1
|
Ferrer I. The Primary Microglial Leukodystrophies: A Review. Int J Mol Sci 2022; 23:ijms23116341. [PMID: 35683020 PMCID: PMC9181167 DOI: 10.3390/ijms23116341] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 06/03/2022] [Indexed: 11/17/2022] Open
Abstract
Primary microglial leukodystrophy or leukoencephalopathy are disorders in which a genetic defect linked to microglia causes cerebral white matter damage. Pigmented orthochromatic leukodystrophy, adult-onset orthochromatic leukodystrophy associated with pigmented macrophages, hereditary diffuse leukoencephalopathy with (axonal) spheroids, and adult-onset leukoencephalopathy with axonal spheroids and pigmented glia (ALSP) are different terms apparently used to designate the same disease. However, ALSP linked to dominantly inherited mutations in CSF1R (colony stimulating factor receptor 1) cause CSF-1R-related leukoencephalopathy (CRP). Yet, recessive ALSP with ovarian failure linked to AARS2 (alanyl-transfer (t)RNA synthase 2) mutations (LKENP) is a mitochondrial disease and not a primary microglial leukoencephalopathy. Polycystic membranous lipomembranous osteodysplasia with sclerosing leukoencephalopathy (PLOSL; Nasu–Hakola disease: NHD) is a systemic disease affecting bones, cerebral white matter, selected grey nuclei, and adipose tissue The disease is caused by mutations of one of the two genes TYROBP or TREM2, identified as PLOSL1 and PLOSL2, respectively. TYROBP associates with receptors expressed in NK cells, B and T lymphocytes, dendritic cells, monocytes, macrophages, and microglia. TREM2 encodes the protein TREM2 (triggering receptor expressed on myeloid cells 2), which forms a receptor signalling complex with TYROBP in macrophages and dendritic cells. Rather than pure microglial leukoencephalopathy, NHD can be considered a multisystemic “immunological” disease.
Collapse
Affiliation(s)
- Isidro Ferrer
- Network Centre of Biomedical Research of Neurodegenerative Diseases (CIBERNED), Department of Pathology and Experimental Therapeutics, Bellvitge Biomedical Research Institute (IDIBELL), University of Barcelona, 08907 Barcelona, L'Hospitalet de Llobregat, Spain
| |
Collapse
|
2
|
Shixing X, Wei W, Xueyan H, Wei T. Pathogenicity analysis and a novel case report of intronic mutations in CSF1R gene. Neurocase 2022; 28:251-257. [PMID: 35503975 DOI: 10.1080/13554794.2022.2071625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Colony-stimulating factor 1 receptor-associated leukoencephalopathy (CSF1R-related leukoencephalopathy) is a genetic disorder mutated in a single allele. It is characterized by an adult-onset along with predominantly cognitive impairment, accompanied by neuropsychiatric symptoms as well as motor symptoms such as Parkinsonism. In the current study, we confirmed a case of CSF1R-related leukoencephalopathy pedigree by genetic screening, and a new intron c. 1858 + 5 G > A mutation was detected in affected patients. After reviewing all previous reports of introns, we found that symptoms and clinical manifestations of the patients were typical and met the features of previous intron reports.
Collapse
Affiliation(s)
- Xue Shixing
- Department of Neurology, Dalian University Affiliated Xinhua Hospital, Dalian, Liaoning, China
| | - Wang Wei
- Department of Rehabilitation Medicine, Dalian University Affiliated Zhongshan Hospital, Dalian, Liaoning, China
| | - Hou Xueyan
- Department of Medical Imaging, Dalian University Affiliated Xinhua Hospital, Dalian, Liaoning, China
| | - Tang Wei
- Department of Neurology, Dalian University Affiliated Xinhua Hospital, Dalian, Liaoning, China
| |
Collapse
|
3
|
Zarekiani P, Nogueira Pinto H, Hol EM, Bugiani M, de Vries HE. The neurovascular unit in leukodystrophies: towards solving the puzzle. Fluids Barriers CNS 2022; 19:18. [PMID: 35227276 PMCID: PMC8887016 DOI: 10.1186/s12987-022-00316-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 02/11/2022] [Indexed: 12/11/2022] Open
Abstract
The neurovascular unit (NVU) is a highly organized multicellular system localized in the brain, formed by neuronal, glial (astrocytes, oligodendrocytes, and microglia) and vascular (endothelial cells and pericytes) cells. The blood-brain barrier, a complex and dynamic endothelial cell barrier in the brain microvasculature that separates the blood from the brain parenchyma, is a component of the NVU. In a variety of neurological disorders, including Alzheimer's disease, multiple sclerosis, and stroke, dysfunctions of the NVU occurs. There is, however, a lack of knowledge regarding the NVU function in leukodystrophies, which are rare monogenic disorders that primarily affect the white matter. Since leukodystrophies are rare diseases, human brain tissue availability is scarce and representative animal models that significantly recapitulate the disease are difficult to develop. The introduction of human induced pluripotent stem cells (hiPSC) now makes it possible to surpass these limitations while maintaining the ability to work in a biologically relevant human context and safeguarding the genetic background of the patient. This review aims to provide further insights into the NVU functioning in leukodystrophies, with a special focus on iPSC-derived models that can be used to dissect neurovascular pathophysiology in these diseases.
