1
|
Lozano LP, Jensen R, Jennisch M, Pandala NG, Jamshidi F, Boldt HC, Tucker BA, Binkley EM. Genetics and current research models of Mendelian tumor predisposition syndromes with ocular involvement. Prog Retin Eye Res 2025; 106:101359. [PMID: 40274012 DOI: 10.1016/j.preteyeres.2025.101359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 04/17/2025] [Accepted: 04/18/2025] [Indexed: 04/26/2025]
Abstract
In this review, we aim to provide a survey of hereditable tumor predisposition syndromes with a Mendelian inheritance pattern and ocular involvement. We focus our discussion on von Hippel-Lindau disease, neurofibromatosis type 1, NF2-related schwannomatosis, tuberous sclerosis complex, retinoblastoma, and the BAP1 tumor predisposition syndrome. For each of the six diseases, we discuss the clinical presentation and the molecular pathophysiology. We emphasize the genetics, current research models, and therapeutic developments. After reading each disease section, readers should possess an understanding of the clinical presentation, genetic causes and inheritance patterns, and current state of research in disease modeling and treatment.
Collapse
Affiliation(s)
- Lola P Lozano
- Institute for Vision Research, The University of Iowa, Iowa City, IA, 52242, USA.
| | - Renato Jensen
- Institute for Vision Research, The University of Iowa, Iowa City, IA, 52242, USA.
| | - Madeleine Jennisch
- Institute for Vision Research, The University of Iowa, Iowa City, IA, 52242, USA.
| | - Narendra G Pandala
- Institute for Vision Research, The University of Iowa, Iowa City, IA, 52242, USA.
| | - Farzad Jamshidi
- Department of Ophthalmology, University of Pittsburgh/UPMC, Pittsburgh, PA, 15213, USA.
| | - H Culver Boldt
- Institute for Vision Research, The University of Iowa, Iowa City, IA, 52242, USA; Department of Ophthalmology and Visual Sciences, The University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA.
| | - Budd A Tucker
- Institute for Vision Research, The University of Iowa, Iowa City, IA, 52242, USA; Department of Ophthalmology and Visual Sciences, The University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA.
| | - Elaine M Binkley
- Institute for Vision Research, The University of Iowa, Iowa City, IA, 52242, USA; Department of Ophthalmology and Visual Sciences, The University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA.
| |
Collapse
|
2
|
Jang HJ, Park JW. Microenvironmental Drivers of Glioma Progression. Int J Mol Sci 2025; 26:2108. [PMID: 40076738 PMCID: PMC11900340 DOI: 10.3390/ijms26052108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 02/22/2025] [Accepted: 02/25/2025] [Indexed: 03/14/2025] Open
Abstract
Gliomas, particularly glioblastoma (GBM), are among the most challenging brain tumors due to their complex and dynamic tumor microenvironment (TME). The TME plays a pivotal role in tumor progression, immune evasion, and resistance to therapy through intricate interactions among glioma cells, immune components, neurons, astrocytes, the extracellular matrix, and the blood-brain barrier. Targeting the TME has demonstrated potential, with immunotherapies such as checkpoint inhibitors and neoadjuvant therapies enhancing immune responses. Nonetheless, overcoming the immunosuppressive landscape and metabolic adaptations continues to pose significant challenges. This review explores the diverse cellular and molecular mechanisms that shape the glioma TME. A deeper understanding of these mechanisms holds promise for providing novel therapeutic opportunities to improve glioma treatment outcomes.
Collapse
Affiliation(s)
- Hyun Ji Jang
- Department of Life Sciences, College of BioNano Technology, Gachon University, Seongnam 13120, Republic of Korea
| | - Jong-Whi Park
- Department of Life Sciences, College of BioNano Technology, Gachon University, Seongnam 13120, Republic of Korea
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, Republic of Korea
| |
Collapse
|
3
|
Laurenge A, Castro-Vega LJ, Huberfeld G. Reciprocal interactions between glioma and tissue-resident cells fueling tumor progression. HANDBOOK OF CLINICAL NEUROLOGY 2025; 210:177-190. [PMID: 40148044 DOI: 10.1016/b978-0-443-19102-2.00007-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
Gliomas are the most frequent primary brain tumor and are essentially incurable. While nondiffuse gliomas are circumscribed, diffuse gliomas display an aggressive behavior characterized by tumor cell migration over large distances into the brain parenchyma, thereby precluding curative surgical resection. Almost all diffuse gliomas progress and recur as higher grades and become resistant to standard-of-care treatments. It is being increasingly recognized that glioma cells establish functional interactions with cells residing in the tumor microenvironment. Of these, tumor-associated microglia and macrophages (TAMs) play critical roles in immunosuppression through modulation of the extracellular matrix, and the secretion of molecules such as cytokines, neurotrophic factors, and micro-RNAs (miRNAs). Conversely, glioma cell signals influence cell states and drive the metabolic reprogramming of TAMs. Similarly, emergent evidence indicates that neuronal activity influences glioma by released factors and by establishing functional synapses with glioma cells to promote tumor growth and invasion. Glioma cells also affect local neuronal activities and maintain connections through microtube gap junctions to amplify local effects. Here, we discuss the molecular mechanisms underlying bidirectional interactions between glioma cells and TAMs, as well as between glioma cells and neurons. A better understanding of these cellular cross talks is crucial for the development of novel therapeutic strategies for diffuse gliomas.
Collapse
Affiliation(s)
- Alice Laurenge
- Genetics & Development of Brain Tumors Laboratory, ICM - Paris Brain Institute, Sorbonne University, UMR S 1127, Inserm U 1127, CNRS UMR 7225, F-75013, Paris, France; AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Neuro-Oncology Department, F-75013, Paris, France
| | - Luis Jaime Castro-Vega
- Genetics & Development of Brain Tumors Laboratory, ICM - Paris Brain Institute, Sorbonne University, UMR S 1127, Inserm U 1127, CNRS UMR 7225, F-75013, Paris, France
| | - Gilles Huberfeld
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Neuronal Signaling in Epilepsy and Glioma, Paris, France; Department of Neurology, Hôpital Fondation Adolphe de Rothschild, Paris, France.
| |
Collapse
|
4
|
Cole JJ, Ferner RE, Gutmann DH. Neurofibromatosis type 1. ROSENBERG'S MOLECULAR AND GENETIC BASIS OF NEUROLOGICAL AND PSYCHIATRIC DISEASE 2025:231-249. [DOI: 10.1016/b978-0-443-19176-3.00017-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
5
|
Chan A, Zhang K, Martin G, Bano S, Chatterjee J, Mahto S, Wang A, Gutmann DH, Brossier NM. Maternal obesogenic diet operates at the tumor cell of origin to increase incidence and decrease latency of neurofibromatosis type 1 optic pathway glioma. Neuro Oncol 2024; 26:2339-2351. [PMID: 39023130 PMCID: PMC11630557 DOI: 10.1093/neuonc/noae136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Indexed: 07/20/2024] Open
Abstract
BACKGROUND Pediatric low-grade glioma incidence has been rising in the United States, mirroring the rising rates of pediatric and maternal obesity. Recently, children of obese mothers were demonstrated to develop brain tumors at higher rates. Importantly, obesity in the United States is largely driven by diet, given the prevalence of high-fat and high-sugar (HFHS) food choices. Since high-fat diet exposure can increase embryonic neuroglial progenitor cell (NPC) proliferation, the potential cells of origin for a low-grade glioma, we hypothesized that in utero exposure to an obesogenic diet would modify pediatric brain penetrance and latency by affecting the tumor cell of origin. METHODS We employed several murine models of the neurofibromatosis type 1 (NF1) pediatric brain tumor predisposition syndrome, in which optic pathway gliomas (Nf1-OPGs) arise from neuroglial progenitor cells in the embryonic third ventricular zone (TVZ). We exposed dams and offspring to an obesogenic HFHS diet or control chow and analyzed fetal neurodevelopment at E19.5 and tumor formation at 6 weeks-3 months. RESULTS Progeny from HFHS diet-exposed dams demonstrated increased TVZ NPC proliferation and glial differentiation. Dietary switch cohorts confirmed that these effects were dependent upon maternal diet, rather than maternal weight. Obesogenic diet (Ob) similarly accelerated glioma formation in a high-penetrance Nf1-OPG strain and increased glioma penetrance in 2 low-penetrance Nf1-OPG strains. In contrast, Ob exposure in the postnatal period alone did not recapitulate these effects. CONCLUSIONS These findings establish maternal obesogenic diet as a risk factor for murine Nf1-OPG formation, acting in part through in utero effects on the tumor cell of origin.
Collapse
Affiliation(s)
- Ambrose Chan
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Kailong Zhang
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Gemma Martin
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Sabiha Bano
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Jit Chatterjee
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Sarvika Mahto
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Avery Wang
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - David H Gutmann
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Nicole M Brossier
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
6
|
Barakat R, Chatterjee J, Mu R, Qi X, Gu X, Smirnov I, Cobb O, Gao K, Barnes A, Kipnis J, Gutmann DH. Human single cell RNA-sequencing reveals a targetable CD8 + exhausted T cell population that maintains mouse low-grade glioma growth. Nat Commun 2024; 15:10312. [PMID: 39609412 PMCID: PMC11605098 DOI: 10.1038/s41467-024-54569-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 11/14/2024] [Indexed: 11/30/2024] Open
Abstract
In solid cancers, T cells typically function as cytotoxic effectors to limit tumor growth, prompting therapies that capitalize upon this antineoplastic property (immune checkpoint inhibition; ICI). Unfortunately, ICI treatments have been largely ineffective for high-grade brain tumors (gliomas; HGGs). Leveraging several single-cell RNA sequencing datasets, we report greater CD8+ exhausted T cells in human pediatric low-grade gliomas (LGGs) relative to adult and pediatric HGGs. Using several preclinical mouse LGG models (Nf1-OPG mice), we show that these PD1+/TIGIT+ CD8+ exhausted T cells are restricted to the tumor tissue, where they express paracrine factors necessary for OPG growth. Importantly, ICI treatments with α-PD1 and α-TIGIT antibodies attenuate Nf1-OPG tumor proliferation through suppression of two cytokine (Ccl4 and TGFβ)-mediated mechanisms, rather than by T cell-mediated cytotoxicity, as well as suppress monocyte-controlled T cell chemotaxis. Collectively, these findings establish a previously unrecognized function for CD8+ exhausted T cells as specialized regulators of LGG maintenance.
Collapse
Affiliation(s)
- Rasha Barakat
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Jit Chatterjee
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Rui Mu
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Xuanhe Qi
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Xingxing Gu
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Igor Smirnov
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Olivia Cobb
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Karen Gao
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Angelica Barnes
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Jonathan Kipnis
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - David H Gutmann
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| |
Collapse
|
7
|
Zhang L, Wang Y, Cai X, Mao X, Sun H. Deciphering the CNS-glioma dialogue: Advanced insights into CNS-glioma communication pathways and their therapeutic potential. J Cent Nerv Syst Dis 2024; 16:11795735241292188. [PMID: 39493257 PMCID: PMC11528668 DOI: 10.1177/11795735241292188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 09/23/2024] [Indexed: 11/05/2024] Open
Abstract
The field of cancer neuroscience has rapidly evolved, shedding light on the complex interplay between the nervous system and cancer, with a particular focus on the relationship between the central nervous system (CNS) and gliomas. Recent advancements have underscored the critical influence of CNS activity on glioma progression, emphasizing the roles of neurons and neuroglial cells in both the onset and evolution of gliomas. This review meticulously explores the primary communication pathways between the CNS and gliomas, encompassing neuro-glioma synapses, paracrine mechanisms, extracellular vesicles, tunneling nanotubes, and the integrative CNS-immune-glioma axis. It also evaluates current and emerging therapeutic interventions aimed at these pathways and proposes forward-looking perspectives for research in this domain.
Collapse
Affiliation(s)
- Lu Zhang
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital and the Second Clinical Medical College, Southern Medical University, Guangzhou, China
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yajing Wang
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital and the Second Clinical Medical College, Southern Medical University, Guangzhou, China
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaoxi Cai
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital and the Second Clinical Medical College, Southern Medical University, Guangzhou, China
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xinyuan Mao
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital and the Second Clinical Medical College, Southern Medical University, Guangzhou, China
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Haitao Sun
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital and the Second Clinical Medical College, Southern Medical University, Guangzhou, China
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong–Hong Kong–Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Southern Medical University, Guangzhou, China
| |
Collapse
|
8
|
Chen Y, Yu J, Ge S, Jia R, Song X, Wang Y, Fan X. An Overview of Optic Pathway Glioma With Neurofibromatosis Type 1: Pathogenesis, Risk Factors, and Therapeutic Strategies. Invest Ophthalmol Vis Sci 2024; 65:8. [PMID: 38837168 PMCID: PMC11160950 DOI: 10.1167/iovs.65.6.8] [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: 01/07/2024] [Accepted: 05/14/2024] [Indexed: 06/06/2024] Open
Abstract
Optic pathway gliomas (OPGs) are most predominant pilocytic astrocytomas, which are typically diagnosed within the first decade of life. The majority of affected children with OPGs also present with neurofibromatosis type 1 (NF1), the most common tumor predisposition syndrome. OPGs in individuals with NF1 primarily affect the optic pathway and lead to visual disturbance. However, it is challenging to assess risk in asymptomatic patients without valid biomarkers. On the other hand, for symptomatic patients, there is still no effective treatment to prevent or recover vision loss. Therefore, this review summarizes current knowledge regarding the pathogenesis of NF1-associated OPGs (NF1-OPGs) from preclinical studies to seek potential prognostic markers and therapeutic targets. First, the loss of the NF1 gene activates 3 distinct Ras effector pathways, including the PI3K/AKT/mTOR pathway, the MEK/ERK pathway, and the cAMP pathway, which mediate glioma tumorigenesis. Meanwhile, non-neoplastic cells from the tumor microenvironment (microglia, T cells, neurons, etc.) also contribute to gliomagenesis via various soluble factors. Subsequently, we investigated potential genetic risk factors, molecularly targeted therapies, and neuroprotective strategies for tumor prevention and vision recovery. Last, potential directions and promising preclinical models of NF1-OPGs are presented for further research. On the whole, NF1-OPGs develop as a result of the interaction between glioma cells and the tumor microenvironment. Developing effective treatments require a better understanding of tumor molecular characteristics, as well as multistage interventions targeting both neoplastic cells and non-neoplastic cells.
