1
|
Taïb S, Durand J, Dehais V, Boulay AC, Martin S, Blugeon C, Jourdren L, Freydier R, Cohen-Salmon M, Hazan J, Brunet I. Vascular dysfunction is at the onset of oxaliplatin-induced peripheral neuropathy symptoms in mice. Life Sci Alliance 2025; 8:e202402791. [PMID: 39578077 PMCID: PMC11584327 DOI: 10.26508/lsa.202402791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 11/12/2024] [Accepted: 11/12/2024] [Indexed: 11/24/2024] Open
Abstract
Oxaliplatin-induced peripheral neuropathy (OIPN) is an adverse side effect of this chemotherapy used for gastrointestinal cancers. The continuous pain experienced by OIPN patients often results in the reduction or discontinuation of chemotherapy, thereby affecting patient survival. Several pathogenic mechanisms involving sensory neurons were shown to participate in the occurrence of OIPN symptoms. However, the dysfunction of the blood-nerve barrier as a source of nerve alteration had not been thoroughly explored. To characterise the vascular contribution to OIPN symptoms, we undertook two comparative transcriptomic analyses of mouse purified brain and sciatic nerve blood vessels (BVs) and nerve BVs after oxaliplatin or control administration. These analyses reveal distinct molecular landscapes between brain and nerve BVs and the up-regulation of transcripts involved in vascular contraction after oxaliplatin treatment. Anatomical examination of the nerve yet shows the preservation of BV architecture in the acute OIPN mouse model, although treated mice exhibit both neuropathic symptoms and enhanced vasoconstriction reflected by hypoxia. Moreover, vasodilators significantly reduce oxaliplatin-induced neuropathic symptoms and endoneurial hypoxia, establishing the key involvement of nerve blood flow in OIPN.
Collapse
Affiliation(s)
- Sonia Taïb
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS UMR 7241, INSERM U1050, Université PSL, Paris, France
| | - Juliette Durand
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS UMR 7241, INSERM U1050, Université PSL, Paris, France
| | - Vianney Dehais
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS UMR 7241, INSERM U1050, Université PSL, Paris, France
| | - Anne-Cécile Boulay
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS UMR 7241, INSERM U1050, Université PSL, Paris, France
| | - Sabrina Martin
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS UMR 7241, INSERM U1050, Université PSL, Paris, France
| | - Corinne Blugeon
- Genomic Facility, Institut de Biologie de l'ENS (IBENS), École Normale Supérieure, CNRS UMR 8197, INSERM U1024, Université PSL, Paris, France
| | - Laurent Jourdren
- Genomic Facility, Institut de Biologie de l'ENS (IBENS), École Normale Supérieure, CNRS UMR 8197, INSERM U1024, Université PSL, Paris, France
| | - Rémi Freydier
- HydroSciences Montpellier, Université de Montpellier, CNRS, IRD, Montpellier, France
| | - Martine Cohen-Salmon
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS UMR 7241, INSERM U1050, Université PSL, Paris, France
| | - Jamilé Hazan
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS UMR 7241, INSERM U1050, Université PSL, Paris, France
| | - Isabelle Brunet
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS UMR 7241, INSERM U1050, Université PSL, Paris, France
| |
Collapse
|
2
|
Ru D, Zhang Z, Liu M, Fan X, Wang Y, Yan Y, Wang E. Downregulation of Notch Signaling-Stimulated Genes in Neurovascular Unit Alterations Induced by Chronic Cerebral Hypoperfusion. Immun Inflamm Dis 2024; 12:e70082. [PMID: 39607309 PMCID: PMC11603426 DOI: 10.1002/iid3.70082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 10/09/2024] [Accepted: 11/13/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND Chronic cerebral hypoperfusion (CCH) is a key contributor to vascular cognitive impairment (VCI) and is typically associated with blood-brain barrier (BBB) damage. This study investigates the pathological mechanisms underlying CCH-induced neurovascular unit (NVU) alterations. METHODS A mouse model of CCH was established using the bilateral common carotid artery stenosis (BCAS) procedure. Decreased cerebral blood flow (CBF) and impaired BBB integrity were assessed. Brain microvessel (BMV)-specific transcriptome profiles were analyzed using RNA-seq, supplemented with published single-cell RNA-seq data. RESULTS RNA-seq revealed neuroinflammation-related gene activation and significant downregulation of Notch signaling pathway genes in BMVs post-BCAS. Upregulated differentially expressed genes (DEGs) were enriched in microglia/macrophages, while downregulated DEGs were prominent in endothelial cells and pericytes. Enhanced activation of vascular-associated microglia (VAM) was linked to neurovascular alterations. CONCLUSION CCH induces significant NVU changes, marked by microglia-associated neuroinflammation and Notch signaling downregulation. These insights highlight potential therapeutic targets for treating neuroinflammatory and vascular-related neurodegenerative diseases.
Collapse
Affiliation(s)
- Dewen Ru
- Department of Neurosurgery, Jinshan HospitalFudan UniversityShanghaiChina
- Department of Neurosurgery, Huashan HospitalFudan UniversityShanghaiChina
- Shanghai Medical CollegeFudan UniversityShanghaiChina
| | - Zengyu Zhang
- Shanghai Medical CollegeFudan UniversityShanghaiChina
- Department of Neurology, Minhang HospitalFudan UniversityShanghaiChina
| | - Meng Liu
- Department of Neurosurgery, Jinshan HospitalFudan UniversityShanghaiChina
| | - Xuhui Fan
- Department of Neurosurgery, Jinshan HospitalFudan UniversityShanghaiChina
| | - Yuqi Wang
- Department of Neurosurgery, Jinshan HospitalFudan UniversityShanghaiChina
| | - Yufeng Yan
- Department of Neurosurgery, Jinshan HospitalFudan UniversityShanghaiChina
| | - Ersong Wang
- Department of Neurosurgery, Jinshan HospitalFudan UniversityShanghaiChina
| |
Collapse
|
3
|
Wang N, Ma Q, Zhang J, Wang J, Li X, Liang Y, Wu X. Transcriptomics-based anti-tuberculous mechanism of traditional Chinese polyherbal preparation NiuBeiXiaoHe intermediates. Front Pharmacol 2024; 15:1415951. [PMID: 39364045 PMCID: PMC11446850 DOI: 10.3389/fphar.2024.1415951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 07/29/2024] [Indexed: 10/05/2024] Open
Abstract
Background Integrated traditional Chinese medicine and biomedicine is an effective method to treat tuberculosis (TB). In our previous research, traditional Chinese medicine preparation NiuBeiXiaoHe (NBXH) achieved obvious anti-TB effects in animal experiments and clinical practice. However, the action mechanism of NBXH has not been elucidated. Method Peripheral blood mononuclear cells (PBMCs) were collected to extract mRNA and differentially expressed (DE) genes were obtained using gene microarray technology. Finally, GEO databases and RT-qPCR were used to verify the results of expression profile. Result After MTB infection, most upregulated DE genes in mice were immune-related genes, including cxcl9, camp, cfb, c4b, serpina3g, and ngp. Downregulated DE genes included lrrc74b, sult1d1, cxxc4, and grip2. After treatment with NBXH, especially high-dose NBXH, the abnormal gene expression was significantly corrected. Some DE genes have been confirmed in multiple GEO datasets or in pulmonary TB patients through RT-qPCR. Conclusion MTB infection led to extensive changes in host gene expression and mainly caused the host's anti-TB immune responses. The treatment using high-dose NBXH partially repaired the abnormal gene expression, further enhanced the anti-TB immunity included autophagy and NK cell-mediated cytotoxicity, and had a certain inhibitory effect on overactivated immune responses.
