1
|
Papageorgiou L, Papa L, Papakonstantinou E, Mataragka A, Dragoumani K, Chaniotis D, Beloukas A, Iliopoulos C, Bongcam-Rudloff E, Chrousos GP, Kossida S, Eliopoulos E, Vlachakis D. SNP and Structural Study of the Notch Superfamily Provides Insights and Novel Pharmacological Targets against the CADASIL Syndrome and Neurodegenerative Diseases. Genes (Basel) 2024; 15:529. [PMID: 38790158 PMCID: PMC11120892 DOI: 10.3390/genes15050529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 04/17/2024] [Accepted: 04/22/2024] [Indexed: 05/26/2024] Open
Abstract
The evolutionary conserved Notch signaling pathway functions as a mediator of direct cell-cell communication between neighboring cells during development. Notch plays a crucial role in various fundamental biological processes in a wide range of tissues. Accordingly, the aberrant signaling of this pathway underlies multiple genetic pathologies such as developmental syndromes, congenital disorders, neurodegenerative diseases, and cancer. Over the last two decades, significant data have shown that the Notch signaling pathway displays a significant function in the mature brains of vertebrates and invertebrates beyond neuronal development and specification during embryonic development. Neuronal connection, synaptic plasticity, learning, and memory appear to be regulated by this pathway. Specific mutations in human Notch family proteins have been linked to several neurodegenerative diseases including Alzheimer's disease, CADASIL, and ischemic injury. Neurodegenerative diseases are incurable disorders of the central nervous system that cause the progressive degeneration and/or death of brain nerve cells, affecting both mental function and movement (ataxia). There is currently a lot of study being conducted to better understand the molecular mechanisms by which Notch plays an essential role in the mature brain. In this study, an in silico analysis of polymorphisms and mutations in human Notch family members that lead to neurodegenerative diseases was performed in order to investigate the correlations among Notch family proteins and neurodegenerative diseases. Particular emphasis was placed on the study of mutations in the Notch3 protein and the structure analysis of the mutant Notch3 protein that leads to the manifestation of the CADASIL syndrome in order to spot possible conserved mutations and interpret the effect of these mutations in the Notch3 protein structure. Conserved mutations of cysteine residues may be candidate pharmacological targets for the potential therapy of CADASIL syndrome.
Collapse
Affiliation(s)
- Louis Papageorgiou
- Laboratory of Genetics, Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, 75 Iera Odos, 11855 Athens, Greece; (L.P.); (L.P.); (E.P.); (A.M.); (K.D.); (E.E.)
- Department of Biomedical Sciences, School of Health and Care Sciences, University of West Attica, Agioy Spyridonos, 12243 Egaleo, Greece; (D.C.); (A.B.)
| | - Lefteria Papa
- Laboratory of Genetics, Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, 75 Iera Odos, 11855 Athens, Greece; (L.P.); (L.P.); (E.P.); (A.M.); (K.D.); (E.E.)
| | - Eleni Papakonstantinou
- Laboratory of Genetics, Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, 75 Iera Odos, 11855 Athens, Greece; (L.P.); (L.P.); (E.P.); (A.M.); (K.D.); (E.E.)
- University Research Institute of Maternal and Child Health & Precision Medicine, National and Kapodistrian University of Athens, “Aghia Sophia” Children’s Hospital, 11527 Athens, Greece;
| | - Antonia Mataragka
- Laboratory of Genetics, Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, 75 Iera Odos, 11855 Athens, Greece; (L.P.); (L.P.); (E.P.); (A.M.); (K.D.); (E.E.)
| | - Konstantina Dragoumani
- Laboratory of Genetics, Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, 75 Iera Odos, 11855 Athens, Greece; (L.P.); (L.P.); (E.P.); (A.M.); (K.D.); (E.E.)
| | - Dimitrios Chaniotis
- Department of Biomedical Sciences, School of Health and Care Sciences, University of West Attica, Agioy Spyridonos, 12243 Egaleo, Greece; (D.C.); (A.B.)
| | - Apostolos Beloukas
- Department of Biomedical Sciences, School of Health and Care Sciences, University of West Attica, Agioy Spyridonos, 12243 Egaleo, Greece; (D.C.); (A.B.)
| | - Costas Iliopoulos
- School of Informatics, Faculty of Natural & Mathematical Sciences, King’s College London, Bush House, Strand, London WC2R 2LS, UK;
| | - Erik Bongcam-Rudloff
- Department of Animal Biosciences, Swedish University of Agricultural Sciences, 756 51 Uppsala, Sweden;
| | - George P. Chrousos
- University Research Institute of Maternal and Child Health & Precision Medicine, National and Kapodistrian University of Athens, “Aghia Sophia” Children’s Hospital, 11527 Athens, Greece;
| | - Sofia Kossida
- IMGT, The International ImMunoGenetics Information System, Laboratoire d’ImmunoGénétique Moléculaire LIGM, Institut de Génétique Humaine, (IGH), Centre National de la Recherche Scientifique (CNRS), Université de Montpellier (UM), 34000 Montpellier, France;
| | - Elias Eliopoulos
- Laboratory of Genetics, Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, 75 Iera Odos, 11855 Athens, Greece; (L.P.); (L.P.); (E.P.); (A.M.); (K.D.); (E.E.)
| | - Dimitrios Vlachakis
- Laboratory of Genetics, Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, 75 Iera Odos, 11855 Athens, Greece; (L.P.); (L.P.); (E.P.); (A.M.); (K.D.); (E.E.)
- University Research Institute of Maternal and Child Health & Precision Medicine, National and Kapodistrian University of Athens, “Aghia Sophia” Children’s Hospital, 11527 Athens, Greece;
- School of Informatics, Faculty of Natural & Mathematical Sciences, King’s College London, Bush House, Strand, London WC2R 2LS, UK;
| |
Collapse
|
2
|
Cai J, Qiao Y, Chen L, Lu Y, Zheng D. Regulation of the Notch signaling pathway by natural products for cancer therapy. J Nutr Biochem 2024; 123:109483. [PMID: 37848105 DOI: 10.1016/j.jnutbio.2023.109483] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 09/13/2023] [Accepted: 10/11/2023] [Indexed: 10/19/2023]
Abstract
The Notch signaling pathway is an evolutionarily conserved pathway that modulates normal biological processes involved in cellular differentiation, apoptosis, and stem cell self-renewal in a context-dependent fashion. Attributed to its pleiotropic physiological roles, both overexpression and silencing of the pathway are associated with the emergence, progression, and poorer prognosis in various types of cancer. To decrease disease incidence and promote survival, targeting Notch may have chemopreventive and anti-cancer effects. Natural products with profound historical origins have distinguished themselves from other therapies due to their easy access, high biological compatibility, low toxicity, and reliable effects at specific physiological sites in vivo. This review describes the Notch signaling pathway, particularly its normal activation process, and some main illnesses related to Notch signaling pathway dysregulation. Emphasis is placed on the effects and mechanisms of natural products targeting the Notch signaling pathway in diverse cancer types, including curcumin, ellagic acid (EA), resveratrol, genistein, epigallocatechin-3-gallate (EGCG), quercetin, and xanthohumol and so on. Existing evidence indicates that natural products are feasible solution to fight against cancer by targeting Notch signaling, either alone or in combination with current therapeutic agents.
Collapse
Affiliation(s)
- Jiayi Cai
- School of Stomatology, Fujian Medical University, Fuzhou 350122, China
| | - Yajie Qiao
- School of Stomatology, Fujian Medical University, Fuzhou 350122, China
| | - Lingbin Chen
- School of Stomatology, Fujian Medical University, Fuzhou 350122, China
| | - Youguang Lu
- Fujian Key Laboratory of Oral Diseases, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350004, China; Department of Preventive Dentistry, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350001, China
| | - Dali Zheng
- Fujian Key Laboratory of Oral Diseases, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350004, China.
| |
Collapse
|
3
|
Chen Y, Ren P, He X, Yan F, Gu R, Bai J, Zhang X. Olfactory bulb neurogenesis depending on signaling in the subventricular zone. Cereb Cortex 2023; 33:11102-11111. [PMID: 37746807 DOI: 10.1093/cercor/bhad349] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 09/04/2023] [Accepted: 09/06/2023] [Indexed: 09/26/2023] Open
Abstract
Olfaction is a crucial sense that is essential for the well-being and survival of individuals. Olfactory bulb (OB) is the first olfactory relay station, and its function depends on newly generated neurons from the subventricular zone (SVZ). These newly born neurons constantly migrate through the rostral migratory stream to integrate into existing neural networks within the OB, thereby contributing to olfactory information processing. However, the mechanisms underlying the contribution of SVZ adult neurogenesis to OB neurogenesis remain largely elusive. Adult neurogenesis is a finely regulated multistep process involving the proliferation of adult neural stem cells (aNSCs) and neural precursor cells, as well as the migration and differentiation of neuroblasts, and integration of newly generated neurons into preexisting neuronal circuitries. Recently, extensive studies have explored the mechanism of SVZ and OB neurogenesis. This review focused on elucidating various molecules and signaling pathways associated with OB neurogenesis dependent on the SVZ function. A better understanding of the mechanisms underlying the OB neurogenesis on the adult brain is an attractive prospect to induce aNSCs in SVZ to generate new neurons to ameliorate olfactory dysfunction that is involved in various diseases. It will also contribute to developing new strategies for the human aNSCs-based therapies.
Collapse
Affiliation(s)
- Yali Chen
- Laboratory of Molecular Neurobiology, Medical Faculty, Kunming University of Science and Technology, Kunming 650500, China
| | - Peng Ren
- Laboratory of Molecular Neurobiology, Medical Faculty, Kunming University of Science and Technology, Kunming 650500, China
| | - Xiongjie He
- Laboratory of Molecular Neurobiology, Medical Faculty, Kunming University of Science and Technology, Kunming 650500, China
| | - Fang Yan
- Laboratory of Molecular Neurobiology, Medical Faculty, Kunming University of Science and Technology, Kunming 650500, China
| | - Rou Gu
- Laboratory of Molecular Neurobiology, Medical Faculty, Kunming University of Science and Technology, Kunming 650500, China
| | - Jie Bai
- Laboratory of Molecular Neurobiology, Medical Faculty, Kunming University of Science and Technology, Kunming 650500, China
| | - Xianwen Zhang
- Laboratory of Molecular Neurobiology, Medical Faculty, Kunming University of Science and Technology, Kunming 650500, China
| |
Collapse
|
4
|
Vogt M, Unnikrishnan MK, Heinig N, Schumann U, Schmidt MHH, Barth K. c-Cbl Regulates Murine Subventricular Zone-Derived Neural Progenitor Cells in Dependence of the Epidermal Growth Factor Receptor. Cells 2023; 12:2400. [PMID: 37830613 PMCID: PMC10572332 DOI: 10.3390/cells12192400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 09/15/2023] [Accepted: 09/26/2023] [Indexed: 10/14/2023] Open
Abstract
The localization, expression, and physiological role of regulatory proteins in the neurogenic niches of the brain is fundamental to our understanding of adult neurogenesis. This study explores the expression and role of the E3-ubiquitin ligase, c-Cbl, in neurogenesis within the subventricular zone (SVZ) of mice. In vitro neurosphere assays and in vivo analyses were performed in specific c-Cbl knock-out lines to unravel c-Cbl's role in receptor tyrosine kinase signaling, including the epidermal growth factor receptor (EGFR) pathway. Our findings suggest that c-Cbl is significantly expressed within EGFR-expressing cells, playing a pivotal role in neural stem cell proliferation and differentiation. However, c-Cbl's function extends beyond EGFR signaling, as its loss upon knock-out stimulated progenitor cell proliferation in neurosphere cultures. Yet, this effect was not detected in hippocampal progenitor cells, reflecting the lack of the EGFR in the hippocampus. In vivo, c-Cbl exerted only a minor proneurogenic influence with no measurable impact on the formation of adult-born neurons. In conclusion, c-Cbl regulates neural stem cells in the subventricular zone via the EGFR pathway but, likely, its loss is compensated by other signaling modules in vivo.
Collapse
|
5
|
Lampada A, Taylor V. Notch signaling as a master regulator of adult neurogenesis. Front Neurosci 2023; 17:1179011. [PMID: 37457009 PMCID: PMC10339389 DOI: 10.3389/fnins.2023.1179011] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 06/05/2023] [Indexed: 07/18/2023] Open
Abstract
Neurogenesis ceases in most regions of the mammalian brain before or shortly after birth, however, in a few restricted brain regions, the production of new neurons proceeds into adulthood. Neural stem cells (NSCs) in these neurogenic zones are integrated into niches that control their activity and fate. Most stem cells in the adult brain are mitotically inactive and these cells can remain quiescent for months or even years. One of the key questions is what are the molecular mechanisms that regulate NSC maintenance and differentiation. Notch signaling has been shown to be a critical regulator of stem cell activity and maintenance in many tissues including in the nervous system. In this mini-review we discuss the roles of Notch signaling and the functions of the different Notch receptors and ligands in regulating neurogenesis in the adult murine brain. We review the functions of Notch signaling components in controlling NSC quiescence and entry into cell cycle and neurogenesis.
Collapse
|
6
|
Maeda Y, Isomura A, Masaki T, Kageyama R. Differential cell-cycle control by oscillatory versus sustained Hes1 expression via p21. Cell Rep 2023; 42:112520. [PMID: 37200191 DOI: 10.1016/j.celrep.2023.112520] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 01/06/2023] [Accepted: 05/02/2023] [Indexed: 05/20/2023] Open
Abstract
Oscillatory Hes1 expression activates cell proliferation, while high and sustained Hes1 expression induces quiescence, but the mechanism by which Hes1 differentially controls cell proliferation depending on its expression dynamics is unclear. Here, we show that oscillatory Hes1 expression down-regulates the expression of the cyclin-dependent kinase inhibitor p21 (Cdkn1a), which delays cell-cycle progression, and thereby activates the proliferation of mouse neural stem cells (NSCs). By contrast, sustained Hes1 overexpression up-regulates p21 expression and inhibits NSC proliferation, although it initially down-regulates p21 expression. Compared with Hes1 oscillation, sustained Hes1 overexpression represses Dusp7, a phosphatase for phosphorylated Erk (p-Erk), and increases the levels of p-Erk, which can up-regulate p21 expression. These results indicate that p21 expression is directly repressed by oscillatory Hes1 expression, but indirectly up-regulated by sustained Hes1 overexpression, suggesting that depending on its expression dynamics, Hes1 differentially controls NSC proliferation via p21.
