1
|
Cahill R, Wang Y, Xian RP, Lee AJ, Zeng H, Yu B, Tasic B, Abbasi-Asl R. Unsupervised pattern identification in spatial gene expression atlas reveals mouse brain regions beyond established ontology. Proc Natl Acad Sci U S A 2024; 121:e2319804121. [PMID: 39226356 PMCID: PMC11406299 DOI: 10.1073/pnas.2319804121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Accepted: 07/24/2024] [Indexed: 09/05/2024] Open
Abstract
The rapid growth of large-scale spatial gene expression data demands efficient and reliable computational tools to extract major trends of gene expression in their native spatial context. Here, we used stability-driven unsupervised learning (i.e., staNMF) to identify principal patterns (PPs) of 3D gene expression profiles and understand spatial gene distribution and anatomical localization at the whole mouse brain level. Our subsequent spatial correlation analysis systematically compared the PPs to known anatomical regions and ontology from the Allen Mouse Brain Atlas using spatial neighborhoods. We demonstrate that our stable and spatially coherent PPs, whose linear combinations accurately approximate the spatial gene data, are highly correlated with combinations of expert-annotated brain regions. These PPs yield a brain ontology based purely on spatial gene expression. Our PP identification approach outperforms principal component analysis and typical clustering algorithms on the same task. Moreover, we show that the stable PPs reveal marked regional imbalance of brainwide genetic architecture, leading to region-specific marker genes and gene coexpression networks. Our findings highlight the advantages of stability-driven machine learning for plausible biological discovery from dense spatial gene expression data, streamlining tasks that are infeasible by conventional manual approaches.
Collapse
Affiliation(s)
- Robert Cahill
- Department of Neurology, University of California, San Francisco, CA 94143
- UCSF Weill Institute for Neurosciences, San Francisco, CA 94143
| | - Yu Wang
- Department of Statistics, University of California, Berkeley, CA 94720
| | - R Patrick Xian
- Department of Neurology, University of California, San Francisco, CA 94143
- UCSF Weill Institute for Neurosciences, San Francisco, CA 94143
| | - Alex J Lee
- Department of Neurology, University of California, San Francisco, CA 94143
- UCSF Weill Institute for Neurosciences, San Francisco, CA 94143
| | - Hongkui Zeng
- Allen Institute for Brain Science, Seattle, WA 98109
| | - Bin Yu
- Department of Statistics, University of California, Berkeley, CA 94720
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, CA 94720
| | | | - Reza Abbasi-Asl
- Department of Neurology, University of California, San Francisco, CA 94143
- UCSF Weill Institute for Neurosciences, San Francisco, CA 94143
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA 94143
| |
Collapse
|
2
|
Xu S, Li X, Fan P, Li X, Hong Y, Han X, Wu S, Chu C, Chen Y, Xu M, Lin M, Guo X, Liu Y. Single-Cell Transcriptome Landscape and Cell Fate Decoding in Human Brain Organoids after Transplantation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2402287. [PMID: 38711218 PMCID: PMC11267311 DOI: 10.1002/advs.202402287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 04/08/2024] [Indexed: 05/08/2024]
Abstract
Human stem cells and derivatives transplantation are widely used to treat nervous system diseases, while the fate determination of transplanted cells is not well elucidated. To explore cell fate changes of human brain organoids before and after transplantation, human brain organoids are transplanted into prefrontal cortex (PFC) and hippocampus (HIP), respectively. Single-cell sequencing is then performed. According to time-series sample comparison, transplanted cells mainly undergo neural development at 2 months post-transplantation (MPT) and then glial development at 4MPT, respectively. A different brain region sample comparison shows that organoids grafted to PFC have obtained cell fate close to those of host cells in PFC, other than HIP, which may be regulated by the abundant expression of dopamine (DA) and acetylcholine (Ach) in PFC. Meanwhile, morphological complexity of human astrocyte grafts is greater in PFC than in HIP. DA and Ach both activate the calcium activity and increase morphological complexity of astrocytes in vitro. This study demonstrates that human brain organoids receive host niche factor regulation after transplantation, resulting in the alignment of grafted cell fate with implanted brain regions, which may contribute to a better understanding of cell transplantation and regenerative medicine.
Collapse
Affiliation(s)
- Shi‐Bo Xu
- State Key Laboratory of Reproductive MedicineInstitute for Stem Cell and Neural RegenerationSchool of PharmacyKey Laboratory of Targeted Intervention of Cardiovascular DiseaseCollaborative Innovation Center for Cardiovascular Disease Translational MedicineNanjing Medical UniversityNanjing211166P. R. China
- State Key Laboratory of Reproductive MedicineDepartment of NeurobiologySchool of Basic Medical SciencesNanjing Medical UniversityNanjing211166P. R. China
| | - Xin‐Rui Li
- State Key Laboratory of Reproductive MedicineInstitute for Stem Cell and Neural RegenerationSchool of PharmacyKey Laboratory of Targeted Intervention of Cardiovascular DiseaseCollaborative Innovation Center for Cardiovascular Disease Translational MedicineNanjing Medical UniversityNanjing211166P. R. China
| | - Pan Fan
- State Key Laboratory of Reproductive MedicineDepartment of NeurobiologySchool of Basic Medical SciencesNanjing Medical UniversityNanjing211166P. R. China
| | - Xiyang Li
- State Key Laboratory of Reproductive MedicineDepartment of NeurobiologySchool of Basic Medical SciencesNanjing Medical UniversityNanjing211166P. R. China
| | - Yuan Hong
- State Key Laboratory of Reproductive MedicineInstitute for Stem Cell and Neural RegenerationSchool of PharmacyKey Laboratory of Targeted Intervention of Cardiovascular DiseaseCollaborative Innovation Center for Cardiovascular Disease Translational MedicineNanjing Medical UniversityNanjing211166P. R. China
| | - Xiao Han
- State Key Laboratory of Reproductive MedicineInstitute for Stem Cell and Neural RegenerationSchool of PharmacyKey Laboratory of Targeted Intervention of Cardiovascular DiseaseCollaborative Innovation Center for Cardiovascular Disease Translational MedicineNanjing Medical UniversityNanjing211166P. R. China
| | - Shanshan Wu
- State Key Laboratory of Reproductive MedicineInstitute for Stem Cell and Neural RegenerationSchool of PharmacyKey Laboratory of Targeted Intervention of Cardiovascular DiseaseCollaborative Innovation Center for Cardiovascular Disease Translational MedicineNanjing Medical UniversityNanjing211166P. R. China
| | - Chu Chu
- State Key Laboratory of Reproductive MedicineInstitute for Stem Cell and Neural RegenerationSchool of PharmacyKey Laboratory of Targeted Intervention of Cardiovascular DiseaseCollaborative Innovation Center for Cardiovascular Disease Translational MedicineNanjing Medical UniversityNanjing211166P. R. China
| | - Yuejun Chen
- Institute of NeuroscienceKey Laboratory of Primate NeurobiologyCAS Center for Excellence in Brain Science and Intelligence TechnologyChinese Academy of SciencesShanghai200031China
| | - Min Xu
- State Key Laboratory of Reproductive MedicineInstitute for Stem Cell and Neural RegenerationSchool of PharmacyKey Laboratory of Targeted Intervention of Cardiovascular DiseaseCollaborative Innovation Center for Cardiovascular Disease Translational MedicineNanjing Medical UniversityNanjing211166P. R. China
| | - Mingyan Lin
- State Key Laboratory of Reproductive MedicineDepartment of NeurobiologySchool of Basic Medical SciencesNanjing Medical UniversityNanjing211166P. R. China
| | - Xing Guo
- State Key Laboratory of Reproductive MedicineDepartment of NeurobiologySchool of Basic Medical SciencesNanjing Medical UniversityNanjing211166P. R. China
- Co‐innovation Center of NeuroregenerationNantong UniversityJiangsu226019China
| | - Yan Liu
- State Key Laboratory of Reproductive MedicineInstitute for Stem Cell and Neural RegenerationSchool of PharmacyKey Laboratory of Targeted Intervention of Cardiovascular DiseaseCollaborative Innovation Center for Cardiovascular Disease Translational MedicineNanjing Medical UniversityNanjing211166P. R. China
| |
Collapse
|
3
|
Davila-Valencia I, Saad M, Olthoff G, Faulkner M, Charara M, Farnum A, Dysko RC, Zhang Z. Sex specific effects of buprenorphine on adult hippocampal neurogenesis and behavioral outcomes during the acute phase after pediatric traumatic brain injury in mice. Neuropharmacology 2024; 245:109829. [PMID: 38159797 DOI: 10.1016/j.neuropharm.2023.109829] [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: 10/05/2023] [Revised: 12/15/2023] [Accepted: 12/23/2023] [Indexed: 01/03/2024]
Abstract
Traumatic brain injury (TBI) in children often causes cognitive and mental dysfunctions, as well as acute and chronic pain. Adult hippocampal neurogenesis plays a key role in cognition, depression, and pain. Adult hippocampal neurogenesis can be modulated by genetic and environmental factors, such as TBI and opioids. Buprenorphine (BPN), a semisynthetic opioid, is commonly used for pain management in children, however, the effects of BPN on adult hippocampal neurogenesis after pediatric TBI are still unclear. This study investigated the sex-specific effects of BPN on adult hippocampal neurogenesis during acute phase after pediatric TBI. Male and female littermates were randomized on postnatal day 20-21(P20-21) into Sham, TBI+saline and TBI+BPN groups. BPN was administered intraperitoneally to the TBI+BPN mice at 30 min after injury, and then every 6-12 h (h) for 2 days (d). Bromodeoxyuridine (BrdU) was administered intraperitoneally to all groups at 2, 4, 6, and 8-h post-injury. All outcomes were evaluated at 3-d post-BrdU administration. We found that TBI induced significant cognitive impairment, depression, and reduced adult hippocampal neurogenesis in both male and female mice, with more prominent effects in females. BPN significantly improved adult hippocampal neurogenesis and depression in males, but not in females. We further demonstrated that differential expressions of opioid receptors, transcription factors and neuroinflammatory markers at the neurogenic niche might be responsible for the differential effects of BPN in males and females. In conclusion, this study elucidates the effects of BPN on adult hippocampal neurogenesis and behavioral outcomes at the acute phase after pediatric TBI.
Collapse
Affiliation(s)
- Ivan Davila-Valencia
- Department of Natural Sciences, College of Arts, Sciences, and Letters, University of Michigan-Dearborn, 4901 Evergreen Rd, Dearborn, MI, 48128, USA.
| | - Mark Saad
- Department of Natural Sciences, College of Arts, Sciences, and Letters, University of Michigan-Dearborn, 4901 Evergreen Rd, Dearborn, MI, 48128, USA.
| | - Grace Olthoff
- Department of Natural Sciences, College of Arts, Sciences, and Letters, University of Michigan-Dearborn, 4901 Evergreen Rd, Dearborn, MI, 48128, USA.
| | - Megan Faulkner
- Department of Natural Sciences, College of Arts, Sciences, and Letters, University of Michigan-Dearborn, 4901 Evergreen Rd, Dearborn, MI, 48128, USA.
| | - Maysoun Charara
- Department of Natural Sciences, College of Arts, Sciences, and Letters, University of Michigan-Dearborn, 4901 Evergreen Rd, Dearborn, MI, 48128, USA.
| | - Abigail Farnum
- Department of Natural Sciences, College of Arts, Sciences, and Letters, University of Michigan-Dearborn, 4901 Evergreen Rd, Dearborn, MI, 48128, USA.
| | - Robert C Dysko
- Unit for Laboratory Animal Medicine, University of Michigan-Ann Arbor, 2800 Plymouth Rd, Ann Arbor, MI, 48109, USA.
| | - Zhi Zhang
- Department of Natural Sciences, College of Arts, Sciences, and Letters, University of Michigan-Dearborn, 4901 Evergreen Rd, Dearborn, MI, 48128, USA.
| |
Collapse
|
4
|
Šimončičová E, Henderson Pekarik K, Vecchiarelli HA, Lauro C, Maggi L, Tremblay MÈ. Adult Neurogenesis, Learning and Memory. ADVANCES IN NEUROBIOLOGY 2024; 37:221-242. [PMID: 39207695 DOI: 10.1007/978-3-031-55529-9_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Neural plasticity can be defined as the ability of neural circuits to be shaped by external and internal factors. It provides the brain with a capacity for functional and morphological remodelling, with many lines of evidence indicating that these changes are vital for learning and memory formation. The basis of this brain plasticity resides in activity- and experience-driven modifications of synaptic strength, including synaptic formation, elimination or weakening, as well as of modulation of neuronal population, which drive the structural reorganization of neural networks. Recent evidence indicates that brain-resident glial cells actively participate in these processes, suggesting that mechanisms underlying plasticity in the brain are multifaceted. Establishing the 'tripartite' synapse, the role of astrocytes in modulating synaptic transmission in response to neuronal activity was recognized first. Further redefinition of the synapse as 'quad-partite' followed to acknowledge the contribution of microglia which were revealed to affect numerous brain functions via dynamic interactions with synapses, acting as 'synaptic sensors' that respond to neuronal activity and neurotransmitter release, as well as crosstalk with astrocytes. Early studies identified microglial ability to dynamically survey their local brain environment and established their integral role in the active interfacing of environmental stimuli (both internal and external), with brain plasticity and remodelling. Following the introduction to neurogenesis, this chapter details the role that microglia play in regulating neurogenesis in adulthood, specifically as it relates to learning and memory, as well as factors involved in modulation of microglia. Further, a microglial perspective is introduced for the context of environmental enrichment impact on neurogenesis, learning and memory across states of stress, ageing, disease and injury.
Collapse
Affiliation(s)
- Eva Šimončičová
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | | | | | - Clotilde Lauro
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| | - Laura Maggi
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| | - Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada.
| |
Collapse
|
5
|
Sarrafha L, Neavin DR, Parfitt GM, Kruglikov IA, Whitney K, Reyes R, Coccia E, Kareva T, Goldman C, Tipon R, Croft G, Crary JF, Powell JE, Blanchard J, Ahfeldt T. Novel human pluripotent stem cell-derived hypothalamus organoids demonstrate cellular diversity. iScience 2023; 26:107525. [PMID: 37646018 PMCID: PMC10460991 DOI: 10.1016/j.isci.2023.107525] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 06/19/2023] [Accepted: 07/31/2023] [Indexed: 09/01/2023] Open
Abstract
The hypothalamus is a region of the brain that plays an important role in regulating body functions and behaviors. There is a growing interest in human pluripotent stem cells (hPSCs) for modeling diseases that affect the hypothalamus. Here, we established an hPSC-derived hypothalamus organoid differentiation protocol to model the cellular diversity of this brain region. Using an hPSC line with a tyrosine hydroxylase (TH)-TdTomato reporter for dopaminergic neurons (DNs) and other TH-expressing cells, we interrogated DN-specific pathways and functions in electrophysiologically active hypothalamus organoids. Single-cell RNA sequencing (scRNA-seq) revealed diverse neuronal and non-neuronal cell types in mature hypothalamus organoids. We identified several molecularly distinct hypothalamic DN subtypes that demonstrated different developmental maturities. Our in vitro 3D hypothalamus differentiation protocol can be used to study the development of this critical brain structure and can be applied to disease modeling to generate novel therapeutic approaches for disorders centered around the hypothalamus.