Collapse
Affiliation(s)
- Nan Wang
- Tuberculosis Prevention and Control Key Laboratory, Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Department of Tuberculosis, The Eighth Medical Center of PLA General Hospital, PLA General Hospital, Beijing, China
| | - Qianqian Ma
- Tuberculosis Prevention and Control Key Laboratory, Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Department of Tuberculosis, The Eighth Medical Center of PLA General Hospital, PLA General Hospital, Beijing, China
- Graduate School, Hebei North University, Zhangjiakou, Hebei, China
| | - Junxian Zhang
- Tuberculosis Prevention and Control Key Laboratory, Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Department of Tuberculosis, The Eighth Medical Center of PLA General Hospital, PLA General Hospital, Beijing, China
| | - Jie Wang
- Tuberculosis Prevention and Control Key Laboratory, Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Department of Tuberculosis, The Eighth Medical Center of PLA General Hospital, PLA General Hospital, Beijing, China
| | - Xiaojun Li
- Graduate School, Hebei North University, Zhangjiakou, Hebei, China
| | - Yan Liang
- Tuberculosis Prevention and Control Key Laboratory, Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Department of Tuberculosis, The Eighth Medical Center of PLA General Hospital, PLA General Hospital, Beijing, China
| | - Xueqiong Wu
- Tuberculosis Prevention and Control Key Laboratory, Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Department of Tuberculosis, The Eighth Medical Center of PLA General Hospital, PLA General Hospital, Beijing, China
| |
Collapse
|
4
|
Dupré N, Gueniot F, Domenga-Denier V, Dubosclard V, Nilles C, Hill-Eubanks D, Morgenthaler-Roth C, Nelson MT, Keime C, Danglot L, Joutel A. Protein aggregates containing wild-type and mutant NOTCH3 are major drivers of arterial pathology in CADASIL. J Clin Invest 2024; 134:e175789. [PMID: 38386425 PMCID: PMC11014667 DOI: 10.1172/jci175789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 02/20/2024] [Indexed: 02/24/2024] Open
Abstract
Loss of arterial smooth muscle cells (SMCs) and abnormal accumulation of the extracellular domain of the NOTCH3 receptor (Notch3ECD) are the 2 core features of CADASIL, a common cerebral small vessel disease caused by highly stereotyped dominant mutations in NOTCH3. Yet the relationship between NOTCH3 receptor activity, Notch3ECD accumulation, and arterial SMC loss has remained elusive, hampering the development of disease-modifying therapies. Using dedicated histopathological and multiscale imaging modalities, we could detect and quantify previously undetectable CADASIL-driven arterial SMC loss in the CNS of mice expressing the archetypal Arg169Cys mutation. We found that arterial pathology was more severe and Notch3ECD accumulation greater in transgenic mice overexpressing the mutation on a wild-type Notch3 background (TgNotch3R169C) than in knockin Notch3R170C/R170C mice expressing this mutation without a wild-type Notch3 copy. Notably, expression of Notch3-regulated genes was essentially unchanged in TgNotch3R169C arteries. We further showed that wild-type Notch3ECD coaggregated with mutant Notch3ECD and that elimination of 1 copy of wild-type Notch3 in TgNotch3R169C was sufficient to attenuate Notch3ECD accumulation and arterial pathology. These findings suggest that Notch3ECD accumulation, involving mutant and wild-type NOTCH3, is a major driver of arterial SMC loss in CADASIL, paving the way for NOTCH3-lowering therapeutic strategies.
Collapse
Affiliation(s)
- Nicolas Dupré
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Paris, France
| | - Florian Gueniot
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Paris, France
| | - Valérie Domenga-Denier
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Paris, France
| | - Virginie Dubosclard
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Paris, France
| | - Christelle Nilles
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Paris, France
| | - David Hill-Eubanks
- Department of Pharmacology, College of Medicine, University of Vermont, Burlington, Vermont, USA
| | - Christelle Morgenthaler-Roth
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS UMR 7104, INSERM U1258, Université de Strasbourg, Illkirch, France
| | - Mark T. Nelson
- Department of Pharmacology, College of Medicine, University of Vermont, Burlington, Vermont, USA
- Division of Cardiovascular Sciences, University of Manchester, Manchester, United Kingdom
| | - Céline Keime
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS UMR 7104, INSERM U1258, Université de Strasbourg, Illkirch, France
| | - Lydia Danglot
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Paris, France
| | - Anne Joutel
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Paris, France
- Department of Pharmacology, College of Medicine, University of Vermont, Burlington, Vermont, USA
- GHU Paris Psychiatrie et Neurosciences, Hôpital Sainte Anne, Paris, France
| |
Collapse
|
5
|
Mizuta I, Nakao-Azuma Y, Yoshida H, Yamaguchi M, Mizuno T. Progress to Clarify How NOTCH3 Mutations Lead to CADASIL, a Hereditary Cerebral Small Vessel Disease. Biomolecules 2024; 14:127. [PMID: 38254727 PMCID: PMC10813265 DOI: 10.3390/biom14010127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 01/09/2024] [Accepted: 01/16/2024] [Indexed: 01/24/2024] Open
Abstract
Notch signaling is conserved in C. elegans, Drosophila, and mammals. Among the four NOTCH genes in humans, NOTCH1, NOTCH2, and NOTCH3 are known to cause monogenic hereditary disorders. Most NOTCH-related disorders are congenital and caused by a gain or loss of Notch signaling activity. In contrast, cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) caused by NOTCH3 is adult-onset and considered to be caused by accumulation of the mutant NOTCH3 extracellular domain (N3ECD) and, possibly, by an impairment in Notch signaling. Pathophysiological processes following mutant N3ECD accumulation have been intensively investigated; however, the process leading to N3ECD accumulation and its association with canonical NOTCH3 signaling remain unknown. We reviewed the progress in clarifying the pathophysiological process involving mutant NOTCH3.
Collapse
Affiliation(s)
- Ikuko Mizuta
- Department of Neurology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan; (I.M.)
| | - Yumiko Nakao-Azuma
- Department of Neurology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan; (I.M.)
- Department of Rehabilitation Medicine, Gunma University Graduate School of Medicine, Showa-machi, Maebashi, Gunma 371-8511, Japan
| | - Hideki Yoshida
- Department of Applied Biology, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto 606-8585, Japan
| | - Masamitsu Yamaguchi
- Department of Applied Biology, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto 606-8585, Japan
- Kansai Gakken Laboratory, Kankyo Eisei Yakuhin Co., Ltd., 3-6-2 Hikaridai, Seika-cho, Kyoto 619-0237, Japan
| | - Toshiki Mizuno
- Department of Neurology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan; (I.M.)
| |
Collapse
|
6
|
Specification of CNS macrophage subsets occurs postnatally in defined niches. Nature 2022; 604:740-748. [PMID: 35444273 DOI: 10.1038/s41586-022-04596-2] [Citation(s) in RCA: 163] [Impact Index Per Article: 54.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 02/28/2022] [Indexed: 02/08/2023]
Abstract
All tissue-resident macrophages of the central nervous system (CNS)-including parenchymal microglia, as well as CNS-associated macrophages (CAMs1) such as meningeal and perivascular macrophages2-7-are part of the CNS endogenous innate immune system that acts as the first line of defence during infections or trauma2,8-10. It has been suggested that microglia and all subsets of CAMs are derived from prenatal cellular sources in the yolk sac that were defined as early erythromyeloid progenitors11-15. However, the precise ontogenetic relationships, the underlying transcriptional programs and the molecular signals that drive the development of distinct CAM subsets in situ are poorly understood. Here we show, using fate-mapping systems, single-cell profiling and cell-specific mutants, that only meningeal macrophages and microglia share a common prenatal progenitor. By contrast, perivascular macrophages originate from perinatal meningeal macrophages only after birth in an integrin-dependent manner. The establishment of perivascular macrophages critically requires the presence of arterial vascular smooth muscle cells. Together, our data reveal a precisely timed process in distinct anatomical niches for the establishment of macrophage subsets in the CNS.
Collapse
|
7
|
Ruchoux MM, Kalaria RN, Román GC. The pericyte: A critical cell in the pathogenesis of CADASIL. CEREBRAL CIRCULATION - COGNITION AND BEHAVIOR 2021; 2:100031. [PMID: 34950895 PMCID: PMC8661128 DOI: 10.1016/j.cccb.2021.100031] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 10/12/2021] [Accepted: 10/12/2021] [Indexed: 12/22/2022]
Abstract
CADASIL is the most common hereditary small vessel disease presenting with strokes and subcortical vascular dementia caused by mutations in the NOTCH3 gene. CADASIL is a vasculopathy primarily involving vascular smooth-muscle cells. Arteriolar and capillary pericyte damage or deficiency is a key feature in disease pathogenesis. Pericyte-mediated cerebral venous insufficiency may explain white matter lesions and increased perivascular spaces. Central role of the pericyte offers novel approaches to the treatment of CADASIL.
Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) is a hereditary small vessel disease presenting with migraine, mood and cognitive disorders, focal neurological deficits, recurrent ischemic attacks, lacunar infarcts and brain white matter changes. As they age, CADASIL patients invariably develop cognitive impairment and subcortical dementia. CADASIL is caused by missense mutations in the NOTCH3 gene resulting in a profound cerebral vasculopathy affecting primarily arterial vascular smooth muscle cells, which target the microcirculation and perfusion. Based on a thorough review of morphological lesions in arteries, veins, and capillaries in CADASIL, we surmise that arteriolar and capillary pericyte damage or deficiency appears a key feature in the pathogenesis of the disease. This may affect critical pericyte-endothelial interactions causing stroke injury and vasomotor disturbances. Changes in microvascular permeability due to perhaps localized blood-brain barrier alterations and pericyte secretory dysfunction likely contribute to delayed neuronal as well as glial cell death. Moreover, pericyte-mediated cerebral venous insufficiency may explain white matter lesions and the dilatation of Virchow-Robin perivascular spaces typical of CADASIL. The postulated central role of the pericyte offers some novel approaches to the study and treatment of CADASIL and enable elucidation of other forms of cerebral small vessel diseases and subcortical vascular dementia.
