1
|
Milan M, Maiullari F, Chirivì M, Ceraolo MG, Zigiotto R, Soluri A, Maiullari S, Landoni E, Silvestre DD, Brambilla F, Mauri P, De Paolis V, Fratini N, Crosti MC, Cordiglieri C, Parisi C, Calogero A, Seliktar D, Torrente Y, Lanzuolo C, Dotti G, Toccafondi M, Bombaci M, De Falco E, Bearzi C, Rizzi R. Macrophages producing chondroitin sulfate proteoglycan-4 induce neuro-cardiac junction impairment in Duchenne muscular dystrophy. J Pathol 2025; 265:1-13. [PMID: 39523812 PMCID: PMC11638662 DOI: 10.1002/path.6362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 08/12/2024] [Accepted: 10/01/2024] [Indexed: 11/16/2024]
Abstract
Duchenne muscular dystrophy (DMD) is caused by the absence of the full form of the dystrophin protein, which is essential for maintaining the structural integrity of muscle cells, including those in the heart and respiratory system. Despite progress in understanding the molecular mechanisms associated with DMD, myocardial insufficiency persists as the primary cause of mortality, and existing therapeutic strategies remain limited. This study investigates the hypothesis that a dysregulation of the biological communication between infiltrating macrophages (MPs) and neurocardiac junctions exists in dystrophic cardiac tissue. In a mouse model of DMD (mdx), this phenomenon is influenced by the over-release of chondroitin sulfate proteoglycan-4 (CSPG4), a key inhibitor of nerve sprouting and a modulator of the neural function, by MPs infiltrating the cardiac tissue and associated with dilated cardiomyopathy, a hallmark of DMD. Givinostat, the histone deacetylase inhibitor under current development as a clinical treatment for DMD, is effective at both restoring a physiological microenvironment at the neuro-cardiac junction and cardiac function in mdx mice in addition to a reduction in cardiac fibrosis, MP-mediated inflammation, and tissue CSPG4 content. This study provides novel insight into the pathophysiology of DMD in the heart, identifying potential new biological targets. © 2024 The Author(s). The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Marika Milan
- Neurology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore PoliclinicoMilanItaly
- Fondazione Istituto Nazionale di Genetica Molecolare (INGM) ‘Romeo ed Enrica Invernizzi’MilanItaly
| | - Fabio Maiullari
- Fondazione Istituto Nazionale di Genetica Molecolare (INGM) ‘Romeo ed Enrica Invernizzi’MilanItaly
- PhD Program in Cellular and Molecular Biology, Department of BiologyUniversity of Rome ‘Tor Vergata’RomeItaly
| | - Maila Chirivì
- Neurology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore PoliclinicoMilanItaly
- Department of Molecular MedicineSapienza UniversityRomeItaly
| | - Maria Grazia Ceraolo
- Neurology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore PoliclinicoMilanItaly
- Fondazione Istituto Nazionale di Genetica Molecolare (INGM) ‘Romeo ed Enrica Invernizzi’MilanItaly
| | - Rebecca Zigiotto
- Fondazione Istituto Nazionale di Genetica Molecolare (INGM) ‘Romeo ed Enrica Invernizzi’MilanItaly
| | - Andrea Soluri
- Unit of Molecular NeurosciencesUniversity Campus Bio‐Medico, RomeRomeItaly
- Institute of Biochemistry and Cell BiologyNational Research CouncilRomeItaly
| | - Silvia Maiullari
- Institute of Biochemistry and Cell BiologyNational Research CouncilRomeItaly
| | - Elisa Landoni
- Lineberger Comprehensive Cancer CenterUniversity of North Carolina at Chapel HillChapel HillNCUSA
| | | | | | - Pierluigi Mauri
- Institute of Biomedical TechnologiesNational Research CouncilMilanItaly
| | - Veronica De Paolis
- Institute of Biochemistry and Cell BiologyNational Research CouncilRomeItaly
| | - Nicole Fratini
- Department of Molecular MedicineSapienza UniversityRomeItaly
| | - Maria Cristina Crosti
- Fondazione Istituto Nazionale di Genetica Molecolare (INGM) ‘Romeo ed Enrica Invernizzi’MilanItaly
| | - Chiara Cordiglieri
- Fondazione Istituto Nazionale di Genetica Molecolare (INGM) ‘Romeo ed Enrica Invernizzi’MilanItaly
| | - Chiara Parisi
- Institute of Biochemistry and Cell BiologyNational Research CouncilRomeItaly
| | - Antonella Calogero
- Department of Medical‐Surgical Sciences and BiotechnologiesSapienza University of RomeLatinaItaly
| | - Dror Seliktar
- Department of Biomedical EngineeringTechnion InstituteHaifaIsrael
| | - Yvan Torrente
- Neurology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore PoliclinicoMilanItaly
| | - Chiara Lanzuolo
- Fondazione Istituto Nazionale di Genetica Molecolare (INGM) ‘Romeo ed Enrica Invernizzi’MilanItaly
- Institute of Biomedical TechnologiesNational Research CouncilMilanItaly
| | - Gianpietro Dotti
- Lineberger Comprehensive Cancer CenterUniversity of North Carolina at Chapel HillChapel HillNCUSA
| | - Mirco Toccafondi
- Fondazione Istituto Nazionale di Genetica Molecolare (INGM) ‘Romeo ed Enrica Invernizzi’MilanItaly
| | - Mauro Bombaci
- Fondazione Istituto Nazionale di Genetica Molecolare (INGM) ‘Romeo ed Enrica Invernizzi’MilanItaly
| | - Elena De Falco
- Department of Medical‐Surgical Sciences and BiotechnologiesSapienza University of RomeLatinaItaly
| | - Claudia Bearzi
- Fondazione Istituto Nazionale di Genetica Molecolare (INGM) ‘Romeo ed Enrica Invernizzi’MilanItaly
- Institute of Biomedical TechnologiesNational Research CouncilMilanItaly
| | - Roberto Rizzi
- Fondazione Istituto Nazionale di Genetica Molecolare (INGM) ‘Romeo ed Enrica Invernizzi’MilanItaly
- Department of Medical‐Surgical Sciences and BiotechnologiesSapienza University of RomeLatinaItaly
| |
Collapse
|
2
|
Ma Z, Zhang W, Wang C, Su Y, Yi C, Niu J. A New Acquaintance of Oligodendrocyte Precursor Cells in the Central Nervous System. Neurosci Bull 2024; 40:1573-1589. [PMID: 39042298 PMCID: PMC11422404 DOI: 10.1007/s12264-024-01261-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 03/21/2024] [Indexed: 07/24/2024] Open
Abstract
Oligodendrocyte precursor cells (OPCs) are a heterogeneous multipotent population in the central nervous system (CNS) that appear during embryogenesis and persist as resident cells in the adult brain parenchyma. OPCs could generate oligodendrocytes to participate in myelination. Recent advances have renewed our knowledge of OPC biology by discovering novel markers of oligodendroglial cells, the myelin-independent roles of OPCs, and the regulatory mechanism of OPC development. In this review, we will explore the updated knowledge on OPC identity, their multifaceted roles in the CNS in health and diseases, as well as the regulatory mechanisms that are involved in their developmental stages, which hopefully would contribute to a further understanding of OPCs and attract attention in the field of OPC biology.
Collapse
Affiliation(s)
- Zexuan Ma
- Department of Histology and Embryology, College of basic medicine, Third Military Medical University, Chongqing, 400038, China
| | - Wei Zhang
- Department of Histology and Embryology, College of basic medicine, Third Military Medical University, Chongqing, 400038, China
| | - Chenmeng Wang
- Department of Histology and Embryology, College of basic medicine, Third Military Medical University, Chongqing, 400038, China
- Research Centre, Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| | - Yixun Su
- Research Centre, Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| | - Chenju Yi
- Research Centre, Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China.
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Guangzhou, China.
- Shenzhen Key Laboratory of Chinese Medicine Active substance screening and Translational Research, Shenzhen, 518107, China.
| | - Jianqin Niu
- Department of Histology and Embryology, College of basic medicine, Third Military Medical University, Chongqing, 400038, China.
- Chongqing Key Laboratory of Neurobiology, Chongqing, 400038, China.
| |
Collapse
|
3
|
Bottero M, Pessina G, Bason C, Vigo T, Uccelli A, Ferrara G. Nerve-Glial antigen 2: unmasking the enigmatic cellular identity in the central nervous system. Front Immunol 2024; 15:1393842. [PMID: 39136008 PMCID: PMC11317297 DOI: 10.3389/fimmu.2024.1393842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 07/05/2024] [Indexed: 08/15/2024] Open
Abstract
Chondroitin sulfate proteoglycans (CSPGs) are fundamental components of the extracellular matrix in the central nervous system (CNS). Among these, the Nerve-Glial antigen 2 (NG2) stands out as a transmembrane CSPG exclusively expressed in a different population of cells collectively termed NG2-expressing cells. These enigmatic cells, found throughout the developing and adult CNS, have been indicated with various names, including NG2 progenitor cells, polydendrocytes, synantocytes, NG2 cells, and NG2-Glia, but are more commonly referred to as oligodendrocyte progenitor cells. Characterized by high proliferation rates and unique morphology, NG2-expressing cells stand apart from neurons, astrocytes, and oligodendrocytes. Intriguingly, some NG2-expressing cells form functional glutamatergic synapses with neurons, challenging the long-held belief that only neurons possess the intricate machinery required for neurotransmission. In the CNS, the complexity surrounding NG2-expressing cells extends to their classification. Additionally, NG2 expression has been documented in pericytes and immune cells, suggesting a role in regulating brain innate immunity and neuro-immune crosstalk in homeostasis. Ongoing debates revolve around their heterogeneity, potential as progenitors for various cell types, responses to neuroinflammation, and the role of NG2. Therefore, this review aims to shed light on the enigma of NG2-expressing cells by delving into their structure, functions, and signaling pathways. We will critically evaluate the literature on NG2 expression across the CNS, and address the contentious issues surrounding their classification and roles in neuroinflammation and neurodegeneration. By unraveling the intricacies of NG2-expressing cells, we hope to pave the way for a more comprehensive understanding of their contributions to CNS health and during neurological disorders.
Collapse
Affiliation(s)
- Marta Bottero
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Giada Pessina
- Department of Experimental Medicine, University of Genoa, Genoa, Italy
| | | | - Tiziana Vigo
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Antonio Uccelli
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Department of Neurology, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Genoa, Italy
| | | |
Collapse
|
4
|
Alsabbagh R, Ahmed M, Alqudah MAY, Hamoudi R, Harati R. Insights into the Molecular Mechanisms Mediating Extravasation in Brain Metastasis of Breast Cancer, Melanoma, and Lung Cancer. Cancers (Basel) 2023; 15:cancers15082258. [PMID: 37190188 DOI: 10.3390/cancers15082258] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 04/08/2023] [Accepted: 04/11/2023] [Indexed: 05/17/2023] Open
Abstract
Brain metastasis is an incurable end-stage of systemic cancer associated with poor prognosis, and its incidence is increasing. Brain metastasis occurs through a multi-step cascade where cancer cells spread from the primary tumor site to the brain. The extravasation of tumor cells through the blood-brain barrier (BBB) is a critical step in brain metastasis. During extravasation, circulating cancer cells roll along the brain endothelium (BE), adhere to it, then induce alterations in the endothelial barrier to transmigrate through the BBB and enter the brain. Rolling and adhesion are generally mediated by selectins and adhesion molecules induced by inflammatory mediators, while alterations in the endothelial barrier are mediated by proteolytic enzymes, including matrix metalloproteinase, and the transmigration step mediated by factors, including chemokines. However, the molecular mechanisms mediating extravasation are not yet fully understood. A better understanding of these mechanisms is essential as it may serve as the basis for the development of therapeutic strategies for the prevention or treatment of brain metastases. In this review, we summarize the molecular events that occur during the extravasation of cancer cells through the blood-brain barrier in three types of cancer most likely to develop brain metastasis: breast cancer, melanoma, and lung cancer. Common molecular mechanisms driving extravasation in these different tumors are discussed.