Collapse
Affiliation(s)
- Lily Sarrafha
- Nash Family Department of Neuroscience, Mount Sinai, New York, NY 10029, USA
- Department of Neurology, Mount Sinai, New York, NY 10029, USA
- Department of Cell, Developmental and Regenerative Biology, Mount Sinai, New York, NY 10029, USA
- Ronald M. Loeb Center for Alzheimer’s Disease, Mount Sinai, New York, NY 10029, USA
- Friedman Brain Institute, Mount Sinai, New York, NY 10029, USA
- Black Family Stem Cell Institute, Mount Sinai, New York, NY 10029, USA
| | - Drew R. Neavin
- Garvan-Weizmann Centre for Cellular Genomics, Garvan Institute of Medical Research, Darlinghurst, Sydney, NSW 2010, Australia
| | - Gustavo M. Parfitt
- Nash Family Department of Neuroscience, Mount Sinai, New York, NY 10029, USA
- Ronald M. Loeb Center for Alzheimer’s Disease, Mount Sinai, New York, NY 10029, USA
- Friedman Brain Institute, Mount Sinai, New York, NY 10029, USA
- Black Family Stem Cell Institute, Mount Sinai, New York, NY 10029, USA
| | | | - Kristen Whitney
- Nash Family Department of Neuroscience, Mount Sinai, New York, NY 10029, USA
- Ronald M. Loeb Center for Alzheimer’s Disease, Mount Sinai, New York, NY 10029, USA
- Friedman Brain Institute, Mount Sinai, New York, NY 10029, USA
- Department of Pathology, Molecular, and Cell-Based Medicine, Mount Sinai, New York, NY 10029, USA
| | - Ricardo Reyes
- Nash Family Department of Neuroscience, Mount Sinai, New York, NY 10029, USA
- Department of Neurology, Mount Sinai, New York, NY 10029, USA
- Department of Cell, Developmental and Regenerative Biology, Mount Sinai, New York, NY 10029, USA
- Ronald M. Loeb Center for Alzheimer’s Disease, Mount Sinai, New York, NY 10029, USA
- Friedman Brain Institute, Mount Sinai, New York, NY 10029, USA
- Black Family Stem Cell Institute, Mount Sinai, New York, NY 10029, USA
| | - Elena Coccia
- Nash Family Department of Neuroscience, Mount Sinai, New York, NY 10029, USA
- Department of Neurology, Mount Sinai, New York, NY 10029, USA
- Department of Cell, Developmental and Regenerative Biology, Mount Sinai, New York, NY 10029, USA
- Ronald M. Loeb Center for Alzheimer’s Disease, Mount Sinai, New York, NY 10029, USA
- Friedman Brain Institute, Mount Sinai, New York, NY 10029, USA
- Black Family Stem Cell Institute, Mount Sinai, New York, NY 10029, USA
| | - Tatyana Kareva
- Nash Family Department of Neuroscience, Mount Sinai, New York, NY 10029, USA
- Department of Neurology, Mount Sinai, New York, NY 10029, USA
- Department of Cell, Developmental and Regenerative Biology, Mount Sinai, New York, NY 10029, USA
- Ronald M. Loeb Center for Alzheimer’s Disease, Mount Sinai, New York, NY 10029, USA
- Friedman Brain Institute, Mount Sinai, New York, NY 10029, USA
- Black Family Stem Cell Institute, Mount Sinai, New York, NY 10029, USA
| | - Camille Goldman
- Nash Family Department of Neuroscience, Mount Sinai, New York, NY 10029, USA
- Department of Neurology, Mount Sinai, New York, NY 10029, USA
- Department of Cell, Developmental and Regenerative Biology, Mount Sinai, New York, NY 10029, USA
- Ronald M. Loeb Center for Alzheimer’s Disease, Mount Sinai, New York, NY 10029, USA
- Friedman Brain Institute, Mount Sinai, New York, NY 10029, USA
- Black Family Stem Cell Institute, Mount Sinai, New York, NY 10029, USA
| | - Regine Tipon
- New York Stem Cell Foundation, New York, NY 10019, USA
| | - Gist Croft
- New York Stem Cell Foundation, New York, NY 10019, USA
| | - John F. Crary
- Nash Family Department of Neuroscience, Mount Sinai, New York, NY 10029, USA
- Ronald M. Loeb Center for Alzheimer’s Disease, Mount Sinai, New York, NY 10029, USA
- Friedman Brain Institute, Mount Sinai, New York, NY 10029, USA
- Department of Pathology, Molecular, and Cell-Based Medicine, Mount Sinai, New York, NY 10029, USA
- Windreich Department of Artificial Intelligence and Human Health, Mount Sinai, New York, NY 10029, USA
| | - Joseph E. Powell
- Garvan-Weizmann Centre for Cellular Genomics, Garvan Institute of Medical Research, Darlinghurst, Sydney, NSW 2010, Australia
- UNSW Cellular Genomics Futures Institute, University of New South Wales, Kensington, Sydney, NSW 2052, Australia
| | - Joel Blanchard
- Nash Family Department of Neuroscience, Mount Sinai, New York, NY 10029, USA
- Department of Cell, Developmental and Regenerative Biology, Mount Sinai, New York, NY 10029, USA
- Ronald M. Loeb Center for Alzheimer’s Disease, Mount Sinai, New York, NY 10029, USA
- Friedman Brain Institute, Mount Sinai, New York, NY 10029, USA
- Black Family Stem Cell Institute, Mount Sinai, New York, NY 10029, USA
| | - Tim Ahfeldt
- Nash Family Department of Neuroscience, Mount Sinai, New York, NY 10029, USA
- Department of Neurology, Mount Sinai, New York, NY 10029, USA
- Department of Cell, Developmental and Regenerative Biology, Mount Sinai, New York, NY 10029, USA
- Ronald M. Loeb Center for Alzheimer’s Disease, Mount Sinai, New York, NY 10029, USA
- Friedman Brain Institute, Mount Sinai, New York, NY 10029, USA
- Black Family Stem Cell Institute, Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
6
|
Spatio-temporal dynamics enhance cellular diversity, neuronal function and further maturation of human cerebral organoids. Commun Biol 2023; 6:173. [PMID: 36788328 PMCID: PMC9926461 DOI: 10.1038/s42003-023-04547-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 02/02/2023] [Indexed: 02/16/2023] Open
Abstract
The bioengineerined and whole matured human brain organoids stand as highly valuable three-dimensional in vitro brain-mimetic models to recapitulate in vivo brain development, neurodevelopmental and neurodegenerative diseases. Various instructive signals affecting multiple biological processes including morphogenesis, developmental stages, cell fate transitions, cell migration, stem cell function and immune responses have been employed for generation of physiologically functional cerebral organoids. However, the current approaches for maturation require improvement for highly harvestable and functional cerebral organoids with reduced batch-to-batch variabilities. Here, we demonstrate two different engineering approaches, the rotating cell culture system (RCCS) microgravity bioreactor and a newly designed microfluidic platform (µ-platform) to improve harvestability, reproducibility and the survival of high-quality cerebral organoids and compare with those of traditional spinner and shaker systems. RCCS and µ-platform organoids have reached ideal sizes, approximately 95% harvestability, prolonged culture time with Ki-67 + /CD31 + /β-catenin+ proliferative, adhesive and endothelial-like cells and exhibited enriched cellular diversity (abundant neural/glial/ endothelial cell population), structural brain morphogenesis, further functional neuronal identities (glutamate secreting glutamatergic, GABAergic and hippocampal neurons) and synaptogenesis (presynaptic-postsynaptic interaction) during whole human brain development. Both organoids expressed CD11b + /IBA1 + microglia and MBP + /OLIG2 + oligodendrocytes at high levels as of day 60. RCCS and µ-platform organoids showing high levels of physiological fidelity a high level of physiological fidelity can serve as functional preclinical models to test new therapeutic regimens for neurological diseases and benefit from multiplexing.
Collapse
|
7
|
Wang YC, Meng WT, Zhang HF, Zhu J, Wang QL, Mou FF, Guo HD. Lymphangiogenesis, a potential treatment target for myocardial injury. Microvasc Res 2023; 145:104442. [PMID: 36206847 DOI: 10.1016/j.mvr.2022.104442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 07/26/2022] [Accepted: 09/29/2022] [Indexed: 11/07/2022]
Abstract
The lymphatic vascular system is crucial for the regulation of tissue fluid homeostasis, lipid metabolism, and immune function. Cardiac injury quickly leads to myocardial edema, cardiac lymphatic dysfunction, which ultimately results in myocardial fluid imbalance and cardiac dysfunction. Therefore, lymphangiogenesis-targeted therapy may improve the recovery of myocardial function post cardiac ischemia as observed in myocardial infarction (MI). Indeed, a promising strategy for the clinical treatment of MI relies on vascular endothelial growth factor-C (VEGF-C)-targeted therapy, which promotes lymphangiogenesis. However, much effort is needed to identify the mechanisms of lymphatic transport in response to heart disease. This article reviews regulatory factors of lymphangiogenesis, and discusses the effects of lymphangiogenesis on cardiac function after cardiac injury and its regulatory mechanisms. The involvement of stem cells on lymphangiogenesis was also discussed as stem cells could differentiate into lymphatic endothelial cells (LECs) and stimulate phenotype of LECs.
Collapse
Affiliation(s)
- Ya-Chao Wang
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Department of Anatomy, School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Wan-Ting Meng
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Department of Anatomy, School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Hai-Feng Zhang
- Department of Human Anatomy, Xuzhou Medical University, Xuzhou 221004, China
| | - Jing Zhu
- Department of Anatomy, School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Qiang-Li Wang
- School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Fang-Fang Mou
- Department of Anatomy, School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Hai-Dong Guo
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Department of Anatomy, School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
8
|
Jeong M, Jung E, Oh S, Shin SY. Homeobox Protein PROX1 Expression is Negatively Regulated by Histone Deacetylase 1 and c-JUN Complex in MDA-MB-231 Human Breast Cancer Cells. Folia Biol (Praha) 2023; 69:81-90. [PMID: 38206773 DOI: 10.14712/fb2023069030081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2024]
Abstract
Prospero homeobox 1 (PROX1) is a member of the homeobox transcription factor family that plays a critical role in the development of multiple tissues and specification of cell fate. PROX1 expression is differentially regulated based on the cellular context and plays an antagonistic role as a tumour promoter or suppressor in different tumour types. In human breast cancer, PROX1 expression is suppress-ed; however, the molecular mechanism by which it is down-regulated remains poorly understood. Here, we show that ectopic expression of PROX1 reduces the motility and invasiveness of MDA-MB-231 human breast cancer cells, suggesting that PROX1 functions as a negative regulator of tumour invasion in MDA-MB-231 cells. Treatment with histone deacetylase (HDAC) inhibitors up-regulates PROX1 mRNA and protein expression levels. Knockdown of HDAC1 using short hairpin RNA also up-regulates PROX1 mRNA and protein expression levels. We found that HDAC1 interacted with c-JUN at the activator protein (AP)-1-binding site located at -734 to -710 in the PROX1 promoter region to suppress PROX1 expression. In addition, c-JUN N-terminal kinase-mediated c-JUN phosphorylation was found to be crucial for silencing PROX1 expression. In conclusion, PROX1 expression can be silenced by the epigenetic mechanism involved in the complex formation of HDAC1 and c-JUN at the AP-1 site in the PROX1 promoter region in MDA-MB-231 human breast cancer cells. Therefore, this study revealed the epigenetic regulatory mechanism involved in the suppression of PROX1 expression in breast cancer cells.
Collapse
Affiliation(s)
- Munki Jeong
- Department of Biological Sciences, Sanghuh College of Life Sciences, Konkuk University, Seoul 05029, Republic of Korea
| | - Euitaek Jung
- Department of Biological Sciences, Sanghuh College of Life Sciences, Konkuk University, Seoul 05029, Republic of Korea
| | - Sukjin Oh
- Department of Biological Sciences, Sanghuh College of Life Sciences, Konkuk University, Seoul 05029, Republic of Korea
| | - Soon Young Shin
- Department of Biological Sciences, Sanghuh College of Life Sciences, Konkuk University, Seoul 05029, Republic of Korea.
| |
Collapse
|
9
|
Lin Q, Wu L, Chatla S, Chowdhury FA, Atale N, Joseph J, Du W. Hematopoietic stem cell regeneration through paracrine regulation of the Wnt5a/Prox1 signaling axis. J Clin Invest 2022; 132:155914. [PMID: 35703178 PMCID: PMC9197516 DOI: 10.1172/jci155914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 05/05/2022] [Indexed: 11/20/2022] Open
Abstract
The crosstalk between the BM microenvironment (niche) and hematopoietic stem cells (HSCs) is critical for HSC regeneration. Here, we show that in mice, deletion of the Fanconi anemia (FA) genes Fanca and Fancc dampened HSC regeneration through direct effects on HSCs and indirect effects on BM niche cells. FA HSCs showed persistent upregulation of the Wnt target Prox1 in response to total body irradiation (TBI). Accordingly, lineage-specific deletion of Prox1 improved long-term repopulation of the irradiated FA HSCs. Forced expression of Prox1 in WT HSCs mimicked the defective repopulation phenotype of FA HSCs. WT mice but not FA mice showed significant induction by TBI of BM stromal Wnt5a protein. Mechanistically, FA proteins regulated stromal Wnt5a expression, possibly through modulating the Wnt5a transcription activator Pax2. Wnt5a treatment of irradiated FA mice enhanced HSC regeneration. Conversely, Wnt5a neutralization inhibited HSC regeneration after TBI. Wnt5a secreted by LepR+CXCL12+ BM stromal cells inhibited β-catenin accumulation, thereby repressing Prox1 transcription in irradiated HSCs. The detrimental effect of deregulated Wnt5a/Prox1 signaling on HSC regeneration was also observed in patients with FA and aged mice. Irradiation induced upregulation of Prox1 in the HSCs of aged mice, and deletion of Prox1 in aged HSCs improved HSC regeneration. Treatment of aged mice with Wnt5a enhanced hematopoietic repopulation. Collectively, these findings identified the paracrine Wnt5a/Prox1 signaling axis as a regulator of HSC regeneration under conditions of injury and aging.
