1
|
Dolique T, Baudet S, Charron F, Ferent J. A central role for Numb/Nbl in multiple Shh-mediated axon repulsion processes. iScience 2025; 28:112293. [PMID: 40276749 PMCID: PMC12018091 DOI: 10.1016/j.isci.2025.112293] [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: 11/25/2024] [Revised: 02/12/2025] [Accepted: 03/21/2025] [Indexed: 04/26/2025] Open
Abstract
Sonic hedgehog (Shh) is an axon guidance molecule that can act as either a chemorepellent or a chemoattractant, depending on the neuron type and their developmental stage. In the developing spinal cord, Shh initially attracts commissural axons to the floor plate and later repels them after they cross the midline. In the developing visual system, Shh repels ipsilateral retinal ganglion cell (iRGC) axons at the optic chiasm. Although Shh requires the endocytic adaptor Numb for attraction of spinal commissural axons, the molecular mechanisms underlying Shh dual function in attraction and repulsion are still unclear. In this study, we show that Numb is essential for two Shh-mediated repulsion processes: iRGC axon repulsion at the optic chiasm and antero-posterior commissural axon repulsion in the spinal cord. Therefore, Numb is required for Shh-mediated attraction and repulsion. These results position Numb as a central player in the non-canonical Shh signaling pathway mediating axon repulsion.
Collapse
Affiliation(s)
- Tiphaine Dolique
- Montreal Clinical Research Institute (IRCM), 110 Pine Avenue West, Montreal, QC H2W 1R7, Canada
- Department of Anatomy and Cell Biology, Division of Experimental Medicine, McGill University, Montreal, QC H3A 0G4, Canada
- Inovarion, 75005 Paris, France
| | - Sarah Baudet
- Institut du Fer à Moulin, Inserm, Sorbonne Université, Paris, France
- Sorbonne Université, CNRS, Inserm, Center of Neuroscience Neuro-SU, 75005 Paris, France
- Sorbonne Université, CNRS, Inserm, Institut de Biologie Paris-Seine, IBPS, 75005 Paris, France
| | - Frederic Charron
- Montreal Clinical Research Institute (IRCM), 110 Pine Avenue West, Montreal, QC H2W 1R7, Canada
- Department of Anatomy and Cell Biology, Division of Experimental Medicine, McGill University, Montreal, QC H3A 0G4, Canada
- Department of Medicine, University of Montreal, Montreal QC H3T 1J4, Canada
| | - Julien Ferent
- Montreal Clinical Research Institute (IRCM), 110 Pine Avenue West, Montreal, QC H2W 1R7, Canada
- Institut du Fer à Moulin, Inserm, Sorbonne Université, Paris, France
- Sorbonne Université, CNRS, Inserm, Center of Neuroscience Neuro-SU, 75005 Paris, France
- Sorbonne Université, CNRS, Inserm, Institut de Biologie Paris-Seine, IBPS, 75005 Paris, France
| |
Collapse
|
2
|
Dai W, Nian X, Zhou Z, Du A, Liu Q, Jia S, Lu Y, Li D, Lu X, Zhu Y, Huang Q, Lu J, Xiao Y, Zheng L, Lei W, Sheng N, Zang X, Hou Y, Qiu Z, Xu R, Xu S, Zhang X, Zhang L. A neuronal Slit1-dependent program rescues oligodendrocyte differentiation and myelination under chronic hypoxic conditions. Cell Rep 2025; 44:115467. [PMID: 40117292 DOI: 10.1016/j.celrep.2025.115467] [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: 12/01/2024] [Revised: 01/25/2025] [Accepted: 03/05/2025] [Indexed: 03/23/2025] Open
Abstract
Oligodendrocyte maturation arrest in hypoxia-induced white matter injury (WMI) results in long-term neurofunctional disabilities of preterm infants. Although neurons are closely linked to myelination regulation, how neurons respond to the above process remains elusive. Here, we identify a compensatory role of neuronal Slit1-dependent signaling in protecting against hypoxia-induced hypomyelination and ameliorating motor and cognitive disabilities. Conditional ablation of Slit1 in neurons exacerbates hypoxia-induced hypomyelination but is negligible for developmental myelination. Secreted Slit1 from hypoxic neurons directly targets oligodendrocyte, acting through Robo2-srGAP1-RhoA signaling. Pharmacological inhibition of RhoA restores myelination and promotes neurofunctional recovery in adolescent mice. Notably, natural selection analysis and functional validation indicate an adaptive variant with higher Slit1 gene expression in the Tibetan population, which has low oxygen availability. Collectively, these findings show a neuronal Slit1-dependent program of OL differentiation and suggest that targeting the Slit1-Robo2 signaling axis may have therapeutic potential for treatment of preterm infants with hypoxic WMI.
Collapse
Affiliation(s)
- Wenxiu Dai
- Department of Neurology, Songjiang Research Institute, Shanghai Key Laboratory of Emotions and Affective Disorders, Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Ximing Nian
- Department of Neurology, Songjiang Research Institute, Shanghai Key Laboratory of Emotions and Affective Disorders, Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Zhihao Zhou
- State Key Laboratory of Cellular Stress Biology, School of Life Science, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Ailian Du
- Department of Neurology, Songjiang Research Institute, Shanghai Key Laboratory of Emotions and Affective Disorders, Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Qi Liu
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Center for Evolutionary Biology, School of Life Sciences, Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200433, China
| | - Shufang Jia
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Center for Evolutionary Biology, School of Life Sciences, Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200433, China
| | - Yan Lu
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Center for Evolutionary Biology, School of Life Sciences, Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200433, China
| | - Daopeng Li
- State Key Laboratory of Cellular Stress Biology, School of Life Science, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Xiaoyun Lu
- State Key Laboratory of Cellular Stress Biology, School of Life Science, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Yanqin Zhu
- State Key Laboratory of Cellular Stress Biology, School of Life Science, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Qiuying Huang
- State Key Laboratory of Cellular Stress Biology, School of Life Science, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Jiaquan Lu
- Department of Neurology, Songjiang Research Institute, Shanghai Key Laboratory of Emotions and Affective Disorders, Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yunshan Xiao
- Department of Obstetrics and Gynecology, Women and Children's Hospital Affiliated to Xiamen University, State Key Laboratory of Cellular Stress Biology, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Liangkai Zheng
- Department of Pathology, Women and Children's Hospital Affiliated to Xiamen University, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Wanying Lei
- Institutes of Brain Science, Fudan University, Shanghai 200433, China
| | - Nengyin Sheng
- State Key Laboratory of Genetic Evolution and Animal Models, Key Laboratory of Animal Models and Human Disease Mechanisms of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
| | - Xiujuan Zang
- Department of Nephrology, Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yanqiang Hou
- Department of Clinical Laboratory, Songjiang Research Institute, Shanghai Key Laboratory of Emotions and Affective Disorders, Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Zilong Qiu
- Department of Neurology, Songjiang Research Institute, Shanghai Key Laboratory of Emotions and Affective Disorders, Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Ren Xu
- State Key Laboratory of Cellular Stress Biology, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Shuhua Xu
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Center for Evolutionary Biology, School of Life Sciences, Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200433, China; School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Xueqin Zhang
- Department of Obstetrics and Gynecology, Women and Children's Hospital Affiliated to Xiamen University, State Key Laboratory of Cellular Stress Biology, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian 361102, China.
| | - Liang Zhang
- Department of Neurology, Songjiang Research Institute, Shanghai Key Laboratory of Emotions and Affective Disorders, Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Naval Medical University, Shanghai 200433, China.
| |
Collapse
|
3
|
Sun F, Shuai Y, Wang J, Yan J, Lin B, Li X, Zhao Z. Hippocampal gray matter volume alterations in patients with first-episode and recurrent major depressive disorder and their associations with gene profiles. BMC Psychiatry 2025; 25:134. [PMID: 39955494 PMCID: PMC11829352 DOI: 10.1186/s12888-025-06562-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 01/31/2025] [Indexed: 02/17/2025] Open
Abstract
BACKGROUND Recent studies indicate that patients with first-episode drug-naïve (FEDN) and recurrent major depressive disorder (R-MDD) exhibit distinct atrophy patterns in the hippocampal subregions along the proximal-distal axis. However, it remains unclear whether such differences occur along the long axis and how they may relate to specific genes. METHODS In the present study, we analyzed T1-weighted images from 421 patients (FEDN: n = 232; R-MDD: n = 189) and 544 normal controls (NC) as part of the REST-meta-MDD consortium. Additionally, transcriptome maps and structural Magnetic Resonance Imaging (MRI) data of six donated brains were obtained from the Allen Human Brain Atlas (AHBA). We first identified changes in gray matter volume (GMV) within the hippocampus of both FEDN and R-MDD patients and then integrated these findings with AHBA transcriptome data to investigate the genes associated with hippocampal GMV changes. RESULTS Compared to NC, FEDN patients displayed reduced GMV in the left hippocampal tail, whereas R-MDD patients exhibited decreased GMV in the bilateral hippocampal body and increased GMV in the bilateral hippocampal tail. Further analysis revealed that expression levels of SYTL2 positively correlated with GMV changes in the hippocampus of FEDN patients, while SORCS3 and SLIT2 positively correlated with those in R-MDD. CONCLUSIONS Our results suggest that GMV alterations in hippocampal subfields along the long axis differ between FEDN and R-MDD, reflecting progressive hippocampal deterioration with prolonged depression, potentially supported by the expression of specific genes. These findings offer valuable insights into the distinct neural and genetic mechanisms underlying FEDN and R-MDD, which may aid in the development of more targeted and effective treatment strategies for MDD subtypes.
Collapse
Affiliation(s)
- Fenfen Sun
- Center for Brain, Mind and Education, Shaoxing University, Shaoxing, China
- Department of Psychology, Shaoxing University, Shaoxing, China
| | - Yifan Shuai
- Key Laboratory for Biomedical Engineering of Ministry of Education, Department of Biomedical Engineering, College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou, China
| | - Jingru Wang
- Department of Psychology, Shaoxing University, Shaoxing, China
| | - Jin Yan
- Department of Psychology, Shaoxing University, Shaoxing, China
| | - Bin Lin
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xinyun Li
- School of Rehabilitation, Hangzhou Medical College, Hangzhou, China
| | - Zhiyong Zhao
- Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Binjiang Campus, 3333 Binsheng Rd, Hangzhou, China.
| |
Collapse
|
4
|
Delpech C, Schaeffer J, Vilallongue N, Delaunay A, Benadjal A, Blot B, Excoffier B, Plissonnier E, Gascon E, Albert F, Paccard A, Saintpierre A, Gasnier C, Zagar Y, Castellani V, Belin S, Chédotal A, Nawabi H. Axon guidance during mouse central nervous system regeneration is required for specific brain innervation. Dev Cell 2024; 59:3213-3228.e8. [PMID: 39353435 DOI: 10.1016/j.devcel.2024.09.005] [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: 10/10/2023] [Revised: 07/11/2024] [Accepted: 09/05/2024] [Indexed: 10/04/2024]
Abstract
Reconstructing functional neuronal circuits is one major challenge of central nervous system repair. Through activation of pro-growth signaling pathways, some neurons achieve long-distance axon regrowth. Yet, functional reconnection has hardly been obtained, as these regenerating axons fail to resume their initial trajectory and reinnervate their proper target. Axon guidance is considered to be active only during development. Here, using the mouse visual system, we show that axon guidance is still active in the adult brain in regenerative conditions. We highlight that regenerating retinal ganglion cell axons avoid one of their primary targets, the suprachiasmatic nucleus (SCN), due to Slit/Robo repulsive signaling. Together with promoting regeneration, silencing Slit/Robo in vivo enables regenerating axons to enter the SCN and form active synapses. The newly formed circuit is associated with neuronal activation and functional recovery. Our results provide evidence that axon guidance mechanisms are required to reconnect regenerating axons to specific brain nuclei.
Collapse
Affiliation(s)
- Céline Delpech
- Université Grenoble Alpes, Inserm U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Julia Schaeffer
- Université Grenoble Alpes, Inserm U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Noemie Vilallongue
- Université Grenoble Alpes, Inserm U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Apolline Delaunay
- Université Grenoble Alpes, Inserm U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Amin Benadjal
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Beatrice Blot
- Université Grenoble Alpes, Inserm U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Blandine Excoffier
- Université Grenoble Alpes, Inserm U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Elise Plissonnier
- Université Grenoble Alpes, Inserm U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Eduardo Gascon
- Aix Marseille University, CNRS, INT, Institute of Neurosci Timone, Marseille, France
| | - Floriane Albert
- Université Grenoble Alpes, Inserm U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Antoine Paccard
- Université Grenoble Alpes, Inserm U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Ana Saintpierre
- Université Grenoble Alpes, Inserm U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Celestin Gasnier
- Université Grenoble Alpes, Inserm U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Yvrick Zagar
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Valérie Castellani
- University Claude Bernard Lyon 1, MeLiS, CNRS UMR5284, INSERM U1314, Lyon, France
| | - Stephane Belin
- Université Grenoble Alpes, Inserm U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Alain Chédotal
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France; University Claude Bernard Lyon 1, MeLiS, CNRS UMR5284, INSERM U1314, Lyon, France; Institut de pathologie, groupe hospitalier Est, Hospices Civils de Lyon, Lyon, France
| | - Homaira Nawabi
- Université Grenoble Alpes, Inserm U1216, Grenoble Institut Neurosciences, 38000 Grenoble, France.
| |
Collapse
|
5
|
Grudet F, Martinot E, Godin P, Bérubé M, Chédotal A, Boerboom D. Slit1 inhibits ovarian follicle development and female fertility in mice†. Biol Reprod 2024; 111:834-844. [PMID: 38943353 PMCID: PMC11473917 DOI: 10.1093/biolre/ioae106] [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: 03/12/2024] [Revised: 06/17/2024] [Accepted: 06/27/2024] [Indexed: 07/01/2024] Open
Abstract
Previous in vitro studies have suggested that SLIT ligands could play roles in regulating ovarian granulosa cell proliferation and gene expression, as well as luteolysis. However, no in vivo study of Slit gene function has been conducted to date. Here, we investigated the potential role of Slit1 in ovarian biology using a Slit1-null mouse model. Female Slit1-null mice were found to produce larger litters than their wild-type counterparts due to increased ovulation rates. Increased ovarian weights in Slit1-null animals were found to be due to the presence of greater numbers of healthy antral follicles with similar numbers of atretic ones, suggesting both an increased rate of follicle recruitment and a decreased rate of atresia. Consistent with this, treatment of cultured granulosa cells with exogenous SLIT1 induced apoptosis in presence or absence of follicle-stimulating hormone, but had no effect on cell proliferation. Although few alterations in the messenger RNA levels of follicle-stimulating hormone-responsive genes were noted in granulosa cells of Slit1-null mice, luteinizing hormone target gene mRNA levels were greatly increased. Finally, increased phospho-AKT levels were found in granulosa cells isolated from Slit1-null mice, and SLIT1 pretreatment of cultured granulosa cells inhibited the ability of both follicle-stimulating hormone and luteinizing hormone to increase AKT phosphorylation, suggesting a mechanism whereby SLIT1 could antagonize gonadotropin signaling. These findings therefore represent the first evidence for a physiological role of a SLIT ligand in the ovary, and define Slit1 as a novel autocrine/paracrine regulator of follicle development.