Collapse
Affiliation(s)
- Rama Alsabbagh
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Munazza Ahmed
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Mohammad A Y Alqudah
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Rifat Hamoudi
- Clinical Sciences Department, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
- Division of Surgery and Interventional Science, University College London, London W1W 7EJ, UK
| | - Rania Harati
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| |
Collapse
|
5
|
Lee HJ, Jung DH, Kim NK, Shin HK, Choi BT. Effects of electroacupuncture on the functionality of NG2-expressing cells in perilesional brain tissue of mice following ischemic stroke. Neural Regen Res 2021; 17:1556-1565. [PMID: 34916441 PMCID: PMC8771106 DOI: 10.4103/1673-5374.330611] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Neural/glial antigen 2 (NG2)-expressing cells has multipotent stem cell activity under cerebral ischemia. Our study examined the effects of electroacupuncture (EA) therapy (2 Hz, 1 or 3 mA, 20 minutes) at the Sishencong acupoint on motor function after ischemic insult in the brain by investigating the rehabilitative potential of NG2-derived cells in a mouse model of ischemic stroke. EA stimulation alleviated motor deficits caused by ischemic stroke, and 1 mA EA stimulation was more efficacious than 3 mA EA stimulation or positive control treatment with edaravone, a free radical scavenger. The properties of NG2-expressing cells were altered with 1 mA EA stimulation, enhancing their survival in perilesional brain tissue via reduction of tumor necrosis factor alpha expression. EA stimulation robustly activated signaling pathways related to proliferation and survival of NG2-expressing cells and increased the expression of neurotrophic factors such as brain-derived neurotrophic factor, tumor growth factor beta, and neurotrophin 3. In the perilesional striatum, EA stimulation greatly increased the number of NG2-expressing cells double-positive for oligodendrocyte, endothelial cell, and microglia/macrophage markers (CC1, CD31, and CD68). EA therapy also greatly activated brain-derived neurotrophic factor/tropomyosin receptor kinase B and glycogen synthase kinase 3 beta signaling. Our results indicate that EA therapy may prevent functional loss at the perilesional site by enhancing survival and differentiation of NG2-expressing cells via the activation of brain-derived neurotrophic factor -induced signaling, subsequently ameliorating motor dysfunction. The animal experiments were approved by the Animal Ethics Committee of Pusan National University (approval Nos. PNU2019-2199 and PNU2019-2884) on April 8, 2019 and June 19, 2019.
Collapse
Affiliation(s)
- Hong Ju Lee
- Department of Korean Medical Science, School of Korean Medicine; Graduate Training Program of Korean Medicine for Healthy Aging, Pusan National University, Yangsan, Republic of Korea
| | - Da Hee Jung
- Department of Korean Medical Science, School of Korean Medicine; Graduate Training Program of Korean Medicine for Healthy Aging, Pusan National University, Yangsan, Republic of Korea
| | - Nam Kwen Kim
- Department of Korean Ophthalmology, Otolaryngology and Dermatology, School of Korean Medicine, Pusan National University, Yangsan, Republic of Korea
| | - Hwa Kyoung Shin
- Department of Korean Medical Science, School of Korean Medicine; Graduate Training Program of Korean Medicine for Healthy Aging, Pusan National University, Yangsan, Republic of Korea
| | - Byung Tae Choi
- Department of Korean Medical Science, School of Korean Medicine; Graduate Training Program of Korean Medicine for Healthy Aging, Pusan National University, Yangsan, Republic of Korea
| |
Collapse
|
6
|
An HJ, Ko KR, Baek M, Jeong Y, Lee HH, Kim H, Kim DK, Lee SY, Lee S. Pro-Angiogenic and Osteogenic Effects of Adipose Tissue-Derived Pericytes Synergistically Enhanced by Nel-like Protein-1. Cells 2021; 10:cells10092244. [PMID: 34571892 PMCID: PMC8470876 DOI: 10.3390/cells10092244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 08/19/2021] [Accepted: 08/24/2021] [Indexed: 11/16/2022] Open
Abstract
An important objective of vascularized tissue regeneration is to develop agents for osteonecrosis. We aimed to identify the pro-angiogenic and osteogenic efficacy of adipose tissue-derived (AD) pericytes combined with Nel-like protein-1 (NELL-1) to investigate the therapeutic effects on osteonecrosis. Tube formation and cell migration were assessed to determine the pro-angiogenic efficacy. Vessel formation was evaluated in vivo using the chorioallantoic membrane assay. A mouse model with a 2.5 mm necrotic bone fragment in the femoral shaft was used as a substitute for osteonecrosis in humans. Bone formation was assessed radiographically (plain radiographs, three-dimensional images, and quantitative analyses), and histomorphometric analyses were performed. To identify factors related to the effects of NELL-1, analysis using microarrays, qRT-PCR, and Western blotting was performed. The results for pro-angiogenic efficacy evaluation identified synergistic effects of pericytes and NELL-1 on tube formation, cell migration, and vessel formation. For osteogenic efficacy analysis, the mouse model for osteonecrosis was treated in combination with pericytes and NELL-1, and the results showed maximum bone formation using radiographic images and quantitative analyses, compared with other treatment groups and showed robust bone and vessel formation using histomorphometric analysis. We identified an association between FGF2 and the effects of NELL-1 using array-based analysis. Thus, combinatorial therapy using AD pericytes and NELL-1 may have potential as a novel treatment for osteonecrosis.
Collapse
Affiliation(s)
- Hyun-Ju An
- Department of Orthopaedic Surgery, CHA Bundang Medical Center, CHA University School of Medicine, 59 Yatap-Ro, Seongnam-si 13496, Gyeonggi-do, Korea; (H.-J.A.); (M.B.); (Y.J.); (H.H.L.)
| | - Kyung Rae Ko
- Department of Orthopaedic Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-Ro, Gangnam-gu, Seoul 06351, Korea;
| | - Minjung Baek
- Department of Orthopaedic Surgery, CHA Bundang Medical Center, CHA University School of Medicine, 59 Yatap-Ro, Seongnam-si 13496, Gyeonggi-do, Korea; (H.-J.A.); (M.B.); (Y.J.); (H.H.L.)
| | - Yoonhui Jeong
- Department of Orthopaedic Surgery, CHA Bundang Medical Center, CHA University School of Medicine, 59 Yatap-Ro, Seongnam-si 13496, Gyeonggi-do, Korea; (H.-J.A.); (M.B.); (Y.J.); (H.H.L.)
| | - Hyeon Hae Lee
- Department of Orthopaedic Surgery, CHA Bundang Medical Center, CHA University School of Medicine, 59 Yatap-Ro, Seongnam-si 13496, Gyeonggi-do, Korea; (H.-J.A.); (M.B.); (Y.J.); (H.H.L.)
| | - Hyungkyung Kim
- Department of Pathology, Kyung Hee University Hospital at Gangdong, Kyung Hee University, College of Medicine, 892 Dongnam-Ro, Gangdong-gu, Seoul 05278, Korea;
| | - Do Kyung Kim
- CHA Graduate School of Medicine, 120 Hyeryong-Ro, Pocheon-si 11160, Gyeonggi-do, Korea;
| | - So-Young Lee
- Department of Internal Medicine, CHA Bundang Medical Center, CHA University School of Medicine, 59 Yatap-Ro, Seongnam-si 13496, Gyeonggi-do, Korea;
| | - Soonchul Lee
- Department of Orthopaedic Surgery, CHA Bundang Medical Center, CHA University School of Medicine, 59 Yatap-Ro, Seongnam-si 13496, Gyeonggi-do, Korea; (H.-J.A.); (M.B.); (Y.J.); (H.H.L.)
- Correspondence: ; Tel.: +82-317-805-289; Fax: +82-317-083-578
| |
Collapse
|
7
|
Riew TR, Jin X, Kim S, Kim HL, Lee MY. Temporal dynamics of cells expressing NG2 and platelet-derived growth factor receptor-β in the fibrotic scar formation after 3-nitropropionic acid-induced acute brain injury. Cell Tissue Res 2021; 385:539-555. [PMID: 33864501 DOI: 10.1007/s00441-021-03438-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 02/18/2021] [Indexed: 12/22/2022]
Abstract
Neuron-glia antigen 2 (NG2) proteoglycan and platelet-derived growth factor receptor beta (PDGFR-β) are widely used markers of pericytes, which are considered cells that form fibrotic scars in response to central nervous system insults. However, the exact phenotypes of NG2- and PDGFR-β-expressing cells, as well as the origin of the fibrotic scar after central nervous system insults, are still elusive. In the present study, we directly examined the identities and distributions of NG2- and PDGFR-β-positive cells in the control and lesioned striatum injured by the mitochondrial toxin 3-nitropropionic acid. Immunoelectron microscopy and correlative light and electron microscopy clearly distinguished NG2 and PDGFR-β expression in the vasculature during the post-injury period. Vascular smooth muscle cells and pericytes expressed NG2, which was prominently increased after the injury. NG2 expression was restricted to these vascular mural cells until 14 days post-lesion. By contrast, PDGFR-β-positive cells were perivascular fibroblasts located abluminal to smooth muscle cells or pericytes. These PDGFR-β-expressing cells formed extravascular networks associated with collagen fibrils at 14 days post-lesion. We also found that in the injured striatal parenchyma, PDGFR-β could be used as a complementary marker of resting and reactive NG2 glia because activated microglia/macrophages shared only the NG2 expression with NG2 glia in the lesioned striatum. These data indicate that NG2 and PDGFR-β label different vascular mural and parenchymal cells in the healthy and injured brain, suggesting that fibrotic scar-forming cells most likely originate in PDGFR-β-positive perivascular fibroblasts rather than in NG2-positive pericytes.
Collapse
Affiliation(s)
- Tae-Ryong Riew
- Department of Anatomy, Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Xuyan Jin
- Department of Anatomy, Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea.,Department of Biomedicine and Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Soojin Kim
- Department of Anatomy, Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Hong Lim Kim
- Integrative Research Support Center, Laboratory of Electron Microscope, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Mun-Yong Lee
- Department of Anatomy, Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea. .,Department of Biomedicine and Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea.
| |
Collapse
|
8
|
Asiry S, Kim G, Filippou PS, Sanchez LR, Entenberg D, Marks DK, Oktay MH, Karagiannis GS. The Cancer Cell Dissemination Machinery as an Immunosuppressive Niche: A New Obstacle Towards the Era of Cancer Immunotherapy. Front Immunol 2021; 12:654877. [PMID: 33927723 PMCID: PMC8076861 DOI: 10.3389/fimmu.2021.654877] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 03/23/2021] [Indexed: 12/13/2022] Open
Abstract
Although cancer immunotherapy has resulted in unpreceded survival benefits to subsets of oncology patients, accumulating evidence from preclinical animal models suggests that the immunosuppressive tumor microenvironment remains a detrimental factor limiting benefit for many patient subgroups. Recent efforts on lymphocyte-mediated immunotherapies are primarily focused on eliminating cancer foci at primary and metastatic sites, but few studies have investigated the impact of these therapies on the highly complex process of cancer cell dissemination. The metastatic cascade involves the directional streaming of invasive/migratory tumor cells toward specialized blood vessel intravasation gateways, called TMEM doorways, to the peripheral circulation. Importantly, this process occurs under the auspices of a specialized tumor microenvironment, herewith referred to as "Dissemination Trajectory", which is supported by an ample array of tumor-associated macrophages (TAMs), skewed towards an M2-like polarization spectrum, and which is also vital for providing microenvironmental cues for cancer cell invasion, migration and stemness. Based on pre-existing evidence from preclinical animal models, this article outlines the hypothesis that dissemination trajectories do not only support the metastatic cascade, but also embody immunosuppressive niches, capable of providing transient and localized immunosubversion cues to the migratory/invasive cancer cell subpopulation while in the act of departing from a primary tumor. So long as these dissemination trajectories function as "immune deserts", the migratory tumor cell subpopulation remains efficient in evading immunological destruction and seeding metastatic sites, despite administration of cancer immunotherapy and/or other cytotoxic treatments. A deeper understanding of the molecular and cellular composition, as well as the signaling circuitries governing the function of these dissemination trajectories will further our overall understanding on TAM-mediated immunosuppression and will be paramount for the development of new therapeutic strategies for the advancement of optimal cancer chemotherapies, immunotherapies, and targeted therapies.