Collapse
Affiliation(s)
- Parand Zarekiani
- Department of Pathology, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, de Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Leukodystrophy Center, Amsterdam UMC, Amsterdam, The Netherlands
- Department of Molecular Cell Biology and Immunology, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Henrique Nogueira Pinto
- Department of Molecular Cell Biology and Immunology, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Elly M Hol
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Marianna Bugiani
- Department of Pathology, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, de Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Leukodystrophy Center, Amsterdam UMC, Amsterdam, The Netherlands
| | - Helga E de Vries
- Department of Molecular Cell Biology and Immunology, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands.
| |
Collapse
|
4
|
Wu X, Sun C, Wang X, Liu Y, Wu W, Jia G. Identification of a de novo splicing mutation in the CSF1R gene in a Chinese patient with hereditary diffuse leukoencephalopathy with spheroids. Neurol Sci 2021; 43:3265-3272. [PMID: 34791569 PMCID: PMC9018673 DOI: 10.1007/s10072-021-05755-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Accepted: 11/12/2021] [Indexed: 11/26/2022]
Abstract
Objective To report a de novo splicing mutation in the CSF1R gene in a patient with hereditary diffuse leukoencephalopathy with spheroids (HDLS). Methods A 42-year-old Chinese woman with constant weakness on her left lower extremity was recruited in the current study. Detail medical history and clinical characteristics were reviewed. Brain magnetic resonance imaging (MRI), whole-exome sequencing, and Sanger sequencing were performed with bioinformatics analysis. Results The Chinese HDLS patient with no HDLS family history exhibited a de novo splicing mutation (c.1754-10 T > A) in the CSF1R gene. This mutation was located at the splice site of intron 12 and resulted in the skipping of exon 13 from the CSF1R mRNA. This finding constitutes the first de novo splicing mutation ever reported in HDLS. Furthermore, MRI abnormalities had been reported at least 6 months prior to the onset of the patient’s clinical phenotype. Conclusion Our study indicates that the diagnosis of HDLS should be considered even in the absence of a family history and can help deepen the clinical and genetic understanding of HDLS.
Collapse
Affiliation(s)
- Xinwei Wu
- Department of Geriatrics, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Shan Dong, Jinan, 250012, China
| | - Congcong Sun
- Department of Neurology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Shan Dong, Jinan, 250012, China
| | - Xingbang Wang
- Department of Geriatrics, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Shan Dong, Jinan, 250012, China
| | - Ying Liu
- Department of Neurology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Shan Dong, Jinan, 250012, China
| | - Wei Wu
- Department of Neurology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Shan Dong, Jinan, 250012, China
| | - Guoyong Jia
- Department of Neurology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Shan Dong, Jinan, 250012, China.
| |
Collapse
|
5
|
A novel mutation in CSF1R associated with hereditary diffuse leukoencephalopathy with spheroids. Neurol Sci 2021; 43:411-417. [PMID: 33948764 DOI: 10.1007/s10072-021-05296-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 04/28/2021] [Indexed: 02/05/2023]
Abstract
BACKGROUND Hereditary diffuse leukoencephalopathy with axonal spheroids (HDLS) is a rare autosomal-dominant disorder with high penetrance characterized by progressive cognitive and motor dysfunction. The objective of the study was to describe a new variant of the colony stimulating factor-1 receptor (CSF1R) gene causing HDLS in a Chinese family. METHODS Physical examinations, laboratory tests, structural neuroimaging studies, and whole-exome sequence analysis were carried out. RESULTS Three patients in this family exhibited typical manifestations of HDLS, including progressive cognitive impairment, language and motor dysfunctions, and urinary and bowel incontinence. Genetic analysis identified a heterozygous missense mutation (c.2264T>C, p.L755P) in exon 17 of the CSF1R gene that cosegregated with the HDLS phenotype in an autosomal-dominant pattern. Brain MRI of the proband and her father showed diffuse white matter changes. The proband's 10-year-old son, a gene carrier, remains clinically asymptomatic at present. CONCLUSIONS Our findings identify a novel missense mutation, p.L755P, in the CSF1R gene within a Chinese family with autosomal-dominant HDLS and broaden the genetic spectrum of CSF1R-associated HDLS.
Collapse
|
6
|
Benn CL, Dawson LA. Clinically Precedented Protein Kinases: Rationale for Their Use in Neurodegenerative Disease. Front Aging Neurosci 2020; 12:242. [PMID: 33117143 PMCID: PMC7494159 DOI: 10.3389/fnagi.2020.00242] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 07/13/2020] [Indexed: 12/12/2022] Open
Abstract
Kinases are an intensively studied drug target class in current pharmacological research as evidenced by the large number of kinase inhibitors being assessed in clinical trials. Kinase-targeted therapies have potential for treatment of a broad array of indications including central nervous system (CNS) disorders. In addition to the many variables which contribute to identification of a successful therapeutic molecule, drug discovery for CNS-related disorders also requires significant consideration of access to the target organ and specifically crossing the blood-brain barrier (BBB). To date, only a small number of kinase inhibitors have been reported that are specifically designed to be BBB permeable, which nonetheless demonstrates the potential for success. This review considers the potential for kinase inhibitors in the context of unmet medical need for neurodegenerative disease. A subset of kinases that have been the focus of clinical investigations over a 10-year period have been identified and discussed individually. For each kinase target, the data underpinning the validity of each in the context of neurodegenerative disease is critically evaluated. Selected molecules for each kinase are identified with information on modality, binding site and CNS penetrance, if known. Current clinical development in neurodegenerative disease are summarized. Collectively, the review indicates that kinase targets with sufficient rationale warrant careful design approaches with an emphasis on improving brain penetrance and selectivity.