Collapse
Affiliation(s)
- Qiqi Lin
- Division of Hematology and Oncology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Limei Wu
- Division of Hematology and Oncology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Srinivas Chatla
- Fels Cancer Institute for Personalized Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, USA
| | - Fabliha A Chowdhury
- Division of Hematology and Oncology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Neha Atale
- Division of Hematology and Oncology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Jonathan Joseph
- University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center, Pittsburgh, Pennsylvania, USA.,University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Wei Du
- Division of Hematology and Oncology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
10
|
Jin Y, Gao X, Lu M, Chen G, Yang X, Ren N, Song Y, Hou C, Li J, Liu Q, Gao J. Loss of BAF (mSWI/SNF) chromatin-remodeling ATPase Brg1 causes multiple malformations of cortical development in mice. Hum Mol Genet 2022; 31:3504-3520. [PMID: 35666215 DOI: 10.1093/hmg/ddac127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 05/12/2022] [Accepted: 05/26/2022] [Indexed: 11/13/2022] Open
Abstract
Mutations in genes encoding subunits of the BAF (BRG1/BRM-associated factor) complex cause various neurodevelopmental diseases. However, the underlying pathophysiology remains largely unknown. Here, we analyzed the function of Brg1, a core ATPase of BAF complexes, in the developing cerebral cortex. Loss of Brg1 causes several morphological defects resembling human malformations of cortical development (MCDs), including microcephaly, cortical dysplasia, cobblestone lissencephaly, and periventricular heterotopia. We demonstrated that neural progenitor cell (NPC) renewal, neuronal differentiation, neuronal migration, apoptotic cell death, pial basement membrane, and apical junctional complexes, which are associated with MCD formation, were impaired after Brg1 deletion. Furthermore, transcriptome profiling indicated that a large number of genes were deregulated. The deregulated genes were closely related to MCD formation, and most of these genes were bound by Brg1. Cumulatively, our study indicates an essential role of Brg1 in cortical development and provides a new possible pathogenesis underlying Brg1-based BAF complex-related neurodevelopmental disorders.
Collapse
Affiliation(s)
- Yecheng Jin
- Key Laboratory for Experimental Teratology of the Ministry of Education and Department of Medical Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Xiaotong Gao
- School of Life Science and Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong University, Jinan, Shandong 250100, China
| | - Miaoqing Lu
- Department of Neurology, Ningbo Medical Center Lihuili Hospital, Ningbo University, Ningbo, Zhejiang 315040, China
| | - Ge Chen
- School of Life Science and Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong University, Jinan, Shandong 250100, China
| | - Xiaofan Yang
- Key Laboratory for Experimental Teratology of the Ministry of Education and Department of Medical Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.,Department of Pediatrics, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Naixia Ren
- School of Life Science and Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong University, Jinan, Shandong 250100, China
| | - Yuning Song
- School of Life Science and Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong University, Jinan, Shandong 250100, China
| | - Congzhe Hou
- Department of Reproductive medicine, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250033, China
| | - Jiangxia Li
- Key Laboratory for Experimental Teratology of the Ministry of Education and Department of Medical Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Qiji Liu
- Key Laboratory for Experimental Teratology of the Ministry of Education and Department of Medical Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Jiangang Gao
- School of Laboratory Animal Science, Shandong First Medical University, Jinan, Shandong 250117, China
| |
Collapse
|
11
|
Liang HB, Lai ZH, Tu XQ, Ding KQ, He JR, Yang GY, Sheng H, Zeng LL. MicroRNA-140-5p exacerbates vascular cognitive impairment by inhibiting neurogenesis in the adult mouse hippocampus after global cerebral ischemia. Brain Res Bull 2022; 183:73-83. [DOI: 10.1016/j.brainresbull.2022.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 01/19/2022] [Accepted: 03/01/2022] [Indexed: 11/26/2022]
|
12
|
The Role of Lymphatic Marker Prox-1 in Relation to Brain Tumours. FOLIA VETERINARIA 2021. [DOI: 10.2478/fv-2021-0040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
The homeobox gene, Prox-1 is a transcription factor essential for lymphatic development (lymphangiogenesis) during embryogenesis. It also performs different functions in various tissues such as: retina, lens, liver, pancreas and the central nervous system. Intense expression of Prox-1 has been demonstrated in the developing spinal cord and brain. In adulthood its expression continues in the hippocampus and cerebellum. In adult tissues the process of lymphatic vasculature formation is accompanied under certain pathological conditions such as inflammation, tissue repair and tumour growth. Prox-1 expression is typical for lymphatic vessels; thus it belongs to one of the most specific and widely used mammalian lymphatic endothelial marker in the detection of lymphangiogenesis and lymphatic vessel invasion in oncogenesis. It has been shown that Prox-1 is involved in cancer development and progression. It’s tumour suppressive and oncogenic properties are proven in several human cancers, including brain tumours. Among all body cancers the brain tumours represent the most feared tumours with very limited treatment options and a poor diagnosis. The aim of this paper was to show the current knowledge of the gene Prox-1 with an emphasis on brain tumours, especially in gliomas.
Collapse
|
13
|
Kenney JW, Steadman PE, Young O, Shi MT, Polanco M, Dubaishi S, Covert K, Mueller T, Frankland PW. A 3D adult zebrafish brain atlas (AZBA) for the digital age. eLife 2021; 10:69988. [PMID: 34806976 PMCID: PMC8639146 DOI: 10.7554/elife.69988] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 11/21/2021] [Indexed: 01/19/2023] Open
Abstract
Zebrafish have made significant contributions to our understanding of the vertebrate brain and the neural basis of behavior, earning a place as one of the most widely used model organisms in neuroscience. Their appeal arises from the marriage of low cost, early life transparency, and ease of genetic manipulation with a behavioral repertoire that becomes more sophisticated as animals transition from larvae to adults. To further enhance the use of adult zebrafish, we created the first fully segmented three-dimensional digital adult zebrafish brain atlas (AZBA). AZBA was built by combining tissue clearing, light-sheet fluorescence microscopy, and three-dimensional image registration of nuclear and antibody stains. These images were used to guide segmentation of the atlas into over 200 neuroanatomical regions comprising the entirety of the adult zebrafish brain. As an open source, online (azba.wayne.edu), updatable digital resource, AZBA will significantly enhance the use of adult zebrafish in furthering our understanding of vertebrate brain function in both health and disease.
Collapse
Affiliation(s)
- Justin W Kenney
- Department of Biological Sciences, Wayne State University, Detroit, United States.,Program in Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, Canada
| | - Patrick E Steadman
- Program in Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, Canada
| | - Olivia Young
- Department of Biological Sciences, Wayne State University, Detroit, United States
| | - Meng Ting Shi
- Department of Biological Sciences, Wayne State University, Detroit, United States
| | - Maris Polanco
- Department of Biological Sciences, Wayne State University, Detroit, United States
| | - Saba Dubaishi
- Department of Biological Sciences, Wayne State University, Detroit, United States
| | - Kristopher Covert
- Department of Biological Sciences, Wayne State University, Detroit, United States
| | - Thomas Mueller
- Division of Biology, Kansas State University, Manhattan, United States
| | - Paul W Frankland
- Program in Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, Canada.,Department of Physiology, University of Toronto, Toronto, Canada.,Institute of Medical Sciences, University of Toronto, Toronto, Canada.,Department of Psychology, University of Toronto, Toronto, Canada
| |
Collapse
|
14
|
Charlton-Perkins MA, Friedrich M, Cook TA. Semper's cells in the insect compound eye: Insights into ocular form and function. Dev Biol 2021; 479:126-138. [PMID: 34343526 PMCID: PMC8410683 DOI: 10.1016/j.ydbio.2021.07.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 07/26/2021] [Accepted: 07/28/2021] [Indexed: 11/28/2022]
Abstract
The arthropod compound eye represents one of two major eye types in the animal kingdom and has served as an essential experimental paradigm for defining fundamental mechanisms underlying sensory organ formation, function, and maintenance. One of the most distinguishing features of the compound eye is the highly regular array of lens facets that define individual eye (ommatidial) units. These lens facets are produced by a deeply conserved quartet of cuticle-secreting cells, called Semper cells (SCs). Also widely known as cone cells, SCs were originally identified for their secretion of the dioptric system, i.e. the corneal lens and underlying crystalline cones. Additionally, SCs are now known to execute a diversity of patterning and glial functions in compound eye development and maintenance. Here, we present an integrated account of our current knowledge of SC multifunctionality in the Drosophila compound eye, highlighting emerging gene regulatory modules that may drive the diverse roles for these cells. Drawing comparisons with other deeply conserved retinal glia in the vertebrate single lens eye, this discussion speaks to glial cell origins and opens new avenues for understanding sensory system support programs.
Collapse
Affiliation(s)
- Mark A Charlton-Perkins
- Department of Paediatrics, Wellcome-MRC Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Puddicombe Way, Cambridge, CB2 0AW, United Kingdom
| | - Markus Friedrich
- Department of Biological Sciences, Wayne State University, 5047 Gullen Mall, Detroit, MI, 48202, USA; Department of Ophthalmological, Visual, and Anatomical Sciences, Wayne State University, School of Medicine, 540 East Canfield Avenue, Detroit, MI, 48201, USA
| | - Tiffany A Cook
- Department of Ophthalmological, Visual, and Anatomical Sciences, Wayne State University, School of Medicine, 540 East Canfield Avenue, Detroit, MI, 48201, USA; Center of Molecular Medicine and Genetics, Wayne State University School of Medicine, 540 East Canfield Avenue, Detroit, MI, 48201, USA.
| |
Collapse
|
15
|
Fusz K, Kovács-Öller T, Kóbor P, Szabó-Meleg E, Völgyi B, Buzás P, Telkes I. Regional Variation of Gap Junctional Connections in the Mammalian Inner Retina. Cells 2021; 10:2396. [PMID: 34572046 PMCID: PMC8466939 DOI: 10.3390/cells10092396] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 09/04/2021] [Accepted: 09/07/2021] [Indexed: 11/24/2022] Open
Abstract
The retinas of many species show regional specialisations that are evident in the differences in the processing of visual input from different parts of the visual field. Regional specialisation is thought to reflect an adaptation to the natural visual environment, optical constraints, and lifestyle of the species. Yet, little is known about regional differences in synaptic circuitry. Here, we were interested in the topographical distribution of connexin-36 (Cx36), the major constituent of electrical synapses in the retina. We compared the retinas of mice, rats, and cats to include species with different patterns of regional specialisations in the analysis. First, we used the density of Prox1-immunoreactive amacrine cells as a marker of any regional specialisation, with higher cell density signifying more central regions. Double-labelling experiments showed that Prox1 is expressed in AII amacrine cells in all three species. Interestingly, large Cx36 plaques were attached to about 8-10% of Prox1-positive amacrine cell somata, suggesting the strong electrical coupling of pairs or small clusters of cell bodies. When analysing the regional changes in the volumetric density of Cx36-immunoreactive plaques, we found a tight correlation with the density of Prox1-expressing amacrine cells in the ON, but not in the OFF sublamina in all three species. The results suggest that the relative contribution of electrical synapses to the ON- and OFF-pathways of the retina changes with retinal location, which may contribute to functional ON/OFF asymmetries across the visual field.
Collapse
Affiliation(s)
- Katalin Fusz
- Institute of Physiology, Medical School, University of Pécs, 7624 Pécs, Hungary; (K.F.); (P.K.); (I.T.)
- Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary; (T.K.-Ö.); (E.S.-M.); (B.V.)
- Centre for Neuroscience, University of Pécs, 7624 Pécs, Hungary
| | - Tamás Kovács-Öller
- Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary; (T.K.-Ö.); (E.S.-M.); (B.V.)
- Centre for Neuroscience, University of Pécs, 7624 Pécs, Hungary
- MTA-PTE NAP-2 Retinal Electrical Synapses Research Group, 7624 Pécs, Hungary
| | - Péter Kóbor
- Institute of Physiology, Medical School, University of Pécs, 7624 Pécs, Hungary; (K.F.); (P.K.); (I.T.)
- Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary; (T.K.-Ö.); (E.S.-M.); (B.V.)
- Centre for Neuroscience, University of Pécs, 7624 Pécs, Hungary
| | - Edina Szabó-Meleg
- Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary; (T.K.-Ö.); (E.S.-M.); (B.V.)
- Institute of Biophysics, Medical School, University of Pécs, 7624 Pécs, Hungary
| | - Béla Völgyi
- Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary; (T.K.-Ö.); (E.S.-M.); (B.V.)
- Centre for Neuroscience, University of Pécs, 7624 Pécs, Hungary
- MTA-PTE NAP-2 Retinal Electrical Synapses Research Group, 7624 Pécs, Hungary
- Department of Experimental Zoology and Neurobiology, University of Pécs, 7624 Pécs, Hungary
| | - Péter Buzás
- Institute of Physiology, Medical School, University of Pécs, 7624 Pécs, Hungary; (K.F.); (P.K.); (I.T.)
- Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary; (T.K.-Ö.); (E.S.-M.); (B.V.)
- Centre for Neuroscience, University of Pécs, 7624 Pécs, Hungary
| | - Ildikó Telkes
- Institute of Physiology, Medical School, University of Pécs, 7624 Pécs, Hungary; (K.F.); (P.K.); (I.T.)
- Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary; (T.K.-Ö.); (E.S.-M.); (B.V.)
- Centre for Neuroscience, University of Pécs, 7624 Pécs, Hungary
| |
Collapse
|
16
|
Gustorff C, Scheuer T, Schmitz T, Bührer C, Endesfelder S. GABA B Receptor-Mediated Impairment of Intermediate Progenitor Maturation During Postnatal Hippocampal Neurogenesis of Newborn Rats. Front Cell Neurosci 2021; 15:651072. [PMID: 34421540 PMCID: PMC8377254 DOI: 10.3389/fncel.2021.651072] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 07/12/2021] [Indexed: 12/19/2022] Open
Abstract
The neurotransmitter GABA and its receptors assume essential functions during fetal and postnatal brain development. The last trimester of a human pregnancy and early postnatal life involves a vulnerable period of brain development. In the second half of gestation, there is a developmental shift from depolarizing to hyperpolarizing in the GABAergic system, which might be disturbed by preterm birth. Alterations of the postnatal GABA shift are associated with several neurodevelopmental disorders. In this in vivo study, we investigated neurogenesis in the dentate gyrus (DG) in response to daily administration of pharmacological GABAA (DMCM) and GABAB (CGP 35348) receptor inhibitors to newborn rats. Six-day-old Wistar rats (P6) were daily injected (i.p.) to postnatal day 11 (P11) with DMCM, CGP 35348, or vehicle to determine the effects of both antagonists on postnatal neurogenesis. Due to GABAB receptor blockade by CGP 35348, immunohistochemistry revealed a decrease in the number of NeuroD1 positive intermediate progenitor cells and a reduction of proliferative Nestin-positive neuronal stem cells at the DG. The impairment of hippocampal neurogenesis at this stage of differentiation is in line with a significantly decreased RNA expression of the transcription factors Pax6, Ascl1, and NeuroD1. Interestingly, the number of NeuN-positive postmitotic neurons was not affected by GABAB receptor blockade, although strictly associated transcription factors for postmitotic neurons, Tbr1, Prox1, and NeuroD2, displayed reduced expression levels, suggesting impairment by GABAB receptor antagonization at this stage of neurogenesis. Antagonization of GABAB receptors decreased the expression of neurotrophins (BDNF, NT-3, and NGF). In contrast to the GABAB receptor blockade, the GABAA receptor antagonization revealed no significant changes in cell counts, but an increased transcriptional expression of Tbr1 and Tbr2. We conclude that GABAergic signaling via the metabotropic GABAB receptor is crucial for hippocampal neurogenesis at the time of rapid brain growth and of the postnatal GABA shift. Differentiation and proliferation of intermediate progenitor cells are dependent on GABA. These insights become more pertinent in preterm infants whose developing brains are prematurely exposed to spostnatal stress and predisposed to poor neurodevelopmental disorders, possibly as sequelae of early disruption in GABAergic signaling.