Collapse
Affiliation(s)
- Saeed Asiry
- Department of Pathology, Montefiore Medical Center, Albert Einstein College of Medicine, New York City, NY, United States
| | - Gina Kim
- Department of Surgery, Montefiore Medical Center, Albert Einstein College of Medicine, New York City, NY, United States
| | - Panagiota S. Filippou
- School of Health and Life Sciences, Teesside University, Middlesbrough, United Kingdom
- National Horizons Centre, Teesside University, Darlington, United Kingdom
| | - Luis Rivera Sanchez
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, New York City, NY, United States
| | - David Entenberg
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, New York City, NY, United States
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, New York City, NY, United States
- Integrated Imaging Program, Albert Einstein College of Medicine, New York City, NY, United States
| | - Douglas K. Marks
- Department of Medicine, NYU Long Island School of Medicine, Mineola, NY, United States
| | - Maja H. Oktay
- Department of Pathology, Montefiore Medical Center, Albert Einstein College of Medicine, New York City, NY, United States
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, New York City, NY, United States
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, New York City, NY, United States
- Integrated Imaging Program, Albert Einstein College of Medicine, New York City, NY, United States
| | - George S. Karagiannis
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, New York City, NY, United States
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, New York City, NY, United States
- Integrated Imaging Program, Albert Einstein College of Medicine, New York City, NY, United States
| |
Collapse
|
9
|
Girolamo F, de Trizio I, Errede M, Longo G, d'Amati A, Virgintino D. Neural crest cell-derived pericytes act as pro-angiogenic cells in human neocortex development and gliomas. Fluids Barriers CNS 2021; 18:14. [PMID: 33743764 PMCID: PMC7980348 DOI: 10.1186/s12987-021-00242-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 02/13/2021] [Indexed: 02/07/2023] Open
Abstract
Central nervous system diseases involving the parenchymal microvessels are frequently associated with a ‘microvasculopathy’, which includes different levels of neurovascular unit (NVU) dysfunction, including blood–brain barrier alterations. To contribute to the understanding of NVU responses to pathological noxae, we have focused on one of its cellular components, the microvascular pericytes, highlighting unique features of brain pericytes with the aid of the analyses carried out during vascularization of human developing neocortex and in human gliomas. Thanks to their position, centred within the endothelial/glial partition of the vessel basal lamina and therefore inserted between endothelial cells and the perivascular and vessel-associated components (astrocytes, oligodendrocyte precursor cells (OPCs)/NG2-glia, microglia, macrophages, nerve terminals), pericytes fulfil a central role within the microvessel NVU. Indeed, at this critical site, pericytes have a number of direct and extracellular matrix molecule- and soluble factor-mediated functions, displaying marked phenotypical and functional heterogeneity and carrying out multitasking services. This pericytes heterogeneity is primarily linked to their position in specific tissue and organ microenvironments and, most importantly, to their ontogeny. During ontogenesis, pericyte subtypes belong to two main embryonic germ layers, mesoderm and (neuro)ectoderm, and are therefore expected to be found in organs ontogenetically different, nonetheless, pericytes of different origin may converge and colonize neighbouring areas of the same organ/apparatus. Here, we provide a brief overview of the unusual roles played by forebrain pericytes in the processes of angiogenesis and barriergenesis by virtue of their origin from midbrain neural crest stem cells. A better knowledge of the ontogenetic subpopulations may support the understanding of specific interactions and mechanisms involved in pericyte function/dysfunction, including normal and pathological angiogenesis, thereby offering an alternative perspective on cell subtype-specific therapeutic approaches. ![]()
Collapse
Affiliation(s)
- Francesco Girolamo
- Department of Basic Medical Sciences, Neuroscience and Sensory Organs, Human Anatomy and Histology Unit, University of Bari School of Medicine, Bari, Italy.
| | - Ignazio de Trizio
- Department of Basic Medical Sciences, Neuroscience and Sensory Organs, Human Anatomy and Histology Unit, University of Bari School of Medicine, Bari, Italy.,Intensive Care Unit, Department of Intensive Care, Regional Hospital of Lugano, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Mariella Errede
- Department of Basic Medical Sciences, Neuroscience and Sensory Organs, Human Anatomy and Histology Unit, University of Bari School of Medicine, Bari, Italy
| | - Giovanna Longo
- Department of Basic Medical Sciences, Neuroscience and Sensory Organs, Molecular Biology Unit, University of Bari School of Medicine, Bari, Italy
| | - Antonio d'Amati
- Department of Basic Medical Sciences, Neuroscience and Sensory Organs, Human Anatomy and Histology Unit, University of Bari School of Medicine, Bari, Italy.,Department of Emergency and Organ Transplantation, Pathology Section, University of Bari School of Medicine, Bari, Italy
| | - Daniela Virgintino
- Department of Basic Medical Sciences, Neuroscience and Sensory Organs, Human Anatomy and Histology Unit, University of Bari School of Medicine, Bari, Italy
| |
Collapse
|
10
|
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
|
11
|
Liu Y, Hammel G, Shi M, Cheng Z, Zivkovic S, Wang X, Xu P, He X, Guo B, Ren Y, Zuo L. Myelin Debris Stimulates NG2/CSPG4 Expression in Bone Marrow-Derived Macrophages in the Injured Spinal Cord. Front Cell Neurosci 2021; 15:651827. [PMID: 33815067 PMCID: PMC8017290 DOI: 10.3389/fncel.2021.651827] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 03/02/2021] [Indexed: 12/20/2022] Open
Abstract
Although the increased expression of members of the chondroitin sulfate proteoglycan family, such as neuron-glial antigen 2 (NG2), have been well documented after an injury to the spinal cord, a complete picture as to the cellular origins and function of this NG2 expression has yet to be made. Using a spinal cord injury (SCI) mouse model, we describe that some infiltrated bone marrow-derived macrophages (BMDMΦ) are early contributors to NG2/CSPG4 expression and secretion after SCI. We demonstrate for the first time that a lesion-related form of cellular debris generated from damaged myelin sheaths can increase NG2/CSPG4 expression in BMDMΦ, which then exhibit enhanced proliferation and decreased phagocytic capacity. These results suggest that BMDMΦ may play a much more nuanced role in secondary spinal cord injury than previously thought, including acting as early contributors to the NG2 component of the glial scar.
Collapse
Affiliation(s)
- Yang Liu
- Department of Immunology, Guizhou Medical University, Guiyang, China.,Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL, United States.,Department of Anesthesiology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Grace Hammel
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL, United States
| | - Minjun Shi
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL, United States.,Department of Pathology, Guizhou Medical University, Guiyang, China
| | - Zhijian Cheng
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL, United States.,Department of Orthopedics, The Second Affiliated Hospital of Xian Jiaotong University, Xian, China
| | - Sandra Zivkovic
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL, United States
| | - Xiaoqi Wang
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL, United States
| | - Pingyi Xu
- Department of Neurology, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xijing He
- Department of Orthopedics, The Second Affiliated Hospital of Xian Jiaotong University, Xian, China
| | - Bing Guo
- Department of Pathology, Guizhou Medical University, Guiyang, China
| | - Yi Ren
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL, United States
| | - Li Zuo
- Department of Immunology, Guizhou Medical University, Guiyang, China
| |
Collapse
|
12
|
Uusitalo-Kylmälä L, Santo Mendes AC, Polari L, Joensuu K, Heino TJ. An In Vitro Co-Culture Model of Bone Marrow Mesenchymal Stromal Cells and Peripheral Blood Mononuclear Cells Promotes the Differentiation of Myeloid Angiogenic Cells and Pericyte-Like Cells. Stem Cells Dev 2021; 30:309-324. [PMID: 33499756 DOI: 10.1089/scd.2019.0171] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) are known to stimulate the survival and growth of endothelial cells (ECs) by producing paracrine signals, as well as to differentiate into pericytes and thereby support blood vessel formation and stability. On the other hand, cells with an EC-like phenotype have been found within the CD14+ and CD34+ cell populations of peripheral blood (PB) mononuclear cells (MNCs). The aim of this study was to investigate the proangiogenic differentiation potential of human MSC-MNC co-cultures. Bone marrow-derived MSCs (2,500 cells/cm2) were co-cultured with MNCs (50,000 cells/cm2), which were isolated from the PB of healthy donors. MSCs and MNCs cultured alone at same cell densities were used as controls. Cells in MNC fraction and in co-cultures were isolated for CD14, CD34, and CD31 surface markers with magnetic-activated cell sorting. Co-cultures were analyzed for cell proliferation and morphology, as well as for the expression of various hematopoietic, endothelial, and pericyte markers by immunocytochemistry, quantitative PCR (qPCR), and flow cytometry. Vascular endothelial growth factor (VEGF) expression and secretion was measured with qPCR and enzyme-linked immunosorbent assay, respectively. Our results show that in co-cultures with MSCs, CD14+CD45+ MNCs differentiated into spindle-shaped, nonproliferative, EC-like, myeloid angiogenic cells (MACs) expressing CD31, but also into pericyte-like cells expressing neural/glial antigen 2 (NG2) and CD146. Functionality of the isolated MACs was demonstrated in co-cultures with human umbilical vein endothelial cells, where they supported the formation of tube-like structures. NG2+ cells of MNC-origin were found among both CD34-CD14+ and CD34-CD14- cell populations, indicating the existence of different subtypes of pericyte-like cells. In addition, VEGF was shown to be secreted in MSC-MNC co-cultures, mainly by MSCs. In conclusion, MSCs were shown to possess proangiogenic capacity in MSC-MNC co-cultures as they supported the differentiation of functional MACs, as well as the differentiation of pericyte-like cells of MNC origin. This phenomenon was mediated at least partially via secreted VEGF.
Collapse
Affiliation(s)
| | - Ana Carolina Santo Mendes
- Institute of Biomedicine, University of Turku, Turku, Finland.,Department of Life Sciences, Faculty of Science and Technology, University of Coimbra, Coimbra, Portugal
| | - Lauri Polari
- Department of Biosciences, Cell Biology, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
| | - Katriina Joensuu
- Institute of Biomedicine, University of Turku, Turku, Finland.,Department of Plastic Surgery, Tampere University Hospital, Tampere, Finland
| | - Terhi J Heino
- Institute of Biomedicine, University of Turku, Turku, Finland
| |
Collapse
|
13
|
Schmitt BM, Boewe AS, Becker V, Nalbach L, Gu Y, Götz C, Menger MD, Laschke MW, Ampofo E. Protein Kinase CK2 Regulates Nerve/Glial Antigen (NG)2-Mediated Angiogenic Activity of Human Pericytes. Cells 2020; 9:cells9061546. [PMID: 32630438 PMCID: PMC7348826 DOI: 10.3390/cells9061546] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 06/15/2020] [Accepted: 06/23/2020] [Indexed: 12/14/2022] Open
Abstract
Protein kinase CK2 is a crucial regulator of endothelial cell proliferation, migration and sprouting during angiogenesis. However, it is still unknown whether this kinase additionally affects the angiogenic activity of other vessel-associated cells. In this study, we investigated the effect of CK2 inhibition on primary human pericytes. We found that CK2 inhibition reduces the expression of nerve/glial antigen (NG)2, a crucial factor which is involved in angiogenic processes. Reporter gene assays revealed a 114 bp transcriptional active region of the human NG2 promoter, whose activity was decreased after CK2 inhibition. Functional analyses demonstrated that the pharmacological inhibition of CK2 by CX-4945 suppresses pericyte proliferation, migration, spheroid sprouting and the stabilization of endothelial tubes. Moreover, aortic rings of NG2−/− mice showed a significantly reduced vascular sprouting when compared to rings of NG2+/+ mice, indicating that NG2 is an important regulator of the angiogenic activity of pericytes. In vivo, implanted Matrigel plugs containing CX-4945-treated pericytes exhibited a lower microvessel density when compared to controls. These findings demonstrate that CK2 regulates the angiogenic activity of pericytes through NG2 gene expression. Hence, the inhibition of CK2 represents a promising anti-angiogenic strategy, because it does not only target endothelial cells, but also vessel-associated pericytes.