Collapse
Affiliation(s)
- Florine Grudet
- Centre de Recherche en Reproduction et Fertilité (CRRF), Département de Biomédecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC, Canada
| | - Emmanuelle Martinot
- Centre de Recherche en Reproduction et Fertilité (CRRF), Département de Biomédecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC, Canada
| | - Philippe Godin
- Centre de Recherche en Reproduction et Fertilité (CRRF), Département de Biomédecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC, Canada
| | - Michael Bérubé
- Centre de Recherche en Reproduction et Fertilité (CRRF), Département de Biomédecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC, Canada
| | - Alain Chédotal
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Derek Boerboom
- Centre de Recherche en Reproduction et Fertilité (CRRF), Département de Biomédecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC, Canada
| |
Collapse
|
6
|
Cazares O, Chen M, Menendez J, Molinuevo R, Thomas G, Cervantes J, Yee M, Cadell M, Durham M, Zhu Y, Strietzel C, Bubolz JW, Hinck L. SLIT Loss or Sequestration Increases Mammary Alveologenesis and Lactogenesis. MICROPUBLICATION BIOLOGY 2024; 2024:10.17912/micropub.biology.001264. [PMID: 39381643 PMCID: PMC11461027 DOI: 10.17912/micropub.biology.001264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Figures] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 07/17/2024] [Accepted: 09/04/2024] [Indexed: 10/10/2024]
Abstract
SLITs comprise a family of secreted proteins that function as ligands for Roundabout (ROBO) receptors. Previous research showed that ROBO1 promotes the differentiation of milk-producing alveolar cells by inhibiting Notch signaling in mammary luminal cells. Here, we show enhanced alveolar development and increased milk production in Slit2-/-;Slit3-/- knockout mammary gland epithelia. This result can also be achieved by intraperitoneal delivery of recombinant ROBO1 extracellular domain fragment, ROBO1-5Ig-Fc, which sequesters SLITs. Together, our phenotypic studies suggest that SLITs restrict alveologenesis and lactogenesis by inhibiting ROBO1.
Collapse
Affiliation(s)
| | - Min Chen
- University of California, Santa Cruz, CA, USA
| | | | | | - Gwen Thomas
- University of California, Santa Cruz, CA, USA
| | | | - Michael Yee
- University of California, Santa Cruz, CA, USA
| | | | | | - Yaqi Zhu
- Zoetis (United States), Kalamazoo, MI, United States
| | | | | | | |
Collapse
|
7
|
Curran BM, Nickerson KR, Yung AR, Goodrich LV, Jaworski A, Tessier-Lavigne M, Ma L. Multiple guidance mechanisms control axon growth to generate precise T-shaped bifurcation during dorsal funiculus development in the spinal cord. eLife 2024; 13:RP94109. [PMID: 39159057 PMCID: PMC11333043 DOI: 10.7554/elife.94109] [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: 08/21/2024] Open
Abstract
The dorsal funiculus in the spinal cord relays somatosensory information to the brain. It is made of T-shaped bifurcation of dorsal root ganglion (DRG) sensory axons. Our previous study has shown that Slit signaling is required for proper guidance during bifurcation, but loss of Slit does not affect all DRG axons. Here, we examined the role of the extracellular molecule Netrin-1 (Ntn1). Using wholemount staining with tissue clearing, we showed that mice lacking Ntn1 had axons escaping from the dorsal funiculus at the time of bifurcation. Genetic labeling confirmed that these misprojecting axons come from DRG neurons. Single axon analysis showed that loss of Ntn1 did not affect bifurcation but rather altered turning angles. To distinguish their guidance functions, we examined mice with triple deletion of Ntn1, Slit1, and Slit2 and found a completely disorganized dorsal funiculus. Comparing mice with different genotypes using immunolabeling and single axon tracing revealed additive guidance errors, demonstrating the independent roles of Ntn1 and Slit. Moreover, the same defects were observed in embryos lacking their cognate receptors. These in vivo studies thus demonstrate the presence of multi-factorial guidance mechanisms that ensure proper formation of a common branched axonal structure during spinal cord development.
Collapse
Affiliation(s)
- Bridget M Curran
- Department of Neuroscience, Jefferson Synaptic Biology Center, Vickie and Jack Farber, Institute for Neuroscience, Sydney Kimmel Medical College, Thomas Jefferson UniversityPhiladelphiaUnited States
| | - Kelsey R Nickerson
- Department of Neuroscience, Brown UniversityProvidenceUnited States
- Robert J. and Nancy D. Carney Institute for Brain ScienceProvidenceUnited States
| | - Andrea R Yung
- Department of Neurobiology, Harvard Medical SchoolBostonUnited States
| | - Lisa V Goodrich
- Department of Neurobiology, Harvard Medical SchoolBostonUnited States
| | - Alexander Jaworski
- Department of Neuroscience, Brown UniversityProvidenceUnited States
- Robert J. and Nancy D. Carney Institute for Brain ScienceProvidenceUnited States
| | | | - Le Ma
- Department of Neuroscience, Jefferson Synaptic Biology Center, Vickie and Jack Farber, Institute for Neuroscience, Sydney Kimmel Medical College, Thomas Jefferson UniversityPhiladelphiaUnited States
| |
Collapse
|
8
|
Li Z, Shi B, Li N, Sun J, Zeng X, Huang R, Bok S, Chen X, Han J, Yallowitz AR, Debnath S, Cung M, Ling Z, Zhong CQ, Hong Y, Li G, Koenen M, Cohen P, Su X, Lu H, Greenblatt MB, Xu R. Bone controls browning of white adipose tissue and protects from diet-induced obesity through Schnurri-3-regulated SLIT2 secretion. Nat Commun 2024; 15:6697. [PMID: 39107299 PMCID: PMC11303806 DOI: 10.1038/s41467-024-51155-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 07/31/2024] [Indexed: 08/10/2024] Open
Abstract
The skeleton has been suggested to function as an endocrine organ controlling whole organism energy balance, however the mediators of this effect and their molecular links remain unclear. Here, utilizing Schnurri-3-/- (Shn3-/-) mice with augmented osteoblast activity, we show Shn3-/-mice display resistance against diet-induced obesity and enhanced white adipose tissue (WAT) browning. Conditional deletion of Shn3 in osteoblasts but not adipocytes recapitulates lean phenotype of Shn3-/-mice, indicating this phenotype is driven by skeleton. We further demonstrate osteoblasts lacking Shn3 can secrete cytokines to promote WAT browning. Among them, we identify a C-terminal fragment of SLIT2 (SLIT2-C), primarily secreted by osteoblasts, as a Shn3-regulated osteokine that mediates WAT browning. Lastly, AAV-mediated Shn3 silencing phenocopies the lean phenotype and augmented glucose metabolism. Altogether, our findings establish a novel bone-fat signaling axis via SHN3 regulated SLIT2-C production in osteoblasts, offering a potential therapeutic target to address both osteoporosis and metabolic syndrome.
Collapse
Affiliation(s)
- Zan Li
- State Key Laboratory of Cellular Stress Biology, Cancer Research Center, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, People's Republic of China
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, People's Republic of China
- PET Center, Department of Nuclear Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Baohong Shi
- State Key Laboratory of Cellular Stress Biology, Cancer Research Center, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, People's Republic of China
- Xiamen Key Laboratory of Regeneration Medicine, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, China
| | - Na Li
- State Key Laboratory of Cellular Stress Biology, Cancer Research Center, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, People's Republic of China
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
- Xiamen Key Laboratory of Regeneration Medicine, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, China
| | - Jun Sun
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Xiangchen Zeng
- State Key Laboratory of Cellular Stress Biology, Cancer Research Center, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, People's Republic of China
| | - Rui Huang
- State Key Laboratory of Cellular Stress Biology, Cancer Research Center, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, People's Republic of China
| | - Seoyeon Bok
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Xiaohui Chen
- State Key Laboratory of Cellular Stress Biology, Cancer Research Center, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, People's Republic of China
- Xiamen Key Laboratory of Regeneration Medicine, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, China
| | - Jie Han
- State Key Laboratory of Cellular Stress Biology, Cancer Research Center, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, People's Republic of China
| | - Alisha R Yallowitz
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Shawon Debnath
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Michelle Cung
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Zheng Ling
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Chuan-Qi Zhong
- College of Life Science, Xiamen University, Xiamen, China
| | - Yixang Hong
- Xiamen Cardiovascular Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Gang Li
- Xiamen Cardiovascular Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Mascha Koenen
- Laboratory of Molecular Metabolism, The Rockefeller University, New York, NY, USA
| | - Paul Cohen
- Laboratory of Molecular Metabolism, The Rockefeller University, New York, NY, USA
| | - Xinhui Su
- PET Center, Department of Nuclear Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hongbin Lu
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, People's Republic of China.
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China.
- Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| | - Matthew B Greenblatt
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA.
- Research Division, Hospital for Special Surgery, New York, NY, USA.
| | - Ren Xu
- State Key Laboratory of Cellular Stress Biology, Cancer Research Center, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, People's Republic of China.
- Xiamen Key Laboratory of Regeneration Medicine, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, China.
| |
Collapse
|
9
|
Herrera E, Chédotal A, Mason C. Development of the Binocular Circuit. Annu Rev Neurosci 2024; 47:303-322. [PMID: 38635868 DOI: 10.1146/annurev-neuro-111020-093230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/20/2024]
Abstract
Seeing in three dimensions is a major property of the visual system in mammals. The circuit underlying this property begins in the retina, from which retinal ganglion cells (RGCs) extend to the same or opposite side of the brain. RGC axons decussate to form the optic chiasm, then grow to targets in the thalamus and midbrain, where they synapse with neurons that project to the visual cortex. Here we review the cellular and molecular mechanisms of RGC axonal growth cone guidance across or away from the midline via receptors to cues in the midline environment. We present new views on the specification of ipsi- and contralateral RGC subpopulations and factors implementing their organization in the optic tract and termination in subregions of their targets. Lastly, we describe the functional and behavioral aspects of binocular vision, focusing on the mouse, and discuss recent discoveries in the evolution of the binocular circuit.
Collapse
Affiliation(s)
- Eloísa Herrera
- Instituto de Neurociencias (CSIC-UMH), Consejo Superior de Investigaciones Científicas and Universidad Miguel Hernández, Alicante, Spain;
| | - Alain Chédotal
- Université Claude Bernard Lyon 1, MeLiS, CNRS UMR5284, INSERM U1314, Lyon, France
- Institut de Pathologie, Groupe Hospitalier Est, Hospices Civils de Lyon, Lyon, France
- Institut de la Vision, INSERM, Sorbonne Université, Paris, France;
| | - Carol Mason
- Departments of Pathology and Cell Biology, Neuroscience, and Ophthalmology, Zuckerman Institute, Columbia University, New York, NY, USA;
| |
Collapse
|
10
|
Ventriglia S, Kalcheim C. From neural tube to spinal cord: The dynamic journey of the dorsal neuroepithelium. Dev Biol 2024; 511:26-38. [PMID: 38580174 DOI: 10.1016/j.ydbio.2024.04.001] [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: 02/07/2024] [Revised: 03/21/2024] [Accepted: 04/02/2024] [Indexed: 04/07/2024]
Abstract
In a developing embryo, formation of tissues and organs is remarkably precise in both time and space. Through cell-cell interactions, neighboring progenitors coordinate their activities, sequentially generating distinct types of cells. At present, we only have limited knowledge, rather than a systematic understanding, of the underlying logic and mechanisms responsible for cell fate transitions. The formation of the dorsal aspect of the spinal cord is an outstanding model to tackle these dynamics, as it first generates the peripheral nervous system and is later responsible for transmitting sensory information from the periphery to the brain and for coordinating local reflexes. This is reflected first by the ontogeny of neural crest cells, progenitors of the peripheral nervous system, followed by formation of the definitive roof plate of the central nervous system and specification of adjacent interneurons, then a transformation of roof plate into dorsal radial glia and ependyma lining the forming central canal. How do these peripheral and central neural branches segregate from common progenitors? How are dorsal radial glia established concomitant with transformation of the neural tube lumen into a central canal? How do the dorsal radial glia influence neighboring cells? This is only a partial list of questions whose clarification requires the implementation of experimental paradigms in which precise control of timing is crucial. Here, we outline some available answers and still open issues, while highlighting the contributions of avian models and their potential to address mechanisms of neural patterning and function.
Collapse
Affiliation(s)
- Susanna Ventriglia
- Department of Medical Neurobiology, Institute of Medical Research Israel-Canada (IMRIC) and the Edmond and Lily Safra Center for Brain Sciences (ELSC), Hebrew University of Jerusalem-Hadassah Medical School, Jerusalem, 9112102, P.O.Box 12272, Israel.
| | - Chaya Kalcheim
- Department of Medical Neurobiology, Institute of Medical Research Israel-Canada (IMRIC) and the Edmond and Lily Safra Center for Brain Sciences (ELSC), Hebrew University of Jerusalem-Hadassah Medical School, Jerusalem, 9112102, P.O.Box 12272, Israel.
| |
Collapse
|
11
|
Gong H, Zhu C, Han D, Liu S. Secreted Glycoproteins That Regulate Synaptic Function: the Dispatchers in the Central Nervous System. Mol Neurobiol 2024; 61:2719-2727. [PMID: 37924485 DOI: 10.1007/s12035-023-03731-y] [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: 02/07/2023] [Accepted: 10/17/2023] [Indexed: 11/06/2023]
Abstract
Glycoproteins are proteins that contain oligosaccharide chains. As widely distributed functional proteins in the body, glycoproteins are essential for cellular development, cellular function maintenance, and intercellular communication. Glycoproteins not only play a role in the cell and the membrane, but they are also secreted in the intercell. These secreted glycoproteins are critical to the central nervous system for neurodevelopment and synaptic transmission. More specifically, secreted glycoproteins play indispensable roles in neurite growth mediation, axon guiding, synaptogenesis, neuronal differentiation, the release of synaptic vesicles, subunit composition of neurotransmitter receptors, and neurotransmitter receptor trafficking among other things. Abnormal expressions of secreted glycoproteins in the central nervous system are associated with abnormal neuron development, impaired synaptic organization/transmission, and neuropsychiatric disorders. This article reviews the secreted glycoproteins that regulate neuronal development and synaptic function in the central nervous system, and the molecular mechanism of these regulations, providing reference for research about synaptic function regulation and related central nervous system diseases.
