51
|
Quick S, Moss J, Rajani RM, Williams A. A Vessel for Change: Endothelial Dysfunction in Cerebral Small Vessel Disease. Trends Neurosci 2020; 44:289-305. [PMID: 33308877 DOI: 10.1016/j.tins.2020.11.003] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 09/24/2020] [Accepted: 11/11/2020] [Indexed: 01/08/2023]
Abstract
The blood vessels of the brain are lined with endothelial cells and it has been long known that these help to regulate blood flow to the brain. However, there is increasing evidence that these cells also interact with the surrounding brain tissue. These interactions change when the endothelial cells become dysfunctional and have an impact in diseases such as cerebral small vessel disease, the leading cause of vascular dementia. In this review, we focus on what endothelial dysfunction is, what causes it, how it leads to surrounding brain pathology, how researchers can investigate it with current models, and where this might lead in the future for dementia therapies.
Collapse
Affiliation(s)
- Sophie Quick
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Jonathan Moss
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Rikesh M Rajani
- UK Dementia Research Institute at UCL, University College London, London, UK
| | - Anna Williams
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, UK.
| |
Collapse
|
52
|
Fu X, Zhang P, Song H, Wu C, Li S, Li S, Yan C. LTBP1 plays a potential bridge between depressive disorder and glioblastoma. J Transl Med 2020; 18:391. [PMID: 33059753 PMCID: PMC7566028 DOI: 10.1186/s12967-020-02509-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 08/26/2020] [Indexed: 12/27/2022] Open
Abstract
Background Glioblastoma multiforme (GBM) is the most malignant tumor in human brain. Diagnosis and treatment of GBM may lead to psychological disorders such as depressive and anxiety disorders. There was no research focusing on the correlation between depressive/anxiety disorder and the outcome of GBM. Thus, the aim of this study was to investigate the possibility of depressive/anxiety disorder correlated with the outcome of GBM patients, as well as the overlapped mechanism bridge which could link depressive/anxiety disorders and GBM. Methods Patient Health Questionnaire (PHQ-9) and Generalized Anxiety Disorder (GAD-7) were used to investigate the psychological condition of GBM patients in our department. To further explore the potential mechanism, bioinformatic methods were used to screen out genes that could be indicators of outcome in GBM, followed by gene ontology (GO) analysis, Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis, and protein–protein interaction (PPI) analysis. Further, cellular experiments were conducted to evaluate the proliferation, migration capacity of primary GBM cells from the patients. Results It was revealed that patients with higher PHQ-9 and GAD-7 scores had significantly worse prognosis than their lower-scored counterparts. Bioinformatic mining revealed that LTBP1 could be a potential genetic mechanism in both depressive/anxiety disorder and GBM. Primary GBM cells with different expression level of LTBP1 should significantly different proliferation and migration capacity. GO, KEGG analysis confirmed that extracellular matrix (ECM) was the most enriched function of LTBP1. PPI network showed the interaction of proteins altered by LTBP1. Hub genes COL1A2, COL5A1 and COL10A1, as well as mesenchymal marker CD44 and Vimentin were statistically higher expressed in LTBP1 high group; while proneural marker E-cadherin was significantly higher expressed in low LTBP1 group. Conclusion There is closely correlation between depressive/anxiety disorders and GBM. LTBP1 could be a potential bridge linking the two diseases through the regulation of ECM.
Collapse
Affiliation(s)
- Xiaojun Fu
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Xiangshanyikesong 50#, HaiDian District, Beijing, 100093, China.,Capital Medical University, Beijing, People's Republic of China
| | - Pei Zhang
- Beijing Institute of Technology, Beijing, China
| | - Hongwang Song
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, People's Republic of China
| | - Chenxing Wu
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Xiangshanyikesong 50#, HaiDian District, Beijing, 100093, China
| | | | - Shouwei Li
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Xiangshanyikesong 50#, HaiDian District, Beijing, 100093, China.
| | - Changxiang Yan
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Xiangshanyikesong 50#, HaiDian District, Beijing, 100093, China.
| |
Collapse
|
53
|
Altay L, Subiras X, Lorés de Motta L, Schick T, Berghold A, Hoyng CB, den Hollander AI, Fauser S, Sadda SR, Liakopoulos S. Genetic and environmental risk factors for extramacular drusen. Mol Vis 2020; 26:661-669. [PMID: 33088170 PMCID: PMC7553721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 10/02/2020] [Indexed: 12/02/2022] Open
Abstract
PURPOSE To analyze risk factors for extramacular drusen (EMD) in patients with age-related macular degeneration (AMD) and healthy control individuals. METHODS This case-control study included 1,520 patients from the prospective multicenter European Genetic Database (EUGENDA). Color fundus photographs and optical coherence tomography scans were evaluated for the presence of AMD and EMD. EMD was considered present if ten or fewer drusen including at least one intermediate-sized drusen were detected outside the macula. Association of EMD was evaluated with various genetic and non-genetic risk factors (31 single nucleotide polymorphisms, systemic complement activation, smoking, cardiovascular factors, and sunlight exposure) using logistic regression models adjusted for age, gender, and AMD. RESULTS EMD was found in 608 subjects (40%) and AMD in 763 (50%) of 1,520 participants. EMD was strongly associated with AMD (p = 2.83 × 10-63, odds ratio [OR] 7.63). After adjustment for AMD, age (p = 0.06, OR 1.02), female gender (p = 3.34 × 10-24, OR 4.44), history of sunlight exposure ≥ 8 h /day (p = 0.0004, OR 1.99), serum complement activation (p = 0.004, OR 1.61), and polymorphisms in ARMS2 (p = 0.00016, OR 1.43) and CFI (p = 0.043, OR 1.20) were identified as risk factors for EMD. The final prediction model including these variants showed an area under the curve of 0.820. CONCLUSIONS The comprehensive analysis of various risk factors revealed a common genetic and pathological pathway of EMD with AMD. Future longitudinal studies are needed to evaluate the role of EMD in otherwise healthy subjects as an expanded phenotype of AMD.
Collapse
Affiliation(s)
- Lebriz Altay
- Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Germany
| | - Xavier Subiras
- Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Germany,Cologne Image Reading Center, Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Germany
| | - Laura Lorés de Motta
- Department of Ophthalmology, Donders Institute for Brain, Cognition and Behaviour, Radboud university medical center, Nijmegen, the Netherlands,Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud university medical center, Nijmegen, the Netherlands
| | - Tina Schick
- Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Germany,Cologne Image Reading Center, Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Germany,AugenZentrum Siegburg, MVZ ADTC Siegburg GmbH, Siegburg, Germany
| | - Aileen Berghold
- Cologne Image Reading Center, Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Germany
| | - Carel B. Hoyng
- Department of Ophthalmology, Donders Institute for Brain, Cognition and Behaviour, Radboud university medical center, Nijmegen, the Netherlands
| | - Anneke I. den Hollander
- Department of Ophthalmology, Donders Institute for Brain, Cognition and Behaviour, Radboud university medical center, Nijmegen, the Netherlands,Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud university medical center, Nijmegen, the Netherlands
| | - Sascha Fauser
- Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Germany
| | | | - Sandra Liakopoulos
- Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Germany,Cologne Image Reading Center, Department of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Germany
| |
Collapse
|
54
|
Fasano A, Formichi P, Taglia I, Bianchi S, Di Donato I, Battisti C, Federico A, Dotti MT. HTRA1 expression profile and activity on TGF-β signaling in HTRA1 mutation carriers. J Cell Physiol 2020; 235:7120-7127. [PMID: 32017060 DOI: 10.1002/jcp.29609] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 01/13/2020] [Indexed: 11/06/2022]
Abstract
High temperature requirement A1 (HTRA1) is a serine protease playing a modulatory role in various cell processes, particularly in the regulation of transforming growth factor-β (TGF-β) signaling. A deleterious role in late-onset cerebral small vessel diseases (CSVDs) of heterozygous HTRA1 mutations, otherwise causative in homozygosity of cerebral autosomal recessive arteriopathy with subcortical infarcts and leukoencephalopathy, was recently suggested. However, the pathomechanism of these heterozygous mutations is still undefined. Our aim is to evaluate the expression profile and activity of HTRA1 on TGF-β signaling in fibroblasts from four subjects carrying the HTRA1 heterozygous mutations-p.E42Dfs*173, p.A321T, p.G295R, and p.Q151K. We found a 50% reduction of HTRA1 expression in HTRA1 mutation carriers compared to the control. Moreover, we showed no changes in TGF-β signaling pathway downstream intermediate, Phospho Smad2/3. However, we found overexpression of genes involved in the extracellular matrix formation in two heterozygous HTRA1 carriers. Our results suggest that each heterozygous HTRA1 missense mutation displays a different and peculiar HTRA1 expression pattern and that CSVD phenotype may also result from 50% of HTRA1 expression.
Collapse
Affiliation(s)
- Alessandro Fasano
- Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy
| | - Patrizia Formichi
- Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy
| | - Ilaria Taglia
- Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy
| | - Silvia Bianchi
- Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy
| | - Ilaria Di Donato
- Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy
| | - Carla Battisti
- Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy
| | - Antonio Federico
- Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy
| | - Maria Teresa Dotti
- Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy
| |
Collapse
|
55
|
Zhang Y, Yang X. The Roles of TGF-β Signaling in Cerebrovascular Diseases. Front Cell Dev Biol 2020; 8:567682. [PMID: 33072751 PMCID: PMC7530326 DOI: 10.3389/fcell.2020.567682] [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: 06/03/2020] [Accepted: 08/31/2020] [Indexed: 12/13/2022] Open
Abstract
Cerebrovascular diseases are one of the leading causes of death worldwide, however, little progress has been made in preventing or treating these diseases to date. The transforming growth factor-β (TGF-β) signaling pathway plays crucial and highly complicated roles in cerebrovascular development and homeostasis, and dysregulated TGF-β signaling contributes to cerebrovascular diseases. In this review, we provide an updated overview of the functional role of TGF-β signaling in the cerebrovascular system under physiological and pathological conditions. We discuss the current understanding of TGF-β signaling in cerebral angiogenesis and the maintenance of brain vessel homeostasis. We also review the mechanisms by which disruption of TGF-β signaling triggers or promotes the progression of cerebrovascular diseases. Finally, we briefly discuss the potential of targeting TGF-β signaling to treat cerebrovascular diseases.
Collapse
Affiliation(s)
- Yizhe Zhang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences, Beijing Institute of Lifeomics, Beijing, China
| | - Xiao Yang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences, Beijing Institute of Lifeomics, Beijing, China
| |
Collapse
|
56
|
Dugaucquier L, Feyen E, Mateiu L, Bruyns TAM, De Keulenaer GW, Segers VFM. The role of endothelial autocrine NRG1/ERBB4 signaling in cardiac remodeling. Am J Physiol Heart Circ Physiol 2020; 319:H443-H455. [PMID: 32618511 DOI: 10.1152/ajpheart.00176.2020] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Neuregulin-1 (NRG1) is a paracrine growth factor, secreted by cardiac endothelial cells (ECs) in conditions of cardiac overload/injury. The current concept is that the cardiac effects of NRG1 are mediated by activation of erythroblastic leukemia viral oncogene homolog (ERBB)4/ERBB2 receptors on cardiomyocytes. However, recent studies have shown that paracrine effects of NRG1 on fibroblasts and macrophages are equally important. Here, we hypothesize that NRG1 autocrine signaling plays a role in cardiac remodeling. We generated EC-specific Erbb4 knockout mice to eliminate endothelial autocrine ERBB4 signaling without affecting paracrine NRG1/ERBB4 signaling in the heart. We first observed no basal cardiac phenotype in these mice up to 32 wk. We next studied these mice following transverse aortic constriction (TAC), exposure to angiotensin II (ANG II), or myocardial infarction in terms of cardiac performance, myocardial hypertrophy, myocardial fibrosis, and capillary density. In general, no major differences between EC-specific Erbb4 knockout mice and control littermates were observed. However, 8 wk following TAC both myocardial hypertrophy and fibrosis were attenuated by EC-specific Erbb4 deletion, albeit these responses were normalized after 20 wk. Similarly, 4 wk after ANG II treatment, myocardial fibrosis was less pronounced compared with control littermates. These observations were supported by RNA-sequencing experiments on cultured endothelial cells showing that NRG1 controls the expression of various hypertrophic and fibrotic pathways. Overall, this study shows a role of endothelial autocrine NRG1/ERBB4 signaling in the modulation of hypertrophic and fibrotic responses during early cardiac remodeling. This study contributes to understanding the spatiotemporal heterogeneity of myocardial autocrine and paracrine responses following cardiac injury.NEW & NOTEWORTHY The role of NRG1/ERBB signaling in endothelial cells is not completely understood. Our study contributes to the understanding of spatiotemporal heterogeneity of myocardial autocrine and paracrine responses following cardiac injury and shows a role of endothelial autocrine NRG1/ERBB4 signaling in the modulation of hypertrophic and fibrotic responses during early cardiac remodeling.
Collapse
Affiliation(s)
| | - Eline Feyen
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Ligia Mateiu
- VIB Center for Molecular Neurology, University of Antwerp, Antwerp, Belgium
| | | | - Gilles W De Keulenaer
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium.,Department of Cardiology, Middelheim Hospital, Antwerp, Belgium
| | - Vincent F M Segers
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium.,Department of Cardiology, University Hospital Antwerp, Edegem, Belgium
| |
Collapse
|
57
|
Kandasamy M, Anusuyadevi M, Aigner KM, Unger MS, Kniewallner KM, de Sousa DMB, Altendorfer B, Mrowetz H, Bogdahn U, Aigner L. TGF-β Signaling: A Therapeutic Target to Reinstate Regenerative Plasticity in Vascular Dementia? Aging Dis 2020; 11:828-850. [PMID: 32765949 PMCID: PMC7390515 DOI: 10.14336/ad.2020.0222] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 02/22/2020] [Indexed: 12/11/2022] Open
Abstract
Vascular dementia (VaD) is the second leading form of memory loss after Alzheimer's disease (AD). Currently, there is no cure available. The etiology, pathophysiology and clinical manifestations of VaD are extremely heterogeneous, but the impaired cerebral blood flow (CBF) represents a common denominator of VaD. The latter might be the result of atherosclerosis, amyloid angiopathy, microbleeding and micro-strokes, together causing blood-brain barrier (BBB) dysfunction and vessel leakage, collectively originating from the consequence of hypertension, one of the main risk factors for VaD. At the histopathological level, VaD displays abnormal vascular remodeling, endothelial cell death, string vessel formation, pericyte responses, fibrosis, astrogliosis, sclerosis, microglia activation, neuroinflammation, demyelination, white matter lesions, deprivation of synapses and neuronal loss. The transforming growth factor (TGF) β has been identified as one of the key molecular factors involved in the aforementioned various pathological aspects. Thus, targeting TGF-β signaling in the brain might be a promising therapeutic strategy to mitigate vascular pathology and improve cognitive functions in patients with VaD. This review revisits the recent understanding of the role of TGF-β in VaD and associated pathological hallmarks. It further explores the potential to modulate certain aspects of VaD pathology by targeting TGF-β signaling.
Collapse
Affiliation(s)
- Mahesh Kandasamy
- Laboratory of Stem Cells and Neuroregeneration, Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli, Tamil Nadu, India.
- Faculty Recharge Programme, University Grants Commission (UGC-FRP), New Delhi, India.
| | - Muthuswamy Anusuyadevi
- Molecular Gerontology Group, Department of Biochemistry, School of Life Sciences, Bharathidhasan University, Tiruchirappalli, Tamil Nadu, India.
| | - Kiera M Aigner
- Institute of Molecular Regenerative Medicine, Salzburg, Paracelsus Medical University.
- Spinal Cord Injury and Tissue Regeneration Center, Salzburg, Paracelsus Medical University, Salzburg, Austria.
| | - Michael S Unger
- Institute of Molecular Regenerative Medicine, Salzburg, Paracelsus Medical University.
- Spinal Cord Injury and Tissue Regeneration Center, Salzburg, Paracelsus Medical University, Salzburg, Austria.
| | - Kathrin M Kniewallner
- Institute of Molecular Regenerative Medicine, Salzburg, Paracelsus Medical University.
- Spinal Cord Injury and Tissue Regeneration Center, Salzburg, Paracelsus Medical University, Salzburg, Austria.
| | - Diana M Bessa de Sousa
- Institute of Molecular Regenerative Medicine, Salzburg, Paracelsus Medical University.
- Spinal Cord Injury and Tissue Regeneration Center, Salzburg, Paracelsus Medical University, Salzburg, Austria.
| | - Barbara Altendorfer
- Institute of Molecular Regenerative Medicine, Salzburg, Paracelsus Medical University.
- Spinal Cord Injury and Tissue Regeneration Center, Salzburg, Paracelsus Medical University, Salzburg, Austria.
| | - Heike Mrowetz
- Institute of Molecular Regenerative Medicine, Salzburg, Paracelsus Medical University.
- Spinal Cord Injury and Tissue Regeneration Center, Salzburg, Paracelsus Medical University, Salzburg, Austria.
| | - Ulrich Bogdahn
- Institute of Molecular Regenerative Medicine, Salzburg, Paracelsus Medical University.
- Spinal Cord Injury and Tissue Regeneration Center, Salzburg, Paracelsus Medical University, Salzburg, Austria.
- Velvio GmbH, Regensburg, Germany.
| | - Ludwig Aigner
- Institute of Molecular Regenerative Medicine, Salzburg, Paracelsus Medical University.
- Spinal Cord Injury and Tissue Regeneration Center, Salzburg, Paracelsus Medical University, Salzburg, Austria.
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| |
Collapse
|
58
|
Rutten-Jacobs LCA, Rost NS. Emerging insights from the genetics of cerebral small-vessel disease. Ann N Y Acad Sci 2020; 1471:5-17. [PMID: 30618052 PMCID: PMC6614021 DOI: 10.1111/nyas.13998] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 12/04/2018] [Accepted: 12/12/2018] [Indexed: 01/09/2023]
Abstract
Cerebral small-vessel disease (cSVD) is a common cause of stroke, functional decline, vascular cognitive impairment, and dementia. Pathological processes in the brain's microcirculation are tightly interwoven with pathology in the brain parenchyma, and this interaction has been conceptualized as the neurovascular unit (NVU). Despite intensive research efforts to decipher the NVU's structure and function to date, molecular mechanisms underlying cSVD remain poorly understood, which hampers the development of cSVD-specific therapies. Important steps forward in understanding the disease mechanisms underlying cSVD have been made using genetic approaches in studies of both monogenic and sporadic SVD. We provide an overview of the NVU's structure and function, the implications for cSVD, and the underlying molecular mechanisms of dysfunction that have emerged from recent genetic studies of both monogenic and sporadic diseases of the small cerebral vasculature.