Collapse
|
7
|
Lopes C, Duarte S, Santos E, Reis C, Pinto M. Intrafamilial heterogeneity in hereditary diffuse leukoencephalopathy with axonal spheroids. Neurol Clin Pract 2020; 9:500-502. [PMID: 32042490 DOI: 10.1212/cpj.0000000000000647] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 02/14/2019] [Indexed: 11/15/2022]
Affiliation(s)
- Carolina Lopes
- Department of Neurology (CL, MP), Centro Hospitalar e Universitário de São João; Department of Neurology (SD, ES), Centro Hospitalar e Universitário do Porto; and Department of Neuroradiology (CR), Centro Hospitalar e Universitário de São João
| | - Sara Duarte
- Department of Neurology (CL, MP), Centro Hospitalar e Universitário de São João; Department of Neurology (SD, ES), Centro Hospitalar e Universitário do Porto; and Department of Neuroradiology (CR), Centro Hospitalar e Universitário de São João
| | - Ernestina Santos
- Department of Neurology (CL, MP), Centro Hospitalar e Universitário de São João; Department of Neurology (SD, ES), Centro Hospitalar e Universitário do Porto; and Department of Neuroradiology (CR), Centro Hospitalar e Universitário de São João
| | - Carina Reis
- Department of Neurology (CL, MP), Centro Hospitalar e Universitário de São João; Department of Neurology (SD, ES), Centro Hospitalar e Universitário do Porto; and Department of Neuroradiology (CR), Centro Hospitalar e Universitário de São João
| | - Madalena Pinto
- Department of Neurology (CL, MP), Centro Hospitalar e Universitário de São João; Department of Neurology (SD, ES), Centro Hospitalar e Universitário do Porto; and Department of Neuroradiology (CR), Centro Hospitalar e Universitário de São João
| |
Collapse
|
8
|
Xie JJ, Ni W, Wei Q, Ma H, Bai G, Shen Y, Wu ZY. New clinical characteristics and novel pathogenic variants of patients with hereditary leukodystrophies. CNS Neurosci Ther 2019; 26:567-575. [PMID: 31885218 PMCID: PMC7163788 DOI: 10.1111/cns.13284] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Revised: 12/05/2019] [Accepted: 12/12/2019] [Indexed: 11/28/2022] Open
Abstract
Aim Leukodystrophies are a group of inherited white matter disorders with clinical, genetic, and imaging heterogeneity, which usually pose a diagnostic challenge for physicians. We aimed to identify new clinical characteristics and novel pathogenic variants of hereditary leukodystrophies in this study. Methods Whole exome sequencing (WES) was performed in 28 unrelated patients clinically suspected with leukodystrophies. Leukocytes enzyme activity test, electroencephalogram (EEG), electromyography (EMG), and brain MRI were conducted. Functional analysis was performed, and the pathogenicity of variants was classified according to the American College of Medical Genetics and Genomics (ACMG) standards and guidelines. Results We made definite diagnosis in 8 probands with 12 pathogenic variants and reported new clinical characteristics and imaging features of these patients. Three novel pathogenic variants were identified, including a microdeletion variant c.2654_2654+3del within CSF1R, a nonsense variant c.1321C>T, and a missense variant c.166G>C within GALC. Conclusion Our results have deepened the understanding of clinical, genetic, and imaging heterogeneity of hereditary leukodystrophies, and expanded the spectrum of pathogenic variants and clinical features.
Collapse
Affiliation(s)
- Juan-Juan Xie
- Department of Neurology and Research Center of Neurology in Second Affiliated Hospital, Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Wang Ni
- Department of Neurology and Research Center of Neurology in Second Affiliated Hospital, Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Qiao Wei
- Department of Neurology and Research Center of Neurology in Second Affiliated Hospital, Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Huan Ma
- Institute of Neuroscience, Zhejiang University School of Medicine, Hangzhou, China
| | - Ge Bai
- Institute of Neuroscience, Zhejiang University School of Medicine, Hangzhou, China
| | - Ying Shen
- Institute of Neuroscience, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhi-Ying Wu
- Department of Neurology and Research Center of Neurology in Second Affiliated Hospital, Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China.,CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai, China
| |
Collapse
|
9
|
Cochran JN, McKinley EC, Cochran M, Amaral MD, Moyers BA, Lasseigne BN, Gray DE, Lawlor JMJ, Prokop JW, Geier EG, Holt JM, Thompson ML, Newberry JS, Yokoyama JS, Worthey EA, Geldmacher DS, Love MN, Cooper GM, Myers RM, Roberson ED. Genome sequencing for early-onset or atypical dementia: high diagnostic yield and frequent observation of multiple contributory alleles. Cold Spring Harb Mol Case Stud 2019; 5:mcs.a003491. [PMID: 31836585 PMCID: PMC6913143 DOI: 10.1101/mcs.a003491] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 10/25/2019] [Indexed: 12/14/2022] Open
Abstract
We assessed the results of genome sequencing for early-onset dementia. Participants were selected from a memory disorders clinic. Genome sequencing was performed along with C9orf72 repeat expansion testing. All returned sequencing results were Sanger-validated. Prior clinical diagnoses included Alzheimer's disease, frontotemporal dementia, and unspecified dementia. The mean age of onset was 54 (41–76). Fifty percent of patients had a strong family history, 37.5% had some, and 12.5% had no known family history. Nine of 32 patients (28%) had a variant defined as pathogenic or likely pathogenic (P/LP) by American College of Medical Genetics and Genomics standards, including variants in APP, C9orf72, CSF1R, and MAPT. Nine patients (including three with P/LP variants) harbored established risk alleles with moderate penetrance (odds ratios of ∼2–5) in ABCA7, AKAP9, GBA, PLD3, SORL1, and TREM2. All six patients harboring these moderate penetrance variants but not P/LP variants also had one or two APOE ε4 alleles. One patient had two APOE ε4 alleles with no other established contributors. In total, 16 patients (50%) harbored one or more genetic variants likely to explain symptoms. We identified variants of uncertain significance (VUSs) in ABI3, ADAM10, ARSA, GRID2IP, MME, NOTCH3, PLCD1, PSEN1, TM2D3, TNK1, TTC3, and VPS13C, also often along with other variants. In summary, genome sequencing for early-onset dementia frequently identified multiple established or possible contributory alleles. These observations add support for an oligogenic model for early-onset dementia.