Collapse
Affiliation(s)
- Haiying Gong
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Conglei Zhu
- Department of Pharmacy, Fuyang People's Hospital, Fuyang, Anhui, China
| | - Di Han
- Department of Respiratory and Critical Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Sen Liu
- School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China.
| |
Collapse
|
12
|
Johansson Y, Andreassen M, Hartsch M, Wagner S, Forsby A. Attenuated neuronal differentiation caused by acrylamide is not related to oxidative stress in differentiated human neuroblastoma SH-SY5Y cells. Food Chem Toxicol 2024; 187:114623. [PMID: 38554842 DOI: 10.1016/j.fct.2024.114623] [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: 01/08/2024] [Revised: 03/15/2024] [Accepted: 03/24/2024] [Indexed: 04/02/2024]
Abstract
Acrylamide (ACR) is a known neurotoxicant and developmental neurotoxicant. As a soft electrophile, ACR reacts with thiol groups in cysteine. One hypothesis of ACR induced neurotoxicity and developmental neurotoxicity (DNT) is conjugation with reduced glutathione (GSH) leading to GSH depletion, increased reactive oxygen species (ROS) production and further oxidative stress and cellular damage. In this regard, we have investigated the effect of ACR on neuronal differentiation, glutathione levels and ROS production in the human neuroblastoma SH-SY5Y cell model. After 9 days of differentiation and exposure, ACR significantly impaired area neurites per cell at non-cytotoxic concentrations (0.33 μM and 10 μM). Furthermore, 10 μM ACR dysregulated 9 mRNA markers important for neuronal development, 5 of them being associated with cytoskeleton organization and axonal guidance. At the non-cytotoxic concentrations that significantly attenuate neuronal differentiation, ACR did neither decrease the level of GSH or total glutathione levels, nor increased ROS production. In addition, the expression of 5 mRNA markers for cellular stress was assessed with no significant altered regulation after ACR exposure up to 320 μM. Thus, ACR-induced DNT is not due to GSH depletion and increased ROS production, neither at non-cytotoxic nor cytotoxic concentrations, in the SH-SH5Y model during differentiation.
Collapse
Affiliation(s)
- Ylva Johansson
- Department of Biochemistry and Biophysics, Stockholm University, 106 91, Stockholm, Sweden.
| | - Mathilda Andreassen
- Department of Biochemistry and Biophysics, Stockholm University, 106 91, Stockholm, Sweden
| | - Muriel Hartsch
- Department of Biochemistry and Biophysics, Stockholm University, 106 91, Stockholm, Sweden
| | - Stella Wagner
- Department of Biochemistry and Biophysics, Stockholm University, 106 91, Stockholm, Sweden
| | - Anna Forsby
- Department of Biochemistry and Biophysics, Stockholm University, 106 91, Stockholm, Sweden
| |
Collapse
|
13
|
Curran BM, Nickerson KR, Yung AR, Goodrich LV, Jaworski A, Tessier-Lavigne M, Ma L. Multiple Guidance Mechanisms Control Axon Growth to Generate Precise T-shaped Bifurcation during Dorsal Funiculus Development in the Spinal Cord. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.11.17.567638. [PMID: 38014092 PMCID: PMC10680847 DOI: 10.1101/2023.11.17.567638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
The dorsal funiculus in the spinal cord relays somatosensory information to the brain. It is made of T-shaped bifurcation of dorsal root ganglion (DRG) sensory axons. Our previous study has shown that Slit signaling is required for proper guidance during bifurcation, but loss of Slit does not affect all DRG axons. Here, we examined the role of the extracellular molecule Netrin-1 (Ntn1). Using wholemount staining with tissue clearing, we showed that mice lacking Ntn1 have axons escaping from the dorsal funiculus at the time of bifurcation. Genetic labeling confirmed that these misprojecting axons come from DRG neurons. Single axon analysis showed that loss of Ntn1 does not affect bifurcation but rather alters turning angles. To distinguish their guidance functions, we examined mice with triple deletion of Ntn1, Slit1, and Slit2 and found a completely disorganized dorsal funiculus. Comparing mice with different genotypes using immunolabeling and single axon tracing revealed additive guidance errors, demonstrating the independent roles of Ntn1 and Slit. Moreover, the same defects were observed in embryos lacking their cognate receptors. These in vivo studies thus demonstrate the presence of multi-factorial guidance mechanisms that ensure proper formation of a common branched axonal structure during spinal cord development.
Collapse
Affiliation(s)
- Bridget M Curran
- Department of Neuroscience, Jefferson Synaptic Biology Center, Vickie and Jack Farber Institute for Neuroscience, Sydney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107
| | - Kelsey R Nickerson
- Department of Neuroscience, Brown University, Providence, RI 02912
- Robert J. and Nancy D. Carney Institute for Brain Science, Providence, RI 02912
| | - Andrea R Yung
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115
| | - Lisa V Goodrich
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115
| | - Alexander Jaworski
- Department of Neuroscience, Brown University, Providence, RI 02912
- Robert J. and Nancy D. Carney Institute for Brain Science, Providence, RI 02912
| | | | - Le Ma
- Department of Neuroscience, Jefferson Synaptic Biology Center, Vickie and Jack Farber Institute for Neuroscience, Sydney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107
| |
Collapse
|
14
|
Karas BF, Terez KR, Mowla S, Battula N, Flannery KP, Gural BM, Aboussleman G, Mubin N, Manzini MC. Removal of pomt1 in zebrafish leads to loss of α-dystroglycan glycosylation and dystroglycanopathy phenotypes. Hum Mol Genet 2024; 33:709-723. [PMID: 38272461 PMCID: PMC11000664 DOI: 10.1093/hmg/ddae006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 11/28/2023] [Accepted: 12/22/2023] [Indexed: 01/27/2024] Open
Abstract
Biallelic mutations in Protein O-mannosyltransferase 1 (POMT1) are among the most common causes of a severe group of congenital muscular dystrophies (CMDs) known as dystroglycanopathies. POMT1 is a glycosyltransferase responsible for the attachment of a functional glycan mediating interactions between the transmembrane glycoprotein dystroglycan and its binding partners in the extracellular matrix (ECM). Disruptions in these cell-ECM interactions lead to multiple developmental defects causing brain and eye malformations in addition to CMD. Removing Pomt1 in the mouse leads to early embryonic death due to the essential role of dystroglycan during placental formation in rodents. Here, we characterized and validated a model of pomt1 loss of function in the zebrafish showing that developmental defects found in individuals affected by dystroglycanopathies can be recapitulated in the fish. We also discovered that pomt1 mRNA provided by the mother in the oocyte supports dystroglycan glycosylation during the first few weeks of development. Muscle disease, retinal synapse formation deficits, and axon guidance defects can only be uncovered during the first week post fertilization by generating knock-out embryos from knock-out mothers. Conversely, maternal pomt1 from heterozygous mothers was sufficient to sustain muscle, eye, and brain development only leading to loss of photoreceptor synapses at 30 days post fertilization. Our findings show that it is important to define the contribution of maternal mRNA while developing zebrafish models of dystroglycanopathies and that offspring generated from heterozygous and knock-out mothers can be used to differentiate the role of dystroglycan glycosylation in tissue formation and maintenance.
Collapse
Affiliation(s)
- Brittany F Karas
- Department of Neuroscience and Cell Biology, Child Health Institute of New Jersey, Rutgers University-Robert Wood Johnson Medical School, 89 French Street, New Brunswick, NJ 08901, United States
| | - Kristin R Terez
- Department of Neuroscience and Cell Biology, Child Health Institute of New Jersey, Rutgers University-Robert Wood Johnson Medical School, 89 French Street, New Brunswick, NJ 08901, United States
| | - Shorbon Mowla
- Department of Neuroscience and Cell Biology, Child Health Institute of New Jersey, Rutgers University-Robert Wood Johnson Medical School, 89 French Street, New Brunswick, NJ 08901, United States
| | - Namarata Battula
- Department of Neuroscience and Cell Biology, Child Health Institute of New Jersey, Rutgers University-Robert Wood Johnson Medical School, 89 French Street, New Brunswick, NJ 08901, United States
| | - Kyle P Flannery
- Department of Neuroscience and Cell Biology, Child Health Institute of New Jersey, Rutgers University-Robert Wood Johnson Medical School, 89 French Street, New Brunswick, NJ 08901, United States
| | - Brian M Gural
- Department of Neuroscience and Cell Biology, Child Health Institute of New Jersey, Rutgers University-Robert Wood Johnson Medical School, 89 French Street, New Brunswick, NJ 08901, United States
| | - Grace Aboussleman
- Department of Neuroscience and Cell Biology, Child Health Institute of New Jersey, Rutgers University-Robert Wood Johnson Medical School, 89 French Street, New Brunswick, NJ 08901, United States
| | - Numa Mubin
- Department of Neuroscience and Cell Biology, Child Health Institute of New Jersey, Rutgers University-Robert Wood Johnson Medical School, 89 French Street, New Brunswick, NJ 08901, United States
| | - M Chiara Manzini
- Department of Neuroscience and Cell Biology, Child Health Institute of New Jersey, Rutgers University-Robert Wood Johnson Medical School, 89 French Street, New Brunswick, NJ 08901, United States
| |
Collapse
|
15
|
Le VH, Orniacki C, Murcia-Belmonte V, Denti L, Schütz D, Stumm R, Ruhrberg C, Erskine L. CXCL12 promotes the crossing of retinal ganglion cell axons at the optic chiasm. Development 2024; 151:dev202446. [PMID: 38095299 PMCID: PMC10820821 DOI: 10.1242/dev.202446] [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: 10/20/2023] [Accepted: 12/07/2023] [Indexed: 01/13/2024]
Abstract
Binocular vision requires the segregation of retinal ganglion cell (RGC) axons extending from the retina into the ipsilateral and contralateral optic tracts. RGC axon segregation occurs at the optic chiasm, which forms at the ventral diencephalon midline. Using expression analyses, retinal explants and genetically modified mice, we demonstrate that CXCL12 (SDF1) is required for axon segregation at the optic chiasm. CXCL12 is expressed by the meninges bordering the optic pathway, and CXCR4 by both ipsilaterally and contralaterally projecting RGCs. CXCL12 or ventral diencephalon meninges potently promoted axon outgrowth from both ipsilaterally and contralaterally projecting RGCs. Further, a higher proportion of axons projected ipsilaterally in mice lacking CXCL12 or its receptor CXCR4 compared with wild-type mice as a result of misrouting of presumptive contralaterally specified RGC axons. Although RGCs also expressed the alternative CXCL12 receptor ACKR3, the optic chiasm developed normally in mice lacking ACKR3. Our data support a model whereby meningeal-derived CXCL12 helps drive axon growth from CXCR4-expressing RGCs towards the diencephalon midline, enabling contralateral axon growth. These findings further our understanding of the molecular and cellular mechanisms controlling optic pathway development.
Collapse
Affiliation(s)
- Viet-Hang Le
- School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen,Foresterhill, Aberdeen AB25 2ZD, UK
| | - Clarisse Orniacki
- School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen,Foresterhill, Aberdeen AB25 2ZD, UK
| | - Verónica Murcia-Belmonte
- School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen,Foresterhill, Aberdeen AB25 2ZD, UK
- Instituto de Neurociencias de Alicante (Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández, CSIC-UMH), Campus San Juan, Av. Ramón y Cajal s/n, Alicante 03550, Spain
| | - Laura Denti
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK
| | - Dagmar Schütz
- Institute for Pharmacology/Toxicology, Jena University Hospital,Drackendorfer Str. 1, D-07747 Jena, Germany
| | - Ralf Stumm
- Institute for Pharmacology/Toxicology, Jena University Hospital,Drackendorfer Str. 1, D-07747 Jena, Germany
| | - Christiana Ruhrberg
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK
| | - Lynda Erskine
- School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen,Foresterhill, Aberdeen AB25 2ZD, UK
| |
Collapse
|
16
|
Gong L, Si MS. SLIT3-mediated fibroblast signaling: a promising target for antifibrotic therapies. Am J Physiol Heart Circ Physiol 2023; 325:H1400-H1411. [PMID: 37830982 PMCID: PMC11932536 DOI: 10.1152/ajpheart.00216.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 09/13/2023] [Accepted: 09/27/2023] [Indexed: 10/14/2023]
Abstract
The SLIT family (SLIT1-3) of highly conserved glycoproteins was originally identified as ligands for the Roundabout (ROBO) family of single-pass transmembrane receptors, serving to provide repulsive axon guidance cues in the nervous system. Intriguingly, studies involving SLIT3 mutant mice suggest that SLIT3 might have crucial biological functions outside the neural context. Although these mutant mice display no noticeable neurological abnormalities, they present pronounced connective tissue defects, including congenital central diaphragmatic hernia, membranous ventricular septal defect, and osteopenia. We recently hypothesized that the phenotype observed in SLIT3-deficient mice may be tied to abnormalities in fibrillar collagen-rich connective tissue. Further research by our group indicates that both SLIT3 and its primary receptor, ROBO1, are expressed in fibrillar collagen-producing cells across various nonneural tissues. Global and constitutive SLIT3 deficiency not only reduces the synthesis and content of fibrillar collagen in various organs but also alleviates pressure overload-induced fibrosis in both the left and right ventricles. This review delves into the known phenotypes of SLIT3 mutants and the debated role of SLIT3 in vasculature and bone. Present evidence hints at SLIT3 acting as an autocrine regulator of fibrillar collagen synthesis, suggesting it as a potential antifibrotic treatment. However, the precise pathway and mechanisms through which SLIT3 regulates fibrillar collagen synthesis remain uncertain, presenting an intriguing avenue for future research.
Collapse
Affiliation(s)
- Lianghui Gong
- The Second Department of Thoracic Surgery, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, People's Republic of China
| | - Ming-Sing Si
- Division of Cardiac Surgery, Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, California, United States
| |
Collapse
|
17
|
Kerstein PC, Agreda YS, Curran BM, Ma L, Wright KM. Gbx2 controls amacrine cell dendrite stratification through Robo1/2 receptors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.03.551861. [PMID: 37577554 PMCID: PMC10418232 DOI: 10.1101/2023.08.03.551861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
Within the neuronal classes of the retina, amacrine cells (ACs) exhibit the greatest neuronal diversity in morphology and function. We show that the selective expression of the transcription factor Gbx2 is required for cell fate specification and dendritic stratification of an individual AC subtype in the mouse retina. We identify Robo1 and Robo2 as downstream effectors that when deleted, phenocopy the dendritic misprojections seen in Gbx2 mutants. Slit1 and Slit2, the ligands of Robo receptors, are localized to the OFF layers of the inner plexiform layer where we observe the dendritic misprojections in both Gbx2 and Robo1/2 mutants. We show that Robo receptors also are required for the proper dendritic stratification of additional AC subtypes, such as Vglut3+ ACs. These results show both that Gbx2 functions as a terminal selector in a single AC subtype and identify Slit-Robo signaling as a developmental mechanism for ON-OFF pathway segregation in the retina.
Collapse
|
18
|
Pretzsch CM, Ecker C. Structural neuroimaging phenotypes and associated molecular and genomic underpinnings in autism: a review. Front Neurosci 2023; 17:1172779. [PMID: 37457001 PMCID: PMC10347684 DOI: 10.3389/fnins.2023.1172779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 06/09/2023] [Indexed: 07/18/2023] Open
Abstract
Autism has been associated with differences in the developmental trajectories of multiple neuroanatomical features, including cortical thickness, surface area, cortical volume, measures of gyrification, and the gray-white matter tissue contrast. These neuroimaging features have been proposed as intermediate phenotypes on the gradient from genomic variation to behavioral symptoms. Hence, examining what these proxy markers represent, i.e., disentangling their associated molecular and genomic underpinnings, could provide crucial insights into the etiology and pathophysiology of autism. In line with this, an increasing number of studies are exploring the association between neuroanatomical, cellular/molecular, and (epi)genetic variation in autism, both indirectly and directly in vivo and across age. In this review, we aim to summarize the existing literature in autism (and neurotypicals) to chart a putative pathway from (i) imaging-derived neuroanatomical cortical phenotypes to (ii) underlying (neuropathological) biological processes, and (iii) associated genomic variation.