Collapse
Affiliation(s)
- Charlotte Gustorff
- Department of Neonatology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Till Scheuer
- Department of Neonatology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Thomas Schmitz
- Department of Neonatology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Christoph Bührer
- Department of Neonatology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | | |
Collapse
|
17
|
Olmeda D, Cerezo-Wallis D, Castellano-Sanz E, García-Silva S, Peinado H, Soengas MS. Physiological models for in vivo imaging and targeting the lymphatic system: Nanoparticles and extracellular vesicles. Adv Drug Deliv Rev 2021; 175:113833. [PMID: 34147531 DOI: 10.1016/j.addr.2021.113833] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 05/24/2021] [Accepted: 06/11/2021] [Indexed: 02/06/2023]
Abstract
Imaging of the lymphatic vasculature has gained great attention in various fields, not only because lymphatic vessels act as a key draining system in the body, but also for their implication in autoimmune diseases, organ transplant, inflammation and cancer. Thus, neolymphangiogenesis, or the generation of new lymphatics, is typically an early event in the development of multiple tumor types, particularly in aggressive ones such as malignant melanoma. Still, the understanding of how lymphatic endothelial cells get activated at distal (pre)metastatic niches and their impact on therapy is still unclear. Addressing these questions is of particular interest in the case of immune modulators, because endothelial cells may favor or halt inflammatory processes depending on the cellular context. Therefore, there is great interest in visualizing the lymphatic vasculature in vivo. Here, we review imaging tools and mouse models used to analyze the lymphatic vasculature during tumor progression. We also discuss therapeutic approaches based on nanomedicines to target the lymphatic system and the potential use of extracellular vesicles to track and target sentinel lymph nodes. Finally, we summarize main pre-clinical models developed to visualize the lymphatic vasculature in vivo, discussing their applications with a particular focus in metastatic melanoma.
Collapse
Affiliation(s)
- David Olmeda
- Melanoma Laboratory, Molecular Oncology Programme, Spanish National Cancer Research Center (CNIO), Madrid, Spain
| | - Daniela Cerezo-Wallis
- Melanoma Laboratory, Molecular Oncology Programme, Spanish National Cancer Research Center (CNIO), Madrid, Spain; Area of Cell & Developmental Biology, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, 28029, Spain
| | - Elena Castellano-Sanz
- Microenvironment and Metastasis Laboratory, Molecular Oncology Programme, Spanish National Cancer Research Center (CNIO), Madrid, Spain
| | - Susana García-Silva
- Microenvironment and Metastasis Laboratory, Molecular Oncology Programme, Spanish National Cancer Research Center (CNIO), Madrid, Spain
| | - Héctor Peinado
- Microenvironment and Metastasis Laboratory, Molecular Oncology Programme, Spanish National Cancer Research Center (CNIO), Madrid, Spain.
| | - María S Soengas
- Melanoma Laboratory, Molecular Oncology Programme, Spanish National Cancer Research Center (CNIO), Madrid, Spain.
| |
Collapse
|
18
|
Vohra M, Sharma AR, Satyamoorthy K, Rai PS. Pharmacogenomic considerations for repurposing of dexamethasone as a potential drug against SARS-CoV-2 infection. Per Med 2021; 18:389-398. [PMID: 34086487 PMCID: PMC8186476 DOI: 10.2217/pme-2020-0183] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 04/27/2021] [Indexed: 12/15/2022]
Abstract
Immunomodulatory and analgesic effects of dexamethasone are clinically well established, and this synthetic corticosteroid acts as an agonist of glucocorticoid receptors. Early results of the RECOVERY Trial from the United Kingdom and others suggest certain benefits of dexamethasone against COVID-19 chronic patients. The efforts have been acknowledged by World Health Organization with an interim guideline to use in patients with a severe and critical illness. The inherent genetic variations in genes such as CYP3A5, NR3C1, NR3C2, etc., involved in the pharmacokinetic and pharmacodynamic processes may influence dexamethasone's effects as an anti-inflammatory drug. Besides, the drug may influence transcriptome or metabolic changes in the individuals. In the present review, we summarize the reported genetic variations that impact dexamethasone response and discuss dexamethasone-induced changes in transcriptome and metabolome that may influence potential treatment outcome against COVID-19.
Collapse
Affiliation(s)
- Manik Vohra
- Department of Biotechnology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Anu Radha Sharma
- Department of Biotechnology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Kapaettu Satyamoorthy
- Department of Cell & Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Padmalatha S Rai
- Department of Biotechnology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| |
Collapse
|
19
|
Miyashita S, Owa T, Seto Y, Yamashita M, Aida S, Sone M, Ichijo K, Nishioka T, Kaibuchi K, Kawaguchi Y, Taya S, Hoshino M. Cyclin D1 controls development of cerebellar granule cell progenitors through phosphorylation and stabilization of ATOH1. EMBO J 2021; 40:e105712. [PMID: 34057742 PMCID: PMC8280807 DOI: 10.15252/embj.2020105712] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 03/28/2021] [Accepted: 04/01/2021] [Indexed: 12/27/2022] Open
Abstract
During development, neural progenitors are in proliferative and immature states; however, the molecular machinery that cooperatively controls both states remains elusive. Here, we report that cyclin D1 (CCND1) directly regulates both proliferative and immature states of cerebellar granule cell progenitors (GCPs). CCND1 not only accelerates cell cycle but also upregulates ATOH1 protein, an essential transcription factor that maintains GCPs in an immature state. In cooperation with CDK4, CCND1 directly phosphorylates S309 of ATOH1, which inhibits additional phosphorylation at S328 and consequently prevents S328 phosphorylation-dependent ATOH1 degradation. Additionally, PROX1 downregulates Ccnd1 expression by histone deacetylation of Ccnd1 promoter in GCPs, leading to cell cycle exit and differentiation. Moreover, WNT signaling upregulates PROX1 expression in GCPs. These findings suggest that WNT-PROX1-CCND1-ATOH1 signaling cascade cooperatively controls proliferative and immature states of GCPs. We revealed that the expression and phosphorylation levels of these molecules dynamically change during cerebellar development, which are suggested to determine appropriate differentiation rates from GCPs to GCs at distinct developmental stages. This study contributes to understanding the regulatory mechanism of GCPs as well as neural progenitors.
Collapse
Affiliation(s)
- Satoshi Miyashita
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, NCNP, Tokyo, Japan
| | - Tomoo Owa
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, NCNP, Tokyo, Japan
| | - Yusuke Seto
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, NCNP, Tokyo, Japan.,Laboratory of Developmental Systems, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Mariko Yamashita
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, NCNP, Tokyo, Japan
| | - Shogo Aida
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, NCNP, Tokyo, Japan.,Department of Biomolecular Science, Faculty of Science, Toho University, Chiba, Japan
| | - Masaki Sone
- Department of Biomolecular Science, Faculty of Science, Toho University, Chiba, Japan
| | - Kentaro Ichijo
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, NCNP, Tokyo, Japan.,Department of Otolaryngology and Head and Neck Surgery, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Tomoki Nishioka
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kozo Kaibuchi
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoshiya Kawaguchi
- Department of Life Science Frontiers, Center for iPS cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Shinichiro Taya
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, NCNP, Tokyo, Japan
| | - Mikio Hoshino
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, NCNP, Tokyo, Japan
| |
Collapse
|
20
|
The Participation of Microglia in Neurogenesis: A Review. Brain Sci 2021; 11:brainsci11050658. [PMID: 34070012 PMCID: PMC8157831 DOI: 10.3390/brainsci11050658] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 05/12/2021] [Accepted: 05/15/2021] [Indexed: 01/17/2023] Open
Abstract
Adult neurogenesis was one of the most important discoveries of the last century, helping us to better understand brain function. Researchers recently discovered that microglia play an important role in this process. However, various questions remain concerning where, at what stage, and what types of microglia participate. In this review, we demonstrate that certain pools of microglia are determinant cells in different phases of the generation of new neurons. This sheds light on how cells cooperate in order to fine tune brain organization. It also provides us with a better understanding of distinct neuronal pathologies.
Collapse
|
21
|
Kaltezioti V, Foskolou IP, Lavigne MD, Ninou E, Tsampoula M, Fousteri M, Margarity M, Politis PK. Prox1 inhibits neurite outgrowth during central nervous system development. Cell Mol Life Sci 2021; 78:3443-3465. [PMID: 33247761 PMCID: PMC11072475 DOI: 10.1007/s00018-020-03709-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 11/06/2020] [Accepted: 11/11/2020] [Indexed: 12/24/2022]
Abstract
During central nervous system (CNS) development, proper and timely induction of neurite elongation is critical for generating functional, mature neurons, and neuronal networks. Despite the wealth of information on the action of extracellular cues, little is known about the intrinsic gene regulatory factors that control this developmental decision. Here, we report the identification of Prox1, a homeobox transcription factor, as a key player in inhibiting neurite elongation. Although Prox1 promotes acquisition of early neuronal identity and is expressed in nascent post-mitotic neurons, it is heavily down-regulated in the majority of terminally differentiated neurons, indicating a regulatory role in delaying neurite outgrowth in newly formed neurons. Consistently, we show that Prox1 is sufficient to inhibit neurite extension in mouse and human neuroblastoma cell lines. More importantly, Prox1 overexpression suppresses neurite elongation in primary neuronal cultures as well as in the developing mouse brain, while Prox1 knock-down promotes neurite outgrowth. Mechanistically, RNA-Seq analysis reveals that Prox1 affects critical pathways for neuronal maturation and neurite extension. Interestingly, Prox1 strongly inhibits many components of Ca2+ signaling pathway, an important mediator of neurite extension and neuronal maturation. In accordance, Prox1 represses Ca2+ entry upon KCl-mediated depolarization and reduces CREB phosphorylation. These observations suggest that Prox1 acts as a potent suppressor of neurite outgrowth by inhibiting Ca2+ signaling pathway. This action may provide the appropriate time window for nascent neurons to find the correct position in the CNS prior to initiation of neurites and axon elongation.
Collapse
Affiliation(s)
- Valeria Kaltezioti
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, 4 Soranou Efesiou Street, 115 27, Athens, Greece
| | - Iosifina P Foskolou
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, 4 Soranou Efesiou Street, 115 27, Athens, Greece
| | - Matthieu D Lavigne
- Institute for Fundamental Biomedical Research, BSRC 'Alexander Fleming', 34 Fleming Street, Vari, 16672, Athens, Greece
| | - Elpinickie Ninou
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, 4 Soranou Efesiou Street, 115 27, Athens, Greece
| | - Matina Tsampoula
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, 4 Soranou Efesiou Street, 115 27, Athens, Greece
| | - Maria Fousteri
- Institute for Fundamental Biomedical Research, BSRC 'Alexander Fleming', 34 Fleming Street, Vari, 16672, Athens, Greece
| | - Marigoula Margarity
- Laboratory of Human and Animal Physiology, Department of Biology, School of Natural Sciences, University of Patras, 26500, Rio Achaias, Greece
| | - Panagiotis K Politis
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, 4 Soranou Efesiou Street, 115 27, Athens, Greece.
| |
Collapse
|
22
|
Sanders M, Petrasch-Parwez E, Habbes HW, Düring MV, Förster E. Postnatal Developmental Expression Profile Classifies the Indusium Griseum as a Distinct Subfield of the Hippocampal Formation. Front Cell Dev Biol 2021; 8:615571. [PMID: 33511122 PMCID: PMC7835525 DOI: 10.3389/fcell.2020.615571] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 12/07/2020] [Indexed: 11/13/2022] Open
Abstract
The indusium griseum (IG) is a cortical structure overlying the corpus callosum along its anterior–posterior extent. It has been classified either as a vestige of the hippocampus or as an extension of the dentate gyrus via the fasciola cinerea, but its attribution to a specific hippocampal subregion is still under debate. To specify the identity of IG neurons more precisely, we investigated the spatiotemporal expression of calbindin, secretagogin, Necab2, PCP4, and Prox1 in the postnatal mouse IG, fasciola cinerea, and hippocampus. We identified the calcium-binding protein Necab2 as a first reliable marker for the IG and fasciola cinerea throughout postnatal development into adulthood. In contrast, calbindin, secretagogin, and PCP4 were expressed each with a different individual time course during maturation, and at no time point, IG or fasciola cinerea principal neurons expressed Prox1, a transcription factor known to define dentate granule cell fate. Concordantly, in a transgenic mouse line expressing enhanced green fluorescent protein (eGFP) in dentate granule cells, neurons of IG and fasciola cinerea were eGFP-negative. Our findings preclude that IG neurons represent dentate granule cells, as earlier hypothesized, and strongly support the view that the IG is an own hippocampal subfield composed of a distinct neuronal population.
Collapse
Affiliation(s)
- Marie Sanders
- Department of Neuroanatomy and Molecular Brain Research, Ruhr-University Bochum, Bochum, Germany
| | | | - Hans-Werner Habbes
- Department of Neuroanatomy and Molecular Brain Research, Ruhr-University Bochum, Bochum, Germany
| | - Monika V Düring
- Department of Neuroanatomy and Molecular Brain Research, Ruhr-University Bochum, Bochum, Germany
| | - Eckart Förster
- Department of Neuroanatomy and Molecular Brain Research, Ruhr-University Bochum, Bochum, Germany
| |
Collapse
|
23
|
Bui K, Hong YK. Ras Pathways on Prox1 and Lymphangiogenesis: Insights for Therapeutics. Front Cardiovasc Med 2020; 7:597374. [PMID: 33263009 PMCID: PMC7688453 DOI: 10.3389/fcvm.2020.597374] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 10/12/2020] [Indexed: 12/12/2022] Open
Abstract
Over the past couple of decades, lymphatics research has accelerated and gained a much-needed recognition in pathophysiology. As the lymphatic system plays heavy roles in interstitial fluid drainage, immune surveillance and lipid absorption, the ablation or excessive growth of this vasculature could be associated with many complications, from lymphedema to metastasis. Despite their growing importance in cancer, few anti-lymphangiogenic therapies exist today, as they have yet to pass phase 3 clinical trials and acquire FDA approval. As such, many studies are being done to better define the signaling pathways that govern lymphangiogenesis, in hopes of developing new therapeutic approaches to inhibit or stimulate this process. This review will cover our current understanding of the Ras signaling pathways and their interactions with Prox1, the master transcriptional switch involved in specifying lymphatic endothelial cell fate and lymphangiogenesis, in hopes of providing insights to lymphangiogenesis-based therapies.