Collapse
Affiliation(s)
| | - Emily C McKinley
- Alzheimer's Disease Center, Department of Neurology, University of Alabama at Birmingham, Birmingham, Alabama 35294, USA
| | - Meagan Cochran
- HudsonAlpha Institute for Biotechnology, Huntsville, Alabama 35806, USA
| | - Michelle D Amaral
- HudsonAlpha Institute for Biotechnology, Huntsville, Alabama 35806, USA
| | - Bryan A Moyers
- HudsonAlpha Institute for Biotechnology, Huntsville, Alabama 35806, USA
| | | | - David E Gray
- HudsonAlpha Institute for Biotechnology, Huntsville, Alabama 35806, USA
| | - James M J Lawlor
- HudsonAlpha Institute for Biotechnology, Huntsville, Alabama 35806, USA
| | - Jeremy W Prokop
- HudsonAlpha Institute for Biotechnology, Huntsville, Alabama 35806, USA.,Department of Pediatrics and Human Development, Michigan State University, East Lansing, Michigan 48824, USA
| | - Ethan G Geier
- Department of Neurology, Memory and Aging Center, University of California San Francisco, San Francisco, California 94158, USA
| | - James M Holt
- HudsonAlpha Institute for Biotechnology, Huntsville, Alabama 35806, USA
| | | | - J Scott Newberry
- HudsonAlpha Institute for Biotechnology, Huntsville, Alabama 35806, USA
| | - Jennifer S Yokoyama
- Department of Neurology, Memory and Aging Center, University of California San Francisco, San Francisco, California 94158, USA
| | | | - David S Geldmacher
- Alzheimer's Disease Center, Department of Neurology, University of Alabama at Birmingham, Birmingham, Alabama 35294, USA
| | - Marissa Natelson Love
- Alzheimer's Disease Center, Department of Neurology, University of Alabama at Birmingham, Birmingham, Alabama 35294, USA
| | - Gregory M Cooper
- HudsonAlpha Institute for Biotechnology, Huntsville, Alabama 35806, USA
| | - Richard M Myers
- HudsonAlpha Institute for Biotechnology, Huntsville, Alabama 35806, USA
| | - Erik D Roberson
- Alzheimer's Disease Center, Department of Neurology, University of Alabama at Birmingham, Birmingham, Alabama 35294, USA
| |
Collapse
|
10
|
Tian WT, Zhan FX, Liu Q, Luan XH, Zhang C, Shang L, Zhang BY, Pan SJ, Miao F, Hu J, Zhong P, Liu SH, Zhu ZY, Zhou HY, Sun S, Liu XL, Huang XJ, Jiang JW, Ma JF, Wang Y, Chen SF, Tang HD, Chen SD, Cao L. Clinicopathologic characterization and abnormal autophagy of CSF1R-related leukoencephalopathy. Transl Neurodegener 2019; 8:32. [PMID: 31827782 PMCID: PMC6886209 DOI: 10.1186/s40035-019-0171-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Accepted: 08/21/2019] [Indexed: 01/13/2023] Open
Abstract
Background CSF1R-related leukoencephalopathy, also known as hereditary diffuse leukoencephalopathy with spheroids (HDLS), is a rare white-matter encephalopathy characterized by motor and neuropsychiatric symptoms due to colony-stimulating factor 1 receptor (CSF1R) gene mutation. Few of CSF1R mutations have been functionally testified and the pathogenesis remains unknown. Methods In order to investigate clinical and pathological characteristics of patients with CSF1R-related leukoencephalopathy and explore the potential impact of CSF1R mutations, we analyzed clinical manifestations of 15 patients from 10 unrelated families and performed brain biopsy in 2 cases. Next generation sequencing was conducted for 10 probands to confirm the diagnosis. Sanger sequencing, segregation analysis and phenotypic reevaluation were utilized to substantiate findings. Functional examination of identified mutations was further explored. Results Clinical and neuroimaging characteristics were summarized. The average age at onset was 35.9 ± 6.4 years (range 24–46 years old). Younger age of onset was observed in female than male (34.2 vs. 39.2 years). The most common initial symptoms were speech dysfunction, cognitive decline and parkinsonian symptoms. One patient also had marked peripheral neuropathy. Brain biopsy of two cases showed typical pathological changes, including myelin loss, axonal spheroids, phosphorylated neurofilament and activated macrophages. Electron microscopy disclosed increased mitochondrial vacuolation and disorganized neurofilaments in ballooned axons. A total of 7 pathogenic variants (4 novel, 3 documented) were identified with autophosphorylation deficiency, among which c.2342C > T remained partial function of autophosphorylation. Western blotting disclosed the significantly lower level of c.2026C > T (p.R676*) than wild type. The level of microtubule associated protein 1 light chain 3-II (LC3-II), a classical marker of autophagy, was significantly lower in mutants expressed cells than wild type group by western blotting and immunofluorescence staining. Conclusions Our findings support the loss-of-function and haploinsufficiency hypothesis in pathogenesis. Autophagy abnormality may play a role in the disease. Repairing or promoting the phosphorylation level of mutant CSF1R may shed light on therapeutic targets in the future. However, whether peripheral polyneuropathy potentially belongs to CSF1R-related spectrum deserves further study with longer follow-up and more patients enrolled. Trial registration ChiCTR, ChiCTR1800015295. Registered 21 March 2018. Electronic supplementary material The online version of this article (10.1186/s40035-019-0171-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Wo-Tu Tian
- 1Department of Neurology, Rui Jin Hospital & Rui Jin Hospital North, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Fei-Xia Zhan
- 1Department of Neurology, Rui Jin Hospital & Rui Jin Hospital North, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Qing Liu
- 2Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing, 100032 China
| | - Xing-Hua Luan
- 1Department of Neurology, Rui Jin Hospital & Rui Jin Hospital North, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Chao Zhang