Collapse
Affiliation(s)
- Ying Chen
- Department of Ophthalmology, Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, P.R. China
| | - Jie Yu
- Department of Ophthalmology, Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, P.R. China
| | - Shengfang Ge
- Department of Ophthalmology, Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, P.R. China
| | - Renbing Jia
- Department of Ophthalmology, Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, P.R. China
| | - Xin Song
- Department of Ophthalmology, Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, P.R. China
| | - Yefei Wang
- Department of Ophthalmology, Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, P.R. China
| | - Xianqun Fan
- Department of Ophthalmology, Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, P.R. China
| |
Collapse
|
9
|
Chatterjee J, Koleske JP, Chao A, Sauerbeck AD, Chen JK, Qi X, Ouyang M, Boggs LG, Idate R, Marco Y Marquez LI, Kummer TT, Gutmann DH. Brain injury drives optic glioma formation through neuron-glia signaling. Acta Neuropathol Commun 2024; 12:21. [PMID: 38308315 PMCID: PMC10837936 DOI: 10.1186/s40478-024-01735-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 01/15/2024] [Indexed: 02/04/2024] Open
Abstract
Tissue injury and tumorigenesis share many cellular and molecular features, including immune cell (T cells, monocytes) infiltration and inflammatory factor (cytokines, chemokines) elaboration. Their common pathobiology raises the intriguing possibility that brain injury could create a tissue microenvironment permissive for tumor formation. Leveraging several murine models of the Neurofibromatosis type 1 (NF1) cancer predisposition syndrome and two experimental methods of brain injury, we demonstrate that both optic nerve crush and diffuse traumatic brain injury induce optic glioma (OPG) formation in mice harboring Nf1-deficient preneoplastic progenitors. We further elucidate the underlying molecular and cellular mechanisms, whereby glutamate released from damaged neurons stimulates IL-1β release by oligodendrocytes to induce microglia expression of Ccl5, a growth factor critical for Nf1-OPG formation. Interruption of this cellular circuit using glutamate receptor, IL-1β or Ccl5 inhibitors abrogates injury-induced glioma progression, thus establishing a causative relationship between injury and tumorigenesis.
Collapse
Affiliation(s)
- Jit Chatterjee
- Department of Neurology, Washington University School of Medicine, 660 S. Euclid Avenue, Box 8111, St. Louis, MO, 63110, USA
| | - Joshua P Koleske
- Department of Neurology, Washington University School of Medicine, 660 S. Euclid Avenue, Box 8111, St. Louis, MO, 63110, USA
| | - Astoria Chao
- Department of Neurology, Washington University School of Medicine, 660 S. Euclid Avenue, Box 8111, St. Louis, MO, 63110, USA
| | - Andrew D Sauerbeck
- Department of Neurology, Washington University School of Medicine, 660 S. Euclid Avenue, Box 8111, St. Louis, MO, 63110, USA
| | - Ji-Kang Chen
- Department of Neurology, Washington University School of Medicine, 660 S. Euclid Avenue, Box 8111, St. Louis, MO, 63110, USA
| | - Xuanhe Qi
- Department of Neurology, Washington University School of Medicine, 660 S. Euclid Avenue, Box 8111, St. Louis, MO, 63110, USA
| | - Megan Ouyang
- Department of Neurology, Washington University School of Medicine, 660 S. Euclid Avenue, Box 8111, St. Louis, MO, 63110, USA
| | - Lucy G Boggs
- Department of Neurology, Washington University School of Medicine, 660 S. Euclid Avenue, Box 8111, St. Louis, MO, 63110, USA
| | - Rujuta Idate
- Department of Neurology, Washington University School of Medicine, 660 S. Euclid Avenue, Box 8111, St. Louis, MO, 63110, USA
| | - Lara Isabel Marco Y Marquez
- Department of Neurology, Washington University School of Medicine, 660 S. Euclid Avenue, Box 8111, St. Louis, MO, 63110, USA
| | - Terrence T Kummer
- Department of Neurology, Washington University School of Medicine, 660 S. Euclid Avenue, Box 8111, St. Louis, MO, 63110, USA
| | - David H Gutmann
- Department of Neurology, Washington University School of Medicine, 660 S. Euclid Avenue, Box 8111, St. Louis, MO, 63110, USA.
| |
Collapse
|
10
|
Abstract
Gliomas are a diverse group of primary central nervous system tumors that affect both children and adults. Recent studies have revealed a dynamic cross talk that occurs between glioma cells and components of their microenvironment, including neurons, astrocytes, immune cells, and the extracellular matrix. This cross talk regulates fundamental aspects of glioma development and growth. In this review, we discuss recent discoveries about the impact of these interactions on gliomas and highlight how tumor cells actively remodel their microenvironment to promote disease. These studies provide a better understanding of the interactions in the microenvironment that are important in gliomas, offer insight into the cross talk that occurs, and identify potential therapeutic vulnerabilities that can be utilized to improve clinical outcomes.
Collapse
Affiliation(s)
- Maya Anjali Jayaram
- Department of Neurological Surgery, Brain Tumor Center, University of California, San Francisco, California, USA;
| | - Joanna J Phillips
- Department of Neurological Surgery, Brain Tumor Center, University of California, San Francisco, California, USA;
- Division of Neuropathology, Department of Pathology, University of California, San Francisco, California, USA
| |
Collapse
|
11
|
Kuhrt LD, Motta E, Elmadany N, Weidling H, Fritsche-Guenther R, Efe IE, Cobb O, Chatterjee J, Boggs LG, Schnauß M, Diecke S, Semtner M, Anastasaki C, Gutmann DH, Kettenmann H. Neurofibromin 1 mutations impair the function of human induced pluripotent stem cell-derived microglia. Dis Model Mech 2023; 16:dmm049861. [PMID: 37990867 PMCID: PMC10740172 DOI: 10.1242/dmm.049861] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 11/10/2023] [Indexed: 11/23/2023] Open
Abstract
Neurofibromatosis type 1 (NF1) is an autosomal dominant condition caused by germline mutations in the neurofibromin 1 (NF1) gene. Children with NF1 are prone to the development of multiple nervous system abnormalities, including autism and brain tumors, which could reflect the effect of NF1 mutation on microglia function. Using heterozygous Nf1-mutant mice, we previously demonstrated that impaired purinergic signaling underlies deficits in microglia process extension and phagocytosis in situ. To determine whether these abnormalities are also observed in human microglia in the setting of NF1, we leveraged an engineered isogenic series of human induced pluripotent stem cells to generate human microglia-like (hiMGL) cells heterozygous for three different NF1 gene mutations found in patients with NF1. Whereas all NF1-mutant and isogenic control hiMGL cells expressed classical microglia markers and exhibited similar transcriptomes and cytokine/chemokine release profiles, only NF1-mutant hiMGL cells had defects in P2X receptor activation, phagocytosis and motility. Taken together, these findings indicate that heterozygous NF1 mutations impair a subset of the functional properties of human microglia, which could contribute to the neurological abnormalities seen in children with NF1.
Collapse
Affiliation(s)
- Leonard D. Kuhrt
- Cellular Neurosciences, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany
- Technology Platform Pluripotent Stem Cells, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany
- Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Edyta Motta
- Cellular Neurosciences, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany
- Department of Neurosurgery, University Medical Center Schleswig-Holstein, 24105 Kiel, Germany
| | - Nirmeen Elmadany
- Cellular Neurosciences, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany
- German Cancer Consortium (DKTK), Clinical Cooperation Unit (CCU), Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Department of Neurology, Medical Faculty Mannheim (MCTN), University of Heidelberg, 68167 Mannheim, Germany
| | - Hannah Weidling
- Cellular Neurosciences, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany
- Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Raphaela Fritsche-Guenther
- Berlin Institute of Health (BIH) at Charité – Universitätsmedizin Berlin, BIH Metabolomics Platform, 13353 Berlin, Germany
| | - Ibrahim E. Efe
- Cellular Neurosciences, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany
- Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Olivia Cobb
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jit Chatterjee
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Lucy G. Boggs
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Marina Schnauß
- Cellular Neurosciences, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany
| | - Sebastian Diecke
- Technology Platform Pluripotent Stem Cells, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany
| | - Marcus Semtner
- Cellular Neurosciences, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany
- Klinik für Augenheilkunde, Charité – Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Corina Anastasaki
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - David H. Gutmann
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Helmut Kettenmann
- Cellular Neurosciences, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China, 518000
| |
Collapse
|
12
|
Milde T, Fangusaro J, Fisher MJ, Hawkins C, Rodriguez FJ, Tabori U, Witt O, Zhu Y, Gutmann DH. Optimizing preclinical pediatric low-grade glioma models for meaningful clinical translation. Neuro Oncol 2023; 25:1920-1931. [PMID: 37738646 PMCID: PMC10628935 DOI: 10.1093/neuonc/noad125] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/24/2023] Open
Abstract
Pediatric low-grade gliomas (pLGGs) are the most common brain tumor in young children. While they are typically associated with good overall survival, children with these central nervous system tumors often experience chronic tumor- and therapy-related morbidities. Moreover, individuals with unresectable tumors frequently have multiple recurrences and persistent neurological symptoms. Deep molecular analyses of pLGGs reveal that they are caused by genetic alterations that converge on a single mitogenic pathway (MEK/ERK), but their growth is heavily influenced by nonneoplastic cells (neurons, T cells, microglia) in their local microenvironment. The interplay between neoplastic cell MEK/ERK pathway activation and stromal cell support necessitates the use of predictive preclinical models to identify the most promising drug candidates for clinical evaluation. As part of a series of white papers focused on pLGGs, we discuss the current status of preclinical pLGG modeling, with the goal of improving clinical translation for children with these common brain tumors.
Collapse
Affiliation(s)
- Till Milde
- Hopp Children’s Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
- KiTZ Clinical Trial Unit (ZIPO), Department of Pediatric Hematology, Oncology, Immunology and Pulmonology, Heidelberg University Hospital, Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Jason Fangusaro
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Michael J Fisher
- Division of Oncology, Children’s Hospital of Philadelphia, Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Cynthia Hawkins
- Department of Laboratory Medicine and Pathobiology, Hospital for Sick Children, Toronto, Canada
| | - Fausto J Rodriguez
- Department of Pathology, University of California Los Angeles, Los Angeles, California, USA
| | - Uri Tabori
- Department of Medical Biophysics, Institute of Medical Science and Paediatrics, University of Toronto, Toronto, Canada
| | - Olaf Witt
- Hopp Children’s Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
- KiTZ Clinical Trial Unit (ZIPO), Department of Pediatric Hematology, Oncology, Immunology and Pulmonology, Heidelberg University Hospital, Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Yuan Zhu
- Gilbert Family Neurofibromatosis Institute Center for Cancer and Immunology Research, Children’s National Hospital, Washington, DC, USA
| | - David H Gutmann
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
13
|
Irshad K, Huang YK, Rodriguez P, Lo J, Aghoghovwia BE, Pan Y, Chang KC. The Neuroimmune Regulation and Potential Therapeutic Strategies of Optic Pathway Glioma. Brain Sci 2023; 13:1424. [PMID: 37891793 PMCID: PMC10605541 DOI: 10.3390/brainsci13101424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 09/28/2023] [Accepted: 10/03/2023] [Indexed: 10/29/2023] Open
Abstract
Optic pathway glioma (OPG) is one of the causes of pediatric visual impairment. Unfortunately, there is as yet no cure for such a disease. Understanding the underlying mechanisms and the potential therapeutic strategies may help to delay the progression of OPG and rescue the visual morbidities. Here, we provide an overview of preclinical OPG studies and the regulatory pathways controlling OPG pathophysiology. We next discuss the role of microenvironmental cells (neurons, T cells, and tumor-associated microglia and macrophages) in OPG development. Last, we provide insight into potential therapeutic strategies for treating OPG and promoting axon regeneration.
Collapse
Affiliation(s)
- Khushboo Irshad
- Department of Symptom Research, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (K.I.); (B.E.A.)
| | - Yu-Kai Huang
- Division of Neurosurgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan;
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Paul Rodriguez
- Department of Ophthalmology, Louis J. Fox Center for Vision Restoration, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA;
| | - Jung Lo
- Department of Ophthalmology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan;
| | - Benjamin E. Aghoghovwia
- Department of Symptom Research, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (K.I.); (B.E.A.)
| | - Yuan Pan
- Department of Symptom Research, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (K.I.); (B.E.A.)