Collapse
Affiliation(s)
- Yuki Maeda
- RIKEN Center for Brain Science, Wako 351-0198, Japan; Graduate School of Biostudies, Kyoto University, Kyoto 606-8501, Japan
| | - Akihiro Isomura
- Institute for Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan; Institute for Integrated Cell-Material Sciences, Kyoto University, Kyoto 606-8501, Japan; Japan Science and Technology Agency, PRESTO, Saitama 332-0012, Japan
| | - Taimu Masaki
- RIKEN Center for Brain Science, Wako 351-0198, Japan
| | - Ryoichiro Kageyama
- RIKEN Center for Brain Science, Wako 351-0198, Japan; Graduate School of Biostudies, Kyoto University, Kyoto 606-8501, Japan; Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan.
| |
Collapse
|
7
|
Li L, Li X, Han R, Wu M, Ma Y, Chen Y, Zhang H, Li Y. Therapeutic Potential of Chinese Medicine for Endogenous Neurogenesis: A Promising Candidate for Stroke Treatment. Pharmaceuticals (Basel) 2023; 16:ph16050706. [PMID: 37242489 DOI: 10.3390/ph16050706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 04/26/2023] [Accepted: 05/03/2023] [Indexed: 05/28/2023] Open
Abstract
Strokes are a leading cause of morbidity and mortality in adults worldwide. Extensive preclinical studies have shown that neural-stem-cell-based treatments have great therapeutic potential for stroke. Several studies have confirmed that the effective components of traditional Chinese medicine can protect and maintain the survival, proliferation, and differentiation of endogenous neural stem cells through different targets and mechanisms. Therefore, the use of Chinese medicines to activate and promote endogenous nerve regeneration and repair is a potential treatment option for stroke patients. Here, we summarize the current knowledge regarding neural stem cell strategies for ischemic strokes and the potential effects of these Chinese medicines on neuronal regeneration.
Collapse
Affiliation(s)
- Lin Li
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Ministry of Education, Tianjin 301617, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xiao Li
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Ministry of Education, Tianjin 301617, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Rui Han
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Ministry of Education, Tianjin 301617, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Meirong Wu
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Ministry of Education, Tianjin 301617, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yaolei Ma
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Ministry of Education, Tianjin 301617, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yuzhao Chen
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Ministry of Education, Tianjin 301617, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Han Zhang
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Ministry of Education, Tianjin 301617, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yue Li
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Ministry of Education, Tianjin 301617, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| |
Collapse
|
8
|
Molecular Mechanisms Involved in the Regulation of Neurodevelopment by miR-124. Mol Neurobiol 2023; 60:3569-3583. [PMID: 36840845 DOI: 10.1007/s12035-023-03271-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 02/04/2023] [Indexed: 02/26/2023]
Abstract
miR-124 is a miRNA predominantly expressed in the nervous system and accounts for more than a quarter of the total miRNAs in the brain. It regulates neurogenesis, neuronal differentiation, neuronal maturation, and synapse formation and is the most important miRNA in the brain. Furthermore, emerging evidence has suggested miR-124 may be associated with the pathogenesis of various neurodevelopmental and neuropsychiatric disorders. Here, we provide an overview of the role of miR-124 in neurodevelopment and the underling mechanisms, and finally, we prospect the significance of miR-124 research to the field of neuroscience.
Collapse
|
9
|
Ghafil FA, Majeed SA, Qassam H, Mardan HW, Hadi NR. NEPHROPROTECTIVE EFFECT OF GAMMA-SECRETASE INHIBITOR ON SEPSIS- INDUCED RENAL INJURY IN MOUSE MODEL OF CLP. WIADOMOSCI LEKARSKIE (WARSAW, POLAND : 1960) 2023; 76:122-130. [PMID: 36883500 DOI: 10.36740/wlek202301117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/09/2023]
Abstract
OBJECTIVE The aim: This study was set out to assess the potential protective impact of MK0752 (a gamma secretase inhibitor) on sepsis-induced renal injury through modulation of inflammatory and oxidative stress pathways. PATIENTS AND METHODS Materials and methods: Twenty-four Swiss-albino mice aged between eight and twelve week and weighted twenty to thirty-seven grams were randomly allocated into four groups (n=6 in each group). Sham group (laparotomy without cecal ligation and puncture (CLP), sepsis group (laparotomy with CLP), vehicle-treated group (equivalent volume of DMSO before the CLP), MK0752 treated group (5 mg/kg) single daily dose for three days before the CLP. Blood samples were used to assess the serum levels of urea and creatinine. The kidneys were used to assess tissue levels of the TNF-α, IL-10, IL-6, TNFR1, VEGF, notch1, jagged1 and tissue damage by histopathological analysis. RESULTS Results: The current study shows that pretreatment with MK0752 ameliorates the renal damage by significantly reducing the proinflammatory cytokines and notch1 signaling. CONCLUSION Conclusions: Taken together, these results suggest that MK0752 could be protective against the renal injury induced by sepsis through its ameliorative impact on renal architecture and modulating cytokines and Notch1 singling pathway. Further studies regarding the role of Notch signaling pathways would be worthwhile.
Collapse
Affiliation(s)
- Fadha Abdulameer Ghafil
- DEPARTMENT OF PHARMACOLOGY AND THERAPEUTICS, FACULTY OF MEDICINE, UNIVERSITY OF KUFA, NAJAF, IRAQ
| | - Sahar A Majeed
- DEPARTMENT OF PHARMACOLOGY AND THERAPEUTICS, FACULTY OF MEDICINE, UNIVERSITY OF KUFA, NAJAF, IRAQ
| | - Heider Qassam
- DEPARTMENT OF PHARMACOLOGY AND THERAPEUTICS, FACULTY OF MEDICINE, UNIVERSITY OF KUFA, NAJAF, IRAQ
| | - Haider W Mardan
- MIDDLE EUPHRATES CENTER OF NEUROSCIENCES, AL-SADDER TEACHING HOSPITAL, NAJAF, IRAQ
| | - Najah R Hadi
- MIDDLE EUPHRATES CENTER OF NEUROSCIENCES, AL-SADDER TEACHING HOSPITAL, NAJAF, IRAQ
| |
Collapse
|
10
|
Souilhol C, Tardajos Ayllon B, Li X, Diagbouga MR, Zhou Z, Canham L, Roddie H, Pirri D, Chambers EV, Dunning MJ, Ariaans M, Li J, Fang Y, Jørgensen HF, Simons M, Krams R, Waltenberger J, Fragiadaki M, Ridger V, De Val S, Francis SE, Chico TJA, Serbanovic-Canic J, Evans PC. JAG1-NOTCH4 mechanosensing drives atherosclerosis. SCIENCE ADVANCES 2022; 8:eabo7958. [PMID: 36044575 PMCID: PMC9432841 DOI: 10.1126/sciadv.abo7958] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 07/15/2022] [Indexed: 06/15/2023]
Abstract
Endothelial cell (EC) sensing of disturbed blood flow triggers atherosclerosis, a disease of arteries that causes heart attack and stroke, through poorly defined mechanisms. The Notch pathway plays a central role in blood vessel growth and homeostasis, but its potential role in sensing of disturbed flow has not been previously studied. Here, we show using porcine and murine arteries and cultured human coronary artery EC that disturbed flow activates the JAG1-NOTCH4 signaling pathway. Light-sheet imaging revealed enrichment of JAG1 and NOTCH4 in EC of atherosclerotic plaques, and EC-specific genetic deletion of Jag1 (Jag1ECKO) demonstrated that Jag1 promotes atherosclerosis at sites of disturbed flow. Mechanistically, single-cell RNA sequencing in Jag1ECKO mice demonstrated that Jag1 suppresses subsets of ECs that proliferate and migrate. We conclude that JAG1-NOTCH4 sensing of disturbed flow enhances atherosclerosis susceptibility by regulating EC heterogeneity and that therapeutic targeting of this pathway may treat atherosclerosis.
Collapse
Affiliation(s)
- Celine Souilhol
- Department of Infection, Immunity and Cardiovascular Disease, INSIGNEO Institute for In Silico Medicine, and the Bateson Centre, University of Sheffield, Sheffield, UK
- Biomolecular Sciences Research Centre, Sheffield Hallam University, Sheffield, UK
| | - Blanca Tardajos Ayllon
- Department of Infection, Immunity and Cardiovascular Disease, INSIGNEO Institute for In Silico Medicine, and the Bateson Centre, University of Sheffield, Sheffield, UK
| | - Xiuying Li
- School of Pharmacy, Southwest Medical University, LuZhou, Sichuan 646000, P.R. China
| | - Mannekomba R. Diagbouga
- Department of Infection, Immunity and Cardiovascular Disease, INSIGNEO Institute for In Silico Medicine, and the Bateson Centre, University of Sheffield, Sheffield, UK
| | - Ziqi Zhou
- Department of Infection, Immunity and Cardiovascular Disease, INSIGNEO Institute for In Silico Medicine, and the Bateson Centre, University of Sheffield, Sheffield, UK
| | - Lindsay Canham
- Department of Infection, Immunity and Cardiovascular Disease, INSIGNEO Institute for In Silico Medicine, and the Bateson Centre, University of Sheffield, Sheffield, UK
| | - Hannah Roddie
- Department of Infection, Immunity and Cardiovascular Disease, INSIGNEO Institute for In Silico Medicine, and the Bateson Centre, University of Sheffield, Sheffield, UK
| | - Daniela Pirri
- Department of Infection, Immunity and Cardiovascular Disease, INSIGNEO Institute for In Silico Medicine, and the Bateson Centre, University of Sheffield, Sheffield, UK
| | - Emily V. Chambers
- Sheffield Bioinformatics Core, Sheffield Institute of Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Mark J. Dunning
- Sheffield Bioinformatics Core, Sheffield Institute of Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Mark Ariaans
- Department of Infection, Immunity and Cardiovascular Disease, INSIGNEO Institute for In Silico Medicine, and the Bateson Centre, University of Sheffield, Sheffield, UK
| | - Jin Li
- Biological Sciences Division, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Yun Fang
- Biological Sciences Division, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Helle F. Jørgensen
- Division of Cardiovascular Medicine, University of Cambridge, Addenbrooke’s Centre for Clinical Investigation, Addenbrooke’s Hospital, Cambridge, UK
| | - Michael Simons
- Department of Internal Medicine, Yale Cardiovascular Research Center, New Haven, CT, USA
| | - Rob Krams
- Department of Bioengineering, Queen Mary University of London, London, UK
| | - Johannes Waltenberger
- Department of Cardiovascular Medicine, Medical Faculty, University of Münster, Münster, Germany
- Hirslanden Klinik im Park, Cardiovascular Medicine, Diagnostic and Therapeutic Heart Center AG, 8002 Zürich, Switzerland
| | - Maria Fragiadaki
- Department of Infection, Immunity and Cardiovascular Disease, INSIGNEO Institute for In Silico Medicine, and the Bateson Centre, University of Sheffield, Sheffield, UK
| | - Victoria Ridger
- Department of Infection, Immunity and Cardiovascular Disease, INSIGNEO Institute for In Silico Medicine, and the Bateson Centre, University of Sheffield, Sheffield, UK
| | - Sarah De Val
- BHF Centre of Regenerative Medicine, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
- Ludwig Institute for Cancer Research Ltd, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7DQ, UK
| | - Sheila E. Francis
- Department of Infection, Immunity and Cardiovascular Disease, INSIGNEO Institute for In Silico Medicine, and the Bateson Centre, University of Sheffield, Sheffield, UK
| | - Timothy JA Chico
- Department of Infection, Immunity and Cardiovascular Disease, INSIGNEO Institute for In Silico Medicine, and the Bateson Centre, University of Sheffield, Sheffield, UK
| | - Jovana Serbanovic-Canic
- Department of Infection, Immunity and Cardiovascular Disease, INSIGNEO Institute for In Silico Medicine, and the Bateson Centre, University of Sheffield, Sheffield, UK
| | - Paul C. Evans
- Department of Infection, Immunity and Cardiovascular Disease, INSIGNEO Institute for In Silico Medicine, and the Bateson Centre, University of Sheffield, Sheffield, UK
| |
Collapse
|
11
|
Endogenous Neural Stem Cell Mediated Oligodendrogenesis in the Adult Mammalian Brain. Cells 2022; 11:cells11132101. [PMID: 35805185 PMCID: PMC9265817 DOI: 10.3390/cells11132101] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 06/28/2022] [Accepted: 06/30/2022] [Indexed: 02/08/2023] Open
Abstract
Oligodendrogenesis is essential for replacing worn-out oligodendrocytes, promoting myelin plasticity, and for myelin repair following a demyelinating injury in the adult mammalian brain. Neural stem cells are an important source of oligodendrocytes in the adult brain; however, there are considerable differences in oligodendrogenesis from neural stem cells residing in different areas of the adult brain. Amongst the distinct niches containing neural stem cells, the subventricular zone lining the lateral ventricles and the subgranular zone in the dentate gyrus of the hippocampus are considered the principle areas of adult neurogenesis. In addition to these areas, radial glia-like cells, which are the precursors of neural stem cells, are found in the lining of the third ventricle, where they are called tanycytes, and in the cerebellum, where they are called Bergmann glia. In this review, we will describe the contribution and regulation of each of these niches in adult oligodendrogenesis.
