/ROCK1 pathway is activated. However, the relation between miR-146b-5p and the Notch1/PDGFRβ/ROCK1 pathway in lung fibrosis remains unclear. To investigate the function of miR-146b-5p in lung fibrosis, an in vivo model of lung fibrosis was established in mice by bleomycin. The fibrosis in lung tissues of mice was observed by HE, Masson and Sirius Red staining. Lung pericytes were isolated and identified by fluorescence microscopy. Immunofluorescence staining and Western blot were used to investigate the expression of desmin, NG2, collagen I and α-SMA. CCK8 assay was used to assess the cell viability, and flow cytometry was performed to evaluate the cell cycle in pericytes. Furthermore, the correlation between miR-146b-5p and Notch1 was analysed by Spearman analysis. The mechanism by which miR-146b-5p affects pericytes and lung fibrosis via the Notch1/ PDGFRβ/ROCK1 pathway was explored by RT-qPCR, Western blot, immunofluorescence staining and dual luciferase reporter gene assay. In bleomycin-treated mice, miR-146b-5p was down-regulated, while Notch1 was up-regulated. Up-regulation of miR-146b-5p significantly inhibited the viability and induced G1 phase arrest of lung pericytes. MiR-146b-5p mimics up-regulated miR-146b-5p, desmin, and NG2 and down-regulated α-SMA and collagen I in the lung pericytes. Additionally, miR-146b-5p was negatively correlated with Notch1, and miR-146b-5p interacted with Notch1. Over-expression of miR-146b-5p inactivated the Notch1/PDGFRβ/ROCK1 pathway. Our results indicate that up-regulation of miR-146b-5p inhibits fibrosis in lung pericytes via modulation of the Notch1/PDGFRβ/ROCK1 pathway. Thus, our study might provide a novel target against lung fibrosis.
Collapse
Affiliation(s)
- W Shuai
- Departments of Critical Care Medicine, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha 410005, Hunan, China
| | - Q Chen
- Department of Nephrology, Shanghai General Hospital, Shanghai 200000, China
| | - X Zhou
- Departments of Critical Care Medicine, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha 410005, Hunan, China
| |
Collapse
|
11
|
Phon BWS, Kamarudin MNA, Bhuvanendran S, Radhakrishnan AK. Transitioning pre-clinical glioblastoma models to clinical settings with biomarkers identified in 3D cell-based models: A systematic scoping review. Biomed Pharmacother 2022; 145:112396. [PMID: 34775238 DOI: 10.1016/j.biopha.2021.112396] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 10/25/2021] [Accepted: 11/02/2021] [Indexed: 11/02/2022] Open
Abstract
Glioblastoma (GBM) remains incurable despite the overwhelming discovery of 2-dimensional (2D) cell-based potential therapeutics since the majority of them have met unsatisfactory results in animal and clinical settings. Incremental empirical evidence has laid the widespread need of transitioning 2D to 3-dimensional (3D) cultures that better mimic GBM's complex and heterogenic nature to allow better translation of pre-clinical results. This systematic scoping review analyses the transcriptomic data involving 3D models of GBM against 2D models from 22 studies identified from four databases (PubMed, ScienceDirect, Medline, and Embase). From a total of 499 genes reported in these studies, 313 (63%) genes were upregulated across 3D models cultured using different scaffolds. Our analysis showed that 4 of the replicable upregulated genes are associated with GBM stemness, epithelial to mesenchymal transition (EMT), hypoxia, and migration-related genes regardless of the type of scaffolds, displaying close resemblances to primitive undifferentiated tumour phenotypes that are associated with decreased overall survival and increased hazard ratio in GBM patients. The upregulation of drug response and drug efflux genes (e.g. cytochrome P450s and ABC transporters) mirrors the GBM genetic landscape that contributes to in vivo and clinical treatment resistance. These upregulated genes displayed strong protein-protein interactions when analysed using an online bioinformatics software (STRING). These findings reinforce the need for widespread transition to 3D GBM models as a relatively inexpensive humanised pre-clinical tool with suitable genetic biomarkers to bridge clinical gaps in potential therapeutic evaluations.
Collapse
Affiliation(s)
- Brandon Wee Siang Phon
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, 47500 Bandar Sunway, Selangor, Malaysia
| | - Muhamad N A Kamarudin
- Brain Research Institute Monash Sunway, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500 Bandar Sunway, Selangor, Malaysia.
| | - Saatheeyavaane Bhuvanendran
- Brain Research Institute Monash Sunway, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500 Bandar Sunway, Selangor, Malaysia
| | - Ammu K Radhakrishnan
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, 47500 Bandar Sunway, Selangor, Malaysia
| |
Collapse
|
12
|
Díaz-Flores L, Gutiérrez R, González-Gómez M, García MDP, Díaz-Flores L, González-Marrero I, Ávila J, Martín-Vasallo P. Disproportion in Pericyte/Endothelial Cell Proliferation and Mechanisms of Intussusceptive Angiogenesis Participate in Bizarre Vessel Formation in Glioblastoma. Cells 2021; 10:cells10102625. [PMID: 34685606 PMCID: PMC8534221 DOI: 10.3390/cells10102625] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 09/24/2021] [Accepted: 09/28/2021] [Indexed: 12/14/2022] Open
Abstract
Glioblastoma (GBM) is the most malignant tumor in the brain. In addition to the vascular pattern with thin-walled vessels and findings of sprouting angiogenesis, GBM presents a bizarre microvasculature (BM) formed by vascular clusters, vascular garlands, and glomeruloid bodies. The mechanisms in BM morphogenesis are not well known. Our objective was to assess the role of pericyte/endothelial proliferation and intussusceptive angiogenic mechanisms in the formation of the BM. For this purpose, we studied specimens of 66 GBM cases using immunochemistry and confocal microscopy. In the BM, the results showed (a) transitional forms between the BM patterns, mostly with prominent pericytes covering all the abluminal endothelial cell (EC) surface of the vessels, (b) a proliferation index high in the prominent pericytes and low in ECs (47.85 times higher in pericytes than in ECs), (c) intravascular pillars (hallmark of intussusceptive angiogenesis) formed by transcapillary interendothelial bridges, endothelial contacts of opposite vessel walls, and vessel loops, and (d) the persistence of these findings in complex glomeruloid bodies. In conclusion, disproportion in pericyte/EC proliferation and mechanisms of intussusceptive angiogenesis participate in BM formation. The contributions have morphogenic and clinical interest since pericytes and intussusceptive angiogenesis can condition antiangiogenic therapy in GBM.
Collapse
Affiliation(s)
- Lucio Díaz-Flores
- Department of Basic Medical Sciences, Faculty of Medicine, University of La Laguna, 38071 Tenerife, Spain; (L.D.-F.); (R.G.); (M.G.-G.); (L.D.-F.J.); (I.G.-M.)
| | - Ricardo Gutiérrez
- Department of Basic Medical Sciences, Faculty of Medicine, University of La Laguna, 38071 Tenerife, Spain; (L.D.-F.); (R.G.); (M.G.-G.); (L.D.-F.J.); (I.G.-M.)
| | - Miriam González-Gómez
- Department of Basic Medical Sciences, Faculty of Medicine, University of La Laguna, 38071 Tenerife, Spain; (L.D.-F.); (R.G.); (M.G.-G.); (L.D.-F.J.); (I.G.-M.)
- Instituto de Tecnologías Biomédicas de Canarias, University of La Laguna, 38071 Tenerife, Spain;
| | - María-del-Pino García
- Department of Pathology, Eurofins Megalab–Hospiten Hospitals, 38100 Tenerife, Spain;
| | - Lucio Díaz-Flores
- Department of Basic Medical Sciences, Faculty of Medicine, University of La Laguna, 38071 Tenerife, Spain; (L.D.-F.); (R.G.); (M.G.-G.); (L.D.-F.J.); (I.G.-M.)
| | - Ibrahim González-Marrero
- Department of Basic Medical Sciences, Faculty of Medicine, University of La Laguna, 38071 Tenerife, Spain; (L.D.-F.); (R.G.); (M.G.-G.); (L.D.-F.J.); (I.G.-M.)
| | - Julio Ávila
- Instituto de Tecnologías Biomédicas de Canarias, University of La Laguna, 38071 Tenerife, Spain;
- Department of Bioquímica, Microbiología, Biología Celular y Genética, University of La Laguna, 38206 Tenerife, Spain
| | - Pablo Martín-Vasallo
- Instituto de Tecnologías Biomédicas de Canarias, University of La Laguna, 38071 Tenerife, Spain;
- Department of Bioquímica, Microbiología, Biología Celular y Genética, University of La Laguna, 38206 Tenerife, Spain
- Correspondence: ; Tel.: +34-922-318358; Fax: +34-922-319279
| |
Collapse
|
13
|
Comba A, Faisal SM, Varela ML, Hollon T, Al-Holou WN, Umemura Y, Nunez FJ, Motsch S, Castro MG, Lowenstein PR. Uncovering Spatiotemporal Heterogeneity of High-Grade Gliomas: From Disease Biology to Therapeutic Implications. Front Oncol 2021; 11:703764. [PMID: 34422657 PMCID: PMC8377724 DOI: 10.3389/fonc.2021.703764] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 07/19/2021] [Indexed: 12/13/2022] Open
Abstract
Glioblastomas (GBM) are the most common and aggressive tumors of the central nervous system. Rapid tumor growth and diffuse infiltration into healthy brain tissue, along with high intratumoral heterogeneity, challenge therapeutic efficacy and prognosis. A better understanding of spatiotemporal tumor heterogeneity at the histological, cellular, molecular, and dynamic levels would accelerate the development of novel treatments for this devastating brain cancer. Histologically, GBM is characterized by nuclear atypia, cellular pleomorphism, necrosis, microvascular proliferation, and pseudopalisades. At the cellular level, the glioma microenvironment comprises a heterogeneous landscape of cell populations, including tumor cells, non-transformed/reactive glial and neural cells, immune cells, mesenchymal cells, and stem cells, which support tumor growth and invasion through complex network crosstalk. Genomic and transcriptomic analyses of gliomas have revealed significant inter and intratumoral heterogeneity and insights into their molecular pathogenesis. Moreover, recent evidence suggests that diverse dynamics of collective motion patterns exist in glioma tumors, which correlate with histological features. We hypothesize that glioma heterogeneity is not stochastic, but rather arises from organized and dynamic attributes, which favor glioma malignancy and influences treatment regimens. This review highlights the importance of an integrative approach of glioma histopathological features, single-cell and spatially resolved transcriptomic and cellular dynamics to understand tumor heterogeneity and maximize therapeutic effects.