- Department of Neuro-Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Kun-Che Chang
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Ophthalmology, Louis J. Fox Center for Vision Restoration, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA;
- Department of Neurobiology, Center of Neuroscience, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| |
Collapse
|
14
|
Tang Y, Gutmann DH. Neurofibromatosis Type 1-Associated Optic Pathway Gliomas: Current Challenges and Future Prospects. Cancer Manag Res 2023; 15:667-681. [PMID: 37465080 PMCID: PMC10351533 DOI: 10.2147/cmar.s362678] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 06/06/2023] [Indexed: 07/20/2023] Open
Abstract
Optic pathway glioma (OPG) occurs in as many as one-fifth of individuals with the neurofibromatosis type 1 (NF1) cancer predisposition syndrome. Generally considered low-grade and slow growing, many children with NF1-OPGs remain asymptomatic. However, due to their location within the optic pathway, ~20-30% of those harboring NF1-OPGs will experience symptoms, including progressive vision loss, proptosis, diplopia, and precocious puberty. While treatment with conventional chemotherapy is largely effective at attenuating tumor growth, it is not clear whether there is much long-term recovery of visual function. Additionally, because these tumors predominantly affect young children, there are unique challenges to NF1-OPG diagnosis, monitoring, and longitudinal management. Over the past two decades, the employment of authenticated genetically engineered Nf1-OPG mouse models have provided key insights into the function of the NF1 protein, neurofibromin, as well as the molecular and cellular pathways that contribute to optic gliomagenesis. Findings from these studies have resulted in the identification of new molecular targets whose inhibition blocks murine Nf1-OPG growth in preclinical studies. Some of these promising compounds have now entered into early clinical trials. Future research focused on defining the determinants that underlie optic glioma initiation, expansion, and tumor-induced optic nerve injury will pave the way to personalized risk assessment strategies, improved tumor monitoring, and optimized treatment plans for children with NF1-OPG.
Collapse
Affiliation(s)
- Yunshuo Tang
- Department of Ophthalmology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - David H Gutmann
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
15
|
Winkler F, Venkatesh HS, Amit M, Batchelor T, Demir IE, Deneen B, Gutmann DH, Hervey-Jumper S, Kuner T, Mabbott D, Platten M, Rolls A, Sloan EK, Wang TC, Wick W, Venkataramani V, Monje M. Cancer neuroscience: State of the field, emerging directions. Cell 2023; 186:1689-1707. [PMID: 37059069 PMCID: PMC10107403 DOI: 10.1016/j.cell.2023.02.002] [Citation(s) in RCA: 190] [Impact Index Per Article: 95.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/01/2023] [Accepted: 02/01/2023] [Indexed: 04/16/2023]
Abstract
The nervous system governs both ontogeny and oncology. Regulating organogenesis during development, maintaining homeostasis, and promoting plasticity throughout life, the nervous system plays parallel roles in the regulation of cancers. Foundational discoveries have elucidated direct paracrine and electrochemical communication between neurons and cancer cells, as well as indirect interactions through neural effects on the immune system and stromal cells in the tumor microenvironment in a wide range of malignancies. Nervous system-cancer interactions can regulate oncogenesis, growth, invasion and metastatic spread, treatment resistance, stimulation of tumor-promoting inflammation, and impairment of anti-cancer immunity. Progress in cancer neuroscience may create an important new pillar of cancer therapy.
Collapse
Affiliation(s)
- Frank Winkler
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg and Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Humsa S Venkatesh
- Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA
| | - Moran Amit
- Department of Head and Neck Surgery, MD Anderson Cancer Center and The University of Texas Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Tracy Batchelor
- Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA
| | - Ihsan Ekin Demir
- Department of Surgery, Technical University of Munich, Munich, Germany
| | - Benjamin Deneen
- Center for Stem Cells and Regenerative Medicine, Baylor College of Medicine, Houston, TX, USA
| | - David H Gutmann
- Department of Neurology, Washington University, St Louis, MO, USA
| | - Shawn Hervey-Jumper
- Department of Neurosurgery, University of California, San Francisco, San Francisco, CA, USA
| | - Thomas Kuner
- Department of Functional Neuroanatomy, University of Heidelberg, Heidelberg, Germany
| | - Donald Mabbott
- Department of Psychology, University of Toronto and Neuroscience & Mental Health Program, Research Institute, The Hospital for Sick Children, Toronto, Canada
| | - Michael Platten
- Department of Neurology, Medical Faculty Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Asya Rolls
- Department of Immunology, Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Erica K Sloan
- Monash Institute of Pharmaceutical Sciences, Drug Discovery Biology Theme, Monash University, Parkville, VIC, Australia
| | - Timothy C Wang
- Department of Medicine, Division of Digestive and Gastrointestinal Diseases, Columbia University, New York, NY, USA
| | - Wolfgang Wick
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg and Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Varun Venkataramani
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg and Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Functional Neuroanatomy, University of Heidelberg, Heidelberg, Germany.
| | - Michelle Monje
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA; Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA.
| |
Collapse
|
16
|
Liu J, Poojary MM, Zhu L, Williams AR, Lund MN. Phenolic Acid-Amino Acid Adducts Exert Distinct Immunomodulatory Effects in Macrophages Compared to Parent Phenolic Acids. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:2344-2355. [PMID: 36715127 DOI: 10.1021/acs.jafc.2c06658] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Caffeic acid (CA) and chlorogenic acid (CGA) are commonly found phenolic acids in plant-derived foods and beverages. Their corresponding adducts with cysteine (Cys) have been detected in coffee-containing beverages. However, despite the well-documented antioxidant and anti-inflammatory activity of CA and CGA, the immunomodulatory activities of the Cys adducts (CA-Cys and CGA-Cys) are unknown. The adducts were therefore synthesized, and their immunomodulatory effects were studied in lipopolysaccharide (LPS)-treated RAW 264.7 cells and compared to the activity of the parent phenolic acids. CA and CGA generally down-regulated the inflammatory responses. However, RNA-sequencing showed that the LPS-induced pathways related to Toll-like receptor signaling, chemokine signaling, and NOD-like receptor signaling, and JAK-STAT/MAPK signaling pathways were upregulated in adduct-treated cells relative to parent phenolic acids, while neurodegenerative disorder-related pathways and metabolic pathways were downregulated. Production of prostaglandin E2 (PGE2), interleukin-6, tumor necrosis factor-α (TNF-α), and reactive oxygen species (ROS) was all inhibited by CA and CGA (P < 0.05). PGE2 and TNF-α were further suppressed in adduct-stimulated cells (P < 0.05), but ROS production was increased. For example, TNF-α produced by 100 μM CGA-stimulated cells and 100 μM CGA-Cys adduct-stimulated cells were 4.46 ± 0.23 and 1.61 ± 0.18 ng/mL, respectively. Thus, the addition of the Cys moiety drastically alters the anti-inflammatory activity of phenolic compounds.
Collapse
Affiliation(s)
- Jingyuan Liu
- Department of Food Science, Faculty of Science, University of Copenhagen, Rolighedsvej 26, 1958 Frederiksberg C, Denmark
| | - Mahesha M Poojary
- Department of Food Science, Faculty of Science, University of Copenhagen, Rolighedsvej 26, 1958 Frederiksberg C, Denmark
| | - Ling Zhu
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 1870 Frederiksberg, Denmark
| | - Andrew R Williams
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 1870 Frederiksberg, Denmark
| | - Marianne N Lund
- Department of Food Science, Faculty of Science, University of Copenhagen, Rolighedsvej 26, 1958 Frederiksberg C, Denmark
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen N, Denmark
| |
Collapse
|
17
|
Role of nerves in neurofibromatosis type 1-related nervous system tumors. Cell Oncol (Dordr) 2022; 45:1137-1153. [PMID: 36327093 DOI: 10.1007/s13402-022-00723-3] [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] [Accepted: 09/22/2022] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Neurofibromatosis type 1 (NF1) is an autosomal dominant genetic disorder that affects nearly 1 in 3000 infants. Neurofibromin inactivation and NF1 gene mutations are involved in various aspects of neuronal function regulation, including neuronal development induction, electrophysiological activity elevation, growth factor expression, and neurotransmitter release. NF1 patients often exhibit a predisposition to tumor development, especially in the nervous system, resulting in the frequent occurrence of peripheral nerve sheath tumors and gliomas. Recent evidence suggests that nerves play a role in the development of multiple tumor types, prompting researchers to investigate the nerve as a vital component in and regulator of the initiation and progression of NF1-related nervous system tumors. CONCLUSION In this review, we summarize existing evidence about the specific effects of NF1 mutation on neurons and emerging research on the role of nerves in neurological tumor development, promising a new set of selective and targeted therapies for NF1-related tumors.
Collapse
|
18
|
Xia H, Deng L, Meng S, Liu X, Zheng C. Single-Cell Transcriptome Profiling Signatures and Alterations of Microglia Associated With Glioblastoma Associate Microglia Contribution to Tumor Formation. Pathol Oncol Res 2022; 28:1610067. [PMID: 35693633 PMCID: PMC9176381 DOI: 10.3389/pore.2022.1610067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 05/05/2022] [Indexed: 11/13/2022]
Abstract
Glioblastoma (GBM), which occasionally occurs in pediatric patients, is the most common tumor of the central nervous system in adults. Clinically, GBM is classified as low-grade to high-grade (from 1 to 4) and is characterized by late discovery, limited effective treatment methods, and poor efficacy. With the development of immunotherapy technology, effective GBM treatment strategies are of great significance. The main immune cells found in the GBM tumor microenvironment are macrophages and microglia (MG). Both these monocytes play important roles in the occurrence and development of GBM. Macrophages are recruited during tumorigenesis, whereas MG is present in the brain during embryonic development. Interestingly, the accumulation of these monocytes is inversely proportional to the survival of adult GBM patients but not the pediatric GBM patients. This study used single-cell RNA-seq data to reveal the heterogeneity of MG in tumor lesions and to explore the role of different MG subtypes in the occurrence and development of GBM. The results may help find new targets for immunotherapy of GBM.
Collapse
Affiliation(s)
- Hailong Xia
- Department of Neurosurgery, Chongqing Red Cross Hospital (Jiangbei District People's Hospital), Chongqing, China
| | - Lei Deng
- Department of Neurosurgery, Bishan District People's Hospital, Chongqing, China
| | - Shu Meng
- Internal Medicine, Chongqing Red Cross Hospital (Jiangbei District People's Hospital), Chongqing, China
| | - Xipeng Liu
- Department of Neurosurgery, Chongqing Red Cross Hospital (Jiangbei District People's Hospital), Chongqing, China
| | - Chao Zheng
- Department of Neurosurgery, Chongqing Red Cross Hospital (Jiangbei District People's Hospital), Chongqing, China
| |
Collapse
|
19
|
Anastasaki C, Mo J, Chen JK, Chatterjee J, Pan Y, Scheaffer SM, Cobb O, Monje M, Le LQ, Gutmann DH. Neuronal hyperexcitability drives central and peripheral nervous system tumor progression in models of neurofibromatosis-1. Nat Commun 2022; 13:2785. [PMID: 35589737 PMCID: PMC9120229 DOI: 10.1038/s41467-022-30466-6] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 05/03/2022] [Indexed: 11/16/2022] Open
Abstract
Neuronal activity is emerging as a driver of central and peripheral nervous system cancers. Here, we examined neuronal physiology in mouse models of the tumor predisposition syndrome Neurofibromatosis-1 (NF1), with different propensities to develop nervous system cancers. We show that central and peripheral nervous system neurons from mice with tumor-causing Nf1 gene mutations exhibit hyperexcitability and increased secretion of activity-dependent tumor-promoting paracrine factors. We discovered a neurofibroma mitogen (COL1A2) produced by peripheral neurons in an activity-regulated manner, which increases NF1-deficient Schwann cell proliferation, establishing that neurofibromas are regulated by neuronal activity. In contrast, mice with the Arg1809Cys Nf1 mutation, found in NF1 patients lacking neurofibromas or optic gliomas, do not exhibit neuronal hyperexcitability or develop these NF1-associated tumors. The hyperexcitability of tumor-prone Nf1-mutant neurons results from reduced NF1-regulated hyperpolarization-activated cyclic nucleotide-gated (HCN) channel function, such that neuronal excitability, activity-regulated paracrine factor production, and tumor progression are attenuated by HCN channel activation. Collectively, these findings reveal that NF1 mutations act at the level of neurons to modify tumor predisposition by increasing neuronal excitability and activity-regulated paracrine factor production. Neuronal activity is emerging as a driver of nervous system tumors. Here, the authors show in mouse models of Neurofibromatosis-1 (NF1) that Nf1 mutations differentially drive both central and peripheral nervous system tumor growth in mice through reduced hyperpolarization-activated cyclic nucleotide-gated (HCN) channel function.
Collapse
Affiliation(s)
- Corina Anastasaki
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Juan Mo
- Department of Dermatology, University of Texas, Southwestern, Dallas, TX, 75390, USA
| | - Ji-Kang Chen
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Jit Chatterjee
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Yuan Pan
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, 94305, USA
| | - Suzanne M Scheaffer
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Olivia Cobb
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Michelle Monje
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, 94305, USA.,Howard Hughes Medical Institute, Stanford University, Stanford, CA, 94305, USA
| | - Lu Q Le
- Department of Dermatology, University of Texas, Southwestern, Dallas, TX, 75390, USA
| | - David H Gutmann
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| |
Collapse
|
20
|
Codrici E, Popescu ID, Tanase C, Enciu AM. Friends with Benefits: Chemokines, Glioblastoma-Associated Microglia/Macrophages, and Tumor Microenvironment. Int J Mol Sci 2022; 23:ijms23052509. [PMID: 35269652 PMCID: PMC8910233 DOI: 10.3390/ijms23052509] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 02/17/2022] [Accepted: 02/22/2022] [Indexed: 12/19/2022] Open
Abstract
Glioma is the most common primary intracranial tumor and has the greatest prevalence of all brain tumors. Treatment resistance and tumor recurrence in GBM are mostly explained by considerable alterations within the tumor microenvironment, as well as extraordinary cellular and molecular heterogeneity. Soluble factors, extracellular matrix components, tissue-resident cell types, resident or newly recruited immune cells together make up the GBM microenvironment. Regardless of many immune cells, a profound state of tumor immunosuppression is supported and developed, posing a considerable hurdle to cancer cells' immune-mediated destruction. Several studies have suggested that various GBM subtypes present different modifications in their microenvironment, although the importance of the microenvironment in treatment response has yet to be determined. Understanding the microenvironment and how it changes after therapies is critical because it can influence the remaining invasive GSCs and lead to recurrence. This review article sheds light on the various components of the GBM microenvironment and their roles in tumoral development, as well as immune-related biological processes that support the interconnection/interrelationship between different cell types. Also, we summarize the current understanding of the modulation of soluble factors and highlight the dysregulated inflammatory chemokine/specific receptors cascades/networks and their significance in tumorigenesis, cancer-related inflammation, and metastasis.