Collapse
Affiliation(s)
- Marie-Magdeleine Ruchoux
- Former researcher, Université d'Artois, Blood-Brain-Barrier Laboratory Lens France, Former advisor, Alzheimer's Clinic Methodist Neurological Institute, Houston TX, USA
| | - Raj N Kalaria
- Translational and Clinical Research Institute, Newcastle University, Newcastle-upon-Tyne, United Kingdom
| | - Gustavo C Román
- Methodist Neurological Institute, Department of Neurology, Houston Methodist Hospital Houston TX 77030, USA, Weill Cornell Medical College, New York NY, USA and Texas A&M Medical School, Bryan TX, USA
| |
Collapse
|
8
|
Matsuura N, Tanaka K, Yamasaki M, Yamashita K, Saito T, Makino T, Yamamoto K, Takahashi T, Kurokawa Y, Nakajima K, Eguchi H, Nakagawa H, Doki Y. NOTCH3 limits the epithelial-mesenchymal transition and predicts a favorable clinical outcome in esophageal cancer. Cancer Med 2021; 10:3986-3996. [PMID: 34042293 PMCID: PMC8209574 DOI: 10.1002/cam4.3933] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 03/30/2021] [Accepted: 04/08/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Esophageal squamous cell carcinoma (ESCC) is the deadliest of all human squamous cell carcinomas and is characterized by chemotherapy resistance and poor prognosis associated with the epithelial-mesenchymal transition (EMT). A subset of ESCC displays loss-of-function mutations in genes encoding Notch receptor family members, including NOTCH3. Although Notch signaling regulates EMT in ESCC cells, the role of NOTCH3 in EMT and chemotherapy resistance remains elusive. This study aimed to examine the role of NOTCH3 in EMT and chemotherapy resistance, and determine whether NOTCH3 expression can be used to predict the response to chemotherapy. METHODS In vitro and in vivo assays were conducted to clarify the contribution of NOTCH3 to chemotherapy resistance. Using specimens from 120 ESCC patients treated with neoadjuvant chemotherapy, we compared the expression levels of NOTCH3 and genes involved in EMT according to the degree of chemotherapy sensitivity. RESULTS In ESCC cells, chemotherapy resistance was associated with NOTCH3 downregulation and concurrent activation of EMT. RNA interference to silence NOTCH3 resulted in induction of the EMT marker Vimentin (VIM), leading to chemotherapy resistance in ESCC cells. Conversely, ectopic expression of the activated form of NOTCH3 suppressed EMT and sensitized cells to chemotherapy. Results of chromatin immunoprecipitation assays suggested that NOTCH3 may repress transcription of the VIM. CONCLUSIONS Our findings suggest that NOTCH3 may control chemotherapy sensitivity by regulating EMT. NOTCH3 may serve as a novel biomarker to predict better clinical outcomes in ESCC patients.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Animals
- Antimetabolites, Antineoplastic/pharmacology
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/physiology
- Down-Regulation
- Drug Resistance, Neoplasm/genetics
- Epithelial-Mesenchymal Transition/drug effects
- Epithelial-Mesenchymal Transition/genetics
- Esophageal Neoplasms/drug therapy
- Esophageal Neoplasms/genetics
- Esophageal Neoplasms/surgery
- Esophageal Squamous Cell Carcinoma/drug therapy
- Esophageal Squamous Cell Carcinoma/genetics
- Esophageal Squamous Cell Carcinoma/surgery
- Esophagectomy
- Female
- Fluorouracil/pharmacology
- Gene Silencing
- Humans
- Loss of Function Mutation
- Mice, Inbred BALB C
- Mice, Nude
- Middle Aged
- Receptor, Notch3/drug effects
- Receptor, Notch3/genetics
- Receptor, Notch3/metabolism
- Vimentin/metabolism
- Mice
Collapse
Affiliation(s)
- Norihiro Matsuura
- Department of Gastroenterological SurgeryGraduate School of MedicineOsaka UniversitySuitaOsakaJapan
| | - Koji Tanaka
- Department of Gastroenterological SurgeryGraduate School of MedicineOsaka UniversitySuitaOsakaJapan
| | - Makoto Yamasaki
- Department of Gastroenterological SurgeryGraduate School of MedicineOsaka UniversitySuitaOsakaJapan
| | - Kotaro Yamashita
- Department of Gastroenterological SurgeryGraduate School of MedicineOsaka UniversitySuitaOsakaJapan
| | - Takuro Saito
- Department of Gastroenterological SurgeryGraduate School of MedicineOsaka UniversitySuitaOsakaJapan
| | - Tomoki Makino
- Department of Gastroenterological SurgeryGraduate School of MedicineOsaka UniversitySuitaOsakaJapan
| | - Kazuyoshi Yamamoto
- Department of Gastroenterological SurgeryGraduate School of MedicineOsaka UniversitySuitaOsakaJapan
| | - Tsuyoshi Takahashi
- Department of Gastroenterological SurgeryGraduate School of MedicineOsaka UniversitySuitaOsakaJapan
| | - Yukinori Kurokawa
- Department of Gastroenterological SurgeryGraduate School of MedicineOsaka UniversitySuitaOsakaJapan
| | - Kiyokazu Nakajima
- Department of Gastroenterological SurgeryGraduate School of MedicineOsaka UniversitySuitaOsakaJapan
| | - Hidetoshi Eguchi
- Department of Gastroenterological SurgeryGraduate School of MedicineOsaka UniversitySuitaOsakaJapan
| | - Hiroshi Nakagawa
- Herbert Irving Comprehensive Cancer CenterColumbia UniversityNew YorkNYUSA
| | - Yuichiro Doki
- Department of Gastroenterological SurgeryGraduate School of MedicineOsaka UniversitySuitaOsakaJapan
| |
Collapse
|
9
|
Tseng WC, Johnson Escauriza AJ, Tsai-Morris CH, Feldman B, Dale RK, Wassif CA, Porter FD. The role of Niemann-Pick type C2 in zebrafish embryonic development. Development 2021; 148:dev194258. [PMID: 33722902 PMCID: PMC8077516 DOI: 10.1242/dev.194258] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 03/04/2021] [Indexed: 12/11/2022]
Abstract
Niemann-Pick disease type C (NPC) is a rare, fatal, neurodegenerative lysosomal disease caused by mutations of either NPC1 or NPC2. NPC2 is a soluble lysosomal protein that functions in coordination with NPC1 to efflux cholesterol from the lysosomal compartment. Mutations of either gene result in the accumulation of unesterified cholesterol and other lipids in the late endosome/lysosome, and reduction of cellular cholesterol bioavailability. Zygotic null npc2m/m zebrafish showed significant unesterified cholesterol accumulation at larval stages, a reduction in body size, and motor and balance defects in adulthood. However, the phenotype at embryonic stages was milder than expected, suggesting a possible role of maternal Npc2 in embryonic development. Maternal-zygotic npc2m/m zebrafish exhibited significant developmental defects, including defective otic vesicle development/absent otoliths, abnormal head/brain development, curved/twisted body axes and no circulating blood cells, and died by 72 hpf. RNA-seq analysis conducted on 30 hpf npc2+/m and MZnpc2m/m embryos revealed a significant reduction in the expression of notch3 and other downstream genes in the Notch signaling pathway, suggesting that impaired Notch3 signaling underlies aspects of the developmental defects observed in MZnpc2m/m zebrafish.