Collapse
Affiliation(s)
- Charlotte M. Pretzsch
- Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology, and Neuroscience, King’s College London, London, United Kingdom
| | - Christine Ecker
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital Frankfurt, Goethe University, Frankfurt, Germany
| |
Collapse
|
19
|
Baudet S, Zagar Y, Roche F, Gomez-Bravo C, Couvet S, Bécret J, Belle M, Vougny J, Uthayasuthan S, Ros O, Nicol X. Subcellular second messenger networks drive distinct repellent-induced axon behaviors. Nat Commun 2023; 14:3809. [PMID: 37369692 PMCID: PMC10300027 DOI: 10.1038/s41467-023-39516-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
Second messengers, including cAMP, cGMP and Ca2+ are often placed in an integrating position to combine the extracellular cues that orient growing axons in the developing brain. This view suggests that axon repellents share the same set of cellular messenger signals and that axon attractants evoke opposite cAMP, cGMP and Ca2+ changes. Investigating the confinement of these second messengers in cellular nanodomains, we instead demonstrate that two repellent cues, ephrin-A5 and Slit1, induce spatially segregated signals. These guidance molecules activate subcellular-specific second messenger crosstalk, each signaling network controlling distinct axonal morphology changes in vitro and pathfinding decisions in vivo.
Collapse
Affiliation(s)
- Sarah Baudet
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, F-75012, Paris, France
| | - Yvrick Zagar
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, F-75012, Paris, France
| | - Fiona Roche
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, F-75012, Paris, France
| | - Claudia Gomez-Bravo
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, F-75012, Paris, France
| | - Sandrine Couvet
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, F-75012, Paris, France
| | - Johann Bécret
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, F-75012, Paris, France
| | - Morgane Belle
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, F-75012, Paris, France
| | - Juliette Vougny
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, F-75012, Paris, France
| | | | - Oriol Ros
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, F-75012, Paris, France
- Department of Cell Biology, Physiology and Immunology, Universitat de Barcelona, 08028, Barcelona, Catalonia, Spain
| | - Xavier Nicol
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, F-75012, Paris, France.
| |
Collapse
|
20
|
Kwak HJ, Medina-Jiménez BI, Park SC, Kim JH, Jeong GH, Jeon MJ, Kim S, Kim JW, Weisblat DA, Cho SJ. Slit-Robo expression in the leech nervous system: insights into eyespot evolution. Cell Biosci 2023; 13:70. [PMID: 37013648 PMCID: PMC10071614 DOI: 10.1186/s13578-023-01019-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 03/26/2023] [Indexed: 04/05/2023] Open
Abstract
BACKGROUND Slit and Robo are evolutionarily conserved ligand and receptor proteins, respectively, but the number of slit and robo gene paralogs varies across recent bilaterian genomes. Previous studies indicate that this ligand-receptor complex is involved in axon guidance. Given the lack of data regarding Slit/Robo in the Lophotrochozoa compared to Ecdysozoa and Deuterostomia, the present study aims to identify and characterize the expression of Slit/Robo orthologs in leech development. RESULTS We identified one slit (Hau-slit), and two robo genes (Hau-robo1 and Hau-robo2), and characterized their expression spatiotemporally during the development of the glossiphoniid leech Helobdella austinensis. Throughout segmentation and organogenesis, Hau-slit and Hau-robo1 are broadly expressed in complex and roughly complementary patterns in the ventral and dorsal midline, nerve ganglia, foregut, visceral mesoderm and/or endoderm of the crop, rectum and reproductive organs. Before yolk exhaustion, Hau-robo1 is also expressed where the pigmented eye spots will later develop, and Hau-slit is expressed in the area between these future eye spots. In contrast, Hau-robo2 expression is extremely limited, appearing first in the developing pigmented eye spots, and later in the three additional pairs of cryptic eye spots in head region that never develop pigment. Comparing the expression of robo orthologs between H. austinensis and another glossiphoniid leech, Alboglossiphonia lata allows to that robo1 and robo2 operate combinatorially to differentially specify pigmented and cryptic eyespots within the glossiphoniid leeches. CONCLUSIONS Our results support a conserved role in neurogenesis, midline formation and eye spot development for Slit/Robo in the Lophotrochozoa, and provide relevant data for evo-devo studies related to nervous system evolution.
Collapse
Affiliation(s)
- Hee-Jin Kwak
- Department of Biological Sciences and Biotechnology, College of Natural Sciences, Chungbuk National University, Cheongju, Chungbuk, 28644, Republic of Korea
- Department of Ecology, Evolution and Behavior, Faculty of Science, Alexander Silberman Institute of Life Sciences, Hebrew University of Jerusalem, 9190401, Jerusalem, Israel
| | - Brenda I Medina-Jiménez
- Department of Biological Sciences and Biotechnology, College of Natural Sciences, Chungbuk National University, Cheongju, Chungbuk, 28644, Republic of Korea
- Department of Earth Sciences, Paleobiology, Geocentrum, Uppsala University, Villavägen 16, 75236, Uppsala, Sweden
| | - Soon Cheol Park
- Department of Life Science, College of Natural Sciences, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Jung-Hyeuk Kim
- Department of Biological Sciences and Biotechnology, College of Natural Sciences, Chungbuk National University, Cheongju, Chungbuk, 28644, Republic of Korea
- Wildlife Disease Response Team, National Institute of Wildlife Disease Control and Prevention, Incheon, 22689, Republic of Korea
| | - Geon-Hwi Jeong
- Department of Biological Sciences and Biotechnology, College of Natural Sciences, Chungbuk National University, Cheongju, Chungbuk, 28644, Republic of Korea
| | - Mi-Jeong Jeon
- National Institute of Biological Resources, Environmental Research Complex, Incheon, 22689, Republic of Korea
| | - Sangil Kim
- Museum of Comparative Zoology and Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Jung-Woong Kim
- Department of Life Science, College of Natural Sciences, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - David A Weisblat
- Department of Molecular and Cell Biology, University of California, 385 Weill Hall, Berkeley, CA, 94720-3200, USA.
| | - Sung-Jin Cho
- Department of Biological Sciences and Biotechnology, College of Natural Sciences, Chungbuk National University, Cheongju, Chungbuk, 28644, Republic of Korea.
| |
Collapse
|
21
|
Fu X, Chang J, Jiao D, Zhu M, Ma Y. SLIT3 knockdown inhibited TGF-β-induced hepatic stellate cells activation by down-regulating YAP signal. Mol Cell Toxicol 2023. [DOI: 10.1007/s13273-023-00336-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
Abstract
Objective
Liver fibrosis is a chronic liver disease caused by a variety of pathophysiological. However, there are no effective treatments to combat it. HSCs are a major source of fibrotic cells and exploring the mechanisms of HSC activation may provide new strategies for the treatment of liver fibrosis.
Objectives
To explore the role and underlying mechanism of SLIT3 in HSCs fibrosis.
Results
GSE163211 dataset analysis identified aberrant expression of SLIT3 in NASH F1-F4 tissues and SLIT3 expression level was positively correlated with fibrosis-related proteins. In vitro experiments showed that TGF-β induced upregulation of SLIT3 in LX-2 cells. Knockdown of SLIT3 significantly inhibited TGF-β-induced α-SMA, COL1A2, and COL1A1 expression, inhibited excessive cell proliferation and migration, and suppressed YAP activity.
Conclusion
Collectively, our findings suggest that SLIT3 deficiency alleviates TGF-β-induced HSCs activation by inhibiting YAP activity.
Collapse
|
22
|
Zebrafish Slit2 and Slit3 Act Together to Regulate Retinal Axon Crossing at the Midline. J Dev Biol 2022; 10:jdb10040041. [PMID: 36278546 PMCID: PMC9590056 DOI: 10.3390/jdb10040041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 09/09/2022] [Accepted: 09/20/2022] [Indexed: 11/17/2022] Open
Abstract
Slit-Robo signaling regulates midline crossing of commissural axons in different systems. In zebrafish, all retinofugal axons cross at the optic chiasm to innervate the contralateral tectum. Here, the mutant for the Robo2 receptor presents severe axon guidance defects, which were not completely reproduced in a Slit2 ligand null mutant. Since slit3 is also expressed around this area at the stage of axon crossing, we decided to analyze the possibility that it collaborates with Slit2 in this process. We found that the disruption of slit3 expression by sgRNA-Cas9 injection caused similar, albeit slightly milder, defects than those of the slit2 mutant, while the same treatment in the slit2−/−mz background caused much more severe defects, comparable to those observed in robo2 mutants. Tracking analysis of in vivo time-lapse experiments indicated differential but complementary functions of these secreted factors in the correction of axon turn errors around the optic chiasm. Interestingly, RT-qPCR analysis showed a mild increase in slit2 expression in slit3-deficient embryos, but not the opposite. Our observations support the previously proposed “repulsive channel” model for Slit-Robo action at the optic chiasm, with both Slits acting in different manners, most probably relating to their different spatial expression patterns.
Collapse
|
23
|
László ZI, Lele Z. Flying under the radar: CDH2 (N-cadherin), an important hub molecule in neurodevelopmental and neurodegenerative diseases. Front Neurosci 2022; 16:972059. [PMID: 36213737 PMCID: PMC9539934 DOI: 10.3389/fnins.2022.972059] [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: 06/17/2022] [Accepted: 08/31/2022] [Indexed: 12/03/2022] Open
Abstract
CDH2 belongs to the classic cadherin family of Ca2+-dependent cell adhesion molecules with a meticulously described dual role in cell adhesion and β-catenin signaling. During CNS development, CDH2 is involved in a wide range of processes including maintenance of neuroepithelial integrity, neural tube closure (neurulation), confinement of radial glia progenitor cells (RGPCs) to the ventricular zone and maintaining their proliferation-differentiation balance, postmitotic neural precursor migration, axon guidance, synaptic development and maintenance. In the past few years, direct and indirect evidence linked CDH2 to various neurological diseases, and in this review, we summarize recent developments regarding CDH2 function and its involvement in pathological alterations of the CNS.
Collapse
Affiliation(s)
- Zsófia I. László
- Momentum Laboratory of Molecular Neurobiology, Institute of Experimental Medicine, Budapest, Hungary
- Division of Cellular and Systems Medicine, School of Medicine, University of Dundee, Dundee, United Kingdom
| | - Zsolt Lele
- Momentum Laboratory of Molecular Neurobiology, Institute of Experimental Medicine, Budapest, Hungary
| |
Collapse
|
24
|
A diffusion MRI-based spatiotemporal continuum of the embryonic mouse brain for probing gene-neuroanatomy connections. Proc Natl Acad Sci U S A 2022; 119:2111869119. [PMID: 35165149 PMCID: PMC8851557 DOI: 10.1073/pnas.2111869119] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/07/2021] [Indexed: 11/18/2022] Open
Abstract
We established an ultra high-resolution diffusion MRI atlas of the embryonic mouse brains from E10.5 to E15.5, which characterizes the continuous changes of brain morphology and microstructures at mesoscopic scale. By integrating gene-expression data into the spatiotemporal continuum, we can navigate the evolving landscape of gene expression and neuroanatomy across both spatial and temporal dimensions to visualize their interactions in normal and abnormal embryonic brain development. We also identified regional clusters with distinct developmental trajectories and identified gene-expression profiles that matched to these regional domains. The diffusion MRI–based continuum of the embryonic brain and the computational techniques presented in this study offer a valuable tool for systematic study of the genetic control of brain development. The embryonic mouse brain undergoes drastic changes in establishing basic anatomical compartments and laying out major axonal connections of the developing brain. Correlating anatomical changes with gene-expression patterns is an essential step toward understanding the mechanisms regulating brain development. Traditionally, this is done in a cross-sectional manner, but the dynamic nature of development calls for probing gene–neuroanatomy interactions in a combined spatiotemporal domain. Here, we present a four-dimensional (4D) spatiotemporal continuum of the embryonic mouse brain from E10.5 to E15.5 reconstructed from diffusion magnetic resonance microscopy (dMRM) data. This study achieved unprecedented high-definition dMRM at 30- to 35-µm isotropic resolution, and together with computational neuroanatomy techniques, we revealed both morphological and microscopic changes in the developing brain. We transformed selected gene-expression data to this continuum and correlated them with the dMRM-based neuroanatomical changes in embryonic brains. Within the continuum, we identified distinct developmental modes comprising regional clusters that shared developmental trajectories and similar gene-expression profiles. Our results demonstrate how this 4D continuum can be used to examine spatiotemporal gene–neuroanatomical interactions by connecting upstream genetic events with anatomical changes that emerge later in development. This approach would be useful for large-scale analysis of the cooperative roles of key genes in shaping the developing brain.
Collapse
|
25
|
Freeman AK, Glendining KA, Jasoni CL. Developmental genes controlling neural circuit formation are expressed in the early postnatal hypothalamus and cellular lining of the third ventricle. J Neuroendocrinol 2021; 33:e13020. [PMID: 34423876 DOI: 10.1111/jne.13020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 07/20/2021] [Accepted: 07/27/2021] [Indexed: 02/07/2023]
Abstract
The arcuate nucleus of the hypothalamus is central in the regulation of body weight homeostasis through its ability to sense peripheral metabolic signals and relay them, through neural circuits, to other brain areas, ultimately affecting physiological and behavioural changes. The early postnatal development of these neural circuits is critical for normal body weight homeostasis, such that perturbations during this critical period can lead to obesity. The role for peripheral regulators of body weight homeostasis, including leptin, insulin and ghrelin, in this postnatal development is well described, yet some of the fundamental processes underpinning axonal and dendritic growth remain unclear. Here, we hypothesised that molecules known to regulate axonal and dendritic growth processes in other areas of the developing brain would be expressed in the postnatal arcuate nucleus and/or target nuclei where they would function to mediate the development of this circuitry. Using state-of-the-art RNAscope® technology, we have revealed the expression patterns of genes encoding Dcc/Netrin-1, Robo1/Slit1 and Fzd5/Wnt5a receptor/ligand pairs in the early postnatal mouse hypothalamus. We found that individual genes had unique expression patterns across developmental time in the arcuate nucleus, paraventricular nucleus of the hypothalamus, ventromedial nucleus of the hypothalamus, dorsomedial nucleus of the hypothalamus, median eminence and, somewhat unexpectedly, the third ventricle epithelium. These observations indicate a number of new molecular players in the development of neural circuits regulating body weight homeostasis, as well as novel molecular markers of tanycyte heterogeneity.