Collapse
Affiliation(s)
- Elena Codrici
- Victor Babes National Institute of Pathology, 050096 Bucharest, Romania;
- Correspondence: (E.C.); (I.-D.P.); (A.-M.E.)
| | - Ionela-Daniela Popescu
- Victor Babes National Institute of Pathology, 050096 Bucharest, Romania;
- Correspondence: (E.C.); (I.-D.P.); (A.-M.E.)
| | - Cristiana Tanase
- Victor Babes National Institute of Pathology, 050096 Bucharest, Romania;
- Department of Clinical Biochemistry, Faculty of Medicine, Titu Maiorescu University, 031593 Bucharest, Romania
| | - Ana-Maria Enciu
- Victor Babes National Institute of Pathology, 050096 Bucharest, Romania;
- Department of Cell Biology and Histology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Correspondence: (E.C.); (I.-D.P.); (A.-M.E.)
| |
Collapse
|
21
|
Coleman DM, Wang Y, Yang ML, Hunker KL, Birt I, Bergin IL, Li JZ, Stanley JC, Ganesh SK. Molecular genetic evaluation of pediatric renovascular hypertension due to renal artery stenosis and abdominal aortic coarctation in neurofibromatosis type 1. Hum Mol Genet 2022; 31:334-346. [PMID: 34476477 PMCID: PMC8825256 DOI: 10.1093/hmg/ddab241] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 08/05/2021] [Accepted: 08/06/2021] [Indexed: 12/30/2022] Open
Abstract
The etiology of renal artery stenosis (RAS) and abdominal aortic coarctation (AAC) causing the midaortic syndrome (MAS), often resulting in renovascular hypertension (RVH), remains ill-defined. Neurofibromatosis type 1 (NF-1) is frequently observed in children with RVH. Consecutive pediatric patients (N = 102) presenting with RVH secondary to RAS with and without concurrent AAC were prospectively enrolled in a clinical data base, and blood, saliva and operative tissue, when available, were collected. Among the 102 children, 13 were having a concurrent clinical diagnosis of NF-1 (12.5%). Whole exome sequencing was performed for germline variant detection, and RNA-Seq analysis of NF1, MAPK pathway genes and MCP1 levels were undertaken in five NF-1 stenotic renal arteries, as well as control renal and mesenteric arteries from children with no known vasculopathy or NF-1. In 11 unrelated children with sequencing data, 11 NF1 genetic variants were identified, of which 10 had not been reported in gnomAD. Histologic analysis of NF-1 RAS specimens consistently revealed intimal thickening, disruption of the internal elastic lamina and medial thinning. Analysis of transcript expression in arterial lesions documented an approximately 5-fold reduction in NF1 expression, confirming heterozygosity, MAPK pathway activation and increased MCP1 expression. In summary, NF-1-related RVH in children is rare but often severe and progressive and, as such, important to recognize. It is associated with histologic and molecular features consistent with an aggressive adverse vascular remodeling process. Further research is necessary to define the mechanisms underlying these findings.
Collapse
Affiliation(s)
- Dawn M Coleman
- Vascular Surgery Section, Department of Surgery, Michigan Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Yu Wang
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Min-Lee Yang
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Kristina L Hunker
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Isabelle Birt
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Ingrid L Bergin
- Unit for Laboratory Animal Medicine, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Jun Z Li
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - James C Stanley
- Vascular Surgery Section, Department of Surgery, Michigan Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Santhi K Ganesh
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| |
Collapse
|
22
|
De Andrade Costa A, Chatterjee J, Cobb O, Sanapala S, Scheaffer S, Guo X, Dahiya S, Gutmann DH. RNA sequence analysis reveals ITGAL/CD11A as a stromal regulator of murine low-grade glioma growth. Neuro Oncol 2022; 24:14-26. [PMID: 34043012 PMCID: PMC8730775 DOI: 10.1093/neuonc/noab130] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Emerging insights from numerous laboratories have revealed important roles for nonneoplastic cells in the development and progression of brain tumors. One of these nonneoplastic cellular constituents, glioma-associated microglia (GAM), represents a unique population of brain monocytes within the tumor microenvironment that have been reported to both promote and inhibit glioma proliferation. To elucidate the role of GAM in the setting of low-grade glioma (LGG), we leveraged RNA sequencing meta-analysis, genetically engineered mouse strains, and human biospecimens. METHODS Publicly available disease-associated microglia (DAM) RNA-seq datasets were used, followed by immunohistochemistry and RNAScope validation. CD11a-deficient mouse microglia were used for in vitro functional studies, while LGG growth in mice was assessed using anti-CD11a neutralizing antibody treatment of Neurofibromatosis type 1 (Nf1) optic glioma mice in vivo. RESULTS We identified Itgal/CD11a enrichment in GAM relative to other DAM populations, which was confirmed in several independently generated murine models of Nf1 optic glioma. Moreover, ITGAL/CD11A expression was similarly increased in human LGG (pilocytic astrocytoma) specimens from several different datasets, specifically in microglia from these tumors. Using CD11a-knockout mice, CD11a expression was shown to be critical for murine microglia CX3CL1 receptor (Cx3cr1) expression and CX3CL1-directed motility, as well as glioma mitogen (Ccl5) production. Consistent with an instructive role for CD11a+ microglia in stromal control of LGG growth, antibody-mediated CD11a inhibition reduced mouse Nf1 LGG growth in vivo. CONCLUSIONS Collectively, these findings establish ITGAL/CD11A as a critical microglia regulator of LGG biology relevant to future stroma-targeted brain tumor treatment strategies.
Collapse
Affiliation(s)
| | - Jit Chatterjee
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Olivia Cobb
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Shilpa Sanapala
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Suzanne Scheaffer
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Xiaofan Guo
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Sonika Dahiya
- Department of Pathology, Washington University School of Medicine, St. Louis, MO, USA
| | - David H Gutmann
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
23
|
de Andrade Costa A, Chatterjee J, Cobb O, Cordell E, Chao A, Schaeffer S, Goldstein A, Dahiya S, Gutmann DH. Immune deconvolution and temporal mapping identifies stromal targets and developmental intervals for abrogating murine low-grade optic glioma formation. Neurooncol Adv 2022; 4:vdab194. [PMID: 35187488 PMCID: PMC8852255 DOI: 10.1093/noajnl/vdab194] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Brain tumor formation and progression are dictated by cooperative interactions between neoplastic and non-neoplastic cells. This stromal dependence is nicely illustrated by tumors arising in the Neurofibromatosis type 1 (NF1) cancer predisposition syndrome, where children develop low-grade optic pathway gliomas (OPGs). Using several authenticated Nf1-OPG murine models, we previously demonstrated that murine Nf1-OPG growth is regulated by T cell function and microglia Ccl5 production, such that their inhibition reduces tumor proliferation in vivo. While these interactions are critical for established Nf1-OPG tumor growth, their importance in tumor formation has not been explored. METHODS A combination of bulk and single-cell RNA mouse optic nerve sequencing, immunohistochemistry, T cell assays, and pharmacologic and antibody-mediated inhibition methods were used in these experiments. RESULTS We show that T cells and microglia are the main non-neoplastic immune cell populations in both murine and human LGGs. Moreover, we demonstrate that CD8+ T cells, the predominant LGG-infiltrating lymphocyte population, are selectively recruited through increased Ccl2 receptor (Ccr4) expression in CD8+, but not CD4+, T cells, in a NF1/RAS-dependent manner. Finally, we identify the times during gliomagenesis when microglia Ccl5 production (3-6 weeks of age) and Ccl2-mediated T cell infiltration (7-10 weeks of age) occur, such that temporally-restricted Ccl2 or Ccl5 inhibition abrogates tumor formation >3.5 months following the cessation of treatment. CONCLUSIONS Collectively, these findings provide proof-of-concept demonstrations that targeting stromal support during early gliomagenesis durably blocks murine LGG formation.
Collapse
Affiliation(s)
| | - Jit Chatterjee
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Olivia Cobb
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Elizabeth Cordell
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Astoria Chao
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Suzanne Schaeffer
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Andrea Goldstein
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Sonika Dahiya
- Department of Pathology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - David H Gutmann
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
24
|
Asthma reduces glioma formation by T cell decorin-mediated inhibition of microglia. Nat Commun 2021; 12:7122. [PMID: 34880260 PMCID: PMC8654836 DOI: 10.1038/s41467-021-27455-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 11/23/2021] [Indexed: 01/17/2023] Open
Abstract
To elucidate the mechanisms underlying the reduced incidence of brain tumors in children with Neurofibromatosis type 1 (NF1) and asthma, we leverage Nf1 optic pathway glioma (Nf1OPG) mice, human and mouse RNAseq data, and two different experimental asthma models. Following ovalbumin or house dust mite asthma induction at 4-6 weeks of age (WOA), Nf1OPG mouse optic nerve volumes and proliferation are decreased at 12 and 24 WOA, indicating no tumor development. This inhibition is accompanied by reduced expression of the microglia-produced optic glioma mitogen, Ccl5. Human and murine T cell transcriptome analyses reveal that inhibition of microglia Ccl5 production results from increased T cell expression of decorin, which blocks Ccl4-mediated microglia Ccl5 expression through reduced microglia NFκB signaling. Decorin or NFκB inhibitor treatment of Nf1OPG mice at 4-6 WOA inhibits tumor formation at 12 WOA, thus establishing a potential mechanistic etiology for the attenuated glioma incidence observed in children with asthma.
Collapse
|
25
|
Bornhorst M. New insights into low grade glioma tumor microenvironment for improved patient management. Neuro Oncol 2021; 24:27-28. [PMID: 34515792 DOI: 10.1093/neuonc/noab224] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Miriam Bornhorst
- Department of Pediatric Hematology-Oncology, Children's National Hospital, Washington, DC.,Gilbert Family Neurofibromatosis Institute, Children's National Hospital, Washington, DC.,Center for Genetics Research, Children's National Hospital, Washington, DC
| |
Collapse
|
26
|
Rabab’h O, Gharaibeh A, Al-Ramadan A, Ismail M, Shah J. Pharmacological Approaches in Neurofibromatosis Type 1-Associated Nervous System Tumors. Cancers (Basel) 2021; 13:cancers13153880. [PMID: 34359780 PMCID: PMC8345673 DOI: 10.3390/cancers13153880] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 07/28/2021] [Accepted: 07/28/2021] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Neurofibromatosis type 1 (NF1) is a common cancer predisposition genetic disease that is associated with significant morbidity and mortality. In this literature review, we discuss the major pathways in the nervous system that are affected by NF1, tumors that are associated with NF1, drugs that target these pathways, and genetic models of NF1. We also summarize the latest updates from clinical trials that are evaluating pharmacological agents to treat these tumors and discuss the efforts that are being made to cure the disease in the future Abstract Neurofibromatosis type 1 is an autosomal dominant genetic disease and a common tumor predisposition syndrome that affects 1 in 3000 to 4000 patients in the USA. Although studies have been conducted to better understand and manage this disease, the underlying pathogenesis of neurofibromatosis type 1 has not been completely elucidated, and this disease is still associated with significant morbidity and mortality. Treatment options are limited to surgery with chemotherapy for tumors in cases of malignant transformation. In this review, we summarize the advances in the development of targeted pharmacological interventions for neurofibromatosis type 1 and related conditions.
Collapse
Affiliation(s)
- Omar Rabab’h
- Insight Research Institute, Flint, MI 48507, USA; (O.R.); (A.G.); (A.A.-R.); (M.I.)
- Center for Cognition and Neuroethics, University of Michigan-Flint, Flint, MI 48502, USA
| | - Abeer Gharaibeh
- Insight Research Institute, Flint, MI 48507, USA; (O.R.); (A.G.); (A.A.-R.); (M.I.)
- Center for Cognition and Neuroethics, University of Michigan-Flint, Flint, MI 48502, USA
- Insight Institute of Neurosurgery & Neuroscience, Flint, MI 48507, USA
- Insight Surgical Hospital, Warren, MI 48091, USA
| | - Ali Al-Ramadan
- Insight Research Institute, Flint, MI 48507, USA; (O.R.); (A.G.); (A.A.-R.); (M.I.)
- Center for Cognition and Neuroethics, University of Michigan-Flint, Flint, MI 48502, USA
| | - Manar Ismail
- Insight Research Institute, Flint, MI 48507, USA; (O.R.); (A.G.); (A.A.-R.); (M.I.)
| | - Jawad Shah
- Insight Research Institute, Flint, MI 48507, USA; (O.R.); (A.G.); (A.A.-R.); (M.I.)