Collapse
Affiliation(s)
- Loes C A Rutten-Jacobs
- Population Health Sciences, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Natalia S Rost
- Department of Neurology, J. Philip Kistler Stroke Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
59
|
Zhuo Z, Cong L, Zhang J, Zhao X. A novel heterozygous HTRA1 mutation is associated with autosomal dominant hereditary cerebral small vessel disease. Mol Genet Genomic Med 2020; 8:e1111. [PMID: 32239807 PMCID: PMC7284040 DOI: 10.1002/mgg3.1111] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 12/16/2019] [Accepted: 12/16/2019] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND We investigated whether a heterozygous mutation that we newly identified in HTRA1 (high-temperature requirement serine protease A1 gene) in a pedigree with autosomal dominant hereditary cerebral small vessel disease (SVD) reduces the function of HTRA1 and affects the transforming growth factor-β1 (TGF-β1)/Smad signaling. METHODS Whole-exome sequence from the proband and her two sisters was examined using whole-exome enrichment and sequencing. Expression of HTRA1 and TGF-β1/Smad and HTRA1 activity were assayed using sodium dodecyl sulfate-polyacrylamide gel electrophoresis and western blotting analyses after transfecting wild-type and mutant HTRA1 genes into HEK293 cells. RESULTS A new heterozygous mutation (c.614C>G:p.Ser205Cys) in HTRA1 was identified in the sequence encoding the trypsin-like serine protease domain. The mutation was predicted to be deleterious by in silico tools. Moreover, in vitro activity and protein analyses revealed a loss-of-function effect of the mutation: the proteolytic activity of mutant HTRA1 was decreased, and, notably, this was accompanied by an increase in TGF-β1/Smad protein levels. CONCLUSIONS The heterozygous mutation HTRA1 S205C causing diminished protease activity is associated with-and could represent a cause of-autosomal dominant hereditary cerebral SVD. Our results also indicate a relationship between HTRA1 and TGF-β1/Smad signaling.
Collapse
Affiliation(s)
- Zhong‐ling Zhuo
- Department of Clinical LaboratoryPeking University People's HospitalBeijingChina
| | - Lu Cong
- Department of NeurologyPeking University People's HospitalBeijingChina
| | - Jun Zhang
- Department of NeurologyPeking University People's HospitalBeijingChina
| | - Xiao‐tao Zhao
- Department of Clinical LaboratoryPeking University People's HospitalBeijingChina
| |
Collapse
|
60
|
Tom I, Pham VC, Katschke KJ, Li W, Liang WC, Gutierrez J, Ah Young A, Figueroa I, Eshghi ST, Lee CV, Kanodia J, Snipas SJ, Salvesen GS, Lai P, Honigberg L, van Lookeren Campagne M, Kirchhofer D, Baruch A, Lill JR. Development of a therapeutic anti-HtrA1 antibody and the identification of DKK3 as a pharmacodynamic biomarker in geographic atrophy. Proc Natl Acad Sci U S A 2020; 117:9952-9963. [PMID: 32345717 PMCID: PMC7211935 DOI: 10.1073/pnas.1917608117] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Genetic polymorphisms in the region of the trimeric serine hydrolase high-temperature requirement 1 (HTRA1) are associated with increased risk of age-related macular degeneration (AMD) and disease progression, but the precise biological function of HtrA1 in the eye and its contribution to disease etiologies remain undefined. In this study, we have developed an HtrA1-blocking Fab fragment to test the therapeutic hypothesis that HtrA1 protease activity is involved in the progression of AMD. Next, we generated an activity-based small-molecule probe (ABP) to track target engagement in vivo. In addition, we used N-terminomic proteomic profiling in preclinical models to elucidate the in vivo repertoire of HtrA1-specific substrates, and identified substrates that can serve as robust pharmacodynamic biomarkers of HtrA1 activity. One of these HtrA1 substrates, Dickkopf-related protein 3 (DKK3), was successfully used as a biomarker to demonstrate the inhibition of HtrA1 activity in patients with AMD who were treated with the HtrA1-blocking Fab fragment. This pharmacodynamic biomarker provides important information on HtrA1 activity and pharmacological inhibition within the ocular compartment.
Collapse
Affiliation(s)
- Irene Tom
- OMNI Biomarker Development, Genentech, Inc., South San Francisco, CA 94080
| | - Victoria C Pham
- Department of Microchemistry, Proteomics & Lipidomics, Genentech, Inc., South San Francisco, CA 94080
| | - Kenneth J Katschke
- Department of Immunology, Genentech, Inc., South San Francisco, CA 94080
| | - Wei Li
- Department of Early Discovery Biochemistry, Genentech, Inc., South San Francisco, CA 94080
| | - Wei-Ching Liang
- Department of Antibody Discovery, Genentech, Inc., South San Francisco, CA 94080
| | - Johnny Gutierrez
- OMNI Biomarker Development, Genentech, Inc., South San Francisco, CA 94080
| | - Andrew Ah Young
- Department of Early Discovery Biochemistry, Genentech, Inc., South San Francisco, CA 94080
| | - Isabel Figueroa
- Drug Metabolism, Pharmacokinetics, and Bioanalysis, AbbVie, South San Francisco, CA 94090
| | - Shadi Toghi Eshghi
- OMNI Biomarker Development, Genentech, Inc., South San Francisco, CA 94080
| | - ChingWei V Lee
- Biology Core Support, Gilead Sciences, Foster City, CA 94404
| | - Jitendra Kanodia
- Clinical and Translational Pharmacology, Theravance Biopharma, Inc., South San Francisco, CA 94080
| | - Scott J Snipas
- National Cancer Institute-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037
| | - Guy S Salvesen
- National Cancer Institute-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037
| | - Phillip Lai
- Early Clinical Development OMNI Department, Genentech, Inc., South San Francisco, CA 94080
| | - Lee Honigberg
- OMNI Biomarker Development, Genentech, Inc., South San Francisco, CA 94080
| | | | - Daniel Kirchhofer
- Department of Early Discovery Biochemistry, Genentech, Inc., South San Francisco, CA 94080
| | - Amos Baruch
- Biomarker Development, Calico Life Sciences, LLC, South San Francisco, CA 94080
| | - Jennie R Lill
- Department of Microchemistry, Proteomics & Lipidomics, Genentech, Inc., South San Francisco, CA 94080;
| |
Collapse
|
61
|
Müller SJ, Khadhraoui E, Allam I, Argyriou L, Hehr U, Liman J, Hasenfuß G, Bähr M, Riedel CH, Koch JC. CARASIL with coronary artery disease and distinct cerebral microhemorrhage: A case report and literature review. CLINICAL AND TRANSLATIONAL NEUROSCIENCE 2020. [DOI: 10.1177/2514183x20914182] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Cerebral Autosomal Recessive Arteriopathy with Subcortical Infarcts and Leukoencephalopathy (CARASIL, Maeda syndrome) is an extremely rare autosomal-recessive genetic disorder with a serious arteriopathy causing subcortical infarcts and leukoencephalopathy. In less than 20 cases, a genetic mutation was proven. Patients suffer from alopecia, disc herniations, and spondylosis. Between the age of 30 and 40, the patients typically develop severe cerebral infarcts. Clinical symptoms, genetic study, magnetic resonance imaging (MRI), and coronary angiography of a patient with proven CARASIL are presented. The patient showed the typical phenotype with cerebral small-vessel disease, cerebral infarcts, spondylosis, and abnormal hair loss. Additionally, distinct cerebral microhemorrhage and a severe coronary artery disease (CAD) were found, which have not been reported before for CARASIL. Mutation screening revealed the presence of a homozygous c.1022G > T substitution in the HTRA1 gene. Evidence from other publications supports a pathogenetic link between the HTRA1 mutation and CAD as a new feature of CARASIL. This is the first report about CARASIL with a concomitant severe CAD. Thus, in patients with CARASIL, other vessel diseases should also be considered.
Collapse
Affiliation(s)
- Sebastian J Müller
- Institute of Neuroradiology, Georg-August-University Göttingen, Göttingen, Germany
| | - Eya Khadhraoui
- Institute of Neuroradiology, Georg-August-University Göttingen, Göttingen, Germany
| | - Ibrahim Allam
- Department of Neurology, Georg-August-University Göttingen, Göttingen, Germany
| | - Loukas Argyriou
- Institute of Human Genetics, Georg-August-University Göttingen, Göttingen, Germany
| | - Ute Hehr
- Center for Human Genetics Regensburg, Regensburg, Germany
| | - Jan Liman
- Department of Neurology, Georg-August-University Göttingen, Göttingen, Germany
| | - Gerd Hasenfuß
- Department of Cardiology, Georg-August-University Göttingen, Göttingen, Germany
| | - Mathias Bähr
- Department of Neurology, Georg-August-University Göttingen, Göttingen, Germany
| | - Christian H Riedel
- Institute of Neuroradiology, Georg-August-University Göttingen, Göttingen, Germany
| | - Jan C Koch
- Department of Neurology, Georg-August-University Göttingen, Göttingen, Germany
| |
Collapse
|
62
|
Cai R, Wang P, Zhao X, Lu X, Deng R, Wang X, Su Z, Hong C, Lin J. LTBP1 promotes esophageal squamous cell carcinoma progression through epithelial-mesenchymal transition and cancer-associated fibroblasts transformation. J Transl Med 2020; 18:139. [PMID: 32216815 PMCID: PMC7098101 DOI: 10.1186/s12967-020-02310-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 03/17/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Esophageal squamous cell carcinoma (ESCC) is one of the most prevalent cancers worldwide. Due to its high morbidity and mortality rates, it is urgent to find a molecular target that contributes to esophageal carcinogenesis and progression. In this research, we aimed to investigate the functions of Latent transforming growth factor β binding protein 1(LTBP1) in ESCC progression and elucidate the underlying mechanisms. METHODS The tandem mass tag-based quantitative proteomic approach was applied to screen the differentially expressed proteins (DEPs) between 3 cases of ESCC tumor samples and paired normal tissues. Then the DEPs were validated in human ESCC tissues using western blot assays and GEPIA database respectively. The expression level of LTBP1 was detected in 152 cases of ESCC tissues and paired normal tissues. Loss-of-function assays were performed to detect the function of LTBP1 in vivo and in vitro. Immunofluorescence and Western blot assays were used to detect the expression of apoptosis, epithelial-mesenchymal transition (EMT) and cancer-associated fibroblasts (CAFs) markers. RESULTS A total of 39 proteins were screened to be up-regulated (ratio > 2.0) in all three ESCC tissues. The results of immunohistochemistry assays indicated that the expression level of LTBP1 was higher in ESCC tissues than that in paired normal tissues (p < 0.001). Overexpression of LTBP1 was positively associated with lymphatic metastasis in ESCC (p = 0.002). Down-regulation of LTBP1 inhibited the invasion and migration as well as metastatic abilities in vitro and in vivo. It was also observed the down-regulation of LTBP1 not only decreased the mesenchymal phenotypes but also inhibited TGFβ-induced EMT in ESCC cells. We further found that down-regulation of LTBP1 enhanced ESCC cells' sensitivity to 5-FU treatment. Inhibition of LTBP1 expression could also attenuate induction of CAFs transformation and restrain fibroblast express fibronectin (FN1) in ESCC cells. CONCLUSION Overexpression of LTBP1 was associated with lymph node metastasis in ESCC. Our results indicated that LTBP1 not only increased the malignant behaviors of ESCC cells but also induced EMT and CAFs transformation. Our studies suggested an oncogenic role of LTBP1 in ESCC progression and it may serve as a potential therapeutic target for ESCC patients.
Collapse
Affiliation(s)
- Rui Cai
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China.,Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou, Guangdong province, People's Republic of China
| | - Ping Wang
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China.,Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou, Guangdong province, People's Republic of China
| | - Xin Zhao
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China.,Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou, Guangdong province, People's Republic of China
| | - Xiansheng Lu
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China.,Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou, Guangdong province, People's Republic of China
| | - Ruxia Deng
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China.,Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou, Guangdong province, People's Republic of China
| | - Xiaoyu Wang
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China.,Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou, Guangdong province, People's Republic of China
| | - Zhaoji Su
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China.,Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou, Guangdong province, People's Republic of China
| | - Chang Hong
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China.,Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou, Guangdong province, People's Republic of China
| | - Jie Lin
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China. .,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China. .,Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou, Guangdong province, People's Republic of China.
| |
Collapse
|
63
|
Li Y, Yuan J, Rothzerg E, Wu X, Xu H, Zhu S, Xu J. Molecular structure and the role of high-temperature requirement protein 1 in skeletal disorders and cancers. Cell Prolif 2019; 53:e12746. [PMID: 31867863 PMCID: PMC7048211 DOI: 10.1111/cpr.12746] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 11/26/2019] [Accepted: 11/26/2019] [Indexed: 01/09/2023] Open
Abstract
Human high‐temperature requirement protein 1 (HTRA1) is a member of serine proteases and consists of four well‐defined domains—an IGFBP domain, a Kazal domain, a protease domain and a PDZ domain. HTRA1 is a secretory protein and also present intracellularly and associated with microtubules. HTRA1 regulates a broad range of physiological processes via its proteolytic activity. This review examines the role of HTRA1 in bone biology, osteoarthritis, intervertebral disc (IVD) degeneration and tumorigenesis. HTRA1 mediates diverse pathological processes via a variety of signalling pathways, such as TGF‐β and NF‐κB. The expression of HTRA1 is increased in arthritis and IVD degeneration, suggesting that HTRA1 protein is attributed to cartilage degeneration and disease progression. Emerging evidence also suggests that HTRA1 has a role in tumorigenesis. Further understanding the mechanisms by which HTRA1 displays as an extrinsic and intrinsic regulator in a cell type–specific manner will be important for the development of HTRA1 as a therapeutic target.
Collapse
Affiliation(s)
- Yihe Li
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Division of Regenerative Biology, School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia
| | - Jinbo Yuan
- Division of Regenerative Biology, School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia
| | - Emel Rothzerg
- Division of Regenerative Biology, School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia
| | - Xinghuo Wu
- Division of Regenerative Biology, School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia.,Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huazi Xu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Sipin Zhu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Division of Regenerative Biology, School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia
| | - Jiake Xu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Division of Regenerative Biology, School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia
| |
Collapse
|
64
|
Affiliation(s)
- Hugh S Markus
- From the Stroke Research Group, Department of Clinical Neurosciences, University of Cambridge, United Kingdom (H.S.M.)
| | - Reinhold Schmidt
- Department of Neurology, Medical University of Graz, Austria (R.S.)
| |
Collapse
|
65
|
Haffner C. Proteostasis in Cerebral Small Vessel Disease. Front Neurosci 2019; 13:1142. [PMID: 31798396 PMCID: PMC6874119 DOI: 10.3389/fnins.2019.01142] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 10/10/2019] [Indexed: 01/02/2023] Open
Abstract
Maintaining the homeostasis of proteins (proteostasis) by controlling their synthesis, folding and degradation is a central task of cells and tissues. The gradual decline of the capacity of the various proteostasis machineries, frequently in combination with their overload through mutated, aggregation-prone proteins, is increasingly recognized as an important catalyst of age-dependent pathologies in the brain, most prominently neurodegenerative disorders. A dysfunctional proteostasis might also contribute to neurovascular disease as indicated by the occurrence of excessive protein accumulation or massive extracellular matrix expansion within vessel walls in conditions such as cerebral small vessel disease (SVD), a major cause of ischemic stroke, and cerebral amyloid angiopathy. Recent advances in brain vessel isolation techniques and mass spectrometry methodology have facilitated the analysis of cerebrovascular proteomes and fueled efforts to determine the proteomic signatures associated with neurovascular disease. In several studies in humans and mice considerable differences between healthy and diseased vessel proteomes were observed, emphasizing the critical contribution of an impaired proteostasis to disease pathogenesis. These findings highlight the important role of a balanced proteostasis for cerebrovascular health.
Collapse
Affiliation(s)
- Christof Haffner
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-Universität München, Munich, Germany
| |
Collapse
|
66
|
Mustapha M, Nassir CMNCM, Aminuddin N, Safri AA, Ghazali MM. Cerebral Small Vessel Disease (CSVD) - Lessons From the Animal Models. Front Physiol 2019; 10:1317. [PMID: 31708793 PMCID: PMC6822570 DOI: 10.3389/fphys.2019.01317] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Accepted: 09/30/2019] [Indexed: 12/28/2022] Open
Abstract
Cerebral small vessel disease (CSVD) refers to a spectrum of clinical and imaging findings resulting from pathological processes of various etiologies affecting cerebral arterioles, perforating arteries, capillaries, and venules. Unlike large vessels, it is a challenge to visualize small vessels in vivo, hence the difficulty to directly monitor the natural progression of the disease. CSVD might progress for many years during the early stage of the disease as it remains asymptomatic. Prevalent among elderly individuals, CSVD has been alarmingly reported as an important precursor of full-blown stroke and vascular dementia. Growing evidence has also shown a significant association between CSVD's radiological manifestation with dementia and Alzheimer's disease (AD) pathology. Although it remains contentious as to whether CSVD is a cause or sequelae of AD, it is not far-fetched to posit that effective therapeutic measures of CSVD would mitigate the overall burden of dementia. Nevertheless, the unifying theory on the pathomechanism of the disease remains elusive, hence the lack of effective therapeutic approaches. Thus, this chapter consolidates the contemporary insights from numerous experimental animal models of CSVD, to date: from the available experimental animal models of CSVD and its translational research value; the pathomechanical aspects of the disease; relevant aspects on systems biology; opportunities for early disease biomarkers; and finally, converging approaches for future therapeutic directions of CSVD.
Collapse
Affiliation(s)
- Muzaimi Mustapha
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | | | - Niferiti Aminuddin
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
- Department of Basic Medical Sciences, Kulliyyah of Pharmacy, International Islamic University Malaysia, Kuantan, Malaysia
| | - Amanina Ahmad Safri
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | - Mazira Mohamad Ghazali
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| |
Collapse
|
67
|
Giau VV, Bagyinszky E, Youn YC, An SSA, Kim SY. Genetic Factors of Cerebral Small Vessel Disease and Their Potential Clinical Outcome. Int J Mol Sci 2019; 20:ijms20174298. [PMID: 31484286 PMCID: PMC6747336 DOI: 10.3390/ijms20174298] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 08/27/2019] [Accepted: 09/01/2019] [Indexed: 12/23/2022] Open
Abstract
Cerebral small vessel diseases (SVD) have been causally correlated with ischemic strokes, leading to cognitive decline and vascular dementia. Neuroimaging and molecular genetic tests could improve diagnostic accuracy in patients with potential SVD. Several types of monogenic, hereditary cerebral SVD have been identified: cerebral autosomal recessive arteriopathy with subcortical infarcts and leukoencephalopathy (CARASIL), cerebral autosomal-dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL), cathepsin A-related arteriopathy with strokes and leukoencephalopathy (CARASAL), hereditary diffuse leukoencephalopathy with spheroids (HDLS), COL4A1/2-related disorders, and Fabry disease. These disorders can be distinguished based on their genetics, pathological and imaging findings, clinical manifestation, and diagnosis. Genetic studies of sporadic cerebral SVD have demonstrated a high degree of heritability, particularly among patients with young-onset stroke. Common genetic variants in monogenic disease may contribute to pathological progress in several cerebral SVD subtypes, revealing distinct genetic mechanisms in different subtype of SVD. Hence, genetic molecular analysis should be used as the final gold standard of diagnosis. The purpose of this review was to summarize the recent discoveries made surrounding the genetics of cerebral SVD and their clinical significance, to provide new insights into the pathogenesis of cerebral SVD, and to highlight the possible convergence of disease mechanisms in monogenic and sporadic cerebral SVD.