Collapse
Affiliation(s)
- Andrea Comba
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States.,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States.,Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Syed M Faisal
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States.,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States.,Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Maria Luisa Varela
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States.,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States.,Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Todd Hollon
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Wajd N Al-Holou
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Yoshie Umemura
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Felipe J Nunez
- Laboratory of Molecular and Cellular Therapy, Fundación Instituto Leloir, Buenos Aires, Argentina
| | - Sebastien Motsch
- School of Mathematical and Statistical Sciences, Arizona State University, Tempe, AZ, United States
| | - Maria G Castro
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States.,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States.,Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Pedro R Lowenstein
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States.,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States.,Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, United States
| |
Collapse
|
14
|
Li L, Chen J, Ming Y, Li B, Fu R, Duan D, Li Z, Ni R, Wang X, Zhou Y, Zhang L. The Application of Peptides in Glioma: a Novel Tool for Therapy. Curr Pharm Biotechnol 2021; 23:620-633. [PMID: 34182908 DOI: 10.2174/1389201022666210628114042] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 04/25/2021] [Accepted: 04/26/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Glioma is the most aggressive and lethal tumor of the central nervous system. Owing to the cellular heterogeneity, the invasiveness, and blood-brain barrier (BBB), current therapeutic approaches, such as chemotherapy and radiotherapy, are poorly to obtain great anti-tumor efficacy. However, peptides, a novel type of therapeutic agent, displayed excellent ability in the tumor, which becomes a new molecule for glioma treatment. METHOD We review the current knowledge on peptides for the treatment of glioma through a PubMed-based literature search. RESULTS In the treatment of glioma, peptides can be used as (i) decoration on the surface of the delivery system, facilitating the distribution and accumulation of the anti-tumor drug in the target site;(ii) anti-tumor active molecules, inhibiting the growth of glioma and reducing solid tumor volume; (iii) immune-stimulating factor, and activating immune cells in the tumor microenvironment or recruiting immune cells to the tumor for breaking out the immunosuppression by glioma cells. CONCLUSION The application of peptides has revolutionized the treatment of glioma, which is based on targeting, penetrating, anti-tumor activities, and immunostimulatory. Moreover, better outcomes have been discovered in combining different kinds of peptides rather than a single one. Until now, more and more preclinical studies have been developed with multifarious peptides, which show promising results in vitro or vivo with the model of glioma.
Collapse
Affiliation(s)
- Li Li
- Department of Pharmacy, Daping Hospital, Army Medical University, Chongqing, China
| | - Jianhong Chen
- Department of Pharmacy, Daping Hospital, Army Medical University, Chongqing, China
| | - Yue Ming
- Department of Pharmacy, Daping Hospital, Army Medical University, Chongqing, China
| | - Bin Li
- Department of Pharmacy, Daping Hospital, Army Medical University, Chongqing, China
| | - Ruoqiu Fu
- Department of Pharmacy, Daping Hospital, Army Medical University, Chongqing, China
| | - Dongyu Duan
- Department of Pharmacy, Daping Hospital, Army Medical University, Chongqing, China
| | - Ziwei Li
- Department of Pharmacy, Daping Hospital, Army Medical University, Chongqing, China
| | - Rui Ni
- Department of Pharmacy, Daping Hospital, Army Medical University, Chongqing, China
| | - Xianfeng Wang
- Department of Pharmacy, Daping Hospital, Army Medical University, Chongqing, China
| | - Yueling Zhou
- Department of Pharmacy, Daping Hospital, Army Medical University, Chongqing, China
| | - Lin Zhang
- Department of Pharmacy, Daping Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
15
|
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: 26] [Impact Index Per Article: 6.5] [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
|
16
|
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
|
17
|
Lucero R, Zappulli V, Sammarco A, Murillo OD, Cheah PS, Srinivasan S, Tai E, Ting DT, Wei Z, Roth ME, Laurent LC, Krichevsky AM, Breakefield XO, Milosavljevic A. Glioma-Derived miRNA-Containing Extracellular Vesicles Induce Angiogenesis by Reprogramming Brain Endothelial Cells. Cell Rep 2021; 30:2065-2074.e4. [PMID: 32075753 DOI: 10.1016/j.celrep.2020.01.073] [Citation(s) in RCA: 109] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 09/29/2019] [Accepted: 01/22/2020] [Indexed: 12/13/2022] Open
Abstract
Glioblastoma (GBM) is characterized by aberrant vascularization and a complex tumor microenvironment. The failure of anti-angiogenic therapies suggests pathways of GBM neovascularization, possibly attributable to glioblastoma stem cells (GSCs) and their interplay with the tumor microenvironment. It has been established that GSC-derived extracellular vesicles (GSC-EVs) and their cargoes are proangiogenic in vitro. To further elucidate EV-mediated mechanisms of neovascularization in vitro, we perform RNA-seq and DNA methylation profiling of human brain endothelial cells exposed to GSC-EVs. To correlate these results to tumors in vivo, we perform histoepigenetic analysis of GBM molecular profiles in the TCGA collection. Remarkably, GSC-EVs and normal vascular growth factors stimulate highly distinct gene regulatory responses that converge on angiogenesis. The response to GSC-EVs shows a footprint of post-transcriptional gene silencing by EV-derived miRNAs. Our results provide insights into targetable angiogenesis pathways in GBM and miRNA candidates for liquid biopsy biomarkers.
Collapse
Affiliation(s)
- Rocco Lucero
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Valentina Zappulli
- Department of Comparative Biomedicine and Food Science, University of Padua, Padua, Italy; Departments of Neurology and Radiology, Massachusetts General Hospital, Boston, MA 02114, USA; Neuroscience Program, Harvard Medical School, Boston, MA 02115, USA.
| | - Alessandro Sammarco
- Department of Comparative Biomedicine and Food Science, University of Padua, Padua, Italy; Departments of Neurology and Radiology, Massachusetts General Hospital, Boston, MA 02114, USA; Neuroscience Program, Harvard Medical School, Boston, MA 02115, USA
| | - Oscar D Murillo
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Pike See Cheah
- Departments of Neurology and Radiology, Massachusetts General Hospital, Boston, MA 02114, USA; Neuroscience Program, Harvard Medical School, Boston, MA 02115, USA; Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Seri Kembangan, Selangor, Malaysia
| | - Srimeenakshi Srinivasan
- Department of Obstetrics, Gynecology, and Reproductive Sciences and Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, CA 92037, USA
| | - Eric Tai
- Massachusetts General Hospital Cancer Center, Boston, MA 02114, USA; Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - David T Ting
- Massachusetts General Hospital Cancer Center, Boston, MA 02114, USA; Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Zhiyun Wei
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Matthew E Roth
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Louise C Laurent
- Department of Obstetrics, Gynecology, and Reproductive Sciences and Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, CA 92037, USA
| | - Anna M Krichevsky
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Xandra O Breakefield
- Departments of Neurology and Radiology, Massachusetts General Hospital, Boston, MA 02114, USA; Neuroscience Program, Harvard Medical School, Boston, MA 02115, USA
| | | |
Collapse
|
18
|
Su X, Vasilkovska T, Fröhlich N, Garaschuk O. Characterization of cell type-specific S100B expression in the mouse olfactory bulb. Cell Calcium 2021; 94:102334. [PMID: 33460952 DOI: 10.1016/j.ceca.2020.102334] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 12/11/2020] [Accepted: 12/11/2020] [Indexed: 12/17/2022]
Abstract
S100B is an EF-hand type Ca2+-binding protein of the S100 family, known to support neurogenesis and to promote the interactions between brain's nervous and immune systems. Here, we characterized the expression of S100B in the mouse olfactory bulb, a neurogenic niche comprising mature and adult-born neurons, astrocytes, oligodendrocytes and microglia. Besides astrocytes, for which S100B is a classical marker, S100B was also expressed in NG2 cells and, surprisingly, in APC-positive myelinating oligodendrocytes but not in mature/adult-born neurons or microglia. Various layers of the bulb differed substantially in the composition of S100B-positive cells, with the highest fraction of the APC-positive oligodendrocytes found in the granule cell layer. Across all layers, ∼50 % of NG2 cells were S100B-negative. Finally, our data revealed a strong correlation between the fraction of myelinating oligodendrocytes among the S100B-positive cells and the oligodendrocyte density in different brain areas, underscoring the importance of S100B for the establishment and maintenance of myelin sheaths.
Collapse
Affiliation(s)
- Xin Su
- Institute of Physiology, Department of Neurophysiology, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Tamara Vasilkovska
- Institute of Physiology, Department of Neurophysiology, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Nicole Fröhlich
- Institute of Physiology, Department of Neurophysiology, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Olga Garaschuk
- Institute of Physiology, Department of Neurophysiology, Eberhard Karls University of Tübingen, Tübingen, Germany.
| |
Collapse
|
19
|
Cole AP, Hoffmeyer E, Chetty SL, Cruz-Cruz J, Hamrick F, Youssef O, Cheshier S, Mitra SS. Microglia in the Brain Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1273:197-208. [PMID: 33119883 DOI: 10.1007/978-3-030-49270-0_11] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Microglia are the brain resident phagocytes that act as the primary form of the immune defense in the central nervous system. These cells originate from primitive macrophages that arise from the yolk sac. Advances in imaging and single-cell RNA-seq technologies provided new insights into the complexity of microglia biology.Microglia play an essential role in the brain development and maintenance of brain homeostasis. They are also crucial in injury repair in the central nervous system. The tumor microenvironment is complex and includes neoplastic cells as well as varieties of host and infiltrating immune cells. Microglia are part of the glioma microenvironment and play a critical part in initiating and maintaining tumor growth and spread. Microglia can also act as effector cells in treatments against gliomas. In this chapter, we summarize the current knowledge of how and where microglia are generated. We also discuss their functions during brain development, injury repair, and homeostasis. Moreover, we discuss the role of microglia in the tumor microenvironment of gliomas and highlight their therapeutic implications.