Collapse
|
12
|
Del Valle L, Reiss K, Parker Struckhoff A. Culture and Phenotyping of Glial Cell Cultures, Gliospheres, and Neurospheres. Methods Mol Biol 2022; 2422:217-232. [PMID: 34859409 DOI: 10.1007/978-1-0716-1948-3_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Cell cultures constitute an important tool for research as a way to reproduce pathological processes in a controlled system. However, the culture of brain-derived cells in monolayer presents significant challenges that obscure the fidelity of in vitro results. After a few number of passages, glial and neuronal cells begin to lose their morphological characteristics, and most importantly, their specific cellular markers and phenotype. In recent years, the discovery of neural progenitor cells, and the methodology to culture them in suspension maintaining their potentiality while still retaining the ability to differentiate into astrocytes, oligodendrocytes and neurons has been a significant contribution to the fields of neuroscience and neuropathology.In the brain, progenitor cells are located in the Germinal Matrix, the subventricular zone in what later would become the basal ganglia, and play an essential role in the homeostasis of the brain by providing the source to replace differentiated cells that have been lost or damaged by different pathological processes, such as senescence, injury, genetic conditions, or disease. The discovery of these neural stem cells in an organ traditionally thought to have limited or no regenerative capacity has opened the door to the development of novel treatments, which include cell replacement therapy. Here we describe the culture and differentiation of neural progenitor cells into neurospheres, and the phenotyping of the resulting cells using immunocytochemistry . The immunocytological methods outlined are not restricted to the analysis of neurosphere-derived cultures but are also applicable for cell typing of primary glial or cell line-derived samples.
Collapse
Affiliation(s)
- Luis Del Valle
- Department of Pathology and Medicine & Louisiana Cancer Research Center, Louisiana State University Health Sciences Center, New Orleans, LA, USA.
| | - Krzysztof Reiss
- Departments of Medicine & Neurological Cancer Research, Louisiana Cancer Research Center, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Amanda Parker Struckhoff
- Neurological Cancer Research, Louisiana Cancer Research Center, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| |
Collapse
|
13
|
Kaise T, Fukui M, Sueda R, Piao W, Yamada M, Kobayashi T, Imayoshi I, Kageyama R. Functional rejuvenation of aged neural stem cells by Plagl2 and anti-Dyrk1a activity. Genes Dev 2022; 36:23-37. [PMID: 34916302 PMCID: PMC8763050 DOI: 10.1101/gad.349000.121] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Accepted: 11/29/2021] [Indexed: 11/27/2022]
Abstract
The regenerative potential of neural stem cells (NSCs) declines during aging, leading to cognitive dysfunctions. This decline involves up-regulation of senescence-associated genes, but inactivation of such genes failed to reverse aging of hippocampal NSCs. Because many genes are up-regulated or down-regulated during aging, manipulation of single genes would be insufficient to reverse aging. Here we searched for a gene combination that can rejuvenate NSCs in the aged mouse brain from nuclear factors differentially expressed between embryonic and adult NSCs and their modulators. We found that a combination of inducing the zinc finger transcription factor gene Plagl2 and inhibiting Dyrk1a, a gene associated with Down syndrome (a genetic disorder known to accelerate aging), rejuvenated aged hippocampal NSCs, which already lost proliferative and neurogenic potential. Such rejuvenated NSCs proliferated and produced new neurons continuously at the level observed in juvenile hippocampi, leading to improved cognition. Epigenome, transcriptome, and live-imaging analyses indicated that this gene combination induces up-regulation of embryo-associated genes and down-regulation of age-associated genes by changing their chromatin accessibility, thereby rejuvenating aged dormant NSCs to function like juvenile active NSCs. Thus, aging of NSCs can be reversed to induce functional neurogenesis continuously, offering a way to treat age-related neurological disorders.
Collapse
Affiliation(s)
- Takashi Kaise
- Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
- Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| | - Masahiro Fukui
- Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
- Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| | - Risa Sueda
- Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
- Research Center for Dynamic Living Systems, Graduate School of Biostudies, Kyoto University, Kyoto 606-8501, Japan
| | - Wenhui Piao
- Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
- Research Center for Dynamic Living Systems, Graduate School of Biostudies, Kyoto University, Kyoto 606-8501, Japan
| | - Mayumi Yamada
- Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
- Research Center for Dynamic Living Systems, Graduate School of Biostudies, Kyoto University, Kyoto 606-8501, Japan
- Institute for Integrated Cell-Material Sciences, Kyoto University, Kyoto 606-8501, Japan
| | - Taeko Kobayashi
- Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
- Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
- Research Center for Dynamic Living Systems, Graduate School of Biostudies, Kyoto University, Kyoto 606-8501, Japan
| | - Itaru Imayoshi
- Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
- Research Center for Dynamic Living Systems, Graduate School of Biostudies, Kyoto University, Kyoto 606-8501, Japan
- Institute for Integrated Cell-Material Sciences, Kyoto University, Kyoto 606-8501, Japan
| | - Ryoichiro Kageyama
- Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
- Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
- Research Center for Dynamic Living Systems, Graduate School of Biostudies, Kyoto University, Kyoto 606-8501, Japan
- Institute for Integrated Cell-Material Sciences, Kyoto University, Kyoto 606-8501, Japan
| |
Collapse
|
14
|
Liu LP, Zheng DX, Xu ZF, Zhou HC, Wang YC, Zhou H, Ge JY, Sako D, Li M, Akimoto K, Li YM, Zheng YW. Transcriptomic and Functional Evidence Show Similarities between Human Amniotic Epithelial Stem Cells and Keratinocytes. Cells 2021; 11:70. [PMID: 35011631 PMCID: PMC8750621 DOI: 10.3390/cells11010070] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/14/2021] [Accepted: 12/23/2021] [Indexed: 01/06/2023] Open
Abstract
Amniotic epithelial stem cells (AESCs) are considered as potential alternatives to keratinocytes (KCs) in tissue-engineered skin substitutes used for treating skin damage. However, their clinical application is limited since similarities and distinctions between AESCs and KCs remain unclear. Herein, a transcriptomics analysis and functional evaluation were used to understand the commonalities and differences between AESCs and KCs. RNA-sequencing revealed that AESCs are involved in multiple epidermis-associated biological processes shared by KCs and show more similarity to early stage immature KCs than to adult KCs. However, AESCs were observed to be heterogeneous, and some possessed hybrid mesenchymal and epithelial features distinct from KCs. A functional evaluation revealed that AESCs can phagocytose melanosomes transported by melanocytes in both 2D and 3D co-culture systems similar to KCs, which may help reconstitute pigmented skin. The overexpression of TP63 and activation of NOTCH signaling could promote AESC stemness and improve their differentiation features, respectively, bridging the gap between AESCs and KCs. These changes induced the convergence of AESC cell fate with KCs. In future, modified reprogramming strategies, such as the use of small molecules, may facilitate the further modulation human AESCs for use in skin regeneration.
Collapse
Affiliation(s)
- Li-Ping Liu
- Institute of Regenerative Medicine, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang 212001, China; (L.-P.L.); (H.-C.Z.); (Y.-C.W.); (H.Z.); (M.L.)
- Department of Dermatology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
- Faculty of Medicine, University of Tsukuba, Tsukuba 305-8575, Ibaraki, Japan; (D.-X.Z.); (J.-Y.G.); (D.S.)
| | - Dong-Xu Zheng
- Faculty of Medicine, University of Tsukuba, Tsukuba 305-8575, Ibaraki, Japan; (D.-X.Z.); (J.-Y.G.); (D.S.)
- Department of Human Genetics, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Zheng-Fang Xu
- Department of Obstetrics and Gynaecology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China;
| | - Hu-Cheng Zhou
- Institute of Regenerative Medicine, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang 212001, China; (L.-P.L.); (H.-C.Z.); (Y.-C.W.); (H.Z.); (M.L.)
- Department of Dermatology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
| | - Yun-Cong Wang
- Institute of Regenerative Medicine, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang 212001, China; (L.-P.L.); (H.-C.Z.); (Y.-C.W.); (H.Z.); (M.L.)
- Department of Dermatology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
| | - Hang Zhou
- Institute of Regenerative Medicine, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang 212001, China; (L.-P.L.); (H.-C.Z.); (Y.-C.W.); (H.Z.); (M.L.)
- Department of Dermatology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
| | - Jian-Yun Ge
- Faculty of Medicine, University of Tsukuba, Tsukuba 305-8575, Ibaraki, Japan; (D.-X.Z.); (J.-Y.G.); (D.S.)
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, China
| | - Daisuke Sako
- Faculty of Medicine, University of Tsukuba, Tsukuba 305-8575, Ibaraki, Japan; (D.-X.Z.); (J.-Y.G.); (D.S.)
- Department of Medicinal and Life Sciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda 278-8510, Japan;
| | - Mi Li
- Institute of Regenerative Medicine, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang 212001, China; (L.-P.L.); (H.-C.Z.); (Y.-C.W.); (H.Z.); (M.L.)
- Department of Dermatology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
| | - Kazunori Akimoto
- Department of Medicinal and Life Sciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda 278-8510, Japan;
| | - Yu-Mei Li
- Institute of Regenerative Medicine, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang 212001, China; (L.-P.L.); (H.-C.Z.); (Y.-C.W.); (H.Z.); (M.L.)
- Department of Dermatology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
| | - Yun-Wen Zheng
- Institute of Regenerative Medicine, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang 212001, China; (L.-P.L.); (H.-C.Z.); (Y.-C.W.); (H.Z.); (M.L.)
- Faculty of Medicine, University of Tsukuba, Tsukuba 305-8575, Ibaraki, Japan; (D.-X.Z.); (J.-Y.G.); (D.S.)
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, China
- School of Medicine, Yokohama City University, Yokohama 236-0004, Kanagawa, Japan
| |
Collapse
|
15
|
Bicker F, Nardi L, Maier J, Vasic V, Schmeisser MJ. Criss-crossing autism spectrum disorder and adult neurogenesis. J Neurochem 2021; 159:452-478. [PMID: 34478569 DOI: 10.1111/jnc.15501] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 08/05/2021] [Accepted: 08/28/2021] [Indexed: 12/19/2022]
Abstract
Autism spectrum disorder (ASD) comprises a group of multifactorial neurodevelopmental disorders primarily characterized by deficits in social interaction and repetitive behavior. Although the onset is typically in early childhood, ASD poses a lifelong challenge for both patients and caretakers. Adult neurogenesis (AN) is the process by which new functional neurons are created from neural stem cells existing in the post-natal brain. The entire event is based on a sequence of cellular processes, such as proliferation, specification of cell fate, maturation, and ultimately, synaptic integration into the existing neural circuits. Hence, AN is implicated in structural and functional brain plasticity throughout life. Accumulating evidence shows that impaired AN may underlie some of the abnormal behavioral phenotypes seen in ASD. In this review, we approach the interconnections between the molecular pathways related to AN and ASD. We also discuss existing therapeutic approaches targeting such pathways both in preclinical and clinical studies. A deeper understanding of how ASD and AN reciprocally affect one another could reveal important converging pathways leading to the emergence of psychiatric disorders.
Collapse
Affiliation(s)
- Frank Bicker
- Institute for Microscopic Anatomy and Neurobiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Leonardo Nardi
- Institute for Microscopic Anatomy and Neurobiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Jannik Maier
- Institute for Microscopic Anatomy and Neurobiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Verica Vasic
- Institute for Microscopic Anatomy and Neurobiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Michael J Schmeisser
- Institute for Microscopic Anatomy and Neurobiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany.,Focus Program Translational Neurosciences (FTN), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| |
Collapse
|
16
|
Giri A, Sengupta D, Kar S. Deciphering the Role of Fluctuation Dependent Intercellular Communication in Neural Stem Cell Development. ACS Chem Neurosci 2021; 12:2360-2372. [PMID: 34170103 DOI: 10.1021/acschemneuro.1c00116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Neural stem cells (NPCs) efficiently communicate in an intercellular manner to govern specific cell fate decisions during the developmental process despite withstanding the fluctuating cellular environment. How these fluctuations from diverse origins functionally affect the precise cell fate decision making remains elusive. By taking a stochastic mathematical modeling approach, we unravel that the transcriptional variability arising within an NPC population due to intermittent cell cycle events significantly influences the neuron to NPC ratio during development. Our model proficiently quantifies the impact of different sources of heterogeneities in maintaining an exact neuron to NPC ratio and predicts plausible experimental ways to fine-tune the development of NPCs. In the future, these modeling insights may lead to better therapeutic avenues to regenerate neurons from NPCs.
Collapse
Affiliation(s)
- Amitava Giri
- Department of Chemistry, IIT Bombay, Powai, Mumbai 400076, India
| | - Dola Sengupta
- Department of Chemistry, Techno India University, Salt Lake, Kolkata 700091, India
| | - Sandip Kar
- Department of Chemistry, IIT Bombay, Powai, Mumbai 400076, India
| |
Collapse
|
17
|
Parker Struckhoff A, Del Valle L. Neurospheres and Glial Cell Cultures; from Plating to Cell Phenotyping. Methods Mol Biol 2021; 2311:131-145. [PMID: 34033081 DOI: 10.1007/978-1-0716-1437-2_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Cell cultures constitute an important tool for research as a way to reproduce pathological processes in a controlled system. However, the culture of brain-derived cells in monolayer presents significant challenges that obscure the fidelity of in vitro results. This is because after a few number of passages, glial and neuronal cells begin to lose their morphological characteristics, and most importantly, their specific cellular markers and phenotype. In recent years, the discovery of neural progenitor cells, and the methodology to culture them in suspension maintaining their potentiality while still retaining the ability to differentiate into astrocytes, oligodendrocytes, and neurons has made significant contributions to the fields of neuroscience and neuropathology.In the brain, progenitor cells are located in the germinal matrix, in the subventricular zone and play an essential role in the homeostasis of the brain by providing the source to replace differentiated cells that have been lost or damaged by different pathological processes, such as injury, genetic conditions, or disease. The discovery of these Neural Stem Cells in an organ traditionally thought to have limited or no regenerative capacity has opened the door to the development of novel treatments, which include cell replacement therapy. Here we describe the culture and differentiation of neural progenitor cells from Neurospheres, and the phenotyping of the resulting cells using immunocytochemistry. The immunocytological methods outlined are not restricted to the analysis of neurosphere-derived cultures but are also applicable for cell typing of primary glial or cell line-derived samples.