Collapse
Affiliation(s)
- Khoa Bui
- Department of Surgery, Department of Biochemistry and Molecular Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Young-Kwon Hong
- Department of Surgery, Department of Biochemistry and Molecular Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
24
|
Pomeshchik Y, Klementieva O, Gil J, Martinsson I, Hansen MG, de Vries T, Sancho-Balsells A, Russ K, Savchenko E, Collin A, Vaz AR, Bagnoli S, Nacmias B, Rampon C, Sorbi S, Brites D, Marko-Varga G, Kokaia Z, Rezeli M, Gouras GK, Roybon L. Human iPSC-Derived Hippocampal Spheroids: An Innovative Tool for Stratifying Alzheimer Disease Patient-Specific Cellular Phenotypes and Developing Therapies. Stem Cell Reports 2020; 15:256-273. [PMID: 32589876 PMCID: PMC7363942 DOI: 10.1016/j.stemcr.2020.06.001] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 06/01/2020] [Accepted: 06/01/2020] [Indexed: 12/12/2022] Open
Abstract
The hippocampus is important for memory formation and is severely affected in the brain with Alzheimer disease (AD). Our understanding of early pathogenic processes occurring in hippocampi in AD is limited due to tissue unavailability. Here, we report a chemical approach to rapidly generate free-floating hippocampal spheroids (HSs), from human induced pluripotent stem cells. When used to model AD, both APP and atypical PS1 variant HSs displayed increased Aβ42/Aβ40 peptide ratios and decreased synaptic protein levels, which are common features of AD. However, the two variants differed in tau hyperphosphorylation, protein aggregation, and protein network alterations. NeuroD1-mediated gene therapy in HSs-derived progenitors resulted in modulation of expression of numerous genes, including those involved in synaptic transmission. Thus, HSs can be harnessed to unravel the mechanisms underlying early pathogenic changes in the hippocampi of AD patients, and provide a robust platform for the development of therapeutic strategies targeting early stage AD.
Collapse
Affiliation(s)
- Yuriy Pomeshchik
- iPSC Laboratory for CNS Disease Modeling, Department of Experimental Medical Science, BMC D10, Lund University, Lund SE-221 84, Sweden; Strategic Research Area MultiPark, Lund University, Lund SE-221 84, Sweden; Lund Stem Cell Center, Lund University, Lund SE-221 84, Sweden
| | - Oxana Klementieva
- Strategic Research Area MultiPark, Lund University, Lund SE-221 84, Sweden; Medical Microspectroscopy, Department of Experimental Medical Science, BMC B11, Lund University, Lund SE-221 84, Sweden; Experimental Dementia Research Unit, Department of Experimental Medical Science, BMC B11, Lund University, Lund SE-221 84, Sweden
| | - Jeovanis Gil
- Clinical Protein Science and Imaging, Department of Biomedical Engineering, BMC D13, Lund University, Lund SE-221 84, Sweden
| | - Isak Martinsson
- Strategic Research Area MultiPark, Lund University, Lund SE-221 84, Sweden; Experimental Dementia Research Unit, Department of Experimental Medical Science, BMC B11, Lund University, Lund SE-221 84, Sweden
| | - Marita Grønning Hansen
- Lund Stem Cell Center, Lund University, Lund SE-221 84, Sweden; Laboratory of Stem Cells and Restorative Neurology, Department of Clinical Sciences, BMC B10, Lund University, Lund SE-221 84, Sweden
| | - Tessa de Vries
- iPSC Laboratory for CNS Disease Modeling, Department of Experimental Medical Science, BMC D10, Lund University, Lund SE-221 84, Sweden; Strategic Research Area MultiPark, Lund University, Lund SE-221 84, Sweden; Lund Stem Cell Center, Lund University, Lund SE-221 84, Sweden
| | - Anna Sancho-Balsells
- iPSC Laboratory for CNS Disease Modeling, Department of Experimental Medical Science, BMC D10, Lund University, Lund SE-221 84, Sweden; Strategic Research Area MultiPark, Lund University, Lund SE-221 84, Sweden; Lund Stem Cell Center, Lund University, Lund SE-221 84, Sweden
| | - Kaspar Russ
- iPSC Laboratory for CNS Disease Modeling, Department of Experimental Medical Science, BMC D10, Lund University, Lund SE-221 84, Sweden; Strategic Research Area MultiPark, Lund University, Lund SE-221 84, Sweden; Lund Stem Cell Center, Lund University, Lund SE-221 84, Sweden
| | - Ekaterina Savchenko
- iPSC Laboratory for CNS Disease Modeling, Department of Experimental Medical Science, BMC D10, Lund University, Lund SE-221 84, Sweden; Strategic Research Area MultiPark, Lund University, Lund SE-221 84, Sweden; Lund Stem Cell Center, Lund University, Lund SE-221 84, Sweden
| | - Anna Collin
- Department of Clinical Genetics and Pathology, Office for Medical Services, Lund SE-221 85, Sweden
| | - Ana Rita Vaz
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal; Department of Biochemistry and Human Biology, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Silvia Bagnoli
- Laboratorio di Neurogenetica, Dipartimento di Neuroscienze, Psicologia, Area del Farmaco e Salute del Bambino- NEUROFARBA, Università degli Studi di Firenze, Florence 50134, Italy; IRCCS Fondazione Don Carlo Gnocchi, Florence, Italy
| | - Benedetta Nacmias
- Laboratorio di Neurogenetica, Dipartimento di Neuroscienze, Psicologia, Area del Farmaco e Salute del Bambino- NEUROFARBA, Università degli Studi di Firenze, Florence 50134, Italy; IRCCS Fondazione Don Carlo Gnocchi, Florence, Italy
| | - Claire Rampon
- Centre de Recherches sur la Cognition Animale (CRCA), Centre de Biologie Intégrative (CBI), Université de Toulouse; CNRS, UPS, Toulouse Cedex 9, France
| | - Sandro Sorbi
- Laboratorio di Neurogenetica, Dipartimento di Neuroscienze, Psicologia, Area del Farmaco e Salute del Bambino- NEUROFARBA, Università degli Studi di Firenze, Florence 50134, Italy; IRCCS Fondazione Don Carlo Gnocchi, Florence, Italy
| | - Dora Brites
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal; Department of Biochemistry and Human Biology, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - György Marko-Varga
- Clinical Protein Science and Imaging, Department of Biomedical Engineering, BMC D13, Lund University, Lund SE-221 84, Sweden
| | - Zaal Kokaia
- Lund Stem Cell Center, Lund University, Lund SE-221 84, Sweden; Laboratory of Stem Cells and Restorative Neurology, Department of Clinical Sciences, BMC B10, Lund University, Lund SE-221 84, Sweden
| | - Melinda Rezeli
- Clinical Protein Science and Imaging, Department of Biomedical Engineering, BMC D13, Lund University, Lund SE-221 84, Sweden
| | - Gunnar K Gouras
- Strategic Research Area MultiPark, Lund University, Lund SE-221 84, Sweden; Experimental Dementia Research Unit, Department of Experimental Medical Science, BMC B11, Lund University, Lund SE-221 84, Sweden
| | - Laurent Roybon
- iPSC Laboratory for CNS Disease Modeling, Department of Experimental Medical Science, BMC D10, Lund University, Lund SE-221 84, Sweden; Strategic Research Area MultiPark, Lund University, Lund SE-221 84, Sweden; Lund Stem Cell Center, Lund University, Lund SE-221 84, Sweden.
| |
Collapse
|
25
|
Roy A, Millen KJ, Kapur RP. Hippocampal granule cell dispersion: a non-specific finding in pediatric patients with no history of seizures. Acta Neuropathol Commun 2020; 8:54. [PMID: 32317027 PMCID: PMC7171777 DOI: 10.1186/s40478-020-00928-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 04/03/2020] [Indexed: 12/13/2022] Open
Abstract
Chronic epilepsy has been associated with hippocampal abnormalities like neuronal loss, gliosis and granule cell dispersion. The granule cell layer of a normal human hippocampal dentate gyrus is traditionally regarded as a compact neuron-dense layer. Histopathological studies of surgically resected or autopsied hippocampal samples primarily from temporal lobe epilepsy patients, as well as animal models of epilepsy, describe variable patterns of granule cell dispersion including focal cell clusters, broader thick segments, and bilamination or “tram-tracking”. Although most studies have implicated granule cell dispersion as a specific feature of chronic epilepsy, very few “non-seizure” controls were included in these published investigations. Our retrospective survey of 147 cadaveric pediatric human hippocampi identified identical morphological spectra of granule cell dispersion in both normal and seizure-affected brains. Moreover, sections across the entire antero-posterior axis of a control cadaveric hippocampus revealed repetitive occurrence of different morphologies of the granule cell layer – compact, focally disaggregated and bilaminar. The results indicate that granule cell dispersion is within the spectrum of normal variation and not unique to patients with epilepsy. We speculate that sampling bias has been responsible for an erroneous dogma, which we hope to rectify with this investigation.
Collapse
|
26
|
Morona R, Bandín S, López JM, Moreno N, González A. Amphibian thalamic nuclear organization during larval development and in the adult frog Xenopus laevis: Genoarchitecture and hodological analysis. J Comp Neurol 2020; 528:2361-2403. [PMID: 32162311 DOI: 10.1002/cne.24899] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 03/03/2020] [Accepted: 03/04/2020] [Indexed: 12/18/2022]
Abstract
The early patterning of the thalamus during embryonic development defines rostral and caudal progenitor domains, which are conserved from fishes to mammals. However, the subsequent developmental mechanisms that lead to the adult thalamic configuration have only been investigated for mammals and other amniotes. In this study, we have analyzed in the anuran amphibian Xenopus laevis (an anamniote vertebrate), through larval and postmetamorphic development, the progressive regional expression of specific markers for the rostral (GABA, GAD67, Lhx1, and Nkx2.2) and caudal (Gbx2, VGlut2, Lhx2, Lhx9, and Sox2) domains. In addition, the regional distributions at different developmental stages of other markers such as calcium binding proteins and neuropeptides, helped the identification of thalamic nuclei. It was observed that the two embryonic domains were progressively specified and compartmentalized during premetamorphosis, and cell subpopulations characterized by particular gene expression combinations were located in periventricular, intermediate and superficial strata. During prometamorphosis, three dorsoventral tiers formed from the caudal domain and most pronuclei were defined, which were modified into the definitive nuclear configuration through the metamorphic climax. Mixed cell populations originated from the rostral and caudal domains constitute most of the final nuclei and allowed us to propose additional subdivisions in the adult thalamus, whose main afferent and efferent connections were assessed by tracing techniques under in vitro conditions. This study corroborates shared features of early gene expression patterns in the thalamus between Xenopus and mouse, however, the dynamic changes in gene expression observed at later stages in the amphibian support mechanisms different from those of mammals.
Collapse
Affiliation(s)
- Ruth Morona
- Department of Cell Biology, Faculty of Biology, University Complutense of Madrid, Madrid, Spain
| | - Sandra Bandín
- Department of Cell Biology, Faculty of Biology, University Complutense of Madrid, Madrid, Spain
| | - Jesús M López
- Department of Cell Biology, Faculty of Biology, University Complutense of Madrid, Madrid, Spain
| | - Nerea Moreno
- Department of Cell Biology, Faculty of Biology, University Complutense of Madrid, Madrid, Spain
| | - Agustín González
- Department of Cell Biology, Faculty of Biology, University Complutense of Madrid, Madrid, Spain
| |
Collapse
|
27
|
Ho YC, Srinivasan RS. Lymphatic Vasculature in Energy Homeostasis and Obesity. Front Physiol 2020; 11:3. [PMID: 32038308 PMCID: PMC6987243 DOI: 10.3389/fphys.2020.00003] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 01/03/2020] [Indexed: 12/14/2022] Open
Abstract
Obesity is a leading cause of cardiovascular diseases and cancer. Body mass is regulated by the balance between energy uptake and energy expenditure. The etiology of obesity is determined by multiple factors including genetics, nutrient absorption, and inflammation. Lymphatic vasculature is starting to be appreciated as a critical modulator of metabolism and obesity. The primary function of lymphatic vasculature is to maintain interstitial fluid homeostasis. Lymphatic vessels absorb fluids that extravasate from blood vessels and return them to blood circulation. In addition, lymphatic vessels absorb digested lipids from the intestine and regulate inflammation. Hence, lymphatic vessels could be an exciting target for treating obesity. In this article, we will review our current understanding regarding the relationship between lymphatic vasculature and obesity, and highlight some open questions.
Collapse
Affiliation(s)
- Yen-Chun Ho
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
| | - R. Sathish Srinivasan
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| |
Collapse
|
28
|
Margarido AS, Le Guen L, Falco A, Faure S, Chauvet N, de Santa Barbara P. PROX1 is a specific and dynamic marker of sacral neural crest cells in the chicken intestine. J Comp Neurol 2019; 528:879-889. [PMID: 31658363 DOI: 10.1002/cne.24801] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 08/31/2019] [Accepted: 10/16/2019] [Indexed: 01/29/2023]
Abstract
The enteric nervous system (ENS) is a complex network constituted of neurons and glial cells that ensures the intrinsic innervation of the gastrointestinal tract. ENS cells originate from vagal and sacral neural crest cells that are initially located at the border of the neural tube. In birds, sacral neural crest cells (sNCCs) first give rise to an extramural ganglionated structure (the so-called Nerve of Remak [NoR]) and to the pelvic plexus. Later, sNCCs enter the colon mesenchyme to colonize and contribute to the intrinsic innervation of the caudal part of the gut. However, no specific sNCC marker has been described. Here, we report the expression pattern of prospero-related homeobox 1 (PROX1) in the developing chick colon. PROX1 is a homeobox domain transcription factor that plays a role in cell type specification in various tissues. Using in situ hybridization and immunofluorescence techniques, we showed that PROX1 is expressed in sNCCs localized in the NoR and in the pelvic plexus. Then, using real-time quantitative PCR we found that PROX1 displays a strong and highly dynamic expression pattern during NoR development. Moreover, we demonstrated using in vivo cell tracing, that sNCCs are the source of the PROX1-positive cells within the NoR. Our results indicate that PROX1 is the first marker that specifically identifies sNCCs. This might help to better identify the role of the different neural crest cell populations in distal gut innervation, and consequently to improve the diagnosis of diseases linked to incomplete ENS formation, such as Hirschsprung's disease.