Collapse
Affiliation(s)
- Allison P Cole
- Department of Pediatrics, Division of Hematology/Oncology/Bone Marrow Transplant, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Eric Hoffmeyer
- Department of Pediatrics, Division of Hematology/Oncology/Bone Marrow Transplant, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Senthilnath Lakshmana Chetty
- Department of Pediatrics, Division of Hematology/Oncology/Bone Marrow Transplant, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Joselyn Cruz-Cruz
- Department of Pediatrics, Division of Hematology/Oncology/Bone Marrow Transplant, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Forrest Hamrick
- Department of Neurosurgery, Division of Pediatric Neurosurgery, Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Osama Youssef
- Department of Neurosurgery, Division of Pediatric Neurosurgery, Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Samuel Cheshier
- Department of Neurosurgery, Division of Pediatric Neurosurgery, Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT, USA.
| | - Siddhartha S Mitra
- Department of Pediatrics, Division of Hematology/Oncology/Bone Marrow Transplant, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
20
|
Li M, Li G, Kiyokawa J, Tirmizi Z, Richardson LG, Ning J, Das S, Martuza RL, Stemmer-Rachamimov A, Rabkin SD, Wakimoto H. Characterization and oncolytic virus targeting of FAP-expressing tumor-associated pericytes in glioblastoma. Acta Neuropathol Commun 2020; 8:221. [PMID: 33308315 PMCID: PMC7730751 DOI: 10.1186/s40478-020-01096-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 11/26/2020] [Indexed: 12/16/2022] Open
Abstract
Cancer-associated fibroblasts (CAFs) are activated fibroblasts constituting the major stromal components in many types of cancer. CAFs contribute to hallmarks of cancer such as proliferation, invasion and immunosuppressive tumor microenvironment, and are associated with poor prognosis of patients with cancer. However, in glioblastoma (GBM), the most common and aggressive primary malignant brain tumor, our knowledge about CAFs or CAF-like stromal cells is limited. Here, using commonly accepted CAF markers, we characterized CAF-like cell populations in clinical glioma specimens and datasets along with mouse models of GBM. We found that tumor-associated pericytes marked by co-expression of fibroblast activation protein α (FAP) and PDGFRβ represent major stromal cell subsets in both human GBM and mouse GBM models, while a fraction of mesenchymal neoplastic cells also express FAP in patient tumors. Since oncolytic viruses can kill cancer cells and simultaneously modulate the tumor microenvironment by impacting non-neoplastic populations such as immune cells and tumor vasculature, we further investigated the ability of oncolytic viruses to target GBM-associated stromal cells. An oncolytic adenovirus, ICOVIR15, carrying ∆24-E1A and an RGD-fiber, infects and depletes FAP+ pericytes as well as GBM cells in murine GBM. Our study thus identifies FAP+/PDGFRβ+ pericytes as a major CAF-like stromal cell population in GBM, and highlights the unique property of this oncolytic adenovirus to target both GBM cells and GBM-associated stromal FAP+ cells.
Collapse
|
21
|
Valtorta S, Salvatore D, Rainone P, Belloli S, Bertoli G, Moresco RM. Molecular and Cellular Complexity of Glioma. Focus on Tumour Microenvironment and the Use of Molecular and Imaging Biomarkers to Overcome Treatment Resistance. Int J Mol Sci 2020; 21:E5631. [PMID: 32781585 PMCID: PMC7460665 DOI: 10.3390/ijms21165631] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 07/31/2020] [Accepted: 08/03/2020] [Indexed: 02/08/2023] Open
Abstract
This review highlights the importance and the complexity of tumour biology and microenvironment in the progression and therapy resistance of glioma. Specific gene mutations, the possible functions of several non-coding microRNAs and the intra-tumour and inter-tumour heterogeneity of cell types contribute to limit the efficacy of the actual therapeutic options. In this scenario, identification of molecular biomarkers of response and the use of multimodal in vivo imaging and in particular the Positron Emission Tomography (PET) based molecular approach, can help identifying glioma features and the modifications occurring during therapy at a regional level. Indeed, a better understanding of tumor heterogeneity and the development of diagnostic procedures can favor the identification of a cluster of patients for personalized medicine in order to improve the survival and their quality of life.
Collapse
Affiliation(s)
- Silvia Valtorta
- Department of Medicine and Surgery and Tecnomed Foundation, University of Milano—Bicocca, 20900 Monza, Italy; (S.V.); (D.S.); (P.R.)
- Nuclear Medicine Department, San Raffaele Scientific Institute (IRCCS), 20132 Milan, Italy;
| | - Daniela Salvatore
- Department of Medicine and Surgery and Tecnomed Foundation, University of Milano—Bicocca, 20900 Monza, Italy; (S.V.); (D.S.); (P.R.)
- Nuclear Medicine Department, San Raffaele Scientific Institute (IRCCS), 20132 Milan, Italy;
| | - Paolo Rainone
- Department of Medicine and Surgery and Tecnomed Foundation, University of Milano—Bicocca, 20900 Monza, Italy; (S.V.); (D.S.); (P.R.)
- Nuclear Medicine Department, San Raffaele Scientific Institute (IRCCS), 20132 Milan, Italy;
| | - Sara Belloli
- Nuclear Medicine Department, San Raffaele Scientific Institute (IRCCS), 20132 Milan, Italy;
- Institute of Molecular Bioimaging and Physiology (IBFM), CNR, 20090 Segrate, Italy
| | - Gloria Bertoli
- Institute of Molecular Bioimaging and Physiology (IBFM), CNR, 20090 Segrate, Italy
| | - Rosa Maria Moresco
- Department of Medicine and Surgery and Tecnomed Foundation, University of Milano—Bicocca, 20900 Monza, Italy; (S.V.); (D.S.); (P.R.)
- Nuclear Medicine Department, San Raffaele Scientific Institute (IRCCS), 20132 Milan, Italy;
- Institute of Molecular Bioimaging and Physiology (IBFM), CNR, 20090 Segrate, Italy
| |
Collapse
|
22
|
Téglási V, Csűry DT, Dezső K, Bugyik E, Szabó V, Szállási Z, Paku S, Reiniger L. Origin and Distribution of Connective Tissue and Pericytes Impacting Vascularization in Brain Metastases With Different Growth Patterns. J Neuropathol Exp Neurol 2020; 78:326-339. [PMID: 30816955 DOI: 10.1093/jnen/nlz007] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The impact of growth pattern on the distribution of connective tissue and on the vascularization of brain metastases (40 colon, lung and breast carcinoma samples) was analyzed. Most of the cases showed either a "pushing-type" (18/40, mostly colon and lung carcinomas) or a "papillary-type" (19/40, mostly breast carcinomas) growth pattern. There was a striking difference in the growth pattern and vascularization of colon/lung versus breast carcinoma metastases. Pushing-type brain metastases incorporated fewer vessels and accumulated more collagen in the adjacent brain parenchyma, whereas papillary-type brain metastases incorporated more vessels and accumulated collagen in the center of the tumor. We observed duplication of the PDGFRβ-positive pericyte layer accompanied by an increase in the amount of collagen within the vessel walls. The outer layer of pericytes and the collagen was removed from the vessel by invasive activity of the tumors, which occurred either peri- or intratumorally, depending on the growth pattern of the metastasis. Our findings suggest that pericytes are the main source of the connective tissue in brain metastases. Vascularization and connective tissue accumulation of the brain metastases largely depend on the growth pattern of the tumors.
Collapse
Affiliation(s)
- Vanda Téglási
- 1st Department of Pathology and Experimental Cancer Research
| | - Dániel T Csűry
- 1st Department of Pathology and Experimental Cancer Research
| | - Katalin Dezső
- 1st Department of Pathology and Experimental Cancer Research
| | - Edina Bugyik
- 1st Department of Pathology and Experimental Cancer Research
| | - Vanessza Szabó
- 1st Department of Pathology and Experimental Cancer Research
| | - Zoltán Szállási
- Brain Metastasis Research Group, Hungarian Academy of Sciences, 2nd Department of Pathology, Semmelweis University, Budapest, Hungary.,Computational Health Informatics Program, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts.,Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Sándor Paku
- 1st Department of Pathology and Experimental Cancer Research
| | - Lilla Reiniger
- 1st Department of Pathology and Experimental Cancer Research.,Brain Metastasis Research Group, Hungarian Academy of Sciences, 2nd Department of Pathology, Semmelweis University, Budapest, Hungary
| |
Collapse
|
23
|
Cheng M, Zhang ZW, Ji XH, Xu Y, Bian E, Zhao B. Super-enhancers: A new frontier for glioma treatment. Biochim Biophys Acta Rev Cancer 2020; 1873:188353. [PMID: 32112817 DOI: 10.1016/j.bbcan.2020.188353] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 02/21/2020] [Accepted: 02/21/2020] [Indexed: 01/17/2023]
Abstract
Glioma is the most common primary malignant tumor in the human brain. Although there are a variety of treatments, such as surgery, radiation and chemotherapy, glioma is still an incurable disease. Super-enhancers (SEs) are implicated in the control of tumor cell identity, and they promote oncogenic transcription, which supports tumor cells. Inhibition of the SE complex, which is required for the assembly and maintenance of SEs, may repress oncogenic transcription and impede tumor growth. In this review, we discuss the unique characteristics of SEs compared to typical enhancers, and we summarize the recent advances in the understanding of their properties and biological role in gene regulation. Additionally, we highlight that SE-driven lncRNAs, miRNAs and genes are involved in the malignant phenotype of glioma. Most importantly, the application of SE inhibitors in different cancer subtypes has introduced new directions in glioma treatment.
Collapse
Affiliation(s)
- Meng Cheng
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; Cerebral Vascular Disease Research Center, Anhui Medical University, Hefei 230601, China
| | - Zheng Wei Zhang
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; Cerebral Vascular Disease Research Center, Anhui Medical University, Hefei 230601, China
| | - Xing Hu Ji
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; Cerebral Vascular Disease Research Center, Anhui Medical University, Hefei 230601, China
| | - Yadi Xu
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; Cerebral Vascular Disease Research Center, Anhui Medical University, Hefei 230601, China
| | - Erbao Bian
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; Cerebral Vascular Disease Research Center, Anhui Medical University, Hefei 230601, China.
| | - Bing Zhao
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; Cerebral Vascular Disease Research Center, Anhui Medical University, Hefei 230601, China.
| |
Collapse
|
24
|
Mitrofanova L, Hazratov A, Galkovsky B, Gorshkov A, Bobkov D, Gulyaev D, Shlyakhto E. Morphological and immunophenotypic characterization of perivascular interstitial cells in human glioma: Telocytes, pericytes, and mixed immunophenotypes. Oncotarget 2020; 11:322-346. [PMID: 32064038 PMCID: PMC6996916 DOI: 10.18632/oncotarget.27340] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 10/26/2019] [Indexed: 12/12/2022] Open
Abstract
Telocytes (Tcs) and pericytes (Pcs) are two types of perivascular interstitial cell known to be widespread in various organs and tissues, including the brain. We postulated that Tcs and Pcs may be involved in glioblastoma (GBM) neovascularization.