- 1Department of Neurology, Rui Jin Hospital & Rui Jin Hospital North, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China.,3Anhui University of Science and Technology School of Medicine, Huainan, 232001 Anhui Province China
| | - Liang Shang
- 2Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing, 100032 China
| | - Ben-Yan Zhang
- 4Department of Pathology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Si-Jian Pan
- 5Department of Neurosurgery, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Fei Miao
- 6Department of Radiology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Jiong Hu
- 7Department of Hematology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Ping Zhong
- 8Suzhou Municipal Hospital, Suzhou, 234000 Anhui Province China
| | - Shi-Hua Liu
- 8Suzhou Municipal Hospital, Suzhou, 234000 Anhui Province China
| | - Ze-Yu Zhu
- 1Department of Neurology, Rui Jin Hospital & Rui Jin Hospital North, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Hai-Yan Zhou
- 1Department of Neurology, Rui Jin Hospital & Rui Jin Hospital North, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Suya Sun
- 1Department of Neurology, Rui Jin Hospital & Rui Jin Hospital North, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Xiao-Li Liu
- 9Department of Neurology, Shanghai Fengxian District Central Hospital, Shanghai Jiao Tong University Affiliated Sixth People's Hospital South Campus, Shanghai, 201406 China
| | - Xiao-Jun Huang
- 1Department of Neurology, Rui Jin Hospital & Rui Jin Hospital North, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Jing-Wen Jiang
- 1Department of Neurology, Rui Jin Hospital & Rui Jin Hospital North, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Jian-Fang Ma
- 1Department of Neurology, Rui Jin Hospital & Rui Jin Hospital North, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Ying Wang
- 1Department of Neurology, Rui Jin Hospital & Rui Jin Hospital North, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Shu-Fen Chen
- 1Department of Neurology, Rui Jin Hospital & Rui Jin Hospital North, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Hui-Dong Tang
- 1Department of Neurology, Rui Jin Hospital & Rui Jin Hospital North, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Sheng-Di Chen
- 1Department of Neurology, Rui Jin Hospital & Rui Jin Hospital North, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Li Cao
- 1Department of Neurology, Rui Jin Hospital & Rui Jin Hospital North, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| |
Collapse
|
11
|
Golde TE. Harnessing Immunoproteostasis to Treat Neurodegenerative Disorders. Neuron 2019; 101:1003-1015. [PMID: 30897353 DOI: 10.1016/j.neuron.2019.02.027] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 02/05/2019] [Accepted: 02/15/2019] [Indexed: 12/12/2022]
Abstract
Immunoproteostasis is a term used to reflect interactions between the immune system and the proteinopathies that are presumptive "triggers" of many neurodegenerative disorders. The study of immunoproteostasis is bolstered by several observations. Mutations or rare variants in genes expressed in microglial cells, known to regulate immune functions, or both can cause, or alter risk for, various neurodegenerative disorders. Additionally, genetic association studies identify numerous loci harboring genes that encode proteins of known immune function that alter risk of developing Alzheimer's disease (AD) and other neurodegenerative proteinopathies. Further, preclinical studies reveal beneficial effects and liabilities of manipulating immune pathways in various neurodegenerative disease models. Although there are concerns that manipulation of the immune system may cause more harm than good, there is considerable interest in developing immune modulatory therapies for neurodegenerative disorders. Herein, I highlight the promise and challenges of harnessing immunoproteostasis to treat neurodegenerative proteinopathies.
Collapse
Affiliation(s)
- Todd E Golde
- McKnight Brain Institute, Center for Translational Research in Neurodegenerative Disease, Department of Neuroscience and Neurology, University of Florida, Gainesville, FL 32607, USA.
| |
Collapse
|
12
|
Stadelmann C, Timmler S, Barrantes-Freer A, Simons M. Myelin in the Central Nervous System: Structure, Function, and Pathology. Physiol Rev 2019; 99:1381-1431. [PMID: 31066630 DOI: 10.1152/physrev.00031.2018] [Citation(s) in RCA: 325] [Impact Index Per Article: 65.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Oligodendrocytes generate multiple layers of myelin membrane around axons of the central nervous system to enable fast and efficient nerve conduction. Until recently, saltatory nerve conduction was considered the only purpose of myelin, but it is now clear that myelin has more functions. In fact, myelinating oligodendrocytes are embedded in a vast network of interconnected glial and neuronal cells, and increasing evidence supports an active role of oligodendrocytes within this assembly, for example, by providing metabolic support to neurons, by regulating ion and water homeostasis, and by adapting to activity-dependent neuronal signals. The molecular complexity governing these interactions requires an in-depth molecular understanding of how oligodendrocytes and axons interact and how they generate, maintain, and remodel their myelin sheaths. This review deals with the biology of myelin, the expanded relationship of myelin with its underlying axons and the neighboring cells, and its disturbances in various diseases such as multiple sclerosis, acute disseminated encephalomyelitis, and neuromyelitis optica spectrum disorders. Furthermore, we will highlight how specific interactions between astrocytes, oligodendrocytes, and microglia contribute to demyelination in hereditary white matter pathologies.