Collapse
Affiliation(s)
- Beate M. Schmitt
- Institute for Clinical & Experimental Surgery, Saarland University, 66421 Homburg, Germany; (B.M.S.); (A.S.B.); (V.B.); (L.N.); (Y.G.); (M.D.M.); (M.W.L.)
| | - Anne S. Boewe
- Institute for Clinical & Experimental Surgery, Saarland University, 66421 Homburg, Germany; (B.M.S.); (A.S.B.); (V.B.); (L.N.); (Y.G.); (M.D.M.); (M.W.L.)
| | - Vivien Becker
- Institute for Clinical & Experimental Surgery, Saarland University, 66421 Homburg, Germany; (B.M.S.); (A.S.B.); (V.B.); (L.N.); (Y.G.); (M.D.M.); (M.W.L.)
| | - Lisa Nalbach
- Institute for Clinical & Experimental Surgery, Saarland University, 66421 Homburg, Germany; (B.M.S.); (A.S.B.); (V.B.); (L.N.); (Y.G.); (M.D.M.); (M.W.L.)
| | - Yuan Gu
- Institute for Clinical & Experimental Surgery, Saarland University, 66421 Homburg, Germany; (B.M.S.); (A.S.B.); (V.B.); (L.N.); (Y.G.); (M.D.M.); (M.W.L.)
| | - Claudia Götz
- Medical Biochemistry and Molecular Biology, Saarland University, 66421 Homburg, Germany;
| | - Michael D. Menger
- Institute for Clinical & Experimental Surgery, Saarland University, 66421 Homburg, Germany; (B.M.S.); (A.S.B.); (V.B.); (L.N.); (Y.G.); (M.D.M.); (M.W.L.)
| | - Matthias W. Laschke
- Institute for Clinical & Experimental Surgery, Saarland University, 66421 Homburg, Germany; (B.M.S.); (A.S.B.); (V.B.); (L.N.); (Y.G.); (M.D.M.); (M.W.L.)
| | - Emmanuel Ampofo
- Institute for Clinical & Experimental Surgery, Saarland University, 66421 Homburg, Germany; (B.M.S.); (A.S.B.); (V.B.); (L.N.); (Y.G.); (M.D.M.); (M.W.L.)
- Correspondence: ; Tel.: +49-6841-16-26561; Fax: +49-6841-16-26553
| |
Collapse
|
14
|
Mellai M, Annovazzi L, Bisogno I, Corona C, Crociara P, Iulini B, Cassoni P, Casalone C, Boldorini R, Schiffer D. Chondroitin Sulphate Proteoglycan 4 (NG2/CSPG4) Localization in Low- and High-Grade Gliomas. Cells 2020; 9:E1538. [PMID: 32599896 PMCID: PMC7349878 DOI: 10.3390/cells9061538] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 06/05/2020] [Accepted: 06/16/2020] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Neuron glial antigen 2 or chondroitin sulphate proteoglycan 4 (NG2/CSPG4) is expressed by immature precursors/progenitor cells and is possibly involved in malignant cell transformation. The aim of this study was to investigate its role on the progression and survival of sixty-one adult gliomas and nine glioblastoma (GB)-derived cell lines. METHODS NG2/CSPG4 protein expression was assessed by immunohistochemistry and immunofluorescence. Genetic and epigenetic alterations were detected by molecular genetic techniques. RESULTS NG2/CSPG4 was frequently expressed in IDH-mutant/1p19q-codel oligodendrogliomas (59.1%) and IDH-wild type GBs (40%) and rarely expressed in IDH-mutant or IDH-wild type astrocytomas (14.3%). Besides tumor cells, NG2/CSPG4 immunoreactivity was found in the cytoplasm and/or cell membranes of reactive astrocytes and vascular pericytes/endothelial cells. In GB-derived neurospheres, it was variably detected according to the number of passages of the in vitro culture. In GB-derived adherent cells, a diffuse positivity was found in most cells. NG2/CSPG4 expression was significantly associated with EGFR gene amplification (p = 0.0005) and poor prognosis (p = 0.016) in astrocytic tumors. CONCLUSION The immunoreactivity of NG2/CSPG4 provides information on the timing of the neoplastic transformation and could have prognostic and therapeutic relevance as a promising tumor-associated antigen for antibody-based immunotherapy in patients with malignant gliomas.
Collapse
Affiliation(s)
- Marta Mellai
- Dipartimento di Scienze della Salute, Scuola di Medicina, Università del Piemonte Orientale (UPO), Via Solaroli 17, 28100 Novara, Italy; (M.M.); (R.B.)
- Centro Interdipartimentale di Ricerca Traslazionale sulle Malattie Autoimmuni e Allergiche (CAAD), Università del Piemonte Orientale (UPO), Corso Trieste 15A, 28100 Novara, Italy
- Fondazione Edo ed Elvo Tempia Valenta—ONLUS, Via Malta 3, 13900 Biella, Italy
| | - Laura Annovazzi
- Ex Centro Ricerche/Fondazione Policlinico di Monza, Via P. Micca 29, 13100 Vercelli, Italy; (L.A.); (I.B.); (D.S.)
| | - Ilaria Bisogno
- Ex Centro Ricerche/Fondazione Policlinico di Monza, Via P. Micca 29, 13100 Vercelli, Italy; (L.A.); (I.B.); (D.S.)
| | - Cristiano Corona
- Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle d’Aosta, Via Bologna 148, 10154 Torino, Italy; (C.C.); (P.C.); (B.I.)
| | - Paola Crociara
- Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle d’Aosta, Via Bologna 148, 10154 Torino, Italy; (C.C.); (P.C.); (B.I.)
| | - Barbara Iulini
- Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle d’Aosta, Via Bologna 148, 10154 Torino, Italy; (C.C.); (P.C.); (B.I.)
| | - Paola Cassoni
- Dipartimento di Scienze Mediche, Università di Torino/Città della Salute e della Scienza, Via Santena 7, 10126 Torino, Italy;
| | - Cristina Casalone
- Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle d’Aosta, Via Bologna 148, 10154 Torino, Italy; (C.C.); (P.C.); (B.I.)
| | - Renzo Boldorini
- Dipartimento di Scienze della Salute, Scuola di Medicina, Università del Piemonte Orientale (UPO), Via Solaroli 17, 28100 Novara, Italy; (M.M.); (R.B.)
| | - Davide Schiffer
- Ex Centro Ricerche/Fondazione Policlinico di Monza, Via P. Micca 29, 13100 Vercelli, Italy; (L.A.); (I.B.); (D.S.)
| |
Collapse
|
15
|
Finocchiaro G, Pellegatta S. NG2/CSPG4 in glioblastoma: about flexibility. Neuro Oncol 2020; 21:697-698. [PMID: 30918960 DOI: 10.1093/neuonc/noz055] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Affiliation(s)
- Gaetano Finocchiaro
- Unit of Molecular Neuro-Oncology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Serena Pellegatta
- Unit of Molecular Neuro-Oncology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| |
Collapse
|
16
|
Yuan K, Shamskhou EA, Orcholski ME, Nathan A, Reddy S, Honda H, Mani V, Zeng Y, Ozen MO, Wang L, Demirci U, Tian W, Nicolls MR, de Jesus Perez VA. Loss of Endothelium-Derived Wnt5a Is Associated With Reduced Pericyte Recruitment and Small Vessel Loss in Pulmonary Arterial Hypertension. Circulation 2020; 139:1710-1724. [PMID: 30586764 DOI: 10.1161/circulationaha.118.037642] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Pulmonary arterial hypertension (PAH) is a life-threatening disorder of the pulmonary circulation associated with loss and impaired regeneration of microvessels. Reduced pericyte coverage of pulmonary microvessels is a pathological feature of PAH and is caused partly by the inability of pericytes to respond to signaling cues from neighboring pulmonary microvascular endothelial cells (PMVECs). We have shown that activation of the Wnt/planar cell polarity pathway is required for pericyte recruitment, but whether production and release of specific Wnt ligands by PMVECs are responsible for Wnt/planar cell polarity activation in pericytes is unknown. METHODS Isolation of pericytes and PMVECs from healthy donor and PAH lungs was carried out with 3G5 or CD31 antibody-conjugated magnetic beads. Wnt expression profile of PMVECs was documented via quantitative polymerase chain reaction with a Wnt primer library. Exosome purification from PMVEC media was carried out with the ExoTIC device. Hemodynamic profile, right ventricular function, and pulmonary vascular morphometry were obtained in a conditional endothelium-specific Wnt5a knockout ( Wnt5aECKO) mouse model under normoxia, chronic hypoxia, and hypoxia recovery. RESULTS Quantification of Wnt ligand expression in healthy PMVECs cocultured with pericytes demonstrated a 35-fold increase in Wnt5a, a known Wnt/planar cell polarity ligand. This Wnt5a spike was not seen in PAH PMVECs, which correlated with an inability to recruit pericytes in Matrigel coculture assays. Exosomes purified from media demonstrated an increase in Wnt5a content when healthy PMVECs were cocultured with pericytes, a finding that was not observed in exosomes of PAH PMVECs. Furthermore, the addition of either recombinant Wnt5a or purified healthy PMVEC exosomes increased pericyte recruitment to PAH PMVECs in coculture studies. Although no differences were noted in normoxia and chronic hypoxia, Wnt5aECKO mice demonstrated persistent pulmonary hypertension and right ventricular failure 4 weeks after recovery from chronic hypoxia, which correlated with significant reduction, muscularization, and decreased pericyte coverage of microvessels. CONCLUSIONS We identify Wnt5a as a key mediator for the establishment of pulmonary endothelium-pericyte interactions, and its loss could contribute to PAH by reducing the viability of newly formed vessels. We speculate that therapies that mimic or restore Wnt5a production could help prevent loss of small vessels in PAH.
Collapse
Affiliation(s)
- Ke Yuan
- Division of Pulmonary and Critical Care Medicine (K.Y., E.A.S., M.E.O., A.N., V.A.d.J.P.), Stanford University, Palo Alto, CA.,Stanford Cardiovascular Institute (K.Y., E.A.S., M.E.O., A.N., S.R., M.O.O, L.W., W.T., M.R.N., V.A.d.J.P.), Stanford University, Palo Alto, CA.,Wall Center for Pulmonary Vascular Research (K.Y., E.A.S., M.E.O., A.N., L.W., W.T., M.R.N., V.A.d.J.P.), Stanford University, Palo Alto, CA
| | - Elya A Shamskhou
- Division of Pulmonary and Critical Care Medicine (K.Y., E.A.S., M.E.O., A.N., V.A.d.J.P.), Stanford University, Palo Alto, CA.,Stanford Cardiovascular Institute (K.Y., E.A.S., M.E.O., A.N., S.R., M.O.O, L.W., W.T., M.R.N., V.A.d.J.P.), Stanford University, Palo Alto, CA.,Wall Center for Pulmonary Vascular Research (K.Y., E.A.S., M.E.O., A.N., L.W., W.T., M.R.N., V.A.d.J.P.), Stanford University, Palo Alto, CA
| | - Mark E Orcholski
- Division of Pulmonary and Critical Care Medicine (K.Y., E.A.S., M.E.O., A.N., V.A.d.J.P.), Stanford University, Palo Alto, CA.,Stanford Cardiovascular Institute (K.Y., E.A.S., M.E.O., A.N., S.R., M.O.O, L.W., W.T., M.R.N., V.A.d.J.P.), Stanford University, Palo Alto, CA.,Wall Center for Pulmonary Vascular Research (K.Y., E.A.S., M.E.O., A.N., L.W., W.T., M.R.N., V.A.d.J.P.), Stanford University, Palo Alto, CA
| | - Abinaya Nathan
- Division of Pulmonary and Critical Care Medicine (K.Y., E.A.S., M.E.O., A.N., V.A.d.J.P.), Stanford University, Palo Alto, CA.,Stanford Cardiovascular Institute (K.Y., E.A.S., M.E.O., A.N., S.R., M.O.O, L.W., W.T., M.R.N., V.A.d.J.P.), Stanford University, Palo Alto, CA.,Wall Center for Pulmonary Vascular Research (K.Y., E.A.S., M.E.O., A.N., L.W., W.T., M.R.N., V.A.d.J.P.), Stanford University, Palo Alto, CA
| | - Sushma Reddy
- Stanford Cardiovascular Institute (K.Y., E.A.S., M.E.O., A.N., S.R., M.O.O, L.W., W.T., M.R.N., V.A.d.J.P.), Stanford University, Palo Alto, CA.,Division of Pediatric Cardiology (S.R.), Stanford University, Palo Alto, CA
| | - Hiroaki Honda
- Field of Human Disease Models, Major in Advanced Life Sciences and Medicine, Institute of Laboratory Animals, Tokyo Women's Medical University, Japan (H.H.)