- Center for Cognition and Neuroethics, University of Michigan-Flint, Flint, MI 48502, USA
- Insight Institute of Neurosurgery & Neuroscience, Flint, MI 48507, USA
- Insight Surgical Hospital, Warren, MI 48091, USA
- Department of Medicine, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA
- Correspondence:
| |
Collapse
|
27
|
Milde T, Rodriguez FJ, Barnholtz-Sloan JS, Patil N, Eberhart CG, Gutmann DH. Reimagining Pilocytic Astrocytomas in the Context of Pediatric Low-Grade Gliomas. Neuro Oncol 2021; 23:1634-1646. [PMID: 34131743 DOI: 10.1093/neuonc/noab138] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Pediatric low-grade gliomas (pLGGs) are the most common brain tumor in children, and are associated with life-long clinical morbidity. Relative to their high-grade adult counterparts or other malignant childhood brain tumors, there is a paucity of authenticated preclinical models for these pediatric low-grade gliomas and an incomplete understanding of their molecular and cellular pathogenesis. While large scale genomic profiling efforts have identified the majority of pathogenic driver mutations, which converge on the MAPK/ERK signaling pathway, it is now appreciated that these events may not be sufficient by themselves for gliomagenesis and clinical progression. In light of the recent World Health Organization reclassification of pLGGs, and pilocytic astrocytoma (PA) in particular, we review our current understanding of these pediatric brain tumors, provide a conceptual framework for future mechanistic studies, and outline the challenges and pressing needs for the pLGG clinical and research communities.
Collapse
Affiliation(s)
- Till Milde
- Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany.,Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany.,Department of Pediatric Hematology and Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - Fausto J Rodriguez
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore MD, USA
| | - Jill S Barnholtz-Sloan
- Department of Population and Quantitative Health Sciences, Case Western Reserve School of Medicine, Cleveland OH, USA.,University Hospitals, Cleveland OH, USA.,Central Brain Tumor Registry of the United States (CBTRUS), Hinsdale, IL, USA
| | - Nirav Patil
- University Hospitals, Cleveland OH, USA.,Central Brain Tumor Registry of the United States (CBTRUS), Hinsdale, IL, USA
| | - Charles G Eberhart
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore MD, USA
| | - David H Gutmann
- Department of Neurology, Washington University School of Medicine, St. Louis MO, USA
| |
Collapse
|
28
|
Packer RJ, Iavarone A, Jones DTW, Blakeley JO, Bouffet E, Fisher MJ, Hwang E, Hawkins C, Kilburn L, MacDonald T, Pfister SM, Rood B, Rodriguez FJ, Tabori U, Ramaswamy V, Zhu Y, Fangusaro J, Johnston SA, Gutmann DH. Implications of new understandings of gliomas in children and adults with NF1: report of a consensus conference. Neuro Oncol 2021; 22:773-784. [PMID: 32055852 PMCID: PMC7283027 DOI: 10.1093/neuonc/noaa036] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Gliomas are the most common primary central nervous system tumors occurring in children and adults with neurofibromatosis type 1 (NF1). Over the past decade, discoveries of the molecular basis of low-grade gliomas (LGGs) have led to new approaches for diagnosis and treatments. However, these new understandings have not been fully applied to the management of NF1-associated gliomas. A consensus panel consisting of experts in NF1 and gliomas was convened to review the current molecular knowledge of NF1-associated low-grade “transformed” and high-grade gliomas; insights gained from mouse models of NF1-LGGs; challenges in diagnosing and treating older patients with NF1-associated gliomas; and advances in molecularly targeted treatment and potential immunologic treatment of these tumors. Next steps are recommended to advance the management and outcomes for NF1-associated gliomas.
Collapse
Affiliation(s)
- Roger J Packer
- Center for Neuroscience and Behavioral Medicine, Washington, DC, USA.,Gilbert Family Neurofibromatosis Institute, Brain Tumor Institute, and Children's National Hospital, Washington, DC, USA
| | - Antonio Iavarone
- Departments of Neurology and Pathology Institute for Cancer Genetics Columbia University Medical Center, New York, New York, USA
| | - David T W Jones
- Division of Pediatric Neuro-Oncology German Cancer Research Center Hopp Children's Cancer Center Heidelberg, Germany
| | - Jaishri O Blakeley
- Departments of Neurology; Oncology; Neurosurgery, Baltimore, Maryland, USA
| | - Eric Bouffet
- Pediatric Neuro-Oncology Program; Research Institute; and The Arthur and Sonia Labatt; Brain Tumor Research Centre, Hospital for Sick Children, Toronto, Canada
| | - Michael J Fisher
- Department of Pediatric Oncology; Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Eugene Hwang
- Gilbert Family Neurofibromatosis Institute, Brain Tumor Institute, and Children's National Hospital, Washington, DC, USA
| | - Cynthia Hawkins
- Pediatric Neuro-Oncology Program; Research Institute; and The Arthur and Sonia Labatt; Brain Tumor Research Centre, Hospital for Sick Children, Toronto, Canada
| | - Lindsay Kilburn
- Gilbert Family Neurofibromatosis Institute, Brain Tumor Institute, and Children's National Hospital, Washington, DC, USA
| | - Tobey MacDonald
- Department of Pediatrics; Emory University School of Medicine, Atlanta, Georgia, USA
| | - Stefan M Pfister
- Division of Pediatric Neuro-Oncology German Cancer Research Center Hopp Children's Cancer Center Heidelberg, Germany
| | - Brian Rood
- Gilbert Family Neurofibromatosis Institute, Brain Tumor Institute, and Children's National Hospital, Washington, DC, USA
| | - Fausto J Rodriguez
- Pathology; The Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Uri Tabori
- Pediatric Neuro-Oncology Program; Research Institute; and The Arthur and Sonia Labatt; Brain Tumor Research Centre, Hospital for Sick Children, Toronto, Canada
| | - Vijay Ramaswamy
- Pediatric Neuro-Oncology Program; Research Institute; and The Arthur and Sonia Labatt; Brain Tumor Research Centre, Hospital for Sick Children, Toronto, Canada
| | - Yuan Zhu
- Gilbert Family Neurofibromatosis Institute, Brain Tumor Institute, and Children's National Hospital, Washington, DC, USA
| | - Jason Fangusaro
- Department of Pediatrics; Emory University School of Medicine, Atlanta, Georgia, USA
| | - Stephen A Johnston
- Center for Innovations in Medicine; Biodesign Institute; Arizona State University, Tempe, Arizona, USA
| | - David H Gutmann
- Department of Neurology; Washington University, St Louis, Missouri, USA
| |
Collapse
|
29
|
Lah Turnšek T, Jiao X, Novak M, Jammula S, Cicero G, Ashton AW, Joyce D, Pestell RG. An Update on Glioblastoma Biology, Genetics, and Current Therapies: Novel Inhibitors of the G Protein-Coupled Receptor CCR5. Int J Mol Sci 2021; 22:4464. [PMID: 33923334 PMCID: PMC8123168 DOI: 10.3390/ijms22094464] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 04/20/2021] [Accepted: 04/21/2021] [Indexed: 02/07/2023] Open
Abstract
The mechanisms governing therapeutic resistance of the most aggressive and lethal primary brain tumor in adults, glioblastoma, have increasingly focused on tumor stem cells. These cells, protected by the periarteriolar hypoxic GSC niche, contribute to the poor efficacy of standard of care treatment of glioblastoma. Integrated proteogenomic and metabolomic analyses of glioblastoma tissues and single cells have revealed insights into the complex heterogeneity of glioblastoma and stromal cells, comprising its tumor microenvironment (TME). An additional factor, which isdriving poor therapy response is the distinct genetic drivers in each patient's tumor, providing the rationale for a more individualized or personalized approach to treatment. We recently reported that the G protein-coupled receptor CCR5, which contributes to stem cell expansion in other cancers, is overexpressed in glioblastoma cells. Overexpression of the CCR5 ligand CCL5 (RANTES) in glioblastoma completes a potential autocrine activation loop to promote tumor proliferation and invasion. CCL5 was not expressed in glioblastoma stem cells, suggesting a need for paracrine activation of CCR5 signaling by the stromal cells. TME-associated immune cells, such as resident microglia, infiltrating macrophages, T cells, and mesenchymal stem cells, possibly release CCR5 ligands, providing heterologous signaling between stromal and glioblastoma stem cells. Herein, we review current therapies for glioblastoma, the role of CCR5 in other cancers, and the potential role for CCR5 inhibitors in the treatment of glioblastoma.
Collapse
Affiliation(s)
- Tamara Lah Turnšek
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, 1000 Ljubljana, Slovenia;
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Xuanmao Jiao
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, 3805 Old Easton Road, Doylestown, PA 18902, USA;
- School of Medicine, Xavier University, Santa Helenastraat #23, Oranjestad, Aruba; (S.J.); (G.C.); (A.W.A.)
| | - Metka Novak
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, 1000 Ljubljana, Slovenia;
| | - Sriharsha Jammula
- School of Medicine, Xavier University, Santa Helenastraat #23, Oranjestad, Aruba; (S.J.); (G.C.); (A.W.A.)
| | - Gina Cicero
- School of Medicine, Xavier University, Santa Helenastraat #23, Oranjestad, Aruba; (S.J.); (G.C.); (A.W.A.)
| | - Anthony W. Ashton
- School of Medicine, Xavier University, Santa Helenastraat #23, Oranjestad, Aruba; (S.J.); (G.C.); (A.W.A.)
- Division of Perinatal Research, Kolling Institute, Northern Sydney Local Health District, St Leonards, NSW 2065, Australia
- Sydney Medical School Northern, University of Sydney, Sydney, NSW 2006, Australia
- Lankenau Institute for Medical Research Philadelphia, 100 East Lancaster Ave., Wynnewood, PA 19069, USA
| | - David Joyce
- Medical School, Faculty of Health and Medical Sciences, The University of Western Australia, 35 Stirling Highway, Crawley, WA 6009, Australia;
| | - Richard G. Pestell
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, 3805 Old Easton Road, Doylestown, PA 18902, USA;
- School of Medicine, Xavier University, Santa Helenastraat #23, Oranjestad, Aruba; (S.J.); (G.C.); (A.W.A.)
- The Wistar Cancer Center, Philadelphia, PA 19107, USA
| |
Collapse
|
30
|
Gu YH, Cui XW, Ren JY, Long MM, Wang W, Wei CJ, Aimaier R, Li YH, Chung MH, Gu B, Li QF, Wang ZC. Selection of internal references for RT-qPCR assays in Neurofibromatosis type 1 (NF1) related Schwann cell lines. PLoS One 2021; 16:e0241821. [PMID: 33630851 PMCID: PMC7906369 DOI: 10.1371/journal.pone.0241821] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 02/02/2021] [Indexed: 11/20/2022] Open
Abstract
Real-time quantitative PCR (RT-qPCR) has been widely applied in uncovering disease mechanisms and screening potential biomarkers. Internal reference gene selection determines the accuracy and reproducibility of data analyses. The aim of this study was to identify the optimal reference genes for the relative quantitative analysis of RT-qPCR in fourteen NF1 related cell lines, including non-tumor, benign and malignant Schwann cell lines. The expression characteristics of eleven candidate reference genes (RPS18, ACTB, B2M, GAPDH, PPIA, HPRT1, TBP, UBC, RPLP0, TFRC and RPL32) were screened and analyzed by four software programs: geNorm, NormFinder, BestKeeper and RefFinder. Results showed that GAPDH, the most frequently used internal reference gene, was significantly unstable between various cell lines. The combinational use of two reference genes (PPIA and TBP) was optimal in malignant Schwann cell lines and the use of single reference genes (PPIA or PRLP0) alone or in combination was optimal in benign Schwann cell lines. These recommended internal reference gene selections may improve the accuracy and reproducibility of RT-qPCR in gene expression analyses of NF1 related tumors.
Collapse
Affiliation(s)
- Yi-Hui Gu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xi-Wei Cui
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie-Yi Ren
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Man-Mei Long
- Department of Pathology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Cheng-Jiang Wei
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rehanguli Aimaier
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yue-Hua Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Man-Hon Chung
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bin Gu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qing-Feng Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- * E-mail: (QFL); (ZCW)
| | - Zhi-Chao Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- * E-mail: (QFL); (ZCW)
| |
Collapse
|
31
|
Brosseau JP, Sathe AA, Wang Y, Nguyen T, Glass DA, Xing C, Le LQ. Human cutaneous neurofibroma matrisome revealed by single-cell RNA sequencing. Acta Neuropathol Commun 2021; 9:11. [PMID: 33413690 PMCID: PMC7792184 DOI: 10.1186/s40478-020-01103-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 12/13/2020] [Indexed: 12/12/2022] Open
Abstract
Neurofibromatosis Type I (NF1) is a neurocutaneous genetic syndrome characterized by a wide spectrum of clinical presentations, including benign peripheral nerve sheath tumor called neurofibroma. These tumors originate from the Schwann cell lineage but other cell types as well as extracellular matrix (ECM) in the neurofibroma microenvironment constitute the majority of the tumor mass. In fact, collagen accounts for up to 50% of the neurofibroma's dry weight. Although the presence of collagens in neurofibroma is indisputable, the exact repertoire of ECM genes and ECM-associated genes (i.e. the matrisome) and their functions are unknown. Here, transcriptome profiling by single-cell RNA sequencing reveals the matrisome of human cutaneous neurofibroma (cNF). We discovered that classic pro-fibrogenic collagen I myofibroblasts are rare in neurofibroma. In contrast, collagen VI, a pro-tumorigenic ECM, is abundant and mainly secreted by neurofibroma fibroblasts. This study also identified potential cell type-specific markers to further elucidate the biology of the cNF microenvironment.