Collapse
Affiliation(s)
- Wei-Chia Tseng
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, USA
| | - Ana J. Johnson Escauriza
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, USA
| | - Chon-Hwa Tsai-Morris
- Zebrafish Core, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, USA
| | - Benjamin Feldman
- Zebrafish Core, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, USA
| | - Ryan K. Dale
- Bioinformatics and Scientific Programming Core, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, USA
| | - Christopher A. Wassif
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, USA
| | - Forbes D. Porter
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, USA
| |
Collapse
|
10
|
Chabriat H, Joutel A, Tournier‐Lasserve E, Bousser MG. CADASIL: yesterday, today, tomorrow. Eur J Neurol 2020; 27:1588-1595. [DOI: 10.1111/ene.14293] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 04/28/2020] [Indexed: 12/27/2022]
Affiliation(s)
- H. Chabriat
- Department of Neurology and CERVCO Reference Center for Rare Vascular Diseases of the Eye and Brain Hôpital Lariboisiére, APHP Paris France
- INSERM U 1141 Paris France
- University of Paris Paris France
| | - A. Joutel
- University of Paris Paris France
- Institute of Psychiatry and Neurosciences of Paris INSERM U1266 Paris France
| | - E. Tournier‐Lasserve
- INSERM U 1141 Paris France
- University of Paris Paris France
- Molecular Genetics Department and CERVCO Reference Center for Rare Vascular Diseases of the Eye and Brain Hopital Lariboisiére, APHP Paris France
| | - M. G. Bousser
- Department of Neurology and CERVCO Reference Center for Rare Vascular Diseases of the Eye and Brain Hôpital Lariboisiére, APHP Paris France
- University of Paris Paris France
| |
Collapse
|
11
|
Ratelade J, Klug NR, Lombardi D, Angelim MKSC, Dabertrand F, Domenga-Denier V, Al-Shahi Salman R, Smith C, Gerbeau JF, Nelson MT, Joutel A. Reducing Hypermuscularization of the Transitional Segment Between Arterioles and Capillaries Protects Against Spontaneous Intracerebral Hemorrhage. Circulation 2020; 141:2078-2094. [PMID: 32183562 DOI: 10.1161/circulationaha.119.040963] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Spontaneous deep intracerebral hemorrhage (ICH) is a devastating subtype of stroke without specific treatments. It has been thought that smooth muscle cell (SMC) degeneration at the site of arteriolar wall rupture may be sufficient to cause hemorrhage. However, deep ICHs are rare in some aggressive small vessel diseases that are characterized by significant arteriolar SMC degeneration. Here we hypothesized that a second cellular defect may be required for the occurrence of ICH. METHODS We studied a genetic model of spontaneous deep ICH using Col4a1+/G498V and Col4a1+/G1064D mouse lines that are mutated for the α1 chain of collagen type IV. We analyzed cerebroretinal microvessels, performed genetic rescue experiments, vascular reactivity analysis, and computational modeling. We examined postmortem brain tissues from patients with sporadic deep ICH. RESULTS We identified in the normal cerebroretinal vasculature a novel segment between arterioles and capillaries, herein called the transitional segment (TS), which is covered by mural cells distinct from SMCs and pericytes. In Col4a1 mutant mice, this TS was hypermuscularized, with a hyperplasia of mural cells expressing more contractile proteins, whereas the upstream arteriole exhibited a loss of SMCs. TSs mechanistically showed a transient increase in proliferation of mural cells during postnatal maturation. Mutant brain microvessels, unlike mutant arteries, displayed a significant upregulation of SM genes and Notch3 target genes, and genetic reduction of Notch3 in Col4a1+/G498V mice protected against ICH. Retina analysis showed that hypermuscularization of the TS was attenuated, but arteriolar SMC loss was unchanged in Col4a1+/G498V, Notch3+/- mice. Moreover, hypermuscularization of the retinal TS increased its contractility and tone and raised the intravascular pressure in the upstream feeding arteriole. We similarly found hypermuscularization of the TS and focal arteriolar SMC loss in brain tissues from patients with sporadic deep ICH. CONCLUSIONS Our results suggest that hypermuscularization of the TS, through increased Notch3 activity, is involved in the occurrence of ICH in Col4a1 mutant mice, by raising the intravascular pressure in the upstream feeding arteriole and promoting its rupture at the site of SMC loss. Our human data indicate that these 2 mutually reinforcing vascular defects may represent a general mechanism of deep ICH.
Collapse
Affiliation(s)
- Julien Ratelade
- Institute of Psychiatry and Neurosciences of Paris (IPNP), Inserm U1266, University of Paris, France (J.R., M.K.S.C.A., V.D-D., A.J.)
| | - Nicholas R Klug
- Department of Pharmacology, College of Medicine, University of Vermont, Burlington (N.R.K., F.D., M.T.N., A.J.)
| | - Damiano Lombardi
- Inria Paris, Sorbonne University, Laboratory Jacques-Louis Lions, France (D.L., J.-F.G.)
| | | | - Fabrice Dabertrand
- Department of Pharmacology, College of Medicine, University of Vermont, Burlington (N.R.K., F.D., M.T.N., A.J.).,Department of Anesthesiology, Department of Pharmacology, Anschutz Medical Campus, University of Colorado, Aurora (F.D.)
| | - Valérie Domenga-Denier
- Institute of Psychiatry and Neurosciences of Paris (IPNP), Inserm U1266, University of Paris, France (J.R., M.K.S.C.A., V.D-D., A.J.)
| | - Rustam Al-Shahi Salman
- Centre for Clinical Brain Sciences, University of Edinburgh, United Kingdom (R.A.-S.S., C.S.)
| | - Colin Smith
- Centre for Clinical Brain Sciences, University of Edinburgh, United Kingdom (R.A.-S.S., C.S.)
| | - Jean-Frédéric Gerbeau
- Inria Paris, Sorbonne University, Laboratory Jacques-Louis Lions, France (D.L., J.-F.G.)
| | - Mark T Nelson
- Department of Pharmacology, College of Medicine, University of Vermont, Burlington (N.R.K., F.D., M.T.N., A.J.).,Division of Cardiovascular Sciences, University of Manchester, United Kingdom (M.T.N.)
| | - Anne Joutel
- Institute of Psychiatry and Neurosciences of Paris (IPNP), Inserm U1266, University of Paris, France (J.R., M.K.S.C.A., V.D-D., A.J.).,Department of Pharmacology, College of Medicine, University of Vermont, Burlington (N.R.K., F.D., M.T.N., A.J.).,DHU NeuroVasc, Sorbonne Paris Cité, France (A.J.)
| |
Collapse
|
12
|
Dunn PJ, Maksemous N, Smith RA, Sutherland HG, Haupt LM, Griffiths LR. Investigating diagnostic sequencing techniques for CADASIL diagnosis. Hum Genomics 2020; 14:2. [PMID: 31915071 PMCID: PMC6950909 DOI: 10.1186/s40246-019-0255-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 12/30/2019] [Indexed: 11/18/2022] Open
Abstract
Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) is a cerebral small vessel disease caused by mutations in the NOTCH3 gene. Our laboratory has been undertaking genetic diagnostic testing for CADASIL since 1997. Work originally utilised Sanger sequencing methods targeting specific NOTCH3 exons. More recently, next-generation sequencing (NGS)-based technologies such as a targeted gene panel and whole exome sequencing (WES) have been used for improved genetic diagnostic testing. In this study, data from 680 patient samples was analysed for 764 tests utilising 3 different sequencing technologies. Sanger sequencing was performed for 407 tests, a targeted NGS gene panel which includes NOTCH3 exonic regions accounted for 354 tests, and WES with targeted analysis was performed for 3 tests. In total, 14.7% of patient samples (n = 100/680) were determined to have a mutation. Testing efficacy varied by method, with 10.8% (n = 44/407) of tests using Sanger sequencing able to identify mutations, with 15.8% (n = 56/354) of tests performed using the NGS custom panel successfully identifying mutations and a likely non-NOTCH3 pathogenic variant (n = 1/3) identified through WES. Further analysis was then performed through stratification of the number of mutations detected at our facility based on the number of exons, level of pathogenicity and the classification of mutations as known or novel. A systematic review of NOTCH3 mutation testing data from 1997 to 2017 determined the diagnostic rate of pathogenic findings and found the NGS-customised panel increases our ability to identify disease-causing mutations in NOTCH3.
Collapse
Affiliation(s)
- P J Dunn
- Genomics Research Centre, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - N Maksemous
- Genomics Research Centre, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - R A Smith
- Genomics Research Centre, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - H G Sutherland
- Genomics Research Centre, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - L M Haupt
- Genomics Research Centre, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia.
| | - L R Griffiths
- Genomics Research Centre, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| |
Collapse
|
13
|
Rippe C, Albinsson S, Guron G, Nilsson H, Swärd K. Targeting transcriptional control of soluble guanylyl cyclase via NOTCH for prevention of cardiovascular disease. Acta Physiol (Oxf) 2019; 225:e13094. [PMID: 29754438 DOI: 10.1111/apha.13094] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 05/04/2018] [Accepted: 05/04/2018] [Indexed: 12/18/2022]
Abstract
Soluble guanylyl cyclase (sGC) is an effector enzyme of nitric oxide (NO). Recent work has unravelled how levels of this enzyme are controlled, and highlighted a role in vascular disease. We provide a timely summary of available knowledge on transcriptional regulation of sGC, including influences from the NOTCH signalling pathway and genetic variants. It is speculated that hypertension-induced repression of sGC starts a vicious circle that can be initiated by periods of stress, diet or genetic factors, and a key tenet is that reduction in sGC further raises blood pressure. The idea that dysregulation of sGC contributes to syndromes caused by defective NOTCH signalling is advanced, and we discuss drug repositioning for vascular disease prevention. The advantage of targeting sGC expression rather than activity is also considered. It is argued that transcriptional inputs on sGC arise from interactions with other cells, the extracellular matrix and microRNAs (miRNAs), and concluded that the promise of sGC as a target for prevention of cardiovascular disease has increased in recent time.