Collapse
Affiliation(s)
| | - Ludovic Le Guen
- PHYMEDEXP, Université de Montpellier, INSERM, CNRS, Montpellier, France
| | - Amandine Falco
- PHYMEDEXP, Université de Montpellier, INSERM, CNRS, Montpellier, France
| | - Sandrine Faure
- PHYMEDEXP, Université de Montpellier, INSERM, CNRS, Montpellier, France
| | - Norbert Chauvet
- PHYMEDEXP, Université de Montpellier, INSERM, CNRS, Montpellier, France
| | | |
Collapse
|
29
|
Saglietti C, La Rosa S, Sykiotis GP, Letovanec I, Bulliard JL, Piana S, Mermod M, Petrova T, Uccella S, Sessa F, Bongiovanni M. Expression of Prox1 in Medullary Thyroid Carcinoma Is Associated with Chromogranin A and Calcitonin Expression and with Ki67 Proliferative Index, but Not with Prognosis. Endocr Pathol 2019; 30:138-145. [PMID: 31001799 DOI: 10.1007/s12022-019-9576-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Medullary thyroid carcinoma (MTC) has been shown to express Prospero homeobox protein 1 (Prox1), a transcription factor whose expression is altered in a variety of human cancers. We conducted a retrospective study on a series of 32 patients with MTC to test the correlation of Prox1 expression in MTC with clinicopathological features and to evaluate its prognostic significance. Correlation of Prox1 immunohistochemical expression with tumor size, proliferative index (Ki67), and calcitonin and CEA serum levels prior to surgery was tested for significant correlations. The difference in Prox1 and Ki67 immunohistochemical expression according to the immunohistochemical staining intensity of CEA, chromogranin A, and calcitonin was tested using the Kruskal-Wallis H test and linear regression analysis. The prognostic value of Prox1 and Ki67 for our patient cohort was assessed by Kaplan-Meier log rank survival analysis. We demonstrated a positive correlation between Prox1 expression and Ki67 index. Prox1 also showed significant difference in expression according to chromogranin A and calcitonin immunohistochemical expression, with higher Prox1 expression in tumors with stronger chromogranin A or calcitonin staining. Prox1 expression did not correlate with PFS or OS based on Kaplan-Meier log rank survival analysis. In conclusion, Prox1 expression in MTC is positively correlated with Ki67 and with the immunohistochemical expression of chromogranin A and calcitonin. However, the present study does not support a role for Prox1 in MTC prognosis.
Collapse
Affiliation(s)
- Chiara Saglietti
- Service of Clinical Pathology, Institute of Pathology, Lausanne University Hospital, Rue du Bugnon 25, CH-1011, Lausanne, Switzerland
| | - Stefano La Rosa
- Service of Clinical Pathology, Institute of Pathology, Lausanne University Hospital, Rue du Bugnon 25, CH-1011, Lausanne, Switzerland
| | - Gerasimos P Sykiotis
- Service of Endocrinology, Diabetology and Metabolism, Lausanne University Hospital, Lausanne, Switzerland
| | - Igor Letovanec
- Service of Clinical Pathology, Institute of Pathology, Lausanne University Hospital, Rue du Bugnon 25, CH-1011, Lausanne, Switzerland
| | - Jean-Luc Bulliard
- Institute of Social and Preventive Medicine, Lausanne University Hospital, Lausanne, Switzerland
| | - Simonetta Piana
- Pathology Unit, Arcispedale Santa Maria Nuova, Azienda USL-IRCCS Reggio Emilia, Reggio Emilia, Italy
| | - Maxime Mermod
- Department of Otolaryngology-Head and Neck Surgery, CHUV, University of Lausanne, Lausanne, Switzerland
| | - Tatiana Petrova
- Department of Oncology, CHUV and University of Lausanne, Lausanne, Switzerland
- Ludwig Institute for Cancer Research Lausanne, Division of Experimental Pathology, CHUV and Swiss Institute for Cancer Research, EPFL, Lausanne, Switzerland
| | - Silvia Uccella
- Pathology Unit, Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Fausto Sessa
- Pathology Unit, Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Massimo Bongiovanni
- Service of Clinical Pathology, Institute of Pathology, Lausanne University Hospital, Rue du Bugnon 25, CH-1011, Lausanne, Switzerland.
| |
Collapse
|
30
|
Duan Z, Deng S, Ji X, Zhao J, Yuan C, Gao H. Nuclear localization of Newcastle disease virus matrix protein promotes virus replication by affecting viral RNA synthesis and transcription and inhibiting host cell transcription. Vet Res 2019; 50:22. [PMID: 30894203 PMCID: PMC6425612 DOI: 10.1186/s13567-019-0640-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 02/11/2019] [Indexed: 01/24/2023] Open
Abstract
Nuclear localization of paramyxovirus proteins is crucial for virus life cycle, including the regulation of viral replication and the evasion of host immunity. We previously showed that a recombinant Newcastle disease virus (NDV) with nuclear localization signal mutation in the matrix (M) protein results in a pathotype change and attenuates viral pathogenicity in chickens. However, little is known about the nuclear localization functions of NDV M protein. In this study, the potential functions of the M protein in the nucleus were investigated. We first demonstrate that nuclear localization of the M protein could not only promote the cytopathogenicity of NDV but also increase viral RNA synthesis and transcription efficiency in DF-1 cells. Using microarray analysis, we found that nuclear localization of the M protein might inhibit host cell transcription, represented by numerous up-regulating genes associated with transcriptional repressor activity and down-regulating genes associated with transcriptional activator activity. The role of representative up-regulated gene prospero homeobox 1 (PROX1) and down-regulated gene aryl hydrocarbon receptor (AHR) in the replication of NDV was then evaluated. The results show that siRNA-mediated knockdown of PROX1 or AHR significantly reduced or increased the viral RNA synthesis and viral replication, respectively, demonstrating the important roles of the expression changes of these genes in NDV replication. Together, our findings demonstrate for the first time that nuclear localization of NDV M protein promotes virus replication by affecting viral RNA synthesis and transcription and inhibiting host cell transcription, improving our understanding of the molecular mechanism of NDV replication and pathogenesis.
Collapse
Affiliation(s)
- Zhiqiang Duan
- Key Laboratory of Animal Genetics, Breeding and Reproduction in The Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang, China. .,College of Animal Science, Guizhou University, Guiyang, China.
| | - Shanshan Deng
- College of Animal Science, Guizhou University, Guiyang, China
| | - Xinqin Ji
- Key Laboratory of Animal Genetics, Breeding and Reproduction in The Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang, China.,College of Animal Science, Guizhou University, Guiyang, China
| | - Jiafu Zhao
- Key Laboratory of Animal Genetics, Breeding and Reproduction in The Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang, China.,College of Animal Science, Guizhou University, Guiyang, China
| | - Chao Yuan
- Key Laboratory of Animal Genetics, Breeding and Reproduction in The Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang, China.,College of Animal Science, Guizhou University, Guiyang, China
| | - Hongbo Gao
- Key Laboratory of Animal Genetics, Breeding and Reproduction in The Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang, China.,College of Animal Science, Guizhou University, Guiyang, China
| |
Collapse
|
31
|
Arimura N, Dewa KI, Okada M, Yanagawa Y, Taya SI, Hoshino M. Comprehensive and cell-type-based characterization of the dorsal midbrain during development. Genes Cells 2018; 24:41-59. [PMID: 30422377 DOI: 10.1111/gtc.12656] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 10/15/2018] [Accepted: 10/24/2018] [Indexed: 01/09/2023]
Abstract
The layer structure has been intensively characterized in the developing neocortex and cerebellum based on the various molecular markers. However, as to the developing dorsal midbrain, comprehensive analyses have not been intensely carried out, and thus, the name as well as the definition of each layer is not commonly shared. Here, we redefined the three layers, such as the ventricular zone, intermediate zone and marginal zone, based on various markers for proliferation and differentiation in embryonic dorsal midbrain. Biphasic Ki67 expression defines the classical VZ, in which there is clear separation of the mitotic and interphase zones. Next, we mapped the distribution of immature neurons to the defined layers, based on markers for glutamatergic and GABAergic lineage. Interestingly, Tbr2 and Neurog2 were expressed in the postmitotic neurons. We also report that active (phosphorylated) JNK is a useful marker to demarcate layers during the embryonic stage. Finally, we validated the final arrival layers of the migratory glutamatergic and GABAergic neurons. These results form a foundation for analyses of brain development, especially in the proliferation and migration of excitatory and inhibitory neurons in the dorsal midbrain.
Collapse
Affiliation(s)
- Nariko Arimura
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, NCNP, Tokyo, Japan
| | - Ken-Ichi Dewa
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, NCNP, Tokyo, Japan.,Department of Pharmacology, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Yamanashi, Japan
| | - Mako Okada
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, NCNP, Tokyo, Japan.,Department of Pharmacology, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Yamanashi, Japan
| | - Yuchio Yanagawa
- Department of Genetic and Behavioral Neuroscience, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Shin-Ichiro Taya
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, NCNP, Tokyo, Japan
| | - Mikio Hoshino
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, NCNP, Tokyo, Japan
| |
Collapse
|
32
|
Atoji Y, Sarkar S. Prox1 mRNA expression in the medial cortex of the turtle (Pseudemys scripta elegans). Neurosci Lett 2018; 687:285-289. [PMID: 30218766 DOI: 10.1016/j.neulet.2018.09.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 09/11/2018] [Indexed: 11/18/2022]
Abstract
The medial cortex of the cerebrum in reptiles is thought to be homologous to the mammalian dentate gyrus, based on cytoarchitectures, fiber connections, and neurochemical profiles. To support this hypothesis, we examined the mRNA expression of vesicular glutamate transporter 1 (vGluT1), a glutamatergic gene marker, and Prox1, a selective gene marker for granule cells of the dentate gyrus, in the turtle medial cortex (zone 2). Reverse transcription-polymerase chain reaction revealed the presence of both mRNAs in the turtle cerebrum. In situ hybridization of zone 2, which is a layer of densely packed neurons in Nissl stains, intensely expressed vGluT1 and Prox1. In zone 3, which is a loosely packed layer, vGluT1 was intensely expressed, whereas Prox1 signals gradated from strong to negative toward zone 4. These findings demonstrate that zone 2 contains glutamatergic neurons and expresses Prox1 mRNA and suggest that zone 2 in the turtle cerebrum is homologous to the mammalian dentate gyrus.
Collapse
Affiliation(s)
- Yasuro Atoji
- Laboratory of Veterinary anatomy, Faculty of Applied Biological Sciences, Gifu University, Gifu 501-1193, Japan.
| | - Sonjoy Sarkar
- Laboratory of Veterinary anatomy, Faculty of Applied Biological Sciences, Gifu University, Gifu 501-1193, Japan
| |
Collapse
|
33
|
The Key Transcription Factor Expression in the Developing Vestibular and Auditory Sensory Organs: A Comprehensive Comparison of Spatial and Temporal Patterns. Neural Plast 2018; 2018:7513258. [PMID: 30410537 PMCID: PMC6205106 DOI: 10.1155/2018/7513258] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 07/31/2018] [Accepted: 09/06/2018] [Indexed: 11/17/2022] Open
Abstract
Inner ear formation requires that a series of cell fate decisions and morphogenetic events occur in a precise temporal and spatial pattern. Previous studies have shown that transcription factors, including Pax2, Sox2, and Prox1, play important roles during the inner ear development. However, the temporospatial expression patterns among these transcription factors are poorly understood. In the current study, we present a comprehensive description of the temporal and spatial expression profiles of Pax2, Sox2, and Prox1 during auditory and vestibular sensory organ development in mice. Using immunohistochemical analyses, we show that Sox2 and Pax2 are both expressed in the prosensory cells (the developing hair cells), but Sox2 is later restricted to only the supporting cells of the organ of Corti. In the vestibular sensory organ, however, the Pax2 expression is localized in hair cells at postnatal day 7, while Sox2 is still expressed in both the hair cells and supporting cells at that time. Prox1 was transiently expressed in the presumptive hair cells and developing supporting cells, and lower Prox1 expression was observed in the vestibular sensory organ compared to the organ of Corti. The different expression patterns of these transcription factors in the developing auditory and vestibular sensory organs suggest that they play different roles in the development of the sensory epithelia and might help to shape the respective sensory structures.
Collapse
|
34
|
Development of lentiviral vectors for efficient glutamatergic-selective gene expression in cultured hippocampal neurons. Sci Rep 2018; 8:15156. [PMID: 30310105 PMCID: PMC6181963 DOI: 10.1038/s41598-018-33509-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 10/01/2018] [Indexed: 01/11/2023] Open
Abstract
Targeting gene expression to a particular subset of neurons helps study the cellular function of the nervous system. Although neuron-specific promoters, such as the synapsin I promoter and the α-CaMKII promoter, are known to exhibit selectivity for excitatory glutamatergic neurons in vivo, the cell type-specificity of these promoters has not been thoroughly tested in culture preparations. Here, by using hippocampal culture preparation from the VGAT-Venus transgenic mice, we examined the ability of five putative promoter sequences of glutamatergic-selective markers including synapsin I, α-CaMKII, the vesicular glutamate transporter 1 (VGLUT1), Dock10 and Prox1. Among these, a genomic fragment containing a 2.1 kb segment upstream of the translation start site (TSS) of the VGLUT1 implemented in a lentiviral vector with the Tet-Off inducible system achieved the highest preferential gene expression in glutamatergic neurons. Analysis of various lengths of the VGLUT1 promoter regions identified a segment between −2.1 kb and −1.4 kb from the TSS as a responsible element for the glutamatergic selectivity. Consistently, expression of channelrhodopsin under this promoter sequence allowed for selective light-evoked activation of excitatory neurons. Thus, the lentiviral system carrying the VGLUT1 promoter fragment can be used to effectively target exogenous gene expression to excitatory glutamatergic neurons in cultures.
Collapse
|
35
|
Louveau A, Herz J, Alme MN, Salvador AF, Dong MQ, Viar KE, Herod SG, Knopp J, Setliff JC, Lupi AL, Da Mesquita S, Frost EL, Gaultier A, Harris TH, Cao R, Hu S, Lukens JR, Smirnov I, Overall CC, Oliver G, Kipnis J. CNS lymphatic drainage and neuroinflammation are regulated by meningeal lymphatic vasculature. Nat Neurosci 2018; 21:1380-1391. [PMID: 30224810 PMCID: PMC6214619 DOI: 10.1038/s41593-018-0227-9] [Citation(s) in RCA: 561] [Impact Index Per Article: 93.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 08/02/2018] [Indexed: 12/25/2022]
Abstract
Neuroinflammatory diseases, such as multiple sclerosis, are characterized by invasion of the brain by autoreactive T cells. The mechanism for how T cells acquire their encephalitogenic phenotype and trigger disease remains, however, unclear. The existence of lymphatic vessels in the meninges indicates a relevant link between the CNS and peripheral immune system, perhaps affecting autoimmunity. Here we demonstrate that meningeal lymphatics fulfill two critical criteria: they assist in the drainage of cerebrospinal fluid components and enable immune cells to enter draining lymph nodes in a CCR7-dependent manner. Unlike other tissues, meningeal lymphatic endothelial cells do not undergo expansion during inflammation, and they express a unique transcriptional signature. Notably, the ablation of meningeal lymphatics diminishes pathology and reduces the inflammatory response of brain-reactive T cells during an animal model of multiple sclerosis. Our findings demonstrate that meningeal lymphatics govern inflammatory processes and immune surveillance of the CNS and pose a valuable target for therapeutic intervention.
Collapse
Affiliation(s)
- Antoine Louveau
- Center for Brain Immunology and Glia (BIG), University of Virginia, Charlottesville, VA, USA.