Collapse
|
32
|
Guo X, Pan Y, Gutmann DH. Genetic and genomic alterations differentially dictate low-grade glioma growth through cancer stem cell-specific chemokine recruitment of T cells and microglia. Neuro Oncol 2020; 21:1250-1262. [PMID: 31111915 DOI: 10.1093/neuonc/noz080] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND One of the clinical hallmarks of low-grade gliomas (LGGs) arising in children with the neurofibromatosis type 1 (NF1) cancer predisposition syndrome is significant clinical variability with respect to tumor growth, associated neurologic deficits, and response to therapy. Numerous factors could contribute to this clinical heterogeneity, including the tumor cell of origin, the specific germline NF1 gene mutation, and the coexistence of additional genomic alterations. Since human specimens are rarely acquired, and have proven difficult to maintain in vitro or as xenografts in vivo, we have developed a series of Nf1 mutant optic glioma mouse strains representing each of these contributing factors. METHODS Optic glioma stem cells (o-GSCs) were generated from this collection of Nf1 genetically engineered mice, and analyzed for their intrinsic growth properties, as well as the production of chemokines that could differentially attract T cells and microglia. RESULTS The observed differences in Nf1 optic glioma growth are not the result of cell autonomous growth properties of o-GSCs, but rather the unique patterns of o-GSC chemokine expression, which differentially attract T cells and microglia. This immune profile collectively dictates the levels of chemokine C-C ligand 5 (Ccl5) expression, the key stromal factor that drives murine Nf1 optic glioma growth. CONCLUSIONS These findings reveal that genetic and genomic alterations create murine LGG biological heterogeneity through the differential recruitment of T cells and microglia by o-GSC-produced chemokines, which ultimately determine the expression of stromal factors that drive tumor growth.
Collapse
Affiliation(s)
- Xiaofan Guo
- Department of Neurology, Washington University School of Medicine, St Louis, Missouri
| | - Yuan Pan
- Department of Neurology, Washington University School of Medicine, St Louis, Missouri
| | - David H Gutmann
- Department of Neurology, Washington University School of Medicine, St Louis, Missouri
| |
Collapse
|
33
|
DeCordova S, Shastri A, Tsolaki AG, Yasmin H, Klein L, Singh SK, Kishore U. Molecular Heterogeneity and Immunosuppressive Microenvironment in Glioblastoma. Front Immunol 2020; 11:1402. [PMID: 32765498 PMCID: PMC7379131 DOI: 10.3389/fimmu.2020.01402] [Citation(s) in RCA: 199] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 06/01/2020] [Indexed: 12/14/2022] Open
Abstract
Glioblastoma (GBM) is the most aggressive primary brain tumor in adults, with a poor prognosis, despite surgical resection combined with radio- and chemotherapy. The major clinical obstacles contributing to poor GBM prognosis are late diagnosis, diffuse infiltration, pseudo-palisading necrosis, microvascular proliferation, and resistance to conventional therapy. These challenges are further compounded by extensive inter- and intra-tumor heterogeneity and the dynamic plasticity of GBM cells. The complex heterogeneous nature of GBM cells is facilitated by the local inflammatory tumor microenvironment, which mostly induces tumor aggressiveness and drug resistance. An immunosuppressive tumor microenvironment of GBM provides multiple pathways for tumor immune evasion. Infiltrating immune cells, mostly tumor-associated macrophages, comprise much of the non-neoplastic population in GBM. Further understanding of the immune microenvironment of GBM is essential to make advances in the development of immunotherapeutics. Recently, whole-genome sequencing, epigenomics and transcriptional profiling have significantly helped improve the prognostic and therapeutic outcomes of GBM patients. Here, we discuss recent genomic advances, the role of innate and adaptive immune mechanisms, and the presence of an established immunosuppressive GBM microenvironment that suppresses and/or prevents the anti-tumor host response.
Collapse
Affiliation(s)
- Syreeta DeCordova
- Biosciences, College of Health and Life Sciences, Brunel University London, London, United Kingdom
| | - Abhishek Shastri
- Central and North West London NHS Foundation Trust, London, United Kingdom
| | - Anthony G Tsolaki
- Biosciences, College of Health and Life Sciences, Brunel University London, London, United Kingdom
| | - Hadida Yasmin
- Immunology and Cell Biology Laboratory, Department of Zoology, Cooch Behar Panchanan Barma University, Cooch Behar, India
| | - Lukas Klein
- Department of Gastroenterology and Gastroenterology Oncology, University Medical Centre, Göttingen, Germany
| | - Shiv K Singh
- Department of Gastroenterology and Gastroenterology Oncology, University Medical Centre, Göttingen, Germany
| | - Uday Kishore
- Biosciences, College of Health and Life Sciences, Brunel University London, London, United Kingdom
| |
Collapse
|
34
|
The CCL5/CCR5 Axis in Cancer Progression. Cancers (Basel) 2020; 12:cancers12071765. [PMID: 32630699 PMCID: PMC7407580 DOI: 10.3390/cancers12071765] [Citation(s) in RCA: 250] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 06/25/2020] [Accepted: 06/30/2020] [Indexed: 02/07/2023] Open
Abstract
Tumor cells can “hijack” chemokine networks to support tumor progression. In this context, the C-C chemokine ligand 5/C-C chemokine receptor type 5 (CCL5/CCR5) axis is gaining increasing attention, since abnormal expression and activity of CCL5 and its receptor CCR5 have been found in hematological malignancies and solid tumors. Numerous preclinical in vitro and in vivo studies have shown a key role of the CCL5/CCR5 axis in cancer, and thus provided the rationale for clinical trials using the repurposed drug maraviroc, a CCR5 antagonist used to treat HIV/AIDS. This review summarizes current knowledge on the role of the CCL5/CCR5 axis in cancer. First, it describes the involvement of the CCL5/CCR5 axis in cancer progression, including autocrine and paracrine tumor growth, ECM (extracellular matrix) remodeling and migration, cancer stem cell expansion, DNA damage repair, metabolic reprogramming, and angiogenesis. Then, it focuses on individual hematological and solid tumors in which CCL5 and CCR5 have been studied preclinically. Finally, it discusses clinical trials of strategies to counteract the CCL5/CCR5 axis in different cancers using maraviroc or therapeutic monoclonal antibodies.
Collapse
|
35
|
Brosseau JP, Liao CP, Le LQ. Translating current basic research into future therapies for neurofibromatosis type 1. Br J Cancer 2020; 123:178-186. [PMID: 32439933 PMCID: PMC7374719 DOI: 10.1038/s41416-020-0903-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 03/25/2020] [Accepted: 05/01/2020] [Indexed: 12/12/2022] Open
Abstract
Neurofibromatosis type 1 (NF1) is a hereditary tumour syndrome that predisposes to benign and malignant tumours originating from neural crest cells. Biallelic inactivation of the tumour-suppressor gene NF1 in glial cells in the skin, along a nerve plexus or in the brain results in the development of benign tumours: cutaneous neurofibroma, plexiform neurofibroma and glioma, respectively. Despite more than 40 years of research, only one medication was recently approved for treatment of plexiform neurofibroma and no drugs have been specifically approved for the management of other tumours. Work carried out over the past several years indicates that inhibiting different cellular signalling pathways (such as Hippo, Janus kinase/signal transducer and activator of transcription, mitogen-activated protein kinase and those mediated by sex hormones) in tumour cells or targeting cells in the microenvironment (nerve cells, macrophages, mast cells and T cells) might benefit NF1 patients. In this review, we outline previous strategies aimed at targeting these signalling pathways or cells in the microenvironment, agents that are currently in clinical trials, and the latest advances in basic research that could culminate in the development of novel therapeutics for patients with NF1.
Collapse
Affiliation(s)
- Jean-Philippe Brosseau
- Department of Dermatology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, 75390-9069, USA.
- Department of Biochemistry and Functional Genomics, University of Sherbrooke, Sherbrooke, QC, J1E 4K8, Canada.
| | - Chung-Ping Liao
- Department of Dermatology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, 75390-9069, USA
| | - Lu Q Le
- Department of Dermatology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, 75390-9069, USA.
- Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, 75390-9069, USA.
- UTSW Comprehensive Neurofibromatosis Clinic, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, 75390-9069, USA.
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, 75390-9069, USA.
| |
Collapse
|
36
|
Midkine activation of CD8 + T cells establishes a neuron-immune-cancer axis responsible for low-grade glioma growth. Nat Commun 2020; 11:2177. [PMID: 32358581 PMCID: PMC7195398 DOI: 10.1038/s41467-020-15770-3] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 03/26/2020] [Indexed: 12/17/2022] Open
Abstract
Brain tumors (gliomas) are heterogeneous cellular ecosystems, where non-neoplastic monocytic cells have emerged as key regulators of tumor maintenance and progression. However, relative to macrophages/microglia, comparatively less is known about the roles of neurons and T cells in glioma pathobiology. Herein, we leverage genetically engineered mouse models and human biospecimens to define the axis in which neurons, T cells, and microglia interact to govern Neurofibromatosis-1 (NF1) low-grade glioma (LGG) growth. NF1-mutant human and mouse brain neurons elaborate midkine to activate naïve CD8+ T cells to produce Ccl4, which induces microglia to produce a key LGG growth factor (Ccl5) critical for LGG stem cell survival. Importantly, increased CCL5 expression is associated with reduced survival in patients with LGG. The elucidation of the critical intercellular dependencies that constitute the LGG neuroimmune axis provides insights into the role of neurons and immune cells in controlling glioma growth, relevant to future therapeutic targeting. The role of neurons and T cells in glioma progression remains poorly understood. Here the authors show that midkine-dependent activation of a neuron-T cell-microglia axis promotes the growth of optic pathway gliomas.
Collapse
|
37
|
Gutmann DH, Kettenmann H. Microglia/Brain Macrophages as Central Drivers of Brain Tumor Pathobiology. Neuron 2020; 104:442-449. [PMID: 31697921 DOI: 10.1016/j.neuron.2019.08.028] [Citation(s) in RCA: 194] [Impact Index Per Article: 38.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 07/18/2019] [Accepted: 08/16/2019] [Indexed: 12/21/2022]
Abstract
One of the most common brain tumors in children and adults is glioma or astrocytoma. There are few effective therapies for these cancers, and patients with malignant glioma fare poorly, even after aggressive surgery, chemotherapy, and radiation. Over the past decade, it is now appreciated that these tumors are composed of numerous distinct neoplastic and non-neoplastic cell populations, which could each influence overall tumor biology and response to therapy. Among these noncancerous cell types, monocytes (microglia and macrophages) predominate. In this Review, we discuss the complex interactions involving microglia and macrophages relevant to glioma formation, progression, and response to therapy.
Collapse
Affiliation(s)
- David H Gutmann
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Helmut Kettenmann
- Cellular Neurosciences, Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin 13125, Germany.
| |
Collapse
|
38
|
Ni J, Liu S, Qi F, Li X, Yu S, Feng J, Zheng Y. Screening TCGA database for prognostic genes in lower grade glioma microenvironment. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:209. [PMID: 32309356 PMCID: PMC7154476 DOI: 10.21037/atm.2020.01.73] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Background To identify prognostic hub genes which associated with tumor microenvironment (TME) in lower grade glioma (LGG) of central nervous system. Methods We downloaded LGG patients gene transcriptome profiles of the central nervous system in The Cancer Genome Atlas (TCGA) database. Clinical characteristics and survival data through the Genomic Data Commons (GDC) tool were extracted. We used limma package for normalization processing. Scores of immune, stromal and ESTIMATE were calculated using ESTIMATE algorithm. Then, box plots were applied to explore the association between immune scores, stromal scores, ESTIMATE scores and histological type, tumor grade. Kaplan-Meier (K-M) analysis was utilized to explore the prognostic value of scores. Furthermore, heatmaps and volcano plots were applied for visualizing expression of differential expressed-gene screening and cluster analysis. Venn plots were constructed to screen the intersected differentially expressed genes (DEGs). In addition, enrichment of functions and signaling pathways and Gene Set Enrichment Analysis (GESA) of the DEGs were performed. Then we used protein-protein interaction (PPI) network and Cytoscape software to identify hub genes. We evaluated the prognostic value of hub genes and risk score (RS) calculated based on multivariate cox regression analysis. Finally, relationships of hub genes with the TME of LGG patients were evaluated based on tumor immune estimation resource (TIMER) database. Results Gene expression profiles and clinical data of 514 LGG samples were extracted and the results revealed that higher scores were significantly related with histological types and higher tumor grade (P<0.0001, respectively). Besides, higher scores were associated with worse survival outcomes in immune scores (P=0.0167), stromal scores (P=0.0035) and ESTIMATE scores (P=0.0190). Then, 785 up-regulated intersected genes and 357 down-regulated intersected genes were revealed. Functional enrichment analysis revealed that intersected genes were associated with immune response, inflammatory response, plasma membrane and receptor activity. After PPI network construction and cytoHubba analysis, 25 tumor immune-related hub genes were identified and enriched pathways were identified by GSEA. Besides, receiver operating characteristic (ROC) curves showed significantly predictive accuracy [area under curve (AUC) =0.771] of RS. Furthermore, significant prognostic values of hub genes were observed, and the relationships between hub genes and LGG TME were demonstrated. Conclusions We identified 25 TME-related genes which significantly associated with overall survival in patients with central nervous system LGG from TCGA database.