Collapse
Affiliation(s)
- Alice Katherine Freeman
- Centre for Neuroendocrinology, Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Kelly A Glendining
- Centre for Neuroendocrinology, Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Christine L Jasoni
- Centre for Neuroendocrinology, Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| |
Collapse
|
26
|
Laroche S, Stil A, Germain P, Cherif H, Chemtob S, Bouchard JF. Participation of L-Lactate and Its Receptor HCAR1/GPR81 in Neurovisual Development. Cells 2021; 10:1640. [PMID: 34208876 PMCID: PMC8303161 DOI: 10.3390/cells10071640] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 06/18/2021] [Accepted: 06/22/2021] [Indexed: 11/16/2022] Open
Abstract
During the development of the retina and the nervous system, high levels of energy are required by the axons of retinal ganglion cells (RGCs) to grow towards their brain targets. This energy demand leads to an increase of glycolysis and L-lactate concentrations in the retina. L-lactate is known to be the endogenous ligand of the GPR81 receptor. However, the role of L-lactate and its receptor in the development of the nervous system has not been studied in depth. In the present study, we used immunohistochemistry to show that GPR81 is localized in different retinal layers during development, but is predominantly expressed in the RGC of the adult rodent. Treatment of retinal explants with L-lactate or the exogenous GPR81 agonist 3,5-DHBA altered RGC growth cone (GC) morphology (increasing in size and number of filopodia) and promoted RGC axon growth. These GPR81-mediated modifications of GC morphology and axon growth were mediated by protein kinases A and C, but were absent in explants from gpr81-/- transgenic mice. Living gpr81-/- mice showed a decrease in ipsilateral projections of RGCs to the dorsal lateral geniculate nucleus (dLGN). In conclusion, present results suggest that L-lactate and its receptor GPR81 play an important role in the development of the visual nervous system.
Collapse
Affiliation(s)
- Samuel Laroche
- Neuropharmacology Laboratory, School of Optometry, Université de Montréal, Montreal, QC H3T 1P1, Canada; (S.L.); (A.S.); (P.G.); (H.C.)
| | - Aurélie Stil
- Neuropharmacology Laboratory, School of Optometry, Université de Montréal, Montreal, QC H3T 1P1, Canada; (S.L.); (A.S.); (P.G.); (H.C.)
| | - Philippe Germain
- Neuropharmacology Laboratory, School of Optometry, Université de Montréal, Montreal, QC H3T 1P1, Canada; (S.L.); (A.S.); (P.G.); (H.C.)
| | - Hosni Cherif
- Neuropharmacology Laboratory, School of Optometry, Université de Montréal, Montreal, QC H3T 1P1, Canada; (S.L.); (A.S.); (P.G.); (H.C.)
| | - Sylvain Chemtob
- Department of Pediatrics, Research Center-CHU Sainte-Justine, Montreal, QC H3T 1C5, Canada;
- Department of Ophtalmology, Faculty of Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada
| | - Jean-François Bouchard
- Neuropharmacology Laboratory, School of Optometry, Université de Montréal, Montreal, QC H3T 1P1, Canada; (S.L.); (A.S.); (P.G.); (H.C.)
| |
Collapse
|
27
|
Wang L, Sun F, Wen Y, Yue GH. Effects of Ocean Acidification on Transcriptomes in Asian Seabass Juveniles. MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2021; 23:445-455. [PMID: 33993358 DOI: 10.1007/s10126-021-10036-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 04/28/2021] [Indexed: 06/12/2023]
Abstract
Ocean acidification is changing the fate of marine organisms. It is essential to predict the biological responses and evolutionary processes driven by ocean acidification, to maintain the equilibrium of the marine ecosystem and to facilitate aquaculture. However, how marine organisms, particularly the marine fish species, respond to ocean acidification, is still poorly understood. Consequences of ocean acidification on finfish aquaculture are largely not well known. We studied the effects of ocean acidification for 7 days on growth, behaviour and gene expression profiles in the brain, gill and kidney of Asian seabass juveniles. Results showed that growth and behaviour were not affected by short-term ocean acidification. We found tissue-specific differentially expressed genes (DEGs) involving many molecular processes, such as organ development, growth, muscle development, ion homeostasis and neurogenesis and development, as well as behaviours. Most of the DEGs, which were functionally enriched in ion homeostasis, were related to calcium transport, followed by sodium/potassium channels. We found that genes associated with neurogenesis and development were significantly enriched, implying that ocean acidification has also adversely affected the neural regulatory mechanism. Our results indicate that although the short-term ocean acidification does not cause obvious phenotypic and behavioural changes, it causes substantial changes of gene expressions in all three analysed tissues. All these changes of gene expressions may eventually affect physiological fitness. The DEGs identified here should be further investigated to discover DNA markers associated with adaptability to ocean acidification to improve fish's capability to adapt to ocean acidification.
Collapse
Affiliation(s)
- Le Wang
- Molecular Population Genetics and Breeding Group, Temasek Life Sciences Laboratory, 1 Research Link, National University of Singapore, Singapore, 117604, Singapore
| | - Fei Sun
- Molecular Population Genetics and Breeding Group, Temasek Life Sciences Laboratory, 1 Research Link, National University of Singapore, Singapore, 117604, Singapore
| | - Yanfei Wen
- Molecular Population Genetics and Breeding Group, Temasek Life Sciences Laboratory, 1 Research Link, National University of Singapore, Singapore, 117604, Singapore
| | - Gen Hua Yue
- Molecular Population Genetics and Breeding Group, Temasek Life Sciences Laboratory, 1 Research Link, National University of Singapore, Singapore, 117604, Singapore.
- Department of Biological Sciences, National University of Singapore, 14 Science Drive 4, Singapore, 117543, Singapore.
| |
Collapse
|
28
|
Kaya TB, Aydemir O, Ceylaner S, Ceylaner G, Tekin AN. Isolated congenital diaphragm hernia associated with homozygous SLIT3 gene variant in dizygous twins. Eur J Med Genet 2021; 64:104215. [PMID: 33933663 DOI: 10.1016/j.ejmg.2021.104215] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 03/22/2021] [Accepted: 04/04/2021] [Indexed: 11/16/2022]
Abstract
Congenital diaphragmatic hernia (CDH) is a serious life-threatening birth defect characterized by abnormal development in the muscular or tendinous portion of the diaphragm during embryogenesis. Despite its high incidence, the etiology of CDH hasn't been fully understood. Genetic factors are important in pathogenesis; however, few single genes have been definitively implicated in human CDH. SLIT1, SLIT2, and SLIT3 (slit guidance ligand) are three human homologs of the drosophila Slit gene. They interact with roundabout (Robo) homolog receptors to affect cell migration, adhesion, cell motility, and angiogenesis and play important roles in cell signaling pathways including the guidance of axons. In this report, we presented dizygous twin babies with CDH related to the SLIT3 gene variant. Previous studies showed that Slit3 null mice had congenital diaphragmatic hernias on or near the ventral midline portion of the central tendon. This is the first report of homozygous SLIT3 variant associated with CDH in humans.
Collapse
Affiliation(s)
- Tugba Barsan Kaya
- Eskişehir Osmangazi University Faculty of Medicine, Department of Neonatology, Eskişehir, Turkey.
| | - Ozge Aydemir
- Eskişehir Osmangazi University Faculty of Medicine, Department of Neonatology, Eskişehir, Turkey
| | | | | | - Ayse Neslihan Tekin
- Eskişehir Osmangazi University Faculty of Medicine, Department of Neonatology, Eskişehir, Turkey
| |
Collapse
|
29
|
Knickmeyer MD, Mateo JL, Heermann S. BMP Signaling Interferes with Optic Chiasm Formation and Retinal Ganglion Cell Pathfinding in Zebrafish. Int J Mol Sci 2021; 22:ijms22094560. [PMID: 33925390 PMCID: PMC8123821 DOI: 10.3390/ijms22094560] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/19/2021] [Accepted: 04/24/2021] [Indexed: 11/17/2022] Open
Abstract
Decussation of axonal tracts is an important hallmark of vertebrate neuroanatomy resulting in one brain hemisphere controlling the contralateral side of the body and also computing the sensory information originating from that respective side. Here, we show that BMP interferes with optic chiasm formation and RGC pathfinding in zebrafish. Experimental induction of BMP4 at 15 hpf results in a complete ipsilateral projection of RGC axons and failure of commissural connections of the forebrain, in part as the result of an interaction with shh signaling, transcriptional regulation of midline guidance cues and an affected optic stalk morphogenesis. Experimental induction of BMP4 at 24 hpf, resulting in only a mild repression of forebrain shh ligand expression but in a broad expression of pax2a in the diencephalon, does not per se prevent RGC axons from crossing the midline. It nevertheless shows severe pathologies of RGC projections e.g., the fasciculation of RGC axons with the ipsilateral optic tract resulting in the innervation of one tectum by two eyes or the projection of RGC axons in the direction of the contralateral eye.
Collapse
Affiliation(s)
- Max D. Knickmeyer
- Department of Molecular Embryology, Institute of Anatomy and Cell Biology, Faculty of Medicine, University Freiburg, 79104 Freiburg, Germany;
- Faculty of Biology, University of Freiburg, Schaenzlestrasse 1, 79104 Freiburg, Germany
| | - Juan L. Mateo
- Departamento de Informática, Universidad de Oviedo, Jesús Arias de Velasco, 33005 Oviedo, Spain;
| | - Stephan Heermann
- Department of Molecular Embryology, Institute of Anatomy and Cell Biology, Faculty of Medicine, University Freiburg, 79104 Freiburg, Germany;
- Correspondence:
| |
Collapse
|
30
|
Serres-Armero A, Davis BW, Povolotskaya IS, Morcillo-Suarez C, Plassais J, Juan D, Ostrander EA, Marques-Bonet T. Copy number variation underlies complex phenotypes in domestic dog breeds and other canids. Genome Res 2021; 31:762-774. [PMID: 33863806 PMCID: PMC8092016 DOI: 10.1101/gr.266049.120] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 02/26/2021] [Indexed: 01/02/2023]
Abstract
Extreme phenotypic diversity, a history of artificial selection, and socioeconomic value make domestic dog breeds a compelling subject for genomic research. Copy number variation (CNV) is known to account for a significant part of inter-individual genomic diversity in other systems. However, a comprehensive genome-wide study of structural variation as it relates to breed-specific phenotypes is lacking. We have generated whole genome CNV maps for more than 300 canids. Our data set extends the canine structural variation landscape to more than 100 dog breeds, including novel variants that cannot be assessed using microarray technologies. We have taken advantage of this data set to perform the first CNV-based genome-wide association study (GWAS) in canids. We identify 96 loci that display copy number differences across breeds, which are statistically associated with a previously compiled set of breed-specific morphometrics and disease susceptibilities. Among these, we highlight the discovery of a long-range interaction involving a CNV near MED13L and TBX3, which could influence breed standard height. Integration of the CNVs with chromatin interactions, long noncoding RNA expression, and single nucleotide variation highlights a subset of specific loci and genes with potential functional relevance and the prospect to explain trait variation between dog breeds.
Collapse
Affiliation(s)
- Aitor Serres-Armero
- IBE, Institut de Biologia Evolutiva (Universitat Pompeu Fabra/CSIC), Ciencies Experimentals i de la Salut, Barcelona 08003, Spain
| | - Brian W Davis
- Cancer Genetics and Comparative Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland 20892, USA.,Department of Veterinary Integrative Biosciences, College of Veterinary Medicine, Texas A&M University, College Station, Texas 77843, USA
| | - Inna S Povolotskaya
- Veltischev Research and Clinical Institute for Pediatrics of the Pirogov Russian National Research Medical University, Moscow 117997, Russia
| | - Carlos Morcillo-Suarez
- IBE, Institut de Biologia Evolutiva (Universitat Pompeu Fabra/CSIC), Ciencies Experimentals i de la Salut, Barcelona 08003, Spain
| | - Jocelyn Plassais
- Cancer Genetics and Comparative Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - David Juan
- IBE, Institut de Biologia Evolutiva (Universitat Pompeu Fabra/CSIC), Ciencies Experimentals i de la Salut, Barcelona 08003, Spain
| | - Elaine A Ostrander
- Cancer Genetics and Comparative Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Tomas Marques-Bonet
- IBE, Institut de Biologia Evolutiva (Universitat Pompeu Fabra/CSIC), Ciencies Experimentals i de la Salut, Barcelona 08003, Spain.,CNAG-CRG, Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology (BIST), Barcelona 08028, Spain.,Institucio Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Catalonia 08010, Spain.,Institut Català de Paleontologia Miquel Crusafont, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Catalonia 08201, Spain
| |
Collapse
|
31
|
Slit2 is necessary for optic axon organization in the zebrafish ventral midline. Cells Dev 2021; 166:203677. [PMID: 33994352 DOI: 10.1016/j.cdev.2021.203677] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 03/25/2021] [Accepted: 03/26/2021] [Indexed: 02/07/2023]
Abstract
Slit-Robo signaling has been implicated in regulating several steps of retinal ganglion cell axon guidance, with a central role assigned to Slit2. We report here the phenotypical characterization of a CRISPR-Cas9-generated zebrafish null mutant for this gene, along with a detailed analysis of its expression pattern by WM-FISH. All evident defects in the optic axons in slit2-/- mutants were detected outside the retina, coincident with the major sites of expression at the ventral forebrain, around the developing optic nerve and anterior to the optic chiasm/proximal tract. Anterograde axon tracing experiments in zygotic and maternal-zygotic mutants, as well as morphants, showed the occurrence of axon sorting defects, which appeared mild at the optic nerve level, but more severe in the optic chiasm and the proximal tract. A remarkable sorting defect was the usual splitting of one of the optic nerves in two branches that surrounded the contralateral nerve at the chiasm. Although all axons eventually crossed the midline, the retinotopic order appeared lost at the proximal optic tract, to eventually correct distally. Time-lapse analysis demonstrated the sporadic occurrence of axon misrouting at the chiasm level, which could be responsible for the sorting errors. Our results support previous evidence of a channeling role for Slit molecules in retinal ganglion cell axons at the optic nerve, in addition to a function in the segregation of axons coming from each nerve and from different retinal regions at the medio-ventral area of the forebrain.
Collapse
|
32
|
Morphogenesis of the Islets of Langerhans Is Guided by Extraendocrine Slit2 and Slit3 Signals. Mol Cell Biol 2021; 41:e0045120. [PMID: 33318057 PMCID: PMC8088276 DOI: 10.1128/mcb.00451-20] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The spatial architecture of the islets of Langerhans is vitally important for their correct function, and alterations in islet morphogenesis often result in diabetes mellitus. We have previously reported that Roundabout (Robo) receptors are required for proper islet morphogenesis. As part of the Slit-Robo signaling pathway, Robo receptors function in conjunction with Slit ligands to mediate axon guidance, cell migration, and cell positioning in development. However, the role of Slit ligands in islet morphogenesis has not yet been determined. Here, we report that Slit ligands are expressed in overlapping and distinct patterns in both endocrine and nonendocrine tissues in late pancreas development. We show that the function of either Slit2 or Slit3, which are predominantly expressed in the pancreatic mesenchyme, is required and sufficient for islet morphogenesis, while Slit1, which is predominantly expressed in the β cells, is dispensable for islet morphogenesis. We further show that Slit functions as a repellent signal to β cells. These data suggest that clustering of endocrine cells during islet morphogenesis is guided, at least in part, by repelling Slit2/3 signals from the pancreatic mesenchyme.