Collapse
Affiliation(s)
- Amanda Parker Struckhoff
- Department of Pathology & Stanley S. Scott Cancer Center, Louisiana State University Health, New Orleans, LA, USA
| | - Luis Del Valle
- Department of Pathology & Stanley S. Scott Cancer Center, Louisiana State University Health, New Orleans, LA, USA.
| |
Collapse
|
18
|
Zhang R, Boareto M, Engler A, Louvi A, Giachino C, Iber D, Taylor V. Id4 Downstream of Notch2 Maintains Neural Stem Cell Quiescence in the Adult Hippocampus. Cell Rep 2020; 28:1485-1498.e6. [PMID: 31390563 DOI: 10.1016/j.celrep.2019.07.014] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 04/14/2019] [Accepted: 07/02/2019] [Indexed: 12/31/2022] Open
Abstract
Neural stem cells (NSCs) in the adult mouse hippocampal dentate gyrus (DG) are mostly quiescent, and only a few are in cell cycle at any point in time. DG NSCs become increasingly dormant with age and enter mitosis less frequently, which impinges on neurogenesis. How NSC inactivity is maintained is largely unknown. Here, we found that Id4 is a downstream target of Notch2 signaling and maintains DG NSC quiescence by blocking cell-cycle entry. Id4 expression is sufficient to promote DG NSC quiescence and Id4 knockdown rescues Notch2-induced inhibition of NSC proliferation. Id4 deletion activates NSC proliferation in the DG without evoking neuron generation, and overexpression increases NSC maintenance while promoting astrogliogenesis at the expense of neurogenesis. Together, our findings indicate that Id4 is a major effector of Notch2 signaling in NSCs and a Notch2-Id4 axis promotes NSC quiescence in the adult DG, uncoupling NSC activation from neuronal differentiation.
Collapse
Affiliation(s)
- Runrui Zhang
- Embryology and Stem Cell Biology Lab, Department of Biomedicine, University of Basel, Mattenstrasse 28, 4058 Basel, Switzerland
| | - Marcelo Boareto
- Computational Biology Group, D-BSSE, ETH Zürich, Mattenstrasse 26, 4058 Basel, Switzerland; Swiss Institute of Bioinformatics (SIB), Mattenstrasse 26, 4058 Basel, Switzerland
| | - Anna Engler
- Embryology and Stem Cell Biology Lab, Department of Biomedicine, University of Basel, Mattenstrasse 28, 4058 Basel, Switzerland
| | - Angeliki Louvi
- Departments of Neurosurgery and Neuroscience, Yale Program on Neurogenetics, Yale School of Medicine, New Haven, CT 06520, USA
| | - Claudio Giachino
- Embryology and Stem Cell Biology Lab, Department of Biomedicine, University of Basel, Mattenstrasse 28, 4058 Basel, Switzerland
| | - Dagmar Iber
- Computational Biology Group, D-BSSE, ETH Zürich, Mattenstrasse 26, 4058 Basel, Switzerland; Swiss Institute of Bioinformatics (SIB), Mattenstrasse 26, 4058 Basel, Switzerland
| | - Verdon Taylor
- Embryology and Stem Cell Biology Lab, Department of Biomedicine, University of Basel, Mattenstrasse 28, 4058 Basel, Switzerland.
| |
Collapse
|
19
|
Yokoi Y, Nishimura SI. Effect of Site-Specific O-Glycosylation on the Structural Behavior of NOTCH1 Receptor Extracellular EGF-like Domains 11 and 10. Chemistry 2020; 26:12363-12372. [PMID: 32632967 DOI: 10.1002/chem.202002652] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Indexed: 12/16/2022]
Abstract
Human NOTCH1 receptor contains 36 epidermal growth factor (EGF)-like repeating domains, in which O-glycosylation status of EGF12 domain regulates the interaction with Notch ligands. Our interest is focused on the effect of specific O-glycosylation states on the structural behavior of EGF11 and EGF10, because they appeared to affect molecular mechanism in receptor-ligand interactions by inducing some conformational alterations in these domains and/or the regions connecting two domains. To understand the structural impact of various O-glycosylation patterns on the pivotal EGF-like repeats 10, 11, and 12, we performed chemical synthesis and NMR studies of site-specifically O-glycosylated EGF11 and EGF10. Our strategy enabled us to synthesize four EGF11 and five EGF10 modules. The specific O-glycosylation states affected in vitro folding of EGF10 more than EGF11, while calcium ion had a larger effect on EGF11 folding. Comprehensive NMR studies shed light on the new type "sugar bridges" crosslinking Thr-O-GlcNAc in the consensus sequence C5-X-X-G-X-(T/S)-G-X-X-C6 and an amino acid in the hinge region between the domains, 445Thr-O-GlcNAc-IIe451 in domain 11 and 405Thr-O-GlcNAc-Gln411 in domain 10, respectively.
Collapse
Affiliation(s)
- Yasuhiro Yokoi
- Graduate School of Life Science and Faculty of Advanced Life Science, Hokkaido University, N21, W11, Kita-ku, Sapporo, 001-0021, Japan
| | - Shin-Ichiro Nishimura
- Graduate School of Life Science and Faculty of Advanced Life Science, Hokkaido University, N21, W11, Kita-ku, Sapporo, 001-0021, Japan
| |
Collapse
|
20
|
Häussinger D, Kordes C. Space of Disse: a stem cell niche in the liver. Biol Chem 2020; 401:81-95. [PMID: 31318687 DOI: 10.1515/hsz-2019-0283] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 07/08/2019] [Indexed: 02/06/2023]
Abstract
Recent evidence indicates that the plasticity of preexisting hepatocytes and bile duct cells is responsible for the appearance of intermediate progenitor cells capable of restoring liver mass after injury without the need of a stem cell compartment. However, mesenchymal stem cells (MSCs) exist in all organs and are associated with blood vessels which represent their perivascular stem cell niche. MSCs are multipotent and can differentiate into several cell types and are known to support regenerative processes by the release of immunomodulatory and trophic factors. In the liver, the space of Disse constitutes a stem cell niche that harbors stellate cells as liver resident MSCs. This perivascular niche is created by extracellular matrix proteins, sinusoidal endothelial cells, liver parenchymal cells and sympathetic nerve endings and establishes a microenvironment that is suitable to maintain stellate cells and to control their fate. The stem cell niche integrity is important for the behavior of stellate cells in the normal, regenerative, aged and diseased liver. The niche character of the space of Disse may further explain why the liver can become an organ of extra-medullar hematopoiesis and why this organ is frequently prone to tumor metastasis.
Collapse
Affiliation(s)
- Dieter Häussinger
- Clinic of Gastroenterology, Hepatology and Infectious Diseases, Heinrich Heine University Düsseldorf, Moorenstraße 5, D-40225 Düsseldorf, Germany
| | - Claus Kordes
- Clinic of Gastroenterology, Hepatology and Infectious Diseases, Heinrich Heine University Düsseldorf, Moorenstraße 5, D-40225 Düsseldorf, Germany
| |
Collapse
|
21
|
Blackwood CA, Bailetti A, Nandi S, Gridley T, Hébert JM. Notch Dosage : Jagged1 Haploinsufficiency Is Associated With Reduced Neuronal Division and Disruption of Periglomerular Interneurons in Mice. Front Cell Dev Biol 2020; 8:113. [PMID: 32161758 PMCID: PMC7054221 DOI: 10.3389/fcell.2020.00113] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Accepted: 02/10/2020] [Indexed: 11/13/2022] Open
Abstract
Neural stem cells in the lateral ganglionic eminence (LGE) generate progenitors that migrate through the rostral migratory stream (RMS) to repopulate olfactory bulb (OB) interneurons, but the regulation of this process is poorly defined. The evolutionarily conserved Notch pathway is essential for neural development and maintenance of neural stem cells. Jagged1, a Notch ligand, is required for stem cell maintenance. In humans, heterozygous mutations in JAGGED1 cause Alagille syndrome, a genetic disorder characterized by complications such as cognitive impairment and reduced number of bile ducts in the liver, suggesting the presence of a JAGGED1 haploinsufficient phenotype. Here, we examine the role of Jagged1 using a conditional loss-of-function allele in the nervous system. We show that heterozygous Jagged1 mice possess a haploinsufficient phenotype that is associated with a reduction in size of the LGE, a reduced proliferative state, and fewer progenitor cells in the LGE and RMS. Moreover, loss of Jagged1 leads to deficits in periglomerular interneurons in the OB. Our results support a dose-dependent role for Jagged1 in maintaining progenitor division within the LGE and RMS.
Collapse
Affiliation(s)
- Christopher A. Blackwood
- Molecular Neuropsychiatry Research Branch, National Institutes of Health/National Institute on Drug Abuse Intramural Research Program, Baltimore, MD, United States
- Department of Biomedical Sciences, Cornell University, Ithaca, NY, United States
- Departments of Neuroscience and Genetics, Albert Einstein College of Medicine, New York, NY, United States
| | - Alessandro Bailetti
- Department of Biomedical Sciences, Cornell University, Ithaca, NY, United States
| | - Sayan Nandi
- Departments of Neuroscience and Genetics, Albert Einstein College of Medicine, New York, NY, United States
| | - Thomas Gridley
- Maine Medical Center Research Institute, Scarborough, ME, United States
| | - Jean M. Hébert
- Departments of Neuroscience and Genetics, Albert Einstein College of Medicine, New York, NY, United States
| |
Collapse
|
22
|
Bacigaluppi M, Sferruzza G, Butti E, Ottoboni L, Martino G. Endogenous neural precursor cells in health and disease. Brain Res 2019; 1730:146619. [PMID: 31874148 DOI: 10.1016/j.brainres.2019.146619] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 11/25/2019] [Accepted: 12/19/2019] [Indexed: 12/15/2022]
Abstract
Neurogenesis persists in the adult brain of mammals in the subventricular zone (SVZ) of the lateral ventricles and in the subgranular zone (SGZ) of the dentate gyrus (DG). The complex interactions between intrinsic and extrinsic signals provided by cells in the niche but also from distant sources regulate the fate of neural stem/progenitor cells (NPCs) in these sites. This fine regulation is perturbed in aging and in pathological conditions leading to a different NPC behavior, tailored to the specific physio-pathological features. Indeed, NPCs exert in physiological and pathological conditions important neurogenic and non-neurogenic regulatory functions and participate in maintaining and protecting brain tissue homeostasis. In this review, we discuss intrinsic and extrinsic signals that regulate NPC activation and NPC functional role in various homeostatic and non-homeostatic conditions.
Collapse
Affiliation(s)
- Marco Bacigaluppi
- Neuroimmunology Unit and Department of Neurology, Institute of Experimental Neurology, San Raffaele Hospital and Università Vita- Salute San Raffaele, Via Olgettina 60, 20132 Milano, Italy.
| | - Giacomo Sferruzza
- Neuroimmunology Unit and Department of Neurology, Institute of Experimental Neurology, San Raffaele Hospital and Università Vita- Salute San Raffaele, Via Olgettina 60, 20132 Milano, Italy
| | - Erica Butti
- Neuroimmunology Unit and Department of Neurology, Institute of Experimental Neurology, San Raffaele Hospital and Università Vita- Salute San Raffaele, Via Olgettina 60, 20132 Milano, Italy
| | - Linda Ottoboni
- Neuroimmunology Unit and Department of Neurology, Institute of Experimental Neurology, San Raffaele Hospital and Università Vita- Salute San Raffaele, Via Olgettina 60, 20132 Milano, Italy
| | - Gianvito Martino
- Neuroimmunology Unit and Department of Neurology, Institute of Experimental Neurology, San Raffaele Hospital and Università Vita- Salute San Raffaele, Via Olgettina 60, 20132 Milano, Italy
| |
Collapse
|
23
|
Abstract
The transition between proliferating and quiescent states must be carefully regulated to ensure that cells divide to create the cells an organism needs only at the appropriate time and place. Cyclin-dependent kinases (CDKs) are critical for both transitioning cells from one cell cycle state to the next, and for regulating whether cells are proliferating or quiescent. CDKs are regulated by association with cognate cyclins, activating and inhibitory phosphorylation events, and proteins that bind to them and inhibit their activity. The substrates of these kinases, including the retinoblastoma protein, enforce the changes in cell cycle status. Single cell analysis has clarified that competition among factors that activate and inhibit CDK activity leads to the cell's decision to enter the cell cycle, a decision the cell makes before S phase. Signaling pathways that control the activity of CDKs regulate the transition between quiescence and proliferation in stem cells, including stem cells that generate muscle and neurons. © 2020 American Physiological Society. Compr Physiol 10:317-344, 2020.
Collapse
Affiliation(s)
- Hilary A Coller
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, California, USA.,Department of Biological Chemistry, David Geffen School of Medicine, and the Molecular Biology Institute, University of California, Los Angeles, California, USA.,Molecular Biology Institute, University of California, Los Angeles, California, USA
| |
Collapse
|
24
|
Blackwood C. Quantitative approach to numbers and sizes: Generation of primary neurospheres from the dorsal lateral ganglionic eminence of late embryonic mice. F1000Res 2019; 8:1983. [PMID: 32266058 PMCID: PMC7101018 DOI: 10.12688/f1000research.21208.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/09/2020] [Indexed: 11/21/2022] Open
Abstract
Background: The neurosphere assay is a powerful
in vitro tool to investigate neural stem cells in the dorsal lateral ventricle (dLGE). In the dLGE, metrics of sizes and numbers of neurospheres generated using this assay has not been completely characterized. The objective of this protocol is to provide a stepwise method from a single isolation that predicts the average number of neurospheres generated and to estimate an approximation of its sizes after several days
in vitro. The advantage of this protocol is that no expensive and specialized equipment is needed for tissue isolation. Estimates about the numbers and sizes of neurospheres will provide investigators with quantitative data to advise on how much starting dLGE tissue is required to generate the appropriate number of spheres for the implementation of downstream applications, including immunocytochemistry, self-renewal and differentiation assays. Methods: Our method is based on a simple dissection technique, where tissue surrounding the dorsal lateral ventricle from a single mouse embryo is trimmed away to enrich for neural stem cell and progenitor populations. Following this dissection, tissue is mechanically dissociated by trituration. Cells are then cultured in media containing epidermal growth factor and other supplements to generate healthy primary neurospheres. Results: Using this approach, we found reproducible number of primary neurospheres after 7 days
in vitro (DIV). Furthermore, we observed that this method yields an average range of neurospheres sizes greater than 50 μm, but less than 100 μm after 7 DIV. Lastly, using an anti-GFAP antibody, we show that these neurospheres can be stained, confirming their use in future immunocytochemistry studies. Conclusions: Future use of this protocol provides metrics on the generation of primary neurospheres that will be useful for further advances in the area of stem cell biology.