| | - Vigneshwaran Mani
- Department of Radiology, Canary Center for Early Cancer Detection (V.M., M.O.O., U.D.), Stanford University, Palo Alto, CA
| | - Yitian Zeng
- Department of Materials Science and Engineering (Y.Z.), Stanford University, Palo Alto, CA
| | - Mehmet O Ozen
- Stanford Cardiovascular Institute (K.Y., E.A.S., M.E.O., A.N., S.R., M.O.O, L.W., W.T., M.R.N., V.A.d.J.P.), Stanford University, Palo Alto, CA.,Department of Radiology, Canary Center for Early Cancer Detection (V.M., M.O.O., U.D.), Stanford University, Palo Alto, CA
| | - Lingli Wang
- Stanford Cardiovascular Institute (K.Y., E.A.S., M.E.O., A.N., S.R., M.O.O, L.W., W.T., M.R.N., V.A.d.J.P.), Stanford University, Palo Alto, CA.,Wall Center for Pulmonary Vascular Research (K.Y., E.A.S., M.E.O., A.N., L.W., W.T., M.R.N., V.A.d.J.P.), Stanford University, Palo Alto, CA.,Department of Pediatrics (L.W.), Stanford University, Palo Alto, CA
| | - Utkan Demirci
- Department of Radiology, Canary Center for Early Cancer Detection (V.M., M.O.O., U.D.), Stanford University, Palo Alto, CA
| | - Wen Tian
- Stanford Cardiovascular Institute (K.Y., E.A.S., M.E.O., A.N., S.R., M.O.O, L.W., W.T., M.R.N., V.A.d.J.P.), Stanford University, Palo Alto, CA.,Wall Center for Pulmonary Vascular Research (K.Y., E.A.S., M.E.O., A.N., L.W., W.T., M.R.N., V.A.d.J.P.), Stanford University, Palo Alto, CA.,Department of Medicine, VA Palo Alto Health Care System/Stanford University, CA (W.T., M.R.N.)
| | - Mark R Nicolls
- Stanford Cardiovascular Institute (K.Y., E.A.S., M.E.O., A.N., S.R., M.O.O, L.W., W.T., M.R.N., V.A.d.J.P.), Stanford University, Palo Alto, CA.,Wall Center for Pulmonary Vascular Research (K.Y., E.A.S., M.E.O., A.N., L.W., W.T., M.R.N., V.A.d.J.P.), Stanford University, Palo Alto, CA.,Department of Medicine, VA Palo Alto Health Care System/Stanford University, CA (W.T., M.R.N.)
| | - Vinicio A de Jesus Perez
- Division of Pulmonary and Critical Care Medicine (K.Y., E.A.S., M.E.O., A.N., V.A.d.J.P.), Stanford University, Palo Alto, CA.,Stanford Cardiovascular Institute (K.Y., E.A.S., M.E.O., A.N., S.R., M.O.O, L.W., W.T., M.R.N., V.A.d.J.P.), Stanford University, Palo Alto, CA.,Wall Center for Pulmonary Vascular Research (K.Y., E.A.S., M.E.O., A.N., L.W., W.T., M.R.N., V.A.d.J.P.), Stanford University, Palo Alto, CA
| |
Collapse
|
17
|
Chondroitin Sulphate Proteoglycans in the Tumour Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1272:73-92. [PMID: 32845503 DOI: 10.1007/978-3-030-48457-6_5] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Proteoglycans are macromolecules that are essential for the development of cells, human diseases and malignancies. In particular, chondroitin sulphate proteoglycans (CSPGs) accumulate in tumour stroma and play a key role in tumour growth and invasion by driving multiple oncogenic pathways in tumour cells and promoting crucial interactions in the tumour microenvironment (TME). These pathways involve receptor tyrosine kinase (RTK) signalling via the mitogen-activated protein kinase (MAPK) cascade and integrin signalling via the activation of focal adhesion kinase (FAK), which sustains the activation of extracellular signal-regulated kinases 1/2 (ERK1/2).Human CSPG4 is a type I transmembrane protein that is associated with the growth and progression of human brain tumours. It regulates cell signalling and migration by interacting with components of the extracellular matrix, extracellular ligands, growth factor receptors, intracellular enzymes and structural proteins. Its overexpression by tumour cells, perivascular cells and precursor/progenitor cells in gliomas suggests that it plays a role in their origin, progression and neo-angiogenesis and its aberrant expression in tumour cells may be a promising biomarker to monitor malignant progression and patient survival.The aim of this chapter is to review and discuss the role of CSPG4 in the TME of human gliomas, including its potential as a druggable therapeutic target.
Collapse
|
18
|
Liu Q, Yang Y, Fan X. Microvascular pericytes in brain-associated vascular disease. Biomed Pharmacother 2020; 121:109633. [DOI: 10.1016/j.biopha.2019.109633] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 10/31/2019] [Accepted: 11/01/2019] [Indexed: 01/01/2023] Open
|
19
|
Boshans LL, Factor DC, Singh V, Liu J, Zhao C, Mandoiu I, Lu QR, Casaccia P, Tesar PJ, Nishiyama A. The Chromatin Environment Around Interneuron Genes in Oligodendrocyte Precursor Cells and Their Potential for Interneuron Reprograming. Front Neurosci 2019; 13:829. [PMID: 31440130 PMCID: PMC6694778 DOI: 10.3389/fnins.2019.00829] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Accepted: 07/25/2019] [Indexed: 12/13/2022] Open
Abstract
Oligodendrocyte precursor cells (OPCs), also known as NG2 glia, arise from neural progenitor cells in the embryonic ganglionic eminences that also generate inhibitory neurons. They are ubiquitously distributed in the central nervous system, remain proliferative through life, and generate oligodendrocytes in both gray and white matter. OPCs exhibit some lineage plasticity, and attempts have been made to reprogram them into neurons, with varying degrees of success. However, little is known about how epigenetic mechanisms affect the ability of OPCs to undergo fate switch and whether OPCs have a unique chromatin environment around neuronal genes that might contribute to their lineage plasticity. Our bioinformatic analysis of histone posttranslational modifications at interneuron genes in OPCs revealed that OPCs had significantly fewer bivalent and repressive histone marks at interneuron genes compared to astrocytes or fibroblasts. Conversely, OPCs had a greater degree of deposition of active histone modifications at bivalently marked interneuron genes than other cell types, and this was correlated with higher expression levels of these genes in OPCs. Furthermore, a significantly higher proportion of interneuron genes in OPCs than in other cell types lacked the histone posttranslational modifications examined. These genes had a moderately high level of expression, suggesting that the "no mark" interneuron genes could be in a transcriptionally "poised" or "transitional" state. Thus, our findings suggest that OPCs have a unique histone code at their interneuron genes that may obviate the need for erasure of repressive marks during their fate switch to inhibitory neurons.
Collapse
Affiliation(s)
- Linda L. Boshans
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, United States
- Connecticut Institute for Brain and Cognitive Sciences, University of Connecticut, Storrs, CT, United States
| | - Daniel C. Factor
- Department of Genetics and Genome Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Vijender Singh
- Computational Biology Core, University of Connecticut, Storrs, CT, United States
| | - Jia Liu
- Advanced Science Research Center at the Graduate Center, Neuroscience Initiative, The City University of New York, New York, NY, United States
| | - Chuntao Zhao
- Department of Pediatrics, University of Cincinnati, Cincinnati, OH, United States
| | - Ion Mandoiu
- Department of Computer Science and Engineering, University of Connecticut, Storrs, CT, United States
| | - Q. Richard Lu
- Department of Pediatrics, University of Cincinnati, Cincinnati, OH, United States
| | - Patrizia Casaccia
- Advanced Science Research Center at the Graduate Center, Neuroscience Initiative, The City University of New York, New York, NY, United States
| | - Paul J. Tesar
- Department of Genetics and Genome Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Akiko Nishiyama
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, United States
- Connecticut Institute for Brain and Cognitive Sciences, University of Connecticut, Storrs, CT, United States
- Institute for Systems Genomics, University of Connecticut, Storrs, CT, United States
| |
Collapse
|
20
|
Abstract
Besides seminal functions in angiogenesis and blood pressure regulation, microvascular pericytes possess a latent tissue regenerative potential that can be revealed in culture following transition into mesenchymal stem cells. Endowed with robust osteogenic potential, pericytes and other related perivascular cells extracted from adipose tissue represent a potent and abundant cell source for refined bone tissue engineering and improved cell therapies of fractures and other bone defects. The use of diverse bone formation assays in vivo, which include mouse muscle pocket osteogenesis and calvaria replenishment, rat and dog spine fusion, and rat non-union fracture healing, has confirmed the superiority of purified perivascular cells for skeletal (re)generation. As a surprising observation though, despite strong endogenous bone-forming potential, perivascular cells drive bone regeneration essentially indirectly, via recruitment by secreted factors of local osteo-progenitors.
Collapse
|
21
|
Mandoj C, Tomao L, Conti L. Coagulation in Brain Tumors: Biological Basis and Clinical Implications. Front Neurol 2019; 10:181. [PMID: 30949114 PMCID: PMC6436068 DOI: 10.3389/fneur.2019.00181] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 02/13/2019] [Indexed: 12/31/2022] Open
Abstract
Cancer patients commonly show abnormal laboratory coagulation tests, indicating a subclinical hypercoagulable condition that contribute to morbidity and mortality. The hypercoagulation status not only increases the risk of thromboembolic events but also influences the tumor biology promoting its growth and progression by stimulating intracellular signaling pathways. Recent molecular studies characterized the role of oncogene and suppressor gene in activating clotting pathways, as an integral feature of the neoplastic transformation. It is now clear how haemostatic processes, activated by cancer cells harboring oncogenic mutations, rely on the molecular profile of a particular malignancy, an aspect particularly evident in the differential coagulome profiles showed by different molecular subtypes of brain tumors, such as glioblastoma and medulloblastoma. This review focuses on the biological and clinical aspects of haemostasis in cancer with particular regard on brain tumors.
Collapse
Affiliation(s)
- Chiara Mandoj
- Clinical Pathology, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Luigi Tomao
- Department of Hematology/Oncology, IRCCS Bambino Gesù Children Hospital, Rome, Italy
| | - Laura Conti
- Clinical Pathology, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| |
Collapse
|
22
|
Schiffer D, Annovazzi L, Casalone C, Corona C, Mellai M. Glioblastoma: Microenvironment and Niche Concept. Cancers (Basel) 2018; 11:cancers11010005. [PMID: 30577488 PMCID: PMC6357107 DOI: 10.3390/cancers11010005] [Citation(s) in RCA: 141] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 12/07/2018] [Accepted: 12/07/2018] [Indexed: 01/11/2023] Open
Abstract
The niche concept was originally developed to describe the location of normal neural stem cells (NSCs) in the subependymal layer of the sub-ventricular zone. In this paper, its significance has been extended to the location of tumor stem cells in glioblastoma (GB) to discuss the relationship between GB stem cells (GSCs) and endothelial cells (ECs). Their interaction is basically conceived as responsible for tumor growth, invasion and recurrence. Niches are described as the points of utmost expression of the tumor microenvironment (TME), therefore including everything in the tumor except for tumor cells: NSCs, reactive astrocytes, ECs, glioma-associated microglia/macrophages (GAMs), myeloid cells, pericytes, fibroblasts, etc. and all intrinsic and extrinsic signaling pathways. Perivascular (PVNs), perinecrotic (PNNs) and invasive niches were described from the pathological point of view, highlighting the basic significance of the EC/tumor stem cell couple. PNN development was reinterpreted based on the concept that hyperproliferative areas of GB are composed of GSCs/progenitors. TME was depicted in its function as the main regulator of everything that happens in the tumor. A particular emphasis was given to GAMs, pericytes and reactive astrocytes as important elements affecting proliferation, growth, invasion and resistance to therapies of tumor cells.