Collapse
Affiliation(s)
- Christine Stadelmann
- Institute of Neuropathology, University Medical Center Göttingen , Göttingen , Germany ; Institute of Neuronal Cell Biology, Technical University Munich , Munich , Germany ; German Center for Neurodegenerative Diseases (DZNE), Munich , Germany ; Department of Neuropathology, University Medical Center Leipzig , Leipzig , Germany ; Munich Cluster of Systems Neurology (SyNergy), Munich , Germany ; and Max Planck Institute of Experimental Medicine, Göttingen , Germany
| | - Sebastian Timmler
- Institute of Neuropathology, University Medical Center Göttingen , Göttingen , Germany ; Institute of Neuronal Cell Biology, Technical University Munich , Munich , Germany ; German Center for Neurodegenerative Diseases (DZNE), Munich , Germany ; Department of Neuropathology, University Medical Center Leipzig , Leipzig , Germany ; Munich Cluster of Systems Neurology (SyNergy), Munich , Germany ; and Max Planck Institute of Experimental Medicine, Göttingen , Germany
| | - Alonso Barrantes-Freer
- Institute of Neuropathology, University Medical Center Göttingen , Göttingen , Germany ; Institute of Neuronal Cell Biology, Technical University Munich , Munich , Germany ; German Center for Neurodegenerative Diseases (DZNE), Munich , Germany ; Department of Neuropathology, University Medical Center Leipzig , Leipzig , Germany ; Munich Cluster of Systems Neurology (SyNergy), Munich , Germany ; and Max Planck Institute of Experimental Medicine, Göttingen , Germany
| | - Mikael Simons
- Institute of Neuropathology, University Medical Center Göttingen , Göttingen , Germany ; Institute of Neuronal Cell Biology, Technical University Munich , Munich , Germany ; German Center for Neurodegenerative Diseases (DZNE), Munich , Germany ; Department of Neuropathology, University Medical Center Leipzig , Leipzig , Germany ; Munich Cluster of Systems Neurology (SyNergy), Munich , Germany ; and Max Planck Institute of Experimental Medicine, Göttingen , Germany
| |
Collapse
|
13
|
Hume DA, Caruso M, Ferrari-Cestari M, Summers KM, Pridans C, Irvine KM. Phenotypic impacts of CSF1R deficiencies in humans and model organisms. J Leukoc Biol 2019; 107:205-219. [PMID: 31330095 DOI: 10.1002/jlb.mr0519-143r] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 06/20/2019] [Accepted: 07/01/2019] [Indexed: 12/12/2022] Open
Abstract
Mϕ proliferation, differentiation, and survival are controlled by signals from the Mϕ CSF receptor (CSF1R). Mono-allelic gain-of-function mutations in CSF1R in humans are associated with an autosomal-dominant leukodystrophy and bi-allelic loss-of-function mutations with recessive skeletal dysplasia, brain disorders, and developmental anomalies. Most of the phenotypes observed in these human disease states are also observed in mice and rats with loss-of-function mutations in Csf1r or in Csf1 encoding one of its two ligands. Studies in rodent models also highlight the importance of genetic background and likely epistatic interactions between Csf1r and other loci. The impacts of Csf1r mutations on the brain are usually attributed solely to direct impacts on microglial number and function. However, analysis of hypomorphic Csf1r mutants in mice and several other lines of evidence suggest that primary hydrocephalus and loss of the physiological functions of Mϕs in the periphery contribute to the development of brain pathology. In this review, we outline the evidence that CSF1R is expressed exclusively in mononuclear phagocytes and explore the mechanisms linking CSF1R mutations to pleiotropic impacts on postnatal growth and development.
Collapse
Affiliation(s)
- David A Hume
- Mater Research Institute, University of Queensland, Woolloongabba, Queensland, Australia
| | - Melanie Caruso
- Mater Research Institute, University of Queensland, Woolloongabba, Queensland, Australia
| | | | - Kim M Summers
- Mater Research Institute, University of Queensland, Woolloongabba, Queensland, Australia
| | - Clare Pridans
- Centre for Inflammation Research, The University of Edinburgh, Edinburgh, United Kingdom
| | - Katharine M Irvine
- Mater Research Institute, University of Queensland, Woolloongabba, Queensland, Australia
| |
Collapse
|
14
|
Yang X, Huang P, Tan Y, Xiao Q. A Novel Splicing Mutation in the CSF1R Gene in a Family With Hereditary Diffuse Leukoencephalopathy With Axonal Spheroids. Front Genet 2019; 10:491. [PMID: 31191609 PMCID: PMC6541038 DOI: 10.3389/fgene.2019.00491] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 05/06/2019] [Indexed: 11/13/2022] Open
Abstract
Hereditary diffuse leukoencephalopathy with axonal spheroids (HDLS) is a rare autosomal dominant disorder that typically presents with early-onset cognitive decline or personality change. The disease is associated with heterozygous mutations in the colony stimulating factor-1 receptor (CSF1R) gene. CSF1R activation regulates microglial survival, proliferation, and differentiation. The different gene mutations may be related to the various clinical phenotypes. Here, we described comprehensive clinical, neuroimaging, neuropathological, and genetic analyses of a family with HDLS. A novel splicing mutation in intron 13 (c.1858+1G>T) of CSF1R was found in this family. It is located at the splice site of intron 13, resulting in a splice donor site leading to exon 13 skipping from the CSF1R mRNA. The mother and two elderly siblings of the proband had the same CSF1R mutation as the proband but showed very mild neuroimaging abnormalities and mild memory loss, which did not affect daily life, indicating very uneven penetrance and distinctly different disease progression among family members. This report provides diverse neuroimaging and clinical characteristics of a novel CSF1R mutation with different disease penetrance. The large clinical heterogeneity in the same family who all had the same mutation indicates that modifying genes and environmental factors may play a role in the pathogenesis of HDLS.