Collapse
|
25
|
Sueda R, Kageyama R. Regulation of active and quiescent somatic stem cells by Notch signaling. Dev Growth Differ 2019; 62:59-66. [PMID: 31489617 PMCID: PMC7027910 DOI: 10.1111/dgd.12626] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 07/31/2019] [Accepted: 08/01/2019] [Indexed: 12/14/2022]
Abstract
Somatic stem/progenitor cells actively proliferate and give rise to different types of mature cells (active state) in embryonic tissues while they are mostly dormant (quiescent state) in many adult tissues. Notch signaling is known to regulate both active and quiescent states of somatic stem cells, but how it regulates these different states is unknown. Recent studies revealed that the Notch effector Hes1 is expressed differently during the active and quiescent states during neurogenesis and myogenesis: high in the quiescent state and oscillatory in the active state. When the Hes1 expression level is high, both Ascl1 and MyoD expression are continuously suppressed. By contrast, when Hes1 expression oscillates, it periodically represses expression of the neurogenic factor Ascl1 and the myogenic factor MyoD, thereby driving Ascl1 and MyoD oscillations. High levels of Hes1 and the resultant Ascl1 suppression promote the quiescent state of neural stem cells, while Hes1 oscillation-dependent Ascl1 oscillations regulate their active state. Similarly, in satellite cells of muscles, known adult muscle stem cells, high levels of Hes1 and the resultant MyoD suppression seem to promote their quiescent state, while Hes1 oscillation-dependent MyoD oscillations activate their proliferation and differentiation. Therefore, the expression dynamics of Hes1 is a key regulatory mechanism of generating and maintaining active/quiescent stem cell states.
Collapse
Affiliation(s)
- Risa Sueda
- Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan.,Kyoto University Graduate School of Biostudies, Kyoto, Japan
| | - Ryoichiro Kageyama
- Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan.,Kyoto University Graduate School of Biostudies, Kyoto, Japan.,Kyoto University Graduate School of Medicine, Kyoto, Japan.,Institute for Integrated Cell-Material Sciences, Kyoto University, Kyoto, Japan
| |
Collapse
|
26
|
Adult Neurogenesis in the Subventricular Zone and Its Regulation After Ischemic Stroke: Implications for Therapeutic Approaches. Transl Stroke Res 2019; 11:60-79. [DOI: 10.1007/s12975-019-00717-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 06/13/2019] [Accepted: 06/27/2019] [Indexed: 12/21/2022]
|
27
|
Blackwood CA. Jagged1 is Essential for Radial Glial Maintenance in the Cortical Proliferative Zone. Neuroscience 2019; 413:230-238. [PMID: 31202705 DOI: 10.1016/j.neuroscience.2019.05.062] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 05/31/2019] [Accepted: 05/31/2019] [Indexed: 12/15/2022]
Abstract
Radial glial maintenance is essential for the proper development of the cortex. It is known that the evolutionarily conserved Notch signaling pathway is required for maintaining the pool of radial glial stem cells although the mechanisms involved are not entirely understood. Here, we study the Notch ligand, Jagged1, in the mouse ventricular zone at a late stage of embryonic development. We use a conditional loss of function allele to show that Jagged1 is required for maintaining radial glial cells and when absent, leads to defects in the cortical proliferation zone and expression of intermediate progenitor cells. Using in vitro approaches, we found that depletion of Jagged1 reduced the size of primary neurospheres and their capacity to self-renewal. Finally, Jagged1 mutants also showed precocious neuronal differentiation and cortical defects. Together, these data support a role for Jagged1 in radial glia maintenance in the neocortex.
Collapse
Affiliation(s)
- Christopher A Blackwood
- Department of Neuroscience, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY, 10461.
| |
Collapse
|
28
|
Sueda R, Imayoshi I, Harima Y, Kageyama R. High Hes1 expression and resultant Ascl1 suppression regulate quiescent vs. active neural stem cells in the adult mouse brain. Genes Dev 2019; 33:511-523. [PMID: 30862661 PMCID: PMC6499325 DOI: 10.1101/gad.323196.118] [Citation(s) in RCA: 122] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 02/26/2019] [Indexed: 01/03/2023]
Abstract
Sueda et al. show that in quiescent neural stem cells, Hes1 levels are oscillatory, although the peaks and troughs are higher than those in active neural stem cells, causing Ascl1 expression to be continuously suppressed. Somatic stem/progenitor cells are active in embryonic tissues but quiescent in many adult tissues. The detailed mechanisms that regulate active versus quiescent stem cell states are largely unknown. In active neural stem cells, Hes1 expression oscillates and drives cyclic expression of the proneural gene Ascl1, which activates cell proliferation. Here, we found that in quiescent neural stem cells in the adult mouse brain, Hes1 levels are oscillatory, although the peaks and troughs are higher than those in active neural stem cells, causing Ascl1 expression to be continuously suppressed. Inactivation of Hes1 and its related genes up-regulates Ascl1 expression and increases neurogenesis. This causes rapid depletion of neural stem cells and premature termination of neurogenesis. Conversely, sustained Hes1 expression represses Ascl1, inhibits neurogenesis, and maintains quiescent neural stem cells. In contrast, induction of Ascl1 oscillations activates neural stem cells and increases neurogenesis in the adult mouse brain. Thus, Ascl1 oscillations, which normally depend on Hes1 oscillations, regulate the active state, while high Hes1 expression and resultant Ascl1 suppression promote quiescence in neural stem cells.
Collapse
Affiliation(s)
- Risa Sueda
- Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan.,Graduate School of Biostudies, Kyoto University, Kyoto 606-8501, Japan
| | - Itaru Imayoshi
- Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan.,Graduate School of Biostudies, Kyoto University, Kyoto 606-8501, Japan.,Research Center for Dynamic Living Systems, Graduate School of Biostudies, Kyoto University, Kyoto 606-8501, Japan.,Precursory Research for Embryonic Science and Technology, Japan Science and Technology Agency, Saitama 332-0012, Japan.,Institute for Integrated Cell-Material Sciences (iCeMS), Kyoto University, Kyoto 606-8501, Japan
| | - Yukiko Harima
- Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
| | - Ryoichiro Kageyama
- Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan.,Graduate School of Biostudies, Kyoto University, Kyoto 606-8501, Japan.,Research Center for Dynamic Living Systems, Graduate School of Biostudies, Kyoto University, Kyoto 606-8501, Japan.,Institute for Integrated Cell-Material Sciences (iCeMS), Kyoto University, Kyoto 606-8501, Japan.,Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| |
Collapse
|
29
|
Bazzoni R, Bentivegna A. Role of Notch Signaling Pathway in Glioblastoma Pathogenesis. Cancers (Basel) 2019; 11:cancers11030292. [PMID: 30832246 PMCID: PMC6468848 DOI: 10.3390/cancers11030292] [Citation(s) in RCA: 125] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Revised: 02/17/2019] [Accepted: 02/25/2019] [Indexed: 12/12/2022] Open
Abstract
Notch signaling is an evolutionarily conserved pathway that regulates important biological processes, such as cell proliferation, apoptosis, migration, self-renewal, and differentiation. In mammals, Notch signaling is composed of four receptors (Notch1–4) and five ligands (Dll1-3–4, Jagged1–2) that mainly contribute to the development and maintenance of the central nervous system (CNS). Neural stem cells (NSCs) are the starting point for neurogenesis and other neurological functions, representing an essential aspect for the homeostasis of the CNS. Therefore, genetic and functional alterations to NSCs can lead to the development of brain tumors, including glioblastoma. Glioblastoma remains an incurable disease, and the reason for the failure of current therapies and tumor relapse is the presence of a small subpopulation of tumor cells known as glioma stem cells (GSCs), characterized by their stem cell-like properties and aggressive phenotype. Growing evidence reveals that Notch signaling is highly active in GSCs, where it suppresses differentiation and maintains stem-like properties, contributing to Glioblastoma tumorigenesis and conventional-treatment resistance. In this review, we try to give a comprehensive view of the contribution of Notch signaling to Glioblastoma and its possible implication as a target for new therapeutic approaches.
Collapse
Affiliation(s)
- Riccardo Bazzoni
- Stem Cell Research Laboratory, Section of Hematology, Department of Medicine, University of Verona, Pz.le Scuro 10, 37134 Verona, Italy.
- Program in Clinical and Experimental Biomedical Sciences, University of Verona, 37134 Verona, Italy.
- NeuroMi, Milan Center for Neuroscience, Department of Neurology and Neuroscience, San Gerardo Hospital, University of Milano-Bicocca, 20900 Monza, Italy.
| | - Angela Bentivegna
- NeuroMi, Milan Center for Neuroscience, Department of Neurology and Neuroscience, San Gerardo Hospital, University of Milano-Bicocca, 20900 Monza, Italy.
- School of Medicine and Surgery, University of Milano-Bicocca, via Cadore 48, 20900 Monza, Italy.
| |
Collapse
|
30
|
Abstract
Purpose Retinal degenerative diseases lead to the death of retinal neurons causing visual impairment and blindness. In lower order vertebrates, the retina and its surrounding tissue contain stem cell niches capable of regenerating damaged tissue. Here we examine these niches and review their capacity to be used as retinal stem/progenitor cells (RSC/RPCs) for retinal repair. Recent Findings Exogenous factors can control the in vitro activation of RSCs/PCs found in several niches within the adult eye including cells in the ciliary margin, the retinal pigment epithelium, iris pigment epithelium as well as the inducement of Müller and amacrine cells within the neural retina itself. Recently, factors have been identified for the activation of adult mammalian Müller cells to a RPC state in vivo. Summary Whereas cell transplantation still holds potential for retinal repair, activation of the dormant native regeneration process may lead to a more successful process including greater integration efficiency and proper synaptic targeting.
Collapse
|
31
|
Rushing GV, Bollig MK, Ihrie RA. Heterogeneity of Neural Stem Cells in the Ventricular-Subventricular Zone. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1169:1-30. [PMID: 31487016 DOI: 10.1007/978-3-030-24108-7_1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
In this chapter, heterogeneity is explored in the context of the ventricular-subventricular zone, the largest stem cell niche in the mammalian brain. This niche generates up to 10,000 new neurons daily in adult mice and extends over a large spatial area with dorso-ventral and medio-lateral subdivisions. The stem cells of the ventricular-subventricular zone can be subdivided by their anatomical position and transcriptional profile, and the stem cell lineage can also be further subdivided into stages of pre- and post-natal quiescence and activation. Beyond the stem cells proper, additional differences exist in their interactions with other cellular constituents of the niche, including neurons, vasculature, and cerebrospinal fluid. These variations in stem cell potential and local interactions are discussed, as well as unanswered questions within this system.
Collapse
Affiliation(s)
- Gabrielle V Rushing
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA.,Neuroscience Program, Vanderbilt University, Nashville, TN, USA
| | - Madelyn K Bollig
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA.,Neuroscience Program, Vanderbilt University, Nashville, TN, USA
| | - Rebecca A Ihrie
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA. .,Neuroscience Program, Vanderbilt University, Nashville, TN, USA. .,Department of Neurological Surgery, Vanderbilt University School of Medicine, Nashville, TN, USA.
| |
Collapse
|
32
|
Kim E, Hwang SU, Yoon JD, Kim H, Lee G, Hyun SH. Isolation and characterization of GFAP-positive porcine neural stem/progenitor cells derived from a GFAP-CreER T2 transgenic piglet. BMC Vet Res 2018; 14:331. [PMID: 30404643 PMCID: PMC6222979 DOI: 10.1186/s12917-018-1660-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2018] [Accepted: 10/22/2018] [Indexed: 01/17/2023] Open
Abstract
Background The porcine brain is gyrencephalic with similar gray and white matter composition and size more comparable to the human rather than the rodent brain; however, there is lack of information about neural progenitor cells derived from this model. Results Here, we isolated GFAP-positive porcine neural stem cells (NSCs) from the brain explant of a transgenic piglet, with expression of CreERT2 under the control of the GFAP promoter (pGFAP-CreERT2). The isolated pGFAP-CreERT2 NSCs showed self-renewal and expression of representative NSC markers such as Nestin and Sox2. Pharmacological inhibition studies revealed that Notch1 signaling is necessary to maintain NSC identity, whereas serum treatment induced cell differentiation into reactive astrocytes and neurons. Conclusions Collectively, these results indicate that GFAP promoter-driven porcine CreERT2 NSCs would be a useful tool to study neurogenesis of the porcine adult central nervous system and furthers our understanding of its potential clinical application in the future. Graphical abstract ᅟ![]() Electronic supplementary material The online version of this article (10.1186/s12917-018-1660-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Eunhye Kim
- Laboratory of Veterinary Embryology and Biotechnology, Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University, 1 Chungdae-ro, Seowon-gu, Cheongju, 28644, Republic of Korea.,Institute of Stem Cell & Regenerative Medicine (ISCRM), Chungbuk National University, Cheongju, 28644, Chungbuk, Republic of Korea
| | - Seon-Ung Hwang
- Laboratory of Veterinary Embryology and Biotechnology, Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University, 1 Chungdae-ro, Seowon-gu, Cheongju, 28644, Republic of Korea.,Institute of Stem Cell & Regenerative Medicine (ISCRM), Chungbuk National University, Cheongju, 28644, Chungbuk, Republic of Korea
| | - Junchul David Yoon
- Laboratory of Veterinary Embryology and Biotechnology, Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University, 1 Chungdae-ro, Seowon-gu, Cheongju, 28644, Republic of Korea.,Institute of Stem Cell & Regenerative Medicine (ISCRM), Chungbuk National University, Cheongju, 28644, Chungbuk, Republic of Korea
| | - Hyunggee Kim
- Department of Biotechnology, School of Life Sciences and Biotechnology, Korea University, 02841, Seoul, Republic of Korea
| | - Gabsang Lee
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sang-Hwan Hyun
- Laboratory of Veterinary Embryology and Biotechnology, Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University, 1 Chungdae-ro, Seowon-gu, Cheongju, 28644, Republic of Korea. .,Institute of Stem Cell & Regenerative Medicine (ISCRM), Chungbuk National University, Cheongju, 28644, Chungbuk, Republic of Korea.
| |
Collapse
|
33
|
Huang S, Park J, Qiu C, Chung KW, Li SY, Sirin Y, Han SH, Taylor V, Zimber-Strobl U, Susztak K. Jagged1/Notch2 controls kidney fibrosis via Tfam-mediated metabolic reprogramming. PLoS Biol 2018; 16:e2005233. [PMID: 30226866 PMCID: PMC6161902 DOI: 10.1371/journal.pbio.2005233] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 09/28/2018] [Accepted: 09/03/2018] [Indexed: 12/14/2022] Open
Abstract
While Notch signaling has been proposed to play a key role in fibrosis, the direct molecular pathways targeted by Notch signaling and the precise ligand and receptor pair that are responsible for kidney disease remain poorly defined. In this study, we found that JAG1 and NOTCH2 showed the strongest correlation with the degree of interstitial fibrosis in a genome-wide expression analysis of a large cohort of human kidney samples. Transcript analysis of mouse kidney disease models, including folic-acid (FA)-induced nephropathy, unilateral ureteral obstruction (UUO), or apolipoprotein L1 (APOL1)-associated kidney disease, indicated that Jag1 and Notch2 levels were higher in all analyzed kidney fibrosis models. Mice with tubule-specific deletion of Jag1 or Notch2 (Kspcre/Jag1flox/flox and Kspcre/Notch2flox/flox) had no kidney-specific alterations at baseline but showed protection from FA-induced kidney fibrosis. Tubule-specific genetic deletion of Notch1 and global knockout of Notch3 had no effect on fibrosis. In vitro chromatin immunoprecipitation experiments and genome-wide expression studies identified the mitochondrial transcription factor A (Tfam) as a direct Notch target. Re-expression of Tfam in tubule cells prevented Notch-induced metabolic and profibrotic reprogramming. Tubule-specific deletion of Tfam resulted in fibrosis. In summary, Jag1 and Notch2 play a key role in kidney fibrosis development by regulating Tfam expression and metabolic reprogramming.