Collapse
|
33
|
Rafipay A, Dun X, Parkinson DB, Erskine L, Vargesson N. Knockdown of slit signaling during limb development leads to a reduction in humerus length. Dev Dyn 2021; 250:1340-1357. [DOI: 10.1002/dvdy.284] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 12/16/2020] [Accepted: 12/16/2020] [Indexed: 12/18/2022] Open
Affiliation(s)
- Alexandra Rafipay
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition University of Aberdeen Aberdeen UK
| | - Xin‐Peng Dun
- Peninsula Medical School, Faculty of Health University of Plymouth Plymouth UK
| | - David B Parkinson
- Peninsula Medical School, Faculty of Health University of Plymouth Plymouth UK
| | - Lynda Erskine
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition University of Aberdeen Aberdeen UK
| | - Neil Vargesson
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition University of Aberdeen Aberdeen UK
| |
Collapse
|
34
|
Yamagata M, Yan W, Sanes JR. A cell atlas of the chick retina based on single-cell transcriptomics. eLife 2021; 10:e63907. [PMID: 33393903 PMCID: PMC7837701 DOI: 10.7554/elife.63907] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 01/01/2021] [Indexed: 12/14/2022] Open
Abstract
Retinal structure and function have been studied in many vertebrate orders, but molecular characterization has been largely confined to mammals. We used single-cell RNA sequencing (scRNA-seq) to generate a cell atlas of the chick retina. We identified 136 cell types plus 14 positional or developmental intermediates distributed among the six classes conserved across vertebrates - photoreceptor, horizontal, bipolar, amacrine, retinal ganglion, and glial cells. To assess morphology of molecularly defined types, we adapted a method for CRISPR-based integration of reporters into selectively expressed genes. For Müller glia, we found that transcriptionally distinct cells were regionally localized along the anterior-posterior, dorsal-ventral, and central-peripheral retinal axes. We also identified immature photoreceptor, horizontal cell, and oligodendrocyte types that persist into late embryonic stages. Finally, we analyzed relationships among chick, mouse, and primate retinal cell classes and types. Our results provide a foundation for anatomical, physiological, evolutionary, and developmental studies of the avian visual system.
Collapse
Affiliation(s)
- Masahito Yamagata
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard UniversityCambridgeUnited States
| | - Wenjun Yan
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard UniversityCambridgeUnited States
| | - Joshua R Sanes
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard UniversityCambridgeUnited States
| |
Collapse
|
35
|
Terzi A, Roeder H, Weaver CJ, Suter DM. Neuronal NADPH oxidase 2 regulates growth cone guidance downstream of slit2/robo2. Dev Neurobiol 2020; 81:3-21. [PMID: 33191581 DOI: 10.1002/dneu.22791] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 10/10/2020] [Accepted: 11/09/2020] [Indexed: 02/06/2023]
Abstract
NADPH oxidases (Nox) are membrane-bound multi-subunit protein complexes producing reactive oxygen species (ROS) that regulate many cellular processes. Emerging evidence suggests that Nox-derived ROS also control neuronal development and axonal outgrowth. However, whether Nox act downstream of receptors for axonal growth and guidance cues is presently unknown. To answer this question, we cultured retinal ganglion cells (RGCs) derived from zebrafish embryos and exposed these neurons to netrin-1, slit2, and brain-derived neurotrophic factor (BDNF). To test the role of Nox in cue-mediated growth and guidance, we either pharmacologically inhibited Nox or investigated neurons from mutant fish that are deficient in Nox2. We found that slit2-mediated growth cone collapse, and axonal retraction were eliminated by Nox inhibition. Though we did not see an effect of either BDNF or netrin-1 on growth rates, growth in the presence of netrin-1 was reduced by Nox inhibition. Furthermore, attractive and repulsive growth cone turning in response to gradients of BDNF, netrin-1, and slit2, respectively, were eliminated when Nox was inhibited in vitro. ROS biosensor imaging showed that slit2 treatment increased growth cone hydrogen peroxide levels via mechanisms involving Nox2 activation. We also investigated the possible relationship between Nox2 and slit2/Robo2 signaling in vivo. astray/nox2 double heterozygote larvae exhibited decreased area of tectal innervation as compared to individual heterozygotes, suggesting both Nox2 and Robo2 are required for establishment of retinotectal connections. Our results provide evidence that Nox2 acts downstream of slit2/Robo2 by mediating growth and guidance of developing zebrafish RGC neurons.
Collapse
Affiliation(s)
- Aslihan Terzi
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA.,Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN, USA
| | - Haley Roeder
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA.,Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN, USA
| | - Cory J Weaver
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA.,Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN, USA
| | - Daniel M Suter
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA.,Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN, USA.,Bindley Bioscience Center, Purdue University, West Lafayette, IN, USA.,Birck Nanotechnology Center, Purdue University, West Lafayette, IN, USA
| |
Collapse
|
36
|
Huang Y, Xie Y, Abel PW, Wei P, Plowman J, Toews ML, Strah H, Siddique A, Bailey KL, Tu Y. TGF-β1-induced miR-424 promotes pulmonary myofibroblast differentiation by targeting Slit2 protein expression. Biochem Pharmacol 2020; 180:114172. [PMID: 32712053 PMCID: PMC8742596 DOI: 10.1016/j.bcp.2020.114172] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 07/20/2020] [Accepted: 07/21/2020] [Indexed: 01/09/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a devastating interstitial lung disease with irreversible loss of lung tissue and function. Myofibroblasts in the lung are key cellular mediators of IPF progression. Transforming growth factor (TGF)-β1, a major profibrogenic cytokine, induces pulmonary myofibroblast differentiation, and emerging evidence has established the importance of microRNAs (miRs) in the development of IPF. The objective of this study was to define the pro-fibrotic roles and mechanisms of miRs in TGF-β1-induced pulmonary myofibroblast differentiation. Using RNA sequencing, we identified miR-424 as an important TGF-β1-induced miR in human lung fibroblasts (HLFs). Quantitative RT-PCR confirmed that miR-424 expression was increased by 2.6-fold in HLFs in response to TGF-β1 and was 1.7-fold higher in human fibrotic lung tissues as compared to non-fibrotic lung tissues. TGF-β1-induced upregulation of miR-424 was blocked by the Smad3 inhibitor SIS3, suggesting the involvement of this canonical TGF-β1 signaling pathway. Transfection of a miR-424 hairpin inhibitor into HLFs reduced TGF-β1-induced expression of classic myofibroblast differentiation markers including ɑ-smooth muscle actin (ɑ-SMA) and connective tissue growth factor (CTGF), whereas a miR-424 mimic significantly enhanced TGF-β1-induced myofibroblast differentiation. In addition, TGF-β1 induced Smad3 phosphorylation in HLFs, and this response was reduced by the miR-424 inhibitor. In silico analysis identified Slit2, a protein that inhibits TGF-β1 profibrogenic signaling, as a putative target of regulation by miR-424. Slit2 is less highly expressed in human fibrotic lung tissues than in non-fibrotic lung tissues, and knockdown of Slit2 by its siRNA enhanced TGF-β1-induced HLF differentiation. Overexpression of a miR-424 mimic down-regulated expression of Slit2 but not the Slit2 major receptor ROBO1 in HLFs. Luciferase reporter assays showed that the miR-424 mimic represses Slit2 3' untranslated region (3'-UTR) reporter activity, and mutations at the seeding regions in the 3'-UTR of Slit2 abolish this inhibition. Together, these data demonstrate a pro-fibrotic role of miR-424 in TGF-β1-induced HLF differentiation. It functions as a positive feed-back regulator of the TGF-β1 signaling pathway by reducing expression of the negative regulator Slit2. Thus, targeting miR-424 may provide a new therapeutic strategy to prevent myofibroblast differentiation and IPF progression.
Collapse
Affiliation(s)
- Yapei Huang
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, NE 68178, USA
| | - Yan Xie
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, NE 68178, USA
| | - Peter W Abel
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, NE 68178, USA
| | - Peng Wei
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, NE 68178, USA
| | - Jocelyn Plowman
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, NE 68178, USA
| | - Myron L Toews
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Heather Strah
- Department of Internal Medicine, Pulmonary Critical Care, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Aleem Siddique
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Kristina L Bailey
- Department of Internal Medicine, Pulmonary Critical Care, University of Nebraska Medical Center, Omaha, NE 68198, USA; VA Nebraska-Western Iowa Health Care Center, Omaha, NE 68105, USA.
| | - Yaping Tu
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, NE 68178, USA.
| |
Collapse
|
37
|
Bhosle VK, Mukherjee T, Huang YW, Patel S, Pang BWF, Liu GY, Glogauer M, Wu JY, Philpott DJ, Grinstein S, Robinson LA. SLIT2/ROBO1-signaling inhibits macropinocytosis by opposing cortical cytoskeletal remodeling. Nat Commun 2020; 11:4112. [PMID: 32807784 PMCID: PMC7431850 DOI: 10.1038/s41467-020-17651-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 07/08/2020] [Indexed: 01/06/2023] Open
Abstract
Macropinocytosis is essential for myeloid cells to survey their environment and for growth of RAS-transformed cancer cells. Several growth factors and inflammatory stimuli are known to induce macropinocytosis, but its endogenous inhibitors have remained elusive. Stimulation of Roundabout receptors by Slit ligands inhibits directional migration of many cell types, including immune cells and cancer cells. We report that SLIT2 inhibits macropinocytosis in vitro and in vivo by inducing cytoskeletal changes in macrophages. In mice, SLIT2 attenuates the uptake of muramyl dipeptide, thereby preventing NOD2-dependent activation of NF-κB and consequent secretion of pro-inflammatory chemokine, CXCL1. Conversely, blocking the action of endogenous SLIT2 enhances CXCL1 secretion. SLIT2 also inhibits macropinocytosis in RAS-transformed cancer cells, thereby decreasing their survival in nutrient-deficient conditions which resemble tumor microenvironment. Our results identify SLIT2 as a physiological inhibitor of macropinocytosis and challenge the conventional notion that signals that enhance macropinocytosis negatively regulate cell migration, and vice versa.
Collapse
Affiliation(s)
- Vikrant K Bhosle
- Program in Cell Biology, The Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, 686 Bay Street, Toronto, ON, M5G 0A4, Canada
| | - Tapas Mukherjee
- Department of Immunology, University of Toronto, Medical Sciences Building, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada
| | - Yi-Wei Huang
- Program in Cell Biology, The Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, 686 Bay Street, Toronto, ON, M5G 0A4, Canada
| | - Sajedabanu Patel
- Program in Cell Biology, The Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, 686 Bay Street, Toronto, ON, M5G 0A4, Canada
| | - Bo Wen Frank Pang
- Program in Cell Biology, The Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, 686 Bay Street, Toronto, ON, M5G 0A4, Canada
- Institute of Medical Science, University of Toronto, Medical Sciences Building, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada
- BenchSci, Suite 201, 559 College Street, Toronto, ON, M6G 1A9, Canada
| | - Guang-Ying Liu
- Program in Cell Biology, The Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, 686 Bay Street, Toronto, ON, M5G 0A4, Canada
| | - Michael Glogauer
- Faculty of Dentistry, University of Toronto, 101 Elm Street, Toronto, ON, M5G 2L3, Canada
- Department of Dental Oncology and Maxillofacial Prosthetics, University Health Network, Princess Margaret Cancer Centre, 610 University Avenue, Toronto, ON, M5G 2C1, Canada
- Centre for Advanced Dental Research and Care, Mount Sinai Hospital, 600 University Avenue, Toronto, ON, M5G 1X5, Canada
| | - Jane Y Wu
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
- Lurie Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Dana J Philpott
- Department of Immunology, University of Toronto, Medical Sciences Building, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada
| | - Sergio Grinstein
- Program in Cell Biology, The Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, 686 Bay Street, Toronto, ON, M5G 0A4, Canada
- Department of Biochemistry, University of Toronto, Medical Sciences Building, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada
- Keenan Research Centre of the Li Ka Shing Knowledge Institute, St. Michael's Hospital, 290 Victoria Street, Toronto, ON, M5C 1N8, Canada
| | - Lisa A Robinson
- Program in Cell Biology, The Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, 686 Bay Street, Toronto, ON, M5G 0A4, Canada.
- Institute of Medical Science, University of Toronto, Medical Sciences Building, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada.
- Division of Nephrology, The Hospital for Sick Children, 555 University Avenue, Toronto, ON, M5G 1X8, Canada.
- Department of Paediatrics, Faculty of Medicine, University of Toronto, 555 University Avenue, Toronto, ON, M5G 1X8, Canada.
| |
Collapse
|
38
|
Deboux C, Spigoni G, Caillava C, Garcia-Diaz B, Ypsilanti A, Sarrazin N, Bachelin C, Chédotal A, Baron-Van Evercooren A. Slit1 Protein Regulates SVZ-Derived Precursor Mobilization in the Adult Demyelinated CNS. Front Cell Neurosci 2020; 14:168. [PMID: 32670024 PMCID: PMC7332780 DOI: 10.3389/fncel.2020.00168] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 05/19/2020] [Indexed: 01/03/2023] Open
Abstract
Slit1 is a secreted axon guidance molecule, also involved in adult neurogenesis. In physiological conditions, Slit1 loss promotes ectopic dispersal of SVZ-derived neural precursors (SVZ-NPCs) into periventricular structures such as the corpus callosum. Demyelination of the corpus callosum triggers SVZ-NPC migration to ectopic locations and their recruitment by the lesion, suggesting a possible role for Slit1 in SVZ-NPCs ectopic dispersal regulation in pathological conditions. Here, we have investigated the function of Slit1 protein in the recruitment of SVZ-NPCs after CNS demyelination. We find that the dynamics of oligodendrogenesis and temporal profile of developmental myelination in Slit1–/– mice are similar to Slit1+/− controls. SVZ micro-dissection and RT-PCR from wild-type mice, show that Slits and Robos are physiologically regulated at the transcriptional level in response to corpus callosum demyelination suggesting their role in the process of SVZ-NPC ectopic migration in demyelinating conditions. Moreover, we find that the number of SVZ-NPCs recruited by the lesion increases in Sli1–/– mice compared to Slit1+/− mice, leading to higher numbers of Olig2+ cells within the lesion. Time-lapse video-microscopy of immuno-purified NPCs shows that Slit1-deficient cells migrate faster and make more frequent directional changes than control NPCs, supporting a cell-autonomous mechanism of action of Slit1 in NPC migration. In conclusion, while Slit1 does not affect the normal developmental process of oligodendrogenesis and myelination, it regulates adult SVZ-NPC ectopic migration in response to demyelination, and consequently oligodendrocyte renewal within the lesion.