Collapse
Affiliation(s)
- Vo Van Giau
- Department of Bionano Technology & Gachon Bionano Research Institute, Gachon University, Seongnam-si, Gyeonggi-do 461-701, Korea
| | - Eva Bagyinszky
- Department of Bionano Technology & Gachon Bionano Research Institute, Gachon University, Seongnam-si, Gyeonggi-do 461-701, Korea
| | - Young Chul Youn
- Department of Neurology, Chung-Ang University College of Medicine, Seoul 06973, Korea.
| | - Seong Soo A An
- Department of Bionano Technology & Gachon Bionano Research Institute, Gachon University, Seongnam-si, Gyeonggi-do 461-701, Korea.
| | - Sang Yun Kim
- Department of Neurology, Seoul National University College of Medicine & Neurocognitive Behavior Center, Seoul National University Bundang Hospital, Seoul 06973, Korea
| |
Collapse
|
68
|
Wenta T, Jarzab M, Rychlowski M, Borysiak M, Latala A, Zurawa-Janicka D, Filipek A, Lipinska B. Cellular substrates and pro-apoptotic function of the human HtrA4 protease. J Proteomics 2019; 209:103505. [PMID: 31470122 DOI: 10.1016/j.jprot.2019.103505] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 07/25/2019] [Accepted: 08/21/2019] [Indexed: 12/19/2022]
Abstract
The human HtrA4 protein, belonging to the HtrA family of proteases/chaperones, participates in oncogenesis and placentation, and plays a role in preeclampsia. As the knowledge concerning the biochemical features of this protein and its role at the molecular level is limited, in this work we characterized the HtrA4 molecule and searched for its cellular function. We found that recombinant HtrA4 composed of the protease and PDZ domains is a trimeric protein of intermediate thermal stability whose activity is considerably lower compared to other human HtrA proteases. By pull-down combined with mass spectrometry we identified a large array of potential HtrA4 partners. Using other experimental approaches, including immunoprecipitation, enzyme-linked immunosorbent assay and fluorescence microscopy we confirmed that HtrA4 formed complexes in vitro and in cellulo with proteins such as XIAP (inhibitor of apoptosis protein), caspases 7 and 9, β-tubulin, actin, TCP1α and S100A6. The recombinant HtrA4 degraded XIAP, the caspases, β-tubulin and actin but not TCP1α or S100A6. Together, these results suggest that HtrA4 may influence various cellular functions, including apoptosis. Furthermore, the panel of potential HtrA4 partners may serve as a basis for future studies of HtrA4 function.
Collapse
Affiliation(s)
- Tomasz Wenta
- Department of General and Medical Biochemistry, Faculty of Biology, University of Gdansk, Poland
| | - Miroslaw Jarzab
- Department of General and Medical Biochemistry, Faculty of Biology, University of Gdansk, Poland
| | - Michal Rychlowski
- Laboratory of Virus Molecular Biology, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Gdansk, Poland
| | - Monika Borysiak
- Department of General and Medical Biochemistry, Faculty of Biology, University of Gdansk, Poland
| | - Anna Latala
- Department of General and Medical Biochemistry, Faculty of Biology, University of Gdansk, Poland
| | - Dorota Zurawa-Janicka
- Department of General and Medical Biochemistry, Faculty of Biology, University of Gdansk, Poland
| | - Anna Filipek
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Barbara Lipinska
- Department of General and Medical Biochemistry, Faculty of Biology, University of Gdansk, Poland.
| |
Collapse
|
69
|
Malik R, Dichgans M. Challenges and opportunities in stroke genetics. Cardiovasc Res 2019; 114:1226-1240. [PMID: 29554300 DOI: 10.1093/cvr/cvy068] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 03/14/2018] [Indexed: 12/13/2022] Open
Abstract
Stroke, ischaemic stroke and subtypes of ischaemic stroke display substantial heritability. When compared with related vascular conditions, the number of established risk loci reaching genome-wide significance for association with stroke is still in the lower range, particularly for aetiological stroke subtypes such as large artery atherosclerotic stroke or small vessel stroke. Nevertheless, for individual loci substantial progress has been made in determining the specific mechanisms mediating stroke risk. In this review, we present a roadmap for functional follow-up of common risk variants associated with stroke. First, we discuss in silico strategies for characterizing signals in non-coding regions and highlight databases providing information on quantitative trait loci for mRNA and protein expression, as well as methylation, focussing on those with presumed relevance for stroke. Next, we discuss experimental strategies for following up on non-coding risk variants and regions such as massively parallel reporter assays, proteome-wide association studies, and chromatin conformation capture (3C) assays. These and other approaches are relevant for gaining insight into the specific variants and mechanisms mediating genetic stroke risk. Finally, we discuss how genetic findings could influence clinical practice by adding to diagnostic algorithms and eventually improve treatment options for stroke.
Collapse
Affiliation(s)
- Rainer Malik
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilians-Universität (LMU) München, Feodor-Lynen-Straße 17, Munich, Germany
| | - Martin Dichgans
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilians-Universität (LMU) München, Feodor-Lynen-Straße 17, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Feodor-Lynen-Straße 17, Munich, Germany
| |
Collapse
|
70
|
Ziaei A, Xu X, Dehghani L, Bonnard C, Zellner A, Jin Ng AY, Tohari S, Venkatesh B, Haffner C, Reversade B, Shaygannejad V, Pouladi MA. Novel mutation in HTRA1 in a family with diffuse white matter lesions and inflammatory features. NEUROLOGY-GENETICS 2019; 5:e345. [PMID: 31403081 PMCID: PMC6659136 DOI: 10.1212/nxg.0000000000000345] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 05/28/2019] [Indexed: 11/24/2022]
Abstract
Objective To investigate the possible involvement of germline mutations in a neurologic condition involving diffuse white matter lesions. Methods The patients were 3 siblings born to healthy parents. We performed homozygosity mapping, whole-exome sequencing, site-directed mutagenesis, and immunoblotting. Results All 3 patients showed clinical manifestations of ataxia, behavioral and mood changes, premature hair loss, memory loss, and lower back pain. In addition, they presented with inflammatory-like features and recurrent rhinitis. MRI showed abnormal diffuse demyelination lesions in the brain and myelitis in the spinal cord. We identified an insertion in high-temperature requirement A (HTRA1), which showed complete segregation in the pedigree. Functional analysis showed the mutation to affect stability and secretion of truncated protein. Conclusions The patients' clinical manifestations are consistent with cerebral autosomal recessive arteriopathy with subcortical infarcts and leukoencephalopathy (CARASIL; OMIM #600142), which is known to be caused by HTRA1 mutations. Because some aspects of the clinical presentation deviate from those reported for CARASIL, our study expands the spectrum of clinical consequences of loss-of-function mutations in HTRA1.
Collapse
Affiliation(s)
- Amin Ziaei
- Translational Laboratory in Genetic Medicine (TLGM) (A. Ziaei, X.X., M.A.P.), Agency for Science, Technology and Research (ASTAR), 8A Biomedical Grove, Immunos, Level 5; Department of Medicine (A. Ziaei, M.A.P.), National University of Singapore; Department of Neurology and Stroke Center (X.X.), the First Affiliated Hospital, Jinan University; Clinical Neuroscience Institute of Jinan University (X.X.), Guangzhou, Guangdong, China; Department of Tissue Engineering and Regenerative Medicine (L.D.), School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Institute of Medical Biology (IMB) (C.B., B.R.), A*STAR, 8A Biomedical Grove, Immunos, Level 5, Singapore; Institute for Stroke and Dementia Research (A. Zellner, C.H.), Klinikum der Universität München, Ludwig Maximilians University, Munich, Germany; Comparative Genomics Laboratory (A.Y.J.N., S.T., B.V.), Institute of Molecular and Cell Biology, A*STAR, Biopolis; Department of Paediatrics (B.V.), National University of Singapore; Department of Neurology (A. Ziaei, V.S.), Isfahan Neurosciences Research Centre, Faculty of Medicine, Isfahan University of Medical Sciences, Iran; and Department of Physiology (M.A.P.), National University of Singapore
| | - Xiaohong Xu
- Translational Laboratory in Genetic Medicine (TLGM) (A. Ziaei, X.X., M.A.P.), Agency for Science, Technology and Research (ASTAR), 8A Biomedical Grove, Immunos, Level 5; Department of Medicine (A. Ziaei, M.A.P.), National University of Singapore; Department of Neurology and Stroke Center (X.X.), the First Affiliated Hospital, Jinan University; Clinical Neuroscience Institute of Jinan University (X.X.), Guangzhou, Guangdong, China; Department of Tissue Engineering and Regenerative Medicine (L.D.), School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Institute of Medical Biology (IMB) (C.B., B.R.), A*STAR, 8A Biomedical Grove, Immunos, Level 5, Singapore; Institute for Stroke and Dementia Research (A. Zellner, C.H.), Klinikum der Universität München, Ludwig Maximilians University, Munich, Germany; Comparative Genomics Laboratory (A.Y.J.N., S.T., B.V.), Institute of Molecular and Cell Biology, A*STAR, Biopolis; Department of Paediatrics (B.V.), National University of Singapore; Department of Neurology (A. Ziaei, V.S.), Isfahan Neurosciences Research Centre, Faculty of Medicine, Isfahan University of Medical Sciences, Iran; and Department of Physiology (M.A.P.), National University of Singapore
| | - Leila Dehghani
- Translational Laboratory in Genetic Medicine (TLGM) (A. Ziaei, X.X., M.A.P.), Agency for Science, Technology and Research (ASTAR), 8A Biomedical Grove, Immunos, Level 5; Department of Medicine (A. Ziaei, M.A.P.), National University of Singapore; Department of Neurology and Stroke Center (X.X.), the First Affiliated Hospital, Jinan University; Clinical Neuroscience Institute of Jinan University (X.X.), Guangzhou, Guangdong, China; Department of Tissue Engineering and Regenerative Medicine (L.D.), School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Institute of Medical Biology (IMB) (C.B., B.R.), A*STAR, 8A Biomedical Grove, Immunos, Level 5, Singapore; Institute for Stroke and Dementia Research (A. Zellner, C.H.), Klinikum der Universität München, Ludwig Maximilians University, Munich, Germany; Comparative Genomics Laboratory (A.Y.J.N., S.T., B.V.), Institute of Molecular and Cell Biology, A*STAR, Biopolis; Department of Paediatrics (B.V.), National University of Singapore; Department of Neurology (A. Ziaei, V.S.), Isfahan Neurosciences Research Centre, Faculty of Medicine, Isfahan University of Medical Sciences, Iran; and Department of Physiology (M.A.P.), National University of Singapore
| | - Carine Bonnard
- Translational Laboratory in Genetic Medicine (TLGM) (A. Ziaei, X.X., M.A.P.), Agency for Science, Technology and Research (ASTAR), 8A Biomedical Grove, Immunos, Level 5; Department of Medicine (A. Ziaei, M.A.P.), National University of Singapore; Department of Neurology and Stroke Center (X.X.), the First Affiliated Hospital, Jinan University; Clinical Neuroscience Institute of Jinan University (X.X.), Guangzhou, Guangdong, China; Department of Tissue Engineering and Regenerative Medicine (L.D.), School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Institute of Medical Biology (IMB) (C.B., B.R.), A*STAR, 8A Biomedical Grove, Immunos, Level 5, Singapore; Institute for Stroke and Dementia Research (A. Zellner, C.H.), Klinikum der Universität München, Ludwig Maximilians University, Munich, Germany; Comparative Genomics Laboratory (A.Y.J.N., S.T., B.V.), Institute of Molecular and Cell Biology, A*STAR, Biopolis; Department of Paediatrics (B.V.), National University of Singapore; Department of Neurology (A. Ziaei, V.S.), Isfahan Neurosciences Research Centre, Faculty of Medicine, Isfahan University of Medical Sciences, Iran; and Department of Physiology (M.A.P.), National University of Singapore
| | - Andreas Zellner
- Translational Laboratory in Genetic Medicine (TLGM) (A. Ziaei, X.X., M.A.P.), Agency for Science, Technology and Research (ASTAR), 8A Biomedical Grove, Immunos, Level 5; Department of Medicine (A. Ziaei, M.A.P.), National University of Singapore; Department of Neurology and Stroke Center (X.X.), the First Affiliated Hospital, Jinan University; Clinical Neuroscience Institute of Jinan University (X.X.), Guangzhou, Guangdong, China; Department of Tissue Engineering and Regenerative Medicine (L.D.), School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Institute of Medical Biology (IMB) (C.B., B.R.), A*STAR, 8A Biomedical Grove, Immunos, Level 5, Singapore; Institute for Stroke and Dementia Research (A. Zellner, C.H.), Klinikum der Universität München, Ludwig Maximilians University, Munich, Germany; Comparative Genomics Laboratory (A.Y.J.N., S.T., B.V.), Institute of Molecular and Cell Biology, A*STAR, Biopolis; Department of Paediatrics (B.V.), National University of Singapore; Department of Neurology (A. Ziaei, V.S.), Isfahan Neurosciences Research Centre, Faculty of Medicine, Isfahan University of Medical Sciences, Iran; and Department of Physiology (M.A.P.), National University of Singapore
| | - Alvin Yu Jin Ng
- Translational Laboratory in Genetic Medicine (TLGM) (A. Ziaei, X.X., M.A.P.), Agency for Science, Technology and Research (ASTAR), 8A Biomedical Grove, Immunos, Level 5; Department of Medicine (A. Ziaei, M.A.P.), National University of Singapore; Department of Neurology and Stroke Center (X.X.), the First Affiliated Hospital, Jinan University; Clinical Neuroscience Institute of Jinan University (X.X.), Guangzhou, Guangdong, China; Department of Tissue Engineering and Regenerative Medicine (L.D.), School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Institute of Medical Biology (IMB) (C.B., B.R.), A*STAR, 8A Biomedical Grove, Immunos, Level 5, Singapore; Institute for Stroke and Dementia Research (A. Zellner, C.H.), Klinikum der Universität München, Ludwig Maximilians University, Munich, Germany; Comparative Genomics Laboratory (A.Y.J.N., S.T., B.V.), Institute of Molecular and Cell Biology, A*STAR, Biopolis; Department of Paediatrics (B.V.), National University of Singapore; Department of Neurology (A. Ziaei, V.S.), Isfahan Neurosciences Research Centre, Faculty of Medicine, Isfahan University of Medical Sciences, Iran; and Department of Physiology (M.A.P.), National University of Singapore
| | - Sumanty Tohari
- Translational Laboratory in Genetic Medicine (TLGM) (A. Ziaei, X.X., M.A.P.), Agency for Science, Technology and Research (ASTAR), 8A Biomedical Grove, Immunos, Level 5; Department of Medicine (A. Ziaei, M.A.P.), National University of Singapore; Department of Neurology and Stroke Center (X.X.), the First Affiliated Hospital, Jinan University; Clinical Neuroscience Institute of Jinan University (X.X.), Guangzhou, Guangdong, China; Department of Tissue Engineering and Regenerative Medicine (L.D.), School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Institute of Medical Biology (IMB) (C.B., B.R.), A*STAR, 8A Biomedical Grove, Immunos, Level 5, Singapore; Institute for Stroke and Dementia Research (A. Zellner, C.H.), Klinikum der Universität München, Ludwig Maximilians University, Munich, Germany; Comparative Genomics Laboratory (A.Y.J.N., S.T., B.V.), Institute of Molecular and Cell Biology, A*STAR, Biopolis; Department of Paediatrics (B.V.), National University of Singapore; Department of Neurology (A. Ziaei, V.S.), Isfahan Neurosciences Research Centre, Faculty of Medicine, Isfahan University of Medical Sciences, Iran; and Department of Physiology (M.A.P.), National University of Singapore
| | - Byrappa Venkatesh
- Translational Laboratory in Genetic Medicine (TLGM) (A. Ziaei, X.X., M.A.P.), Agency for Science, Technology and Research (ASTAR), 8A Biomedical Grove, Immunos, Level 5; Department of Medicine (A. Ziaei, M.A.P.), National University of Singapore; Department of Neurology and Stroke Center (X.X.), the First Affiliated Hospital, Jinan University; Clinical Neuroscience Institute of Jinan University (X.X.), Guangzhou, Guangdong, China; Department of Tissue Engineering and Regenerative Medicine (L.D.), School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Institute of Medical Biology (IMB) (C.B., B.R.), A*STAR, 8A Biomedical Grove, Immunos, Level 5, Singapore; Institute for Stroke and Dementia Research (A. Zellner, C.H.), Klinikum der Universität München, Ludwig Maximilians University, Munich, Germany; Comparative Genomics Laboratory (A.Y.J.N., S.T., B.V.), Institute of Molecular and Cell Biology, A*STAR, Biopolis; Department of Paediatrics (B.V.), National University of Singapore; Department of Neurology (A. Ziaei, V.S.), Isfahan Neurosciences Research Centre, Faculty of Medicine, Isfahan University of Medical Sciences, Iran; and Department of Physiology (M.A.P.), National University of Singapore
| | - Christof Haffner
- Translational Laboratory in Genetic Medicine (TLGM) (A. Ziaei, X.X., M.A.P.), Agency for Science, Technology and Research (ASTAR), 8A Biomedical Grove, Immunos, Level 5; Department of Medicine (A. Ziaei, M.A.P.), National University of Singapore; Department of Neurology and Stroke Center (X.X.), the First Affiliated Hospital, Jinan University; Clinical Neuroscience Institute of Jinan University (X.X.), Guangzhou, Guangdong, China; Department of Tissue Engineering and Regenerative Medicine (L.D.), School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Institute of Medical Biology (IMB) (C.B., B.R.), A*STAR, 8A Biomedical Grove, Immunos, Level 5, Singapore; Institute for Stroke and Dementia Research (A. Zellner, C.H.), Klinikum der Universität München, Ludwig Maximilians University, Munich, Germany; Comparative Genomics Laboratory (A.Y.J.N., S.T., B.V.), Institute of Molecular and Cell Biology, A*STAR, Biopolis; Department of Paediatrics (B.V.), National University of Singapore; Department of Neurology (A. Ziaei, V.S.), Isfahan Neurosciences Research Centre, Faculty of Medicine, Isfahan University of Medical Sciences, Iran; and Department of Physiology (M.A.P.), National University of Singapore
| | - Bruno Reversade
- Translational Laboratory in Genetic Medicine (TLGM) (A. Ziaei, X.X., M.A.P.), Agency for Science, Technology and Research (ASTAR), 8A Biomedical Grove, Immunos, Level 5; Department of Medicine (A. Ziaei, M.A.P.), National University of Singapore; Department of Neurology and Stroke Center (X.X.), the First Affiliated Hospital, Jinan University; Clinical Neuroscience Institute of Jinan University (X.X.), Guangzhou, Guangdong, China; Department of Tissue Engineering and Regenerative Medicine (L.D.), School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Institute of Medical Biology (IMB) (C.B., B.R.), A*STAR, 8A Biomedical Grove, Immunos, Level 5, Singapore; Institute for Stroke and Dementia Research (A. Zellner, C.H.), Klinikum der Universität München, Ludwig Maximilians University, Munich, Germany; Comparative Genomics Laboratory (A.Y.J.N., S.T., B.V.), Institute of Molecular and Cell Biology, A*STAR, Biopolis; Department of Paediatrics (B.V.), National University of Singapore; Department of Neurology (A. Ziaei, V.S.), Isfahan Neurosciences Research Centre, Faculty of Medicine, Isfahan University of Medical Sciences, Iran; and Department of Physiology (M.A.P.), National University of Singapore
| | - Vahid Shaygannejad
- Translational Laboratory in Genetic Medicine (TLGM) (A. Ziaei, X.X., M.A.P.), Agency for Science, Technology and Research (ASTAR), 8A Biomedical Grove, Immunos, Level 5; Department of Medicine (A. Ziaei, M.A.P.), National University of Singapore; Department of Neurology and Stroke Center (X.X.), the First Affiliated Hospital, Jinan University; Clinical Neuroscience Institute of Jinan University (X.X.), Guangzhou, Guangdong, China; Department of Tissue Engineering and Regenerative Medicine (L.D.), School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Institute of Medical Biology (IMB) (C.B., B.R.), A*STAR, 8A Biomedical Grove, Immunos, Level 5, Singapore; Institute for Stroke and Dementia Research (A. Zellner, C.H.), Klinikum der Universität München, Ludwig Maximilians University, Munich, Germany; Comparative Genomics Laboratory (A.Y.J.N., S.T., B.V.), Institute of Molecular and Cell Biology, A*STAR, Biopolis; Department of Paediatrics (B.V.), National University of Singapore; Department of Neurology (A. Ziaei, V.S.), Isfahan Neurosciences Research Centre, Faculty of Medicine, Isfahan University of Medical Sciences, Iran; and Department of Physiology (M.A.P.), National University of Singapore
| | - Mahmoud A Pouladi
- Translational Laboratory in Genetic Medicine (TLGM) (A. Ziaei, X.X., M.A.P.), Agency for Science, Technology and Research (ASTAR), 8A Biomedical Grove, Immunos, Level 5; Department of Medicine (A. Ziaei, M.A.P.), National University of Singapore; Department of Neurology and Stroke Center (X.X.), the First Affiliated Hospital, Jinan University; Clinical Neuroscience Institute of Jinan University (X.X.), Guangzhou, Guangdong, China; Department of Tissue Engineering and Regenerative Medicine (L.D.), School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Institute of Medical Biology (IMB) (C.B., B.R.), A*STAR, 8A Biomedical Grove, Immunos, Level 5, Singapore; Institute for Stroke and Dementia Research (A. Zellner, C.H.), Klinikum der Universität München, Ludwig Maximilians University, Munich, Germany; Comparative Genomics Laboratory (A.Y.J.N., S.T., B.V.), Institute of Molecular and Cell Biology, A*STAR, Biopolis; Department of Paediatrics (B.V.), National University of Singapore; Department of Neurology (A. Ziaei, V.S.), Isfahan Neurosciences Research Centre, Faculty of Medicine, Isfahan University of Medical Sciences, Iran; and Department of Physiology (M.A.P.), National University of Singapore
| |
Collapse
|
71
|
Uemura M, Nozaki H, Koyama A, Sakai N, Ando S, Kanazawa M, Kato T, Onodera O. HTRA1 Mutations Identified in Symptomatic Carriers Have the Property of Interfering the Trimer-Dependent Activation Cascade. Front Neurol 2019; 10:693. [PMID: 31316458 PMCID: PMC6611441 DOI: 10.3389/fneur.2019.00693] [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: 03/21/2019] [Accepted: 06/13/2019] [Indexed: 12/03/2022] Open
Abstract
Background: Mutations in the high-temperature requirement A serine peptidase 1 (HTRA1) cause cerebral autosomal recessive arteriopathy with subcortical infarcts and leukoencephalopathy (CARASIL). Most carriers for HTRA1 mutations are asymptomatic, but more than 10 mutations have been reported in symptomatic carriers. The molecular differences between the mutations identified in symptomatic carriers and mutations identified only in CARASIL patients are unclear. HTRA1 is a serine protease that forms homotrimers, with each HTRA1 subunit activating the adjacent HTRA1 via the sensor domain of loop 3 (L3) and the activation domain of loop D (LD). Previously, we analyzed four HTRA1 mutant proteins identified in symptomatic carriers and found that they were unable to form trimers or had mutations in the LD or L3 domain. The mutant HTRA1s with these properties are presumed to inhibit trimer-dependent activation cascade. Indeed, these mutant HTRA1s inhibited wild-type (WT) protease activity. In this study, we further analyzed 15 missense HTRA1s to clarify the molecular character of mutant HTRA1s identified in symptomatic carriers. Methods: We analyzed 12 missense HTRA1s identified in symptomatic carriers (hetero-HTRA1) and three missense HTRA1s found only in CARASIL (CARASIL-HTRA1). The protease activity of the purified recombinant mutant HTRA1s was measured using fluorescein isothiocyanate-labeled casein as substrate. Oligomeric structure was evaluated by size-exclusion chromatography. The protease activities of mixtures of WT with each mutant HTRA1 were also measured. Results: Five hetero-HTRA1s had normal protease activity and were excluded from further analysis. Four of the seven hetero-HTRA1s and one of the three CARASIL-HTRA1s were unable to form trimers. The other three hetero-HTRA1s had mutations in the LD domain. Together with our previous work, 10 of 11 hetero-HTRA1s and two of six CARASIL-HTRA1s were either defective in trimerization or had mutations in the LD or L3 domain (P = 0.006). By contrast, eight of 11 hetero-HTRA1s and two of six CARASIL-HTRA1 inhibited WT protease activity (P = 0.162). Conclusions: HTRA1 mutations identified in symptomatic carriers have the property of interfering the trimer-dependent activation cascade of HTRA1.