Collapse
Affiliation(s)
- Shizheng Huang
- Renal Electrolyte and Hypertension Division, Department of Medicine, Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Jihwan Park
- Renal Electrolyte and Hypertension Division, Department of Medicine, Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Chengxiang Qiu
- Renal Electrolyte and Hypertension Division, Department of Medicine, Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Ki Wung Chung
- Renal Electrolyte and Hypertension Division, Department of Medicine, Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Szu-yuan Li
- Renal Electrolyte and Hypertension Division, Department of Medicine, Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Yasemin Sirin
- Renal Electrolyte and Hypertension Division, Department of Medicine, Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Seung Hyeok Han
- Renal Electrolyte and Hypertension Division, Department of Medicine, Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Verdon Taylor
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Ursula Zimber-Strobl
- Research Unit Gene Vectors, Helmholtz Zentrum München, German Research Center for Environment and Health, Munich, Germany
| | - Katalin Susztak
- Renal Electrolyte and Hypertension Division, Department of Medicine, Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
34
|
Braccioli L, Vervoort SJ, Adolfs Y, Heijnen CJ, Basak O, Pasterkamp RJ, Nijboer CH, Coffer PJ. FOXP1 Promotes Embryonic Neural Stem Cell Differentiation by Repressing Jagged1 Expression. Stem Cell Reports 2018; 9:1530-1545. [PMID: 29141232 PMCID: PMC5688236 DOI: 10.1016/j.stemcr.2017.10.012] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 10/13/2017] [Accepted: 10/13/2017] [Indexed: 01/11/2023] Open
Abstract
Mutations in FOXP1 have been linked to neurodevelopmental disorders including intellectual disability and autism; however, the underlying molecular mechanisms remain ill-defined. Here, we demonstrate with RNA and chromatin immunoprecipitation sequencing that FOXP1 directly regulates genes controlling neurogenesis. We show that FOXP1 is expressed in embryonic neural stem cells (NSCs), and modulation of FOXP1 expression affects both neuron and astrocyte differentiation. Using a murine model of cortical development, FOXP1-knockdown in utero was found to reduce NSC differentiation and migration during corticogenesis. Furthermore, transplantation of FOXP1-knockdown NSCs in neonatal mice after hypoxia-ischemia challenge demonstrated that FOXP1 is also required for neuronal differentiation and functionality in vivo. FOXP1 was found to repress the expression of Notch pathway genes including the Notch-ligand Jagged1, resulting in inhibition of Notch signaling. Finally, blockade of Jagged1 in FOXP1-knockdown NSCs rescued neuronal differentiation in vitro. Together, these data support a role for FOXP1 in regulating embryonic NSC differentiation by modulating Notch signaling. FOXP1 promotes astrocyte and neuronal differentiation of NSCs in vitro FOXP1 promotes neuronal differentiation of NSCs in vivo FOXP1 transcriptionally regulates pro-neural genes and represses Notch pathway genes FOXP1 promotes neuronal differentiation by limiting Jagged1 expression
Collapse
Affiliation(s)
- Luca Braccioli
- Laboratory of Neuroimmunology and Developmental Origins of Disease (NIDOD), University Medical Center Utrecht, Utrecht 3508 AB, the Netherlands; Center for Molecular Medicine and Regenerative Medicine Center, University Medical Center Utrecht, Utrecht 3584 CT, the Netherlands
| | - Stephin J Vervoort
- Center for Molecular Medicine and Regenerative Medicine Center, University Medical Center Utrecht, Utrecht 3584 CT, the Netherlands
| | - Youri Adolfs
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht 3584 CX, the Netherlands
| | - Cobi J Heijnen
- Laboratory of Neuroimmunology, Department of Symptom Research, Division of Internal Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Onur Basak
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, 3584 CT Utrecht, the Netherlands
| | - R Jeroen Pasterkamp
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht 3584 CX, the Netherlands
| | - Cora H Nijboer
- Laboratory of Neuroimmunology and Developmental Origins of Disease (NIDOD), University Medical Center Utrecht, Utrecht 3508 AB, the Netherlands.
| | - Paul J Coffer
- Center for Molecular Medicine and Regenerative Medicine Center, University Medical Center Utrecht, Utrecht 3584 CT, the Netherlands.
| |
Collapse
|
35
|
Engler A, Rolando C, Giachino C, Saotome I, Erni A, Brien C, Zhang R, Zimber-Strobl U, Radtke F, Artavanis-Tsakonas S, Louvi A, Taylor V. Notch2 Signaling Maintains NSC Quiescence in the Murine Ventricular-Subventricular Zone. Cell Rep 2018; 22:992-1002. [PMID: 29386140 DOI: 10.1016/j.celrep.2017.12.094] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 06/12/2017] [Accepted: 12/22/2017] [Indexed: 12/21/2022] Open
Abstract
Neurogenesis continues in the ventricular-subventricular zone (V-SVZ) of the adult forebrain from quiescent neural stem cells (NSCs). V-SVZ NSCs are a reservoir for new olfactory bulb (OB) neurons that migrate through the rostral migratory stream (RMS). To generate neurons, V-SVZ NSCs need to activate and enter the cell cycle. The mechanisms underlying NSC transition from quiescence to activity are poorly understood. We show that Notch2, but not Notch1, signaling conveys quiescence to V-SVZ NSCs by repressing cell-cycle-related genes and neurogenesis. Loss of Notch2 activates quiescent NSCs, which proliferate and generate new neurons of the OB lineage. Notch2 deficiency results in accelerated V-SVZ NSC exhaustion and an aging-like phenotype. Simultaneous loss of Notch1 and Notch2 resembled the total loss of Rbpj-mediated canonical Notch signaling; thus, Notch2 functions are not compensated in NSCs, and Notch2 is indispensable for the maintenance of NSC quiescence in the adult V-SVZ.
Collapse
Affiliation(s)
- Anna Engler
- Department of Biomedicine, University of Basel, Mattenstrasse 28, 4058 Basel, Switzerland
| | - Chiara Rolando
- Department of Biomedicine, University of Basel, Mattenstrasse 28, 4058 Basel, Switzerland
| | - Claudio Giachino
- Department of Biomedicine, University of Basel, Mattenstrasse 28, 4058 Basel, Switzerland
| | - Ichiko Saotome
- Departments of Neurosurgery and Neuroscience, Yale Program on Neurogenetics, Yale School of Medicine, New Haven, CT 06520, USA
| | - Andrea Erni
- Department of Biomedicine, University of Basel, Mattenstrasse 28, 4058 Basel, Switzerland
| | - Callum Brien
- Department of Biomedicine, University of Basel, Mattenstrasse 28, 4058 Basel, Switzerland; School of Science and Technology, Nottingham Trent University, Clifton Campus, NG11 8NS Nottingham, UK
| | - Runrui Zhang
- Department of Biomedicine, University of Basel, Mattenstrasse 28, 4058 Basel, Switzerland
| | - Ursula Zimber-Strobl
- Department of Gene Vectors, Helmholtz Zentrum München, Marchioninistrasse 25, 81377 Munich, Germany
| | - Freddy Radtke
- EPFL SV ISREC UPRAD, SV 2534 (Bâtiment SV), Station 19, 1015 Lausanne, Switzerland
| | | | - Angeliki Louvi
- Departments of Neurosurgery and Neuroscience, Yale Program on Neurogenetics, Yale School of Medicine, New Haven, CT 06520, USA
| | - Verdon Taylor
- Department of Biomedicine, University of Basel, Mattenstrasse 28, 4058 Basel, Switzerland.
| |
Collapse
|
36
|
Basak O, Krieger TG, Muraro MJ, Wiebrands K, Stange DE, Frias-Aldeguer J, Rivron NC, van de Wetering M, van Es JH, van Oudenaarden A, Simons BD, Clevers H. Troy+ brain stem cells cycle through quiescence and regulate their number by sensing niche occupancy. Proc Natl Acad Sci U S A 2018; 115:E610-E619. [PMID: 29311336 PMCID: PMC5789932 DOI: 10.1073/pnas.1715911114] [Citation(s) in RCA: 114] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The adult mouse subependymal zone provides a niche for mammalian neural stem cells (NSCs). However, the molecular signature, self-renewal potential, and fate behavior of NSCs remain poorly defined. Here we propose a model in which the fate of active NSCs is coupled to the total number of neighboring NSCs in a shared niche. Using knock-in reporter alleles and single-cell RNA sequencing, we show that the Wnt target Tnfrsf19/Troy identifies both active and quiescent NSCs. Quantitative analysis of genetic lineage tracing of individual NSCs under homeostasis or in response to injury reveals rapid expansion of stem-cell number before some return to quiescence. This behavior is best explained by stochastic fate decisions, where stem-cell number within a shared niche fluctuates over time. Fate mapping proliferating cells using a Ki67iresCreER allele confirms that active NSCs reversibly return to quiescence, achieving long-term self-renewal. Our findings suggest a niche-based mechanism for the regulation of NSC fate and number.
Collapse
Affiliation(s)
- Onur Basak
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center Utrecht, 3584 CT, Utrecht, The Netherlands
- Cancer Genomics Netherlands, University Medical Center Utrecht, 3584 GC, Utrecht, The Netherlands
| | - Teresa G Krieger
- The Wellcome Trust/Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge CB2 1QN, United Kingdom
| | - Mauro J Muraro
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center Utrecht, 3584 CT, Utrecht, The Netherlands
- Cancer Genomics Netherlands, University Medical Center Utrecht, 3584 GC, Utrecht, The Netherlands
| | - Kay Wiebrands
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center Utrecht, 3584 CT, Utrecht, The Netherlands
- Cancer Genomics Netherlands, University Medical Center Utrecht, 3584 GC, Utrecht, The Netherlands
| | - Daniel E Stange
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center Utrecht, 3584 CT, Utrecht, The Netherlands
- Cancer Genomics Netherlands, University Medical Center Utrecht, 3584 GC, Utrecht, The Netherlands
| | - Javier Frias-Aldeguer
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center Utrecht, 3584 CT, Utrecht, The Netherlands
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6229ER, Maastricht, The Netherlands
| | - Nicolas C Rivron
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center Utrecht, 3584 CT, Utrecht, The Netherlands
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6229ER, Maastricht, The Netherlands
| | - Marc van de Wetering
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center Utrecht, 3584 CT, Utrecht, The Netherlands
- Cancer Genomics Netherlands, University Medical Center Utrecht, 3584 GC, Utrecht, The Netherlands
- Princess Máxima Centre, 3584 CT, Utrecht, The Netherlands
| | - Johan H van Es
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center Utrecht, 3584 CT, Utrecht, The Netherlands
- Cancer Genomics Netherlands, University Medical Center Utrecht, 3584 GC, Utrecht, The Netherlands
| | - Alexander van Oudenaarden
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center Utrecht, 3584 CT, Utrecht, The Netherlands
- Cancer Genomics Netherlands, University Medical Center Utrecht, 3584 GC, Utrecht, The Netherlands
| | - Benjamin D Simons
- The Wellcome Trust/Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge CB2 1QN, United Kingdom;
- Cavendish Laboratory, Department of Physics, University of Cambridge, Cambridge CB3 0HE, United Kingdom
- Wellcome Trust-Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge CB2 1TN, United Kingdom
| | - Hans Clevers
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center Utrecht, 3584 CT, Utrecht, The Netherlands;
- Cancer Genomics Netherlands, University Medical Center Utrecht, 3584 GC, Utrecht, The Netherlands
- Princess Máxima Centre, 3584 CT, Utrecht, The Netherlands
| |
Collapse
|
37
|
Junghans D, Herzog S. Cnn3 regulates neural tube morphogenesis and neuronal stem cell properties. FEBS J 2018; 285:325-338. [PMID: 29151265 DOI: 10.1111/febs.14338] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 10/25/2017] [Accepted: 11/15/2017] [Indexed: 12/15/2022]
Abstract
Calponin 3 (Cnn3) is a member of the Cnn family of actin-binding molecules that is highly expressed in the mammalian brain and has been shown to control dendritic spine morphology, density, and plasticity by regulating actin cytoskeletal reorganization and dynamics. However, little is known about the role of Cnn3 during embryonic development. In this study, we analyzed mutant animals deficient in Cnn3 to gain a better understanding of its role in brain morphogenesis. Embryos lacking Cnn3 exhibited massive malformation of the developing brain including exoencephaly, closure defects at the rostral neural tube, and strong enlargement of brain tissue. In wild-type animals, we found Cnn3 being localized to the apical lining of the neuroepithelium in close vicinity to beta-Catenin and N-cadherin. By performing immunohistochemistry on beta-Catenin and p-Smad, and furthermore taking advantage of Wnt-reporter animals, we provide evidence that the loss of Cnn3 during development can affect signaling pathways crucial for correct morphogenesis of the neural tube. In addition, we used embryonic neurosphere cultures to investigate the role of Cnn3 in embryonic neuronal stem cells (NSC). Here, we observed that Cnn3 deficiency in NSCs increased the number of newly formed neurospheres and increased neurosphere size without perturbing their differentiation potential. Together, our study provides evidence for an important role of Cnn3 during development of the embryonic brain and in regulating NSC function.