Collapse
Affiliation(s)
- Jie Ni
- Department of Medical Oncology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing 210009, China
| | - Siwen Liu
- Research Center for Clinical Oncology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing 210009, China
| | - Feng Qi
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Xiao Li
- Department of Urology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing 210009, China
| | - Shaorong Yu
- Department of Medical Oncology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing 210009, China
| | - Jifeng Feng
- Department of Medical Oncology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing 210009, China
| | - Yuxiao Zheng
- Department of Urology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing 210009, China
| |
Collapse
|
39
|
Yu-Ju Wu C, Chen CH, Lin CY, Feng LY, Lin YC, Wei KC, Huang CY, Fang JY, Chen PY. CCL5 of glioma-associated microglia/macrophages regulates glioma migration and invasion via calcium-dependent matrix metalloproteinase 2. Neuro Oncol 2020; 22:253-266. [PMID: 31593589 PMCID: PMC7032635 DOI: 10.1093/neuonc/noz189] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 10/03/2019] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Glioma-associated microglia/macrophages (GAMs) comprise macrophages of peripheral origin and brain-intrinsic microglia, which support tumor progression. Chemokine C-C ligand 5 (CCL5) is an inflammatory mediator produced by immune cells and is involved in tumor growth and migration in several cancers, including glioma. However, the mechanisms detailing how CCL5 facilitates glioma invasion remain largely unresolved. METHODS Glioma migration and invasion were determined by wound healing, transwell assay, and 3D µ-slide chemotaxis assay. The expression levels of CCL5, CD68, matrix metalloproteinase 2 (MMP2), phosphorylated Ca2+/calmodulin-dependent protein kinase II (p-CaMKII), p-Akt, and phosphorylated proline-rich tyrosine kinase 2 were determined by cytokine array, quantitative PCR, western blot, or immunohistochemistry. Zymography and intracellular calcium assays were used to analyze MMP2 activity and intracellular calcium levels, respectively. RESULTS CCL5 modulated the migratory and invasive activities of human glioma cells in association with MMP2 expression. In response to CCL5, glioma cells underwent a synchronized increase in intracellular calcium levels and p-CaMKII and p-Akt expression levels. CCL5-directed glioma invasion and increases in MMP2 were suppressed after inhibition of p-CaMKII. Glioma cells tended to migrate toward GAM-conditioned media activated by granulocyte-macrophage colony-stimulating factor (GM-CSF) in which CCL5 was abundant. This homing effect was associated with MMP2 upregulation, and could be ameliorated either by controlling intracellular and extracellular calcium levels or by CCL5 antagonism. Clinical results also revealed the associations between CCL5 and GAM activation. CONCLUSION Our results suggest that modulation of glioma CaMKII may restrict the effect of CCL5 on glioma invasion and could be a potential therapeutic target for alleviating glioma growth.
Collapse
Affiliation(s)
- Caren Yu-Ju Wu
- Graduate Institute of Biomedical Sciences, Pharmaceutics Laboratory, Graduate Institute of Natural Products, Chang Gung University, Taoyuan, Taiwan
- Department of Neurosurgery, Keelung Chang Gung Memorial Hospital, Keelung, Taiwan
| | - Chia-Hua Chen
- Department of Neurosurgery, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Chun-Yen Lin
- Department of Gastroenterology and Hepatology, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
- School of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Li-Ying Feng
- Department of Neurosurgery, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Yung-Chang Lin
- Department of Gastroenterology and Hepatology, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Kuo-Chen Wei
- Department of Neurosurgery, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
- School of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chiung-Yin Huang
- Department of Neurosurgery, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Jia-You Fang
- Graduate Institute of Biomedical Sciences, Pharmaceutics Laboratory, Graduate Institute of Natural Products, Chang Gung University, Taoyuan, Taiwan
- Department of Anesthesiology, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Research Center for Food and Cosmetic Safety and Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, Taoyuan, Taiwan
| | - Pin-Yuan Chen
- Department of Neurosurgery, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Department of Neurosurgery, Keelung Chang Gung Memorial Hospital, Keelung, Taiwan
- School of Medicine, Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
40
|
Abstract
Brain tumors are complex cellular ecosystems, composed of populations of both neoplastic and non-neoplastic cell types. While the contributions of the cancer cells in low-grade and high-grade gliomas have been extensively studied, there is comparatively less known about the contributions of the non-neoplastic cells in these tumors. As such, a large proportion of the non-neoplastic cells in gliomas are resident brain microglia, infiltrating circulating macrophages, and T lymphocytes. These immune system-like stromal cells are recruited into the evolving tumor through the elaboration of chemokines, and are reprogrammed to adopt new cellular identities critical for glioma formation, maintenance, and progression. In this manner, these populations of tumor-associated microglia and macrophages produce growth factors that support gliomagenesis and continued tumor growth. As we begin to characterize these immune cell contributions, future therapies might emerge as adjuvant approaches to glioma treatment.
Collapse
Affiliation(s)
- David H Gutmann
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
41
|
Cheng XQ, Liang XZ, Wei S, Ding X, Han GH, Liu P, Sun X, Quan Q, Tang H, Zhao Q, Shang AJ, Peng J. Protein microarray analysis of cytokine expression changes in distal stumps after sciatic nerve transection. Neural Regen Res 2020; 15:503-511. [PMID: 31571662 PMCID: PMC6921340 DOI: 10.4103/1673-5374.266062] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
A large number of chemokines, cytokines, other trophic factors and the extracellular matrix molecules form a favorable microenvironment for peripheral nerve regeneration. This microenvironment is one of the major factors for regenerative success. Therefore, it is important to investigate the key molecules and regulators affecting nerve regeneration after peripheral nerve injury. However, the identities of specific cytokines at various time points after sciatic nerve injury have not been determined. The study was performed by transecting the sciatic nerve to establish a model of peripheral nerve injury and to analyze, by protein microarray, the expression of different cytokines in the distal nerve after injury. Results showed a large number of cytokines were up-regulated at different time points post injury and several cytokines, e.g., ciliary neurotrophic factor, were downregulated. The construction of a protein-protein interaction network was used to screen how the proteins interacted with differentially expressed cytokines. Kyoto Encyclopedia of Genes and Genomes pathway and Gene ontology analyses indicated that the differentially expressed cytokines were significantly associated with chemokine signaling pathways, Janus kinase/signal transducers and activators of transcription, phosphoinositide 3-kinase/protein kinase B, and notch signaling pathway. The cytokines involved in inflammation, immune response and cell chemotaxis were up-regulated initially and the cytokines involved in neuronal apoptotic processes, cell-cell adhesion, and cell proliferation were up-regulated at 28 days after injury. Western blot analysis showed that the expression and changes of hepatocyte growth factor, glial cell line-derived neurotrophic factor and ciliary neurotrophic factor were consistent with the results of protein microarray analysis. The results provide a comprehensive understanding of changes in cytokine expression and changes in these cytokines and classical signaling pathways and biological functions during Wallerian degeneration, as well as a basis for potential treatments of peripheral nerve injury. The study was approved by the Institutional Animal Care and Use Committee of the Chinese PLA General Hospital, China (approval number: 2016-x9-07) in September 2016.
Collapse
Affiliation(s)
- Xiao-Qing Cheng
- Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, China
| | - Xue-Zhen Liang
- Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Institute of Orthopedics, Chinese PLA General Hospital, Beijing; The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, China
| | - Shuai Wei
- Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, China
| | - Xiao Ding
- Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, China
| | - Gong-Hai Han
- Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, China
| | - Ping Liu
- Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, China
| | - Xun Sun
- Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, China
| | - Qi Quan
- Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, China
| | - He Tang
- Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, China
| | - Qing Zhao
- Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, China
| | - Ai-Jia Shang
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province; Department of Neurosurgery, Chinese PLA General Hospital, Beijing, China
| | - Jiang Peng
- Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Institute of Orthopedics, Chinese PLA General Hospital, Beijing; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| |
Collapse
|
42
|
Cytokine CCL5 and receptor CCR5 axis in glioblastoma multiforme. Radiol Oncol 2019; 53:397-406. [PMID: 31747383 PMCID: PMC6884928 DOI: 10.2478/raon-2019-0057] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 10/15/2019] [Indexed: 12/21/2022] Open
Abstract
Background Glioblastoma is the most frequent and aggressive brain tumour in humans with median survival from 12 to 15 months after the diagnosis. This is mostly due to therapy resistant glioblastoma stem cells in addition to intertumour heterogeneity that is due to infiltration of a plethora of host cells. Besides endothelial cells, mesenchymal stem cells and their differentiated progenies, immune cells of various differentiation states, including monocytes, comprise resident, brain tumour microenvironment. There are compelling evidence for CCL5/CCR5 in the invasive and metastatic behaviour of many cancer types. CCR5, a G-protein coupled receptor, known to function as an essential co-receptor for HIV entry, is now known to participate in driving tumour heterogeneity, the formation of cancer stem cells and the promotion of cancer invasion and metastasis. Clinical trials have recently opened targeting CCR5 using a humanized monoclonal antibody (leronlimab) for metastatic triple negative breast cancer (TNBC) or a small molecule inhibitor (maraviroc) for metastatic colon cancer. There are important CCL5 and CCR5 structure and signalling mechanisms in glioblastoma. In addition, the CCL5/CCR5 axis directs infiltration and interactions with monocytes/macrophages and mesenchymal stem cells, comprising glioblastoma stem cell niches. Conclusions CCR5 is highly expressed in glioblastoma and is associated with poor prognosis of patients. CCL5/CCR5 is suggested to be an excellent new target for glioblastoma therapy. The molecular mechanisms, by which chemoattractant and receptor respond within the complex tissue microenvironment to promote cancer stem cells and tumour heterogeneity, should be considered in forthcoming studies.
Collapse
|
43
|
Optic Pathway Glioma in Type 1 Neurofibromatosis: Review of Its Pathogenesis, Diagnostic Assessment, and Treatment Recommendations. Cancers (Basel) 2019; 11:cancers11111790. [PMID: 31739524 PMCID: PMC6896195 DOI: 10.3390/cancers11111790] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 11/12/2019] [Accepted: 11/12/2019] [Indexed: 12/13/2022] Open
Abstract
Type 1 neurofibromatosis (NF1) is a dominantly inherited condition predisposing to tumor development. Optic pathway glioma (OPG) is the most frequent central nervous system tumor in children with NF1, affecting approximately 15-20% of patients. The lack of well-established prognostic markers and the wide clinical variability with respect to tumor progression and visual outcome make the clinical management of these tumors challenging, with significant differences among distinct centers. We reviewed published articles on OPG diagnostic protocol, follow-up and treatment in NF1. Cohorts of NF1 children with OPG reported in the literature and patients prospectively collected in our center were analyzed with regard to clinical data, tumor anatomical site, diagnostic workflow, treatment and outcome. In addition, we discussed the recent findings on the pathophysiology of OPG development in NF1. This review provides a comprehensive overview about the clinical management of NF1-associated OPG, focusing on the most recent advances from preclinical studies with genetically engineered models and the ongoing clinical trials.
Collapse
|
44
|
Abstract
As a cancer predisposition syndrome, individuals with neurofibromatosis type 1 (NF1) are at increased risk for the development of both benign and malignant tumors. One of the most common locations for these cancers is the central nervous system, where low-grade gliomas predominate in children. During early childhood, gliomas affecting the optic pathway are most frequently encountered, whereas gliomas of the brainstem and other locations are observed in slightly older children. In contrast, the majority of gliomas arising in adults with NF1 are malignant cancers, typically glioblastoma, involving the cerebral hemispheres. Our understanding of the pathogenesis of NF1-associated gliomas has been significantly advanced through the use of genetically engineered mice, yielding new targets for therapeutic drug design and evaluation. In addition, Nf1 murine glioma models have served as instructive platforms for defining the cell of origin of these tumors, elucidating the critical role of the tumor microenvironment in determining tumor growth and vision loss, and determining how cancer risk factors (sex, germline NF1 mutation) impact on glioma formation and progression. Moreover, these preclinical models have permitted early phase analysis of promising drugs that reduce tumor growth and attenuate vision loss, as an initial step prior to translation to human clinical trials.
Collapse
Affiliation(s)
| | - David H Gutmann
- Department of Neurology, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
45
|
Arzt M, Monje M. Inflaming glioma growth. Neuro Oncol 2019; 21:1213-1214. [PMID: 31304975 DOI: 10.1093/neuonc/noz125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Marlene Arzt
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, California
| | - Michelle Monje
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, California
| |
Collapse
|
46
|
Wright-Jin EC, Gutmann DH. Microglia as Dynamic Cellular Mediators of Brain Function. Trends Mol Med 2019; 25:967-979. [PMID: 31597593 DOI: 10.1016/j.molmed.2019.08.013] [Citation(s) in RCA: 110] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 06/27/2019] [Accepted: 08/28/2019] [Indexed: 12/30/2022]
Abstract
Originally hypothesized to function solely as immunologic responders within the central nervous system (CNS), emerging evidence has revealed that microglia have more complex roles in normal brain development and in the context of disease. In health, microglia influence neural progenitor fate decisions, astrocyte activation, neuronal homeostasis, and synaptogenesis. In the setting of brain disease, including autism, brain tumors, and neurodegenerative disorders, microglia undergo substantial morphological, molecular, and functional changes, which establish new biological states relevant to disease pathogenesis and progression. In this review, we discuss the function of microglia in health and disease and outline a conceptual framework for elucidating their specific contributions to nervous system pathobiology.