Collapse
Affiliation(s)
- C Deboux
- Institut du Cerveau et de la Moelle épinière-Groupe Hospitalier Pitié-Salpêtrière, INSERM U1127, CNRS, UMR 7225, Sorbonne Université, UM75, Paris, France
| | - G Spigoni
- Institut du Cerveau et de la Moelle épinière-Groupe Hospitalier Pitié-Salpêtrière, INSERM U1127, CNRS, UMR 7225, Sorbonne Université, UM75, Paris, France
| | - C Caillava
- Institut du Cerveau et de la Moelle épinière-Groupe Hospitalier Pitié-Salpêtrière, INSERM U1127, CNRS, UMR 7225, Sorbonne Université, UM75, Paris, France
| | - B Garcia-Diaz
- Institut du Cerveau et de la Moelle épinière-Groupe Hospitalier Pitié-Salpêtrière, INSERM U1127, CNRS, UMR 7225, Sorbonne Université, UM75, Paris, France
| | - A Ypsilanti
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, Paris, France
| | - N Sarrazin
- Institut du Cerveau et de la Moelle épinière-Groupe Hospitalier Pitié-Salpêtrière, INSERM U1127, CNRS, UMR 7225, Sorbonne Université, UM75, Paris, France
| | - C Bachelin
- Institut du Cerveau et de la Moelle épinière-Groupe Hospitalier Pitié-Salpêtrière, INSERM U1127, CNRS, UMR 7225, Sorbonne Université, UM75, Paris, France
| | - A Chédotal
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, Paris, France
| | - A Baron-Van Evercooren
- Institut du Cerveau et de la Moelle épinière-Groupe Hospitalier Pitié-Salpêtrière, INSERM U1127, CNRS, UMR 7225, Sorbonne Université, UM75, Paris, France
| |
Collapse
|
39
|
Gong L, Wang S, Shen L, Liu C, Shenouda M, Li B, Liu X, Shaw JA, Wineman AL, Yang Y, Xiong D, Eichmann A, Evans SM, Weiss SJ, Si MS. SLIT3 deficiency attenuates pressure overload-induced cardiac fibrosis and remodeling. JCI Insight 2020; 5:136852. [PMID: 32644051 PMCID: PMC7406261 DOI: 10.1172/jci.insight.136852] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 05/06/2020] [Indexed: 01/28/2023] Open
Abstract
In pulmonary hypertension and certain forms of congenital heart disease, ventricular pressure overload manifests at birth and is an obligate hemodynamic abnormality that stimulates myocardial fibrosis, which leads to ventricular dysfunction and poor clinical outcomes. Thus, an attractive strategy is to attenuate the myocardial fibrosis to help preserve ventricular function. Here, by analyzing RNA-sequencing databases and comparing the transcript and protein levels of fibrillar collagen in WT and global-knockout mice, we found that slit guidance ligand 3 (SLIT3) was present predominantly in fibrillar collagen-producing cells and that SLIT3 deficiency attenuated collagen production in the heart and other nonneuronal tissues. We then performed transverse aortic constriction or pulmonary artery banding to induce left and right ventricular pressure overload, respectively, in WT and knockout mice. We discovered that SLIT3 deficiency abrogated fibrotic and hypertrophic changes and promoted long-term ventricular function and overall survival in both left and right ventricular pressure overload. Furthermore, we found that SLIT3 stimulated fibroblast activity and fibrillar collagen production, which coincided with the transcription and nuclear localization of the mechanotransducer yes-associated protein 1. These results indicate that SLIT3 is important for regulating fibroblast activity and fibrillar collagen synthesis in an autocrine manner, making it a potential therapeutic target for fibrotic diseases, especially myocardial fibrosis and adverse remodeling induced by persistent afterload elevation.
Collapse
Affiliation(s)
- Lianghui Gong
- Section of Pediatric Cardiovascular Surgery, Department of Cardiac Surgery, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA.,Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Shuyun Wang
- Section of Pediatric Cardiovascular Surgery, Department of Cardiac Surgery, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Li Shen
- Section of Pediatric Cardiovascular Surgery, Department of Cardiac Surgery, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Catherine Liu
- Section of Pediatric Cardiovascular Surgery, Department of Cardiac Surgery, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Mena Shenouda
- Section of Pediatric Cardiovascular Surgery, Department of Cardiac Surgery, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Baolei Li
- Section of Pediatric Cardiovascular Surgery, Department of Cardiac Surgery, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Xiaoxiao Liu
- Section of Pediatric Cardiovascular Surgery, Department of Cardiac Surgery, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | | | - Alan L. Wineman
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Yifeng Yang
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Dingding Xiong
- Section of Pediatric Cardiovascular Surgery, Department of Cardiac Surgery, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Anne Eichmann
- Yale Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA.,Paris Cardiovascular Research Center, INSERM U970, Paris, France.,Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Sylvia M. Evans
- Skaggs School of Pharmacy and Pharmaceutical Sciences,,Department of Medicine, and,Department of Pharmacology, UCSD, La Jolla, California, USA
| | - Stephen J. Weiss
- Division of Genetic Medicine,,Department of Internal Medicine,,Life Sciences Institute,,Cellular and Molecular Biology Graduate Program, and,Rogel Cancer Center, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Ming-Sing Si
- Section of Pediatric Cardiovascular Surgery, Department of Cardiac Surgery, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
40
|
Romanov RA, Tretiakov EO, Kastriti ME, Zupancic M, Häring M, Korchynska S, Popadin K, Benevento M, Rebernik P, Lallemend F, Nishimori K, Clotman F, Andrews WD, Parnavelas JG, Farlik M, Bock C, Adameyko I, Hökfelt T, Keimpema E, Harkany T. Molecular design of hypothalamus development. Nature 2020; 582:246-252. [PMID: 32499648 PMCID: PMC7292733 DOI: 10.1038/s41586-020-2266-0] [Citation(s) in RCA: 105] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Accepted: 03/05/2020] [Indexed: 12/21/2022]
Abstract
A wealth of specialized neuroendocrine command systems intercalated within the hypothalamus control the most fundamental physiological needs in vertebrates1,2. Nevertheless, we lack a developmental blueprint that integrates the molecular determinants of neuronal and glial diversity along temporal and spatial scales of hypothalamus development3. Here we combine single-cell RNA sequencing of 51,199 mouse cells of ectodermal origin, gene regulatory network (GRN) screens in conjunction with genome-wide association study-based disease phenotyping, and genetic lineage reconstruction to show that nine glial and thirty-three neuronal subtypes are generated by mid-gestation under the control of distinct GRNs. Combinatorial molecular codes that arise from neurotransmitters, neuropeptides and transcription factors are minimally required to decode the taxonomical hierarchy of hypothalamic neurons. The differentiation of γ-aminobutyric acid (GABA) and dopamine neurons, but not glutamate neurons, relies on quasi-stable intermediate states, with a pool of GABA progenitors giving rise to dopamine cells4. We found an unexpected abundance of chemotropic proliferation and guidance cues that are commonly implicated in dorsal (cortical) patterning5 in the hypothalamus. In particular, loss of SLIT-ROBO signalling impaired both the production and positioning of periventricular dopamine neurons. Overall, we identify molecular principles that shape the developmental architecture of the hypothalamus and show how neuronal heterogeneity is transformed into a multimodal neural unit to provide virtually infinite adaptive potential throughout life.
Collapse
Affiliation(s)
- Roman A. Romanov
- Department of Molecular Neurosciences, Center for Brain Research,
Medical University of Vienna, Vienna, Austria
- Department of Neuroscience, Biomedicum D7, Karolinska Institutet,
Solna, Sweden
| | - Evgenii O. Tretiakov
- Department of Molecular Neurosciences, Center for Brain Research,
Medical University of Vienna, Vienna, Austria
| | - Maria Eleni Kastriti
- Department of Molecular Neurosciences, Center for Brain Research,
Medical University of Vienna, Vienna, Austria
- Department of Physiology and Pharmacology, Biomedicum D6, Karolinska
Institutet, Solna, Sweden
| | - Maja Zupancic
- Department of Molecular Neurosciences, Center for Brain Research,
Medical University of Vienna, Vienna, Austria
| | - Martin Häring
- Department of Molecular Neurosciences, Center for Brain Research,
Medical University of Vienna, Vienna, Austria
| | - Solomiia Korchynska
- Department of Molecular Neurosciences, Center for Brain Research,
Medical University of Vienna, Vienna, Austria
| | - Konstantin Popadin
- Human Genomics of Infection and Immunity, School of Life Sciences,
Ecole Polytechnique Federale de Lausanne, Lausanne, Switzerland
| | - Marco Benevento
- Department of Molecular Neurosciences, Center for Brain Research,
Medical University of Vienna, Vienna, Austria
| | - Patrick Rebernik
- Department of Molecular Neurosciences, Center for Brain Research,
Medical University of Vienna, Vienna, Austria
| | - Francois Lallemend
- Department of Neuroscience, Biomedicum D7, Karolinska Institutet,
Solna, Sweden
| | - Katsuhiko Nishimori
- Deptartment of Obesity and Internal Inflammation, Fukushima Medical
University, Fukushima City, Japan
| | - Frédéric Clotman
- Laboratory of Neural Differentiation, Institute of Neuroscience,
Université Catholique de Louvain, Brussels, Belgium
| | - William D. Andrews
- Department of Cell and Developmental Biology, University College
London, London, United Kingdom
| | - John G. Parnavelas
- Department of Cell and Developmental Biology, University College
London, London, United Kingdom
| | - Matthias Farlik
- CeMM Research Center for Molecular Medicine of the Austrian Academy
of Sciences, Vienna, Austria
- Department of Dermatology, Medical University of Vienna, Vienna,
Austria
| | - Christoph Bock
- CeMM Research Center for Molecular Medicine of the Austrian Academy
of Sciences, Vienna, Austria
- Department of Laboratory Medicine, Medical University of Vienna,
Vienna, Austria
| | - Igor Adameyko
- Department of Molecular Neurosciences, Center for Brain Research,
Medical University of Vienna, Vienna, Austria
- Department of Physiology and Pharmacology, Biomedicum D6, Karolinska
Institutet, Solna, Sweden
| | - Tomas Hökfelt
- Department of Neuroscience, Biomedicum D7, Karolinska Institutet,
Solna, Sweden
| | - Erik Keimpema
- Department of Molecular Neurosciences, Center for Brain Research,
Medical University of Vienna, Vienna, Austria
| | - Tibor Harkany
- Department of Molecular Neurosciences, Center for Brain Research,
Medical University of Vienna, Vienna, Austria
- Department of Neuroscience, Biomedicum D7, Karolinska Institutet,
Solna, Sweden
| |
Collapse
|
41
|
Sasaki T, Komatsu Y, Yamamori T. Expression patterns of SLIT/ROBO mRNAs reveal a characteristic feature in the entorhinal-hippocampal area of macaque monkeys. BMC Res Notes 2020; 13:262. [PMID: 32460877 PMCID: PMC7251749 DOI: 10.1186/s13104-020-05100-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Accepted: 05/19/2020] [Indexed: 01/06/2023] Open
Abstract
OBJECTIVE SLITs are secreted glycoproteins that bind to Roundabouts (ROBOs) which are a family member of transmembrane receptors. SLIT signaling has well-conserved roles in mediating axon repulsion in a developing nervous system. We previously reported that SLIT1 mRNA is enriched in middle layers of the prefrontal cortex of macaque monkeys in a developmentally regulated manner. Other SLIT (SLIT2 and SLIT3) mRNAs showed preferential expressions in the prefrontal cortex with a distinct laminar pattern. To obtain further clues to the role of SLIT signaling in the organization of the primate brain, we performed ISH analysis of SLIT and ROBO mRNAs using adult macaque brain tissues. RESULTS In this study, we examined the expression patterns of SLITs and ROBOs (ROBO1 and ROBO2) in other brain regions, and found intense and characteristic expression patterns of these genes in the entorhinal-hippocampal area. In situ hybridization analysis revealed that SLIT1 and SLIT2 mRNAs showed marked complementary distribution in the entorhinal cortex. SLIT and ROBO mRNAs were widely expressed in the hippocampus with modest regional preference. These findings suggest that each SLIT gene has a specialized role that is particularly important for prefrontal as well as hippocampal connectivity in the primate cortex.
Collapse
Affiliation(s)
- Tetsuya Sasaki
- Division of Brain Biology, National Institute for Basic Biology, 38 Nishigonaka Myodaiji, Okazaki, 444-8585, Japan.
- Department of Anatomy and Neuroscience, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8577, Japan.
- Ph.D Program of Neurosciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8577, Japan.
| | - Yusuke Komatsu
- Division of Brain Biology, National Institute for Basic Biology, 38 Nishigonaka Myodaiji, Okazaki, 444-8585, Japan
- Laboratory of Veterinary Hygiene, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, 060-0816, Japan
| | - Tetsuo Yamamori
- Division of Brain Biology, National Institute for Basic Biology, 38 Nishigonaka Myodaiji, Okazaki, 444-8585, Japan.
- Laboratory for Molecular Analysis of Higher Brain Function, RIKEN Center for Brain Science, Wako, Saitama, 351-0198, Japan.
| |
Collapse
|
42
|
Hall CM, Moeendarbary E, Sheridan GK. Mechanobiology of the brain in ageing and Alzheimer's disease. Eur J Neurosci 2020; 53:3851-3878. [DOI: 10.1111/ejn.14766] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 04/20/2020] [Accepted: 04/23/2020] [Indexed: 02/07/2023]
Affiliation(s)
- Chloe M. Hall
- Department of Mechanical Engineering University College London London UK
- School of Pharmacy and Biomolecular Sciences University of Brighton Brighton UK
| | - Emad Moeendarbary
- Department of Mechanical Engineering University College London London UK
- Department of Biological Engineering Massachusetts Institute of Technology Cambridge MA USA
| | - Graham K. Sheridan
- School of Life Sciences Queens Medical Centre University of Nottingham Nottingham UK
| |
Collapse
|
43
|
Abstract
Binocular vision depends on retinal ganglion cell (RGC) axon projection either to the same side or to the opposite side of the brain. In this article, we review the molecular mechanisms for decussation of RGC axons, with a focus on axon guidance signaling at the optic chiasm and ipsi- and contralateral axon organization in the optic tract prior to and during targeting. The spatial and temporal features of RGC neurogenesis that give rise to ipsilateral and contralateral identity are described. The albino visual system is highlighted as an apt comparative model for understanding RGC decussation, as albinos have a reduced ipsilateral projection and altered RGC neurogenesis associated with perturbed melanogenesis in the retinal pigment epithelium. Understanding the steps for RGC specification into ipsi- and contralateral subtypes will facilitate differentiation of stem cells into RGCs with proper navigational abilities for effective axon regeneration and correct targeting of higher-order visual centers.