Collapse
Affiliation(s)
- Davide Schiffer
- Professore Emerito di Neurologia, Università di Torino, Corso Bramante 88/90, 10126 Torino, Italy.
| | - Laura Annovazzi
- Ex Centro Ricerche/Fondazione Policlinico di Monza, Via P. Micca 29, 13100 Vercelli, Italy.
| | - Cristina Casalone
- Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle d'Aosta, Via Bologna 148, 10154 Torino, Italy.
| | - Cristiano Corona
- Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle d'Aosta, Via Bologna 148, 10154 Torino, Italy.
| | - Marta Mellai
- Dipartimento di Scienze della Salute, Scuola di Medicina, Università del Piemonte Orientale "A. Avogadro", Corso Mazzini 18, 28100 Novara, Italy.
- Fondazione Edo ed Elvo Tempia Valenta-Onlus, Via Malta 3, 13900 Biella, Italy.
| |
Collapse
|
23
|
The Significance of Chondroitin Sulfate Proteoglycan 4 (CSPG4) in Human Gliomas. Int J Mol Sci 2018; 19:ijms19092724. [PMID: 30213051 PMCID: PMC6164575 DOI: 10.3390/ijms19092724] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 08/27/2018] [Accepted: 08/28/2018] [Indexed: 12/15/2022] Open
Abstract
Neuron glial antigen 2 (NG2) is a chondroitin sulphate proteoglycan 4 (CSPG4) that occurs in developing and adult central nervous systems (CNSs) as a marker of oligodendrocyte precursor cells (OPCs) together with platelet-derived growth factor receptor α (PDGFRα). It behaves variably in different pathological conditions, and is possibly involved in the origin and progression of human gliomas. In the latter, NG2/CSPG4 induces cell proliferation and migration, is highly expressed in pericytes, and plays a role in neoangiogenesis. NG2/CSPG4 expression has been demonstrated in oligodendrogliomas, astrocytomas, and glioblastomas (GB), and it correlates with malignancy. In rat tumors transplacentally induced by N-ethyl-N-nitrosourea (ENU), NG2/CSPG4 expression correlates with PDGFRα, Olig2, Sox10, and Nkx2.2, and with new vessel formation. In this review, we attempt to summarize the normal and pathogenic functions of NG2/CSPG4, as well as its potential as a therapeutic target.
Collapse
|
24
|
Bardelli D, Dander E, Bugarin C, Cappuzzello C, Pievani A, Fazio G, Pierani P, Corti P, Farruggia P, Dufour C, Cesaro S, Cipolli M, Biondi A, D'Amico G. Mesenchymal stromal cells from Shwachman-Diamond syndrome patients fail to recreate a bone marrow niche in vivo and exhibit impaired angiogenesis. Br J Haematol 2018; 182:114-124. [PMID: 29767474 DOI: 10.1111/bjh.15388] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 03/26/2018] [Indexed: 01/28/2023]
Abstract
Shwachman-Diamond syndrome (SDS) is a rare multi-organ recessive disease mainly characterised by pancreatic insufficiency, skeletal defects, short stature and bone marrow failure (BMF). As in many other BMF syndromes, SDS patients are predisposed to develop a number of haematopoietic malignancies, particularly myelodysplastic syndrome and acute myeloid leukaemia. However, the mechanism of cancer predisposition in SDS patients is only partially understood. In light of the emerging role of mesenchymal stromal cells (MSCs) in the regulation of bone marrow homeostasis, we assessed the ability of MSCs derived from SDS patients (SDS-MSCs) to recreate a functional bone marrow niche, taking advantage of a murine heterotopic MSC transplant model. We show that the ability of semi-cartilaginous pellets (SCPs) derived from SDS-MSCs to generate complete heterotopic ossicles in vivo is severely impaired in comparison with HD-MSC-derived SCPs. Specifically, after in vitro angiogenic stimuli, SDS-MSCs showed a defective ability to form correct networks, capillary tubes and vessels and displayed a marked decrease in VEGFA expression. Altogether, these findings unveil a novel mechanism of SDS-mediated haematopoietic dysfunction based on hampered ability of SDS-MSCs to support angiogenesis. Overall, MSCs could represent a new appealing therapeutic target to treat dysfunctional haematopoiesis in paediatric SDS patients.
Collapse
Affiliation(s)
- Donatella Bardelli
- Paediatric Department, Centro Ricerca Tettamanti, University of Milano-Bicocca, Fondazione MBBM, Monza, Italy
| | - Erica Dander
- Paediatric Department, Centro Ricerca Tettamanti, University of Milano-Bicocca, Fondazione MBBM, Monza, Italy
| | - Cristina Bugarin
- Paediatric Department, Centro Ricerca Tettamanti, University of Milano-Bicocca, Fondazione MBBM, Monza, Italy
| | - Claudia Cappuzzello
- Paediatric Department, Centro Ricerca Tettamanti, University of Milano-Bicocca, Fondazione MBBM, Monza, Italy
| | - Alice Pievani
- Department of Paediatrics, Dulbecco Telethon Institute, Centro Ricerca M. Tettamanti, University of Milano-Bicocca, Monza, Italy
| | - Grazia Fazio
- Paediatric Department, Centro Ricerca Tettamanti, University of Milano-Bicocca, Fondazione MBBM, Monza, Italy
| | - Paolo Pierani
- Department of Paediatric Haemato-Oncology, Ancona, Italy
| | - Paola Corti
- Department of Paediatrics, University of Milano-Bicocca, San Gerardo Hospital/Fondazione MBBM, Monza, Italy
| | - Piero Farruggia
- Department of Paediatric Haemato-Oncology, ARNAS Ospedali Civico, G Di Cristina, Palermo, Italy
| | - Carlo Dufour
- Haematology Unit, Giannina Gaslini Children's Research Hospital, Genoa, Italy
| | - Simone Cesaro
- Department of Paediatrics, Paediatric Haematology Oncology, Azienda Ospedaliera Universitaria Integrata, Verona, Italy
| | - Marco Cipolli
- Azienda Ospedaliero Universitaria Ospedali Riuniti Umberto I - G.M. Lancisi - G. Salesi, Cystic Fibrosis Centre, Ancona, Italy
| | - Andrea Biondi
- Paediatric Department, Centro Ricerca Tettamanti, University of Milano-Bicocca, Fondazione MBBM, Monza, Italy.,Department of Paediatrics, University of Milano-Bicocca, San Gerardo Hospital/Fondazione MBBM, Monza, Italy
| | - Giovanna D'Amico
- Paediatric Department, Centro Ricerca Tettamanti, University of Milano-Bicocca, Fondazione MBBM, Monza, Italy
| |
Collapse
|
25
|
Morphological characterization of NG2 glia and their association with neuroglial cells in the 3-nitropropionic acid-lesioned striatum of rat. Sci Rep 2018; 8:5942. [PMID: 29654253 PMCID: PMC5899159 DOI: 10.1038/s41598-018-24385-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 04/03/2018] [Indexed: 01/18/2023] Open
Abstract
Our aim was to examine the spatiotemporal profiles and phenotypic characteristics of neuron-glia antigen 2 (NG2) glia and their associations with neuroglial cells in striatal lesions due to the mitochondrial toxin 3-nitropropionic acid (3-NP). In control striatum, weak NG2 immunoreactivity was restricted to resting NG2 glia with thin processes, but prominent NG2 expression was noted on activated microglia/macrophages, and reactive NG2 glia in the lesion core after 3-NP injection. Activation of NG2 glia, including enhanced proliferation and morphological changes, had a close spatiotemporal relationship with infiltration of activated microglia into the lesion core. Thick and highly branched processes of reactive NG2 glia formed a cellular network in the astrocyte-free lesion core and primarily surrounded developing cavities 2–4 weeks post-lesion. NG2 glia became associated with astrocytes in the lesion core and the border of cavities over the chronic interval of 4–8 weeks. Immunoelectron microscopy indicated that reactive NG2 glia had large euchromatic nuclei with prominent nucleoli and thick and branched processes that ramified distally. Thus, our data provide detailed information regarding the morphologies of NG2 glia in the lesion core, and support the link between transformation of NG2 glia to the reactive form and microglial activation/recruitment in response to brain insults.
Collapse
|
26
|
Origin and development of septoclasts in endochondral ossification of mice. Histochem Cell Biol 2018; 149:645-654. [PMID: 29464321 DOI: 10.1007/s00418-018-1653-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/15/2018] [Indexed: 12/31/2022]
Abstract
Septoclasts are mononuclear spindle-shaped phagocytes with their long processes in uncalcified cartilage matrices and locate adjacent to the capillary endothelium at the chondro-osseous junction of the growth plate. We have previously revealed a selective expression of epidermal-type fatty acid-binding protein (E-FABP/FABP5) in septoclasts. Although, pericytes are known to distribute along capillaries and directly surround their endothelial cells in a situation similar to septoclasts, no clear evidence is available on the relationship between septoclasts and pericytes. We investigated the chronological localization and morphological change of septoclasts during development of the tibia of mice to clarify the development of septoclasts and the immune-localization of pericyte markers in septoclasts to clarify the origin of septoclasts. E-FABP-immunoreactive septoclasts emerged at the perichondrium in the middle of the cartilaginous templates of the tibia in prenatal development. Septoclasts migrated to the surface of the cartilage adjacent to invading blood vessels. Processes of septoclasts became longer and their apexes attached to Von Kossa-negative uncalcified matrices during the formation process of the primary ossification center. Not only platelet-derived growth factor receptor beta, but also neuron-glial antigen 2 was localized in septoclasts of mice from E15 (embryonic day 15) to P6w (postnatal 6 week). Our results suggest that septoclasts are originated from pericytes and involved in the blood vessel invasion during formation of the primary ossification center.
Collapse
|
27
|
Ilieva KM, Cheung A, Mele S, Chiaruttini G, Crescioli S, Griffin M, Nakamura M, Spicer JF, Tsoka S, Lacy KE, Tutt ANJ, Karagiannis SN. Chondroitin Sulfate Proteoglycan 4 and Its Potential As an Antibody Immunotherapy Target across Different Tumor Types. Front Immunol 2018; 8:1911. [PMID: 29375561 PMCID: PMC5767725 DOI: 10.3389/fimmu.2017.01911] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 12/14/2017] [Indexed: 12/18/2022] Open
Abstract
Overexpression of the chondroitin sulfate proteoglycan 4 (CSPG4) has been associated with the pathology of multiple types of such as melanoma, breast cancer, squamous cell carcinoma, mesothelioma, neuroblastoma, adult and pediatric sarcomas, and some hematological cancers. CSPG4 has been reported to exhibit a role in the growth and survival as well as in the spreading and metastasis of tumor cells. CSPG4 is overexpressed in several malignant diseases, while it is thought to have restricted and low expression in normal tissues. Thus, CSPG4 has become the target of numerous anticancer treatment approaches, including monoclonal antibody-based therapies. This study reviews key potential anti-CSPG4 antibody and immune-based therapies and examines their direct antiproliferative/metastatic and immune activating mechanisms of action.