Collapse
Affiliation(s)
- C. Rippe
- Department of Experimental Medical Science; Lund University; Lund Sweden
| | - S. Albinsson
- Department of Experimental Medical Science; Lund University; Lund Sweden
| | - G. Guron
- Department of Physiology; University of Gothenburg; Gothenburg Sweden
| | - H. Nilsson
- Department of Physiology; University of Gothenburg; Gothenburg Sweden
| | - K. Swärd
- Department of Experimental Medical Science; Lund University; Lund Sweden
| |
Collapse
|
14
|
Isolation and differential transcriptome of vascular smooth muscle cells and mid-capillary pericytes from the rat brain. Sci Rep 2018; 8:12272. [PMID: 30116021 PMCID: PMC6095852 DOI: 10.1038/s41598-018-30739-5] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 07/20/2018] [Indexed: 12/20/2022] Open
Abstract
Brain mural cells form a heterogeneous family which significantly contributes to the maintenance of the blood-brain barrier and regulation of the cerebral blood flow. Current procedures to isolate them cannot specifically separate their distinct subtypes, in particular vascular smooth muscle cells (VSMCs) and mid-capillary pericytes (mcPCs), which differ among others by their expression of smooth muscle actin (SMA). We herein describe an innovative method allowing SMA+ VSMCs and SMA− mcPCs to be freshly isolated from the rat cerebral cortex. Using differential RNA-Seq analysis, we then reveal the specific gene expression profile of each subtype. Our results refine the current description of the role of VSMCs in parenchymal cortical arterioles at the molecular level and provide a unique platform to identify the molecular mechanisms underlying the specific functions of mcPCs in the brain vasculature.
Collapse
|
15
|
Rippe C, Zhu B, Krawczyk KK, Bavel EV, Albinsson S, Sjölund J, Bakker ENTP, Swärd K. Hypertension reduces soluble guanylyl cyclase expression in the mouse aorta via the Notch signaling pathway. Sci Rep 2017; 7:1334. [PMID: 28465505 PMCID: PMC5430981 DOI: 10.1038/s41598-017-01392-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 03/28/2017] [Indexed: 02/06/2023] Open
Abstract
Hypertension is a dominating risk factor for cardiovascular disease. To characterize the genomic response to hypertension, we administered vehicle or angiotensin II to mice and performed gene expression analyses. AngII treatment resulted in a robust increase in blood pressure and altered expression of 235 genes in the aorta, including Gucy1a3 and Gucy1b3 which encode subunits of soluble guanylyl cyclase (sGC). Western blotting and immunohistochemistry confirmed repression of sGC associated with curtailed relaxation via sGC activation. Analysis of transcription factor binding motifs in promoters of differentially expressed genes identified enrichment of motifs for RBPJ, a component of the Notch signaling pathway, and the Notch coactivators FRYL and MAML2 were reduced. Gain and loss of function experiments demonstrated that JAG/NOTCH signaling controls sGC expression together with MAML2 and FRYL. Reduced expression of sGC, correlating with differential expression of MAML2, in stroke prone and spontaneously hypertensive rats was also seen, and RNA-Seq data demonstrated correlations between JAG1, NOTCH3, MAML2 and FRYL and the sGC subunits GUCY1A3 and GUCY1B3 in human coronary artery. Notch signaling thus provides a constitutive drive on expression of the major nitric oxide receptor (GUCY1A3/GUCY1B3) in arteries from mice, rats, and humans, and this control mechanism is disturbed in hypertension.
Collapse
Affiliation(s)
- Catarina Rippe
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Baoyi Zhu
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | | | - Ed Van Bavel
- Department of Biomedical Engineering and Physics, Academic Medical Center, Amsterdam, The Netherlands
| | | | - Jonas Sjölund
- Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Erik N T P Bakker
- Department of Biomedical Engineering and Physics, Academic Medical Center, Amsterdam, The Netherlands
| | - Karl Swärd
- Department of Experimental Medical Science, Lund University, Lund, Sweden.
| |
Collapse
|
16
|
Baron-Menguy C, Domenga-Denier V, Ghezali L, Faraci FM, Joutel A. Increased Notch3 Activity Mediates Pathological Changes in Structure of Cerebral Arteries. Hypertension 2016; 69:60-70. [PMID: 27821617 DOI: 10.1161/hypertensionaha.116.08015] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 07/07/2016] [Accepted: 08/26/2016] [Indexed: 01/24/2023]
Abstract
CADASIL (Cerebral Autosomal Dominant Arteriopathy With Subcortical Infarcts and Leukoencephalopathy), the most frequent genetic cause of stroke and vascular dementia, is caused by highly stereotyped mutations in the NOTCH3 receptor, which is predominantly expressed in vascular smooth muscle. The well-established TgNotch3R169C mouse model develops characteristic features of the human disease, with deposition of NOTCH3 and other proteins, including TIMP3 (tissue inhibitor of metalloproteinase 3), on brain vessels, as well as reduced maximal dilation, and attenuated myogenic tone of cerebral arteries, but without elevated blood pressure. Increased TIMP3 levels were recently shown to be a major determinant of altered myogenic tone. In this study, we investigated the contribution of TIMP3 and Notch3 signaling to the impairment of maximal vasodilator capacity caused by the archetypal R169C mutation. Maximally dilated cerebral arteries in TgNotch3R169C mice exhibited a decrease in lumen diameter over a range of physiological pressures that occurred before myogenic tone deficits. This defect was not prevented by genetic reduction of TIMP3 in TgNotch3R169C mice and was not observed in mice overexpressing TIMP3. Knock-in mice with the R169C mutation (Notch3R170C/R170C) exhibited similar reductions in arterial lumen, and both TgNotch3R169C and Notch3R170C/R170C mice showed increased cerebral artery expression of Notch3 target genes. Reduced maximal vasodilation was prevented by conditional reduction of Notch activity in smooth muscle of TgNotch3R169C mice and mimicked by conditional activation of Notch3 in smooth muscle, an effect that was blood pressure-independent. We conclude that increased Notch3 activity mediates reduction in maximal dilator capacity of cerebral arteries in CADASIL and may contribute to reductions in cerebral blood flow.
Collapse
Affiliation(s)
- Celine Baron-Menguy
- From the Genetics and Pathogenesis of Cerebrovascular Diseases, INSERM, UMRS 1161, and Univ Paris Diderot, Sorbonne Paris Cité, Paris, France (C.B.-M., V.D.-D., L.G., A.J.); DHU NeuroVasc Sorbonne Paris Cité, Paris, France (C.B.-M., V.D.-D., L.G., A.J.); and Departments of Internal Medicine and Pharmacology, Francois M. Abboud Cardiovascular Center, The University of Iowa Carver College of Medicine, Iowa City Veterans Affairs Healthcare System (F.M.F.)
| | - Valérie Domenga-Denier
- From the Genetics and Pathogenesis of Cerebrovascular Diseases, INSERM, UMRS 1161, and Univ Paris Diderot, Sorbonne Paris Cité, Paris, France (C.B.-M., V.D.-D., L.G., A.J.); DHU NeuroVasc Sorbonne Paris Cité, Paris, France (C.B.-M., V.D.-D., L.G., A.J.); and Departments of Internal Medicine and Pharmacology, Francois M. Abboud Cardiovascular Center, The University of Iowa Carver College of Medicine, Iowa City Veterans Affairs Healthcare System (F.M.F.)
| | - Lamia Ghezali
- From the Genetics and Pathogenesis of Cerebrovascular Diseases, INSERM, UMRS 1161, and Univ Paris Diderot, Sorbonne Paris Cité, Paris, France (C.B.-M., V.D.-D., L.G., A.J.); DHU NeuroVasc Sorbonne Paris Cité, Paris, France (C.B.-M., V.D.-D., L.G., A.J.); and Departments of Internal Medicine and Pharmacology, Francois M. Abboud Cardiovascular Center, The University of Iowa Carver College of Medicine, Iowa City Veterans Affairs Healthcare System (F.M.F.)
| | - Frank M Faraci
- From the Genetics and Pathogenesis of Cerebrovascular Diseases, INSERM, UMRS 1161, and Univ Paris Diderot, Sorbonne Paris Cité, Paris, France (C.B.-M., V.D.-D., L.G., A.J.); DHU NeuroVasc Sorbonne Paris Cité, Paris, France (C.B.-M., V.D.-D., L.G., A.J.); and Departments of Internal Medicine and Pharmacology, Francois M. Abboud Cardiovascular Center, The University of Iowa Carver College of Medicine, Iowa City Veterans Affairs Healthcare System (F.M.F.)
| | - Anne Joutel
- From the Genetics and Pathogenesis of Cerebrovascular Diseases, INSERM, UMRS 1161, and Univ Paris Diderot, Sorbonne Paris Cité, Paris, France (C.B.-M., V.D.-D., L.G., A.J.); DHU NeuroVasc Sorbonne Paris Cité, Paris, France (C.B.-M., V.D.-D., L.G., A.J.); and Departments of Internal Medicine and Pharmacology, Francois M. Abboud Cardiovascular Center, The University of Iowa Carver College of Medicine, Iowa City Veterans Affairs Healthcare System (F.M.F.).