Collapse
Affiliation(s)
- Xiaodong Yang
- Department of Neurology, Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Pei Huang
- Department of Neurology, Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yuyan Tan
- Department of Neurology, Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Qin Xiao
- Department of Neurology, Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
15
|
A novel CSF-1R mutation in a family with hereditary diffuse leukoencephalopathy with axonal spheroids misdiagnosed as hydrocephalus. Neurogenetics 2019; 20:155-160. [PMID: 31093799 PMCID: PMC6647879 DOI: 10.1007/s10048-019-00579-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Accepted: 04/13/2019] [Indexed: 01/08/2023]
Abstract
Hereditary diffuse leukoencephalopathy with axonal spheroids (HDLS) is a rare autosomal dominant disease caused by mutations in the colony stimulating factor 1 receptor (CSF1R) gene that often results in cognitive impairment, psychiatric disorders, motor dysfunction and seizure. We report familial cases of a novel CSF1R mutation causing HDLS similar to hydrocephalus. The patients initially presented with a gait disturbance and then developed progressive cognitive decline, urinary incontinence, epileptic seizures and became bedridden as the disease progressed. A brain magnetic resonance imaging (MRI) scan revealed striking ventricular enlargement and diffuse brain atrophy with frontotemporal predominance, which was later accompanied by white matter changes. Genetic testing in this family showed a novel c.2552T>C (p.L851P) mutation in exon 19 of the CSF1R gene. However, three gene carriers in the family remained clinically asymptomatic. Because of its heterogeneous clinical phenotypes, HDLS patients are often misdiagnosed with other diseases. This is the first genetically proven HDLS case resembling hydrocephalus, and the clinical symptoms of HDLS may be related to the specific genetic mutation.
Collapse
|
16
|
A novel mutation in the CSF1R gene causes hereditary diffuse leukoencephalopathy with axonal spheroids. Neurol Sci 2019; 40:1287-1290. [DOI: 10.1007/s10072-018-3693-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 12/19/2018] [Indexed: 10/27/2022]
|
17
|
Abstract
We herein report the case of a 47-year-old female with the colony-stimulating factor 1 receptor (CSF1R) mutation p.G589R, which is related to hereditary leukoencephalopathy with axonal spheroid (HDLS). The patient presented with an early-onset cognitive decline and progressive aphasia. Brain magnetic resonance imaging revealed HDLS-related alterations. In addition, brain computed tomography revealed interspersed spotty calcifications in the frontal and parietal subcortical white matter, while a characteristic "stepping stone" appearance was observed in the frontal pericallosal regions. Our findings emphasize the importance of calcification appearances in establishing an HDLS diagnosis and in screening for CSF1R mutations.
Collapse
Affiliation(s)
- Kensuke Daida
- Department of Neurology, Juntendo University School of Medicine, Japan
| | - Kenya Nishioka
- Department of Neurology, Juntendo University School of Medicine, Japan
| | - Yuanzhe Li
- Department of Neurology, Juntendo University School of Medicine, Japan
| | - Sho Nakajima
- Department of Neurology, Juntendo University School of Medicine, Japan
| | - Ryota Tanaka
- Department of Neurology, Juntendo University School of Medicine, Japan
| | - Nobutaka Hattori
- Department of Neurology, Juntendo University School of Medicine, Japan
| |
Collapse
|
18
|
van der Knaap MS, Bugiani M. Leukodystrophies: a proposed classification system based on pathological changes and pathogenetic mechanisms. Acta Neuropathol 2017; 134:351-382. [PMID: 28638987 PMCID: PMC5563342 DOI: 10.1007/s00401-017-1739-1] [Citation(s) in RCA: 234] [Impact Index Per Article: 33.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 06/06/2017] [Accepted: 06/06/2017] [Indexed: 12/29/2022]
Abstract
Leukodystrophies are genetically determined disorders characterized by the selective involvement of the central nervous system white matter. Onset may be at any age, from prenatal life to senescence. Many leukodystrophies are degenerative in nature, but some only impair white matter function. The clinical course is mostly progressive, but may also be static or even improving with time. Progressive leukodystrophies are often fatal, and no curative treatment is known. The last decade has witnessed a tremendous increase in the number of defined leukodystrophies also owing to a diagnostic approach combining magnetic resonance imaging pattern recognition and next generation sequencing. Knowledge on white matter physiology and pathology has also dramatically built up. This led to the recognition that only few leukodystrophies are due to mutations in myelin- or oligodendrocyte-specific genes, and many are rather caused by defects in other white matter structural components, including astrocytes, microglia, axons and blood vessels. We here propose a novel classification of leukodystrophies that takes into account the primary involvement of any white matter component. Categories in this classification are the myelin disorders due to a primary defect in oligodendrocytes or myelin (hypomyelinating and demyelinating leukodystrophies, leukodystrophies with myelin vacuolization); astrocytopathies; leuko-axonopathies; microgliopathies; and leuko-vasculopathies. Following this classification, we illustrate the neuropathology and disease mechanisms of some leukodystrophies taken as example for each category. Some leukodystrophies fall into more than one category. Given the complex molecular and cellular interplay underlying white matter pathology, recognition of the cellular pathology behind a disease becomes crucial in addressing possible treatment strategies.
Collapse
Affiliation(s)
- Marjo S van der Knaap
- Department of Pediatrics/Child Neurology, VU University Medical Centre, Amsterdam Neuroscience, Amsterdam, The Netherlands
- Department of Functional Genomics, Centre for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, VU University, Amsterdam, The Netherlands
| | - Marianna Bugiani
- Department of Pediatrics/Child Neurology, VU University Medical Centre, Amsterdam Neuroscience, Amsterdam, The Netherlands.