Collapse
Affiliation(s)
- Elizabeth C Wright-Jin
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - David H Gutmann
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
47
|
Haage V, Semtner M, Vidal RO, Hernandez DP, Pong WW, Chen Z, Hambardzumyan D, Magrini V, Ly A, Walker J, Mardis E, Mertins P, Sauer S, Kettenmann H, Gutmann DH. Comprehensive gene expression meta-analysis identifies signature genes that distinguish microglia from peripheral monocytes/macrophages in health and glioma. Acta Neuropathol Commun 2019; 7:20. [PMID: 30764877 PMCID: PMC6376799 DOI: 10.1186/s40478-019-0665-y] [Citation(s) in RCA: 109] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 01/22/2019] [Indexed: 12/21/2022] Open
Abstract
Monocytes/macrophages have begun to emerge as key cellular modulators of brain homeostasis and central nervous system (CNS) disease. In the healthy brain, resident microglia are the predominant macrophage cell population; however, under conditions of blood-brain barrier leakage, peripheral monocytes/macrophages can infiltrate the brain and participate in CNS disease pathogenesis. Distinguishing these two populations is often challenging, owing to a paucity of universally accepted and reliable markers. To identify discriminatory marker sets for microglia and peripheral monocytes/macrophages, we employed a large meta-analytic approach using five published murine transcriptional datasets. Following hierarchical clustering, we filtered the top differentially expressed genes (DEGs) through a brain cell type-specific sequencing database, which led to the identification of eight microglia and eight peripheral monocyte/macrophage markers. We then validated their differential expression, leveraging a published single cell RNA sequencing dataset and quantitative RT-PCR using freshly isolated microglia and peripheral monocytes/macrophages from two different mouse strains. We further verified the translation of these DEGs at the protein level. As top microglia DEGs, we identified P2ry12, Tmem119, Slc2a5 and Fcrls, whereas Emilin2, Gda, Hp and Sell emerged as the best DEGs for identifying peripheral monocytes/macrophages. Lastly, we evaluated their utility in discriminating monocyte/macrophage populations in the setting of brain pathology (glioma), and found that these DEG sets distinguished glioma-associated microglia from macrophages in both RCAS and GL261 mouse models of glioblastoma. Taken together, this unbiased bioinformatic approach facilitated the discovery of a robust set of microglia and peripheral monocyte/macrophage expression markers to discriminate these monocyte populations in both health and disease.
Collapse
|
48
|
D’Angelo F, Ceccarelli M, Tala, Garofano L, Zhang J, Frattini V, Caruso FP, Lewis G, Alfaro KD, Bauchet L, Berzero G, Cachia D, Cangiano M, Capelle L, de Groot J, DiMeco F, Ducray F, Farah W, Finocchiaro G, Goutagny S, Kamiya-Matsuoka C, Lavarino C, Loiseau H, Lorgis V, Marras CE, McCutcheon I, Nam DH, Ronchi S, Saletti V, Seizeur R, Slopis J, Suñol M, Vandenbos F, Varlet P, Vidaud D, Watts C, Tabar V, Reuss DE, Kim SK, Meyronet D, Mokhtari K, Salvador H, Bhat KP, Eoli M, Sanson M, Lasorella A, lavarone A. The molecular landscape of glioma in patients with Neurofibromatosis 1. Nat Med 2019; 25:176-187. [PMID: 30531922 PMCID: PMC6857804 DOI: 10.1038/s41591-018-0263-8] [Citation(s) in RCA: 149] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 10/17/2018] [Indexed: 12/30/2022]
Abstract
Neurofibromatosis type 1 (NF1) is a common tumor predisposition syndrome in which glioma is one of the prevalent tumors. Gliomagenesis in NF1 results in a heterogeneous spectrum of low- to high-grade neoplasms occurring during the entire lifespan of patients. The pattern of genetic and epigenetic alterations of glioma that develops in NF1 patients and the similarities with sporadic glioma remain unknown. Here, we present the molecular landscape of low- and high-grade gliomas in patients affected by NF1 (NF1-glioma). We found that the predisposing germline mutation of the NF1 gene was frequently converted to homozygosity and the somatic mutational load of NF1-glioma was influenced by age and grade. High-grade tumors harbored genetic alterations of TP53 and CDKN2A, frequent mutations of ATRX associated with Alternative Lengthening of Telomere, and were enriched in genetic alterations of transcription/chromatin regulation and PI3 kinase pathways. Low-grade tumors exhibited fewer mutations that were over-represented in genes of the MAP kinase pathway. Approximately 50% of low-grade NF1-gliomas displayed an immune signature, T lymphocyte infiltrates, and increased neo-antigen load. DNA methylation assigned NF1-glioma to LGm6, a poorly defined Isocitrate Dehydrogenase 1 wild-type subgroup enriched with ATRX mutations. Thus, the profiling of NF1-glioma defined a distinct landscape that recapitulates a subset of sporadic tumors.
Collapse
Affiliation(s)
- Fulvio D’Angelo
- Institute for Cancer Genetics, Columbia University Medical Center, New York, NY, USA.,BIOGEM Istituto di Ricerche Genetiche ‘G. Salvatore’, Ariano Irpino, Italy.,These authors contributed equally: F. D’Angelo, M. Ceccarelli
| | - Michele Ceccarelli
- BIOGEM Istituto di Ricerche Genetiche ‘G. Salvatore’, Ariano Irpino, Italy.,Department of Science and Technology, Università degli Studi del Sannio, Benevento, Italy.,These authors contributed equally: F. D’Angelo, M. Ceccarelli
| | - Tala
- Institute for Cancer Genetics, Columbia University Medical Center, New York, NY, USA
| | - Luciano Garofano
- Institute for Cancer Genetics, Columbia University Medical Center, New York, NY, USA.,BIOGEM Istituto di Ricerche Genetiche ‘G. Salvatore’, Ariano Irpino, Italy
| | - Jing Zhang
- Institute for Cancer Genetics, Columbia University Medical Center, New York, NY, USA
| | - Véronique Frattini
- Institute for Cancer Genetics, Columbia University Medical Center, New York, NY, USA
| | - Francesca P. Caruso
- BIOGEM Istituto di Ricerche Genetiche ‘G. Salvatore’, Ariano Irpino, Italy.,Department of Science and Technology, Università degli Studi del Sannio, Benevento, Italy
| | - Genevieve Lewis
- Institute for Cancer Genetics, Columbia University Medical Center, New York, NY, USA
| | - Kristin D. Alfaro
- The University of Texas M.D. Anderson Cancer Center John Mendelsohn Faculty Center (FC7.3025) – Neuro-Oncology – Unit 0431, Houston, TX, USA
| | - Luc Bauchet
- Department of Neurosurgery, Gui de Chauliac Hospital, Montpellier University Medical Center, Montpellier, France
| | - Giulia Berzero
- Sorbonne Universités UPMC Université Paris 06, UMR S 1127, Inserm U 1127, CNRS UMR 7225, ICM, APHP, Paris, France
| | - David Cachia
- Department of Neuro-Oncology, Medical University of South Carolina, Charleston, SC, USA.,Department of Neurosurgery, Medical University of South Carolina, Charleston, SC, USA
| | - Mario Cangiano
- BIOGEM Istituto di Ricerche Genetiche ‘G. Salvatore’, Ariano Irpino, Italy
| | - Laurent Capelle
- AP-HP, Hôpital de la Pitié-Salpêtrière, Service de Neurochirurgie, Paris, France
| | - John de Groot
- The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Francesco DiMeco
- Department of Neurological Surgery, Carlo Besta Neurological Institute, Milan, Italy.,Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy.,Hunterian Brain Tumor Research Laboratory CRB2 2M41, Baltimore, MD, USA
| | - François Ducray
- Service de Neuro-Oncologie, Hospices Civils de Lyon, Université Claude Bernard Lyon 1, Department of Cancer Cell Plasticity, Cancer Research Center of Lyon, INSERM U1052, CNRS UMR5286, Lyon, France
| | - Walid Farah
- Department of Neurosurgery, CHU, Dijon, France
| | - Gaetano Finocchiaro
- Unit of Molecular Neuro-Oncology, IRCCS Foundation, Carlo Besta Neurological Institute, Milan, Italy
| | - Stéphane Goutagny
- Service de Neurochirurgie, Hôpital Beaujon, Assistance PubliqueHôpitaux de Paris, Clichy, France
| | | | - Cinzia Lavarino
- Developmental Tumor Laboratory, Fundación Sant Joan de Déu, Barcelona, Spain
| | - Hugues Loiseau
- Department of Neurosurgery, Bordeaux University Hospital. Labex TRAIL (ANR-10-LABX-57). EA 7435 – IMOTION Bordeaux University, Bordeaux, France
| | - Véronique Lorgis
- Department of Medical Oncology, Centre GF Leclerc, Dijon, France
| | - Carlo E. Marras
- Pediatric Neurosurgery Unit, Department of Neuroscience and Neurorehabilitation, Bambino Gesù Children’s Hospital, Rome, Italy
| | - Ian McCutcheon
- The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Do-Hyun Nam
- Department of Neurosurgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea
| | - Susanna Ronchi
- Sorbonne Universités UPMC Université Paris 06, UMR S 1127, Inserm U 1127, CNRS UMR 7225, ICM, APHP, Paris, France
| | - Veronica Saletti
- Developmental Neurology Unit, IRCCS Foundation, Carlo Besta Neurological Institute, Milan, Italy
| | - Romuald Seizeur
- Service de Neurochirurgie, Hôpital de la Cavale Blanche, CHRU de Brest, Université de Brest, Brest, France
| | - John Slopis
- The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Mariona Suñol
- Department of Pathology, Hospital Sant Joan de Déu, Barcelona, Spain
| | - Fanny Vandenbos
- Central Laboratory of Pathology, Pasteur I University Hospital, Nice, France
| | - Pascale Varlet
- Department of Neuropathology, Sainte-Anne Hospital, Paris, France.,IMA-Brain, Inserm U894, Institute of Psychiatry and Neuroscience of Paris, Paris, France
| | - Dominique Vidaud
- EA7331, Université Paris Descartes, France; Service de Génétique et Biologie Moléculaires, Hôpital Cochin, AP-HP, Paris, France
| | - Colin Watts
- Institute of Cancer and Genomic Sciences University of Birmingham Edgbaston, Birmingham, United Kingdom
| | - Viviane Tabar
- Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - David E. Reuss
- Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Neuropathology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Seung-Ki Kim
- Division of Pediatric Neurosurgery, Seoul National University Children’s Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - David Meyronet
- Centre de Pathologie Et Neuropathologie Est Hospices Civils de Lyon, Lyon, France
| | - Karima Mokhtari
- Sorbonne Universités UPMC Université Paris 06, UMR S 1127, Inserm U 1127, CNRS UMR 7225, ICM, APHP, Paris, France
| | - Hector Salvador
- Pediatric Oncology Unit, Hospital Sant Joan de Déu, Esplugues, Barcelona, Spain
| | - Krishna P. Bhat
- The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Marica Eoli
- Unit of Molecular Neuro-Oncology, IRCCS Foundation, Carlo Besta Neurological Institute, Milan, Italy
| | - Marc Sanson
- Sorbonne Universités UPMC Université Paris 06, UMR S 1127, Inserm U 1127, CNRS UMR 7225, ICM, APHP, Paris, France
| | - Anna Lasorella
- Institute for Cancer Genetics, Columbia University Medical Center, New York, NY, USA. .,Department of Pediatrics, Columbia University Medical Center, New York, NY, USA. .,Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA.
| | - Antonio lavarone
- Institute for Cancer Genetics, Columbia University Medical Center, New York, NY, USA.,Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA.,Department of Neurology, Columbia University Medical Center, New York, NY, USA.,These authors jointly supervised this work: A. Lasorella, A. Iavarone.,Correspondence and requests for materials should be addressed to A.L. or A.I. ;
| |
Collapse
|
49
|
KIAA1549-BRAF Expression Establishes a Permissive Tumor Microenvironment Through NFκB-Mediated CCL2 Production. Neoplasia 2018; 21:52-60. [PMID: 30504064 PMCID: PMC6277251 DOI: 10.1016/j.neo.2018.11.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 11/09/2018] [Accepted: 11/12/2018] [Indexed: 11/23/2022] Open
Abstract
KIAA1549-BRAF is the most frequently identified genetic mutation in sporadic pilocytic astrocytoma (PA), creating a fusion BRAF (f-BRAF) protein with increased BRAF activity. Fusion-BRAF-expressing neural stem cells (NSCs) exhibit increased cell growth and can generate glioma-like lesions following injection into the cerebella of naïve mice. Increased Iba1+ monocyte (microglia) infiltration is associated with murine f-BRAF-expressing NSC-induced glioma-like lesion formation, suggesting that f-BRAF-expressing NSCs attract microglia to establish a microenvironment supportive of tumorigenesis. Herein, we identify Ccl2 as the chemokine produced by f-BRAF-expressing NSCs, which is critical for creating a permissive stroma for gliomagenesis. In addition, f-BRAF regulation of Ccl2 production operates in an ERK- and NFκB-dependent manner in cerebellar NSCs. Finally, Ccr2-mediated microglia recruitment is required for glioma-like lesion formation in vivo, as tumor do not form in Ccr2-deficient mice following f-BRAF-expressing NSC injection. Collectively, these results demonstrate that f-BRAF expression creates a supportive tumor microenvironment through NFκB-mediated Ccl2 production and microglia recruitment.
Collapse
|
50
|
Durón C, Pan Y, Gutmann DH, Hardin J, Radunskaya A. Variability of Betweenness Centrality and Its Effect on Identifying Essential Genes. Bull Math Biol 2018; 81:3655-3673. [PMID: 30350013 DOI: 10.1007/s11538-018-0526-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Accepted: 10/11/2018] [Indexed: 11/27/2022]
Abstract
This paper begins to build a theoretical framework that would enable the pharmaceutical industry to use network complexity measures as a way to identify drug targets. The variability of a betweenness measure for a network node is examined through different methods of network perturbation. Our results indicate a robustness of betweenness centrality in the identification of target genes.
Collapse
Affiliation(s)
- Christina Durón
- Math Department, Claremont Graduate University, Claremont, CA, 91711, USA
| | - Yuan Pan
- Neurology and Neurological Sciences, Stanford University Medical Center, Palo Alto, CA, USA
| | - David H Gutmann
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Johanna Hardin
- Math Department, Pomona College, Claremont, CA, 91711, USA
| | - Ami Radunskaya
- Math Department, Pomona College, Claremont, CA, 91711, USA.
| |
Collapse
|