Collapse
Affiliation(s)
- Dirk Junghans
- Institute of Embryology and Stem Cell Biology, Department of Biomedicine, University of Basel, Switzerland
| | - Sebastian Herzog
- Division of Developmental Immunology, Biocenter, Medical University of Innsbruck, Austria
| |
Collapse
|
38
|
Abstract
Neurogenesis is the process of forming neurons and is essential during vertebrate development to produce most of the neurons of the adult brain. However, neurogenesis continues throughout life at distinct locations in the vertebrate brain. Neural stem cells (NSCs) are the origin of both embryonic and adult neurogenesis, but their activity and fate are tightly regulated by their local milieu or niche. In this chapter, we will discuss the role of Notch signaling in the control of neurogenesis and regeneration in the embryo and adult. Notch-dependence is a common feature among NSC populations, we will discuss how differences in Notch signaling might contribute to heterogeneity among adult NSCs. Understanding the fate of multiple NSC populations with distinct functions could be important for effective brain regeneration.
Collapse
|
39
|
Bigas A, Porcheri C. Notch and Stem Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1066:235-263. [DOI: 10.1007/978-3-319-89512-3_12] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
40
|
Area-Specific Regulation of Quiescent Neural Stem Cells by Notch3 in the Adult Mouse Subependymal Zone. J Neurosci 2017; 37:11867-11880. [PMID: 29101245 DOI: 10.1523/jneurosci.0001-17.2017] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2017] [Revised: 10/17/2017] [Accepted: 10/20/2017] [Indexed: 12/15/2022] Open
Abstract
In the adult mammalian brain, neural stem cells (NSCs) generate new neurons throughout the mammal's lifetime. The balance between quiescence and active cell division among NSCs is crucial in producing appropriate numbers of neurons while maintaining the stem cell pool for a long period. The Notch signaling pathway plays a central role in both maintaining quiescent NSCs (qNSCs) and promoting cell division of active NSCs (aNSCs), although no one knows how this pathway regulates these apparently opposite functions. Notch1 has been shown to promote proliferation of aNSCs without affecting qNSCs in the adult mouse subependymal zone (SEZ). In this study, we found that Notch3 is expressed to a higher extent in qNSCs than in aNSCs while Notch1 is preferentially expressed in aNSCs and transit-amplifying progenitors in the adult mouse SEZ. Furthermore, Notch3 is selectively expressed in the lateral and ventral walls of the SEZ. Knockdown of Notch3 in the lateral wall of the adult SEZ increased the division of NSCs. Moreover, deletion of the Notch3 gene resulted in significant reduction of qNSCs specifically in the lateral and ventral walls, compared with the medial and dorsal walls, of the lateral ventricles. Notch3 deletion also reduced the number of qNSCs activated after antimitotic cytosine β-D-arabinofuranoside (Ara-C) treatment. Importantly, Notch3 deletion preferentially reduced specific subtypes of newborn neurons in the olfactory bulb derived from the lateral walls of the SEZ. These results indicate that Notch isoforms differentially control the quiescent and proliferative steps of adult SEZ NSCs in a domain-specific manner.SIGNIFICANCE STATEMENT In the adult mammalian brain, the subependymal zone (SEZ) of the lateral ventricles is the largest neurogenic niche, where neural stem cells (NSCs) generate neurons. In this study, we found that Notch3 plays an important role in the maintenance of quiescent NSCs (qNSCs), while Notch1 has been reported to act as a regulator of actively cycling NSCs. Furthermore, we found that Notch3 is specifically expressed in qNSCs located in the lateral and ventral walls of the lateral ventricles and regulates neuronal production of NSCs in a region-specific manner. Our results indicate that Notch3, by maintaining the quiescence of a subpopulation of NSCs, confers a region-specific heterogeneity among NSCs in the adult SEZ.
Collapse
|
41
|
Boareto M, Iber D, Taylor V. Differential interactions between Notch and ID factors control neurogenesis by modulating Hes factor autoregulation. Development 2017; 144:3465-3474. [PMID: 28974640 PMCID: PMC5665482 DOI: 10.1242/dev.152520] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 08/14/2017] [Indexed: 12/25/2022]
Abstract
During embryonic and adult neurogenesis, neural stem cells (NSCs) generate the correct number and types of neurons in a temporospatial fashion. Control of NSC activity and fate is crucial for brain formation and homeostasis. Neurogenesis in the embryonic and adult brain differ considerably, but Notch signaling and inhibitor of DNA-binding (ID) factors are pivotal in both. Notch and ID factors regulate NSC maintenance; however, it has been difficult to evaluate how these pathways potentially interact. Here, we combined mathematical modeling with analysis of single-cell transcriptomic data to elucidate unforeseen interactions between the Notch and ID factor pathways. During brain development, Notch signaling dominates and directly regulates Id4 expression, preventing other ID factors from inducing NSC quiescence. Conversely, during adult neurogenesis, Notch signaling and Id2/3 regulate neurogenesis in a complementary manner and ID factors can induce NSC maintenance and quiescence in the absence of Notch. Our analyses unveil key molecular interactions underlying NSC maintenance and mechanistic differences between embryonic and adult neurogenesis. Similar Notch and ID factor interactions may be crucial in other stem cell systems. Summary: Computational analysis of transcriptome data from neural stem cells reveals key differences in the synergistic interactions between Notch and inhibitor of DNA-binding factors during embryonic and adult neurogenesis.
Collapse
Affiliation(s)
- Marcelo Boareto
- Department of Biosystems Science and Engineering (D-BSSE), ETH Zurich, Mattenstrasse 26, 4058 Basel, Switzerland .,Swiss Institute of Bioinformatics, Mattenstrasse 26, 4058 Basel, Switzerland
| | - Dagmar Iber
- Department of Biosystems Science and Engineering (D-BSSE), ETH Zurich, Mattenstrasse 26, 4058 Basel, Switzerland .,Swiss Institute of Bioinformatics, Mattenstrasse 26, 4058 Basel, Switzerland
| | - Verdon Taylor
- Department of Biomedicine, University of Basel, Mattenstrasse 28, 4058 Basel, Switzerland
| |
Collapse
|
42
|
Hao XZ, Tian JQ, Yin LK, Zhang XX, Li CC, Feng XY, Yao ZW, Jiang M, Yang YM. MRI detects protective effects of DAPT treatment with modulation of microglia/macrophages at subacute and chronic stages following cerebral ischemia. Mol Med Rep 2017; 16:4493-4500. [PMID: 28849053 PMCID: PMC5647009 DOI: 10.3892/mmr.2017.7200] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 06/08/2017] [Indexed: 12/24/2022] Open
Abstract
Notch homolog 1 (Notch 1) signaling is regarded as a potential therapeutic target for modulating the inflammatory response and exhibiting neuroprotective effects in cerebral injury following stroke. N-[N-(3,5-difluorophenacetyl)-1-alanyl]-S-phenylglycine t-butylester (DAPT) efficiently inhibits activation of the Notch 1 signaling pathway in microglia and may protect brain tissue from ischemic damage. However, the temporal proliferation and morphological alterations of microglia/macrophages throughout progression of the disease, as well as the comprehensive alterations of the whole brain following DAPT treatment, remain to be elucidated. The present study evaluated the temporal proliferation and the morphological alterations of microglia/macrophages over the period of the subacute and chronic stages, in addition to dynamic alterations of brain tissue, using the magnetic resonance imaging (MRI) method, following DAPT treatment. Sprague-Dawley rats (n=40) were subjected to 90 min of middle cerebral artery occlusion and were treated with DAPT (n=20) or acted as controls with no treatment (n=20). The two groups of rats underwent MRI scans prior to the induction of stroke symptoms and at 24 h, 7, 14, 21 and 28 days following the stroke. A total of five rats from each group were sacrificed at 7, 14, 21 and 28 days following induction of stroke. Compared with control rats, the MRI data of the ipsilateral striatum in treated rats revealed ameliorated brain edema at the subacute stage and recovered brain tissue at the chronic stage. In addition to this, treatment attenuated the round-shape and promoted a ramified-shape of microglia/macrophages. The present study confirmed the protective effect of DAPT treatment by dynamically monitoring the cerebral alterations and indicated the possibility of DAPT treatment to alter microglial characteristics to induce a protective effect, via inhibition of the Notch signaling pathway.
Collapse
Affiliation(s)
- Xiao-Zhu Hao
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Jia-Qi Tian
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Le-Kang Yin
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Xiao-Xue Zhang
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Chan-Chan Li
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Xiao-Yuan Feng
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Zhen-Wei Yao
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Min Jiang
- Institutes of Science and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, P.R. China
| | - Yan-Mei Yang
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| |
Collapse
|
43
|
Marathe S, Jaquet M, Annoni JM, Alberi L. Jagged1 Is Altered in Alzheimer's Disease and Regulates Spatial Memory Processing. Front Cell Neurosci 2017; 11:220. [PMID: 28848392 PMCID: PMC5552758 DOI: 10.3389/fncel.2017.00220] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 07/10/2017] [Indexed: 01/17/2023] Open
Abstract
Notch signaling plays an instrumental role in hippocampus-dependent memory formation and recent evidence indicates a displacement of Notch1 and a reduction its activity in hippocampal and cortical neurons from Alzheimer's disease (AD) patients. As Notch activation depends on ligand availability, we investigated whether Jagged1 expression was altered in brain specimen of AD patients. We found that Jagged1 expression was reduced in the CA fields and that there was a gradual reduction of Jagged1 in the cerebrospinal fluid (CSF) with the progression of dementia. Given the role of Notch signaling in memory encoding, we investigated whether targeted loss of Jagged1 in neurons may be responsible for the memory loss seen in AD patients. Using a transgenic mouse model, we show that the targeted loss of Jagged1 expression during adulthood is sufficient to cause spatial memory loss and a reduction in exploration-dependent Notch activation. We also show that Jagged1 is selectively enriched at the presynaptic terminals in mice. Overall, the present data emphasizes the role of the Notch ligand, Jagged1, in memory formation and the potential deficit of the signaling ligand in AD patients.
Collapse
Affiliation(s)
- Swananda Marathe
- Department of Medicine, University of FribourgFribourg, Switzerland
| | - Muriel Jaquet
- Swiss Integrative Center for Human Health SAFribourg, Switzerland
| | - Jean-Marie Annoni
- Department of Medicine, University of FribourgFribourg, Switzerland.,Neurology Clinic, Cantonal HospitalFribourg, Switzerland
| | - Lavinia Alberi
- Department of Medicine, University of FribourgFribourg, Switzerland.,Swiss Integrative Center for Human Health SAFribourg, Switzerland
| |
Collapse
|
44
|
Karimzadeh F, Modarres Mousavi SM, Alipour F, Hosseini Ravandi H, Kovac S, Gorji A. Developmental changes in Notch1 and NLE1 expression in a genetic model of absence epilepsy. Brain Struct Funct 2017; 222:2773-2785. [PMID: 28210849 DOI: 10.1007/s00429-017-1371-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Accepted: 01/16/2017] [Indexed: 01/08/2023]
Abstract
Childhood absence epilepsy (CAE) is an epilepsy syndrome with seizures occurring in the early childhood, highlighting that seizures susceptibility in CAE is dependent on brain development. The Notch 1 signalling pathway is important in brain development, yet the role of the Notch1 signalling pathway in CAE remains elusive. We here explored Notch1 and its modulator notchless homologue 1 (NLE1) expression in WAG/Rij and control rats using immunohistochemistry. Functional Notch 1 effects were assessed in WAG/Rij rats in vivo. WAG/Rij rats lack the developmental increase in cortical Notch1 and NLE 1 mRNA expression seen in controls, and Notch 1 and NLE1 mRNA and protein expression were lower in somatosensory cortices of WAG/Rij rats when compared to controls. This coincided with an overall decreased cortical GFAP expression in the early development in WAG/Rij rats. These effects were region-specific as they were not observed in thalamic tissues. Neuron-to-glia ratio as a marker of the impact of Notch signalling on differentiation was higher in layer 4 of somatosensory cortex of WAG/Rij rats. Acute application of Notch 1 agonist Jagged 1 suppressed, whereas DAPT, a Notch antagonist, facilitated spike and wave discharges (SWDs) in WAG/Rij rats. These findings point to Notch1 as an important signalling pathway in CAE which likely shapes architectural organization of the somatosensory cortex, a region critically involved in developmental epileptogenesis in CAE. More immediate effects of Notch 1 signalling are seen on in vivo SWDs in CAE, pointing to the Notch 1 pathway as a possible treatment target in CAE.