Collapse
Affiliation(s)
- Masahiro Uemura
- Department of Neurology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Hiroaki Nozaki
- Department of Medical Technology, Graduate School of Health Sciences, Niigata University, Niigata, Japan
| | - Akihide Koyama
- Department of Neurology, Brain Research Institute, Niigata University, Niigata, Japan.,Division of Legal Medicine, Niigata University, Niigata, Japan
| | - Naoko Sakai
- Department of Neurology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Shoichiro Ando
- Department of Neurology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Masato Kanazawa
- Department of Neurology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Taisuke Kato
- Department of System Pathology for Neurological Disorders, Brain Research Institute, Niigata University, Niigata, Japan
| | - Osamu Onodera
- Department of Neurology, Brain Research Institute, Niigata University, Niigata, Japan
| |
Collapse
|
72
|
Schütz R, Rawlings A, Wandeler E, Jackson E, Trevisan S, Monneuse J, Bendik I, Massironi M, Imfeld D. Bio-derived hydroxystearic acid ameliorates skin age spots and conspicuous pores. Int J Cosmet Sci 2019; 41:240-256. [PMID: 30955220 PMCID: PMC6852045 DOI: 10.1111/ics.12529] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 04/03/2019] [Indexed: 12/21/2022]
Abstract
INTRODUCTION We report on the preparation and efficacy of 10-hydroxystearic acid (HSA) that improves facial age spots and conspicuous pores. METHODS The hydration of oleic acid into HSA was catalyzed by the oleate hydratase from Escherichia coli. Following treatment with HSA, collagen type I and type III was assessed in primary human dermal fibroblasts together with collagen type III, p53 protein levels and sunburn cells (SBC) after UVB irradiation (1 J cm-2 ) by immunohistochemistry on human ex vivo skin. UVB-induced expression of matrix metalloprotease-1 (MMP-1) was determined from full thickness skin by RT-qPCR. Modification of the fibroblast secretome by HSA was studied by mass-spectrometry-based proteomics. In a full-face, double blind, vehicle-controlled trial HSA was assessed for its effects on conspicuous facial pore size and degree of pigmentation of age spots in Caucasian women over an 8-week period. RESULTS HSA was obtained in enantiomeric pure, high yield (≥80%). Collagen type I and type III levels were dose-dependently increased (96% and 244%; P < 0.01) in vitro and collagen type III in ex vivo skin by +57% (P < 0.01) by HSA. HSA also inhibited UVB-induced MMP-1 gene expression (83%; P < 0.01) and mitigated SBC induction (-34% vs. vehicle control) and reduced significantly UV-induced p53 up-regulation (-46% vs. vehicle control; P < 0.01) in irradiated skin. HSA modified the fibroblast secretome with significant increases in proteins associated with the WNT pathway that could reduce melanogenesis and proteins that could modify dermal fibroblast activity and keratinocyte differentiation to account for the alleviation of conspicuous pores. Docking studies in silico and EC50 determination in reporter gene assays (EC50 5.5 × 10-6 M) identified HSA as a peroxisomal proliferator activated receptor-α (PPARα) agonist. Clinically, HSA showed a statistically significant decrease of surface and volume of skin pores (P < 0.05) after 8 weeks of application and age spots became significantly less pigmented than the surrounding skin (contrast, P < 0.05) after 4 weeks. CONCLUSION HSA acts as a PPARα agonist to reduce the signs of age spots and conspicuous pores by significantly modulating the expression of p53, SBC, MMP-1 and collagen together with major changes in secreted proteins that modify keratinocyte, melanocyte and fibroblast cell behavior.
Collapse
Affiliation(s)
- R. Schütz
- DSM Nutritional Products Ltd.KaiseraugstSwitzerland
| | | | - E. Wandeler
- DSM Nutritional Products Ltd.KaiseraugstSwitzerland
| | - E. Jackson
- DSM Nutritional Products Ltd.KaiseraugstSwitzerland
| | | | | | - I. Bendik
- DSM Nutritional Products Ltd.KaiseraugstSwitzerland
| | | | - D. Imfeld
- DSM Nutritional Products Ltd.KaiseraugstSwitzerland
| |
Collapse
|
73
|
Dichgans M, Pulit SL, Rosand J. Stroke genetics: discovery, biology, and clinical applications. Lancet Neurol 2019; 18:587-599. [PMID: 30975520 DOI: 10.1016/s1474-4422(19)30043-2] [Citation(s) in RCA: 105] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 01/21/2019] [Accepted: 01/24/2019] [Indexed: 02/07/2023]
Abstract
Stroke, a leading cause of long-term disability and death worldwide, has a heritable component. Recent gene discovery efforts have expanded the number of known single-gene disorders associated with stroke and have linked common variants at approximately 35 genetic loci to stroke risk. These discoveries have highlighted novel mechanisms and pathways implicated in stroke related to large artery atherosclerosis, cardioembolism, and small vessel disease, and defined shared genetic influences with related vascular traits. Genetics has also successfully established causal relationships with risk factors and holds promise for prioritising targets for exploration in clinical trials. Genome-wide polygenic scores enable the identification of high-risk individuals before the emergence of vascular risk factors. Challenges ahead include a better understanding of rare variants and ancestral differences for integration of genetics into precision medicine, integration with other omics data, uncovering the genetic factors that govern stroke recurrence and stroke outcome, and the conversion of genetic discoveries to novel therapies.
Collapse
Affiliation(s)
- Martin Dichgans
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-Universität, Munich, Germany; German Center for Neurodegenerative Diseases, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| | - Sara L Pulit
- Department of Genetics, Centre for Molecular Medicine, University Medical Centre Utrecht, Utrecht, Netherlands; Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, Oxford University, Oxford, UK; Program in Medical Population and Genetics, Broad Institute, Cambridge, MA, USA
| | - Jonathan Rosand
- Program in Medical Population and Genetics, Broad Institute, Cambridge, MA, USA; Henry and Allison McCance Center for Brain Health, and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
74
|
Mishra A, Chauhan G, Violleau MH, Vojinovic D, Jian X, Bis JC, Li S, Saba Y, Grenier-Boley B, Yang Q, Bartz TM, Hofer E, Soumaré A, Peng F, Duperron MG, Foglio M, Mosley TH, Schmidt R, Psaty BM, Launer LJ, Boerwinkle E, Zhu Y, Mazoyer B, Lathrop M, Bellenguez C, Van Duijn CM, Ikram MA, Schmidt H, Longstreth WT, Fornage M, Seshadri S, Joutel A, Tzourio C, Debette S. Association of variants in HTRA1 and NOTCH3 with MRI-defined extremes of cerebral small vessel disease in older subjects. Brain 2019; 142:1009-1023. [PMID: 30859180 PMCID: PMC6439324 DOI: 10.1093/brain/awz024] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 11/30/2018] [Accepted: 12/21/2018] [Indexed: 12/20/2022] Open
Abstract
We report a composite extreme phenotype design using distribution of white matter hyperintensities and brain infarcts in a population-based cohort of older persons for gene-mapping of cerebral small vessel disease. We demonstrate its application in the 3C-Dijon whole exome sequencing (WES) study (n = 1924, nWESextremes = 512), with both single variant and gene-based association tests. We used other population-based cohort studies participating in the CHARGE consortium for replication, using whole exome sequencing (nWES = 2,868, nWESextremes = 956) and genome-wide genotypes (nGW = 9924, nGWextremes = 3308). We restricted our study to candidate genes known to harbour mutations for Mendelian small vessel disease: NOTCH3, HTRA1, COL4A1, COL4A2 and TREX1. We identified significant associations of a common intronic variant in HTRA1, rs2293871 using single variant association testing (Pdiscovery = 8.21 × 10-5, Preplication = 5.25 × 10-3, Pcombined = 4.72 × 10-5) and of NOTCH3 using gene-based tests (Pdiscovery = 1.61 × 10-2, Preplication = 3.99 × 10-2, Pcombined = 5.31 × 10-3). Follow-up analysis identified significant association of rs2293871 with small vessel ischaemic stroke, and two blood expression quantitative trait loci of HTRA1 in linkage disequilibrium. Additionally, we identified two participants in the 3C-Dijon cohort (0.4%) carrying heterozygote genotypes at known pathogenic variants for familial small vessel disease within NOTCH3 and HTRA1. In conclusion, our proof-of-concept study provides strong evidence that using a novel composite MRI-derived phenotype for extremes of small vessel disease can facilitate the identification of genetic variants underlying small vessel disease, both common variants and those with rare and low frequency. The findings demonstrate shared mechanisms and a continuum between genes underlying Mendelian small vessel disease and those contributing to the common, multifactorial form of the disease.
Collapse
Affiliation(s)
- Aniket Mishra
- University of Bordeaux, Inserm, Bordeaux Population Health Research Center, team VINTAGE, UMR 1219, F-33000 Bordeaux, France
| | - Ganesh Chauhan
- University of Bordeaux, Inserm, Bordeaux Population Health Research Center, team VINTAGE, UMR 1219, F-33000 Bordeaux, France
- Centre for Brain Research, Indian Institute of Science, Bangalore, India
| | - Marie-Helene Violleau
- University of Bordeaux, Inserm, Bordeaux Population Health Research Center, team VINTAGE, UMR 1219, F-33000 Bordeaux, France
| | - Dina Vojinovic
- Department of Epidemiology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Xueqiu Jian
- The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Joshua C Bis
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Shuo Li
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Yasaman Saba
- Gottfried Schatz Research Center, Department of Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Benjamin Grenier-Boley
- Inserm, U1167, RID-AGE - Risk factors and molecular determinants of aging-related diseases, F-59000 Lille, France
- Institut Pasteur de Lille, F-59000 Lille, France
- Univ. Lille, U1167 - Excellence Laboratory LabEx DISTALZ, F-59000 Lille, France
| | - Qiong Yang
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
| | - Traci M Bartz
- Cardiovascular Health Research Unit, Departments of Biostatistics and Medicine, University of Washington, Seattle, WA, USA
| | - Edith Hofer
- Clinical Division of Neurogeriatrics, Department of Neurology, Medical University of Graz, Austria
- Institute for Medical Informatics, Statistics and Documentation, Medical University of Graz, Austria
| | - Aïcha Soumaré
- University of Bordeaux, Inserm, Bordeaux Population Health Research Center, team VINTAGE, UMR 1219, F-33000 Bordeaux, France
| | - Fen Peng
- The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Marie-Gabrielle Duperron
- University of Bordeaux, Inserm, Bordeaux Population Health Research Center, team VINTAGE, UMR 1219, F-33000 Bordeaux, France
| | - Mario Foglio
- University of McGill Genome Center, Montreal, Canada
| | - Thomas H Mosley
- Division of Geriatrics, School of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
- Memory Impairment and Neurodegenerative Dementia Center, University of Mississippi Medical Center, Jackson, MS, USA
| | - Reinhold Schmidt
- Clinical Division of Neurogeriatrics, Department of Neurology, Medical University of Graz, Austria
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
- Department of Health Services, University of Washington, Seattle, WA, USA
- Kaiser Permanente Washington Health Research Institute, Seattle, WA, USA
| | - Lenore J Launer
- Intramural Research Program, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Eric Boerwinkle
- The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Yicheng Zhu
- Department of Neurology, Peking Union Medical College Hospital, Beijing, China
| | - Bernard Mazoyer
- University of Bordeaux, Institut des Maladies Neurodégénératives, CNRS-CEA UMR 5293, France
| | - Mark Lathrop
- University of McGill Genome Center, Montreal, Canada
| | - Celine Bellenguez
- Inserm, U1167, RID-AGE - Risk factors and molecular determinants of aging-related diseases, F-59000 Lille, France
- Institut Pasteur de Lille, F-59000 Lille, France
- Univ. Lille, U1167 - Excellence Laboratory LabEx DISTALZ, F-59000 Lille, France
| | - Cornelia M Van Duijn
- Department of Epidemiology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - M Arfan Ikram
- Department of Epidemiology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Helena Schmidt
- Gottfried Schatz Research Center, Department of Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - W T Longstreth
- Department of Neurology and Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Myriam Fornage
- The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Sudha Seshadri
- Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, University of Texas Health Sciences Center, San Antonio, Texas, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
| | - Anne Joutel
- Institute of Psychiatry and Neurosciences of Paris, Inserm, University Paris Descartes, DHU NeuroVasc, Sorbonne Paris Cité, Paris, France
| | - Christophe Tzourio
- University of Bordeaux, Inserm, Bordeaux Population Health Research Center, team VINTAGE, UMR 1219, F-33000 Bordeaux, France
- CHU de Bordeaux, Pole de santé publique, Service d’information médicale, F-33000 Bordeaux, France
| | - Stephanie Debette
- University of Bordeaux, Inserm, Bordeaux Population Health Research Center, team VINTAGE, UMR 1219, F-33000 Bordeaux, France
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
- CHU de Bordeaux, Department of Neurology, F-33000 Bordeaux, France
| |
Collapse
|
75
|
Sweeney MD, Zhao Z, Montagne A, Nelson AR, Zlokovic BV. Blood-Brain Barrier: From Physiology to Disease and Back. Physiol Rev 2019; 99:21-78. [PMID: 30280653 PMCID: PMC6335099 DOI: 10.1152/physrev.00050.2017] [Citation(s) in RCA: 1220] [Impact Index Per Article: 244.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 04/17/2018] [Accepted: 04/17/2018] [Indexed: 12/12/2022] Open
Abstract
The blood-brain barrier (BBB) prevents neurotoxic plasma components, blood cells, and pathogens from entering the brain. At the same time, the BBB regulates transport of molecules into and out of the central nervous system (CNS), which maintains tightly controlled chemical composition of the neuronal milieu that is required for proper neuronal functioning. In this review, we first examine molecular and cellular mechanisms underlying the establishment of the BBB. Then, we focus on BBB transport physiology, endothelial and pericyte transporters, and perivascular and paravascular transport. Next, we discuss rare human monogenic neurological disorders with the primary genetic defect in BBB-associated cells demonstrating the link between BBB breakdown and neurodegeneration. Then, we review the effects of genes underlying inheritance and/or increased susceptibility for Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease, and amyotrophic lateral sclerosis (ALS) on BBB in relation to other pathologies and neurological deficits. We next examine how BBB dysfunction relates to neurological deficits and other pathologies in the majority of sporadic AD, PD, and ALS cases, multiple sclerosis, other neurodegenerative disorders, and acute CNS disorders such as stroke, traumatic brain injury, spinal cord injury, and epilepsy. Lastly, we discuss BBB-based therapeutic opportunities. We conclude with lessons learned and future directions, with emphasis on technological advances to investigate the BBB functions in the living human brain, and at the molecular and cellular level, and address key unanswered questions.