Collapse
Affiliation(s)
- Carol Mason
- Department of Pathology and Cell Biology, Columbia University, New York, NY 10027, USA; .,Department of Neuroscience, Columbia University, New York, NY 10027, USA.,Department of Ophthalmology, Columbia University, New York, NY 10027, USA.,Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA;
| | - Nefeli Slavi
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA;
| |
Collapse
|
44
|
Castro Martínez XH, Moltó Ruiz MD, Morales Marin ME, Flores Lázaro JC, González Fernández J, Gutiérrez Najera NA, Alvarez Amado DE, Nicolini Sánchez JH. FOXP2 and language alterations in psychiatric pathology. SALUD MENTAL 2019; 42:297-308. [DOI: 10.17711/sm.0185-3325.2019.039] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/13/2024]
Abstract
Background. From the first reports of the linguist Noam Chomsky it has become clear that the development of language has an important genetic component. Several reports in families have shown the relationship between language disorders and genetic polymorphisms. The FOXP2 gene has been a fundamental piece for the understanding of language development. This gene codes for a transcription factor containing a forkhead domain of DNA binding and participates in the regulation of the expression of a large number of genes involved in the embryonic development of fundamental neuronal structures needed for the development of speech and language. Objective. To present an updated view of the relationship between FOXP2 and language alterations in psychiatric pathology. Method. Narrative review of information reported in databases on the recent advances supporting genetic participation in language disorders of psychiatric illness. Results. Update of content related to FOXP2 and its participation in language alterations in psychiatric diseases. Discussion and conclusion. Advances in the genetic study of language disorders in psychiatric pathology open up new avenues of investigation that allow us to explore how language emerged and how it evolved, as well as to carry out comparative studies on the structure and functioning of genes to approach the understanding of this complex characteristic that makes us human.
Collapse
|
45
|
Kim M, Lee CH, Barnum SJ, Watson RC, Li J, Mastick GS. Slit/Robo signals prevent spinal motor neuron emigration by organizing the spinal cord basement membrane. Dev Biol 2019; 455:449-457. [PMID: 31356769 PMCID: PMC6842423 DOI: 10.1016/j.ydbio.2019.07.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 07/24/2019] [Accepted: 07/24/2019] [Indexed: 01/30/2023]
Abstract
The developing spinal cord builds a boundary between the CNS and the periphery, in the form of a basement membrane. The spinal cord basement membrane is a barrier that retains CNS neuron cell bodies, while being selectively permeable to specific axon types. Spinal motor neuron cell bodies are located in the ventral neural tube next to the floor plate and project their axons out through the basement membrane to peripheral targets. However, little is known about how spinal motor neuron cell bodies are retained inside the ventral neural tube, while their axons can exit. In previous work, we found that disruption of Slit/Robo signals caused motor neuron emigration outside the spinal cord. In the current study, we investigate how Slit/Robo signals are necessary to keep spinal motor neurons within the neural tube. Our findings show that when Slit/Robo signals were removed from motor neurons, they migrated outside the spinal cord. Furthermore, this emigration was associated with abnormal basement membrane protein expression in the ventral spinal cord. Using Robo2 and Slit2 conditional mutants, we found that motor neuron-derived Slit/Robo signals were required to set up a normal basement membrane in the spinal cord. Together, our results suggest that motor neurons produce Slit signals that are required for the basement membrane assembly to retain motor neuron cell bodies within the spinal cord.
Collapse
Affiliation(s)
- Minkyung Kim
- Department of Biology, University of Nevada, Reno, NV, 89557, USA.
| | - Clare H Lee
- Department of Biology, University of Nevada, Reno, NV, 89557, USA
| | - Sarah J Barnum
- Department of Biology, University of Nevada, Reno, NV, 89557, USA
| | - Roland Cj Watson
- Department of Biology, University of Nevada, Reno, NV, 89557, USA
| | - Jennifer Li
- Department of Biology, University of Nevada, Reno, NV, 89557, USA
| | - Grant S Mastick
- Department of Biology, University of Nevada, Reno, NV, 89557, USA
| |
Collapse
|
46
|
Conceição R, Evans RS, Pearson CS, Hänzi B, Osborne A, Deshpande SS, Martin KR, Barber AC. Expression of Developmentally Important Axon Guidance Cues in the Adult Optic Chiasm. Invest Ophthalmol Vis Sci 2019; 60:4727-4739. [PMID: 31731293 PMCID: PMC6859889 DOI: 10.1167/iovs.19-26732] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Purpose Regeneration of optic nerve axons after injury can be facilitated by several approaches, but misguidance at the optic chiasm is often observed. We characterized guidance cues in the embryonic visual system and adult optic chiasm before and after optic nerve crush (ONC) injury to better understand barriers to optic nerve regeneration in adults. Methods Radial glial (RC2/BLBP/Slit1), developmental (Pax2) and extracellular markers (CSPG: H2B/CS-56) were assessed in C57BL/6J mice by immunohistochemistry. RC2, BLBP, Slit1, and CSPG are known inhibitory guidance cues while Pax2 is a permissive guidance cue. Results At embryonic day 15.5 (E.15.5), RC2 and BLBP were identified superior to, and extending through, the optic chiasm. The optic chiasm was BLBP-ve in adult uninjured mice but BLBP+ve in adult mice 10 days after ONC injury. The reverse was true for RC2. Both BLBP and RC2 were absent in adult mice 6 weeks post-ONC. Slit1 was present in the optic chiasm midline and optic tracts in embryonic samples but was absent in uninjured adult tissue. Slit1 was observed superior to and at the midline of the optic chiasm 10 days post-ONC but absent 6 weeks after injury. Pax2 was expressed at the junction between the optic nerve and optic chiasm in embryonic brain tissue. In embryonic sections, CS-56 was observed at the junction between the optic chiasm and optic tract, and immediately superior to the optic chiasm. Both 2H6 and CS-56 staining was absent in uninjured and ONC-injured adult brains. Conclusion Differences in guidance cue expression during development, in adulthood and after injury may contribute to misguidance of regenerating RGC axons in the adult optic chiasm.
Collapse
Affiliation(s)
- Raquel Conceição
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, United Kingdom
| | - Rachel S Evans
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, United Kingdom
| | - Craig S Pearson
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, United Kingdom.,Laboratory of Developmental Neurobiology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, United States
| | - Barbara Hänzi
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, United Kingdom
| | - Andrew Osborne
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, United Kingdom
| | - Sarita S Deshpande
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, United Kingdom
| | - Keith R Martin
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, United Kingdom.,Centre for Eye Research Australia, Melbourne, Australia.,University of Melbourne, Melbourne, Australia.,Department of Ophthalmology, NIHR Biomedical Research Centre and Wellcome Trust-MRC Cambridge Stem Cell Institute, University of Cambridge, United Kingdom
| | - Amanda C Barber
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, United Kingdom
| |
Collapse
|
47
|
Murcia-Belmonte V, Erskine L. Wiring the Binocular Visual Pathways. Int J Mol Sci 2019; 20:ijms20133282. [PMID: 31277365 PMCID: PMC6651880 DOI: 10.3390/ijms20133282] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 06/29/2019] [Accepted: 07/03/2019] [Indexed: 02/06/2023] Open
Abstract
Retinal ganglion cells (RGCs) extend axons out of the retina to transmit visual information to the brain. These connections are established during development through the navigation of RGC axons along a relatively long, stereotypical pathway. RGC axons exit the eye at the optic disc and extend along the optic nerves to the ventral midline of the brain, where the two nerves meet to form the optic chiasm. In animals with binocular vision, the axons face a choice at the optic chiasm—to cross the midline and project to targets on the contralateral side of the brain, or avoid crossing the midline and project to ipsilateral brain targets. Ipsilaterally and contralaterally projecting RGCs originate in disparate regions of the retina that relate to the extent of binocular overlap in the visual field. In humans virtually all RGC axons originating in temporal retina project ipsilaterally, whereas in mice, ipsilaterally projecting RGCs are confined to the peripheral ventrotemporal retina. This review will discuss recent advances in our understanding of the mechanisms regulating specification of ipsilateral versus contralateral RGCs, and the differential guidance of their axons at the optic chiasm. Recent insights into the establishment of congruent topographic maps in both brain hemispheres also will be discussed.
Collapse
Affiliation(s)
| | - Lynda Erskine
- School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, Scotland AB25 2ZD, UK
| |
Collapse
|
48
|
Denisova K. Failure to attune to language predicts autism in high risk infants. BRAIN AND LANGUAGE 2019; 194:109-120. [PMID: 31133435 DOI: 10.1016/j.bandl.2019.04.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 04/10/2019] [Accepted: 04/10/2019] [Indexed: 06/09/2023]
Abstract
Young humans are typically sensitive to evolutionarily important aspects of information in the surrounding environment in a way that makes us thrive. Seeking to probe the putative disruptions of this process in infancy, I examined the statistical character of head movements in 52 9-10 mo-old infants, half at high familial risk (HR) for Autism Spectrum Disorders (ASD), who underwent an fMRI scan while listening to words spoken with alternating stress patterns on syllables. Relative to low risk (LR) infants, HR infants, in particular those showing the least rapid receptive language progress, had significantly lower noise-to-signal levels and increased symmetry. A comparison of patterns during a native language and a sleep scan revealed the most atypical ordering of signatures on the 3 tasks in a subset of HR infants, suggesting that the biological mechanism of language development is least acquisitive in those HR infants who go on to develop ASD in toddlerhood.
Collapse
Affiliation(s)
- Kristina Denisova
- Sackler Institute for Developmental Psychobiology, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA; Department of Psychiatry, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA; Division of Developmental Neuroscience, New York State Psychiatric Institute, New York, NY 10032, USA.
| |
Collapse
|
49
|
Mason C, Guillery R. Conversations with Ray Guillery on albinism: linking Siamese cat visual pathway connectivity to mouse retinal development. Eur J Neurosci 2019; 49:913-927. [PMID: 30801828 DOI: 10.1111/ejn.14396] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 01/23/2019] [Accepted: 02/12/2019] [Indexed: 02/06/2023]
Abstract
In albinism of all species, perturbed melanin biosynthesis in the eye leads to foveal hypoplasia, retinal ganglion cell misrouting, and, consequently, altered binocular vision. Here, written before he died, Ray Guillery chronicles his discovery of the aberrant circuitry from eye to brain in the Siamese cat. Ray's characterization of visual pathway anomalies in this temperature sensitive mutation of tyrosinase and thus melanin synthesis in domestic cats opened the exploration of albinism and simultaneously, a genetic approach to the organization of neural circuitry. I follow this account with a remembrance of Ray's influence on my work. Beginning with my postdoc research with Ray on the cat visual pathway, through my own work on the mechanisms of retinal axon guidance in the developing mouse, Ray and I had a continuous and rich dialogue about the albino visual pathway. I will present the questions Ray posed and clues we have to date on the still-elusive link between eye pigment and the proper balance of ipsilateral and contralateral retinal ganglion cell projections to the brain.
Collapse
Affiliation(s)
- Carol Mason
- Departments of Pathology and Cell Biology, Neuroscience, and Ophthalmology, Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, Jerome L. Greene Science Center, 3227 Broadway, Room L3-043, Quad 3C, New York, NY, 10027, USA
| | - Ray Guillery
- Departments of Pathology and Cell Biology, Neuroscience, and Ophthalmology, Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, Jerome L. Greene Science Center, 3227 Broadway, Room L3-043, Quad 3C, New York, NY, 10027, USA
| |
Collapse
|
50
|
Murcia-Belmonte V, Coca Y, Vegar C, Negueruela S, de Juan Romero C, Valiño AJ, Sala S, DaSilva R, Kania A, Borrell V, Martinez LM, Erskine L, Herrera E. A Retino-retinal Projection Guided by Unc5c Emerged in Species with Retinal Waves. Curr Biol 2019; 29:1149-1160.e4. [PMID: 30905607 PMCID: PMC6453780 DOI: 10.1016/j.cub.2019.02.052] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 01/04/2019] [Accepted: 02/22/2019] [Indexed: 12/29/2022]
Abstract
The existence of axons extending from one retina to the other has been reported during perinatal development in different vertebrates. However, it has been thought that these axons are either a labeling artifact or misprojections. Here, we show unequivocally that a small subset of retinal ganglion cells (RGCs) project to the opposite retina and that the guidance receptor Unc5c, expressed in the retinal region where the retinal-retinal (R-R) RGCs are located, is necessary and sufficient to guide axons to the opposite retina. In addition, Netrin1, an Unc5c ligand, is expressed in the ventral diencephalon in a pattern that is consistent with impeding the growth of Unc5c-positive retinal axons into the brain. We also have generated a mathematical model to explore the formation of retinotopic maps in the presence and absence of a functional connection between both eyes. This model predicts that an R-R connection is required for the bilateral coordination of axonal refinement in species where refinement depends upon spontaneous retinal waves. Consistent with this idea, the retinal expression of Unc5c correlates with the existence and size of an R-R projection in different species and with the extent of axonal refinement in visual targets. These findings demonstrate that active guidance drives the formation of the R-R projection and suggest an important role for these projections in visual mapping to ensure congruent bilateral refinement. A subset of retinal ganglion cells project to the contralateral retina Unc5c mediates the formation of the retina-retina projection Unc5c retinal expression correlates with extent of refinement in visual targets Congruency of visual maps in species with retinal waves may rely on R-R axons
Collapse
Affiliation(s)
- Verónica Murcia-Belmonte
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández, CSIC-UMH, Av. Santiago Ramón y Cajal s/n, Sant Joan d'Alacant 03550, Alicante, Spain
| | - Yaiza Coca
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández, CSIC-UMH, Av. Santiago Ramón y Cajal s/n, Sant Joan d'Alacant 03550, Alicante, Spain
| | - Celia Vegar
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández, CSIC-UMH, Av. Santiago Ramón y Cajal s/n, Sant Joan d'Alacant 03550, Alicante, Spain
| | - Santiago Negueruela
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández, CSIC-UMH, Av. Santiago Ramón y Cajal s/n, Sant Joan d'Alacant 03550, Alicante, Spain
| | - Camino de Juan Romero
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández, CSIC-UMH, Av. Santiago Ramón y Cajal s/n, Sant Joan d'Alacant 03550, Alicante, Spain
| | - Arturo José Valiño
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández, CSIC-UMH, Av. Santiago Ramón y Cajal s/n, Sant Joan d'Alacant 03550, Alicante, Spain
| | - Salvador Sala
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández, CSIC-UMH, Av. Santiago Ramón y Cajal s/n, Sant Joan d'Alacant 03550, Alicante, Spain
| | - Ronan DaSilva
- Institut de Recherches Cliniques de Montréal (IRCM), 110, ave. des Pins Ouest, Montréal, QC H2W 1R7, Canada
| | - Artur Kania
- Institut de Recherches Cliniques de Montréal (IRCM), 110, ave. des Pins Ouest, Montréal, QC H2W 1R7, Canada; Division of Experimental Medicine, Department of Anatomy and Cell Biology, McGill University, Montréal, QC H3A 2B2, Canada
| | - Víctor Borrell
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández, CSIC-UMH, Av. Santiago Ramón y Cajal s/n, Sant Joan d'Alacant 03550, Alicante, Spain
| | - Luis M Martinez
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández, CSIC-UMH, Av. Santiago Ramón y Cajal s/n, Sant Joan d'Alacant 03550, Alicante, Spain
| | - Lynda Erskine
- School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, Scotland, UK
| | - Eloísa Herrera
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández, CSIC-UMH, Av. Santiago Ramón y Cajal s/n, Sant Joan d'Alacant 03550, Alicante, Spain.
| |
Collapse
|