Collapse
Affiliation(s)
- Kristina M Ilieva
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London & NIHR Biomedical Research Centre at Guy's and St. Thomas' Hospitals and King's College London, Guy's Hospital, London, United Kingdom.,Breast Cancer Now Research Unit, School of Cancer and Pharmaceutical Sciences, King's College London, Guy's Cancer Centre, London, United Kingdom
| | - Anthony Cheung
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London & NIHR Biomedical Research Centre at Guy's and St. Thomas' Hospitals and King's College London, Guy's Hospital, London, United Kingdom.,Breast Cancer Now Research Unit, School of Cancer and Pharmaceutical Sciences, King's College London, Guy's Cancer Centre, London, United Kingdom
| | - Silvia Mele
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London & NIHR Biomedical Research Centre at Guy's and St. Thomas' Hospitals and King's College London, Guy's Hospital, London, United Kingdom
| | - Giulia Chiaruttini
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London & NIHR Biomedical Research Centre at Guy's and St. Thomas' Hospitals and King's College London, Guy's Hospital, London, United Kingdom
| | - Silvia Crescioli
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London & NIHR Biomedical Research Centre at Guy's and St. Thomas' Hospitals and King's College London, Guy's Hospital, London, United Kingdom
| | - Merope Griffin
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London & NIHR Biomedical Research Centre at Guy's and St. Thomas' Hospitals and King's College London, Guy's Hospital, London, United Kingdom
| | - Mano Nakamura
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London & NIHR Biomedical Research Centre at Guy's and St. Thomas' Hospitals and King's College London, Guy's Hospital, London, United Kingdom.,Department of Informatics, Faculty of Natural and Mathematical Sciences, King's College London, London, United Kingdom
| | - James F Spicer
- School of Cancer and Pharmaceutical Sciences, King's College London, Guy's Cancer Centre, London, United Kingdom
| | - Sophia Tsoka
- Department of Informatics, Faculty of Natural and Mathematical Sciences, King's College London, London, United Kingdom
| | - Katie E Lacy
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London & NIHR Biomedical Research Centre at Guy's and St. Thomas' Hospitals and King's College London, Guy's Hospital, London, United Kingdom
| | - Andrew N J Tutt
- Breast Cancer Now Research Unit, School of Cancer and Pharmaceutical Sciences, King's College London, Guy's Cancer Centre, London, United Kingdom.,Breast Cancer Now Toby Robins Research Centre, Institute of Cancer Research, London, United Kingdom
| | - Sophia N Karagiannis
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, King's College London & NIHR Biomedical Research Centre at Guy's and St. Thomas' Hospitals and King's College London, Guy's Hospital, London, United Kingdom.,Breast Cancer Now Research Unit, School of Cancer and Pharmaceutical Sciences, King's College London, Guy's Cancer Centre, London, United Kingdom
| |
Collapse
|
28
|
Theocharis AD, Karamanos NK. Proteoglycans remodeling in cancer: Underlying molecular mechanisms. Matrix Biol 2017; 75-76:220-259. [PMID: 29128506 DOI: 10.1016/j.matbio.2017.10.008] [Citation(s) in RCA: 146] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 10/23/2017] [Accepted: 10/24/2017] [Indexed: 02/07/2023]
Abstract
Extracellular matrix is a highly dynamic macromolecular network. Proteoglycans are major components of extracellular matrix playing key roles in its structural organization and cell signaling contributing to the control of numerous normal and pathological processes. As multifunctional molecules, proteoglycans participate in various cell functions during morphogenesis, wound healing, inflammation and tumorigenesis. Their interactions with matrix effectors, cell surface receptors and enzymes enable them with unique properties. In malignancy, extensive remodeling of tumor stroma is associated with marked alterations in proteoglycans' expression and structural variability. Proteoglycans exert diverse functions in tumor stroma in a cell-specific and context-specific manner and they mainly contribute to the formation of a permissive provisional matrix for tumor growth affecting tissue organization, cell-cell and cell-matrix interactions and tumor cell signaling. Proteoglycans also modulate cancer cell phenotype and properties, the development of drug resistance and tumor stroma angiogenesis. This review summarizes the proteoglycans remodeling and their novel biological roles in malignancies with particular emphasis to the underlying molecular mechanisms.
Collapse
Affiliation(s)
- Achilleas D Theocharis
- Biochemistry, Biochemical Analysis & Matrix Pathobiochemistry Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26500 Patras, Greece.
| | - Nikos K Karamanos
- Biochemistry, Biochemical Analysis & Matrix Pathobiochemistry Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26500 Patras, Greece.
| |
Collapse
|
29
|
Distinct NG2 proteoglycan-dependent roles of resident microglia and bone marrow-derived macrophages during myelin damage and repair. PLoS One 2017; 12:e0187530. [PMID: 29095924 PMCID: PMC5667885 DOI: 10.1371/journal.pone.0187530] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 10/20/2017] [Indexed: 12/21/2022] Open
Abstract
We used a bone marrow transplantation approach to distinguish the activities of bone marrow-derived macrophages from the activities of central nervous system-resident microglia in phenomena associated with axon demyelination and remyelination. We transplanted wild type or germline NG2 null beta-actin-EGFP expressing bone marrow into irradiated wild type or NG2 null recipient mice, followed by analysis of lysolecithin-induced spinal cord demyelination and remyelination and quantification of Iba-1+/ F4/80+/ EGFP+ macrophages and Iba-1+/ F4/80+/ EGFP- microglia. One week after microinjection of 1% lysolecithin into the spinal cord, wild type recipients receiving NG2 null bone marrow exhibit greatly reduced infiltration of macrophages into lesions, compared to wild type recipients receiving wild type bone marrow. Wild type bone marrow recipients also exhibit larger numbers of demyelinated axons than NG2 null recipients, indicative of macrophage participation in the initial myelin damage. However, wild type bone marrow recipients also exhibit superior myelin repair at 6 weeks post-injury, compared to NG2 null bone marrow recipients, demonstrating the additional importance of macrophages in remyelination. Incompletely repaired lesions in NG2 null bone marrow recipients at 6 weeks post-injury retain elevated numbers of macrophages, in contrast to lower numbers of macrophages in more completely repaired lesions in wild type bone marrow recipients. This suggests that NG2 expression renders macrophages more effective in myelin repair and less likely to promote chronic inflammation. Effective macrophage involvement in myelin repair is due in part to effects on the proliferation and/or recruitment of oligodendrocyte progenitor cells. Reduced numbers of oligodendrocyte progenitors are seen in lesions in NG2 null bone marrow recipients, likely due to deficits in macrophage production of oligodendrocyte progenitor-relevant mitogens and in phagocytosis of inhibitory myelin debris. Microglia also appear to be important for clearance of myelin debris, as indicated by reduced phagocytosis in NG2 null recipients receiving wild type bone marrow.
Collapse
|
30
|
Tsidulko AY, Kazanskaya GM, Kostromskaya DV, Aidagulova SV, Kiselev RS, Volkov AM, Kobozev VV, Gaitan AS, Krivoshapkin AL, Grigorieva EV. Prognostic relevance of NG2/CSPG4, CD44 and Ki-67 in patients with glioblastoma. Tumour Biol 2017; 39:1010428317724282. [PMID: 28945172 DOI: 10.1177/1010428317724282] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Neuron-glial antigen 2 (NG2, also known as CSPG4) and hyaluronic acid receptor CD44 are chondroitin sulphate proteoglycans actively involved in brain development and its malignant transformation. Here, we aimed to compare prognostic significances of NG2, CD44 and Ki-67 expression in glioblastoma multiforme patients. Totally, 45 tissue samples and 83 paraffin-embedded tissues for 75 patients were analysed. The prognostic values of the genes were analysed using Kaplan-Meier survival curves. Grade III gliomas showed 2-fold difference in NG2 expression between anaplastic astrocytoma and oligoastrocytoma (10.1 ± 3.5 and 25.5 ± 14.5, respectively). For grade IV gliomas, upregulated NG2 expression (21.0 ± 6.8) was associated with poor glioblastoma multiforme prognosis (overall survival < 12 months) compared with glioblastoma multiforme patients with good prognosis (4.4 ± 3.2; overall survival > 12 months). Multivariate survival analysis using Cox proportional hazards model confirmed that high NG2 expression was associated with low survival of the patients (hazard ratio: 3.43; 95% confidence interval: 1.18-9.93; p = 0.02), whereas age (hazard ratio: 1.02; 95% confidence interval: 0.96-1.09; p = 0.42), tumour resection (hazard ratio: 1.03; 95% confidence interval: 0.98-1.08; p = 0.25) and sex (hazard ratio: 0.62; 95% confidence interval: 0.21-1.86; p = 0.40) did not show significant association with prognosis. Although the positive correlation was shown for NG2 and CD44 expression in the glioblastomas (Pearson coefficient = 0.954), Kaplan-Meier and multivariate survival analyses did not revealed a significant association of the increased CD44 expression (hazard ratio: 2.18; 95% confidence interval: 0.50-9.43; p = 0.30) or high Ki-67 proliferation index (hazard ratio: 1.10; 95% confidence interval: 1.02-1.20; p = 0.02) with the disease prognosis. The results suggest that upregulation of NG2/CSPG4 rather than changes in CD44 or Ki-67 expression is associated with low overall survival in glioblastoma multiforme patients, supporting NG2/CSPG4 as a potential prognostic marker in glioblastoma.
Collapse
Affiliation(s)
| | - Galina M Kazanskaya
- 1 Institute of Molecular Biology and Biophysics, Novosibirsk, Russia.,2 Meshalkin Research Institute of Circulation Pathology, Novosibirsk, Russia
| | | | | | - Roman S Kiselev
- 2 Meshalkin Research Institute of Circulation Pathology, Novosibirsk, Russia.,3 Novosibirsk State Medical University, Novosibirsk, Russia
| | - Alexandr M Volkov
- 2 Meshalkin Research Institute of Circulation Pathology, Novosibirsk, Russia
| | | | | | - Alexei L Krivoshapkin
- 2 Meshalkin Research Institute of Circulation Pathology, Novosibirsk, Russia.,3 Novosibirsk State Medical University, Novosibirsk, Russia.,4 European Medical Center, Moscow, Russia
| | | |
Collapse
|
31
|
You WK, Stallcup WB. Localization of VEGF to Vascular ECM Is an Important Aspect of Tumor Angiogenesis. Cancers (Basel) 2017; 9:cancers9080097. [PMID: 28788063 PMCID: PMC5575600 DOI: 10.3390/cancers9080097] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 07/22/2017] [Accepted: 07/26/2017] [Indexed: 12/02/2022] Open
Abstract
Our research has identified several examples in which reduced VEGF-A binding to deficient vascular extracellular matrix leads to deficits in tumor vascularization and tumor growth: (1) germline ablation of collagen VI in the stroma of intracranial B16F10 melanomas; (2) knockdown of the Tks5 scaffolding protein in MDA-MB-231 mammary tumor cells; (3) germline ablation of NG2 proteoglycan in the stroma of MMTV-PyMT mammary tumors; and (4) myeloid-specific ablation of NG2 in the stroma of intracranial B16F10 melanomas. Tumor hypoxia is increased in each of the four types of experimental mice, accompanied by increases in total VEGF-A. However, while VEGF-A is highly associated with tumor blood vessels in control mice, it is much more diffusely distributed in tumors in all four sets of experimental mice, likely due to reduced extent of the vascular extracellular matrix. In parallel to lost VEGF-A localization, tumor vessels in each case have smaller diameters and are leakier than tumor vessels in control mice. Tumor growth is decreased as a result of this poor vascular function. The fact that the observed vascular changes occur in the absence of alterations in vascular density suggests that examination of vessel structure and function is more useful than vascular density for understanding the importance of angiogenesis in tumor progression.
Collapse
Affiliation(s)
| | - William B Stallcup
- Tumor Microenvironment and Cancer Immunology Program, Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA.
| |
Collapse
|
32
|
The Importance of Pericytes in Healing: Wounds and other Pathologies. Int J Mol Sci 2017; 18:ijms18061129. [PMID: 28538706 PMCID: PMC5485953 DOI: 10.3390/ijms18061129] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 05/12/2017] [Accepted: 05/15/2017] [Indexed: 12/20/2022] Open
Abstract
Much of current research investigates the beneficial properties of mesenchymal stem cells (MSCs) as a treatment for wounds and other forms of injury. In this review, we bring attention to and discuss the role of the pericyte, a cell type which shares much of the differentiation potential and regenerative properties of the MSC as well as specific roles in the regulation of angiogenesis, inflammation and fibrosis. Pericytes have been identified as dysfunctional or depleted in many disease states, and observing the outcomes of pericyte perturbation in models of disease and wound healing informs our understanding of overall pericyte function and identifies these cells as an important target in the development of therapies to encourage healing.