Collapse
Affiliation(s)
- Melanie D Sweeney
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California , Los Angeles, California ; and Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California , Los Angeles, California
| | - Zhen Zhao
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California , Los Angeles, California ; and Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California , Los Angeles, California
| | - Axel Montagne
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California , Los Angeles, California ; and Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California , Los Angeles, California
| | - Amy R Nelson
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California , Los Angeles, California ; and Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California , Los Angeles, California
| | - Berislav V Zlokovic
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California , Los Angeles, California ; and Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California , Los Angeles, California
| |
Collapse
|
76
|
Oura Y, Nakamura M, Takigawa T, Fukushima Y, Wakabayashi T, Tsujikawa M, Nishida K. High-Temperature Requirement A 1 Causes Photoreceptor Cell Death in Zebrafish Disease Models. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:2729-2744. [PMID: 30273602 DOI: 10.1016/j.ajpath.2018.08.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 07/28/2018] [Accepted: 08/23/2018] [Indexed: 02/06/2023]
Abstract
Age-related macular degeneration (AMD) is an important cause of blindness. It is characterized by a retinal pigment epithelium (RPE) disorder that leads to death of photoreceptor cells (PRCs). AMD has a strong genetic association with high-temperature requirement A 1 (HTRA1). The relationship between HTRA1 and the AMD phenotype is unknown. In this study, we show that the expression of HTRA1 in PRCs, as well as in RPE, is increased by the disease-associated HTRA1 mutation and aging. Terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling assay and quantitative PCR of apoptosis-associated caspases confirmed that PRC-specific overexpression of HTRA1 induced PRC death. Transgenic zebrafish overexpressing human HTRA1 in rod PRCs showed morphologic changes of the RPE, including PRC death and lipofuscin accumulation, features similar to those of early AMD. htra1 expression was also increased in a retinitis pigmentosa zebrafish model compared with wild type. In both fish lines, PRC death was rescued by the suppression of htra1 by the inhibitor 6-boroV. AKT-forkhead box O3 signaling downstream of HTRA1 was activated via a tumor growth factor β signal, resulting in PRC death. These findings suggest that HTRA1 derived from PRCs is associated with early AMD via PRC death. HTRA1 is a potentially effective target for neuroprotective therapy of early AMD and other degenerative diseases of PRCs.
Collapse
Affiliation(s)
- Yoshihito Oura
- Department of Ophthalmology, Osaka University Medical School, Suita, Japan
| | - Machiko Nakamura
- Pain and Neuroscience Laboratories, Daiichi Sankyo Co, Ltd, Tokyo, Japan
| | - Tohru Takigawa
- Department of Ophthalmology, Osaka University Medical School, Suita, Japan
| | - Yoko Fukushima
- Department of Ophthalmology, Osaka University Medical School, Suita, Japan
| | - Taku Wakabayashi
- Department of Ophthalmology, Osaka University Medical School, Suita, Japan
| | - Motokazu Tsujikawa
- Department of Ophthalmology, Osaka University Medical School, Suita, Japan.
| | - Kohji Nishida
- Department of Ophthalmology, Osaka University Medical School, Suita, Japan
| |
Collapse
|
77
|
Lin MK, Yang J, Hsu CW, Gore A, Bassuk AG, Brown LM, Colligan R, Sengillo JD, Mahajan VB, Tsang SH. HTRA1, an age-related macular degeneration protease, processes extracellular matrix proteins EFEMP1 and TSP1. Aging Cell 2018; 17:e12710. [PMID: 29730901 PMCID: PMC6052470 DOI: 10.1111/acel.12710] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/07/2017] [Indexed: 01/12/2023] Open
Abstract
High-temperature requirement protein A1 (HTRA1) is a serine protease secreted by a number of tissues including retinal pigment epithelium (RPE). A promoter variant of the gene encoding HTRA1 is part of a mutant allele that causes increased HTRA1 expression and contributed to age-related macular degeneration (AMD) in genomewide association studies. AMD is characterized by pathological development of drusen, extracellular deposits of proteins and lipids on the basal side of RPE. The molecular pathogenesis of AMD is not well understood, and understanding dysregulation of the extracellular matrix may be key. We assess the high-risk genotype at 10q26 by proteomic comparison of protein levels of RPE cells with and without the mutation. We show HTRA1 protein level is increased in high-risk RPE cells along with several extracellular matrix proteins, including known HTRA1 cleavage targets LTBP-1 and clusterin. In addition, two novel targets of HTRA1 have been identified: EFEMP1, an extracellular matrix protein mutated in Doyne honeycomb retinal dystrophy, a genetic eye disease similar to AMD, and thrombospondin 1 (TSP1), an inhibitor of angiogenesis. Our data support the role of RPE extracellular deposition with potential effects in compromised barrier to neovascularization in exudative AMD.
Collapse
Affiliation(s)
- Michael K. Lin
- College of Physicians & SurgeonsColumbia UniversityNew YorkNYUSA
- Edward S. Harkness Eye InstituteNew York‐Presbyterian HospitalNew YorkNYUSA
- Jonas Children's Vision Care, and Bernard & Shirlee Brown Glaucoma LaboratoryColumbia Stem Cell InitiativeDepartments of Ophthalmology, Pathology & Cell BiologyInstitute of Human NutritionHerbert Irving Comprehensive Cancer CenterColumbia UniversityNew YorkNYUSA
| | - Jin Yang
- Edward S. Harkness Eye InstituteNew York‐Presbyterian HospitalNew YorkNYUSA
- Jonas Children's Vision Care, and Bernard & Shirlee Brown Glaucoma LaboratoryColumbia Stem Cell InitiativeDepartments of Ophthalmology, Pathology & Cell BiologyInstitute of Human NutritionHerbert Irving Comprehensive Cancer CenterColumbia UniversityNew YorkNYUSA
- Tianjin Medical University Eye HospitalTianjinChina
| | - Chun Wei Hsu
- Edward S. Harkness Eye InstituteNew York‐Presbyterian HospitalNew YorkNYUSA
- Jonas Children's Vision Care, and Bernard & Shirlee Brown Glaucoma LaboratoryColumbia Stem Cell InitiativeDepartments of Ophthalmology, Pathology & Cell BiologyInstitute of Human NutritionHerbert Irving Comprehensive Cancer CenterColumbia UniversityNew YorkNYUSA
| | - Anuradha Gore
- Omics LaboratoryDepartment of OphthalmologyByers Eye InstituteStanford UniversityPalo AltoCAUSA
| | | | - Lewis M. Brown
- Quantitative Proteomics and Metabolomics CenterDepartment of Biological SciencesColumbia UniversityNew YorkNYUSA
| | - Ryan Colligan
- Quantitative Proteomics and Metabolomics CenterDepartment of Biological SciencesColumbia UniversityNew YorkNYUSA
| | - Jesse D. Sengillo
- Edward S. Harkness Eye InstituteNew York‐Presbyterian HospitalNew YorkNYUSA
- Jonas Children's Vision Care, and Bernard & Shirlee Brown Glaucoma LaboratoryColumbia Stem Cell InitiativeDepartments of Ophthalmology, Pathology & Cell BiologyInstitute of Human NutritionHerbert Irving Comprehensive Cancer CenterColumbia UniversityNew YorkNYUSA
| | - Vinit B. Mahajan
- Omics LaboratoryDepartment of OphthalmologyByers Eye InstituteStanford UniversityPalo AltoCAUSA
- Palo Alto Veterans AdministrationPalo AltoCAUSA
| | - Stephen H. Tsang
- Edward S. Harkness Eye InstituteNew York‐Presbyterian HospitalNew YorkNYUSA
- Jonas Children's Vision Care, and Bernard & Shirlee Brown Glaucoma LaboratoryColumbia Stem Cell InitiativeDepartments of Ophthalmology, Pathology & Cell BiologyInstitute of Human NutritionHerbert Irving Comprehensive Cancer CenterColumbia UniversityNew YorkNYUSA
| |
Collapse
|
78
|
Pati AR, Battisti C, Taglia I, Galluzzi P, Bianchi M, Federico A. A new case of autosomal dominant small vessel disease carrying a novel heterozygous mutation in HTRA1 gene: 2-year follow-up. Neurol Sci 2018; 39:1479-1481. [PMID: 29546604 DOI: 10.1007/s10072-018-3294-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 02/15/2018] [Indexed: 10/17/2022]
Affiliation(s)
- A R Pati
- Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy
| | - C Battisti
- Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy.
| | - I Taglia
- Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy
| | - P Galluzzi
- Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy
| | - M Bianchi
- Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy
| | - A Federico
- Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy
| |
Collapse
|
79
|
Zellner A, Scharrer E, Arzberger T, Oka C, Domenga-Denier V, Joutel A, Lichtenthaler SF, Müller SA, Dichgans M, Haffner C. CADASIL brain vessels show a HTRA1 loss-of-function profile. Acta Neuropathol 2018; 136:111-125. [PMID: 29725820 DOI: 10.1007/s00401-018-1853-8] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 03/29/2018] [Accepted: 04/24/2018] [Indexed: 01/06/2023]
Abstract
Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) and a phenotypically similar recessive condition (CARASIL) have emerged as important genetic model diseases for studying the molecular pathomechanisms of cerebral small vessel disease (SVD). CADASIL, the most frequent and intensely explored monogenic SVD, is characterized by a severe pathology in the cerebral vasculature including the mutation-induced aggregation of the Notch3 extracellular domain (Notch3ECD) and the formation of protein deposits of insufficiently determined composition in vessel walls. To identify key molecules and pathways involved in this process, we quantitatively determined the brain vessel proteome from CADASIL patient and control autopsy samples (n = 6 for each group), obtaining 95 proteins with significantly increased abundance. Intriguingly, high-temperature requirement protein A1 (HTRA1), the extracellular protease mutated in CARASIL, was found to be strongly enriched (4.9-fold, p = 1.6 × 10-3) and to colocalize with Notch3ECD deposits in patient vessels suggesting a sequestration process. Furthermore, the presence of increased levels of several HTRA1 substrates in the CADASIL proteome was compatible with their reduced degradation as consequence of a loss of HTRA1 activity. Indeed, a comparison with the brain vessel proteome of HTRA1 knockout mice (n = 5) revealed a highly significant overlap of 18 enriched proteins (p = 2.2 × 10-16), primarily representing secreted and extracellular matrix factors. Several of them were shown to be processed by HTRA1 in an in vitro proteolysis assay identifying them as novel substrates. Our study provides evidence for a loss of HTRA1 function as a critical step in the development of CADASIL pathology linking the molecular mechanisms of two distinct SVD forms.
Collapse
Affiliation(s)
- Andreas Zellner
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-Universität München, Feodor-Lynen-Straße 17, 81377, Munich, Germany
| | - Eva Scharrer
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-Universität München, Feodor-Lynen-Straße 17, 81377, Munich, Germany
| | - Thomas Arzberger
- Center for Neuropathology and Prion Research, Ludwig-Maximilians-Universität München, Munich, Germany
- Department of Psychiatry and Psychotherapy, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Chio Oka
- Laboratory of Gene Function in Animals, Nara Institute of Science and Technology, Takayama, Ikoma, Nara, Japan
| | - Valérie Domenga-Denier
- Department of Genetics and Pathogenesis of Cerebrovascular Diseases, INSERM, UMRS 1161, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
- DHU NeuroVasc, Sorbonne Paris Cité, Paris, France
| | - Anne Joutel
- Department of Genetics and Pathogenesis of Cerebrovascular Diseases, INSERM, UMRS 1161, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
- DHU NeuroVasc, Sorbonne Paris Cité, Paris, France
| | - Stefan F Lichtenthaler
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
- Institute for Advanced Study, Technische Universität München, Garching, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Stephan A Müller
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Martin Dichgans
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-Universität München, Feodor-Lynen-Straße 17, 81377, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Christof Haffner
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-Universität München, Feodor-Lynen-Straße 17, 81377, Munich, Germany.
| |
Collapse
|
80
|
Lee YC, Chung CP, Chao NC, Fuh JL, Chang FC, Soong BW, Liao YC. Characterization of Heterozygous HTRA1 Mutations in Taiwanese Patients With Cerebral Small Vessel Disease. Stroke 2018; 49:1593-1601. [PMID: 29895533 DOI: 10.1161/strokeaha.118.021283] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 05/08/2018] [Accepted: 05/14/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND PURPOSE Homozygous and compound heterozygous mutations in the high temperature requirement serine peptidase A1 gene (HTRA1) cause cerebral autosomal recessive arteriopathy with subcortical infarcts and leukoencephalopathy. However, heterozygous HTRA1 mutations were recently identified to be associated with autosomal dominant cerebral small vessel disease (SVD). The present study aims at investigating the clinical features, frequency, and spectrum of HTRA1 mutations in a Taiwanese cohort with SVD. METHODS Mutational analyses of HTRA1 were performed by Sanger sequencing in 222 subjects, selected from a cohort of 337 unrelated patients with SVD after excluding those harboring a NOTCH3 mutation. The influence of these mutations on HTRA1 protease activities was characterized. RESULTS Seven novel heterozygous mutations in HTRA1 were identified, including p.Gly120Asp, p.Ile179Asn, p.Ala182Profs*33, p.Ile256Thr, p.Gly276Ala, p.Gln289Ter, and p.Asn324Thr, and each was identified in 1 single index patient. All mutations significantly compromise the HTRA1 protease activities. For the 7 index cases and another 2 affected siblings carrying a heterozygous HTRA1 mutation, the common clinical presentations include lacunar infarction, intracerebral hemorrhage, cognitive decline, and spondylosis at the fifth to sixth decade of life. Among the 9 patients, 4 have psychiatric symptoms as delusion, depression, and compulsive behavior, 3 have leukoencephalopathy in anterior temporal poles, and 2 patients have alopecia. CONCLUSIONS Heterozygous HTRA1 mutations account for 2.08% (7 of 337) of SVD in Taiwan. The clinical and neuroradiological features of HTRA1-related SVD and sporadic SVD are similar. These findings broaden the mutational spectrum of HTRA1 and highlight the pathogenic role of heterozygous HTRA1 mutations in SVD.
Collapse
Affiliation(s)
- Yi-Chung Lee
- From the Departments of Neurology (Y.-C.L., C.-P.C., N.-C.C., J.-L.F., B.-W.S., Y.-C.L.)
- Taipei Veterans General Hospital, Taiwan; and Department of Neurology (Y.-C.L., C.-P.C., J.-L.F., B.-W.S., Y.-C.L.)
- Brain Research Center (Y.-C.L., J.-L.F., B.-W.S.), National Yang-Ming University School of Medicine, Taipei, Taiwan
| | - Chih-Ping Chung
- From the Departments of Neurology (Y.-C.L., C.-P.C., N.-C.C., J.-L.F., B.-W.S., Y.-C.L.)
- Taipei Veterans General Hospital, Taiwan; and Department of Neurology (Y.-C.L., C.-P.C., J.-L.F., B.-W.S., Y.-C.L.)
| | - Nai-Chen Chao
- From the Departments of Neurology (Y.-C.L., C.-P.C., N.-C.C., J.-L.F., B.-W.S., Y.-C.L.)
| | - Jong-Ling Fuh
- From the Departments of Neurology (Y.-C.L., C.-P.C., N.-C.C., J.-L.F., B.-W.S., Y.-C.L.)
- Taipei Veterans General Hospital, Taiwan; and Department of Neurology (Y.-C.L., C.-P.C., J.-L.F., B.-W.S., Y.-C.L.)
- Brain Research Center (Y.-C.L., J.-L.F., B.-W.S.), National Yang-Ming University School of Medicine, Taipei, Taiwan
| | | | - Bing-Wing Soong
- From the Departments of Neurology (Y.-C.L., C.-P.C., N.-C.C., J.-L.F., B.-W.S., Y.-C.L.)
- Taipei Veterans General Hospital, Taiwan; and Department of Neurology (Y.-C.L., C.-P.C., J.-L.F., B.-W.S., Y.-C.L.)
- Brain Research Center (Y.-C.L., J.-L.F., B.-W.S.), National Yang-Ming University School of Medicine, Taipei, Taiwan
| | - Yi-Chu Liao
- From the Departments of Neurology (Y.-C.L., C.-P.C., N.-C.C., J.-L.F., B.-W.S., Y.-C.L.)
- Taipei Veterans General Hospital, Taiwan; and Department of Neurology (Y.-C.L., C.-P.C., J.-L.F., B.-W.S., Y.-C.L.)
| |
Collapse
|
81
|
Tang X, Muhammad H, McLean C, Miotla-Zarebska J, Fleming J, Didangelos A, Önnerfjord P, Leask A, Saklatvala J, Vincent TL. Connective tissue growth factor contributes to joint homeostasis and osteoarthritis severity by controlling the matrix sequestration and activation of latent TGFβ. Ann Rheum Dis 2018; 77:1372-1380. [PMID: 29925506 PMCID: PMC6104679 DOI: 10.1136/annrheumdis-2018-212964] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Revised: 05/14/2018] [Accepted: 05/26/2018] [Indexed: 12/22/2022]
Abstract
OBJECTIVES One mechanism by which cartilage responds to mechanical load is by releasing heparin-bound growth factors from the pericellular matrix (PCM). By proteomic analysis of the PCM, we identified connective tissue growth factor (CTGF) and here investigate its function and mechanism of action. METHODS Recombinant CTGF (rCTGF) was used to stimulate human chondrocytes for microarray analysis. Endogenous CTGF was investigated by in vitro binding assays and confocal microscopy. Its release from cut cartilage (injury CM) was analysed by Western blot under reducing and non-reducing conditions. A postnatal, conditional CtgfcKO mouse was generated for cartilage injury experiments and to explore the course of osteoarthritis (OA) by destabilisation of the medial meniscus. siRNA knockdown was performed on isolated human chondrocytes. RESULTS The biological responses of rCTGF were TGFβ dependent. CTGF displaced latent TGFβ from cartilage and both were released on cartilage injury. CTGF and latent TGFβ migrated as a single high molecular weight band under non-reducing conditions, suggesting that they were in a covalent (disulfide) complex. This was confirmed by immunoprecipitation. Using CtgfcKO mice, CTGF was required for sequestration of latent TGFβ in the matrix and activation of the latent complex at the cell surface through TGFβR3. In vivo deletion of CTGF increased the thickness of the articular cartilage and protected mice from OA. CONCLUSIONS CTGF is a latent TGFβ binding protein that controls the matrix sequestration and activation of TGFβ in cartilage. Deletion of CTGF in vivo caused a paradoxical increase in Smad2 phosphorylation resulting in thicker cartilage that was protected from OA.