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA.
| | - Jasmin Herz
- Center for Brain Immunology and Glia (BIG), University of Virginia, Charlottesville, VA, USA
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
| | - Maria Nordheim Alme
- Center for Brain Immunology and Glia (BIG), University of Virginia, Charlottesville, VA, USA
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
- Department of Neurology, Haukeland University Hospital, Bergen, Norway
| | - Andrea Francesca Salvador
- Center for Brain Immunology and Glia (BIG), University of Virginia, Charlottesville, VA, USA
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
- Neuroscience Graduate Program, University of Virginia, Charlottesville, VA, USA
| | - Michael Q Dong
- Center for Brain Immunology and Glia (BIG), University of Virginia, Charlottesville, VA, USA
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
| | - Kenneth E Viar
- Center for Brain Immunology and Glia (BIG), University of Virginia, Charlottesville, VA, USA
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
| | - S Grace Herod
- Center for Brain Immunology and Glia (BIG), University of Virginia, Charlottesville, VA, USA
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
| | - James Knopp
- Center for Brain Immunology and Glia (BIG), University of Virginia, Charlottesville, VA, USA
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
| | - Joshua C Setliff
- Center for Brain Immunology and Glia (BIG), University of Virginia, Charlottesville, VA, USA
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
| | - Alexander L Lupi
- Center for Brain Immunology and Glia (BIG), University of Virginia, Charlottesville, VA, USA
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
| | - Sandro Da Mesquita
- Center for Brain Immunology and Glia (BIG), University of Virginia, Charlottesville, VA, USA
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
| | - Elizabeth L Frost
- Center for Brain Immunology and Glia (BIG), University of Virginia, Charlottesville, VA, USA
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
| | - Alban Gaultier
- Center for Brain Immunology and Glia (BIG), University of Virginia, Charlottesville, VA, USA
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
| | - Tajie H Harris
- Center for Brain Immunology and Glia (BIG), University of Virginia, Charlottesville, VA, USA
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
| | - Rui Cao
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Song Hu
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - John R Lukens
- Center for Brain Immunology and Glia (BIG), University of Virginia, Charlottesville, VA, USA
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
| | - Igor Smirnov
- Center for Brain Immunology and Glia (BIG), University of Virginia, Charlottesville, VA, USA
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
| | - Christopher C Overall
- Center for Brain Immunology and Glia (BIG), University of Virginia, Charlottesville, VA, USA
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
| | - Guillermo Oliver
- Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - Jonathan Kipnis
- Center for Brain Immunology and Glia (BIG), University of Virginia, Charlottesville, VA, USA.
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA.
- Neuroscience Graduate Program, University of Virginia, Charlottesville, VA, USA.
- Gutenberg Research Fellowship Group of Neuroimmunology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn²), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany.
| |
Collapse
|
36
|
A novel environment-evoked transcriptional signature predicts reactivity in single dentate granule neurons. Nat Commun 2018; 9:3084. [PMID: 30082781 PMCID: PMC6079101 DOI: 10.1038/s41467-018-05418-8] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Accepted: 07/06/2018] [Indexed: 12/21/2022] Open
Abstract
Activity-induced remodeling of neuronal circuits is critical for memory formation. This process relies in part on transcription, but neither the rate of activity nor baseline transcription is equal across neuronal cell types. In this study, we isolated mouse hippocampal populations with different activity levels and used single nucleus RNA-seq to compare their transcriptional responses to activation. One hour after novel environment exposure, sparsely active dentate granule (DG) neurons had a much stronger transcriptional response compared to more highly active CA1 pyramidal cells and vasoactive intestinal polypeptide (VIP) interneurons. Activity continued to impact transcription in DG neurons up to 5 h, with increased heterogeneity. By re-exposing the mice to the same environment, we identified a unique transcriptional signature that selects DG neurons for reactivation upon re-exposure to the same environment. These results link transcriptional heterogeneity to functional heterogeneity and identify a transcriptional correlate of memory encoding in individual DG neurons. Single nuclei RNA-seq has been used to characterize transcriptional signature of environment-related activity in cells of the dentate gyrus. Here the authors use this approach to show that whether a neuron will be reactivated in response to re-exposure to a previous environment can be predicted by its transcriptional signature.
Collapse
|
37
|
Chang W, Teng J. Prox1 is essential for oligodendrocyte survival and regulates oligodendrocyte apoptosis via the regulation of NOXA. Acta Biochim Biophys Sin (Shanghai) 2018; 50:709-717. [PMID: 29931031 DOI: 10.1093/abbs/gmy061] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Indexed: 11/13/2022] Open
Abstract
Demyelinating diseases, such as multiple sclerosis, are known to result from acute or chronic injury to the myelin sheath and inadequate remyelination. Its underlying molecular mechanisms, however, remain unclear. The transcription factor prospero homeobox 1 (Prox1) plays an essential role during embryonic development of the central nervous system and cell differentiation. Thus, we aimed to investigate the role of Prox1 in the survival and differentiation of oligodendrocytes. Cell viability was measured by MTT assay. Flow cytometry was conducted to analyze cell apoptosis. Ectopic-Prox1 and shProx1 were used for the overexpression and knockdown respectively of Prox1 in FBD-102b cells. Real-time reverse transcriptase polymerase chain reaction and western blot analysis were used to assess the alterations of signaling pathway-related mRNAs and proteins, respectively. Results showed that Prox1 was upregulated in differentiating oligodendrocytes, and Prox1 knockdown inhibited the differentiation of oligodendrocytes. In addition, overexpression of Prox1 promoted oligodendrocyte differentiation, as shown by the change in myelin basic protein expression. The overexpression of Prox1 had no effect on oligodendrocyte survival, while Prox1 knockdown impaired cell survival. Further study demonstrated that Prox1 knockdown promoted oligodendrocyte apoptosis and activated NOXA, a pro-apoptotic member of the Bcl-2 protein family. Knockdown of NOXA by siRNA abrogated Prox1 knockdown-induced apoptosis. Our findings indicated that Prox1 regulated the differentiation of oligodendrocyte precursor cells via the regulation of NOXA. Therefore, Prox1 could be a potential modulator of demyelinating diseases in clinical settings.
Collapse
Affiliation(s)
- Wenguang Chang
- Department of Neurology, the Center Hospital of Xinxiang, Xinxiang, China
| | - Junfang Teng
- Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
38
|
Abstract
The transcription factor PROX1 is essential for development and cell fate specification. Its function in cancer is context-dependent since PROX1 has been shown to play both oncogenic and tumour suppressive roles. Here, we show that PROX1 suppresses the transcription of MMP14, a metalloprotease involved in angiogenesis and cancer invasion, by binding and suppressing the activity of MMP14 promoter. Prox1 deletion in murine dermal lymphatic vessels in vivo and in human LECs increased MMP14 expression. In a hepatocellular carcinoma cell line expressing high endogenous levels of PROX1, its silencing increased both MMP14 expression and MMP14-dependent invasion in 3D. Moreover, PROX1 ectopic expression reduced the MMP14-dependent 3D invasiveness of breast cancer cells and angiogenic sprouting of blood endothelial cells in conjunction with MMP14 suppression. Our study uncovers a new transcriptional regulatory mechanism of cancer cell invasion and endothelial cell specification.
Collapse
|
39
|
Kong L, Li J, Liu Y, Sun Z, Zhou S, Tang J, Ye T, Wang J, Rosie Xing H. Neuralized1a regulates asymmetric division in mouse Lewis lung carcinoma cells. Life Sci 2018; 206:70-76. [PMID: 29782871 DOI: 10.1016/j.lfs.2018.05.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 05/10/2018] [Accepted: 05/17/2018] [Indexed: 11/25/2022]
Abstract
Asymmetric division (ASD), the unique characteristic of normal stem cells, is regarded as a stemness marker when applied to the study of cancer stem cells (CSCs). However, the role of ASD in the self-renewal of CSCs and its regulation remain largely unknown. Here, we first established a mouse Lewis lung carcinoma CSC cell line that could undergo asymmetric division (LLC-ASD cells) derived from the parental mouse Lewis lung carcinoma cancer cells (LLC-Parental cells). In vitro assessment of stemness by RT-qPCR and western blot analysis of stem cell markers, clonogenic assay (p < 0.001), single cell spheroid formation assay (p < 0.05) and 96-well-plate single-cell cloning assay (p < 0.01) indicated that the LLC-ASD cells exhibited stronger stemness features in comparison to the LLC-Parental cells. In vivo, tumorigenicity of LLC-ASD cells, transplanted subcutaneously to the nude mice, was increased compared to that of LLC-parental cells (p < 0.05). Further, Neuralized1a, a regulator of ASD in normal stem cells, was highly expressed in the LLC-ASD cells. Silencing Neuralized1a expression in LLC-ASD cells by siRNA weakened the stemness features measured by the in vitro assays listed above (p < 0.05). The tumorigenic ability was also decreased in the nude mice upon Neuralized1a silencing (p < 0.05). Collectively, the present study suggests that Neuralized1a regulates the stemness of LLC-ASD cells which could be the new marker and therapeutic target of CSCs.
Collapse
Affiliation(s)
- Liangsheng Kong
- Laboratory of Translational Cancer Stem Cell Research, Institute of Life Sciences, Chongqing Medical University, Chongqing, China; College of Biomedical Engineering, Chongqing Medical University, Chongqing, China
| | - Jingyuan Li
- Laboratory of Translational Cancer Stem Cell Research, Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Yongli Liu
- Laboratory of Translational Cancer Stem Cell Research, Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Zhiwei Sun
- Laboratory of Translational Cancer Stem Cell Research, Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Shixia Zhou
- Laboratory of Translational Cancer Stem Cell Research, Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Junling Tang
- Laboratory of Translational Cancer Stem Cell Research, Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Ting Ye
- Laboratory of Translational Cancer Stem Cell Research, Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Jianyu Wang
- Laboratory of Translational Cancer Stem Cell Research, Institute of Life Sciences, Chongqing Medical University, Chongqing, China.
| | - H Rosie Xing
- Laboratory of Translational Cancer Stem Cell Research, Institute of Life Sciences, Chongqing Medical University, Chongqing, China; State Key Laboratory of Ultrasound Engineering in Medicine Co-Founded by Chongqing and the Ministry of Science and Technology, Chongqing, China.
| |
Collapse
|
40
|
The role of prospero homeobox 1 (PROX1) expression in follicular thyroid carcinoma cells. Oncotarget 2017; 8:114136-114155. [PMID: 29371975 PMCID: PMC5768392 DOI: 10.18632/oncotarget.23167] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 11/29/2017] [Indexed: 12/17/2022] Open
Abstract
The prospero homeobox 1 (Prox1) transcription factor is a key player during embryogenesis and lymphangiogenesis. Altered Prox1 expression has been found in a variety of human cancers, including papillary thyroid carcinoma (PTC). Interestingly, Prox1 may exert tumor suppressive or tumor promoting effect, depending on the tissue context. In this study, we have analyzed Prox1 expression in normal and malignant human thyroid carcinoma cell lines. Moreover, we determined the effect of Prox1 silencing and overexpression on the cellular processes associated with the metastatic potential of tumor cells: proliferation, migration, invasion, apoptosis and anchorage-independent growth, in the follicular thyroid carcinoma (FTC) FTC-133 cell line. We found that Prox1 expression was significantly higher in FTC-derived cells than in PTC-derived cells and normal thyroid, and it was associated with the PI3K/Akt signaling pathway. In the FTC-133 cells, it was associated with cell invasive potential, motility and wound closure capacities, but not with proliferation or apoptosis. Modifying Prox1 expression also induced substantial changes in the cytoskeleton structure and cell morphology. In conclusion, we have shown that Prox1 plays an important role in the development of FTC and that its suppression prevents, whereas its overexpression promotes, the malignant behavior of thyroid follicular cancer cells.
Collapse
|
41
|
Liu Y, Zhang Y, Wang S, Dong QZ, Shen Z, Wang W, Tao S, Gu C, Liu J, Xie Y, Qin LX. Prospero-related homeobox 1 drives angiogenesis of hepatocellular carcinoma through selectively activating interleukin-8 expression. Hepatology 2017. [PMID: 28646551 DOI: 10.1002/hep.29337] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
UNLABELLED Angiogenesis has been proven to play an important role in the progression of hepatocellular carcinoma (HCC). However, the molecular mechanism underlying HCC angiogenesis is not well understood. In this study, Prospero-related homeobox 1 (PROX1) was identified as a novel proangiogenic factor in HCC cell lines and tissues. A strong positive correlation was found between the levels of PROX1 and microvessel density in HCC tissues. Knockdown of PROX1 expression in HCC cells significantly inhibited the in vitro capillary tube formation by human vascular endothelial cells and in vivo angiogenesis of HCC, while overexpression of PROX1 in HCC cells induced the opposite effects. PROX1 and nuclear factor κB p65 expression levels were positively correlated in both HCC tissues and cell lines. PROX1 enhances the nuclear accumulation of p65 and stabilizes p65 by recruiting ubiquitin-specific protease 7 to prevent p65 ubiquitination. Consequently, PROX1 activated nuclear factor κB signaling and selectively promoted expression of the proangiogenic interleukin-8 (IL-8) by epigenetically stimulating the IL-8 promoter. Finally, progression of high PROX1 expression HCC in tumor xenograft mice could be effectively contained by an anti-IL-8 monoclonal antibody. CONCLUSIONS We have identified PROX1 as a crucial promoter of HCC angiogenesis; our study provides an insight into PROX1's function in HCC progression and the potential therapeutic application of anti-IL-8 antibody in high PROX1 expression HCC patients. (Hepatology 2017;66:1894-1909).
Collapse
Affiliation(s)
- Yanfeng Liu
- Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, Shanghai, China.,Key Laboratory of Medical Molecular Virology (MOE & MOH), Institute of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yonglong Zhang
- Department of Biliary-Pancreatic Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Shenghao Wang
- Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, Shanghai, China
| | - Qiong-Zhu Dong
- Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, Shanghai, China.,Institute of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhongliang Shen
- Department of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Wei Wang
- Key Laboratory of Medical Molecular Virology (MOE & MOH), Institute of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shuai Tao
- Key Laboratory of Medical Molecular Virology (MOE & MOH), Institute of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Chenjian Gu
- Key Laboratory of Medical Molecular Virology (MOE & MOH), Institute of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jing Liu
- Key Laboratory of Medical Molecular Virology (MOE & MOH), Institute of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Youhua Xie
- Key Laboratory of Medical Molecular Virology (MOE & MOH), Institute of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lun-Xiu Qin
- Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, Shanghai, China.,Institute of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
42
|
Escobedo N, Oliver G. The Lymphatic Vasculature: Its Role in Adipose Metabolism and Obesity. Cell Metab 2017; 26:598-609. [PMID: 28844882 PMCID: PMC5629116 DOI: 10.1016/j.cmet.2017.07.020] [Citation(s) in RCA: 120] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 06/22/2017] [Accepted: 07/27/2017] [Indexed: 02/07/2023]
Abstract
Obesity is a key risk factor for metabolic and cardiovascular diseases, and although we understand the mechanisms regulating weight and energy balance, the causes of some forms of obesity remain enigmatic. Despite the well-established connections between lymphatics and lipids, and the fact that intestinal lacteals play key roles in dietary fat absorption, the function of the lymphatic vasculature in adipose metabolism has only recently been recognized. It is well established that angiogenesis is tightly associated with the outgrowth of adipose tissue, as expanding adipose tissue requires increased nutrient supply from blood vessels. Results supporting a crosstalk between lymphatic vessels and adipose tissue, and linking lymphatic function with metabolic diseases, obesity, and adipose tissue, also started to accumulate in the last years. Here we review our current knowledge of the mechanisms by which defective lymphatics contribute to obesity and fat accumulation in mouse models, as well as our understanding of the lymphatic-adipose tissue relationship.