Collapse
Affiliation(s)
- Lubov Mitrofanova
- Almazov National Medical Research Centre, Pathomorphology Research Laboratory, St. Petersburg, Russia
| | - Anton Hazratov
- Almazov National Medical Research Centre, Pathomorphology Research Laboratory, St. Petersburg, Russia
| | - Boris Galkovsky
- Almazov National Medical Research Centre, Pathomorphology Research Laboratory, St. Petersburg, Russia
| | - Andrey Gorshkov
- Almazov National Medical Research Centre, Pathomorphology Research Laboratory, St. Petersburg, Russia.,Smorodintsev Research Institute of Influenza, Laboratory of Intracellular Signaling and Transport, St. Petersburg, Russia
| | - Danila Bobkov
- Smorodintsev Research Institute of Influenza, Laboratory of Intracellular Signaling and Transport, St. Petersburg, Russia.,Institute of Cytology of the Russian Academy of Science, Laboratory of Cell Biology in Culture, St. Petersburg, Russia
| | - Dmitry Gulyaev
- Almazov National Medical Research Centre, Research Department of Neurosurgery, St. Petersburg, Russia
| | - Evgeny Shlyakhto
- Almazov National Medical Research Centre, General Director, St. Petersburg, Russia
| |
Collapse
|
25
|
Behnan J, Finocchiaro G, Hanna G. The landscape of the mesenchymal signature in brain tumours. Brain 2019; 142:847-866. [PMID: 30946477 PMCID: PMC6485274 DOI: 10.1093/brain/awz044] [Citation(s) in RCA: 235] [Impact Index Per Article: 39.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 01/07/2019] [Accepted: 01/09/2019] [Indexed: 12/12/2022] Open
Abstract
The complexity of glioblastoma multiforme, the most common and lethal variant of gliomas, is reflected by cellular and molecular heterogeneity at both the inter- and intra-tumoural levels. Molecular subtyping has arisen in the past two decades as a promising strategy to give better predictions of glioblastoma multiforme evolution, common disease pathways, and rational treatment options. The Cancer Genome Atlas network initially identified four molecular subtypes of glioblastoma multiforme: proneural, neural, mesenchymal and classical. However, further studies, also investigated glioma stem cells, have only identified two to three subtypes: proneural, mesenchymal and classical. The proneural-mesenchymal transition upon tumour recurrence has been suggested as a mechanism of tumour resistance to radiation and chemotherapy treatment. Glioblastoma multiforme patients with the mesenchymal subtype tend to survive shorter than other subtypes when analysis is restricted to samples with low transcriptional heterogeneity. Although the mesenchymal signature in malignant glioma may seem at odds with the common idea of the ectodermal origin of neural-glial lineages, the presence of the mesenchymal signature in glioma is supported by several studies suggesting that it can result from: (i) intrinsic expression of tumour cells affected with accumulated genetic mutations and cell of origin; (ii) tumour micro-environments with recruited macrophages or microglia, mesenchymal stem cells or pericytes, and other progenitors; (iii) resistance to tumour treatment, including radiotherapy, antiangiogenic therapy and possibly chemotherapy. Genetic abnormalities, mainly NF1 mutations, together with NF-κB transcriptional programs, are the main driver of acquiring mesenchymal-signature. This signature is far from being simply tissue artefacts, as it has been identified in single cell glioma, circulating tumour cells, and glioma stem cells that are released from the tumour micro-environment. All these together suggest that the mesenchymal signature in glioblastoma multiforme is induced and sustained via cell intrinsic mechanisms and tumour micro-environment factors. Although patients with the mesenchymal subtype tend to have poorer prognosis, they may have favourable response to immunotherapy and intensive radio- and chemotherapy.
Collapse
Affiliation(s)
- Jinan Behnan
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden.,Duke Preclinical Translational Unit, Duke University Medical Center, Durham, North Carolina
| | - Gaetano Finocchiaro
- Unit of Molecular Neuro-Oncology, Neurological Institute C. Besta, Milan, Italy
| | - Gabi Hanna
- Duke Preclinical Translational Unit, Duke University Medical Center, Durham, North Carolina
| |
Collapse
|
26
|
Su X, Huang L, Qu Y, Xiao D, Mu D. Pericytes in Cerebrovascular Diseases: An Emerging Therapeutic Target. Front Cell Neurosci 2019; 13:519. [PMID: 31824267 PMCID: PMC6882740 DOI: 10.3389/fncel.2019.00519] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 11/05/2019] [Indexed: 12/14/2022] Open
Abstract
Pericytes are functional components of the neurovascular unit (NVU) that are located around the blood vessels, and their roles in the regulation of cerebral health and diseases has been reported. Currently, the potential properties of pericytes as emerging therapeutic targets for cerebrovascular diseases have attracted considerable attention. Nonetheless, few reviews have comprehensively discussed pericytes and their roles in cerebrovascular diseases. Therefore, in this review, we not only summarized and described the basic characteristics of pericytes but also focused on clarifying the new understanding about the roles of pericytes in the pathogenesis of cerebrovascular diseases, including white matter injury (WMI), hypoxic-ischemic brain damage, depression, neovascular insufficiency disease, and Alzheimer's disease (AD). Furthermore, we summarized the current therapeutic strategies targeting pericytes for cerebrovascular diseases. Collectively, this review is aimed at providing a comprehensive understanding of pericytes and new insights about the use of pericytes as novel therapeutic targets for cerebrovascular diseases.
Collapse
Affiliation(s)
- Xiaojuan Su
- Department of Paediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| | - Lingyi Huang
- West China College of Stomatology, Sichuan University, Chengdu, China
| | - Yi Qu
- Department of Paediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| | - Dongqiong Xiao
- Department of Paediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| | - Dezhi Mu
- Department of Paediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| |
Collapse
|
27
|
Ghosh MK, Chakraborty D, Sarkar S, Bhowmik A, Basu M. The interrelationship between cerebral ischemic stroke and glioma: a comprehensive study of recent reports. Signal Transduct Target Ther 2019; 4:42. [PMID: 31637020 PMCID: PMC6799849 DOI: 10.1038/s41392-019-0075-4] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 08/26/2019] [Accepted: 08/29/2019] [Indexed: 12/16/2022] Open
Abstract
Glioma and cerebral ischemic stroke are two major events that lead to patient death worldwide. Although these conditions have different physiological incidences, ~10% of ischemic stroke patients develop cerebral cancer, especially glioma, in the postischemic stages. Additionally, the high proliferation, venous thrombosis and hypercoagulability of the glioma mass increase the significant risk of thromboembolism, including ischemic stroke. Surprisingly, these events share several common pathways, viz. hypoxia, cerebral inflammation, angiogenesis, etc., but the proper mechanism behind this co-occurrence has yet to be discovered. The hypercoagulability and presence of the D-dimer level in stroke are different in cancer patients than in the noncancerous population. Other factors such as atherosclerosis and coagulopathy involved in the pathogenesis of stroke are partially responsible for cancer, and the reverse is also partially true. Based on clinical and neurosurgical experience, the neuronal structures and functions in the brain and spine are observed to change after a progressive attack of ischemia that leads to hypoxia and atrophy. The major population of cancer cells cannot survive in an adverse ischemic environment that excludes cancer stem cells (CSCs). Cancer cells in stroke patients have already metastasized, but early-stage cancer patients also suffer stroke for multiple reasons. Therefore, stroke is an early manifestation of cancer. Stroke and cancer share many factors that result in an increased risk of stroke in cancer patients, and vice-versa. The intricate mechanisms for stroke with and without cancer are different. This review summarizes the current clinical reports, pathophysiology, probable causes of co-occurrence, prognoses, and treatment possibilities.
Collapse
Affiliation(s)
- Mrinal K. Ghosh
- Signal Transduction in Cancer and Stem Cells Laboratory, Division of Cancer Biology and Inflammatory Disorder, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology (CSIR-IICB), 4 Raja S.C. Mullick Road, Kolkata 700032 and CN-06, Sector-V, Salt Lake, Kolkata, 700091 India
| | - Dipankar Chakraborty
- Signal Transduction in Cancer and Stem Cells Laboratory, Division of Cancer Biology and Inflammatory Disorder, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology (CSIR-IICB), 4 Raja S.C. Mullick Road, Kolkata 700032 and CN-06, Sector-V, Salt Lake, Kolkata, 700091 India
| | - Sibani Sarkar
- Signal Transduction in Cancer and Stem Cells Laboratory, Division of Cancer Biology and Inflammatory Disorder, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology (CSIR-IICB), 4 Raja S.C. Mullick Road, Kolkata 700032 and CN-06, Sector-V, Salt Lake, Kolkata, 700091 India
| | - Arijit Bhowmik
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute, 37 S. P. Mukherjee Road, Kolkata, 700 026 India
| | - Malini Basu
- Department of Microbiology, Dhruba Chand Halder College, Dakshin Barasat, South 24, Paraganas, 743372 India
| |
Collapse
|
28
|
Özen I, Roth M, Barbariga M, Gaceb A, Deierborg T, Genové G, Paul G. Loss of Regulator of G-Protein Signaling 5 Leads to Neurovascular Protection in Stroke. Stroke 2019; 49:2182-2190. [PMID: 30354999 PMCID: PMC6116795 DOI: 10.1161/strokeaha.118.020124] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Background and Purpose- In ischemic stroke, breakdown of the blood-brain barrier (BBB) aggravates brain damage. Pericyte detachment contributes to BBB disruption and neurovascular dysfunction, but little is known about its regulation in stroke. Here, we investigated how loss of RGS5 (regulator of G protein signaling 5) in pericytes affects BBB breakdown in stroke and its consequences. Method- We used RGS5 knockout and control mice and applied a permanent middle cerebral occlusion model. We analyzed pericyte numbers, phenotype, and vessel morphology using immunohistochemistry and confocal microscopy. We investigated BBB breakdown by measuring endothelial coverage, tight junctions, and AQP4 (aquaporin 4) in addition to BBB permeability (fluorescent-conjugated dextran extravasation). Tissue hypoxia was assessed with pimonidazole hydrochloride and neuronal death quantified with the terminal deoxynucleotidyl transferase dUTP nick end labeling assay. Results- We demonstrate that loss of RGS5 increases pericyte numbers and their endothelial coverage, which is associated with higher capillary density and length, and significantly less BBB damage after stroke. Loss of RGS5 in pericytes results in reduced vascular leakage and preserved tight junctions and AQP4, decreased cerebral hypoxia, and partial neuronal protection in the infarct area. Conclusions- Our findings show that loss of RGS5 affects pericyte-related BBB preservation in stroke and identifies RGS5 as an important target for neurovascular protection.