Collapse
Affiliation(s)
- Xiaodi Tang
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Hayat Muhammad
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Celia McLean
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | | | - Jacob Fleming
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | | | | | - Andrew Leask
- Department of Dentistry, University of Western Ontario, London, Ontario, Canada
| | | | - Tonia L Vincent
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| |
Collapse
|
82
|
Loss of HtrA1 serine protease induces synthetic modulation of aortic vascular smooth muscle cells. PLoS One 2018; 13:e0196628. [PMID: 29768431 PMCID: PMC5955505 DOI: 10.1371/journal.pone.0196628] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 04/15/2018] [Indexed: 01/01/2023] Open
Abstract
Homozygous mutations of human HTRA1 cause cerebral autosomal recessive arteriopathy with subcortical infarcts and leukoencephalopathy (CARASIL). HtrA1-/- mice were examined for arterial abnormalities. Although their cerebral arteries were normal, the thoracic aorta was affected in HtrA1-/- mice. The number of vascular smooth muscle cells (VSMCs) in the aorta was increased in HtrA1-/- mice of 40 weeks or younger, but decreased thereafter. The cross-sectional area of the aorta was increased in HtrA1-/- mice of 40 weeks or older. Aortic VSMCs isolated from HtrA1-/- mice rapidly proliferated and migrated, produced high MMP9 activity, and were prone to oxidative stress-induced cell death. HtrA1-/- VSMCs expressed less smooth muscle α-actin, and more vimentin and osteopontin, and responded to PDGF-BB more strongly than wild type VSMCs, indicating that HtrA1-/- VSMCs were in the synthetic phenotype. The elastic lamina was disrupted, and collagens were decreased in the aortic media. Calponin in the media was decreased, whereas vimentin and osteopontin were increased, suggesting a synthetic shift of VSMCs in vivo. Loss of HtrA1 therefore skews VSMCs toward the synthetic phenotype, induces MMP9 expression, and expedites cell death. We propose that the synthetic modulation is the primary event that leads to the vascular abnormalities caused by HtrA1 deficiency.
Collapse
|
83
|
Thaler FS, Catak C, Einhäupl M, Müller S, Seelos K, Wollenweber FA, Kümpfel T. Cerebral small vessel disease caused by a novel heterozygous mutation in HTRA1. J Neurol Sci 2018; 388:19-21. [PMID: 29627020 DOI: 10.1016/j.jns.2018.02.043] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 02/25/2018] [Accepted: 02/26/2018] [Indexed: 10/17/2022]
Affiliation(s)
- Franziska S Thaler
- Institute of Clinical Neuroimmunology, Ludwig Maximilians University, Munich, Germany.
| | - Cihan Catak
- Institute for Stroke and Dementia Research, Ludwig Maximilians University, Munich, Germany
| | | | - Susanna Müller
- Institute of Pathology, Ludwig Maximilians University, Munich, Germany
| | - Klaus Seelos
- Department of Neuroradiology, Ludwig Maximilians University, Munich, Germany
| | - Frank A Wollenweber
- Institute for Stroke and Dementia Research, Ludwig Maximilians University, Munich, Germany
| | - Tania Kümpfel
- Institute of Clinical Neuroimmunology, Ludwig Maximilians University, Munich, Germany
| |
Collapse
|
84
|
Klose R, Adam MG, Weis EM, Moll I, Wüstehube-Lausch J, Tetzlaff F, Oka C, Ehrmann M, Fischer A. Inactivation of the serine protease HTRA1 inhibits tumor growth by deregulating angiogenesis. Oncogene 2018; 37:4260-4272. [PMID: 29713059 DOI: 10.1038/s41388-018-0258-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 02/26/2018] [Accepted: 03/27/2018] [Indexed: 11/09/2022]
Abstract
The serine protease HTRA1 is involved in several vascular diseases and its expression is often deregulated in cancer. We aimed at identifying how HTRA1 in the vasculature affects tumor growth. Here we report that silencing of HTRA1 in cultured endothelial cells increased migration rate and tube formation, whereas forced HTRA1 expression impaired sprouting angiogenesis. Mechanistically, endothelial HTRA1 expression enhanced Delta/Notch signaling by reducing the amount of the weak Notch ligand JAG1. HTRA1 physically interacted with JAG1 and cleaved it within the intracellular domain, leading to protein degradation. Expression of a constitutive active Notch1 prevented the hypersprouting phenotype upon silencing of HTRA1. In HtrA1-deficient mice, endothelial Notch signaling was diminished and isolated endothelial cells had increased expression of VEGF receptor-2. Growth of syngeneic tumors was strongly impaired in HtrA1-/- mice. The tumor vasculature was much denser in HtrA1-/- mice and less covered with mural cells. This chaotic and immature vascular network was poorly functional as indicated by large hypoxic tumor areas and low tumor cell proliferation rates. In summary, inhibition of HTRA1 in the tumor stroma impaired tumor progression by deregulating angiogenesis.
Collapse
Affiliation(s)
- Ralph Klose
- Division Vascular Signaling and Cancer (A270), German Cancer Research Center, Heidelberg, 69120, Germany
| | - M Gordian Adam
- Division Vascular Signaling and Cancer (A270), German Cancer Research Center, Heidelberg, 69120, Germany.,Metanomics Health GmbH, Berlin, Germany
| | - Eva-Maria Weis
- Division Vascular Signaling and Cancer (A270), German Cancer Research Center, Heidelberg, 69120, Germany
| | - Iris Moll
- Division Vascular Signaling and Cancer (A270), German Cancer Research Center, Heidelberg, 69120, Germany
| | - Joycelyn Wüstehube-Lausch
- Division Vascular Signaling and Cancer (A270), German Cancer Research Center, Heidelberg, 69120, Germany.,European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68167, Germany.,BioNTech AG, Mainz, Germany
| | - Fabian Tetzlaff
- Division Vascular Signaling and Cancer (A270), German Cancer Research Center, Heidelberg, 69120, Germany
| | - Chio Oka
- Laboratory of Gene Function in Animals, Nara Institute of Science and Technology, 8916-5 Takayama, Ikoma, Nara, 630-0192, Japan
| | - Michael Ehrmann
- Centre of Medical Biotechnology, Faculty of Biology, University Duisburg-Essen, Essen, 45117, Germany
| | - Andreas Fischer
- Division Vascular Signaling and Cancer (A270), German Cancer Research Center, Heidelberg, 69120, Germany. .,European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68167, Germany. .,Medical Clinic I, Endocrinology and Clinical Chemistry, Heidelberg University Hospital, Heidelberg, 69120, Germany.
| |
Collapse
|
85
|
Horsburgh K, Wardlaw JM, van Agtmael T, Allan SM, Ashford MLJ, Bath PM, Brown R, Berwick J, Cader MZ, Carare RO, Davis JB, Duncombe J, Farr TD, Fowler JH, Goense J, Granata A, Hall CN, Hainsworth AH, Harvey A, Hawkes CA, Joutel A, Kalaria RN, Kehoe PG, Lawrence CB, Lockhart A, Love S, Macleod MR, Macrae IM, Markus HS, McCabe C, McColl BW, Meakin PJ, Miller A, Nedergaard M, O'Sullivan M, Quinn TJ, Rajani R, Saksida LM, Smith C, Smith KJ, Touyz RM, Trueman RC, Wang T, Williams A, Williams SCR, Work LM. Small vessels, dementia and chronic diseases - molecular mechanisms and pathophysiology. Clin Sci (Lond) 2018; 132:851-868. [PMID: 29712883 PMCID: PMC6700732 DOI: 10.1042/cs20171620] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 02/08/2018] [Accepted: 02/21/2018] [Indexed: 12/14/2022]
Abstract
Cerebral small vessel disease (SVD) is a major contributor to stroke, cognitive impairment and dementia with limited therapeutic interventions. There is a critical need to provide mechanistic insight and improve translation between pre-clinical research and the clinic. A 2-day workshop was held which brought together experts from several disciplines in cerebrovascular disease, dementia and cardiovascular biology, to highlight current advances in these fields, explore synergies and scope for development. These proceedings provide a summary of key talks at the workshop with a particular focus on animal models of cerebral vascular disease and dementia, mechanisms and approaches to improve translation. The outcomes of discussion groups on related themes to identify the gaps in knowledge and requirements to advance knowledge are summarized.
Collapse
Affiliation(s)
- Karen Horsburgh
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, U.K.
| | - Joanna M Wardlaw
- Centre for Clinical Brain Sciences, UK Dementia Research Institute, University of Edinburgh, Edinburgh, U.K
| | - Tom van Agtmael
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, U.K
| | - Stuart M Allan
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, U.K
| | | | - Philip M Bath
- Stroke Trials Unit, Division of Clinical Neuroscience, University of Nottingham, Nottingham, U.K
| | - Rosalind Brown
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, U.K
| | - Jason Berwick
- Department of Psychology, University of Sheffield, Sheffield, U.K
| | - M Zameel Cader
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Roxana O Carare
- Faculty of Medicine, University of Southampton, Southampton, U.K
| | - John B Davis
- Alzheimer's Research UK Oxford Drug Discovery Institute, University of Oxford, Oxford, U.K
| | - Jessica Duncombe
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, U.K
| | - Tracy D Farr
- School of Life Sciences, Nottingham University, Nottingham, U.K
| | - Jill H Fowler
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, U.K
| | - Jozien Goense
- Institute of Neuroscience and Psychology, University of Glasgow, Glasgow, U.K
| | - Alessandra Granata
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, U.K
| | | | - Atticus H Hainsworth
- Molecular and Clinical Sciences Research Institute, St Georges University of London, London, U.K
| | - Adam Harvey
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, U.K
| | - Cheryl A Hawkes
- Faculty of Science, Technology, Engineering & Mathematics, Open University, Milton Keynes, U.K
| | - Anne Joutel
- Genetics and Pathogenesis of Cerebrovascular Diseases, INSERM, Université Paris Diderot-Paris 7, Paris, France
| | - Rajesh N Kalaria
- Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, U.K
| | | | - Catherine B Lawrence
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, U.K
| | | | - Seth Love
- Clinical Neurosciences, University of Bristol, Bristol, U.K
| | - Malcolm R Macleod
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, U.K
| | - I Mhairi Macrae
- Institute of Neuroscience and Psychology, University of Glasgow, Glasgow, U.K
| | - Hugh S Markus
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, U.K
| | - Chris McCabe
- Institute of Neuroscience and Psychology, University of Glasgow, Glasgow, U.K
| | - Barry W McColl
- The Roslin Institute & R(D)SVS, UK Dementia Research Institute, University of Edinburgh, Edinburgh, U.K
| | - Paul J Meakin
- Division of Molecular & Clinical Medicine, School of Medicine, University of Dundee, Dundee, U.K
| | - Alyson Miller
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, U.K
| | - Maiken Nedergaard
- University of Rochester Medical Center, Rochester, NY, USA and University of Copenhagen's Center of Basic and Translational Neuroscience, Copenhagen, Denmark
| | - Michael O'Sullivan
- Mater Centre for Neuroscience and Queensland Brain Institute, Brisbane, Australia
| | - Terry J Quinn
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, U.K
| | - Rikesh Rajani
- Genetics and Pathogenesis of Cerebrovascular Diseases, INSERM, Université Paris Diderot-Paris 7, Paris, France
| | - Lisa M Saksida
- Robarts Research Institute, Western University, London, Ontario, Canada
| | - Colin Smith
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, U.K
| | - Kenneth J Smith
- Department of Neuroinflammation, UCL Institute of Neurology, London, U.K
| | - Rhian M Touyz
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, U.K
| | | | - Tao Wang
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, U.K
| | - Anna Williams
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, U.K
| | | | - Lorraine M Work
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, U.K
| |
Collapse
|
86
|
Yamawaki S, Naitoh M, Kubota H, Aya R, Katayama Y, Ishiko T, Tamura T, Yoshikawa K, Enoshiri T, Ikeda M, Suzuki S. HtrA1 Is Specifically Up-Regulated in Active Keloid Lesions and Stimulates Keloid Development. Int J Mol Sci 2018; 19:E1275. [PMID: 29695130 PMCID: PMC5983720 DOI: 10.3390/ijms19051275] [Citation(s) in RCA: 6] [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: 03/03/2018] [Revised: 04/04/2018] [Accepted: 04/16/2018] [Indexed: 11/16/2022] Open
Abstract
Keloids occur after failure of the wound healing process; inflammation persists, and various treatments are ineffective. Keloid pathogenesis is still unclear. We have previously analysed the gene expression profiles in keloid tissue and found that HtrA1 was markedly up-regulated in the keloid lesions. HtrA1 is a serine protease suggested to play a role in the pathogenesis of various diseases, including age-related macular degeneration and osteoarthritis, by modulating extracellular matrix or cell surface proteins. We analysed HtrA1 localization and its role in keloid pathogenesis. Thirty keloid patients and twelve unrelated patients were enrolled for in situ hybridization, immunohistochemical, western blot, and cell proliferation analyses. Fibroblast-like cells expressed more HtrA1 in active keloid lesions than in surrounding lesions. The proportion of HtrA1-positive cells in keloids was significantly higher than that in normal skin, and HtrA1 protein was up-regulated relative to normal skin. Silencing HtrA1 gene expression significantly suppressed cell proliferation. HtrA1 was highly expressed in keloid tissues, and the suppression of the HtrA1 gene inhibited the proliferation of keloid-derived fibroblasts. HtrA1 may promote keloid development by accelerating cell proliferation and remodelling keloid-specific extracellular matrix or cell surface molecules. HtrA1 is suggested to have an important role in keloid pathogenesis.
Collapse
Affiliation(s)
- Satoko Yamawaki
- Department of Plastic and Reconstructive Surgery, Japanese Red Cross Fukui Hospital, 2-4-1, Tsukimi, Fukui-City, Fukui 918-8501, Japan.
| | - Motoko Naitoh
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan.
| | - Hiroshi Kubota
- Department of Life Science, Faculty of Engineering Science, Akita University, 1-1 Tegata Gakuenmachi, Akita 010-8502, Japan.
| | - Rino Aya
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan.
| | - Yasuhiro Katayama
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan.
| | - Toshihiro Ishiko
- Department of Plastic and Reconstructive Surgery, Japanese Red Cross Otsu Hospital, 1-1-35, Nagara, Otsu City, Shiga 520-8511, Japan.
| | - Taku Tamura
- Department of Life Science, Faculty of Engineering Science, Akita University, 1-1 Tegata Gakuenmachi, Akita 010-8502, Japan.
| | - Katsuhiro Yoshikawa
- Department of Plastic and Reconstructive Surgery, Shiga Medical Center for Adults, 5-4-30, Moriyama, Moriyama City, Shiga 524-8524, Japan.
| | - Tatsuki Enoshiri
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan.
| | - Mika Ikeda
- Department of Plastic and Reconstructive Surgery, Kobe City Medical Center General Hospital, 2-1-1, Minatojima minami-machi, Cyuou-ku, Kobe City, Hyogo 650-0047, Japan.
| | - Shigehiko Suzuki
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan.
| |
Collapse
|
87
|
HtrA3 is a cellular partner of cytoskeleton proteins and TCP1α chaperonin. J Proteomics 2018; 177:88-111. [PMID: 29477555 DOI: 10.1016/j.jprot.2018.02.022] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 02/13/2018] [Accepted: 02/19/2018] [Indexed: 01/09/2023]
Abstract
The human HtrA3 protease is involved in placentation, mitochondrial homeostasis, stimulation of apoptosis and proposed to be a tumor suppressor. Molecular mechanisms of the HtrA3 functions are poorly understood and knowledge concerning its cellular targets is very limited. There are two HtrA3 isoforms, the long (HtrA3L) and short (HtrA3S). Upon stress, their N-terminal domains are removed, resulting in the more active ΔN-HtrA3. By pull down and mass spectrometry techniques, we identified a panel of putative ΔN-HtrA3L/S substrates. We confirmed that ΔN-HtrA3L/S formed complexes with actin, β-tubulin, vimentin and TCP1α in vitro and in a cell and partially co-localized with the actin and vimentin filaments, microtubules and TCP1α in a cell. In vitro, both isoforms cleaved the cytoskeleton proteins, promoted tubulin polymerization and displayed chaperone-like activity, with ΔN-HtrA3S being more efficient in proteolysis and ΔN-HtrA3L - in polymerization. TCP1α, essential for the actin and tubulin folding, was directly bound by the ΔN-HtrA3L/S but not cleaved. These results indicate that actin, β-tubulin, vimentin, and TCP1α are HtrA3 cellular partners and suggest that HtrA3 may influence cytoskeleton dynamics. They also suggest different roles of the HtrA3 isoforms and a possibility that HtrA3 protease may also function as a co-chaperone. SIGNIFICANCE The HtrA3 protease stimulates apoptosis and is proposed to be a tumor suppressor and a therapeutic target, however little is known about its function at the molecular level and very few HtrA3 physiological substrates have been identified so far. Furthermore, HtrA3 is the only member of the HtrA family of proteins which, apart from the long isoform possessing the PD and PDZ domains (HtrA3L), has a short isoform (HtrA3S) lacking the PDZ domain. In this work we identified a large panel (about 150) of the tentative HtrA3L/S cellular partners which provides a good basis for further research concerning the HtrA3 function. We have shown that the cytoskeleton proteins actin, β-tubulin and vimentin, and the TCP1α chaperonin are cellular partners of both HtrA3 isoforms. Our findings indicate that HtrA3 may promote destabilization of the actin and vimentin cytoskeleton and suggest that it may influence the dynamics of the microtubule network, with the HtrA3S being more efficient in cytoskeleton protein cleavage and HtrA3L - in tubulin polymerization. Also, we have shown for the first time that HtrA3 has a chaperone-like, holdase activity in vitro - activity typical for co-chaperone proteins. The proposed HtrA3 influence on the cytoskeleton dynamics may be one of the ways in which HtrA3 promotes cell death and affects cancerogenesis. We believe that the results of this study provide a new insight into the role of HtrA3 in a cell and further confirm the notion that HtrA3 should be considered as a target of new anti-cancer therapies.
Collapse
|
88
|
BMP-Responsive Protease HtrA1 Is Differentially Expressed in Astrocytes and Regulates Astrocytic Development and Injury Response. J Neurosci 2018; 38:3840-3857. [PMID: 29483282 DOI: 10.1523/jneurosci.2031-17.2018] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 01/23/2018] [Accepted: 01/29/2018] [Indexed: 11/21/2022] Open
Abstract
Astrocytes perform a wide array of physiological functions, including structural support, ion exchange, and neurotransmitter uptake. Despite this diversity, molecular markers that label subpopulations of astrocytes are limited, and mechanisms that generate distinct astrocyte subtypes remain unclear. Here we identified serine protease high temperature requirement A 1 (HtrA1), a bone morphogenetic protein 4 signaling regulated protein, as a novel marker of forebrain astrocytes, but not of neural stem cells, in adult mice of both sexes. Genetic deletion of HtrA1 during gliogenesis accelerates astrocyte differentiation. In addition, ablation of HtrA1 in cultured astrocytes leads to altered chondroitin sulfate proteoglycan expression and inhibition of neurite extension, along with elevated levels of transforming growth factor-β family proteins. Brain injury induces HtrA1 expression in reactive astrocytes, and loss of HtrA1 leads to an impairment in wound closure accompanied by increased proliferation of endothelial and immune cells. Our findings demonstrate that HtrA1 is differentially expressed in adult mouse forebrain astrocytes, and that HtrA1 plays important roles in astrocytic development and injury response.SIGNIFICANCE STATEMENT Astrocytes, an abundant cell type in the brain, perform a wide array of physiological functions. Although characterized as morphologically and functionally diverse, molecular markers that label astrocyte subtypes or signaling pathways that lead to their diversity remain limited. Here, after examining the expression profile of astrocytes generated in response to bone morphogenetic protein signaling, we identify high temperature requirement A 1 (HtrA1) as an astrocyte-specific marker that is differentially expressed in distinct adult mouse brain regions. HtrA1 is a serine protease that has been linked to cerebral autosomal recessive arteriopathy with subcortical infarcts and leukoencephalopathy, a small blood vessel disease in humans. Understanding the role of HtrA1 during development and after injury will provide insights into how distinct astrocyte populations are generated and their unique roles in injury and disease.