Collapse
Affiliation(s)
- Noelia Escobedo
- Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Talca, Chile
| | - Guillermo Oliver
- Center for Vascular and Developmental Biology, Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.
| |
Collapse
|
43
|
Liu W, Crews FT. Persistent Decreases in Adult Subventricular and Hippocampal Neurogenesis Following Adolescent Intermittent Ethanol Exposure. Front Behav Neurosci 2017; 11:151. [PMID: 28855864 PMCID: PMC5557743 DOI: 10.3389/fnbeh.2017.00151] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 07/28/2017] [Indexed: 11/25/2022] Open
Abstract
Neurogenesis in hippocampal dentate gyrus (DG) and subventricular zone (SVZ) matures during adolescence to adult levels. Binge drinking is prevalent in adolescent humans, and could alter brain neurogenesis and maturation in a manner that persists into adulthood. To determine the impact of adolescent binge drinking on adult neurogenesis, Wistar rats received adolescent intermittent ethanol (AIE) exposure (5.0 g/kg/day, i.g., 2 days on/2 days off from postnatal day, P25–P54) and sacrificed on P57 or P95. Neural progenitor cell proliferation, differentiation, survival and maturation using immunohistochemistry was determined in the DG and SVZ. We found that AIE exposure decreased neurogenesis in both brain regions in adulthood (P95). In the DG at P57, AIE exposure resulted in a significant reduction of SOX2+, Tbr2+, Prox1+ and parvalbumin (PV)+IR expression, and at P95 decreased DCX+ and PV+IR expression. AIE exposure also reduced the expression of two cell proliferation markers (Ki67+ and BrdU+IR with 300 mg/kg, 2 h) at P95. The immune signaling molecule β-2 microglobulin+ and the cell death marker activated caspase-3+IR were significantly increased in the DG by AIE exposure. In the SVZ, AIE exposure decreased SOX2+, Mash1+, DCX+ and Dlx2+IR expression at P95, but not at P57. Thus, in adulthood both brain regions have reduced neurogenesis following AIE exposure. To assess progenitor cell survival and maturation, rats were treated with BrdU (150 mg/kg/day, 14 days) to label proliferating cells and were sacrificed weeks later on P95. In the hippocampus DG, AIE exposure increased survival BrdU+ cells which differentiated into Iba1+ microglia. In contrast, SVZ had decreased BrdU+ cells similar to decreased DCX+ neurogenesis. These data indicate that AIE exposure causes a lasting decrease in both adult hippocampal DG and forebrain SVZ neurogenesis with brain regional differences in the AIE response that persist into adulthood.
Collapse
Affiliation(s)
- Wen Liu
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel HillChapel Hill, NC, United States
| | - Fulton T Crews
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel HillChapel Hill, NC, United States
| |
Collapse
|
44
|
Hou H, Fan Q, He W, Suh H, Hu X, Yan R. BACE1 Deficiency Causes Abnormal Neuronal Clustering in the Dentate Gyrus. Stem Cell Reports 2017; 9:217-230. [PMID: 28669600 PMCID: PMC5511112 DOI: 10.1016/j.stemcr.2017.05.030] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Revised: 05/24/2017] [Accepted: 05/24/2017] [Indexed: 12/04/2022] Open
Abstract
BACE1 is validated as Alzheimer's β-secretase and a therapeutic target for Alzheimer's disease. In examining BACE1-null mice, we discovered that BACE1 deficiency develops abnormal clusters of immature neurons, forming doublecortin-positive neuroblasts, in the developing dentate gyrus, mainly in the subpial zone (SPZ). Such clusters were rarely observed in wild-type SPZ and not reported in other mouse models. To understand their origins and fates, we examined how neuroblasts in BACE1-null SPZ mature and migrate during early postnatal development. We show that such neuroblasts are destined to form Prox1-positive granule cells in the dentate granule cell layer, and mainly mature to form excitatory neurons, but not inhibitory neurons. Mechanistically, higher levels of reelin potentially contribute to abnormal neurogenesis and timely migration in BACE1-null SPZ. Altogether, we demonstrate that BACE1 is a critical regulator in forming the dentate granule cell layer through timely maturation and migration of SPZ neuroblasts. BACE1 deficiency causes abnormal neuronal clusters retained in the mouse SPZ Mis-migrated neural progenitor cells in the SPZ are destined to form granule cells Such neural progenitor cells form excitatory neurons but not inhibitor neurons Elevated levels of reelin contribute to abnormal neuronal maturation and migration
Collapse
Affiliation(s)
- Hailong Hou
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue/NC30, Cleveland, OH 44195, USA
| | - Qingyuan Fan
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue/NC30, Cleveland, OH 44195, USA
| | - Wanxia He
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue/NC30, Cleveland, OH 44195, USA
| | - Hoonkyo Suh
- Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Xiangyou Hu
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue/NC30, Cleveland, OH 44195, USA
| | - Riqiang Yan
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue/NC30, Cleveland, OH 44195, USA.
| |
Collapse
|
45
|
Parisot J, Flore G, Bertacchi M, Studer M. COUP-TFI mitotically regulates production and migration of dentate granule cells and modulates hippocampal Cxcr4 expression. Development 2017; 144:2045-2058. [PMID: 28506990 DOI: 10.1242/dev.139949] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Accepted: 04/24/2017] [Indexed: 12/22/2022]
Abstract
Development of the dentate gyrus (DG), the primary gateway for hippocampal inputs, spans embryonic and postnatal stages, and involves complex morphogenetic events. We have previously identified the nuclear receptor COUP-TFI as a novel transcriptional regulator in the postnatal organization and function of the hippocampus. Here, we dissect its role in DG morphogenesis by inactivating it in either granule cell progenitors or granule neurons. Loss of COUP-TFI function in progenitors leads to decreased granule cell proliferative activity, precocious differentiation and increased apoptosis, resulting in a severe DG growth defect in adult mice. COUP-TFI-deficient cells express high levels of the chemokine receptor Cxcr4 and migrate abnormally, forming heterotopic clusters of differentiated granule cells along their paths. Conversely, high COUP-TFI expression levels downregulate Cxcr4 expression, whereas increased Cxcr4 expression in wild-type hippocampal cells affects cell migration. Finally, loss of COUP-TFI in postmitotic cells leads to only minor and transient abnormalities, and to normal Cxcr4 expression. Together, our results indicate that COUP-TFI is required predominantly in DG progenitors for modulating expression of the Cxcr4 receptor during granule cell neurogenesis and migration.
Collapse
Affiliation(s)
| | - Gemma Flore
- Institute of Genetics and Biophysics, CNR, Naples 80131 Italy
| | | | - Michèle Studer
- Université Côte d'Azur, CNRS, Inserm, iBV, Nice 06100, France
| |
Collapse
|
46
|
Furchtgott LA, Melton S, Menon V, Ramanathan S. Discovering sparse transcription factor codes for cell states and state transitions during development. eLife 2017; 6:e20488. [PMID: 28296636 PMCID: PMC5352226 DOI: 10.7554/elife.20488] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 01/31/2017] [Indexed: 12/16/2022] Open
Abstract
Computational analysis of gene expression to determine both the sequence of lineage choices made by multipotent cells and to identify the genes influencing these decisions is challenging. Here we discover a pattern in the expression levels of a sparse subset of genes among cell types in B- and T-cell developmental lineages that correlates with developmental topologies. We develop a statistical framework using this pattern to simultaneously infer lineage transitions and the genes that determine these relationships. We use this technique to reconstruct the early hematopoietic and intestinal developmental trees. We extend this framework to analyze single-cell RNA-seq data from early human cortical development, inferring a neocortical-hindbrain split in early progenitor cells and the key genes that could control this lineage decision. Our work allows us to simultaneously infer both the identity and lineage of cell types as well as a small set of key genes whose expression patterns reflect these relationships.
Collapse
Affiliation(s)
- Leon A Furchtgott
- FAS Center for Systems Biology, Harvard University, Cambridge, United States
- Biophysics Program, Harvard University, Cambridge, United States
| | - Samuel Melton
- FAS Center for Systems Biology, Harvard University, Cambridge, United States
- Harvard Stem Cell Institute, Harvard University, Cambridge, United States
| | - Vilas Menon
- Allen Institute for Brain Science, Seattle, United States
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| | - Sharad Ramanathan
- FAS Center for Systems Biology, Harvard University, Cambridge, United States
- Harvard Stem Cell Institute, Harvard University, Cambridge, United States
- Allen Institute for Brain Science, Seattle, United States
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, United States
- School of Engineering and Applied Sciences, Harvard University, Cambridge, United States
| |
Collapse
|
47
|
Time-lapse imaging reveals highly dynamic structural maturation of postnatally born dentate granule cells in organotypic entorhino-hippocampal slice cultures. Sci Rep 2017; 7:43724. [PMID: 28256620 PMCID: PMC5335612 DOI: 10.1038/srep43724] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 01/27/2017] [Indexed: 12/18/2022] Open
Abstract
Neurogenesis of hippocampal granule cells (GCs) persists throughout mammalian life and is important for learning and memory. How newborn GCs differentiate and mature into an existing circuit during this time period is not yet fully understood. We established a method to visualize postnatally generated GCs in organotypic entorhino-hippocampal slice cultures (OTCs) using retroviral (RV) GFP-labeling and performed time-lapse imaging to study their morphological development in vitro. Using anterograde tracing we could, furthermore, demonstrate that the postnatally generated GCs in OTCs, similar to adult born GCs, grow into an existing entorhino-dentate circuitry. RV-labeled GCs were identified and individual cells were followed for up to four weeks post injection. Postnatally born GCs exhibited highly dynamic structural changes, including dendritic growth spurts but also retraction of dendrites and phases of dendritic stabilization. In contrast, older, presumably prenatally born GCs labeled with an adeno-associated virus (AAV), were far less dynamic. We propose that the high degree of structural flexibility seen in our preparations is necessary for the integration of newborn granule cells into an already existing neuronal circuit of the dentate gyrus in which they have to compete for entorhinal input with cells generated and integrated earlier.
Collapse
|
48
|
Shiraishi A, Muguruma K, Sasai Y. Generation of thalamic neurons from mouse embryonic stem cells. Development 2017; 144:1211-1220. [PMID: 28219951 DOI: 10.1242/dev.144071] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 02/03/2017] [Indexed: 01/08/2023]
Abstract
The thalamus is a diencephalic structure that plays crucial roles in relaying and modulating sensory and motor information to the neocortex. The thalamus develops in the dorsal part of the neural tube at the level of the caudal forebrain. However, the molecular mechanisms that are essential for thalamic differentiation are still unknown. Here, we have succeeded in generating thalamic neurons from mouse embryonic stem cells (mESCs) by modifying the default method that induces the most-anterior neural type in self-organizing culture. A low concentration of the caudalizing factor insulin and a MAPK/ERK kinase inhibitor enhanced the expression of the caudal forebrain markers Otx2 and Pax6. BMP7 promoted an increase in thalamic precursors such as Tcf7l2+/Gbx2+ and Tcf7l2+/Olig3+ cells. mESC thalamic precursors began to express the glutamate transporter vGlut2 and the axon-specific marker VGF, similar to mature projection neurons. The mESC thalamic neurons extended their axons to cortical layers in both organotypic culture and subcortical transplantation. Thus, we have identified the minimum elements sufficient for in vitro generation of thalamic neurons. These findings expand our knowledge of thalamic development.
Collapse
Affiliation(s)
- Atsushi Shiraishi
- Laboratory for Organogenesis and Neurogenesis, RIKEN Center for Developmental Biology, Kobe 650-0047, Japan.,Laboratory of Growth Regulation, Institute for Virus Research, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan.,Laboratory for Cell Asymmetry, RIKEN Center for Developmental Biology, Kobe 650-0047, Japan
| | - Keiko Muguruma
- Laboratory for Organogenesis and Neurogenesis, RIKEN Center for Developmental Biology, Kobe 650-0047, Japan .,Laboratory for Cell Asymmetry, RIKEN Center for Developmental Biology, Kobe 650-0047, Japan
| | - Yoshiki Sasai
- Laboratory for Organogenesis and Neurogenesis, RIKEN Center for Developmental Biology, Kobe 650-0047, Japan
| |
Collapse
|
49
|
Lin F, Li Y, Yan S, Liu S, Qian W, Shen D, Lin Q, Mao W. MicroRNA-181a inhibits tumor proliferation, invasiveness, and metastasis and is downregulated in gastric cancer. Oncol Res 2017; 22:75-84. [PMID: 25706394 PMCID: PMC7838452 DOI: 10.3727/096504014x14024160459203] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
MicroRNAs (miRNAs) play crucial roles in the development and progression of human cancers, including gastric cancer. The discovery of miRNAs may provide a new and powerful tool for studying the mechanism, diagnosis, and treatment of gastric cancer. Here we show that miR-181a levels were significantly downregulated in gastric cancer tissues compared with the adjacent normal regions in 80 paired samples. Moreover, the lower levels of miR-181a were associated with the pM or pTNM stage in clinical gastric cancer patients. In addition, the ectopic expression of miR-181a in the gastric cancer cell line HGC-27 inhibited cell proliferation, cell migration, and invasion by directly interacting with the mRNA encoding the oncogenic factor Prox1. Taken together, our results indicate that miR-181a might act as a tumor suppressor in gastric cancer, which may provide a novel diagnostic and therapeutic option for human gastric cancer in the near future.
Collapse
Affiliation(s)
- Feng Lin
- Department of Oncology, the Affiliated Jiangyin Hospital of Southeast University Medical College, Jiangyin, Jiangsu, PR China
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Transmembrane protein 108 is required for glutamatergic transmission in dentate gyrus. Proc Natl Acad Sci U S A 2017; 114:1177-1182. [PMID: 28096412 DOI: 10.1073/pnas.1618213114] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Neurotransmission in dentate gyrus (DG) is critical for spatial coding, learning memory, and emotion processing. Although DG dysfunction is implicated in psychiatric disorders, including schizophrenia, underlying pathological mechanisms remain unclear. Here we report that transmembrane protein 108 (Tmem108), a novel schizophrenia susceptibility gene, is highly enriched in DG granule neurons and its expression increased at the postnatal period critical for DG development. Tmem108 is specifically expressed in the nervous system and enriched in the postsynaptic density fraction. Tmem108-deficient neurons form fewer and smaller spines, suggesting that Tmem108 is required for spine formation and maturation. In agreement, excitatory postsynaptic currents of DG granule neurons were decreased in Tmem108 mutant mice, indicating a hypofunction of glutamatergic activity. Further cell biological studies indicate that Tmem108 is necessary for surface expression of AMPA receptors. Tmem108-deficient mice display compromised sensorimotor gating and cognitive function. Together, these observations indicate that Tmem108 plays a critical role in regulating spine development and excitatory transmission in DG granule neurons. When Tmem108 is mutated, mice displayed excitatory/inhibitory imbalance and behavioral deficits relevant to schizophrenia, revealing potential pathophysiological mechanisms of schizophrenia.
Collapse
|