Collapse
|
33
|
Inhibition of Lysyl Oxidases Impairs Migration and Angiogenic Properties of Tumor-Associated Pericytes. Stem Cells Int 2017; 2017:4972078. [PMID: 28553358 PMCID: PMC5434472 DOI: 10.1155/2017/4972078] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Accepted: 03/05/2017] [Indexed: 12/30/2022] Open
Abstract
Pericytes are important cellular components of the tumor microenviroment with established roles in angiogenesis and metastasis. These two cancer hallmarks are modulated by enzymes of the LOX family, but thus far, information about LOX relevance in tumor-associated pericytes is lacking. Here, we performed a comparative characterization of normal and tumoral pericytes and report for the first time the modulatory effects of LOX enzymes on activated pericyte properties. Tumoral pericytes isolated from childhood ependymoma and neuroblastoma specimens displayed angiogenic properties in vitro and expressed typical markers, including CD146, NG2, and PDGFRβ. Expression of all LOX family members could be detected in both normal and tumor-associated pericytes. In most pericyte samples, LOXL3 was the family member displaying the highest transcript levels. Inhibition of LOX/LOXL activity with the inhibitor β-aminopropionitrile (βAPN) significantly reduced migration of pericytes, while proliferation rates were kept unaltered. Formation of tube-like structures in vitro by pericytes was also significantly impaired upon inhibition of LOX/LOXL activity with βAPN, which induced more prominent effects in tumor-associated pericytes. These findings reveal a novel involvement of the LOX family of enzymes in migration and angiogenic properties of pericytes, with implications in tumor development and in therapeutic targeting tumor microenvironment constituents.
Collapse
|
34
|
NG2 Proteoglycan Enhances Brain Tumor Progression by Promoting Beta-1 Integrin Activation in both Cis and Trans Orientations. Cancers (Basel) 2017; 9:cancers9040031. [PMID: 28362324 PMCID: PMC5406706 DOI: 10.3390/cancers9040031] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 03/23/2017] [Accepted: 03/29/2017] [Indexed: 12/23/2022] Open
Abstract
By physically interacting with beta-1 integrins, the NG2 proteoglycan enhances activation of the integrin heterodimers. In glioma cells, co-localization of NG2 and 31 integrin in individual cells (cis interaction) can be demonstrated by immunolabeling, and the NG2-integrin interaction can be confirmed by co-immunoprecipitation. NG2-dependent integrin activation is detected via use of conformationally sensitive monoclonal antibodies that reveal the activated state of the beta-1 subunit in NG2-positive versus NG2-negative cells. NG2-dependent activation of beta-1 integrins triggers downstream activation of FAK and PI3K/Akt signaling, resulting in increased glioma cell proliferation, motility, and survival. Similar NG2-dependent cis activation of beta-1 integrins occurs in microvascular pericytes, leading to enhanced proliferation and motility of these vascular cells. Surprisingly, pericyte NG2 is also able to promote beta-1 integrin activation in closely apposed endothelial cells (trans interaction). Enhanced beta-1 signaling in endothelial cells promotes endothelial maturation by inducing the formation of endothelial junctions, resulting in increased barrier function of the endothelium and increased basal lamina assembly. NG2-dependent beta-1 integrin signaling is therefore important for tumor progression by virtue of its affects not only on the tumor cells themselves, but also on the maturation and function of tumor blood vessels.
Collapse
|
35
|
Ampofo E, Schmitt BM, Menger MD, Laschke MW. The regulatory mechanisms of NG2/CSPG4 expression. Cell Mol Biol Lett 2017; 22:4. [PMID: 28536635 PMCID: PMC5415841 DOI: 10.1186/s11658-017-0035-3] [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] [Received: 12/13/2016] [Accepted: 02/22/2017] [Indexed: 12/24/2022] Open
Abstract
Neuron-glial antigen 2 (NG2), also known as chondroitin sulphate proteoglycan 4 (CSPG4), is a surface type I transmembrane core proteoglycan that is crucially involved in cell survival, migration and angiogenesis. NG2 is frequently used as a marker for the identification and characterization of certain cell types, but little is known about the mechanisms regulating its expression. In this review, we provide evidence that the regulation of NG2 expression underlies inflammation and hypoxia and is mediated by methyltransferases, transcription factors, including Sp1, paired box (Pax) 3 and Egr-1, and the microRNA miR129-2. These regulatory factors crucially determine NG2-mediated cellular processes such as glial scar formation in the central nervous system (CNS) or tumor growth and metastasis. Therefore, they are potential targets for the establishment of novel NG2-based therapeutic strategies in the treatment of CNS injuries, cancer and other conditions of these types.
Collapse
Affiliation(s)
- Emmanuel Ampofo
- Institute for Clinical & Experimental Surgery, Saarland University, 66421 Homburg, Germany
| | - Beate M Schmitt
- Institute for Clinical & Experimental Surgery, Saarland University, 66421 Homburg, Germany
| | - Michael D Menger
- Institute for Clinical & Experimental Surgery, Saarland University, 66421 Homburg, Germany
| | - Matthias W Laschke
- Institute for Clinical & Experimental Surgery, Saarland University, 66421 Homburg, Germany
| |
Collapse
|
36
|
Sá da Bandeira D, Casamitjana J, Crisan M. Pericytes, integral components of adult hematopoietic stem cell niches. Pharmacol Ther 2016; 171:104-113. [PMID: 27908803 DOI: 10.1016/j.pharmthera.2016.11.006] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The interest in perivascular cells as a niche for adult hematopoietic stem cells (HSCs) is significantly growing. In the adult bone marrow (BM), perivascular cells and HSCs cohabit. Among perivascular cells, pericytes are precursors of mesenchymal stem/stromal cells (MSCs) that are capable of differentiating into osteoblasts, adipocytes and chondrocytes. In situ, pericytes are recognised by their localisation to the abluminal side of the blood vessel wall and closely associated with endothelial cells, in combination with the expression of markers such as CD146, neural glial 2 (NG2), platelet derived growth factor receptor β (PDGFRβ), α-smooth muscle actin (α-SMA), nestin (Nes) and/or leptin receptor (LepR). However, not all pericytes share a common phenotype: different immunophenotypes can be associated with distinct mesenchymal features, including hematopoietic support. In adult BM, arteriolar and sinusoidal pericytes control HSC behaviour, maintenance, quiescence and trafficking through paracrine effects. Different groups identified and characterized hematopoietic supportive pericyte subpopulations using various markers and mouse models. In this review, we summarize recent work performed by others to understand the role of the perivascular niche in the biology of HSCs in adults, as well as their importance in the development of therapies.
Collapse
Affiliation(s)
- D Sá da Bandeira
- BHF Centre for Cardiovascular Science, MRC Scottish Centre for Regenerative Medicine, The Edinburgh Medical School, University of Edinburgh, Edinburgh, UK
| | - J Casamitjana
- BHF Centre for Cardiovascular Science, MRC Scottish Centre for Regenerative Medicine, The Edinburgh Medical School, University of Edinburgh, Edinburgh, UK
| | - M Crisan
- BHF Centre for Cardiovascular Science, MRC Scottish Centre for Regenerative Medicine, The Edinburgh Medical School, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
37
|
Secomb TW, Pries AR. Microvascular Plasticity: Angiogenesis in Health and Disease--Preface. Microcirculation 2016; 23:93-4. [PMID: 26639099 DOI: 10.1111/micc.12262] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2015] [Accepted: 11/25/2015] [Indexed: 11/26/2022]
Abstract
This Special Topic Issue is concerned with the mechanisms that determine the structure of microvascular networks. The vast number of vessels and the highly plastic character of the microcirculation give evidence that microvascular network structures emerge as a result of responses of individual vessels and cells to the local stimuli that they experience, through a combination of angiogenesis, remodeling and pruning. The articles in this issue of Microcirculation address a range of cellular and molecular mechanisms involved in these processes.
Collapse
Affiliation(s)
- Timothy W Secomb
- Department of Physiology, University of Arizona, Tucson, Arizona, USA
| | - Axel R Pries
- Department of Physiology, Charité Berlin, Berlin, Germany.,Deutsches Herzzentrum Berlin, Berlin, Germany
| |
Collapse
|
38
|
Abstract
Pericytes are contractile mural cells that wrap around the endothelial cells of capillaries and venules. Depending on the triggers by cellular signals, pericytes have specific functionality in tumor microenvironments, properties of potent stem cells, and plasticity in cellular pathology. These features of pericytes can be activated for the promotion or reduction of angiogenesis. Frontier studies have exploited pericyte-targeting drug delivery, using pericyte-specific peptides, small molecules, and DNA in tumor therapy. Moreover, the communication between pericytes and endothelial cells has been applied to the induction of vessel neoformation in tissue engineering. Pericytes may prove to be a novel target for tumor therapy and tissue engineering. The present paper specifically reviews pericyte-specific drug delivery and tissue engineering, allowing insight into the emerging research targeting pericytes.
Collapse
Affiliation(s)
- Eunah Kang
- School of Chemical Engineering and Material Science, Department of Internal Medicine, College of Medicine, Chung-Ang University, Dongjak-Gu, Seoul, South Korea
| | - Jong Wook Shin
- Division of Allergic and Pulmonary Medicine, Department of Internal Medicine, College of Medicine, Chung-Ang University, Dongjak-Gu, Seoul, South Korea
| |
Collapse
|
39
|
Kucharova K, Stallcup WB. NG2-proteoglycan-dependent contributions of oligodendrocyte progenitors and myeloid cells to myelin damage and repair. J Neuroinflammation 2015; 12:161. [PMID: 26338007 PMCID: PMC4559177 DOI: 10.1186/s12974-015-0385-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 08/20/2015] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The NG2 proteoglycan is expressed by several cell types in demyelinated lesions and has important effects on the biology of these cells. Here we determine the cell-type-specific roles of NG2 in the oligodendrocyte progenitor cell (OPC) and myeloid cell contributions to demyelination and remyelination. METHODS We have used Cre-Lox technology to dissect the cell-type-specific contributions of NG2 to myelin damage and repair. Demyelination is induced by microinjection of 1 % lysolecithin into the spinal cord white matter of control, OPC-specific NG2-null (OPC-NG2ko), and myeloid-specific NG2-null (My-NG2ko) mice. The status of OPCs, myeloid cells, axons, and myelin is assessed by light, immunofluorescence, confocal, and electron microscopy. RESULTS In OPC-NG2ko mice 1 week after lysolecithin injection, the OPC mitotic index is reduced by 40 %, resulting in 25 % fewer OPCs at 1 week and a 28 % decrease in mature oligodendrocytes at 6 weeks post-injury. The initial demyelinated lesion size is not affected in OPC-NG2ko mice, but lesion repair is delayed by reduced production of oligodendrocytes. In contrast, both the initial extent of demyelination and the kinetics of lesion repair are decreased in My-NG2ko mice. Surprisingly, the OPC mitotic index at 1 week post-injury is also reduced (by 48 %) in My-NG2ko mice, leading to a 35 % decrease in OPCs at 1 week and a subsequent 34 % reduction in mature oligodendrocytes at 6 weeks post-injury. Clearance of myelin debris is also reduced by 40 % in My-NG2ko mice. Deficits in myelination detected by immunostaining for myelin basic protein are confirmed by toluidine blue staining and by electron microscopy. In addition to reduced myelin repair, fewer axons are found in 6-week lesions in both OPC-NG2ko and My-NG2ko mice, emphasizing the importance of myelination for neuron survival. CONCLUSIONS Reduced generation of OPCs and oligodendrocytes in OPC-NG2ko mice correlates with reduced myelin repair. Diminished demyelination in My-NG2ko mice may stem from a reduction (approximately 70 %) in myeloid cell recruitment to lesions. Reduced macrophage/microglia numbers may then result in decreased myelin repair via diminished clearance of myelin debris and reduced stimulatory effects on OPCs.
Collapse
Affiliation(s)
- Karolina Kucharova
- Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA, 92037, USA.
| | - William B Stallcup
- Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA, 92037, USA
| |
Collapse
|