Collapse
MESH Headings
- Age Factors
- Animals
- Antigens, Nuclear/metabolism
- Brain Waves
- Disease Models, Animal
- Electrocorticography
- Epilepsy, Absence/genetics
- Epilepsy, Absence/metabolism
- Epilepsy, Absence/physiopathology
- Gene Expression Regulation, Developmental
- Genetic Predisposition to Disease
- Glial Fibrillary Acidic Protein/metabolism
- Immunohistochemistry
- Jagged-1 Protein/administration & dosage
- Microfilament Proteins/genetics
- Microfilament Proteins/metabolism
- Nerve Tissue Proteins/metabolism
- Phenotype
- Rats, Wistar
- Real-Time Polymerase Chain Reaction
- Receptor, Notch1/drug effects
- Receptor, Notch1/genetics
- Receptor, Notch1/metabolism
- Somatosensory Cortex/drug effects
- Somatosensory Cortex/growth & development
- Somatosensory Cortex/metabolism
- Somatosensory Cortex/physiopathology
- Thalamus/metabolism
- Thalamus/physiopathology
Collapse
Affiliation(s)
- Fariba Karimzadeh
- Shefa Neuroscience Research Centre, Khatam Alanbia Hospital, Tehran, Iran
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | | | - Fatemeh Alipour
- Shefa Neuroscience Research Centre, Khatam Alanbia Hospital, Tehran, Iran
| | | | - Stjepana Kovac
- Department of Neurology, Westfälische Wilhelms-Universität Münster, Münster, Germany
| | - Ali Gorji
- Shefa Neuroscience Research Centre, Khatam Alanbia Hospital, Tehran, Iran.
- Department of Neurology, Westfälische Wilhelms-Universität Münster, Münster, Germany.
- Epilepsy Research Center, Department of Neurosurgery, Westfälische Wilhelms-Universität Münster, Robert-Koch-Straße 45, 48149, Münster, Germany.
- Department of Neuroscience, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
45
|
Neurovascular EGFL7 regulates adult neurogenesis in the subventricular zone and thereby affects olfactory perception. Nat Commun 2017; 8:15922. [PMID: 28656980 PMCID: PMC5493759 DOI: 10.1038/ncomms15922] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 05/11/2017] [Indexed: 01/19/2023] Open
Abstract
Adult neural stem cells reside in a specialized niche in the subventricular zone (SVZ). Throughout life they give rise to adult-born neurons in the olfactory bulb (OB), thus contributing to neural plasticity and pattern discrimination. Here, we show that the neurovascular protein EGFL7 is secreted by endothelial cells and neural stem cells (NSCs) of the SVZ to shape the vascular stem-cell niche. Loss of EGFL7 causes an accumulation of activated NSCs, which display enhanced activity and re-entry into the cell cycle. EGFL7 pushes activated NSCs towards quiescence and neuronal progeny towards differentiation. This is achieved by promoting Dll4-induced Notch signalling at the blood vessel-stem cell interface. Fewer inhibitory neurons form in the OB of EGFL7-knockout mice, which increases the absolute signal conducted from the mitral cell layer of the OB but decreases neuronal network synchronicity. Consequently, EGFL7-knockout mice display severe physiological defects in olfactory behaviour and perception. The vascular stem cell niche regulates the proliferation and differentiation of neural stem cells (NSCs) in the adult subventricular zone. Here the authors identify EGFL7 as a neurovascular regulator of NSCs in vivo; EGFL7-knockout mice show reduced neurogenesis, and exhibit impaired olfactory perception and behaviour.
Collapse
|
46
|
Abstract
Notch signaling is evolutionarily conserved from Drosophila to human. It plays critical roles in neural stem cell maintenance and neurogenesis in the embryonic brain as well as in the adult brain. Notch functions greatly depend on careful regulation and cross-talk with other regulatory mechanisms. Deregulation of Notch signaling is involved in many neurodegenerative diseases and brain disorders. Here, we summarize the fundamental role of Notch in neuronal development and specification and discuss how epigenetic regulation and pathway cross-talk contribute to Notch function. In addition, we cover aberrant alterations of Notch signaling in the diseased brain. The aim of this review is to provide an insight into how Notch signaling works in different contexts to control neurogenesis and its potential effects in diagnoses and therapies of neurodegeneration, brain tumors and disorders.
Collapse
Affiliation(s)
- Runrui Zhang
- Embryology and Stem Cell Biology, Department of Biomedicine, University of Basel, Mattenstrasse 28, 4058, Basel, Switzerland
| | - Anna Engler
- Embryology and Stem Cell Biology, Department of Biomedicine, University of Basel, Mattenstrasse 28, 4058, Basel, Switzerland
| | - Verdon Taylor
- Embryology and Stem Cell Biology, Department of Biomedicine, University of Basel, Mattenstrasse 28, 4058, Basel, Switzerland.
| |
Collapse
|
47
|
Lawal RA, Zhu X, Batey K, Hoffman CM, Georger MA, Radtke F, Hilton MJ, Xing L, Frisch BJ, Calvi LM. The Notch Ligand Jagged1 Regulates the Osteoblastic Lineage by Maintaining the Osteoprogenitor Pool. J Bone Miner Res 2017; 32:1320-1331. [PMID: 28277610 PMCID: PMC5466455 DOI: 10.1002/jbmr.3106] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 01/30/2017] [Accepted: 02/01/2017] [Indexed: 12/16/2022]
Abstract
Notch signaling is critical for osteoblastic differentiation; however, the specific contribution of individual Notch ligands is unknown. Parathyroid hormone (PTH) regulates the Notch ligand Jagged1 in osteoblastic cells. To determine if osteolineage Jagged1 contributes to bone homeostasis, selective deletion of Jagged1 in osteolineage cells was achieved through the presence of Prx1 promoter-driven Cre recombinase expression, targeting mesenchymal stem cells (MSCs) and their progeny (PJag1 mice). PJag1 mice were viable and fertile and did not exhibit any skeletal abnormalities at 2 weeks of age. At 2 months of age, however, PJag1 mice had increased trabecular bone mass compared to wild-type (WT) littermates. Dynamic histomorphometric analysis showed increased osteoblastic activity and increased mineral apposition rate. Immunohistochemical analysis showed increased numbers of osteocalcin-positive mature osteoblasts in PJag1 mice. Also increased phenotypically defined Lin- /CD45- /CD31- /Sca1- /CD51+ osteoblastic cells were measured by flow cytometric analysis. Surprisingly, phenotypically defined Lin- /CD45- /CD31- /Sca1+ /CD51+ MSCs were unchanged in PJag1 mice as measured by flow cytometric analysis. However, functional osteoprogenitor (OP) cell frequency, measured by Von Kossa+ colony formation, was decreased, suggesting that osteolineage Jagged1 contributes to maintenance of the OP pool. The trabecular bone increases were not due to osteoclastic defects, because PJag1 mice had increased bone resorption. Because PTH increases osteoblastic Jagged1, we sought to understand if osteolineage Jagged1 modulates PTH-mediated bone anabolism. Intermittent PTH treatment resulted in a significantly greater increase in BV/TV in PJag1 hind limbs compared to WT. These findings demonstrate a critical role of osteolineage Jagged1 in bone homeostasis, where Jagged1 maintains the transition of OP to maturing osteoblasts. This novel role of Jagged1 not only identifies a regulatory loop maintaining appropriate populations of osteolineage cells, but also provides a novel approach to increase trabecular bone mass, particularly in combination with PTH, through modulation of Jagged1. © 2017 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Rialnat A Lawal
- Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
- Department of Pathology and Laboratory Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Xichao Zhu
- Department of Pathology and Laboratory Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Kaylind Batey
- Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Corey M Hoffman
- Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
- Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Mary A Georger
- Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Freddy Radtke
- Ecole Polytechnique Fédérale de Lausanne, School of Life Sciences, Swiss Institute for Experimental Cancer Research, Lausanne, Vaud, Switzerland
| | - Matthew J Hilton
- Duke Orthopedic, Cellular, and Developmental and Genome Laboratories, Duke University School of Medicine, Durham, NC, USA
| | - Lianping Xing
- Department of Pathology and Laboratory Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Benjamin J Frisch
- Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Laura M Calvi
- Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| |
Collapse
|
48
|
Sarkar S, Mirzaei R, Zemp FJ, Wei W, Senger DL, Robbins SM, Yong VW. Activation of NOTCH Signaling by Tenascin-C Promotes Growth of Human Brain Tumor-Initiating Cells. Cancer Res 2017; 77:3231-3243. [PMID: 28416488 DOI: 10.1158/0008-5472.can-16-2171] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Revised: 02/10/2017] [Accepted: 04/10/2017] [Indexed: 01/08/2023]
Abstract
Oncogenic signaling by NOTCH is elevated in brain tumor-initiating cells (BTIC) in malignant glioma, but the mechanism of its activation is unknown. Here we provide evidence that tenascin-C (TNC), an extracellular matrix protein prominent in malignant glioma, increases NOTCH activity in BTIC to promote their growth. We demonstrate the proximal localization of TNC and BTIC in human glioblastoma specimens and in orthotopic murine xenografts of human BTIC implanted intracranially. In tissue culture, TNC was superior amongst several extracellular matrix proteins in enhancing the sphere-forming capacity of glioma patient-derived BTIC. Exogenously applied or autocrine TNC increased BTIC growth through an α2β1 integrin-mediated mechanism that elevated NOTCH ligand Jagged1 (JAG1). Microarray analyses and confirmatory PCR and Western analyses in BTIC determined that NOTCH signaling components including JAG1, ADAMTS15, and NICD1/2 were elevated in BITC after TNC exposure. Inhibition of γ-secretase and metalloproteinase proteolysis in the NOTCH pathway, or silencing of α2β1 integrin or JAG1, reduced the proliferative effect of TNC on BTIC. Collectively, our findings identified TNC as a pivotal initiator of elevated NOTCH signaling in BTIC and define the establishment of a TN-α2β1-JAG1-NOTCH signaling axis as a candidate therapeutic target in glioma patients. Cancer Res; 77(12); 3231-43. ©2017 AACR.
Collapse
Affiliation(s)
- Susobhan Sarkar
- Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Reza Mirzaei
- Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Franz J Zemp
- Department of Oncology, University of Calgary, Calgary, Alberta, Canada.,Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, Alberta, Canada
| | - Wu Wei
- Department of Oncology, University of Calgary, Calgary, Alberta, Canada.,Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, Alberta, Canada
| | - Donna L Senger
- Department of Oncology, University of Calgary, Calgary, Alberta, Canada.,Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, Alberta, Canada
| | - Stephen M Robbins
- Department of Oncology, University of Calgary, Calgary, Alberta, Canada.,Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, Alberta, Canada
| | - V Wee Yong
- Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada. .,Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
49
|
Dulken BW, Leeman DS, Boutet SC, Hebestreit K, Brunet A. Single-Cell Transcriptomic Analysis Defines Heterogeneity and Transcriptional Dynamics in the Adult Neural Stem Cell Lineage. Cell Rep 2017; 18:777-790. [PMID: 28099854 PMCID: PMC5269583 DOI: 10.1016/j.celrep.2016.12.060] [Citation(s) in RCA: 278] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Revised: 10/04/2016] [Accepted: 12/19/2016] [Indexed: 12/31/2022] Open
Abstract
Neural stem cells (NSCs) in the adult mammalian brain serve as a reservoir for the generation of new neurons, oligodendrocytes, and astrocytes. Here, we use single-cell RNA sequencing to characterize adult NSC populations and examine the molecular identities and heterogeneity of in vivo NSC populations. We find that cells in the NSC lineage exist on a continuum through the processes of activation and differentiation. Interestingly, rare intermediate states with distinct molecular profiles can be identified and experimentally validated, and our analysis identifies putative surface markers and key intracellular regulators for these subpopulations of NSCs. Finally, using the power of single-cell profiling, we conduct a meta-analysis to compare in vivo NSCs and in vitro cultures, distinct fluorescence-activated cell sorting strategies, and different neurogenic niches. These data provide a resource for the field and contribute to an integrative understanding of the adult NSC lineage.
Collapse
Affiliation(s)
- Ben W Dulken
- Department of Genetics, Stanford University, Stanford, CA 94305, USA; Stanford Medical Scientist Training Program, Stanford University, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA 94305, USA
| | - Dena S Leeman
- Department of Genetics, Stanford University, Stanford, CA 94305, USA; Cancer Biology Program, Stanford University, Stanford, CA 94305, USA
| | | | - Katja Hebestreit
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Anne Brunet
- Department of Genetics, Stanford University, Stanford, CA 94305, USA; Glenn Laboratories for the Biology of Aging at Stanford University, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
50
|
Saraiva C, Ferreira L, Bernardino L. Traceable microRNA-124 loaded nanoparticles as a new promising therapeutic tool for Parkinson's disease. NEUROGENESIS 2016; 3:e1256855. [PMID: 28405588 DOI: 10.1080/23262133.2016.1256855] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 09/22/2016] [Accepted: 11/01/2016] [Indexed: 01/09/2023]
Abstract
Parkinson's disease (PD), a neurodegenerative disorder characterized by the selective degeneration of the nigrostriatal dopaminergic pathway, is a major socio-economic burden in modern society. While there is presently no cure for PD, enhancing the number of neural stem cells (NSCs) and/or stimulating their differentiation into new neurons are promising therapeutic strategies. Many proneurogenic factors have been implicated in controlling NSCs activity, including the microRNA (miR)-124. However, current strategies described for the intracellular delivery of miR involve mostly unspecific or inefficient platforms. In Saraiva et al. we developed miR-124 loaded nanoparticles (NPs) able to efficiently deliver miR-124 into neural stem/progenitor cells and boost neuronal differentiation and maturation in vitro. In vivo, the intracerebroventricular injection of miR-124 NPs increased the number of new neurons in the olfactory bulb of healthy and 6-hydroxidopamine (6-OHDA) lesioned mice, a model for PD. Importantly, miR-124 NPs enhanced the migration of new neurons into the 6-OHDA lesioned striatum, culminating in motor function improvement. Given the recent advent of clinical trials for miR-based therapies and the theranostic applications of our NPs, we expect to support the clinical translation of our delivery platform in the context of PD and other neurodegenerative diseases which may benefit from enhancing miR levels.
Collapse
Affiliation(s)
- Cláudia Saraiva
- Health Sciences Research Center, Faculty of Health Sciences, University of Beira Interior , Covilhã, Portugal
| | - Lino Ferreira
- CNC-Center for Neuroscience and Cell Biology, Coimbra, Portugal; Biocant - Center of Innovation in Biotechnology, Cantanhede, Portugal; Institute for Interdisciplinary Research, University of Coimbra (IIIUC), Coimbra, Portugal
| | - Liliana Bernardino
- Health Sciences Research Center, Faculty of Health Sciences, University of Beira Interior , Covilhã, Portugal
| |
Collapse
|