Collapse
Affiliation(s)
- Ilknur Özen
- From the Translational Neurology Group, Department of Clinical Science (I.O., M.R., M.B., A.G., G.P.)
| | - Michaela Roth
- From the Translational Neurology Group, Department of Clinical Science (I.O., M.R., M.B., A.G., G.P.)
| | - Marco Barbariga
- From the Translational Neurology Group, Department of Clinical Science (I.O., M.R., M.B., A.G., G.P.)
| | - Abderahim Gaceb
- From the Translational Neurology Group, Department of Clinical Science (I.O., M.R., M.B., A.G., G.P.)
| | - Tomas Deierborg
- Experimental Neuroinflammation Laboratory, Department of Experimental Medical Sciences (T.D.)
| | - Guillem Genové
- Lund University, Sweden; Department of Medicine, Integrated Cardio Metabolic Center, Karolinska Institute, Huddinge, Sweden (G.G.)
| | - Gesine Paul
- From the Translational Neurology Group, Department of Clinical Science (I.O., M.R., M.B., A.G., G.P.).,Wallenberg Centre for Molecular Medicine (G.P.).,Department of Neurology, Scania University Hospital, Lund, Sweden (G.P.)
| |
Collapse
|
29
|
Isocitrate dehydrogenase1 mutation reduces the pericyte coverage of microvessels in astrocytic tumours. J Neurooncol 2019; 143:187-196. [PMID: 31004262 DOI: 10.1007/s11060-019-03156-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Accepted: 03/21/2019] [Indexed: 01/20/2023]
Abstract
INTRODUCTION Tumour-associated angiogenesis is associated with the malignancy and poor prognosis of glioma. Isocitrate dehydrogenase (IDH) mutations are present in the majority of lower-grade (WHO grade II and III) and secondary glioblastomas, but their roles in tumour angiogenesis remain unclear. METHODS Using magnetic resonance imaging (MRI), the cerebral blood flow (CBF) of IDH-mutated glioma was measured and compared with the IDH-wildtype glioma. The densities of microvessels in IDH-mutated and wildtype astrocytoma and glioblastoma were assessed by immunohistochemical (IHC) staining with CD34, and the pericytes were labelled with α-smooth muscle antigen (α-SMA), neural-glial antigen 2 (NG2) and PDGF receptor-β (PDGFR-β), respectively. Furthermore, glia-specific mutant IDH1 knock-in mice were generated to evaluate the roles of mutant IDH1 on brain vascular architectures. The transcriptions of the angiogenesis-related genes were assessed in TCGA datasets, including ANGPT1, PDGFB and VEGFA. The expressions of these genes were further determined by western blot in U87-MG cells expressing a mutant IDH1 or treated with 2-HG. RESULTS The MRI results indicated that CBF was reduced in the IDH-mutated gliomas. The IHC staining showed that the pericyte coverages of microvessels were significantly decreased, but the microvessel densities (MVDs) were only slightly decreased in IDH-mutated glioma. The mutant IDH1 knock-in also impeded the pericyte coverage of brain microvessels in mice. Moreover, the TCGA database showed the mRNA levels of angiogenesis factors, including ANGPT1, PDGFB and VEGFA, were downregulated, and their promoters were also highly hyper-methylated in IDH-mutated gliomas. In addition, both mutant IDH1 and D-2-HG could downregulate the expression of these genes in U87-MG cells. CONCLUSIONS Our results suggested that IDH mutations could reduce the pericyte coverage of microvessels in astrocytic tumours by inhibiting the expression of angiogenesis factors. As vascular pericytes play an essential role in maintaining functional blood vessels to support tumour growth, our findings imply a potential avenue of therapeutic strategy for IDH-mutated gliomas.
Collapse
|
30
|
Notch1 promotes the pericyte-myofibroblast transition in idiopathic pulmonary fibrosis through the PDGFR/ROCK1 signal pathway. Exp Mol Med 2019; 51:1-11. [PMID: 30902967 PMCID: PMC6430797 DOI: 10.1038/s12276-019-0228-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 09/02/2018] [Accepted: 09/19/2018] [Indexed: 11/17/2022] Open
Abstract
The goals of this study were to investigate the role of the Notch1/PDGFRβ/ROCK1 signaling pathway in the pathogenesis of pulmonary fibrosis and to explore the possibility of treating fibrosis by targeting Notch1. Lung tissues from patients with pulmonary fibrosis were examined for the expression of Notch1/PDGFRβ/ROCK1 using RT-qPCR, western blotting, and immunostaining. Cultured mouse lung pericytes were transfected with Notch1-overexpressed vectors or shRNA targeting PDGFRβ/ROCK1 to examine cell behaviors, including proliferation, cell cycle arrest, and differentiation toward myofibroblasts. Finally, a mouse pulmonary fibrosis model was prepared, and a Notch1 inhibitor was administered to observe tissue morphology and pericyte cell behaviors. Human pulmonary fibrotic tissues presented with overexpression of Notch1, PDGFRβ, and ROCK1, in addition to a prominent transition of pericytes into myofibroblasts. In cultured mouse lung pericytes, overexpression of Notch1 led to the accelerated proliferation and differentiation of cells, and it also increased the expression of the PDGFRβ and ROCK1 proteins. The knockdown of PDGFRβ/ROCK1 in pericytes remarkably suppressed pericyte proliferation and differentiation. As further substantiation, the administration of a Notch1 inhibitor in a mouse model of lung fibrosis inhibited the PDGFRβ/ROCK1 pathway, suppressed pericyte proliferation and differentiation, and alleviated the severity of fibrosis. Our results showed that the Notch1 signaling pathway was aberrantly activated in pulmonary fibrosis, and this pathway may facilitate disease progression via mediating pericyte proliferation and differentiation. The inhibition of the Notch1 pathway may provide one promising treatment strategy for pulmonary fibrosis. A cell membrane protein called Notch1, which binds to signaling molecules outside cells and then alters the activity of genes inside the cells, might be a promising target for drugs to treat the lung damage of pulmonary fibrosis. This condition, generally of unknown cause, involves thickening, stiffening and scarring of lung tissue. It can lead to serious breathing difficulties and eventually death, especially in people aged over 70. Hui Wang and colleagues at Central South University, Changsha, investigated the significance of the Notch1 signaling pathway by examining lung tissue from patients and manipulating the activity of the pathway in mouse cells. They conclude that Notch1 signaling is activated in pulmonary fibrosis. Drugs that could inhibit the pathway, for example by binding to the Notch1 protein, might open a promising new avenue toward treatment.
Collapse
|
31
|
Sattiraju A, Mintz A. Pericytes in Glioblastomas: Multifaceted Role Within Tumor Microenvironments and Potential for Therapeutic Interventions. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1147:65-91. [PMID: 31147872 DOI: 10.1007/978-3-030-16908-4_2] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Glioblastoma (GBM) is an aggressive and lethal disease that often results in a poor prognosis. Unlike most solid tumors, GBM is characterized by diffuse infiltrating margins, extensive angiogenesis, hypoxia, necrosis, and clonal heterogeneity. Recurrent disease is an unavoidable consequence for many patients as standard treatment options such as surgery, radiotherapy, and chemotherapy have proven to be insufficient in causing long-term survival benefits. Systemic delivery of promising drugs is hindered due to the blood-brain barrier and non-uniform perfusion within GBM tissue. In recent years, many investigations have highlighted the role of GBM stem cells (GSCs) and their microenvironment in the initiation and maintenance of tumor tissue. Preclinical and early clinical studies to target GSCs and microenvironmental components are currently underway. Of these strategies, immunotherapy using checkpoint inhibitors and redirected cytotoxic T cells have shown promising results in early investigations. But, GBM microenvironment is heterogenous and recent investigations have shown cell populations within this microenvironment to be plastic. These studies underline the importance of identifying the role of and targeting multiple cell populations within the GBM microenvironment which could have a synergistic effect when combined with novel therapies. Pericytes are multipotent perivascular cells that play a vital role within the GBM microenvironment by assisting in tumor initiation, survival, and progression. Due to their role in regulating the blood-brain barrier permeability, promoting angiogenesis, tumor growth, clearing extracellular matrix for infiltrating GBM cells and in helping GBM cells evade immune surveillance, pericytes could be ideal therapeutic targets for stymieing or exploiting their role within the GBM microenvironment. This chapter will introduce hallmarks of GBM and elaborate on the contributions of pericytes to these hallmarks by examining recent findings. In addition, the chapter also highlights the therapeutic value of targeting pericytes, while discussing conventional and novel GBM therapies and obstacles to their efficacy.
Collapse
Affiliation(s)
- Anirudh Sattiraju
- Department of Radiology, Columbia University Irving Medical Center, New York, NY, USA
| | - Akiva Mintz
- Department of Radiology, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
32
|
Cheng J, Korte N, Nortley R, Sethi H, Tang Y, Attwell D. Targeting pericytes for therapeutic approaches to neurological disorders. Acta Neuropathol 2018; 136:507-523. [PMID: 30097696 PMCID: PMC6132947 DOI: 10.1007/s00401-018-1893-0] [Citation(s) in RCA: 167] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 07/30/2018] [Accepted: 07/31/2018] [Indexed: 12/13/2022]
Abstract
Many central nervous system diseases currently lack effective treatment and are often associated with defects in microvascular function, including a failure to match the energy supplied by the blood to the energy used on neuronal computation, or a breakdown of the blood–brain barrier. Pericytes, an under-studied cell type located on capillaries, are of crucial importance in regulating diverse microvascular functions, such as angiogenesis, the blood–brain barrier, capillary blood flow and the movement of immune cells into the brain. They also form part of the “glial” scar isolating damaged parts of the CNS, and may have stem cell-like properties. Recent studies have suggested that pericytes play a crucial role in neurological diseases, and are thus a therapeutic target in disorders as diverse as stroke, traumatic brain injury, migraine, epilepsy, spinal cord injury, diabetes, Huntington’s disease, Alzheimer’s disease, diabetes, multiple sclerosis, glioma, radiation necrosis and amyotrophic lateral sclerosis. Here we report recent advances in our understanding of pericyte biology and discuss how pericytes could be targeted to develop novel therapeutic approaches to neurological disorders, by increasing blood flow, preserving blood–brain barrier function, regulating immune cell entry to the CNS, and modulating formation of blood vessels in, and the glial scar around, damaged regions.