| |
Collapse
|
17
|
Ragot H, Monfort A, Baudet M, Azibani F, Fazal L, Merval R, Polidano E, Cohen-Solal A, Delcayre C, Vodovar N, Chatziantoniou C, Samuel JL. Loss of Notch3 Signaling in Vascular Smooth Muscle Cells Promotes Severe Heart Failure Upon Hypertension. Hypertension 2016; 68:392-400. [PMID: 27296994 DOI: 10.1161/hypertensionaha.116.07694] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 05/10/2016] [Indexed: 12/28/2022]
Abstract
Hypertension, which is a risk factor of heart failure, provokes adaptive changes at the vasculature and cardiac levels. Notch3 signaling plays an important role in resistance arteries by controlling the maturation of vascular smooth muscle cells. Notch3 deletion is protective in pulmonary hypertension while deleterious in arterial hypertension. Although this latter phenotype was attributed to renal and cardiac alterations, the underlying mechanisms remained unknown. To investigate the role of Notch3 signaling in the cardiac adaptation to hypertension, we used mice with either constitutive Notch3 or smooth muscle cell-specific conditional RBPJκ knockout. At baseline, both genotypes exhibited a cardiac arteriolar rarefaction associated with oxidative stress. In response to angiotensin II-induced hypertension, the heart of Notch3 knockout and SM-RBPJκ knockout mice did not adapt to pressure overload and developed heart failure, which could lead to an early and fatal acute decompensation of heart failure. This cardiac maladaptation was characterized by an absence of media hypertrophy of the media arteries, the transition of smooth muscle cells toward a synthetic phenotype, and an alteration of angiogenic pathways. A subset of mice exhibited an early fatal acute decompensated heart failure, in which the same alterations were observed, although in a more rapid timeframe. Altogether, these observations indicate that Notch3 plays a major role in coronary adaptation to pressure overload. These data also show that the hypertrophy of coronary arterial media on pressure overload is mandatory to initially maintain a normal cardiac function and is regulated by the Notch3/RBPJκ pathway.
Collapse
Affiliation(s)
- Hélène Ragot
- From the Inserm UMR-S 942 and Paris Diderot University, Paris, France (H.R., A.M., M.B., F.A., L.F., R.M., E.P., A.C.-S., C.D., N.V., J.-L.S.); Department of Cardiology, Lariboisière Hospital, Paris, France (A.C.-S.); and Inserm UMR-S 1155 and Pierre and Marie Curie University, Paris, France (C.C.)
| | - Astrid Monfort
- From the Inserm UMR-S 942 and Paris Diderot University, Paris, France (H.R., A.M., M.B., F.A., L.F., R.M., E.P., A.C.-S., C.D., N.V., J.-L.S.); Department of Cardiology, Lariboisière Hospital, Paris, France (A.C.-S.); and Inserm UMR-S 1155 and Pierre and Marie Curie University, Paris, France (C.C.)
| | - Mathilde Baudet
- From the Inserm UMR-S 942 and Paris Diderot University, Paris, France (H.R., A.M., M.B., F.A., L.F., R.M., E.P., A.C.-S., C.D., N.V., J.-L.S.); Department of Cardiology, Lariboisière Hospital, Paris, France (A.C.-S.); and Inserm UMR-S 1155 and Pierre and Marie Curie University, Paris, France (C.C.)
| | - Fériel Azibani
- From the Inserm UMR-S 942 and Paris Diderot University, Paris, France (H.R., A.M., M.B., F.A., L.F., R.M., E.P., A.C.-S., C.D., N.V., J.-L.S.); Department of Cardiology, Lariboisière Hospital, Paris, France (A.C.-S.); and Inserm UMR-S 1155 and Pierre and Marie Curie University, Paris, France (C.C.)
| | - Loubina Fazal
- From the Inserm UMR-S 942 and Paris Diderot University, Paris, France (H.R., A.M., M.B., F.A., L.F., R.M., E.P., A.C.-S., C.D., N.V., J.-L.S.); Department of Cardiology, Lariboisière Hospital, Paris, France (A.C.-S.); and Inserm UMR-S 1155 and Pierre and Marie Curie University, Paris, France (C.C.)
| | - Régine Merval
- From the Inserm UMR-S 942 and Paris Diderot University, Paris, France (H.R., A.M., M.B., F.A., L.F., R.M., E.P., A.C.-S., C.D., N.V., J.-L.S.); Department of Cardiology, Lariboisière Hospital, Paris, France (A.C.-S.); and Inserm UMR-S 1155 and Pierre and Marie Curie University, Paris, France (C.C.)
| | - Evelyne Polidano
- From the Inserm UMR-S 942 and Paris Diderot University, Paris, France (H.R., A.M., M.B., F.A., L.F., R.M., E.P., A.C.-S., C.D., N.V., J.-L.S.); Department of Cardiology, Lariboisière Hospital, Paris, France (A.C.-S.); and Inserm UMR-S 1155 and Pierre and Marie Curie University, Paris, France (C.C.)
| | - Alain Cohen-Solal
- From the Inserm UMR-S 942 and Paris Diderot University, Paris, France (H.R., A.M., M.B., F.A., L.F., R.M., E.P., A.C.-S., C.D., N.V., J.-L.S.); Department of Cardiology, Lariboisière Hospital, Paris, France (A.C.-S.); and Inserm UMR-S 1155 and Pierre and Marie Curie University, Paris, France (C.C.)
| | - Claude Delcayre
- From the Inserm UMR-S 942 and Paris Diderot University, Paris, France (H.R., A.M., M.B., F.A., L.F., R.M., E.P., A.C.-S., C.D., N.V., J.-L.S.); Department of Cardiology, Lariboisière Hospital, Paris, France (A.C.-S.); and Inserm UMR-S 1155 and Pierre and Marie Curie University, Paris, France (C.C.)
| | - Nicolas Vodovar
- From the Inserm UMR-S 942 and Paris Diderot University, Paris, France (H.R., A.M., M.B., F.A., L.F., R.M., E.P., A.C.-S., C.D., N.V., J.-L.S.); Department of Cardiology, Lariboisière Hospital, Paris, France (A.C.-S.); and Inserm UMR-S 1155 and Pierre and Marie Curie University, Paris, France (C.C.)
| | - Christos Chatziantoniou
- From the Inserm UMR-S 942 and Paris Diderot University, Paris, France (H.R., A.M., M.B., F.A., L.F., R.M., E.P., A.C.-S., C.D., N.V., J.-L.S.); Department of Cardiology, Lariboisière Hospital, Paris, France (A.C.-S.); and Inserm UMR-S 1155 and Pierre and Marie Curie University, Paris, France (C.C.)
| | - Jane-Lise Samuel
- From the Inserm UMR-S 942 and Paris Diderot University, Paris, France (H.R., A.M., M.B., F.A., L.F., R.M., E.P., A.C.-S., C.D., N.V., J.-L.S.); Department of Cardiology, Lariboisière Hospital, Paris, France (A.C.-S.); and Inserm UMR-S 1155 and Pierre and Marie Curie University, Paris, France (C.C.).
| |
Collapse
|
18
|
Pippucci T, Maresca A, Magini P, Cenacchi G, Donadio V, Palombo F, Papa V, Incensi A, Gasparre G, Valentino ML, Preziuso C, Pisano A, Ragno M, Liguori R, Giordano C, Tonon C, Lodi R, Parmeggiani A, Carelli V, Seri M. Homozygous NOTCH3 null mutation and impaired NOTCH3 signaling in recessive early-onset arteriopathy and cavitating leukoencephalopathy. EMBO Mol Med 2016; 7:848-58. [PMID: 25870235 PMCID: PMC4459822 DOI: 10.15252/emmm.201404399] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Notch signaling is essential for vascular physiology. Neomorphic heterozygous mutations in NOTCH3, one of the four human NOTCH receptors, cause cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL). Hypomorphic heterozygous alleles have been occasionally described in association with a spectrum of cerebrovascular phenotypes overlapping CADASIL, but their pathogenic potential is unclear. We describe a patient with childhood-onset arteriopathy, cavitating leukoencephalopathy with cerebral white matter abnormalities presented as diffuse cavitations, multiple lacunar infarctions and disseminated microbleeds. We identified a novel homozygous c.C2898A (p.C966*) null mutation in NOTCH3 abolishing NOTCH3 expression and causing NOTCH3 signaling impairment. NOTCH3 targets acting in the regulation of arterial tone (KCNA5) or expressed in the vasculature (CDH6) were downregulated. Patient's vessels were characterized by smooth muscle degeneration as in CADASIL, but without deposition of granular osmiophilic material (GOM), the CADASIL hallmark. The heterozygous parents displayed similar but less dramatic trends in decrease in the expression of NOTCH3 and its targets, as well as in vessel degeneration. This study suggests a functional link between NOTCH3 deficiency and pathogenesis of vascular leukoencephalopathies.