Collapse
|
89
|
Globus O, Evron T, Caspi M, Siman-Tov R, Rosin-Arbesfeld R. High-Temperature Requirement A1 (Htra1) - A Novel Regulator of Canonical Wnt Signaling. Sci Rep 2017; 7:17995. [PMID: 29269789 PMCID: PMC5740065 DOI: 10.1038/s41598-017-18203-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 12/07/2017] [Indexed: 01/02/2023] Open
Abstract
Different cancer types as well as many other diseases are caused by aberrant activation of the canonical Wnt signal transduction pathway, and it is especially implicated in the development and progression of colorectal cancer (CRC). The main effector protein of the canonical Wnt signaling cascade is β-catenin, which binds to the T- cell factor/lymphoid enhancer factor (TCF/LEF) and triggers the activation of Wnt target genes. Here, we identify the serine protease High-Temperature Requirement A1 (HTRA1) as a novel component of the canonical Wnt pathway. We show that the HTRA1 protein inhibits the Wnt/β-catenin signaling, in both paracrine and autocrine manners, and affects the expression of several Wnt target genes. Moreover, HTRA1 forms a complex with β-catenin and reduces the proliferation rates of cells. Taken together, our findings indicate that HTRA1 functions as a novel suppressor of the canonical Wnt signaling pathway.
Collapse
Affiliation(s)
- Oriane Globus
- Department of Clinical Microbiology and Immunology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Tamar Evron
- Department of Clinical Microbiology and Immunology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Michal Caspi
- Department of Clinical Microbiology and Immunology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Ronen Siman-Tov
- Department of Clinical Microbiology and Immunology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Rina Rosin-Arbesfeld
- Department of Clinical Microbiology and Immunology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, 69978, Israel.
| |
Collapse
|
90
|
Fex Svenningsen Å, Löring S, Sørensen AL, Huynh HUB, Hjæresen S, Martin N, Moeller JB, Elkjær ML, Holmskov U, Illes Z, Andersson M, Nielsen SB, Benedikz E. Macrophage migration inhibitory factor (MIF) modulates trophic signaling through interaction with serine protease HTRA1. Cell Mol Life Sci 2017; 74:4561-4572. [PMID: 28726057 PMCID: PMC5663815 DOI: 10.1007/s00018-017-2592-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Revised: 07/05/2017] [Accepted: 07/11/2017] [Indexed: 02/07/2023]
Abstract
Macrophage migration inhibitory factor (MIF), a small conserved protein, is abundant in the immune- and central nervous system (CNS). MIF has several receptors and binding partners that can modulate its action on a cellular level. It is upregulated in neurodegenerative diseases and cancer although its function is far from clear. Here, we report the finding of a new binding partner to MIF, the serine protease HTRA1. This enzyme cleaves several growth factors, extracellular matrix molecules and is implicated in some of the same diseases as MIF. We show that the function of the binding between MIF and HTRA1 is to inhibit the proteolytic activity of HTRA1, modulating the availability of molecules that can change cell growth and differentiation. MIF is therefore the first endogenous inhibitor ever found for HTRA1. It was found that both molecules were present in astrocytes and that the functional binding has the ability to modulate astrocytic activities important in development and disease of the CNS.
Collapse
Affiliation(s)
- Åsa Fex Svenningsen
- Department of Molecular Medicine-Neurobiology Research, University of Southern Denmark, J.B. Winslows Vej 21.1, 5000, Odense, Denmark.
| | - Svenja Löring
- Department of Molecular Medicine-Neurobiology Research, University of Southern Denmark, J.B. Winslows Vej 21.1, 5000, Odense, Denmark
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Center, Meibergdreef 69-71, 1105 BK, Amsterdam, The Netherlands
| | - Anna Lahn Sørensen
- Department of Molecular Medicine-Neurobiology Research, University of Southern Denmark, J.B. Winslows Vej 21.1, 5000, Odense, Denmark
| | - Ha Uyen Buu Huynh
- Department of Molecular Medicine-Neurobiology Research, University of Southern Denmark, J.B. Winslows Vej 21.1, 5000, Odense, Denmark
| | - Simone Hjæresen
- Department of Molecular Medicine-Neurobiology Research, University of Southern Denmark, J.B. Winslows Vej 21.1, 5000, Odense, Denmark
| | - Nellie Martin
- Department of Neurology, Odense University Hospital, University of Southern Denmark, Sdr. Boulevard 29, 5000, Odense C, Denmark
| | - Jesper Bonnet Moeller
- Department of Molecular Medicine-Cancer and Inflammation, University of Southern Denmark, J.B. Winslows Vej 21.1, 5000, Odense, Denmark
- Weill Cornell Medicine, Cornell University, 413 East 69th Street, New York, 10021, USA
| | - Maria Louise Elkjær
- Department of Neurology, Odense University Hospital, University of Southern Denmark, Sdr. Boulevard 29, 5000, Odense C, Denmark
| | - Uffe Holmskov
- Department of Molecular Medicine-Cancer and Inflammation, University of Southern Denmark, J.B. Winslows Vej 21.1, 5000, Odense, Denmark
| | - Zsolt Illes
- Department of Neurology, Odense University Hospital, University of Southern Denmark, Sdr. Boulevard 29, 5000, Odense C, Denmark
| | - Malin Andersson
- Department of Pharmaceutical Biosciences, Uppsala University, Box 59, 751 24, Uppsala, Sweden
| | - Solveig Beck Nielsen
- Department of Molecular Medicine-Neurobiology Research, University of Southern Denmark, J.B. Winslows Vej 21.1, 5000, Odense, Denmark
| | - Eirikur Benedikz
- Department of Molecular Medicine-Neurobiology Research, University of Southern Denmark, J.B. Winslows Vej 21.1, 5000, Odense, Denmark
- Faculty of Health, University College Zealand, Parkvej 190, 4700, Næstved, Denmark
| |
Collapse
|
91
|
Hollander MC, Latour LL, Yang D, Ishii H, Xiao Z, Min Y, Ray-Choudhury A, Munasinghe J, Merchant AS, Lin PC, Hallenbeck J, Boehm M, Yang L. Attenuation of Myeloid-Specific TGFβ Signaling Induces Inflammatory Cerebrovascular Disease and Stroke. Circ Res 2017; 121:1360-1369. [PMID: 29051340 DOI: 10.1161/circresaha.116.310349] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 10/12/2017] [Accepted: 10/18/2017] [Indexed: 12/20/2022]
Abstract
RATIONALE Cryptogenic strokes, those of unknown cause, have been estimated as high as 30% to 40% of strokes. Inflammation has been suggested as a critical etiologic factor. However, there is lack of experimental evidence. OBJECTIVE In this study, we investigated inflammation-associated stroke using a mouse model that developed spontaneous stroke because of myeloid deficiency of TGF-β (transforming growth factor-β) signaling. METHODS AND RESULTS We report that mice with deletion of Tgfbr2 in myeloid cells (Tgfbr2Myeko) developed cerebrovascular inflammation in the absence of significant pathology in other tissues, culminating in stroke and severe neurological deficits with 100% penetrance. The stroke phenotype can be transferred to syngeneic wild-type mice via Tgfbr2Myeko bone marrow transplant and can be rescued in Tgfbr2Myeko mice with wild-type bone marrow. The underlying mechanisms involved an increased type 1 inflammation and cerebral endotheliopathy, characterized by elevated NF-κB (nuclear factor-κB) activation and TNF (tumor necrosis factor) production by myeloid cells. A high-fat diet accelerated stroke incidence. Anti-TNF treatment, as well as metformin and methotrexate, which are associated with decreased stroke risk in population studies, delayed stroke occurrence. CONCLUSIONS Our studies show that TGF-β signaling in myeloid cells is required for maintenance of vascular health and provide insight into inflammation-mediated cerebrovascular disease and stroke.
Collapse
Affiliation(s)
- M Christine Hollander
- From the Laboratory of Cancer Biology and Genetics, National Cancer Institute (M.C.H., H.I., Z.X., L.Y.), Clinical Stroke Cause and Development, National Institute of Neurological Disorders and Stroke (L.L.L., J.M., J.H.), Center for Molecular Medicine, National Institute of Heart Lung and Blood (D.Y., M.B.), Neuropathology, National Institute of Neurological Disorders and Stroke (A.R.-C.), and Bioinformatics, Center for Cancer Research, National Cancer Institute (A.S.M.), National Institutes of Health, Bethesda, MD; and Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD (Y.M., P.C.L.).
| | - Lawrence L Latour
- From the Laboratory of Cancer Biology and Genetics, National Cancer Institute (M.C.H., H.I., Z.X., L.Y.), Clinical Stroke Cause and Development, National Institute of Neurological Disorders and Stroke (L.L.L., J.M., J.H.), Center for Molecular Medicine, National Institute of Heart Lung and Blood (D.Y., M.B.), Neuropathology, National Institute of Neurological Disorders and Stroke (A.R.-C.), and Bioinformatics, Center for Cancer Research, National Cancer Institute (A.S.M.), National Institutes of Health, Bethesda, MD; and Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD (Y.M., P.C.L.)
| | - Dan Yang
- From the Laboratory of Cancer Biology and Genetics, National Cancer Institute (M.C.H., H.I., Z.X., L.Y.), Clinical Stroke Cause and Development, National Institute of Neurological Disorders and Stroke (L.L.L., J.M., J.H.), Center for Molecular Medicine, National Institute of Heart Lung and Blood (D.Y., M.B.), Neuropathology, National Institute of Neurological Disorders and Stroke (A.R.-C.), and Bioinformatics, Center for Cancer Research, National Cancer Institute (A.S.M.), National Institutes of Health, Bethesda, MD; and Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD (Y.M., P.C.L.)
| | - Hiroki Ishii
- From the Laboratory of Cancer Biology and Genetics, National Cancer Institute (M.C.H., H.I., Z.X., L.Y.), Clinical Stroke Cause and Development, National Institute of Neurological Disorders and Stroke (L.L.L., J.M., J.H.), Center for Molecular Medicine, National Institute of Heart Lung and Blood (D.Y., M.B.), Neuropathology, National Institute of Neurological Disorders and Stroke (A.R.-C.), and Bioinformatics, Center for Cancer Research, National Cancer Institute (A.S.M.), National Institutes of Health, Bethesda, MD; and Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD (Y.M., P.C.L.)
| | - Zhiguang Xiao
- From the Laboratory of Cancer Biology and Genetics, National Cancer Institute (M.C.H., H.I., Z.X., L.Y.), Clinical Stroke Cause and Development, National Institute of Neurological Disorders and Stroke (L.L.L., J.M., J.H.), Center for Molecular Medicine, National Institute of Heart Lung and Blood (D.Y., M.B.), Neuropathology, National Institute of Neurological Disorders and Stroke (A.R.-C.), and Bioinformatics, Center for Cancer Research, National Cancer Institute (A.S.M.), National Institutes of Health, Bethesda, MD; and Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD (Y.M., P.C.L.)
| | - Yongfen Min
- From the Laboratory of Cancer Biology and Genetics, National Cancer Institute (M.C.H., H.I., Z.X., L.Y.), Clinical Stroke Cause and Development, National Institute of Neurological Disorders and Stroke (L.L.L., J.M., J.H.), Center for Molecular Medicine, National Institute of Heart Lung and Blood (D.Y., M.B.), Neuropathology, National Institute of Neurological Disorders and Stroke (A.R.-C.), and Bioinformatics, Center for Cancer Research, National Cancer Institute (A.S.M.), National Institutes of Health, Bethesda, MD; and Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD (Y.M., P.C.L.)
| | - Abhik Ray-Choudhury
- From the Laboratory of Cancer Biology and Genetics, National Cancer Institute (M.C.H., H.I., Z.X., L.Y.), Clinical Stroke Cause and Development, National Institute of Neurological Disorders and Stroke (L.L.L., J.M., J.H.), Center for Molecular Medicine, National Institute of Heart Lung and Blood (D.Y., M.B.), Neuropathology, National Institute of Neurological Disorders and Stroke (A.R.-C.), and Bioinformatics, Center for Cancer Research, National Cancer Institute (A.S.M.), National Institutes of Health, Bethesda, MD; and Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD (Y.M., P.C.L.)
| | - Jeeva Munasinghe
- From the Laboratory of Cancer Biology and Genetics, National Cancer Institute (M.C.H., H.I., Z.X., L.Y.), Clinical Stroke Cause and Development, National Institute of Neurological Disorders and Stroke (L.L.L., J.M., J.H.), Center for Molecular Medicine, National Institute of Heart Lung and Blood (D.Y., M.B.), Neuropathology, National Institute of Neurological Disorders and Stroke (A.R.-C.), and Bioinformatics, Center for Cancer Research, National Cancer Institute (A.S.M.), National Institutes of Health, Bethesda, MD; and Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD (Y.M., P.C.L.)
| | - Anand S Merchant
- From the Laboratory of Cancer Biology and Genetics, National Cancer Institute (M.C.H., H.I., Z.X., L.Y.), Clinical Stroke Cause and Development, National Institute of Neurological Disorders and Stroke (L.L.L., J.M., J.H.), Center for Molecular Medicine, National Institute of Heart Lung and Blood (D.Y., M.B.), Neuropathology, National Institute of Neurological Disorders and Stroke (A.R.-C.), and Bioinformatics, Center for Cancer Research, National Cancer Institute (A.S.M.), National Institutes of Health, Bethesda, MD; and Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD (Y.M., P.C.L.)
| | - P Charles Lin
- From the Laboratory of Cancer Biology and Genetics, National Cancer Institute (M.C.H., H.I., Z.X., L.Y.), Clinical Stroke Cause and Development, National Institute of Neurological Disorders and Stroke (L.L.L., J.M., J.H.), Center for Molecular Medicine, National Institute of Heart Lung and Blood (D.Y., M.B.), Neuropathology, National Institute of Neurological Disorders and Stroke (A.R.-C.), and Bioinformatics, Center for Cancer Research, National Cancer Institute (A.S.M.), National Institutes of Health, Bethesda, MD; and Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD (Y.M., P.C.L.)
| | - John Hallenbeck
- From the Laboratory of Cancer Biology and Genetics, National Cancer Institute (M.C.H., H.I., Z.X., L.Y.), Clinical Stroke Cause and Development, National Institute of Neurological Disorders and Stroke (L.L.L., J.M., J.H.), Center for Molecular Medicine, National Institute of Heart Lung and Blood (D.Y., M.B.), Neuropathology, National Institute of Neurological Disorders and Stroke (A.R.-C.), and Bioinformatics, Center for Cancer Research, National Cancer Institute (A.S.M.), National Institutes of Health, Bethesda, MD; and Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD (Y.M., P.C.L.)
| | - Manfred Boehm
- From the Laboratory of Cancer Biology and Genetics, National Cancer Institute (M.C.H., H.I., Z.X., L.Y.), Clinical Stroke Cause and Development, National Institute of Neurological Disorders and Stroke (L.L.L., J.M., J.H.), Center for Molecular Medicine, National Institute of Heart Lung and Blood (D.Y., M.B.), Neuropathology, National Institute of Neurological Disorders and Stroke (A.R.-C.), and Bioinformatics, Center for Cancer Research, National Cancer Institute (A.S.M.), National Institutes of Health, Bethesda, MD; and Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD (Y.M., P.C.L.)
| | - Li Yang
- From the Laboratory of Cancer Biology and Genetics, National Cancer Institute (M.C.H., H.I., Z.X., L.Y.), Clinical Stroke Cause and Development, National Institute of Neurological Disorders and Stroke (L.L.L., J.M., J.H.), Center for Molecular Medicine, National Institute of Heart Lung and Blood (D.Y., M.B.), Neuropathology, National Institute of Neurological Disorders and Stroke (A.R.-C.), and Bioinformatics, Center for Cancer Research, National Cancer Institute (A.S.M.), National Institutes of Health, Bethesda, MD; and Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD (Y.M., P.C.L.)
| |
Collapse
|
92
|
Ungvari Z, Valcarcel-Ares MN, Tarantini S, Yabluchanskiy A, Fülöp GA, Kiss T, Csiszar A. Connective tissue growth factor (CTGF) in age-related vascular pathologies. GeroScience 2017; 39:491-498. [PMID: 28875415 DOI: 10.1007/s11357-017-9995-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 08/23/2017] [Indexed: 12/20/2022] Open
Abstract
Connective tissue growth factor (CTGF, also known as CCN2) is a matricellular protein expressed in the vascular wall, which regulates diverse cellular functions including cell adhesion, matrix production, structural remodeling, angiogenesis, and cell proliferation and differentiation. CTGF is principally regulated at the level of transcription and is induced by mechanical stresses and a number of cytokines and growth factors, including TGFβ. In this mini-review, the role of age-related dysregulation of CTGF signaling and its role in a range of macro- and microvascular pathologies, including pathogenesis of aorta aneurysms, atherogenesis, and diabetic retinopathy, are discussed. A potential role of CTGF and TGFβ in regulation and non-cell autonomous propagation of cellular senescence is also discussed.
Collapse
Affiliation(s)
- Zoltan Ungvari
- Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, 975 N. E. 10th Street, Oklahoma City, OK, 73104, USA
- Translational Geroscience Laboratory, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Medical Physics and Informatics, Faculty of Medicine and Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Marta Noa Valcarcel-Ares
- Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, 975 N. E. 10th Street, Oklahoma City, OK, 73104, USA
- Translational Geroscience Laboratory, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Stefano Tarantini
- Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, 975 N. E. 10th Street, Oklahoma City, OK, 73104, USA
- Translational Geroscience Laboratory, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Andriy Yabluchanskiy
- Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, 975 N. E. 10th Street, Oklahoma City, OK, 73104, USA
- Translational Geroscience Laboratory, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Gábor A Fülöp
- Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, 975 N. E. 10th Street, Oklahoma City, OK, 73104, USA
- Translational Geroscience Laboratory, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Division of Clinical Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Tamas Kiss
- Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, 975 N. E. 10th Street, Oklahoma City, OK, 73104, USA
- Department of Medical Physics and Informatics, Faculty of Medicine and Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Anna Csiszar
- Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, 975 N. E. 10th Street, Oklahoma City, OK, 73104, USA.