- Department of Pathology, VU University Medical Centre, Amsterdam Neuroscience, Amsterdam, The Netherlands.
| |
Collapse
|
19
|
Arcuri C, Mecca C, Bianchi R, Giambanco I, Donato R. The Pathophysiological Role of Microglia in Dynamic Surveillance, Phagocytosis and Structural Remodeling of the Developing CNS. Front Mol Neurosci 2017; 10:191. [PMID: 28674485 PMCID: PMC5474494 DOI: 10.3389/fnmol.2017.00191] [Citation(s) in RCA: 176] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 05/30/2017] [Indexed: 12/13/2022] Open
Abstract
In vertebrates, during an early wave of hematopoiesis in the yolk sac between embryonic day E7.0 and E9.0, cells of mesodermal leaflet addressed to macrophage lineage enter in developing central nervous system (CNS) and originate the developing native microglial cells. Depending on the species, microglial cells represent 5–20% of glial cells resident in adult brain. Here, we briefly discuss some canonical functions of the microglia, i.e., cytokine secretion and functional transition from M1 to M2 phenotype. In addition, we review studies on the non-canonical functions of microglia such as regulation of phagocytosis, synaptic pruning, and sculpting postnatal neural circuits. In this latter context the contribution of microglia to some neurodevelopmental disorders is now well established. Nasu-Hakola (NHD) disease is considered a primary microgliopathy with alterations of the DNAX activation protein 12 (DAP12)-Triggering receptor expressed on myeloid cells 2 (TREM-2) signaling and removal of macromolecules and apoptotic cells followed by secondary microglia activation. In Rett syndrome Mecp2-/- microglia shows a substantial impairment of phagocytic ability, although the role of microglia is not yet clear. In a mouse model of Tourette syndrome (TS), microglia abnormalities have also been described, and deficient microglia-mediated neuroprotection is obvious. Here we review the role of microglial cells in neurodevelopmental disorders without inflammation and on the complex role of microglia in developing CNS.
Collapse
Affiliation(s)
- Cataldo Arcuri
- Department of Experimental Medicine, Centro Universitario per la Ricerca sulla Genomica Funzionale, Perugia Medical School, University of PerugiaPerugia, Italy
| | - Carmen Mecca
- Department of Experimental Medicine, Centro Universitario per la Ricerca sulla Genomica Funzionale, Perugia Medical School, University of PerugiaPerugia, Italy
| | - Roberta Bianchi
- Department of Experimental Medicine, Centro Universitario per la Ricerca sulla Genomica Funzionale, Perugia Medical School, University of PerugiaPerugia, Italy
| | - Ileana Giambanco
- Department of Experimental Medicine, Centro Universitario per la Ricerca sulla Genomica Funzionale, Perugia Medical School, University of PerugiaPerugia, Italy
| | - Rosario Donato
- Department of Experimental Medicine, Centro Universitario per la Ricerca sulla Genomica Funzionale, Perugia Medical School, University of PerugiaPerugia, Italy
| |
Collapse
|
20
|
Dwyer AR, Greenland EL, Pixley FJ. Promotion of Tumor Invasion by Tumor-Associated Macrophages: The Role of CSF-1-Activated Phosphatidylinositol 3 Kinase and Src Family Kinase Motility Signaling. Cancers (Basel) 2017; 9:E68. [PMID: 28629162 PMCID: PMC5483887 DOI: 10.3390/cancers9060068] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 06/08/2017] [Accepted: 06/12/2017] [Indexed: 12/12/2022] Open
Abstract
Macrophages interact with cells in every organ to facilitate tissue development, function and repair. However, the close interaction between macrophages and parenchymal cells can be subverted in disease, particularly cancer. Motility is an essential capacity for macrophages to be able to carry out their various roles. In cancers, the macrophage's interstitial migratory ability is frequently co-opted by tumor cells to enable escape from the primary tumor and metastatic spread. Macrophage accumulation within and movement through a tumor is often stimulated by tumor cell production of the mononuclear phagocytic growth factor, colony-stimulating factor-1 (CSF-1). CSF-1 also regulates macrophage survival, proliferation and differentiation, and its many effects are transduced by its receptor, the CSF-1R, via phosphotyrosine motif-activated signals. Mutational analysis of CSF-1R signaling indicates that the major mediators of CSF-1-induced motility are phosphatidyl-inositol-3 kinase (PI3K) and one or more Src family kinase (SFK), which activate signals to adhesion, actin polymerization, polarization and, ultimately, migration and invasion in macrophages. The macrophage transcriptome, including that of the motility machinery, is very complex and highly responsive to the environment, with selective expression of proteins and splice variants rarely found in other cell types. Thus, their unique motility machinery can be specifically targeted to block macrophage migration, and thereby, inhibit tumor invasion and metastasis.
Collapse
Affiliation(s)
- Amy R Dwyer
- School of Biomedical Sciences, The University of Western Australia, 35 Stirling Highway, Crawley, WA 6009, Australia.
| | - Eloise L Greenland
- School of Biomedical Sciences, The University of Western Australia, 35 Stirling Highway, Crawley, WA 6009, Australia.
| | - Fiona J Pixley
- School of Biomedical Sciences, The University of Western Australia, 35 Stirling Highway, Crawley, WA 6009, Australia.
| |
Collapse
|
21
|
Advances in clinical neurology through the journal "Neurological Sciences" (2015-2016). Neurol Sci 2017; 38:9-18. [PMID: 28093657 DOI: 10.1007/s10072-017-2815-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|