Collapse
Affiliation(s)
- Tommaso Pippucci
- U.O. Genetica Medica, Policlinico Sant'Orsola-Malpighi, Bologna, Italy Dipartimento di Scienze Mediche Chirurgiche (DIMEC), University of Bologna, Bologna, Italy
| | - Alessandra Maresca
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy Unita' di Neurologia, Dipartimento di Scienze Biomediche e Neuromotorie (DIBINEM), University of Bologna, Bologna, Italy
| | - Pamela Magini
- Dipartimento di Scienze Mediche Chirurgiche (DIMEC), University of Bologna, Bologna, Italy
| | - Giovanna Cenacchi
- Unita' di Neurologia, Dipartimento di Scienze Biomediche e Neuromotorie (DIBINEM), University of Bologna, Bologna, Italy
| | - Vincenzo Donadio
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Flavia Palombo
- Dipartimento di Scienze Mediche Chirurgiche (DIMEC), University of Bologna, Bologna, Italy
| | - Valentina Papa
- Unita' di Neurologia, Dipartimento di Scienze Biomediche e Neuromotorie (DIBINEM), University of Bologna, Bologna, Italy
| | - Alex Incensi
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Giuseppe Gasparre
- Dipartimento di Scienze Mediche Chirurgiche (DIMEC), University of Bologna, Bologna, Italy
| | - Maria Lucia Valentino
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy Unita' di Neurologia, Dipartimento di Scienze Biomediche e Neuromotorie (DIBINEM), University of Bologna, Bologna, Italy
| | - Carmela Preziuso
- Dipartimento di Scienze Radiologiche, Oncologiche ed Anatomopatologiche, Sapienza, University of Rome, Rome, Italy
| | - Annalinda Pisano
- Dipartimento di Scienze Radiologiche, Oncologiche ed Anatomopatologiche, Sapienza, University of Rome, Rome, Italy
| | - Michele Ragno
- Divisione di Neurologia, Ospedale Mazzoni, Azienda Sanitaria Unica Regionale, Ascoli Piceno, Italy
| | - Rocco Liguori
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy Unita' di Neurologia, Dipartimento di Scienze Biomediche e Neuromotorie (DIBINEM), University of Bologna, Bologna, Italy
| | - Carla Giordano
- Dipartimento di Scienze Radiologiche, Oncologiche ed Anatomopatologiche, Sapienza, University of Rome, Rome, Italy
| | - Caterina Tonon
- Unita' di Neurologia, Dipartimento di Scienze Biomediche e Neuromotorie (DIBINEM), University of Bologna, Bologna, Italy Unità Risonanza Magnetica Funzionale, Policlinico S.Orsola-Malpighi, Bologna, Italy
| | - Raffaele Lodi
- Unita' di Neurologia, Dipartimento di Scienze Biomediche e Neuromotorie (DIBINEM), University of Bologna, Bologna, Italy Unità Risonanza Magnetica Funzionale, Policlinico S.Orsola-Malpighi, Bologna, Italy
| | - Antonia Parmeggiani
- Dipartimento di Scienze Mediche Chirurgiche (DIMEC), University of Bologna, Bologna, Italy U.O. Neuropsichiatria Infantile, Policlinico S.Orsola-Malpighi, Bologna, Italy
| | - Valerio Carelli
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy Unita' di Neurologia, Dipartimento di Scienze Biomediche e Neuromotorie (DIBINEM), University of Bologna, Bologna, Italy
| | - Marco Seri
- U.O. Genetica Medica, Policlinico Sant'Orsola-Malpighi, Bologna, Italy Dipartimento di Scienze Mediche Chirurgiche (DIMEC), University of Bologna, Bologna, Italy
| |
Collapse
|
19
|
Potassium channelopathy-like defect underlies early-stage cerebrovascular dysfunction in a genetic model of small vessel disease. Proc Natl Acad Sci U S A 2015; 112:E796-805. [PMID: 25646445 DOI: 10.1073/pnas.1420765112] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL), caused by dominant mutations in the NOTCH3 receptor in vascular smooth muscle, is a genetic paradigm of small vessel disease (SVD) of the brain. Recent studies using transgenic (Tg)Notch3(R169C) mice, a genetic model of CADASIL, revealed functional defects in cerebral (pial) arteries on the surface of the brain at an early stage of disease progression. Here, using parenchymal arterioles (PAs) from within the brain, we determined the molecular mechanism underlying the early functional deficits associated with this Notch3 mutation. At physiological pressure (40 mmHg), smooth muscle membrane potential depolarization and constriction to pressure (myogenic tone) were blunted in PAs from TgNotch3(R169C) mice. This effect was associated with an ∼ 60% increase in the number of voltage-gated potassium (KV) channels, which oppose pressure-induced depolarization. Inhibition of KV1 channels with 4-aminopyridine (4-AP) or treatment with the epidermal growth factor receptor agonist heparin-binding EGF (HB-EGF), which promotes KV1 channel endocytosis, reduced KV current density and restored myogenic responses in PAs from TgNotch3(R169C) mice, whereas pharmacological inhibition of other major vasodilatory influences had no effect. KV1 currents and myogenic responses were similarly altered in pial arteries from TgNotch3(R169C) mice, but not in mesenteric arteries. Interestingly, HB-EGF had no effect on mesenteric arteries, suggesting a possible mechanistic basis for the exclusive cerebrovascular manifestation of CADASIL. Collectively, our results indicate that increasing the number of KV1 channels in cerebral smooth muscle produces a mutant vascular phenotype akin to a channelopathy in a genetic model of SVD.
Collapse
|
20
|
Liu XY, Gonzalez-Toledo ME, Fagan A, Duan WM, Liu Y, Zhang S, Li B, Piao CS, Nelson L, Zhao LR. Stem cell factor and granulocyte colony-stimulating factor exhibit therapeutic effects in a mouse model of CADASIL. Neurobiol Dis 2015; 73:189-203. [DOI: 10.1016/j.nbd.2014.09.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Revised: 08/27/2014] [Accepted: 09/12/2014] [Indexed: 11/28/2022] Open
|
21
|
Joutel A. The NOTCH3ECDcascade hypothesis of cerebral autosomal-dominant arteriopathy with subcortical infarcts and leukoencephalopathy disease. ACTA ACUST UNITED AC 2014. [DOI: 10.1111/ncn3.135] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Anne Joutel
- INSERM; U1161; Paris F-75010 France
- Univ Paris Diderot; Sorbonne Cité; UMR-S1161; Paris F-75010 France
| |
Collapse
|
22
|
Tikka S, Baumann M, Siitonen M, Pasanen P, Pöyhönen M, Myllykangas L, Viitanen M, Fukutake T, Cognat E, Joutel A, Kalimo H. CADASIL and CARASIL. Brain Pathol 2014; 24:525-44. [PMID: 25323668 PMCID: PMC8029192 DOI: 10.1111/bpa.12181] [Citation(s) in RCA: 128] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 07/28/2014] [Indexed: 12/31/2022] Open
Abstract
CADASIL and CARASIL are hereditary small vessel diseases leading to vascular dementia. CADASIL commonly begins with migraine followed by minor strokes in mid-adulthood. Dominantly inherited CADASIL is caused by mutations (n > 230) in NOTCH3 gene, which encodes Notch3 receptor expressed in vascular smooth muscle cells (VSMC). Notch3 extracellular domain (N3ECD) accumulates in arterial walls followed by VSMC degeneration and subsequent fibrosis and stenosis of arterioles, predominantly in cerebral white matter, where characteristic ischemic MRI changes and lacunar infarcts emerge. The likely pathogenesis of CADASIL is toxic gain of function related to mutation-induced unpaired cysteine in N3ECD. Definite diagnosis is made by molecular genetics but is also possible by electron microscopic demonstration of pathognomonic granular osmiophilic material at VSMCs or by positive immunohistochemistry for N3ECD in dermal arteries. In rare, recessively inherited CARASIL the clinical picture and white matter changes are similar as in CADASIL, but cognitive decline begins earlier. In addition, gait disturbance, low back pain and alopecia are characteristic features. CARASIL is caused by mutations (presently n = 10) in high-temperature requirement. A serine peptidase 1 (HTRA1) gene, which result in reduced function of HTRA1 as repressor of transforming growth factor-β (TGF β) -signaling. Cerebral arteries show loss of VSMCs and marked hyalinosis, but not stenosis.