Collapse
Affiliation(s)
- Jinping Cheng
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 107 Yan Jiang Xi Rd, Guangzhou, 510120, People's Republic of China
| | - Nils Korte
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, WC1E 6BT, UK
| | - Ross Nortley
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, WC1E 6BT, UK
| | - Huma Sethi
- Department of Neurosurgery, National Hospital for Neurology and Neurosurgery, Queen Square, London, WC1N 3BG, UK
| | - Yamei Tang
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 107 Yan Jiang Xi Rd, Guangzhou, 510120, People's Republic of China.
| | - David Attwell
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, WC1E 6BT, UK.
| |
Collapse
|
33
|
Schultz N, Brännström K, Byman E, Moussaud S, Nielsen HM, Olofsson A, Wennström M. Amyloid-beta 1-40 is associated with alterations in NG2+ pericyte population ex vivo and in vitro. Aging Cell 2018; 17:e12728. [PMID: 29453790 PMCID: PMC5946076 DOI: 10.1111/acel.12728] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/24/2017] [Indexed: 12/17/2022] Open
Abstract
The population of brain pericytes, a cell type important for vessel stability and blood brain barrier function, has recently been shown altered in patients with Alzheimer's disease (AD). The underlying reason for this alteration is not fully understood, but progressive accumulation of the AD characteristic peptide amyloid‐beta (Aβ) has been suggested as a potential culprit. In the current study, we show reduced number of hippocampal NG2+ pericytes and an association between NG2+ pericyte numbers and Aβ1‐40 levels in AD patients. We further demonstrate, using in vitro studies, an aggregation‐dependent impact of Aβ1‐40 on human NG2+ pericytes. Fibril‐EP Aβ1‐40 exposure reduced pericyte viability and proliferation and increased caspase 3/7 activity. Monomer Aβ1‐40 had quite the opposite effect: increased pericyte viability and proliferation and reduced caspase 3/7 activity. Oligomer‐EP Aβ1‐40 had no impact on either of the cellular events. Our findings add to the growing number of studies suggesting a significant impact on pericytes in the brains of AD patients and suggest different aggregation forms of Aβ1‐40 as potential key regulators of the brain pericyte population size.
Collapse
Affiliation(s)
- Nina Schultz
- Clinical Memory Research Unit; Department of Clinical Sciences Malmö; Lund University; Malmö Sweden
| | | | - Elin Byman
- Clinical Memory Research Unit; Department of Clinical Sciences Malmö; Lund University; Malmö Sweden
| | - Simon Moussaud
- Department of Neurochemistry; Stockholm University; Stockholm Sweden
| | | | - Anders Olofsson
- Department of Medical Biochemistry and Biophysics; Umeå University; Umeå Sweden
| | - Malin Wennström
- Clinical Memory Research Unit; Department of Clinical Sciences Malmö; Lund University; Malmö Sweden
| | | |
Collapse
|
34
|
Microglial SMAD4 regulated by microRNA-146a promotes migration of microglia which support tumor progression in a glioma environment. Oncotarget 2018; 9:24950-24969. [PMID: 29861845 PMCID: PMC5982777 DOI: 10.18632/oncotarget.25116] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Accepted: 03/19/2018] [Indexed: 12/28/2022] Open
Abstract
Glioma tumors constitute a significant portion of microglial cells, which are known to support tumor progression. The present study demonstrates that transforming growth factor-β (TGFβ) signaling pathway in microglia in a glioma environment is involved in tumor progression and pathogenesis. It has been shown that the TGFβ level is elevated in higher grades of gliomas and its signaling pathway regulates tumor progression through phosphorylation of SMAD2 and SMAD3, which form a complex with SMAD4 to regulate target gene transcription. In an in vitro cell line-based model increased protein levels of pSMAD2/3, total SMAD2/3 and SMAD4 were observed in murine BV2 microglia cultured in glioma conditioned medium (GCM), indicative of the activated TGFβ signaling pathway in microglia associated with glioma environment. Immunofluorescence labeling further revealed the expression of SMAD4 in microglial and non-microglial cells of human glioblastomas tissue in vivo. Functional analysis through shRNA-mediated stable knockdown of SMAD4 in microglia revealed the downregulation of the expression of matrix metalloproteinase 9 (MMP9), which has been shown to be involved in tumor progression and cell migration. Further, knockdown of SMAD4 in microglia decreased the migration of microglial cells towards GCM, indicating that SMAD4 promotes microglial migration in glioma environment. In addition, SMAD4 has been shown to be post-transcriptionally regulated by microRNA-146a, which was downregulated in microglia treated with GCM. Overexpression of miR-146a resulted in decreased expression of SMAD4 together with tumor supportive gene MMP9 in microglia, and subsequently suppressed microglial migration towards GCM, possibly through regulation of SMAD4. On the other hand, the cell viability assay revealed decreased viability of glioma cells when they were treated with conditioned medium derived from SMAD4 knockdown microglia or miR-146a overexpressed microglia as compared to glioma cells treated with the medium from control microglial cells. Taken together, the present study suggests that microglial SMAD4 which is epigenetically regulated by miR-146a promotes microglial migration in gliomas and glioma cell viability.
Collapse
|
35
|
Mäder L, Blank AE, Capper D, Jansong J, Baumgarten P, Wirsik NM, Zachskorn C, Ehlers J, Seifert M, Klink B, Liebner S, Niclou S, Naumann U, Harter PN, Mittelbronn M. Pericytes/vessel-associated mural cells (VAMCs) are the major source of key epithelial-mesenchymal transition (EMT) factors SLUG and TWIST in human glioma. Oncotarget 2018; 9:24041-24053. [PMID: 29844871 PMCID: PMC5963615 DOI: 10.18632/oncotarget.25275] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 04/03/2018] [Indexed: 11/25/2022] Open
Abstract
Epithelial-to-mesenchymal transition (EMT) is supposed to be responsible for increased invasion and metastases in epithelial cancer cells. The activation of EMT genes has further been proposed to be important in the process of malignant transformation of primary CNS tumors. Since the cellular source and clinical impact of EMT factors in primary CNS tumors still remain unclear, we aimed at deciphering their distribution in vivo and clinico-pathological relevance in human gliomas. We investigated 350 glioma patients for the expression of the key EMT factors SLUG and TWIST by immunohistochemistry and immunofluorescence related to morpho-genetic alterations such as EGFR-amplification, IDH-1 (R132H) mutation and 1p/19q LOH. Furthermore, transcriptional cluster and survival analyses were performed. Our data illustrate that SLUG and TWIST are overexpressed in gliomas showing vascular proliferation such as pilocytic astrocytomas and glioblastomas. EMT factors are exclusively expressed by non-neoplastic pericytes/vessel-associated mural cells (VAMCs). They are not associated with patient survival but correlate with pericytic/VAMC genes in glioblastoma cluster analysis. In summary, the upregulation of EMT genes in pilocytic astrocytomas and glioblastomas reflects the level of activation of pericytes/VAMCs in newly formed blood vessels. Our results underscore that the negative prognostic potential of the EMT signature in the group of diffuse gliomas of WHO grade II-IV does most likely not derive from glioma cells but rather reflects the degree of proliferating mural cells thereby constituting a potential target for future alternative treatment approaches.
Collapse
Affiliation(s)
- Lisa Mäder
- Edinger Institute (Neurological Institute), Goethe University, Frankfurt, Germany
| | - Anna E Blank
- Edinger Institute (Neurological Institute), Goethe University, Frankfurt, Germany
| | - David Capper
- Department of Neuropathology, University of Heidelberg, Heidelberg, Germany.,Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Neuropathology, Berlin, Germany
| | - Janina Jansong
- Laboratory of Molecular Neuro-Oncology, Department of Vascular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Peter Baumgarten
- Edinger Institute (Neurological Institute), Goethe University, Frankfurt, Germany
| | - Naita M Wirsik
- Edinger Institute (Neurological Institute), Goethe University, Frankfurt, Germany
| | - Cornelia Zachskorn
- Edinger Institute (Neurological Institute), Goethe University, Frankfurt, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jakob Ehlers
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Neuropathology, Berlin, Germany.,Department of Radiation Oncology, University of Tübingen, Tübingen, Germany
| | - Michael Seifert
- Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Institute for Medical Informatics and Biometry (IMB), Dresden, Germany.,National Center for Tumor Diseases (NCT), Dresden, Germany
| | - Barbara Klink
- Institute for Clinical Genetics, Faculty of Medicine Carl Gustav Carus, Dresden University of Technology, Dresden, Germany
| | - Stefan Liebner
- Edinger Institute (Neurological Institute), Goethe University, Frankfurt, Germany
| | - Simone Niclou
- NORLUX Neuro-Oncology Laboratory, Luxembourg Institute of Health (LIH), Strassen, Luxembourg
| | - Ulrike Naumann
- Laboratory of Molecular Neuro-Oncology, Department of Vascular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Patrick N Harter
- Edinger Institute (Neurological Institute), Goethe University, Frankfurt, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Michel Mittelbronn
- Edinger Institute (Neurological Institute), Goethe University, Frankfurt, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany.,NORLUX Neuro-Oncology Laboratory, Luxembourg Institute of Health (LIH), Strassen, Luxembourg.,Luxembourg Centre of Neuropathology (LCNP), Dudelange, Luxembourg.,Laboratoire National de Santé (LNS), Dudelange, Luxembourg.,Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
| |
Collapse
|
36
|
Padel T, Roth M, Gaceb A, Li JY, Björkqvist M, Paul G. Brain pericyte activation occurs early in Huntington's disease. Exp Neurol 2018; 305:139-150. [PMID: 29630897 DOI: 10.1016/j.expneurol.2018.03.015] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 03/03/2018] [Accepted: 03/29/2018] [Indexed: 02/03/2023]
Abstract
Microvascular changes have recently been described for several neurodegenerative disorders, including Huntington's disease (HD). HD is characterized by a progressive neuronal cell loss due to a mutation in the Huntingtin gene. However, the temporal and spatial microvascular alterations in HD remain unclear. Also, knowledge on the implication of pericytes in HD pathology is still sparse and existing findings are contradictory. Here we examine alterations in brain pericytes in the R6/2 mouse model of HD and in human post mortem HD brain sections. To specifically track activated pericytes, we crossbred R6/2 mice with transgenic mice expressing the Green fluorescent protein gene under the Regulator of G-protein signaling 5 (Rgs5) promoter. We demonstrate an increase in activated pericytes in the R6/2 brain and in post mortem HD brain tissue. Importantly, pericyte changes are already detected before striatal neuronal cell loss, weight loss or behavioural deficits occur in R6/2 mice. This is associated with vascular alterations, whereby striatal changes precede cortical changes. Our findings suggest that pericyte activation may be one of the initial steps contributing to the observed vascular modifications in HD. Thus, pericytes may constitute an important target to address early microvascular changes contributing to disease progression in HD.