- Translational Geroscience Laboratory, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- Department of Medical Physics and Informatics, Faculty of Medicine and Faculty of Science and Informatics, University of Szeged, Szeged, Hungary.
| |
Collapse
|
93
|
Di Donato I, Bianchi S, Gallus GN, Cerase A, Taglia I, Pescini F, Nannucci S, Battisti C, Inzitari D, Pantoni L, Zini A, Federico A, Dotti MT. Heterozygous mutations of HTRA1 gene in patients with familial cerebral small vessel disease. CNS Neurosci Ther 2017; 23:759-765. [PMID: 28782182 PMCID: PMC6492684 DOI: 10.1111/cns.12722] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 07/11/2017] [Accepted: 07/12/2017] [Indexed: 01/05/2023] Open
Abstract
AIMS Cerebral small vessel disease (SVD) is the leading cause of vascular dementia. Although the most of cases are sporadic, familial monogenic causes have been identified in a growing minority of patients. CADASIL, due to mutations of NOTCH3 gene, is the most common genetic SVD, and CARASIL, linked to HTRA1 gene mutations, is a rare but well known autosomal recessive SVD. Recently, also heterozygous HTRA1 mutations have been described in patients with familial SVD. To detect a genetic cause of familial SVD, we performed mutational analysis of HTRA1 gene in a large cohort of Italian NOTCH3-negative patients. METHODS We recruited 142 NOTCH3-negative patients and 160 healthy age-matched controls. Additional control data were obtained from five pathogenicity prediction software. RESULTS Five different HTRA1 heterozygous mutations were detected in nine patients from five unrelated families. Clinical phenotype was typical of SVD, and the onset was presenile. Brain magnetic resonance imaging (MRI) showed a subcortical leukoencephalopathy, with involvement of the external and internal capsule, corpus callosum, and multiple lacunar infarcts. Cerebral microbleeds were also seen, while anterior temporal lobes involvement was not present. CONCLUSION Our observation further supports the pathogenic role of the heterozygous HTRA1 mutations in familial SVD.
Collapse
Affiliation(s)
- Ilaria Di Donato
- Department of MedicineSurgery and NeurosciencesMedical SchoolUniversity of SienaSienaItaly
| | - Silvia Bianchi
- Department of MedicineSurgery and NeurosciencesMedical SchoolUniversity of SienaSienaItaly
| | - Gian Nicola Gallus
- Department of MedicineSurgery and NeurosciencesMedical SchoolUniversity of SienaSienaItaly
| | - Alfonso Cerase
- Unit NINT Neuroimaging and NeurointerventionDepartment of Neurological and Sensorineural SciencesAzienda Ospedaliera Universitaria SeneseSienaItaly
| | - Ilaria Taglia
- Department of MedicineSurgery and NeurosciencesMedical SchoolUniversity of SienaSienaItaly
| | - Francesca Pescini
- NEUROFARBA DepartmentNeuroscience sectionUniversity of FlorenceFlorenceItaly
| | - Serena Nannucci
- NEUROFARBA DepartmentNeuroscience sectionUniversity of FlorenceFlorenceItaly
| | - Carla Battisti
- Department of MedicineSurgery and NeurosciencesMedical SchoolUniversity of SienaSienaItaly
| | - Domenico Inzitari
- NEUROFARBA DepartmentNeuroscience sectionUniversity of FlorenceFlorenceItaly
| | - Leonardo Pantoni
- NEUROFARBA DepartmentNeuroscience sectionUniversity of FlorenceFlorenceItaly
| | - Andrea Zini
- Stroke UnitNeurology ClinicDepartment of NeuroscienceNuovo Ospedale Civile S. Agostino‐EstenseUniversity Hospital of ModenaModenaItaly
| | - Antonio Federico
- Department of MedicineSurgery and NeurosciencesMedical SchoolUniversity of SienaSienaItaly
| | - Maria Teresa Dotti
- Department of MedicineSurgery and NeurosciencesMedical SchoolUniversity of SienaSienaItaly
| |
Collapse
|
94
|
Filliat G, Mirsaidi A, Tiaden AN, Kuhn GA, Weber FE, Oka C, Richards PJ. Role of HTRA1 in bone formation and regeneration: In vitro and in vivo evaluation. PLoS One 2017; 12:e0181600. [PMID: 28732055 PMCID: PMC5521800 DOI: 10.1371/journal.pone.0181600] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 07/03/2017] [Indexed: 12/26/2022] Open
Abstract
The role of mammalian high temperature requirement protease A1 (HTRA1) in somatic stem cell differentiation and mineralized matrix formation remains controversial, having been demonstrated to impart either anti- or pro-osteogenic effects, depending on the in vitro cell model used. The aim of this study was therefore to further evaluate the role of HTRA1 in regulating the differentiation potential and lineage commitment of murine mesenchymal stem cells in vitro, and to assess its influence on bone structure and regeneration in vivo. Our results demonstrated that short hairpin RNA-mediated ablation of Htra1 in the murine mesenchymal cell line C3H10T1/2 increased the expression of several osteogenic gene markers, and significantly enhanced matrix mineralization in response to BMP-2 stimulation. These effects were concomitant with decreases in the expression of chondrogenic gene markers, and increases in adipogenic gene expression and lipid accrual. Despite the profound effects of loss-of-function of HTRA1 on this in vitro osteochondral model, these were not reproduced in vivo, where bone microarchitecture and regeneration in 16-week-old Htra1-knockout mice remained unaltered as compared to wild-type controls. By comparison, analysis of femurs from 52-week-old mice revealed that bone structure was better preserved in Htra1-knockout mice than age-matched wild-type controls. These findings therefore provide additional insights into the role played by HTRA1 in regulating mesenchymal stem cell differentiation, and offer opportunities for improving our understanding of how this multifunctional protease may act to influence bone quality.
Collapse
Affiliation(s)
- Gladys Filliat
- Bone and Stem Cell Research Group, CABMM, University of Zurich, Zurich, Switzerland
- Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
| | - Ali Mirsaidi
- Bone and Stem Cell Research Group, CABMM, University of Zurich, Zurich, Switzerland
| | - André N. Tiaden
- Bone and Stem Cell Research Group, CABMM, University of Zurich, Zurich, Switzerland
| | - Gisela A. Kuhn
- Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| | - Franz E. Weber
- Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
- Oral Biotechnology & Bioengineering, Center for Dental Medicine, University of Zurich, Zurich, Switzerland
| | - Chio Oka
- Division of Gene Function in Animals, Nara Institute of Science and Technology, Nara, Japan
| | - Peter J. Richards
- Bone and Stem Cell Research Group, CABMM, University of Zurich, Zurich, Switzerland
- Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
- * E-mail:
| |
Collapse
|
95
|
Endothelial cell-oligodendrocyte interactions in small vessel disease and aging. Clin Sci (Lond) 2017; 131:369-379. [PMID: 28202749 PMCID: PMC5310718 DOI: 10.1042/cs20160618] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 11/28/2016] [Accepted: 12/14/2016] [Indexed: 12/11/2022]
Abstract
Cerebral small vessel disease (SVD) is a prevalent, neurological disease that significantly increases the risk of stroke and dementia. The main pathological changes are vascular, in the form of lipohyalinosis and arteriosclerosis, and in the white matter (WM), in the form of WM lesions. Despite this, it is unclear to what extent the key cell types involved–the endothelial cells (ECs) of the vasculature and the oligodendrocytes of the WM–interact. Here, we describe the work that has so far been carried out suggesting an interaction between ECs and oligodendrocytes in SVD. As these interactions have been studied in more detail in other disease states and in development, we explore these systems and discuss the role these mechanisms may play in SVD.
Collapse
|
96
|
Grand Moursel L, Munting LP, van der Graaf LM, van Duinen SG, Goumans MJTH, Ueberham U, Natté R, van Buchem MA, van Roon-Mom WMC, van der Weerd L. TGFβ pathway deregulation and abnormal phospho-SMAD2/3 staining in hereditary cerebral hemorrhage with amyloidosis-Dutch type. Brain Pathol 2017; 28:495-506. [PMID: 28557134 PMCID: PMC8028662 DOI: 10.1111/bpa.12533] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 05/19/2017] [Indexed: 12/20/2022] Open
Abstract
Hereditary cerebral hemorrhage with amyloidosis‐Dutch type (HCHWA‐D) is an early onset hereditary form of cerebral amyloid angiopathy (CAA) pathology, caused by the E22Q mutation in the amyloid β (Aβ) peptide. Transforming growth factor β1 (TGFβ1) is a key player in vascular fibrosis and in the formation of angiopathic vessels in transgenic mice. Therefore, we investigated whether the TGFβ pathway is involved in HCHWA‐D pathogenesis in human postmortem brain tissue from frontal and occipital lobes. Components of the TGFβ pathway were analyzed with quantitative RT‐PCR. TGFβ1 and TGFβ Receptor 2 (TGFBR2) gene expression levels were significantly increased in HCHWA‐D in comparison to the controls, in both frontal and occipital lobes. TGFβ‐induced pro‐fibrotic target genes were also upregulated. We further assessed pathway activation by detecting phospho‐SMAD2/3 (pSMAD2/3), a direct TGFβ down‐stream signaling mediator, using immunohistochemistry. We found abnormal pSMAD2/3 granular deposits specifically on HCHWA‐D angiopathic frontal and occipital vessels. We graded pSMAD2/3 accumulation in angiopathic vessels and found a positive correlation with the CAA load independent of the brain area. We also observed pSMAD2/3 granules in a halo surrounding occipital vessels, which was specific for HCHWA‐D. The result of this study indicates an upregulation of TGFβ1 in HCHWA‐D, as was found previously in AD with CAA pathology. We discuss the possible origins and implications of the TGFβ pathway deregulation in the microvasculature in HCHWA‐D. These findings identify the TGFβ pathway as a potential biomarker of disease progression and a possible target of therapeutic intervention in HCHWA‐D.
Collapse
Affiliation(s)
- Laure Grand Moursel
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands.,Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Leon P Munting
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands.,Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Linda M van der Graaf
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands.,Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Sjoerd G van Duinen
- Department of Pathology, Leiden University Medical Center, Leiden, the Netherlands
| | - Marie-Jose T H Goumans
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, the Netherlands
| | - Uwe Ueberham
- Paul Flechsig Institute of Brain Research, University of Leipzig, Leipzig, Germany
| | - Remco Natté
- Department of Pathology, Leiden University Medical Center, Leiden, the Netherlands
| | - Mark A van Buchem
- Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | | | - Louise van der Weerd
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands.,Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
97
|
Dichgans M, Leys D. Vascular Cognitive Impairment. Circ Res 2017; 120:573-591. [PMID: 28154105 DOI: 10.1161/circresaha.116.308426] [Citation(s) in RCA: 310] [Impact Index Per Article: 44.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 07/28/2016] [Accepted: 08/29/2016] [Indexed: 01/10/2023]
Abstract
Cerebrovascular disease typically manifests with stroke, cognitive impairment, or both. Vascular cognitive impairment refers to all forms of cognitive disorder associated with cerebrovascular disease, regardless of the specific mechanisms involved. It encompasses the full range of cognitive deficits from mild cognitive impairment to dementia. In principle, any of the multiple causes of clinical stroke can cause vascular cognitive impairment. Recent work further highlights a role of microinfarcts, microhemorrhages, strategic white matter tracts, loss of microstructural tissue integrity, and secondary neurodegeneration. Vascular brain injury results in loss of structural and functional connectivity and, hence, compromise of functional networks within the brain. Vascular cognitive impairment is common both after stroke and in stroke-free individuals presenting to dementia clinics, and vascular pathology frequently coexists with neurodegenerative pathology, resulting in mixed forms of mild cognitive impairment or dementia. Vascular dementia is now recognized as the second most common form of dementia after Alzheimer's disease, and there is increasing awareness that targeting vascular risk may help to prevent dementia, even of the Alzheimer type. Recent advances in neuroimaging, neuropathology, epidemiology, and genetics have led to a deeper understanding of how vascular disease affects cognition. These new findings provide an opportunity for the present reappraisal of vascular cognitive impairment. We further briefly address current therapeutic concepts.
Collapse
Affiliation(s)
- Martin Dichgans
- From the Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-Universität LMU, Munich, Germany (M.D.); German Center for Neurodegenerative Diseases (DZNE), Munich, Germany (M.D.); Munich Cluster for Systems Neurology (SyNergy), Germany (M.D.); and University of Lille, INSERM, CHU Lille, U1171-Degenerative & Vascular Cognitive Disorders, F-59000 Lille, France (D.L.).
| | - Didier Leys
- From the Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-Universität LMU, Munich, Germany (M.D.); German Center for Neurodegenerative Diseases (DZNE), Munich, Germany (M.D.); Munich Cluster for Systems Neurology (SyNergy), Germany (M.D.); and University of Lille, INSERM, CHU Lille, U1171-Degenerative & Vascular Cognitive Disorders, F-59000 Lille, France (D.L.)
| |
Collapse
|
98
|
Zurawa-Janicka D, Wenta T, Jarzab M, Skorko-Glonek J, Glaza P, Gieldon A, Ciarkowski J, Lipinska B. Structural insights into the activation mechanisms of human HtrA serine proteases. Arch Biochem Biophys 2017; 621:6-23. [PMID: 28396256 DOI: 10.1016/j.abb.2017.04.004] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 04/05/2017] [Accepted: 04/06/2017] [Indexed: 12/21/2022]
Abstract
Human HtrA1-4 proteins belong to the HtrA family of evolutionarily conserved serine proteases and function as important modulators of many physiological processes, including maintenance of mitochondrial homeostasis, cell signaling and apoptosis. Disturbances in their action are linked to severe diseases, including oncogenesis and neurodegeneration. The HtrA1-4 proteins share structural and functional features of other members of the HtrA protein family, however there are several significant differences in structural architecture and mechanisms of action which makes each of them unique. Our goal is to present recent studies regarding human HtrAs. We focus on their physiological functions, structure and regulation, and describe current models of activation mechanisms. Knowledge of molecular basis of the human HtrAs' action is a subject of great interest; it is crucial for understanding their relevance in cellular physiology and pathogenesis as well as for using them as targets in future therapies of diseases such as neurodegenerative disorders and cancer.
Collapse
Affiliation(s)
- Dorota Zurawa-Janicka
- Department of General and Medical Biochemistry, Faculty of Biology, University of Gdansk, Wita Stwosza 59, 80-308 Gdansk, Poland.
| | - Tomasz Wenta
- Department of General and Medical Biochemistry, Faculty of Biology, University of Gdansk, Wita Stwosza 59, 80-308 Gdansk, Poland
| | - Miroslaw Jarzab
- Department of General and Medical Biochemistry, Faculty of Biology, University of Gdansk, Wita Stwosza 59, 80-308 Gdansk, Poland
| | - Joanna Skorko-Glonek
- Department of General and Medical Biochemistry, Faculty of Biology, University of Gdansk, Wita Stwosza 59, 80-308 Gdansk, Poland
| | - Przemyslaw Glaza
- Department of General and Medical Biochemistry, Faculty of Biology, University of Gdansk, Wita Stwosza 59, 80-308 Gdansk, Poland
| | - Artur Gieldon
- Department of Theoretical Chemistry, Faculty of Chemistry, University of Gdansk, Wita Stwosza 63, 80-308 Gdansk, Poland
| | - Jerzy Ciarkowski
- Department of Theoretical Chemistry, Faculty of Chemistry, University of Gdansk, Wita Stwosza 63, 80-308 Gdansk, Poland
| | - Barbara Lipinska
- Department of General and Medical Biochemistry, Faculty of Biology, University of Gdansk, Wita Stwosza 59, 80-308 Gdansk, Poland
| |
Collapse
|
99
|
New insights into mechanisms of small vessel disease stroke from genetics. Clin Sci (Lond) 2017; 131:515-531. [DOI: 10.1042/cs20160825] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 01/03/2017] [Accepted: 01/06/2017] [Indexed: 02/02/2023]
Abstract
Cerebral small vessel disease (SVD) is a common cause of lacunar strokes, vascular cognitive impairment (VCI) and vascular dementia. SVD is thought to result in reduced cerebral blood flow, impaired cerebral autoregulation and increased blood–brain barrier (BBB) permeability. However, the molecular mechanisms underlying SVD are incompletely understood. Recent studies in monogenic forms of SVD, such as cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL), and ‘sporadic’ SVD have shed light on possible disease mechanisms in SVD. Proteomic and biochemical studies in post-mortem monogenic SVD patients, as well as in animal models of monogenic disease have suggested that disease pathways are shared between different types of monogenic disease, often involving the impairment of extracellular matrix (ECM) function. In addition, genetic studies in ‘sporadic’ SVD have also shown that the disease is highly heritable, particularly among young-onset stroke patients, and that common variants in monogenic disease genes may contribute to disease processes in some SVD subtypes. Genetic studies in sporadic lacunar stroke patients have also suggested distinct genetic mechanisms between subtypes of SVD. Genome-wide association studies (GWAS) have also shed light on other potential disease mechanisms that may be shared with other diseases involving the white matter, or with pathways implicated in monogenic disease. This review brings together recent data from studies in monogenic SVD and genetic studies in ‘sporadic’ SVD. It aims to show how these provide new insights into the pathogenesis of SVD, and highlights the possible convergence of disease mechanisms in monogenic and sporadic SVD.
Collapse
|
100
|
Søndergaard CB, Nielsen JE, Hansen CK, Christensen H. Hereditary cerebral small vessel disease and stroke. Clin Neurol Neurosurg 2017; 155:45-57. [PMID: 28254515 DOI: 10.1016/j.clineuro.2017.02.015] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Revised: 01/31/2017] [Accepted: 02/20/2017] [Indexed: 12/31/2022]
Abstract
Cerebral small vessel disease is considered hereditary in about 5% of patients and is characterized by lacunar infarcts and white matter hyperintensities on MRI. Several monogenic hereditary diseases causing cerebral small vessel disease and stroke have been identified. The purpose of this systematic review is to provide a guide for determining when to consider molecular genetic testing in patients presenting with small vessel disease and stroke. CADASIL, CARASIL, collagen type IV mutations (including PADMAL), retinal vasculopathy with cerebral leukodystrophy, Fabry disease, hereditary cerebral hemorrhage with amyloidosis, and forkhead box C1 mutations are described in terms of genetics, pathology, clinical manifestation, imaging, and diagnosis. These monogenic disorders are often characterized by early-age stroke, but also by migraine, mood disturbances, vascular dementia and often gait disturbances. Some also present with extra-cerebral manifestations such as microangiopathy of the eyes and kidneys. Many present with clinically recognizable syndromes. Investigations include a thorough family medical history, medical history, neurological examination, neuroimaging, often supplemented by specific examinations e.g of the of vision, retinal changes, as well as kidney and heart function. However molecular genetic analysis is the final gold standard of diagnosis. There are increasing numbers of reports on new monogenic syndromes causing cerebral small vessel disease. Genetic counseling is important. Enzyme replacement therapy is possible in Fabry disease, but treatment options remain overall very limited.
Collapse
Affiliation(s)
| | - Jørgen Erik Nielsen
- Department of Cellular and Molecular Medicine, Section of Neurogenetics, The Panum Institute, University of Copenhagen, Copenhagen, Denmark
| | | | - Hanne Christensen
- Department of Neurology, Copenhagen University Hospital, Bispebjerg, Denmark
| |
Collapse
|