Collapse
Affiliation(s)
- Saara Tikka
- Protein Chemistry Unit, Institute of Biomedicine/AnatomyUniversity of HelsinkiHelsinkiFinland
| | - Marc Baumann
- Protein Chemistry Unit, Institute of Biomedicine/AnatomyUniversity of HelsinkiHelsinkiFinland
| | - Maija Siitonen
- Department of Medical Biochemistry and Genetics, Institute of BiomedicineUniversity of TurkuTurkuFinland
| | - Petra Pasanen
- Department of Medical Biochemistry and Genetics, Institute of BiomedicineUniversity of TurkuTurkuFinland
| | - Minna Pöyhönen
- Department of Clinical GeneticsHelsinki University Hospital, HUSLABHelsinkiFinland
| | - Liisa Myllykangas
- Department of PathologyHaartman InstituteUniversity of HelsinkiHelsinkiFinland
| | - Matti Viitanen
- Turku City HospitalTurkuFinland
- Division of Clinical GeriatricsDepartment of NeurobiologyCare Sciences and SocietyKarolinska InstitutetStockholmSweden
| | - Toshio Fukutake
- Department of NeurologyKameda Medical CenterKamogawaChibaJapan
| | - Emmanuel Cognat
- INSERMU1161ParisFrance
- Université Paris DiderotSorbonne Paris CitéUMRS 1161ParisFrance
| | - Anne Joutel
- INSERMU1161ParisFrance
- Université Paris DiderotSorbonne Paris CitéUMRS 1161ParisFrance
| | - Hannu Kalimo
- Department of PathologyHaartman InstituteUniversity of HelsinkiHelsinkiFinland
- Institute of BiomedicineDepartment of Forensic MedicineUniversity of TurkuTurkuFinland
| |
Collapse
|
23
|
Arboleda-Velasquez JF, Primo V, Graham M, James A, Manent J, D'Amore PA. Notch signaling functions in retinal pericyte survival. Invest Ophthalmol Vis Sci 2014; 55:5191-9. [PMID: 25015359 DOI: 10.1167/iovs.14-14046] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE Pericytes, the vascular cells that constitute the outer layer of capillaries, have been shown to have a crucial role in vascular development and stability. Loss of pericytes precedes endothelial cell dysfunction and vascular degeneration in small-vessel diseases, including diabetic retinopathy. Despite their clinical relevance, the cellular pathways controlling survival of retinal pericytes remain largely uncharacterized. Therefore, we investigated the role of Notch signaling, a master regulator of cell fate decisions, in retinal pericyte survival. METHODS A coculture system of ligand-dependent Notch signaling was developed using primary cultured retinal pericytes and a mesenchymal cell line derived from an inducible mouse model expressing the Delta-like 1 Notch ligand. This model was used to examine the effect of Notch activity on pericyte survival using quantitative PCR (qPCR) and a light-induced cell death assay. The effect of Notch gain- and loss-of-function was analyzed in monocultures of retinal pericytes using antibody arrays to interrogate the expression of apoptosis-related proteins. RESULTS Primary cultured retinal pericytes differentially expressed key molecules of the Notch pathway and displayed strong expression of canonical Notch/RBPJK (recombination signal-binding protein 1 for J-kappa) downstream targets. A gene expression screen using gain- and loss-of-function approaches identified genes relevant to cell survival as downstream targets of Notch activity in retinal pericytes. Ligand-mediated Notch activity protected retinal pericytes from light-induced cell death. CONCLUSIONS Our results have identified signature genes downstream of Notch activity in retinal pericytes and suggest that tight regulation of Notch signaling is crucial for pericyte survival.
Collapse
Affiliation(s)
- Joseph F Arboleda-Velasquez
- Schepens Eye Research Institute, Mass Eye and Ear Infirmary and Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States
| | - Vincent Primo
- Schepens Eye Research Institute, Mass Eye and Ear Infirmary and Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States
| | - Mark Graham
- Schepens Eye Research Institute, Mass Eye and Ear Infirmary and Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States University of Exeter Medical School, Exeter, Devon, United Kingdom
| | - Alexandra James
- Schepens Eye Research Institute, Mass Eye and Ear Infirmary and Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States University of Exeter Medical School, Exeter, Devon, United Kingdom
| | - Jan Manent
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, United States
| | - Patricia A D'Amore
- Schepens Eye Research Institute, Mass Eye and Ear Infirmary and Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States Department of Pathology, Harvard Medical School, Boston, Massachusetts, United States
| |
Collapse
|
24
|
Inhibition of Notch signaling facilitates the differentiation of human-induced pluripotent stem cells into neural stem cells. Mol Cell Biochem 2014; 395:291-8. [PMID: 24972705 DOI: 10.1007/s11010-014-2130-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Accepted: 06/17/2014] [Indexed: 12/19/2022]
Abstract
Neural stem cells (NSCs) derived from induced pluripotent stem cells (iPSCs) are becoming an appealing source of cell-based therapies of brain diseases. As such, it is important to understand the molecular mechanisms that regulate the differentiation of iPSCs toward NSCs. It is well known that Notch signaling governs the retention of stem cell features and drives stem cells fate. However, further studies are required to investigate the role of Notch signaling in the NSCs differentiation of iPSCs. In this study, we successfully generated NSCs from human iPSCs using serum-free medium supplemented with retinoic acid (RA) in vitro. We then assessed changes in the expression of Notch signaling-related molecules and some miRNAs (9, 34a, 200b), which exert their regulation by targeting Notch signaling. Moreover, we used a γ-secretase inhibitor (DAPT) to disturb Notch signaling. Data revealed that the levels of the Notch signaling-related molecules decreased, whereas those miRNAs increased, during this differentiation process. Inhibition of Notch signaling accelerated the formation of the neural rosette structures and the expression of NSC and mature neurocyte marker genes. This suggests that Notch signaling negatively regulated the neuralization of human iPSCs, and that this process may be regulated by some miRNAs.
Collapse
|
25
|
Cognat E, Baron-Menguy C, Domenga-Denier V, Cleophax S, Fouillade C, Monet-Leprêtre M, Dewerchin M, Joutel A. Archetypal Arg169Cys Mutation in NOTCH3 Does Not Drive the Pathogenesis in Cerebral Autosomal Dominant Arteriopathy With Subcortical Infarcts and Leucoencephalopathy via a Loss-of-Function Mechanism. Stroke 2014; 45:842-9. [DOI: 10.1161/strokeaha.113.003339] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Emmanuel Cognat
- From the INSERM, U740 and Univ Paris Diderot, Sorbonne Cité, UMR S740, Paris, France (E.C., C.B.-M., V.D.-D., S.C., C.F., M.M.-L., A.J.); and Laboratory of Angiogenesis & Neurovascular Link, Vesalius Research Center, VIB–KU Leuven, Leuven, Belgium (M.D.)
| | - Céline Baron-Menguy
- From the INSERM, U740 and Univ Paris Diderot, Sorbonne Cité, UMR S740, Paris, France (E.C., C.B.-M., V.D.-D., S.C., C.F., M.M.-L., A.J.); and Laboratory of Angiogenesis & Neurovascular Link, Vesalius Research Center, VIB–KU Leuven, Leuven, Belgium (M.D.)
| | - Valérie Domenga-Denier
- From the INSERM, U740 and Univ Paris Diderot, Sorbonne Cité, UMR S740, Paris, France (E.C., C.B.-M., V.D.-D., S.C., C.F., M.M.-L., A.J.); and Laboratory of Angiogenesis & Neurovascular Link, Vesalius Research Center, VIB–KU Leuven, Leuven, Belgium (M.D.)
| | - Sabine Cleophax
- From the INSERM, U740 and Univ Paris Diderot, Sorbonne Cité, UMR S740, Paris, France (E.C., C.B.-M., V.D.-D., S.C., C.F., M.M.-L., A.J.); and Laboratory of Angiogenesis & Neurovascular Link, Vesalius Research Center, VIB–KU Leuven, Leuven, Belgium (M.D.)
| | - Charles Fouillade
- From the INSERM, U740 and Univ Paris Diderot, Sorbonne Cité, UMR S740, Paris, France (E.C., C.B.-M., V.D.-D., S.C., C.F., M.M.-L., A.J.); and Laboratory of Angiogenesis & Neurovascular Link, Vesalius Research Center, VIB–KU Leuven, Leuven, Belgium (M.D.)
| | - Marie Monet-Leprêtre
- From the INSERM, U740 and Univ Paris Diderot, Sorbonne Cité, UMR S740, Paris, France (E.C., C.B.-M., V.D.-D., S.C., C.F., M.M.-L., A.J.); and Laboratory of Angiogenesis & Neurovascular Link, Vesalius Research Center, VIB–KU Leuven, Leuven, Belgium (M.D.)
| | - Mieke Dewerchin
- From the INSERM, U740 and Univ Paris Diderot, Sorbonne Cité, UMR S740, Paris, France (E.C., C.B.-M., V.D.-D., S.C., C.F., M.M.-L., A.J.); and Laboratory of Angiogenesis & Neurovascular Link, Vesalius Research Center, VIB–KU Leuven, Leuven, Belgium (M.D.)
| | - Anne Joutel
- From the INSERM, U740 and Univ Paris Diderot, Sorbonne Cité, UMR S740, Paris, France (E.C., C.B.-M., V.D.-D., S.C., C.F., M.M.-L., A.J.); and Laboratory of Angiogenesis & Neurovascular Link, Vesalius Research Center, VIB–KU Leuven, Leuven, Belgium (M.D.)
| |
Collapse
|