Collapse
Affiliation(s)
- Thomas Padel
- Translational Neurology group, Department of Clinical Science, Wallenberg Neuroscience Center, Wallenberg Centre for Molecular Medicine at Lund University, Lund University, 22184 Lund, Sweden
| | - Michaela Roth
- Translational Neurology group, Department of Clinical Science, Wallenberg Neuroscience Center, Wallenberg Centre for Molecular Medicine at Lund University, Lund University, 22184 Lund, Sweden
| | - Abderahim Gaceb
- Translational Neurology group, Department of Clinical Science, Wallenberg Neuroscience Center, Wallenberg Centre for Molecular Medicine at Lund University, Lund University, 22184 Lund, Sweden
| | - Jia-Yi Li
- Neural Plasticity and Repair Unit, Department of Experimental Medical Science, Wallenberg Neuroscience Center, Lund University, 22184 Lund, Sweden
| | - Maria Björkqvist
- Biomarkers in Brain Disease, Department of Experimental Medical Science, Wallenberg Neuroscience Center, Lund University, 22184 Lund, Sweden
| | - Gesine Paul
- Translational Neurology group, Department of Clinical Science, Wallenberg Neuroscience Center, Wallenberg Centre for Molecular Medicine at Lund University, Lund University, 22184 Lund, Sweden; Department of Neurology, Scania University Hospital, 22185 Lund, Sweden.
| |
Collapse
|
37
|
STAT3 precedes HIF1α transcriptional responses to oxygen and oxygen and glucose deprivation in human brain pericytes. PLoS One 2018. [PMID: 29518129 PMCID: PMC5843348 DOI: 10.1371/journal.pone.0194146] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Brain pericytes are important to maintain vascular integrity of the neurovascular unit under both physiological and ischemic conditions. Ischemic stroke is known to induce an inflammatory and hypoxic response due to the lack of oxygen and glucose in the brain tissue. How this early response to ischemia is molecularly regulated in pericytes is largely unknown and may be of importance for future therapeutic targets. Here we evaluate the transcriptional responses in in vitro cultured human brain pericytes after oxygen and/or glucose deprivation. Hypoxia has been widely known to stabilise the transcription factor hypoxia inducible factor 1-alpha (HIF1α) and mediate the induction of hypoxic transcriptional programs after ischemia. However, we find that the transcription factors Jun Proto-Oncogene (c-JUN), Nuclear Factor Of Kappa Light Polypeptide Gene Enhancer In B-Cells (NFκB) and signal transducer and activator of transcription 3 (STAT3) bind genes regulated after 2hours (hs) of omitted glucose and oxygen before HIF1α. Potent HIF1α responses require 6hs of hypoxia to substantiate transcriptional regulation comparable to either c-JUN or STAT3. Phosphorylated STAT3 protein is at its highest after 5 min of oxygen and glucose (OGD) deprivation, whereas maximum HIF1α stabilisation requires 120 min. We show that STAT3 regulates angiogenic and metabolic pathways before HIF1α, suggesting that HIF1α is not the initiating trans-acting factor in the response of pericytes to ischemia.
Collapse
|
38
|
Zhao X, Chen R, Liu M, Feng J, Chen J, Hu K. Remodeling the blood-brain barrier microenvironment by natural products for brain tumor therapy. Acta Pharm Sin B 2017; 7:541-553. [PMID: 28924548 PMCID: PMC5595291 DOI: 10.1016/j.apsb.2017.07.002] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 06/08/2017] [Accepted: 07/12/2017] [Indexed: 12/23/2022] Open
Abstract
Brain tumor incidence shows an upward trend in recent years; brain tumors account for 5% of adult tumors, while in children, this figure has increased to 70%. Moreover, 20%-30% of malignant tumors will eventually metastasize into the brain. Both benign and malignant tumors can cause an increase in intracranial pressure and brain tissue compression, leading to central nervous system (CNS) damage which endangers the patients' lives. Despite the many approaches to treating brain tumors and the progress that has been made, only modest gains in survival time of brain tumor patients have been achieved. At present, chemotherapy is the treatment of choice for many cancers, but the special structure of the blood-brain barrier (BBB) limits most chemotherapeutic agents from passing through the BBB and penetrating into tumors in the brain. The BBB microenvironment contains numerous cell types, including endothelial cells, astrocytes, peripheral cells and microglia, and extracellular matrix (ECM). Many chemical components of natural products are reported to regulate the BBB microenvironment near brain tumors and assist in their treatment. This review focuses on the composition and function of the BBB microenvironment under both physiological and pathological conditions, and the current research progress in regulating the BBB microenvironment by natural products to promote the treatment of brain tumors.
Collapse
Affiliation(s)
- Xiao Zhao
- Murad Research Center for Modernized Chinese Medicine, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Rujing Chen
- Murad Research Center for Modernized Chinese Medicine, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Mei Liu
- Murad Research Center for Modernized Chinese Medicine, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jianfang Feng
- Murad Research Center for Modernized Chinese Medicine, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jun Chen
- Key Laboratory of Smart Drug Delivery, Fudan University, Ministry of Education, Shanghai 201203, China
| | - Kaili Hu
- Murad Research Center for Modernized Chinese Medicine, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| |
Collapse
|
39
|
Jackson S, ElAli A, Virgintino D, Gilbert MR. Blood-brain barrier pericyte importance in malignant gliomas: what we can learn from stroke and Alzheimer's disease. Neuro Oncol 2017; 19:1173-1182. [PMID: 28541444 PMCID: PMC5570196 DOI: 10.1093/neuonc/nox058] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The pericyte, a constitutive component of the central nervous system, is a poorly understood cell type that envelops the endothelial cell with the intended purpose of regulating vascular flow and endothelial cell permeability. Previous studies of pericyte function have been limited to a small number of disease processes such as ischemic stroke and Alzheimer's disease. Recently, publications have postulated a link between glioma stem cell differentiation and pericyte function. These studies suggest that there may be an important interaction of pericytes with tumor cells and other components of the tumor microenvironment in malignant primary glial neoplasms, most notably glioblastoma. This potential cellular interaction underscores the need to pursue more investigations of pericytes in malignant brain tumor biology. In this review, we summarize the functional roles of pericytes, particularly focusing on changes in pericyte biology during response to immune cells, inflammation, and hypoxic conditions. The information presented is based on the available data from studies of pericyte function in other central nervous system diseases but will serve as a foundation for research investigations to further understand the role of pericytes in malignant gliomas.
Collapse
Affiliation(s)
- Sadhana Jackson
- National Cancer Institute, Neuro-oncology Branch, Bethesda, Maryland; Research Center of CHU de Québec-Université Laval, Neuroscience Axis, Quebec, Canada; Department of Basic Medical Sciences, Neurosciences, and Sensory Organs, Human Anatomy and Histology Unit, University of Bari School of Medicine, Bari, Italy
| | - Ayman ElAli
- National Cancer Institute, Neuro-oncology Branch, Bethesda, Maryland; Research Center of CHU de Québec-Université Laval, Neuroscience Axis, Quebec, Canada; Department of Basic Medical Sciences, Neurosciences, and Sensory Organs, Human Anatomy and Histology Unit, University of Bari School of Medicine, Bari, Italy
| | - Daniela Virgintino
- National Cancer Institute, Neuro-oncology Branch, Bethesda, Maryland; Research Center of CHU de Québec-Université Laval, Neuroscience Axis, Quebec, Canada; Department of Basic Medical Sciences, Neurosciences, and Sensory Organs, Human Anatomy and Histology Unit, University of Bari School of Medicine, Bari, Italy
| | - Mark R Gilbert
- National Cancer Institute, Neuro-oncology Branch, Bethesda, Maryland; Research Center of CHU de Québec-Université Laval, Neuroscience Axis, Quebec, Canada; Department of Basic Medical Sciences, Neurosciences, and Sensory Organs, Human Anatomy and Histology Unit, University of Bari School of Medicine, Bari, Italy
| |
Collapse
|
40
|
Chen J, Mao S, Li H, Zheng M, Yi L, Lin JM, Lin ZX. The pathological structure of the perivascular niche in different microvascular patterns of glioblastoma. PLoS One 2017; 12:e0182183. [PMID: 28771552 PMCID: PMC5542434 DOI: 10.1371/journal.pone.0182183] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 07/13/2017] [Indexed: 01/22/2023] Open
Abstract
The perivascular niche is critical for intercellular communication between resident cell types in glioblastoma (GBM), and it plays a vital role in maintaining the glioma stem cell (GSC) microenvironment. It is shown in abundant research that different microvascular patterns exist in GBM; and it can be implied that different microvascular patterns are associated with different pathological structures in the perivascular niche. However, the pathological structure of the perivascular niche is still not clear. Here, we investigated the distribution and biological characteristics of different microvascular pattern niches (MVPNs) in GBM by detecting the expression of CD34, CD133, Nestin, α-SMA, GFAP and CD14 in the perivascular niche using multiple -fluorescence. The four basic microvascular patterns are microvascular sprouting (MS), vascular cluster (VC), vascular garland (VG), and glomeruloid vascular proliferation (GVP). By analyzing the proportion of the area of each marker in four types of formations, the results indicated that the expression of CD34, CD133 and Nestin in MS and VC was significantly lower than that in VG and GVP (P<0.05). Furthermore, the results showed that α-SMA expression different in the MS, VC, VG and GVP (P<0.05). However, the expression of GFAP and CD14 in each type of formation exhibited no significant difference (P>0.05). According to the area distributions of different markers, we mapped four precise simulation diagrams to provide an effective foundation for the accurate simulation of glioblastoma in vitro.
Collapse
Affiliation(s)
- Jintao Chen
- Department of Neurosurgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
- Department of Chemistry, Beijing Key Laboratory of Microanalytical Methods and Instrumentation, Tsinghua University, Beijing, China
| | - Sifeng Mao
- Department of Chemistry, Beijing Key Laboratory of Microanalytical Methods and Instrumentation, Tsinghua University, Beijing, China
| | - Haifang Li
- Department of Chemistry, Beijing Key Laboratory of Microanalytical Methods and Instrumentation, Tsinghua University, Beijing, China
| | - Mingcheng Zheng
- Department of Neurosurgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Linglu Yi
- Department of Chemistry, Beijing Key Laboratory of Microanalytical Methods and Instrumentation, Tsinghua University, Beijing, China
| | - Jin-Ming Lin
- Department of Chemistry, Beijing Key Laboratory of Microanalytical Methods and Instrumentation, Tsinghua University, Beijing, China
- * E-mail: (JML); (ZXL)
| | - Zhi-xiong Lin
- Department of Neurosurgery, Beijing Sanbo Brain Hospital, Capital Medical University, Beijing, China
- * E-mail: (JML); (ZXL)
| |
Collapse
|
41
|
Audia A, Conroy S, Glass R, Bhat KPL. The Impact of the Tumor Microenvironment on the Properties of Glioma Stem-Like Cells. Front Oncol 2017; 7:143. [PMID: 28740831 PMCID: PMC5502267 DOI: 10.3389/fonc.2017.00143] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 06/20/2017] [Indexed: 01/22/2023] Open
Abstract
Glioblastoma is the most common and highly malignant primary brain tumor, and patients affected with this disease exhibit a uniformly dismal prognosis. Glioma stem-like cells (GSCs) are a subset of cells within the bulk tumor that possess self-renewal and multi-lineage differentiation properties similar to somatic stem cells. These cells also are at the apex of the cellular hierarchy and cause tumor initiation and expansion after chemo-radiation. These traits make them an attractive target for therapeutic development. Because GSCs are dependent on the brain microenvironment for their growth, and because non-tumorigenic cell types in the microenvironment can influence GSC phenotypes and treatment response, a better understanding of these cell types is needed. In this review, we provide a focused overview of the contributions from the microenvironment to GSC homing, maintenance, phenotypic plasticity, and tumor initiation. The interaction of GSCs with the vascular compartment, mesenchymal stem cells, immune system, and normal brain cell types are discussed. Studies that provide mechanistic insight into each of these GSC–microenvironment interactions are warranted in the future.
Collapse
Affiliation(s)
- Alessandra Audia
- Department of Translational Molecular Pathology, University of Texas, M.D. Anderson Cancer Center, Houston, TX, United States
| | - Siobhan Conroy
- Department of Translational Molecular Pathology, University of Texas, M.D. Anderson Cancer Center, Houston, TX, United States.,Department of Pathology and Medical Biology, University Medical Center, Groningen, Netherlands
| | - Rainer Glass
- Neurosurgical Research, Department of Neurosurgery, University Hospital, LMU Munich, Munich, Germany.,German Cancer Consortium (DKTK) partner site Munich and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Krishna P L Bhat
- Department of Translational Molecular Pathology, University of Texas, M.D. Anderson Cancer Center, Houston, TX, United States.,Department of Neurosurgery, University of Texas, M.D. Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
42
|
Sweeney MD, Ayyadurai S, Zlokovic BV. Pericytes of the neurovascular unit: key functions and signaling pathways. Nat Neurosci 2017; 19:771-83. [PMID: 27227366 DOI: 10.1038/nn.4288] [Citation(s) in RCA: 776] [Impact Index Per Article: 97.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 02/29/2016] [Indexed: 12/12/2022]
Abstract
Pericytes are vascular mural cells embedded in the basement membrane of blood microvessels. They extend their processes along capillaries, pre-capillary arterioles and post-capillary venules. CNS pericytes are uniquely positioned in the neurovascular unit between endothelial cells, astrocytes and neurons. They integrate, coordinate and process signals from their neighboring cells to generate diverse functional responses that are critical for CNS functions in health and disease, including regulation of the blood-brain barrier permeability, angiogenesis, clearance of toxic metabolites, capillary hemodynamic responses, neuroinflammation and stem cell activity. Here we examine the key signaling pathways between pericytes and their neighboring endothelial cells, astrocytes and neurons that control neurovascular functions. We also review the role of pericytes in CNS disorders including rare monogenic diseases and complex neurological disorders such as Alzheimer's disease and brain tumors. Finally, we discuss directions for future studies.
Collapse
Affiliation(s)
- Melanie D Sweeney
- Department of Physiology and Biophysics, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA.,Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Shiva Ayyadurai
- Systems Biology Group, CytoSolve Research Division, Cambridge, Massachusetts, USA
| | - Berislav V Zlokovic
- Department of Physiology and Biophysics, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA.,Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| |
Collapse
|
43
|
Vascular Transdifferentiation in the CNS: A Focus on Neural and Glioblastoma Stem-Like Cells. Stem Cells Int 2016; 2016:2759403. [PMID: 27738435 PMCID: PMC5055959 DOI: 10.1155/2016/2759403] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Accepted: 09/05/2016] [Indexed: 01/12/2023] Open
Abstract
Glioblastomas are devastating and extensively vascularized brain tumors from which glioblastoma stem-like cells (GSCs) have been isolated by many groups. These cells have a high tumorigenic potential and the capacity to generate heterogeneous phenotypes. There is growing evidence to support the possibility that these cells are derived from the accumulation of mutations in adult neural stem cells (NSCs) as well as in oligodendrocyte progenitors. It was recently reported that GSCs could transdifferentiate into endothelial-like and pericyte-like cells both in vitro and in vivo, notably under the influence of Notch and TGFβ signaling pathways. Vascular cells derived from GBM cells were also observed directly in patient samples. These results could lead to new directions for designing original therapeutic approaches against GBM neovascularization but this specific reprogramming requires further molecular investigations. Transdifferentiation of nontumoral neural stem cells into vascular cells has also been described and conversely vascular cells may generate neural stem cells. In this review, we present and discuss these recent data. As some of them appear controversial, further validation will be needed using new technical approaches such as high throughput profiling and functional analyses to avoid experimental pitfalls and misinterpretations.
Collapse
|
44
|
Paolillo M, Serra M, Schinelli S. Integrins in glioblastoma: Still an attractive target? Pharmacol Res 2016; 113:55-61. [PMID: 27498157 DOI: 10.1016/j.phrs.2016.08.004] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 07/29/2016] [Accepted: 08/01/2016] [Indexed: 02/08/2023]
Abstract
Integrin-mediated signaling pathways have been found to promote the invasiveness and survival of glioma cells by modifying the brain microenvironment to support the formation of the tumoral niche. A variety of cells in the niche express integrin receptors, including tumor-associated macrophages, fibroblasts, endothelial cells and pericytes. In particular, RGD-binding integrins have been demonstrated to have an important role in the epithelial-mesenchymal transition process, considered the first step in the infiltration of tissue by cancer cells and molecular markers of which have been found in glioma cells. In simultaneous research, Small Molecule Integrin Antagonists (SMIA) yielded initially promising results in in vitro and in vivo studies, leading to clinical trials to test their safety and efficacy in combination with other anticancer drugs in the treatment of several tumor types. The initially high expectations, especially because of their antiangiogenic activity, which appeared to be a winning strategy against GBM, were not confirmed and this cast serious doubts on the real benefits to be gained from the use of SMIA for the treatment of cancer in humans. In this review, we provide an overview of recent findings concerning the functional roles of integrins, especially RGD-binding integrins, in the processes related to glioma cells survival and brain tissue infiltration. These findings disclose a new scenario in which recently developed SMIA might become useful tools to hinder glioblastoma cell dissemination.
Collapse
Affiliation(s)
- Mayra Paolillo
- Department of Drug Sciences, University of Pavia, Pavia, Italy.
| | - Massimo Serra
- Department of Drug Sciences, University of Pavia, Pavia, Italy
| | | |
Collapse
|
45
|
Pombero A, Garcia-Lopez R, Martinez S. Brain mesenchymal stem cells: physiology and pathological implications. Dev Growth Differ 2016; 58:469-80. [PMID: 27273235 DOI: 10.1111/dgd.12296] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 05/03/2016] [Accepted: 05/03/2016] [Indexed: 12/12/2022]
Abstract
Mesenchymal stem cells (MSCs) are defined as progenitor cells that give rise to a number of unique, differentiated mesenchymal cell types. This concept has progressively evolved towards an all-encompassing concept including multipotent perivascular cells of almost any tissue. In central nervous system, pericytes are involved in blood-brain barrier, and angiogenesis and vascular tone regulation. They form the neurovascular unit (NVU) together with endothelial cells, astrocytes and neurons. This functional structure provides an optimal microenvironment for neural proliferation in the adult brain. Neurovascular niche include both diffusible signals and direct contact with endothelial and pericytes, which are a source of diffusible neurotrophic signals that affect neural precursors. Therefore, MSCs/pericyte properties such as differentiation capability, as well as immunoregulatory and paracrine effects make them a potential resource in regenerative medicine.
Collapse
Affiliation(s)
- Ana Pombero
- Intituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, University of Murcia, Murcia, Spain
| | - Raquel Garcia-Lopez
- Instituto de Neurociencias, Universidad Miguel Hernandez-Consejo Superior de Investigaciones, Av Ramon y Cajal s/n, San Juan de Alicante, 03550, Spain
| | - Salvador Martinez
- Instituto de Neurociencias, Universidad Miguel Hernandez-Consejo Superior de Investigaciones, Av Ramon y Cajal s/n, San Juan de Alicante, 03550, Spain
| |
Collapse
|